3146 PART 12 Endocrinology and Metabolism
Familial Combined Hypolipidemia Nonsense mutations in both
alleles of the gene angiopoietin-like 3 (ANGPTL3) lead to low plasma
levels of all three major lipid fractions—TG, LDL-C, and HDL-C—a
phenotype termed familial combined hypolipidemia. ANGPTL3 is a protein synthesized by the liver and secreted into the bloodstream. It inhibits
LPL, thus delaying clearance of TRLs from the blood and increasing TRL
blood concentrations. Deficiency of ANGPTL3, therefore, raises LPL
activity and lowers blood TG; it also lowers LDL-C and raises HDL-C
levels apparently related to the effects of ANGPTL3 on endothelial lipase.
ANGPTL3 deficiency is associated with a reduced risk for CHD. The discovery of this condition led to the development of therapies that antagonize or silence ANGPTL3 to reduce LDL-C and TG levels (see below).
DISORDERS ASSOCIATED WITH REDUCED
HIGH-DENSITY LIPOPROTEINS
Low levels of HDL-C, generally defined as <50 mg/dL in women and
<40 mg/dL in men, are very common in clinical practice. Low HDL-C
is an important independent predictor of increased cardiovascular
risk and has been used regularly in standardized risk calculators. As
an independent risk factor, it has clinical value in the assessment of
cardiovascular risk, and a patient with low HDL-C should generally
be considered at higher risk of ASCVD. However, it is now considered doubtful that low HDL-C is directly causal for the development
of ASCVD. Thus, while HDL-C remains an important biomarker for
assessing cardiovascular risk, it is not considered a particularly attractive target for therapeutic intervention to raise HDL-C levels in order
to reduce cardiovascular risk. HDL-targeted therapies that, remain in
clinical development include a CETP inhibitor and an intravenous
infusion of a lipidated apoA-I particle.
HDL metabolism is strongly influenced by TG metabolism, insulin
resistance, and inflammation, among other environmental and medical
factors. Thus, the HDL-C measurement integrates a number of cardiovascular risk factors, potentially explaining its strong inverse association
with ASCVD. The majority of patients with low HDL-C have some
combination of genetic predisposition and secondary factors. Variants
in hundreds of genes have been shown to influence HDL-C levels.
Even more important quantitatively, obesity and insulin resistance have
strong suppressive effects on HDL-C, and low HDL-C in these conditions is widely observed. Furthermore, the vast majority of patients
with elevated TGs have reduced levels of HDL-C due to the substantial
interplay between the metabolism of TRLs and HDL (see above). Most
patients with low HDL-C who have been studied in detail have accelerated catabolism of HDL and its associated apoA-I protein as the physiologic basis for the low HDL-C. Single-gene Mendelian disorders that
reduce HDL-C activity have been described (Table 407-2) but are rare;
the vast majority of patients with low HDL-C have a polygenic predisposition with secondary factors like obesity, insulin resistance, or HTG.
■ PRIMARY (GENETIC) CAUSES OF LOW HDL-C
Mutations in three key genes encoding proteins that play critical roles
in HDL synthesis and catabolism result in hypoalphalipoproteinemia
(primary low levels of HDL-C). Unlike the genetic forms of hypercholesterolemia, which are invariably associated with premature coronary
atherosclerosis, genetic forms of hypoalphalipoproteinemia are usually
not associated with clearly increased risk of ASCVD. Nevertheless, in
the clinical setting of an HDL-C level <20 mg/dL without accompanying severe HTG, these rare conditions should be considered.
Gene Deletions and Missense Mutations in APOA1 Complete genetic deficiency of apoA-I due to a complete deletion of the
APOA1 gene results in the virtual absence of circulating HDL, proving
the critical role of apoA-I in HDL biogenesis. The APOA1 gene is part
of a gene cluster on chromosome 11 that includes APOA5, APOC3, and
APOA4. Some patients with no apoA-I have large genomic deletions
that include other genes in the cluster. The rare patient lacking apoA-I
can have cholesterol deposits in the cornea and in the skin, and in
contrast to the other genetic disorders of low HDL-C, premature CHD
has been reported. Heterozygotes for apoA-I deletions have reduced
HDL-C levels but no obvious clinical sequelae.
More common, but still rare, are heterozygous missense mutations
in the APOA1 gene associated with low plasma levels of HDL-C. The
first example reported, and still the best known, is an Arg173Cys substitution in apoA-I (so-called apoA-IMilano), found in multiple residents
of a town in northern Italy. Heterozygotes for this mutation have very
low plasma levels of HDL-C (<25 mg/dL) due to impaired LCAT activation and accelerated clearance of the HDL particles containing the
abnormal apoA-I. Despite having very low plasma levels of HDL-C,
these individuals do not appear have an increased risk of premature CHD (neither are they protected against CHD as was initially
believed). Multiple other rare APOA1 missense mutations causing low
HDL-C have been reported. A few of these mutations in APOA1 (as
well as some mutations in APOA2) promote the formation of amyloid
fibrils, causing systemic amyloidosis.
Tangier Disease (ABCA1 Deficiency) Tangier disease is a rare
autosomal co-dominant form of extremely low plasma HDL-C levels
that is caused by mutations in the ABCA1 gene encoding ABCA1, a
cellular transporter that facilitates efflux of unesterified cholesterol
and phospholipids from cells to apoA-I as an acceptor (Fig. 407-3).
Through transporting cellular lipids, ABCA1 in the hepatocytes and
intestinal enterocytes promotes the extracellular lipidation of the
apoA-I secreted from the basolateral membranes of these tissues. In
the genetic absence of ABCA1, the nascent, poorly lipidated apoA-I
is rapidly cleared from the circulation. Thus, patients with Tangier
disease (both ABCA1 alleles mutated) have extremely low circulating
plasma levels of HDL-C (<5 mg/dL) and apoA-I (<5 mg/dL). Cholesterol accumulates in the reticuloendothelial system of these patients,
resulting in hepatosplenomegaly and pathognomonic enlarged, grayish yellow or orange tonsils. An intermittent peripheral neuropathy
(mononeuritis multiplex) or a sphingomyelia-like neurologic disorder
can also be seen in this disorder. Tangier disease may be associated
with some increased risk of ASCVD, although the association is not
as robust as might have been anticipated, given the extremely low
levels of HDL-C in these patients. Patients with Tangier disease also
have low plasma levels of LDL-C, which may attenuate the atherosclerotic risk. Heterozygotes for ABCA1 mutations have moderately
reduced plasma HDL-C levels (~15–40 mg/dL), and the effect on risk
of ASCVD remains uncertain.
Familial LCAT Deficiency This rare autosomal recessive disorder is caused by mutations in LCAT, an enzyme synthesized in the
liver and secreted into the plasma, where it circulates associated with
lipoproteins (Fig. 407-3). As reviewed above, the enzyme is activated
by apoA-I and mediates the esterification of cholesterol to form CEs.
Consequently, in familial LCAT deficiency, the proportion of free cholesterol in circulating lipoproteins is greatly increased (from ~25% to
>70% of total plasma cholesterol). Deficiency in this enzyme interferes
with the maturation of HDL particles and results in rapid catabolism
of circulating apoA-I.
Two genetic forms of familial LCAT deficiency have been described
in humans: complete deficiency (also called classic LCAT deficiency)
and partial deficiency (also called fish eye disease). Progressive corneal
opacification due to the deposition of free cholesterol in the cornea,
very low plasma levels of HDL-C (usually <10 mg/dL), and variable
HTG are characteristic of both disorders. In partial LCAT deficiency,
there are no other known clinical sequelae. In contrast, patients with
complete LCAT deficiency have hemolytic anemia and progressive
renal insufficiency that eventually leads to end-stage renal disease.
Remarkably, despite the extremely low plasma levels of HDL-C and
apoA-I, premature ASCVD is not a consistent feature of either LCAT
deficiency or fish eye disease. The diagnosis can be confirmed in a specialized laboratory by assaying plasma LCAT activity or by sequencing
the LCAT gene.
Primary Hypoalphalipoproteinemia Primary hypoalphalipoproteinemia is defined as a plasma HDL-C level below the tenth percentile in the setting of relatively normal cholesterol and TG levels, no
apparent secondary causes of low plasma HDL-C, and no clinical signs
of LCAT deficiency or Tangier disease. This syndrome is often referred
3147 Disorders of Lipoprotein Metabolism CHAPTER 407
to as isolated low HDL. A family history of low HDL-C suggests an
inherited condition and may trigger an evaluation of one of the Mendelian causes of hypoalphalipoproteinemia. However, most patients with
isolated low HDL do not have an identifiable single-gene disorder and
likely have a polygenic etiology, possibly exacerbated by a secondary
factor. The physiologic defect appears to be accelerated catabolism of
HDL and its apolipoproteins. Several kindreds with primary hypoalphalipoproteinemia and an increased incidence of premature CHD
have been described, although it is not clear if the low HDL-C level is
the cause of the accelerated atherosclerosis in these families.
■ SECONDARY FACTORS THAT
REDUCE HDL-C LEVELS
Hypertriglyceridemia Low HDL-C is very commonly found in
association with elevated TG levels. The lipolysis of TRLs generates
lipids that transfer to HDL, and therefore, any impairment of lipolysis
(the most common cause of elevated TGs) leads to reduced HDL biosynthesis. In settings of elevated TGs, where the HDL-C is not reduced,
alternative explanations (e.g., FDBL, alcohol, estrogens) should be considered. Conversely, an isolated low HDL-C in the presence of normal
TGs should prompt consideration of a primary genetic etiology (as
above) or specific secondary factors (see below).
Very-Low-Fat Diet Dietary fat is positively associated with HDL-C
levels. Individuals who eat very-low-fat vegan diets or who have
anorexia or severe fat malabsorption often have low levels of HDL-C
that are secondary to low dietary fat. In this setting, LDL-C levels are
also usually low as well. There is no known harm to low HDL-C levels
in this setting and no indication for liberalizing the diet solely for the
purpose of raising the HDL-C.
Sedentary Lifestyle and Obesity Physical activity is known to
have a (generally modest) effect in raising HDL-C levels, and conversely, a sedentary lifestyle is often associated with low HDL-C levels.
Concordant with that observation, obesity is frequently associated with
low HDL-C levels even when overt insulin resistance or HTG is not
present. Increased physical activity and weight loss usually have some
effect in raising HDL-C, which is not the primary reason for recommending these interventions but can have a motivating influence on
the patient.
ANABOLIC STEROIDS AND TESTOSTERONE Anabolic steroids have
a well-established effect on lowering HDL-C levels, sometimes quite
dramatically. Testosterone supplementation can also reduce HDL-C
levels, although not to the degree caused by anabolic steroids. In a
young male patient who presents with unexplained very low HDL-C,
a careful history of medication and supplement use should be taken.
APPROACH TO THE PATIENT
Lipoprotein Disorders
The major goals in the diagnosis and clinical management of lipoprotein disorders are (1) prevention of acute pancreatitis in patients
with severe HTG and (2) prevention of CVD and related cardiovascular events. Given the high prevalence of dyslipidemia and the
proven clinical benefits of early diagnosis and initiation of therapy,
it is essential that physicians screen lipids systematically, rule out
secondary causes of dyslipidemia, suspect inherited disorders of
lipoprotein metabolism where appropriate, actively promote family-based cascade screening, carefully assess risk for ASCVD and
consider additional risk stratification approaches, and be knowledgeable about the wide range of existing therapeutic options for
dyslipidemia. The field of clinical lipidology has matured and is
moving toward a more systematic clinical application of genomic
medicine. Diagnostic DNA sequencing or genotyping in patients
with suspected FCS, FPLD, FH, and FDBL has the potential to
enhance molecular diagnosis, facilitate appropriate therapeutic
interventions, and promote family-based cascade screening.
DIAGNOSIS
A critical first step in managing a lipoprotein disorder is to attempt
to determine the class or classes of lipoproteins that are increased
or decreased in the patient. Once the dyslipidemia is accurately
classified, efforts should be directed to identify or rule out any possible secondary causes (Table 407-3). A careful social, medical, and
family history should be obtained. In patients with elevated TG levels (>150 mg/dL), a fasting glucose and/or hemoglobin A1c should
be obtained to rule out diabetes. In patients with elevated LDL-C
levels (>160 mg/dL), a TSH should be obtained to rule out hypothyroidism and consideration should be given to the possibility of
liver or kidney disease. Once secondary causes have been ruled
out, attempts should be made to diagnose a primary lipid disorder,
because the underlying genetic defect can provide important prognostic information regarding the risk of pancreatitis in severe HTG
and the risk of ASCVD in other dyslipidemias, as well as impact
on the choice of drug therapy and the screening of other family
members. Obtaining the correct diagnosis often requires a detailed
family history, lipid analyses in family members, and sometimes
specialized or genetic testing.
Severe Hypertriglyceridemia If the fasting plasma TG level is
>500 mg/dL, the patient has severe HTG and may be at risk for
pancreatitis. If the TG levels are persistently severely elevated,
especially if they are >1000 mg/dL, and the total cholesterol-to-TG
ratio is >8, FCS should be considered, and genetic testing of an FCS
gene panel may be indicated (Table 407-2). If central obesity, insulin
resistance, and/or fatty liver disease are also present, consideration
should be given to the possibility of FPLD, and an FPLD gene panel
may be indicated (Table 407-2). However, most individuals with
severe HTG do not have a single-gene disorder but have increased
polygenic risk for high TGs often exacerbated by secondary factors (diet, alcohol, obesity, insulin resistance, medications). Such
patients are still at risk for acute pancreatitis and should be treated
to reduce their TG levels and thus their risk of pancreatitis (see
below).
Hypercholesterolemia If the LDL-C levels are >190 mg/dL, the
patient has severe hypercholesterolemia and is at risk for premature
ASCVD. In absence of secondary causes, FH should be considered,
particularly if there is a family history of hypercholesterolemia
and/or premature CHD, and genetic testing of an FH gene panel
may be indicated (Table 407-2). While FH is a clinical diagnosis,
a finding of a causal mutation may appropriately result in earlier
and more aggressive therapy to lower LDL-C and should also
promote family-based cascade screening as per the Centers for
Disease Control and Prevention guidelines labeling FH as a Tier 1
condition. Recessive forms of severe hypercholesterolemia are rare,
but if a patient with severe hypercholesterolemia has parents with
normal cholesterol levels, ARH, sitosterolemia, and LALD should
be considered, and genetic testing may be indicated (Table 407-2).
Patients without an identified genetic variant or who have more
moderate hypercholesterolemia are likely to have polygenic hypercholesterolemia but should still be considered at risk and eligible for
treatment (see below).
Mixed Hyperlipidemia Elevations in fasting plasma levels of both
TGs (>150 mg/dL) and LDL-C (>130 mg/dL), often accompanied
by reduced levels of HDL-C (<40 mg/dL in men and <50 mg/dL in
women), are common, and such patients are often diagnosed as having “mixed hyperlipidemia.” Most such patients are at increased risk
of ASCVD and merit consideration of lifestyle and/or pharmacologic interventions. Secondary factors, particularly obesity, insulin
resistance, and type 2 diabetes, are common in such patients, who
often also have increased polygenic risk for dyslipidemia. The presence of palmar or tuberous xanthomas or an unusual lipid profile
of total cholesterol and TG levels in the same range with an HDL-C
that is not reduced should prompt consideration of FDBL, or type
III hyperlipidemia, and can be diagnosed by a nuclear magnetic
3148 PART 12 Endocrinology and Metabolism
resonance (NMR) lipoprotein profile or genetic testing for the
APOE2 genotype. FDBL patients should be managed aggressively
due to substantially increased risk of ASCVD. More commonly,
patients with mixed hyperlipidemia, particularly those with family
histories of dyslipidemia or premature ASCVD, have familial combined hyperlipidemia (FCHL). ApoB should be measured in such
patients, and the finding of substantially elevated apoB levels can
help identify patients with FCHL, who are at especially increased
risk of ASCVD and require more aggressive treatment.
TREATMENT
Severe Hypertriglyceridemia
There is a well-established observational relationship between
severe HTG, particularly chylomicronemia, and acute pancreatitis;
however, there has never been a clinical trial designed or powered to
definitively prove that intervention to reduce TGs reduces the risk
of pancreatitis. Nevertheless, it is generally considered appropriate
medical practice to intervene in patients with TGs >500 mg/dL in
order to reduce the risk of pancreatitis. It remains uncertain whether
chylomicronemia increases risk for ASCVD per se. Importantly,
moderate HTG (TG 150–500 mg/dL) is associated with increased
ASCVD risk; management of these patients is focused on reducing
risk of ASCVD and on reducing LDL-C, non-HDL-C, and apoB.
LIFESTYLE AND MODIFIABLE FACTORS
In patients with severe HTG, lifestyle modification can be associated
with a significant reduction in plasma TG level. Patients who drink
alcohol should be encouraged to decrease or preferably eliminate
their intake. Patients with severe HTG often benefit from a formal
dietary consultation with a dietician intimately familiar with counseling patients on the dietary management of high TGs. Dietary fat
intake should be restricted to reduce the formation of chylomicrons
in the intestine. The excessive intake of simple carbohydrates should
be discouraged because insulin drives TG production in the liver.
Aerobic exercise and even increase in regular physical activity can
have a positive effect in reducing TG levels and should be strongly
encouraged. For patients who are overweight, weight loss can help
to reduce TG levels. In extreme cases, bariatric surgery has been
shown to not only produce effective weight loss but also substantially reduce plasma TG levels. Many patients with diabetes have
HTG, and better control of diabetes can result in lowering of TGs.
Finally, certain medications can exacerbate HTG (Table 407-3).
PHARMACOLOGIC THERAPY
Despite lifestyle interventions, many patients with severe HTG
require pharmacologic therapy (Table 407-4). Patients who persist
in having fasting TG >500 mg/dL despite active lifestyle management are candidates for pharmacologic therapy. The two major
classes of drugs used for management of these patients are fibrates
and omega-3 fatty acids (fish oils). In addition, statins can reduce
plasma TG levels and also reduce ASCVD risk and should be used
in patients with severe HTG who are at increased risk of ASCVD.
Fibrates Fibric acid derivatives, or fibrates, are agonists of PPARα,
a nuclear receptor involved in the regulation of lipid metabolism.
Fibrates stimulate LPL activity (enhancing TG hydrolysis), reduce
apoC-III synthesis (enhancing lipoprotein remnant clearance),
promote β-oxidation of fatty acids, and may reduce VLDL TG
production. Fibrates reduce TG levels by ~30% in individuals
with severe HTG and are often used as first-line therapy. They
sometimes modestly raise LDL-C levels. Fibrates are generally well
tolerated but can cause myopathy, especially when combined with
statins, can raise creatinine, and are associated with an increase in
gallstones. Fibrates can potentiate the effect of warfarin and certain
oral hypoglycemic agents.
Omega-3 Fatty Acids (Fish Oils) Omega-3 fatty acids, or omega-3
polyunsaturated fatty acids (n-3 PUFAs), commonly known as fish
oils, are present in high concentration in fish and in flaxseed. The
n-3 PUFAs used for the treatment of HTG are eicosapentaenoic
acid (EPA) and docosahexaenoic acid (DHA). n-3 PUFAs have been
concentrated into capsules and in doses of 3–4 g/d are effective at
lowering fasting TG levels by ~30%. Fish oils can cause an increase
in plasma LDL-C levels in some patients. Icosapent ethyl is an EPAonly product that has been shown to reduce cardiovascular events
in patients with HTG. In general, fish oils are well tolerated, with
the major side effect being dyspepsia. They appear to be safe, at least
at doses up to 3–4 g, but can be associated with a prolongation in the
bleeding time. Fish oils can be first-line therapy for the treatment of
severe HTG or can be used in combination with fibrates.
APOC3 Silencing ApoC-III inhibits LPL and TRL uptake, and
genetic variants in the APOC3 gene reduce TG levels and risk of
ASCVD. Volanesorsen is an antisense oligonucleotide targeted
to the APOC3 mRNA in the liver; it significantly reduces plasma
apoC-III and TG levels and is approved in Europe for patients with
FCS. It has been associated with severe thrombocytopenia. Additional therapeutic approaches to APOC3 and other targets for TG
lowering are in development.
Hypercholesterolemia (Elevated LDL-C
with or without Elevated TG)
There are abundant and compelling data that intervention to
reduce LDL-C substantially reduces the risk of ASCVD, including
myocardial infarction and stroke, as well as total mortality. Thus, it
is imperative that patients with hypercholesterolemia be carefully
assessed for cardiovascular risk and need for intervention. It is also
worth emphasizing that patients with or at high risk for ASCVD
who have plasma LDL-C levels in the “normal” or average range
also benefit from intervention to reduce LDL-C levels.
LIFESTYLE AND MODIFIABLE FACTORS
In patients with elevated LDL-C, lifestyle modifications can be
effective but are often less effective than in HTG. Patients should
receive dietary counseling to reduce the content of saturated fats
and trans fats in the diet. Obese patients should make an effort to
lose weight. Regular aerobic exercise has relatively little impact on
reducing plasma LDL-C levels, although it has cardiovascular benefits independent of LDL-C lowering. Patients with hypothyroidism
should be optimally controlled. Finally, certain medications can
elevate LDL-C levels (Table 407-3).
PHARMACOLOGIC THERAPY
The decision to use LDL-lowering drug therapy (Table 407-4)—
with a statin being first-line therapy—depends on the presence
of ASCVD or, if absent, the level of LDL-C as well as the level of
cardiovascular risk. In patients with established ASCVD, drug
therapy to reduce LDL-C is well supported by clinical trial data to
reduce LDL-C as long as it remains >70 mg/dL, using combination
drug therapy if necessary. In the absence of ASCVD, patients with
FH must be treated to reduce the very high lifetime risk of ASCVD,
and treatment should be initiated as early as possible, ideally during
childhood. Otherwise, the decision to initiate LDL-lowering drug
therapy is generally determined by the level of cardiovascular risk.
For patients >40 years old without clinical CVD, the AHA/ACC
risk calculator can be used to determine the 10-year absolute risk
for CVD, and current guidelines suggest that a 10-year risk >7.5%
merits consideration of statin therapy regardless of plasma LDL-C
level. For younger patients, the assessment of lifetime risk of CVD
may help inform the decision to start a statin, as well as a careful
assessment of family history of ASCVD. In patients for whom the
decision to start a statin is uncertain due to borderline ASCVD
risk and/or borderline LDL-C levels, additional risk stratification
might be considered. Blood tests that predict ASCVD risk beyond
3149 Disorders of Lipoprotein Metabolism CHAPTER 407
traditional risk factors include apoB, Lp(a), and high-sensitivity
C-reactive protein (hs-CRP). In patients who are of a sufficient age
(men >40 years and women >50 years), a coronary artery calcium
(CAC) score has been shown to provide independent information
about risk of CAD. Elevated levels of one or more of these biomarkers or an elevated CAC score might be used to justify initiation
of statin therapy in primary prevention for patients who are in a
borderline zone with regard to treatment. Finally, given the strong
polygenic contribution to ASCVD, there is increasing interest in
the concept that a polygenic risk score for CAD might eventually
be of clinical utility in lifetime risk assessment and decision-making
regarding statin therapy in primary prevention.
HMG-CoA Reductase Inhibitors (Statins) Statins inhibit HMGCoA reductase, a key enzyme in cholesterol biosynthesis. By
inhibiting cholesterol synthesis in the liver, statins lead to a counterregulatory increase in the expression of the LDL receptor and thus
accelerated clearance of circulating LDL, resulting in a dose-dependent
reduction in plasma levels of LDL-C. The magnitude of LDL-C
lowering associated with statin treatment (~30–55%) varies by
statin and among individuals, but once a patient is on a statin, the
doubling of the statin dose produces a ~6% further reduction in the
level of plasma LDL-C. An extensive body of randomized clinical
trials has clearly established that statin therapy significantly reduces
major cardiovascular events (and in some cases total mortality) in
both primary and secondary prevention settings. The seven statins
currently available differ in their LDL-C–reducing potency
(Table 407-4). Current recommendations are to use high-intensity
statin therapy in patients with ASCVD or deemed at high risk of
ASCVD. Statins also reduce plasma TGs in a dose-dependent fashion, which is roughly proportional to their LDL-C–lowering effects.
Statins, taken in tablet form once a day, are remarkably safe
and well tolerated. The most important side effect associated
with statin therapy is muscle pain, or myalgia, which occurs in
3–5% of patients, some of whom are unable to tolerate any statin.
Severe myopathy (associated with an increase in plasma creatine
kinase [CK]) and even rhabdomyolysis can occur rarely with statin
treatment. The risk of statin-associated myalgia or myopathy is
increased by the presence of older age, frailty, renal insufficiency,
and coadministration of drugs that interfere with the metabolism
of statins, such as erythromycin and related antibiotics, antifungal agents, immunosuppressive drugs, and fibric acid derivatives
(particularly gemfibrozil). In the event of muscle symptoms, a
plasma CK level may be obtained to differentiate myopathy from
myalgia. Serum CK levels need not be monitored on a routine basis
in patients taking statins because an elevated CK in the absence of
symptoms does not predict the development of myopathy and does
not necessarily suggest the need for discontinuing the drug. Statins
can result in elevation in liver transaminases (alanine aminotransferase [ALT] and aspartate aminotransferase [AST]), but it is usually
mild and transient and generally does not require discontinuation.
Finally, meta-analyses of large randomized controlled clinical trials
with statins indicate a slight excess in incident type 2 diabetes, an
observation as yet not fully understood. However, the cardiovascular benefits associated with statin therapy far outweigh the slight
increase in incident diabetes. Based on their safety and extensively
documented benefit with regard to cardiovascular outcomes, statins
are the clear drug class of choice for LDL-C reduction and are by far
the most widely used class of lipid-lowering drugs.
Cholesterol Absorption Inhibitor Cholesterol within the lumen of
the small intestine is derived from the diet (about one-third) and
the bile (about two-thirds) and is actively absorbed by the enterocyte through a process that involves the protein NPC1L1. Ezetimibe (Table 407-4) is a cholesterol absorption inhibitor that binds
directly to and inhibits NPC1L1 and blocks the intestinal absorption of cholesterol. Ezetimibe (10 mg taken once daily) inhibits
cholesterol absorption by almost 60%, resulting in a reduction in
delivery of dietary sterols in the liver and a compensatory increase
in hepatic LDL receptor expression. The mean reduction in plasma
LDL-C on ezetimibe (10 mg) is 18%, and the effect is additive when
used in combination with a statin. Effects on TG and HDL-C levels
are negligible. Ezetimibe added to a statin has been shown to significantly reduce major cardiovascular events compared with statin
alone. It is generally considered the second-line option for adding
to a statin in order to achieve further LDL-C reduction. Ezetimibe
is very safe and well-tolerated. When used in combination with
a statin, monitoring of liver transaminases is recommended. The
only roles for ezetimibe in monotherapy are in patients who do not
tolerate statins and in some patients with sitosterolemia.
PCSK9 Inhibitors Circulating PCSK9 targets the LDL receptor for
lysosomal degradation, thus reducing its recycling and abundance
at the surface of the hepatocyte. Genetic loss of function of PCSK9
results in low levels of LDL-C and protection from CAD. Antibodies to PCSK9 (Table 407-4) sequester it and functionally increase
the number of LDL receptors available to remove LDL from the
blood. They are highly effective in lowering LDL-C, with an ~60%
reduction in LDL-C. They also reduce plasma levels of Lp(a)
modestly. Both PCSK9 antibodies have been shown to significantly
reduce cardiovascular events when added to a statin in patients with
existing CAD. These antibodies are administered subcutaneously
every 2 weeks. They are generally well tolerated, with the major side
effect being injection site reactions. They are generally indicated
as second-line (added to statin) or third-line (added to statin plus
ezetimibe) therapy in patients with FH or ASCVD in whom LDL-C
is not reduced to acceptable levels with a statin (with or without
ezetimibe) alone. An alternative approach to silencing PCSK9,
inclisiran, is a therapeutic siRNA molecule that targets the PCSK9
mRNA in the liver. In contrast to the antibodies, it is administered
subcutaneously every 6 months. It is effective in reducing LDL-C
by ~60% and appears to be well tolerated and safe; a cardiovascular
outcomes trial is ongoing.
ATP Citrate Lyase Inhibitor Bempedoic acid is a first-in-class
competitive inhibitor of ATP citrate lyase (ACL), which acts on
mitochondrial-derived citrate to generate production of acetylCoA, which is subsequently used for cholesterol synthesis. Thus, it
reduces cholesterol synthesis through a different mechanism than
statins, ultimately upregulating the hepatic LDL receptor. In phase
3 trials, bempedoic acid 180 mg daily reduced LDL-C by ~18%
when added to a statin and by ~23% as monotherapy. A cardiovascular outcomes trial is ongoing. Bempedoic acid is a prodrug
that requires activation by very-long-chain acyl-CoA synthetase-1
(ASCVL1), which is not expressed in skeletal muscle, potentially
explaining why it has less association with myalgias than statins;
indeed, it has been shown to be relatively well tolerated in patients
with statin intolerance. It is available in a fixed-dose combination
with ezetimibe, which reduced LDL-C by ~36%, for patients who
are statin intolerant. It can be used in combination with statins but
should not be used with simvastatin in a dose >20 mg. Bempedoic
acid is associated with increased uric acid levels and incidence of
gout; it was also associated with increased incidence of tendon
rupture in phase 3 trials. Unlike statins, it was not associated with
increased incidence of diabetes.
Bile Acid Sequestrants (Resins) Bile acid sequestrants (BAS)
bind bile acids in the intestine and promote their excretion rather
than reabsorption in the ileum. To maintain the bile acid pool size,
the liver diverts cholesterol to bile acid synthesis. The decreased
hepatic intracellular cholesterol content results in upregulation of
the LDL receptor and enhanced LDL clearance from the plasma.
BAS, including cholestyramine, colestipol, and colesevelam (Table
407-4), primarily reduce plasma LDL-C levels but can cause an
increase in plasma TGs. Therefore, patients with HTG generally
should not be treated with bile acid–binding resins. Cholestyramine
and colestipol are insoluble resins that must be suspended in liquids.
Colesevelam is available as tablets but generally requires up to six to
seven tablets per day for effective LDL-C lowering. BAS are effective
3150 PART 12 Endocrinology and Metabolism
in combination with statins and in combination with ezetimibe.
Side effects of resins are limited to the gastrointestinal tract and
include bloating and constipation. Because BAS are not systemically
absorbed, they are very safe and are the cholesterol-lowering drug
of choice in children and in women who are pregnant, lactating, or
actively trying to conceive. However, they are otherwise fourth- or
fifth-line drugs for LDL-C reduction in other settings.
Specialized Drugs for HoFH Three “orphan” drugs are approved
specifically for the management of HoFH, a rare condition caused
by biallelic mutations in the major genes causing FH in which
patients respond poorly to traditional LDL-lowering medications.
Lomitapide is a small-molecule inhibitor of MTP that reduces
LDL-C by ~50%, and mipomersen is an antisense oligonucleotide
against apoB tgat reduces LDL-C by ~25%. Both of these drugs
reduce hepatic VLDL production and thus LDL-C levels; however,
due to their mechanism of action, each drug causes an increase in
hepatic fat, the long-term consequences of which are unknown.
In addition, lomitapide is associated with gastrointestinal-related
side effects, and mipomersen is associated with skin reactions and
flulike symptoms. Finally, an antibody inhibitor of ANGPTL3,
evinacumab, was approved in 2021 for the treatment of HoFH. In a
phase 3 trial, an intravenous infusion every 4 weeks reduced LDL-C
levels in patients with HoFH by ~50% and was well tolerated. One
of these three drugs should be considered in HoFH patients after
a trial of a high-intensity statin, and possibly a PCSK9 inhibitor, is
shown to be insufficient to reduce LDL-C levels.
LDL Apheresis Patients with severe hypercholesterolemia who
cannot reduce their LDL-C to acceptable levels despite optimally
tolerated combination drug therapy are candidates for LDL apheresis. In this process, the patient’s plasma is passed over a column
that selectively removes the LDL, and the LDL-depleted plasma is
returned to the patient. LDL apheresis is indicated for patients on
maximally tolerated combination drug therapy (including a PCSK9
inhibitor) who have CHD and a plasma LDL-C level >200 mg/dL
or no CHD and a plasma LDL-C level >300 mg/dL; LDL apheresis
could be considered in high-risk patients who have an LDL-C >160
mg/dL on maximal therapy.
■ FURTHER READING
Baass A et al: Familial chylomicronemia syndrome: An under-recognized
cause of severe hypertriglyceridaemia. J Intern Med 287:340, 2020.
Brown EE et al: Genetic testing in dyslipidemia: A scientific statement
from the National Lipid Association. J Clin Lipidol 14:398, 2020.
Feingold KR: Approach to the patient with dyslipidemia. In Endotext.
Available at https://www.ncbi.nlm.nih.gov/books/NBK326736/. Last
updated May 11, 2020.
Hussain I et al: Lipodystrophies, dyslipidaemias and atherosclerotic
cardiovascular disease. Pathology 51:202, 2019.
Li F, Zhang H: Lysosomal acid lipase in lipid metabolism and beyond.
Arterioscler Thromb Vasc Biol 39:850, 2019.
Luirink IK et al: 20-Year follow-up of statins in children with familial
hypercholesterolemia. N Engl J Med 381:1547, 2019.
Schmidt AF et al: PCSK9 monoclonal antibodies for the primary and
secondary prevention of cardiovascular disease. Cochrane Database
Syst Rev 10:CD011748, 2020.
Sniderman AD et al: Apolipoprotein B particles and cardiovascular
disease: A narrative review. JAMA Cardiol 4:1287, 2019.
Sturm AC et al: Clinical genetic testing for familial hypercholesterolemia: JACC scientific expert panel. J Am Coll Cardiol 72:662, 2019.
Trinder M et al: Association of monogenic vs polygenic hypercholesterolemia with risk of atherosclerotic cardiovascular disease. JAMA
Cardiol 5:390, 2020.
The metabolic syndrome (syndrome X, insulin resistance syndrome)
consists of a constellation of metabolic abnormalities that confer
increased risk of cardiovascular disease (CVD) and diabetes mellitus.
Evolution of the criteria for the metabolic syndrome since the original
definition by the World Health Organization in 1998 reflects growing
clinical evidence and analysis by a variety of consensus conferences
and professional organizations. The major features of the metabolic
syndrome include central obesity, hypertriglyceridemia, low levels of
high-density lipoprotein (HDL) cholesterol, hyperglycemia, and hypertension (Table 408-1).
■ GLOBAL HEALTH/EPIDEMIOLOGY
The most challenging feature of the metabolic syndrome to define is
waist circumference. Intraabdominal circumference (visceral adipose
tissue) is most strongly related to insulin resistance and risk of diabetes
and CVD, and for any given waist circumference, the distribution of
adipose tissue between subcutaneous (SC) and visceral depots varies
substantially. Thus, within and between populations, there is a lesser
versus greater risk at the same waist circumference. These differences
in populations reflect the range of waist circumferences considered to
confer risk in different geographic locations (Table 408-1).
The prevalence of the metabolic syndrome varies around the world,
in part reflecting the age and ethnicity of the populations studied and
the diagnostic criteria applied. In general, the prevalence of the metabolic syndrome increases with age. The prevalence of metabolic syndrome in the U.S. adult population meeting the criteria of the National
Cholesterol Education Program (NCEP) and Adult Treatment Panel
III (ATPIII) is ~35%. Greater global industrialization is associated
with rising rates of obesity and expected increase in the prevalence of
the metabolic syndrome, especially as the population ages. Moreover,
the rising prevalence and severity of obesity among children reflect
features of the metabolic syndrome in a younger population, now
estimated to be 12 and 30% among obese and overweight children,
respectively.
Using National Health and Nutrition Examination Survey
(NHANES) data from 2012–2016, the prevalence of metabolic syndrome in the United States was 34.7% and not different between
men (35.1%) and women (34.3%). The highest prevalence was in
“other” race/ethnicity (39.0%) followed by Hispanic (36.3%) and nonHispanic white (36.0%). Although increased from 32.5% in 2010–2012,
the increase was not significant; however, the prevalence did increase
significantly among those aged 20–39 years (from 16.2 to 21.3%) and
in women (from 31.7 to 36.6%), Asian participants (from 19.9 to
26.2%), and Hispanic participants (from 32.9 to 40.4%). As in previous
data, prevalence of metabolic syndrome increased with increasing age
among all subgroups, i.e., from 19.5% among those aged 20–39 years
to 48.6% among those aged ≥60 years.
The frequency distribution of the five components of the syndrome
for the U.S. population (NHANES III) is summarized in Fig. 408-1.
Increases in waist circumference predominate among women, whereas
increases in fasting plasma triglyceride levels (i.e., >150 mg/dL), reductions in HDL cholesterol levels, and hyperglycemia are more likely in
men.
■ RISK FACTORS
Overweight/Obesity The metabolic syndrome was first described
in the early twentieth century; however, the worldwide overweight/
obesity epidemic has recently been the force driving its increasing
recognition. Central adiposity is a key feature of the syndrome, and
the syndrome’s prevalence reflects the strong relationship between
waist circumference and increasing adiposity. However, despite the
importance of obesity, patients who are of normal weight may also be
408 The Metabolic Syndrome
Robert H. Eckel
3151The Metabolic Syndrome CHAPTER 408
insulin-resistant and may have the metabolic syndrome. This phenotype is particularly evident for populations in India, Southeast Asia,
and Central America.
Sedentary Lifestyle Physical inactivity and less cardiorespiratory
fitness are predictors of CVD events and the related risk of death.
Many components of the metabolic syndrome are associated with a
sedentary lifestyle, including increased adipose tissue (predominantly
central), reduced HDL cholesterol, and increased triglycerides, blood
pressure, and glucose in genetically susceptible persons. Compared
with individuals who watch television or videos or use the computer
<1 h daily, those who do so for >4 h daily have a twofold increased risk
of the metabolic syndrome.
Genetics No single gene explains the complex phenotype called the
metabolic syndrome. However, using genome-wide association and
candidate gene approaches, several genetic variants are associated with
the metabolic syndrome. Although many of the loci have unknown
function, many others relate to body weight and composition, insulin resistance, and unfavorable disturbances in lipid and lipoprotein
metabolism.
Aging The metabolic syndrome affects nearly 50% of the U.S.
population aged >60, and at >60 years of age, women are more often
affected. The age dependency of the syndrome’s prevalence is seen in
most populations around the world.
Diabetes Mellitus Diabetes mellitus can be included in both the
NCEP and the harmonizing definitions of the metabolic syndrome,
but the greatest value of the metabolic syndrome, and especially fasting
glucose, is predicting type 2 diabetes. The great majority (~75%) of
patients with type 2 diabetes or impaired glucose tolerance have the
metabolic syndrome. The presence of the metabolic syndrome in these
populations relates to a higher prevalence of CVD than in patients who
have type 2 diabetes or impaired glucose tolerance but do not have the
syndrome.
Cardiovascular Disease Individuals with the metabolic syndrome are twice as likely to die of CVD as those who do not, and their
risk of an acute myocardial infarction or stroke is threefold higher. The
approximate prevalence of the metabolic syndrome among patients
with coronary heart disease (CHD) is 60%, with a prevalence of ~35%
among patients with premature coronary artery disease (age ≤45) and a
particularly high prevalence among women. With appropriate cardiac
rehabilitation and changes in lifestyle (e.g., nutrition, physical activity,
weight reduction, and—in some cases—pharmacologic therapy), the
prevalence of the syndrome can be reduced.
Lipodystrophy Lipodystrophic disorders in general are associated with the metabolic syndrome. Moreover, it is quite common for
such patients to present with the metabolic syndrome. Both genetic
TABLE 408-1 NCEP:ATPIIIa
2001 and Harmonizing Definition Criteria for the Metabolic Syndrome
NCEP:ATPIII 2001 HARMONIZING DEFINITIONb
Three or more of the following:
• Central obesity: waist circumference >102 cm (males),
>88 cm (females)
• Hypertriglyceridemia: triglyceride level ≥150 mg/dL or
specific medication
• Low HDLc
cholesterol: <40 mg/dL and <50 mg/dL for
men and women, respectively, or specific medication
• Hypertension: blood pressure ≥130 mmHg systolic or
≥85 mmHg diastolic or specific medication
• Fasting plasma glucose level ≥100 mg/dL or specific
medication or previously diagnosed type 2 diabetes
Three of the following:
Waist circumference (cm)
Men Women Ethnicity
≥94 ≥80 Europid, sub-Saharan African, Eastern and Middle Eastern
≥90 ≥80 South Asian, Chinese, and ethnic South and Central American
≥85 ≥90 Japanese
• Fasting triglyceride level >150 mg/dL or specific medication
• HDL cholesterol level <40 mg/dL and <50 mg/dL for men and women, respectively, or specific medication
• Blood pressure >130 mm systolic or >85 mm diastolic or previous diagnosis or specific medication
• Fasting plasma glucose level ≥100 mg/dL (alternative indication: drug treatment of elevated glucose levels)
a
National Cholesterol Education Program and Adult Treatment Panel III. b
In this analysis, the following thresholds for waist circumference were used: white men,
≥94 cm; African-American men, ≥94 cm; Mexican-American men, ≥90 cm; white women, ≥80 cm; African-American women, ≥80 cm; Mexican-American women, ≥80 cm. For
participants whose designation was “other race—including multiracial,” thresholds that were once based on Europid cutoffs (≥94 cm for men and ≥80 cm for women) and
on South Asian cutoffs (≥90 cm for men and ≥80 cm for women) were used. For participants who were considered “other Hispanic,” the International Diabetes Federation
thresholds for ethnic South and Central Americans were used. c
High-density lipoprotein.
lipodystrophy (e.g., Berardinelli-Seip congenital lipodystrophy, Dunnigan familial partial lipodystrophy) and acquired lipodystrophy
(e.g., HIV-related lipodystrophy and in HIV patients receiving certain
antiretroviral therapies) may give rise to severe insulin resistance and
many of the components of the metabolic syndrome.
■ ETIOLOGY
Insulin Resistance The most accepted and unifying hypothesis
to describe the pathophysiology of the metabolic syndrome is insulin
resistance, caused systemically by an incompletely understood defect in
insulin action (Chap. 403). The onset of insulin resistance is heralded
by postprandial hyperinsulinemia, which is followed by fasting hyperinsulinemia and ultimately by hyperglycemia.
An early major contributor to the development of insulin resistance
is an overabundance of circulating fatty acids (Fig. 408-2). Plasma
albumin-bound free fatty acids are derived predominantly from
adipose-tissue triglyceride stores released by intracellular lipolytic
enzymes. The lipolysis of triglyceride-rich lipoproteins in tissues by
lipoprotein lipase also produces free fatty acids. Insulin mediates both
anti-lipolysis and the stimulation of lipoprotein lipase in adipose tissue.
Of note, the inhibition of lipolysis in adipose tissue is the most sensitive pathway of insulin action. Thus, when insulin resistance develops,
increased lipolysis produces more fatty acids, which further decrease
the anti-lipolytic effect of insulin. Excessive fatty acids enhance substrate availability and create insulin resistance by modifying downstream signaling. Fatty acids impair insulin-mediated glucose uptake
and accumulate as triglycerides in both skeletal and cardiac muscle,
whereas increased fatty acid flux increases glucose production and
triglyceride production and accumulation in the liver.
Leptin resistance also may be a pathophysiologic mechanism
to explain the metabolic syndrome. Physiologically, leptin reduces
appetite, promotes energy expenditure, and enhances insulin sensitivity when insulin resistance is associated with leptin deficiency. In
addition, leptin may regulate cardiac and vascular function through
a nitric oxide–dependent mechanism. However, when obesity develops, hyperleptinemia ensues, with evidence of leptin resistance in the
brain and other tissues resulting in inflammation, insulin resistance,
hyperlipidemia, and a plethora of cardiovascular disorders, such as
hypertension, atherosclerosis, CHD, and heart failure.
The oxidative stress hypothesis provides a unifying theory for
aging and the predisposition to the metabolic syndrome. In studies of
insulin-resistant individuals with obesity or type 2 diabetes, the offspring of patients with type 2 diabetes, and the elderly, a defect in
mitochondrial oxidative phosphorylation leads to the accumulation of
triglycerides and related lipid molecules in muscle, liver, and perhaps
other tissues, i.e., β-cells.
Recently, the gut microbiome has emerged as an important contributor to the development of obesity and related metabolic disorders,
3152 PART 12 Endocrinology and Metabolism
18–29 y
30–49 y
50–69 y
>70 y
80
70
60
% With metabolic syndrome
C
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
D
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
E
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
F
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
G
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
H
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
A
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
80
70
60
% With metabolic syndrome
B
50
40
30
20
10
0
1988–1994 1999–2006
NHANES time period
2007–2012
Non-Hispanic white women
Non-Hispanic black women
Mexican American women
Standard error
Non-Hispanic white men
Non-Hispanic black men
Mexican American men
Standard error
FIGURE 408-1 Prevalence of metabolic syndrome among US adults over time by race/ethnicity-sex and age group, National Health and Nutrition Examination Survey
(NHANES), 1988-2012. Metabolic syndrome was defined by using the criteria agreed to jointly by the International Diabetes Federation; the US National Heart, Lung, and
Blood Institute in the United States; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study
of Obesity SE, standard error. (Reproduced with permission from JX Moore, N Chaudhary, T Akinyemiju. Metabolic syndrome prevalence by race/ethnicity and sex in the
United States, National Health and Nutrition Examination Survey, 1988-2012. Prev Chronic Dis 14:E24, 2017.)
3153The Metabolic Syndrome CHAPTER 408
including inflammation and components of the metabolic syndrome.
Although the mechanisms remain uncertain, interaction among
genetic predisposition, diet, bile acid metabolism, and the intestinal
flora is important.
Increased Waist Circumference Waist circumference is an
important component of the most recent and frequently applied diagnostic criteria for the metabolic syndrome. However, measuring waist
circumference does not reliably distinguish increases in SC abdominal
adipose tissue from that in visceral fat; this distinction requires CT or
MRI. With increases in visceral adipose tissue, adipose tissue–derived
free fatty acids reach the liver more readily. In contrast, increases in
abdominal SC fat release lipolysis products into the systemic circulation and therefore have fewer direct effects on hepatic metabolism.
Relative increases in visceral versus SC adipose tissue with increasing waist circumference in Asians and Asian Indians may explain
the greater prevalence of the syndrome in those populations than in
African-American men, in whom SC fat predominates. It is also possible that visceral fat is a marker for—but not the source of—excess
postprandial free fatty acids in obesity.
Dyslipidemia (See also Chap. 407) In general, free fatty acid flux
to the liver results in increased production of apolipoprotein (apo) B–
containing, triglyceride-rich, very-low-density lipoproteins (VLDLs).
The effect of insulin on this process is complex, but hypertriglyceridemia is an excellent marker of the insulin-resistant condition. Not
only is hypertriglyceridemia a feature of the metabolic syndrome, but
patients with the metabolic syndrome have elevated levels of apoC-III
carried on VLDLs and other lipoproteins. This increase in apoC-III is
inhibitory to lipoprotein lipase, reduces triglyceride-rich lipoprotein
remnant removal further contributing to hypertriglyceridemia, and
confers more risk for atherosclerotic cardiovascular disease (ASCVD).
The other major lipoprotein disturbance in the metabolic syndrome
is a reduction in HDL cholesterol. This reduction is a consequence
of changes in HDL composition and metabolism. In the presence of
hypertriglyceridemia, a decrease in the cholesterol content of HDL is
a consequence of reduced cholesteryl ester content of the lipoprotein
core in combination with cholesteryl ester transfer protein–mediated
alterations in triglycerides that make the HDL particle small and
dense. This change in lipoprotein composition also results in increased
clearance of HDL from the circulation. These changes in HDL have a
relationship to insulin resistance that is probably indirect, occurring in
concert with the changes in triglyceride-rich lipoprotein metabolism.
In addition to HDLs, low-density lipoproteins (LDLs) have alterations in composition in the metabolic syndrome. With fasting serum
triglycerides at >2.0 mM (~180 mg/dL), there is usually a predominance of small dense LDLs, which are thought to be more atherogenic,
although their association with hypertriglyceridemia and low HDLs
make their independent contribution to ASCVD difficult to assess.
Individuals with hypertriglyceridemia often have increases in cholesterol content of both VLDL1 and VLDL2 subfractions and in LDL particle number. Both lipoprotein changes may contribute to atherogenic
risk in patients with the metabolic syndrome.
Glucose Intolerance (See also Chap. 403) Defects in insulin
action in the metabolic syndrome lead to impaired suppression of
glucose production by the liver (and kidney) and reduced glucose
uptake and metabolism in insulin-sensitive tissues—i.e., muscle and
adipose tissue. There is a strong relationship between impaired fasting
– – –
Hypertension
FFA
HDL cholesterol
Small dense LDL
C-III
C-II
B-100 and
VLDL
CRP
FFA
PAI-1 Adiponectin
FFA
Triglyceride
(intramuscular
droplet)
Glycogen
Glucose
TNF-α
IL-6
CO2
Fibrinogen
Prothrombotic
state
TG Insulin
Insulin
IL-6 SNS
FIGURE 408-2 Pathophysiology of the metabolic syndrome. Free fatty acids (FFAs) are released in abundance from an expanded adipose tissue mass. In the liver, FFAs
result in increased production of glucose and triglycerides and secretion of very-low-density lipoproteins (VLDLs). Associated lipid/lipoprotein abnormalities include
reductions in high-density lipoprotein (HDL) cholesterol and an increased low-density lipoprotein (LDL) particle number. FFAs also reduce insulin sensitivity in muscle by
inhibiting insulin-mediated glucose uptake. Associated defects include a reduction in glucose partitioning to glycogen and increased lipid accumulation in triglyceride
(TG). The increase in circulating glucose, and to some extent FFAs, increases pancreatic insulin secretion, resulting in hyperinsulinemia. Hyperinsulinemia may result in
enhanced sodium reabsorption and increased sympathetic nervous system (SNS) activity and contribute to hypertension, as might higher levels of circulating FFAs. The
proinflammatory state is superimposed and contributory to the insulin resistance produced by excessive FFAs. The enhanced secretion of interleukin 6 (IL-6) and tumor
necrosis factor α (TNF-α) produced by adipocytes and monocyte-derived macrophages results in more insulin resistance and lipolysis of adipose tissue triglyceride stores
to circulating FFAs. IL-6 and other cytokines also enhance hepatic glucose production, VLDL production by the liver, hypertension, and insulin resistance in muscle. Insulin
resistance also contributes to increased triglyceride accumulation in the liver (nonalcoholic fatty liver disease). Cytokines and FFAs also increase hepatic production of
fibrinogen and adipocyte production of plasminogen activator inhibitor 1 (PAI-1), resulting in a pro-thrombotic state. Higher levels of circulating cytokines stimulate hepatic
production of C-reactive protein (CRP). Reduced production of the anti-inflammatory and insulin-sensitizing cytokine adiponectin is also associated with the metabolic
syndrome. (Reproduced with permission from RH Eckel et al: The metabolic syndrome. Lancet 365:1415, 2005.)
3154 PART 12 Endocrinology and Metabolism
glucose or impaired glucose tolerance and insulin resistance in studies of humans, nonhuman primates, and rodents. To compensate for
defects in insulin action, insulin secretion and/or clearance increases
or decreases, respectively, so that euglycemia remains. Ultimately, this
compensatory mechanism fails because of defects in insulin secretion,
resulting in progression from impaired fasting glucose and/or impaired
glucose tolerance to type 2 diabetes mellitus.
Hypertension The relationship between insulin resistance and
hypertension is well established. Paradoxically, under normal physiologic conditions, insulin is a vasodilator with secondary effects on
sodium reabsorption in the kidney. However, in the setting of insulin
resistance, the vasodilatory effect of insulin is lost but the renal effect
on sodium reabsorption is preserved. Sodium reabsorption is increased
in Caucasians with the metabolic syndrome but not in Africans or
Asians. Insulin also increases the activity of the sympathetic nervous
system, an effect that is preserved in the setting of insulin resistance.
Insulin resistance is also associated with pathway-specific impairment
in phosphatidylinositol-3-kinase signaling. In the endothelium, this
impairment may cause an imbalance between the production of nitric
oxide and the secretion of endothelin 1, with a consequent decrease in
blood flow. In addition, increases in angiotensinogen gene expression
in adipose tissue of obese subjects results in increases in circulating
angiotensin II and vasoconstriction. Although these mechanisms are
provocative, the inadequate evaluation of insulin action by measurement of fasting insulin levels or by homeostasis model assessment
shows that insulin resistance contributes only partially to the increased
prevalence of hypertension in the metabolic syndrome.
Another possible mechanism underlying hypertension in the metabolic syndrome is the vasoactive role of perivascular adipose tissue.
Reactive oxygen species released by NADPH oxidase impair endothelial function and result in local vasoconstriction. Other paracrine
effects such as leptin or other proinflammatory cytokines released
from adipose tissue, such as tumor necrosis factor α (TNF-α) may also
be important.
Hyperuricemia is another consequence of insulin resistance in the
metabolic syndrome. There is growing evidence not only that uric acid
is associated with hypertension but also that reduction of uric acid
normalizes blood pressure in hyperuricemic adolescents with hypertension. The mechanism appears to be in part related to an adverse
effect of uric acid on nitric oxide synthase in the macula densa of the
kidney and stimulation of the renin-angiotensin-aldosterone system.
Proinflammatory Cytokines The increases in proinflammatory
cytokines—including interleukins 1, 6, and 18; resistin; TNF-α; and
the systemic biomarker C-reactive protein—reflect overproduction by
the expanded adipose tissue mass (Fig. 408-2). Adipose tissue–derived
macrophages may be the primary source of proinflammatory cytokines
locally and in the systemic circulation. It remains unclear, however,
how much of the insulin resistance is caused by the paracrine effects of
these cytokines and how much by the endocrine effects.
Adiponectin Adiponectin is an anti-inflammatory cytokine produced exclusively by adipocytes. Adiponectin enhances insulin sensitivity and inhibits many steps in the inflammatory process. In the liver,
adiponectin inhibits the expression of gluconeogenic enzymes and the
rate of glucose production. In muscle, adiponectin increases glucose
transport and enhances fatty acid oxidation, partially through the activation of AMP kinase. Reductions in adiponectin levels are common
in the metabolic syndrome. The relative contributions of adiponectin
deficiency and overabundance of the proinflammatory cytokines are
unclear.
■ CLINICAL FEATURES
Symptoms and Signs The metabolic syndrome typically is not
associated with symptoms. On physical examination, waist circumference and blood pressure are often elevated. The presence of either or
both signs should prompt the clinician to search for other biochemical
abnormalities that may be associated with the metabolic syndrome.
Much less frequently, lipoatrophy or acanthosis nigricans is present
on examination. Because these physical findings characteristically are
associated with severe insulin resistance, other components of the metabolic syndrome are much more common.
Associated Diseases • CARDIOVASCULAR DISEASE The relative risk for new-onset CVD in patients with the metabolic syndrome
who do not have diabetes averages 1.5- to 3-fold. However, in INTERHEART, a study of 26,903 subjects from 52 countries, the risk for acute
myocardial infarction in subjects with the metabolic syndrome (World
Health Organization or International Diabetes Federation definition)
is comparable to that conferred by some, but not all, of the component
risk factors. Diabetes mellitus (odds ratio [OR], 2.72) and hypertension
(OR, 2.60) are stronger than other risk factors. Although congestive
heart failure and the metabolic syndrome can occur together, typically
this consequence is secondary to metabolic syndrome–related ASCVD
or hypertension. Metabolic syndrome is also associated with increases
in the risk for stroke, peripheral vascular disease, and Alzheimer’s
disease. However, as for myocardial infarction, the risk beyond the
additive role of the components of the metabolic syndrome remains
debatable. In the Reasons for Geographic and Racial Differences in
Stroke (REGARDS) cohort, an observational study of black and white
adults ≥45 years old across the United States, there were 9741 participants, and 41% had the metabolic syndrome. After adjustment for multiple confounders, metabolic syndrome was associated with increases
in high-sensitivity C-reactive protein (hsCRP), and this relationship
was associated with a 1.34 relative risk for all-cause mortality, but <50%
of deaths were from CVD. The population-attributable risk was 9.5%
for the metabolic syndrome alone and 14.7% for both metabolic syndrome and increased hsCRP. The relationship of metabolic syndrome
and hsCRP to mortality was greater for whites than blacks.
TYPE 2 DIABETES Overall, the risk for type 2 diabetes among patients
with the metabolic syndrome is increased three- to fivefold. In the
Framingham Offspring Study’s 8-year follow-up of middle-aged participants, the population-attributable risk of the metabolic syndrome
for developing type 2 diabetes was 62% among men and 47% among
women, yet increases in fasting plasma glucose explained most, if not
all, of this increased risk.
Other Associated Conditions In addition to the features specifically used to define the metabolic syndrome, other metabolic
alterations are secondary to or accompany insulin resistance. Those
alterations include increases in apoB and apoC-III, uric acid, prothrombotic factors (fibrinogen, plasminogen activator inhibitor 1),
serum viscosity, asymmetric dimethylarginine, homocysteine, white
blood cell count, proinflammatory cytokines, C-reactive protein, urine
albumin/creatinine ratio, nonalcoholic fatty liver disease (NAFLD)
and/or nonalcoholic steatohepatitis (NASH), polycystic ovary syndrome, and obstructive sleep apnea.
NONALCOHOLIC FATTY LIVER DISEASE NAFLD has become the most
common liver disease, in part a consequence of the insulin resistance
of the metabolic syndrome. The mechanism relates to increases in
free fatty acid flux and reductions in intrahepatic fatty acid oxidation
with resultant increases in triglyceride biosynthesis and hepatocellular
accumulation, with variable inflammation and oxidative stress. The
more serious NASH, a consequence of NAFLD in some patients and
a precursor of cirrhosis and end-stage liver disease, includes a more
substantial proinflammatory contribution. NAFLD affects ~25% of
the global population and up to 45% of patients with the metabolic
syndrome; over half of these patients have NASH. As the prevalence of
overweight/obesity and the metabolic syndrome increases, NASH may
become one of the more common causes of end-stage liver disease and
hepatocellular carcinoma.
HYPERURICEMIA (See also Chap. 417) Hyperuricemia reflects defects
in insulin action on the renal tubular reabsorption of uric acid and may
contribute to hypertension through its effect on the endothelium. An
increase in asymmetric dimethylarginine, an endogenous inhibitor
of nitric oxide synthase, also relates to endothelial dysfunction. In
addition, increases in the urine albumin/creatinine ratio may relate to
altered endothelial pathophysiology in the insulin-resistant state.
3155The Metabolic Syndrome CHAPTER 408
POLYCYSTIC OVARY SYNDROME (See also Chap. 392) Polycystic
ovary syndrome is highly associated with insulin resistance (50–80%)
and the metabolic syndrome, with a prevalence of the syndrome
between 12 and 60% based on phenotypes D through A.
OBSTRUCTIVE SLEEP APNEA (See also Chap. 31) Obstructive sleep
apnea is commonly associated with obesity, hypertension, increased
circulating cytokines, impaired glucose tolerance, and insulin resistance. In fact, obstructive sleep apnea may predict metabolic syndrome,
even in the absence of excess adiposity. Moreover, when biomarkers of
insulin resistance are compared between patients with obstructive
sleep apnea and weight-matched controls, insulin resistance is found to
be more severe in those with apnea. Continuous positive airway pressure treatment improves insulin sensitivity in patients with obstructive
sleep apnea.
■ DIAGNOSIS
The diagnosis of the metabolic syndrome relies on fulfillment of the
criteria listed in Table 408-1, as assessed using tools at the bedside and in
the laboratory. The medical history should include evaluation of symptoms for obstructive sleep apnea in all patients and polycystic ovary
syndrome in premenopausal women. Family history will help determine
risk for CVD and diabetes mellitus. Blood pressure and waist circumference measurements provide information necessary for the diagnosis.
Laboratory Tests Measurement of fasting lipids and glucose is
needed in determining whether the metabolic syndrome is present.
The measurement of additional biomarkers associated with insulin
resistance can be individualized. Such tests might include those for
apoB, hsCRP, fibrinogen, uric acid, urinary albumin/creatinine ratio,
and liver function. A sleep study should be performed if symptoms
of obstructive sleep apnea are present. If polycystic ovary syndrome
is suspected based on clinical features and anovulation, testosterone,
luteinizing hormone, and follicle-stimulating hormone should be measured. NAFLD can be further assessed by the NAFLD fibrosis score
(FIB4) or elastography.
TREATMENT
The Metabolic Syndrome
LIFESTYLE (SEE ALSO CHAP. 402)
Obesity, particularly abdominal, is the driving force behind the
metabolic syndrome. Thus, weight reduction is the primary
approach to the disorder. With at least a 5% and more so with
10% weight reduction, improvement in insulin sensitivity results
in favorable modifications in many components of the metabolic
syndrome. In general, recommendations for weight loss include a
combination of caloric restriction, increased physical activity, and
behavior modification. Caloric restriction is the most important
component, whereas increases in physical activity are important
for maintenance of weight loss. Some but not all evidence suggests
that the addition of exercise to caloric restriction may promote
greater weight loss from the visceral depot. The tendency for weight
regain after successful weight reduction underscores the need for
long-lasting behavioral changes.
Diet Before prescribing a weight-loss diet, it is important to
emphasize that it has taken the patient a long time to develop an
expanded fat mass; thus, the correction need not occur quickly.
Given that ~3500 kcal = 1 lb of fat, an ~500-kcal restriction daily
equates to weight reduction of 1 lb per week. Diets restricted in carbohydrate typically provide a more rapid initial weight loss. However, after 1 year, the amount of weight reduction is minimally more
reduced or no different from that with caloric restriction alone.
Thus, adherence to the diet is more important than the chosen diet.
Moreover, there is concern about low-carbohydrate diets enriched
in saturated fat, particularly for patients at risk for ASCVD. Therefore, a high-quality dietary pattern—i.e., a diet enriched in fruits,
vegetables, whole grains, lean poultry, and fish—should be encouraged to maximize overall health benefit.
Physical Activity Before prescribing a physical activity program
to patients with the metabolic syndrome, it is important to ensure
that the increased activity does not incur risk. Some high-risk
patients should undergo formal cardiovascular evaluation before
initiating an exercise program. For an inactive participant, gradual
increases in physical activity should be encouraged to enhance
adherence and avoid injury. Although increases in physical activity
can lead to modest weight reduction, 60–90 min of moderate- to
high-intensity daily activity is required to achieve this goal. Even
if an overweight or obese adult is unable to undertake this level of
activity, a health benefit will follow from at least 30 min of moderate-intensity activity daily. The caloric value of 30 min of a variety of
activities can be found at https://www.health.harvard.edu/diet-andweight-loss/calories-burned-in-30-minutes-of-leisure-and-routineactivities. Of note, a variety of routine activities, such as gardening,
walking, and housecleaning, require moderate caloric expenditure.
Thus, physical activity should not be defined solely in terms of formal exercise such as jogging, swimming, or tennis.
Behavior Modification Behavioral treatment typically includes
recommendations for dietary restriction and more physical activity that predicts sufficient weight loss that benefits metabolic
health. The subsequent challenge is the duration of the program
because weight regain so often follows successful weight reduction.
Improved long-term outcomes often follow a variety of methods,
such as a personal or group counselor, the Internet, social media,
and telephone follow-up to maintain contact between providers
and patients.
Obesity (See also Chap. 402) In some patients with the metabolic syndrome, treatment options need to extend beyond lifestyle
intervention. Weight-loss drugs come in two major classes: appetite
suppressants and absorption inhibitors. Appetite suppressants
approved by the U.S. Food and Drug Administration (FDA)
include phentermine (for short-term use [3 months] only) as well
as the more recent additions phentermine/topiramate, naltrexone/
bupropion, high-dose (3.0 mg) liraglutide (rather than 1.8 mg, the
maximum for treatment of type 2 diabetes), semaglutide (2.4 mg)
which are approved without restrictions on the duration of therapy.
In clinical trials, the phentermine/topiramate extended-release
combination resulted in ~8% weight loss relative to placebo in 50%
of patients. Side effects include palpitations, headache, paresthesias, constipation, and insomnia. Naltrexone/bupropion extended
release reduces body weight by ≥10% in ~20% of patients; however,
the drug combination is contraindicated in patients with seizure
disorders or any condition that predisposes to seizures. Naltrexone/
bupropion also increases pulse and blood pressure and should not
be given to patients with uncontrolled hypertension. High-dose
liraglutide, a glucagon-like peptide 1 (GLP-1) receptor agonist,
results in ~6% weight loss relative to placebo with ~33% of patients
with >10% weight loss. Common side effects are limited to the
upper gastrointestinal tract, including nausea and, less frequently,
emesis. Semaglutide (2.4 mg weekly), recently FDA approved
has been shown to produce an average weight loss of 14.9% over
68 weeks.
Orlistat inhibits fat absorption by ~30% and is moderately effective compared with placebo (~4% more weight loss). Moreover,
orlistat reduced the incidence of type 2 diabetes, an effect that was
especially evident among patients with impaired glucose tolerance
at baseline. This drug is often difficult to take because of oily leakage per rectum. In general, for all weight-loss drugs, greater weight
reduction leads to greater improvement in metabolic syndrome
components, including the conversion from prediabetes to type 2
diabetes.
Metabolic or bariatric surgery is an important option for patients
with the metabolic syndrome who have a body mass index of
>40 kg/m2
or >35 kg/m2
with comorbidities. An evolving application for metabolic surgery includes patients with a body mass
index as low as 30 kg/m2
and type 2 diabetes. Gastric bypass or
vertical sleeve gastrectomy results in dramatic weight reduction
3156 PART 12 Endocrinology and Metabolism
and improvement in most features of the metabolic syndrome. A
survival benefit with gastric bypass has also been realized.
LDL CHOLESTEROL (SEE ALSO CHAP. 407)
The rationale for the development of criteria for the metabolic syndrome by NCEP was to go beyond LDL cholesterol in identifying
and reducing the risk of ASCVD. The working assumption by the
panel was that LDL cholesterol goals had already been achieved
and that increasing evidence supports a linear reduction in ASCVD
events because of progressive lowering of LDL cholesterol with
statins with subsequent benefit using additional LDL cholesterol–
lowering agents. The 2019 American College of Cardiology (ACC)/
American Health Association (AHA) Cholesterol Guidelines have
no specific recommendations for patients with the metabolic syndrome; however, they recommend that patients aged 20–75 years
with LDL cholesterol levels ≥190 mg/dL should use a high-intensity
statin (e.g., atorvastatin 40–80 mg or rosuvastatin 20–40 mg daily)
and those with type 2 diabetes aged 40–75 years should use a
moderate-intensity statin and, if or when risk estimate is high, a
high-intensity statin. For patients with the metabolic syndrome
but without diabetes, the 10-year ASCVD risk estimator should
be employed, and patients with a risk ≥7.5% and ≤20% or patients
aged 20–59 with elevated lifetime risk should have a discussion
with their provider about initiating statin therapy for primary prevention of ASCVD. A coronary calcium score may help in making
this decision.
Diets restricted in saturated fats (<6% of calories) and trans
fats (as few as possible) should be applied aggressively. Although
evidence is controversial, dietary cholesterol can also be restricted.
If LDL cholesterol remains elevated, pharmacologic intervention
is needed. Based on substantial evidence, treatment with statins,
which lower LDL cholesterol by 15–60%, is the first-choice medication intervention. Of note, for each doubling of the statin dose,
LDL cholesterol is further lowered by only ~6%. Hepatotoxicity
(more than a threefold increase in hepatic aminotransferases) is
rare, but myopathy occurs in ~10–20% of patients. The cholesterol
absorption inhibitor ezetimibe is well tolerated and should be the
second-choice medication intervention. Ezetimibe typically reduces
LDL cholesterol by 15–20%. Bempedoic acid alone or in combination with ezetimibe is another option, with up to a 35% lowering
of LDL cholesterol with the combination. Bempedoic acid can
increase plasma uric acid. Proprotein convertase subtilisin/kexin
type 9 (PCSK9) inhibitors are potent LDL cholesterol–lowering
drugs (~45–60%) but are not needed for most patients with the
metabolic syndrome. Of course, if these patients also have familial
hypercholesterolemia or insufficient LDL cholesterol lowering on
statins with or without ezetimibe, a PCSK9 inhibitor should be
considered. The bile acid sequestrants cholestyramine, colestipol,
and colesevelam may be more effective than ezetimibe alone, but
because they can increase triglyceride levels, they must be used
with caution in patients with the metabolic syndrome when fasting
triglycerides are >300 mg/dL. Side effects include gastrointestinal
symptoms (palatability, bloating, belching, constipation, anal irritation). Nicotinic acid has similar LDL cholesterol–lowering capabilities (<20%). Fibrates are best employed to lower LDL cholesterol
when triglycerides are not elevated. Fenofibrate may be more effective than gemfibrozil in this setting.
TRIGLYCERIDES (SEE ALSO CHAP. 407)
The 2019 ACC/AHA Cholesterol Guidelines stated that fasting
triglycerides >500 mg/dL should be treated to prevent more serious
hypertriglyceridemia and pancreatitis. Although a fasting triglyceride value of >150 mg/dL is a component of the metabolic syndrome,
post hoc analyses of multiple fibrate trials have suggested reduction in the primary ASCVD outcome in patients (with or without
concomitant statin therapy) with fasting triglycerides >200 mg/dL,
often in the setting of reduced levels of HDL cholesterol. It remains
uncertain whether triglycerides cause ASCVD or if levels are just
associated with increased ASCVD risk.
A fibrate (gemfibrozil or fenofibrate) is one drug class of choice
to lower fasting triglyceride levels, which are typically reduced by
30–45%. Concomitant administration with drugs metabolized by the
3A4 cytochrome P450 system (including some statins) increases the
risk of myopathy. In these cases, fenofibrate may be preferable to gemfibrozil. In the Veterans Affairs HDL Intervention Trial, gemfibrozil was
administered to men with known CHD and levels of HDL cholesterol
<40 mg/dL. A coronary disease event and mortality rate benefit was
experienced predominantly among men with hyperinsulinemia and/
or diabetes, many of whom were identified retrospectively as having
the metabolic syndrome. Of note, the degree of triglyceride lowering in
this trial or other fibrate trials did not predict benefit.
Other drugs that lower triglyceride levels include statins, nicotinic acid, and prescription omega-3 fatty acids. For this purpose,
an intermediate or high dose of the “more potent” statins (atorvastatin, rosuvastatin) is needed. The effect of nicotinic acid on
fasting triglycerides is dose related and ~20–35%, an effect that is
less pronounced than that of fibrates. In patients with the metabolic
syndrome and diabetes, nicotinic acid may increase fasting glucose
levels, and clinical trials with nicotinic acid plus a statin have failed
to reduce ASCVD events. Prescriptions of omega-3 fatty acid preparations that include high doses of eicosapentaenoic acid (EPA)
with or without docosahexaenoic acid (DHA) (~1.5–4.5 g/d) lower
fasting triglyceride levels by ~25–40%. The two omega-3 randomized controlled trials associated with ASCVD risk reduction, JELIS
and REDUCE-IT, used EPA only, whereas STRENGTH, which was
terminated prematurely because of futility, used EPA plus DHA.
Here, no drug interactions with fibrates or statins occur, and the
main side effect of their use is eructation with a fishy taste. Freezing
the nutraceutical can partially block this unpleasant side effect.
Importantly, lowering triglycerides with any of the pharmaceuticals
has not proven to be an independent predictor of CVD outcomes.
HDL CHOLESTEROL (SEE ALSO CHAP. 407)
Very few lipid-modifying compounds increase HDL cholesterol levels. Statins, fibrates, and bile acid sequestrants have modest effects
(5–10%), whereas ezetimibe and omega-3 fatty acids have no effect.
Nicotinic acid is the only currently available drug with predictable
HDL cholesterol–raising properties. The response is dose related,
and nicotinic acid can increase HDL cholesterol by up to 30%
above baseline. After several trials of nicotinic acid versus placebo
in statin-treated patients, there is no evidence that raising HDL
cholesterol with nicotinic acid beneficially affects ASCVD events in
patients with or without the metabolic syndrome.
BLOOD PRESSURE (SEE ALSO CHAP. 277)
The direct relationship between blood pressure and all-cause mortality rate has been well established in studies comparing patients
with hypertension (>140/90 mmHg), patients with prehypertension (>120/80 mmHg but <140/90 mmHg), and individuals with
normal blood pressure (<120/80 mmHg). In patients who have the
metabolic syndrome without diabetes, the best choice for the initial
antihypertensive medication is an angiotensin-converting enzyme
(ACE) inhibitor or an angiotensin II receptor blocker, as these two
classes of drugs are effective and well tolerated. Additional agents
include a diuretic, calcium channel blocker, beta blocker, and mineralocorticoid inhibitor, such as the recently FDA approved mineralocorticoid receptor antagonist finerenone. In all patients with
hypertension, a sodium-restricted dietary pattern enriched in fruits
and vegetables, whole grains, and low-fat dairy products should be
advocated. Home monitoring of blood pressure may assist in maintaining good blood pressure control.
IMPAIRED FASTING GLUCOSE (SEE ALSO CHAP. 403)
In patients with the metabolic syndrome and type 2 diabetes,
aggressive glycemic control may favorably modify fasting levels
of triglycerides and/or HDL cholesterol. In patients with impaired
fasting glucose who do not have diabetes, a lifestyle intervention
that includes weight reduction, dietary saturated fat restriction, and
increased physical activity has been shown to reduce the incidence
No comments:
Post a Comment
اكتب تعليق حول الموضوع