Lipid-Modifying Drugs





Serum cholesterol level is known to be related to incident atherosclerotic cardiovascular disease (ASCVD), with low-density lipoprotein cholesterol (LDL-C) found to be a dominant contributor to atherosclerosis. Multiple large landmark randomized controlled trials of lipid-lowering therapy have consistently shown that LDL-C lowering reduces risk of incident ASCVD. The widespread availability, convincing clinical evidence, and relative safety of the statins have established pharmacologic control of blood lipids as an increasingly acceptable strategy. Furthermore, aggressive reduction in LDL-C, via intensified statin therapy, has been shown to yield greater reduction in cardiovascular morbidity and mortality. Therefore, measurement of cholesterol level, especially LDL-C, is an important step both for assessing cardiovascular risk and as an indicator of effectiveness of lipid-lowering therapy.


Cardiovascular risk assessment involves a thorough knowledge of both the serum lipid profile and other “traditional” risk factors that have been shown to play a pivotal role in atherosclerosis. The Pooled Cohort Equation, incorporated first in the 2013 American College of Cardiology (ACC)/American Heart Association (AHA) cholesterol guidelines, is a risk prediction tool that integrates the major “traditional” risk factors, which include cigarette smoking, hypertension, dysglycemia, and advancing age along with blood lipid profile, to calculate 10-year risk for ASCVD ( Table 6.1 ). This equation has been derived from five community-based cohorts that provide a broad and diverse representative sample of the U.S. population. The European guidelines recommend the use of the Systematic Coronary Risk Evaluation (SCORE) system for 10-year ASCVD risk prediction, as it was derived from a large representative European cohort data set. The patient’s absolute risk for developing cardiovascular disease (CVD) in the next 10 years determines the aggressiveness of lipid intervention. Since the 2013 ACC/AHA guidelines were published, immense research in individualized risk assessment has resulted in the emergence of several additional risk factors that are strongly associated with atherosclerosis and confer a higher-risk state. These “risk-enhancing factors” (see Table 6.1 ), as defined in the 2018 AHA/ACC guidelines for the management of blood cholesterol, allow for more individualized risk assessment and care for patients. Conditions associated with systemic inflammation, e.g., metabolic syndrome, chronic renal disease, and elevated high-sensitivity C-reactive protein (hs-CRP), contribute to the pathogenesis of atherosclerosis and predispose to atherosclerotic events. Additionally, certain individual characteristics, such as premature menopause, certain ethnicities, and family history of premature ASCVD, confer higher risk. In addition to the standard lipid profile, two additional lipid-related measures, apolipoprotein B (apoB) and lipoprotein(a) [Lp(a)], can also be useful in risk assessment, especially in circumstances such as hypertriglyceridemia and/or LDL-C > 160 mg/dL. Additional characteristics that can increase risk mentioned in the 2019 European Society of Cardiology (ESC)/European Atherosclerosis Society (EAS) Guidelines for the Management of Dyslipidaemias include social deprivation, obesity and central obesity, physical inactivity, psychosocial stress including vital exhaustion, major psychiatric disorders, atrial fibrillation, left ventricular hypertrophy, obstructive sleep apnea syndrome, and nonalcoholic fatty liver disease.



Table 6.1

Traditional risk factors and risk-enhancing factors for ASCVD

Adapted from AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the management of blood cholesterol, page 34.











Traditional risk factors (used in the Pooled-cohort Equation)



  • Age



  • Cigarette smoking



  • Blood pressure



  • Presence or absence of diabetes mellitus



  • Serum TC



  • Serum HDL-C

Risk-enhancing factors



  • Family history of premature ASCVD (males, age < 55 years; females, age < 65 years)



  • Primary hypercholesterolemia (LDL-C, 160–189 mg/dL [4.1–4.8 mmol/L); non–HDL-C 190–219 mg/dL [4.9–5.6 mmol/L])



  • Metabolic syndrome



  • CKD



  • Chronic inflammatory conditions such as such as psoriasis, RA, or HIV/AIDS



  • ABI < 0.9



  • History of premature menopause (before age 40) and history of pregnancy-associated conditions that increase later ASCVD risk such as preeclampsia



  • High-risk race/ethnicities (e.g., South Asian ancestry)



  • Lipid/biomarkers: Associated with increased ASCVD




    • Persistently elevated, primary hypertriglyceridemia (≥ 175 mg/dL);



    • If measured:




      • Elevated high-sensitivity C-reactive protein (≥ 2.0 mg/L)



      • Elevated Lp(a) (≥ 50 mg/dL or ≥ 125 nmol/L)



      • Elevated apoB (≥ 130 mg/dL)




ABI, Ankle-brachial index; AIDS; Acquired immunodeficiency syndrome; apoB, apolipoprotein B; ASCVD, atherosclerotic cardiovascular disease; CKD, chronic kidney disease; eGFR, estimated glomerular filtration rate; HDL-C, high-density lipoprotein cholesterol; HIV: human immunodeficiency virus; LDL-C, low-density lipoprotein cholesterol; Lp(a), lipoprotein (a).


Physicians may help guide younger patients toward long-term cardiovascular health by addressing early risk factors, whereas middle-aged and older patients may need a more-intensive approach because of their near-term risk for coronary heart disease (CHD). Effective strategy for lipid-lowering therapy therefore involves the following important considerations: (1) detailed evaluation of individualized risk for CVD based on lipid parameters as well as genetic and acquired risk factors; (2) review of lifestyle habits (e.g., diet, exercise, tobacco use) and development of individualized recommendations regarding healthy diet and body mass index and regular physical activity; (3) potential benefit of high-intensity therapy to achieve very low LDL-C levels, i.e., “lower is better ”; and (4) growing recognition of newer lipid-lowering therapies and their role in CVD risk reduction. These developments and their translation into clinical practice hold the potential to improve patient outcomes.


Lipid-modifying therapy encompasses several classes of drugs: statins, cholesterol absorption inhibitors, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, bile acid sequestrants, fibrates, and nicotinic acid ( Fig. 6.1 ). These all have been shown to reduce LDL-C.




Fig. 6.1


Sites and targets of lipid-lowering therapies.

Several cholesterol-lowering medications act through their respective mechanisms of actions to increase the number of low-density lipoprotein receptors (LDLR). Sites of action of individual therapies in boxes. (A) Liver. Citrate is converted to acetyl-CoA (AcCoA) by ACL (ATP citrate lyase) for fatty acid (FA) and cholesterol (Chol) biosynthesis; bempedoic acid blocks its activity. Statins inhibit 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). (B) Small intestine. Absorption of dietary cholesterol into enterocytes is blocked by ezetimibe. Bile acid sequestrants bind luminal bile acids (BA) and block their enterohepatic circulation in the terminal ileum, thereby reducing cholesterol delivery to the liver. Reduced levels of intracellular cholesterol in A and B lead to upregulation of LDLR synthesis. (C) Recycling of LDLRs is increased by inhibiting proprotein convertase subtilisin kexin type 9 (PCSK9) either through monoclonal antibodies (mAB) or small interfering RNA (siRNA). ASCVD, atherosclerotic cardiovascular disease; apo B, apolipoprotein B; CAC, coronary artery calcium; CHD, coronary heart disease hsCRP , high-sensitivity C-reactive protein; LDL-C , low-density lipoprotein cholesterol; Lp(a), lipoprotein(a).


Inflammation and Atherogenesis


Atherosclerosis is characterized by a chronic inflammatory process of the arterial wall that results from unbalanced lipid accumulation and the ensuing maladaptive immune responses. Atherosclerosis is triggered when circulating LDL enters the arterial wall and is retained in the subendothelium through interaction with proteoglycans in the extracellular matrix ( Fig. 6.2 ). LDL modification within the arterial wall occurs through a series of oxidative steps, as reactive oxygen species or enzymes such as myeloperoxidase and lipoxygenases are released from inflammatory cells. Oxidized LDL in turn damages the endothelium, which stimulates an immune and inflammatory response, with increased production of chemoattractant molecules, cytokines, and adhesion molecules, driving intimal immune cell infiltration. Subsequently, the dysfunctional endothelium is more permeable to circulating monocytes and T cells; both are transported into the intima, where the monocytes are converted into macrophages. Activated macrophages and T cells release a variety of mediators that collectively exacerbate inflammation and oxidation within the vessel wall. Foam cells are formed when macrophages ingest oxidized LDL through receptors, including CD36. Elevated levels of circulating LDL therefore promote atherosclerosis and ASCVD. Patients with severe hyperlipidemia and postprandial lipemia have been shown to have lipid uptake in circulating monocytes known as “foamy monocytes,” which are activated and accelerate atherosclerosis. Growth of the atherosclerotic lesion is characterized by smooth muscle cell proliferation and increased production of matrix metalloproteinases, which can cause deterioration of elastin and collagen within the extracellular matrix. Mature plaques typically consist of a lipid-rich necrotic core encased by a weakened fibrous cap. Inflammatory cells, such as macrophages, T cells, and mast cells, produce enzymes and proinflammatory mediators, promote the deterioration of fibrous caps, and may make mature plaques more prone to rupture.




Fig. 6.2


Role of lipoproteins in atherosclerosis.

Atherosclerosis is an inflammatory process that involves atherogenic, apolipoprotein B (apoB) –containing lipoproteins (low-density lipoprotein [LDL] , triglyceride-rich lipoprotein [ TGRL ; chylomicrons, very-low-density lipoprotein, and their remnants), and lipoprotein(a) [ Lp(a) ]) and proinflammatory agents such as oxidized LDL (oxLDL) and angiotensin-II (A-II) . OxLDL is taken up by scavenger receptors (SR), and circulating monocytes infiltrate into vascular endothelium and differentiate to macrophages, which take up oxLDL and become lipid-laden foam cells. Platelet-derived growth factor (PDGF) induces smooth muscle cell proliferation, which leads to neointimal hyperplasia.


C-Reactive Protein


Much interest has centered on CRP, a general measure of inflammation that is produced in the liver in response to interleukin-6. This inflammatory marker is one of the “risk-enhancing factors” and is useful in assessment of patients at borderline or intermediate risk (5%–20% 10-year risk) according to traditional risk factors. hs-CRP level < 1 mg/L is considered low risk, and > 3 mg/L is high risk, with an approximate doubling of the relative risk compared with the low-risk category. Elevated CRP is associated with obesity and the metabolic syndrome, and levels can be reduced through weight loss, increased physical activity, and smoking cessation. The Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial (see later), which studied apparently healthy persons at increased ASCVD risk because of age, elevated hs-CRP (> 2 mg/L), and one additional ASCVD risk factor, demonstrated the utility of hs-CRP in identifying individuals at increased risk despite having low to normal levels of LDL-C. Patients in this trial who attained LDL-C levels < 50 mg/dL with rosuvastatin 20 mg daily had greater reductions in cardiovascular morbidity and mortality than the rest of the cohort.


Prevention and Risk Factors


Primary Prevention


Assessment of global CVD risk is a fundamental step in primary prevention. In general, patients without known CHD have a much lower baseline risk of CVD events than patients with known CVD, and their potential absolute risk reduction with treatment for hypercholesterolemia will usually be smaller than for patients with established CVD. Hence, the decision of whether to initiate LDL-C treatment relies heavily on determination of global CVD risk. Based on the 2018 AHA/ACC cholesterol guidelines and 10-year ASCVD risk as estimated by the Pooled Cohort Equation, adults 40–75 years of age in primary prevention can be classified as low risk (10-year risk of ASCVD < 5%), borderline risk (5% to < 7.5%), intermediate risk (7.5% to < 20%), and high risk (≥ 20%). The 2019 ESC/EAS dyslipidemia guidelines, on the other hand, divide patients into low risk (calculated SCORE < 1%), moderate risk (calculated SCORE ≥ 1% but < 5%), high risk (calculated SCORE ≥ 5% but < 10%), or very high risk (calculated SCORE ≥ 10%). Individuals with markedly elevated single risk factors such as familial dyslipidemias (LDL-C > 190 mg/dL or total cholesterol > 310 mg/dL), severe hypertension (blood pressure ≥ 180/110 mmHg), or moderate chronic kidney disease (CKD) with estimated glomerular filtration rate < 60 mL/min/1.73 m 2 are classified as high risk irrespective of SCORE in the ESC/EAS guidelines. Individuals with very high risk are treated similar to secondary prevention. Lifestyle interventions (dietary modification, smoking cessation, and physical activity) are first-line treatment and may achieve meaningful cholesterol reduction in many patients. Clinical trials of statins in the past decade have demonstrated safety and clinical event reduction across a spectrum of cardiovascular risk, even in populations with low baseline risk such as the Japanese. The current 2018 AHA/ACC guidelines recommend statin therapy along with lifestyle intervention for intermediate- to high-risk individuals; the absolute risk for developing CVD in the next 10 years determines the aggressiveness of lipid-lowering therapy. For individuals with intermediate risk, moderate-intensity statin to reduce LDL-C by 30%–49% is recommended, whereas for high risk, high-intensity statin to reduce LDL-C by ≥ 50% is recommended. The 2019 ESC/EAS dyslipidemia guidelines recommend consideration of lipid-lowering therapy in addition to lifestyle interventions in individuals at moderate risk if LDL-C remains > 100 mg/dL and in those at low risk if LDL-C remains > 116 mg/dL. In patients at high risk, statin therapy along with lifestyle intervention is recommended to achieve LDL-C reduction of ≥ 50% from baseline and goal LDL-C goal of < 70 mg/dL (Class IIa). The Heart Outcomes Prevention Evaluation–3 (HOPE-3) trial showed that ASCVD risk reduction in a large diverse population with intermediate risk outweighs the observable risk of treatment. Furthermore, individuals in the high-risk category or with risk-enhancing factors as seen in the JUPITER trial benefit from maximal statin therapy to achieve greater reductions in LDL-C level and ASCVD events. Still debated, however, are the fiscal and ethical issues related to the cost effectiveness of lipid drug therapy in lower-risk primary prevention.


Secondary Prevention


The 2018 AHA/ACC guidelines support aggressive lipid-lowering therapies for patients with established CHD or other ASCVD (including peripheral vascular disease, stroke, and aortic aneurysm) and recommend an LDL-C threshold of 70 mg/dL (1.8 mmol/L) to consider further LDL-C–lowering therapy, with the addition of ezetimibe or, in very-high-risk ASCVD patients, a PCSK9 inhibitor. Very high risk ASCVD was defined as either a history of multiple major ASCVD events or one major ASCVD event and multiple high-risk conditions (age >= 65 years, history of familial hypercholesterolemia, history of CABG or primary cutaneous intervention outside of the major ASCVD event, DM, HTN, CKD (eGFR 15–59 mL/min/1.73 m 2 ), current tobacco smoking, persistently elevated LDL-C >=100 mg/dL despite maximally tolerated statin and ezetimibe or history of congestive HF.


In contrast, the 2019 ESC/EAS dyslipidemia guidelines adopted a broader definition of individuals at very high risk to include anyone with documented ASCVD, either clinically or on imaging. This group includes all of those identified in the 2018 AHA/ACC guideline as secondary prevention but additionally patients with diabetes mellitus and end organ damage, moderate to severe CKD (estimated glomerular filtration rate < 30 mL/min/1.73 m 2 ) even in the absence of ASCVD, familial hypercholesterolemia with ASCVD or with another major risk factor, or a calculated SCORE of ≥ 10% (roughly equivalent to a 30% risk of 10-year ASCVD events according to the Pooled Cohort Equation). Furthermore, unlike the “threshold” of 70 mg/dL set by the AHA/ACC guidelines for considering the addition of a nonstatin lipid-modifying agent in very-high-risk patients, the ESC/EAS guidelines recommend a more aggressive approach: ≥ 50% reduction in LDL-C with an absolute “goal” of < 55 mg/dL (Class IIa), with first ezetimibe and then PCSK9 inhibitors in all patients with ASCVD, even without a recent ASCVD event. This goal is based on LDL-C levels achieved in large-scale trials of PCSK9 inhibitors. For patients with ASCVD who have a second vascular event within 2 years, a more aggressive LDL-C goal of < 40 mg/dL may be considered.


While maximally tolerated high-intensity statin remains the cornerstone of lipid lowering, more recent trials such as the Improved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE-IT), Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk (FOURIER), and Evaluation of Cardiovascular Outcomes after an Acute Coronary Syndrome During Treatment with Alirocumab (ODYSSEY Outcomes) now provide convincing evidence of clinical benefit with the addition of ezetimibe or PCSK9 inhibitors to statin to reduce LDL-C levels further. Although drug-induced LDL-C reduction remains an essential component of cardiovascular risk factor management, total risk can also be reduced through blood pressure control, dietary changes, increased exercise, weight loss, smoking cessation, and treatment of diabetes.


Blood Lipid Profile


Total Cholesterol and Low-Density Lipoprotein Cholesterol


Optimal total blood cholesterol levels are < 150 mg/dL (3.9 mmol/L), but it bears reemphasizing that cholesterol level is only part of the patient’s absolute global risk. Furthermore, LDL-C, not total cholesterol, is the real target of therapy. Both the 2018 AHA/ACC guidelines and 2019 ESC/EAS guidelines emphasize lowering LDL-C as the primary target of therapy.


Every reduction in LDL-C of 40 mg/dL (1 mmol/L) is accompanied by a 22% reduction in vascular events. There is now an overall consensus that “lower LDL is better.” In a large meta-analysis, the Cholesterol Treatment Trialists’ (CTT) Collaboration suggested that lower LDL-C levels with more-intensive statin therapy resulted in greater reductions in cardiovascular events and proposed that aggressive reduction of LDL-C by 2–3 mmol/L (about 80–120 mg/dL) would reduce risk by about 40%–50%. Similarly, the Pravastatin or Atorvastatin Evaluation or Infection Therapy (PROVE IT) trial showed that in patients with recent acute coronary syndrome (ACS), LDL-C levels of 62 mg/dL (1.60 mmol/L) led to convincingly better clinical outcomes than levels of 95 mg/dL (2.46 mmol/L). In primary prevention, the JUPITER study also supported this theory: a subgroup of patients achieving LDL-C levels < 50 mg/dL had a 65% reduction in cardiovascular events compared with placebo, whereas risk reduction was 44% in the study overall. In a study involving patients with stable coronary disease and much lower values of hs-CRP, high-dose atorvastatin reduced atheroma volume at an LDL-C of 79 mg/dL. In another study, an LDL-C value of approximately 75 mg/dL (2 mmol/L) marked the point at which progression and regression of the atheroma volume were in balance.


There had been debate in the past about whether there is a lower limit of LDL-C beyond which no further benefit occurs; this argument is now largely settled with the recent trials. In IMPROVE-IT, mean LDL-C level of 54 mg/dL led to convincingly better clinical outcomes than mean LDL-C level of 70 mg/dL. Furthermore, LDL-C reduction even to very low levels (< 30 mg/dL) appeared to be safe; these patients, in fact, had the lowest event rates. Similarly, in the FOURIER trial (discussed later), the absolute event rate of cardiovascular death, myocardial infarction (MI), or stroke was lowest in patients achieving LDL-C level < 20 mg/dL compared with the group with LDL-C > 100 mg/dL (5.7% versus 7.8%; hazard ratio [HR] 0.69; 95% confidence interval [CI] 0.56–0.85; P < 0.0001). From a safety perspective, there were no differences in drug discontinuation rates or serious adverse events regardless of the achieved LDL-C at 4 weeks. Of the 1839 subjects who achieved ultralow LDL-C levels (< 15 mg/dL), cardiovascular events further declined without any major safety concerns. However, it is important to note these data are limited to only 2.2 years of follow-up. An open-label extension study (FOURIER-OLE; NCT02867813) is currently ongoing in the United States and Europe to determine safety over a 5-year follow-up period.


Based on these substantial data, the 2018 AHA/ACC Guideline on the Management of Blood Cholesterol reintroduced thresholds for LDL-C in secondary prevention. An LDL-C threshold of ≥ 70 mg/dL is recommended for consideration of combination therapy to lower LDL-C further in patients with ASCVD.


High-Density Lipoprotein Cholesterol


High-density lipoprotein (HDL) is postulated to aid in clearing cholesterol from the foam cells that develop in diseased arteries, either by returning cholesteryl esters directly to the liver through scavenger receptor class B type I (SR-BI) or through transfer to the apoB-containing lipoproteins in exchange for triglycerides (reverse cholesterol transport mediated by cholesteryl ester transfer protein). HDL is also hypothesized to exert antiinflammatory and antioxidant effects.


Low HDL-C level is an independent risk factor that is strongly associated with risk for CHD. Many observational studies have shown an inverse relationship between HDL-C levels and incident cardiovascular events. In the Cholesterol and Recurrent Events (CARE) study, every 10 mg/dL decrease in HDL-C led to a 10% increase in risk. HDL-C ≥ 60 mg/dL (1.6 mmol/L) is a negative (protective) risk factor, although it remains to be proven that raising HDL-C is cardioprotective. Low HDL-C is often associated with other lipid abnormalities such as high triglycerides, but there is insufficient evidence to support treating these lipid components separately. The Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides: Impact on Global Health Outcomes (AIM-HIGH) study, which investigated the effect of raising HDL-C with niacin, did not show cardiovascular benefit in CHD patients who were already treated with a statin to a baseline mean LDL-C of 71 mg/dL. Furthermore, genetic studies do not show that genetic variants that can raise HDL-C lower cardiovascular events. Although American and European guidelines do not propose a target value for HDL-C, they do recommend raising low HDL-C when possible by lifestyle modification (exercise, modest alcohol intake, weight loss, smoking cessation).


A low HDL-C level is often part of atherogenic dyslipidemia , with the other two components being elevated triglycerides and small, dense LDL particles. Atherogenic dyslipidemia is a risk factor in its own right and is commonly found in patients with the metabolic syndrome, type 2 diabetes, and premature CHD. Lifestyle modification, combined with omega-3 fatty acids or fibrates, are the recommended treatments for patients with atherogenic dyslipidemia.


Triglycerides


Although triglyceride levels are commonly high in patients with CHD, the specific role of hypertriglyceridemia in atherogenesis remains controversial because it often occurs in conjunction with obesity, hypertension, and diabetes mellitus. Epidemiologically, an elevated triglyceride level can be an independent risk factor, even with adjustment for HDL-C, and in PROVE IT, triglyceride < 150 mg/dL (1.6 mmol/L) was associated with reduced cardiovascular risk even after major reduction of LDL-C. AHA defines normal triglycerides as fasting level < 150 mg/dL and optimal as < 100 mg/dL. The 2018 AHA/ACC guidelines define two categories of elevated triglycerides: moderate hypertriglyceridemia (fasting or nonfasting triglycerides 150–499 mg/dL [1.6–5.6 mmol/L]) and severe hypertriglyceridemia (fasting triglycerides ≥ 500 mg/dL [≥ 5.6 mmol/L]) ( Fig. 6.3 ). The guidelines recommend treatment with intensive dietary and lifestyle therapy for patients with moderate hypertriglyceridemia prior to initiating medications to lower triglyceride levels. A severely elevated triglyceride level (> 500 mg/dL [2.3 mmol/L]) may be viewed with special concern for risk of pancreatitis and should be treated with therapy shown to lower triglycerides most reliably, i.e., fibrates or omega-3 fatty acids. The Reduction of Cardiovascular Events with Icosapent Ethyl–Intervention Trial (REDUCE-IT; discussed later), which was published after the AHA/ACC guidelines and hence was not included in the guidelines, showed favorable reduction in cardiovascular events, including cardiovascular mortality, in patients with established ASCVD or diabetes and moderately elevated triglyceride levels treated with icosapent ethyl. The 2019 ESC/EAS guidelines, which came out afterwards, however, did recommend the addition of omega-3 fatty acids (icosapent ethyl 4 g/day) to statins in high-risk patients with triglyceride of 135–499 mg/dL) despite statin treatment (Class IIa). Similar recommendations are endorsed by both the National Lipid Association (NLA) and American Diabetes Association (ADA) (see below).




Fig. 6.3


Management of hypertriglyceridemia: 2018 AHA/ACC cholesterol guidelines.

For ASCVD prevention, treatment of elevated triglycerides includes lifestyle modification, controlling secondary dyslipidemias, and, if needed, drug therapy. Patients with severe hypertriglyceridemia are also at risk for pancreatitis and should be treated accordingly. ASCVD , Atherosclerotic cardiovascular disease; CLD , chronic liver disease; CKD , chronic kidney disease; DM , diabetes mellitus; MetS , metabolic syndrome; TG , triglycerides.

(Data from Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the management of blood cholesterol: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. J Am Coll Cardiol 2019;73:e285–e350.)


Other Lipoproteins and Lipoprotein Carriers


The combination of cholesterol in LDL and very-low-density lipoprotein (VLDL) is known as non-HDL-C and is a strong predictor of cardiovascular risk, especially in patients with elevated triglycerides or diabetes. Elevated triglycerides result from accumulation of VLDL and other triglyceride-rich lipoproteins, which are highly atherogenic. In such cases, non-HDL-C provides a more accurate measure of the cholesterol content of all atherogenic lipoproteins than LDL-C alone. The 2018 AHA/ACC guidelines support a non-HDL-C threshold of 100 mg/dL (2.6 mmol/L), along with LDL-C, to guide therapy and enhance identification of those at increased ASCVD risk, especially in very-high-risk patients with ASCVD.


Similarly, measurement of apoB may be valuable in patients with moderately elevated triglycerides, as it may be a more accurate indicator of atherogenic potential in these individuals. ApoB level ≥ 130 mg/dL, especially in patients with elevated triglycerides, denotes a high lifetime risk and is a risk-enhancing factor (see Table 6.1 ) in the 2018 AHA/ACC guidelines.


Lp(a), which is structurally similar to LDL with the addition of apo(a) covalently linked to apoB, may also be useful in risk assessment. The 2018 AHA/ACC guidelines define Lp(a) ≥ 50 mg/dL as a risk-enhancing factor and consider family history of premature ASCVD a relative indication for measurement of Lp(a) level. The 2019 ESC/EAS dyslipidemia guidelines recommend Lp(a) measurement in all adults at least once to identify individuals with very high inherited Lp(a) levels > 180 mg/dL, whose lifetime ASCVD risk may be equivalent to individuals with heterozygous familial hypercholesterolemia.


Lipids in Special Population Groups


Metabolic Syndrome


Metabolic syndrome is a cluster of risk factors ( Fig. 6.4 ) that greatly enhances the risk for coronary morbidity and mortality at any level of LDL-C. The underlying pathology of the metabolic syndrome appears to be linked to obesity and insulin resistance, and its prevalence increases with age and with presence of type 2 diabetes mellitus. The 2018 AHA/ACC cholesterol guidelines incorporate metabolic syndrome in the risk-enhancing factors (see Table 6.1 ), which influence initiation or up-titration of lipid-lowering therapy in primary prevention. First-line therapy for metabolic syndrome is weight control and increased physical activity. LDL-C and non-HDL-C should be controlled; achieving a significant increase in HDL-C, although desirable, has not proven to be clinically useful.




Fig. 6.4


Metabolic syndrome.

Metabolic syndrome is a constellation of risk factors, including central obesity, insulin resistance, hypertriglyceridemia, low HDL-C, and hypertension, that increases risk for ASCVD and diabetes. In the 2018 AHA/ACC cholesterol guidelines, metabolic syndrome is diagnosed by ≥ three of the risk factors shown. ASCVD , Atherosclerotic cardiovascular disease; HDL-C, high-density lipoprotein cholesterol.

(Data from Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the management of blood cholesterol: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. J Am Coll Cardiol 2019;73:e285–e350.)


Secondary Dyslipidemias


Diabetes mellitus, hypothyroidism, nephrotic syndrome, and alcoholism should be remedied if possible. Among drugs causing adverse lipid changes are β-blockers and diuretics ( Table 6.2 ), progestogens, and oral retinoids. Nonetheless, cardiac drugs known to be protective should not be withheld on the basis of their lipid effects alone, especially in postinfarct patients when there is clear indication for the expected overall benefit.



Table 6.2

Effects of antihypertensive agents on blood lipid profiles





















































































































































Change, %
Agent TC LDL-C HDL-C TG
Diuretics
TZ 14 10 2 14
Low-dose TZ a 0 0 0 0
Indapamide 0 (+ 9) 0 0 0
Spironolactone 5 ? ? 31
β-Blockers
Grouped (> 1 year) 0 0 –8 22
Propranolol 0 –3 –11 16
Atenolol 0 –2 –7 15
Metoprolol 0 –1 –9 14
Acebutolol a –3 –4 b –3 6
Pindolol –1 –3 –2 7
α-Blockers
Grouped –4 –13 5 –8
Doxazosin a –4 b –5 b 2 –8
αβ-Blocker
Labetalol 2 2 1 8
Carvedilol –4 ? 7 –20
CCBs
Grouped 0 0 0 0
Amlodipine a –1 –1 1 –3
ACE inhibitors
Grouped 0 0 0 0
Enalapril –1 –1 3 –7
Angiotensin receptor blockers
Losartan (0) c (0) c (0) c (0) c
Central agents
MD + TZ 0 0 0 0

ACE , Angiotensin-converting enzyme; CCBs , calcium channel blockers; HDL-C , high-density lipoprotein cholesterol; LDL-C , low-density lipoprotein cholesterol; MD , methyldopa; TC , total cholesterol; TG , triglyceride; TZ , thiazide.

a Chlorthalidone 15 mg/day; acebutolol 400 mg/day; doxazosin 2 mg/day; amlodipine 5 mg/day; enalapril 5 mg/day; data placebo-corrected.


b < 0.01 versus placebo over 4 years.


c no long-term data.



β-Blockers


β-Blockers tend to reduce HDL-C and to increase triglycerides. β-blockers with high intrinsic sympathomimetic activity or high cardioselectivity may have less or no effect (as in the case of carvedilol with added α-blockade). The fact that β-blockers also impair glucose metabolism is an added cause for concern when giving these agents to young patients. Nonetheless, strong evidence supports the protective effects of β-blockers in postinfarct and heart failure patients. Statins appear to counter some of the effects of β-blockers on blood lipids. In stable effort angina, calcium channel blockers may have a more favorable effect on triglycerides and HDL-C than β-blockers. In hypertensive patients, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and calcium channel blockers are all lipid neutral.


Diuretics


Diuretics increase triglycerides and tend to increase total cholesterol unless used in low doses. In the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT), chlorthalidone 12.5–25 mg daily over 5 years increased total cholesterol by 2–3 mg/dL. In the Antihypertensive Treatment and Lipid Profile in a North of Sweden Efficacy Evaluation (ALPINE) study, hydrochlorothiazide 25 mg, combined with atenolol in most patients, increased blood triglycerides and apoB, while decreasing HDL-C.


Oral Contraceptives


When oral contraceptives are given to patients with ischemic heart disease or with risk factors such as smoking, possible atherogenic effects of high-dose estrogen merit attention. In postmenopausal women, the cardiovascular benefits of hormone replacement therapy have not been supported by clinical trials.


Diabetic Patients


Patients with diabetes constitute a high-risk group and warrant aggressive risk reduction. Risk for MI is increased almost fivefold in diabetic women aged 35–54 years and more than twofold in diabetic men aged 35–54 years, compared with age-matched women and men, respectively, without diabetes. In line with this, type 2 diabetes is regarded as a risk category in its own right in the 2018 AHA/ACC cholesterol guidelines, and in the 2019 ESC/EAS guidelines, type 2 diabetes is considered a high-risk category, irrespective of calculated SCORE. Both guidelines recommend initiation of statin therapy along with lifestyle modification in these patients. In recent years, growing awareness of the overlapping pathophysiologic characteristics of CHD and type 2 diabetes has led to increased coordination between cardiologists and endocrinologists in addressing the joint risk. Patients with type 2 diabetes may have a preponderance of smaller, denser, more atherogenic LDL particles, even though the LDL-C level may be relatively normal.


Meta-analysis of 14 randomized trials with a follow-up of at least 2 years indicated that lipid-lowering drug treatment significantly reduced cardiovascular risk in both diabetic and nondiabetic patients. The Collaborative Atorvastatin Diabetes Study (CARDS), a multicenter, randomized primary-prevention trial in patients with type 2 diabetes and at least one other risk factor who were treated with atorvastatin, 10 mg/day, or placebo, was stopped early because of a favorable clinical benefit of statin therapy. Taken together with a large subgroup analysis from the Heart Protection Study (HPS) and Anglo-Scandinavian Cardiac Outcomes Trial–Lipid-Lowering Arm (ASCOT-LLA), there are strong arguments for considering statin therapy, in addition to lifestyle modification and blood pressure control, in all patients with type 2 diabetes. A meta-analysis of all four double-blinded primary-prevention randomized controlled trials with large cohorts with diabetes found that use of moderate-intensity statin therapy in a total of 10,187 participants was associated with a risk reduction of 25%, with no apparent difference in benefit between type 1 and type 2 diabetes mellitus.


Recent trials have provided evidence for the role of nonstatin lipid-lowering therapy in combination with statin to reduce cardiovascular risk among patients with diabetes. In IMPROVE-IT, which evaluated the addition of ezetimibe to statin therapy in patients with recent ACS (27% of whom had diabetes), individuals with diabetes had significantly greater relative and absolute benefit on cardiovascular outcomes than those without diabetes. Subanalysis of the diabetic subgroup in the FOURIER trial showed that median LDL-C levels were reduced to a similar degree with evolocumab relative to placebo (57% in those with diabetes mellitus versus 60% in those without diabetes mellitus). In the ODYSSEY OUTCOMES trial, a similar subgroup analysis compared patients with diabetes (n = 5444; 29%), prediabetes (n = 8246; 43%), and normoglycemia (n = 5234; 28%). Over a median follow-up of 2.8 years, treatment with alirocumab resulted in twice the absolute reduction in cardiovascular events among patients with diabetes as in those without diabetes given higher baseline risk. Similarly, in REDUCE-IT, 30% of subjects enrolled had diabetes and at least one other traditional risk factor, along with elevated triglycerides, and had significant reduction in risk of ischemic events, including cardiovascular death, with icosapent ethyl compared to placebo. Based on this study, the American Diabetes Association updated its comprehensive evidence-based recommendations to endorse the use of icosapent ethyl in patient with diabetes already on statin with elevated triglyceride level (134–499 mg/dL). Similar endorsements were made by the NLA and 2019 ESC/EAS guidelines.


Older Adults


Although the relation between cholesterol and CHD weakens with age, physicians should continue to consider lipids as a modifiable risk factor in older adults. The absolute risk for clinical CHD in older adults is much higher because age is a powerful risk factor and because blood pressure, another risk factor, often increases with age. Furthermore, consider the cumulative effect of lifetime exposure to a coronary risk factor on an older adult patient. While the Prospective Study of Pravastatin in the Elderly at Risk (PROSPER) found coronary but not overall mortality benefit with statin treatment in older adults (see section on pravastatin), this trial may have been too short (3 years) to show major decreases in cerebrovascular disease. In a recent meta-analysis of JUPITER and HOPE-3, benefits on ASCVD reduction with rosuvastatin were similar among those ≥ 70 years of age versus < 70 years of age, with relative risk reduction for nonfatal MI, nonfatal stroke, or cardiovascular death of about 26% and no difference in adverse events between the two age groups. Furthermore, those ≥ 70 years of age had much higher event rates, which along with the comparable relative risk reductions, would mean that larger absolute risk reductions can be achieved with statin treatment and hence a smaller number needed to treat (NNT) to prevent an event in older compared with younger patients. Other meta-analyses similarly support primary prevention for adults in their 70s. The Study Assessing Goals in the Elderly (SAGE) confirmed the safety and benefit of intensive treatment with atorvastatin, 80 mg/day, in older adult patients with stable coronary syndromes, but failed to demonstrate the superiority of intensive versus moderate treatment in reducing the primary endpoint of total ischemia duration from baseline to 1 year. However, data on older subsets (≥ 80 years of age) remain sparse. Furthermore, older adults may be more susceptible to statin-related risk, owing to increasing frailty, multiple comorbidities, cognitive impairment, polypharmacy, and altered pharmacodynamics in older adults. The 2018 AHA/ACC guidelines therefore recommend judicious use of statin therapy in higher-risk older adults and support clinical judgment and thorough risk and benefit discussion between patient and clinician prior to initiating statin. The 2019 ESC/EAS dyslipidemia guidelines recommend initiation of statin in older adults > 75 years old with high risk or above, and suggest that statin be started at a low dose and titrated upwards to achieve LDL-C treatment goals. In individuals whose cumulative risk outweighs benefit or with limited lifespan, the guidelines recommend not initiating therapy and, in individuals already taking statin, deprescription.


Women


Women have a lower baseline risk for CHD than men at all ages except perhaps beyond 80 years. Risk lags by about 10–15 years, perhaps because of a slower rate of increase in LDL-C, higher levels of HDL-C, or ill-understood protective genetic factors in the heart itself. It is not simply a question of being pre- or postmenopausal. In large statin trials such as HPS, women had relative risk reduction comparable to that in men. In the Management of Elevated Cholesterol in the Primary Prevention Group of Adult Japanese (MEGA) trial of low-dose pravastatin (10–20 mg daily) in low-risk Japanese patients, 69% of whom were women, women had marginally less CHD risk reduction than men, possibly because of their lower initial risk. The JUPITER trial, which enrolled 6801 women (38% of the study population), showed that women had similar risk reduction as men, primarily because of reductions in risk for revascularization and unstable angina. A meta-analysis conducted by the JUPITER investigators found that statins reduced cardiovascular events in women in primary prevention trials by one-third.


The 2018 AHA/ACC guidelines address certain conditions specific to women that may augment their baseline risk for CVD and may help clinical decision-making regarding lifestyle intervention and lipid-lowering therapy. These conditions include pregnancy-related complications (hypertensive disorders during pregnancy, preeclampsia, gestational diabetes mellitus, delivering a preterm or low-birth weight infant) and premature menopause, all of which have been shown to increase future risk of CVD and portend increased cardiovascular morbidity and mortality.


Pregnant Women


As a group, lipid-lowering drugs are either completely or relatively contraindicated during pregnancy because of the essential role of cholesterol in fetal development. Bile acid sequestrants may be safest, whereas statins should not be used (see “Contraindications and Pregnancy Warning” in the later section on statins).


Dietary and Other Nondrug Therapy for Dyslipidemia


Lifestyle and Risk Factor Reduction


Nondrug dietary therapy is fundamental to the management of all primary hyperlipidemias and frequently suffices as basic therapy when coupled with weight reduction, exercise, ideal (low) alcohol intake, and treatment of other risk factors such as smoking, hypertension, or diabetes. Regular exercise may also increase insulin sensitivity and lessen the risk of type 2 diabetes. If lifestyle recommendations, including diet, were rigorously followed, CHD would be dramatically reduced in those younger than age 70. However, high-intensity lifestyle modification is required to prevent progression or even to achieve regression of CHD.


Diet


Changes in diet are an absolute cornerstone of lipid-modifying treatment. The 2018 AHA/ACC Guideline on the Management of Blood Cholesterol provides practitioners with evidence-based dietary recommendations to improve cardiovascular health. They stress including nutrient-dense foods with cardioprotective fats while avoiding intake of excessive calories, saturated and trans fats, and refined carbohydrates. The two most commonly employed dietary patterns, also supported by the ACC/AHA, are the Mediterranean dietary pattern and Dietary Approaches to Stop Hypertension (DASH) diet.


The DASH dietary pattern was initially developed for blood pressure management. It puts emphasis on intake of fruits, vegetables, and low-fat dairy products; includes whole grains, poultry, fish, and nuts; and reduces saturated fats, red meat, sweets, and beverages containing added sugars. The Optimal Macronutrient Intake Trial for Heart Health (OmniHeart) study compared three variants of the DASH diet: a diet rich in carbohydrate (like the original DASH diet), a diet higher in protein (about half from plant sources), and a diet higher in unsaturated fat (predominantly monounsaturated fat). Each of the diets was similar to the original DASH diet, and all led to reductions in LDL-C and triglycerides.


In comparison, the typical Mediterranean dietary pattern is lower in dairy products and red and processed meats, higher in olive oil and seafood, and includes moderate wine intake. Total dietary fat is in the range of 32% to ≥ 35% of total energy intake. Similar to the DASH diet, the Mediterranean diet limits saturated fats but includes relatively high amounts of monounsaturated and polyunsaturated fats, with an emphasis on omega-3 fatty acids, instead. Fruits, vegetables, and whole grains provide a high dietary fiber intake. In the Prevención con Dieta Mediterránea (PREDIMED) trial, in nearly 7500 adults with high cardiovascular risk, strict adherence to a Mediterranean diet with added olive oil or nuts reduced the incidence of major cardiovascular events—stroke or heart attack—by nearly one-third. The better the adherence to this diet, the better the survival rate.


Physical Activity


Along with dietary modification, physicians should counsel patients on staying active and incorporating regular physical activity in their weekly routines to help reduce risk of CVD. The 2018 AHA/ACC guidelines recommend at least 120 minutes a week of aerobic physical activity (3–4 sessions per week, ~ 40 minutes per session) and including moderate- to vigorous-intensity physical activity. The Diabetes Prevention Program (DPP) showed that intensive lifestyle interventions focusing on exercise and weight loss prevented the development of diabetes and future microvascular complications over a 15-year follow-up.


Drug Therapy for Dyslipidemia


Statins: 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Inhibitors


Statins are well established as the first drugs of choice in primary and secondary prevention of CHD because of their favorable clinical outcomes, predictable effects on LDL-C, and relatively few side effects across multiple large clinical trials. Available statins include lovastatin, pravastatin, simvastatin, fluvastatin, atorvastatin, rosuvastatin, and pitavastatin. All the statins decrease hepatic cholesterol synthesis by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. They are highly effective in reducing total cholesterol and LDL-C, they usually increase HDL-C, and long-term safety and efficacy is well established. Many are now available in generic form. The landmark Scandinavian Simvastatin Survival Study (4S) showed that simvastatin used in secondary prevention achieved a reduction in total mortality and in coronary events. This was soon followed by a successful primary-prevention study with pravastatin in high-risk men (CARE). Successful primary prevention of common events has been found in patients with LDL-C values near the U.S. national average. An interesting concept is that lipid-lowering drugs may act in ways beyond regression of the atheromatous plaque, for example, by improving endothelial function, stabilizing platelets, reducing fibrinogen (strongly correlated with triglyceride levels), or inhibiting the inflammatory response associated with atherogenesis.


Class Indications for Statins


ASCVD Prevention


In general, depending on the drug chosen, the large statin trials ( Table 6.3 ) show beyond doubt that cardiovascular endpoints are reduced, total mortality is reduced in primary and secondary prevention, and the NNT to prevent any given major endpoint makes statins cost effective, especially in secondary prevention. In patients with clinical ASCVD, statins may be used to slow the progression of coronary atherosclerosis, again as part of an overall treatment strategy. In patients with primary hypercholesterolemia, homozygous familial hypercholesterolemia, or mixed dyslipidemias, statins reduce levels of total cholesterol, LDL-C, apoB, and triglycerides.



Table 6.3

Key statin trials with major significant outcomes












































































Trial, statin, 1° or 2° prevention Initial blood cholesterol (mean) Duration and numbers Comparator events per trial (%) Statin events per trial (%) Absolute risk reduction per trial Number needed to treat per trial
4S
Simvastatin 40 mg
2° prevention
260 mg/dL (6.75 mmol/L) 5.4 yr, median
(Placebo: 2223; statin: 2221)
Total deaths
1° end point: 256 (11.5%)
2° end point: 502 (22.6%)
182 (8.2%)
353 (15.9%)
74 (3.3%)
149 (30%)
30 (162/yr)
15 (80/yr)
WOSCOPS
Pravastatin
1° prevention
272 mg/dL (7.03 mmol/L) 4.9 yr (mean)
(Placebo: 3293; statin: 3302)
Deaths: 135 (4.1%)
1° end point: 248 (7.5%)
106 (3.2%)
174 (5.3%)
29 (0.9%)
74 (2.2%)
114 (558/yr)
45 (217/yr)
AFCAPS/TexCAPS
Lovastatin
1° prevention
221 mg/dL (5.71 mmol/L) 5.2 yr (mean)
(Placebo: 3301; statin: 3304)
CAD deaths: 15 (0.5%)
AMI a 81 (2.5%)
1° end point: 183 (5.5%)
11 (0.3%)
45 (1.4%)
116 (3.5%)
4(0.12%)
39 (1.3%)
67 (2.0%)
826 (4295/yr)
85 (441/yr)
49 (256/yr)
HPS
Simvastatin 40 mg
65% with CHD
228 mg/dL (5.9 mmol/L) 5 yr (mean)
(Placebo: 10,267; statin: 10,269)
Mortality: 1507 (14.7%)
Vascular deaths: 937 (9.1%)
Total Ml: 1212 (11.8%)
1328 (12.9%)
781 (7.6%)
898 (8.7%)
179 (1.8%)
156 (1.5%)
314 (3.1%)
56 (280/yr)
66 (330/yr)
32 (160/yr)
PROSPER
Pravastatin
High-risk older adults
221 mg/dL (5.7 mmol/L) 3.2 yr (mean)
(Placebo: 2913; statin: 2891)
Primary end point CHD death, nonfatal MI, + stroke: 473 (16.2%) 408 (14.1%) 65 (2.1%) 48 (152/yr)
ASCOT-LLA
Atorvastatin 10 mg
1° prevention; hypertensive
212 mg/dL (5.48 mmol/L) 3.3 yr (median)
(Placebo: 5137; statin: 5168)
Primary end point nonfatal MI + CHD death: 154 (3.0%) 100 (1.9%) 54 (1.1%) 90 (297/yr)
PROVE IT b
Atorvastatin 80 mg; pravastatin 40 mg
Recent ACS, 2° prevention
180 mg/dL (4.65 mmol/L) 2 yr (median)
(Pravastatin: 2063; atorvastatin 2099)
Primary composite endpoint (death plus cardiovascular events): pravastatin, 543 (26.3%) Atorvastatin, 470 (22.4%) 73 (3.7%) 29 (58/yr)
JUPITER
Rosuvastatin 20 mg
1° prevention
186 mg/dL (4.81 mmol/L) 1.9 yr (median)
(Placebo: 8901; rosuvastatin: 8901)
Primary composite endpoint (MI, stroke, revascularization, hospitalization for angina, CV death): 251 (2.8%) 142 (1.6%) 109 (1.2%) 29 (5/yr) c

ACS , Acute coronary syndrome; AMI , (nonfatal) acute myocardial infarction; CAD , coronary artery disease; CHD , coronary heart disease; CV , cardiovascular; MI , myocardial infarction.

a Estimated.


b PROVE IT compares atorvastatin versus pravastatin, not versus placebo.


c Endpoint of myocardial infarction, stroke, or death.



Stroke Prevention and Transient Ischemic Attack


Patients with a history of stroke or CHD equivalent should be considered for statin therapy. In CARDS, stroke risk in diabetic patients was reduced by 48% with only a 10-mg daily dose of atorvastatin. In the Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) study, high-dose atorvastatin (80 mg/day) reduced fatal and nonfatal strokes (2.2% absolute risk reduction; HR 0.84) and major cardiovascular events (3.5% absolute risk reduction; HR 0.80) in patients with a history of stroke or transient ischemic attack but no clinical ischemic heart disease. These benefits outweighed the slight increase in nonfatal hemorrhagic stroke (22 in 2365 patients; absolute increase 0.9%). A meta-analysis of more than 120,000 persons found powerful statin-related reductions of ischemic stroke and associated mortality that were not linked to the degree of LDL-C reduction, which suggested that stroke reduction was related to pleiotropic effects of statins. However, PCSK9 inhibitors have also been shown to reduce stroke (discussed later).


How Intensive Should Statin Therapy Be?


A meta-analysis of more than 90,000 subjects with clinical vascular disease on standard statin therapy showed significant reduction in cardiovascular events with statin use. The authors estimated that, for every 1- mmol/L decrease in LDL-C (approximately 40 mg/dL), the 5-year relative risk for major coronary events is reduced by about one-fifth, with the absolute risk reduction dependent on the initial level of risk, and they projected that sustained statin therapy for 5 years might reduce the incidence of major vascular events by approximately one-third. An updated meta-analysis of trials comparing intensive- versus moderate-intensity statin therapy from the same group, including 170,000 subjects, found that intensive statin treatment further reduced the risk of major vascular events, so that the relation between absolute LDL-C reduction and proportional risk reduction remained consistent in the trials of intensive statin therapy. These findings support a strategy to achieve the largest LDL-C reduction possible in high-risk patients without increasing risk for myopathy.


Benefit Versus Possible Harm With Very-High-Dose Statins


A retrospective analysis of possible adverse effects with very-high-dose statin therapy suggested a small increased risk of cancer, equivalent to only 1.5% per 5 years. However, in an analysis of more than 6000 patients with LDL-C levels < 60 mg/dL, those with very low levels (< 40 mg/dL) had improved survival without any increased risk of cancer or rhabdomyolysis. Myopathy remains a definite risk, especially in the case of high-dose simvastatin, and new diabetes is more common in high- than in medium-dose statin therapy (see “Class Warnings”).


Class Warnings


Liver Damage, Myopathy, New Diabetes, and Cognitive Side Effects


The package inserts for statins were revised by the U.S. Food and Drug Administration (FDA) in early 2012. Pretreatment liver function tests are recommended, but routine periodic monitoring of liver enzymes is not, as it was in the past, because serious liver injury with statins rarely occurs. Warnings regarding myopathy and rhabdomyolysis remain in place. Skeletal muscle effects range from muscle pains to objective myopathy to severe myocyte breakdown that in turn can cause potentially fatal renal failure by way of myoglobinuria. Myopathy is diagnosed when creatine kinase blood levels exceed 10 times normal. The patient should be warned that muscle pain, tenderness, or weakness must immediately be reported to the physician and the statin stopped. Abnormal enzyme values usually resolve with cessation of treatment. Thereafter follows a monitored rechallenge at a lower dose or a change to low-dose fluvastatin or alternate-day low-dose rosuvastatin (because these may cause less myopathy) or nonstatin therapy. A trial of added coenzyme Q10 may help. However, in HPS, with more than 10,000 patients in each treatment group, enzyme-diagnosed myopathy over 5 years occurred in only 0.11% of statin-treated patients versus 0.06% in controls, and rhabdomyolysis in only 0.05% versus 0.03% in controls. The absolute rates of myopathy, much less rhabdomyolysis, are low in reported clinical surveys, although in clinical practice, these complaints are more common. Fatal cases are extremely rare, occurring in only 0.2 or fewer instances per million prescriptions. Risk for myopathy is greater with high-dose simvastatin (see section on simvastatin) and coadministration with fibrates, niacin, cyclosporine, erythromycin, or azole antifungal agents. Although not contraindicated, the combination of a statin and a fibrate increases the risk for myopathy to an incidence of approximately 0.12%, and physicians are cautioned to be mindful of this risk.


Interactions with protease inhibitors are potentially myopathic. The FDA warns as follows (statins in alphabetical order):




  • Atorvastatin: caution with telaprevir and ritonavir; use lowest dose with lopinavir + ritonavir; limit dose to 20 mg/day with darunavir + ritonavir, fosamprenavir, fosamprenavir + ritonavir, saquinavir + ritonavir; limit dose to 40 mg/day with nelfinavir



  • Fluvastatin: no data available



  • Lovastatin: contraindicated with human immunodeficiency virus (HIV) protease inhibitors, boceprevir, telaprevir



  • Pitavastatin: no dose limitations



  • Pravastatin: no dose limitations



  • Rosuvastatin: limit dose to 10 mg/day with atazanavir ± ritonavir, lopinavir + ritonavir



  • Simvastatin: contraindicated with HIV protease inhibitors, boceprevir, telaprevir



Thus, pravastatin and pitavastatin are the safest and simvastatin the least safe to use with HIV and hepatitis C protease inhibitors.


New-onset diabetes is a more recently discovered side effect, first reported with rosuvastatin and now recognized as a generalized problem of high-dose statins. In a meta-analysis of 91,140 persons in 13 trials, statin therapy was associated with a slightly increased risk of new diabetes (9%, odds ratio 1.09; 95% CI 1.02–1.17). Based on these data, statin therapy used in 255 patients for 4 years led to one extra case of diabetes and prevented 5.4 coronary events (coronary deaths, nonfatal MI). In another meta-analysis, which compared intensive- with moderate-dose statin therapy in five studies that included 32,752 persons without diabetes at baseline, the NNT annually was 498 for new-onset diabetes versus 155 for reduced cardiovascular events. Thus, the approximate ratio of benefit to harm for high versus medium doses was approximately 3:1. An observational study of 161,808 postmenopausal women found an increase in risk for diabetes (48%, multivariate-adjusted HR 1.48; 95% CI 1.38–1.59). As a result of this cumulative evidence, the FDA has added information concerning an effect of statins on incident diabetes and increases in hemoglobin A1c and fasting plasma glucose to all statin labels.


Additional information about potential nonserious and reversible cognitive side effects (memory loss, confusion, etc.) were also added to statin labels, based on postmarketing reports. However in the PROSPER trial, cognitive function was assessed repeatedly in all 5804 participants, and no difference was found in cognitive decline in subjects treated with pravastatin compared with placebo during a 3-year follow-up period. Similarly, a systematic review and meta-analysis found no difference in cognitive performance related to procedural memory, attention, or motor speed.


Contraindications and Pregnancy Warning


Statins are contraindicated in patients with active liver disease or unexplained persistent elevations of serum transaminases. Statins must not be prescribed to women who are pregnant or who are planning to become pregnant because cholesterol is essential to fetal development. Statins are excreted in the mother’s milk, so women taking statins should not breast feed. Women desiring to become pregnant should stop statins for approximately 6 months before conception. If a patient becomes pregnant while taking statins, therapy should be discontinued, and the patient apprised of the potential hazard to the fetus.


Lovastatin (Altoprev, Mevacor)


Lovastatin (Altoprev, Mevacor) was the first statin to be approved and marketed in the United States and was the first generically available. In the landmark primary prevention Air Force/Texas Coronary Atherosclerosis Prevention Study (AFCAPS/TexCAPS), lovastatin reduced clinical cardiac events, including MI, by 37% in individuals with baseline LDL-C values considered “normal” within the general American population (221 mg/dL; 5.71 mmol/L), but with low HDL-C levels (36 mg/dL; 1.03 mmol/L).


Dose, Effects, and Side Effects


The usual starting dose for lovastatin is 20 mg once daily with the evening meal, going up to 80 mg in one or two doses. In 2012, the FDA revised the labeling of lovastatin with new contraindications and dose limitations for concomitant medications, because of an increased risk for myopathy with strong inhibitors of the hepatic cytochrome P-450 3A4 (CYP3A4) substrate. Lovastatin is contraindicated with itraconazole, ketoconazole, posaconazole, erythromycin, clarithromycin, telithromycin, HIV protease inhibitors, boceprevir, telaprevir, and nefazodone. Regarding drug interactions, concomitant therapy with cyclosporine and gemfibrozil should be avoided; a 20-mg dose of lovastatin should not be exceeded when the patient is taking danazol, diltiazem, or verapamil; and the 40-mg dose should not be exceeded with amiodarone. Large quantities of grapefruit juice should also be avoided. There are no significant interactions between lovastatin and the common antihypertensive drugs, nor with digoxin. The same cautions concerning hepatotoxicity, myopathy, and rhabdomyolysis that affect other statins also apply to lovastatin.


Fluvastatin (Lescol, Lescol XL)


Fluvastatin was the first synthetic statin approved by the FDA. In the Lipoprotein and Coronary Atherosclerosis Study (LCAS), fluvastatin in patients with mildly to moderately elevated LDL-C reduced CHD progression, and the Lescol Intervention Prevention Study (LIPS) showed reduced risk for cardiac events with early initiation of fluvastatin in patients with average cholesterol levels following percutaneous coronary intervention.


Dose, Effects, and Side Effects


The dosing range of fluvastatin is 20–80 mg/day, taken in the evening or at bedtime, and the recommended starting dosage may be determined by the degree of LDL-C reduction needed. Fluvastatin is metabolized mainly by the CYP2C9 isoenzyme, making it less likely to interact with drugs that compete for the CYP3A4 pathway, such as the fibrates. However, phenytoin and warfarin share metabolism by CYP2C9, raising the risk for interactions. The same cautions concerning hepatotoxicity, myopathy, and rhabdomyolysis that affect other statins also apply to fluvastatin.


Pravastatin (Pravachol, Lipostat)


In the primary-prevention West of Scotland Coronary Prevention Study (WOSCOPS), pravastatin reduced the risk for coronary morbidity and mortality in high-risk men. In the secondary-prevention Long-term Intervention with Pravastatin in Ischemic Disease (LIPID) trial, pravastatin therapy reduced the risk for death from any cause by 22% ( P < 0.001) and also decreased the risks for nonfatal MI or CHD death, stroke, and coronary revascularization. In PROVE IT, pravastatin at 40 mg daily was inferior to atorvastatin at 80 mg daily in the reduction of LDL-C and clinical events. The PROSPER trial, which enrolled older adult patients with a mean cholesterol of 212 mg/dL (5.7 mmol/L) and high coronary risk, found that pravastatin, 40 mg/day, reduced the relative risk for CHD death by 24% ( P = 0.043), chiefly when given for secondary prevention; results for primary prevention were nonsignficant. There was, however, an increased incidence of cancer in PROSPER. Longer-term follow-up from WOSCOPS found no increase in cancer at 10 or 20 years after the trial.


Indications


Besides its class indications (see previous), pravastatin is approved for primary prevention in patients with hypercholesterolemia to reduce the risk for MI, revascularization, and cardiovascular mortality. In patients with previous MI, it is indicated to reduce total mortality by reducing coronary deaths and to reduce recurrent MI, revascularization, and stroke or transient ischemic attack.


Dose and Effects


The recommended starting dose for pravastatin is 40 mg at any time of the day, increasing to 80 mg if needed. As with the other statins, liver damage and myopathy are rare but serious side effects. Cautions and contraindications are similar to other statins. There is no drug interaction with digoxin. Pravastatin is not metabolized by the CYP3A4 pathway, so there may be a lower risk for interactions with agents such as erythromycin and ketoconazole. Importantly, there is no interaction with antiretrovirals.


Simvastatin (Zocor)


Major Trials


The landmark 4S paved the way to widespread acceptance of statins as the cornerstone of lipid-lowering drug therapy. In this study in 4444 patients with severely elevated cholesterol levels, mostly men with past MI, simvastatin reduced LDL-C by 35% over 4 years, total mortality by 30%, cardiac death rate by 42%, and revascularization by 37%. There was no evidence of increased suicide or violent death, previously thought to be a potential hazard of cholesterol reduction. Differences between simvastatin and placebo arms started to emerge after 1–2 years of treatment, and most curves were still diverging at 4 years. Longer-term follow-up (up to 8 years) after the trial suggested that benefits were maintained.


HPS evaluated the role of simvastatin versus placebo in 20,536 high-risk patients for whom guidelines at the time would not have recommended drug intervention. Included patients were 40 to 80 years of age and had total serum cholesterol concentrations of at least 135 mg/dL (3.49 mmol/L). Only 65% of the patients had a history of CHD at baseline, and HPS included many high-risk “primary-prevention” individuals who had never had a coronary event (n = 7150), although a significant number had a CHD risk equivalent: diabetes, peripheral vascular disease, or cerebrovascular disease. Simvastatin reduced the risk for any major vascular event by 24% ( P < 0.0001) and all-cause mortality by 13% ( P = 0.0003), with a 17% reduction in deaths attributed to any vascular cause. No safety issues were observed with treatment, and myopathy incidence was only 0.01%. The similar clinical benefit in patients with baseline LDL-C level of less than 116 mg/dL (3 mmol/L) and those with higher levels supports treatment decisions based on clinical risk rather than baseline lipids, and initiation of statin therapy in high-risk patients regardless of initial LDL-C level. In the Study of the Effectiveness of Additional Reductions in Cholesterol and Homocysteine (SEARCH), 12,064 participants were randomized to either 80 mg or 20 mg simvastatin daily. The 6% reduction in major vascular events with a further 13.5 mg/dL (0.35 mmol/L) reduction in LDL-C was consistent with previous trials. However, myopathy was increased with 80 mg simvastatin daily, which led to new FDA recommendations (see later).


Indications


Simvastatin has additional, specific indications in patients with CHD and hypercholesterolemia, for (1) reduction of coronary and total mortality, (2) reduction of nonfatal MI, (3) reduction of myocardial revascularization procedures, and (4) reduction of stroke or transient ischemic attack. Simvastatin is also indicated for increasing HDL-C in patients with hypercholesterolemia or combined dyslipidemias, without claiming an effect independent of LDL-C lowering. Based on the results of HPS, the FDA approved revised labeling for simvastatin in 2003 that emphasized high-risk status rather than LDL-C alone as the primary determinant of treatment. Essentially, the labeling states that simvastatin may be started simultaneously with dietary therapy in patients with CHD or at high risk for CHD.


Dose, Side Effects, and Safety


The usual starting dosage for simvastatin is 20 mg once daily in the evening. In 4S, the initial dose was 20 mg once daily just before the evening meal, increased to 40 mg if cholesterol lowering was inadequate after 6 weeks (37% of subjects). For patients at high risk, the starting dosage is 40 mg/day as in HPS. The previous maximum dosage of 80 mg daily is now linked to a substantial risk of myopathy; therefore, the FDA recommends that patients should not be started on or switched to this dose, and patients already on this dose should be carefully monitored for myopathy. FDA recommendations to reduce myopathy with simvastatin , which is broken down by the hepatic enzyme CYP3A4 system, are that simvastatin should not be used with the conazole group of drugs (itraconazole, ketoconazole, posaconazole), some antibiotics (erythromycin, clarithromycin, telithromycin), nefazodone, gemfibrozil, cyclosporine, and danazol. Specifically contraindicated by the FDA are the HIV protease inhibitors, boceprevir, and telaprevir. The 10-mg dose should not be exceeded in patients taking amiodarone, verapamil, and diltiazem. The 20-mg dose should not be exceeded with amlodipine and ranolazine (Ranexa). The 80-mg dose should not be started.


The 11-year follow-up study of HPS found no increase in cancer incidence, cancer mortality, or other nonvascular mortality with simvastatin. The original concerns about the long-term safety of statins have thus been dispelled.


Atorvastatin (Lipitor)


Secondary Prevention


Atorvastatin is one of the best tested and most prescribed of the statins. The Myocardial Ischemia Reduction and Aggressive Cholesterol Lowering (MIRACL) trial and PROVE IT examined the premise that early treatment with high-dose (80 mg daily) atorvastatin therapy following ACS would give clinical benefits. In MIRACL, atorvastatin, compared to placebo, produced modestly significant relative risk reductions for symptomatic ischemia. In the large PROVE IT study in more than 4000 patients, atorvastatin reduced LDL-C to 62 mg/dL (1.60 mmol/L) and decreased the composite primary endpoint when compared with pravastatin 40 mg daily. In patients with stable coronary disease in the Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) study, a similar vigorous reduction of LDL-C with atorvastatin versus pravastatin decreased atheroma volume. In the Treating to New Targets (TNT) trial, high-dose atorvastatin (80 mg daily) reduced mean LDL-C from approximately 100 mg/dL (2.6 mmol/L) to 77 mg/dL (2 mmol/L), and major cardiovascular events fell by 22% versus low-dose atorvastatin (10 mg daily). In the Incremental Decrease in Endpoints through Aggressive Lipid lowering (IDEAL) study in 8888 patients with prior MI, atorvastatin 80 mg daily reduced the secondary endpoint of any coronary event, when compared with simvastatin taken at mostly 20 mg daily. However, the primary endpoint of major coronary events was not different between treatment groups nor was mortality reduced. The final lower LDL-C level of 81 mg/dL (2.1 mmol/L) in the atorvastatin group versus 100 mg/dL (2.6 mmol/L) in the simvastatin group modestly supports the “lower is better” hypothesis, at the cost of approximately double the rate of adverse events leading to drug discontinuation (9.6% for atorvastatin versus 4.2% for simvastatin).


Primary Prevention


ASCOT-LLA assessed the clinical effect of atorvastatin, 10 mg/day, versus placebo in 10,305 hypertensive patients with mean total cholesterol of 212 mg/dL (5.5 mmol/L), mean LDL-C of 130 mg/dL (3.4 mmol/L), and a high-risk profile. Originally planned with a follow-up of 5 years, ASCOT ended early because of clear benefit. Atorvastatin reduced the relative risk for cardiovascular events by 36% ( P = 0.0005) and for stroke by 27% ( P = 0.024). There was no effect on the low total mortality rate, and the adverse event rates did not differ between the treatment groups. CARDS, in high-risk diabetics, was similarly stopped early because of improved clinical endpoints in those treated with atorvastatin, 10 mg daily, versus placebo. An analysis from TNT suggests that atorvastatin may improve glomerular filtration rate in patients with kidney disease.


Indications


Besides class indications (see previous), atorvastatin is approved by the FDA for primary prevention in patients with multiple risk factors to reduce the risk for MI, stroke, revascularization, or angina. For primary prevention in those with type 2 diabetes and multiple risk factors, atorvastatin is indicated for reduction of MI and stroke. For patients with CHD, atorvastatin is indicated for reduction of nonfatal MI, stroke, revascularization, hospitalization for congestive heart failure, and angina.


Dosage, Effects, and Side Effects


Atorvastatin is available as 10-, 20-, 40-, and 80-mg tablets, which can be given once daily at any time of the day, with or without food. ASCOT and CARDS suggested that a dosage of only 10 mg daily may help prevent clinical events. The PROVE IT study showed that high-dose atorvastatin, 80 mg/day, reduces LDL-C to very low levels and reduces clinical events in patients with recent ACS. A 10-mg starting dose of atorvastatin provides good reductions in total cholesterol, LDL-C, apoB, and triglyceride, and a modest increase in HDL-C. Blood lipid levels should be checked 2–4 weeks after starting therapy and the dosage adjusted accordingly. As with the other statins, liver damage and myopathy are rare but serious side effects .


Drug Interactions


Patients on potent inhibitors of hepatic CYP3A4, such as ketoconazole, erythromycin, or HIV protease inhibitors, should in principle not be given any statin that is metabolized through this enzyme (atorvastatin, fluvastatin, lovastatin). Specifically, the FDA warns as follows: avoid atorvastatin with tipranavir and ritonavir, use lowest dose with lopinavir and ritonavir, and use care with other antiretrovirals. Erythromycin inhibits hepatic CYP3A4 to increase blood atorvastatin levels by approximately 40%. The interaction with clopidogrel has not been clinically evident. Atorvastatin increases blood levels of some oral contraceptives . There is no interaction with warfarin. Other drug interactions are similar to the other statins, including cotherapy with fibrates and niacin.


Rosuvastatin (Crestor)


Rosuvastatin is a hydrophilic compound with high uptake into and selectivity for its site of action in the liver, leading to substantial reductions in total cholesterol and LDL-C. Rosuvastatin’s half-life is approximately 19 hours, and it can be taken at any time of the day. It is not metabolized by the CYP3A4 system, thus lessening the risk for certain key drug interactions. However, there are interactions with antiretrovirals.


Major Trials


A Study to Evaluate the Effect of Rosuvastatin on Intravascular Ultrasound-Derived Coronary Atheroma Burden (ASTEROID), conducted in 349 patients with coronary atherosclerosis, found that high-intensity rosuvastatin, 40 mg/day, achieved a mean LDL-C of 61 mg/dL (1.6 mmol/L) and increased HDL-C by 14.7%, with regression of coronary atherosclerosis as measured by intravascular ultrasound. In Measuring Effects on Intima-Media Thickness: an Evaluation of Rosuvastatin (METEOR), in low-risk men with modest carotid intimal–medial thickening and mean LDL-C values of 154 mg/dL, 40-mg/day rosuvastatin for 2 years substantially reduced the rate of progression of carotid atherosclerosis. Results from the JUPITER study have established the efficacy of rosuvastatin in primary prevention, particularly for individuals at increased risk because of elevated levels of hs-CRP but with low levels of LDL-C. JUPITER, which enrolled 17,802 middle-aged adults free of heart disease and diabetes with LDL-C < 130 mg/dL and hs-CRP ≥ 2 mg/L, compared rosuvastatin 20 mg versus placebo and was stopped after 1.9 years because of efficacy. Rosuvastatin reduced LDL-C levels by 50% to a median of 55 mg/dL and decreased hs-CRP levels by 37%, which translated to a 44% relative reduction in major cardiovascular events and a 20% reduction in all-cause mortality compared with placebo.


Indications


In addition to its class indications, rosuvastatin has a favorable effect on triglycerides in patients with elevated serum triglyceride levels and is indicated to slow the progression of atherosclerosis. In primary prevention, based on JUPITER, rosuvastatin is indicated to reduce the risk for stroke, MI, and revascularization in patients at increased risk because of age, hs-CRP of at least 2 mg/L, and one additional cardiovascular risk factor. Rosuvastatin can be safely used in systolic heart failure without any specific antifailure benefit.


Dosage, Effects, and Side Effects


Rosuvastatin is supplied in 5-, 10-, 20-, and 40-mg tablets. The usual starting dosage is 10 mg/day (5 mg for Asian patients) taken any time with or without food. At this dosage, in patients with primary hypercholesterolemia, the expected LDL-C reduction is 52%, with approximately 10% increase in HDL-C and 24% decrease in triglycerides. For patients of advanced age or with renal insufficiency, the recommended starting dose of rosuvastatin is 5 mg/day. In renal patients, rosuvastatin may be titrated up to 10 mg/day; at this dose, rosuvastatin did not increase adverse events and reduced lipid parameters in patients with end-stage renal disease, although it had no effect on cardiovascular outcomes. In patients receiving concomitant cyclosporine, rosuvastatin should be limited to 5 mg/day. In combination with gemfibrozil, rosuvastatin should be limited to 10 mg/day. Its side effects and warnings are similar to those of other statins. The maximum 40-mg dose of rosuvastatin is reserved for patients who have an inadequate response to 20 mg/day. Findings of increased risk for new diabetes were first observed with rosuvastatin in the JUPITER trial and subsequently extended to the other statins. Whereas a large meta-analysis of statin trials found a 9% increased risk for incident diabetes with statin treatment over a 4-year period, the risk with rosuvastatin was 18%, based on the results of JUPITER and two other clinical trials. Uncommon instances of proteinuria with microscopic hematuria have been reported, and the frequency may be greater at the 40 mg dose.


Drug Interactions


Like fluvastatin, rosuvastatin is metabolized by way of the CYP2C9 isoenzyme and therefore may be less likely to interact with common drugs that use the CYP3A4 pathway, such as ketoconazole or erythromycin. The FDA warns that the rosuvastatin dose should be limited to 10 mg daily with atazanavir with or without ritonavir, or lopinavir with ritonavir. Warfarin interaction is a risk. The standard statin warnings against cotherapy with fibrates or niacin remain, although fenofibrate appears safe. Coadministration of cyclosporine or gemfibrozil with rosuvastatin results in reduced rosuvastatin clearance from the circulation; therefore, the rosuvastatin dose should be reduced. An antacid (aluminum and magnesium hydroxide combination) decreases plasma concentrations of rosuvastatin and should be taken 2 hours after and not before rosuvastatin.


Pitavastatin (Livalo)


Pitavastatin, a low-dose statin, has shown in noninferiority studies of equivalent doses to produce LDL-C reductions comparable to those of atorvastatin and simvastatin and greater than those of pravastatin. It also favorably affects HDL-C and triglycerides. The Japan Assessment of Pitavastatin and Atorvastatin in Acute Coronary Syndrome (JAPAN-ACS) study demonstrated that pitavastatin reduced plaque volume similar to atorvastatin. In the Stabilization and Regression of Coronary Plaque Treated with Pitavastatin Proved by Angioscopy and Intravascular Ultrasound (TOGETHAR) trial, pitavastatin improved plaque composition as assessed by intravascular ultrasound in coronary segments in patients with ACS. High-Dose Versus Low-Dose Pitavastatin in Japanese Patients with Stable Coronary Artery Disease (REAL-CAD), a large prospective, multicenter clinical trial in which 13,054 Japanese patients with stable CHD were randomized to receive pitavastatin 4 mg/day (high dose) or pitavastatin 1 mg/day (low dose), confirmed that high-dose pitavastatin compared to low-dose pitavastatin safely and significantly reduced cardiovascular events in Asian patients.


Indications, Dose, Effects, and Side Effects


Pitavastatin is indicated as an adjunct to diet to reduce elevated total cholesterol, LDL-C, apoB, and triglyceride levels and to increase HDL-C in patients with primary hyperlipidemia or mixed dyslipidemia. It is supplied in 1-, 2-, and 4-mg tablets, with a usual starting dose of 2 mg/day taken at any time of day and a maximum dose of 4 mg/day. For patients with renal disease, the recommended starting dose is 1 mg/day up to a maximum of 2 mg/day. Depending on the dose, pitavastatin can be expected to reduce LDL-C by 31%–45%, reduce triglycerides by 13%–22%, and increase HDL-C by 1%–8%. The side effects and warnings for pitavastatin are similar to those of other statins.


Drug Interactions


Pitavastatin is not a substrate for CYP3A4, so it may be less likely to interact with drugs that inhibit the CYP3A4 system. It is minimally metabolized by CYP2C9, which appears to have little clinical effect on drug clearance. Importantly, there is no interaction with antiretrovirals. It is primarily metabolized via glucuronidation, so concomitant treatment with gemfibrozil and other fibrates should only be used with caution, as gemfibrozil has the potential to inhibit the glucuronidation and clearance of statins. Coadministration of cyclosporine is contraindicated because of reduced clearance of pitavastatin, and dosages of pitavastatin should be reduced with coadministration of erythromycin and rifampin for the same reason. Pitavastatin has not been studied with the protease inhibitor combination lopinavir–ritonavir, so should not be used with this combination. As with other statins, combination treatment with niacin and fibrates increases risk for myopathy.


Combination Therapy


Despite the widespread availability of effective statin therapy, observational studies have shown that 16%–53% of patients fail to attain their recommended LDL-C targets in clinical practice, and even in patients on ideal statin dosing the risk of major vascular events is reduced by only around one-third. These limitations may be due to suboptimal treatment because of insufficient starting doses or failure to up-titrate therapy, poor LDL-C response to statin, or issues with medication adherence, especially among patients who cannot tolerate the recommended intensity of statin because of adverse effects. The recent large outcome studies combining statin with ezetimibe (IMPROVE-IT ), PCSK9 inhibitor (FOURIER and ODYSSEY ), and most recently icosapent ethyl (REDUCE-IT ) provide strong support for combination strategies in secondary prevention. Both the 2018 AHA/ACC guidelines and 2019 ESC/EAS guidelines recommend the sequential addition of nonstatin lipid-modifying agents, ezetimibe and then PCSK9 inhibitors, to maximally tolerated statin in high-risk statin-intolerant or statin-unresponsive secondary-prevention patients. Furthermore, though increased risk of myopathy was previously a feared complication of combination therapy, myopathy is a rare event with combination therapy. Two reservations regarding combination therapy are the lack of data on primary prevention and, for PCSK9 inhibitors, economic feasibility and long-term safety, which is still being established (discussed later).


Cholesterol Absorption Inhibitors: Ezetimibe (Zetia, Vytorin, Nexlizet)


Although statins remain the mainstay for treatment of hypercholesterolemia and secondary prevention of ASCVD, recent data suggest that ezetimibe may serve as a valuable addition to the armamentarium of lipid-lowering drugs. Cholesterol absorption inhibitors selectively interrupt intestinal absorption of cholesterol and phytosterols. Ezetimibe acts at the brush border of the small intestine and inhibits the absorption of cholesterol, leading to decreased delivery of intestinal cholesterol to the liver, which reduces hepatic cholesterol and increases cholesterol clearance from the blood. This mechanism is complementary to that of statins. Ezetimibe has a half-life of 22 hours and is not metabolized by the CYP system.


A meta-analysis of eight randomized, double-blind, placebo-controlled trials found that ezetimibe significantly reduced LDL-C levels by 18.5% when used as monotherapy. The use of ezetimibe in conjunction with a statin has been shown to be an effective strategy for not only reducing LDL-C levels but also preventing cardiovascular events. Early trials demonstrated that statin and ezetimibe combination therapy reduced LDL-C by 12%–19% more than statin monotherapy. The Plaque Regression With Cholesterol Absorption Inhibitor or Synthesis Inhibitor Evaluated by Intravascular Ultrasound (PRECISE-IVUS) trial in patients with CHD demonstrated greater coronary plaque regression, assessed by serial volumetric intravascular ultrasound, in patients treated with ezetimibe in combination with atorvastatin than in patients treated with atorvastatin monotherapy (78% versus 58%; P = 0.004). However, the Ezetimibe and Simvastatin in Hypercholesterolemia Enhances Atherosclerosis Regression (ENHANCE) study did not find significant reduction of carotid atherosclerosis in patients with familial hypercholesterolemia with combined ezetimibe and simvastatin compared with simvastatin monotherapy, despite greater LDL-C reduction.


The Study of Heart and Renal Protection (SHARP) was the first major trial that assessed cardiovascular outcomes using combination therapy of statin and ezetimibe. In a wide range of patients with CKD, simvastatin 20 mg daily plus ezetimibe 10 mg daily compared to placebo significantly reduced both the LDL-C levels (33-mg/dL [0.85-mmol/L] difference between treatment groups) and the rate of cardiovascular events (11.3% versus 13.4%; rate ratio 0.83; absolute risk reduction 2.1%; NNT 48) at median 5-year follow-up. However, because the study lacked a statin-only comparison arm, it was unclear whether the benefit of adding ezetimibe was independent of the LDL-lowering effects of the statin. Note that this study could equally well argue for lipid lowering with a statin in dialysis patients. The FDA updated the prescribing information for ezetimibe to include data from SHARP. Although the FDA approved the ezetimibe–simvastatin combination for use in CKD as a new indication, ezetimibe without simvastatin was not approved because the relative contributions of simvastatin and ezetimibe were not assessed in the trial.


IMPROVE-IT was the first trial to provide compelling evidence that a nonstatin medication, ezetimibe, resulted in greater reduction in cardiovascular events when added to statin therapy than statin monotherapy in high-risk patients. Patients with known CHD, within 10 days of a recent MI or ACS and with low LDL-C levels (< 125 mg/dL [3.2 mmol/L]), were randomized to ezetimibe therapy in conjunction with simvastatin or to simvastatin monotherapy and followed for a median of 6 years. The combination therapy group had a significantly lower rate of major cardiovascular events (32.7% versus 34.7% in the simvastatin monotherapy group; absolute risk reduction 2.0%; HR 0.936; 95% CI 0.89–0.99; P = 0.016) but no difference in mortality. Adverse effects were similar in the two groups, demonstrating the safety profile of ezetimibe. The application of the findings of IMPROVE-IT are twofold. First, it supports the theory of “lower is better” for LDL-C levels to reduce cardiovascular risk. Second, it demonstrated a significant add-on effect of ezetimibe to statin in terms of both LDL-C reduction and reduction of cardiovascular events.


Subanalysis of IMPROVE-IT demonstrated greater reduction in LDL-C among patients with diabetes within the first year of the trial: 43 mg/dL (1.1 mmol/L) reduction in the simvastatin–ezetimibe arm versus 23 mg/dL (0.6 mmol/L) reduction in the simvastatin monotherapy arm. Addition of ezetimibe also conferred a larger protective cardiovascular benefit in high-risk patients with diabetes. Therefore, combination therapy of statin and ezetimibe may be an effective option for patients with diabetes who are unable to tolerate high-intensity statin or those requiring large LDL-C reductions.


The results of the open-label Ezetimibe Lipid Lowering Trial on Prevention of Atherosclerosis in 75 or Older (EWTOPIA75) showed that in 4000 elderly Japanese patients, without prior history of CHD but with LDL-C level ≥ 140 mg/dL and one or more other cardiovascular risk factors (including diabetes, hypertension, prior cerebral infarction, or peripheral artery disease), patients receiving ezetimibe monotherapy had significantly lower cardiovascular events, including strokes, over a 5-year follow-up than patients not receiving ezetimibe.


Indications


Current indications for ezetimibe approved by the FDA include use in primary hypercholesterolemia (heterozygous familial and nonfamilial), as monotherapy or as combination therapy with statins, as adjunctive therapy to diet for the reduction of elevated total cholesterol, LDL-C, and apoB. Combination therapy with atorvastatin or simvastatin is approved for lipid-lowering treatment in homozygous familial hypercholesterolemia, as an adjunct to other lipid-lowering treatments (e.g. LDL apheresis), or used if such treatments are unavailable. Ezetimibe can be combined with fenofibrate for reduction of elevated total cholesterol, LDL-C, apoB, and non-HDL-C in patients with mixed hyperlipidemia. The FDA also approved ezetimibe as adjunctive therapy to diet for the reduction of elevated sitosterol and campesterol levels in patients with homozygous familial sitosterolemia. Both the 2018 AHA/ACC guidelines and 2019 ESC/EAS guidelines recommend the use of ezetimibe as add-on therapy in patients with ASCVD who are unable to achieve recommended LDL-C with maximally tolerated statin therapy, especially very-high-risk patients. The 2019 ESC/EAS guidelines extend the use of ezetimibe as an add-on therapy to statin even in primary-prevention patients unable to achieve the individualized LDL-C goals set for the specific level of risk (Class Ib).


Dosage and Effect


The recommended dosage of ezetimibe is 10 mg once daily, administered with or without food. It may be taken at the same time as a statin. As fixed-dose monotherapy, ezetimibe produces an approximate 18% reduction in LDL-C and modest beneficial effects on triglycerides and HDL-C, with no apparent safety concerns. No dosage adjustment is necessary in patients with mild hepatic insufficiency, but the effects of ezetimibe have not been examined in patients with moderate or severe hepatic insufficiency. No dosage adjustment is necessary in patients with renal insufficiency or in geriatric patients. As cotherapy , the lipid effects of ezetimibe and a statin appear to be additive. For example, with pravastatin, 10–40 mg, LDL-C fell by 34%–41% and triglycerides by 21%–23%, and HDL-C rose by 7.8%–8.4%, with a safety profile similar to pravastatin alone. Coadministration of a resin may decrease the bioavailability of ezetimibe; therefore, ezetimibe should be administered either 2 or more hours before or 4 or more hours after administration of the resin.


The FDA recommendations to reduce myopathy with simvastatin are also applicable to combined simvastatin–ezetimibe (Vytorin). In brief, simvastatin–ezetimibe should not be used with the conazole group of drugs, some antibiotics, HIV protease inhibitors, cyclosporine, and gemfibrozil.


Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors (Repatha, Praluent)


PCSK9 inhibitors have been shown to be the most potent LDL-lowering class of drug. The FDA has approved two monoclonal antibodies in this class of drugs for LDL-C reduction and secondary prevention: evolocumab (Repatha) and alirocumab (Praluent).


PCSK9 is a hepatic protease that attaches to and internalizes LDL receptors into lysosomes, promoting LDL receptor degradation. PCSK9 inhibitors bind and inactivate extracellular PCSK9 and prevent its interaction with the LDL receptor, thereby preventing trafficking of LDL receptors to lysosomes and therefore increasing the number of LDL receptors on the surface of liver cells available to clear LDL, which in turn lowers LDL-C levels in the blood. Additionally, PCSK9 inhibitors significantly reduce total cholesterol, apoB, triglycerides, and Lp(a).


Despite widespread use of the statins, a large proportion of high-risk patients are not able to achieve targeted LDL-C levels and have residual risk. Previously, options were limited for patients who develop CVD despite being on maximally tolerated statin therapy, were intolerant to statin therapy, or had severe hypercholesterolemia. The PCSK9 inhibitors evolocumab and alirocumab have been shown in multiple phase III and IV clinical trials to provide consistent and substantial LDL-C reductions of 50%–70% across a broad range of CVD risk, pretreatment LDL-C levels, and background therapy and have been studied as monotherapy (MENDEL-2, ODYSSEY COMBO I ), as an add-on to statin therapy (LAPLACE-2, ODYSSEY CHOICE I ), or in individuals with heterozygous familial hypercholesterolemia (RUTHERFORD-2, ODYSSEY-FH ). The Goal Achievement After Utilizing an Anti-PCSK9 Antibody in Statin Intolerant Subjects 3 (GAUSS-3) randomized clinical trial also demonstrated tolerability of PCSK9 inhibitor therapy in patients with muscle-related statin intolerance. These agents can be used as both adjunctive and alternative therapy for reducing LDL-C and have ushered in a new era of lipid-lowering therapy


Data from two large landmark outcome trials, FOURIER and ODYSSEY OUTCOMES, provided robust proof of the clinical safety and efficacy of evolocumab and alirocumab in reducing ASCVD events when used in combination with a statin compared to statin monotherapy. FOURIER was a large randomized, double-blind, placebo-controlled clinical trial investigating the efficacy and safety of evolocumab when added to high-intensity or moderate-intensity statin therapy in patients with stable clinical ASCVD. Patients were randomized to receive evolocumab or matching placebo as subcutaneous injections for 26 months. The majority of the 27,564 patients (69.3%) were on a high-intensity statin, 30.4% were on moderate-intensity statin, and 5.2% were on ezetimibe. At 48 weeks, evolocumab reduced LDL-C levels by 59% from baseline compared with placebo for a mean absolute reduction of 56 mg/dL. The primary efficacy endpoint (composite of cardiovascular death, MI, stroke, hospitalization for unstable angina, or coronary revascularization) occurred in 9.8% of patients in the evolocumab group and 11.3% of patients in the placebo group, indicating a 15% risk reduction with evolocumab (HR 0.85; 95% CI 0.79–0.92; P < 0.001) and NNT of 74. No effect was observed on hospitalization for unstable angina, cardiovascular death, or all-cause death but there were significant reductions in the risk for nonfatal MI (HR 0.73; 95% CI 0.65–0.82; P < 0.001), nonfatal stroke (HR 0.79; 95% CI 0.66–0.95; P = 0.01), and coronary revascularization (HR 0.78; 95% CI 0.71–0.86; P < 0.001).


ODYSSEY OUTCOMES assessed the efficacy of adding alirocumab to maximally tolerated statins on cardiovascular outcomes in 18,924 patients who had an ACS within a year of enrolling in the trial. At 48 weeks, alirocumab reduced LDL-C levels by 54.7% compared to placebo for an absolute reduction of 48.1 mg/dL. Patients who received alirocumab had significant reduction in major cardiovascular events; the primary composite endpoint (composite of cardiovascular death, MI, stroke, or hospitalization for unstable angina) occurred in 9.5% of patients in the alirocumab group and 11.1% of patients in the placebo group, resulting in a 15% risk reduction (HR 0.85; 95% CI 0.78–0.93; P = 0.0003) and NNT of 62. There were also significant reductions in nonfatal MI (6.6% versus 7.6%; P = 0.006), stroke (1.2% versus 1.6%; P = 0.01), and all-cause death (3.5% versus 4.1%; nominal P = 0.026) but no difference in cardiovascular death. This was the first trial to show mortality benefit with the addition of a nonstatin medication. Whereas alirocumab significantly reduced total deaths, cardiovascular death or noncardiovascular death analyzed separately was not significantly reduced. Patients with nonfatal cardiovascular events were at increased risk for both cardiovascular and noncardiovascular deaths. Because alirocumab reduced total nonfatal cardiovascular events ( P < 0.001), the authors postulated that this may have attenuated the number of cardiovascular and noncardiovascular deaths and thus led to a reduction in total mortality.


Patients with peripheral artery disease are a very-high-risk group and often undertreated. A subanalysis from FOURIER demonstrated that evolocumab reduced both major adverse cardiovascular events and major adverse limb events, defined as acute limb ischemia, major amputation, or urgent peripheral revascularization in patients with history of peripheral artery disease. Furthermore, the subgroup of patients with peripheral artery disease were shown to derive the most benefit from alirocumab therapy, as this subgroup achieved the highest absolute risk reduction in both adverse cardiovascular and adverse limb events. Evolocumab also reduced the risk of major adverse limb events in all patients, regardless of peripheral artery disease diagnosis at baseline, with reduction in lower limb events shown to be directly proportional to achieved LDL-C level, down to an LDL-C level of 10 mg/dL. Thus, PCSK9 inhibitor therapy should be strongly considered to reduce risk for cardiovascular and peripheral artery disease events.


Indications


FDA approved use of alirocumab (Praluent) and evolocumab (Repatha) for adult patients with heterozygous familial hypercholesterolemia or in patients with clinically significant ASCVD requiring additional LDL-C lowering after diet and maximally tolerated statin therapy. Evolocumab has also been approved for use in patients with homozygous familial hypercholesterolemia. The FDA has also approved use of both alirocumab and evolocumab to reduce the risk of MI, stroke, and unstable angina requiring hospitalization in adults with established CVD based on FOURIER and ODYSSEY OUTCOMES. The 2018 AHA/ACC Guideline on the Management of Blood Cholesterol reserve PCSK9 inhibitors for the treatment of patients with very high ASCVD risk on maximally tolerated statins and ezetimibe (Class IIa recommendation) and for primary prevention patients with heterozygous familial hypercholesterolemia on maximally tolerated statins and ezetimibe (Class IIb recommendation). These conservative recommendations are based on issues related to cost effectiveness, insurance coverage, affordability, and patient acceptance of subcutaneous administration. However, after publication of the guideline, the price of both drugs was reduced by > 60% by their respective pharmaceutical companies. The subsequently published 2019 ESC/EAS dyslipidemia guidelines, on the other hand, recommend the use of PCSK9 inhibitors in any patients with a documented ASCVD event, even if the event is not recent, if unable to achieve goal LDL-C < 55 mg/dL with statin and ezetimibe (Class IIa).


Dosage and Side Effects


Alirocumab is administered subcutaneously 75 mg every 2 weeks and can be titrated to 150 mg every 2 weeks; alternatively, alirocumab can also be administered 300 mg once every 4 weeks (monthly), according to patient preference. Evolocumab is administered subcutaneously 140 mg every 2 weeks or 420 mg every 4 weeks. Pooled data from multiple large clinical trials showed that these agents are well tolerated with no difference in serious adverse effects compared to placebo. For both alirocumab and evolocumab, the most common adverse events reported in clinical trials were injection-site reactions (erythema, itchiness, swelling, pain, or tenderness), nasopharyngitis, and upper respiratory tract infection. The most common adverse events that led to drug discontinuation were allergic reactions with alirocumab and myalgia, nausea, and dizziness with evolocumab. PCSK9 inhibitors seem to provoke fewer muscle-related adverse effects than statins and do not appear to cause muscle toxicity or elevated liver enzymes. The Evaluating PCSK9 Binding Antibody Influence on Cognitive Health in High Cardiovascular Risk Subjects (EBBINGHAUS) study in a subgroup of patients from FOURIER found no significant difference in cognitive function in patients who received evolocumab or placebo in addition to statin over a median of 19 months.


In Development: Inclisiran


In contrast with these monoclonal antibodies against PCSK9, inclisiran is a novel, synthetic, small interfering double-stranded RNA (siRNA) molecule that inhibits intracellular PCSK9 synthesis in hepatocytes. siRNA binds intracellularly to RNA-induced silencing complex, affecting the degradation of mRNA posttranscription, thus preventing translation. Inclisiran is a long-acting, synthetic siRNA directed against mRNA coding for PCSK9. It is conjugated to triantennary N-acetylgalactosamine carbohydrates, which specifically bind to abundant liver-expressed asialoglycoprotein receptors, leading to the uptake of inclisiran specifically into the hepatocytes. As mentioned for the previous PCSK9 inhibitors, any therapeutic approach to reduce circulating levels of PCSK9 offers an additional route through which plasma LDL-C levels can be controlled. This is especially important in clinical practice because of significant variability in individual responses to statins, with many individuals at risk for or with ASCVD failing to achieve LDL-C goals or exhibiting intolerance to statins. Such individuals may benefit from additional LDL-C lowering by other therapeutic means, as evidenced in the FOURIER and ODYSSEY OUTCOMES trials.


In two randomized, single-blind, placebo-controlled, phase I studies of inclisiran in healthy adult volunteers, significant, dose-dependent, long-term mean reductions in circulating PCSK9 and LDL-C levels were demonstrated, with similar safety profile and tolerability to placebo. ORION-1 was the first phase II, multicenter, double-blind, placebo-controlled, multiple-ascending-dose trial of inclisiran, conducted in 501 patients at high risk for or with history of ASCVD. The greatest reductions in LDL-C and PCSK9 levels were attained with the two-dose 300-mg regimen of inclisiran: 52.6%, and 69.1%, respectively, at 180 days. Serious adverse events occurred in 11% of the patients who received inclisiran and in 8% of the patients who received placebo. A follow-up study of ORION-1, in which participants were followed up to 1 year after initial injection, showed that treatment with inclisiran resulted in durable reductions in LDL-C over 1 year, with similar incidence of adverse events between inclisiran and placebo.


Three phase III clinical trials, ORION-9 (NCT03397121), ORION-10 (NCT03399370), and ORION-11 (NCT03400800), designed to evaluate the safety and efficacy of inclisiran in people with ASCVD and elevated LDL-C despite the maximum tolerated dose of LDL-C–lowering therapies, as well as in individuals with familial hypercholesterolemia, have been completed. All three studies demonstrated that inclisiran produced significant reductions in LDL-C and PCSK9 levels with an acceptable side effect profile, as compared to placebo. In ORION-9, a randomized trial in 482 patients with heterozygous familial hypercholesterolemia who were already taking statins and ezetimibe, inclisiran 300 mg administered as a subcutaneous injection on days 1, 90, 270, and 450 was superior to placebo in reducing LDL-C. In ORION-10, a randomized, parallel-group, double-blind, clinical trial in 1561 US patients with ASCVD who were taking maximally tolerated statin therapy, twice-yearly inclisiran injections (300 mg) reduced LDL-C by 56% over a follow-up of 18 months; serious and treatment-emergent side effects were similar between the two groups. ORION-11 showed similar results in 1617 European patients with ASCVD or at high risk for ASCVD who received inclisiran 300 mg on days 1, 90, 270, and 450 and had 50% reduction in LDL-C over 18 months; the overall adverse event profiles of the placebo- and inclisiran-treated groups in ORION-11 were similar. The ongoing ORION-4 trial (NCT03705234; HPS-4/TIMI 65/ORION-4) is designed to evaluate cardiovascular outcomes in 15,000 people with ASCVD; primary results are expected in 2024 and final completion in 2049.


Bempedoic Acid (Nexletol, Nexlizet [US]; Nilemdo, Nustendi [EU])


Bempedoic acid is a recently approved nonstatin therapy designed to inhibit cholesterol biosynthesis primarily in the liver. Bempedoic acid is administered as a prodrug that is converted to its active moiety primarily in the liver and inhibits adenosine triphosphate citrate lyase (ACL), an enzyme two steps upstream from HMG-CoA reductase along the cholesterol biosynthesis pathway, thereby effectively reducing cholesterol synthesis, resulting in LDL receptor upregulation and increased clearance of LDL from the bloodstream. Bempedoic acid can only converted to its active moiety by the enzyme ACSVL1, which is present in hepatocytes but not present in skeletal muscle ; therefore, based on its pharmacological profile, bempedoic acid may have fewer serious muscle adverse effects.


Five randomized, double-blind, placebo-controlled, parallel-group, multicenter phase III clinical trials known as Cholesterol Lowering via Bempedoic Acid, an ACL-inhibiting Regimen (CLEAR) have established safety, tolerability, and LDL-C–lowering efficacy of bempedoic acid in a total 3623 participants. In the largest of these trials, CLEAR Harmony, which studied bempedoic acid only in 2230 patients with ASCVD and/or heterozygous familial hypercholesterolemia on background statin therapy (85% using moderate- to high-intensity statin), bempedoic acid resulted in a significant 16.5% reduction in LDL-C and did not lead to higher incidence of overall adverse events compared to placebo over 52 weeks’ follow-up. In CLEAR Tranquility, in which 269 statin-intolerant patients on stable background therapy and open-label ezetimibe 10 mg were randomized to receive bempedoic acid 180 mg or placebo for 12 weeks, bempedoic acid + ezetimibe resulted in 28.5% greater LDL-C reduction than placebo + ezetimibe. In another phase III clinical trial (not part of the CLEAR program), the safety and efficacy of a fixed-dose combination tablet containing bempedoic acid 180 mg and ezetimibe 10 mg was evaluated in 301 patients with hypercholesterolemia and ASCVD and/or heterozygous familial hypercholesterolemia on background maximally tolerated statin therapy (35% on high-intensity statin, 35% on no statin). Fixed-dose combination therapy lowered LDL-C by 38% compared with placebo at week 12 and had a generally similar safety profile compared with bempedoic acid, ezetimibe, or placebo. . The ongoing CLEAR Outcomes trial (NCT02993406) is a cardiovascular event–driven, multinational, randomized, double-blind, placebo-controlled study in approximately 12,600 patients, with an estimated study duration of 4.75 years.


Indications


Bempedoic acid is indicated as an adjunct to diet and maximally tolerated statin therapy for the treatment of adults with heterozygous familial hypercholesterolemia or established ASCVD who require additional lowering of LDL-C. The European Medicines Agency indications also include use in patients who cannot tolerate statins.


Dosage, Effects, and Side Effects


Bempedoic acid is available as a tablet containing 180 mg of bempedoic acid or as a combination tablet containing 180 mg of bempedoic acid and 10 mg of ezetimibe. Either tablet is taken orally once a day with or without food.


The combined tablet is contraindicated in individuals with a known hypersensitivity to ezetimibe. Bempedoic acid may increase blood uric acid levels and may lead to gout, especially in patients with a history of gout. Bempedoic acid therapy may also be associated with an increased risk of tendon rupture, which may occur more frequently in patients over 60 years of age, patients taking corticosteroid or fluoroquinolone drugs, patients with renal failure, and patients with previous tendon disorders. In clinical trials of bempedoic acid, the most commonly reported adverse events were upper respiratory tract infection, muscle spasms, hyperuricemia, back pain, abdominal pain or discomfort, bronchitis, pain in extremity, anemia, and elevated liver enzymes; events reported less frequently, but still more often than with placebo, included benign prostatic hyperplasia and atrial fibrillation. For the combination bempedoic acid/ezetimibe tablet, the most commonly reported adverse events that were not observed in clinical trials of bempedoic acid or ezetimibe and occurred more frequently than with placebo were urinary tract infection, nasopharyngitis, and constipation.


Treatment with bempedoic acid has been associated with persistent changes in laboratory tests within the first four weeks of treatment, including increases in creatinine, blood urea nitrogen, platelet counts, liver enzymes, and creatine kinase and decreases in hemoglobin and leukocytes. Laboratory abnormalities generally return to baseline after discontinuation of treatment.


Bempedoic acid should not be taken during breastfeeding. Pregnant patients should consult their healthcare provider about whether to continue treatment during pregnancy. The safety and efficacy of bempedoic acid has not been established in patients under the age of 18. No adjustments in dosing are required for advanced age, mild or moderate renal impairment, or mild hepatic impairment; in patients with moderate hepatic impairment, no dosing adjustment is required for bempedoic acid, but the combination bempedoic acid/ezetimibe tablet is not recommended for patients with moderate or severe hepatic impairment.


Drug Interactions


Concomitant use of bempedoic acid with simvastatin or pravastatin results in increased statin concentration and increased risk for statin-related myopathy. Use of bempedoic acid with > 20 mg of simvastatin or > 40 mg of pravastatin should be avoided. Caution should be exercised when using combination bempedoic acid/ezetimibe with cyclosporine because of increased exposure to both ezetimibe and cyclosporine; cyclosporine concentrations should be monitored, and the potential risk/benefit ratio of concomitant use should be carefully considered. Coadministration of combination bempedoic acid/ezetimibe with fibrates other than fenofibrate is not recommended. Fenofibrate and ezetimibe may increase cholesterol excretion into the bile, leading to cholelithiasis; if cholelithiasis is suspected, gallbladder studies are indicated and alternative lipid-lowering therapy should be considered. Concomitant use of combination bempedoic acid/ezetimibe with cholestyramine decreases ezetimibe concentration, which may reduce efficacy; bempedoic acid/ezetimibe should be administered at least 2 hours before or at least 4 hours after bile acid sequestrants.


Omega-3 Fatty Acids (Fish Oils; Lovaza, Vascepa, Epanova)


Omega-3 fatty acids are a major class of polyunsaturated fatty acids. Studies show that omega-3 fatty acids in general decrease blood triglyceride and VLDL levels in hyperlipidemic individuals but may have no effect or may increase LDL-C in patients with very high triglycerides. These effects are most prominently seen with high-dose supplements (> 2 to 44 g/day). Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are two major types of omega-3 fatty acids. EPA and DHA have been shown to reduce inflammation, with production of resolvins and decrease in proinflammatory compounds. A number of other beneficial actions for atherosclerosis prevention have been reported, relating to endothelial function, oxidative stress, foam-cell formation, plaque formation/progression, platelet aggregation, thrombus formation, and plaque rupture. DHA is the major polyunsaturated fatty acid found in the brain and is important for brain development and function.


In the Multi-center, Placebo-controlled, Randomized, Double-blind, 12-Week Study with an Open-label Extension (MARINE) trial, in adult patients with very high fasting triglyceride levels between 500 mg/dL and 2000 mg/dL, patients treated with EPA ethyl ester (icosapent ethyl) 4 g/day for 12 weeks had a statistically significant placebo-adjusted median triglyceride reduction of 33% ( P < 0.0001) and a small reduction in LDL-C levels (5%). In addition, treatment with icosapent ethyl 4 g/day led to statistically significant placebo-adjusted median reductions from baseline in non-HDL-C (18%), total cholesterol (16%), VLDL-C (29%), and apoB (8.5%).


The efficacy of icosapent ethyl was also evaluated in ANCHOR, a phase III placebo-controlled randomized clinical trial in high-risk statin-treated patients (n = 702) with triglyceride levels between 200 mg/dL and 500 mg/dL and with LDL-C levels of 40–100 mg/dL. After 12 weeks, treatment with icosapent ethyl 4 g/day resulted in a significant median placebo-adjusted change from baseline in triglyceride levels of 21.5% ( P < 0.0001).


Historically, clinical trials assessing the role of fish oil supplementation or low-dose prescription omega-3 fatty acids showed variable results without clear benefit in prevention of cardiovascular events. However, both the Japan EPA Lipid Intervention Study (JELIS) and REDUCE-IT provided valuable findings to support clinical benefit of EPA as an add-on to statins in high-risk patients. In JELIS, 18,645 Japanese patients with hypercholesterolemia were randomly assigned to receive either low-intensity statin therapy plus EPA 1.8 g/day or statin therapy alone (there was no placebo group). The risk of major coronary events was significantly reduced by 19% in the group that received EPA plus statin therapy compared with the group that received statin therapy alone. Additional evidence was obtained from REDUCE-IT, which was the first large multinational cardiovascular outcomes study that evaluated the effect of prescription EPA therapy as an add-on to statins. Over 8000 patients with high cardiovascular risk who, despite stable statin therapy, had residual hypertriglyceridemia (fasting triglyceride of at least 135 mg/dL) were randomized to 2 g of icosapent ethyl twice daily (total daily dose, 4 g) or placebo. Patients in the icosapent ethyl group had significantly reduced risk for the primary endpoint of cardiovascular events (cardiovascular death, nonfatal MI, nonfatal stroke, coronary revascularization, or unstable angina), which occurred in 17.2% of the icosapent ethyl group versus 22.0% of the placebo group ( P < 0.001) for an absolute risk reduction of 4.8%. Cardiovascular death was also significantly reduced with icosapent ethyl compared with placebo (4.3% versus 5.2%, respectively; P < 0.001).


Indications


Two prescription-strength omega-3 fatty acids, omega-3-acid ethyl esters (Lovaza) and icosapent ethyl (Vascepa), are approved by the FDA to treat severe hypertriglyceridemia (triglycerides levels 500 mg/dL or more) at a dose of 4 g/day. A third formulation, omega-3 carboxylic acids (Epanova), was being evaluated in A Long-Term Outcomes Study to Assess Statin Residual Risk Reduction with Epanova in High Cardiovascular Risk Patients with Hypertriglyceridemia (STRENGTH), which was discontinued due to low likelihood of benefit.


Each 1-g capsule of omega-3-acid ethyl esters contains ≥ 900 mg of ethyl esters of omega-3 fatty acids, predominantly a combination of ethyl esters of EPA (~ 465 mg) and DHA (~ 375 mg), whereas icosapent ethyl contains only EPA and no DHA. The suggested daily dosage of both omega-3-acid ethyl esters and icosapent ethyl is 4 g twice a day with food. Each gram of omega-3 carboxylic acids contains 850 mg of polyunsaturated fatty acids, including multiple omega-3 fatty acids (EPA and DHA being most abundant). Both EPA and DHA reduce triglyceride levels; however, DHA also raises LDL-C.


Icosapent ethyl is also approved by the FDA as an adjunctive therapy to statins to reduce the risk of ASCVD events among adults with established ASCVD or with diabetes and ≥ 2 risk factors who also have elevated triglyceride levels ≥ 150 mg/dL. It is the first and only FDA-approved medication to reduce cardiovascular risk beyond cholesterol-lowering therapy in high-risk patients. The 2019 ESC/EAS guidelines recommend the addition of icosapent ethyl 4 g/day in high-risk patients with triglyceride of 135–499 mg/dL despite high-intensity or maximally tolerated statin treatment (Class IIa). The NLA issued a scientific statement and the ADA amended its Standard of Care to endorse the use of icosapent ethyl in individuals with clinical ASCVD or diabetes mellitus with other cardiovascular risk factors to treat elevated triglycerides, as an add-on to statin therapy.


Side Effects


Because omega-3 fatty acids are obtained from the oil of fish, they should be used cautiously in patients with a fish allergy and/or shellfish allergy. Some data suggest that omega-3 fatty acids may prolong bleeding time; therefore, patients receiving omega-3 fatty acids with other drugs that affect coagulation should be monitored periodically. In clinical trials, treatment-emergent adverse events were similar in those treated with omega-3 fatty acids compared with placebo. In REDUCE-IT, the most common reported adverse reaction was arthralgia (2.3% for icosapent ethyl, 1.0% for placebo); other common adverse events occurring more frequently with icosapent ethyl than placebo were peripheral edema (6.5% versus 5.0%, respectively), constipation (5.4% versus 3.6%), and atrial fibrillation (5.3% versus 3.9% placebo). A larger percentage of patients in the icosapent ethyl group than in the placebo group were hospitalized for atrial fibrillation or flutter (3.1% versus 2.1%, P = 0.004). Icosapent ethyl patients had reductions in rates of cardiac arrest, sudden death, and myocardial infarctions. Serious adverse bleeding events occurred in 2.7% of the icosapent ethyl group and 2.1% of the placebo group ( P = 0.06), but the rate of anemia was significantly lower in the icosapent ethyl group than in the placebo group (4.7% versus 5.8%; P = 0.03).


Fibrates: Fibric Acid Derivatives


Fibrates are highly effective in reducing triglycerides, but as a rule, none of the fibrates reduce LDL-C as much as do the statins or PCSK9 inhibitors. Unlike statins, which have demonstrated clinical efficacy across a broad range of LDL-C levels, fibrates have primarily shown reductions in cardiovascular events in a subset of patients with high triglycerides (≥ 200 mg/dL [2.2 mmol/L]) and low HDL-C (< 40 mg/dL [1.0 mmol/L]). The primary role of fibrates is to decrease triglyceride level, atherogenic triglyceride-rich lipoproteins, and concentration of small, dense LDL particles. They are therefore suitable for use in atherogenic dyslipidemia. Fibrates are first-line therapy to reduce the risk for pancreatitis in patients with very high levels of plasma triglycerides and may be useful with more modest triglyceride elevations or when the primary dyslipidemia is low HDL-C.


At a molecular level, fibrates are agonists for the nuclear transcription factor peroxisome proliferator–activated receptor–α (PPAR-α), which stimulates the synthesis of the enzymes of fatty acid oxidation, thereby reducing VLDL triglycerides. Although all fibrates belong to the same class of drugs, structural differences between the compounds seem important because of the very different results between large-scale trials of clofibrate (unfavorable) and gemfibrozil (favorable; see below).


Combined Statin Plus Fibrate


In primary prevention, for patients with severe hypercholesterolemia or familial combined hyperlipidemia with marked triglyceride elevations, combination of a statin with a fibrate is an option. The statin is effective in the reduction of LDL-C, whereas the fibrate reduces triglycerides and triglyceride-rich lipoproteins. Statins metabolized through CYP3A4 have a greater risk of adverse interaction with fibrates during cotherapy with erythromycin, azole antifungals, and antiretrovirals. A logical combination would be a statin and a fibrate that are metabolized by noncompeting pathways, for example, fluvastatin or rosuvastatin combined with fenofibrate.


Class Warnings


There are five warnings or reservations for this class of drugs. First, the early experience with clofibrate suggested that fibrates may increase mortality. This fear has not been borne out by trials of other fibrates, and gemfibrozil has demonstrated significant coronary benefits. Second, hepatotoxicity may occur, with a pooled analysis of 10 placebo-controlled trials showing elevated transaminases in 5.3% of patients given fenofibrate compared to 1.1% on placebo. Third, cholelithiasis is a risk, because fibrates act in part by increasing biliary secretion of cholesterol; however, this was not found in the Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial (VA-HIT). Fourth, fibrates have an important drug interaction with concomitant oral anticoagulants; therefore, warfarin dose needs to be reduced by about 30%. Fifth, combined therapy with statins should be avoided unless the potential beneficial effect on lipids outweighs the increased risk for myopathy via competitively inhibiting CYP3A4, which leads to a reduction in statin metabolism.


Fenofibrate (Tricor, Trilipix, Lipofen, Antara, Lofibra)


Fenofibrate is a prodrug converted to fenofibric acid in the tissues. The FDA-approved indications are as adjunctive therapy to diet to reduce LDL-C and total cholesterol, triglycerides, and apoB and to increase HDL-C in severe hypertriglyceridemia or mixed dyslipidemia. Although indicated for treatment of hypertriglyceridemia, its effect on the risk for pancreatitis in patients with very high triglyceride levels, typically exceeding 1000 mg/dL, has not been well studied. The Trilipix formulation, which contains fenofibric acid rather than the ester, has an indication for mixed dyslipidemia in combination with statin therapy. Tricor is available in 48- and 145-mg tablets and is dosed at 48–145 mg once daily (half-life of 20 hours), taken with food to optimize bioavailability. Other formulations have slightly altered dosing. Predisposing diseases such as obesity, diabetes, CKD, chronic liver disease, nephrotic syndrome, and hypothyroidism need to be excluded and treated prior to initiating treatment. The Diabetes Atherosclerosis Intervention Study (DAIS) suggests that treatment with fenofibrate in patients with type 2 diabetes reduces progression of atherosclerosis, with a nonsignificant trend toward cardiovascular event reduction. The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study similarly attempted to assess the effect of fenofibrate on CVD events in patients with type 2 diabetes, but failed to show a benefit on the primary endpoint of coronary events (MI and CHD death), possibly because the study design allowed for initiation of statin therapy in both the placebo and fenofibrate treatment arms. Despite these null findings, FIELD did show a decrease in total cardiovascular events, primarily caused by significant reductions in nonfatal MI and revascularizations, as well as a significant benefit on the primary endpoint in the subgroup with high triglycerides and low HDL-C. The Action to Control Cardiovascular Risk in Diabetes Lipid (ACCORD Lipid) trial was conducted in 5500 patients with type 2 diabetes on statin therapy who were randomized to receive fenofibrate or placebo. Combination therapy with fenofibrate and statin did not significantly reduce major cardiovascular events (HR 0.92; 95% CI 0.79–1.08; P = 0.32) as compared to statin alone, and no cardiovascular benefit was found with the drug combination, except in a subgroup of individuals with low HDL-C and high triglycerides at baseline. Post hoc analyses of three other fibrate trials, including the Helsinki Heart Study, the Bezafibrate Infarction Prevention study (BIP), and FIELD, similarly suggested benefit with a fibrate in a subgroup of patients with atherogenic dyslipidemia. Thus the cumulative body of evidence indicates that the prime lipid-lowering therapy for prevention of macrovascular complications in most diabetic patients remains a statin.


Weight reduction, increased exercise, and elimination of excess alcohol are essential steps in the overall control of triglyceride levels. Fenofibrate coadministered with cyclosporine may cause renal damage with decreased excretion of fenofibrate and increased blood levels. Use with caution in patients taking oral coumarin-type anticoagulants; anticoagulant dosage may need to be adjusted. Animal data suggest a deleterious effect in pregnancy. Avoid in nursing mothers (carcinogenic potential in animals). Use with caution in older adults or patients with renal dysfunction (renal excretion).


Gemfibrozil (Lopid)


Major Trials


Gemfibrozil was used in the large primary-prevention Helsinki Heart Study in 4081 apparently healthy men with modest hypercholesterolemia (non-HDL-C ≥ 200 mg/dL) observed for 5 years. Gemfibrozil 600 mg twice daily led to a major increase in HDL-C (10%), decreases in total cholesterol, LDL-C, and non-HDL-C (11%, 10%, and 14%, respectively), and a substantial reduction in triglycerides (43%), with a 34% reduction in coronary events (fatal and nonfatal MI and cardiac death). Although the total death rate was not different between treatment groups, the study was not powered to assess mortality. An open-label follow-up study found mortality reduction after 13 years. Despite the theoretical risk of gallstone formation with fibrate therapy, none was reported during the study.


VA-HIT was a secondary-prevention trial in 2531 men with CHD whose primary abnormality was low HDL-C: < 40 mg/dL (1 mmol/L), with a mean of 32 mg/dL. The entry criterion for LDL-C was ≤ 140 mg/dL (3.6 mmol/L), with a mean of 112 mg/dL. Over 5 years, mean HDL-C was 6% higher, mean triglyceride 31% lower, and total cholesterol 4% lower with gemfibrozil than with placebo, whereas mean LDL-C level was not different between treatment groups. The primary outcome of nonfatal MI or coronary death was reduced by 22% with gemfibrozil (event rates 17.3% in the gemfibrozil group versus 21.7% in the placebo group, P < 0.001). The 5-year NNT was 23, which compared well with that of the major statin trials. It must be noted that gemfibrozil was only studied as monotherapy and not as an add-on to statin in this clinical trial. Therefore, with statins currently the first-line option and multiple other drugs showing benefit as an add-on therapy (discussed previously), gemfibrozil probably has limited clinical utility.


Dose, Side Effects, Contraindications


Gemfibrozil is currently approved in the United States for treatment of severe hypertriglyceridemia and mixed dyslipidemia (elevated LDL-C, decreased HDL-C, and increased triglycerides). The dose is 1200 mg given in two divided doses 30 minutes before the morning and evening meals. Contraindications are hepatic or severe renal dysfunction, preexisting gallbladder disease (possible risk of increased gallstones, not found in VA-HIT), and coadministration with simvastatin, repaglinide, dasabuvir, or selexipag. There are drug interactions to consider. Because it is highly protein bound, gemfibrozil potentiates warfarin. When combined with statins, there is an increased risk for myopathy with myoglobinuria and a further rare risk for acute renal failure


Bezafibrate


Bezafibrate (Bezalip in the United Kingdom; not available in the United States) resembles gemfibrozil in its overall effects, side effects, and alterations in blood lipid profile. Uniquely among fibrates, bezafibrate is also a PPAR-γ agonist, thereby theoretically stimulating the enzymes that regulate glucose metabolism. Hence, plasma glucose tends to fall with bezafibrate, which may be useful in patients with diabetes or abnormal glucose metabolic patterns. In patients with CHD, bezafibrate slows the development of insulin resistance. As with other fibrates, warfarin potentiation is possible, and cotherapy with statins should ideally be avoided. In addition, myositis, renal failure, alopecia, and loss of libido have occurred. Bezafibrate is dosed at 200 mg two to three times daily; however, once daily is nearly as effective, and a slow-release formulation is available (in the United Kingdom: Bezalip-Mono , 400 mg once daily). Some increase in plasma creatinine is very common and of unknown consequence. The major limitation with bezafibrate is that, unlike gemfibrozil and the statins, no major long-term outcome trials have provided clear results. In BIP, conducted in 3090 patients with previous MI or angina and low HDL-C combined with modestly elevated LDL-C, HDL-C was increased by 18% and triglyceride decreased by 21% with bezafibrate, but the primary endpoint of fatal or nonfatal MI or sudden death was not significantly different between treatment groups (13.6% with bezafibrate versus 15.0% with placebo; P = 0.26), except post hoc in a subgroup of patients with initial triglyceride levels ≥ 200 mg/dL.


Bile Acid Sequestrants: Resins (Questran, Welchol, Colestid)


Bile acid sequestrants— cholestyramine (Questran), colesevelam (Welchol) , and colestipol (Colestid) —bind to bile acids to promote the secretion of bile acids into the intestine, resulting in increased loss of hepatic cholesterol into bile acids and hepatic cellular cholesterol depletion. The latter leads to a compensatory increase in hepatic LDL receptors, increasing the removal of LDL from the circulation and decreasing total cholesterol and LDL-C. There may be a transitory compensatory rise in plasma triglycerides that is usually mild but may require cotherapy or discontinuation of the agent. Colesevelam has an additional FDA indication for glycemic control in the treatment of type 2 diabetes, as combination therapy with metformin, sulfonylureas, or insulin. The major outcome trial conducted with resins was the Lipid Research Clinics Coronary Primary Prevention Trial, in which cholestyramine modestly reduced CHD (primary endpoint: CHD death or nonfatal MI) in hypercholesterolemic patients and improved blood lipid profiles but had no effect on overall mortality. Drug interactions include interference with the absorption of digoxin, warfarin, thyroxine, and thiazides, which need to be taken 1 hour before or 4 hours after the sequestrant. Impaired absorption of vitamin K may lead to bleeding and sensitization to warfarin. Poor palatability is the major problem. Combination therapy is often undertaken, and coadministration with a statin may exploit the complementary mechanisms of action of these two drug classes. Resins may increase triglycerides, so a second agent such as nicotinic acid or a fibrate may be required to lower triglycerides. Resins should be used with caution in patients with hypertriglyceridemia.


Nicotinic Acid (Niacin; Niaspan)


Nicotinic acid was the first hypolipidemic drug shown to reduce overall mortality, in 15-year follow-up from the Coronary Drug Project. The basic effect of nicotinic acid may be decreased mobilization of free fatty acids from adipose tissue, so that there is less substrate for hepatic synthesis of lipoprotein lipid. Consequently, there is less secretion of lipoproteins so that LDL particles, including triglyceride-rich VLDL, are reduced. Nicotinic acid also increases HDL-C and reduces Lp(a).


In the angiographic Familial Atherosclerosis Treatment Study (FATS), men with apoB ≥ 125 mg/dL, CHD, and family history of CVD received either lovastatin (20 mg BID) or nicotinic acid (1 g QID), combined with colestipol (10 g TID). Both regimens were equally effective on blood lipids, and angiographically measured coronary stenosis was lessened, although side effects were worse, with nicotinic acid.


The AIM-HIGH study, an outcomes study examining the effect of adding extended-release niacin to simvastatin in patients with cardiovascular disease, was stopped early because of lack of clinically meaningful efficacy. Niacin failed to demonstrate incremental benefit in cardiovascular event reduction for patients already optimally treated with lipid-lowering therapy to a mean LDL-C of 71 mg/dL at baseline; in addition, an unexplained increase in ischemic stroke was observed in the niacin arm. After 36 months, the difference in HDL-C between treatment groups was only 4 mg/dL; the study may have been underpowered to show benefit of niacin on top of statin therapy.


Laropiprant was developed in an attempt to reduce the side effects of niacin that led to poor patient compliance but was discontinued. In the Heart Protection Study 2–Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE), a large multicenter, double-blind, controlled clinical trial, 25,673 patients with prior CVD, all on LDL-lowering therapy with simvastatin 40 mg, were randomized to receive either extended-release niacin–laropiprant combination tablets (a total of 2 g of niacin and 40 mg of laropiprant) daily or matching placebo. Over a median follow-up of 4 years, HDL-C levels increased by 6 mg/dL, triglycerides levels decreased by 33 mg/dL, and LDL-C levels decreased by 10 mg/dL in the niacin–laropiprant group. However, no significant difference in the incidence of major vascular events (nonfatal MI, stroke, coronary or noncoronary revascularization, or death from coronary causes) was demonstrated between patients assigned to niacin–laropiprant and those assigned to placebo (13.2% versus 13.6%, respectively, P = 0.29). Lack of efficacy was uniform in all subgroups defined according to different types of vascular disease or diabetes. Patients assigned to niacin–laropiprant were more likely to have disturbances in diabetes control (11.1% versus 7.5%; P < 0.001) and a new diagnosis of diabetes (5.7% versus 4.3%; P < 0.001) than those assigned to placebo. Study drug was discontinued in 25.4% of the niacin–laropiprant group compared with 16.6% of the placebo group ( P < 0.001). Furthermore, the niacin–laropiprant group had a significant excess in serious adverse events associated with the gastrointestinal system (mostly bleeding and ulcerations; 4.8% versus 3.8%; P < 0.001), myopathy (3.7% versus 3.0%; P < 0.001), and skin rash/ulceration (0.7% versus 0.4%; P = 0.003), as well as excess bleeding events (mostly gastrointestinal and intracranial; 2.5% versus 1.9%; P < 0.001) and infections (8.0% versus 6.6%; P < 0.001).


Both AIM-HIGH and HPS2-THRIVE failed to show any incremental clinical benefit from niacin added to standard LDL-lowering therapy and raised doubts about the safety profile of niacin. With widespread acceptance of statin therapy and recent landmark trials supporting the safety and clinical benefit of ezetimibe and PCSK9 inhibitors, the role of niacin as lipid-modifying therapy has become very limited.


Dose, Side Effects, and Contraindications


The dosage required for lipid lowering is up to 4 g of immediate-release (crystalline) niacin daily, achieved gradually with a low starting dose (100 mg twice daily with meals to avoid gastrointestinal discomfort) that is increased until the lipid target is reached, or side effects occur. The extended-release formulation (Niaspan) is available in an initiation package that up-titrates the dose to reduce side effects. The recommended dose of extended-release niacin is 1–2 g once daily at bedtime. Because of the difference in dosing, care must be taken in switching patients between immediate-release and extended-release formulations.


Niacin has numerous side effects , which can be lessened by carefully building up the dose. Nicotinic acid causes prostaglandin-mediated symptoms such as flushing, dizziness, and palpitations. Flushing, which is very common, lessens with time and with use of the extended-release formulation; flushing is also reduced by taking niacin with food. Caution should be used in patients with peptic ulcer, diabetes, liver disease, or a history of gout. Impaired glucose tolerance and increased blood urate are reminiscent of thiazide side effects, also with an unknown basis. Hepatotoxicity may be linked to some long-acting preparations (sustained-release formulations), whereas flushing and pruritus are reduced. Myopathy is rare. Use in pregnant women is questionable.


Non–LDL-C Therapies in Development


Considerable residual cardiovascular persists despite well-controlled LDL-C levels, and epidemiological and genetic studies have established the role of other lipid parameters, most notably triglycerides, triglyceride-rich lipoproteins (VLDL, chylomicrons, and remnants), and Lp(a), in residual cardiovascular risk, especially among patients on maximal statin therapy. Advances in gene-silencing technology through antisense oligonucleotide inhibition or siRNA provide novel approaches to target lipid parameters, by degrading mRNA transcripts of specific genes to reduce protein production and plasma lipoprotein levels. As discussed previously with inclisiran, the most recently developed agents have been modified to target the liver, allowing much lower doses to be used, for improved safety profiles compared with prior delivery approaches. Pharmacological use of monoclonal antibodies is also being examined for non–LDL-C targets.


Targeting Lp(a)


Therapies targeting Lp(a) currently in development include an siRNA to apo(a) called AMG 890 (formerly ARO-LPA; Amgen), which is currently completing phase I testing (NCT03626662 ) and beginning phase II trials, and an antisense oligonucleotide targeted to apo(a) called TQJ230 (formerly AKCEA-APO(a)-L Rx ; Novartis). Results of a phase II clinical study of TQJ230 in 286 patients with established CVD and elevated levels of Lp(a) showed significant Lp(a) reductions that were dose dependent and without any serious adverse effects such as thrombocytopenia. Novartis is conducting a phase III cardiovascular outcomes trial called Lp(a) HORIZON (NCT04023552 ).


Targeting Triglycerides


To reduce triglycerides, gene silencing can be employed to target proteins that are involved in the production or clearance of triglyceride-rich lipoproteins, such as apoC-III (present on triglyceride-rich lipoproteins) and angiopoetin-related protein 3 (ANGPTL3, which inhibits lipoprotein lipase and catabolism of triglyceride-rich lipoproteins), thereby reducing circulating triglyceride levels. Antisense oligonucleotides and siRNAs that target each of these proteins are in development.


Volanesorsen (AKCEA-APOCIII Rx ) is a second-generation antisense oligonucleotide developed to reduce circulating apoC-III and triglyceride levels. In the phase III, double-blind, randomized, 52-week APPROACH Study trial conducted in 66 patients with familial chylomicronemia syndrome, volanesorsen reduced mean triglyceride level by 77%, compared with an 18% increase in triglyceride level with placebo; thrombocytopenia and injection-site reactions were significant adverse events. Another phase III study of volanesorsen, the COMPASS Study conducted in patients with severe hypertriglyceridemia (triglycerides ≥ 500 mg/dL), also included patients with familial chylomicronemia syndrome, and based on these studies, volanesorsen was approved for treatment of familial chylomicronemia syndrome in the European Union but not the United States. Volanesorsen is undergoing further phase III testing in the ongoing APPROACH Open-Label Study in patients with familial chylomicronemia syndrome and the BROADEN Study in patients with familial partial lipodystrophy.


AKCEA-APOCIII-L Rx , a modified version of volanesorsen, is a second-generation ligand-conjugated antisense oligonucleotide that targets the liver; it is more potent and has a much better safety profile than volanesorsen. In a phase I/IIa study in 67 healthy volunteers, triglyceride levels were significantly reduced by up to 77%, with one injection-site reaction and no platelet count reductions. Phase II results are expected in 2020, and a phase III study is under way.


ARO-APOC3 is a hepatocyte-targeted siRNA to apoC-III. Results of a phase I/IIa trial showed that single doses in 40 healthy volunteers reduced serum apoC-III levels by 70%–91% and reduced serum triglycerides by 41%–55% at week 16. A phase I trial that includes patients with hypertriglyceridemia and patients familial chylomicronemia syndrome is under way.


ANGPTL3 is another promising protein target for triglyceride reduction. Evinacumab (REGN1500; Regeneron) is a human monoclonal antibody against ANGPTL3 that reduced fasting triglyceride levels by up to 70% and LDL-C levels by up to 23% in a phase I trial in healthy adults. In a small phase II study in nine patients with homozygous familial hypercholesterolemia, evinacumab reduced LDL-C by 49% and triglycerides by 47%. Another phase II clinical trial (NCT03175367), in 252 patients with heterozygous familial hypercholesterolemia or with hypercholesterolemia and ASCVD, is currently under way.


AKCEA-ANGPTL3-L Rx (Pfizer), an antisense therapy against hepatic ANGPTL3, similarly reduced triglycerides by up to 63% and LDL-C by 33% without any serious adverse events in a phase I, randomized, double-blind, placebo-controlled trial in 44 healthy adults. It is currently being evaluated in phase II studies in patients with familial chylomicronemia syndrome, familial partial lipodystrophy, or type 2 diabetes, hypertriglyceridemia, and nonalcoholic fatty liver disease.


ARO-ANG3 is a hepatocyte-targeting siRNA against ANGPTL3 under development by Arrowhead. Phase I/IIa safety and efficacy data showed that a single dose in normal healthy volunteers led to dose-dependent reductions in ANGPTL3 levels of 43%–75% and in triglyceride levels of 47%–53% by week 16, with no serious adverse events reported.


Although the findings for these developing drugs are promising, evidence from large cardiovascular outcome studies and long-term safety trials is needed before these novel, targeted, gene-silencing technologies can be evaluated for use in clinical practice.


Summary


Primary Prevention


In primary prevention of CVD, global risk factor assessment and correction are the recommended approach. The atherogenic components of blood lipids, especially LDL-C, are an important part of an overall risk factor profile that includes factors that cannot be changed, such as age, sex, and family history of premature disease, and those that can, such as blood pressure, diet, smoking, exercise, and weight (see Table 6.1 ). The Pooled Cohort Equation should be used to guide treatment and identify individuals for whom aggressive lifestyle modification and statin therapy are indicated. For a heart-healthy diet, a Mediterranean or DASH diet is currently recommended. Risk-enhancing factors (see Table 6.1 ) can help identify individuals at increased risk who may benefit most from initiating or maximizing statin therapy. The ideal blood cholesterol and LDL-C levels appear to be falling lower and lower, based on clinical trial results.


Secondary Prevention


In secondary prevention, strict LDL-C lowering with high-intensity statin therapy, along with lifestyle modification, is an essential part of a comprehensive program of risk-factor modification. The 2018 AHA/ACC guidelines make strong recommendation to use a threshold of LDL-C ≥ 70 mg/dL in patients with established ASCVD for augmenting statin therapy with add-on ezetimibe or (in very-high-risk patients) PCSK9 inhibitor therapy. In individuals already on statin therapy who have residual hypertriglyceridemia, strong evidence supports add-on icosapent ethyl. Strict dietary modification and exercise (minimum of 150 minutes of moderate- or 75 minutes of high-intensity exercise per week) must be emphasized in all patients.


Diabetes


Diabetes is regarded as a risk category in its own right in the 2018 AHA/ACC cholesterol guidelines, as this high-risk group warrants aggressive risk reduction. Guidelines recommend initiation of moderate-intensity statin irrespective of LDL-C levels in patients with diabetes aged 40–75 years.


Statins as Initial Treatment


Statin trials have shown substantial reductions in total and cardiac mortality as well as major adverse cardiovascular events, and statins are recommended for four patient management groups (according to the 2018 AHA/ACC guidelines): 1) secondary prevention for patients with established ASCVD; 2) patients with severe hypercholesterolemia (LDL-C ≥ 190 mg/dL); 3) patients with diabetes; and 4) primary prevention. Statins have few serious side effects or contraindications. In primary prevention, the Pooled Cohort Equation can be used to stratify risk, with 10-year risk < 5% considered low, 5 to < 7.5% borderline, 7.5 to < 20% intermediate, and ≥ 20% high. In intermediate- and high-risk groups, statin therapy is appropriate, with high-intensity statin preferred for the latter group (Class I recommendation). In the borderline-risk group, risk-enhancing factors (see Table 6.1 ) may be considered to guide risk discussion.


Combination Therapy


Combination therapy is now increasingly used to achieve reductions in LDL-C and non-HDL-C. The principle is to combine two different classes of agents with different mechanisms of action, such as a statin and ezetimibe or a statin and a PCSK9 inhibitor. Recent trials have shown overwhelming evidence of efficacious LDL lowering as well as reduction of adverse cardiovascular events with these combined agents. The consensus is that judicious use of combination therapy, when required, is likely to confer more benefit than harm.


Other lipid-regulating agents, which are used less frequently because they lack clinical trial evidence of incremental clinical benefit and increase risk for adverse events, include fibrates, bile acid sequestrants, and niacin. The combination of fibrates and statin increases risk of myopathy and hepatotoxity. Niacin is usually not well tolerated because of multiple side effects including flushing, pruritus, skin rashes, and gastrointestinal issues. Bile acid sequestrants require caution in patients with high triglycerides.


Hypertriglyceridemia


Intensive diet and lifestyle modification remains the cornerstone of therapy for elevated triglycerides, especially in patients with moderate hypertriglyceridemia (triglycerides 150–499 mg/dL [1.7–2.3 mmol/L]). Secondary causes of elevated triglycerides, including obesity, metabolic syndrome, chronic kidney or liver disease, nephrotic syndrome, diabetes, or hypothyroidism, must be evaluated and addressed. Triglyceride levels of more than 1000 mg/dL (11.3 mmol/L) confer increased risk for pancreatitis and require treatment with prescription-strength omega-3 fatty acids or a fibrate (see Fig. 6.4 ).



References

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jan 3, 2021 | Posted by in CARDIOLOGY | Comments Off on Lipid-Modifying Drugs

Full access? Get Clinical Tree

Get Clinical Tree app for offline access