You are on page 1of 29

AACE/ACE Consensus Statement

CONSENSUS STATEMENT BY THE AMERICAN ASSOCIATION OF


CLINICAL ENDOCRINOLOGISTS AND AMERICAN COLLEGE OF
ENDOCRINOLOGY ON THE MANAGEMENT OF DYSLIPIDEMIA AND
PREVENTION OF CARDIOVASCULAR DISEASE ALGORITHM –
2020 EXECUTIVE SUMMARY

Yehuda Handelsman, MD, Chair1; Paul S. Jellinger, MD, Co-Chair2; Chris K. Guerin, MD, Co-Chair3;
Zachary T. Bloomgarden, MD4; Eliot A. Brinton, MD5; Matthew J. Budoff, MD6;
Michael H. Davidson, MD7; Daniel Einhorn, MD8; Sergio Fazio, MD9; Vivian A. Fonseca, MD10;
Alan J. Garber, MD, PhD11; George Grunberger, MD12; Ronald M. Krauss, MD13;
Jeffrey I. Mechanick, MD14; Paul D. Rosenblit, MD, PhD15; Donald A. Smith, MD, MPH16;
Kathleen L. Wyne, MD, PhD17

This document represents the official position of the American Association of Clinical Endocrinologists and American
College of Endocrinology. Where there were no randomized controlled trials or specific United States Food and Drug
Administration (FDA) labeling for issues in clinical practice, the participating clinical experts utilized their judgment and
experience. Every effort was made to achieve consensus among the committee members. Position statements are meant to
provide guidance, but they are not to be considered prescriptive for any individual patient and cannot replace the judgment
of a clinician.

Submitted for publication July 18, 2020 American Association of Clinical Endocrinologists, Bloomfield Hills,
Accepted for publication August 10, 2020 Michigan, 13Professor of Pediatrics and Medicine, UCSF, Adjunct Professor,
From the 1Medical Director & Principal Investigator, Metabolic Institute of Department of Nutritional Sciences, University of California, Berkeley,
America, Tarzana, California, 2Professor of Clinical Medicine, Voluntary Dolores Jordan Endowed Chair, UCSF Benioff Children’s Hospital Oakland,
14Professor of Medicine, Medical Director, The Marie-Josee and Henry R.
Faculty, University of Miami Miller School of Medicine, Center for Diabetes
& Endocrine Care, Hollywood, Florida, 3Clinical Assistant Professor of Kravis Center for Clinical Cardiovascular Health at Mount Sinai Heart,
Medicine, Voluntary Faculty, University of California San Diego, San Diego, Director, Metabolic Support, Divisions of Cardiology and Endocrinology,
California, 4Editor, the Journal of Diabetes, Clinical Professor, Icahn School Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai Heart,
of Medicine at Mount Sinai, New York, New York, 5President, Utah Lipid Director, Metabolic Support, Divisions of Cardiology and Endocrinology,
Center, Salt Lake City, Utah, Past President, American Board of Clinical Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New
Lipidology, 6Professor of Medicine, UCLA Endowed Chair of Preventive York, New York, 15Clinical Professor, Medicine (Division of Endocrinology,
Cardiology, Los Angeles Biomedical Research Institute, Torrance, California, Diabetes, Metabolism), University California, Irvine, School of Medicine,
7Professor, Director of the Lipid Clinic, University of Chicago Pritzker Irvine, California, Co-Director, Diabetes Out-Patient Clinic, UCI Medical
School of Medicine, Chicago, Illinois, 8Associate Editor, the Journal of Center, Orange, California, Director & Site Principal Investigator, Diabetes/
Diabetes, Medical Director, Scripps Whittier Diabetes Institute, Clinical Lipid Management & Research Center, Huntington Beach, California,
16Endocrinologist, Clinical Lipidologist, Associate Professor of Medicine,
Professor of Medicine, UCSD, President, Diabetes and Endocrine Associates,
San Diego, California, 9The William and Sonja Connor Chair of Preventive Icahn School of Medicine Mount Sinai, Director Lipids and Metabolism,
Cardiology, Professor of Medicine and Physiology & Pharmacology, Mount Sinai Heart, New York, New York, 17Director, Adult Type 1 Diabetes
Director, Center for Preventive Cardiology, Knight Cardiovascular Institute, Program, Division of Endocrinology, Diabetes, and Metabolism, The Ohio
Oregon Health & Science University, Portland, Oregon, 10Professor of State University Wexner Medical Center, Columbus, Ohio.
Medicine and Pharmacology, Assistant Dean for Clinical Research, Tullis Address correspondence to Dr. Yehuda Handelsman, Medical Director &
Tulane Alumni Chair in Diabetes, Chief, Section of Endocrinology, Tulane Principal Investigator, Metabolic Institute of America, 18372 Clark Street,
University Health Sciences Center, New Orleans, Louisiana, 11Professor, #212, Tarzana, CA 91356.
Departments of Medicine, Biochemistry and Cell and Molecular Biology, E-mail: yhandelsman@gmail.com.
Baylor College of Medicine, Houston, Texas, 12Chairman, Grunberger Published as a Rapid Electronic Article in Press at http://www.endocrine
Diabetes Institute, Clinical Professor, Internal Medicine and Molecular practice.org. DOI: 10.4158/CS-2020-0490
Medicine & Genetics, Wayne State University School of Medicine, To purchase reprints of this article, please visit: https://www.aace.com/
Professor, Internal Medicine, Oakland University William Beaumont publications/journal-reprints-copyrights-permissions.
School of Medicine, Visiting Professor, Internal Medicine, First Faculty of Copyright © 2020 AACE.
Medicine, Charles University, Prague, Czech Republic, Past President,

ENDOCRINE PRACTICE Vol 26 No. 10 October 2020 1


2 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

ABSTRACT
of Cardiovascular Events with EPA-Intervention Trial;
The treatment of lipid disorders begins with lifestyle UKPDS = United Kingdom Prospective Diabetes
therapy to improve nutrition, physical activity, weight, and Study; U.S. = United States; VLDL = very-low-density
other factors that affect lipids. Secondary causes of lipid lipoproteins
disorders should be addressed, and pharmacologic therapy
initiated based on a patient’s risk for atherosclerotic cardio-
vascular disease (ASCVD). Patients at extreme ASCVD EXECUTIVE SUMMARY
risk should be treated with high-intensity statin therapy to
achieve a goal low-density lipoprotein cholesterol (LDL-C) This algorithm for the comprehensive management
of <55 mg/dL, and those at very high ASCVD risk should of dyslipidemia and prevention of cardiovascular disease
be treated to achieve LDL-C <70 mg/dL. Treatment for (CVD) complements the 2017 American Association
moderate and high ASCVD risk patients may begin with of Clinical Endocrinologists/American College of
a moderate-intensity statin to achieve an LDL-C <100 mg/ Endocrinology (AACE/ACE) Guidelines for Management
dL, while the LDL-C goal is <130 mg/dL for those at low of Dyslipidemia and Prevention of Cardiovascular Disease
risk. In all cases, treatment should be intensified, includ- (1) and provides clinicians with a practical guide that
ing the addition of other LDL-C-lowering agents (i.e., considers the whole patient, their spectrum of risks and
proprotein convertase subtilisin/kexin type 9 inhibitors, complications, and evidence-based approaches to treat-
ezetimibe, colesevelam, or bempedoic acid) as needed to ment. However, the algorithm has incorporated newer data
achieve treatment goals. When targeting triglyceride levels, that were not available when the 2017 guidelines were
the desirable goal is <150 mg/dL. Statin therapy should be drafted but which are necessary for contemporary lipid
combined with a fibrate, prescription-grade omega-3 fatty management. Despite recent improvements in the overall
acid, and/or niacin to reduce triglycerides in all patients rates of lipid disorders and heart disease, atherosclerotic
with triglycerides ≥500 mg/dL, and icosapent ethyl should cardiovascular disease (ASCVD) remains the leading
be added to a statin in any patient with established ASCVD cause of death throughout the world (2,3). In the United
or diabetes with ≥2 ASCVD risk factors and triglycerides States (U.S.), coronary heart disease (CHD), heart failure,
between 135 and 499 mg/dL to prevent ASCVD events. and stroke together affect 24.3 million people (9% of the
Management of additional risk factors such as elevated population), and 29% and 26% of adults have elevations in
lipoprotein(a) and statin intolerance is also described. low-density lipoprotein cholesterol (LDL-C) and triglyc-
erides, respectively, putting them at risk of major adverse
Abbreviations: cardiovascular events (MACE) (3,4).
AACE = American Association of Clinical Controlling atherogenic cholesterol particle concen-
Endocrinologists; ACE = American College of trations is fundamental to prevention of ASCVD, includ-
Endocrinology; ACS = acute coronary syndrome; ing CHD (5). However, only a small percentage of the U.S.
apo B = apolipoprotein B; ASCVD = atheroscle- population who clearly would benefit from a statin (i.e.,
rotic cardiovascular disease; BA = bempedoic acid; hydroxymethylglutaryl-coenzyme A [HMG-CoA] reduc-
CAC = coronary artery calcium; CHD = coronary tase inhibitor) currently takes one (6). Moreover, the aver-
heart disease; CK = creatine kinase; CKD = chronic age LDL-C among U.S. adults, 112 mg/dL (3), is higher
kidney disease; DHA = docosahexaenoic acid; EPA = than the recommended LDL-C goal of <100 mg/dL for
eicosapentaenoic acid; FCS = familial chylomicrone- those with moderate ASCVD risk and well above the goals
mia syndrome; FDA = United States Food and Drug for people at high risk of ASCVD (1).
Administration; FOURIER = Further Cardiovascular Risk factors for dyslipidemia, including obesity,
Outcomes Research with PCSK9 Inhibition in Subjects chronic kidney disease (CKD), and diabetes, have risen
with Elevated Risk; HDL-C = high-density lipoprotein steadily over recent decades and are expected to continue
cholesterol; HeFH = heterozygous familial hypercho- to increase (7-9). Most notably, 44% of U.S. adults now
lesterolemia; HoFH = homozygous familial hyper- have obesity (defined by a body mass index [BMI] >30 kg/
cholesterolemia; hsCRP = high-sensitivity C reac- m2), without any difference in the proportions of younger
tive protein; IDL = intermediate-density lipoproteins; versus older adults (7). Early exposure to elevated lipids,
IMPROVE-IT = Improved Reduction of Outcomes: prior to age 55 years, has a greater impact on CHD risk
Vytorin Efficacy International Trial; IPE = icosapent than do elevations later in life (10), indicating that care-
ethyl; LDL-C = low-density lipoprotein cholesterol; ful management of patients with dyslipidemia is important
Lp(a) = lipoprotein a; MACE = major adverse cardio- across the lifespan.
vascular events; MI = myocardial infarction; OSA = This executive summary expands on the information
obstructive sleep apnea; PCSK9 = proprotein conver- in the Dyslipidemia and Prevention of Cardiovascular
tase subtilisin/kexin type 9; REDUCE-IT = Reduction Disease Algorithm slides and provides the supporting refer-
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 3

ences. The algorithm is organized into discrete sections, Familial hypercholesterolemia is caused by major gene
as follows: mutations affecting LDL receptor function with codomi-
I. Dyslipidemic States nant inheritance, meaning the milder form is passed on
II. Secondary Causes of Lipid Disorders by 1 parent (heterozygous familial hypercholesterolemia;
III. Screening for and Assessing Lipid Disorders and HeFH) and the more severe form by both parents (homozy-
ASCVD Risk gous familial hypercholesterolemia; HoFH). HeFH is asso-
IV. ASCVD Risk Categories and Treatment Goals ciated with LDL-C levels of >190 mg/dL (>160 mg/dL in
V. Lifestyle Recommendations children) and HoFH with LDL-C >500 mg/dL in the absence
VI. Treating LDL-C to Goal of secondary causes (1). HeFH affects approximately 1 in
VII. Managing Statin Intolerance and Safety 250 to 500 persons, whereas HoFH occurs at a rate of 1 to
VIII. Management of Hypertriglyceridemia and the Role 4 per 1 million, although rates as high as 1/160,000 have
of Icosapent Ethyl been reported in some genetically isolated populations
IX. Assessment and Management of Elevated (1,16). Other rare genetic dyslipidemic syndromes include
Lipoprotein(a) familial hypoalphalipoproteinemia, familial chylomicrone-
X. Profiles of Medications for Dyslipidemia mia syndrome (FCS), beta-sitosterolemia, lysosomal acid
lipase deficiency, and lipodystrophy. Clinicians may refer
I. Dyslipidemic States patients to lipid specialists for further investigations and
Dyslipidemia comprises a range of conditions, management as appropriate, including genetic testing.
primarily defined by elevations in lipoprotein choles-
terol, including LDL-C and non-high-density lipoprotein II. Secondary Causes of Lipid Disorders
cholesterol (HDL-C), as well as elevated triglycerides (see Once dyslipidemia has been diagnosed, secondary
Dyslipidemia and Prevention of Cardiovascular Disease causes of the disorder must be excluded to rule out cases
Algorithm Slide I. Dyslipidemic States). All of these condi- that could be treated or cured with approaches that do not
tions independently increase the risk of ASCVD (11,12). involve cholesterol- or triglyceride-lowering medications
In the U.S., 38% of adults have a total cholesterol (1,11,17,18).
>200 mg/dL and 29% have LDL-C ≥130 mg/dL (3). Diagnosis of secondary causes should begin with a
Hypertriglyceridemia, defined as triglycerides ≥150 mg/ complete medical, family, and nutrition history. A physi-
dL, affects 26% of U.S. adults (4). Severe hypertriglyc- cal examination must be undertaken to identify additional
eridemia (triglycerides >500 mg/dL) increases the risk of risk factors, including genetic features. Laboratory testing
acute pancreatitis and chylomicronemia syndrome (1,13). for glucose, thyroid, liver, and renal function should also
Lipoprotein(a), or Lp(a), is a low-density lipoprotein be conducted. Finally, a list of all prescription and over
(LDL) particle with an apolipoprotein(a) attached to the the counter medications, as well as dietary supplements,
apolipoprotein B (apo B) component. Approximately 20% should be compiled, as many of these affect lipids. Treating
of the population has increased levels of this pro-athero- an underlying contributing disease or discontinuing a
sclerotic particle (14). contributing medication, if medically appropriate, may
Hypercholesterolemia and hypertriglyceridemia are ameliorate dyslipidemia.
frequently secondary to other medical conditions or medi- In developed nations, the most common secondary
cations (see Dyslipidemia and Prevention of Cardiovascular cause of dyslipidemia is a sedentary lifestyle with lack
Disease Algorithm Slide II. Secondary Causes of Lipid of physical activity and a diet high in carbohydrates and/
Disorders), and there are also a wide variety of primary or simple sugars. A diet high in saturated fats and exces-
dyslipidemias. The most common primary dyslipidemias sive alcohol intake may also lead to dyslipidemia. Other
are elevated Lp(a) and mixed dyslipidemia, a combina- common secondary causes include medical conditions
tion of elevated levels of both cholesterol and triglycer- such as overweight or obesity and associated metabolic
ides. This combination is also seen in 2 much less common syndrome or prediabetes; uncontrolled diabetes; hypothy-
states: familial combined hyperlipidemia and dysbetalipo- roidism; pregnancy; stage ≥3 CKD (ie, estimated glomeru-
proteinemia. Familial combined hyperlipidemia is defined lar filtration rate [eGFR] ≤59 mL/min/1.73 m2), especially
as elevations in either cholesterol or triglycerides in ≥2 with albuminuria; nephrotic syndrome; cholestatic diseases
first-degree relatives (often occurring sequentially rather of the liver; lipodystrophy; paraproteinemia (e.g., dysgam-
than simultaneously), along with a strong family history of maglobulinemia, multiple myeloma); and chronic inflam-
premature ASCVD, and has an estimated population prev- matory conditions (e.g., rheumatoid arthritis, systemic
alence of 1 to 3% (3,15). Dysbetalipoproteinemia (type III lupus erythematosus) (1,18).
dyslipidemia) consists of an excess of cholesterol enriched Medications that may contribute to dyslipidemia
triglyceride-remnant lipoproteins. It is also associated with include oral estrogens and progestins, anabolic steroids,
premature ASCVD but infrequently with a strong family selective estrogen receptor modulators, highly active anti-
history due to generally recessive inheritance. retroviral agents such as protease inhibitors for the treat-
4 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

ment of HIV, immunosuppressive medications (e.g., cyclo- Laboratory evaluations include fasting levels of the
sporine, mammalian target of rapamycin [mTOR] kinase lipid profile, including total cholesterol, HDL-C, triglyc-
inhibitor), glucocorticoids, retinoids, interferon, taxol erides, LDL-C, and calculated non-HDL-C; a compre-
derivatives, L-asparaginase, cyclophosphamide, atypical hensive medical panel, including uric acid (which is a
antipsychotic agents, beta-blockers, and thiazide diuret- cardiovascular risk factor); hemoglobin A1C (A1C); and
ics. Although bile acid sequestrants are used mainly to thyroid-stimulating hormone. Assessment of apo B or LDL
reduce cholesterol, these agents may also increase triglyc- particles, Lp(a), and high-sensitivity C reactive protein
eride levels and should be used cautiously in patients with (hsCRP) should also be considered based on individual
triglyceride elevations (1,18). patient clinical circumstances.
Monitoring of lipid levels should continue after a Diagnostic procedures may include resting electro-
secondary cause of dyslipidemia has been diagnosed, as cardiogram as well as treadmill, chemical, and/or nuclear
some conditions such as diabetes also increase the risk stress tests, as appropriate. Beyond the CAC score, carot-
of ASCVD, and more aggressive lipid-lowering therapy id ultrasound for plaque formation may be informative.
is warranted. Simultaneous treatment of the secondary Measurement of carotid intima-media thickness was used
cause of dyslipidemia and the dyslipidemic state may for predicting risk for years, but in the Multiethnic Study of
be necessary. Atherosclerosis (MESA), values above the 75th percentile
did not predict risk of transient ischemic attack or stroke,
III. Screening for and Assessing whereas the presence of carotid plaques did (19).
Lipid Disorders and ASCVD Risk The Framingham Risk Equation (https://framingham
Screening for lipid disorders should be based on each heartstudy.org/fhs-risk-functions/cardiovascular-disease-
patient’s personal and family medical history, and assess- 10-year-risk/) was the first widely used risk calculator (20).
ments should include these findings as well as the results More currently used risk calculators include the MESA
of a physical examination; laboratory evaluations; and risk calculator incorporating a CAC score (https://www.
diagnostic procedures as appropriate, including electro- mesa-nhlbi.org/CAC-Tools.aspx), Reynold’s Risk Score
cardiograms and imaging. These results could be supple- incorporating hsCRP (http://www.reynoldsriskscore.org),
mented with an ASCVD risk calculator chosen based on and the United Kingdom Prospective Diabetes Study Risk
the patient’s individual characteristics. In particular, a Engine for patients with diabetes (https://www.dtu.ox.ac.
coronary artery calcium (CAC) score is very useful for uk/riskengine/) (20-22). The recently enhanced American
risk stratification. College of Cardiology/American Heart Association
Medical conditions that increase a patient’s risk of pooled-cohort ASCVD Risk Estimator (www.cvrisk
dyslipidemia and/or ASCVD include impaired glucose calculator.com) is another option (23).
tolerance, metabolic syndrome, diabetes, obesity, hyper- More detailed information on screening for lipid
tension, prior cardiovascular or cerebrovascular events, disorders and ASCVD risk can be found in the AACE/
CKD, nonalcoholic fatty liver disease (NAFLD) or nonal- ACE 2017 Guidelines for Management of Dyslipidemia
coholic steatohepatitis (NASH), autoimmune or inflam- and Prevention of Cardiovascular Disease (1).
matory disease (e.g., lupus, rheumatoid arthritis, psoria-
sis, periodontal disease), hepatitis C, a history of pancre- IV. ASCVD Risk Categories and Goals
atitis, and medications that alter lipids (e.g., steroids, In the clinical management of dyslipidemia, a reason-
retinoids, HIV therapy, antirejection medications; see able goal is to strive for lipid levels in the normal range;
Dyslipidemia and Prevention of Cardiovascular Disease however, more aggressive goals need to be set for higher-
Algorithm Slide II. Secondary Causes of Lipid Disorders). risk individuals. As shown in Table 1, AACE has defined 5
Behavioral factors include smoking, a sedentary lifestyle, risk categories based on the number and severity of major
and diets high in saturated fat (1). Patients whose family risk factors (see Table 2). Each category has goals for
members have ASCVD, hypertension, or dyslipidemia LDL-C, non-HDL-C, and apo B levels, proportional to the
should also be screened. A personal history or family degree of risk. A goal for triglyceride level is also offered
history of tendon xanthomas, corneal arcus, or xanthe- (see Dyslipidemia and Prevention of Cardiovascular
lasma are clues suggesting hypercholesterolemia. Eruptive Disease Algorithm Slide IV. ASCVD Risk Categories and
xanthomas and lipemia retinalis are suggestive of severe Goals) (1).
hypertriglyceridemia. Individuals at extreme risk for ASCVD events include
The physical examination should include each patient’s those already diagnosed with progressive ASCVD, includ-
height and weight (for calculation of BMI), waist circum- ing unstable angina after achieving an LDL-C <70 mg/dL;
ference, blood pressure, and peripheral and carotid pulses. those with established clinical ASCVD plus diabetes, stage
Further advanced evaluation may include a cardiac evalua- ≥3 CKD, or HeFH; or those with a history of premature
tion; vascular bruits; ankle-brachial index; and assessment ASCVD (age <55 years, male; <65 years, female).
of tendon xanthomas, eruptive xanthomas, lipemia retina- Very-high-risk criteria include established or recent
lis, corneal arcus, and xanthelasma. hospitalization for acute coronary syndrome (ACS) or for
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 5

coronary, carotid, or peripheral vascular disease with a International Trial (IMPROVE-IT) trial, wherein lower-
10-year ASCVD risk >20%. Individuals with diabetes who ing of LDL-C to 53 mg/dL with the addition of ezetimibe
have 1 or more major risk factor(s) for ASCVD (Table 2), to simvastatin resulted in further ASCVD benefit (26).
those with stage ≥3 CKD with albuminuria, or those with However, because an isolated focus on LDL-C is not
HeFH are also at very high risk. always sufficient to prevent ASCVD in at-risk individuals
Individuals at high risk include those with ≥2 risk or to treat existing atherosclerosis, goals for non-HDL-C,
factors and a 10-year risk between 10% and 20% or who apo B, and triglycerides are also included in the risk assess-
have diabetes or stage ≥3 CKD with no other risk factors. ment and goals. Non-HDL (total cholesterol minus HDL-C)
Moderate risk individuals have <2 risk factors and a reflects the total atherogenic burden, including particles
10-year risk <10%, and those at low risk have no ASCVD contained within very-low-density lipoproteins (VLDL),
risk factors. intermediate-density lipoproteins (IDL), and LDL as well
LDL-C has been, and remains, the main focus of as chylomicron remnants and Lp(a). The non-HDL-C goal
efforts to improve lipid profiles in individuals at risk for is 25 to 30 mg/dL above the LDL-C goal (i.e., <80 mg/dL
ASCVD. Numerous LDL-C-lowering trials utilizing statins for extreme risk, <100 mg/dL for very high risk, <130 mg/
over many years have consistently demonstrated that lower dL for medium to high risk, and <160 mg/dL for low risk)
LDL-C levels result in improved ASCVD outcomes (1). and is a more precise indicator of ASCVD risk than LDL-C
Outcomes trials with proprotein convertase subtilisin/kexin (1). Non-HDL-C reached a level of <65 mg/dL and apo
type 9 (PCSK9) inhibitors in which achieved LDL-C levels B of <50 mg/dL in the Further Cardiovascular Outcomes
were substantially lower than those in statin trials further Research with PCSK9 Inhibition in Subjects with Elevated
reinforced the notion that “lower is better” (24,25). Thus, Risk (FOURIER) trial, validating the non-HDL-C and apo
LDL-C goals range from <130 mg/dL for low-risk indi- B goals for patients at extreme risk (25).
viduals to <55 mg/dL for those at extreme risk for ASCVD. The term apo B refers to the total plasma concentra-
The goal for the extreme risk category was derived from tion of apo B-100, plus apo B-48. Apo B may be elevated
the Improved Reduction of Outcomes: Vytorin Efficacy in individuals with optimal LDL-C when small, dense

Table 1
ASCVD Risk Categories and Treatment Goals
Treatment goals (mg/dL)
Risk category Risk factors and 10-year risk LDL-C Non-HDL-C Apo B TG
• Progressive ASCVD including unstable
angina

• Established clinical ASCVD plus diabetes or


Extreme risk <55 <80 <70 <150
CKD ≥3 or HeFH

• History of premature ASCVD (<55 y, male;


<65 y, female)
• Established clinical ASCVD or recent
hospitalization for ACS, carotid, or
peripheral vascular disease, or 10-year risk
>20%
Very high risk <70 <100 <80 <150
• Diabetes with ≥1 risk factor(s)

• CKD ≥3 with albuminuria

• HeFH
• ≥2 risk factors and 10-year risk 10-20%
High risk <100 <130 <90 <150
• Diabetes or CKD ≥3 with no other risk
factors
Moderate risk • <2 risk factors and 10-year risk <10% <100 <130 <90 <150
Low risk • No risk factors <130 <160 NR <150
Abbreviations: ACS = acute coronary syndrome; Apo B = apolipoprotein B; ASCVD = atherosclerotic cardiovascular disease;
CKD ≥3 = stage 3-5 chronic kidney disease (estimated glomerular filtration rate ≤59 mL/min/1.73 m2); HeFH = heterozygous familial
hypercholesterolemia; NR = not recommended; TG = triglycerides; y = years.
Adapted from Jellinger et al (1).
6 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

Table 2
Major Atherosclerotic Cardiovascular Disease Risk Factors
Major risk factors Additional risk factors Nontraditional risk factors
Advancing agea,b,c,d Obesity, abdominal obesityc,d ­ ↑Lipoprotein (a)
↑Total serum cholesterol levela,b,d Family history of ­ ↑Clotting factors
­↑Non–HDL-Cd hyperlipidemiad ­ ↑Inflammation markers
­↑LDL-Ca,d ­ ↑Small, dense LDL-Cd (hsCRP; Lp-PLA2)
Low HDL-Ca,d,e ­ ↑Apo Bd ­ ↑Homocysteine levels
Diabetes mellitusa,b,c,d ­ ↑LDL particle concentration Apo E4 isoform
Hypertensiona,b,c,d Fasting/postprandial ­ ↑Uric acid
Chronic kidney disease 3,4h hypertriglyceridemiad ­ ↑TG-rich remnants
Cigarette smokinga,b,c,d PCOSd
Family history of ASCVDa,d,g Dyslipidemic triadf
Abbreviations: apo = apolipoprotein; ASCVD = atherosclerotic cardiovascular disease; HDL-C = high-density lipoprotein cholesterol;
hsCRP = high sensitivity C-reactive protein; LDL = low-density lipoprotein; LDL-C = low-density lipoprotein cholesterol; Lp-PLA2
= lipoprotein-associated phospholipase; PCOS = polycystic ovary syndrome.
aRisk factors identified in the Framingham Heart study.
bRisk factors identified in the MRFIT study (Multiple Risk Factor Intervention Trial).
cRisk factors identified in the INTERHEART study.
dRisk factors identified in guidelines and position statements (National Cholesterol Education Program Adult Treatment Panel

III, American Association of Clinical Endocrinologists Polycystic Ovary Syndrome Position Statement, American Association of
Clinical Endocrinologists Insulin Resistance Syndrome Position Statement, American Diabetes Association Standards of Care,
American Diabetes Association/American College of Cardiology Consensus Statement on Lipoprotein Management in Patients
with Cardiometabolic Risk, National Lipid Association, Clinical Utility of Inflammatory Markers and Advanced Lipoprotein
Testing).
eElevated high-density lipoprotein cholesterol is a negative risk factor.
fHypertriglyceridemia; low high-density lipoprotein cholesterol; and an excess of small, dense low-density lipoproteins.
gDefinite myocardial infarction or sudden death before 55 years of age in father or other male first-degree relative or before 65 years

of age in mother or other female first-degree relative.


hBased on a pooled analysis of community-based studies (N = 22,634).

Reprinted with permission from Jellinger et al (1).

LDL particles are present. This often occurs in individuals rate (32-34). Additionally, a subgroup analysis of several
with insulin resistance who manifest hypertriglyceridemia, triglyceride-lowering trials utilizing fibrates demon-
diabetes, metabolic syndrome, or obesity (12,27-31). Apo strated improved ASCVD outcomes when baseline
B-100 measurement may provide a more accurate assess- triglycerides are >200 mg/dL and HDL-C is <40 mg/dL
ment of atherogenicity because all atherogenic particles (32,35-37).
(i.e., VLDL, IDL, and LDL) contain 1 apo B-100 molecule. Low HDL-C (HDL-C <40 mg/dL in men and <50 mg/
AACE supports an apo B goal of <90 mg/dL for individu- dL in women, without accompanying hypertriglyceride-
als at risk of ASCVD, including those with diabetes, and mia) is not included as a factor in the risk category table.
of <80 mg/dL for those with established ASCVD or diabe- However, epidemiologic evidence and findings from the
tes plus ≥1 additional risk factor (12,31). An apo B goal Veterans Affairs High-Density Lipoprotein Cholesterol
<70 mg/dL is recommended for patients in the extreme Intervention Trial study support a cardioprotective role of
risk category and should be considered in clinical situa- HDL-C (1,38,39). Increasing HDL-C pharmacologically
tions characterized by persistent ASCVD. Furthermore, utilizing fibrates or niacin, however, has not demonstrated
measurement of apo B is useful in assessing the success of a primary ASCVD benefit. Therefore, AACE recommends
lipid-lowering therapy, since apo B may remain above goal that, after aggressive lifestyle interventions to raise HDL-C
after achieving the LDL-C goal. as high as possible, the focus should be on reducing LDL-C
Goal triglyceride levels are <150 mg/dL; levels rang- by pharmacologic therapy, especially if HDL-C levels are
ing from 150 to 199 mg/dL are classified as borderline low and other risk factors are present (including border-
high; levels 200 to 499 mg/dL are high, and levels ≥500 line elevated LDL-C levels, a family history of premature
mg/dL are considered very high (11). Several studies ASCVD, or a personal history of ASCVD) (1).
including the long-term follow up of the Helsinki Heart
Study, the Japan EPA Lipid Intervention Study, and the V. Lifestyle Recommendations
Reduction of Cardiovascular Events with EPA-Intervention The management of dyslipidemia requires a compre-
Trial (REDUCE-IT), showed that treating patients with hensive strategy to control lipid levels and address associ-
elevated triglycerides significantly improved cardiovas- ated metabolic abnormalities and modifiable risk factors.
cular outcomes and/or lowered the ASCVD mortality Managing individuals with lipid disorders begins with
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 7

implementation of lifestyle changes including medical intensity physical activity (48). Daily physical activity
nutrition therapy, physical activity, smoking cessation, and goals may be met in a single session or multiple sessions
assessment of sleep and mental health issues. The intensity throughout the course of a day (10 minutes minimum); for
of these interventions should be stratified by the degree some individuals, breaking activity up throughout the day
of cardiovascular risk, type of dyslipidemia, and related may help improve adherence to physical activity programs
complications. (49). Additional studies also suggest that weight and resis-
Diet can have a substantial effect on lipid levels and is tance training may be beneficial to some individuals with
an important determinant of ASCVD risk. Experts have yet the insulin resistance syndrome, independent of body fat or
to reach consensus on what is the optimal diet for prevention aerobic fitness (50). Therefore, in addition to aerobic activ-
and treatment of ASCVD. Several dietary patterns, howev- ity, muscle-strengthening activity is recommended at least
er, appear to be beneficial, including the Mediterranean and 2 days a week (51).
Dietary Approaches to Stop Hypertension (DASH), which Individuals who are nonadherent to regular physical
improve cardiovascular outcomes (40,41). A single dietary activity should be repeatedly encouraged, and practitio-
pattern may not be optimal for all populations, and logistic ners should apply a variety of strategies as necessary to
feasibility and cultural appeal of specific diets vary widely improve adherence. Strategies may include individually
among the many regions of the world. Even though medi- tailored advice, identification of adherence barriers, refer-
cal nutrition therapy continues to be a challenging area, ral to instructor-led exercise classes, and routine follow-up
there are certain commonalities between diets which have and consultation.
shown benefit. Evidence supports an association of 6 to 8 hours of
Nutritional guidelines for the reduction of cardio- sleep per night with a reduction in cardiometabolic risk
vascular risk usually recommend diets rich in fruits and factors, whereas sleep deprivation aggravates insulin resis-
vegetables, whole grains, legumes, and high soluble fiber, tance, hypertension, hyperglycemia, and dyslipidemia,
and avoiding processed foods, with a reduction in total and increases inflammatory cytokines (52-57). Daytime
calories. The Mediterranean and DASH diets support these drowsiness, a frequent symptom of sleep disorders such as
principles. Generally, reduced fat dairy products, fish, lean sleep apnea, is associated with an increased risk of acci-
meats, and skinless poultry are preferable to traditional dents, errors in judgment, and diminished performance.
high fat alternatives, and salt intake should be decreased Basic sleep hygiene recommendations should be provided
(41,42). Soluble fiber lowers LDL-C with reductions of 8 to all patients with potential cardiovascular problems. The
to 24%. Sources of insoluble fiber (such as whole wheat) most common type of sleep apnea, obstructive sleep apnea
are also associated with low ASCVD risk. The recom- (OSA), is caused by physical obstruction of the airway
mendation for a total fat intake of 25 to 35% of calories during sleep. The resulting lack of oxygen causes the
comes from the observation that a low-fat diet can lead patient to awaken and snore, snort, and grunt throughout
to elevations of triglycerides and lowering of HDL-C, the night. The awakenings may happen hundreds of times
especially when excessive simple carbohydrates or alco- per night, often without the patient’s awareness. OSA is
hol are consumed. Paradoxically, studies have shown that more common in males, the elderly, and persons with
limiting fat intake to ~10% of calories is associated with obesity. People with suspected OSA should be referred for
ASCVD regression and decreased ASCVD events (43,44). a home study in lower risk settings or to a sleep specialist
In the European Prospective Investigation into Cancer and for formal evaluation and treatment in higher-risk settings.
Nutrition (EPIC)–Oxford study, mortality from ischemic Evidence supports an association between ASCVD
heart disease was lower in vegetarians than in nonvegetar- and mental health issues. Depression is an independent risk
ians (45). Studies looking at the incidence of stroke have factor, as well as more prevalent, in patients with ASCVD.
revealed a benefit to whole-food, plant-based, high-fiber Other mental health issues, such as schizophrenia, bipolar
diets with little or no animal products (46). One mecha- disorder, anxiety, and post-traumatic stress disorder have
nism might be that meat and dairy cause higher levels of been found to increase the risk for ASCVD. The associa-
trimethylamine N-oxide, which has been associated with tion is unclear but may be on the basis of genetic, envi-
development and progression of atherosclerosis, primarily ronmental, psychologic, or other mechanisms (58). Having
by altering the gut microbiome (47). a positive outlook, in contrast, has been associated with
Physical activity is associated with improvements in improved ASCVD outcomes (59). This has been referred
risk factors such as obesity, waist circumference, glucose to as a happiness factor but emphasizes the importance of a
intolerance, hypertension, and dyslipidemia. Specific close social structure and community involvement.
lipid-level improvements associated with regular exercise Although some meta-analyses have shown decreased
include reduced triglyceride and VLDL-C levels, increased cardiovascular outcomes in persons who drink limited
HDL-C, reductions in hsCRP, and in some individuals, quantities of alcohol compared to abstainers (60,61), drink-
decreased LDL-C levels (1). Exercise programs generally ing alcohol is not recommended as an intervention due to
should include 150 to 300 minutes weekly of moderate- potential adverse effects. Drinking too much alcohol can
8 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

raise triglycerides. Increases in HDL-C may occur, but in therapy in some patients (69). Due to these limitations,
not the HDL-C subfraction associated with a cardioprotec- some patients may require the addition of other agents to
tive effect. Excess alcohol can also lead to hypertension, achieve LDL-C goals, and the evidence from recent combi-
cardiomyopathy, and atrial fibrillation with subsequent nation therapy studies have proven that reducing LDL-C
strokes (62). Substance abuse with cocaine has also been by any means leads to ASCVD event reductions. In the
associated with both acute cardiovascular events (e.g., IMPROVE-IT trial, which involved 18,144 patients with
hypertension and arrhythmias) and atherosclerosis and ACS, an LDL-C of 53 mg/dL achieved with the combi-
cardiomyopathy in the long-term (63). nation of ezetimibe plus moderate-dose statin significantly
Smoking is a major risk factor for ASCVD and may reduced the risk of MACE (a composite of cardiovascu-
triple the risk of death due to atherosclerosis. Fortunately, lar death, nonfatal MI, unstable angina requiring rehospi-
smoking cessation mitigates the risk rapidly and signifi- talization, coronary revascularization, or nonfatal stroke)
cantly. One year of abstinence will decrease the risk of compared with an LDL-C of 70 mg/dL achieved with the
heart attacks significantly, and 5 years will decrease the statin alone, with the most pronounced benefits in patients
risk of stroke to a level comparable to that of nonsmokers with diabetes and those older than 75 years (26). The
(3,64). Smoking cessation is perhaps the most important FOURIER and Evaluation of Cardiovascular Outcomes
component of lifestyle therapy and involves avoidance of After an Acute Coronary Syndrome During Treatment
all tobacco products. Transient nicotine replacement ther- With Alirocumab Study (ODYSSEY Outcomes) trials also
apy and other pharmacologic interventions (e.g., sustained showed that reducing LDL-C to low levels with combina-
release bupropion and varenicline) should be considered tion therapy improves outcomes over high-intensity statin
in patients having difficulty with smoking cessation. alone (24,25). In the FOURIER trial, evolocumab added to
Structured programs should be recommended for patients high-intensity statin therapy reduced LDL-C to 30 mg/dL
unable to stop smoking on their own (1). (59% difference from the statin-only group), which after
2.2 years was associated with a 15% decrease in a compos-
VI. Treating LDL-C to Goal ite of cardiovascular death, MI, stroke, hospitalization for
As described in Section IV, ASCVD Risk Categories unstable angina, or coronary revascularization (25). As
and Goals, AACE has established LDL-C goals rang- individual endpoints, MI, stroke, and coronary revascu-
ing from <55 mg/dL to <130 mg/dL according to indi- larization were reduced by 27%, 21%, and 22%, respec-
viduals’ ASCVD risk based on a large body of evidence tively. Likewise, after 4 years of therapy in ODYSSEY
of numerous outcomes trials with statins, ezetimibe, and Outcomes, LDL-C levels were 55% lower with alirocumab
PCSK9 inhibitors (1,24-26). The findings from these trials plus a high-intensity statin than with statin alone (66 mg/
consistently demonstrate that ASCVD risk decreases with dL versus 103 mg/dL), and this reduction was associated
LDL-C along a continuum, supporting a “lower is better” with a statistically significant 15% decrease in a composite
approach. In a meta-analysis of 26 prospective statin trials of death from CHD, nonfatal MI, fatal or nonfatal ischemic
involving close to 170,000 participants by the Cholesterol stroke, or unstable angina requiring hospitalization (24).
Treatment Trialists group (CTT), each 38 mg/dL (1 MI, ischemic stroke, and unstable angina were reduced
mmol/L) reduction in LDL-C led to a 29% decrease in as individual events by 14%, 27%, and 39%, respective-
major vascular events (nonfatal myocardial infarction [MI] ly. Post-hoc analysis of prespecified groups in FOURIER
or ASCVD death) when baseline LDL-C was <77 mg/dL suggests a progressive composite MACE benefit with
and a 37% reduction when baseline was <70 mg/dL. There progressive LDL-C reductions to very low levels (70).
was a 19% reduction in coronary revascularizations and a To date, cardiovascular outcomes trials (CVOTs)
16% reduction in cerebrovascular accident (65). Another with colesevelam or bempedoic acid (BA) have not been
meta-analysis of 8 major statin trials demonstrated that published, but the CVOTs with statins plus ezetimibe or
those individuals achieving an LDL-C <50 mg/dL, a non- a PCSK9 inhibitor suggest that further LDL-C lowering
HDL-C <75 mg/dL, and apo B <50 mg/dL have the lowest with any combination of agents would confer ASCVD
ASCVD events (66). benefits. On this basis, the algorithm advocates progres-
A statin should be used as first-line cholesterol- sively increasing the intensity of therapy to achieve
lowering therapy, unless contraindicated; current evidence LDL-C goals.
supports a moderate- to high-intensity statin (1,65,67). Lifestyle management is the foundation of all lipid-
However, considerable residual risk often persists even reduction regimens, and statin therapy should be started in
after aggressive statin monotherapy in primary prevention low-risk patients unable to maintain LDL-C <130 mg/dL
patients with multiple cardiovascular risk factors and in on lifestyle therapy and given to all patients at moderate to
secondary prevention patients with stable clinical ASCVD extreme risk regardless of their initial LDL-C level. Lipids
or ACS (30,67,68). Statin intolerance (see Section VII, should be checked every 3 months or more frequently
Managing Statin Intolerance and Safety) or the inability when necessary. The table at the bottom of Slide VI lists
to reach LDL-C goal may limit the use of intensive statin the dosages of the different LDL-C-lowering options
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 9

recommended in each ASCVD risk column, and the range VII. Managing Statin Intolerance and Safety
of LDL-C reductions in primary hyperlipidemia or familial For the great majority of patients, statins are safe and
hypercholesterolemia reported in the prescribing informa- well-tolerated, and for those at heightened risk of ASCVD,
tion for each agent appear in Table 3 (71-82). the ASCVD benefit far outweighs the likelihood of adverse
If patients are not at goal, address adherence to therapy effects. Nevertheless, statin intolerance may occur in a
first. When intensification is necessary, the order of agents significant subset of statin-treated patients.
listed indicates the AACE preference for consideration. Statin-associated muscle symptoms are the most
For example, treatment for patients at extreme risk should common reason for statin intolerance, and may include
begin with lifestyle therapy plus a high-intensity statin pain, weakness, cramps, or stiffness. Observational stud-
(atorvastatin 40 to 80 mg or rosuvastatin 20 to 40 mg, or ies and experience in clinical practice have suggested an
the highest tolerated statin dose) to achieve an LDL-C incidence ranging from ~5 to >20%, although placebo-
goal of <55 mg/dL. If LDL-C remains above goal after 3 controlled clinical trials have reported the risk to be much
months, a PCSK9 inhibitor, ezetimibe, colesevelam, or BA lower. Assessment is based on the temporal association of
should be added, depending on required LDL-C lowering, symptoms with onset of treatment, cessation upon statin
and a third agent should be added if the combination fails discontinuation, and if not severe, recurrence of symp-
to achieve the goal. Because the cost of ezetimibe is low, toms following re-challenge with the same and/or up to
it may be preferred over PCSK9 inhibitors as second-line 2 other statins. An elevated creatine kinase (CK) level is
therapy to achieve an LDL-C <70 mg/dL for patients who not required for establishing the diagnosis, although it
require no more than 15 to 20% further reduction to reach may be corroborated by CK ≥4 times the upper limit of
goals. For patients at moderate or high risk, lipid manage- normal. More severe myopathy leading to rhabdomy-
ment should begin with a moderate-intensity statin and be olysis, with CK >10 times the upper limit of normal, is
increased to a high-intensity statin before adding a second rare (~1 per 10,000 patient-years) and requires immedi-
lipid-lowering medication to reach an LDL-C <100 mg/dL. ate statin cessation, hydration, and monitoring of renal

Table 3
Effects of LDL-C Lowering Agentsa
Agent LDL-C reductions
Moderate-intensity HMG-CoA reductase inhibitors (statins)
Lovastatin 40 mg –31% to –42%
Pravastatin 40-80 mg –34% to –37%
Fluvastatin 40 mg BID –36%
Fluvastatin XL 80 mg –35%
Pitavastatin 2-4 mg –38% to –45%
Simvastatin 20-40 mg –29% to –41%
Atorvastatin 10-20 mg –29% to –33%
Rosuvastatin 5-10 mg –45% to –52%
High-intensity HMG-CoA reductase inhibitors (statins)
Atorvastatin 40-80 mg –50% to –60%
Rosuvastatin 20-40 mg –55% to –63%
Cholesterol absorption inhibitor: ezetimibeb –12% to –17%
PCSK9 inhibitors
Evolocumab 140 mg Q2W or 420 mg Q4Wb –63% to –71%
Alirocumab 75-150 mg Q2Wb –48% to –58%
Bile acid sequestrant: colesevelamb –8% to –16%
ACL inhibitor: bempedoic acidb –17% to –18%
Abbreviations: ACL = adenosine triphosphate-citrate lyase; BID = twice daily; HMG-CoA = hydroxymethylglutaryl-coenzyme A;
LDL-C = low-density lipoprotein cholesterol; PCSK9 = proprotein convertase subtilisin/kexin type 9; Q2W = once every 2 weeks;
Q4W = once every 4 weeks; XL = extended release.
aRefer to references 71 to 82.
bIn combination with statin therapy.
10 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

function. Its occurrence is a contraindication for future be measured fasting or nonfasting, and optimum levels are
statin therapy (83-85). considered to be well below 100 mg/dL (91).
Physicians should be aware of the factors that may The prevalence of hypertriglyceridemia, defined as
increase the risk for statin-induced myopathy. These triglyceride levels exceeding 150 mg/dL, is approximate-
include female sex, small body size, age >65 years, frail- ly 26% in the U.S. (4). Combinations of genetic defects
ty, East Asian ancestry, a personal or family history of and environmental effects contribute to the development
myopathy, poorly controlled hypothyroidism, subnormal of hypertriglyceridemia; increased production of triglyc-
vitamin D, and the use of medications that raise circulat- erides occurs in obesity, insulin resistance, and diabetes,
ing levels of statins and/or their active metabolites (e.g., whereas various genetic conditions cause decreased clear-
erythromycin, fluconazole). In such cases consider using ance of triglycerides. Hypertriglyceridemia may present
smaller statin doses and/or less potent statins with lower with increased numbers or concentrations of atherogenic
incidence of myopathy (e.g., pitavastatin, extended-release small, dense LDL particles and apo B-100-associated
fluvastatin), along with cautious up-titration of dose. It is triglyceride-rich lipoprotein remnant cholesterol particles,
important to advise patients to report muscle symptoms of which increase ASCVD risk (92).
any severity that occur with initiation of statin, increase in Prior to publication of REDUCE-IT, only a few stud-
dose, or change to a different statin type, and to stop treat- ies with inconsistent results supported lowering triglycer-
ment if these are deemed to be clinically significant. Once ides to reduce ASCVD events (32,35-37), whereas several
symptoms resolve, therapy may be resumed as a trial, with large-scale clinical trials failed to prove that managing
consideration of a lower dose of the same statin, an alter- triglycerides reduces ASCVD (93,94). No medication that
nate statin such as those noted above, or others with rela- lowers triglycerides was approved by the United States
tively low lipid solubility (pravastatin, rosuvastatin). Other Food and Drug Administration (FDA) to reduce ASCVD
approaches to consider for alleviating symptoms include until 2019, when icosapent ethyl (IPE), a highly puri-
less frequent dosing (e.g., 1 to 3 per week), and/or a trial fied, nonoxidized formulation of eicosapentaenoic acid
of coenzyme Q10 therapy, with addition or substitution of (EPA), received an indication to prevent ASCVD morbid-
nonstatin lipid lowering medications as needed (86). ity and mortality in patients with triglycerides ≥150 mg/
Statins may accelerate hyperglycemia progression dL and established ASCVD or diabetes with ≥2 ASCVD
minimally as an on-target effect (average A1C increase risk factors on maximally tolerated statins (95). The
of ~0.1%), leading to onset of diabetes in those already indication was granted on the strength of the REDUCE-
at highest diabetes risk. Overall, the incidence of statin- IT results; however, because the triglyceride decrease in
associated diabetes is ~10% greater than that with placebo the trial was only 18%, the cardiovascular outcome does
over the usual 2- to 6-year time course of clinical trials not seem to be related to this reduction. Therefore, in
(87). However, in view of the demonstrated benefits of the discussion below, we separate the role of managing
statin therapy for prevention of ASCVD, the leading cause high triglycerides with lifestyle and medications (includ-
of death in patients with diabetes, the risk for new onset ing IPE and other omega-3 fatty acids) from the role of
diabetes or modest worsening of glycemic control in IPE to prevent cardiovascular events, which appears to be
established diabetes does not justify withholding statins in largely, if not entirely independent of its effect on triglyc-
patients with significant ASCVD risk (88,89). erides. This represents a paradigm shift in the management
Statin use may be suspected of triggering less common of people with established ASCVD or those with diabetes
adverse symptoms in individual patients, and, as for at high risk for ASCVD: preventing the next event rather
muscle symptoms, a trial of discontinuation and resump- than simply further controlling traditional cardiovascular
tion of statin therapy can help in assessing the likelihood risk biomarkers.
that such symptoms are statin related. However, there is Management of hypertriglyceridemia should begin
no convincing clinical trial evidence for a significant risk with lifestyle modification, including weight reduction
of hemorrhagic stroke, arthritis, tendonitis, or cataracts, and/or macronutrient modification (i.e., restricted intake of
or for adverse effects on cognition or liver or kidney simple carbohydrates, fat, and alcohol) and increased phys-
function (87). ical activity (see Section V. Lifestyle Recommendations)
(1). Secondary causes of hypertriglyceridemia, including
VIII. Management of Hypertriglyceridemia medical conditions such as diabetes and medications that
and the Role of Icosapent Ethyl increase triglycerides, should be addressed first.
In 2001, the Adult Treatment Panel III (ATP III) of the Pharmacologic agents used to reduce triglycerides
National Cholesterol Education Program suggested a level include fibrates, omega-3 fatty acids, and nicotinic acid
of <150 mg/dL as the therapeutic goal for triglycerides (niacin). Along with their primary LDL-C-lowering effects,
(90). Although this threshold was not very well validated, statins and PCSK9 inhibitors also moderately reduce
it is generally thought to be a reasonable goal for triglycer- triglycerides, and ezetimibe may have a mild triglyceride-
ide reduction and has been adopted by AACE, the National lowering effect (see Section X. Profiles of Medications for
Lipid Association (NLA), and others. Triglycerides may Dyslipidemia).
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 11

Fibrates are generally considered the most potent nations of triglyceride-lowering agents to substantially
triglyceride-lowering agents, with reductions of 45 to 55% reduce triglycerides. Although rare, this disorder is also
(1,35,96,97). Fibrates bind to and activate hepatic peroxi- associated with an increased incidence of acute and chron-
some proliferator-activated receptor alpha, which complex- ic pancreatitis; affected individuals should be referred to a
es with the retinoid X receptor to regulate gene expression lipid specialist.
involved with activation of beta-oxidation of fatty acids, Management of ASCVD risk in people with triglycer-
thus reducing the availability of free fatty acids for triglyc- ides >150 mg/dL should begin with statin therapy, which
eride synthesis for the formation of VLDL (98). Subgroup yields triglyceride reductions of up to 35% (1,106-108).
analyses from various fibrate trials such as ACCORD, Moderate- to high-intensity statin therapy should be initi-
FIELD, and others, though nonsignificant, suggest fibrates ated according to ASCVD risk category (see Section VI.
may confer cardiovascular benefits in patients with triglyc- Treating LDL-C to Goal). Based on the REDUCE-IT
erides ≥200 mg/dL and HDL-C ≤40 mg/dL (1,32,35). findings (34), AACE recommends adding IPE to prevent
Prescription-grade omega-3 fatty acids, which include ASCVD if triglycerides are between 135 and 499 mg/dL
formulations of EPA alone or in combination with doco- on maximally tolerated statin therapy in patients who have
sahexaenoic acid (DHA), are approved to treat triglycer- ASCVD (secondary prevention cohort of REDUCE-IT)
ide elevations ≥500 mg/dL. These agents reduce triglyc- or diabetes plus ≥2 cardiovascular risk factors (primary
erides by approximately 20 to 45%, depending on base- prevention cohort). In REDUCE-IT, IPE 2,000 mg twice
line triglyceride levels (34,95,99,100). Prescription-grade daily was evaluated in patients who had triglycerides of 135
omega-3 fatty acids also demonstrated anti-inflammatory, to 499 mg/dL and LDL-C of 41 to 100 mg/dL on a stable
antioxidant, and antiplatelet mechanisms; reduced platelet statin dose with or without ezetimibe. At a median 4.9
activity; and reduced thrombosis, and EPA seems to also years of follow-up, an 18% reduction from baseline triglyc-
improve plaque stability (1,101). erides in the IPE group was associated with a profoundly
Niacin, or nicotinic acid, reduces triglyceride by 20 to significant 25% relative risk reduction and a 4.8% abso-
30% in a dose-dependent fashion (1). Niacin likely reduces lute risk reduction in the primary composite endpoint of
triglycerides by reducing lipolysis and thereby decreasing cardiovascular death, nonfatal MI, nonfatal stroke, coro-
the supply of free fatty acids, which depresses triglyceride nary revascularization, or unstable angina. Relative risk
synthesis in the liver. Niacin may also reduce the hepatic of the key secondary 3-point composite hard endpoints of
synthesis of apo CIII, thus removing a potent inhibitor of cardiovascular death, nonfatal MI, or nonfatal stroke was
lipoprotein lipase activity. This leads to increased lipo- reduced by 26%, and absolute risk of these endpoints by
protein lipase hydrolysis and clearance of triglycerides, 3.6% (34). Overall, there was a remarkable 30% reduc-
VLDL, IDL, and chylomicrons (102,103). tion in total ASCVD events (109). The mechanism behind
For patients with hypertriglyceridemia who do not the IPE benefits is not clear, as neither its triglyceride-
have established ASCVD or diabetes with ≥2 risk factors, lowering capabilities, its reduction of inflammation, nor
a triglyceride level of <150 mg/dL is the goal. If this cannot its mild effect on platelets can explain the remarkable
be achieved with statin therapy, then a fibrate, omega-3 benefits observed at the end of the trial (34). A prespecified
fatty acid, or niacin may be considered. substudy indicates that the benefits were strongly related
To reduce the risk of acute pancreatitis, a fibrate, to serum levels of EPA. After 1 year, IPE significantly
prescription-grade omega-3 fatty acid (IPE, EPA, or increased serum EPA levels by 386% from baseline. The
EPA-DHA formulation), and/or niacin should be given to on-treatment levels correlated strongly with reductions
all patients with severe hypertriglyceridemia (>500 mg/ in cardiovascular death, MI, stroke, coronary revascular-
dL), with the goal of reducing triglycerides to well below ization, unstable angina, sudden cardiac death, cardiac
500 mg/dL (1,91,104,105). The higher the baseline triglyc- arrest, new heart failure, and all-cause mortality (110).
eride level, the greater the percent and absolute triglyc- Patients whose triglycerides are <150 mg/dL on a
eride lowering, but the less likely any single agent will statin plus IPE should continue lifestyle and statin thera-
sufficiently reduce triglycerides to goal. One may add one py, as indicated, and triglycerides and other lipids should
agent at a time or combinations of agents depending on be monitored every 3 to 12 months, along with diabetes
the degree of triglyceride lowering required (1,91,104). In risk, which is increased in patients with high triglycer-
patients with diabetes, pioglitazone and/or insulin can also ides (1,89). If the patient’s triglycerides remain >150
be utilized in an additive fashion to reduce triglycerides mg/dL on the statin-IPE regimen, a fibrate or niacin may
(1,88,89,105). be considered. Over-the-counter fish oil dietary supple-
Aggressive triglyceride lowering is also required ments are not FDA-approved for lowering triglycerides
to treat chylomicronemia, which usually presents with and are not recommended for this purpose because they
triglycerides >880 mg/dL. This condition may be a sign contain very low amounts of polyunsaturated omega-
of lipoprotein lipase activity deficiency, a manifestation 3 fatty acids as well as trans fatty acids and saturated
of FCS. FCS can be identified by the inability of combi- fat (111-113).
12 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

IX. Assessment and Management established the efficacy and safety profile of statins, which
of Elevated Lipoprotein(a) decrease plasma LDL-C in a dose-dependent fashion by
Increased Lp(a) is the most common genetic dyslipid- 20 to 55%, triglycerides by up to 35%, and raise HDL-C
emia, with a prevalence in the general population of 20%, by 2 to 10% (1). As mentioned, a 2010 meta-analysis by
and is an independent, genetic, and causal risk factor for the CTT strongly confirmed the benefit of LDL-C lowering
ASCVD (MI, stroke, peripheral arterial disease, recurrent with a statin. Robust event reduction correlated with the
events, and calcific aortic stenosis) (14,114-116). It possess- degree of LDL-C lowering (123).
es homology with the fibrin-binding domains of plasmino- Statin therapy is associated with a modestly increased
gen and plasmin with prothrombotic and antifibrinolytic risk of increased glucose levels and new-onset diabetes
effects, as well as the oxidized phospholipids that are (87,124,125). However, any increases in glucose levels
pro-inflammatory. A Lp(a) level >50 mg/dL is associated that occur are not enough to offset the benefits of reduced
with increased risk of recurrent events in patients on statin cardiovascular morbidity and mortality. Despite initial
therapy (117). observations of elevated liver enzymes in clinical trials,
Measurement of Lp(a) in patients should be consid- the FDA has concluded that statins as a class do not have
ered in the following settings: negative impacts on the liver, and liver monitoring is not
• All patients with clinical ASCVD, especially prema- necessary (126). However, statins are contraindicated
ture or recurrent ASCVD despite LDL-C lowering; in active liver disease (127). See Section VII, Managing
• Individuals with a family history of premature ASCVD Statin Intolerance and Safety, for a discussion of other side
and/or increased Lp(a); effects such as statin intolerance and myalgias.
• Individuals with South Asian or African ancestry, espe- The unique characteristics of various statins emerge
cially with a family history of ASCVD or increased from differences in their metabolism, bioavailability,
Lp(a); potency, and duration of action. For example, some statins
• Individuals with a 10-year ASCVD risk ≥10% (prima- are potent inhibitors of cytochrome P450, which can lead to
ry prevention setting), in order to stratify risk; interactions with other agents such as cyclosporin, rifampin,
• Patients with a personal or family history of aortic protease inhibitors, and other agents. For more details,
valve stenosis; see the AACE/ACE 2017 Guidelines for Management
• Patients with refractory elevations of LDL-C despite of Dyslipidemia and Prevention of Cardiovascular
aggressive LDL-C-lowering therapy (i.e., statin resis- Disease (1).
tance). Cholesterol absorption inhibitors primarily reduce
No medications are FDA approved to lower Lp(a). LDL-C by 10 to 25% and may also have beneficial effects
Agents that have been demonstrated to reduce Lp(a) levels on triglycerides, apo B, and HDL-C. These effects are
include PCSK9 inhibitors, niacin, oral estrogen, and aspi- often synergistic in combination therapy with statins,
rin (118-121). Outcomes trials focusing on selective lower- colesevelam, or BA. Ezetimibe is the only member of this
ing of Lp(a) by an antisense oligonucleotide are underway. drug class; it acts by reducing cholesterol absorption at
Lipoprotein apheresis, although infrequently performed the brush border of enterocytes via cholesterol transporter
today, can be considered in extreme cases (122). Currently, interference (1). The IMPROVE-IT trial demonstrated a
recommended treatment of patients with elevated Lp(a) is significant reduction in the ASCVD endpoints in individu-
aggressive lowering of LDL-C, which mitigates the risk als treated with ezetimibe plus simvastatin versus simvas-
associated with the Lp(a) elevation (1). tatin alone (26). Ezetimibe has minimal adverse effects and
a strong safety profile (1).
X. Profiles of Medications for Dyslipidemia Alirocumab and evolocumab are monoclonal antibod-
The pharmacologic management of dyslipidemia ies that inhibit PCSK9, a protein that regulates the recy-
requires a comprehensive strategy to control lipid levels cling of LDL, thereby reducing LDL-C up to 71% when
and more importantly reduce cardiovascular events. This added to maximum statin therapy or given as monotherapy
field is constantly evolving as new studies are published (1,73,74). Given by subcutaneous injection every 2 to 4
and new agents are developed. weeks, they have a favorable safety and tolerability profile.
Statins (i.e., HMG-CoA reductase inhibitors) are the They have demonstrated favorable ASCVD outcomes,
initial medication of choice for LDL-C reduction. The best- especially in high-risk patients (24,25), although high cost
studied class of lipid lowering medications, they signifi- and need for injections have limited their use.
cantly and consistently improve ASCVD outcomes, not At full dosage, bile acid sequestrants reduce LDL-C
just lipid values. Statins have a relatively low side effect by 15 to 25% and increase HDL-C by 4 to 11%. In patients
profile and have been studied across all ages and comor- with elevated triglycerides, they may further increase
bidities. Currently available options include atorvastatin, triglycerides. In fact, colesevelam is contraindicated in
fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvas- patients with triglycerides >500 mg/dL. One agent, cole-
tatin, and simvastatin. Numerous large clinical trials have sevelam, is approved for glucose-lowering therapy in
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 13

adults with type 2 diabetes. They can be added to a choles- fibrate, and fenofibric acid. Fibrates lower triglycerides by
terol absorption inhibitor to achieve LDL-C reductions up to 55% and increase HDL-C by 6 to 18% (1,35,96,97).
comparable to moderate-dose statins. However, there are Outcomes studies with fibrates have had mixed results,
no outcomes data as of this time. Important side effects especially in statin-treated populations and in individuals
are bloating, constipation, and interference with absorp- treated with fibrates who have less severe triglyceride and
tion and action of other medications such as thyroid HDL-C abnormalities (32,35,39,93). Maximum dosages
hormone (1). of certain statin-fibrate combinations such as simvastatin
BA is a novel LDL-C-lowering agent that is effective with fenofibrate and other statins with gemfibrozil need to
in combination with statins and with ezetimibe, acting in be considered. There is a small but significant increase in
a fashion that potentiates statin action. In clinical trials, creatinine with fibrates (1). However, despite initially and
BA reduced LDL-C by 15 to 24% (128-131). When given reversibly increasing serum creatinine, fenofibrate reduced
as a fixed-dose combination, BA plus ezetimibe reduced albuminuria and slowed eGFR loss over 5 years (133).
LDL-C by 36% compared with a 2% increase with placebo Niacin is a potent LDL-C- and triglyceride-lowering
(132). No evidence is, as yet, available on cardiovascular medication that also substantially increases HDL-C. Niacin
outcomes benefit. BA does not lead to increases in glucose lowers LDL-C by up to 20% in a dose-dependent manner.
concentrations or the development of diabetes (131). Uric In combination with statins or bile acid sequestrants,
acid was increased in 26% of treated patients versus 10% niacin has been associated with angiographic evidence
of placebo; 11% of persons having a past history of gout of reduced progression and some regression of atheroma-
versus 2% of placebo had an acute gout episode, which tous plaques (1). However, large-scale clinical trials such
in the total trial population was 1.5% on BA versus 0.4% as Atherothrombosis Intervention in Metabolic Syndrome
on placebo. Another infrequent but noteworthy complica- With Low HDL/High Triglycerides (AIM-HIGH) have
tion is tendon rupture. Tendon rupture in the rotator cuff, not demonstrated cardiovascular benefit with niacin when
biceps tendon, or Achilles tendon may occur more often in individuals are well-controlled on statin therapy, and in
people who are older, have CKD, or have been treated with some instances, the combination may increase the side
glucocorticoids or with fluoroquinolone antibiotics. BA is effect profile (134,135).
contraindicated in combination with simvastatin >20 mg Blood glucose elevations have been associated with
and pravastatin >40 mg (71). higher dosages of niacin, particularly in individuals with
IPE, a purified formulation of the omega-3 fatty acid diabetes, but these are mild and temporary. Flushing
EPA, is indicated to reduce cardiovascular risk among may occur, especially at the beginning of niacin therapy;
patients with elevated triglyceride levels as an add-on however, this effect often diminishes with continued use.
to maximally tolerated statin therapy, as established by Flushing occurs less frequently with extended-release
REDUCE-IT. In this trial, IPE reduced the risk of cardio- niacin and can be ameliorated by pretreating with aspirin
vascular events (cardiovascular death, nonfatal MI, nonfatal or a nonsteroidal anti-inflammatory agent and by slowly
stroke, coronary revascularization, or unstable angina) by titrating the dosage upward. Hepatoxicity is associated
25% and cardiovascular death by 20%, which correspond- with sustained release niacin. Increases in uric acid and
ed to absolute risk reductions of 4.8% and 0.9%, respec- gout can occur, along with an increased possibility of statin
tively, at 4.9 years in patients with hypertriglyceridemia muscle toxicity (1). Niacin has been shown to decrease
who were treated with IPE and maximally tolerated statins Lp(a), although the clinical impact of this is unknown and
(see Section VIII. Management of Hypertriglyceridemia may only occur with certain phenotypic expression (136).
and the Role of Icosapent Ethyl) (34). In clinical trials, IPE Lomitapide, a microsomal triglyceride transfer protein
was associated with muscle and joint pain, swelling of the (MTP) inhibitor, is a treatment option for HoFH. This
extremities, constipation, gout, increased bleeding, and agent, which may be associated with hepatotoxicity, is
increased risk of atrial fibrillation or flutter (1). available with restricted distribution and may be useful for
Both IPE and combination omega-3-acid ethyl esters, individuals with HoFH not responsive to PCSK9 inhibitor
in which EPA is coupled with DHA, are indicated as therapy (137). It is usually only utilized by lipid experts.
adjuncts to diet for the reduction of triglyceride levels in Another HoFH treatment, mipomersen (an antisense apo
adults with severe (≥500 mg/dL) hypertriglyceridemia. B oligonucleotide), was recently withdrawn from the
It should be noted that these effects are for prescription U.S. market. Treatment of HoFH is outside the scope of
strength omega-3 fatty acids and not over-the-counter this algorithm.
supplements (1).
Fibrates are effective for treating individuals with ACKNOWLEDGMENT
severe hypertriglyceridemia and for individuals at risk of
ASCVD who have elevated triglycerides and low HDL-C We thank Amanda M. Justice, BA, for providing edito-
levels as their primary lipid abnormality. Currently avail- rial support and medical writing assistance in the prepara-
able fibrates are gemfibrozil, fenofibrate, micronized feno- tion of this document.
14 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

DISCLOSURES prevention of atherosclerosis. Endocr Pract. 2017;23(suppl 2):


1-87.
2. Bhatnagar P, Wickramasinghe K, Wilkins E, Townsend N.
Chair Trends in the epidemiology of cardiovascular disease in the UK.
Yehuda Handelsman, MD, FACP, FNLA, MACE: Heart. 2016;102:1945-1952.
3. Virani SS, Alonso A, Benjamin EJ, et al. Heart disease and
Amarin, BI-Lilly, Sanofi, consultant and/or speaker; stroke statistics–2020 update: a report from the American Heart
Amgen, Applied Therapeutics, AstraZeneca, BMS, Gan & Association. Circulation. 2020;141:e139-e596.
Lee, Novo Nordisk, Sanofi, research grant support. 4. Toth PP, Fazio S, Wong ND, Hull M, Nichols GA. Risk of cardio-
vascular events in patients with hypertriglyceridaemia: A review of
real-world evidence. Diabetes Obes Metab. 2020;22:279-289.
Co-Chairs 5. Ference BA, Ginsberg HN, Graham I, et al. Low-density lipo-
Paul S. Jellinger, MD, MACE: Amgen, Amarin, proteins cause atherosclerotic cardiovascular disease. 1. Evidence
from genetic, epidemiologic, and clinical studies. A consensus
AstraZeneca, Esperion, speaker. statement from the European Atherosclerosis Society Consensus
Chris Guerin, MD, FACE: Novo Nordisk, speaker. Panel. Eur Heart J. 2017;38:2459-2472.
6. Hales CM, Servais J, Martin CB, Kohen D. Prescription drug
use among adults aged 40-79 in the United States and Canada.
Task Force Members NCHS Data Brief. 2019:1-8.
Zachary Bloomgarden, MD, MACE: Novartis, 7. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of
stock ownership. obesity and severe obesity among adults: United States, 2017–
2018. NCHS Data Brief. 2020;360:1-7.
Eliot A. Brinton, MD, FAHA, FNLA, FACE: 8. Centers for Disease Control and Prevention. Chronic kidney
Amarin, Amgen, Kowa, consultant and/or speaker; Kowa. disease surveillance system–United States. Available at: https://
Matthew Budoff, MD: AstraZeneca, Boehringer nccd.cdc.gov/CKD/. Accessed August 13, 2020.
9. Centers for Disease Control and Prevention. Diabetes atlas.
Ingelheim, Janssen, speaker; AstraZeneca, Novo Nordisk, Available at: https://gis.cdc.gov/grasp/diabetes/DiabetesAtlas.
Boehringer Ingelheim, research grant support. html#. Accessed August 13, 2020.
Michael Davidson, MD: Amgen, Regeneron, Sanofi, 10. Navar-Boggan AM, Peterson ED, D’Agostino RB, Sr., Neely B,
Sniderman AD, Pencina MJ. Hyperlipidemia in early adulthood
AstraZeneca, Novo Nordisk, consultant and/or speaker; increases long-term risk of coronary heart disease. Circulation.
Chief Medical Officer, Corvidia Therapeutics. 2015;131:451-458.
Alan J. Garber, MD, PhD, MACE; Donald Smith, 11. National Cholesterol Education Program. Third Report of the
National Cholesterol Education Program (NCEP) Expert Panel on
MD, MPH, FACE; Jeffrey I. Mechanick, MD, FACP, Detection, Evaluation, and Treatment of High Blood Cholesterol
FACN, ECNU, MACE; Kathleen Wyne, MD, PhD, in Adults (Adult Treatment Panel III) final report. Circulation.
FNLA, FACE; Ronald Krauss, MD: report no potential 2002;106:3143-3421.
12. Brunzell JD, Davidson M, Furberg CD, et al. Lipoprotein
conflicts of interest. management in patients with cardiometabolic risk: consensus state-
Daniel Einhorn, MD, FACP, FACE: Lilly, Novo ment from the American Diabetes Association and the American
Nordisk, Sanofi, AstraZeneca, Janssen, Abbott, Boehringer College of Cardiology Foundation. Diabetes Care. 2008;31:
811-822.
Ingelheim, consultant, speaker, and/or research grant 13. Yuan G, Al-Shali KZ, Hegele RA. Hypertriglyceridemia: its
support. etiology, effects and treatment. CMAJ. 2007;176:1113-1120.
Sergio Fazio, MD, PhD: Amgen, Esperion, consul- 14. Wilson DP, Jacobson TA, Jones PH, et al. Use of Lipoprotein(a)
in clinical practice: A biomarker whose time has come. A scien-
tant. tific statement from the National Lipid Association. J Clin Lipidol.
Vivian Fonseca, MD: Amgen, Microbiome 2019;13:374-392.
Technologies, Insulin Algorithms, BRAVO4Health, stock 15. Gaddi A, Cicero AF, Odoo FO, Poli AA, Paoletti R. Practical
guidelines for familial combined hyperlipidemia diagnosis: an
ownership; Bayer, Boehringer Ingelheim, research grant up-date. Vasc Health Risk Manag. 2007;3:877-886.
support; Gilead, Takeda, Novo Nordisk, Sanofi-Aventis, 16. Raal FJ, Santos RD. Homozygous familial hypercholesterolemia:
Abbott, AstraZeneca, Intarcia, Asahi, consultant and/or current perspectives on diagnosis and treatment. Atherosclerosis.
2012;223:262-268.
speaker. 17. Davidson MH. Dyslipidemia. 2019. Available at: www.merck
George Grunberger, MD, FACP, FACE: Lilly, manuals.com.
Sanofi, Novo Nordisk, speaker. 18. Vodnala D, Rubenfire M, Brook RD. Secondary causes of
dyslipidemia. Am J Cardiol. 2012;110:823-825.
Paul D. Rosenblit, MD, PhD, FACE, FNLA: 19. Gepner AD, Young R, Delaney JA, et al. Comparison of coro-
Dexcom, Ionis (Akcea), Lilly, Mylan., Novo Nordisk, clin- nary artery calcium presence, carotid plaque presence, and carotid
ical research trial site support. intima-media thickness for cardiovascular disease prediction in the
Multi-Ethnic Study of Atherosclerosis. Circ Cardiovasc Imaging.
Amanda M. Justice (medical writer): Asahi Kasei 2015;8:e002262.
Pharma Corp., Lexicon Pharmaceuticals Inc., Metavant 20. Lloyd-Jones DM, Wilson PW, Larson MG, et al. Framingham
Sciences Inc., and Sanofi. risk score and prediction of lifetime risk for coronary heart disease.
Am J Cardiol. 2004;94:20-24.
21. Ridker PM, Buring JE, Rifai N, Cook NR. Development and
validation of improved algorithms for the assessment of global
REFERENCES cardiovascular risk in women: the Reynolds Risk Score. JAMA.
2007;297:611-619.
1. Jellinger PS, Handelsman Y, Rosenblit PD, et al. American 22. Stevens RJ, Kothari V, Adler AI, Stratton IM. The UKPDS risk
Association of Clinical Endocrinologists and American College engine: a model for the risk of coronary heart disease in type II
of Endocrinology guidelines for management of dyslipidemia and diabetes (UKPDS 56). Clin Sci. 2001;101:671-679.
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 15

23. Goff DC Jr, Lloyd-Jones DM, Bennett G, et al. 2013 ACC/AHA 41. Chiavaroli L, Viguiliouk E, Nishi SK, et al. DASH dietary
guideline on the assessment of cardiovascular risk: a report of the pattern and cardiometabolic outcomes: an umbrella review of
American College of Cardiology/American Heart Association systematic reviews and meta-analyses. Nutrients. 2019;11:338.
Task Force on Practice Guidelines. Circulation. 2013;129: 42. Bazzano LA, He J, Ogden LG, et al. Fruit and vegetable intake
S49-S73. and risk of cardiovascular disease in US adults: the first National
24. Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and cardio- Health and Nutrition Examination Survey Epidemiologic Follow-
vascular outcomes after acute coronary syndrome. N Engl J Med. up Study. Am J Clin Nutr. 2002;76:93-99.
2018;379:2097-2107. 43. Esselstyn CB, Jr., Gendy G, Doyle J, Golubic M, Roizen MF.
25. Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and A way to reverse CAD? J Fam Pract. 2014;63:356b-364b.
clinical outcomes in patients with cardiovascular disease. N Engl J 44. Ornish D, Scherwitz LW, Billings JH, et al. Intensive life-
Med. 2017;376:1713-1722. style changes for reversal of coronary heart disease. JAMA.
26. Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added 1998;280:2001-2007.
to statin therapy after acute coronary syndromes. N Engl J Med. 45. Crowe FL, Appleby PN, Travis RC, Key TJ. Risk of hospitaliza-
2015;372:2387-2397. tion or death from ischemic heart disease among British vegetar-
27. Heart Protection Study Collaborative Group. MRC/BHF Heart ians and nonvegetarians: results from the EPIC-Oxford cohort
Protection Study of cholesterol lowering with simvastatin in study. Am J Clin Nutr. 2013;97:597-603.
20,536 high-risk individuals: a randomised placebo-controlled 46. Campbell T. A plant-based diet and stroke. J Geriatr Cardiol.
trial. Lancet. 2002;360:7-22. 2017;14:321-326.
28. Shepherd J, Blauw GJ, Murphy MB, et al. Pravastatin in elderly 47. Zeisel SH, Warrier M. Trimethylamine N-oxide, the microbiome,
individuals at risk of vascular disease (PROSPER): a randomised and heart and kidney disease. Annu Rev Nutr. 2017;37:157-181.
controlled trial. Lancet. 2002;360:1623-1630. 48. U.S. Department of Health and Human Services. Physical
29. Sever PS, Dahlof B, Poulter NR, et al. Prevention of coronary Activity Guidelines for Americans. 2nd ed. Washington, DC: U.S.
and stroke events with atorvastatin in hypertensive patients who Department of Health and Human Services; 2018.
have average or lower-than-average cholesterol concentrations, in 49. Jakicic JM, Wing RR, Butler BA, Robertson RJ. Prescribing
the Anglo-Scandinavian Cardiac Outcomes Trial--Lipid Lowering exercise in multiple short bouts versus one continuous bout: effects
Arm (ASCOT-LLA): a multicentre randomised controlled trial. on adherence, cardiorespiratory fitness, and weight loss in over-
Lancet. 2003;361:1149-1158. weight women. Int J Obes Relat Metab Disord. 1995;19:893-901.
30. Ridker PM, Morrow DA, Rose LM, Rifai N, Cannon CP, 50. Levinger I, Goodman C, Hare DL, Jerums G, Selig S. The effect
Braunwald E. Relative efficacy of atorvastatin 80 mg and pravas- of resistance training on functional capacity and quality of life in
tatin 40 mg in achieving the dual goals of low-density lipoprotein individuals with high and low numbers of metabolic risk factors.
cholesterol <70 mg/dL and C-reactive protein <2 mg/L: an analy- Diabetes Care. 2007;30:2205-2210.
sis of the PROVE-IT TIMI-22 trial. J Am Coll Cardiol. 2005;45: 51. Kelley GA, Kelley KS. Impact of progressive resistance training
1644-1648. on lipids and lipoproteins in adults: a meta-analysis of randomized
31. Barter PJ, Ballantyne CM, Carmena R, et al. Apo B versus controlled trials. Prev Med. 2009;48:9-19.
cholesterol in estimating cardiovascular risk and in guiding ther- 52. McNeil J, Doucet E, Chaput JP. Inadequate sleep as a contributor
apy: report of the thirty-person/ten-country panel. J Intern Med. to obesity and type 2 diabetes. Can J Diabetes. 2013;37:103-108.
2006;259:247-258. 53. Cappuccio FP, Cooper D, D’Elia L, Strazzullo P, Miller MA.
32. Tenkanen L, Manttari M, Kovanen PT, Virkkunen H, Sleep duration predicts cardiovascular outcomes: a systematic
Manninen V. Gemfibrozil in the treatment of dyslipidemia: an review and meta-analysis of prospective studies. Eur Heart J.
18-year mortality follow-up of the Helsinki Heart Study. Arch 2011;32:1484-1492.
Intern Med. 2006;166:743-748. 54. Patel SR, Malhotra A, White DP, Gottlieb DJ, Hu FB.
33. Saito Y, Yokoyama M, Origasa H, et al. Effects of EPA on coro- Association between reduced sleep and weight gain in women. Am
nary artery disease in hypercholesterolemic patients with multiple J Epidemiol. 2006;164:947-954.
risk factors: sub-analysis of primary prevention cases from the 55. Gottlieb DJ, Redline S, Nieto FJ, et al. Association of usual sleep
Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis. duration with hypertension: the Sleep Heart Health Study. Sleep.
2008;200:135-140. 2006;29:1009-1014.
34. Bhatt DL, Steg PG, Miller M, et al. Cardiovascular risk reduc- 56. Chaput JP, Despres JP, Bouchard C, Tremblay A. Short sleep
tion with icosapent ethyl for hypertriglyceridemia. N Engl J Med. duration is associated with reduced leptin levels and increased
2019;380:11-22. adiposity: Results from the Quebec family study. Obesity.
35. Keech A, Simes RJ, Barter P, et al. Effects of long-term fenofi- 2007;15:253-261.
brate therapy on cardiovascular events in 9795 people with type 2 57. Ayas NT, White DP, Manson JE, et al. A prospective study of
diabetes mellitus (the FIELD study): randomised controlled trial. sleep duration and coronary heart disease in women. Arch Intern
Lancet. 2005;366:1849-1861. Med. 2003;163:205-209.
36. Arbel Y, Klempfner R, Erez A, et al. Bezafibrate for the treat- 58. De Hert M, Detraux J, Vancampfort D. The intriguing rela-
ment of dyslipidemia in patients with coronary artery disease: tionship between coronary heart disease and mental disorders.
20-year mortality follow-up of the BIP randomized control trial. Dialogues Clin Neurosci. 2018;20:31-40.
Cardiovasc Diabetol. 2016;15:11. 59. Sin NL. The Protective Role of Positive Well-being in cardiovas-
37. Bezafibrate Infarction Prevention (BIP) Study. Secondary cular disease: review of current evidence, mechanisms, and clini-
prevention by raising HDL cholesterol and reducing triglycerides cal implications. Curr Cardiol Rep. 2016;18:106.
in patients with coronary artery disease. Circulation. 2000;102: 60. Corrao G, Bagnardi V, Zambon A, La Vecchia C. A meta-anal-
21-27. ysis of alcohol consumption and the risk of 15 diseases. Prev Med.
38. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the 2004;38:613-619.
secondary prevention of coronary heart disease in men with low 61. Costanzo S, Di Castelnuovo A, Donati MB, Iacoviello L, de
levels of high-density lipoprotein cholesterol. Veterans Affairs Gaetano G. Cardiovascular and overall mortality risk in relation
High-Density Lipoprotein Cholesterol Intervention Trial Study to alcohol consumption in patients with cardiovascular disease.
Group. N Engl J Med. 1999;341:410-418. Circulation. 2010;121:1951-1959.
39. Robins SJ, Collins D, Wittes JT, et al. Relation of gemfibrozil 62. American Heart Association. Is drinking alcohol part of a healthy
treatment and lipid levels with major coronary events: VA-HIT: lifestyle? 2019. Available at: https://www.heart.org/en/healthy-
a randomized controlled trial. JAMA. 2001;285:1585-1591. living/healthy-eating/eat-smart/nutrition-basics/alcohol-and-heart-
40. Estruch R, Ros E, Salas-Salvado J, et al. Primary prevention of health. Accessed August 13, 2020.
cardiovascular disease with a Mediterranean diet supplemented 63. Kim ST, Park T. Acute and chronic effects of cocaine on cardio-
with extra-virgin olive oil or nuts. N Engl J Med. 2018;378:e34. vascular health. Int J Mol Sci. 2019;20:584.
16 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)

64. U.S. Department of Health and Human Services. The Health and American College of Endocrinology on the Comprehensive
Consequences of Smoking–50 Years of Progress: A Report of the Type 2 Diabetes Management Algorithm–2020 Executive
Surgeon General. Rockville, MD: U.S. Department of Health and Summary. Endocr Pract. 2020;26:107-139.
Human Services; 2014. Available at: https://pubmed.ncbi.nlm.nih. 89. Handelsman Y, Bloomgarden ZT, Grunberger G, et al.
gov/24455788/. Accessed August 13, 2020. American Association of Clinical Endocrinologists and American
65. Cholesterol Treatment Trialists (CTT) Collaboration, Baigent College of Endocrinology: clinical practice guidelines for devel-
C, Blackwell L, et al. Efficacy and safety of more intensive lower- oping a diabetes mellitus comprehensive care plan–2015. Endocr
ing of LDL cholesterol: a meta-analysis of data from 170,000 Pract. 2015;21:1-87.
participants in 26 randomised trials. Lancet. 2010;376:1670-1681. 90. Expert Panel on Detection, Evaluation, and Treatment of High
66. Boekholdt SM, Hovingh GK, Mora S, et al. Very low levels of Blood Cholesterol in Adults. Executive Summary of The Third
atherogenic lipoproteins and the risk for cardiovascular events: a Report of The National Cholesterol Education Program (NCEP)
meta-analysis of statin trials. J Am Coll Cardiol. 2014;64:485-494. Expert Panel on Detection, Evaluation, and Treatment of High
67. Cholesterol Treatment Trialists (CTT) Collaborators, Kearney Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA.
PM, Blackwell L, et al. Efficacy of cholesterol-lowering therapy 2001;285:2486-2497.
in 18,686 people with diabetes in 14 randomised trials of statins: a 91. Nordestgaard BG, Varbo A. Triglycerides and cardiovascular
meta-analysis. Lancet. 2008;371:117-125. disease. Lancet. 2014;384:626-635.
68. de Lemos JA, Blazing MA, Wiviott SD, et al. Early intensive 92. Ference BA, Kastelein JJP, Ray KK, et al. Association of
vs a delayed conservative simvastatin strategy in patients with triglyceride-lowering LPL variants and LDL-C-lowering LDLR
acute coronary syndromes: phase Z of the A to Z trial. JAMA. variants with risk of coronary heart disease. JAMA. 2019;321:
2004;292:1307-1316. 364-373.
69. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild to 93. ACCORD Study Group, Ginsberg HN, Elam MB, et al. Effects
moderate muscular symptoms with high-dosage statin therapy of combination lipid therapy in type 2 diabetes mellitus. N Engl J
in hyperlipidemic patients–the PRIMO study. Cardiovasc Drugs Med. 2010;362:1563-1574.
Ther. 2005;19:403-414. 94. FIELD Study Investigators. Effects of long-term fenofibrate
70. Giugliano RP, Pedersen TR, Park JG, et al. Clinical efficacy therapy on cardiovascular events in 9795 people with type 2
and safety of achieving very low LDL-cholesterol concentrations diabetes mellitus (the FIELD study): randomised controlled trial.
with the PCSK9 inhibitor evolocumab: a prespecified secondary Lancet. 2005;366:1849-1861.
analysis of the FOURIER trial. Lancet. 2017;390:1962-1971. 95. Vascepa (icosapent ethyl) prescribing information. Bridgewater,
71. Nexletol (bempedoic acid) prescribing information. Ann Arbor, NJ: Amarin Pharma, Inc.; 2019.
MI: Esperion Therapeutics, Inc.; 2020. 96. Woo Y, Shin JS, Shim CY, et al. Effect of fenofibrate in 1113
72. Welchol (colesevelam hydrochloride) prescribing information. patients at low-density lipoprotein cholesterol goal but high
Basking Ridge, NJ: Daiichi Sankyo, Inc.; 2019. triglyceride levels: Real-world results and factors associated with
73. Praluent (alirocumab) prescribing information. Bridgewater, NJ: triglyceride reduction. PLoS One. 2018;13:e0205006.
sanofi-aventis U.S. LLC; 2020. 97. Nerbrand C, Nyberg P, Nordstrom L, Samsioe G. Effects of a
74. Repatha (evolocumab) prescribing information. Thousand Oaks, lipid lowering fibrate and hormone replacement therapy on serum
CA: Amgen Inc.; 2019. lipids and lipoproteins in overweight postmenopausal women with
75. Zetia (ezetimibe) prescribing information. Whitehouse Station, elevated triglycerides. Maturitas. 2002;42:55-62.
NJ: Merck & Co., Inc.; 2013. 98. Chapman MJ, Ginsberg HN, Amarenco P, et al. Triglyceride-
76. Crestor (rosuvastatin) prescribing information. Wilmington, DE: rich lipoproteins and high-density lipoprotein cholesterol in
AstraZeneca; 2018. patients at high risk of cardiovascular disease: evidence and guid-
77. Lipitor (atorvastatin) prescribing information. New York, NY: ance for management. Eur Heart J. 2011;32:1345-1361.
Pfizer Inc; 2019. 99. Davidson MH, Stein EA, Bays HE, et al. Efficacy and tolerabil-
78. Zocor (simvastatin) prescribing information. Whitehouse Station, ity of adding prescription omega-3 fatty acids 4 g/d to simvastatin
NJ: Merck & Co., Inc.; 2019. 40 mg/d in hypertriglyceridemic patients: an 8-week, random-
79. Livalo (pitavastatin) prescribing information. Montgomery, AL: ized, double-blind, placebo-controlled study. Clin Ther. 2007;29:
Kowa Pharmaceuticals America, Inc.; 2019. 1354-1367.
80. Lescol/Lescol XL (fluvastatin/fluvastatin extended release) 100. Lovaza (omega-3-acid ethyl esters) prescribing information.
prescribing information. East Hanover, NJ: Novartis; 2017. Research Triangle Park, NC: GlaxoSmithKline; 2019.
81. Pravachol (pravastatin) prescribing information. Princeton, NJ: 101. Calder PC. Omega-3 polyunsaturated fatty acids and inflamma-
Bristol-Meyers-Squibb Company; 2016. tory processes: nutrition or pharmacology? Br J Clin Pharmacol.
82. Mevacor (lovastatin) prescribing information. Morgantown, WV: 2013;75:645-662.
Mylan Pharmaceuticals Inc.; 2012. 102. Song WL, FitzGerald GA. Niacin, an old drug with a new twist.
83. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, preven- J Lipid Res. 2013;54:2586-2594.
tion, and management of statin adverse effects and intolerance: 103. Superko HR, Zhao XQ, Hodis HN, Guyton JR. Niacin and heart
Canadian Consensus Working Group update (2016). Can J disease prevention: Engraving its tombstone is a mistake. J Clin
Cardiol. 2016;32:S35-S65. Lipidol. 2017;11:1309-1317.
84. Newman CB, Preiss D, Tobert JA, et al. Statin safety and asso- 104. Zafrir B, Jubran A, Hijazi R, Shapira C. Clinical features and
ciated adverse events: a scientific statement from the American outcomes of severe, very severe, and extreme hypertriglyceridemia
Heart Association. Arterioscler Thromb Vasc Biol. 2019;39: in a regional health service. J Clin Lipidol. 2018;12:928-936.
e38-e81. 105. Christian JB, Arondekar B, Buysman EK, Jacobson TA,
85. Stroes ES, Thompson PD, Corsini A, et al. Statin-associated Snipes RG, Horwitz RI. Determining triglyceride reductions
muscle symptoms: impact on statin therapy–European needed for clinical impact in severe hypertriglyceridemia. Am J
Atherosclerosis Society Consensus Panel statement on assessment, Med. 2014;127:36-44.
aetiology and management. Eur Heart J. 2015;36:1012-1022. 106. Jones PH, Davidson MH, Stein EA, et al. Comparison of the
86. Qu H, Guo M, Chai H, Wang WT, Gao ZY, Shi DZ. Effects efficacy and safety of rosuvastatin versus atorvastatin, simvastatin,
of coenzyme Q10 on statin-induced myopathy: an updated meta- and pravastatin across doses (STELLAR Trial). Am J Cardiol.
analysis of randomized controlled trials. J Am Heart Assoc. 2003;92:152-160.
2018;7:e009835. 107. Jones P, Kafonek S, Laurora I, Hunninghake D. Comparative
87. Mach F, Ray KK, Wiklund O, et al. Adverse effects of statin dose efficacy study of atorvastatin versus simvastatin, pravastatin,
therapy: perception vs. the evidence–focus on glucose homeosta- lovastatin, and fluvastatin in patients with hypercholesterolemia
sis, cognitive, renal and hepatic function, haemorrhagic stroke and (the CURVES Study). Am J Cardiol. 1998;81:582-587.
cataract. Eur Heart J. 2018;39:2526-2539. 108. Diabetes Atorvastin Lipid Intervention (DALI) Study Group.
88. Garber AJ, Handelsman Y, Grunberger G, et al. Consensus The effect of aggressive versus standard lipid lowering by atorv-
statement by the American Association of Clinical Endocrinologists astatin on diabetic dyslipidemia: the DALI study: a double-blind,
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 17

randomized, placebo-controlled trial in patients with type 2 diabe- 124. Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes
tes and diabetic dyslipidemia. Diabetes Care. 2001;24:1335-1341. with intensive-dose compared with moderate-dose statin therapy:
109. Bhatt DL, Steg PG, Miller M, et al. Effects of icosapent ethyl a meta-analysis. JAMA. 2011;305:2556-2564.
on total ischemic events: From REDUCE-IT. J Am Coll Cardiol. 125. Cederberg H, Stancakova A, Yaluri N, Modi S, Kuusisto J,
2019;73:2791-2802. Laakso M. Increased risk of diabetes with statin treatment is
110. Bhatt DL, Miller M, Steg G, et al. EPA levels and cardiovascu- associated with impaired insulin sensitivity and insulin secretion:
lar outcomes in the reduction of cardiovascular events with icosa- a 6 year follow-up study of the METSIM cohort. Diabetologia.
pent ethyl–intervention trial (REDUCE-IT). Presented at the 69th 2015;58:1109-1117.
Annual Convocation of the American College of Cardiology. 2020. 126. US Food and Drug Administration. FDA drug safety commu-
Available at: https://www.acc.org/education-and-meetings/image- nication: important safety label changes to cholesterol-lowering
and-slide-gallery/media-detail?id=062fc9e4b3a74a9fb1c196a35d statin drugs. 2012. Available at: https://www.fda.gov/drugs/
ad8f3b. Accessed August 13, 2020. drug-safety-and-availability/fda-drug-safety-communication-
111. Harris WS. n-3 fatty acids and serum lipoproteins: human studies. important-safety-label-changes-cholesterol-lowering-statin-drugs.
Am J Clin Nutr. 1997;65:1645S-1654S. Accessed August 13, 2020.
112. Pownall HJ, Brauchi D, Kilinc C, et al. Correlation of serum 127. Vargas JI, Arrese M, Shah VH, Arab JP. Use of statins in
triglyceride and its reduction by omega-3 fatty acids with lipid patients with chronic liver disease and cirrhosis: current views and
transfer activity and the neutral lipid compositions of high-density prospects. Curr Gastroenterol Rep. 2017;19:43.
and low-density lipoproteins. Atherosclerosis. 1999;143:285-297. 128. Ballantyne CM, Banach M, Mancini GBJ, et al. Efficacy and
113. Sherratt SCR, Lero M, Mason RP. Are dietary fish oil supple- safety of bempedoic acid added to ezetimibe in statin-intolerant
ments appropriate for dyslipidemia management? A review of the patients with hypercholesterolemia: A randomized, placebo-
evidence. Curr Opin Lipidol. 2020;31:94-100. controlled study. Atherosclerosis. 2018;277:195-203.
114. Erqou S, Kaptoge S, Perry PL, et al. Lipoprotein(a) concentra- 129. Ray KK, Bays HE, Catapano AL, et al. Safety and efficacy
tion and the risk of coronary heart disease, stroke, and nonvascular of bempedoic acid to reduce ldl cholesterol. N Engl J Med.
mortality. JAMA. 2009;302:412-423. 2019;380:1022-1032.
115. Willeit P, Kiechl S, Kronenberg F, et al. Discrimination and net 130. Goldberg AC, Leiter LA, Stroes ESG, et al. Effect of bempedoic
reclassification of cardiovascular risk with lipoprotein(a): prospec- acid vs placebo added to maximally tolerated statins on low-densi-
tive 15-year outcomes in the Bruneck Study. J Am Coll Cardiol. ty lipoprotein cholesterol in patients at high risk for cardiovascular
2014;64:851-860. disease: The CLEAR Wisdom randomized clinical trial. JAMA.
116. Tsimikas S. Lipoprotein(a): novel target and emergence of 2019;322:1780-1788.
novel therapies to lower cardiovascular disease risk. Curr Opin 131. Burke AC, Telford DE, Huff MW. Bempedoic acid: effects on
Endocrinol Diabetes Obes. 2016;23:157-164. lipoprotein metabolism and atherosclerosis. Curr Opin Lipidol.
117. Willeit P, Ridker PM, Nestel PJ, et al. Baseline and on-statin 2019;30:1-9.
treatment lipoprotein(a) levels for prediction of cardiovascular 132. Ballantyne CM, Laufs U, Ray KK, et al. Bempedoic acid plus
events: individual patient-data meta-analysis of statin outcome ezetimibe fixed-dose combination in patients with hypercholester-
trials. Lancet. 2018;392:1311-1320. olemia and high CVD risk treated with maximally tolerated statin
118. O’Donoghue ML, Fazio S, Giugliano RP, et al. Lipoprotein(a), therapy. Eur J Prev Cardiol. 2020;27:593-603.
PCSK9 Inhibition, and Cardiovascular Risk. Circulation. 133. Davis TM, Ting R, Best JD, et al. Effects of fenofibrate on renal
2019;139:1483-1492. function in patients with type 2 diabetes mellitus: the Fenofibrate
119. Ranga GS, Kalra OP, Tandon H, Gambhir JK, Mehrotra G. Intervention and Event Lowering in Diabetes (FIELD) Study.
Effect of aspirin on lipoprotein(a) in patients with ischemic stroke. Diabetologia. 2011;54:280-290.
J Stroke Cerebrovasc Dis. 2007;16:220-224. 134. Boden WE, Probstfield JL, Anderson T, et al. Niacin in patients
120. Suk Danik J, Rifai N, Buring JE, Ridker PM. Lipoprotein(a), with low HDL cholesterol levels receiving intensive statin therapy.
hormone replacement therapy, and risk of future cardiovascular N Engl J Med. 2011;365:2255-2267.
events. J Am Coll Cardiol. 2008;52:124-131. 135. HPS2-THRIVE Collaborative Group, Landray MJ, Haynes R,
121. Sahebkar A, Reiner Z, Simental-Mendia LE, Ferretti G, Cicero et al. Effects of extended-release niacin with laropiprant in high-
AF. Effect of extended-release niacin on plasma lipoprotein(a) risk patients. N Engl J Med. 2014;371:203-212.
levels: A systematic review and meta-analysis of randomized 136. Artemeva NV, Safarova MS, Ezhov MV, Afanasieva OI,
placebo-controlled trials. Metabolism. 2016;65:1664-1678. Dmitrieva OA, Pokrovsky SN. Lowering of lipoprotein(a) level
122. Pokrovsky SN, Afanasieva OI, Safarova MS, et al. Specific under niacin treatment is dependent on apolipoprotein(a) pheno-
Lp(a) apheresis: A tool to prove lipoprotein(a) atherogenicity. type. Atheroscler Suppl. 2015;18:53-58.
Atheroscler Suppl. 2017;30:166-173. 137. Juxtapid (lomitapide) prescribing information. Cambridge, MA:
123. Baigent C, Blackwell L, Emberson J, et al. Efficacy and safety Aegerion Pharmaceuticals, Inc.; 2017.
of more intensive lowering of LDL cholesterol: a meta-analysis
of data from 170,000 participants in 26 randomised trials. Lancet.
2010;376:1670-1681.
18 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 19
20 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 21
22 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 23
24 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 25
26 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 27
28 Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10)
Lipid Management Algorithm, Endocr Pract. 2020;26(No. 10) 29

You might also like