ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Low-density lipoprotein cholesterol lowering with drugs other than statins and PCSK9 inhibitors

Low-density lipoprotein cholesterol lowering with drugs other than statins and PCSK9 inhibitors
Literature review current through: Jan 2024.
This topic last updated: Mar 22, 2023.

INTRODUCTION — Lipid (or lipoprotein) altering agents encompass several classes of drugs including statins, cholesterol absorbing inhibitors, fibric acid derivatives, bile acid sequestrants, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, nicotinic acid, and others. These drugs differ with respect to mechanism of action and to the degree and type of lipid altering.

Most patients for whom a prescription drug therapy is deemed advisable will have an elevation in their low-density lipoprotein cholesterol (LDL-C) level and a statin is the established first line therapy. Other lipid lowering drugs are used to augment statin effects on LDL-C, substitute for statins when that class cannot be used, or to treat non-LDL-C disorders, primarily hypertriglyceridemia. The decision to use a non-statin drug can be influenced by clinical parameters other than the lipid values themselves.

The characteristics, efficacy, and safety of the lipid-lowering drugs other than the statins and PCSK9 inhibitors will be reviewed here. The efficacy of statins and PCSK9 inhibitors is discussed elsewhere. (See "Statins: Actions, side effects, and administration" and "PCSK9 inhibitors: Pharmacology, adverse effects, and use".)

Additionally, therapeutic decision making in patients with elevated lipid levels is discussed in detail separately. (See "Low-density lipoprotein cholesterol-lowering therapy in the primary prevention of cardiovascular disease" and "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

MECHANISMS OF BENEFIT — The mechanisms of benefit seen with these drugs are incompletely understood. Potential mechanisms include regressions of atherosclerosis, plaque stabilization, reversal of endothelial dysfunction, inhibition of inflammatory pathways, and decreased thrombogenicity. (See "Mechanisms of benefit of lipid-lowering drugs in patients with coronary heart disease".)

While the drugs discussed in this topic, as well as the statins and PCSK9 inhibitors, lower LDL-C to variable degrees, it has not yet been established that LDL-C lowering is the only mechanism by which these drugs lead to improved patient outcomes [1]. There are numerous in vitro studies that suggest statins have pleiotropic effects that could affect the atherosclerosis process beyond their LDL-C lowering effect.

DOSING REGIMENS/SIDE EFFECTS — Conventional dosing regimens and common adverse reactions are summarized in a table (table 1).

IMPACT ON LIPID VALUES — The range of expected changes in the lipid profile with these drugs are summarized in a table (table 2).

SPECIFIC DRUGS

Ezetimibe — Ezetimibe is a cholesterol absorption inhibitor that impairs dietary and biliary cholesterol absorption at the brush border of the intestine [2]. It is the most commonly prescribed LDL-C lowering agent after statins. Addition discussion of the clinical use of ezetimibe are presented separately. (See "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

Its mechanism involves inhibition of Niemann-Pick C1 like 1 (NPC1L1) protein, which is expressed both in the intestine and the liver, with this inhibition resulting in a net decrease in cholesterol absorption from the intestine [3-5]. A genetic study found an association between having a single inactivating mutation (heterozygous state) for NPC1L1 and lower LDL-C, and also lower risk of developing coronary heart disease (CHD), providing biologic support that a medication that blocks NPC1L1 might be expected to reduce the risk of CHD [6].

In two randomized, double-blind, placebo-controlled trials, ezetimibe at a dose of 10 mg/day reduced LDL-C by approximately 17 percent [7,8]. However, the reduction in LDL-C is bimodal with one in eight patients experiencing an average 36 percent reduction in LDL-C. The hyper-responsiveness to ezetimibe occurs in patients who lack the common NPC1L1 haplotype 1735C-25342A-27677T [9]. Ezetimibe is also effective as adjunctive therapy to a statin [10-14]; in one study, a 10 mg dose of ezetimibe lowered LDL-C by 14 percent below the LDL lowering effect of simvastatin [12].

IMPROVE-IT, the first large trial to directly assess clinical outcomes with ezetimibe plus a statin compared with a statin alone, found that after a median follow-up of six years, patients with an acute coronary syndrome randomized to ezetimibe/simvastatin had a lower rate of the primary composite cardiovascular outcome (cardiovascular death, myocardial infarction, hospital admission for unstable angina, coronary revascularization 30 or more days after randomization, or stroke) than those randomized to simvastatin alone (hazard ratio [HR] 0.94, 95% CI 0.89-0.99; seven-year event rates 32.7 versus 34.7 percent) [15]. All-cause mortality (HR 0.99) and cardiovascular mortality (HR 1.00) were not reduced. Adverse events were similar in the two arms. The benefits on cardiovascular end points provide support for LDL-C lowering with this drug. (See "Low-density lipoprotein-cholesterol (LDL-C) lowering after an acute coronary syndrome", section on 'Ezetimibe' and "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

Randomized trials, such as the SHARP trial in patients with chronic kidney disease, found reductions in cardiovascular end points with the combination of ezetimibe and simvastatin compared with placebo [16]. However, trials of this sort cannot be used to demonstrate additional clinical benefit from ezetimibe beyond what would be expected with a statin alone.

A trial of the combination of simvastatin and ezetimibe in patients with aortic stenosis, SEAS, found an increased risk of cancer and cancer death in the active treatment arm [17]. Statins do not appear to increase the risk of cancer, which raised the possibility that ezetimibe treatment could cause cancer. Because of this concern, an interim analysis of two other trials of simvastatin and ezetimibe (IMPROVE-IT and SHARP) was performed [18]. This analysis found no increased risk of incident cancer but a trend toward an increase in cancer deaths. However, an early increase in cancer deaths in a trial without an increase in incident cancer would be somewhat unusual. We feel the observed effects on cancer and cancer deaths are most likely due to chance. In a preliminary report of the final analysis of the SHARP trial, there continued to be no increase in cancer incidence with ezetimibe but there was a statistically nonsignificant increase in cancer deaths (3.2 versus 2.8 percent) [16]; the final analysis of IMPROVE-IT showed no increase in cancer (10.2 percent in each arm) [15].

The precise role of ezetimibe relative to other lipid lowering drugs is unclear. Similar reductions in LDL-C can often be achieved simply by maximizing the dose of statins. As an example, one randomized trial found that the reduction in LDL-C concentration was the same with atorvastatin 10 mg plus ezetimibe as with atorvastatin 80 mg alone; however, adding ezetimibe to atorvastatin 80 mg did result in an additional 9 percent reduction in LDL-C concentration [19]. Ezetimibe plus atorvastatin also produced a greater reduction in serum C-reactive protein than atorvastatin alone.

Ezetimibe may be helpful for avoiding high doses of statins (and potentially increased susceptibility to muscle injury) in patients who do not meet cholesterol goals on low-dose statin therapy alone. (See "Statin muscle-related adverse events".)

Ezetimibe has been well tolerated in clinical trials [7,8,13].

Gemfibrozil and fenofibrate have been noted to increase ezetimibe levels [20,21], although the clinical significance of this is uncertain. Ezetimibe and fenofibrate have been safely used in combination [22,23], and we use this combination clinically.

Bempedoic acid — Bempedoic acid is an inhibitor of adenosine triphosphate citrate lyase, an enzyme upstream of 3-hydroxy-3methylglutarly-CoA reductase (the target of statins) in the cholesterol biosynthesis pathway (see "Statins: Actions, side effects, and administration", section on 'Mechanism of action'). Bempedoic acid alone or in combination with statin or ezetimibe lowers LDL-C as well as other atherogenic proteins [24-26].

Bempedoic acid may be used in statin-intolerant patients who require modest lipid lowering, but side effects must be monitored.

In a trial of 13,970 patients who were unable or unwilling to take statins due to adverse effects and had or were at high risk for cardiovascular disease, patients were assigned either bempedoic acid 180 mg daily or placebo [27]. The following findings were noted after 40 months of follow-up:

People assigned bempedoic acid compared with placebo had lower rates of these outcomes:

Composite of death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization was lower (11.7 versus 13.3 percent; HR 0.87; 95% CI 0.79-0.96).

Composite of death from cardiovascular causes, nonfatal stroke, or nonfatal myocardial infarction (8.2 versus 9.5 percent); fatal or nonfatal myocardial infarction (3.7 versus 4.8 percent); and coronary revascularization (6.2 versus 7.6 percent).

People assigned bempedoic acid had similar rates of fatal or nonfatal stroke, death from cardiovascular causes, and death from any cause compared with placebo.

People assigned bempedoic acid had higher rates of gout (3.1 versus 2.1 percent) and cholelithiasis (2.2 versus 1.2 percent). Rates of serum creatinine, uric acid, and hepatic-enzyme levels were also modestly higher in those assigned bempedoic acid.

In 2019, two smaller randomized trials that evaluated the safety and LDL-C-lowering efficacy of this drug were published. In both trials, patients with either established cardiovascular disease or heterozygous familial hypercholesterolemia were randomly assigned to bempedoic acid (180 mg once daily) or placebo in a 2:1 ratio and followed for 52 weeks:

The CLEAR Harmony trial enrolled 2230 patients taking maximally-tolerated doses of a statin that included 6.6 percent taking low-intensity statin therapy, 43.5 percent taking moderate-intensity statin therapy, and 49.9 percent taking high-intensity statin therapy [28]. Baseline LDL-C had to be above 70 mg/dL.

The primary endpoint of safety (any adverse event) was similar in the two groups (78.5 versus 78.7 percent, respectively), as was the endpoint of serious adverse events (14.5 versus 14.0, respectively). The most common adverse events included nasopharyngitis, myalgia, and upper respiratory tract infection after 52 weeks of therapy. At week 12, bempedoic acid reduced the mean LDL-C by 19.2 mg per deciliter (0.50 mmol per liter), representing a change of -16.5 percent from baseline (difference versus placebo in change from baseline, -18.1 percent; p<0.001). There was a trend toward a greater LDL-C lowering response of bempedoic acid patients receiving low- to moderate-intensity statin (-20.0 percent) compared with high-intensity statin (-17.5 percent).

The CLEAR Wisdom trial enrolled 779 patients receiving maximally-tolerated lipid-lowering therapies and whose baseline LDL-C was at least 100 mg/dL [29]. The primary efficacy endpoint of the percent change from baseline to week 12 in LDL-C was significantly lower with bempedoic acid compared with placebo (-15.1 versus 2.4 percent; 97.6 versus 122.8 mg/dL, respectively). Adverse events were similar to CLEAR Harmony and included an elevated uric acid level in a higher percent of patients with active treatment (4.2 versus 1.9 percent).

We measure serum uric acid and stabilize patients with active gout before starting bempedoic acid.

In February 2020, the US Food and Drug Administration (FDA) approved the drug for the treatment of adults with heterozygous familial hypercholesterolemia or established atherosclerotic cardiovascular disease who require additional lowering of LDL-C [30]. A combination single tablet of bempedoic acid-ezetimibe has also been approved.

Monoclonal antibody against ANGPTL3 — Angiopoietin-like proteins (ANGPTLs) are regulators of lipoprotein metabolism. ANGPTL3 is a hormone produced by the liver that inhibits lipoprotein lipase, an enzyme that breaks down plasma triglycerides (see "Lipoprotein classification, metabolism, and role in atherosclerosis", section on 'Endogenous pathway of lipid metabolism'). In addition, it lowers LDL-C using a low-density lipoprotein-independent mechanism [31].

Evinacumab is a fully human monoclonal antibody against ANGPTL3, and it has been shown to significantly decrease LDL-C when given either subcutaneously or intravenously. A 2020 phase 2 trial randomly assigned 272 patients with refractory hypercholesterolemia and either heterozygous familial hypercholesterolemia (FH) or non-heterozygous FH with clinical atherosclerotic cardiovascular disease to various doses of evinacumab or placebo [32]. All patients were treated with a PCSK9 inhibitor and a statin at a maximum-tolerated dose, with or without ezetimibe. At week 16, evinacumab significantly reduced LDL-C by 50 percent with intravenous (IV) administration (15 mg/kg IV every four weeks) and 56 percent with subcutaneous administration (450 mg subcutaneously every week). Through its effects on endothelial lipase, evinacumab reduces hepatic very-low-density lipoprotein cholesterol production and secretion and, consequently, LDL-C [33]. (See "Lipoprotein classification, metabolism, and role in atherosclerosis", section on 'Very low-density lipoprotein'.)

The potential use of monoclonal antibodies to decrease ANGLPT3 in patients with homozygous FH is discussed separately. (See "Familial hypercholesterolemia in adults: Treatment", section on 'Homozygous individuals'.)

Fibrates — We do not recommend fibrate therapy, also known as "fibrates" or "fibric acid," to lower LDL-C or to raise high-density lipoprotein cholesterol (HDL-C) in the absence of hypertriglyceridemia. Fibrates lower triglyceride levels by as much as 50 percent and reduce LDL particle number, but the available evidence does not support their routine use as add-on drugs to statin therapy [34]. Unlike statins, which have demonstrated clinical efficacy across a broad range of LDL-C levels, fibrates have shown reductions in cardiovascular events primarily in subsets of patients with high triglycerides (above 200 mg/dL [2.2 mmol/L]) and/or low HDL-C (below 40 mg/dL [1.0 mmol/L]) [35-37]. However, a prospective randomized, placebo-controlled trial of pemafibrate in dyslipidemic patients was unaccompanied by a reduction in cardiovascular outcomes [38]. Thus, we do not recommend fibrates for cardiovascular event reduction. The potential role for fibrate therapy in patients with hypertriglyceridemia to prevent acute pancreatitis is discussed separately. (See "Hypertriglyceridemia in adults: Management", section on 'Fibrates'.)

The impact of fibrates on the lipid profile are a reduction of serum triglycerides by as much as 50 percent and an elevation of HDL-C by 5 to 20 percent (table 2) [37,39]. Increases in HDL-C of as much as 20 percent are seen in patients with very high triglyceride levels (>500 mg/dL [5.7 mmol/L]), while increases of 5 percent are more typical with fibrate monotherapy in patients with lower triglyceride levels. Fibrates reduce LDL particle number (mean decrease of 10 percent) despite a mild increase in LDL-C (mean increase of 6 percent) [40-43]. Fibrates have variable effects on serum lipoprotein(a) (Lp(a)): In one study, as an example, bezafibrate reduced serum Lp(a) by approximately 26 percent overall and by 39 percent in patients with values above 30 mg/dL [44], but most studies show no effect on Lp(a) levels.

Nicotinic acid (niacin) — Nicotinic acid (niacin) is uncommonly used as therapy for patients with elevated LDL-C. Other therapies such as ezetimibe or a PCSK9 inhibitor are added to statin therapy in patients for whom a further reduction in LDL-C is needed. While nicotinic acid raises HDL-C significantly, there is no evidence that this use leads to improved patient outcomes [45-47]. In addition, the use of nicotinic acid is often limited by poor tolerability, and there are concerns about the safety of nicotinic acid as well as its efficacy for clinical end points.

Niacin lowers lipoprotein(a) (Lp(a)) levels by an average 25 percent [48], and thus it is used for patients with Lp(a) excess and hypercholesterolemia who have elevated LDL-C levels on treatment with maximum tolerated statin therapy plus ezetimibe [48]. (See "Lipoprotein(a)", section on 'Next steps' and "Lipoprotein(a)", section on 'Rarely used drugs'.) There is no evidence that a reduction in Lp(a) with niacin reduces clinical events.

Large randomized trials of niacin in patients with low HDL-C levels and LDL-C levels of around 70 mg/dL have raised serious concerns about its safety and efficacy in combination with statin therapy, and by extension concerns about niacin monotherapy:

The AIM-HIGH trial of 3414 patients with established cardiovascular disease, low HDL-C, elevated triglycerides, and treated with a statin (mean LDL-C of 70 mg/dL) found no additional benefit to treatment with extended-release niacin [49]. The study was stopped early for futility and because of a concern about increased numbers of ischemic strokes in patients treated with niacin, which were not significantly different from placebo after final adjudication. However, HDL-C levels in the "placebo" arm (which received 100 to 200 mg of niacin daily) increased more than expected, which may have reduced the ability of the trial to detect a real benefit with niacin therapy. (See "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

The AIM-HIGH trial does not answer the question of whether niacin would be of value in patients on statin therapy but much higher initial LDL-C levels.

HPS2-THRIVE randomly assigned 25,673 adults ages 50 to 80 with vascular disease to receive extended–release niacin 2 grams daily plus laropiprant (to reduce flushing from niacin) or placebo; all patients received simvastatin 40 mg daily, and if LDL-C reduction was inadequate with simvastatin, ezetimibe 10 mg daily was added [50]. After a median follow-up of 3.9 years, there was no reduction with niacin/laropiprant in the primary end point of first major vascular event (13.2 versus 13.7 percent; risk ratio [RR] 0.96, 95% CI 0.90-1.03) and there was also no benefit for this end point in the subgroup of patients with low HDL-C and elevated triglycerides. There was a statistically nonsignificant increase in mortality (6.2 versus 5.7 percent, RR 1.09, CI 0.99-1.21). Additionally, and despite the run-in period, there was an increase in serious adverse events. These included myopathy (RR 3.54), but with a much greater increased risk in patients from study centers in China (RR 5.2) than in Europe (RR 1.5), gastrointestinal side effects, and rash. Niacin/laropiprant worsened glucose control with both an increase in new cases of diabetes (RR 1.32) and serious disturbance in diabetes control (11.5 versus 7.5 percent, RR 1.55; most of these led to hospitalization). Additionally, there were unanticipated increases in serious infections (8.0 versus 6.6 percent; RR 1.22, CI 1.12-1.34) and bleeding (2.5 versus 1.9 percent; RR 1.38, CI 1.17-1.62).

Niacin was administered with laropiprant in HPS2-THRIVE, and so it is not possible to completely sort out the effects that might have been due to laropiprant, particularly with regard to the unexpected findings of excess bleeding and infection. However, given the results of HPS2-THRIVE, infection and bleeding were subsequently analyzed in AIM-HIGH, which studied niacin without laropiprant [51]. Infections were increased with niacin (8.1 versus 5.8 percent; p = 0.008), and there were only small numbers to assess rates of bleeding (3.4 versus 2.9 percent; p = 0.36). This explanation for the observation of an increased infection risk is not known [52].

Given these results of these randomized trials, we would not administer niacin to most patients receiving statin therapy. Exceptions to this general approach would include patients at extremely high cardiovascular risk such as homozygous or heterozygous familial hypercholesterolemia patients who are not at goal with maximum dosage statin plus ezetimibe or colesevelam, particularly if they have Lp(a) excess and a family history of early onset of atherosclerotic cardiovascular disease. Patients who are unable to take other lipid-lowering therapies may consider long-term niacin therapy if their LDL-C is significantly elevated and is lowered substantially by a trial of niacin treatment.

Niacin has a role in patients with high Lp(a) levels. (See "Lipoprotein(a)", section on 'Management' and "Lipoprotein(a)", section on 'Rarely used drugs'.)

While niacin can raise HDL-C, we generally do not recommend efforts to do so. (See "HDL cholesterol: Clinical aspects of abnormal values", section on 'Role of niacin, fibrates, or estrogen'.)

Use — The HDL-C raising properties of nicotinic acid occur with dosages as low as 1 to 1.5 g/day [46]. In contrast, the very-low-density lipoprotein cholesterol and LDL-C lowering effects are typically seen with higher doses (3 g/day) [45,47]. In one study, for example, nicotinic acid in a dose of 500 mg three times daily raised HDL-C by 20 percent but reduced LDL-C by only 5 percent. In comparison, a higher dose of 1.5 g three times daily produced more prominent changes of 33 percent (HDL-C elevation) and 23 percent (LDL-C reduction). At higher doses, nicotinic acid can also lower Lp(a) levels by as much as 35 percent [47].

For those patients in whom niacin is chosen, therapy with crystalline nicotinic acid is initiated at 100 mg three times daily and gradually increased to the targeted dosage as tolerated [46]. (See 'Bile acid sequestrants' below.)

Extended-release niacin, is a controlled release formulation of nicotinic acid that is administered once daily. It is initiated at a dose of 500 mg nightly for one month and the dosage is titrated to 1000 mg. The standard dosage is 1 to 2 grams nightly. It is advised that the medication be given with a night-time snack, but our experience suggests improved tolerability with dosing after the evening meal.

Side effects — The use of nicotinic acid is often limited by poor tolerability. At standard doses (1.5 to 4.5 g/day), flushing occurs in 80 percent of patients taking the crystalline preparation, and pruritus, paresthesias, and nausea each occur in about 20 percent [47]. Symptoms after a dose of nicotinic acid can last from 10 to 20 minutes up to several hours.

With the crystalline preparation, pretreatment with aspirin (325 to 650 mg) 30 minutes prior to dosing or ibuprofen (200 mg) 60 minutes prior to dosing can minimize flushing and other prostaglandin-mediated side effects associated with niacin. This adverse reaction often diminishes in 7 to 10 days, gradually improving over time. Symptoms may recur episodically on a stable dose or with dose titration. Nicotinic acid is better tolerated when ingested with food, which minimizes gastrointestinal side effects.

Flushing appears to be less common with controlled-release Niaspan. In one study of 269 patients receiving a median dose of 2000 mg/day for 48 weeks, 4.8 percent discontinued the drug because of flushing [53]. In another report in which both formulations were given in a dose of 1500 mg/day for four months, Niaspan was accompanied by fewer flushing episodes per month (1.9 versus 8.6) [54]. A newer formulation of Niaspan (coated caplets) appears to further reduce flushing [55].

Laropiprant is a highly selective prostaglandin D2 receptor 1 antagonist that can decrease niacin-induced flushing [56,57]. It has not been approved in the United States, and this agent was withdrawn from the market worldwide after the results of HPS2-THRIVE. (See 'Nicotinic acid (niacin)' above.)

Additionally, although the absolute increase in risk appears to be small, these results suggest that niacin may increase the risk of myopathy in patients receiving simvastatin, and perhaps other statins (see "Statin muscle-related adverse events", section on 'Concurrent drug therapy'). This increased risk may be a particular concern in patients from China, and we suggest avoiding the combination of simvastatin and niacin in Chinese patients and only using niacin with caution in patients receiving other statins. (See "Statin muscle-related adverse events", section on 'Niacin'.)

Elevations in hepatocellular enzymes are also common with nicotinic acid and may lead to severe hepatotoxicity, jaundice, and fulminant hepatitis. The onset of hepatocellular injury is not predictable; therefore, regular monitoring of biochemical studies is mandatory. Crystalline niacin is preferred to most sustained-release preparations, since the former is associated with a greater hypolipidemic effect and seemingly less hepatotoxicity [46,58]. An exception may be extended release Niaspan, which has been found to minimally raise transaminases in clinical trials, but not cause significant hepatotoxicity [59].

Other important problems with nicotinic acid include [46]:

Nicotinic acid can increase glucose levels, particularly at moderate to high doses [60]. As a result, hyperglycemia may develop in susceptible patients and the glycemic state may be worsened in those already being treated for overt diabetes mellitus [61,62]. This effect appears to be greatest with some extended-release preparations, and minimized with crystalline niacin and perhaps Niaspan. (See "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

Nicotinic acid can induce hyperuricemia and precipitate acute gouty arthritis; it should therefore be avoided in any patient with a history of gout.

Nicotinic acid can produce hypotension in subjects treated with vasodilators, and can exacerbate unstable angina pectoris [63].

Nicotinic acid causes a dose-dependent elevation in plasma homocysteine levels that may negate its favorable effects on the lipid profile in certain subsets of patients [64]. (See "Overview of homocysteine".)

As discussed above, nicotinic acid appears to increase the risk of infection and perhaps bleeding. (See 'Nicotinic acid (niacin)' above.)

Bile acid sequestrants — Bile acid sequestrants are rarely used for elevated LDL-C. This category of drug includes cholestyramine, colestipol, and colesevelam [65]. These agents bind bile acids in the intestine, resulting in interruption of the reabsorption of bile acids which is usually 90 percent efficient. The ensuing reduction in the cholesterol pool lowers intrahepatic cholesterol, which promotes the synthesis of apo B/E (LDL) receptors. The apo B/E (LDL) receptors bind LDL from the plasma, causing a further reduction in blood cholesterol. The bile acid sequestrants also induce a minimal elevation in HDL-C [66]. (See "Lipoprotein classification, metabolism, and role in atherosclerosis".)

Low doses (8 g/day of cholestyramine or 10 g/day of colestipol) can reduce LDL-C by 10 to 15 percent. A more pronounced reduction (about 24 percent) can be achieved at maximal recommended doses (24 and 30 g/day, respectively). A similar reduction in LDL-C can be achieved with 1.5 to 4.5 g/day of colesevelam [65,67].

Bile acid sequestrants are also effective when used in combination with statins or nicotinic acid in patients with markedly elevated plasma levels of LDL-C. As an example, bile acid sequestrants and statins have synergistic actions to lower LDL-C (about 50 percent) and raise HDL–C (11 to 18 percent) [68-70]. Maximal doses of bile acid sequestrants with nicotinic acid (4 g/day) can reduce LDL-C by 32 percent and elevate HDL-C by 43 percent [68].

In addition to the relatively minor (compared with statin) effect on LDL-C, the use of a bile acid sequestrant is often limited by side effects. The major adverse reactions are gastrointestinal, including nausea, bloating, cramping, and an increase in liver enzymes; colesevelam is better tolerated and less likely to cause gastrointestinal side effects [65]. Bile acid sequestrants can also bind to and impair the absorption of other drugs, such as digoxin, warfarin, and fat soluble vitamins (table 1). This effect can be minimized by administering the other drugs one hour before or four hours after the bile acid sequestrants.

Probucol — Probucol is of historical interest. It modestly lowers LDL-C, but more prominently reduces HDL-C (table 2). It is not available for clinical use. (See "Lipoprotein classification, metabolism, and role in atherosclerosis", section on 'Lipoproteins and atherosclerosis'.)

Estrogen replacement therapy — We do not prescribe estrogen replacement therapy (ERT) for the purpose of improving the lipid profile. ERT in postmenopausal women may lead to reductions in LDL-C (15 percent) and Lp(a) (20 percent) and elevations in HDL-C (10 to 15 percent) and triglycerides (24 percent) [71-74]. (See "Menopausal hormone therapy and cardiovascular risk", section on 'Lipids'.)

It had been thought that these lipid changes might contribute to a cardioprotective effect of estrogen replacement therapy. However, such a benefit was not confirmed in the Women's Health Initiative, mostly of primary prevention, and the HERS trials of secondary prevention. Estrogen-progestin replacement had no cardioprotective effect and may have produced harm. (See "Menopausal hormone therapy and cardiovascular risk".)

Thyromimetics — Eprotirome appears to be able to reduce serum lipid levels. Long-term studies with clinical end points are required to determine whether eprotirome provides clinical benefits in patients with hypercholesterolemia.

Eprotirome is a thyroid hormone analog that has minimal uptake in nonhepatic tissue [75]. A trial in 184 patients, nearly all without known cardiovascular disease, who were receiving statin therapy but had an LDL-C ≥116 mg/dL (3.0 mmol/L) randomly assigned patients to three different doses of eprotirome or to placebo [75]. After 12 weeks, eprotirome reduced LDL-C in a dose-dependent fashion; at a dose of 100 mcg daily, LDL-C was reduced by 32 percent, compared with a 7 percent reduction with placebo. Similar changes were seen in levels of triglycerides, lipoprotein(a), and apolipoprotein B. Clinical end points were not assessed, and, over the 12 weeks, eprotirome did not appear to produce clinical hyperthyroidism, hypothyroidism, or have adverse effects on the heart or bone; some patients treated with eprotirome did have elevations in serum aminotransferase levels.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Lipid disorders in adults".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Beyond the Basics topics (see "Patient education: High cholesterol and lipids (Beyond the Basics)" and "Patient education: High cholesterol and lipid treatment options (Beyond the Basics)")

SUMMARY

Although statins are the preferred therapy for most patients requiring treatment of dyslipidemia, other agents are available with varying levels of evidence for clinical benefits. (See "Statins: Actions, side effects, and administration".)

In patients who do not achieve the desired low-density lipoprotein cholesterol (LDL-C) goal with statin therapy, we add ezetimibe more frequently than any other lipid altering drug. (See 'Ezetimibe' above.)

The principal use of fibrate therapy is in the management of patients with hypertriglyceridemia. (See "Hypertriglyceridemia in adults: Management", section on 'Fibrates'.)

Nicotinic acid (niacin) is uncommonly used in the treatment of LDL-C. It can be used to lower lipoprotein(a). (see "Lipoprotein(a)", section on 'Next steps' and "Lipoprotein(a)")

ACKNOWLEDGMENT — The UpToDate editorial staff thank John J P Kastelein, MD, PhD, FESC, who contributed to an earlier version of this topic review.

  1. Rosenson RS, Hegele RA, Koenig W. Cholesterol-Lowering Agents. Circ Res 2019; 124:364.
  2. Sudhop T, Lütjohann D, Kodal A, et al. Inhibition of intestinal cholesterol absorption by ezetimibe in humans. Circulation 2002; 106:1943.
  3. Altmann SW, Davis HR Jr, Zhu LJ, et al. Niemann-Pick C1 Like 1 protein is critical for intestinal cholesterol absorption. Science 2004; 303:1201.
  4. Klett EL, Patel SB. Biomedicine. Will the real cholesterol transporter please stand up. Science 2004; 303:1149.
  5. Temel RE, Tang W, Ma Y, et al. Hepatic Niemann-Pick C1-like 1 regulates biliary cholesterol concentration and is a target of ezetimibe. J Clin Invest 2007; 117:1968.
  6. Myocardial Infarction Genetics Consortium Investigators, Stitziel NO, Won HH, et al. Inactivating mutations in NPC1L1 and protection from coronary heart disease. N Engl J Med 2014; 371:2072.
  7. Dujovne CA, Ettinger MP, McNeer JF, et al. Efficacy and safety of a potent new selective cholesterol absorption inhibitor, ezetimibe, in patients with primary hypercholesterolemia. Am J Cardiol 2002; 90:1092.
  8. Knopp RH, Gitter H, Truitt T, et al. Effects of ezetimibe, a new cholesterol absorption inhibitor, on plasma lipids in patients with primary hypercholesterolemia. Eur Heart J 2003; 24:729.
  9. Hegele RA, Guy J, Ban MR, Wang J. NPC1L1 haplotype is associated with inter-individual variation in plasma low-density lipoprotein response to ezetimibe. Lipids Health Dis 2005; 4:16.
  10. Gagné C, Gaudet D, Bruckert E, Ezetimibe Study Group. Efficacy and safety of ezetimibe coadministered with atorvastatin or simvastatin in patients with homozygous familial hypercholesterolemia. Circulation 2002; 105:2469.
  11. Gagné C, Bays HE, Weiss SR, et al. Efficacy and safety of ezetimibe added to ongoing statin therapy for treatment of patients with primary hypercholesterolemia. Am J Cardiol 2002; 90:1084.
  12. Davidson MH, McGarry T, Bettis R, et al. Ezetimibe coadministered with simvastatin in patients with primary hypercholesterolemia. J Am Coll Cardiol 2002; 40:2125.
  13. Melani L, Mills R, Hassman D, et al. Efficacy and safety of ezetimibe coadministered with pravastatin in patients with primary hypercholesterolemia: a prospective, randomized, double-blind trial. Eur Heart J 2003; 24:717.
  14. Goldberg AC, Sapre A, Liu J, et al. Efficacy and safety of ezetimibe coadministered with simvastatin in patients with primary hypercholesterolemia: a randomized, double-blind, placebo-controlled trial. Mayo Clin Proc 2004; 79:620.
  15. Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387.
  16. Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 2011; 377:2181.
  17. Rossebø AB, Pedersen TR, Boman K, et al. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N Engl J Med 2008; 359:1343.
  18. Peto R, Emberson J, Landray M, et al. Analyses of cancer data from three ezetimibe trials. N Engl J Med 2008; 359:1357.
  19. Ballantyne CM, Houri J, Notarbartolo A, et al. Effect of ezetimibe coadministered with atorvastatin in 628 patients with primary hypercholesterolemia: a prospective, randomized, double-blind trial. Circulation 2003; 107:2409.
  20. Kosoglou T, Statkevich P, Fruchart JC, et al. Pharmacodynamic and pharmacokinetic interaction between fenofibrate and ezetimibe. Curr Med Res Opin 2004; 20:1197.
  21. Gustavson LE, Schweitzer SM, Burt DA, et al. Evaluation of the potential for pharmacokinetic interaction between fenofibrate and ezetimibe: A phase I, open-label, multiple-dose, three-period crossover study in healthy subjects. Clin Ther 2006; 28:373.
  22. Farnier M, Freeman MW, Macdonell G, et al. Efficacy and safety of the coadministration of ezetimibe with fenofibrate in patients with mixed hyperlipidaemia. Eur Heart J 2005; 26:897.
  23. McKenney JM, Farnier M, Lo KW, et al. Safety and efficacy of long-term co-administration of fenofibrate and ezetimibe in patients with mixed hyperlipidemia. J Am Coll Cardiol 2006; 47:1584.
  24. Ballantyne CM, Davidson MH, Macdougall DE, et al. Efficacy and safety of a novel dual modulator of adenosine triphosphate-citrate lyase and adenosine monophosphate-activated protein kinase in patients with hypercholesterolemia: results of a multicenter, randomized, double-blind, placebo-controlled, parallel-group trial. J Am Coll Cardiol 2013; 62:1154.
  25. Thompson PD, Rubino J, Janik MJ, et al. Use of ETC-1002 to treat hypercholesterolemia in patients with statin intolerance. J Clin Lipidol 2015; 9:295.
  26. Ballantyne CM, Banach M, Mancini GBJ, et al. Efficacy and safety of bempedoic acid added to ezetimibe in statin-intolerant patients with hypercholesterolemia: A randomized, placebo-controlled study. Atherosclerosis 2018; 277:195.
  27. Nissen SE, Lincoff AM, Brennan D, et al. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353.
  28. Ray KK, Bays HE, Catapano AL, et al. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med 2019; 380:1022.
  29. Goldberg AC, Leiter LA, Stroes ESG, et al. Effect of Bempedoic Acid vs Placebo Added to Maximally Tolerated Statins on Low-Density Lipoprotein Cholesterol in Patients at High Risk for Cardiovascular Disease: The CLEAR Wisdom Randomized Clinical Trial. JAMA 2019; 322:1780.
  30. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/211616s000lbl.pdf.
  31. Banerjee P, Chan KC, Tarabocchia M, et al. Functional Analysis of LDLR (Low-Density Lipoprotein Receptor) Variants in Patient Lymphocytes to Assess the Effect of Evinacumab in Homozygous Familial Hypercholesterolemia Patients With a Spectrum of LDLR Activity. Arterioscler Thromb Vasc Biol 2019; 39:2248.
  32. Rosenson RS, Burgess LJ, Ebenbichler CF, et al. Evinacumab in Patients with Refractory Hypercholesterolemia. N Engl J Med 2020; 383:2307.
  33. Adam RC, Mintah IJ, Alexa-Braun CA, et al. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res 2020; 61:1271.
  34. Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation 2011; 123:2292.
  35. Frick MH, Elo O, Haapa K, et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med 1987; 317:1237.
  36. Bezafibrate Infarction Prevention (BIP) study. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation 2000; 102:21.
  37. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 1999; 341:410.
  38. Das Pradhan A, Glynn RJ, Fruchart JC, et al. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med 2022; 387:1923.
  39. Keech A, Simes RJ, Barter P, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005; 366:1849.
  40. Rosenson RS, Wolff DA, Huskin AL, et al. Fenofibrate therapy ameliorates fasting and postprandial lipoproteinemia, oxidative stress, and the inflammatory response in subjects with hypertriglyceridemia and the metabolic syndrome. Diabetes Care 2007; 30:1945.
  41. Otvos JD, Collins D, Freedman DS, et al. Low-density lipoprotein and high-density lipoprotein particle subclasses predict coronary events and are favorably changed by gemfibrozil therapy in the Veterans Affairs High-Density Lipoprotein Intervention Trial. Circulation 2006; 113:1556.
  42. Rosenson RS. Fenofibrate: treatment of hyperlipidemia and beyond. Expert Rev Cardiovasc Ther 2008; 6:1319.
  43. Rosenson RS, Underberg JA. Systematic review: Evaluating the effect of lipid-lowering therapy on lipoprotein and lipid values. Cardiovasc Drugs Ther 2013; 27:465.
  44. Bimmermann A, Boerschmann C, Schwartzkopff W, et al. Effective therapeutic measures for reducing lipoprotein(a) in patients with dyslipidemia. Lipoprotein(a) reduction with sustained-release bezafibrate. Curr Ther Res 1991; 49:635.
  45. Grundy SM, Mok HY, Zech L, Berman M. Influence of nicotinic acid on metabolism of cholesterol and triglycerides in man. J Lipid Res 1981; 22:24.
  46. Probstfield JL, Hunninghake DB. Nicotinic acid as a lipoprotein-altering agent. Therapy directed by the primary physician. Arch Intern Med 1994; 154:1557.
  47. Illingworth DR, Stein EA, Mitchel YB, et al. Comparative effects of lovastatin and niacin in primary hypercholesterolemia. A prospective trial. Arch Intern Med 1994; 154:1586.
  48. Stein EA, Raal F. Future Directions to Establish Lipoprotein(a) as a Treatment for Atherosclerotic Cardiovascular Disease. Cardiovasc Drugs Ther 2016; 30:101.
  49. AIM-HIGH Investigators, Boden WE, Probstfield JL, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255.
  50. HPS2-THRIVE Collaborative Group, Landray MJ, Haynes R, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014; 371:203.
  51. Anderson TJ, Boden WE, Desvigne-Nickens P, et al. Safety profile of extended-release niacin in the AIM-HIGH trial. N Engl J Med 2014; 371:288.
  52. Lloyd-Jones DM. Niacin and HDL cholesterol--time to face facts. N Engl J Med 2014; 371:271.
  53. Guyton JR, Goldberg AC, Kreisberg RA, et al. Effectiveness of once-nightly dosing of extended-release niacin alone and in combination for hypercholesterolemia. Am J Cardiol 1998; 82:737.
  54. Knopp RH, Alagona P, Davidson M, et al. Equivalent efficacy of a time-release form of niacin (Niaspan) given once-a-night versus plain niacin in the management of hyperlipidemia. Metabolism 1998; 47:1097.
  55. Cefali EA, Simmons PD, Stanek EJ, Shamp TR. Improved control of niacin-induced flushing using an optimized once-daily, extended-release niacin formulation. Int J Clin Pharmacol Ther 2006; 44:633.
  56. Maccubbin D, Koren MJ, Davidson M, et al. Flushing profile of extended-release niacin/laropiprant versus gradually titrated niacin extended-release in patients with dyslipidemia with and without ischemic cardiovascular disease. Am J Cardiol 2009; 104:74.
  57. Maccubbin D, Bays HE, Olsson AG, et al. Lipid-modifying efficacy and tolerability of extended-release niacin/laropiprant in patients with primary hypercholesterolaemia or mixed dyslipidaemia. Int J Clin Pract 2008; 62:1959.
  58. Etchason JA, Miller TD, Squires RW, et al. Niacin-induced hepatitis: a potential side effect with low-dose time-release niacin. Mayo Clin Proc 1991; 66:23.
  59. Goldberg A, Alagona P Jr, Capuzzi DM, et al. Multiple-dose efficacy and safety of an extended-release form of niacin in the management of hyperlipidemia. Am J Cardiol 2000; 85:1100.
  60. Grundy SM, Vega GL, McGovern ME, et al. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Arch Intern Med 2002; 162:1568.
  61. Garg A, Grundy SM. Nicotinic acid as therapy for dyslipidemia in non-insulin-dependent diabetes mellitus. JAMA 1990; 264:723.
  62. Rosenson RS. Measure for measure--sugar or fats? Reconciling cardiovascular and diabetes risk with niacin therapy. Nat Clin Pract Endocrinol Metab 2007; 3:72.
  63. Pasternak RC, Kolman BS. Unstable myocardial ischemia after the initiation of niacin therapy. Am J Cardiol 1991; 67:904.
  64. Garg R, Malinow M, Pettinger M, et al. Niacin treatment increases plasma homocyst(e)ine levels. Am Heart J 1999; 138:1082.
  65. Davidson MH, Dillon MA, Gordon B, et al. Colesevelam hydrochloride (cholestagel): a new, potent bile acid sequestrant associated with a low incidence of gastrointestinal side effects. Arch Intern Med 1999; 159:1893.
  66. Shepherd J, Packard CJ, Morgan HG, et al. The effects of cholestyramine on high density lipoprotein metabolism. Atherosclerosis 1979; 33:433.
  67. Insull W Jr, Toth P, Mullican W, et al. Effectiveness of colesevelam hydrochloride in decreasing LDL cholesterol in patients with primary hypercholesterolemia: a 24-week randomized controlled trial. Mayo Clin Proc 2001; 76:971.
  68. Brown G, Albers JJ, Fisher LD, et al. Regression of coronary artery disease as a result of intensive lipid-lowering therapy in men with high levels of apolipoprotein B. N Engl J Med 1990; 323:1289.
  69. Comparative efficacy and safety of pravastatin and cholestyramine alone and combined in patients with hypercholesterolemia. Pravastatin Multicenter Study Group II. Arch Intern Med 1993; 153:1321.
  70. Knapp HH, Schrott H, Ma P, et al. Efficacy and safety of combination simvastatin and colesevelam in patients with primary hypercholesterolemia. Am J Med 2001; 110:352.
  71. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA 1995; 273:199.
  72. Kim CJ, Jang HC, Cho DH, Min YK. Effects of hormone replacement therapy on lipoprotein(a) and lipids in postmenopausal women. Arterioscler Thromb 1994; 14:275.
  73. Darling GM, Johns JA, McCloud PI, Davis SR. Estrogen and progestin compared with simvastatin for hypercholesterolemia in postmenopausal women. N Engl J Med 1997; 337:595.
  74. Binder EF, Williams DB, Schechtman KB, et al. Effects of hormone replacement therapy on serum lipids in elderly women. a randomized, placebo-controlled trial. Ann Intern Med 2001; 134:754.
  75. Ladenson PW, Kristensen JD, Ridgway EC, et al. Use of the thyroid hormone analogue eprotirome in statin-treated dyslipidemia. N Engl J Med 2010; 362:906.
Topic 4562 Version 65.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟