Download Maintaining Cardiovascular Health in Patients With Mixed

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

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

Document related concepts

Remote ischemic conditioning wikipedia , lookup

Niacin wikipedia , lookup

Antihypertensive drug wikipedia , lookup

Quantium Medical Cardiac Output wikipedia , lookup

Management of acute coronary syndrome wikipedia , lookup

Myocardial infarction wikipedia , lookup

Saturated fat and cardiovascular disease wikipedia , lookup

Cardiovascular disease wikipedia , lookup

Coronary artery disease wikipedia , lookup

Transcript
Maintaining Cardiovascular Health in Patients With
Mixed Dyslipidemia: Optimizing the Management of
Hypertriglyceridemia and Non-HDL Cholesterol
Matthew K. Ito, PharmD, FCCP, CLS
James M. McKenney, PharmD
Eliot A. Brinton, MD
Supplement
February 2009
Vol. 15, No. 1-c
Continuing Education Activity
F a c u lt y
Editor-in-Chief
Frederic R. Curtiss, PhD, RPh, CEBS
830.935.4319, [email protected]
Associate Editor
Kathleen A. Fairman, MA
602.867.1343, [email protected]
Peer Review Administrator
Jennifer A. Booker, 703.317.0725
[email protected]
Graphic Designer
Margie Hunter, 703.297.9319
[email protected]
February Supplement Editor
Peter Whittaker, PhD
[email protected]
Account Manager
Peter Palmer, 800.486.5454, ext. 13
[email protected]
Publisher
Judith A. Cahill, CEBS
Executive Director
Academy of Managed Care Pharmacy
This supplement to the Journal of Managed Care Pharmacy
(ISSN 1083–4087) is a publication of the Academy of Managed
Care Pharmacy, 100 North Pitt St., Suite 400, Alexandria, VA
22314; 703.683.8416; 703.683.8417 (fax).
Copyright © 2009, Academy of Managed Care Pharmacy.
All rights reserved. No part of this publication may be
reproduced or transmitted in any form or by any means,
electronic or mechanical, without written permission from
the Academy of Managed Care Pharmacy.
POSTMASTER: Send address changes to JMCP,
100 North Pitt St., Suite 400, Alexandria, VA 22314.
Supplement Policy Statement
Standards for Supplements to the
Journal of Managed Care Pharmacy
Supplements to the Journal of Managed Care Pharmacy are
intended to support medical education and research in areas
of clinical practice, health care quality improvement, or
efficient administration and delivery of health benefits. The
following standards are applied to all JMCP supplements to
ensure quality and assist readers in evaluating potential
bias and determining alternate explanations for findings
and results.
1. Disclose the principal sources of funding in a manner that
permits easy recognition by the reader.
2. Disclose the existence of all potential conflicts of interest
among supplement contributors, including financial or personal bias.
3. Describe all drugs by generic name unless the use of
the brand name is necessary to reduce the opportunity for
confusion among readers.
4. Identify any off-label (unapproved) use by drug name and
specific off-label indication.
5. Strive to report subjects of current interest to managed
care pharmacists and other managed care professionals.
6. Seek and publish content that does not duplicate content
in the Journal of Managed Care Pharmacy.
7. Subject all supplements to expert peer review.
Matthew K. Ito, PharmD, FCCP, CLS, is Professor and Chair of Pharmacy Practice
at Oregon State University College of Pharmacy in Corvallis and Oregon Health &
Science University in Portland. He received his doctorate in pharmacy in 1986 from
the University of Southern California, Los Angeles, where he also completed a postdoctoral residency and fellowship. Dr. Ito began his academic career as an Assistant
Professor at the University of the Pacific (UOP) Thomas J. Long School of Pharmacy
and Health Sciences in Stockton, California, and as the Regional Coordinator in San
Diego for the Advanced Experiential Program. In 2002, he became Vice Chair of
Pharmacy Practice at UOP. During his tenure at UOP, he received numerous faculty
awards for teaching and scholarship. Dr. Ito was appointed Professor and Chair of
Pharmacy Practice at Oregon State University in 2005.
Dr. Ito’s research focuses on the effects of lipid-modifying agents on endothelial dysfunction, pharmacokinetics, pharmacodynamics, and clinical outcomes
research. He has garnered close to $750,000 in research grants; published more
than 80 research papers, review articles, book chapters, and abstracts; given more
than 300 presentations to professional audiences at the local, state, and national
levels; and trained research fellows since 1995.
He is a member and fellow of the American College of Clinical Pharmacy; a
member of the Council on Arteriosclerosis, Thrombosis, and Vascular Biology of the
American Heart Association; an officer for the Pacific Lipid Association; an editorial
board member of the Journal of Clinical Lipidology and The Annals of Pharmacotherapy;
and a diplomate of the American Council on Clinical Lipidology.
James M. McKenney, PharmD, is President and CEO of National Clinical Research
and Professor Emeritus of the School of Pharmacy of Virginia Commonwealth
University, both in Richmond, Virginia. Dr. McKenney is the founder of an ambulatory clinical pharmacy service based on patient referral for primary care centers at
Medical College of Virginia hospitals and the first accredited ambulatory care training program for pharmacy residents in the United States.
Dr. McKenney received his doctor of pharmacy degree from Wayne State
University in Detroit, Michigan. His numerous accomplishments include appointment to the Adult Treatment Panels II and III to develop guidelines for the evaluation and treatment of hyperlipidemia, fellowships from the American Society of
Hospital Pharmacists and the American Heart Association, and the Clinical Practice
Award for lifetime contributions to pharmacy practice from the American College
of Clinical Pharmacy. He has authored or coauthored more than 160 original peerreviewed research papers and articles on hypertension, patient compliance, and
hyperlipidemia; been engaged as a principal or coinvestigator in more than 650
clinical trials; and given more than 1,600 professional and scientific presentations.
Dr. McKenney is a founding member and former President of the National Lipid
Association and has been a member of the Coordinating and Executive Committees
of the National Cholesterol Education Program since its inception in 1985.
Eliot A. Brinton, MD, is Director of the Metabolism Section of Cardiovascular
Genetics and Associate Professor at the University of Utah School of Medicine
(UUSOM) in Salt Lake City. He is also Attending Physician at the University of
Utah’s Cardiovascular Disease Prevention Clinic.
Dr. Brinton earned his medical degree from UUSOM and trained in internal
medicine at Duke University, Durham, North Carolina, and in endocrinology at
the University of Washington in Seattle. Prior to joining the faculty of UUSOM, he
had been on the faculty of The Rockefeller University in New York, New York, and
Wake Forest University in Winston-Salem, North Carolina, and was Chief of the
Section of Metabolism, Endocrinology, and Nutrition at the Phoenix VA Medical
Center in Arizona.
Dr. Brinton has received research funding from the National Institutes of Health,
the U.S. Department of Veterans Affairs, and various foundations and has published
several research articles over the past 25 years. Among the numerous scientific advisory positions he has held, Dr. Brinton was a member of the National Grant Review
Panel of the American Diabetes Association. He currently serves on the editorial
boards of the Journal of Clinical Lipidology, Lipids Online, the Journal of Managed Care
Pharmacy, and Future Lipidology and has been Section Editor of “Clinical Trials and
Their Interpretation” for Current Atherosclerosis Reports. Dr. Brinton is President of
the Utah Atherosclerosis Society, President-Elect of the Pacific Lipid Association,
and a founding board member of both the National Lipid Association and the
American Board of Clinical Lipidology.
Table of Contents
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
Matthew K. Ito, PharmD, FCCP, CLS; James M. McKenney, PharmD; Eliot A. Brinton, MD
S3 Abstract and Faculty Correspondence
S3 Causes and Consequences of Mixed Dyslipidemia: Mitigating Coronary Heart Disease Risk
Matthew K. Ito, PharmD, FCCP, CLS
S8 Making Informed Choices: Assessing Efficacy and Cost-Benefit of Therapeutic Options for the
Management of Mixed Dyslipidemia
James M. McKenney, PharmD
S14 Clinical Decision Making: Improving Outcomes for Patients With Mixed Dyslipidemia
Eliot A. Brinton, MD
S20 Continuing Education: CE Submission Instructions and Posttest Questions
Target Audience
This program is designed to meet the educational needs of pharmacists, managed care professionals, and other health
care professionals who are interested in learning more about the management of hypertriglyceridemia-associated
mixed dyslipidemia and the reduction of cardiovascular risk.
Educational Objectives
After completing this knowledge activity, participants should be better able to:
1. Assess the impact of hypertriglyceridemia and non-HDL cholesterol on cardiovascular risk.
2. Describe the pharmacologic properties and rationale for selecting agents used in the treatment of mixed dyslipidemia.
3. Evaluate the cost-benefits of pharmacologic agents used in the management of mixed dyslipidemia.
4. Apply best-evidence when developing clinical recommendations designed to optimize the pharmacologic management of mixed dyslipidemia.
Source of Funding
Sponsored by
Developed in conjunction with
Supported by an educational grant from
Continuing Education Credit
This supplement is based on the live program titled, “Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol” (ACPE Universal Program Number 073999-08-082-L01-P), held as a dinner meeting on April 17, 2008, at the Intercontinental San Francisco in San Francisco,
California. Anyone who received CE credit for the live program is not eligible to receive CE credit for this supplement, which
represents the same content material.
Extension Services in Pharmacy at the University of Wisconsin-Madison School of Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of pharmacy continuing education. This program
has been approved for a maximum of 2 hours (0.2 CEUs) of pharmacy continuing education credit. A Statement of Credit
can be obtained from the AMCP.org Online Learning Center following completion of materials documenting successful
participation in this program (completion of a program evaluation and posttest with a minimum score of 70%). Universal Program
Number (UPN) 073-999-08-082-H01-P.
Activity Release Date: April 17, 2008
Activity Expiration Date: April 17, 2011
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
Faculty Disclosures
All program personnel (faculty and staff) in a position to control the content of an educational activity sponsored by the University of WisconsinMadison School of Pharmacy must disclose all relevant financial relationships with any commercial interest. Extension Services in Pharmacy shall
determine whether identified relationships present a conflict to an individual’s role in this program.
Faculty and Sponsor Disclosures
When asked to report any potential conflict(s) of interest, the faculty reported the following:
Matthew K. Ito, PharmD, FCCP, CLS: No relevant personal relationships to disclose.
James M. McKenney, PharmD: Consultant to Abbott Laboratories, Aegerion Pharmaceuticals Inc., AstraZeneca, Merck & Co., Inc., Reliant
Pharmaceuticals, Inc., and Daiichi Sankyo Co., Ltd.; speakers’ bureaus for AstraZeneca and Merck & Co., Inc.
Eliot A. Brinton, MD: Grant/research support from Abbott Laboratories, AstraZeneca, GlaxoSmithKline, Merck & Co., Inc., Pfizer Inc., and
Takeda Pharmaceuticals North America, Inc.; consultant to Abbott Laboratories, AstraZeneca, GlaxoSmithKline, Merck & Co., Inc., Pfizer Inc.,
and Takeda Pharmaceuticals North America, Inc.; speakers’ bureaus for Abbott Laboratories, AstraZeneca, GlaxoSmithKline, Merck & Co., Inc.,
Pfizer Inc., and Takeda Pharmaceuticals North America, Inc.
Sponsor and Medical Education Company
All SciMed personnel involved in the development of content for this activity have no relevant conflicts to report. The materials for this activity
were peer reviewed, and the reviewer had no relevant conflicts to report. All Extension Services in Pharmacy personnel involved in the development and review of content for this activity have no relevant conflicts to report. Further, Extension Services in Pharmacy has determined that
there are no faculty conflicts associated with this activity.
Disclosure of Off-Label Use
This educational activity contains discussion of unapproved (investigational) uses of laropiprant in the management of dyslipidemia. The authors
have determined to the best of their ability that all other drugs of the statin class (rosuvastatin, pravastatin, atorvastatin, lovastatin, fluvastatin,
and simvastatin), purified omega-3 acid ethyl esters, niacin, ezetimibe, and all drugs of the fibrate class (gemfibrozil, bezafibrate, clofibrate, and
fenofibrate) discussed in this supplement are FDA approved, either alone or in combination at the doses described, for the management of elevated
non–HDL-C associated with hypertriglyceridemia and mixed dyslipidemia. Please refer to the official prescribing information for each product
for a description of approved indications, contraindications, and warnings.
Disclaimer
The opinions or views expressed in this pharmacy CE activity are those of the presenters and do not necessarily reflect the opinions or recommendations of SciMed, the University of Wisconsin-Madison School of Pharmacy, or the commercial supporter. Participants should critically appraise
the information presented and are encouraged to consult appropriate resources for information surrounding any product or device mentioned.
S2 Supplement to Journal of Managed Care Pharmacy
JMCP
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed
Dyslipidemia: Optimizing the Management of Hypertriglyceridemia
and Non-HDL Cholesterol
Matthew K. Ito, PharmD, FCCP, CLS; James M. McKenney, PharmD; Eliot A. Brinton, MD
Abstract
OBJECTIVE: To review the role of elevated levels of serum cholesterol and
triglycerides in coronary heart disease (CHD) and the increasing recognition of the need to improve the overall lipid profile of patients with mixed
dyslipidemia to further mitigate CHD risk.
SUMMARY: Hypercholesterolemia, represented by elevated levels of lowdensity lipoprotein cholesterol (LDL-C), is a well-known and studied dyslipidemia associated with increased risk for CHD. Statin treatment is highly
effective for lowering serum levels of LDL-C. Nevertheless, mixed dyslipidemia associated with low levels of high-density lipoprotein cholesterol
(HDL-C) and/or elevated triglycerides, which are risk factors independent
of high LDL-C levels, are common. Although interventions using statins are
the standard-of-care in mixed dyslipidemia, statin treatment alone does not
adequately address these components of the lipid profile in many patients,
resulting in residual dyslipidemia and considerable CHD risk. Fibrates, niacin, and omega-3 fatty acids are efficacious additional potential treatment
options for patients with mixed dyslipidemia.
J Manag Care Pharm. 2009;15(1)(Suppl S-c):S3-S21
Copyright © 2009, Academy of Managed Care Pharmacy. All rights reserved.
Faculty
MATTHEW K. ITO, PharmD, FCCP, CLS, is Professor and Chair
of Pharmacy Practice at the College of Pharmacy at Oregon State
University/Oregon Health & Science University in Portland, Oregon.
CORRESPONDENCE: Matthew K. Ito, PharmD, FCCP, CLS,
Professor and Chair of Pharmacy Practice, Oregon State University/
Oregon Health & Science University, College of Pharmacy, Portland
Campus at OHSU, 3303 SW Bond Avenue, Portland, OR 97239. Tel:
503.494.3657; Fax: 503.418.9360; E-mail: [email protected]
JAMES M. MCKENNEY, PharmD, is President and CEO of
National Clinical Research and Professor Emeritus of Virginia
Commonwealth University School of Pharmacy in Richmond,
Virginia.
CORRESPONDENCE: James M. McKenney, PharmD, National
Clinical Research, Inc., 2809 Emerywood Parkway, Suite 140,
Richmond, VA 23294. Tel: 804.672.2133; Fax: 804.672.3369;
E-mail: [email protected]
ELIOT A. BRINTON, MD, is Director of the Metabolism Section,
Cardiovascular Genetics, and Associate Professor at the University of
Utah School of Medicine in Salt Lake City, Utah.
CORRESPONDENCE: Eliot A. Brinton, MD, Cardiovascular
Genetics, 420 Chipeta Way, Room 1160, University of Utah School
of Medicine, Salt Lake City, UT 84108. Tel: 801.581.3888; Fax:
801.581.6862; E-mail: [email protected]
www.amcp.org
Vol. 15, No. 1, S-c
■■ Causes and Consequences of Mixed Dyslipidemia:
Mitigating Coronary Heart Disease Risk
Matthew Ito, PharmD, FCCP, CLS
Summary
Data from the Framingham Heart Study show a direct relationship between the risk of developing coronary heart disease
(CHD) and total serum cholesterol levels. Epidemiologic studies
have also established specific lipid parameters as important risk
factors for CHD. For example, in the Prospective Cardiovascular
Münster (PROCAM) study, the incidence of CHD was positively
correlated with increasing serum levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TGs) and negatively
correlated with increasing high-density lipoprotein cholesterol
(HDL-C) serum concentrations. Nevertheless, there are patients
with CHD who have plasma LDL-C levels within the normal
range. Variation in size, density, and composition of LDL-C
particles affect an individual’s likelihood of developing CHD.
Smaller, denser LDL-C particles are more prevalent in patients
with atherogenic metabolic syndrome (low HDL-C and high TG
levels) and in those with CHD. Despite the availability of therapies to reduce dyslipidemia-associated CHD, the risk remains
high for many patients. Therapeutic decision making and treatment monitoring for mixed dyslipidemia is complex and, when
TGs are elevated, should include assessment of the non–HDL-C
level, which is a better predictor of cardiovascular events than the
LDL-C level in this patient population.
Overview of Dyslipidemia
Dyslipidemia associated with high levels of LDL-C is a wellknown risk factor for CHD and is the primary focus of lipidlowering therapy with statins. Individuals with mixed dyslipidemia may also have high levels of TGs, low levels of HDL-C,
and elevated non–HDL-C.1 In clinical trials, patients treated with
statins to normalize LDL-C levels have considerable residual risk
for CHD, explained in part by the recognition that elevated TGs
and low HDL-C levels are LDL-C–independent risk factors for
CHD. Therefore, further CHD risk reduction might be achieved
by normalizing TGs and HDL-C levels.
A brief review of the major components of the atherosclerotic
process will emphasize the relationship between dyslipidemia
and CHD.
In addition to cholesterol, TGs and lipoproteins that constitute
the serum lipids play an important role. TGs, the most prevalent
fat in the body and diet, are composed of 3 fatty acids linked to a
glycerol molecule. Plasma TGs are diet derived or are synthesized
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S3
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
FIGURE 1
Cholesterol-Carrying Lipoproteins
VLDL
VLDL
VLDL VLDL
Remnants
Density in gm per mL
0.95
1.006
IDL
1.02
1.06
1.10
1.20
Chylomicron
Chylomicron PP TG
Remnants
LDL
HDL2
HDL3
HDL
Cholesterol
5
10
20
Mainly TG
LDL
Cholesterol
Lp(a)
40
60
Diameter (nm)
80
1000
Reprinted from Segrest JP et al. Adv Protein Chem. 1994;45:303-69.4
HDL = high-density lipoprotein; IDL = intermediate-density lipoprotein; LDL = lowdensity lipoprotein; Lp(a) = lipoprotein A; nm = nanometer; PP = postprandial
plasma; TG = triglyceride; VLDL = very low-density lipoprotein.
in the body from carbohydrates and other energy sources. Excess
dietary calories are converted to TGs and stored in adipocytes.2
TGs are a heterogeneous group of compounds because many fatty
acids of varying length, branching, and bond saturation exist.
Cholesterol, a waxy, fat-like steroid alcohol, is produced by the
liver or ingested from animal-derived food sources. It normally
functions as a regulator of membrane fluidity and is also a precursor in the synthesis of biomolecules, such as steroid hormones
and bile acids. Pathogenically, when associated with lipoprotein
particles, cholesterol is a key component of the atherogenic process.3
Lipoproteins are particulate accumulations of proteins and
fat that carry lipids and lipid-soluble substances, such as cholesterol, through the blood. Lipoproteins have an outer membrane
consisting of phospholipids, unesterified cholesterol, and various
surface apolipoproteins; the core of the particle is composed
of cholesteryl ester, TGs, and other neutral lipids.4 Lipoprotein
classification is based on density, particle size, surface apolipoproteins, and TG and cholesterol content (Figure 1). The terminology used to describe lipoprotein particle density includes
very low (VLDL), low (LDL), intermediate (IDL), and high (HDL).
When describing the cholesterol component of lipoprotein, “-C”
is added to the acronym (e.g., LDL-C). Non–HDL-C, a measure
of atherogenic cholesterol, represents the cholesterol carried by
VLDL, IDL, and large and small LDL particles and is calculated
as total cholesterol minus HDL-C.
Diet-derived cholesterol as well as other sterols and cholesterol
from biliary recirculation enter the gastrointestinal tract and are
incorporated into mixed micelles. Cholesterol and other sterols
are transported from the micelles into enterocytes by interacting
with Niemann-Pick C1-Like 1 transporters.5 Some cholesterol
S4 Supplement to Journal of Managed Care Pharmacy
JMCP
February 2009
and most plant sterols, which are structurally similar to cholesterol, are exported back from the enterocyte into the intestinal
lumen by the adenosine triphosphate–binding cassette transporters G5 and G8 (ABC G5/G8).6 Absorbed cholesterol is packaged
with TGs and various surface proteins into chylomicrons. These
large, buoyant, TG-rich particles are then transported across
the enterocytes and secreted into the blood. The content of the
chylomicrons is then hydrolyzed by apolipoprotein C-II (apoCII)–activated lipoprotein lipase into chylomicron remnants.
During this process, surface lipids and proteins are transferred
between chylomicron remnants and HDL particles. Chylomicron
remnants are then taken up by endocytosis in the liver by LDLrelated protein. In the liver, cholesterol and TGs are incorporated
in VLDL together with phospholipids and apolipoprotein B-100.
VLDLs are released into the circulation, where they acquire apolipoproteins E and C-II from HDL. A reduction in the TG content
of VLDLs occurs in the circulation by the hydrolytic action of
lipoprotein lipase to form VLDL remnants and IDLs. IDLs can
be cleared from the circulation by hepatic LDL receptors or
converted to LDL by further depletion of TGs through the action
of hepatic lipases.7 All of the cholesterol-containing LDLs are
atherogenic; however, small dense LDL-C particles are particularly pathogenic. LDL particles can migrate to artery walls and
bind to proretentive proteoglycans in the subendothelium where
they are oxidized. Oxidized lipoproteins promote endothelial
dysfunction, resulting in monocyte entry into the subendothelium.8 These monocytes differentiate into macrophages and
ingest the oxidized lipoproteins to become cholesterol-laden
foam cells that form fatty streaks. These streaks can progress to
unstable necrotic plaques that block arteries and contribute to
cardiovascular disease.6,9-11
Lipids, Cholesterol, and Coronary Heart Disease
Increased levels of total cholesterol are positively correlated with
CHD risk. Reduction of LDL-C levels has long been the gold
standard for measuring the efficacy of lipid-lowering therapy.
However, placebo-controlled trials of various statins (hydroxymethylglutaryl-coenzyme A reductase inhibitors) have shown
that a 20%-40% reduction in LDL-C levels results in a similar
reduction in coronary risk over a treatment duration of approximately 5 years.12-16 Therefore, considerable coronary risk remains
after treatment targets for LDL-C are achieved with statin therapy,
yet the potential exists for further risk reduction by modifying
other factors associated with dyslipidemia.
A subgroup analysis of the Framingham Heart Study clarified the role of LDL-C, HDL-C, and TG levels in determining
coronary risk.17 Specifically, study participants with LDL-C levels
< 100 mg per dL were stratified by increasing TG concentrations.
These data showed that, even though LDL-C levels remained
fairly constant throughout the various subgroups, the number
of LDL-C particles increased with increasing levels of serum
TGs, the size of those particles decreased, and the number of
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
cholesterol molecules per LDL-C particle decreased—all consistent with the observed increase in small dense LDL-C particles.
These changes were accompanied by an increase in non–HDL-C
levels.18 The Prospective Cardiovascular Münster (PROCAM)
study was undertaken to assess global CHD risk. The study
recruited more than 4,800 middle-aged men between 1979 and
1985 to determine the rate of CHD events over 8 years. The
patients’ CHD risk factors were recorded at study entry; those
with ratios of LDL-C/HDL-C > 5 had a 19.2% chance of experiencing a CHD event. When hypertriglyceridemia was also
present, the risk increased to 26.9%.19 Results of these studies
suggest that the existence of large numbers of LDL particles better predicts coronary risk than does high LDL-C levels, that low
HDL-C levels and high TG levels are CHD risk factors independent of LDL-C levels, and that non–HDL-C levels may be a better
marker for coronary risk than those of LDL-C. The mechanistic
roles of LDL-C, HDL-C, and TGs in coronary risk are detailed in
the following sections.
LDL-C. While the goal of lipid-lowering therapy with statins
is the reduction of LDL-C levels, the atherogenicity of LDL-C is
determined not only by its concentration, but also by the size and
density of the particles. Even though they carry less cholesterol,
small dense LDL particles are more atherogenic than their larger
counterparts because they bind more tightly to proteoglycans
in arterial walls and are more easily oxidized. These particles
first need to be oxidized to be taken up by scavenger receptors
on macrophages. An increase in foam cell formation occurs
and, eventually, so does development of atherosclerotic plaques.
Because small dense LDL particles carry fewer cholesterol molecules, there are more of these particles present at any given LDL-C
concentration. It is these LDL particles that migrate to the arterial
wall, not the LDL itself. Because this is a gradient-driven process,
the more particles that are present in the serum, the greater the
drive for particles to migrate from the lumen to the arterial wall.20
In fact, the decreased risk of CHD associated with high HDL-C
levels and the increased risk associated with high TG levels probably act as surrogate markers for the primary culprit—an increase
in atherogenic small dense LDL-C particles.
The creation of these small dense LDL-C particles occurs
when there is an increased flux of free fatty acids delivered to
the liver. The liver increases production of TG-rich VLDL particles from fatty acids. These VLDL particles then interact with
HDL and LDL through the action of cholesteryl ester transfer
protein and exchange the TG content for cholesterol and HDL
or LDL. The end result is a TG-rich, cholesterol-depleted LDL or
HDL particle; following hydrolysis with hepatic lipase, a small
dense LDL particle and a cholesterol-depleted HDL particle are
formed.21 In patients with the metabolic syndrome, after dissociation from HDL particles, apolipoprotein A-1 (apoA-1) is
eliminated by the kidneys, further reducing levels of HDL-C and
increasing coronary risk.7
Data supporting the differential atherogenicity of various
www.amcp.org
Vol. 15, No. 1, S-c
forms of LDL-C were provided by the Québec Cardiovascular
Study, which followed patients initially free of coronary disease
to determine CHD risk. Data on the size and number of LDL
particles were collected for 5 years, and CHD-related outcomes
were analyzed. The results demonstrated that patients with
numerous small dense LDL particles had a 6-fold increase in risk
of ischemic heart disease compared with patients with fewer, but
larger, LDL particles.22
HDL-C. In contrast, HDL-C is cardioprotective, and higher
levels of HDL-C are associated with reduced CHD risk. The
role of HDL-C in reverse cholesterol transport partly explains
this effect. In reverse transport, lipid-poor apoA-1 interacts with
ABCA1, a specialized transporter, and moves cholesteryl ester
out of macrophages to apoA-1, resulting in a discoidal immature
HDL particle. With further elution of cholesteryl ester, a mature
HDL particle is formed that interacts with scavenger receptors
on hepatocytes to deliver their cholesteryl ester content, which is
then excreted in bile.7 Other mechanisms that contribute to the
antiatherogenic effects of HDL include its antioxidative effects,
inhibition of adhesion molecule expression, platelet activation
inhibition, prostacyclin stabilization, and promotion of nitric
oxide production. Low HDL-C levels are associated with additional CHD risk factors including smoking, sedentary lifestyle,
obesity, insulin resistance and diabetes, hypertriglyceridemia,
and chronic inflammatory disorders.23
Triglycerides. There are a number of primary and secondary causes of elevated serum TG levels. Primary causes include
familial disorders of combined hypertriglyceridemia and hypercholesterolemia (e.g., familial combined hyperlipidemia, familial
dysbetalipoproteinemia, familial hepatic lipase deficiency) and
familial disorders of hypertriglyceridemia, such as familial
chylomicronemia (e.g., lipoprotein lipase deficiency, apoC-II
deficiency). Secondary causes include physical inactivity, a highcholesterol diet, excessive alcohol and/or carbohydrate intake,
obesity, insulin resistance (type 2 diabetes or the metabolic syndrome), chronic kidney disease, Cushing’s syndrome, nephritic
syndrome, hypothyroidism, and use of certain medications (e.g.,
tamoxifen, steroids, beta-blockers, thiazides, retinoids, atypical
antipsychotics, immunosuppressants, human immunodeficiency
virus protease inhibitors, isotretinoin).24-26
The various familial disorders can cause severe elevations in
serum TG levels, usually > 1,000 mg per dL; the primary target
for intervention in individuals with these disorders is to reduce
TG levels and the associated risk of pancreatitis. The most
worrisome disorders associated with elevated TG levels and
CHD risk are those that increase levels of both cholesterol and
TGs (insulin resistance and familial combined hyperlipidemia,
dysbetalipoproteinemia, and hepatic lipase deficiency). These
conditions occur in approximately 1%-6% of the patient population, but are probably associated with 20% of the individuals
that present with CHD. Approximately 25% of the patients with
elevated TGs are diabetic or have the metabolic syndrome.24-26
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S5
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
Hypertriglyceridemia contributes to coronary risk in several
ways. Excessive TG levels cause the accumulation of cholesterolrich remnants from the hydrolysis of chylomicrons or VLDL
particles. These remnants create a proinflammatory and oxidative environment that may exacerbate the atherosclerotic process
by enhancing adhesion molecule expression, foam cell formation, and smooth muscle cell toxicity. High TG levels may also
increase the amount of TG-rich HDL particles; evidence suggests
that these are less efficient at the reverse transport of cholesterol.
There is also an increase in the proportion of atherogenic small
dense LDL particles associated with hypertriglyceridemia.27
The Metabolic Syndrome and CHD Risk
Patients with the metabolic syndrome are at an increased risk for
cardiovascular death. The metabolic syndrome is a constellation
of various lipid abnormalities (low HDL-C levels, elevated TGs,
and an increase in small dense LDL-C particles) as well as other
factors such as increased waist circumference or visceral adiposity, elevated blood pressure, and an increase in fasting serum glucose levels. The presence of 3 of these factors is necessary to meet
the National Cholesterol Education Program Adult Treatment
Panel III (NCEP ATP III) diagnostic criteria for the metabolic
syndrome.28 The condition is highly prevalent: 1 of 4 adults in
the United States meet the NCEP ATP III diagnostic criteria for
the metabolic syndrome. By age 60, approximately 40% of individuals have the condition; prevalence is higher in men than in
women, and the risk is greater in certain ethnic populations. As
rates of obesity continue to rise, the incidence of the metabolic
syndrome is expected to increase.29
The association of the metabolic syndrome with cardiovascular risk was demonstrated in a large family study of patients with
type 2 diabetes in Finland and Sweden that followed participants
for 7 years. After controlling for traditional coronary risk factors, those with the metabolic syndrome had a 6-fold increase in
cardiovascular mortality versus patients without the condition.30
Another analysis of data from the Framingham Heart Study
stratified male patients by the number of the metabolic syndrome
components present (0 to 5) and compared LDL-C levels in each
group. As the number of the metabolic syndrome risk factors
increased, LDL-C levels remained relatively constant, yet the
number of atherogenic small dense LDL-C particles increased;
results were similar in women.31
■■ Treatment Goals for Patients with Dyslipidemias
Cholesterol
Desirable serum levels of total cholesterol and LDL-C are < 200
mg per dL and < 100 mg per dL, respectively. A normal level
for serum TGs is considered to be < 150 mg per dL and that for
HDL-C is ≥ 40 mg per dL. The goal for non–HDL-C levels is 30
mg per dL higher than the patient’s LDL-C level. A number of
traditional coronary risk factors modify specific LDL-C and non–
HDL-C goals in patients without preexisting coronary disease.
S6 Supplement to Journal of Managed Care Pharmacy
JMCP
February 2009
These factors include advancing age, family history of premature CHD, ongoing cigarette smoking, elevated blood pressure
(≥ 140/90 mm Hg), and low levels of HDL-C.32
NCEP ATP III treatment guidelines for LDL-C target levels
categorize each patient by their number of CHD risk factors. In
general, the threshold to initiate therapeutic lifestyle changes
occurs when a patient’s LDL-C level is just above target, whereas
the threshold to initiate drug therapy occurs when LDL-C levels
are 30 mg per dL over the target. Initiation of therapeutic lifestyle
changes is generally recommended for patients with 0-1 CHD
risk factor and LDL-C levels ≥ 160 mg per dL; however, if the
LDL-C level is ≥ 190 mg per dL, drug therapy should be considered. The threshold values for intervention and the LDL-C target
levels steadily decline as the CHD risk factors increase. If a patient
has 2 risk factors for CHD, the LDL-C goal is < 130 mg per dL
and the non–HDL-C goal is < 160 mg per dL.32,33 In the highestrisk patients, the LDL-C target is < 70 mg per dL, and thresholds
for lifestyle changes and drug therapy are ≥ 100 mg per dL and
≥ 130 mg per dL, respectively.32 These values were derived from
results of the Pravastatin or Atorvastatin Evaluation and Infection
Therapy (PROVE IT) trial34 and updated per the Treating to New
Targets (TNT) trial.35 In patients with mixed dyslipidemia associated with high levels of LDL-C and TGs, lowering LDL-C levels
is the primary goal of intervention and non–HDL-C becomes a
secondary target.32
Triglycerides
Patients with TG levels ≥ 500 mg per dL are at an increased risk
for pancreatitis, and lowering TG levels is the primary target for
intervention. Once TG levels are normalized, therapy can refocus
on LDL-C and, if necessary, non–HDL-C levels.
Management of Mixed Dyslipidemia
Both nonpharmacologic and pharmacologic therapies are available to treat mixed dyslipidemia. Recommended therapeutic
lifestyle changes include increased physical activity (moderate
exercise sufficient to expend ≥ 200 kcal per day); increased intake
of omega-3 fatty acids, plant stanols/sterols, and soluble fiber; a
caloric intake limited to that needed to maintain an appropriate
body weight and prevent weight gain; and limited dietary intake
of saturated fats, cholesterol, and trans fatty acids. Limiting saturated fat intake to < 7% of total calories and dietary cholesterol to
< 200 mg per day should be the primary recommendation in all
patients with elevated LDL-C levels. Other therapeutic options
include intake of 2 gm per day of plant sterols/stanols and 10-25
gm per day of soluble fiber.32,36 In patients with dyslipidemia
associated with elevated TG levels, recommendations for lifestyle
changes include increasing physical activity and decreasing total
caloric intake to levels that reduce weight and maintain an appropriate body weight.32
The American Heart Association (AHA) recommends that
all individuals increase the amount of fish incorporated into the
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
diet as a source of omega-3 fatty acids that may lower CHD risk.
People without coronary disease should be consuming at least 2
servings of fish per week. In patients with preexisting CHD, the
AHA recommends the equivalent of 1 gm per day of omega-3
fatty acids—a level shown to decrease the risk of cardiovascular
death. In patients with very high TG levels (> 500 mg per dL), 2-4
grams of omega-3 fatty acids can be ingested daily under the care
of a physician to help reduce serum TG levels.37
Prescription medications available to treat mixed dyslipidemia
include statins, purified omega-3 fatty acids, nicotinic acid, and
fibrates. Most patients with mixed dyslipidemia will require combination therapy with a statin plus one or more additional lipidlowering agents. These pharmacotherapies will be discussed in
further detail later in this supplement.
Conclusions
Dyslipidemia increases the risk for CHD. Mixed dyslipidemia
with elevated TG levels as a component is commonly seen, especially among patients with the metabolic syndrome or diabetes.
As the U.S. population ages and obesity rates increase, a greater
number of patients will have elevated TG level–associated lipid
disorders. In patients with mixed dyslipidemia associated with
increased TG levels, treatment recommendations now include the
lowering of non–HDL-C levels as a secondary intervention goal.
Patients with TG levels > 500 mg per dL should first be treated for
hypertriglyceridemia, with secondary goals of lowering LDL-C
and non–HDL-C levels.
REFERENCES
1. Fazio S. Management of mixed dyslipidemia in patients with or at risk
for cardiovascular disease: a role for combination fibrate therapy. Clin Ther.
2008;30(2):294-306.
2. American Heart Association. Triglycerides. What are triglycerides?
Available at: http://www.americanheart.org/presenter.jhtml?identifier=4778.
Accessed June 11, 2008.
3. National Institutes of Health. Medline Medical Dictionary: cholesterol.
http://www.nlm.nih.gov/medlineplus/mplusdictionary.html. Accessed June
11, 2008.
4. Segrest JP, Garber DW, Brouillette CG, Harvey SC, Anantharamaiah GM.
The amphipathic alpha helix: a multifunctional structural motif in plasma
apolipoproteins. Adv Protein Chem. 1994;45:303-69.
5. Garcia-Calvo M, Lisnock J, Bull HG, et al., The target of ezetimibe
is Niemann-Pick C1-Like 1 (NPC1L1). Proc Natl Acad Sci USA.
2005;102(23):8132-37.
6. Goldstein JL, Brown MS. Molecular medicine. The cholesterol quartet.
Science. 2001;292(5520):1310-12.
7. Genest J. Lipoprotein disorders and cardiovascular risk. J Inherit Metab
Dis. 2003;26(2-3):267-87.
8. Tabas I, Williams KJ, Borén J. Subendothelial lipoprotein retention as the
initiating process in atherosclerosis: update and therapeutic implications.
Circulation. 2007;116(16):1832-44.
11. Yu KC, Cooper AD. Postprandial lipoproteins and atherosclerosis. Front
Biosci. 2001;6:D332-D354.
12. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection
Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7-22.
13. Shepherd J, Cobbe SM, Ford I, et al., for the West of Scotland Coronary
Prevention Study Group. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med. 1995;333(20):1301-07.
14. Scandinavian Simvastatin Survival Study Group. Randomised trial
of cholesterol lowering in 4444 patients with coronary heart disease: the
Scandinavian Simvastatin Survival Study (4S). Lancet. 1994;344(8934):138389.
15. Sever PS, Dahlöf B, Poulter NR, et al., for the ASCOT Investigators.
Prevention of coronary and stroke events with atorvastatin in hypertensive
patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering
Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet.
2003;361(9364):1149-58.
16. Colhoun HM, Betteridge DJ, Durrington PN, et al., on behalf of the
CARDS investigators. Primary prevention of cardiovascular disease with
atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes
Study (CARDS): multicentre randomised placebo-controlled trial. Lancet.
2004;364(9435):685-96.
17. Kannel WB. Importance of hypertension as a major risk factor in cardiovascular disease. In: Genest J, Koiw E, Kuchel O, eds. Hypertension:
Physiopathology and Treatment. New York, NY: McGraw-Hill; 1977:888-10.
18. Cromwell WC, Otvos JD, Keyes MJ, et al., LDL particle number and
risk of future cardiovascular disease in the Framingham Offspring Study—
implications for LDL management. J Clin Lipidol. 2007;1(6):583-92.
19. Assmann G, Schulte H, Funke H, Von Eckardstein A. The emergence of
triglycerides as a significant independent risk factor in coronary artery disease. Eur Heart J. 1998;19(suppl M):M8-M14.
20. Weissberg PL, Rudd JH. Atherosclerotic biology and epidemiology
of disease. In: Topol EJ, ed. Textbook of Cardiovascular Medicine. 2nd ed.
Philadelphia, PA: Lippincott Williams & Wilkins; 2002:3-14.
21. Bays H, Stein EA. Pharmacotherapy for dyslipidaemia—current therapies and future agents. Expert Opin Pharmacother. 2003;4(11):1901-38.
22. Lamarche B, Tchernof A, Moorjani S, et al., Small, dense low-density
lipoprotein particles as a predictor of the risk of ischemic heart disease in
men. Prospective results from the Québec Cardiovascular Study. Circulation.
1997;95(1):69-75.
23. Baylor College of Medicine. Lipids online: educational resources in atherosclerosis. Available at: http://www.lipidsonline.org. Accessed June 26,
2008.
24. Fung MA, Frohlich JJ. Common problems in the management of hypertriglyceridemia. CMAJ. 2002;167(11):1261-66.
25. Hachem SB, Mooradian AD. Familial dyslipidaemias: an overview of
genetics, pathophysiology and management. Drugs. 2006;66(15):1949-69.
26. Dunbar RL, Rader DJ. Demystifying triglycerides: a practical approach
for the clinician. Cleve Clin J Med. 2005;72(8):661-66, 670-72, 674-75.
27. Miller M, Cannon CP, Murphy SA, Qin J, Ray KK, Braunwald E; for the
PROVE IT-TIMI 22 Investigators. Impact of triglyceride levels beyond lowdensity lipoprotein cholesterol after acute coronary syndrome in the PROVE
IT-TIMI 22 Trial. J Am Coll Cardiol. 2008;51(7):724-30.
28. Grundy SM, Cleeman JI, Daniels SR, et al., Diagnosis and management of the metabolic syndrome: an American Heart Association/National
Heart, Lung, and Blood Institute scientific statement. Curr Opin Cardiol.
2006;21(1):1-6.
29. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome
among US adults: findings from the Third National Health and Nutrition
Examination Survey. JAMA. 2002;287(3):356-59.
9. Shepherd J. The role of the exogenous pathway in hypercholesterolaemia.
Eur Heart J. 2001;3(suppl E):E2-E5.
30. Isomaa B, Almgren P, Tuomi T, et al., Cardiovascular morbidity
and mortality associated with the metabolic syndrome. Diabetes Care.
2001;24(4):683-89.
10. Davis HR Jr, Zhu LJ, Hoos LM, et al. Niemann-Pick C1 Like 1 (NPC1L1)
is the intestinal phytosterol and cholesterol transporter and a key modulator
of whole-body cholesterol homeostasis. J Biol Chem. 2004;279(32):33586-92.
31. Kathiresan S, Otvos JD, Sullivan LM, et al., Increased small low-density
lipoprotein particle number: a prominent feature of the metabolic syndrome
in the Framingham Heart Study. Circulation. 2006;113(1):20-29.
www.amcp.org
Vol. 15, No. 1, S-c
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S7
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
32. National Heart, Lung, and Blood Institute. Third Report of the National
Cholesterol Education Program (NCEP) Expert Panel on Detection,
Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult
Treatment Panel III): final report. Bethesda, MD: National Institutes of
Health; 2002. NIH Publication No. 02-5215.
33. Grundy SM, Cleeman JI, Merz CN, et al., for the National Heart, Blood,
and Lung Institute; American College of Cardiology Foundation; American
Heart Association. Implications of recent clinical trials for the National
Cholesterol Education Program Adult Treatment Panel III Guidelines.
Circulation. 2004;110(2):227-39.
34. Ridker PM, Morrow DA, Rose LM, Rifai N, Cannon CP, Braunwald E.
Relative efficacy of atorvastatin 80 mg and pravastatin 40 mg in achieving the dual goals of low-density lipoprotein cholesterol < 70 mg/dl and
C-reactive protein < 2 mg/l: an analysis of the PROVE-IT TIMI-22 trial. J Am
Coll Cardiol. 2005;45(10):1644-48.
35. LaRosa JC, Grundy SM, Waters DD, et al., for the Treating to New
Targets (TNT) Investigators. Intensive lipid lowering with atorvastatin in
patients with stable coronary disease. N Engl J Med. 2005;352(14):1425-35.
36. Kris-Etherton PM, Harris WS, Appel LJ; for the American Heart
Association Nutrition Committee. Fish consumption, fish oil, omega-3 fatty
acids, and cardiovascular disease. Circulation. 2002;106(21):2747-57.
37. American Heart Association. Fish and omega-3 fatty acids: AHA recommendation. Available at: http://www.americanheart.org/presenter.
jhtml?identifier=4632. Accessed June 10, 2008.
■■ Making Informed Choices: Assessing Efficacy and
Cost-Benefit of Therapeutic Options for the Management
of Mixed Dyslipidemia
James M. McKenney, PharmD
Summary
Atherosclerosis leading to cardiovascular disease (CVD) is the
most common cause of death and disability in developed nations.
The economic burden of CVD is also significant; in 2006, the
estimated total cost of coronary heart disease alone was $142.5
billion in the United States. In addition, the total cost of medical
expenses and lost productivity associated with CVD was in excess
of $400 billion. Although the lowering of low-density lipoprotein
cholesterol (LDL-C) levels is the first step in managing hypercholesterolemia, even when LDL-C is controlled, significant residual
cardiovascular risk remains in many patients. Myriad lipid disorders contribute to this risk and are characterized by high levels
of triglycerides, very low-density lipoprotein remnant cholesterol,
small dense LDL particles, and low levels of high-density lipoprotein cholesterol. The availability of an array of cost-effective
lipid-lowering agents enables clinicians to individualize therapy.
Thus, fibrates, niacin, and prescription omega-3 fatty acids can
be combined with statins to reduce CVD in these patients. Such
combination therapy to manage atherogenic dyslipidemia can
achieve greater protection against cardiovascular events than is
currently realized by statin monotherapy.
Overview of Management of Dyslipidemia
Patients treated with statins to achieve National Cholesterol
Education Program (NCEP)–recommended goals for low-density
lipoprotein cholesterol (LDL-C) levels may still have an elevated
risk for cardiovascular disease (CVD) (Figure 2).1-5 This residual
S8 Supplement to Journal of Managed Care Pharmacy
JMCP
February 2009
FIGURE 2
0
-20
Patients Treated to LDL-C Targets May
Benefit From Further Risk Reduction
HPS
WOSCOPS
4S
-27b
-31b
-34b
-40
ASCOT-LLA CARDSa
-36b
-37b
-60
-80
-100
Reduction in major coronary events vs. placebo (%)
Potential for further risk reduction
a Includes
stroke.
bP < 0.001.
ASCOT-LLA = Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering
Arm. Sever PS et al. Lancet. 2003;361:1149-58.4
CARDS = Collaborative Atorvastatin Diabetes Study. Colhoun HM et al. Lancet.
2004;364:685-96.5
HPS = Heart Protection Study Collaborative Group. Lancet. 2002;360:7-22.1
4S = Scandinavian Simvastatin Survival Study Group. Lancet. 1994;344:1383-89.3
WOSCOPS = West of Scotland Coronary Prevention Study. Shepherd J et al. N Engl
J Med. 1995;333:1301-07.2
cardiovascular risk may be related to a persistent dyslipidemia,
typically characterized by increased triglyceride (TG) levels, low
levels of high-density lipoprotein cholesterol (HDL-C) and the
presence of LDL particle remnants, small dense LDL particles,
and excessive levels of lipoprotein particles in general. Further
treatment with more aggressive statin therapy does not directly
address the residual dyslipidemia.6 The National Health and
Nutrition Examination Survey, conducted between 2003 and
2004, analyzed serum lipid levels of more than 2,800 adults aged
≥ 20 years. Of those without CVD or related comorbid diseases,
85%-89% were at recommended levels for LDL-C, non–HDL-C,
HDL-C, and TGs. Only 36%-37% of individuals with CVD or
related comorbidities met goals for LDL-C and non–HDL-C, and
only 17% were at recommended levels for all lipids tested. Those
undergoing treatment for dyslipidemia had significantly lower
LDL-C and non–HDL-C levels compared with untreated individuals; however, HDL-C and TGs remained uncontrolled.7 This
survey reinforces the need to address the residual dyslipidemia
found in statin-treated mixed dyslipidemia. Fibrates, niacin, and
omega-3 fatty acids—agents that lower TGs, reduce remnant
cholesterol and particle number, and raise HDL-C levels—are
reviewed, with a focus on their utility and cost-effectiveness for
reducing coronary heart disease (CHD) risk in patients with
mixed dyslipidemia.
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
Fibrates
Fibrates have been used to lower serum TG levels for many years;
however, their mechanism of action is complex and remains
only partially elucidated. It is known that fibrates stimulate the
nuclear receptor peroxisome proliferator-activated receptor-alpha
(PPAR-alpha), resulting in the transcriptional regulation of a family of genes. Fibrates bind to PPAR-alpha, which forms a complex
with the retinoid X receptor (RXR) ligand. This complex then
binds to a PPAR response element in DNA to either suppress or
enhance the transcription of many genes.8 In the liver, the fibrate/
PPAR-alpha/RXR ligand complex upregulates acyl-coenzyme A
(CoA) synthase, an enzyme in hepatocytes that reduces free fatty
acids to produce acetyl-CoA.9 This process results in reduced
TG synthesis and, ultimately, lower TG content in secreted very
low-density lipoprotein (VLDL) and LDL particles. There is also
upregulation of the adenosine triphosphate–binding cassette
genes ABCA1 and ABCG1, which code for proteins that transport
cholesterol from within the cell to the cell surface where they may
be picked up by circulating HDL particles for transport to the
liver for elimination.10 A modest increase in apolipoprotein A-1
(apoA-1) production has also been demonstrated.9 The net effect
of fibrate treatment is a modest increase in HDL-C levels, a substantial reduction in VLDL-C levels, and a decrease in number
and increase in size of LDL particles.11,12 Fenofibrate treatment
may increase LDL-C levels because it results in an increase of
LDL particles that are not rapidly cleared from the serum; however, non–HDL-C levels drop, with the net effect of a reduction
in the number of atherogenic particles and VLDL-C levels along
with an increase in HDL-C levels.13
However, fibrates are associated with several poorly understood limitations: (a) increases in creatinine and/or homocysteine
levels and hence a need for cautious use in patients with impaired
renal function, especially the elderly; (b) increased rates of myopathy, rhabdomyolysis, cholelithiasis, and cholescytectomy; and
(c) a trend toward an increased risk of noncardiovascular and
total mortality.14-17 When fibrates are coadministered with a statin, an additive myotoxic effect is possible.18 Furthermore, fibrate
treatment is associated with an increased risk for venothromboembolic disease19 (e.g., pulmonary embolism, deep venous
thrombosis) potentially due to increased homocysteine levels.20
Gemfibrozil can interfere with the glucuronidation of statins,
resulting in increased serum concentrations of lovastatin, pravastatin, rosuvastatin, and simvastatin; this effect is not significant
with atorvastatin and fluvastatin.21 Because such interactions
do not occur with fenofibrate, it is often used when combination therapy with statins is indicated. The effects of fibrates
on mortality were mixed in 4 placebo-controlled, randomized
end-point studies that enrolled a combined total of more than
19,000 patients. CHD-related and total mortality increased with
fenofibrate use by 19% and 11%, respectively, in the Fenofibrate
Intervention and Event Lowering in Diabetes (FIELD) study and
increased by 7% and 5%, respectively, with bezafibrate in the
www.amcp.org
Vol. 15, No. 1, S-c
FIGURE 3
Field (n = 9,795)
BIP (n = 3,090)
VA-HIT (n = 2,531)
Helsinki (n = 4,081)
Mortality Rate Changes in
Large Fibrate Trials
Fibrate
Fenofibrate
Bezafibrate
Gemfibrozil
Gemfibrozil
CHD Mortality Total Mortality
+19%
+7%
-22%
-26%
+11%
+5%
-11%
+7%
Source: McKenney JM. Future Lipidol. 2006;1:275-81.22
No change was statistically significant.
BIP = Bezafibrate Infarction Prevention study; CHD = coronary heart disease;
FIELD = Fenofibrate Intervention and Event Lowering in Diabetes study;
VA-HIT = Veterans Affairs High-Density Lipoprotein Intervention Trial.
Bezafibrate Infarction Prevention (BIP) study. In contrast, mortality was reduced by 22% and 11%, respectively, with gemfibrozil
in the Veterans Affairs High-Density Lipoprotein Intervention
Trial (VA-HIT) and decreased by 26% and increased by 7%,
respectively, with gemfibrozil in the Helsinki Heart Study fibrate
trial (Figure 3).22 Although these differences were not statistically significant, there is a troubling trend toward increased
mortality with fibrate therapy. In the World Health Organization
cooperative trial of primary prevention of ischemic heart disease,
clofibrate use was associated with 25% more deaths than in the
control groups (P < 0.01), with excess mortality attributable to cancer and diseases of the gallbladder, liver, and intestines—further
adding to the controversy surrounding the use of fibrates.23 A
meta-analysis of 10 placebo-controlled trials examined the role
of long-term fibrate treatment in the prevention of CVD. Results
indicated that, compared with placebo, fibrate treatment was
associated with an increased risk of all-cause mortality that was
not statistically significant and a significantly increased risk of
noncardiovascular mortality (P = 0.004). When clofibrate trials
were removed from the analysis, there was no significant difference in either value. This analysis also demonstrated that fibrates
did not significantly lower the risk of cardiovascular mortality,
fatal myocardial infarction (MI), or stroke, but did reduce the risk
of nonfatal MI by approximately one-fifth (P < 0.001).17
Currently, there is a lack of data concerning the incremental
cost-benefit of adding fibrates to statin therapy for primary and
secondary prevention of CHD. Outcome studies are needed
specifically to assess the pharmacoeconomics of this treatment
strategy.
Niacin
Although niacin has been used to lower TG levels for approximately 50 years, little was known about its mechanism of action.
However, we now appreciate that niacin acts during the last step
in the hepatocytic synthesis of TGs by inhibiting the conversion
of diacylglycerols to TGs by the enzyme diacylglycerol acyltransferase-2.24 Normally, the body anticipates the production of TGs
and thus begins to produce apolipoprotein B (apoB) to form
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S9
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
FIGURE 4
Niacin-Associated Reduction in
Cardiovascular Events and
All-Cause Mortality
50
Coronary Drug Project
45
Event rate (%)
40
35
30
25
P<0.05
P= NS
30.1
25.6
25.4
24.4
P < 0.05
20
13.8
15
10
10.2
5
0
Nonfatal MI/
CHD death
Nonfatal MI
Placebo
All-cause
mortality
Niacin
Source: Coronary Drug Project Research Group. JAMA. 1975;231:360-81.26
Copyright (2005), American Medical Association; all rights reserved.
CHD = coronary heart disease; MI = myocardial infarction; NS = not significant.
VLDL particles. When niacin is administered and TG synthesis is inhibited, the apoB that was produced degrades. The net
results of niacin therapy are fewer LDL-C particles, a reduction in
TG-rich VLDL particles, and lower serum LDL-C levels.
Niacin is thought to raise HDL-C levels via a unique mechanism. Kinetic studies of niacin-treated patients indicate a
decreased catabolism of apoA-1. At least 2 liver receptors are
responsible for the metabolism of HDL particles: the scavenger
receptor B1 facilitates HDL-C uptake, and the HDL holoparticle
catabolism receptor takes up HDL–apoA-1 particles by endocytosis. Niacin interferes with the latter receptor and leads to
a reduced removal of HDL, thus preserving HDL–apoA-1 for
additional reverse cholesterol transport from macrophage cells
to the liver. In this manner, apoA-1 rises—not because more
is produced, but because the apoA-1 that has been produced is
retained longer.24
Niacin was evaluated as part of the Coronary Drug Project
study for secondary prevention of cardiovascular events in men
with previous MI. In this study, niacin was associated with a
small but significant reduction in nonfatal MI and CHD-related
death versus placebo (Figure 4).25 In a side-by-side comparison
study, niacin was shown to be the better HDL-raising agent,
while gemfibrozil was a better TG-lowering agent. Niacin usually does not increase LDL-C levels, and both therapies have a
positive effect on non–HDL-C.26 In the Arterial Biology for the
S10 Supplement to Journal of Managed Care Pharmacy
JMCP
Investigation of the Treatment Effects of Reducing Cholesterol
(ARBITER) 2 study, 167 patients with prior MI were first treated
with statins to achieve a mean LDL-C level of approximately 90
mg per dL (below the < 100-mg-per-dL treatment goal) and an
HDL-C level of approximately 40 mg per dL. Extended-release
(ER) niacin at a dose of 1,000 mg per day or placebo was then
randomly added to the statin therapy. Progression of atherosclerosis, as measured by carotid artery intima-media thickness
(IMT), was followed for 12 months. There was less increase from
baseline to study end in IMT in patients who received a statin
plus ER niacin (P = 0.23) compared with those who received the
statin-plus-placebo regimen (P < 0.001); however, the betweengroup difference was not statistically significant (P = 0.08). There
was an additional 2.3% decrease in LDL-C, a 21.0% increase in
HDL-C, and a 7% decrease in non–HDL-C when ER niacin was
part of the treatment regimen.
Niacin is associated with an increased risk of hepatotoxicity
and a 4-mg-per-dL increase in fasting blood glucose (FBG); the
latter may be important when managing patients with impaired
FBG.27 Flushing is the most common side effect of niacin and,
while not dangerous, can lead to noncompliance or discontinuation of the drug.24 Recent research on the mechanism of niacininduced flushing may reduce the impact of this side effect. A
niacin receptor has been identified on adipocytes as well as on
immune and skin cells. Flushing is triggered by stimulation of
the niacin receptor in epidermal Langerhans cells, resulting in
increased production of arachidonic acid and, subsequently, an
array of prostaglandins (PG), including PGD2. Flushing is caused
by the dilation of capillary vessels due to the binding of PGD2
to peripheral DP-1 receptors.24 Laropiprant, a DP-1 receptor
antagonist, was developed to block the niacin-induced flushing
response. When laropiprant was added to ER niacin therapy,
approximately 90% of patients had moderate to no flushing
compared with 65% of patients on ER niacin alone in the first
week of niacin therapy, the period when flushing is usually most
severe.28
A cost-effectiveness analysis of ER niacin–augmented statin
therapy was undertaken using the Framingham Heart Study
dataset and data from a population of ARBITER 2 patients of
similar age, gender, and lipid profile. Rates of MI, angina, and
death from CHD were compared for both therapies. The incidence of all 3 measures declined modestly when ER niacin was
added to therapy. The patients who received both a statin and
ER niacin therapy gained an average of 0.17 life-years, which
translates to approximately €20,645 in incremental costs per
life-year gained.29 This figure is within the acceptable range for
expenditures to prevent cardiovascular disease.30
Omega-3 Fatty Acids
Populations with fish-rich diets have a lower risk of heart disease.
This phenomenon has been linked to the omega-3 fatty acids
eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA),
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
which are present in high concentrations in fatty fish. EPA and
DHA lower serum TG levels, but also have a number of other
effects that may explain their cardiovascular benefits. Linolenic
acid, a plant-derived omega-3 fatty acid, can be converted to
DHA, but the rate at which this occurs is neither sufficient to
raise DHA levels significantly nor contribute to TG lowering.
Although the primary dietary source of EPA and DHA is fatty
fish, fish oil supplements are sold over-the-counter, and a purified and concentrated form of omega-3 fatty acids is available as a
pharmaceutical-grade product by prescription.
Omega-3 fatty acids are thought to lower TG levels by acting
early in the TG biosynthetic pathway, most likely by stimulating
sterol regulatory element binding protein-1c (SREBP-1c), a transcriptional activator.31 When stimulated, SREBP-1c upregulates
the transcription of the enzymes—acetyl-CoA carboxylase and
fatty acid synthase—needed for the production of fatty acids,
which in turn are used to produce TGs. Omega-3 fatty acids
apparently inhibit binding of the liver X receptor (LXR) alpha/
RXR alpha heterodimer to the LXR-responsive elements in the
SREBP-1c promoter region, which suppresses SREBP-1c expression and ultimately leads to decreased TG levels.31
The American Heart Association (AHA) recommendation for
omega-3 fatty acid intake in individuals who do not have CHD is
consumption of a variety of fatty fish about twice a week, which
provides approximately 1 gm per day of omega-3 fatty acids. For
patients with CHD, the recommended AHA dose is also EPA/
DHA 1 gm per day from either fatty fish or supplements in consultation with their physician. For treatment of patients with very
high TG levels (≥ 500 mg per dL), 2-4 gm per day of omega-3 fatty
acids is recommended to prevent pancreatitis.32
Potential adverse effects associated with the consumption of
omega-3 fatty acids include increased caloric intake, consumption of cholesterol from low-potency dietary fish oil supplements,
and gastrointestinal disturbances, such as dyspepsia. Fishy
aftertaste and belching are common complaints associated with
supplement use. In the manufacture of supplements, omega-3
fatty acids are extracted from fish, raising the potential concern
for exposure to environmental toxins (e.g., methylmercury, polychlorinated biphenyls, organochlorine pesticides) found in the
waters from which the fish were caught. However, because they
are water soluble, mercury and other heavy metals are eliminated
when the oil is extracted from the fish during the manufacturing process; therefore, exposure to these toxins is generally not
a problem with supplement use.33,34 On the other hand, if eating
fish is a person’s sole source of omega-3 fatty acids, exposure to
toxins may occur because of the large quantity of fish required to
obtain the appropriate amount of fatty acids.
Several products are available to supplement the intake of
omega-3 fatty acids other than consumption of fatty fish. Overthe-counter fish oil supplements generally deliver 200-300 mg
of EPA/DHA in a 1-gram tablet; the remainder (700-800 mg)
www.amcp.org
Vol. 15, No. 1, S-c
FIGURE 5
Low-Dose P-OM3a– Associated
Reduction in Cardiovascular and
Cerebrovascular Events
Better
Death
Nonfatal MI
Nonfatal strokeb
Worse
RR 0.85 (0.74-0.98)
Cardiovascular death
Nonfatal MI
Nonfatal strokeb
RR 0.80 (0.68-0.95)
0.5
0.75
1.0
1.25
Relative risk of main end points
1.5
a Low-dose
P-OM3 = 1 gm per day.
protection evident beginning with month 9.
Source: [No authors listed] results of the GISSI-Prevenzione trial. Lancet.
1999;354(9177):447-55.38 Marchioli et al. Circulation. 2002;105:1897-903.39
MI = myocardial infarction; P-OM3 = purified omega-3; RR = relative risk.
bEarly
is composed of saturated fats, other fatty acids, and cholesterol.
A major issue with these supplements is the large number of
tablets or capsules that the patient must consume to receive the
4-gm-per-day dosage required to lower TG levels.35,36 A second
problem is the consumption of saturated fats, cholesterol, and
calories derived from the 80% of the dose that is not composed
of omega-3 fatty acids.
The U.S. Food and Drug Administration (FDA) approved
a concentrated, highly purified form of omega-3 fatty acids
(P-OM3) in November of 2004. P-OM3 is available by prescription and provides at least 900 mg of omega-3 fatty acid ethyl
esters per capsule (predominantly EPA/DHA) with only 10%
derived from other fatty acids, saturated fats, and cholesterol. Two
randomized, double-blind, placebo-controlled 6- and 16-week
studies of 4 gm per day (recommended dosage) of P-OM3 were
undertaken in 84 patients whose median TG level was 792 mg
per dL. In this population, P-OM3 reduced median TG, VLDL-C,
and non–HDL-C levels and increased median HDL-C levels
compared with placebo.37 P-OM3 is indicated as an adjunct to
diet modification for patients with very high TG levels (≥ 500 mg
per dL) associated with a high risk for pancreatitis.37 The FDAmonitored manufacturing process ensures a known source of
fish oil in a consistent, stable product with all contents analyzed
and reported.
A large, long-term trial was conducted to determine the effects
of omega-3 fatty acid therapy on secondary prevention of CVD.
The open-label Gruppo Italiano per lo Studio della Sopravvivenza
nell’Infarto Miocardico (GISSI)-Prevenzione study followed
11,324 patients for 3.5 years who had suffered an MI within 3
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S11
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
months prior to study entry. Patients were randomly assigned
to supplements of P-OM3, vitamin E, both supplements, or no
treatment (controls), and all patients were maintained on standard
care of pharmacologic treatment and lifestyle recommendations
that included a Mediterranean diet for most patients. The primary
combined efficacy end points of the study were (a) the cumulative rate of all-cause death, non-fatal MI, and non-fatal stroke,
and (b) the cumulative rate of cardiovascular death, non-fatal MI,
and non-fatal stroke. Using a 4-way factorial analysis comparing the efficacy of P-OM3 supplements, vitamin E supplements,
combined treatment versus controls, and combined treatment
versus individual interventions, the addition of P-OM3 therapy
reduced end point 1 from 14.6% to 12.3%, which indicates a 15%
reduction in risk (P = 0.023). End point 2 was 11.4% and 9.2% in
the control and P-OM3 treatment groups, respectively, which
represents a risk reduction of 20% (P = 0.008) (Figure 5). When
the components of the end points were analyzed separately, significant reductions were found in the risk for cardiovascular mortality (P = 0.024), coronary mortality (P = 0.023), fatal plus non-fatal
coronary events (P = 0.024), and sudden death (P = 0.010). The total
mortality rate was 10.4% with standard care alone and 8.3% with
the addition of P-OM3, a reduction of 20%. The rate of sudden
death was 3.5% with standard care alone and 1.9% with standard
care plus P-OM3, a 45% reduction. In addition, the cardiovascular event rate decreased from 26.5% with standard care alone to
18.9% with the addition of P-OM3, a reduction of 30%.38 Timecourse analysis of the results from the GISSI-Prevenzione trial
showed that total mortality was reduced by 41% after 3 months of
treatment with P-OM3 (P = 0.037). The risk reduction for sudden
death contributed to this decrease in total mortality, consistent
with either an antiarrhythmic or antithrombotic effect.39
A comparison study of P-OM3 (4 gm per day) and gemfibrozil
(1,200 mg per day) demonstrated similar efficacy for both agents
in terms of lowering TG (37%-40%) and VLDL-C (33%-39%)
levels. Both also raised LDL-C levels slightly; however, there was
also a net reduction in non–HDL-C with omega-3 fatty acids.40
Furthermore, P-OM3 has been studied in patients scheduled
for coronary artery bypass graft without concomitant cardiac
surgery, in which it reduced the incidence of vein graft occlusion.
Also, progression of renal insufficiency was slowed by P-OM3 in
patients with immunoglobulin A (IgA) nephropathy. In contrast,
P-OM3 had no effect on restenosis in elective coronary angioplasty patients.41
A pharmacoeconomic analysis of P-OM3 therapy was undertaken using data from the GISSI CHD Secondary Prevention
Trial. Patients who experienced a recent MI received either
P-OM3 or standard secondary prevention therapy (antiplatelet
agents, beta-blockers, angiotensin-converting enzyme inhibitors,
and/or lipid-lowering agents) for 3.5 years. Results indicated that
the cost-effectiveness of P-OM3 was comparable with recently
introduced medications in routine use for the secondary prevention of MI. The authors concluded that P-OM3 should be added
S12 Supplement to Journal of Managed Care Pharmacy
JMCP
to currently employed secondary prevention measures because of
its potential additive benefits.42 Similar cost-benefits were found
using comparable study methodologies applied to patients in
Australia, Belgium, Canada, Germany, Poland,43 and the United
Kingdom.44 In addition, investigators in the United States found
that P-OM3 can be expected to reduce overall costs and improve
outcomes when used for secondary prevention of cardiovascular
disease.45
Conclusions
Optimum lipid management strategies for patients with mixed
dyslipidemia associated with hypertriglyceridemia will lower TG
and non–HDL-C levels, in addition to lowering remnant VLDL-C
and LDL-C and raising HDL-C levels. Addressing this dyslipidemia should further reduce CHD risk in patients whose statin
treatment has brought LDL-C levels to goal; however, this risk
reduction has yet to be demonstrated in controlled clinical trials. Fibrates, niacin, and omega-3 fatty acids are all useful agents
for improving lipid profiles in patients with mixed dyslipidemia.
Nevertheless, the TG-lowering effects of fibrates are accompanied
by considerable tolerability issues, and there is concern over the
increase in mortality seen with these agents. Niacin is effective for
lowering VLDL-C and LDL-C levels and is the best agent available to raise HDL-C levels; its potential may be maximized if the
bothersome flushing seen with this agent can be controlled with
novel formulations or coadministered agents. P-OM3 is generally well tolerated and offers significant improvement in serum
lipid profiles by reducing TG, VLDL-C, and non–HDL-C levels
and increasing levels of HDL-C. P-OM3 has also been shown
to be efficacious and cost-effective when added to usual care for
the secondary prevention of cardiovascular and cerebrovascular
events including sudden death, MI, and stroke.
REFERENCES
1. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection
Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7-22.
2. Shepherd J, Cobbe SM, Ford I, et al., for the West of Scotland Coronary
Prevention Study Group. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med. 1995;333(20):1301-07.
3. Scandinavian Simvastatin Survival Study Group. Randomised trial of
cholesterol lowering in 4444 patients with coronary heart disease: the
Scandinavian Simvastatin Survival Study (4S). Lancet. 1994;344(8934):138389.
4. Sever PS, Dahlöf B, Poulter NR, et al., for the ASCOT Investigators.
Prevention of coronary and stroke events with atorvastatin in hypertensive
patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering
Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet.
2003;361(9364):1149-58.
5. Colhoun HM, Betteridge DJ, Durrington PN, et al., on behalf of the
CARDS investigators. Primary prevention of cardiovascular disease with
atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes
Study (CARDS): multicentre randomised placebo-controlled trial. Lancet.
2004;364(9435):685-96.
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
6. Tenenbaum A, Fisman EZ, Motro M, Adler Y. Optimal management of
combined dyslipidemia: what have we behind statins monotherapy? Adv
Cardiol. 2008;45:127-53.
28. Paolini JF, Mitchel YB, Reyes R, et al., Effects of laropiprant on nicotinic acid-induced flushing in patients with dyslipidemia. Am J Cardiol.
2008;101(5):625-30.
7. Ghandehari H, Kamal-Bahl S, Wong ND. Prevalence and extent of dyslipidemia and recommended lipid levels in US adults with and without cardiovascular comorbidities: the National Health and Nutrition Examination
Survey 2003-2004. Am Heart J. 2008;156(1):112-19.
29. Roze S, Ferrières J, Bruckert E, et al., Cost-effectiveness of raising HDL
cholesterol by adding prolonged-release nicotinic acid to statin therapy
in the secondary prevention setting: a French perspective. Int J Clin Pract.
2007;61(11):1805-11.
8. Touyz RM, Schiffrin EL. Peroxisome proliferator-activated receptors in
vascular biology—molecular mechanisms and clinical implications. Vascul
Pharmacol. 2006;45(1):19-28.
30. Kahn R, Robertson RM, Smith R, Eddy D. The impact of prevention on
reducing the burden of cardiovascular disease. Circulation. 2008;118(5):57685.
9. Fruchart JC, Duriez P, Staels B. Peroxisome proliferator-activated receptor-alpha activators regulate genes governing lipoprotein metabolism, vascular inflammation and atherosclerosis. Curr Opin Lipidol. 1999;10(3):245-57.
31. Harris WS, Miller M, Tighe AP, Davidson MH, Schaefer EJ. Omega-3
fatty acids and coronary heart disease risk: clinical and mechanistic perspectives. Atherosclerosis. 2008;197(1):12-24.
10. Wójcicka G, Jamroz-Wiśniewska A, Horoszewicz K, Bełtowski J. Liver X
receptors (LXRs). Part I: structure, function, regulation of activity, and role
in lipid metabolism. Postepy Hig Med Dosw. 2007;61:736-59.
32. Kris-Etherton PM, Harris WS, Appel LJ; for the American Heart
Association Nutrition Committee. Fish consumption, fish oil, omega-3 fatty
acids, and cardiovascular disease. Circulation. 2002;106(21):2747-57.
11. Duval C, Chinetti G, Trottein F, Fruchart JC, Staels B. The role of PPARs
in atherosclerosis. Trends Mol Med. 2002;8(9):422-30.
33. Foran SE, Flood JG, Lewandrowski KB. Measurement of mercury levels
in concentrated over-the-counter fish oil preparations: is fish oil healthier
than fish? Arch Pathol Lab Med. 2003;127(12):1603-05.
12. Lee CH, Olson P, Evans RM. Minireview: lipid metabolism, metabolic
diseases, and peroxisome proliferator-activated receptors. Endocrinology.
2003;144(6):2201-07.
13. Davidson MH, Bays HE, Stein E, Maki KC, Shalwitz RA, Doyle R;
TRIMS Investigators. Effects of fenofibrate on atherogenic dyslipidemia in
hypertriglyceridemic subjects. Clin Cardiol. 2006;29(6):268-73.
14. Unal A, Torun E, Sipahioglu MH, et al., Fenofibrate-induced acute renal
failure due to massive rhabdomyolysis after coadministration of statin in two
patients. Intern Med. 2008;47(11):1017-19.
15. Davidson MH, Armani A, McKenney JM, Jacobson TA. Safety considerations with fibrate therapy. Am J Cardiol. 2007;99(6A):3C-18C.
16. Raedsch R, Plachky J, Wolf N, Simonis G. Biliary lipids, lithogenic index
and biliary drug concentrations during etofibrate and bezafibrate treatment.
Eur J Drug Metab Pharmacokinet. 1995;20(2):113-18.
17. Saha SA, Kizhakepunnur LG, Bahekar A, Arora RR. The role of fibrates
in the prevention of cardiovascular disease—a pooled meta-analysis
of long-term randomized placebo-controlled clinical trials. Am Heart J.
2007;154(5):943-53.
20. Dierkes J, Westphal S, Luley C. The effect of fibrates and other lipidlowering drugs on plasma homocysteine levels. Expert Opin Drug Saf.
2004;3(2):101-11.
21. Davidson MH. Statin/fibrate combination in patients with metabolic
syndrome or diabetes: evaluating the risks of pharmacokinetic drug interactions. Expert Opin Drug Saf. 2006;5(1):145-56.
41. Bays H. Clinical overview of Omacor: a concentrated formulation of
omega-3 polyunsaturated fatty acids. Am J Cardiol. 2006;98(4A):71i-76i.
23. WHO Cooperative Trial Committee of Principal Investigators. W.H.O.
cooperative trial on primary prevention of ischaemic heart disease
using clofibrate to lower serum cholesterol: mortality follow-up. Lancet.
1980;2(8191):379-85.
24. Kamanna VS, Kashyap ML. Nicotinic acid (niacin) receptor agonists: will
they be useful therapeutic agents? Am J Cardiol. 2007;100(11 A):S53-61.
www.amcp.org
Vol. 15, No. 1, S-c
37. Reliant Pharmaceuticals, Inc. Lovaza [package insert]. Liberty Corner,
NJ: 2007.
40. Stalenhoef AF, de Graaf J, Wittekoek ME, Bredie SJ, Demacker PN,
Kastelein JJ. The effect of concentrated n-3 fatty acids versus gemfibrozil on
plasma lipoproteins, low density lipoprotein heterogeneity, and oxidizability
in patients with hypertriglyceridemia. Atherosclerosis. 2000;153(1):129-38.
22. McKenney JM. When it comes to the FIELD study, what is . . . is. Future
Lipidol. 2006;1(3):275-81.
27. Taylor AJ, Sullenberger LE, Lee HJ, Lee JK, Grace KA. Arterial Biology
for the Investigation of the Treatment Effects of Reducing Cholesterol
(ARBITER) 2: a double-blind, placebo-controlled study of extended-release
niacin on atherosclerosis progression in secondary prevention patients treated with statins. Circulation. 2004;110(23):3512-17. Erratum in: Circulation.
2004;110(23):3615.
36. Zucker ML, Bilyeu DS, Helmkamp GM, Harris WS, Dujovne CA. Effects
of dietary fish oil on platelet function and plasma lipids in hyperlipoproteinemic and normal subjects. Atherosclerosis. 1988;73(1):13-22.
39. Marchioli R, Barzi F, Bomba E, et al., on behalf of the GISSI-Prevenzione
Investigators. Early protection against sudden death by n-3 polyunsaturated fatty acids after myocardial infarction: time-course analysis of the
results of the Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto
Miocardico (GISSI)-Prevenzione. Circulation. 2002;105(16):1897-903.
Available at: http://circ.ahajournals.org/cgi/reprint/105/16/1897. Accessed
January 7, 2009.
19. Brown WV. Expert commentary: the safety of fibrates in lipid-lowering
therapy. Am J Cardiol. 2007;99(6A):19C-21C.
26. Guyton JR, Blazing MA, Hagar J, et al., for the Niaspan-Gemfibrozil
Study Group. Extended-release niacin vs gemfibrozil for the treatment
of low levels of high-density lipoprotein cholesterol. Arch Intern Med.
2000;160(8):1177-84.
35. Sanders TA, Sullivan DR, Reeve J, Thompson GR. Triglyceride-lowering
effect of marine polyunsaturates in patients with hypertriglyceridemia.
Arteriosclerosis. 1985;5(5):459-65.
38. Anonymous. Dietary supplementation with n-3 polyunsaturated fatty
acids and vitamin E after myocardial infarction: results of the GISSIPrevenzione trial. Lancet. 1999;354(9177):447-55.
18. Jones PH, Davidson MH. Reporting rate of rhabdomyolysis with fenofibrate + statin versus gemfibrozil + any statin. Am J Cardiol. 2005;95(1):12022.
25. Coronary Drug Project Research Group. Clofibrate and niacin in coronary heart disease. JAMA. 1975;231(4):360-81.
34. ConsumerLab.com. Product Review: Fish Oil/Omega-3 Supplements and
EPA/DHA Fortified Foods & Beverages. July 29, 2008. www.consumerlab.
com/reviews/Omega-3_Fatty_Acids_EPA_and_DHA_from_Fish_Marine_
Oils/omega3/. Accessed September 15, 2008.
42. Franzosi MG, Brunetti M, Marchioli R, Marfisi RM, Tognoni G,
Valagussa F; GISSI-Prevenzione Investigators. Cost-effectiveness analysis of
n-3 polyunsaturated fatty acids (PUFA) after myocardial infarction: results
from Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto (GISSI)Prevenzione Trial. Pharmacoeconomics. 2001;19(4):411-20.
43. Lamotte M, Annemans L, Kawalec P, Zoellner Y. A multi-country health
economic evaluation of highly concentrated n-3 polyunsaturated fatty acids
in secondary prevention after myocardial infarction. Pharmacoeconomics.
2006;24(8):783-95.
44. Quilici S, Martin M, McGuire A, Zoellner Y. A cost-effectiveness analysis of n-3 PUFA (Omacor) treatment in post-MI patients. Int J Clin Pract.
2006;60(8):922-32.
45. Schmier JK, Rachman NJ, Halpern MT. The cost-effectiveness of omega3 supplements for prevention of secondary coronary events. Manag Care.
2006;15(4):43-50.
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S13
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
■■ Clinical Decision Making: Improving
Outcomes for Patients With Mixed Dyslipidemia
Eliot A. Brinton, MD
Summary
Pharmacologic management of dyslipidemia reduces atherosclerotic coronary heart disease (CHD) risk and prolongs patient
survival. Statins are often the treatment of choice for patients
with elevated levels of low-density lipoprotein cholesterol
(LDL-C); however, many statin-treated individuals do not achieve
the target LDL-C goals set by the National Cholesterol Education
Program. In addition, many patients have mixed dyslipidemia
typified by elevated LDL-C and triglyceride (TG) levels. Because
statin monotherapy is only modestly effective in treating TG
elevations, combination modalities are often required for optimal
management of mixed dyslipidemia. Several recent clinical trials have evaluated the potential clinical benefits of combination
therapy for CHD risk reduction. For example, the COMBOS
(COMBination of prescription Omega-3 with Simvastatin) study
demonstrated the safety and efficacy of statins in combination
with purified omega-3 acid ethyl esters for improving lipid
parameters and achieving non–high-density lipoprotein cholesterol goals in patients with mixed dyslipidemia. The HDLAtherosclerosis Treatment Study (HATS) trial showed that the
combination of a statin plus niacin reduced the progression of
atherosclerosis and incidence of cardiovascular events. Several
large clinical trials are under way to assess the value of adding
either a fibrate (ACCORD) or extended-release niacin (AIMHIGH, HPS2-THRIVE) to a statin to reduce cardiovascular events
in high-risk patients.
Overview
There is increasing recognition, supported by clinical studies,
that elevated serum triglyceride (TG) levels are associated with an
increased risk for cardiovascular disease (CVD), independent of
the risk due to high levels of low-density lipoprotein cholesterol
(LDL-C) and low levels of high-density lipoprotein cholesterol
(HDL-C). Hypertriglyceridemia is usually associated with a
number of proatherogenic conditions such as insulin resistance,
endothelial dysfunction, inflammation, and thrombosis. Often,
hypertriglyceridemia occurs with elevated LDL-C levels and is
termed “mixed dyslipidemia.” Frequently, the high TG component of mixed dyslipidemia is undertreated because the usual
treatment is statin monotherapy. There is growing evidence that
the addition of fibrates, niacin, or purified omega-3 fatty acids
(P-OM3) to statin therapy is safe and effective not only for control
of lipid abnormalities but also for prevention of atherosclerosis.
A review of this evidence and treatment recommendations for
mixed dyslipidemia are included in this article.
S14 Supplement to Journal of Managed Care Pharmacy
JMCP
Coronary Risk Associated With Hypertriglyceridemia
A widespread misconception exists that high TG levels are not
associated with an increased risk for CVD. Although it is true that
the TG level itself does not contribute directly to atherogenesis,
TG-rich lipoproteins do carry cholesterol and can directly promote atherosclerosis. There appear to be at least 3 ways in which
hypertriglyceridemia makes lipoproteins more atherogenic—or
less antiatherogenic. First, in the presence of high TG levels,
the action of cholesteryl ester transfer protein drives excess TGs
from very low-density lipoproteins (VLDLs) into low-density
lipoproteins (LDLs) and high-density lipoproteins (HDLs) in
exchange for excess transfer of cholesteryl ester (CE) into VLDLs.
CE enrichment of VLDLs appears to make it more atherogenic.1
Second, the resulting CE depletion and TG enrichment of LDLs
result in greater susceptibility to TG lipolysis of LDLs by lipoprotein lipase and hepatic lipase, which in turn results in shrinkage
of both the lipid core and the lipoprotein itself. The resulting
small dense LDL particles are thought to be more atherogenic
than larger, less dense particles.2 Finally, the same sequence of
core lipid exchange and TG lipolysis also occurs in HDLs, resulting not only in smaller, denser particles, but also, more importantly, in the shedding of lipid-free or lipid-poor apolipoprotein
A-1 from HDLs,3 resulting in its permanent loss through renal
glomerular filtration.4 This loss is associated with the reduction
of HDL levels and its antiatherogenic effects.
The strong metabolic connection between TGs and HDLs
(mentioned previously) results in a strong inverse relationship
between their levels; nevertheless, several studies have confirmed
the direct association of hypertriglyceridemia to increased coronary risk. One study compared the serum lipid profiles of 653
patients who had premature familial coronary artery disease
(CAD) with those of 1,029 control subjects. Results indicate
that the risk associated with high TG levels is independent of
the risk related to high levels of HDL-C; therefore, patients with
elevated TG levels and average or high HDL-C values also have an
increased risk for CAD. The CAD risk for a patient with TG levels
≥500 mg per dL is more than 10 times that of a person with a
TG level < 100 mg per dL. At TG levels between 200 and 299 mg
per dL (levels commonly seen in patients with dyslipidemia), the
CAD risk is double that of patients with TG levels < 100 mg per
dL.5 The Prospective Cardiovascular Münster (PROCAM) study
followed 4,849 middle-aged men for 8 years to determine the
incidence of coronary heart disease (CHD) events in accordance
with the risk factors present at the start of the study. Results
indicated that TG levels have an impact on coronary risk that
is independent of LDL-C or HDL-C levels.6 The Pravastatin or
Atorvastatin Evaluation and Infection Therapy-Thrombolysis
in Myocardial Infarction 22 (PROVE IT-TIMI 22) trial, one of
the classic studies of statin therapy, compared treatment with
pravastatin 40 mg per day and atorvastatin 80 mg per day. Data
indicated that LDL-C levels achieved with statins predicted the
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
The Role of Statins in the Treatment of Mixed Dyslipidemia
Patients with mixed dyslipidemia are often undertreated. Many
patients with dyslipidemia and TG levels ≥200 mg per dL are
not at their LDL-C and non–HDL-C treatment goals. A survey
using electronic data-capture was undertaken to determine the
success rates of dyslipidemia treatment, as defined in the National
Cholesterol Education Program (NCEP) Adult Treatment Panel
III (ATP III) guidelines. For patients with hypertriglyceridemia,
both treatment goals were reached by 64% of the patients who
had ≤1 risk factor for CHD, by 52% of those with ≥2 risk factors,
and by only 27% of patients with CHD and CHD risk equivalents
(Figure 6). Lipid therapies tend to be more effective at reducing
LDL-C levels than non–HDL-C levels, especially on a percentage
basis.11 Because non–HDL-C levels predict CVD risk better than
LDL-C levels, lipid therapy in mixed dyslipidemia should focus
on closing this gap in efficacy; meeting the LDL-C goal may leave
the patient inadequately treated with regard to achievement of the
non–HDL-C goal.12
Increasing the dosage in statin monotherapy is one approach to
achieve additional reduction of TG levels in mixed dyslipidemia.
However, the dose-response curve of statins is relatively flat, and
higher statin doses are not always well tolerated. Statin intolerance
due to myopathy and other causes tends to be more prevalent in
elderly patients, and lower statin doses should be considered for
this population.13 For example, half of the normal statin dose
might be used in patients aged > 75 years. Similar considerations
apply to females and to patients with a small body frame, multisystem disease, or a family history of statin intolerance.14 Patients
who have hepatitis,15 myopathy,16 or fibromyalgia prior to statin
administration are at a higher risk for statin-induced muscle
and liver problems. Polypharmacy also increases risk because
www.amcp.org
Vol. 15, No. 1, S-c
FIGURE 6
Patients With Hypertriglyceridemia
Not at LDL-C and Non–HDL-C Goals
100
80
Goals achieved (%)
risk for death, myocardial infarction (MI), or recurrent acute
coronary syndrome. However, when TG levels were considered,
a substantial elevation of risk for future cardiovascular events
remained among patients with levels > 150 mg per dL who were
receiving aggressive statin monotherapy—a fact that is often
missed in clinical practice.7 Therefore, lipid-lowering treatments
that target elevated TG levels should be considered when developing a comprehensive therapeutic plan for reducing CVD risk in
patients with mixed dyslipidemia.
Because all lipoproteins other than HDLs are probably atherogenic, the total amount of cholesterol carried by these particles,
termed non–HDL-C, is a powerful and convenient index of atherosclerosis risk. Non–HDL-C is invariably elevated in hypertriglyceridemia, with or without mixed dyslipidemia, and predicts
CVD risk more strongly than does LDL-C.8 In addition (and
at least partially independent of the above), a number of lipidrelated proatherogenic conditions are linked to high TG levels
including insulin resistance, endothelial dysfunction, inflammation, thrombosis, and increased oxidation of lipoproteins and
other components of atherosclerotic plaque.7,9,10
60
78
64
71
52
57
52
40
27
50
33
44
25
20
0
17
CHD Diabetes Other
0-1 Risk ≥ 2 Risk CHD +
factors CHD risk (N =320) (no CHD) CHD risk
factor
equivalents
equivalents
(no CHD)
Achieved LDL-C goal
Achieved LDL-C and non–HDL-C goals
Source: Davidson MH et al. Am J Cardiol. 2005;96:556-63.11
CHD = coronary heart disease; LDL-C = low-density lipoprotein cholesterol;
non–HDL-C = non–high-density lipoprotein cholesterol.
of a greater potential for drug-drug interactions.14 Strenuous
physical activity also increases the risk for myopathy,17,18 which
is problematic because most patients are encouraged to exercise
regularly.19,20 The most common liabilities associated with statin
therapy are reviewed in the following sections.
Muscle Effects. Muscle-related problems with statin therapy
are well known and include muscle weakness, pain, inflammation, and, in some cases, rhabdomyolysis. While rhabdomyolysis
is rare, it is associated with myoglobinuria and acute renal failure
and is fatal in approximately 10% of cases.21
Liver Effects. A reversible elevation of liver transaminase is
sometimes seen with the use of statins, although serious cases of
liver damage are rarely, if ever, reported.22
Kidney Effects. The perspective on statin-related renal effects
has gradually evolved. Some studies, especially those involving
rosuvastatin, suggested proteinuria and hematuria.23 Because it is
highly water soluble, early concerns were raised that rosuvastatin
could cause renal damage; indeed, the highest dose requested
was not approved by the U.S. Food and Drug Administration due
to evidence of proteinuria. However, since then, accumulated
evidence suggests that rosuvastatin and other statins may actually
improve renal function.24
Neurologic Effects. Statin-related effects on sleep disorders
and cognition are controversial due to conflicting evidence.25-31
Drug-Drug Interactions. As discussed, drug-drug
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S15
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
interactions between the statins and gemfibrozil may increase the
risk of myopathy. Other agents, such as cyclosporine and certain
anti-infective agents, may interact adversely with statins; thus,
caution is required when combining such treatments. Likewise,
cardiac medicines, such as amiodarone and verapamil as well as
the antidepressant nefazodone, are known to interact negatively
with statins. Finally, if a patient consumes a large quantity of
grapefruit juice (i.e., more than 1 quart per day), the naringenin
contained in the juice can delay statin catabolism and thus
increase blood levels and potential toxicity.32-35
Medication Adherence With Statin Therapy. A number
of factors can negatively influence a patient’s adherence to statin therapy, which has been reported to drop to 50% or lower
after 1 year of treatment.36,37 For example, medication phobia
due primarily to perceived, rather than actual, safety issues can
impact adherence. Numerous factors contribute to statin phobia.
Patients who use the Internet or consult nonmedical professionals are often misinformed about the safety of statins. Those who
are skeptical of Western medicine may assume that they will
have a bad reaction to any drug, and others may be reluctant to
take statins if they subscribe to “conspiracy theories” about the
pharmaceutical industry. Others may overemphasize the use of
natural products and dietary supplements, falsely assuming that
anything derived from plant or animal sources must be safe.38-41
Some patients rely on dietary supplements and trust the claims
made about these products, yet they can be reluctant to believe
the benefits of prescription drugs.42 Nonadherence to statin therapy is a major problem. All medical professionals should strive to
overcome the misconceptions and other factors that are common
to medication nonadherence in general.
Combination Therapy for Mixed Dyslipidemia
Increasing the statin dose for patients with mixed dyslipidemia
may improve lipid levels to some extent, but not necessarily to recommended goals. Ezetimibe, an agent with a distinct mechanism
of action from that of the statins, was shown to reduce LDL-C
levels by inhibiting intestinal absorption of dietary cholesterol.
However, when added to statin therapy in the Effect of Ezetimibe
Plus Simvastatin Versus Simvastatin Alone on Atherosclerosis in
the Carotid Artery (ENHANCE) trial, ezetimibe failed to show
additional antiatherosclerotic benefits beyond statin use alone.43
An array of therapies is available to improve lipid levels in
patients with mixed dyslipidemia. Nonpharmacologic approaches
involve therapeutic lifestyle changes, such as increased exercise
and dietary changes.44-46 A number of diverse agents, such as
interferon,47 olanzapine,48 propofol,49 and human immunodeficiency virus protease inhibitors,50 have the potential to promote hypertriglyceridemia; therefore, removal and replacement
of these agents should be considered in therapeutic planning.
Statins have only modest effects on lowering TG and raising
HDL-C levels; thus, in treating mixed dyslipidemia, the addition
of fibrates, niacin, or P-OM3 is often required to bring TGs and
S16 Supplement to Journal of Managed Care Pharmacy
JMCP
HDL-C to target levels. Combination regimens that are useful
for the treatment of hypertriglyceridemia are summarized in the
following sections.
Fibrates Plus Statins. While fibrates have long been used
for the successful treatment of hypertriglyceridemia, evidence of
their cardiovascular benefits is inconsistent. Further research on
fibrate/statin combination therapy is ongoing and includes the
Action to Control Cardiovascular Risk in Diabetes (ACCORD)
trial, which may help to clarify the current and sometimes
conflicting evidence regarding the efficacy and cardioprotective
effects of fibrates. This study is investigating the control of coronary events in more than 10,000 high-risk patients with type 2
diabetes. Approximately half of the subjects are receiving lipid
treatment with simvastatin either with or without fibrates, while
the other half is being treated with various blood pressure–lowering agents. The outcome under investigation in the ACCORD trial
is the rate of major cardiovascular disease events (i.e., nonfatal
MI, stroke, or CVD death).51-55
Niacin Plus Statins. Niacin and the statins have different and
complementary effects on lipid profiles; therefore, combinations
of these agents would be expected to improve LDL-C, HDL-C,
and TG levels and further reduce coronary risk.56
The Comparative Effects on Lipid Levels (COMPELL) study
compared statin monotherapy using rosuvastatin (40 mg per
day) with combination therapy using atorvastatin (40 mg per
day) plus niacin (2,000 mg per day) and rosuvastatin (20 mg per
day) plus niacin (1,000 mg per day). When niacin was added to
statins, TG and HDL-C levels improved significantly compared
with statin monotherapy. HDL-C levels approximately tripled
with combination therapy versus monotherapy, and TG levels
were reduced by 33%-41% with combination therapy versus 19%
with rosuvastatin alone.57
The Safety and Efficacy of a Combination of ExtendedRelease Niacin and Simvastatin in Patients with Dyslipidemia
(SEACOAST) trial compared the effects of simvastatin 20 mg per
day alone with simvastatin 20 mg per day plus extended-release
niacin either 1 gm or 2 gm per day. The included patients were
at goal for LDL-C but not non–HDL-C levels. After 24 weeks
of therapy, niacin provided as much incremental improvement
in HDL-C and TG levels as simvastatin achieved in lowering
LDL-C levels. Compared with patients with normal TG levels,
those with elevated TG levels demonstrated a greater reduction
in non–HDL-C levels with niacin treatment. These data predict,
but do not prove, better cardiovascular outcomes with combination therapy.56
The cardiovascular benefits of treatment with simvastatin plus
niacin were compared with placebo in the HDL Atherosclerosis
Treatment Study (HATS). A major improvement in atherosclerosis was seen with the combination therapy; regression of stenosis
was demonstrated in the group treated with combination therapy
compared with progression in those who received placebo. While
these results are positive, the drug combination was not compared with statin monotherapy; thus, in this trial, nothing can be
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
FIGURE 7
FIGURE 8
Effect of Adding P-OM3 to Simvastatin
Therapy (COMBOS Trial)
60
Patients achieving
non–HDL-C goal (%)
50
Favors Treatment
Favors Control
51.6
Risk ratio (95% CI)
30
23.8
20
0.5
10
P-OM3 (4 gm per day)
+ simvastatin
Davidson MH et al. Clin Ther. 2007;29:1354-67.63
COMBOS = COMBination of prescription Omega-3 with Simvastatin; non–HDLC = non–high-density lipoprotein cholesterol; P-OM3 = purified omega-3.
concluded about the superiority of either therapy.58 The doubleblind, randomized, placebo-controlled Arterial Biology for the
Investigation of the Treatment Effects of Reducing Cholesterol
(ARBITER) 2 trial studied extended-release niacin added to statin
therapy in 167 patients with known CHD and low HDL-C levels.
The results demonstrated a downward trend in cardiovascular
events, but the difference in this very small study was not statistically significant.59
Two large ongoing studies of similar design should help verify
the efficacy of niacin plus statin combination therapy. In both trials, all participants are treated with a statin; half of these groups
also receive niacin and the other half is randomized to placebo
treatment. The Atherothrombosis Intervention in Metabolic
Syndrome with Low HDL/High Triglycerides and Impact on
Global Health Outcomes (AIM-HIGH) trial is being undertaken
in the United States with the target of following cardiovascular
end points in 3,300 patients with vascular disease for 3 to 5
years.60 The Heart Protection Study 2, a much larger European
trial enrolling 20,000 patients, has a similar design, but compares
simvastatin monotherapy with simvastatin combined with niacin
plus laropiprant (an agent under investigation for the control of
niacin-related flushing), with follow-up planned for 3-6 years.61
Omega-3 Fatty Acids Plus Statins. The Japan Eicosapentaenoic
Acid (EPA) Lipid Intervention Study (JELIS) compared statin
monotherapy with statins plus a purified form of EPA (not available in the United States). Nearly 19,000 patients received either
usual therapy with statins or 1.8 gm per day of EPA added to their
statin therapy and were followed for 4.6 years. A 19% reduction
in major coronary events was seen with combination therapy
Vol. 15, No. 1, S-c
0.8
1.0
1.25
Risk ratio
2.0
Adapted from Studer M et al. Arch Intern Med. 2005;165:725-30.65 Copyright
(2005), American Medical Association; all rights reserved.
n = number of studies; P-OM3 = purified omega-3. Trial of P-OM3 used different
dietary and nondietary sources with food supplements of n-3 fatty acids or n-3
fatty acid precursors.
Placebo +
simvastatin
www.amcp.org
0.68 (0.52-0.90), P <0.001
0.78 (0.72-0.84), P =0.42
0.91 (0.82-1.02), P =0.14
0.93 (0.81-1.08), P =0.13
0.95 (0.82-1.10), P =0.75
P-OM3 (n =12)
Statins ( n=33)
Diet (n =18)
Fibrates (n = 17)
Niacin (n =2)
40
0
Effect of Antilipidemic Agents and
Diet on Cardiac Mortality
compared with statin monotherapy, driven mainly by lower incidence rates of unstable angina. However, because this trial was
performed in Japan where the diet is typically rich in fish and
because the study product used (containing only EPA) is unavailable in the United States, the results may be difficult to translate
to the American population. Despite these factors, it is encouraging evidence that higher doses of P-OM3 acids added to statin
therapy can provide further cardiovascular risk reduction.62
When P-OM3 was added to simvastatin in the COMBination
of prescription Omega-3 with Simvastatin (COMBOS) trial,
statistically significant improvements were seen in non–HDLC, TG, VLDL-C, and LDL-C levels compared with simvastatin
monotherapy. The proportion of patients who achieved non–
HDL-C treatment goals in this study is illustrated in Figure 7.
Combination therapy also increased HDL levels comparable with
those seen with statins, but did not elicit as much improvement
as seen with niacin combinations.63 The COMBOS trial also
demonstrated that the combined agents increase the size of LDL
particles, thereby decreasing atherogenicity—an effect not seen
with fish oil alone.64
A meta-analysis of 97 randomized, controlled clinical trials
was undertaken to assess the efficacy and safety of lipid-lowering
interventions based on all-cause, cardiac, and noncardiovascular
mortality data. The studies included a combined total of more
than 270,000 patients. Results of this analysis are summarized in
Figures 8 and 9. The authors concluded that statins and omega-3
fatty acids offer the best lipid-lowering interventions and reduce
the risk of overall and cardiac mortality. The reduction in cardiac
mortality that was demonstrated by the use of fibrates is offset by
increased mortality from noncardiovascular causes.65
Several trials of omega-3 fatty acids are ongoing, primarily studying cardiovascular end points in addition to other
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S17
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
FIGURE 9
Effect of Antilipidemic Agents and
Diet on Overall Mortality
Favors Treatment
Favors Control
Risk ratio (95% CI)
0.77 (0.63-0.94), P<0.01
0.87 (0.81-0.94), P =0.05
0.96 (0.86-1.08), P =0.81
0.97 (0.91-1.04), P =0.19
1.00 (0.91-1.11), P =0.01
P-OM3 (n =14)
Statins (n=35)
Niacin (n =2)
Diet (n =18)
Fibrates (n= 17)
0.5
0.8
1.0
1.25
Risk ratio
2.0
Adapted from Studer M et al. Arch Intern Med. 2005;165:725-30.65 Copyright
(2005), American Medical Association; all rights reserved.
n = number of studies; P-OM3 = purified omega-3. Trial of P-OM3 used different
dietary and nondietary sources with food supplements of n-3 fatty acids or n-3
fatty acid precursors.
potential benefits. Clinical research is currently being undertaken
to study the effects of P-OM3 on congestive heart failure mortality
(Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto
Miocardico [GISSI]-Heart Failure [HF]), CVD in prediabetes
(Outcome Reduction With an Initial Glargine Intervention
[ORIGIN] trial), and primary prevention of CVD in diabetes mellitus (A Study of Cardiovascular Events in Diabetes [ASCEND]).66
This evolving field of study has the potential to provide clinically
relevant developments.
Conclusions
Hypertriglyceridemia is associated with an increased risk for
pancreatitis and CVD. Mixed dyslipidemia with high LDL-C levels associated with high TG and low HDL-C levels are common.
Medications for lowering TGs are available as monotherapy or
in varying combinations of statins, fibrates, P-OM3, and niacin.
When LDL-C levels are reduced to treatment goals with statin
monotherapy, considerable cardiovascular risk still remains.
Published evidence already suggests that the addition of niacin
or P-OM3 to a statin may improve cardiovascular outcomes
over statin monotherapy; however, definitive data are lacking.
Meanwhile, at present, there is no published evidence for additional cardiovascular benefits when either fibrates or ezetimibe
are added to statins compared with statin monotherapy. Ongoing
clinical trials should soon provide more conclusive data regarding
these clinically important questions.
REFERENCES
1. Shepherd J. Does statin monotherapy address the multiple lipid abnormalities in type 2 diabetes? Atheroscler Suppl. 2005;6(3):15-19.
S18 Supplement to Journal of Managed Care Pharmacy
JMCP
2. Satoh N, Wada H, Ono K, et al., Small dense LDL-cholesterol relative to
LDL-cholesterol is a strong independent determinant of hypoadiponectinemia in metabolic syndrome. Circ J. 2008;72(6):932-39.
3. Genest J. Lipoprotein disorders and cardiovascular risk. J Inherit Metab
Dis. 2003;26(2-3):267-87.
4. Rader DJ. Molecular regulation of HDL metabolism and function: implications for novel therapies. J Clin Invest. 2006;116(12):3090-100.
5. Hopkins PN, Wu LL, Hunt SC, Brinton EA. Plasma triglycerides and type
III hyperlipidemia are independently associated with premature familial
coronary artery disease. J Am Coll Cardiol. 2005;45(7):1003-12.
6. Assmann G, Schulte H, Funke H, von Eckardstein A. The emergence of
triglycerides as a significant independent risk factor in coronary artery disease. Eur Heart J. 1998;19(suppl M):M8-M14.
7. Miller M, Cannon CP, Murphy SA, Qin J, Ray KK, Braunwald E, for the
PROVE IT-TIMI 22 Investigators. Impact of triglyceride levels beyond lowdensity lipoprotein cholesterol after acute coronary syndrome in the PROVE
IT-TIMI 22 Trial. J Am Coll Cardiol. 2008;51(7):724-30.
8. Miller M, Ginsberg HN, Schaefer EJ. Relative atherogenicity and predictive value of non–high-density lipoprotein cholesterol for coronary heart
disease. Am J Cardiol. 2008;101(7):1003-08.
9. López S, Bermúdez B, Pacheco YM, Villar J, Abia R, Muriana FJ.
Distinctive postprandial modulation of beta cell function and insulin sensitivity by dietary fats: monounsaturated compared with saturated fatty acids.
Am J Clin Nutr. 2008;88(3):638-44.
10. Benítez MB, Cuniberti L, Fornari MC, et al., Endothelial and leukocyte adhesion molecules in primary hypertriglyceridemia. Atherosclerosis.
2008;197(2):679-87.
11. Davidson MH, Maki KC, Pearson TA, et al., Results of the National
Cholesterol Education (NCEP) Program Evaluation Project Utilizing
Novel E-Technology (NEPTUNE) II survey and implications for treatment
under the recent NCEP Writing Group Recommendations. Am J Cardiol.
2005;96(4):556-63.
12. Liu J, Sempos CT, Donahue RP, Dorn J, Trevisan M, Grundy SM.
Non–high-density lipoprotein and very-low-density lipoprotein cholesterol
and their risk predictive values in coronary heart disease. Am J Cardiol.
2006;98(10):1363-68.
13. Walker DB, Jacobson TA. Initiating statins in the elderly: the evolving
challenge. Curr Opin Endocrinol Diabetes Obes. 2008;15(2):182-87.
14. Sewright KA, Clarkson PM, Thompson PD. Statin myopathy: incidence,
risk factors, and pathophysiology. Curr Atheroscler Rep. 2007;9(5):389-96.
15. Chen YW, Lai HW, Wang TD. Marked elevation of liver transaminases
after high-dose fluvastatin unmasks chronic hepatitis C: safety and re-challenge. Acta Neurol Taiwan. 2007;16(3):163-67.
16. Toth PP, Harper CR, Jacobson TA. Clinical characterization and
molecular mechanisms of statin myopathy. Expert Rev Cardiovasc Ther.
2008;6(7):955-69.
17. Brancaccio P, Maffulli N, Limongelli FM. Creatine kinase monitoring in
sport medicine. Br Med Bull. 2007;81-82:209-30.
18. Clarkson PM. Exertional rhabdomyolysis and acute renal failure in
marathon runners. Sports Med. 2007;37(4-5):361-63.
19. Bruckert E, Hayem G, Dejager S, Yau C, Bégaud B. Mild to moderate
muscular symptoms with high-dosage statin therapy in hyperlipidemic
patients —The PRIMO study. Cardiovasc Drugs Ther. 2005;19(6):403–14.
20. Thompson PD, Clarkson P, Karas RH. Statin-associated myopathy.
JAMA. 2003:289(13):1681-90.
21. Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol.
2006;97(8A):52C-60C.
22. Onusko E. Statins and elevated liver tests: what’s the fuss? J Fam Pract.
2008;57(7):449-52.
23. Scott LJ, Curran MP, Figgitt DP. Rosuvastatin: a review of its use in
the management of dyslipidemia. Am J Cardiovasc Drugs. 2004;4(2):117-38.
Erratum in: Am J Cardiovasc Drugs. 2005;5(2):130.
24. Shepherd J, Vidt DG, Miller E, Harris S, Blasetto J. Safety of rosuvastatin:
update on 16,876 rosuvastatin-treated patients in a multinational clinical
trial program. Cardiology. 2007;107(4):433-43.
25. Boriani G, Biffi M, Strocchi E, Branzi A. Nightmares and sleep disturbances with simvastatin and metoprolol [editorial]. Ann Pharmacother.
2001;35(10):1292.
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
26. Partinen M, Pihl S, Strandberg T, et al., Comparison of effects on
sleep of lovastatin and pravastatin in hypercholesterolemia. Am J Cardiol.
1994;73(12):876-80.
27. Eckernäs SA, Roos BE, Kvidal P, et al., The effects of simvastatin and
pravastatin on objective and subjective measures of nocturnal sleep: a
comparison of two structurally different HMG CoA reductase inhibitors in
patients with primary moderate hypercholesterolaemia. Br J Clin Pharmacol.
1993;35(3):284-89.
28. Muldoon MF, Ryan CM, Sereika SM, Flory JD, Manuck SB. Randomized
trial of the effects of simvastatin on cognitive functioning in hypercholesterolemic adults. Am J Med. 2004;117(11):823-29.
29. Ehrenberg BL, Lamon-Fava S, Corbett KE, McNamara JR, Dallal GE,
Schaefer EJ. Comparison of the effects of pravastatin and lovastatin on sleep
disturbance in hypercholesterolemic subjects. Sleep. 1999;22(1):117-21.
30. Kostis JB, Rosen RC, Wilson AC. Central nervous system effects of HMG
CoA reductase inhibitors: lovastatin and pravastatin on sleep and cognitive performance in patients with hypercholesterolemia. J Clin Pharmacol.
1994;34(10):989-96.
31. Agostini JV, Tinetti ME, Han L, McAvay G, Foody JM, Concato J. Effects
of statin use on muscle strength, cognition, and depressive symptoms in
older adults. J Am Geriatr Soc. 2007;55(3):420-25.
32. Merck & Co., Inc. Zocor [package insert]. Whitehouse Station, NJ: 2008.
33. AstraZeneca Pharmaceuticals, LP. Crestor [package insert]. Wilmington,
DE: 2007.
34. Bristol-Myers Squibb Co. Pravachol [package insert]. Princeton, NJ:
2007.
35. Parke-Davis Division of Pfizer, Inc. Lipitor [package insert]. New York,
NY: 2007.
36. Ellis JJ, Erickson SR, Stevenson JG, Bernstein SJ, Stiles RA, Fendrick
AM. Suboptimal statin adherence and discontinuation in primary and secondary prevention populations: should we target patients with the most to
gain? J Gen Intern Med. 2004;19(6):638-45.
37. National Institutes of Health. Third report of the National Cholesterol
Education Program Expert Panel (NCEP) on Detection, Evaluation, and
Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III)
final report. National Heart, Lung, and Blood Institute; September 2002.
NIH Publication No. 02-5215. Available at: www.nhlbi.nih.gov/guidelines/
cholesterol/atp3full.pdf. Accessed January 22, 2009.
38. Lim J, Wong M, Chan MY, et al., Use of complementary and alternative medicine in paediatric oncology patients in Singapore. Ann Acad Med
Singapore. 2006;35(11):753-58.
39. Sharma P, Herrmann N, Rochon PA, et al., Perceptions of natural health
products among patients attending a memory clinic. Am J Alzheimers Dis
Other Demen. 2006;21(3):156-63.
40. Krasuski RA, Michaelis K, Eckart RE. The cardiovascular patient’s
perceptions of complementary and alternative medicine. Clin Cardiol.
2006;29(4):161-64.
41. Shahrokh LE, Lukaszuk JM, Prawitz AD. Elderly herbal supplement
users less satisfied with medical care than nonusers. J Am Diet Assoc.
2005;105(7):1138-40.
42. Khanderia U, Townsend KA, Erickson SR, Vlasnik J, Prager RL, Eagle
KA. Medication adherence following coronary artery bypass graft surgery:
assessment of beliefs and attitudes. Ann Pharmacother. 2008;42(2):192-99.
43. Kastelein JJ, Akdim F, Stroes ES, et al., for the ENHANCE Investigators.
Simvastatin with or without ezetimibe in familial hypercholesterolemia. N
Engl J Med. 2008;358(14):1431-43.
44. Buemann B, Tremblay A. Effects of exercise training on abdominal obesity and related metabolic complications. Sports Med. 1996;21(3):191-212.
45. Westheim A, Os I. Physical activity and the metabolic cardiovascular
syndrome. J Cardiovasc Pharmacol. 1992;20(suppl 8):S49-S53.
46. Matía Martín P, Lecumberri Pascual E, Calle Pascual AL. Nutrition and
metabolic syndrome [in Spanish]. Rev Esp Salud Publica. 2007;81(5):489-505.
47. Wong SF, Jakowatz JG, Taheri R. Management of hypertriglyceridemia
in patients receiving interferon for malignant melanoma. Ann Pharmacother.
2004;38(10):1655-59.
48. Markham-Abedi C, de Leon J. Hypertriglyceridemia associated with
direct effects of olanzapine rather than with weight gain: a case report. J Clin
Psychiatry. 2006;67(9):1473-74.
www.amcp.org
Vol. 15, No. 1, S-c
49. Devlin JW, Lau AK, Tanios MA. Propofol-associated hypertriglyceridemia and pancreatitis in the intensive care unit: an analysis of frequency
and risk factors. Pharmacotherapy. 2005;25(10):1348-52.
50. Thomas JC, Lopes-Virella MF, Del Bene VE, et al., Use of fenofibrate
in the management of protease inhibitor-associated lipid abnormalities.
Pharmacotherapy. 2000;20(6):727-34.
51. ACCORD Study Group. Action to Control Cardiovascular Risk
in Diabetes (ACCORD) trial: design and methods. Am J Cardiol.
2007;99(suppl):21i-33i.
52. Goff DC Jr, Gerstein HC, Ginsberg HN, et al., for the ACCORD
Study Group. Prevention of cardiovascular disease in persons with type
2 diabetes mellitus: current knowledge and rationale for the Action to
Control Cardiovascular Risk in Diabetes (ACCORD) trial. Am J Cardiol.
2007;99(12A):4i-20i.
53. Cushman WC, Grimm RH Jr, Cutler JA, et al., for the ACCORD Study
Group. Rationale and design for the blood pressure intervention of the
Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Am J
Cardiol. 2007;99(12A):44i-55i.
54. Williamson JD, Miller ME, Bryan RN, et al., for the ACCORD Study
Group. The Action to Control Cardiovascular Risk in Diabetes Memory in
Diabetes Study (ACCORD-MIND): rationale, design, and methods. Am J
Cardiol. 2007;99(12A):112i-22i.
55. National Institutes of Health. National Heart, Lung, and Blood Institute.
NIH News. For safety, NHLBI changes intensive blood sugar treatment strategy in clinical trial of diabetes and cardiovascular disease. www.nih.gov/
news/health/feb2008/nhlbi-06.htm. Accessed June 11, 2008.
56. Ballantyne CM, Davidson MH, McKenney J, Keller LH, Bajorunas DR,
Karas RH. Comparison of the safety and efficacy of a combination tablet
of niacin extended release and simvastatin vs simvastatin monotherapy in
patients with increased non-HDL cholesterol (from the SEACOAST I Study).
Am J Cardiol. 2008;101(10):1428-36.
57. McKenney JM, Jones PH, Bays HE, et al., Comparative effects on lipid
levels of combination therapy with a statin and extended-release niacin
or ezetimibe versus a statin alone (the COMPELL study). Atherosclerosis.
2007;192(2):432-37.
58. Brown BG, Zhao XQ, Chait A, et al., Simvastatin and niacin, antioxidant
vitamins, or the combination for the prevention of coronary disease. N Engl J
Med. 2001;345(22):1583-92.
59. Taylor AJ, Sullenberger LE, Lee HJ, Lee JK, Grace KA. Arterial Biology
for the Investigation of the Treatment Effects of Reducing Cholesterol
(ARBITER) 2: a double-blind, placebo-controlled study of extended-release
niacin on atherosclerosis progression in secondary prevention patients treated with statins. Circulation. 2004;110(23):3512-17. Erratum in: Circulation.
2004;110(23):3615.
60. National Institutes of Health. National Heart, Lung, and Blood Institute.
Niacin plus statin to prevent vascular events. www.clinicaltrials.gov/ct/
show/NCT00120289. Accessed June 10, 2008.
61. University of Oxford. Treatment of HDL to reduce the incidence
of vascular events: HPS2-THRIVE. www.clinicaltrials.gov/ct/show/
NCT00461630?order=1. Accessed June 10, 2008.
62. Yokoyama M, Origasa H, Matsuzaki M, et al., for the Japan EPA
Lipid Intervention Study (JELIS) Investigators. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients
(JELIS): a randomised open-label, blinded endpoint analysis. Lancet.
2007;369(9567):1090-98.
63. Davidson MH, Stein EA, Bays HE, et al., for the COMBination of prescription Omega-3 with Simvastatin (COMBOS) Investigators. Efficacy and
tolerability of adding prescription omega-3 fatty acids 4 g/d to simvastatin
40 mg/d in hypertriglyceridemic patients: an 8-week, randomized, doubleblind, placebo-controlled study. Clin Ther. 2007;29(7):1354-67.
64. Maki KC, Davidson MH, Bays HE, Stein EA, Shalwitz RA, Doyle R.
Effects of omega-3-acid ethyl esters on LDL particle size in subjects with
hypertriglyceridemia despite statin therapy. Poster presented at: 2007
Experimental Biology Annual Meeting; April 28-May 2, 2007; Washington,
DC.
65. Studer M, Briel M, Leimenstoll B, Glass TR, Bucher HC. Effect of different antilipidemic agents and diets on mortality: a systematic review. Arch
Intern Med. 2005;165(7):725-30.
66. ClinicalTrials.gov. Available at: http://clinicaltrials.gov. Accessed
September 16, 2008.
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S19
C O N T I N U I N G E D UCAT I O N
Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
Activity Release Date: April 17, 2008
Activity Expiration Date: April 17, 2011
Accreditation Statement and Credit Designation
Extension Services in Pharmacy at the University of Wisconsin-Madison School of Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of pharmacy continuing education. This program
has been approved for a maximum of 2 hours (0.2 CEUs) of pharmacy continuing education credit.
Credit Requirements
There are no fees for participating in and receiving CE credit for this activity. To obtain CE credit for participating in this activity
during the period April 17, 2008 through April 17, 2011, participants must:
1.Read the educational objectives and disclosure statements.
2.Study the educational content of the activity.
3.Complete/submit the posttest by recording the best answer to each question.
4. Submit a completed activity evaluation form.
To receive CE credit for this program, you must complete the following forms online:
1.Posttest form for this program, “Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol” on the AMCP.org Online
Learning Center site. To receive CE credit, you must receive a score of at least 70%. You will have 2 opportunities
to pass the posttest.
2.Activity evaluation form.
Upon successful completion of this activity, you will automatically receive your CE statement. Your CE credits will
be automatically archived and tracked for you on the AMCP.org (CE/CME Center) site. All information is kept confidential.
Note: There is no fee to participate in this activity.
S20 Supplement to Journal of Managed Care Pharmacy
JMCP
February 2009
Vol. 15, No. 1, S-c www.amcp.org
Posttest Worksheet: Maintaining Cardiovascular Health in Patients With Mixed Dyslipidemia:
Optimizing the Management of Hypertriglyceridemia and Non-HDL Cholesterol
1. Which of the following is not a diagnostic criterion for the
metabolic syndrome?
a. Increased waist circumference
b. Elevated blood pressure
c. Low triglyceride levels
d. Low levels of HDL-C
6. Over-the-counter fish oil supplements generally contain
about ____% EPA/DHA.
a. 30
b. 50
c. 70
d. 90
2. According to the NCEP ATP-III guidelines, a normal
triglyceride level is
a. <300 mg per dL
b. <250 mg per dL
c. <200 mg per dL
d. <150 mg per dL
7. Which of the following is not thought to be involved in
the triglyceride-lowering effects of the fibrates?
a. Peroxisome proliferator-activated receptor-alpha
b. Retinoid X receptor ligand
c. Acyl-CoA synthase
d. HMG-CoA reductase
3. Which of the following describes non–HDL-C?
a. It is a primary target for intervention in patients with
elevated LDL-C.
b. It may be a better marker of cardiovascular risk than
LDL-C.
c. Increased levels are consistently associated with
increases in LDL-C.
d. It is measured by calculating total cholesterol minus
LDL-C.
8. A flushing reaction is commonly associated with which of
the following medications?
a. P-OM3
b. Naringenin
c. Laropiprant
d. Niacin
4. Approximately what percentage of patients with
cardiovascular disease is at LDL-C and non–HDL-C
treatment goals?
a. 10%
b. 20%
c. 40%
d. 60%
5. According to American Heart Association
recommendations, what amount of omega-3 fatty acids
should be ingested by individuals with cardiovascular
disease?
a. 0.5 gm EPA/DHA per day
b. 1 gm EPA/DHA per day
c. 4 gm EPA/DHA per day
d. 6 gm EPA/DHA per day
9. Which of the following is not a proatherogenic condition
associated with hypertriglyceridemia?
a. Insulin resistance
b. Decreased lipoprotein oxidation
c. Endothelial dysfunction
d. Inflammation
10. When P-OM3 was added to statin therapy, significant
improvement was seen in which of the following lipid
parameters?
a. Triglycerides
b. Non–HDL-C
c. HDL-C
d. All of the above
To complete this activity, go to http://www.amcp.org,
where you will access the posttest and evaluation form.
www.amcp.org
Vol. 15, No. 1, S-c
February 2009
JMCP
Supplement to Journal of Managed Care Pharmacy S21
Supplement