Chapter 31 - Dyslipidemia
Chapter 31 - Dyslipidemia
KEY CONCEPTS
KEY CONCEPTS
Lipid abnormalities increase the risk for coronary heart disease (CHD) and cerebrovascular morbidity and mortality.
Genetic abnormalities and environmental factors are involved in the development of dyslipidemia.
If therapeutic lifestyle changes are insufficient, lipidlowering agents should be chosen based on which lipid is at an undesirable level and
the degree to which it is expected to increase the risk of atherosclerotic cardiovascular disease (ASCVD).
Statins are the drugs of choice for dyslipidemia because of potency and costeffectiveness.
If statin monotherapy is insufficient, patients may be treated with evidencebased combination therapy but should be monitored closely for
drug–drug interactions.
Reducing total cholesterol and LDLC reduces CHD and total mortality.
Several novel medications including antisense oligonucleotide inhibitors of apoB, microsomal triglyceride transport protein inhibitors,
adenosine triphosphatecitrate lyase (ACL) inhibitors, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors can be used as add
on therapy or in lieu of statin therapy in select highrisk populations.
PRECLASS ACTIVITY
Watch these YouTube videos to learn about cholesterol basics as well as the physiology of lipoprotein cholesterol and metabolism:
PRECLASS ACTIVITY
Watch these YouTube videos to learn about cholesterol basics as well as the physiology of lipoprotein cholesterol and metabolism:
INTRODUCTION
Cholesterol, triglycerides, and phospholipids are the major lipids that combine with proteins to be transported as complexes of lipid and proteins
known as lipoproteins. Lipids, such as cholesterol and triglycerides, are insoluble in plasma, which is why the lipoproteins are required for
transportation (Fig. 311).1,2
FIGURE 311
Intestinal cholesterol absorption and transportation. Cholesterol from food and bile enters the gut lumen and is emulsified by bile acids into micelles.
Micelles bind to intestinal enterocytes and cholesterol, and other sterols are transported from the micelles to the enterocytes by sterol transporters.
Triglycerides (TG) synthesized by absorbed fatty acids (FA) are incorporated into chylomicrons. Chylomicrons are released into lymphatic circulation
and converted to chylomicron remnants (by losing triglyceride), and are then taken up by hepatic LDLreceptor–related protein. (Apo, apolipoprotein;
ABC, ATPbinding cassette; CE, cholesterol ester; FA, fatty acid; NPC1L1, NiemannPick C1Like1 protein; TG, triglyceride.) (Reproduced, with
permission, from ChisholmBurns MA, Schwinghammer TL, Malone PM, Kolesar JM, Bookstaver PB, Lee KC, eds. Pharmacotherapy Principles &
Practice. 5th ed. New York: McGrawHill; 2019.)
There are three major classes of lipoproteins in the serum. These include lowdensity lipoproteins (LDL), highdensity lipoproteins (HDL), and very
lowdensity lipoproteins (VLDL). VLDL is the primary carrier of triglycerides (TG) in the circulation. Intermediatedensity lipoprotein (IDL) is between
VLDL and LDL and is included in LDLC measurement (Fig. 312).3
FIGURE 312
Lipoprotein structure, which contains variable amounts of core cholesterol esters and triglycerides and have varying numbers and types of surface
Downloaded 2025213 10:53 P Your IP is
apolipoproteins.
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 2 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
lowdensity lipoproteins (VLDL). VLDL is the primary carrier of triglycerides (TG) in the circulation. Intermediatedensity lipoprotein (IDL) is between
California Northstate University
VLDL and LDL and is included in LDLC measurement (Fig. 312).3
Access Provided by:
FIGURE 312
Lipoprotein structure, which contains variable amounts of core cholesterol esters and triglycerides and have varying numbers and types of surface
apolipoproteins.
Lipid abnormalities increase the risk of coronary, cerebrovascular, and peripheral vascular arterial disease collectively known as
atherosclerotic cardiovascular disease (ASCVD). The ASCVDrisk assessment evaluates a 10year atherosclerotic cardiovascular disease incident.
Developing a first ASCVD event is defined as nonfatal myocardial infarction or coronary heart disease (CHD) death, or fatal or nonfatal stroke, over a
10year period. Premature coronary atherosclerosis is the most common and significant consequence of dyslipidemia. The 2014 guidelines from the
American Heart Association (AHA) and American College of Cardiology (ACC) state there is not sufficient evidence to recommend treating to specific
lipid targets but suggests four statin benefit patient populations instead. Lowdensity lipoprotein cholesterol (LDLC) is the primary target of lipid
lowering therapy. Methods for risk assessment were also updated, which increases the number of patients that would qualify for therapy. Primary and
secondary CHD prevention measures are provided as well.4–7
There are several subtypes of dyslipidemias, including hypertriglyceridemia, low HDL cholesterol (HDLC), and diabetic dyslipidemia.
Hypertriglyceridemia can lead to pancreatitis when very high TG levels (>500 mg/dL [5.65 mmol/L]) are seen. High serum triglycerides should primarily
be treated by achieving desirable body weight, consumption of low saturated fat and cholesterol diet, regular exercise, smoking cessation, and
restriction of alcohol.3,4 In patients with borderlinehigh TG but with accompanying risk factors of established congenital heart disease, family history
of premature CHD, concomitant LDLC elevation or low HDLC, and genetic forms of hypertriglyceridemia associated with CHD, lipidlowering therapy
should be considered.8–10
Low HDLC is another dyslipidemia subtype that can occur due to insulin resistance, physical inactivity, diabetes, cigarette smoking, high carbohydrate
intake, and some medications. In patients with low HDLC, the primary target remains LDLC, but an emphasis on weight loss, increased physical
activity, and smoking cessation are recommended. No randomized controlled trials (RCT) have shown a reduction in ASCVD risk by raising HDLC
levels.3,11 Hypertriglyceridemia, low HDLC, and minimally elevated LDLC characterize diabetic dyslipidemia. Because the primary target is LDLC in
diabetic dyslipidemia, statins are considered the drugs of choice.3,4,12,13
EPIDEMIOLOGY
Total cholesterol and LDLC increase throughout life in both men and women. According to AHA estimates, approximately 45% of American adults
aged 20 or older have total cholesterol levels exceeding 200 mg/dL (5.17 mmol/L).14 The prevalence of elevated total cholesterol in adults has remained
the same over the past decade but has improved in children. Westernized societal diets high in cholesterol are a strong contributor to the increase in
Downloaded 2025213
total and LDLC 10:53
cholesterol. P Your
In 2011, CHDIPcaused
is one in every seven deaths in the United States. More than half of individuals at borderlinehigh risk
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 3 / 37
remain unaware that they have dyslipidemia, and fewer than half of the highest risk persons (those with symptomatic CHD) are receiving lipidlowering
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
treatment. About onethird of treated patients are achieving their LDLC goal; fewer than 20% of CHD patients are at their LDLC goal. Estimates from
the National Cholesterol Education Program (NCEP) state that only 26% of patients have an optimal LDLC (<100 mg/dL [2.59 mmol/L]), and that large
EPIDEMIOLOGY California Northstate University
Access Provided by:
Total cholesterol and LDLC increase throughout life in both men and women. According to AHA estimates, approximately 45% of American adults
aged 20 or older have total cholesterol levels exceeding 200 mg/dL (5.17 mmol/L).14 The prevalence of elevated total cholesterol in adults has remained
the same over the past decade but has improved in children. Westernized societal diets high in cholesterol are a strong contributor to the increase in
total and LDLC cholesterol. In 2011, CHD caused one in every seven deaths in the United States. More than half of individuals at borderlinehigh risk
remain unaware that they have dyslipidemia, and fewer than half of the highest risk persons (those with symptomatic CHD) are receiving lipidlowering
treatment. About onethird of treated patients are achieving their LDLC goal; fewer than 20% of CHD patients are at their LDLC goal. Estimates from
the National Cholesterol Education Program (NCEP) state that only 26% of patients have an optimal LDLC (<100 mg/dL [2.59 mmol/L]), and that large
numbers of patients are either untreated or undertreated. Patients at highest risk are less likely to be treated to desirable LDLC values.15–17 When
patients who are at risk but who have not yet experienced initial cardiovascular (eg, myocardial infarction [MI]) or cerebrovascular (eg, ischemic stroke)
events are treated, it is termed primary prevention. Treatment for those with manifest ASCVD is termed secondary prevention.17 Studies, such as the
Framingham Heart study, show that risk for developing cardiovascular disease is related to the degree of LDLC elevation in a continuous fashion.3
Hypercholesterolemia, cigarette smoking, hypertension, diabetes, and low HDLC levels are all additive risk factors for CHD. Risk of MI increases five to
seven times with any preexisting CHD or previous MI compared to patients with no history of these. Patients with a history of CHD or MI should be
screened, identified, and treated for dyslipidemias. Fifty percent of all MIs and 70% of all deaths due to CHD occur in patients with known CHD.
ETIOLOGY
Genetitc abnormalities and environmental factors are involved in the development of dyslipidemia. The underlying causes of dyslipidemias can be
categorized into two types: primary or secondary. Genetic factors that increase lipid levels can be inherited and cause primary or familial dyslipidemia.
By contrast, lifestyles, diseases, medications, and diet can all lead to abnormal lipid levels and cause secondary or “acquired” dyslipidemia.
Primary or familial dyslipidemias account for a large number of cases of increased total cholesterol, LDLC, TGs, or decreased HDLC. There are certain
familial or genetic defects that can contribute. Genetic disorders can cause an increase or decrease in different lipoproteins. Primary dyslipidemias
result in an increased risk of premature ASCVD due to significant elevations in cholesterol levels. There are different types of familial dyslipidemias,
including hypercholesterolemia, hypertriglyceridemia, combined hyperlipidemia, and disorders of HDLC metabolism and an excess of lipoproteins.
Two other primary disorders include homozygous familial hypercholesterolemia (HoFH) and heterozygous familial hypercholesterolemia (HeFH).
HeFH is more common with one case per 250 people versus HoFH with one case per one million people. In familial hypertriglyceridemia, TGs are
elevated in the range of 200 to 500 mg/dL (2.26 to 5.65 mmol/L), but at times can be greater than 1,000 mg/dL (11.3 mmol/L). Patients presenting with
TG concentrations greater than or equal to 500 mg/dL (5.65 mmol/L) can have eruptive xanthomas and/or acute pancreatitis. Heterozygous gene
dysfunction usually causes elevations in LDLC between 250 and 450 mg/dL (6.47 to 11.64 mmol/L), and homozygous patients may present with LDLC
concentrations above 500 mg/dL (12.93 mmol/L). Tendon xanthomas are thick cholesterol deposits. Xanthelasmas and arcus cornea can also occur,
and these are cholesterol deposits in the eyelids and around the corneal rim.18–21
Secondary or acquired dyslipidemias can accompany genetic disorders or cause lipid imbalances. “The 4D classification” of secondary causes of
dyslipidemia include, diet, drugs, disorder, and diseases.19 Regarding diet, an increase in cholesterol can be caused from excessive alcohol use,
anorexia, weight gain, excessive carbohydrate intake, and high saturated fat intake. Certain medications can also contribute. For example, some
medications that can increase both LDLC and TGs include atypical antipsychotics, diuretics, beta blockers, glucocorticoids, oral estrogen and
progestin, tacrolimus, and cyclosporine.
Certain metabolism disorders can contribute to cholesterol imbalances. Nephrotic syndrome, renal failure, biliary obstruction, hypothyroidism, and
pregnancy can all potentially contribute. Comorbid conditions or diseases such as hypothyroidism, pregnancy, obesity, polycystic ovarian syndrome
(PCOS), uncontrolled diabetes, liver disease, chronic kidney disease can also play a role.
Although we classify the lipid disorders into primary and secondary dyslipidemias based on etiologies, most dyslipidemias are a result of a
combination of both.4,19
PATHOPHYSIOLOGY
Lipoproteins and Cholesterol
Downloaded 2025213 10:53 P Your Synthesis
IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 4 / 37
©2025 McGraw
There are Hill.of
four types Alllipoproteins:
Rights Reserved. Terms of
chylomicrons, UseLDL,
VLDL, • Privacy Policy
and HDL. • Notice
These • Accessibility
lipoproteins vary in content of lipid and protein. The ratio of protein
and lipid in these lipoproteins contribute to the function of each. Chylomicrons contain the most lipid and very little protein. HDL contains the most
protein and a small amount of lipids. The small amount of lipid, and in turn cholesterol, in HDL lipoproteins gives HDL the role of picking up extra
Although we classify the lipid disorders into primary and secondary dyslipidemias based on etiologies, most dyslipidemias are a result of a
California Northstate University
combination of both.4,19
Access Provided by:
PATHOPHYSIOLOGY
Lipoproteins and Cholesterol Synthesis
There are four types of lipoproteins: chylomicrons, VLDL, LDL, and HDL. These lipoproteins vary in content of lipid and protein. The ratio of protein
and lipid in these lipoproteins contribute to the function of each. Chylomicrons contain the most lipid and very little protein. HDL contains the most
protein and a small amount of lipids. The small amount of lipid, and in turn cholesterol, in HDL lipoproteins gives HDL the role of picking up extra
cholesterol from the tissue. LDL is not necessarily “bad” cholesterol, but in excess, this generates the problem. We need cholesterol for the
transportation of fats that are absorbed in our diet and delivered to our tissues. Chylomicrons are not normally in plasma during periods of fasting. In
the small intestine, fats are digested and emulsified into micelles. Cholesterol is also absorbed. Fatty acids, cholesterol, and proteins or apoproteins
are packaged and form the chylomicrons. The chylomicrons circulate around the body and deliver lipids and TGs to tissues in need. The remaining
chylomicrons are transported to the liver and bind to LDL receptors. Glucose that has also been absorbed from our diet is delivered to the liver. In the
liver, glucose is converted to pyruvate and then to acetylCoA. AcetylCoA is eventually converted to cholesterol through 3hydroxy3methylglutaryl
coenzyme A (HMGCoA) reductase. HMGCoA reductase is a target for statins, thus stopping cholesterol synthesis. Glycolysis also synthesizes glycerol.
Combining glycerol and one fatty acid forms monoacylglycerol, and two more fatty acids form triglycerides. Triglycerides and apoproteins are packed
through the Golgi apparatus and form lipoproteins. Lipoproteins contain proteins, apoproteins, TGs, phospholipids, and cholesterol. The liver does
not make all lipoproteins. In fact, only empty HDL and VLDL are made in the liver. VLDL has more TGs and lipids than HDL. VLDL transports these lipids
and TGs to tissues in need of energy or storage. Adipose tissue stores fat, and many tissues use fatty acids for energy. VLDL is transported across
lipase, which changes the VLDL to IDL or intermediatedensity lipoproteins. IDL can then be converted to LDL. LDL mainly transports cholesterol to
body tissues, which is why LDL contains the most cholesterol. Tissues need cholesterol to make hormones and maintain cell membrane integrity. Once
LDL gives these tissues cholesterol, it returns to the liver. LDL binds to LDL receptors and is then either recycled to make more lipoproteins or excreted
into the bile. Any excess cholesterol is excreted into the bile, so the body maintains cholesterol balance. HDL or empty HDL plays a role in picking up
any or excess cholesterol and returning it to the liver. The full HDL containing the picked up cholesterol binds to scavenger receptors. They are then
either recycled or excreted depending on how much cholesterol is needed.22
Cholesterol is waterinsoluble so it cannot circulate through blood without help. Lipoproteins are large carrier proteins to help with transport because
they are watersoluble. This allows major lipids to be circulated through the blood. Lipoproteins vary in characteristics depending on the amount of
cholesterol, TG, and apolipoproteins. All lipoproteins also have something called apolipoproteins on its surface. Apolipoproteins are necessary for the
assembly and secretion of lipoproteins. They are also major structural components of lipoproteins that have ligands for binding to receptors or cell
surfaces. These are the cofactors for the activation of enzymes. Apolipoproteins have various functions that transport lipids from sites of absorption to
sites where they are used. Apolipoprotein B containing lipoproteins, known as nonHDL, make up the lipiddelivery pathway. Apolipoprotein A1 or
HDL participates in reverse cholesterol transport. ApoB containing lipoproteins arise from two sources, one being intestinal ApoB48 and the other
hepatic ApoB100 lineage. ApoB containing lipoproteins are secreted from the intestine or liver into the plasma. Apolipoproteins E, CII, and CIII are
secreted with them. These also may be acquired from HDL. Apolipoprotein remodeling begins, and ApoCII activates lipoprotein lipase (LPL), which
hydrolyzes the lipoprotein core TGs into free fatty acids. The fatty acids exit and the lipoproteins become smaller and smaller. Remodeling of the ApoB
100 hepatic lipoprotein, another step by hepatic lipase (HL) is needed to convert IDL to LDL. Most ApoB remnants are recycled to the liver by the LDL
receptor–related protein (LRP). They can have other metabolic requirements, too. Excess ApoB particles can invade arterial walls and become oxidized.
Once oxidized, they are taken up by macrophage scavenger receptors creating foam cells that lead to atheroma.
ApoA1 or HDL pathways are believed to protect our bodies from atherogenesis. HDL has two major protective roles in preventing atherogenesis.
Reverse cholesterol transport is the transfer of excess cholesterol from peripheral tissues by HDL. ApoA1 is secreted from the liver or intestine and is
transported to the cells to remove excess cholesterol. HDL has several cholesterol removing mechanisms. Upregulation of the ATPbinding cassette
transporter or ABCA1 transporter is triggered by excess cholesterol in the macrophages. ABCA1 harvests free cholesterol and delivers it to the cell
membrane. The free cholesterol is esterified by lecithincholesterol acyltransferase or LCAT. The cholesterol ester moves to the core of the lipoprotein
forming the mature HDL3. Further removal of cholesterol by HDL3 occurs through scavenger receptor class B type 1, or SRB1 receptors and is acted on
by LCAT, which expands to HDL2. ABCA1 and SRB1 are key for cholesterol efflux. HDL2 cholesterol is transferred to ApoB containing lipoproteins. HDL
now has one of three options: HDL triglycerides may be hydrolyzed by HL back into HDL3; HDL2 can return to the liver and through SRB1 converted
back to HDL3; or HDL2 may be catabolized by the liver. All of these systems work together to maintain cholesterol homeostasis (Figs. 313 and 314).23
FIGURE 313
FIGURE 313
Biosynthetic pathway for cholesterol. The ratelimiting enzyme in this pathway is 3hydroxy3methylglutarylcoenzyme A reductase (HMGCoA
reductase). (CETP, cholesterol ester transfer protein; HDL, highdensity lipoprotein; IDL, intermediatedensity lipoprotein; LDL, lowdensity
lipoprotein; LPL, lipoprotein lipase; VLDL, verylowdensity lipoprotein.) (A) Exogenous pathway; (B) Endogenous pathway; (C) Reverse cholesterol
transport. (Adapted from Breslow JL. Genetic basis of lipoprotein disorders. J Clin Invest 1989;84:373.)
FIGURE 314
Endogenous lipoprotein metabolism. In the liver, the cholesterol and triglycerides are packed into VLDL particles and sent into the blood. They are
then converted into IDL, which can be cleared by hepatic IDL receptor or metabolized into LDL. LDL can be cleared by LDL receptors or it can enter the
arterial walls and contribute to the development of atherosclerotic plaques and cardiovascular disease. HDLC is responsible for transporting
cholesterol and phospholipids from the arterial cell wall or other extrahepatic tissues back to the liver. Cholesterol that is returned back to the liver can
then be excreted into the bile. This process is known as reverse cholesterol transport. (Apo, apolipoprotein; CE, cholesteryl ester; FA, fatty acid; HMG
CoA, βHydroxy βmethylglutarylCoA; HL, hepatic lipase; LDL, lowdensity lipoprotein; LPL, lipoprotein lipase; VLDL, very lowdensity lipoprotein)
(Reprinted, with permission, from Marrs JC. Pathophysiology of Atherosclerotic Cardiovascular Disease. In: Wiggins BS, Saseen JJ, eds. Pharmacist’s
Guide to Lipid Management. 2nd ed. Lenexa, KS: American College of Clinical Pharmacy, 2014:2.)
Downloaded 2025213 10:53 P Your IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 6 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
cholesterol and phospholipids from the arterial cell wall or other extrahepatic tissues back to the liver. Cholesterol that is returned back to the liver can
California Northstate University
then be excreted into the bile. This process is known as reverse cholesterol transport. (Apo, apolipoprotein; CE, cholesteryl ester; FA, fatty acid; HMG
Access Provided by:
CoA, βHydroxy βmethylglutarylCoA; HL, hepatic lipase; LDL, lowdensity lipoprotein; LPL, lipoprotein lipase; VLDL, very lowdensity lipoprotein)
(Reprinted, with permission, from Marrs JC. Pathophysiology of Atherosclerotic Cardiovascular Disease. In: Wiggins BS, Saseen JJ, eds. Pharmacist’s
Guide to Lipid Management. 2nd ed. Lenexa, KS: American College of Clinical Pharmacy, 2014:2.)
Familial Hypercholesterolemia
Familial hypercholesterolemia is characterized by (a) a selective elevation in the plasma level of LDL; (b) deposition of LDLderived cholesterol in
tendons (xanthomas) and arteries (atheromas); and (c) inheritance as an autosomal dominant trait with homozygotes more severely affected than
heterozygotes. The primary defect in familial hypercholesterolemia is the inability to bind LDL to the LDL receptor (LDLR) or, rarely, a defect of
internalizing the LDLR complex into the cell after normal binding. This leads to a lack of LDL degradation by cells and unregulated biosynthesis of
cholesterol, with total cholesterol and LDLC being inversely proportional to the deficit in LDL receptors. Homozygotes (prevalence 1 in 1,000,000) have
severe hypercholesterolemia (6501,000 mg/dL [16.825.9 mmol/L]), with the early appearance of cutaneous xanthomas and fatal CHD generally before
the age of 20 and have essentially no functional LDL receptors. Heterozygotes have only about onehalf of the normal number of LDL receptors, total
cholesterol levels in the range of 300 to 600 mg/dL (7.7615.52 mmol/L), and cardiovascular events beginning in the third and fourth decades of life.
Secondary causes of dyslipidemia exist and that several drugs and conditions may contribute to abnormal lipid levels (Table 311). These secondary
forms of dyslipidemia should be managed by addressing the underlying abnormality, including the modification of drug therapy when appropriate.
TABLE 311
Secondary Causes of Lipoprotein Abnormalities
Hypercholesterolemia Hypothyroidism
Obstructive liver disease
Nephrotic syndrome
Anorexia nervosa
Acute Intermittent Porphyria
Drugs: progestins, thiazide diuretics, glucocorticoids, betablockers, isotretinoin, protease inhibitors, cyclosporine,
mirtazapine, sirolimus
Hypertriglyceridemia Obesity
Diabetes mellitus
Lipodystrophy
Glycogen storage disease
Ileal bypass surgery
Downloaded 2025213 10:53 P Your IP is
Sepsis
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 7 / 37
Pregnancy
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
Acute Hepatitis
Systemic lupus erythematosus
the age of 20 and have essentially no functional LDL receptors. Heterozygotes have only about onehalf of the normal number of LDL receptors, total
cholesterol levels in the range of 300 to 600 mg/dL (7.7615.52 mmol/L), and cardiovascular events beginning in the third and fourth
California decades University
Northstate of life.
Access Provided by:
Secondary causes of dyslipidemia exist and that several drugs and conditions may contribute to abnormal lipid levels (Table 311). These secondary
forms of dyslipidemia should be managed by addressing the underlying abnormality, including the modification of drug therapy when appropriate.
TABLE 311
Secondary Causes of Lipoprotein Abnormalities
Hypercholesterolemia Hypothyroidism
Obstructive liver disease
Nephrotic syndrome
Anorexia nervosa
Acute Intermittent Porphyria
Drugs: progestins, thiazide diuretics, glucocorticoids, betablockers, isotretinoin, protease inhibitors, cyclosporine,
mirtazapine, sirolimus
Hypertriglyceridemia Obesity
Diabetes mellitus
Lipodystrophy
Glycogen storage disease
Ileal bypass surgery
Sepsis
Pregnancy
Acute Hepatitis
Systemic lupus erythematosus
Monoclonal gammopathy: multiple myeloma, lymphoma
Drugs: Alcohol, estrogens, isotretinoin, betablockers, glucocorticoids, bileacid resins, thiazides, asparaginase,
interferons, azole antifungals, mirtazapine, anabolic steroids, sirolimus, bexarotene
Hypercholesterolemia Malnutrition
Malabsorption
Myeloproliferative diseases
Chronic infectious diseases: human immunodeficiency virus (HIV), tuberculosis
Monoclonal gammopathy
Chronic Liver Disease
The “responsetoinjury” hypothesis states that risk factors such as oxidized LDL, mechanical injury to the endothelium, excessive homocysteine,
immunologic attacks, or infectioninduced changes in endothelial and intimal function lead to endothelial dysfunction and a series of cellular
interactions that culminate in atherosclerosis. Creactive protein (CRP) is an acutephase reactant and a marker for inflammation. Measuring one’s CRP
levels by means of a highsensitivity CRP may be useful in identifying patients at risk for developing CAD. Lipid abnormalities increase the risk for CHD
and cerebrovascular morbidity and mortality. The eventual outcomes of this atherogenic cascade are clinical events such as angina, MI, arrhythmias,
stroke, peripheral arterial disease, abdominal aortic aneurysm, and sudden death. Atherosclerotic lesions are thought to arise from transport and
retention of plasma LDLC through the endothelial cell layer into the extracellular matrix of the subendothelial space. Once in the artery wall, LDLC is
chemically modified through oxidation and nonenzymatic glycation. Mildly oxidized LDLC then recruits monocytes which transform into macrophages
in the artery wall. Macrophages accelerate LDLC oxidation and apolipoprotein B accumulation and alter the receptormediated uptake of LDLC into
Downloaded 2025213 10:53 P Your IP is
the artery wall from the usual LDLR to a “scavenger receptor” not regulated by cell content of cholesterol. Oxidized LDLC increases plasminogen
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 8 / 37
inhibitor levels (promotion
©2025 McGraw of coagulation),
Hill. All Rights Reserved. induces
Terms ofthe
Useexpression
• Privacyof endothelin
Policy (vasoconstrictive
• Notice • Accessibility substance), inhibits the expression of nitric oxide (a
vasodilator and platelet inhibitor), and is toxic to macrophages if highly oxidized. As oxidation of biologically active lipids proceeds, other lipids
breakdown products of fatty acids and oxysterol are formed, which continue the reaction within the tissue. These events lead to a massive
levels by means of a highsensitivity CRP may be useful in identifying patients at risk for developing CAD. Lipid abnormalities increase the risk for CHD
and cerebrovascular morbidity and mortality. The eventual outcomes of this atherogenic cascade are clinical events such California Northstate
as angina, University
MI, arrhythmias,
stroke, peripheral arterial disease, abdominal aortic aneurysm, and sudden death. Atherosclerotic lesions are thought to arise
Access fromby:transport and
Provided
retention of plasma LDLC through the endothelial cell layer into the extracellular matrix of the subendothelial space. Once in the artery wall, LDLC is
chemically modified through oxidation and nonenzymatic glycation. Mildly oxidized LDLC then recruits monocytes which transform into macrophages
in the artery wall. Macrophages accelerate LDLC oxidation and apolipoprotein B accumulation and alter the receptormediated uptake of LDLC into
the artery wall from the usual LDLR to a “scavenger receptor” not regulated by cell content of cholesterol. Oxidized LDLC increases plasminogen
inhibitor levels (promotion of coagulation), induces the expression of endothelin (vasoconstrictive substance), inhibits the expression of nitric oxide (a
vasodilator and platelet inhibitor), and is toxic to macrophages if highly oxidized. As oxidation of biologically active lipids proceeds, other lipids
breakdown products of fatty acids and oxysterol are formed, which continue the reaction within the tissue. These events lead to a massive
accumulation of cholesterol. The cholesterolladen macrophages become foam cells; foam cells are the earliest recognized cells of the arterial fatty
streak. Oxidized LDLC provokes an inflammatory response, which is mediated by a number of chemoattractants and cytokines. The process of aging
may lead to lipoproteins that are more susceptible to oxidation and have longer resident time in the vascular compartment. Repeated injury and repair
within an atherosclerotic plaque eventually leads to fibrous cap protecting the underlying core of lipids, collagen, calcium, and inflammatory cells such
as Tlymphocytes. Maintenance of the fibrous plaque is critical to prevent plaque rupture and subsequent coronary thrombosis.24 An imbalance
between plaque synthesis and degradation may lead to a weakened or vulnerable plaque prone to rupture. The fibrous cap may become weakened
through decreased synthesis of the extracellular matrix or increased degradation of the matrix (Fig. 315).
FIGURE 315
Atherogenesis is initiated by the migration of LDL and remnant lipoprotein particles into the vessel walls. These particles undergo oxidation and are
taken up by macrophages in an unregulated fashion, which induces endothelial cell dysfunction. This, in turn, reduces the ability of the endothelium to
dilate the artery and cause a prothrombotic state. Unregulated uptake of cholesterol by macrophages leads to foam cell formation, and thus the
development of atherosclerotic plaques. Macrophages eventually produce and secrete matric metalloproteinases, which degrade the collagen matrix
of the plaques and cause them to be unstable. This can potentially lead to a myocardial infarction. This is a progressive process. (IDL, intermediate
density lipoprotein; LDL, lowdensity lipoprotein; MMP, matrix metalloproteinase; NO, nitric oxide; SRB1, scavenger receptor class B type 1)
(Reproduced, with permission, from ChisholmBurns MA, Schwinghammer TL, Malone PM, Kolesar JM, Bookstaver PB, Lee KC, eds. Pharmacotherapy
Principles & Practice. 5th ed. New York: McGrawHill; 2019.)
CLINICAL PRESENTATION
General
Most patients are asymptomatic for years before they develop ASCVD; the initial presentation may be sudden death due to a CHD event.
Many patients with dyslipidemia also present with one or more of the following abnormalities*:
Downloaded 2025213 10:53 P Your IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche
Abdominal obesity
Page 9 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
Atherogenic dyslipidemia
CLINICAL PRESENTATION DYSLIPIDEMIAS
California Northstate University
General Access Provided by:
Most patients are asymptomatic for years before they develop ASCVD; the initial presentation may be sudden death due to a CHD event.
Many patients with dyslipidemia also present with one or more of the following abnormalities*:
Abdominal obesity
Atherogenic dyslipidemia
*Patients with three or more of these abnormalities are considered to have the metabolic syndrome
Symptoms of ASCVD
Chest pain
Palpitations
Sweating
Anxiety
Shortness of breath
Loss of consciousness
Abdominal pain
Signs
Abdominal pain
Pancreatitis
Eruptive xanthomas
Peripheral polyneuropathy
Laboratory Tests
Decreased HDLC
Screenings for manifestations of vascular disease, including carotid ultrasound, coronary calcium score, anklebrachial index, and heart
catheterization.
TREATMENT
Desired Outcomes
Desired levels2025213
Downloaded of TC, LDLC, HDLC,
10:53 and TG
P Your are provided in Table 312 for adults and Table 313 for children. While abnormalities in these surrogate
IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel
markers may impart an increased risk for ASCVD M. Riche
events, Page 10 / 37
the goal of treatment is to not merely correct lab abnormalities but prevent the development
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
and progression of ASCVD. Thus, the desired outcome is to prevent ASCVDrelated morbidity and mortality, including revascularization procedures,
MI, and ischemic stroke. Initiation of lipidlowering therapies primarily involves the use of those agents shown in RCT to reduce ASCVD risk.20,25
California Northstate University
TREATMENT Access Provided by:
Desired Outcomes
Desired levels of TC, LDLC, HDLC, and TG are provided in Table 312 for adults and Table 313 for children. While abnormalities in these surrogate
markers may impart an increased risk for ASCVD events, the goal of treatment is to not merely correct lab abnormalities but prevent the development
and progression of ASCVD. Thus, the desired outcome is to prevent ASCVDrelated morbidity and mortality, including revascularization procedures,
MI, and ischemic stroke. Initiation of lipidlowering therapies primarily involves the use of those agents shown in RCT to reduce ASCVD risk.20,25
TABLE 312
Classification of Total, LDL, HDLCholesterol, and Triglycerides in Adults
Triglycerides Normal
<150 mg/dL (1.70 mmol/L) Borderline high
150199 mg/dL (1.702.25 mmol/L) High
200499 mg/dL (2.265.64 mmol/L) Very high
≥500 mg/dL (5.65 mmol/L)
Table 313
Classification of Total, LDL, HDLCholesterol and Triglycerides in Children
Triglycerides Acceptable
150199 mg/dL (1.702.25 mmol/L) High
200499 mg/dL (2.265.64 mmol/L) Very high California Northstate University
≥500 mg/dL (5.65 mmol/L) Access Provided by:
Table 313
Classification of Total, LDL, HDLCholesterol and Triglycerides in Children
Triglycerides Acceptable
09 years Borderline
<75 mg/dL (0.8 mmol/L) High
7599 mg/dL (0.81.1 mmol/L) Acceptable
≥100 mg/dL (1.1 mmol/L) Borderline
1019 years High
<90 mg/dL (1 mmol/L)
90129 mg/dL (11.5 mmol/L)
≥130 mg/dL (1.5 mmol/L)
Data from Daniels SR, Benuck I, Christakis DA, et al. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents:
Full report, 2011. National Heart Lung and Blood Institute.
General Approach
A comprehensive approach to treating dyslipidemia and all modifiable major risk ASCVD factors is required to significantly reduce the risk of first
and recurrent ASCVD events. Therapeutic lifestyle change is the firstline therapy for any lipoprotein disorder. A healthy lifestyle should be
implemented in all patients with the general components including a reduction in the percent of calories from saturated and trans fats, increased
intake of soluble fiber, weight reduction if overweight or obese, increased physical activity, and avoiding or quitting tobacco use.25 Additionally,
patients with a diagnosis of hypertension should achieve optimal blood pressure control based on the 2017 ACC/AHA Guidelines for control of
hypertension (see Chapter 30, “Hypertension”).26 Persons with diabetes mellitus, especially those with established ASCVD, should receive glucose
lowering therapies that have been shown to reduce ASCVD risk (see Chapter 91, “Diabetes Mellitus”).27
If therapeutic lifestyle changes are insufficient, lipidlowering agents should be chosen based on which lipid is at an undesirable level and the
degree to which it is expected to increase the risk of ASCVD. The decision to initiate lipidlowering therapy in the management of dyslipidemia should
be based on an individual’s ASCVD risk and not merely plasma levels of atherogenic lipoproteins (such as LDLC) alone.28 Patients with established
Downloaded 2025213
ASCVD are at highest risk10:53 P Your
and most likelyIPtoisbenefit from select lipidlowering therapies (such as statins). Risk assessment in patients without
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 12 / 37
established ASCVD is more of an art that requires careful consideration of traditional (eg, age, hypertension) and nontraditional (eg, autoimmune
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
diseases, socioeconomic status) risk factors, the risks of lipidlowering therapy, and patient preference. For patients between 40 and 79 years of age
and no history of ASCVD, the ASCVD Risk Estimator Plus (available at: www.tools.acc.org/ascvdriskestimatorplus) should be used to facilitate a
hypertension (see Chapter 30, “Hypertension”).26 Persons with diabetes mellitus, especially those with established ASCVD, should receive glucose
lowering therapies that have been shown to reduce ASCVD risk (see Chapter 91, “Diabetes Mellitus”).27 California Northstate University
Access Provided by:
If therapeutic lifestyle changes are insufficient, lipidlowering agents should be chosen based on which lipid is at an undesirable level and the
degree to which it is expected to increase the risk of ASCVD. The decision to initiate lipidlowering therapy in the management of dyslipidemia should
be based on an individual’s ASCVD risk and not merely plasma levels of atherogenic lipoproteins (such as LDLC) alone.28 Patients with established
ASCVD are at highest risk and most likely to benefit from select lipidlowering therapies (such as statins). Risk assessment in patients without
established ASCVD is more of an art that requires careful consideration of traditional (eg, age, hypertension) and nontraditional (eg, autoimmune
diseases, socioeconomic status) risk factors, the risks of lipidlowering therapy, and patient preference. For patients between 40 and 79 years of age
and no history of ASCVD, the ASCVD Risk Estimator Plus (available at: www.tools.acc.org/ascvdriskestimatorplus) should be used to facilitate a
clinicianpatient discussion regarding the benefit and risks of lipidlowering therapy, especially in patients whose 10year risk is 7.5% or greater (see
Table 314). The Risk Estimator is comprised of the patient’s age, gender, race, TC, HDLC, blood pressure, diabetes status, smoking status, and use of
antihypertensives, statins, and aspirin. Importantly, the Risk Estimator is based on data from large population studies of mostly AfricanAmerican and
nonHispanic white men and women. The Risk Estimator can be used for other ethnic groups if they are designated as nonHispanic white; however,
the Risk Estimator will underestimate the risk of American Indians and Asian Americans of South Asian ancestry, while overestimating the risk of Asian
Americans of East Asian ancestry and some Hispanics (eg, Mexican Americans).28 An estimated lifetime risk for ASCVD can also be performed for
patients between age 20 and 39, yet these results should only be used to justify the need for lifestyle change and not the initiation of lipidlowering
therapy. Additional tools for ASCVDrisk assessment include highsensitivity Creactive protein (hsCRP), apolipoprotein B, and lipoprotein(a) [Lp(a)]
levels that may be obtained to inform decision making in lowintermediate risk patients or those with recurrent ASCVD events despite appropriate
lipidlowering therapy.28
TABLE 314
Key Recommendations to Reduce the Risk of Atherosclerotic Cardiovascular Disease (ASCVD) in Adults
1. Clinical ASCVDi
a. Very highriskii
Highintensity or maximally tolerated statin therapy (Class I)
b. Not at very highrisk
Age <75 years: Highintensity statin therapy (Class I)
If highintensity not tolerated, use moderateintensity statin therapy (Class I)
Age >75 years: Initiation of moderate or highintensity statin is reasonable (Class IIa)
3. Primary prevention—Diabetes 4075 years and LDLC 70189 mg/dL (1.81 4.89 mmol/L)
a. Moderateintensity statin therapy regardless of 10year ASCVD risk (Class I)
b. Highintensity statin therapy is reasonable in patients with diabetes and multiple ASCVDrisk factors (Class IIa)
c. It is reasonable to continue statin therapy in adults ≥75 years with diabetes previously taking statin therapy (Class IIa)
d. It is reasonable to initiate statin therapy in adults ≥75 years with diabetes after a discussion of risks and benefits (Class IIb)
e. It is reasonable to initiate statin therapy in adults 2030 years with diabetes if any of the following apply: (Class IIb)
Long duration of diabetes (≥10 years for type 2 diabetes; ≥20 years for type 1 diabetes)
Albuminuria (≥30 mg/g albumin/mg creatinine [3.4 mg/mmol creatinine])
Retinopathy
Neuropathy
Anklebrachial index <0.9
4. Primary prevention—no diabetes, 4075 years and LDLC 70189 mg/dL (1.81 4.89 mmol/L)
1. Emphasize hearthealthy lifestyle and address other risk factors
2. Estimate 10year ASCVDrisk score based on Pooled Cohort Equation
High Risk (≥20%): Initiate highintensity statin therapy (Class I)
Intermediate Risk (≥7.5% to <20%): If risk enhancersiii present, consider moderateintensity statin therapy (Class I)
If risk decision is uncertain, consider measuring CAC in selected adults
Consider no statin if CAC = 0 unless patient has diabetes or significant family history of ASCVD
Consider initiating statin therapy if CAC 199, especially if age >55 years
Initiate statin therapy if CAC ≥100
Borderline Risk (5% to <7.5%): If risk enhancersiii present, risk discussion regarding moderateintensity statin therapy (Class IIb)
Low Risk (<5%): Emphasize hearthealthy lifestyle (Class I)
5. Primary prevention—no diabetes, 2039 years and LDLC 70189 mg/dL (1.81 4.89 mmol/L)
a. Estimate lifetime ASCVDrisk based on Pooled Cohort Equation to encourage lifestyle to reduce ASCVD risk
b. Consider statin therapy if family history of premature ASCVD and LDLC ≥160 mg/dL (4.14 mmol/L)
iClinical ASCVD includes nonfatal MI, CHD death, and nonfatal and fatal stroke, TIA or peripheral arterial disease presumed to be of atherosclerotic origin.
iiVery high risk include history of >1 major ASCVD events including recent ACS (past 12 months), history of MI, ischemic stroke, or peripheral arterial disease; O R one
major ASCVD event p l u s multiple highrisk conditions: >65 years, familial hypercholesterolemia, history of coronary bypass graft surgery, diabetes mellitus,
hypertension, CKD, current smoking, LDLC >100 mg/dL (2.59 mmol/L) despite maximally tolerated statin and ezetimibe, or history of heart failure.
iiiRisk Enhancers include: LDLC 160189 mg/dL (4.144.89 mmol/L), family history of premature ASCVD, metabolic syndrome, chronic kidney disease, chronic
inflammatory conditions (eg, rheumatoid arthritis), premature menopause (before age 40), preeclampsia, highrisk race/ethnicities (eg, South Asian), triglycerides
≥175 mg/dL (1.98 mmol/L), hsCRP ≥2 mg/L, lipoprotein(a) ≥50 mg/dL (0.5 g/L), apoB ≥130 mg/dL, (1.3 g/L) or anklebrachial index <0.9.
ABI, ankle brachial index; ACC/AHA, American College of Cardiology/American Heart Association; ASCVD, Atherosclerotic cardiovascular disease; CAC, coronary artery
calcium; COR, Class of recommendation; hsCRP, highsensitivity C reactive protein; LDLC, lowdensity lipoprotein cholesterol; LOE, Level of evidence; NYHA, New
York Heart Association.
The HMGCoA reductase inhibitors or “statins” are the drugs of choice for most patients with dyslipidemia.20 A large body of evidence from
randomized, doubleblind, placebocontrolled trials has demonstrated the effectiveness of statins on reducing first and recurrent cardiovascular
events, cardiovascular mortality, and allcause mortality.29 The 2018 ACC/AHA Blood Cholesterol Guideline identified four statin benefit groups where
the data from RCT demonstrate clear evidence that the benefit of statin therapy outweighs the potential risks (Table 314).20 Nonstatin lipidlowering
therapies (such as ezetimibe, bempedoic acid, and PCSK9 inhibitors) play a supportive role in the management of dyslipidemia and are primarily used
in combination with statins when adequate LDLC lowering cannot be achieved with statins alone, or in patients unable to tolerate the recommended
dose of a statin (Table 315).30
TABLE 315
Key Recommendations on Role of Nonstatin Therapies to Reduce ASCVD Risk in Adults
b. Consider adding or replacing with PCSK9 inhibitor or adding bempedoic acidiv second
in combination with statins when adequate LDLC lowering cannot be achieved with statins alone, or in patients unable to tolerate the recommended
dose of a statin (Table 315).30 California Northstate University
Access Provided by:
TABLE 315
Key Recommendations on Role of Nonstatin Therapies to Reduce ASCVD Risk in Adults
1. Clinical ASCVD without comorbidities on maximally tolerated statin ≥70(1.81 ≥100 (2.59 mmol/L)
a. Consider adding ezetimibe first mmol/L)
b. Consider adding or replacing with PCSK9 inhibitor or adding bempedoic acidiv second
2. Clinical ASCVD with comorbidities on maximally tolerated statin ≥70(1.81 ≥100 (2.59 mmol/L)
a. Consider adding ezetimibe, bempedoic acid, or PCSK9 inhibitor as initial therapy and addition of mmol/L)
another agent if necessary
3. Clinical ASCVD and baseline LDLC >190 mg/dL (4.91 mmol/L) on maximally tolerated statin ≥70(1.81 ≥100 (2.59 mmol/L)
a. Consider adding ezetimibe, bempedoic acid, or PCSK9 inhibitor as initial therapy and addition of mmol/L)
another agent if necessary
4. Primary prevention and baseline LDLC >190 mg/dL (4.91 mmol/L) on maximally tolerated statin ≥100 (2.59 ≥130 (3.36 mmol/L)
a. Consider adding ezetimibe, bempedoic acid, or PCSK9 inhibitor as initial therapy and addition of mmol/L)
another agent if necessary
5. Primary prevention (age 4075 years) with diabetes and baseline LDLC 70189 mg/dL (1.814.89 ≥100 (2.59 ≥130 (3.36 mmol/L)
mmol/L) on maximally tolerated statin mmol/L)
a. Consider ezetimibe
iComorbidities include diabetes, recent ASCVD event (<3 months), ASCVD event while on a statin, poorly controlled ASCVDrisk factors, elevated lipoprotein(a),
chronic kidney disease, symptomatic heart failure, maintenance hemodialysis, baselines LDLC >190 mg/dL (4.91 mmol/L), age >65 years, prior MI or
nonhemorrhagic stroke, current daily cigarette smoking, symptomatic peripheral artery disease with prior history of heart attack or stroke, history of
nonmyocardial infarction–related coronary revascularizations, low HDLC, highsensitivity Creactive protein >2 mg/L, or metabolic syndrome.
iiEzetimibe is preferred if <25% additional LDLC needed, patients <3 months postacute coronary syndrome, cost considerations, patient preference.
iiiPCSK9 inhibitor is preferred if >25% additional LDLC needed and patient willing to administer subcutaneous injections.
Clinical ASCVD includes nonfatal MI, CHD death, and nonfatal and fatal stroke, TIA or peripheral arterial disease presumed to be of atherosclerotic origin.
ivLongterm cardiovascular outcome data is not yet available for bempedoic acid. Medications with documented cardiovascular benefits, when used in combination
ASCVD, Atherosclerotic cardiovascular disease; LDLC, lowdensity lipoprotein cholesterol; nonHDLC, nonhighdensity lipoprotein cholesterol; PCSK9, proprotein
subtilisin/kexin type 9.
Numerous clinical outcome trials have been performed to determine whether lipidlowering therapies reduce ASCVD risk in primary and secondary
prevention populations. Reducing total cholesterol and LDLC reduces CHD and total mortality. It is beyond the scope of this chapter to discuss each of
these trials in detail. Table 316 summarizes select trials supporting the use of statins and select nonstatins (ezetimibe and PCSK9 inhibitors) to reduce
Downloaded 2025213 10:53 P Your IP is
ASCVD risk.
Chapter 31:Most of the primary
Dyslipidemia, Daveand secondary
L. Dixon; prevention
Daniel M. Richestudies were doubleblinded, randomized, and placebocontrolled, lasting 2 toPage 7 years,
15 /and
37
©2025
most hadMcGraw Hill.patient
sufficient All Rights Reserved.
numbers Terms of Use
to be meaningful. The• body
Privacy Policy • Notice
of evidence • Accessibility
supporting the role of statin therapy to reduce ASCVD is significant and
metaanalysis data confirms these agents as the firstline therapy in dyslipidemia management.31 As mentioned previously in this chapter, not all lipid
lowering therapies have translated to reducing ASCVD risk, despite having favorable effects on the lipid profile.
subtilisin/kexin type 9.
Numerous clinical outcome trials have been performed to determine whether lipidlowering therapies reduce ASCVD risk in primary and secondary
prevention populations. Reducing total cholesterol and LDLC reduces CHD and total mortality. It is beyond the scope of this chapter to discuss each of
these trials in detail. Table 316 summarizes select trials supporting the use of statins and select nonstatins (ezetimibe and PCSK9 inhibitors) to reduce
ASCVD risk. Most of the primary and secondary prevention studies were doubleblinded, randomized, and placebocontrolled, lasting 2 to 7 years, and
most had sufficient patient numbers to be meaningful. The body of evidence supporting the role of statin therapy to reduce ASCVD is significant and
metaanalysis data confirms these agents as the firstline therapy in dyslipidemia management.31 As mentioned previously in this chapter, not all lipid
lowering therapies have translated to reducing ASCVD risk, despite having favorable effects on the lipid profile.
TABLE 316
Selected Landmark Clinical Trials with LipidLowering Drugs
Primary Prevention
Secondary Prevention
ARR, absolute risk reduction; AFCAPS/TexCAPS, Air Force/Texas Coronary Atherosclerosis Prevention Study32; CARDS, Collaborative Atorvastatin Diabetes Study12;
FOURIER, Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Patients with Elevated Risk33; HPS, Heart Protection Study34; IMPROVEIT, Improved
Reduction of Outcomes: Vytorin Efficacy International Trail35; JUPITER, Justification for the Use of Statins in Prevention36; NNT, Number Needed to Treat; RRR,
relative risk reduction; TNT, Treat to New Targets Trial37; WOSCOPS, The West of Scotland Coronary Prevention Study.38
Nonpharmacologic Therapy
Lifestyle modification is the cornerstone of ASCVDrisk reduction and is recommended in all patients, including those receiving lipidlowering therapy.
Weight and body mass index (BMI) should be determined at each visit and lifestyle patterns to induce a weight loss of 5% to 10% should be discussed in
persons who are overweight or obese. Moderatetovigorous intensity physical activity is recommended three to four times per week with each session
lasting 40 minutes on average. All patients should also be counseled to stop smoking and avoid tobacco products altogether.25
It is important to recognize that there is no single diet suitable for every patient. Instead, advise patients to reduce the percent of calories from
saturated and trans fats by following a dietary pattern that emphasizes vegetables, fruits, whole grains, lowfat dairy, poultry, fish, legumes, and nuts;
while limiting the intake of sweets, sugary beverages, and red meat. Plans that closely mirror this dietary pattern and effectively lower LDLC include
DASH, the USDA Food Pattern, and AHA Diet. Although the Mediterraneanstyle diet has no consistent effect on LDLC levels, it has been shown to
reduce major 2025213
Downloaded cardiovascular events
10:53 among
P Your IP ispersons at high cardiovascular risk when compared to a control diet. Any recommended dietary pattern
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche
should be adapted to a patient’s caloric requirements, cultural food preferences, and for other medical conditions (eg, diabetes mellitus).Page 16 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
Individualized diet counseling that provides acceptable substitutions for unhealthy foods and ongoing reinforcement by a registered dietitian are
necessary for maximal effect. It is also important to involve all family members, especially if the patient is not the primary person preparing food.25
lasting 40 minutes on average. All patients should also be counseled to stop smoking and avoid tobacco products altogether.25
California Northstate University
It is important to recognize that there is no single diet suitable for every patient. Instead, advise patients to reduce the percent of calories
Access Provided by: from
saturated and trans fats by following a dietary pattern that emphasizes vegetables, fruits, whole grains, lowfat dairy, poultry, fish, legumes, and nuts;
while limiting the intake of sweets, sugary beverages, and red meat. Plans that closely mirror this dietary pattern and effectively lower LDLC include
DASH, the USDA Food Pattern, and AHA Diet. Although the Mediterraneanstyle diet has no consistent effect on LDLC levels, it has been shown to
reduce major cardiovascular events among persons at high cardiovascular risk when compared to a control diet. Any recommended dietary pattern
should be adapted to a patient’s caloric requirements, cultural food preferences, and for other medical conditions (eg, diabetes mellitus).
Individualized diet counseling that provides acceptable substitutions for unhealthy foods and ongoing reinforcement by a registered dietitian are
necessary for maximal effect. It is also important to involve all family members, especially if the patient is not the primary person preparing food.25
Less than onethird of Americans meet the 20152020 Dietary Guidelines for Americans limit of less than 10% of calories from saturated fats.39 In
patients with lipid disorders, the 2013 AHA/ACC Lifestyle Management Guideline recommended only 5% to 6% of total calories from saturated fat.25
This can be achieved by recommending patients limit or avoid fast food, highfat dairy products, and sweets. Previous dietary guideline
recommendations to limit dietary cholesterol to 300 mg/day was omitted in 2015. However, it is still recommended that individuals limit their daily
dietary cholesterol intake. A dietary pattern low in saturated and trans fats will typically result in a reduction in dietary cholesterol since foods high in
saturated and trans fats are often high in cholesterol. A 12week trial of lifestyle modification is generally recommended before considering lipid
lowering therapy in patients without evidence of ASCVD, diabetes, or other highrisk features. Importantly, lifestyle modification alone is inappropriate
for patients with established ASCVD or diabetes given the benefit of statins in these highrisk patients.
Dietary Supplements
Select dietary supplements may be useful to augment diet and lipidlowering therapy. Increased intake of soluble fiber in the form of oat bran, pectins,
certain gums, and psyllium products can reduce total and LDLC, but have little or no effect on HDLC or TG levels. Soluble fiber binds cholesterol and
bile acids in the small intestine, which decrease absorption and reabsorption. Total daily fiber intake should be about 25 g/day, yet most Americans
average only half of the recommended amount.40 Dietary supplements containing fiber may be used to supplement the diet and achieve the
recommended daily intake. An intake of 3 to 12 g/day show reductions in total and LDLC of 10 mg/dL (0.26 mmol/L) and 12 mg/dL (0.31 mmol/L),
respectively, compared to control.30 It remains unknown if soluble fiber supplements have any impact on cardiovascular morbidity and mortality.
Although seemingly safe, patients should be advised to stay well hydrated to avoid gastrointestinal distress. These products may also be useful in
managing constipation associated with the bile acid sequestrants.
In epidemiologic studies, ingestion of large amounts of oily, coldwater fish (such as salmon) is associated with a reduction in ASCVD risk. Modest
consumption (12 servings per week) reduces the risk of cardiovascular death and total mortality.41 The American Heart Association recommends
eating oily fish at least twice a week; however, there are concerns with some types of fish (such as tuna) that often have high levels of environmental
contaminants. There are also concerns about environmental sustainability. Fish oil supplementation is an alternative option that provides a consistent
daily intake of omega3 PUFA such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Fish oil supplementation significantly reduces TG
levels and VLDLC, but may increase total cholesterol and LDLC. Other potentially favorable cardiovascular effects of fish oil supplementation include
antiarrhythmic, antiplatelet, and antiinflammatory properties. Despite this, a metaanalysis of 10 RCT found no benefit with low doses (less than 2
g/day) of omega3 PUFA supplementation.42 Still, current recommendations suggest lowdose omega3 PUFA supplementation is reasonable to
consider in secondary prevention patients with heart failure or at high risk of sudden cardiac death.43 Low dose omega3 PUFA supplementation is not
recommended in primary prevention patients, especially those with diabetes who do not have established ASCVD.43,44 Additional details regarding
prescription omega3 PUFA products are further discussed under drug therapy.
Phytosterols, including plant sterols and stanols isolated from vegetable oils, also reduce LDLC levels. Ingestion of 2 g/day will reduce LDLC by 5% to
15%, while doses above 3 g/day confer no additional LDLC lowering.28 The efficacy of plant sterols and plant stanols is considered to be comparable.
The mechanism by which phytosterols reduce LDLC remains unclear but may decrease the transport of cholesterol in the intestinal brush border
membrane and affect cholesterol uptake via NiemannPick C1Like 1 (NPC1L1) and other transporters. Because lipids are needed to solubilize
stanol/sterol esters, they are usually available in commercial butterlike spreads (such as Benecol®). The presence of plant stanols/sterols is listed on
the food label. Phytosterols should be administered 2 to 4 hours before or after bile acid sequestrants to avoid binding of phytosterols in the gut.
Although phytosterols are generally recognized as safe (GRAS) in the United States, they can cause gastrointestinal distress and should be avoided in
patients with sitosterolemia, a rare genetic disorder characterized by 50 to 100fold increase in plant sterol levels and rapid onset of atherosclerosis.45
The effects of longterm phytosterol supplementation on ASCVD risk remains unknown.
Red yeast rice is a commonly used dietary supplement in the United States that originates from Chinese medicine. The active ingredient of red yeast
rice is monacolin K, which is chemically identical to lovastatin. This leads some patients to believe red yeast rice is a “natural” statin and, therefore,
Downloaded 2025213 10:53 P Your IP is
safer
Chapterthan31:
statins currently available
Dyslipidemia, only byDaniel
Dave L. Dixon; prescription;
M. Richehowever, the active ingredient in overthecounter (OTC) red yeast rice productsPage
vary 17
by /over
37
©2025
120fold.McGraw Hill. All Rights
Many products containReserved.
little to no monacolin. 46
Terms of UseConversely,
• Privacy Policy • Noticeof•rhabdomyolysis,
case reports Accessibility liver toxicity, and renal failure have raised
concerns about some red yeast rice formulations containing significantly higher levels of monacolin K than described on the label.47 Red yeast rice is
Although phytosterols are generally recognized as safe (GRAS) in the United States, they can cause gastrointestinal distress and should be avoided in
California Northstate University
patients with sitosterolemia, a rare genetic disorder characterized by 50 to 100fold increase in plant sterol levels and rapid onset of atherosclerosis.45
Access Provided by:
The effects of longterm phytosterol supplementation on ASCVD risk remains unknown.
Red yeast rice is a commonly used dietary supplement in the United States that originates from Chinese medicine. The active ingredient of red yeast
rice is monacolin K, which is chemically identical to lovastatin. This leads some patients to believe red yeast rice is a “natural” statin and, therefore,
safer than statins currently available only by prescription; however, the active ingredient in overthecounter (OTC) red yeast rice products vary by over
120fold. Many products contain little to no monacolin.46 Conversely, case reports of rhabdomyolysis, liver toxicity, and renal failure have raised
concerns about some red yeast rice formulations containing significantly higher levels of monacolin K than described on the label.47 Red yeast rice is
not recommended as a suitable alternative to statins. However, if patients choose to take red yeast rice, it is recommended they purchase it from a
reputable supplier and avoid concurrent use with prescription statins.
Pharmacologic Therapy
There are numerous randomized, doubleblinded clinical trials demonstrating that reduction of LDLC reduces ASCVDevent rates in the setting of
primary and secondary prevention.27 Epidemiological studies suggest that every 38 mg/dL (0.98 mmol/L) reduction in LDLC produces a 21% reduction
in ASCVD event rates over 5 years.48 Additional findings from large prospective cohort studies and Mendelian randomization studies have also
demonstrated a dosedependent loglinear association between LDLC and ASCVD risk49 and that lower levels of LDLC achieve significant reductions
in ASCVD risk. These studies provide a strong rationale for attempting to lower plasma cholesterol and LDLC in patients at risk for ASCVD.49 It should
be noted, however, that not all lipidlowering agents that reduce LDLC have resulted in a reduction in ASCVD events (eg, CETP inhibitors). Thus, LDLC
lowering alone should not be the sole basis for selecting an appropriate agent.20,28 Lipidlowering drugs can be broadly divided into agents that
primarily decrease atherogenic cholesterolcontaining lipoprotein particles (such as statins) and those that primarily decrease TG levels (such as
fibrates).
Familial Hypercholesterolemia
Individuals with familial hypercholesterolemia (FH) have a very high lifetime risk of developing ASCVD. Compared to the general population, FH is
associated with a 24fold higher risk of developing acute MI before age 40.50 FH should be suspected in adults with untreated LDLC levels of 190 mg/dL
(4.91 mmol/L) or greater or nonHDL cholesterol levels of 220 mg/dL (5.69 mmol/L) or greater who have a family history of high cholesterol or ASCVD in
firstdegree relatives. Physical findings (such as xanthomas or corneal arcus) may be present in some patients with FH, but their absence does not rule
out a diagnosis of FH. In clinical practice, one of several validated tools, including the Dutch Lipid Clinic Network, US Make Early Diagnosis Prevent Early
Death (MEDPED), and SimonBroome Registry, are used to make a formal diagnosis of FH. Genetic testing is available but is only necessary when the
diagnosis is uncertain or if it is needed to obtain prior authorization for select lipidlowering therapies, such as PCSK9 inhibitors. Approximately 20%
of patients with clinically definite FH will not have an identifiable mutation; therefore, a negative genetic test does not exclude FH.51 Importantly,
cascade screening of all firstdegree relatives of diagnosed FH patients is highly recommended as an effective strategy to identify previously
undiagnosed FH patients.
Mipomersen and lomitapide are orphan drugs indicated for use in patients with HoFH and reduce LDLC levels by ∼25% and ∼40%, respectively.52
Mipomersen is an oligonucleotide inhibitor of apolipoprotein B100 synthesis administered via subcutaneous injection, while the orally administered
lomitapide is a microsomal triglyceride transfer protein, or microsomal TG transfer protein inhibitor, which reduces the level of cholesterol that the
liver and intestines assemble and secrete into the circulation. Mipomersen is associated with injection site pain and reactions, while both mipomersen
and lomitapide have a black box warning for severe hepatotoxicity and are only available through restricted Risk Evaluation and Mitigation Strategy
(REMS) programs.52 Other treatment options for HoFH patients include LDL apheresis (a process similar to dialysis that removes LDL from the blood)
and liver transplant.50
Hypertriglyceridemia
Elevated TG levels are strongly associated with an increased risk of ASCVD; however, the direct role of TG in the development of ASCVD is debed.53 All
patients with elevated TG levels (see Tables 311 and 312) should be advised to implement lifestyle interventions that reduce TG levels, including a 5%
to 10% reduction in body weight, reducing consumption of sugar and refined carbohydrates, increasing physical activity, smoking cessation, and
restricting alcohol. Secondary causes of hypertriglyceridemia should also be identified and addressed. Uncontrolled diabetes and chronic kidney
disease are common causes of elevated TG levels, along with certain medications (such as protease inhibitors and atypical antipsychotics). The best
approach to managing
Downloaded 2025213patients
10:53 Pwhose
YourTGIP levels
is remain elevated after optimizing lifestyle interventions and addressing secondary causes remains
unclear, but
Chapter 31: statins are generally
Dyslipidemia, Dave considered firstline
L. Dixon; Daniel M. given
Richethey can reduce TG levels by up to 30% at higher doses and help achieve desired levels
Page 18 / of
37
©202554McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
LDLC. The role of TGlowering therapies such as fibrates and omega3 PUFA in patients with TG of 200 to 499 mg/dL (2.26 5.64 mmol/L) is less clear.
Clinical trials are underway to determine if fibrates and omega3 PUFA reduce ASCVD risk in this population.55
Elevated TG levels are strongly associated with an increased risk of ASCVD; however, the direct role of TG in the development of ASCVD is debed.53 All
California
patients with elevated TG levels (see Tables 311 and 312) should be advised to implement lifestyle interventions that reduce Northstate
TG levels, University
including a 5%
Access Provided by:
to 10% reduction in body weight, reducing consumption of sugar and refined carbohydrates, increasing physical activity, smoking cessation, and
restricting alcohol. Secondary causes of hypertriglyceridemia should also be identified and addressed. Uncontrolled diabetes and chronic kidney
disease are common causes of elevated TG levels, along with certain medications (such as protease inhibitors and atypical antipsychotics). The best
approach to managing patients whose TG levels remain elevated after optimizing lifestyle interventions and addressing secondary causes remains
unclear, but statins are generally considered firstline given they can reduce TG levels by up to 30% at higher doses and help achieve desired levels of
LDLC.54 The role of TGlowering therapies such as fibrates and omega3 PUFA in patients with TG of 200 to 499 mg/dL (2.26 5.64 mmol/L) is less clear.
Clinical trials are underway to determine if fibrates and omega3 PUFA reduce ASCVD risk in this population.55
Fasting TG levels exceeding 500 mg/dL (5.65 mmol/L) are more commonly associated with pancreatitis and other consequences of
hyperchylomicronemia (such as eruptive xanthomas). At this level of elevated TG, a genetic form of hypertriglyceridemia often coexists with other
causes of elevated triglycerides such as diabetes. Dietary fat restriction is a basic element of treatment as this reduces the synthesis and entry of
additional chylomicrons into the circulation. Lipidlowering therapies that primarily lower TG levels (such as fibrates, omega3 PUFA, and niacin) are
recommended as firstline agents.54 Statins may be reasonable firstline options in those patients with an ASCVD risk of 7.5% or greater. If TG levels are
persistently over 500 mg/dL (5.65 mmol/L), it is reasonable to consider adding omega3 PUFA or fibrate therapy. Success in treatment is defined as a
reduction in triglycerides below 500 mg/dL (5.65 mmol/L) and preventing pancreatitis.
Low HDLC is a strong independent risk predictor of ASCVD.56 Low HDLC is defined as less than 40 mg/dL (1.03 mmol/L) for men and less than 50
mg/dL (1.29 mmol/L) for women, but there is no specified goal for HDLC raising. Low HDLC may be a consequence of insulin resistance, physical
inactivity, diabetes, cigarette smoking, very high carbohydrate intake, and certain drugs. In patients with low HDLC levels, the primary target remains
LDLC. Niacin has the potential for the greatest increase in HDLC compared to other lipidlowering therapies and the effect is more pronounced with
regular or immediaterelease forms than with sustainedrelease forms. However, no RCT has resulted in areduction in ASCVD risk by increasing HDL
C.57,58 Additionally, several CETP inhibitors capable of raising HDLC levels as much as 135% were evaluated in randomized, placebocontrolled trials
but no additional benefit was found when these drugs were added to background statin therapy.56 Due to the lack of pharmacological agents
demonstrating an improvement in clinical outcomes by focusing on raising HDLC, lifestyle modification (such as smoking cessation and increasing
physical activity) remains the preferred approach. Although alcohol consumption has been shown to increase HDLC, it is not acceptable to
recommend this to patients who do not already consume alcohol.
Statins (such as atorvastatin) are considered the firstline lipidlowering therapies for managing dyslipidemia due to robust evidence from multiple
RCTs demonstrating that statins significantly decrease the risk of first (primary prevention) and recurrent (secondary prevention) cardiovascular
events.20,48 Statins significantly reduce LDLC levels (20%60%), modestly increase HDLC (6%12%) and decrease TG levels (10%29%).59 Statins
interrupt the conversion of HMGCoA to mevalonate, the ratelimiting step in de novo cholesterol biosynthesis, by inhibiting HMGCoA reductase (see
Fig. 314). Metabolic studies with statins in normal volunteers and patients with hypercholesterolemia suggest a reduced synthesis of LDLC, as well as
enhanced catabolism of LDL mediated through LDL receptors, as the principal mechanisms for lipidlowering effects. Statin selection is primarily
based on the patients individual ASCVD risk and indicated intensity (see Tables 314 and 317). Currently available products in order of decreasing LDL
C lowering potency, include rosuvastatin, atorvastatin, pitavastatin, simvastatin, lovastatin, pravastatin, and fluvastatin.59 The plasma halflives for all
the statins are relatively short (13 hours) except for atorvastatin, pitavastatin, and rosuvastatin, which may account for their potency.59 Statins are
generally well tolerated but are not without adverse effects. However, discontinuation rates due to adverse effects in randomized, doubleblind,
placebocontrolled trials have often been similar between statin and placebo.60
TABLE 317
Intensity of Statin Therapy by Drug and Daily Dose
Lowers LDL on average by ≥50% Lowers LDL on average by 30% to <50% Lowers LDL on average by <30%
Atorvastatin 4080 mg Atorvastatin 1020 mg Simvastatin 10 mg
DownloadedRosuvastatin 2040 P
2025213 10:53 mgYour IP is Rosuvastatin 510 mg Pravastatin 1020 mg
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche
Simvastatin 2040 mgi Lovastatin 20 mg Page 19 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility Fluvastatin 2040 mg
Pravastatin 4080 mg
Lovastatin 40 mg Pitavastatin 1 mg
C lowering potency, include rosuvastatin, atorvastatin, pitavastatin, simvastatin, lovastatin, pravastatin, and fluvastatin.59 The plasma halflives for all
California Northstate University
the statins are relatively short (13 hours) except for atorvastatin, pitavastatin, and rosuvastatin, which may account for their potency.59 Statins are
Access Provided by:
generally well tolerated but are not without adverse effects. However, discontinuation rates due to adverse effects in randomized, doubleblind,
placebocontrolled trials have often been similar between statin and placebo.60
TABLE 317
Intensity of Statin Therapy by Drug and Daily Dose
Lowers LDL on average by ≥50% Lowers LDL on average by 30% to <50% Lowers LDL on average by <30%
Atorvastatin 4080 mg Atorvastatin 1020 mg Simvastatin 10 mg
Rosuvastatin 2040 mg Rosuvastatin 510 mg Pravastatin 1020 mg
iSimvastatin is not recommended by the FDA to be initiated at 80 mg/day due to increased risk of myopathy and rarely rhabdomyolysis.
Boldface type indicates medications that have cardiovascular outcome data from RCTs when given in the specified dose.
Statinassociated muscle symptoms (SAMS) are reported by 10% to 25% of statin users and are frequently reported by patients as a reason for statin
discontinuation.61 While various definitions of SAMS exist, the clinical diagnosis of SAMS is based on a subjective clinical assessment. Myalgia is the
most commonly reported musclerelated adverse effect with statin therapy and refers to bilateral muscle achiness, weakness, or cramps affecting
larger muscle groups (such as thighs and back). Myopathy is often used interchangeably with myalgia, but myopathy is a general term used for any
musclerelated symptoms. The most concerning of SAMS is rhabdomyolysis, which is a rapid breakdown of skeletal muscle resulting in creatine kinase
(CK) elevations greater than 10 times the upper limit of normal. The release of myoglobin from damaged muscle tissue may also compromise renal
function and lead to acute kidney injury. Patients presenting with rhabdomyolysis will often describe their urine as dark or “teacolored” and present
with nausea, vomiting, confusion, coma, cardiac arrhythmias, electrolyte disturbances, and even death. Fortunately, rhabdomyolysis in statintreated
patients is exceedingly rare occurring in only 0.1% of patients in RCT compared to 0.04% of patients receiving placebo. Rhabdomyolysis is not only
caused by statins but can also be induced by extreme physical exercise, certain metabolism disorders (eg, diabetic ketoacidosis), other drugs (eg,
colchicine), toxins, and infection.
Certain risk factors are known to increase the risk of developing SAMS and recognition of these risk factors at the time of statin initiation may minimize
the risk of SAMS. Known risk factors include advanced age, female gender, low body mass index, frequent heavy exercisers, comorbidities (eg, kidney
disease, hypothyroidism), and increased serum statin concentrations due to drug–drug interactions.61 A lower dose might be necessary for patients
with multiple risk factors for SAMS, and once the starting dose is tolerated, the dose can be titrated to the desired potency. Avoiding major drug–drug
interactions is a significant modifiable risk factor for SAMS that pharmacists can directly impact. Nearly 80% of all medications are metabolized in the
liver by the cytochrome P450 system (CYP) with CYP3A4 being the most predominant.62 Statins are no different as nearly all statins, except pravastatin,
are metabolized to some degree by CYP isoenzymes. Lovastatin, simvastatin, and atorvastatin are associated with more significant drug–drug
interactions since they are predominantly metabolized by CYP3A4, while fluvastatin, pitavastatin, and rosuvastatin rely on other CYP isoenzymes (eg,
CYP2C9, CYP2C8, CYP2C19).63 The coprescribing of medications that compete with or inhibit the same CYP isoenzyme (such as verapamil) can increase
serum statin concentrations and the risk for SAMS. The concurrent use of medications such as gemfibrozil that interfere with statin glucuronidation,
which is responsible for statin clearance, increases the risk of SAMS.
The management of SAMS requires a multifaceted approach. Documentation of the patient’s reported symptoms and determining the probability of
SAMS is an important first step. A Statin Intolerance App (available at: http://www.acc.org/statinintoleranceapp) created by the ACC is a helpful
resource that can be used to determine the possibility of SAMS and provide guidance on managing patients with possible SAMS. Statin therapy should
be generally discontinued in patients with intolerable symptoms. If symptoms resolve, initiate a different statin at a lower dose.61,64 Additionally,
Downloaded 2025213 10:53 P Your IP is
hydrophilic
Chapter 31:statins (such as Dave
Dyslipidemia, rosuvastatin)
L. Dixon;may be better
Daniel tolerated than lipophilic statins (such as simvastatin). In patients where symptomsPage
M. Riche do not
20 / 37
©2025 McGraw
improve, Hill. All Rights
other potential causesReserved. Terms
of muscle pain of Use
should • Privacy including
be excluded, Policy • Notice and vitamin D deficiency, before a statin rechallenge.64
• Accessibility
hypothyroidism
Alternative dosing strategies (eg, every other day) using statins with long halflives (atorvastatin, rosuvastatin, and likely pitavastatin) may also be
considered. Nonstatin therapies may be considered in patients who fail multiple statins. While routine CK monitoring is not recommended, a CK
which is responsible for statin clearance, increases the risk of SAMS.
California Northstate University
The management of SAMS requires a multifaceted approach. Documentation of the patient’s reported symptoms and determining
Access Provided the
by: probability of
SAMS is an important first step. A Statin Intolerance App (available at: http://www.acc.org/statinintoleranceapp) created by the ACC is a helpful
resource that can be used to determine the possibility of SAMS and provide guidance on managing patients with possible SAMS. Statin therapy should
be generally discontinued in patients with intolerable symptoms. If symptoms resolve, initiate a different statin at a lower dose.61,64 Additionally,
hydrophilic statins (such as rosuvastatin) may be better tolerated than lipophilic statins (such as simvastatin). In patients where symptoms do not
improve, other potential causes of muscle pain should be excluded, including hypothyroidism and vitamin D deficiency, before a statin rechallenge.64
Alternative dosing strategies (eg, every other day) using statins with long halflives (atorvastatin, rosuvastatin, and likely pitavastatin) may also be
considered. Nonstatin therapies may be considered in patients who fail multiple statins. While routine CK monitoring is not recommended, a CK
measurement prompted by patient symptoms can be used to exclude rhabdomyolysis and can assist with identifying those with definite myalgia.
Importantly, patients should be reassured that statins are effective and safe, and SAMS is reversible with statin discontinuation.
Other notable adverse effects of statins include mild elevations in serum transaminase levels (primarily alanine aminotransferase [ALT]). Liver
enzymes are not, however, an accurate measure of liver function and there is no causal relationship between statin use and liver failure. Therefore,
routine periodic monitoring of liver enzymes is not required, but liver enzyme tests should be obtained before starting statin therapy to have a
baseline value for comparison if liver enzymes are later discovered to be elevated. Other potential causes for elevated liver enzymes, including
excessive alcohol intake, infection, and select medications should also be evaluated. Statins may be initiated in patients with chronic liver disease,
compensated cirrhosis, and nonalcoholic fatty liver disease; however, statins are contraindicated in patients with decompensated cirrhosis or acute
liver failure.64
Statin use is also associated with a small increased risk of newonset diabetes.61 This was first observed in the JUPITER (Justification for the Use of
Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin) trial, where the number of newonset diabetes cases was 0.6% higher in those
receiving rosuvastatin 20 mg/day compared to placebo.36 Subsequent metaanalyses of statin trials have also found a modest increase in the number
of newonset diabetes cases among statintreated patients compared to placebo, but the absolute risk increase is <1%.65,66 Common attributes of
statin users who develop newonset diabetes include receiving higher doses of statins and having other risk factors for diabetes, including obesity,
impaired fasting glucose, HbA1c >6% (0.06; 42 mmol/mol Hb) or metabolic syndrome.67 Mechanisms to explain the association between statin use and
newonset diabetes remains unclear. However, observational data suggest that higher cholesterol levels are protective against developing diabetes.
This may be attributable to changes at the cellular level involving disruption of cholesterolsensitive cellular functions that affect insulin secretion and
insulin sensitivity.61 Ultimately, the benefit of statin therapy greatly outweighs the risk of newonset diabetes as statin use in patients at high ASCVD risk
will prevent approximately three ASCVD events for every new case of diabetes.
Ezetimibe is a preferred adjunct therapy given it has been shown to modestly reduce the risk of recurrent cardiovascular events in a secondary
prevention population when used in combination with statin therapy.35 The primary lipidlowering effect of ezetimibe is a modest reduction in LDLC
of 15% to 24%; with higher reductions achievable when used in combination with statin therapy.35,68 Ezetimibe reduces LDLC by inhibiting the NPC1L1
protein, an important transporter of cholesterol absorption in the small intestine and hepatocytes.68 Known polymorphisms of NPC1L1 are associated
with lower LDLC levels and decreased ASCVD risk; thus, providing a rational why ezetimibe reduces ASCVD risk.69 Other than mild gastrointestinal
complaints (such as diarrhea) and postmarketing reports of myalgia and mild ALT elevations when used in combination with statins, ezetimibe is
generally well tolerated. Previous concerns over a potential increased risk of cancer have been nullified given recent prospective clinical trial data
showing there is no increased risk of cancer with ezetimibe use.35 Ezetimibe has no effects on the CYP450 enzyme system; however, concomitant use
with cyclosporine can lead to increased exposure to both ezetimibe and cyclosporine.68
The bile acid sequestrants (BAS), such as colesevelam, modestly reduce LDLC (13%20%) and reduce cardiovascular events when used as
monotherapy.70 There is currently no data to determine if the benefits observed with BAS monotherapy translate to its use in combination with statin
therapy. As such, BAS are generally used as adjunct therapy with statins when desired LDLC levels are not achieved with statins alone. Importantly,
BAS are considered first line during pregnancy since they are not systemically absorbed and pose no risk to the fetus. The primary action of BAS is to
bind bile acids in the intestinal lumen, with a concurrent interruption of enterohepatic circulation of bile acids and a markedly increased excretion of
acidic steroids in the feces. This decreases the bile acid pool size and stimulates the hepatic synthesis of bile acids from cholesterol. Depletion of the
hepatic pool of cholesterol results in an increase in cholesterol biosynthesis and an increase in the number of LDLR on the hepatocyte membrane.
The increased number of receptors stimulates an enhanced rate of catabolism from plasma and lowers LDLC levels. The increase in hepatic
cholesterol biosynthesis may be paralleled by increased hepatic VLDL production and, consequently, BAS may aggravate hypertriglyceridemia and
should be avoided in those with TG levels exceeding 300 mg/dL (3.39 mmol/L).30
Downloaded 2025213 10:53 P Your IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 21 / 37
One of the main barriers to BAS is their poor tolerability profile. Early BAS (such as cholestyramine) were developed as powders that require mixing
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
with water or juice to create a slurry for oral administration. Gastrointestinal complaints of constipation, bloating, epigastric fullness, nausea, and
flatulence are commonly reported with these formulations.30 These adverse effects can be minimized by increasing fluid intake, modifying the diet to
bind bile acids in the intestinal lumen, with a concurrent interruption of enterohepatic circulation of bile acids and a markedly increased excretion of
acidic steroids in the feces. This decreases the bile acid pool size and stimulates the hepatic synthesis of bile acids fromCalifornia Northstate
cholesterol. DepletionUniversity
of the
hepatic pool of cholesterol results in an increase in cholesterol biosynthesis and an increase in the number of LDLR on Access
the hepatocyte
Provided by: membrane.
The increased number of receptors stimulates an enhanced rate of catabolism from plasma and lowers LDLC levels. The increase in hepatic
cholesterol biosynthesis may be paralleled by increased hepatic VLDL production and, consequently, BAS may aggravate hypertriglyceridemia and
should be avoided in those with TG levels exceeding 300 mg/dL (3.39 mmol/L).30
One of the main barriers to BAS is their poor tolerability profile. Early BAS (such as cholestyramine) were developed as powders that require mixing
with water or juice to create a slurry for oral administration. Gastrointestinal complaints of constipation, bloating, epigastric fullness, nausea, and
flatulence are commonly reported with these formulations.30 These adverse effects can be minimized by increasing fluid intake, modifying the diet to
increase bulk, and using stool softeners. Tablet forms of BAS (such as colesevelam) are generally better tolerated than resin powders and associated
with lower overall discontinuation rates.71 Other potential adverse effects include impaired absorption of fatsoluble vitamins A, D, E, and K;
gastrointestinal obstruction; and reduced bioavailability of other drugs such as warfarin, levothyroxine, and phenytoin.30 Drug–drug interactions may
be avoided by taking other medications 1 hour before or 4 hours after the BAS.30 Colesevelam is not only approved as a lipidlowering agent but also as
an antihyperglycemic that modestly lowers glucose levels in patients with type 2 diabetes mellitus.72 Given the better safety and tolerability profile of
ezetimibe, BAS should be reserved only for those patients unable to tolerate ezetimibe who need additional LDLC lowering despite maximally
tolerated statin therapy.30
The PCSK9 inhibitors (e.g. alirocumab) reduce LDLC by as much as 60% when added to background statin therapy. Inhibiting PCSK9 promotes LDLR
recycling to the cell surface, which increases LDLC clearance from the circulation. Both alirocumab and evolocumab are fully human monoclonal
antibodies to PCSK9 and were approved by the FDA in 2015.30 Randomized, doubleblind clinical trials have also shown these agents effectively reduce
recurrent cardiovascular events in patients following an acute coronary event and secondary prevention populations when added to background
statin therapy.33,73Alirocumab and evolocumab are both administered by subcutaneous injection. Although this may be a barrier for some patients,
PCSK9 inhibitors can be administered biweekly or oncemonthly. The most common adverse effect reported are injection site reactions, which can be
minimized by allowing the injection to come to room temperature before use and icing the site before injecting. Some patients may also report “flu
like” symptoms after the injection. There were initial concerns over a potential increased risk for neurocognitive adverse effects; however, a
randomized trial found no difference in cognitive function between those randomized to evolocumab versus placebo over 19 months of followup.74
Furthermore, those patients who reach very low levels of LDLC (less than 20 mg/dL [0.53 mmol/L]) do not appear to be an increased risk of adverse
events.75 Despite this favorable data, the longterm effects of achieving very low levels of LDLC with PCSK9 inhibitors remains unknown. Despite their
LDLC lowering potency and favorable safety profile, PCSK9 inhibitor use has been limited due to their high cost. Although PCSK9 inhibitors should
primarily be used in combination with maximally tolerated statins in highrisk patients unable to achieve desired LDLC levels with a statin alone,
evolocumab is FDAapproved for use as monotherapy in patients with primary hyperlipidemia (such as heterozygous familial
hypercholesterolemia).30,72
Inclisiran is a small interfering RNA (siRNA) molecule that reduces the production of PCSK9 by inhibiting messenger RNA.76 This novel biological agent,
which pending approved by the FDA, has a sustained effect on LDLlowering and is given subcutaneously every 3 to 6 months. In Phase 3 clinical trials,
inclisiran reduced LDLC by an average of 50% when given as addon therapy in patients who were treated with a highintensity statin but had not
achieved their LDLC goal. Similar to alirocumab and evolocumab, inclisiran therapy should be reserved for patients with familial
hypercholesterolemia and those with established ASVCD who do not achieve LDLC targets despite the use of a highintensity statin or unable to
tolerate statin therapy. Injection site reactions were uncommon (<3% of study participants), transient, and generally mild. Whether inclisiran use
produces a significant reduction in cardiovascular event rates has not been firmly established.
Adenosine triphosphatecitrate lyase (ACL) is a cytoplasmic enzyme responsible for generating acetyl coenzyme A which is needed during the de novo
synthesis of fatty acids and cholesterol. ACL inhibitors prevent cholesterol production upstream from HMG CoA reductase inhibitors (i.e. statins) and
the two therapeutic strategies can be used in combination. Bempedoic acid is an orally administered ACL inhibitor that has been approved for use in
combination with maximally tolerated statin therapy in patients with established ASCVD or familial hypercholesterolemia who require additional LDLC
lowering. In Phase 3 clinical trials, bempedoic acid produced modest reductions in LDLC (1520%) when combined with statin therapy or used as
monotherapy in patients who are unable to tolerate statins.77 It can also be combined with a cholesterol absorption inhibitor (i.e. ezetimibe).
Bempedoic acid plus ezetimibe resulted in a 36% reduction in LDLC from baseline in a Phase 3 clinical trial.78 Bempedoic acid is generally well
tolerated and does not appear to cause or contribute to muscle symptoms. However, bempedoic acid may cause hyperuricemia and tendon rupture.
Downloaded 2025213
Increased blood 10:53 PofYour
concentrations IP iswere seen in some patients treated with bempedoic acid and this may increase the risk of acute gout in
uric acid
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 22 / 37
those
©2025with a history
McGraw Hill.ofAll
gouty arthritis.
Rights Mechanistically,
Reserved. bempedoic
Terms of Use • Privacyacid inhibits
Policy the renal
• Notice tubular organic anion transporter 2 (OAT2), which plays a role
• Accessibility
in the renal uptake of uric acid from the blood. Though rare, bempedoic acid was associated with an increased risk of tendon rupture or injury (0.5%)
in clinical trials. This uncommon but potentially debilitating adverse effect was not reported in patients who received placebo. Risk factors for tendon
the two therapeutic strategies can be used in combination. Bempedoic acid is an orally administered ACL inhibitor that has been approved for use in
combination with maximally tolerated statin therapy in patients with established ASCVD or familial hypercholesterolemia who require
California additional
Northstate LDLC
University
lowering. In Phase 3 clinical trials, bempedoic acid produced modest reductions in LDLC (1520%) when combined withAccess
statin therapy
Provided by: or used as
monotherapy in patients who are unable to tolerate statins.77 It can also be combined with a cholesterol absorption inhibitor (i.e. ezetimibe).
Bempedoic acid plus ezetimibe resulted in a 36% reduction in LDLC from baseline in a Phase 3 clinical trial.78 Bempedoic acid is generally well
tolerated and does not appear to cause or contribute to muscle symptoms. However, bempedoic acid may cause hyperuricemia and tendon rupture.
Increased blood concentrations of uric acid were seen in some patients treated with bempedoic acid and this may increase the risk of acute gout in
those with a history of gouty arthritis. Mechanistically, bempedoic acid inhibits the renal tubular organic anion transporter 2 (OAT2), which plays a role
in the renal uptake of uric acid from the blood. Though rare, bempedoic acid was associated with an increased risk of tendon rupture or injury (0.5%)
in clinical trials. This uncommon but potentially debilitating adverse effect was not reported in patients who received placebo. Risk factors for tendon
rupture appear to be age greater than 60 years, concurrent use of corticosteroids or fluoroquinolones, renal failure, and history of tendon disorders.
Until the results of the longterm cardiovascular outcome trial are published, the role of bempedoic acid in the management of dyslipidemia should be
limited.
Although fibrates, such as gemfibrozil, have been shown to reduce cardiovascular events when used as monotherapy, there is less evidence to support
their use in combination with statin therapy.53,54 Fibrates are primarily used in patients with TG levels that exceed 500 mg/dL (5.65 mmol/L) to reduce
the risk of acute pancreatitis. The two available fibrates, gemfibrozil and fenofibrate, are potent TGlowering therapies (20%50%), but may cause a
modest reciprocal rise in LDLC in patients with severely elevated TG levels.55 Plasma HDLC concentrations may rise 10% to 15% or more with fibrates.
Gemfibrozil increases the activity of LPL and reduces to a lesser extent the synthesis or secretion of VLDL from the liver into the plasma. Fenofibrate
increases LPL activity and reduces apoprotein CIII (an inhibitor of LPL) by activating peroxisome proliferatoractivated receptor α (PPARα), which
regulates the expression of genes involved in the regulation of lipids and other metabolic processes.
Fibrates are generally well tolerated, but gastrointestinal complaints and transient elevations in transaminase levels have been reported.79 Both
gemfibrozil and fenofibrate require dose adjustments for significant renal impairment and fenofibrate has been reported to worsen renal function,
although this is usually transient and selflimiting.20,80 Musclerelated adverse effects can occur with both gemfibrozil and fenofibrate alone but is
more common when used in combination with statins to manage complex dyslipidemia or elevated TG levels. Gemfibrozil, and its glucuronide
metabolite, has potent effects on CYP450 enzymes (such as CYP3A4), intestinal, hepatic, and renal transporters making it highly prone to significantly
increase serum statin concentrations and the risk of SAMS.63 For this reason, current guidelines do not recommend gemfibrozil to be initiated in
patients receiving statin therapy; fenofibrate is favored instead.20 Fenofibrate and gemfibrozil and may enhance the formation of gallstones, but this
occurs rarely.81 Fibrates may potentiate the effects of warfarin and the international normalized ratio (INR) should be monitored very closely with this
combination.82
High doses of omega3 fatty PUFA (24 g/day of EPA/DHA) significantly reduce TG and VLDL cholesterol levels (20%50%) with lesser effects on other
lipoproteins.55 The mechanisms by which omega3 PUFA reduce TG levels include increasing hepatic oxidation of free fatty acids, increasing LDL
hydrolysis by activating PPARα and inhibiting apoprotein CIII. The omega3 PUFA formulations approved by the FDA for treating TG levels of 500
mg/dL (5.65 mmol/L) or greater include an omega3acid ethyl ester of EPA/DHA (Lovaza®), omega3carboxylic acid of EPA/DHA (Epanova®), and ethyl
ester of EPA only (Vascepa®). DHA and EPA have different effects on LDLC as EPA prevents LDL oxidation and promotes LDL clearance, whereas DHA
does not; however, the clinical significance of this remains unclear.83 Prescription omega3 PUFA products contain approximately 1 g of EPA/DHA per
capsule, whereas the EPA/DHA content of OTC “fish oil” supplements is often less than 300 mg per capsule and are not regulated by the FDA. Unless
patient affordability is an issue, prescription omega3 PUFA are preferred to minimize pill burden and ensure product quality. Randomized clinical
trials of omega3 PUFA have shown mixed results due to the lack of generalizability due to the population studied, the background lipidlowering
therapy used, and dose taken.55,83 The recently published REDUCEIT study, a landmark trial, evaluated the effects of icosapent ethyl (4g/day), a high
potency EPA derivative, used as addon therapy to statins.84 At baseline, patients enrolled in the REDUCEIT study had a median TG level of 216 mg/dL
(2.44 mmol/L) and the majority had a history of ASCVD. Icosapent ethyl reduced the rate of ischemic events by 25% and significantly reduced the risk of
cardiovascular death when compared to placebo. However, icosapent ethyl also increased the risk of hospitalization for atrial fibrillation or flutter. The
REDUCEIT study was released shortly after the 2018 ACC/AHA guidelines were published and, thus, icosapent ethyl was not discussed or
recommended. Future guidelines should bring clarity to the role of icosapent ethyl for the treatment of hypertriglyceridemia in patients at high risk of
ASCVD events.
Downloaded 2025213 10:53 P Your IP is
Chapter 31: Dyslipidemia,
Gastrointestinal complaintsDave
(suchL.
asDixon; Daniel
abdominal M.and
pain Riche
“fishy burps”) are common with OTC omega3 PUFA products but may be minimized Pageby23 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
refrigerating the capsules. However, they should not be kept frozen in the freezer. Caution is advised in patients with known sensitivities or allergies to
fish or shellfish. Drug–drug interactions are minimal with omega3 PUFA, although caution is advised when used concomitantly with antiplatelet
potency EPA derivative, used as addon therapy to statins.84 At baseline, patients enrolled in the REDUCEIT study had a median TG level of 216 mg/dL
(2.44 mmol/L) and the majority had a history of ASCVD. Icosapent ethyl reduced the rate of ischemic events by 25% and California Northstate
significantly University
reduced the risk of
cardiovascular death when compared to placebo. However, icosapent ethyl also increased the risk of hospitalization forAccess
atrialProvided
fibrillation
by: or flutter. The
REDUCEIT study was released shortly after the 2018 ACC/AHA guidelines were published and, thus, icosapent ethyl was not discussed or
recommended. Future guidelines should bring clarity to the role of icosapent ethyl for the treatment of hypertriglyceridemia in patients at high risk of
ASCVD events.
Gastrointestinal complaints (such as abdominal pain and “fishy burps”) are common with OTC omega3 PUFA products but may be minimized by
refrigerating the capsules. However, they should not be kept frozen in the freezer. Caution is advised in patients with known sensitivities or allergies to
fish or shellfish. Drug–drug interactions are minimal with omega3 PUFA, although caution is advised when used concomitantly with antiplatelet
agents or anticoagulants since omega3 PUFA may prolong bleeding time.
Niacin
Niacin (nicotinic acid) increases HDLC (5%30%), and lowers TG (20%50%) and LDLC (5%20%). Despite these favorable changes in the lipid profile,
niacin has not been shown to improve cardiovascular outcomes in patients on background statin therapy with relatively wellcontrolled lipids at
baseline.57,58 Niacin primarily lowers TG levels by inhibiting lipolysis with a decrease in free fatty acids in plasma and decreased hepatic esterification
of TG. It also significantly raises HDLC by reducing its catabolism and selectively decreasing hepatic removal of HDL apoAI but not the removal of
cholesterol esters, thereby increasing the capacity of retained apoAI to augment reverse cholesterol transport in isolated hepatic cells. Niacin also
reduces the hepatic synthesis of VLDL, which, in turn, leads to a reduction in the synthesis of LDL. However, the modest decrease in serum LDLC levels
is dosedependent.
Niacin has many adverse drug reactions that frequently limit its use. Cutaneous flushing and itching appear to be prostaglandin mediated and can be
reduced by administering aspirin 325 mg given shortly before niacin ingestion.85 Flushing seems to be related to rising plasma concentrations of niacin
and the use of immediaterelease formulations; taking the dose with meals and slowly titrating the dose upward may also minimize these effects.
Extended or sustainedrelease products may minimize these complaints in some patients. The only legend form of niacin, Niaspan® (Abbott), is an
extendedrelease form of niacin with pharmacokinetics intermediate between immediate and sustainedrelease products that are sold as food
supplements rather than legend products. In controlled trials, Niaspan® is reported to have fewer dermatologic reactions and has a lower risk for
hepatoxicity.85 Potentially important laboratory abnormalities occurring with niacin therapy include elevated liver function tests, hyperuricemia, and
hyperglycemia. With less than 3 g/day, the degree of liver function test elevation is generally not marked and often transient, and a temporary
reduction in dosage frequently corrects the problem. Preexisting gout and diabetes may be exacerbated by niacin; these patients should be
monitored more closely and their medication titrated appropriately.86,87 Niacin is contraindicated in patients with active liver disease and active peptic
ulcer disease. Concomitant alcohol and hot beverages may magnify flushing and pruritus with niacin and they should be avoided at the time of
ingestion. Nicotinamide should not be used in the treatment of hyperlipidemia, as it does not effectively lower cholesterol or TG levels.
Special Populations
Older Adults
Dyslipidemia is an independent risk factor for ASCVD in older adults (greater than 65 years old), as it is in the younger patient.88 The attributable risk,
which is the difference in absolute rates of cardiovascular events between segments of the population with higher or lower serum cholesterol levels,
increases with age. Drug therapy in principle differs little from younger patients, and older patients respond to lipidlowering therapies as well as
younger patients. The gain in life expectancy may be small depending on the age at the start of treatment and the magnitude of LDLC reduction. The
benefits of moderatetohigh intensity statin therapy in older adults for secondary prevention is quite clear, while the benefit of statins in older adults
for primary prevention is more controversial.88 This is especially true in individuals greater than 75 years of age since this age group is poorly
represented in RCTs.20 Primary prevention in younger patients requires about 2 years before reduction in ASCVD risk is apparent, and this lag time
should be taken into consideration, along with life expectancy, in patient selection for statin therapy in older adults. To help address this gap in the
evidence, the ongoing Statin Therapy for Reducing Events in the Elderly (STAREE) trial (NCT02099123) is evaluating the efficacy of atorvastatin 40
mg/day for primary prevention in those 70 years or older without diabetes.
The risks of statin therapy in older adults must also be considered. Changes in body composition, renal function, and other physiologic changes of
aging may make older patients more susceptible to the adverse effects of lipidlowering drug therapy.88 Older adults are more prone to developing
SAMS and the effects of SAMS on the risk of falls and functional status remains unclear.83 There is also concern regarding the potential negative effects
of statins on cognitive function; however, data from metaanalyses has suggested statins are not associated with adverse cognitive effects.89,90 Statin
use has also been associated with increased risk of cataracts, which is highly prevalent among older adults, yet a metaanalysis found no clear evidence
Downloaded 2025213 10:53 P Your IP is 91
showing
Chapter statins increase theDave
31: Dyslipidemia, risk ofL.cataracts. Older
Dixon; Daniel M. adults
Riche are also more likely to develop type 2 diabetes and the impact of statin therapy on new
Page 24 / 37
onset diabetes in older adults is a concern warranting further study.
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
Children
The risks of statin therapy in older adults must also be considered. Changes in body composition, renal function, and other physiologic
California changes
Northstate of
University
aging may make older patients more susceptible to the adverse effects of lipidlowering drug therapy.88 Older adults areAccess
more prone
Provided by:to developing
SAMS and the effects of SAMS on the risk of falls and functional status remains unclear.83 There is also concern regarding the potential negative effects
of statins on cognitive function; however, data from metaanalyses has suggested statins are not associated with adverse cognitive effects.89,90 Statin
use has also been associated with increased risk of cataracts, which is highly prevalent among older adults, yet a metaanalysis found no clear evidence
showing statins increase the risk of cataracts.91 Older adults are also more likely to develop type 2 diabetes and the impact of statin therapy on new
onset diabetes in older adults is a concern warranting further study.
Children
While cardiovascular events rarely occur in those under 18 years of age, the process of atherosclerosis often begins during childhood.92 Early
identification and management of risk factors is critical for primordial prevention of ASCVD. Dyslipidemia in children can develop from secondary
causes, similar to adults, or may present as primary dyslipidemia (such as FH). Universal lipid screening is recommended between age 9 and 11 as this
is a stable time for lipid assessment before the onset of puberty, which decreases cholesterol levels 10% to 20%.92 Lipid screening before age 9 is only
recommended in children with a significant family history of premature ASCVD, known firstdegree relatives with dyslipidemia, or other cardiovascular
risk factors (such as diabetes, obesity, or hypertension).92
Drug therapy in children is not recommended until the age of 10 years or older and the guidelines for initiation of therapy and acceptable levels of
cholesterol and lipoproteins are quite different that adults (see Table 313).92 Children younger than 10 years should only receive drug therapy if they
have a genetic lipid disorder (such as FH) or highrisk ASCVD condition (such as diabetes); these children should be referred to a pediatric lipid
specialist.92 Lifestyle interventions are generally the mainstay of therapy, yet children with FH will often require drug therapy. Pravastatin may be used
in children as young as 8 years old, while all other statins are indicated for use in children 10 years of age and older.92 Start with the lowest available
statin dose and titrate every 3 months as necessary to achieve treatment goals. Appropriate contraception strategies are recommended in females
taking statins who are sexually active. Ezetimibe and BAS also have data suggesting they are safe and effective to use in children 10 years or older. At
this time, evolocumab is the only PCSK9 inhibitor with safety and efficacy data in children 13 years or older with HoFH.93 The safety and efficacy of
fibrates and omega3 PUFA have not been established.
Women
The leading cause of death in women is ASCVD and as many women as men die of ASCVD annually. This is mostly due to a longer average life
expectancy and a higher lifetime risk of ischemic stroke than men.94 Age is an important factor when estimating cardiovascular risk in women as most
ASCVD events occur in postmenopausal women. The decline in estrogen levels that occurs during menopause is associated with increased
cardiovascular risk, yet hormone replacement therapy is not recommended as studies have shown it does not reduce cardiovascular risk.95,96 Other
agerelated changes, including increases in blood pressure and LDLC, play a significant role as well in both women and men. Women have been
underrepresented in RCT of lipidlowering therapies; however, a metaanalysis of 27 RCTs found statin therapy is equally effective in men and
women.97 Nonstatin therapies also appear to be equally effective except for fenofibrate, which is associated with an increased ASCVD risk in women
when combined with simvastatin.98 This finding was not, however, observed in another RCT comparing fenofibrate to placebo.99 The clinical
significance of this subgroup finding remains unknown.
Pregnancy is associated with a progressive rise in cholesterol and TG levels, yet dietary therapy is the mainstay of treatment, with emphasis on
maintaining a nutritionally balanced diet as per the needs of pregnancy.100 If the patient is very high risk or has FH, a BAS may be considered during
pregnancy since there is no systemic drug exposure.100 Statins are pregnancy category X and contraindicated due to potential teratogenic effects.
Women of childbearing age who are on statin therapy and are sexually active should use a reliable form of contraception to prevent pregnancy.
Women who plan to become pregnant should discontinue the statin 1 to 2 months before pregnancy is attempted. Ezetimibe and niacin are pregnancy
category C drugs but no data are available in humans. Increased intake of omega3 PUFA, particularly DHA, during pregnancy is important for fetal
brain development; however, prescription omega3 PUFA products are pregnancy category C.101 There is currently no information on the safety of
PCSK9 inhibitors in pregnant women.
Diabetes is a major risk factor for ASCVD and persons with diabetes are at greater risk of morbidity and mortality following an ASCVD event.20 The
dyslipidemia commonly found in persons with diabetes is often characterized by hypertriglyceridemia, low HDLC, and modestly elevated, but dense,
LDLC that are highly atherogenic.102 Despite the modest elevation in LDLC observed in these patients, statins are the firstline therapy given the
Downloaded 2025213 10:53 P Your IP is
significant body of evidence from RCT demonstrating that statins reduce ASCVD events and mortality in persons with diabetes.20 However,Page individual
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche 25 / 37
risk among those with diabetes who have no history of ASCVD is not homogenous, so the 10year
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility ASCVDrisk score may be used to determine the
appropriate statin intensity (see Table 314).20 Highintensity statin therapy is preferred in those with diabetes and a history of ASCVD (secondary
prevention) given these patients are at very high risk of recurrent ASCVD events.
Patients with Diabetes California Northstate University
Access Provided by:
Diabetes is a major risk factor for ASCVD and persons with diabetes are at greater risk of morbidity and mortality following an ASCVD event.20 The
dyslipidemia commonly found in persons with diabetes is often characterized by hypertriglyceridemia, low HDLC, and modestly elevated, but dense,
LDLC that are highly atherogenic.102 Despite the modest elevation in LDLC observed in these patients, statins are the firstline therapy given the
significant body of evidence from RCT demonstrating that statins reduce ASCVD events and mortality in persons with diabetes.20 However, individual
risk among those with diabetes who have no history of ASCVD is not homogenous, so the 10year ASCVDrisk score may be used to determine the
appropriate statin intensity (see Table 314).20 Highintensity statin therapy is preferred in those with diabetes and a history of ASCVD (secondary
prevention) given these patients are at very high risk of recurrent ASCVD events.
The role of nonstatin therapies in persons with diabetes is complex but has become clearer in recent years. In the Improved Reduction of Outcomes:
Vytorin Efficacy International Trial (IMPROVEIT) the benefit of adding ezetimibe to simvastatin was significantly enhanced in those with diabetes
compared to patients without diabetes.103 The addition of evolocumab to background statin therapy in the Further Cardiovascular Outcomes Research
With PCSK9 Inhibition in Subjects With Elevated Risk (FOURIER) trial was equally effective in those with, and without, diabetes.104 Given the mixed
dyslipidemia associated with diabetes, there has been considerable interest in the potential of fibrates to reduce ASCVD risk. However, in the Action to
Control Cardiovascular Risk in Diabetes (ACCORD) the combination of fenofibrate and a statin in patients with type 2 diabetes did not reduce the rate of
fatal cardiovascular events, nonfatal myocardial infarction, or nonfatal stroke compared to simvastatin alone.98 Subgroup analyses from two RCTs
have suggested a potential benefit with fenofibrate in those with TG levels >204 mg/dL (2.31 mmol/L) and HDLC <34 mg/dL (0.88 mmol/L), but this has
not been evaluated in a prospective RCT.98,99 Additionally, fenofibrate appears to reduce the progression of diabetic retinopathy, as well as the need
for laser treatment.105 The BAS colesevelam is FDAapproved to improve both glycemic and lipid control, but it can exacerbate hypertriglyceridemia,
which is commonly observed in those with diabetes.55 Niacin modestly increases fasting plasma glucose (∼4%5%) and HbA1c levels (∼0.25%).86 As
such, niacin should not be routinely used in persons with diabetes especially given the lack of evidence to support its use.
Dyslipidemia is highly prevalent among patients with kidney disease.106 The dyslipidemia pattern in patients with kidney disease includes
hypertriglyceridemia, slightly elevated total cholesterol and LDLC and low HDLC levels.106 These abnormalities are thought to be caused by a
deficiency in apolipoprotein CII, perhaps as a result of sustained use of heparin during hemodialysis and depletion of LPL, carbohydrateinduced
obesity and hypertriglyceridemia, loss of carnitine during hemodialysis, use of acetate buffer (acetate is a precursor to fatty acid synthesis) during
hemodialysis, and decreased LCAT activity during hemodialysis.107 Dialysis does not correct the lipid abnormalities. Renal transplantation may correct
lipid abnormalities in some patients; however, in others, the use of transplantationrelated medications, such as corticosteroids and cyclosporine may
aggravate lipid abnormalities.107
Statins effectively reduce LDLC in patients with kidney disease, yet the cardiovascular event reduction is less robust in patients kidney disease.106
Notably, rosuvastatin failed to prevent cardiovascular events in a RCT of patients undergoing hemodialysis suggesting statins should not be initiated in
this population.108 Statins are generally continued, however, in patients who are on statins before progressing to endstage renal disease and
requiring dialysis.106 Moderateintensity statins are generally preferred in patients with kidney disease to minimize the risk of adverse effects (such as
SAMS).106 Kidney transplant recipients are at considerably high risk of future cardiovascular events and should receive statin therapy; however,
appropriate statin selection is important given the potential for drug–drug interactions with antirejection therapies (such as cyclosporine).106
Ezetimibe may also be used in combination with statin therapy based on RCT evidence showing this combination reduces cardiovascular events
compared to placebo in patients at various stages of advanced kidney disease.109 Current guidelines do not advocate for routine use of other nonstatin
therapies at this time given the paucity of efficacy data and safety concerns.106
It is well established that the chronic inflammation and immune activation seen with chronic inflammatory disorders (eg, rheumatoid arthritis, lupus)
and human immunodeficiency virus (HIV) accelerate the development and progression of atherosclerosis. These nontraditional risk factors are not
included in the ASCVDrisk estimator, but they should be considered when assessing individual ASCVD risk. After a 3 to 6month trial of lifestyle
interventions, these patients should have their 10year ASCVD risk estimated. In those with a 10year ASCVD risk of 5% or greater, it is reasonable to
initiate moderateintensity statin therapy.
In addition to considering statin therapy, there are additional considerations with the treatments used for chronic inflammatory disorders and HIV.
Longterm use2025213
Downloaded of antiretroviral
10:53 therapy,
P Your IPforisexample, has been shown to mediate atherosclerosis progression and development in patients with HIV.
Chapter
With that31: Dyslipidemia,
said, Dave L. (eg,
many antiretrovirals Dixon; Danielinhibitors)
protease Page 26 / 37
M. Riche can significantly increase TG levels. Antiinflammatory therapies (eg, tociluzimab,
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
methotrexate) used in the management of rheumatoid arthritis have produced mixed results in terms of their effects on lipid levels and ASCVD risk.
included in the ASCVDrisk estimator, but they should be considered when assessing individual ASCVD risk. After a 3 to 6month trial of lifestyle
California Northstate University
interventions, these patients should have their 10year ASCVD risk estimated. In those with a 10year ASCVD risk of 5% or greater, it is reasonable to
Access Provided by:
initiate moderateintensity statin therapy.
In addition to considering statin therapy, there are additional considerations with the treatments used for chronic inflammatory disorders and HIV.
Longterm use of antiretroviral therapy, for example, has been shown to mediate atherosclerosis progression and development in patients with HIV.
With that said, many antiretrovirals (eg, protease inhibitors) can significantly increase TG levels. Antiinflammatory therapies (eg, tociluzimab,
methotrexate) used in the management of rheumatoid arthritis have produced mixed results in terms of their effects on lipid levels and ASCVD risk.
Collect
Patients history: Past medical (eg, HTN), family (eg, earlyonset coronary heart disease), social
Current medications (including overthecounter [OTC]) and prior lipidlowering medication use
Socioeconomic factors that may affect access to treatment or other aspects of care
Objective data
Labs (eg, AST/ALT, urinalysis, TSH, glucose, Serum Creatinine, and BUN at baseline)
Assess
Potential secondary causes (eg, diabetes mellitus, alcohol abuse, renal dysfunction, liver disease, druginduced, thyroid disorder)
Downloaded
Special 2025213 10:53 patient
needs of specific P Yourpopulations
IP is such as children/adolescents, pregnant or menopausal women, older adults, ethnic/racial
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 27 / 37
groups, or highrisk conditions/residual risks (eg, patients with rheumatoid arthritis or residual risk despite statin and lifestyle therapy)
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
Presence of highrisk comorbid conditions: diabetes mellitus, peripheral arterial disease, coronary artery disease, chronic kidney disease,
carotid artery stenosis, abdominal aortic aneurysm
Labs (eg, AST/ALT, urinalysis, TSH, glucose, Serum Creatinine, and BUN at baseline)
California Northstate University
Access Provided by:
Assess
Potential secondary causes (eg, diabetes mellitus, alcohol abuse, renal dysfunction, liver disease, druginduced, thyroid disorder)
Special needs of specific patient populations such as children/adolescents, pregnant or menopausal women, older adults, ethnic/racial
groups, or highrisk conditions/residual risks (eg, patients with rheumatoid arthritis or residual risk despite statin and lifestyle therapy)
Presence of highrisk comorbid conditions: diabetes mellitus, peripheral arterial disease, coronary artery disease, chronic kidney disease,
carotid artery stenosis, abdominal aortic aneurysm
LDLC reduction based on statin benefit group, if applicable (see Table 314)
Plan*
Drug therapy regimen including specific lipidlowering medication, dose, route, frequency, and duration; specify the continuation and
discontinuation of existing therapies (see Table 314)
Monitoring parameters including efficacy (eg, lipid panel, cardiovascular events), safety (medicationspecific adverse effects), and time frame
(3month initial followup intervals, followed by 612 month intervals once at goal)
Patient education (eg, purpose of treatment, dietary and lifestyle modification, drug therapy)
Referrals to other providers when appropriate for coordination of care (eg, physician, dietician)
Implement*
Provide patient education regarding all elements of the treatment plan, including selfmanagement training
should be noted that although total cholesterol (TC) HDLC, and TG levels are directly measured, LDLC is typically estimated using the Friedewald
equation, LDLC = TC – HDLC – (TG/5) (or LDLC = TC – HDLC – (TG/2.2) when lipid levels are all expressed in mmol/L), which does not provide an
California Northstate University
EVALUATION OF THERAPEUTIC OUTCOMES Access Provided by:
Shortterm evaluation of therapy for dyslipidemia is based on a complete lipid panel obtained 4 to 12 weeks after initiation or following a dose
adjustment of lipidlowering therapy to evaluate therapeutic response.20 This is especially important with statin therapy given there are numerous
pharmacokinetic and pharmacodynamic differences among statins that give rise to variable response to therapy.110 Longterm evaluation is based on
a repeat lipid panel obtained every 3 to 12 months to ensure adherence to lipidlowering therapy and maintenance of desired levels of LDLC.20 It
should be noted that although total cholesterol (TC) HDLC, and TG levels are directly measured, LDLC is typically estimated using the Friedewald
equation, LDLC = TC – HDLC – (TG/5) (or LDLC = TC – HDLC – (TG/2.2) when lipid levels are all expressed in mmol/L), which does not provide an
accurate estimate of VLDLC.111 As such, the Friedewald equation can underestimate LDLC in patients with high TG levels as well as those with very low
LDLC levels. Given VLDLC concentrations are typically small in comparison to LDLC, the inaccuracy of VLDLC has previously been accepted. However,
given the increased prevalence of obesity, metabolic syndrome, and diabetes, more patients have elevated levels of VLDLC. Useful alternatives in these
patients include nonHDLC (TC minus HDLC) and direct LDLC measurements, which are more accurate than estimated LDLC using the Friedewald
equation.111 A nonfasting lipid panel is generally acceptable, except in patients with hypertriglyceridemia, where a fasting lipid panel is preferred to
minimize interference from chylomicrons.112 Routine safety monitoring of hepatic function and CK levels is not recommended in statintreated
patients, but these may be obtained if the patient has signs or symptoms suggestive of liver or muscle injury.20 Patients taking niacin, on the other
hand, should have hepatic function tests performed at baseline, after each dosage increase, and every 6 months thereafter while taking a stable
dose.20 Periodic monitoring of A1c is warranted in persons with diabetes receiving niacin and patients treated with statins who are at high risk for
developing diabetes.20
In patients treated with lipidlowering therapy for secondary prevention, symptoms such as angina or intermittent claudication may improve over
months to years. If patients have xanthomas or other external manifestations of dyslipidemia, these lesions should regress with therapy.50 Modifiable
risk factors such as hypertension, smoking, exercise and weight control, and glycemic control in persons with diabetes should also be monitored and
evaluated.20,28 Dietary therapy is an important part of treating dyslipidemia and a dietitian should be consulted to perform an initial evaluation with
periodic followup thereafter if the goals of therapy are not achieved.25 Use of food diaries and recall surveys enable the collection of information
about diet in a systematic manner and may improve patient adherence to dietary recommendations.
POSTCLASS ACTIVITY
Watch the video entitled “Strategies for Optimizing Statin Therapy: Focus on Drug Interactions” from Pharmacy Times by Mary Bridgeman. This 14
minute video provides a detailed overview of potential drug–drug interactions that can occur with statin therapy through the CYP3A4 pathway.
Create a table of the strong CYP3A4 inhibitors and inducers, the effect each would have on a patient’s statin concentrations, and what potential side
effects could occur from those interactions. This information will help students be prepared to participate in the PLAN and FOLLOWUP steps in the
patient care process.
ABBREVIATIONS
Apo apolipoprotein
CE cholesterol ester
HTN hypertension
OTC overthecounter
TC total cholesterol
California Northstate University
PUFA polyunsaturated fatty acid
Access Provided by:
TC total cholesterol
TG triglyceride
UA urinalysis
REFERENCES
1. Rosenson R. Lipoprotein classification, metabolism, and role in atherosclerosis. UpToDate . May 5, 2017.
2. Wiggins BS, Saseen JJ. Pharmacist’s Guide to Lipid Management. 2nd. Lenexa, KS: American College of Clinical Pharmacy; 2014.
3. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive Summary of the Third Report of the National
Cholesterol Education Program (NCEP). (Adult Treatment Panel III). JAMA. 2001;285:2486–2497. [PubMed: 11368702]
4. Grundy SM, Cleeman JI, Merz CN, et al. Implications of recent clinical trials for the national cholesterol education program adult treatment panel III
guidelines. Circulation. 2004;110:227–239. [PubMed: 15249516]
5. Smith SC Jr, Allen J, Blair SN, et al. AHA/ACC guidelines for secondary prevention for patients with coronary and other atherosclerotic vascular
disease: 2006 update: Endorsed by the National Heart, Lung, and Blood Institute. Circulation. 2006;113:2363–2372. [PubMed: 16702489]
6. Mosca L, Banka CL, Benjamin EJ, et al. Evidencebased guidelines for cardiovascular disease prevention in women: 2007 Update. Circulation.
2007;115:1481–1501. [PubMed: 17309915]
7. Fletcher B, Berra K, Ades P, et al. Managing abnormal blood lipids: A collaborative approach. Circulation. 2005;112:3184–3209. [PubMed:
16286609]
8. Oh RC, Lanier JB. Management of hypertriglyceridemia. Am Fam Physician. 2007;75:1365–1371. [PubMed: 17508532]
9. Yuan G, AlShali KZ, Hegele RA. Hypertriglyceridemia: Its etiology, effects, and treatment. CMAJ Can Med Assoc J. 2007;176:1113–1120.
10. Mckenney JM, Sica D. Prescription Omega3 fatty acids for the treatment of hypertriglyceridemia. Am J Health Syst Pharm. 2007;64:595–605.
[PubMed: 17353568]
11. Mckenney J. New perspectives on the use of niacin in the treatment of lipid disorders. Arch Intern Med. 2004;164:697–705. [PubMed: 15078639]
12. Colhoun HM, Betteridge DJ, Durrington PN, et al. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the
Collaborative Atorvastatin Diabetes Study (CARDS): Multicentre randomised placebocontrolled trial. Lancet. 2004;364:685–696. [PubMed: 15325833]
13. Collins R, Armitage J, Parish S, et al. Heart protection study collaborative G. effects of cholesterollowering with simvastatin on stroke and other
major vascular events in 20536 people with cerebrovascular disease or other highrisk conditions. Lancet. 2004;363:757–767. [PubMed: 15016485]
Downloaded 2025213
14. Ford ES, Mokdad AH,10:53
Giles P Your
WH, IP is GA. Serum total cholesterol concentrations and awareness, treatment, and control of
Mensah
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 31 / 37
hypercholesterolemia among US adults: Findings from the National Health and Nutrition Examination Survey, 1999 to 2000. Circulation.
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
2003;107(17):2185–2189. [PubMed: 12719276]
12. Colhoun HM, Betteridge DJ, Durrington PN, et al. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the
Collaborative Atorvastatin Diabetes Study (CARDS): Multicentre randomised placebocontrolled trial. Lancet. 2004;364:685–696.
California [PubMed:
Northstate15325833]
University
Access Provided by:
13. Collins R, Armitage J, Parish S, et al. Heart protection study collaborative G. effects of cholesterollowering with simvastatin on stroke and other
major vascular events in 20536 people with cerebrovascular disease or other highrisk conditions. Lancet. 2004;363:757–767. [PubMed: 15016485]
14. Ford ES, Mokdad AH, Giles WH, Mensah GA. Serum total cholesterol concentrations and awareness, treatment, and control of
hypercholesterolemia among US adults: Findings from the National Health and Nutrition Examination Survey, 1999 to 2000. Circulation.
2003;107(17):2185–2189. [PubMed: 12719276]
15. Foley Ka, Denke MA, KamaBahl S, et al. The impact of physician attitudes and beliefs on treatment decisions: Lipid therapy in highrisk patients.
Med Care. 2006;44:421–428. [PubMed: 16641660]
16. Arnett DK, Jacobs RD Jr, Luepker RV, et al. Twentyyear trends in serum cholesterol, hypercholesterolemia, and cholesterol medication use: The
minnesota heart survey, 19801982 to 20002002. Circulation. 2005;112:3884–3891. [PubMed: 16344385]
17. Mozaffarian D, Benjamin EJ, Go AS, et al. Executive summary: Heart disease and stroke statistics—2015 update: A report from the American Heart
Association. Circulation. 2015;131:434–441.
18. Menotti A, Lanti M, Nedeljkovic S, et al. The relationship of age, blood pressure, serum cholesterol, and smoking habits with the risk of typical and
atypical coronary heart disease death in the European Cohorts of seven countries study. Int J Cardiol. 2006;106:157–163. [PubMed: 16321686]
19. Stone NJ. Secondary causes of hyperlipidemia. Med Clin North Am. 1994;78:117–141. [PubMed: 8283927]
20. 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. Nov 2018;1097:S1. doi: 10.1016/j.jacc.2018.11.003.
21. Yuan F, Wang J, Hegele RA. Heterozygous familial hypercholesterolemia: an underrecognized cause of early cardiovascular disease. CMAJ.
2006;174:1124–1129. [PubMed: 16606962]
22. Hasudungan, Armando. “Physiology of Lipoproteins Cholesterol.” YouTube . June 1, 2015. Available at: https://www.youtube.com/watch?
v=PkKH8lTxvzA.
23. “Physiology of Lipoprotein Metabolism.” YouTube. August 17, 2009. Available at: https://www.youtube.com/watch?v=97uiV4RiSAY.
24. Libby P. How our growing understanding of inflammation has reshaped the way we think of disease and drug development. Clin Pharmacol Ther.
2010;87:389–391. [PubMed: 20305670]
25. Eckel RH, Jakicic JM, Ard JD, et al. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk. J Am Coll Cardiol.
2014;53(25):2960–2984.
26. Whelton PK, Carey RM, Aronow WS. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASPC/NMA/PCNA guideline for the prevention, detection,
evaluation, and management of high blood pressure in adults. J Am Coll Cardiol. 71(19):e127–e248. doi: 10.1016/j.jacc.2017.11.006.
27. American Diabetes Association. 2018 Standards of Medical Care in Diabetes. Diabetes Care . 2018.
28. Goff DC, LloydJones DM, Bennett G, et al. 2013 ACC/AHA Guideline on the assessment of cardiovascular risk: A report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63(25):2935–2959. [PubMed: 24239921]
29. Navarese EP, Robinson JG, Kowalewski M, et al. Association between baseline LDLC level and total and cardiovascular mortality after LDLC
lowering: A systematic review and metaanalysis. JAMA. 2018;319(15):1566–1579. [PubMed: 29677301]
30. LloydJones DM, Morris PB, Ballantyne CM et al. 2017 Focused update on the 2016 ACC expert consensus decision pathway on the role of non
statin therapies for LDLCholesterol lowering in the management of atherosclerotic cardiovascular disease risk. J Am Coll Cardiol. 2017;70(14):1785–
1822. [PubMed: 28886926]
31. Cholesterol
Downloaded Treatment
2025213 Trialists
10:53 Collaboration.
P Your IP is Efficacy and safety of more intensive lowering of LDL cholesterol: A metaanalysis of data from
170,000 participants
Chapter in 26 randomised
31: Dyslipidemia, trials.
Dave L. Dixon; Lancet.
Daniel M.2010;376:1670–1681.
Riche [PubMed: 21067804] Page 32 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
32. Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol
levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study. JAMA. 1998;279(20):1615–1622. [PubMed: 9613910]
CaliforniaonNorthstate
30. LloydJones DM, Morris PB, Ballantyne CM et al. 2017 Focused update on the 2016 ACC expert consensus decision pathway the role ofUniversity
non
Access Provided by:
statin therapies for LDLCholesterol lowering in the management of atherosclerotic cardiovascular disease risk. J Am Coll Cardiol. 2017;70(14):1785–
1822. [PubMed: 28886926]
31. Cholesterol Treatment Trialists Collaboration. Efficacy and safety of more intensive lowering of LDL cholesterol: A metaanalysis of data from
170,000 participants in 26 randomised trials. Lancet. 2010;376:1670–1681. [PubMed: 21067804]
32. Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol
levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study. JAMA. 1998;279(20):1615–1622. [PubMed: 9613910]
33. Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med.
2017;376(18):1713–1722. [PubMed: 28304224]
34. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 highrisk individuals: a randomised placebocontrolled trial.
Lancet. 2002;360(9326):7–22.
[PubMed: 12114036] .
35. Cannon CP, Blazing mA, Giugliano RP et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015; 372:2387–
2397. [PubMed: 26039521]
36. Ridker PM, Danielson E, Fonseca FAH, et al. Rosuvastatin to prevent vascular events in men and women with elevated Creactive protein. N Engl J
Med. 2008;359(21):2195–2207. [PubMed: 18997196]
37. LaRosa JC, Grundy SM, Waters DD, et al. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med.
2005;352(14):1425–1435. [PubMed: 15755765]
38. Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland
Coronary Prevention Study Group. N Engl J Med. 1995;333(20):1301–1307. [PubMed: 7566020]
39. U.S. Department of Health and Human Services and U.S. Department of Agriculture. 20152020 Dietary Guidelines for Americans . 8th ed. December
2015. Available at: http://health.gov/dietaryguidelines/2015/guidelines.
40. Hoy MK, Goldman JD. Fiber intake of the U.S. population: What we eat in America, NHANGES 20092010. Food Surveys Research Group Dietary Data
Brief No. 12. September 2014.
41. Mozaffarian D, Rimm EB. Fish intake, contaminants and human health: Evaluating the risks and benefits. JAMA. 2006;296(15):1885–1899.
[PubMed: 17047219]
42. Aung T, Halsey J, Kromhout D, et al. Associations of Omega3 fatty acid supplement use with cardiovascular disease risks: Metaanalysis of 10
trials involving 77,917 individuals. JAMA Cardiol. 2018;3(3):225–234. [PubMed: 29387889]
43. Siscovick DS, Barringer TA, Fretts AM, et al. Omega3 polyunsaturated fatty acid (Fish Oil) supplementation and the prevention of clinical
cardiovascular disease: A science advisory from the american heart association. Circulation. 2017;135:e867–e884. [PubMed: 28289069]
44. Bowman L, Mafham M, Wallendszus K, et al. ASCEND Study Collaborative Group. Effects of n3 fatty acid supplements in diabetes mellitus. N Engl J
Med. 2018;379(16):1540–1550. [PubMed: 30146932]
45. Yoo EG. Sitosterolemia: A review and update of pathophysiology, clinical spectrum, diagnosis, and management. Ann Pediatr Endocrinol Metab.
2016;21(1):7–14. [PubMed: 27104173]
46. Cohen PA, Avula B, Khan IA. Variability in strength of red yeast rice supplements purchased from mainstream retailers. Eur J Prev Cardiol.
2017;24(13):1431–1434. [PubMed: 28641460]
47. Dujovne CA. Red yeast rice preparations: Are they suitable substitutions for statins? Am J Med. 2017;130(10):1148–1150. [PubMed: 28601545]
48. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterollowering treatment: Prospective metaanalysis of data from 90,056
Downloaded 2025213
participants in 10:53trials
14 randomized P Your IP is Lancet. 2005;366:1267–1278. [PubMed: 16214597]
of statins.
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 33 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
49. Ference BA, Ginsberg HN, Graham I, et al. Lowdensity lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic,
epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur H J. 2017;38(32):2459–
46. Cohen PA, Avula B, Khan IA. Variability in strength of red yeast rice supplements purchased from mainstream retailers. Eur J Prev Cardiol.
California Northstate University
2017;24(13):1431–1434. [PubMed: 28641460]
Access Provided by:
47. Dujovne CA. Red yeast rice preparations: Are they suitable substitutions for statins? Am J Med. 2017;130(10):1148–1150. [PubMed: 28601545]
48. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterollowering treatment: Prospective metaanalysis of data from 90,056
participants in 14 randomized trials of statins. Lancet. 2005;366:1267–1278. [PubMed: 16214597]
49. Ference BA, Ginsberg HN, Graham I, et al. Lowdensity lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic,
epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur H J. 2017;38(32):2459–
2472.
50. Robinson JG, Goldberg AC. Treatment of adults with Familial Hypercholesterolemia and evidence for treatment: Recommendations from the
National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011;5:S18–S29. [PubMed: 21600526]
51. Hopkins PN, Toth PP, Ballantyne CM. Rader DJ national lipid association expert panel on familial hypercholesterolemia. Familial
hypercholesterolemias: Prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial
Hypercholesterolemia. J Clin Lipidol. 2011;5(3 Suppl):S9–S17. [PubMed: 21600530]
52. Dixon DL, Sisson EM, Butler M, et al. Lomitapide and Mipomersen: Novel lipidlowering therapies for the management of familial
hypercholesterolemia. J Cardiovasc Nurs. 2014;29(5):E7–E12. [PubMed: 24231894]
53. Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: A Scientific Statement from the American Heart Association.
Circulation. 2011;123(20):2292–2333. [PubMed: 21502576]
54. Jacobson TA, Ito MK, Maki KC, et al. National lipid association recommendations for patientcentered management of dyslipidemia: Part 1—full
report. J Clin Lipidol. 2015;9:129–169. [PubMed: 25911072]
55. Kelly MS, Beavers C, Bucheit JD, et al. Pharmacologic approaches for the management of patients with moderately elevated triglycerides (150–499
mg/dL). J Clin Lipidol. 2017;11:872–879. [PubMed: 28669686]
56. Toth PP, Barter PJ, Rosenson RS, et al. Highdensity lipoproteins: A consensus statement from the National Lipid Association. J Clin Lipidol.
2013;7(5):484–525. [PubMed: 24079290]
57. Landray MJ, Haynes R, Hopewell JC, et al. HPS2THRIVE Collaborative Group. Effects of extendedrelease niacin with laropiprant in highrisk
patients. N Engl J Med. 2014;371(3):203–212. [PubMed: 25014686]
58. Boden WE, Probstfield JL, Anderson T, et al. AIMHIGH Investigators. Niacin in patients with low HDL cholesterol levels receiving intensive statin
therapy. N Engl J Med. 2012;365(24):2255–2267.
59. Schachter M. Chemical, pharmacokinetics and pharmacodynamic properties of statins: An update. Fundam Clin Pharmacol. 2005;19(1):117–125.
[PubMed: 15660968]
60. Tobert JA, Newman CB. Statin tolerability: In defense of placebocontrolled trials. Eur J Prev Cardiol. 2016;23:891–896. [PubMed: 26318980]
61. Thompson PD, Panza G, Zaleski A, Taylor B. StatinAssociated side effects. J Am Coll Cardiol. 2016;67(20):2396–2410.
62. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic
variation. Pharmacol Ther. 2013;138(1):103–141. [PubMed: 23333322]
63. Wiggins BS, Saseen JJ, Page RL, et al. Recommendations for management of clinically significant drugdrug interactions with statins and select
agents used in patients with cardiovascular disease. Circulation. 2016;134(21):e468–e495. [PubMed: 27754879]
64. Rosenson RS, Baker SK, Jacobson TA, et al. An assessment by the statin muscle safety task force: 2014 Update. J Clin Lipidol. 2014;8(3):S58–S71.
[PubMed: 24793443]
65. Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes with intensivedose compared with moderatedose statin therapy: A metaanalysis.
Downloaded 2025213 10:53
JAMA. 2011;305:2556–2564. P Your 21693744]
[PubMed: IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 34 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
66. Sattar N, Preiss D, Murray HM, et al. Statins and risk of incident diabetes: A collaborative metaanalysis of randomised statin trials. Lancet.
2010;375:735–742. [PubMed: 20167359]
agents used in patients with cardiovascular disease. Circulation. 2016;134(21):e468–e495. [PubMed: 27754879]
California Northstate University
Access Provided by:
64. Rosenson RS, Baker SK, Jacobson TA, et al. An assessment by the statin muscle safety task force: 2014 Update. J Clin Lipidol. 2014;8(3):S58–S71.
[PubMed: 24793443]
65. Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes with intensivedose compared with moderatedose statin therapy: A metaanalysis.
JAMA. 2011;305:2556–2564. [PubMed: 21693744]
66. Sattar N, Preiss D, Murray HM, et al. Statins and risk of incident diabetes: A collaborative metaanalysis of randomised statin trials. Lancet.
2010;375:735–742. [PubMed: 20167359]
67. Ridker PM, Pradhan A, MacFadyen JG, et al. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: An analysis from
the JUPITER trial. Lancet. 2012;380(9841):565–571. [PubMed: 22883507]
68. Ezetimibe (Zetia®) [package insert]. Whitehouse Station, NJ: Merck & Co., Inc; 2007.
69. The Myocardial Infarction Genetics Consortium Investigators. Inactivating mutations in NPC1L1 and protection from coronary heart disease. N Engl
J Med. 2014;371:2072–2082. [PubMed: 25390462]
70. The lipid research clinics coronary primary prevention trial results. I. Reduction in incidence of coronary heart disease. JAMA. 1984;251(3):351–
364. [PubMed: 6361299]
71. Brunetti L, DeSantis EH. Patient tolerance and acceptance of colesevelam hydrochloride: Focus on type2 diabetes mellitus. Pharmacy and
Therapeutics. 2015;40(1):62–67.
72. Fonseca VA, Handelsman Y, Staels B. Coleseveal lowers glucose and lipid levels in type 2 diabetes: The clinical evidence. Diabetes Obes Metab.
2010;12(5):384–392. [PubMed: 20415686]
73. Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med. November 29 ,
2018;379(22):2097–2107. [PubMed: 30403574]
74. Giugliano RP, Mach F, Zavitz K, et al. Cognitive function in a randomized trial of evolocumab. N Engl J Med. 2017;377:633–643. [PubMed:
28813214]
75. Giugliano RP, Pedersen TR, Park JG, et al. Clinical efficacy and safety of achieving very low LDLcholesterol concentrations with the PCSK9
inhibitor evolocumab: A prespecified secondary analysis of the FOURIER trial. Lancet. 2017;390:1692–1671.
76. Ray KK, Wright RS, Kallend D et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N Engl J Med 2020;382(16):1507–1519.
10.1056/NEJMoa1912387
[PubMed: 32187462] .
78. Ballantyne CM, Banach M, Mancini GBJ Efficacy and safety of bempedoic acid added to ezetimibe in statinintolerant patients with
hypercholesterolemia: A randomized, placebocontrolled study. Atherosclerosis 2018;277:195–203. 10.1016/j.atherosclerosis.2018.06.002
[PubMed: 29910030] .
79. Fenofibrate (Tricor®) [package insert]. North Chicago, IL: Abbott Laboratories; 2010.
80. Mychaleckyj JC, Craven T, Nayak U, et al. Reversibility of fenofibrate therapyinduced renal function impairment in ACCORD type 2 diabetic
participants. Diabetes Care. 2012;35(5):1008–1014. [PubMed: 22432114]
81. CaroliBosc FX, Le Gall P, Pugliese P, et al. Role of fibrates and HMGCoA reductase inhibitors in gallstone formation. Digestive Diseases and
Sciences. 2001;46(3):540–544. [PubMed: 11318529]
82. Dixon DL, Williams VG. Interaction between gemfibrozil and warfarin: Case report and review of the literature. Pharmacotherapy. 2009;29(6):744–
Downloaded
748. [PubMed:2025213
19476425]10:53 P Your IP is
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 35 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
83. Bradberry JC, Hilleman DE. Overview of Omega3 fatty acid therapies. Pharmacy and Therapeutics. 2013;38(11):681–691.
participants. Diabetes Care. 2012;35(5):1008–1014. [PubMed: 22432114]
California Northstate University
Access Provided by:
81. CaroliBosc FX, Le Gall P, Pugliese P, et al. Role of fibrates and HMGCoA reductase inhibitors in gallstone formation. Digestive Diseases and
Sciences. 2001;46(3):540–544. [PubMed: 11318529]
82. Dixon DL, Williams VG. Interaction between gemfibrozil and warfarin: Case report and review of the literature. Pharmacotherapy. 2009;29(6):744–
748. [PubMed: 19476425]
83. Bradberry JC, Hilleman DE. Overview of Omega3 fatty acid therapies. Pharmacy and Therapeutics. 2013;38(11):681–691.
84. Bhatt DL, Steg PG, Miller M, et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med. 3 , 2019;380(1):11–22.
doi: 10.1056/NEJMoa1812792.
85. Jacobson TA. A “Hot” topic in dyslipidemia management—”How to Beat a Flush”: Optimizing niacin tolerability to promote longterm treatment
adherence and coronary disease prevention. Mayo Clin Proc. 2010;85(4):365–379. [PubMed: 20360295]
86. Goldberg RB, Jacobson TA. Effects of niacin on glucose control in patients with dyslipidemia. Mayo Clin Proc. 2008 Apr;83(4):470–478. [PubMed:
18380993]
87. Salem CB, Slim R, Fathallah N, Hmouda H. Druginduced hyperuricemia and gout. Rheumatology. 2017;56(5):679–688.
88. Dixon DL, Donohoe KL, Ogbonna KC, Barden SM. Current drug treatment of hyperlipidemia in older adults. Drugs Aging. 2015;32:127–138.
[PubMed: 25637391]
89. Swiger KJ, Manalac RJ, Blumenthal RS, et al. Statins and cognition: A systematic review and metaanalysis of short and longterm cognitive
effects. Mayo Clin Proc. 2013;88(11):1213–1221. [PubMed: 24095248]
90. Ott BR, Daiella LA, Dahabreh IJ, et al. Do statins impair cognition? A systematic review and metaanalysis of randomized controlled trials J Gen
Intern Med. 2015;30(3):348–358. [PubMed: 25575908]
91. Yu S, Chu Y, Li G, et al. Statin use and the risk of cataracts: A systematic review and metaanalysis. J Am Heart Assoc. 2017;6(3):pii: e004180. doi:
10.1161/JAHA.116.004180.
92. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents, National Heart, Lung, and Blood
Institute. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: Summary report. Pediatrics.
2011;128(suppl 5):S213–S256. [PubMed: 22084329]
93. Evolocumab (Repatha®) [package insert]. Thousand Oaks, CA: Amgen; 2015.
94. Benjamin EJ, Virani SS, Callaway CW, et al. Heart disease and stroke statistics2018 update: A report from the American Heart Association.
Circulation. 2018;137(12):e67–e492. [PubMed: 29386200]
95. Manson JE, Hsia J, Johnson KC, et al. Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med. 2003;349:523–534. [PubMed:
12904517]
96. Hulley S, Grady D, Bush T, et al. For the heart and estrogen/progestin replacement study (HERS) research group. Randomized trial of estrogen
plus progestin for secondary prevention of coronary heart disease in postmenopausal women. JAMA. 1998;280:605–613. [PubMed: 9718051]
97. Cholesterol Treatment Trialists Collaboration. Efficacy and safety of LDLlowering therapy among men and women: Metaanalysis of individual data
from 174 000 participants in 27 randomised trials. Lancet. 2015;385:1397–1405. [PubMed: 25579834]
98. Ginsberg HN, Elam MB, Lovato LC, et al. Effects of combination lipid therapy in Type 2 diabetes mellitus. N Engl J Med. 2010;362:1563–1574.
[PubMed: 20228404]
99. Keech A, Simes RJ, Barter P, et al. Effects of longterm fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus
(the FIELD study): Randomised controlled trial. Lancet. 2005;366(9500):1849–1861. [PubMed: 16310551]
Downloaded 2025213
100. Wild R, Weedin 10:53 PD.Your
EA, Wilson IP is
Dyslipidemia in pregnancy. Cardiol Clin. 2015;33:209–215. [PubMed: 25939294]
Chapter 31: Dyslipidemia, Dave L. Dixon; Daniel M. Riche Page 36 / 37
©2025 McGraw Hill. All Rights Reserved. Terms of Use • Privacy Policy • Notice • Accessibility
101. Coletta JM, Bell SJ, Roman AS. Omega3 fatty acids and pregnancy. Rev Obstet Gynecol. 2010;3(4):163–171. [PubMed: 21364848]
102. Goldberg IJ. Diabetic dyslipidemia: Causes and consequences. J Clin Endocrinol Metab. 2001;86(3):965–971. [PubMed: 11238470]
98. Ginsberg HN, Elam MB, Lovato LC, et al. Effects of combination lipid therapy in Type 2 diabetes mellitus. N Engl J Med. 2010;362:1563–1574.
[PubMed: 20228404] California Northstate University
Access Provided by:
99. Keech A, Simes RJ, Barter P, et al. Effects of longterm fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus
(the FIELD study): Randomised controlled trial. Lancet. 2005;366(9500):1849–1861. [PubMed: 16310551]
100. Wild R, Weedin EA, Wilson D. Dyslipidemia in pregnancy. Cardiol Clin. 2015;33:209–215. [PubMed: 25939294]
101. Coletta JM, Bell SJ, Roman AS. Omega3 fatty acids and pregnancy. Rev Obstet Gynecol. 2010;3(4):163–171. [PubMed: 21364848]
102. Goldberg IJ. Diabetic dyslipidemia: Causes and consequences. J Clin Endocrinol Metab. 2001;86(3):965–971. [PubMed: 11238470]
103. Giugliano RP, Cannon CP, Blazing MA, et al. Benefit of adding ezetimibe to statin therapy on cardiovascular outcomes and safety in patients with
versus without diabetes mellitus: Results from IMPROVEIT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation.
2018;137(151):1571–1582. [PubMed: 29263150]
104. Sabatine MS, Leiter LA, Wiviott SD, et al. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without
diabetes and the effect of evolocumab on glycaemia and risk of newonset diabetes: A prespecified analysis of the FOURIER randomised controlled
trial. Lancet Diabetes Endocrinol. 2017;5(12):941–950. [PubMed: 28927706]
105. Chew EY, Ambrosius WT, Davis MD, et al. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med.
2010;363(3):233–244. [PubMed: 20587587]
106. Kidney Disease: Improving Global Outcomes (KDIGO) Lipid Work Group. Clinical practice guidelines for lipid management in CKD: summary of
recommendation statements and clinical approach to the patient. Kindey Int Suppl. 2013;3(3):259–305.
107. Vaziri ND. Dyslipidemia of chronic renal failure: The nature, mechanisms, and potential consequences. Am J Physiol Renal Physiol.
2006;290(2):F262–F272. [PubMed: 16403839]
108. Fellstrom BC, Jardine AG, Schmieder RE, et al. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med.
2009;360:1395–1407. [PubMed: 19332456]
109. Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney
disease (Study of heart and renal protection): A randomized placebocontrolled trial. Lancet. 2011;377(9784):2181–2192. [PubMed: 21663949]
110. Karlson BW, Wiklund O, Palmer MK, et al. Variability of lowdensity lipoprotein cholesterol response with different doses of atorvastatin,
rosuvastatin, and simvastatin: Results from VOYAGER. Eur Heart J Cardiovasc Pharmacother. 2016;2(4):212–217. [PubMed: 27533947]
111. Martin SS, Blaha MJ, Elshazly MB, et al. Friedewaldestimated versus directly measured lowdensity lipoprotein cholesterol and treatment
implications. J Am Coll Cardiol. 2013;62(8):732–739. [PubMed: 23524048]
112. Nordestgaard BG, Langsted A, Mora S, et al. Fasting is not routinely required for determination of a lipid profile: Clinical and laboratory
implications including flagging at desirable concentration cutpointsa joint consensus statement from the European Atherosclerosis Society and
European Federation of Clinical Chemistry and Laboratory Medicine. Eur Heart J. 2016;37(25):1944–1958. [PubMed: 27122601]