ArticlePDF AvailableLiterature Review

Metabolic and Cardiovascular Complications of Highly Active Antiretroviral Therapy for HIV Infection

Authors:

Abstract and Figures

Highly active antiretroviral therapy (HAART) regimens, especially those including protease inhibitors have been shown to cause, in a high proportion of HIV-infected patients, a metabolic syndrome (lipodystrophy/lipoatrophy, dyslipidemia, type 2 diabetes mellitus, insulin resistance) that may be associated with an increased risk of cardiovascular disease. A careful stratification of the cardiovascular risk of HIV-infected patients under HAART is needed according to the most recent clinical guidelines.
Content may be subject to copyright.
Current HIV Research, 2006, 4, 79-85 79
1570-162X/06 $50.00+.00 © 2006 Bentham Science Publishers Ltd.
Metabolic and Cardiovascular Complications of Highly Active Antiretroviral
Therapy for HIV Infection
Giuseppe Barbaro*
Department of Medical Pathophysiology, University “La Sapienza”, Rome, Italy
Abstract: Highly active antiretroviral therapy (HAART) regimens, especially those including protease
inhibitors have been shown to cause, in a high proportion of HIV-infected patients, a metabolic syndrome
(lipodystrophy/lipoatrophy, dyslipidemia, type 2 diabetes mellitus, insulin resistance) that may be associated
with an increased risk of cardiovascular disease. A careful stratification of the cardiovascular risk of HIV-
infected patients under HAART is needed according to the most recent clinical guidelines.
Keywords: Human immunodeficiency virus, highly active antiretroviral therapy, nucleoside reverse transcriptase inhibitors,
protease inhibitors, metabolic syndome, cardiovascular disease.
INTRODUCTION
The introduction of highly active antiretroviral therapy
(HAART) has significantly modified the course of human
immunodeficiency virus (HIV) disease, with longer survival
and improved quality of life in HIV-infected subjects.
However, HAART regimens, especially those including
protease inhibitors (PIs) have shown to cause in a high
proportion of HIV-infected patients metabolic
(dyslipidemia,insulin-resistance) and somatic (lipodystrophy
/lipoatrophy) changes that in the general population are
associated with an increased risk of cardiovascular disease
(coronary artery disease and stroke), producing an intriguing
clinical scenery.
The purpose of this review is to analyse the metabolic
and cardiovascular complications of drugs used to treat HIV
infections and to define the clinical guidelines for stratifying
the cardiovascular risk and cardiovascular monitoring of
HIV-infected patients receiving HAART.
HAART-ASSOCIATED METABOLIC SYNDROME
Molecular Mechanisms
The development of HAART-associated metabolic
syndrome is complex and a number of factors are involved,
including direct effects of HAART on lipid metabolism,
endothelial and adipocyte cell function, and mitochondria.
PIs-Associated Somatic and 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 has been
hypothesized, although without strong experimental support,
that this homology may allow PIs to interfere with these
proteins, which may be the cause of the metabolic and
somatic alterations that develop in PIs-treated patients
*Address correspondence to this author at the Viale Anicio Gallo 63,
00174 Rome, Italy; Tel/Fax: +39-6-7102889; E-mail: g.barbaro@tin.it
[13,14]. The hypothesis is that PIs inhibit CRABP-1-
modified and cytochrome P450-3A-mediated synthesis of
cis-9-retinoic acid and peroxisome proliferator-activated
receptor type-gamma heterodimer. The inhibition increases
the rate of apoptosis of adipocytes and reduces the rate at
which pre-adipocytes differentiate into adipocytes, with the
final effect of reducing triglyceride storage and increasing
lipid release. PIs-binding to LRP would impair hepatic
chylomicron uptake and endothelial triglyceride clearance,
resulting in hyperlipidemia and insulin resistance [13,14].
Ultrastructural analysis of adipocytes in PI-induced
lipodystrophy reveals changes including disruption of cell
membranes, fragmented cytoplasmic rims, irregular cell
outlines, and eventually fat droplets laying free in the
connective tissue, with macrophages around them. Many
adipocytes show variable compartmentalization of fat
droplets with decrease in cell size and abundant,
mitochondria-rich cytoplasm. These findings suggest that
HAART-associated lipodystrophy may be the result of
adipocyte remodeling involving variable combinations of
apoptosis, defective lipogenesis, and increased metabolic
activity in different adipose areas of the body [14].
Vernochet et al. assessed the effect of six different PIs on
the differentiation of cells from four clonal lines. They also
studied the capacity of ritonavir to accumulate both into
drug-sensitive and drug-resistant cultured adipocytes [52]. In
this study adipocyte differentiation of mouse 3T3-F442A,
3T3-L1 and Ob1771 cells as well as embryonic stem cells
were investigated at pharmacological concentrations of
indinavir, saquinavir, ritonavir, amprenavir, nelfinavir and
lopinavir [52]. The authors used a sensitive ELISA to
determine intracellular concentration of ritonavir from 3T3-
L1 and Ob1771 preadipocytes. Nelfinavir and lopinavir
inhibited adipocyte differentiation whereas amprenavir was
ineffective. Indinavir, saquinavir and ritonavir inhibited
differentiation of 3T3-L1 and 3T3-F442A cells but did not
alter differentiation of either Ob1771 or embryonic stem
cells. The authors showed that ritonavir accumulated in
preadipocytes and fully differentiated 3T3-L1 adipocytes as a
function of its extracellular concentration [52]. Although
Ob1771 cells were resistant and 3T3-L1 cells were sensitive
to ritonavir, the drug accumulated to similar levels in both
cases. According to the results of this experimetal study PIs
80 Current HIV Research, 2006, Vol. 4, No. 1 Giuseppe Barbaro
inhibit adipocyte differentiation depending on the cell model
used. Ritonavir seems to accumulate into preadipocytes and
adipocytes, suggesting a direct effect on intracellular targets.
However, intracellular accumulation was clearly not
sufficient as Ob1771 cells remained resistant to the
inhibitory effect of ritonavir [52].
Some data indicate that PIs-associated dyslipidemia may
be caused, at least in part, either by PIs-mediated inhibition
of proteasome activity and accumulation of the active
portion of sterol regulatory element-binding protein
(SREBP)-1c in liver cells and adipocytes [40], or by apo C-
III polymorphisms in HIV-infected patients [19]. Bonnet et
al. have described a two- to three- fold increase in apo-E and
apo C-III, essentially recovered as associated to apo B-
containing lipoparticles [5]. In this study, multivariate
analysis revealed that, among the investigated parameters,
apo C-III was the only parameter strongly associated with
the occurrence of lipodystrophy [5].
Sequence homologies have been described between HIV
protease and human site-1 protease (S1P), which activates
SREBP-1c and SREBP-2 pathways. A polymorphism in the
S1P/SREBP-1c gene confers a difference in risk for
development of an increase in total cholesterol with PIs
therapy. This suggests the presence of a genetic
predisposition to hyperlipoproteinemia in PIs-treated
patients [9]. There is also evidence that PIs directly inhibit
the uptake of glucose in insulin-sensitive tissues, such as fat
and skeletal muscle, by selectively inhibiting the glucose
transporter Glut4 [41].
Tumor Necrosis Factor-Alpha (TNF-alpha) and
Lipodystrophy
The relationship between the degree of insulin resistance
and levels of soluble type-2-TNF-alpha receptor suggests
that an inflammatory stimulus may contribute to the
development of HIV-associated lipodystrophy [42]. TNF-
alpha activates 11-beta-hydroxysteroid dehydrogenase type-1,
which converts inactive cortisone to active cortisol. The
activity of this enzyme is higher in visceral fat compared to
subcutaneous fat. Visceral fat is able to locally produce
cortisol which could act inside adipocytes and increase lipid
accumulation [25].
Mitochondrial Dysfunction and Lipodystrophy
There is evidence for nucleoside-induced mitochondrial
dysfunction in HIV-infected patients treated with nucleoside-
containing HAART because lipodystrophy with peripheral
fat wasting is associated with a decrease in subcutaneous
adipose tissue mitochondrial DNA content [8]. This effect
has been especially described with the use of stavudine and
was correlated with the length of exposure to this drug [47].
HAART regimens with didanosine plus stavudine are more
likely to produce a greater increase in serum lactate and
lipodystrophy than therapies based on zidovudine plus
lamivudine within the first year of therapy [8]. Sobstitution
of stavudine with abacavir or zidovudine improves
mitochondrial indices and fat apoptosis in the setting of
lipoatrophy [38].
Adipocytokines and HAART-Associated Metabolic
Syndrome
Adipocytes secrete a range of adipocytokines which
control insulin sensitivity [53]. There is evidence that an
adipocytokine, adiponectin, a protein product of the apM1
gene, which is expressed exclusively in adipocytes [30],
plays a role in development of HIV-associated
lipodystrophy.
In vitro and animal studies and cross-sectional studies in
humans have shown that adiponectin is inversely correlated
with features of this metabolic syndrome including obesity,
insulin resistance, type 2 diabetes, and coronary heart
disease, as well as congenital and acquired lipodystrophies
in non-HIV infected subjects. This syndrome has recently
been linked to a quantitative trait locus on chromosome
3q27, the location of the apM1 gene [1].
In a study of 112 HIV-infected patients receiving
HAART, adiponectin was significantly correlated with
triglycerides, abdominal visceral fat, extremity fat, insulin
resistance, nucleoside-reverse transcriptase inhibitors
(NRTIs) use, and high density lipoprotein (HDL) cholesterol
levels using bivariate analysis [1]. The association of
adiponectin with insulin resistance became nonsignificant
after adjusting for NRTIs use, suggesting that changes in
adiponectin levels may underlie the worsening effect of
NRTIs use on insulin resistance. This latter effect has been
recently correlated to the length of exposure to NRTIs (odds
ratio: 1.08 for each additional year of exposure to NRTIs)
[7]. The associations of adiponectin with triglycerides and
insulin resistance were also slightly weakened after adjusting
for visceral and extremity fat, indicating that adiponectin
may, in part, mediate the effect of lipodystrophy on
triglycerides and insulin resistance in HIV-infected patients
receiving HAART [1].
In a study of 131 consecutive HIV-infected males under
PIs-based HAART insulin sensitivity correlated positively
with adiponectin and negatively with leptin and interleukin-
6 [53]. Insulin resistance, metabolic defects and
cardiovascular risk markers were strongly correlated with the
adiponectin/leptin ratio, suggesting that adiponectin is
related to lipodystrophy, insulin resistance and metabolic
alterations in HIV-infected patients under PIs-based HAART
and that adiponectin/leptin ratio could predict insulin
sensitivity and potential cardiovascular risk in these patients
[53]. Similar results have been reported also in a cross-
sectional study by Kosmiski et al. [35]. In this study
adiponectin levels were significantly and positively
correlated with insuline sensitivity after adjustment for fat
mass and adiponectin level was also an independent
determinant of insuline sensitivity [35].
HAART and Endothelial Dysfunction
In vitro data suggest that some HAART regimens, such
as those including zidovudine, some non-nucleoside reverse
transcriptase inhibitors (e.g. efavirenz) and PIs disrupt
endothelial cell junctions and cytoskeleton actin of the
endothelial cells leading to endothelial dysfunction [20].
These findings are in agreement with those previously
reported In vivo by Stein et al. [48] and in vitro by Zhong et
al. [56].
Metabolic and Cardiovascular Complications of HAART Current HIV Research, 2006, Vol. 4, No. 1 81
Fig. (1). Prevalence of the principal clinical and laboratory findings observed in HIV-infected patients after the introduction of
HAART compared to the pre-HAART period.
CLINICAL FINDINGS AND THERAPEUTIC
GUIDELINES
The prevalence of the principal clinical and laboratory
findings observed in HIV-infected patients after the
introduction of HAART compared to the pre-HAART period
is represented in Fig. 1.
Fat redistribution. HIV-associated lipodystrophy or
lipoatrophy, unreported before the introduction of HAART,
was first described in 1998 [13]. It is characterized by the
presence of a dorsocervical fat pad (also known as buffalo
hump), increased abdominal girth and breast size,
lipoatrophy of subcutaneous fat of the face, buttocks and
limbs, and prominence of veins on the limbs. The overall
prevalence of at least one physical abnormality is thought to
be about 50% in otherwise healthy HIV-infected patients
receiving HAART, although reported rates range from 18%
to 83% [26]. Differences in rates might be influenced by age,
sex, the type and duration of antiretroviral therapy, and the
lack of an objective and validated case definition [26]. As in
genetic lipodystrophy syndromes [23], fat redistribution may
precede the development of metabolic complications in HIV-
infected patients receiving HAART. The severity of these
metabolic abnormalities increases with increasing severity of
lipodystrophy, and they are associated with a raised risk of
cardiovascular events: approximately 1.4 cardiac events per
1000 years of therapy according to the Framingham score
[26]. There is no clinically proven therapy for any feature of
lipodystrophy [10]. One approach to treatment of
lipodystrophy in patients treated with PIs is switching to
PIs-free combination regimens. Although large randomized
trials are lacking, some favourable effects have been shown
[11,15,45].
Dyslipidemia. Increased serum total and low density
lipoprotein (LDL) cholesterol and triglyceride levels have
been observed in about 70% of HIV-infected patients with
lipodystrophy [10]. Guidelines for the management of
dyslipidemias in the general population, such as those of the
National Cholesterol Education Program [18,18] also
represent the basis for therapeutic recommendations in HIV-
infected individuals, as reported by the HIV Medicine
Association of the Infectious Disease Society of America and
Adult AIDS Clinical Trial Group [31] and by the Pavia
Consensus Statement [54].
Fibrates and statins can lower HAART-associated
increases in cholesterol and triglyceride levels [31], although
further data on potential interactions between these drugs and
PIs are needed. Most statins are metabolized through the
CYP3A4 pathway and, therefore, inhibition of CYP3A4 by
PIs could lead to notably raised concentrations of statins,
thus increasing the risk of skeletal muscle or hepatic toxic
effects. The statins least influenced by the CYP3A4
metabolic pathway are pravastatin and fluvastatin [31,54].
Fibrates are unlikely to have significant interactions with
PIs, since their principal metabolic pathway is CYP4A.
Omega-3-fatty acids may be helpful, even though not already
tested in this subset of patients.
As for lipodystrophy, an option in the treatment of
dyslipidemia in patients receiving PIs-including HAART is
to switch to a combination regimen without PIs [11,15,45].
Atazanavir, a novel azapeptide PI, seems not to be associated
with significant dyslipidaemia and insulin resistance as seen
with other PIs [44]. The results of a phase 2 randomized trial
that compared lipid changes after 32 weeks of therapy with
atazanavir with those with nelfinavir (each in combination
with stavudine and lamivudine) showed that levels of total
cholesterol and LDL cholesterol increased significantly more
among patients who used nelfinavir (+24% and +28%,
respectively) than among those who used atazanavir (+4%
and +1%, respectively) [27]. However, these data need to be
confirmed by further controlled prospective trials. Of interest
are data indicating that patients never treated with HAART,
who started a PIs-sparing regimen including nevirapine
showed a significant increase of HDL-cholesterol [51].
82 Current HIV Research, 2006, Vol. 4, No. 1 Giuseppe Barbaro
Table 1. Cardiovascular Risk Stratification of HIV-Infected Patients Receiving Haart [54]
1. All HIV-infected patients should have cardiovascular risk factors evaluated according to the Framingham score before receiving HAART. The use
of Framingham score and other non-invasive investigations of cardiovascular risk may help in the decision regarding the use of antiretrovirals and
other treatment. Blood tests for preventive cardiology (eg, complete blood count to determine anemia and/or other hematologic abnormalities,
determination of serum electrolytes, assessment of renal and hepatic function, assessment of thyroid function) should be routine before and during
HAART (every 3-6 months).
2. Routine assessment (at least twice a week) of blood pressure in HIV-infected patients is important because these patients seem to be at higher risk of
developing hypertension and of developing it at a younger age than the general population. Predisposing conditions including vasculitis, acquired
glucocorticoid resistance, acute and chronic renal failure, and drug interactions should be carefully assessed.
3. Before HAART is started, lipid profiles should be measured after an 8-12 hour fast to establish a baseline, and the measurements should be repeated
routinely during the HAART therapy. Serum glucose and hemoglobin A1C measurements are especially indicated for patients on HAART.
4. Fasting lipids and glucose should be measured before the initiation of HAART and at regular 3-6 month intervals thereafter. For patients with
elevated triglyceride levels at baseline, lipid measurements should be repeated within 1-2 months of starting HAART.
5. The routine evaluation of coagulation parameters is probably not advisable until the benefit of widespread screening is assessed in prospective
studies. However, clinicians should be aware of the increased risk of coagulative disorders in patients on HAART, especially in those with HAART-
associated metabolic syndrome, and should check coagulative parameters (D-dimer, plasminogen activator inhibitor-1, tissue-type plasminogen
activator antigen, protein S, protein C and antithrombin III) at least once a year in patients on HAART.
6. Elevated plasma homocysteine level is recognized as independent factor for atherosclerosis and cardiovascular disease. It is caused by genetic
variants [homozygous mutations (C677T and A1298C) of the MTHFR gene], malnutrition, drugs or renal failure. Especially when above 10
micromol/l, it can be treated with dietary supplementation of folic acid, vitamin B6, and vitamin B12. Plasma homocysteine level should be checked
at least once a year in HIV-infected patients with at least two major cardiovascolar risk factors and in those who receive PIs.
According to a recent study, lipid profile improves by
replacing PIs with efavirenz, nevirapine or abacavir with
more marked results in non-lipodystrophic patients. In
contrast, this strategy does not seem to be effective for
reversing body fat abnormalities [21]. According to another
study the efavirenz-induced increase in HDL-cholesterol is
influenced by the multidrug resistance gene 1 C3435T
polymorphism [2].
Dysglycemia
Insulin resistance (elevated C-peptide and insulin) and
type 2 diabetes mellitus have been observed in 8% to 10%
of the patients with lipodystrophy [10].In managing glucose
abnormalities in HIV-infected patients receiving HAART, it
is important to remember that some glitazones are
metabolized by the CY3A4 pathway, which could increase
the risk of developing myositis and hepatitis if used
simultaneously with PIs [54]. Although some studies have
shown that the use of glitazones for the treatment of HIV-
associated lipodistrophy or lipoatrophy leads to an
improvement in insulin resistance, contrasting findings
suggest that further work is needed [12].
Hypertension
The prevalence of hypertension in HIV disease has been
estimated to be about 20-25% before the introduction of
HAART [3]. Hypertension is currently considered part of
HAART-associated metabolic syndrome [24]. It appears to
be related to PIs-induced lipodystrophy and metabolic
disorders, especially to elevated fasting triglyceride and
insulin resistance, with a prevalence of up to 74% in patients
with HAART-associated metabolic syndrome [24,46]. When
treating hypertension in HIV-infected patients with HAART-
associated metabolic syndrome, it should be noted that beta-
blockers and diuretics might worsen the metabolic profiles
of these patients. In addition, calcium-channel blockers
should be used with caution, since they may interact with
PIs. No interactions between NRTIs and nucleotide reverse
transcriptase inhibitors and antiarrhythmics, statins or
anticoagulants diseases have, however, been documented.
Angiotensin-converting-enzyme inhibitors and angiotensin
II-receptor blockers might be considered, but data from
controlled clinical trials assessing these drugs in this subset
of patients are lacking.
HAART AND RISK OF CARDIOVASCULAR
DISEASE
Coronary heart disease. HIV-infected patients with pre-
existing additional risk factors (e.g. hypertension, diabetes
or increased plasma homocysteine levels) might be at raised
risk of developing coronary heart disease because of
accelerated atherosclerosis. Cardiovascular risk stratification
of HIV-infected patients receiving HAART according to the
Pavia Consensus Statement [54] has been reported in Table
1. Conflicting data exist, however, on the relationship
between HAART and the incidence of acute coronary
syndromes, such as unstable angina or myocardial infarction,
among HIV-infected patients receiving PIs-containing
HAART [4,6,22,32,37]. Differences in the study design,
selection of the patients and statistical analyses might
explain this disparity. However, longer exposure to HAART
and/or PIs seem to increase the risk of myocardial infarction.
The results of the Data Collection on Adverse Events of
Anti-HIV Drugs study showed that HAART therapy is
associated with a 26% relative risk increase in the rate of
myocardial infarction per year of HAART exposure [22].
Peripheral vascular disease. Also the issue of surrogate
markers of subclinical atherosclerosis has been addressed. A
study was performed on a cohort of 168 HIV-infected
patients to measure the intima-media thickness (IMT) and
Metabolic and Cardiovascular Complications of HAART Current HIV Research, 2006, Vol. 4, No. 1 83
assess indirectly the cardiovascular risk. In this population a
high prevalence of atherosclerotic plaques within the femoral
or carotid arteries was observed, but their presence was not
associated with the use of PIs [17]. Similar results were
reported also by Hsue et al. [33] and by Currier et al. [16] in
case-control studies suggesting that traditional risk factors
may contribute to atherosclerosis in HIV-infected patients
independently of PIs exposure.Different results were reported
by Maggi et al. who observed a higher than expected
prevalence of premature carotid lesions in PIs-treated patients
compared to PIs-naive patients [36]. The impact of
individual measures to reduce the cardiovascular risk and the
progression of atherosclerosis has been addressed by
Thiebault et al. [50]. According to these authors, the
increased use of lipi-lowering agents, of PIs-free HAART
regimes and the reduction of smoking may decrease the IMT
in HIV-infected patients over time [50]. In spite of these
different results, markers of subclinical atherosclerosis
should be carefully assessed in HIV-infected patients
receiving HAART, especially in those with lipodystrophy
[54].
Vasculitis
Drug-induced hypersensitivity vasculitis is common in
HIV-infected patients receiving HAART. The vasculitis
associated with drug reactions typically involves small
vessels and has a lymphocytic or leukocytoclastic
histopathology. The pathologic mechanisms include T-cell
recognition of haptenated proteins or the deposition of
immune complexes in blood-vessel walls. Medical
practitioners need to be especially aware of abacavir
hypersensitivity reactions because of the potential for fatal
outcomes. Hypersensitivity reactions of this type should
always be considered as a possible etiology for a vasculitic
syndrome in an HIV-infected patient [34].
Coagulation Disorders
HIV-infected patients, especially those with fat
redistribution, might develop coagulation abnormalities,
including increased levels of fibrinogen, D-dimer,
plasminogen activator inhibitor-1, and tissue-type
plasminogen activator antigen, or deficiency of protein S
[29,55]. For instance, protein S deficiency has been reported
in up to 73% of HIV-infected men [29,55]. These
abnormalities have been associated with thromboses
involving veins and arteries and seem to be related to
HAART regimens that include PIs [49]. Thrombocytosis has
been reported in 9% of patients receiving HAART, with
cardiovascular complications in up to 25% of cases [39].
CONCLUSIONS
HAART-associated metabolic syndrome is an
increasingly recognized clinical entity. A better
understanding of the molecular mechanisms responsible for
this syndrome will enable the development of new drugs
with reduced metabolic and cardiovascular side effects.
Careful cardiac screening is warranted for patients who are
being evaluated for, or who are receiving, HAART regimens,
particularly for those with known underlying cardiovascular
risk factors. The atherogenic effects of PIs-containing
HAART might synergistically promote the development of
coronary or cerebrovascular disease and increase the risk of
death from myocardial infarction or stroke even in young
HIV-infected people. A close collaboration between
cardiologists and infectious disease specialists is needed for
management decisions regarding the use of antiretrovirals
and other therapies, for a careful stratification of the
cardiovascular risk and cardiovascular monitoring according
to the most recent clinical guidelines.
REFERENCES
[1] Addy CL, Gavrila A, Tsiodras S, Brodovicz B, Karchmer AW,
Mantzoros CS. (2003). Hypoadiponectinemia is associated with
insulin resistance,hypertriglyceridemia, and fat redistribution in
human immunodeficiency virus-infected patients treated with
highly active antiretroviral therapy. Journal of Clinical
Endocrinology and Metabolism 88: 627-636.
[2] Alonso-Villaverde C, Coll B, Gomez F, Parra S, Camps J, Joven J,
Masana L. (2005). The efavirenz-induced increase in HDL-
cholesterol is influenced by the multidrug resistance gene 1
C3435T polymorpism. AIDS 19: 341-342.
[3] Aoun S, Ramos E. (2000). Hypertension in the HIV-infected
patient. Current Hypertension Reports 2: 478-481.
[4] Barbaro G, Di Lorenzo G, Cirelli A, Grisorio B, Lucchini A,
Hazra C, Barbarini G. (2003). An open-label, prospective,
observational study of the incidence of coronary artery disease in
patients with HIV receiving highly active antiretroviral therapy.
Clinical Therapeutics 25: 2405-2418.
[5] Bonnet E, Ruidavets JB, Tuech J, Ferrieres J, Collet X, Fauvel J,
Massip P, Perret B. (2001). Apoprotein c-III and E-containing
lipoparticles are markedly increased in HIV-infected patients
treated with protease inhibitors: association with the development
of lipodystrophy. Journal of Clinical Endocrinology and
Metabolism 86: 296-302.
[6] Bozzette SA, Ake CF, Tam HK, Chang SW, Louis TA. (2003).
Cardiovascular and cerebrovascular events in patients treated for
human immunodeficiency virus infection. New England Journal
of Medicine 348: 702-710.
[7] Brown T, Li X, Cole S, Kingsley L, Palella F, Riddler S, Chmiel J,
Visscher B, Margolick J, Dobs A. (2005). Cumulative exposure to
nucleoside analogue reverse transcriptase inhibitors is associated
with insulin resistance markers in the Multicenter AIDS Cohort
Study. AIDS 19: 1375-1383.
[8] Caron M, Auclair M, Lagathu C, Lombes A, Walker U,
Kornprobst M, Capeau J. (2004). The HIV-1 nucleoside reverse
transcriptase inhibitors stavudine and zidovudine alter adipocyte
functions In vitro. AIDS 18: 2127-2136.
[9] Caron M, Auclair M, Sterlingot H, Kornprobst M, Capeau J.
(2003). Some HIV protease inhibitors alter lamin A/C maturation
and stability, SREBP-1 nuclear localization and adipocyte
differentiation. AIDS 17: 2437-2444.
[10] Carr A. (2003). HIV lipodystrophy: risk factors,pathogenesis,
diagnosis and management. AIDS 17 (S1): S141-S148.
[11] Carr A, Hudson J, Chuan J, Mallal S, Law M, Hoj J, Doong N,
French M, Smith D, Cooper DA. (2001). HIV protease inhibitor
substitution in patients with lipodystrophy: a randomized,
controlled, open-label, multicentre study. AIDS 15: 1811-1822.
[12] Carr A, Workman C, Carey D, Rogers G, Martin A, Baker D,
Wand H, Law M, Samaras K, Emery S, Cooper DA, Rosey
investigators. (2004). No effect of rosiglitazone for treatment of
HIV-1 lipoatrophy: randomised, double-blind, placebo-controlled
trial. Lancet 363: 429-438.
[13] Carr A, Samaras K, Burton S, Law M, Freund J, Chisholm DJ,
Cooper DA. (1998). A syndrome of peripheral lipodystrophy,
hyperlipidaemia and insulin resistance in patients receiving HIV
protease inhibitors. AIDS 12: F51-F58.
[14] Carr A, Samaras K, Chisholm DJ, Cooper DA. (1998).
Pathogenesis of HIV-1-protease inhibitor-associated peripheral
lipodystrophy, hyperlipidaemia, and insulin resistance. Lancet
351: 1881-1883.
84 Current HIV Research, 2006, Vol. 4, No. 1 Giuseppe Barbaro
[15] Clumeck N, Goebel F, Rozenbaum W, Gerstoft J, Staszewski S,
Montaner J, Johnson M, Gazzard BG, Stone C, Athisegaran R,
Moore S. (2001). Simplification with abacavir-based triple
nucleoside therapy versus continued protease inhibitor-based
highly active antiretroviral therapy in HIV-1-infected patients
with undetectable plasma HIV-1 RNA. AIDS 15: 1517-1526.
[16] Currier JS, Kendall MA, Zackin R, Henry WK, Alston-Smith B,
Torriani F, Schouten J, Mickelberg K, Li Y, Hodis HN for the
AACTG 5078 Study Team. (2005). Carotid artery intima-media
thickness and HIV infection: traditional risk factors overshadow
impact of protease inhibitor exposure. AIDS 19: 927-933.
[17] Depairon M, Chessex S, Sudre P, Rodondi N, Doser N, Chave JP,
Riesen W, Nicod P, Darioli R, Telenti A, Mooser VwtSHCS.
(2001). Premature atherosclerosis in HIV-infected individuals:
focus on protease inhibitor therapy. AIDS 15: 329-334.
[18] Executive summary of the 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). Journal of the American
Medical Association 285: 2486-2497.
[19] Fauvel J, Bonnet E, Ruidavets JB, Ferrières J, Toffoletti A, Massip
P, Chap H, Perret B. (2001). An interaction between apo C-III
variants and protease inhibitors contributes to high tiglyceride/low
HDL levels in treated HIV patients. AIDS 15: 2397-2406.
[20] Fiala M, Murphy T, MacDougall J, Yang W, Luque A, Iruela-
Arispe L, Cashman J, Buga G, Byrns RE, Barbaro G, Arthos J.
(2004). HAART drugs induce mitochondrial damage and
intracellular gaps and gp120 causes apoptosis. Cardiovascular
Toxicology 4: 327-338.
[21] Fisac C, Fumero E, Crespo M, Roson B, Ferrer E, Virgili N,
Ribera E, Gatell JM, Podzamczer D. (2005). Metabolic benefits
24 months after replacing a protease inhibitor with
abacavir,efavirenz or nevirapine. AIDS 19: 917-925.
[22] Friis-Moller N, Weber R, Reiss P, Thiebaut R, Kirk O, D'Arminio
Monforte A, Pradier C, Morfeldt L, Mateu S, Law M, El-Sadr W,
De Witt S, Sabin CA, Phillips AN, Lundgren JD for the DAD
Study Group. (2003). Cardiovascular risk factors in HIV patients-
association with antiretroviral therapy. Results from DAD study.
AIDS 17: 1179-1193.
[23] Garg A. (2005). Acquired and inherited lipodystrophies. New
England Journal of Medicine 350: 1220-1234.
[24] Gazzaruso C, Bruno R, Garzaniti A, Giordanetti S, Fratino P,
Sacchi P, Filice G. (2003). Hypertension among HIV patients:
prevalence and relationship to insulin resistance and metabolic
syndrome. Journal of Hypertension 21: 1377-1382.
[25] Gougeon ML, Penicaud L, Fromenty B, Leclercq P, Viard JP,
Capeau J. (2004). Adipocytes targets and actors in the
pathogenesis of HIV-associated lipodystrophy and metabolic
alterations. Antiviral Therapy 9: 161-177.
[26] Grinspoon S, Carr A. (2005). Cardiovascular risk and body-fat
abnormalities in HIV-infected adults. New England Journal of
Medicine 352: 48-62.
[27] Grover SA, Coupal L, Gilmore N, Mukherjee J. (2005). Impact of
dyslipidemia associated with highly active antiretroviral therapy
(HAART) on cardiovascular risk and life expectancy. American
Journal of Cardiology 95: 586-591.
[28] Hadigan C, Yawetz S, Thomas A, Havers F, Sax PE, Grinspoon S.
(2004). Metabolic effects of rosiglitazone in HIV lipodystrophy: a
randomized, controlled trial. Annals of Internal Medicine 140:
786-794.
[29] Hadigan C, Meigs JB, Rabe J, D'Agostino RB, Wilson PW,
Lipinska I, Tofler GH, Grinspoon S. (2001). Increased PAI-1 and
tPA Antigen Levels Are Reduced with Metformin Therapy in
HIV-Infected Patients with Fat Redistribution and Insulin
Resistance. Journal of Clinical Endocrinology and Metabolism 86:
939-943.
[30] Haque WA, Shimomura I, Matsuzawa Y, Garg A. (2002). Serum
adiponectin and leptin levels in patients with lipodystrophies.
Journal of Clinical Endocrinology and Metabolism 87: 2395-2398.
[31] HIV Medicine Association of the Infectious Disease Society of
America and the Adult AIDS Clinical Trial Groups. (2004).
Guidelines for the Evaluation and Management of Dyslipidemia in
Human Immunodeficiency Virus (HIV)-Infected Adults
Receiving Antiretrovial Therapy: Recommendations of the HIV
Medicine Association of the Infectious Disease Society of
America and the Adult AIDS Clinical Trial Groups. Clinical
Infectious Diseases 37: 613-627.
[32] Holmberg SD, Moorman AC, Williamson JM, Tong TC, Ward DJ,
Wood KC, Greenberg AE, Janssen RS and the HIV Outpatient
Study (HOPS) Investigators. (2002). Protease inhibitors and
cardiovascular outcomes in patients with HIV-1. Lancet 360:
1747-1748.
[33] Hsue PY, Lo JC, Franklin A, Bolger AF, Martin JN, Deeks SG,
Waters DD. (2004). Progression of atherosclerosis as assessed by
carotid intima-media thickness in patients with HIV infection.
Circulation 109: 1603-1608.
[34] Johnson RM, Barbarini G, Barbaro G. (2003). Kawasaki-like
syndromes and other vasculitic syndromes in HIV-infected
patients. AIDS 17 (S1): S77-S82.
[35] Kosmiski L, Kuritzkes D, Lichtenstein K, Eckel R. (2003).
Adipocyte-derived hormone levels in HIV lipodystrophy.
Antiviral Therapy 8: 9-15.
[36] Maggi P, Serio G, Epifani G, Fiorentino G, Saracino A, Fico C,
Perilli F, Lillo A, Ferraro S, Gargiulo M, Chirianni A, Angarano
G, Regina G, Pastore G. (2000). Premature lesions of the carotid
vessels in HIV-1-infected patients treated with protease inhibitors.
AIDS 14: F123-F128.
[37] Mary-Krause M, Cotte L, Simon A, Partisani M, Costagliola D and
the Clinical Epidemiology Group from the French Hospital
Database. (2003). Increased risk of myocardial infarction with
duration of protease inhibitor therapy in HIV-infected men. AIDS
17: 2479-2486.
[38] McComsey GA, Paulsen D, Lonergan JT, Hessenthaler SM,
Hoppel CL, Williams VC, Fisher RL, Cherry CL, White-Owen C,
Thompson KA, Ross ST, Hernandez JE, Ross LL. (2005).
Improvements in lipoatrophy, mitochondrial DNA levels and fat
apoptosis after replacing stavudine with abacavir or zidovudine.
AIDS 19: 15-23.
[39] Miguez-Burbano MJ, Burbano X, Rodriguez A, Lecusay R,
Rodriguez N, Shor-Posner G. (2002). Development of
thrombocytosis in HIV+ drug users: impact of antiretroviral
therapy. Platelets 13: 183-185.
[40] Mooser V, Carr A. (2001). Antiretroviral therapy-associated
hyperlipidemia in HIV disease. Current Opinion in Lipidology 12:
313-319.
[41] Murata H, Hruz PW, Mueckler M. (2000). The mechanism of
insulin resistance caused by HIV protease inhibitor therapy.
Journal of Biological Chemistry 275: 20251-20254.
[42] Mynarcik DC, McNurlan MA, Steigbigel RT, Fuhrer J, Gelato
MC. (2000). Association of severe insulin resistance with both loss
of limb fat and elevated serum tumor necrosis factor receptor
levels in HIV lipodystrophy. Journal of Acquired Immune
Deficiency Syndromes 25: 312-321.
[43] Nair R, Robbs JV, Chetty R, Naidoo NG, Woolgar J. (2000).
Occlusive arterial disease in HIV-infected patients: a preliminary
report. European Journal of Vascular and Endovascular Surgery
20: 353-357.
[44] Piliero PJ. (2002). Atazanavir: a novel HIV-protease inhibitor.
Expert Opinion on Investigational Drugs 11: 1295-301.
[45] Ruiz L, Negredo E, Domingo P, Paredes R, Francia E, Balague M,
Gel S, Bonjoch A, Fumaz CR, Johnston S, Romeu J, Lange J,
Clotet B. (2001). Antiretroviral treatment simplification with
nevirapine in protease inhibitor-experienced patients with hiv-
associated lipodystrophy: 1-year prospective follow-up of a
multicenter, randomized, controlled study. Journal of Acquired
Immune Deficiency Syndromes 27: 229-236.
[46] Sattler FR, Qian D, Louie S, Johnson D, Briggs W, DeQuattro V,
Dube MP. (2001). Elevated blood pressure in subjects with
lipodystrophy. AIDS 15: 2001-2010.
[47] Seminari E, Tinelli C, Minoli L, Sacchi P, Filice G, Zocchetti C,
Meneghetti G, Bruno R, Maserati R. (2002). Evaluation of the risk
factors associated with lipodystriphy development in a cohort of
HIV-positive patients. Antiviral Therapy 7: 175-180.
[48] Stein JH, Klein MA, Bellehumeur JL, Mc Bride PE, Wiebe DA,
Otvos JD, Sosman JM. (2001). Use of human immunodeficiency
virus-1 protease inhbitors is associated with atherogenic
lipoprotein changes and endothelial dysfunction. Circulation 104:
257-262.
[49] Sullivan PS, Dworkin MS, Jones JL, Hooper WC. (2000).
Epidemiology of thrombosis in HIV-infected individuals. The
Adult/Adolescent Spectrum of HIV Disease Project. AIDS 14:
321-324.
[50] Thiebaut R, Aurillac-Lavignole V, Bonnet F, Ibrahim N, Cipriano
C, Neau D, Dupon M, Dabis F, Mercie P and the Group
Metabolic and Cardiovascular Complications of HAART Current HIV Research, 2006, Vol. 4, No. 1 85
d'Epidemiologie Clinique du Sida en Aquitaine (GECSA). (2005).
Change in atherosclerosis progression in HIV-infected patients:
ANRS Aquitane Cohort, 1999-2004. AIDS 19: 729-731.
[51] van der Valk M, Kastelein JJP, Murphy RL, van Leth F, Katlama
C, Horban A, Glesby M, Behrens G, Clotet B, Stellato RK,
Molhuizen HOF, Reiss P on behalf of the Atlantic Study Team.
(2001). Nevirapine-containing antiretroviral therapy in HIV-1
infected patients results in an anti-atherogenic lipid profile. AIDS
15: 2407-2414.
[52] Vernochet C, Azoulay S, Duval D, Guedj R, Ailhaud G, Dani C.
(2003). Differential effect of HIV protease inhibitors on
adipogenesis: intracellular ritonavir is not sufficient to inhibit
differentiation. AIDS 17: 2177-2180.
[53] Vigouroux C, Maachi M, Nguyen TH, Coussieu C, Gharakhanian
S, Funahashi T, Matsuzawa Y, Shimomura I, Rozenbaum W,
Capeau J, Bastard JP. (2003). Serum adipocytokines are related to
lipodystrophy and metabolic disorders in HIV-infected men under
antiretroviral therapy. AIDS 17: 1503-1511.
[54] Volberding P, Murphy R, Barbaro G, Barbarini G, Bruno R, Cirelli
A, Currie P, Di Lorenzo G, Fantoni M, Filice G, Galli M, Grisorio
B, Moroni M, Recusani F, Sacchi P, Scevola D, Torre D, Vittecoq
D. (2003). The Pavia Consensus Statement. AIDS 17 (S1): S170-
S179.
[55] Witz M, Lehmann J, Korzets Z. (2000). Acute brachial artery
thrombosis as the initial manifestation of human
immunodeficiency virus infection. American Journal of
Hematology 64: 137-139.
[56] Zhong DS, Lu XH, Conklin BS, Lin PH, Lumsden AB, Yao Q,
Chen C. (2002). HIV protease ritonavir induces cytotoxicity of
human endothelial cells. Arteriosclerosis, Thrombosis and
Vascular Biology 22: 1560-1566.
Received: August 3, 2005 Revised: August 26, 2005 Accepted: August 30, 2005
... Public health programs, especially the HIV/AIDS, Tuberculosis and Malaria have the greatest risk of ADRs because millions of people are treated with a wide range of drugs, some of which have serious/life threatening adverse reactions [35][36][37][38]. In the western countries where ARVs have been used for many years, cases of rising obesity, weight gain and cardio-metabolic diseases are persistently being reported in association with the use of ARVs [39][40]. The demand for drug safety surveillance has therefore become a major consideration in the global scale up of ART to end HIV/AIDS and TB. ...
... Until the SPHAR-TI course, majority (71.0%) of the healthcare workers that participated in the course have never received training in pharmacovigilance but were nonetheless working in public health institutions or hospitals directly treating HIV/ AIDS, Tuberculosis and Malaria through the respective public health disease control programs. Some studies have reported high prevalence of ADRs emanating from the HIV/AIDS, TB and Malaria public health programs [13,35,37,38,39,40], thus, justifying the SPHARTI model. ...
... Malawi has a worse situation; there are 12 million people but only 350 medical doctors are available to cater for all the health needs including the reporting of ADRs [68]. Nigeria appears to have the highest density of healthcare workers in Africa [37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54] but the large population size and the lack of capacity for reporting ADRs are major constrains. However, the training of healthcare workers as has been shown in several studies can improve the reporting of ADRs. ...
Article
Full-text available
Background: Adverse Drug Reactions (ADRs) are a major clinical and public health problem world-wide. The prompt reporting of suspected ADRs to regulatory authorities to activate drug safety surveillance and regulation appears to be the most pragmatic measure for addressing the problem. This paper evaluated a pharmacovigilance (PV) training model that was designed to improve the reporting of ADRs in public health programs treating the Human Immunodeficiency Virus (HIV), Tuberculosis (TB) and Malaria. Methods: A Structured Pharmacovigilance and Training Initiative (SPHAR-TI) model based on the World Health Organization accredited Structured Operational Research and Training Initiative (SOR-IT) model was designed and implemented over a period of 12 months. A prospective cohort design was deployed to evaluate the outcomes of the model. The primary outcomes were knowledge gained and Individual Case Safety Reports (ICSR) (completed adverse drug reactions monitoring forms) submitted, while the secondary outcomes were facility based Pharmacovigilance Committees activated and health facility healthcare workers trained by the participants. Results: Fifty-five (98%) participants were trained and followed up for 12 months. More than three quarter of the participants have never received training on pharmacovigilance prior to the course. Yet, a significant gain in knowledge was observed after the participants completed a comprehensive training for six days. In only seven months, 3000 ICSRs (with 100% completeness) were submitted, 2,937 facility based healthcare workers trained and 46 Pharmacovigilance Committees activated by the participants. Overall, a 273% increase in ICSRs submission to the National Agency for Food and Drug Administration and Control (NAFDAC) was observed. Conclusion: Participants gained knowledge, which tended to increase the reporting of ADRs. The SPHAR-TI model could be an option for strengthening the continuous reporting of ADRs in public health programs in resource limited settings.
... [11] Recent studies of HIV-infected persons have revealed a high prevalence of metabolic syndrome among patients receiving HAART. [12][13][14] HAART itself in a high proportion of patients causes metabolic disorders characterized by lipodystrophy, dyslipidaemia and insulin resistance, which may be associated with an increase in coronary artery disease and stroke. [15] The prevention of this cardiometabolic morbidity by using antiretroviral (ARV) drugs with a low metabolic toxicity and the treatment after its early detection are the current control strategies. ...
... [11] Recent studies of HIV-infected persons have revealed a high prevalence of metabolic syndrome among patients receiving HAART. [12][13][14] HAART itself in a high proportion of patients causes metabolic disorders characterized by lipodystrophy, dyslipidaemia and insulin resistance, which may be associated with an increase in coronary artery disease and stroke. [15] The prevention of this cardiometabolic morbidity by using antiretroviral (ARV) drugs with a low metabolic toxicity and the treatment after its early detection are the current control strategies. ...
... With time, there has also been an increase in first-line ART regimens. [5][6][7] Following the WHO guidelines, the first-line anti-retroviral regimen is widely used in Africa. This includes NRTI with an NNRTI. ...
Article
Objective: To determine lipid derangements in HIV-positive patients receiving anti-retroviral therapy (ART). Study Design: Comparative cross-sectional study. Place and Duration of study: Chemical Pathology Department, in cooperation with the Department of Virology at Armed Forces Institute of Pathology (AFIP), Rawalpindi Pakistan, from Mar 2019 to Mar 2020. Methodology: In this study, two groups were made. In Group-1, lipid profile estimation was done in HIV patients without treatment. In contrast, lipid profile estimation was done after one year of treatment with anti-retroviral drugs in Group-2. Results: One hundred and two HIV-positive subjects were included in our study. Of these patients, 88(86.27%) were males,and 14(13.73%) were females. Independent sample t-test revealed a statistically significant difference (p-value<0.05) in serum cholesterol, serum triglycerides, serum high-density lipoprotein (HDL-c), low-density lipoproteins (LDL) and very lowdensity lipoproteins (VLDL) between the two groups. Conclusion: Hyperlipidemia in HIV patients getting anti-retroviral therapy (ART) exposes them to a high risk of coronary artery diseases and myocardial infarction. Hence lipid profile of patients getting ART must be regularly monitored, and hyperlipidemia, if present, should be managed.
... As the life expectancy of PWH increases, there has been an increased prevalence of diabetes, hypertension, dyslipidemia, and other comorbidities predisposing to CVD. 36 Rates of myocardial infarction or coronary heart disease in PWH are higher with approximately 2-fold increased relative risk compared with HIV-negative controls. 7,37-42 This increased risk remained even when controlled for potential confounders such as socioeconomic status or viral load. ...
Article
Full-text available
Background Persons with HIV have a higher prevalence of coronary artery disease compared with their HIV‐negative counterparts. Earlier identification of subclinical atherosclerosis may provide a greater opportunity for cardiovascular disease risk reduction. We investigated coronary cross‐sectional area (CorCSA) by noncontrasted computed tomography imaging as a noninvasive measure of arterial remodeling among virally suppressed persons with HIV. Methods and Results We assessed 105 persons with HIV with a spectrum of cardiometabolic health. All participants underwent computed tomography imaging to assess the mean corCSA of the proximal left anterior descending artery and 28 participants underwent additional coronary computed tomography angiography. Partial Spearman rank correlations adjusted for cardiovascular disease risk factors were used to assess relationships of corCSA with anthropometric measurements, HIV‐related factors, and plasma cytokines. Mean corCSA measured by noncontrast computed tomography and coronary computed tomography angiography were strongly correlated (ρ=0.91, P <0.0001). Higher mean corCSA was present in those with coronary artery calcium ( P =0.005) and it correlated with participants' atherosclerotic cardiovascular disease risk score (ρ=0.35, P =0.01). After adjusting for established cardiovascular disease risk factors, we observed an inverse relationship between corCSA and CD4 ⁺ T‐cell count (ρ=−0.2, P =0.047). Removal of age from the model strengthened the relationships between corCSA and antiretroviral therapy duration (from ρ=0.19, P =0.08 to ρ=0.3, P =0.01). CorCSA was also inversely correlated with plasma IL‐10 (ρ=−0.25, P =0.03) but had no relationship with IL‐6 (ρ=0.11, P =0.4) or IL‐1β (ρ=0.08, P =0.5). Conclusions Positive coronary arterial remodeling, an imaging marker of subclinical atherosclerosis, is associated with a lower CD4 T‐cell count, lower circulating IL‐10, and possibly a longer antiretroviral therapy duration in persons with HIV. Registration Clinicaltrials.gov; Unique identifier: NCT04451980.
... [11] Recent studies of HIV-infected persons have revealed a high prevalence of metabolic syndrome among patients receiving HAART. [12][13][14] HAART itself in a high proportion of patients causes metabolic disorders characterized by lipodystrophy, dyslipidaemia and insulin resistance, which may be associated with an increase in coronary artery disease and stroke. [15] The prevention of this cardiometabolic morbidity by using antiretroviral (ARV) drugs with a low metabolic toxicity and the treatment after its early detection are the current control strategies. ...
... The introduction of antiretroviral therapy (ART) has led to a dramatic declining in acquired immunodeficiency syndrome (AIDS)-associated diseases and fatality, and the condition directly changed from killer to a chronic, controllable infectious disease. 1,2 Nowadays, the standard and recommended treatments for HIV are a combinations of three-drugs that comprise a non-nucleoside reverse transcriptase inhibitor (NNRTI), a protease inhibitor (PI), or an integrase strand transfer inhibitor (INSTI) plus two nucleoside reverse transcriptase inhibitors (NRTIs), typically abacavir/lamivudine (ABC/3TC) or either tenofovir alafenamide/emtricitabine (TAF/FTC) or tenofovir disoproxil fumarate/lamivudine (TDF/3TC). 3,4 Meanwhile, with the introduction of ART, HIV-infected patients have started to live longer; however, some co-morbid conditions have emerged. ...
Article
Full-text available
HIV-infected people have started to live longer since the introduction of antiretroviral therapy, however various co-morbid illnesses have emerged. Three HIV-infected individuals, all at least 43 years old, reported with a new onset of type 2 diabetes after switching to dolutegravir-combined antiretroviral therapy regimen. These three people were switched to integrase strand transfer inhibitor (dolutegravir)-based first-line antiretroviral treatment after receiving non-nucleoside reverse transcriptase inhibitor-combined first-line antiretroviral treatment for at least 6 years, as recommended by the World Health Organization for Sub-Saharan African countries, including Ethiopia.All of the given cases had normal plasma fasting sugar (fasting blood sugar <100 mg/dL) at the time of switching. Polyuria, polydipsia, considerable weight loss, and fatigue were all classified as signs of diabetes mellitus in the two male cases. In addition, their laboratory results demonstrated hyperglycemia (plasma fasting blood sugar > 200 mg/dL and urine glucose level ⩾2+) with no ketonuria after switching to dolutegravir for 4–10 months. A glycemic control was achieved, and metformin medication was continued. After 6 months of dolutegravir treatment, the third female case developed diabetic ketoacidosis and severe hyperglycemia (fasting blood glucose level 600 mg/dL, urine glucose level 3+, and ketonuria 3+). To recover from diabetic ketoacidosis, the patient was given intravenous normal saline and regular insulin. Her glycemic control was then restored, and she was switched to NPH insulin. For all of the cases presented, the dolutegravir-based regimen was maintained. Antiretroviral regimens using dolutegravir have the potential to cause hyperglycemia and other side effects. As a result, blood glucose monitoring is required throughout treatment initiation and regularly throughout treatment follow-up, particularly for those on dolutegravir-combined antiretroviral therapy regimens.
... Overall, the finding shown the existence of contradicted knowledge regarding the typical influences of specific ART regimen with the incidence of MS arose from the drug combination given together as one time dose . Also, it might also dictate the direct impact of exposure to any ART regimens on lipid metabolism, endothelial and adipocyte cell function, and mitochondria dysfunction[42,58], along with the link of the treatment with the incidences of each traits[42,59] of MS, might be the possible reason for the differences encountered. This implicates to initiates further studies with strong design to differentiate the specific effects each drug regimen with MS incidence.On top of that, with regards to the HIV/AIDS-related factors, this study was revealed that the odds of MS much times higher in those PLHIVs with more than or equal to three WHO staging, compared with less than three WHO staging. ...
Preprint
Full-text available
Background Metabolic syndrome (MS) among people living with HIV (PLHIVs) is a global public health issue. However, there is no primary data about predictors of MS in the SSA and Ethiopia. Therefore, the aim of this study was to determine predictors of MS, among PLHIVs in the Gedeo-Zone,Southern-Ethiopia. Methods Unmatched case-control study approach, among PLHIVs who served at randomly chosen two hospitals and health centers in the zone, in between (December 29th-2017 and January 22nd-2019) was done. WHO-steps tools were used to gather the data, finally handled with (Epidata-V-3.1 and SPSS-V-22) software’s. Lastly, using a multivariable conditional-LR-model, 4-models with AOR (95%CI) were computed to arrived at the final model, and then variables accepted as significant at (p-value < 0.05)level. Result Overall, 633 PLHIVs (139 cases and 494 controls) were included in the analysis. The leading factors associated with MS were Age(>/=45years-old)(AOR=4.0,95%CI: 1.4-11.9), completed secondary school education (AOR=0.1,95% CI:0.01-0.5), un-employed(a home-maker AOR=0.1,95%CI:0.03-0.7 vs able to work AOR=0.1,95%CI:0.06-0.5)),Antiretroviral-therapy-exposed(AOR=0.1,95%CI:1.0-8.5),WHO-stage(>/=III)(AOR=4.4,95% CI:1.4-13.4), total physically activity (AOR=0.1,95% CI:0.04- 0.35), history of blood sugar measured (AOR=10.7,95%CI:3.3-34.6), elevated waist-circumference(AOR=6.9,95%CI:2.5-18.7),raised body mass index(AOR=5.4 95% CI: 1.6,18.4),fasting-glucose(AOR=29.3,95%CI:10.0-85.4),raisedtriglyceride(AOR=4.8,95%CI:2.0-11.3), and low high density lipoprotein(AOR=12.3 95% CI: 5.2,29.3). Conclusion The finding implicated, the significance of planning intervention actions that targets the above factors in to account.
... NRTIs act as false substrates to HIV-1 reverse transcriptase which lack an essential motif, a 3'-hydroxyl group which after incorporation into a growing cDNA cannot mount nucleophilic attack on the incoming dNTP. This leads to abortion of proviral cDNA chain elongation process Barbaro, 2006;El Safadi et al., 2007) (Fig. 1). NRTIs are initially activated intracellularly by tri-phosphorylation (except tenofovir, which already has an attached phosphate group, hence is classified as nucleotide and only undergoes bi-phosphorylation) (Fig. 1). ...
Preprint
Full-text available
Background Hypertension is a public health problem in sub-Saharan Africa, with considerable under- diagnosis, poor management, and lack of community-wide preventive strategies. Hypertension is a common condition in South Africa and is a risk factor for heart attacks, stroke, left ventricular hypertrophy, renal disease, and blindness. In this research study, we identify and examine the trend of physical activity within South Africa's HIV-positive and negative rural communities. Methods This was a secondary analysis of cross sectional survey data from the Agincourt Health and Socio-Demography Surveillance System Site (AHDSS) that were collected over a period of 10 months, from August 2010 to May 2011. All 4436 individuals participating in the AHDSS survey were included. The inclusion criteria for the survey were from aged 15 years or older, and a permanent resident according to the 2009 census. The participants were interview (approximately 45 minutes) on chronic disease risk factor [applying] a questionnaire; anthropometric measurement were taken, blood pressure (BP); point-of care analysis of lipids and glucose; and dried blood spot collections for HIV and ELISA testing were conducted. Results The research findings showed a significant/strong association between hypertension and HIV in both univariate and multivariate analysis. In many developed countries, the prevalence of hypertension among HIV-positive individuals varies between 8% and 39%. A study in Kenya reported a lower prevalence of 7.4%–11.2%, compared with 22.1%– 32.2% in study. Conclusion Hypertension is common in HIV-infected adults and is likely because of a combination of known risk factors, levels of physical activities/sedentary way of life, HIV-specific factors, and ART. There is a need for broad, global cohort studies to improve our knowledge and understanding of the above named subject matter, interventional studies are needed to discover new approaches in hypertension and HIV-infected individuals in South Africa and sub-Saharan Africa to avoid and manage hypertension and hypertension-related cardiovascular diseases among HIV infected persons especially in this era of epidemiologic and demographic transition in LMICs.
Article
Full-text available
Retroviral protease inhibitors used as therapy for HIV-1 infection have been causally associated with serious metabolic side effects, including peripheral lipodystrophy, hyperlipidemia, insulin resistance, and in some cases, overt type 2 diabetes. The etiology of this characteristic clinical syndrome remains unknown. We demonstrate that the HIV protease inhibitor, indinavir, dramatically inhibits insulin-stimulated glucose uptake in 3T3-L1 adipocytes in a dose-dependent manner (63% inhibition observed with 100 μm indinavir). Indinavir treatment did not affect early insulin signaling events or the translocation of intracellular Glut1 or Glut4 glucose transporters to the cell surface. To determine whether indinavir may be directly affecting the intrinsic transport activity of glucose transporters, the Glut1 and Glut4 isoforms were heterologously expressed and analyzed inXenopus laevis oocytes. Indinavir at 100 μmhad no effect on Glut1 transport activity in Xenopusoocytes, whereas Glut4 activity was significantly inhibited (45% inhibition). Similar effects on glucose transport were observed for other HIV protease inhibitors. We conclude that HIV protease inhibitors as a class are capable of selectively inhibiting the transport function of Glut4 and that this effect may be responsible for a major iatrogenic complication frequently observed in HIV patients.
Article
Objective: To describe the incidence of clinically recognized thrombosis and associated factors among individuals infected with HIV. Design: A longitudinal medical record review. Setting: Over 100 medical clinics in nine US cities. Patients: A total of 42 935 individuals aged 13 years or older with HIV infection, observed for an average of 2.4 years. Main outcome measures: The incidence of thrombosis among HIV-infected individuals; adjusted odds ratios for factors associated with thrombosis. Results: The incidence of thrombosis among HIV-infected individuals was 2.6/1000 person-years (PY). Factors significantly associated with thrombosis included: age of 45 or more years (adjusted odds ratio [AOR], 1.9; 95% confidence interval [CI], 1.4-2.7); a diagnosis of cytomegalovirus disease or retinitis (AOR, 1.9; Cl, 1.2-2.9), or other AIDS-defining opportunistic illness (AOR, 1.5; CI, 1.1 -2.2); hospitalization (AOR, 3.3; CI, 2.5-4.4); and the prescription of megestrol acetate (AOR, 2.0; CI 1.3-2.9) or indinavir (AOR, 2.4; Cl 1.4-4.3). The prescription of other protease inhibitors, sex, race, and mode of HIV exposure were not associated with thrombosis. Conclusion: Among HIV-infected individuals, clinically detected thrombosis is more common in those who have opportunistic illnesses, for whom megestrol acetate or indinavir have been prescribed, who have been hospitalized, and who are aged 45 years or older. Physicians should be aware of the risks of thrombosis in order to promote the early identification and appropriate treatment or prophylaxis. Further study is needed to characterize the association between indinavir and thrombosis. (C) 2000 Lippincott Williams & Wilkins.
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
LR: 20061115; JID: 7501160; 0 (Antilipemic Agents); 0 (Cholesterol, HDL); 0 (Cholesterol, LDL); 57-88-5 (Cholesterol); CIN: JAMA. 2001 Nov 21;286(19):2401; author reply 2401-2. PMID: 11712930; CIN: JAMA. 2001 Nov 21;286(19):2400-1; author reply 2401-2. PMID: 11712929; CIN: JAMA. 2001 Nov 21;286(19):2400; author reply 2401-2. PMID: 11712928; CIN: JAMA. 2001 Nov 21;286(19):2400; author reply 2401-2. PMID: 11712927; CIN: JAMA. 2001 May 16;285(19):2508-9. PMID: 11368705; CIN: JAMA. 2003 Apr 16;289(15):1928; author reply 1929. PMID: 12697793; CIN: JAMA. 2001 Aug 1;286(5):533-5. PMID: 11476650; CIN: JAMA. 2001 Nov 21;286(19):2401-2. PMID: 11712931; ppublish
Article
HIV-1 protease-inhibitor treatments are associated with a syndrome of peripheral lipodystrophy, central adiposity, breast hypertrophy in women, hyperlipidaemia, and insulin resistance. The catalytic region of HIV-1 protease, to which protease inhibitors bind, has approximately 60% homology to regions within two proteins that regulate lipid metabolism: cytoplasmic retinoic-acid binding protein type 1 (CRABP-1) and low density lipoprotein-receptor-related protein (LRP). We hypothesise that protease inhibitors inhibit CRABP-1-modified, and cytochrome P450 3A-mediated synthesis of cis-9-retinoic acid, a key activator of the retinoid X receptor; and peroxisome proliferator activated receptor type gamma (PPAR-gamma) heterodimer, an adipocyte receptor that regulates peripheral adipocyte differentiation and apoptosis. Protease-inhibitor binding to LRP would impair hepatic chylomicron uptake and triglyceride clearance by the endothelial LRP-lipoprotein lipase complex. The resulting hyperlipidaemia contributes to central fat deposition (and in the breasts in the presence of oestrogen), insulin resistance, and, in susceptible individuals, type 2 diabetes. Understanding the syndrome's pathogenesis should lead to treatment strategies and to the design of protease inhibitors that do not cause this syndrome.
Article
To describe the incidence of clinically recognized thrombosis and associated factors among individuals infected with HIV. A longitudinal medical record review. Over 100 medical clinics in nine US cities. A total of 42 935 individuals aged 13 years or older with HIV infection, observed for an average of 2.4 years. The incidence of thrombosis among HIV-infected individuals; adjusted odds ratios for factors associated with thrombosis. Results: The incidence of thrombosis among HIV-infected individuals was 2.6/1000 person-years (PY). Factors significantly associated with thrombosis included: age of 45 or more years (adjusted odds ratio [AOR], 1.9; 95% confidence interval [CI], 1.4-2.7); a diagnosis of cytomegalovirus disease or retinitis (AOR, 1.9; CI, 1.2-2.9), or other AIDS-defining opportunistic illness (AOR, 1.5; CI, 1.1-2.2); hospitalization (AOR, 3.3; CI, 2.5-4.4); and the prescription of megestrol acetate (AOR, 2.0; CI 1.3-2.9) or indinavir (AOR, 2.4; CI 1.4-4.3). The prescription of other protease inhibitors, sex, race, and mode of HIV exposure were not associated with thrombosis. Among HIV-infected individuals, clinically detected thrombosis is more common in those who have opportunistic illnesses, for whom megestrol acetate or indinavir have been prescribed, who have been hospitalized, and who are aged 45 years or older. Physicians should be aware of the risks of thrombosis in order to promote the early identification and appropriate treatment or prophylaxis. Further study is needed to characterize the association between indinavir and thrombosis.
Article
Thrombosis of upper extremity arteries is most commonly due to atherosclerosis of the proximal subclavian artery, trauma, or catheter-related injury. In the absence of an identifiable cause, a search for a hypercoagulable state is indicated. Hematologic manifestations of human immunodeficiency virus (HIV) infection and AIDS are frequent occurrences (Coyle TE. Med Clin N Am 1997;81:449-476). The most important of these are cytopenias (anemia, neutropenia, and thrombocytopenia). The incidence and severity of cytopenia are generally correlated to the stage of the HIV infection. In addition, various coagulation abnormalities have been reported in HIV-infected patients. Apart from thrombocytopenia, these have included a prolonged APTT due to the presence of lupus anticoagulant, an increased prevalence of protein S and heparin cofactor II deficiency, and hypoalbuminemia-related fibrin polymerization defects (Toulon P. Ann Bio Clin (Paris) 1998;56:153-160). HIV infection has also been associated with endothelial dysfunction. Although for the most part asymptomatic, elevated D-dimer levels have been found in HIV-infected patients, suggesting the existence of a prethrombotic state. In fact, clinical thrombosis eventuates in 2% of these patients (Toulon, 1988). Documented thromboses have involved both veins and arteries. We hereby present a patient who developed an acute thrombosis of his brachial artery as the initial manifestation of HIV infection.
Article
HIV infection has reached endemic proportions in many African countries. In addition, HIV infection is a significant cause of renal dysfunction in the United States. HIV patients are at higher risk of developing hypertension at a younger age than the general population. Predisposing factors for developing hypertension include vasculitis in small, medium, and large vessels in the form of leukocytoclastic vasculitis, and aneurysms of the large vessels such as the carotid, femoral, and abdominal aorta with impairment of flow to the renal arteries. A syndrome of acquired glucocorticoid resistance has been described in patients with HIV with hypercortisolism and a lower affinity of the glucocorticoid receptors. The syndrome is characterized clinically by weakness, hypertension or hypotension, and skin pigmentation changes. Acute and chronic renal failure is often associated with HIV infection. The associated dysfunction in water and salt handling often induces hypertension. Finally, atherosclerosis has been described in young adults with HIV infection secondary to receiving highly active antiretroviral therapy.
Article
to preliminarily describe the clinical features and management of arterial occlusive disease in human immunodeficiency virus (HIV) infected patients. twenty HIV positive patients with symptomatic large-vessel arterial occlusion treated by a tertiary vascular unit in a 3-year period. retrospective review of clinical case records. patients were noted to be young (median age 37 years), with preponderance of males. Twelve patients had evidence of advanced HIV infection. All patients had critical ischaemia, involving the upper limbs in four and the lower limbs in 16. Coagulation abnormalities were noted in two cases. Operative intervention in 18 patients included revascularisation in seven. Thrombotic occlusion of normal-looking arteries was noted. Arterial biopsy revealed leucoIcytoclastic vasculitis indicative of HIV arteritis in three of five cases examined. initial experience with large-vessel occlusive disease in HIV positive patients suggests an underlying arteritic aetiology, with clinical and pathological features distinct from atherosclerosis. Further in-depth study is necessary to clarify the pathophysiological basis thereof.