Download Lipid Abnormalities - AIDS Education and Training Centers

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Microbicides for sexually transmitted diseases wikipedia , lookup

Transcript
SUPPLEMENT ARTICLE
Lipid Abnormalities
Michael Dube1 and Marcy Fenton2
1
Indiana University School of Medicine, Wishard Memorial Hospital, Division of Infectious Diseases, Indianapolis, Indiana; and 2AIDS Project Los
Angeles HIV Nutrition Program, Los Angeles, California
Dyslipidemia is an important clinical problem in individuals infected with human immunodeficiency virus
(HIV) who are receiving antiretroviral therapy. Data suggest that increased cardiovascular disease is occurring
in this population. HIV-infected individuals should undergo evaluation and treatment regimens based on the
current National Cholesterol Education Program guidelines. In most situations, the first interventions should
be nonpharmacological and should include diet, exercise, and management of other hygienic risk factors. If
pharmacologic therapy becomes necessary, the choices of lipid-lowering agents should be limited to agents
with the least likelihood of adverse drug interactions.
PREVALENCE
Abnormalities of lipid metabolism are common in HIVinfected patients and tend to be accentuated in those
receiving antiretroviral therapy, particularly with protease inhibitors (PIs). Forty-seven percent of PI recipients at one clinic had lipid abnormalities, according
to the National Cholesterol Education Program
(NCEP) guidelines for intervention [1]. Behrens et al.
[2] reported that 56 (57%) of 98 PI recipients experienced hyperlipidemia. Among those, 19% had elevation of low-density lipoprotein cholesterol (LDL-C)
alone, 44% had hypertriglyceridemia alone, and 37%
had both abnormalities. Thus, dyslipidemia is a common problem among HIV-infected individuals receiving treatment.
ETIOLOGY
Abnormalities of lipid metabolism reported before the
use of HIV-1 PIs include increases in serum triglycerides [3, 4] and decreases in total and high-density lipoprotein cholesterol (HDL-C) [4, 5] that occurred
Reprints or correspondence: Dr. Michael Dube, Indiana University School of
Medicine, Wishard Memorial Hospital, Division of Infectious Diseases, 1001 W.
10th St., Suite OPW-430, Indianapolis, IN 46202-2879 ([email protected]).
Clinical Infectious Diseases 2003; 36(Suppl 2):S79–83
2003 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2003/3607S2-0005$15.00
with disease progression. During therapy with PIs, increases in serum triglycerides may be extreme [6], particularly with ritonavir therapy. In contrast to the prePI era, increases in cholesterol have occurred with PIs
[1, 2, 7–9]. A mean increase in serum cholesterol of 32
mg/dL (23%) and a 27% increase in LDL-C occurred
3.4 months after initiation of therapy with a PI [8]. In
HIV-negative volunteers, ritonavir therapy increased
total cholesterol by 24% and triglycerides by 137%
within 2 weeks [10]. In addition, significant elevations
of both total cholesterol and HDL-C have also occurred
during therapy with the nonnucleoside reverse transcriptase inhibitors (NNRTI) efavirenz [11] and nevirapine [12]. Whether these increases in HDL-C are beneficial remains speculative.
POTENTIAL FOR CARDIOVASCULAR
MORBIDITY AND MORTALITY
Anecdotal reports suggest that serious premature vascular events may be related to PI therapy and abnormal
lipids [2, 13–15]. The incidence of cardiovascular morbidity was increased among HIV-infected subjects in
general but not among PI recipients, in one abstract
[16]. However, duration of PI use was positively associated with cardiac events in another preliminary report [17]. Although there is currently no definitive evidence that antiviral drug–associated lipid disturbances
will result in increased cardiovascular morbidity and
Lipid Abnormalities • CID 2003:36 (Suppl 2) • S79
mortality, many experts speculate that HIV treatment-related
dyslipidemia will increase the atherogenic tendency, particularly
when combined with other HIV-associated metabolic abnormalities such as insulin resistance [7, 8, 18], visceral adiposity
[19], impaired fibrinolysis [20], and chronic infection and immune activation. Consequently, rationales exist for intervention
in many individuals.
factor for cardiovascular disease, even when values are only
modestly elevated (200–400 mg/dL) [23]. When triglyceride
levels exceed 200 mg/dL, calculation of non–HDL-C (total cholesterol minus HDL-C) should be performed and considered
a secondary target for intervention [22]. In addition, the high
frequency of low HDL-C in persons with HIV warrants attention [2].
EVALUATION OF CARDIOVASCULAR DISEASE
RISKS
MANAGEMENT: DIET AND NONDRUG
THERAPIES
Evaluation of serum lipids should be performed after fasting
for a minimum of 8 h, and preferably 12 h. The standard
screening lipid profile should include measurement of total
cholesterol, HDL-C, and triglycerides, with calculation of LDLC, and should be obtained before therapy [21]. This should be
repeated 3–6 months after the initiation of HAART, and then
yearly. All patients should be screened for other cardiovascular
risk factors including family history, smoking, hypertension,
menopausal status, physical inactivity, obesity, and diabetes, in
addition to potential exacerbating factors such as certain medical illnesses and medications [22].
Nondrug interventions should be the first approach for management of abnormal lipid levels. Clinicians should consult with
dietary specialists as a first step, when initial attempts at dietary
intervention fail to achieve the desired effects, or when intensive
dietary modification becomes necessary. Other nondrug therapies are expected to be beneficial, as they are in persons without HIV. For example, structured exercise plus diet resulted in
a 21% decrease in triglyceride levels in HIV-infected patients
[1], and resistance training improved triglyceride levels [24].
Smoking cessation and weight reduction for obesity also improve the overall cardiovascular risk profile.
Diet. After assessment of existing dietary habits, the Therapeutic Lifestyle Changes (TLC) diet should be prescribed (table
2). For many patients, these changes can be achieved without
radical alterations in dietary habits. These diets also reduce
serum triglycerides. For patients with low HDL-C, reduction
of dietary fat will further reduce HDL-C. Monounsaturated fats
(e.g., canola and olive oil) should be substituted for saturated
fats. Moreover, if carbohydrate intake is increased as ingestion
of fat is reduced, this may increase triglyceride levels. The management of severe hypertriglyceridemia and hyperchylomicronemia requires a very–low-fat diet and avoidance of simple
sugars (i.e., use of low–glycemic-index carbohydrates) and alcohol. Fish oils (omega-3 fatty acid supplements) variably decrease triglyceride synthesis and may be tried in patients with
severe hypertriglyceridemia.
Weight reduction. In subjects with central (abdominal)
obesity, weight reduction through diet modifications and in-
THRESHOLDS FOR TREATMENT INTERVENTION
Dyslipidemia in patients with well-controlled HIV should have
similar, and perhaps greater, long-term consequences for cardiovascular complications compared with the general population. For the purposes of initiating therapy for dyslipidemia,
the Panel recommends that the NCEP guidelines [22] should
generally be followed for HIV-infected patients. Of note, the
new NCEP guidelines now include a category termed coronary
heart disease (CHD) “risk equivalents,” which include diabetes
mellitus, other atherosclerotic disease, and multiple risk factors
that confer a 10-year risk of CHD of 120%. Because of the
high risk of CHD in these groups, these individuals should be
treated as aggressively as those with established CHD [22]. The
NCEP guidelines target primarily LDL-C (table 1). However,
elevated triglyceride levels also represent an independent risk
Table 1.
Target goals for LDL cholesterol in patients with HIV.
LDL-C level, mg/dL
Risk
Goal
Initiate therapeutic
lifestyle changes
Consider
drug therapy
0–1 risk factors for CHD
!160
⭓160
⭓190
⭓2 risk factors for CHD
(10-y risk ⭐0%)
!130
⭓130
⭓160
With CHD, or CHD risk equivalent
(10-y risk 120%)
!100
⭓100
⭓130 (100–129,
drug optional)
NOTE. Adapted from [22]. CHD, coronary heart disease; LDL-C, low-density lipoprotein cholesterol. See text for definition of CHD risk equivalent.
S80 • CID 2003:36 (Suppl 2) • Dube and Fenton
Table 2.
diet.
National Cholesterol Education Program Therapeutic Lifestyle Changes (TLC)
Nutrient
Recommendation
Total calories
Adjust to achieve desired weight
Total fat
25%–35% of total calories; keep trans fatty acids low
Saturated fat
!7% of total calories
Polyunsaturated fat
Up to 10% of total calories
Monounsaturated fat
Up to 20% of total calories
Cholesterol
!200 mg/d
Carbohydrates
50%–60% of total calories, predominantly complex carbohydrates
Protein
∼15% of total calories
NOTE.
Adapted from [22].
creased physical activity is an important element of therapy for
abnormal lipid levels. Weight reduction enhances the LDL-C
lowering that can be achieved by diet alone. Both weight reduction and intensive aerobic exercise also reduce serum triglycerides, raise HDL-C, lower blood pressure, and decrease
the risk for diabetes mellitus [25]. However, the effects of weight
reduction on peripheral adipose tissues stores have not been
established in patients with HIV but are expected to be minimal
in the presence of central obesity because intra-abdominal fat
is more sensitive to lipolysis than peripheral fat [26].
Patients with CHD or CHD risk equivalents. The goal
for LDL-C is !100 mg/dL in patients with CHD or CHD risk
equivalents. In contrast to subjects without CHD, prolonged
attempts to lower LDL-C with diet may not be desirable before
initiation of drug therapy. If LDL-C is 1130 mg/dL, diet and
lipid-lowering therapy may be instituted simultaneously in
these high-risk individuals.
Monitoring the response to therapy. In patients without
CHD, after initiation of the therapeutic diet, lipids should be
measured and adherence to the diet assessed after 4 to 6 weeks
and again after 3 months.
MANAGEMENT: LIPID-LOWERING DRUGS
As for patients without HIV, if lipid levels remain above the
target after intensive dietary and lifestyle interventions have
been attempted for at least 12 weeks, drug therapy should be
considered. In the absence of other CHD risk factors, especially
in patients !35 years of age, a longer period of nondrug therapies may be appropriate. Shorter periods of time may be considered in patients with severe elevations of LDL-C (1220 mg/
dL) or with multiple risk factors. Drug therapy should only be
added to dietary therapy, never substituted for it.
Lipid-lowering therapies for HIV-infected patients with dyslipidemia are conceptually problematic because of the potential
for drug interactions (reviewed in [21]). Both increased toxicity
due to increased levels of lipid-lowering drugs and reduced
efficacy of antiretroviral drugs are a concern.
Drug therapy for elevated LDL-C or non–HDL-C. The
hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, or statins, reduce the risk of CHD in patients without
previous CHD (primary prevention) and risk of recurrent CHD
events (secondary prevention) [27] in patients without HIV.
On the basis of limited pharmacokinetic data [28] and the
likelihood of significant interactions, the Panel recommends
that when statin therapy is necessary, low initial dosages of
either pravastatin (20 mg daily) or atorvastatin (10 mg daily)
be used, along with careful clinical monitoring. Fluvastatin is
an alternative statin, but there are no data on fluvastatin interactions with PIs. Therapy with lovastatin and simvastatin
should be avoided.
The fibrates (gemfibrozil and fenofibrate) are well-tolerated
alternative agents when hypercholesterolemia is accompanied
by elevated triglycerides, but fibrates are less effective than statins for reducing LDL-C. Niacin lowers LDL-C and triglyceride
levels but produces frequent cutaneous flushing and pruritus.
Because niacin causes insulin resistance [29, 30], niacin should
be avoided as first-line therapy in patients receiving PIs or in
patients with fat redistribution, because both conditions are
associated with reduced insulin sensitivity. Bile-sequestering
resins are not recommended because their effects on antiviral
drug absorption are not known.
Drug therapy for elevated triglycerides. The absolute value
at which drug therapy should be given for isolated hypertriglyceridemia has not been defined, but in the absence of CHD
risk factors or hypercholesterolemia, elevations 1500 mg/dL
(unresponsive to nondrug therapies) carry an increased risk of
pancreatitis. Drug therapy should therefore be considered for
these individuals. For patients with extreme elevations (i.e.,
triglycerides 12000 mg/dL), drug therapies may be initiated
concurrently with nondrug interventions.
Fibrates should be prescribed if drug therapy is necessary for
Lipid Abnormalities • CID 2003:36 (Suppl 2) • S81
hypertriglyceridemia. Treatment is with gemfibrozil [1] (600
mg bid) or fenofibrate (54–160 mg once daily). If treatment
with a fibrate results in inadequate lowering of serum triglycerides, or if LDL-C levels remain elevated, a cautious trial of
niacin or a statin (see above) may be considered, with careful
monitoring for symptoms of skeletal muscle toxicity.
8.
9.
10.
CONSIDERATION FOR SWITCHING
ANTIRETROVIRAL THERAPIES
Where virologically appropriate, substitution of a new antiviral
agent with a lesser tendency to induce dyslipidemia for an
existing agent with a greater tendency may be considered. In
NNRTI-naive patients, substitution of nevirapine for a PI improves serum lipids in some studies [31] but not in others [32,
33]. The substitution of efavirenz for a PI has not consistently
had a beneficial effect [34]. Improvement in lipid levels have
also been reported with the substitution of abacavir for PIs [35,
36]. Rates of virologic relapse when an NNRTI or abacavir is
substituted for PIs have been acceptable. When triglyceride
levels remain markedly elevated despite aggressive interventions, it may be reasonable to change ritonavir to another PI
or non–PI-based regimen or to select the lowest effective dose
of ritonavir when it is used to increase the levels of other PIs.
To our knowledge, there are no studies that compare the
effects of switching antiretroviral treatment with the effects of
adding lipid-lowering agents to ongoing successful therapy. Clinicians will need to weigh the potential risks of new treatmentrelated toxicities and the possibility of virologic relapse when
switching antiviral drugs against the potential risks of drug
interactions and toxicities from lipid-lowering agents added to
antiretroviral drug regimens.
11.
12.
13.
14.
15.
16.
17.
18.
19.
References
1. Henry K, Melroe H, Huebesch J, Hermundson J, Simpson J. Atorvastatin and gemfibrozil for protease-inhibitor–related lipid abnormalities. Lancet 1998; 352:1031–2.
2. Behrens G, Dejam A, Schmidt H, et al. Impaired glucose tolerance,
beta cell function and lipid metabolism in HIV patients under treatment with protease inhibitors. AIDS 1999; 13:F63–70.
3. Grunfeld C, Kotler DP, Shigenaga JK, et al. Circulating interferonalpha levels and hypertriglyceridemia in the acquired immunodeficiency syndrome. Am J Med 1991; 90:154–62.
4. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold
KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human
immunodeficiency virus infection and the acquired immunodeficiency
syndrome. J Clin Endocrinol Metab 1992; 74:1045–52.
5. Shor Posner G, Basit A, Lu Y, et al. Hypocholesterolemia is associated
with immune dysfunction in early human immunodeficiency virus-1
infection. Am J Med 1993; 94:515–9.
6. Danner SA, Carr A, Leonard JM, et al. A short-term study of the safety,
pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease. N Engl J Med 1995; 333:1528–33.
7. Carr A, Samaras K, Burton S, et al. A syndrome of peripheral lipo-
S82 • CID 2003:36 (Suppl 2) • Dube and Fenton
20.
21.
22.
23.
24.
25.
dystrophy, hyperlipidaemia and insulin resistance due to HIV protease
inhibitors. AIDS 1998; 12:F51–8.
Mulligan K, Grunfeld C, Tai VW, et al. Hyperlipidemia and insulin
resistance are induced by protease inhibitors independent of changes
in body composition in patients with HIV infection. J Acquir Immune
Defic Syndr 2000; 23:35–43.
Periard D, Telenti A, Sudre P, et al. Atherogenic dyslipidemia in HIVinfected individuals treated with protease inhibitors. the Swiss HIV
cohort study. Circulation 1999; 100:700–5.
Purnell JQ, Zambon A, Knopp RH, et al. Effect of ritonavir on lipids
and post-heparin lipase activities in normal subjects. AIDS 2000; 14:
51–7.
Tashima K, Stryker R, Skiest D, et al. Lipid profiles and clinical lipodystrophy in study 006 patients [abstract 1304]. In: Program and
abstracts of the 39th Interscience Conference on Antimicrobial Agents
and Chemotherapy (San Diego). Washington, DC: American Society
for Microbiology, 1999; 501.
van der Valk M, Kastelein JJ, Murphy RL, et al. Nevirapine-containing
antiretroviral therapy in HIV-1 infected patients results in an antiatherogenic lipid profile. AIDS 2001; 15:2407–14.
Henry K, Melroe H, Huebsch J, et al. Severe premature coronary artery
disease with protease inhibitors. Lancet 1998; 351:1328.
Gallet B, Pulik M, Genet P, Chedin P, Hiltgen M. Vascular complications
associated with use of HIV protease inhibitors. Lancet 1998; 351:
1958–9.
Vittecoq D, Escaut L, Monsuez JJ. Vascular complications associated
with use of HIV protease inhibitors. Lancet 1998; 351:1959.
Klein D, Hurley L, Sorel M, Sidney S. Do protease inhibitors increase
the risk for coronary heart disease among HIV positive patients? Follow-up through June 2000 [abstract 655]. In: Program and abstracts
of the 8th Conference on Retroviruses and Opportunistic Infections
(Chicago). Washington, DC: Foundation for Retrovirology and Human
Health; 2001.
Mary-Krause M, Cotte L, Partisani M, Simon A, Costagliola D. Impact
of treatment with protease inhibitor (PI) on myocardial infarction (MI)
occurrence in HIV-infected men [abstract 657]. In: Program and abstracts of the 8th Conference on Retroviruses and Opportunistic Infections (Chicago). Washington, DC: Foundation for Retrovirology and
Human Health; 2001.
Walli R, Herfort O, Michl GM, et al. Treatment with protease inhibitors
associated with peripheral insulin resistance and impaired oral glucose
tolerance in HIV-1–infected patients. AIDS 1998; 12:F167–73.
Miller KD, Jones E, Yanovski JA, Shankar R, Feuerstein I, Falloon J.
Visceral abdominal-fat accumulation associated with use of indinavir.
Lancet 1998; 351:871–5.
Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and
cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis 2001; 32:
130–9.
Dubé MP, Sprecher D, Henry WK, et al. Preliminary guidelines for
the evaluation and management of dyslipidemia in HIV-infected adults
receiving antiretroviral therapy. Recommendations of the Adult ACTG
Cardiovascular Disease Focus Group. Clin Infect Dis 2000; 31:1216–24.
Expert Panel on Detection Evaluation and Treatment of High Blood
Cholesterol in Adults. 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). JAMA 2001; 285:2486–97.
Hokanson JE, Austin MA. Plasma triglyceride level is a risk factor for
cardiovascular disease independent of HDL cholesterol level: a metaanalysis of population-based prospective studies. J Cardiovasc Risk
1996; 3:213–9.
Yarasheski KE, Tebas P, Stanerson B, et al. Resistance exercise training
reduces hypertriglyceridemia in HIV-infected men treated with antiviral therapy. J Appl Physiol 2001; 90:133–8.
Franz MJ, Horton ES, Bantle JP, et al. Nutrition principles for the
26.
27.
28.
29.
30.
31.
32.
management of diabetes and related complications. Diabetes Care
1994; 17:490–518.
Smith SR, Zachwieja JJ. Visceral adipose tissue: a critical review of
intervention strategies. Int J Obes Relat Metab Disord 1999; 23:329–35.
LaRosa JC, He J, Vupputuri S. Effect of statins on risk of coronary
disease: a meta-analysis of randomized controlled trials. JAMA 1999;
282:2340–6.
Fichtenbaum CJ, Gerber JG, Rosenkranz SL, et al. Pharmacokinetic
interactions between protease inhibitors and statins in HIV seronegative volunteers: ACTG Study A5047. AIDS 2002; 16:569–77.
Garg A, Grundy SM. Nicotinic acid as therapy for dyslipidemia in noninsulin-dependent diabetes mellitus. JAMA 1990; 264:723–6.
Kahn SE, Beard JC, Schwartz MW, et al. Increased B-cell secretory
capacity as a mechanism for islet cell adaptation to nicotinic acidinduced insulin resistance. Diabetes 1989; 38:562–8.
Martinez E, Conget I, Lozano L, Casamitjana R, Gatell JM. Reversion
of metabolic abnormalities after switching from HIV-1 protease inhibitors to nevirapine. AIDS 1999; 13:805–10.
Barreiro P, Soriano V, Blanco F, Casimiro C, de la Cruz JJ, Gonzalez-
33.
34.
35.
36.
Lahoz J. Risks and benefits of replacing protease inhibitors by nevirapine in HIV-infected subjects under long-term successful triple combination therapy. AIDS 2000; 14:807–12.
Tebas P, Yarasheski K, Powderly WG, et al. A prospective, open-label
pilot trial of a maintenance nevirapine-containing regimen in patients
with undetectable viral loads on protease inhibitor regimens for at least
6 months [abstract 45]. In: Program and abstracts of the 7th Conference
on Retroviruses and Opportunistic Infections (San Francisco). Washington, DC: Foundation for Retrovirology and Human Health; 2000.
Martinez E, Garcia-Viejo MA, Blanco JL, et al. Impact of switching
from human immunodeficiency virus type 1 protease inhibitors to
efavirenz in successfully treated adults with lipodystrophy. Clin Infect
Dis 2000; 31:1266–73.
Walli RK, Michl GM, Bogner JR, Goebel FD. Improvement of HAARTassociated insulin resistance and dyslipidemia after replacement of protease inhibitors with abacavir. Eur J Med Res 2001; 6:413–21.
Carr A, Hudson J, Chuah J, et al. HIV protease inhibitor substitution
in patients with lipodystrophy: a randomized, controlled, open-label,
multicentre study. AIDS 2001; 15:1811–22.
Note added in proof. Since submission of this paper, the following articles have been published with regard to cardiovascular
disease outcomes and HIV: (1) Bozzette SA, Ake CF, Tam HK, Chang SW, Louis TA. Cardiovascular and cerebrovascular events
in patients treated for human immunodeficiency virus infection. N Engl J Med 2003; 702–10; (2) Holmberg SSD, Moorman AC,
Williamson JM, et al. Protease inhibitors and cardiovascular outcomes in patients with HIV-1. Lancet 2002; 360:1747–8; and (3)
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? JAIDS J Acquired Immune Defic Syndr 2002; 30: 471–7. The following reports of controlled clinical trials
of lipid-lowering drugs in HIV-infected subjects have also been recently published: (1) Doser N, Kübli S, Telentic A, et al. Efficacy
and safety of fluvastatin in hyperlipidemic protease inhibitor–treated HIV-infected patients. AIDS 2002; 16:1982–3; and (2) Miller
J, Brown D, Amin J, et al. A randomized, double-blind study of gemfibrozil for the treatment of protease inhibitor–associated
hypertriglyceridaemia. AIDS 2002; 16:2195–200.
Lipid Abnormalities • CID 2003:36 (Suppl 2) • S83