Abstract
Pharmacologic reduction of low-density lipoprotein (LDL) cholesterol using statin drugs is foundational therapy to reduce cardiovascular disease (CVD) risk. Here we consider the place of non-statin therapies that also reduce LDL cholesterol in prevention of CVD. Among conventional non-statins, placebo-controlled randomized clinical trials showed that bile acid sequestrants, niacin and fibrates given as monotherapy each reduce CVD end points. From trials in which patients’ LDL cholesterol was already well-controlled on a statin, adding ezetimibe incrementally reduced CVD end points, while adding a fibrate or niacin showed no incremental benefit. Among emerging non-statins, monoclonal antibodies against proprotein convertase subtilisin kexin type 9 (PCSK9) added to a statin and given for up to 78 weeks showed preliminary evidence of reductions in CVD outcomes. While these promising early findings contributed to the recent approval of these agents in Europe and the US, much larger and longer duration outcomes studies are ongoing for definitive proof of CVD benefits. Other non-statin agents recently approved in the US include lomitapide and mipomersen, which both act via distinctive LDL-receptor independent mechanisms to substantially reduce LDL cholesterol in homozygous familial hypercholesterolemia. We also address some unanswered questions, including measuring alternative biochemical variables to LDL cholesterol, evidence for treating children with monitoring of subclinical atherosclerosis, and potential risks of extremely low LDL cholesterol. As evidence for benefit in CVD prevention accumulates, we anticipate that clinical practice will shift towards more assertive LDL-lowering treatment, using both statins and non-statins initiated earlier in appropriately selected patients.
Introduction
Statins disrupt the atherosclerotic process and have made regression of atherosclerosis possible for many. Akira Endo’s painstaking pharmacological screening of compounds that interfered with cholesterol biosynthesis led to isolation of compactin in 1973 from the rice mold P. citrinum Pen-51 (1). Compactin’s descendants - lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin and pitavastatin - are used by tens of millions of patients to reduce death and disability from cardiovascular disease (CVD), following from irrefutable randomized clinical trial (RCT) evidence of their benefits (2). Statins’ benefits are inextricably linked to low-density lipoprotein (LDL) cholesterol lowering: for each 1.0 mmol/L (~40 mg/dL) reduction in LDL cholesterol, major vascular events and all-cause mortality are reduced by 22% and 10%, respectively, across all patient subgroups (2). Statins’ other putative biological effects include improving endothelial dysfunction; antioxidant, anticoagulant and anti-inflammatory effects; inhibiting cell proliferation; anti-carcinogenic actions; atherosclerotic plaque stabilization and inhibiting graft rejection after organ transplantation (3). Elevated LDL cholesterol per se influences these processes adversely; it remains controversial whether proposed “LDL independent” effects of statins could result from putative pleiotropic direct effects of statin molecules and their metabolites upon non-LDL-related pathways or simply from their LDL-lowering effects (3,4).
If LDL-lowering by statins is integral to their ability to reduce CVD events, then non-statin-based LDL cholesterol reductions should also be beneficial in the absence of unrelated detrimental effects (4). With the recent approval of inhibitors of proprotein convertase subtilisin kexin 9 (PCSK9), a review of all non-statin LDL-cholesterol lowering therapies was timely. Mechanistic understanding is important: if statins’ benefits are in part LDL-independent, then drug development programs should pursue these alternate pathways as well. This review will define the known benefits of lower LDL-cholesterol concentrations from favorable genetic endowment and various clinical interventions.
LDL as a causal factor in atherosclerosis
Epidemiology, pathology and molecular biology
It is beyond the scope here to recapitulate the non-RCT evidence supporting LDL’s causal role in atherosclerosis. Guideline writers have aggregated much of this evidence already (5). The centrality of LDL cholesterol in predicting CVD risk has been confirmed repeatedly in classic epidemiological (6–8) and observational (9) studies. Furthermore, thousands of pathology, and cell and molecular biology experiments implicate perturbed lipid metabolism, particularly quantitative and qualitative abnormalities of LDL, as drivers of dysfunctional immune and inflammatory responses in atherogenesis (10–12). Even as new pro- and anti-inflammatory pathways are being discovered, the foundational role of LDL in atherogenesis remains axiomatic (12).
Atherosclerosis development early in life
Atherosclerosis begins early in life, with higher levels of LDL cholesterol contributing to early atherogenesis (13). The Pathobiological Determinants of Atherosclerosis in Youth Study (PDAY) and the Bogalusa Heart Study showed that every 0.26–0.39 mmol/L (10–15 mg/dL) increase of non-HDL cholesterol is associated with an additional year of vascular aging (14,15). Thus, a 15 year old with heterozygous familial hypercholesterolemia (FH) has essentially the same amount of atherosclerosis as a 20–35 year old with an average lipid profile, depending on the presence of additional risk factors. Four longitudinal studies confirm that lipids measured in youth better predict subclinical atherosclerosis measured in middle age than do risk factors measured in middle age concurrently with arterial imaging (14, 16–18). This implies that when atherosclerosis prevention is started later in life, not only must risk factors be lowered, but also existing advanced disease must be regressed to be completely effective.
Importance of LDL cholesterol from human genetics
Two types of human genetic evidence support LDL’s role in atherosclerosis. First, in the rare single-gene disorder FH, lifelong elevations of plasma LDL cholesterol lead to early atherosclerosis (19). Second, Mendelian randomization studies of common DNA polymorphisms with modest effects on LDL cholesterol suggest a causative role in CVD. Within the Mendelian randomization framework, if a biomarker is causally associated with a disease, its genetic determinants are also associated with disease risk (20). Mendelian randomization avoids confounding and reverse causation (20), and assumes that the culprit genetic variants influence only the biomarker of interest. This is not always the case for plasma lipid traits, where multiple lipid effects are evident for several gene loci, including CETP, LPL and APOA5. However, genetic variants at the PCSK9, HMGCR, and NPC1L1 loci associate specifically with LDL cholesterol; these variants also predict coronary heart disease (CHD) risk.
For instance, Cohen et al. showed loss of function (LOF) mutations in PCSK9 were associated with reduced LDL cholesterol and substantial reductions in CHD risk (21). Similarly, the Myocardial Infarction Genetics Consortium Investigators re-sequenced the NPC1L1 gene and showed that the p.Arg406X LOF mutation was associated with 10% lower LDL cholesterol and 50% decreased CHD risk in a large replication sample (22). Ference et al. studied 108,376 subjects from 14 RCTs and reported that genetic variants at NPC1L1 and HMGCR loci were associated with reductions in LDL cholesterol of 0.06 and 0.07 mmol/L (2.4 and 2.9 mg/dL) and in lifetime CHD risk of 4.8 and 5.3%, respectively (23). Since PCSK9, NPC1L1 and HMGCR genes have minimal effects on other variables, these studies support the direct causal relationship between LDL cholesterol and CHD. Furthermore, reductions in CHD risk in these studies for this degree of LDL reduction are at least twice as large as would be predicted from short-term statin RCTs, presumably because genetic influences are present from birth.
Importance of LDL cholesterol from randomized clinical trials
Early RCTs of LDL lowering studied diet, ileal bypass and various non-statin drugs. Of the latter, estrogen and dextrothyroxine had undesirable physiological effects and failed to reduce CVD despite cholesterol reduction (24). Fibrates and niacin have shown some benefit when used as monotherapy (25,26), but no incremental protection when added to a statin in patients who had achieved low LDL cholesterol levels (27,28). All other LDL-reducing interventions have benefits, including reduced saturated fat and increased polyunsaturated fat intake (29), interrupting enterohepatic circulation by a bile acid sequestrant (30) or surgical intervention (31), blocking intestinal cholesterol absorption by ezetimibe (32), inhibiting cholesterol biosynthesis by statins (2) and removing LDL-cholesterol by repeated LDL-apheresis (33). Early statin RCTs such as the Scandinavian Simvastatin Survival Study (4S) (34) and West of Scotland Coronary Prevention Study (WOSCOPS) (35) used less potent statins, i.e. simvastatin and pravastatin, respectively, and achieved 30 – 36 % reductions in major coronary events. Stronger statins, such as rosuvastatin, lower risk by 45% (36). The Lipid Research Clinics (LRC) (30,37) and Cholesterol Treatment Trialists Collaboration (CTTC) each concurred that “lower is better”, whether achieved with a statin or non-statin mechanism (2,38).
Conventional non-statins and CVD risk
Ezetimibe
Ezetimibe blocks intestinal sterol absorption by interfering with Niemann Pick C1 like receptor 1 (NPC1L1), with LDL cholesterol reduction of ~20%. Convincing evidence of reduced CVD events with ezetimibe has been slow to emerge (39). In 2011, the Study of Heart and Renal Protection (SHARP) showed ezetimibe plus simvastatin vs double placebo reduced CVD events in patients with renal impairment (40). In 2015, The Improved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE-IT) showed that, compared with simvastatin alone, ezetimibe plus simvastatin significantly reduced major cardiovascular events by ~7% when started within 10 days of an acute coronary syndrome, which was commensurate with its incremental LDL cholesterol-lowering effect of ~0.4 mmol/L (16 mg/dL) (32). A subgroup analysis showed proportionally greater benefit in diabetic patients (32). A study employing intravascular ultrasound showed that ezetimibe plus atorvastatin induced significantly greater regression of coronary artery plaque volume than atorvastatin alone (41). Collectively, these results support LDL cholesterol reduction by ezetimibe as having beneficial effects on atherosclerotic CVD.
Bile acid sequestrants
Bile acid sequestrants bind bile acids in the intestinal lumen, diverting them from the enterohepatic circulation (42), depleting the liver of bile, which upregulates bile synthesis from cholesterol by 7-alpha-hydroxylase. This depletes the intra-hepatic cholesterol pool, upregulating LDL receptor activity, which reduces LDL cholesterol levels. Increased 7-alpha-hydoxylase activity raises triglyceride (TG) (43). At daily doses of 24 g cholestyramine, 20 g colestipol or 4.5 g colesevelam, LDL cholesterol is reduced 18–25% (44). Bile acid sequestrants also augment the LDL-lowering effects of other drugs, notably statins (45). Colesevelam improves glycemic control modestly in patients with type 2 diabetes (46). Gastro-intestinal side effects and drug interactions (perhaps somewhat lower with colesevelam) limit the use of these agents (47). In the LRC Coronary Primary Prevention Trial (CPPT), bile acid sequestrants reduced CHD events in treated hypercholesterolemic subjects, with benefit proportional to the degree of LDL cholesterol lowering (30,37).
Niacin
Pharmacologic doses of niacin (nicotinic acid), through incompletely defined mechanisms, lower LDL cholesterol and TG by up to 25% and 50% respectively, and raise HDL cholesterol by up to 30% (48). At high doses, niacin also lowers Lp(a) by up to 30% (49). As monotherapy, niacin administered to hypercholesterolemic men reduced the risk of recurrent myocardial infarction in the Coronary Drug Project (50), and also reduced total mortality in a 15-year follow-up (26). Decreased atherosclerosis progression has also been observed in imaging studies of niacin’s effects (51).
More recently, the Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes (AIM-HIGH) study and Heart Protection Study 2–Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) showed no reductions in CVD events or mortality when niacin was added to statin therapy in patients with pre-existing CVD and well-controlled LDL-C (28,52). These results undermined the hypothesis that HDL directly protects from atherosclerosis (53), and when considered together with niacin’s side effects (skin flushing, hepatotoxicity, hyperuricemia and hyperglycemia), have stifled enthusiasm for its use (54), although it may still have utility in severe FH until newer agents become widely available.
Fibrates
Individuals with the metabolic syndrome, with or without type 2 diabetes, are at higher risk for CVD events and mortality (55,56). Lowering LDL cholesterol with statins in this group is associated with expected reductions in CVD risk, although absolute on-treatment CVD event rates are higher than in people free of metabolic syndrome or type 2 diabetes (55,57–59). The dyslipidemia characterized by high TG and reduced HDL cholesterol increases CVD risk (60), with both on-treatment TG and HDL cholesterol levels predicting CVD events in statin RCTs (61–63). Fibrates lower TG and raise HDL cholesterol via moderate agonism of peroxisome proliferator activated receptor-alpha; however results of CVD outcome trials with fibrate monotherapy have been mixed (64–67). Two studies (one each in primary and secondary prevention) were positive (25,64), but three (two in primary and one in secondary prevention) were negative (65–67). In monotherapy studies, post hoc subgroup analyses suggested a benefit of fibrates in subjects with high TG with or without low HDL cholesterol (68).
In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial (27) of 5518 individuals with type 2 diabetes, adding fenofibrate on top of stable background simvastatin therapy (LDL cholesterol 2.1 mmol/L [80 mg/dL]) reduced TG and increased HDL cholesterol by ~16% and ~2.4%, respectively, compared to placebo, but had no effect on CVD end points (risk reduction 8%, P=0.32). Subgroup analyses showed: 1) women did significantly worse than men; and 2) 17% of the cohort, with a pre-specified group with both upper tertile of TG (>2.3 mmol/L or 204 mg/dL) and lower tertile of HDL cholesterol (<0.9 mmol/L or 34 mg/dL), trended toward greater CVD event reduction (p=0.06). Adverse musculoskeletal events were not increased with fenofibrate plus simvastatin, while raised serum creatinine on fenofibrate returned to baseline levels with cessation of treatment (69). There is no evidence supporting the use of fenofibrate in non-dyslipidemic people and in women without dyslipidemia, fenofibrate may increase risk of CVD events. Subgroup analyses suggesting benefit in individuals with higher TG and HDL cholesterol (68) must be confirmed in a dedicated trial.
Non-statins in children
Limited RCT data support ezetimibe use in children and adolescents (70). As monotherapy, after 12 weeks of treatment, ezetimibe lowered LDL cholesterol by 27% in 6 – 10 year old children with FH. In adolescents, in combination with simvastatin, ezetimibe lowered LDL cholesterol by an additional 10–15% (71). While colesevelam modestly lowered LDL cholesterol (6–12%) irrespective of background statin use, children on statins at trial entry sometimes discontinued this treatment during the open label phase, and LDL cholesterol actually increased on colesevelam alone (72). This highlights the importance of compliance to achieve therapeutic goals, and also the need for additional RCT evidence in this group.
Newer non-statin agents
PCSK9 inhibitors
PCSK9 interrupts the recycling of the LDL receptor by diverting it towards lysosomal degradation after receptor-mediated endocytosis of LDL particles. Gain-of-function mutations in the PCSK9 gene cause autosomal dominant hypercholesterolemia (73), while loss-of-function mutations are associated with lower LDL cholesterol and reduced CVD risk (21,74). Monoclonal antibodies (mAbs) against PCSK9 were recently approved in Europe and the US (75). Alirocumab and evolocumab, both fully human mAbs, have completed the majority of their Phase 3 efficacy and safety trials, while these have not yet been completed for bococizumab, a humanized mAb (76). Large RCTs of CVD outcomes are under way for all three PCSK9 mAbs.
LDL cholesterol lowering efficacy varies from 50–65% for PCSK9 mAbs (Table 1). LDL-lowering efficacy is comparable for alirocumab 150 mg biweekly and evolocumab 140 mg biweekly and 420 mg every 4 weeks. On maximal statin therapy, mean LDL cholesterol levels of ~0.9 mmol/L (35 mg/dL) are achievable with PCSK9 mAbs, and many patients achieve LDL cholesterol <0.64 mmol/L (25 mg/dL) (77,78). Efficacy appears similar across most patient subgroups, including heterozygous FH patients (78,79). PCSK9 mAbs allowed a substantial proportion of FH patients to achieve LDL cholesterol <1.8 mmol/L (<70 mg/dL) for the first time (80,81). In patients with homozygous FH on maximal lipid-lowering therapy, evolocumab 420 mg every 4 weeks reduced LDL cholesterol by ~30%, with efficacy related to the degree of residual LDL receptor activity (82). In addition to robust LDL cholesterol lowering, evolocumab and alirocumab improve other lipid parameters, e.g. Lp(a) is reduced up to 25% (78,79).
Table 1.
class | examples | typical % LDL cholesterol reduction | evidence for clinical CVD end point reduction | key reference(s) | comments |
---|---|---|---|---|---|
statins | lovastatin, simvastatin, pravastatin, fluvastatin atorvastatin, rosuvastatin, pitavastatin | 35–55% | incontrovertible: 22% CVD risk reduction for 1 mmol/L (~40 mg/dL) LDL cholesterol reduction, plus ~10% reduced all-cause mortality | 2 | reference comparator for non-statin agents in this table |
cholesterol absorption inhibitor | ezetimibe | 18–25% | strong: modest incremental ~7% reduction when added to statin therapy in acute coronary syndrome patients | 32 | no studies with ezetimibe as monotherapy |
bile acid sequestrants | cholestyramine, colestipol, colesevelam | 18–25% | good evidence vs placebo: ~20% CVD risk reduction in events in primary prevention | 30,37 | no studies with resins added to statins |
niacin-based preparations | crystalline niacin (short acting), extended release niacin | 20–25% | good evidence vs placebo: ~20% CVD risk reduction in events, with reductions in mortality over longer term; no incremental CVD risk reduction when added to statin therapy in patients with well-treated LDL cholesterol | 49–51 | multiple effects on lipid profile; neutral outcomes when given to patients well-treated on statin |
fibrates | gemfibrozil, fenofibrate, bezafibrate | 5–15%, varies depending on baseline triglyceride levels | good evidence vs placebo: ~20% CVD risk reduction in events; no incremental CVD risk reduction when added to statin therapy in patients with well-treated LDL cholesterol | 64,66–68 | multiple effects on lipid profile; subgroup analyses suggest benefit in men with high TG, low HDL cholesterol |
PCSK9 inhibitors | evolocumab, alirocumab, bococizumab | 40–65% | promising evidence: ~50% reduction in CVD events when added to statin therapy in aggregated small early phase study results | 78,81 | possible new standard of therapy; long term outcomes data pending |
APOB antisense oligonucleotide | mipomersen | 30–45% | no evidence: good efficacy for LDL cholesterol reduction in homozygous familial hypercholesterolemia | 94 | also lowers Lp(a); injection site reactions, flu-like symptoms |
MTP inhibitor | lomitapide | 35–50% | no evidence: good efficacy for LDL cholesterol reduction in homozygous familial hypercholesterolemia | 97, 98 | GI symptoms, fatty liver |
CETP inhibitors | anacetrapib, evacetrapib | 20–30% | no evidence: awaiting large scale outcomes studies | 102, 103 | also raise HDL cholesterol by 80–100% |
ATP citrate lysase inhibitor | bempedoic acid | 20–28% | no evidence: early in development | 107 | no long term data; apparently well tolerated |
abbreviations: APOB, apolipoprotein B; ATP, adenosine triphosphate; CETP, cholesteryl ester transfer protein; CVD, cardiovascular disease; GI, gastrointestinal; HDL, high density lipoprotein; LDL, low density lipoprotein; MTP, microsomal triglyceride transfer protein; PCSK9, proprotein convertase subtilisin kexin 9; TG, triglyceride.
Evolocumab and alirocumab appear well tolerated in trials up to 78 weeks in duration (78, 79, 83). Injection site reactions are relatively infrequent and mild. Myalgia was slightly more frequent with alirocumab compared to placebo. A small, nonsignificant increase in neurocognitive events was reported for alirocumab and evolocumab, as well as a small nonsignificant increase in ophthalmologic events for alirocumab. No excess of adverse events has emerged in patients with LDL cholesterol <0.65 mmol/L (<25 mg/dL) or 0.39 mmol/L (<15 mg/dL) on ≥2 occasions over 78 weeks of treatment. Both evolocumab and alirocumab are well accepted by statin-intolerant patients, with muscle-related adverse event comparable to those seen with ezetimibe (84, 85).
Preliminary data suggest that PCSK9 mAbs reduce CVD events over 1 to 1.5 years (78,83). A meta-analysis of phase 2 and 3 trials found reduced total mortality with alirocumab and evolocumab, in trials ranging from 12 to 78 weeks (86). Four large, ongoing trials of CVD events and safety are evaluating alirocumab, evolocumab, and bococizumab in high risk patient populations (87–90). Patients with FH and high risk patients unable to tolerate high-intensity statin therapy, or unable to achieve >50% LDL cholesterol reduction on statin therapy, will likely benefit from PCSK9 mAbs (91). While long-term CVD outcomes data are pending, PCSK9 mAbs represent an option for patients who might benefit from an additional 50–60% reduction in LDL cholesterol, such as those with severe heterozygous FH and evidence of atherosclerosis or individuals with recurrent CVD events despite maximally tolerated doses of oral therapies. The availability of PCSK9 inhibitors might stimulate reconsideration of the concept of LDL cholesterol targets as part of lipid management guidelines.
Apolipoprotein B antisense therapy
Mipomersen is a second-generation antisense oligonucleotide that concentrates in the liver and specifically binds to human apolipoprotein (apo) B mRNA (92). The bound mRNA is degraded by hepatic RNAse, preventing the translation of apo B and synthesis of apo B-containing lipoproteins. Mipomersen 200 mg administered subcutaneously once weekly reduces LDL cholesterol, apo B and Lp(a) by ~30% (93). Since mipomersen does not require functional LDL receptors, equivalent responsiveness is seen in LDL receptor negative HoFH patients (94). Side effects include injection site reactions and influenza-like symptoms; discontinuation rates in RCTs have been high. Increases in transaminase levels and hepatic steatosis are also of concern. Longer term, larger studies are needed to evaluate its efficacy and potential hepatotoxicity. Mipomersen is approved in the US only for use in homozygous FH (95).
Lomitapide
Microsomal triglyceride transfer protein (MTP) is essential for assembly and secretion of hepatic and intestinal lipoproteins (96). MTP facilitates the incorporation of cholesteryl ester and triglyceride into VLDL in hepatocytes and chylomicrons in enterocytes by interacting with hepatic apo B-100 or intestinal apo B-48, respectively. Inhibiting MTP targets synthesis of apo B-containing lipoproteins (93) independent of the LDL receptor. Thus, lomitapide, an oral MTP inhibitor, reduces LDL cholesterol by up to 50% in subjects with homozygous FH (97,98). Lomitapide has FDA and European Medicines Agency (EMA) approval for the treatment of homozygous FH as an adjunct to diet and drug therapy, with concerns over long term effects, i.e. elevated liver enzymes and hepatic steatosis (99).
Cholesteryl ester transfer protein inhibition
Cholesteryl ester transfer protein (CETP) mediates neutral lipid transfer between lipoproteins, with net reduction in HDL cholesterol and cholesterol enrichment of apo B-containing lipoproteins. Thus CETP has received considerable attention as a drug target, although two CETP inhibitors have failed in large CVD outcome trial (100,101). Torcetrapib-treated individuals experienced increased CV events and mortality despite substantial reductions in LDL cholesterol (100), putatively due to off-target increases in blood pressure. An outcomes trial using dalcetrapib, an agent with modest HDL-raising effects and essentially no effect on LDL cholesterol, was stopped for futility (101). Two other compounds, anacetrapib and evacetrapib, remain under active investigation; both have substantial HDL-raising and LDL-lowering effects (102,103) without apparent off-target effects (104). Ongoing RCTs will determine whether additional beneficial effects of CETP inhibition translate into improved outcomes (105,106).
Bempedoic acid
Bempedoic acid (previously known as ETC-1002) is a small molecule inhibitor of ATP citrate lyase (ACL), a cytoplasmic enzyme that generates acetyl coenzyme A for de novo synthesis of fatty acids and cholesterol (107). Phase 2 trials in patients with hypercholesterolemia (108) and type 2 diabetes (109) show that bempedoic acid 80 and 120 mg daily reduced LDL cholesterol by 25% and 43%, respectively, over a very short term. Bempedoic acid may also result in incremental reductions in LDL cholesterol when co-administered with a statin or ezetimibe. Larger and longer phase 3 trials are needed to assess the durability and safety of this drug’s novel mechanism of action.
Remaining unanswered questions
Are there better alternatives to LDL cholesterol as measures of atherogenicity?
While the concept of LDL cholesterol as agent provocateur, chief epidemiological analyte and target for treatment is embedded in the cardiovascular field, LDL as a tangible clinical entity has its limitations (110). For instance, methods to directly measure LDL are either labor intensive or incompletely validated. LDL cholesterol in the real-world is often indirectly calculated from other lipid and lipoprotein fractions and requires a relatively long period of fasting. Furthermore, its measurement incompletely captures the total burden of atherogenic particles and accuracy of its determination is affected when LDL levels are very low or triglycerides are high (110). Thus, pursuit of alternatives to LDL cholesterol has been a focus of epidemiologic and mechanistic research.
Atherosclerosis was long recognized as more closely related to the total number of apo B–containing particles rather than LDL cholesterol concentration (111). One apo B molecule is present on the surface of chylomicrons (CMs), very low-density lipoprotein (VLDL), intermediate density lipoprotein (IDL), LDL and Lp(a) (112). Thus, apo B may more directly measure circulating atherogenic lipoproteins than LDL cholesterol (110, 113). Apo B has analytic and biological stability and is valid in non-fasting samples, which is useful for epidemiological studies (114).
Non-HDL cholesterol is the sum of VLDL-, IDL- and LDL cholesterol, and is calculated by subtracting HDL cholesterol from total cholesterol. Non-HDL cholesterol quantifies cholesterol content of all atherogenic apo B-containing lipoproteins and is highly correlated with apo B levels (112). The superiority of non-HDL cholesterol to LDL cholesterol in CHD prediction was shown in the Health Professionals Follow-up Study (115), the Framingham Heart Study (116) and Framingham Offspring Study (117), and the Women’s Health Study (118). Non-HDL cholesterol and apo B had equivalent predictive value and were both superior to LDL cholesterol in the Emerging Risk Factors Collaboration (119).
While RCTs have predominantly used LDL cholesterol reduction as a primary biochemical end point, many also measured non-HDL cholesterol and apo B, and found these to also be excellent markers of CHD risk reduction. For instance, the Treating to New Targets (TNT) and Incremental Decrease in End Points through Aggressive Lipid Lowering (IDEAL) showed that on-treatment apo B and non-HDL cholesterol were better predictors of reduced CVD events than levels of LDL cholesterol (120). In the Collaborative Atorvastatin Diabetes Study (CARDS) trial, both apo B and non-HDL cholesterol predicted CHD better than LDL cholesterol (121). Meta-analyses show apo B and non-HDL cholesterol as being superior to LDL cholesterol in predicting CHD events (122–124). Because non-HDL cholesterol and apo B can be determined from non-fasting samples and better predict atherogenicity than LDL cholesterol, several guidelines recommend including these measurements as adjuncts or alternatives to LDL cholesterol for risk assessment and monitoring treatment (13, 125, 126).
What is the role of LDL treatment in children?
Current recommended lipid lowering therapy in childhood is directed towards those with either FH (LDL cholesterol > 4.9 mmol/L [> 190 mg/dL] or > 4.1 mmol/L [>160 mg/dL] after diet management) or those with elevated LDL cholesterol in association with diabetes or multiple other known major risk factors (127). Because outcomes-linked RCTs initiated in youth are logistically challenging, treatment goals attempt to balance the value of LDL cholesterol lowering against risk of long term side effects. Statins are first choice treatments with a goal of achieving LDL cholesterol reduction of ≥50% or achieving a goal of LDL cholesterol ≤3.36 mmol/L (130 mg/dL). Treatment is generally initiated at ages 8 – 10 with FDA approved medications at doses utilized in pediatric RCTs conducted in FH patients. Trials of newer agents, including rosuvastatin and pitavastatin, may extend indications to lower ages. Few LDL cholesterol lowering RCTs in contexts other than FH have been performed in children; these have been of short duration in children with type 1 diabetes, lupus, or Kawasaki disease (128–130).
Carotid intima media thickness measures in children affected with heterozygous FH compared to their siblings suggests accelerated atherosclerosis can be appreciated early in the second decade (131); perhaps treatment below this age is not beneficial. Conversely, initiating statin treatment in the third to fourth decade may be insufficient to reverse advanced atherosclerosis that developed in adolescence or young adulthood (132). Optimal benefit may require initiation of lipid lowering treatment at the age at which plaque development is most likely to commence; regression of early atherosclerosis may restore vessels to optimum health. Conversely, a small subgroup of children will not achieve satisfactory LDL cholesterol lowering with statins, particularly those with homozygous or severe heterozygous FH (133). LDL apheresis is an adjunct to pharmacologic treatment in homozygous FH (33,133). FH homozygotes in particular require treatment at diagnosis and regardless of age because of risk of adverse events in childhood; LDL cholesterol reduction is greater with the addition of ezetimibe or colesevelam (71,72,134,135). Clinical trials utilizing newer agents discussed above are desperately needed to determine pediatric safety and efficacy in children with FH.
What is the role of monitoring subclinical atherosclerosis?
Subclinical atherosclerosis assessment has convincingly shown that populations with higher LDL cholesterol have more atherosclerosis and that presence of subclinical atherosclerosis, particularly coronary artery calcium, improves risk classification. Nonetheless, the role of monitoring subclinical markers in clinical practice is not established (136, 137), and a full discussion is beyond the scope of this paper. In FH or other conditions with elevated lifetime risk, noninvasive subclinical atherosclerosis assessment can be taken into account when determining baseline risk (138), and could be monitored serially as a surrogate for response to treatment, which in turn might affect treatment decisions. If changes in subclinical disease markers could be shown to predict outcomes independently of LDL cholesterol lowering, use of such tests could one day be justified in clinical practice. However, current use is limited by logistical issues such as cost and invasiveness (139,140), as well as lack of evidence.
Can LDL cholesterol be too low?
There are some signals of adverse effects of very low LDL cholesterol that should be monitored now that we have therapies capable of driving levels to such depths. For instance, fatty liver disease leading to hepatic fibrosis in children (141) and cirrhosis and hepatocellular carcinoma in adults (142–145) have been seen in heterozygous familial hypobetalipoproteinemia, where patients have lifetime LDL cholesterol <0.78 mmol/L (<30 mg/dL). Severe fatty liver has also been described in patients with ANGPTL3 mutations with similarly low LDL cholesterol levels (146). In the OSLER study, among those on evolocumab, three patients (1%) reported amnesia and five (1%) reported either memory or mental impairment (unrelated to achieved LDL cholesterol), whereas no cases were reported in controls (147). The FDA issued a directive for neurocognitive assessment in ongoing PCSK9 mAb RCTs (147). Also, subjects randomized to rosuvastatin who achieved LDL cholesterol <0.78 mmol/L (<30 mg/dL) had higher rates of diabetes, hematuria, hepatobiliary disorders and insomnia (148). Longer term follow-up is needed to better define the risk profile when LDL cholesterol is reduced to <0.78 mmol/L (<30 mg/dL).
Does diabetes risk with statins extend to non-statins?
Statins effectively reduce CVD in people with diabetes (2,59), who have a two-fold long-term increase in CVD morbidity and mortality. However, several studies suggest that statins increase risk of developing type 2 diabetes in pre-diabetic individuals (148–151). A meta-analysis of 13 statin trials (150) reported that standard-dose statin therapy was associated with 9% higher type 2 diabetes risk over four years, with greater risk associated with intensive statin therapy (151) and pre-existing risk factors for diabetes (152). No compelling evidence indicates differences in the risk of incident diabetes between statins. There is intense interest in identifying underlying mechanisms (153), with no definitive results to date. Recent genetic data from carriers of variants that reduce the activity of 3-hydroxy-3-methyl-glutaryl-CoA (HMGCoA) reductase (154) or cause FH (155), have raised the question as to whether statins per se, cholesterol synthesis, or LDL receptor function is actually causative. However, one CVD event is prevented for each 100–150 people treated with a statin while 500 people must be treated to cause one new case of type 2 diabetes, emphasizing risk-benefit considerations of statin therapy (150,151). So far, similar risks have not been observed with non-statins, except for niacin (28,52). Finally, the incidence of type 2 diabetes in IMPROVE-IT (32) and the ongoing trials of CETP inhibitors and PCSK9 inhibitors will provide important mechanistic clues.
Can newer non-statins regress lesions?
In the statin RCTs, greater relative CVD risk reductions occurred with progressively lower achieved LDL cholesterol levels; atherosclerosis regression continues as LDL cholesterol levels reach 0.39 mmol/L (15 mg/dL) (156,157). This suggests that combinations of maximally tolerated statins, ezetimibe, and new drugs such as PCSK9 or CETP inhibitors to dramatically reduce LDL cholesterol may have profound effects on atherosclerosis stabilization and regression. The opportunity exists to explore two new approaches to cardiovascular prevention. First, long-term follow-up of statin trials demonstrates persistently reduced CVD risk in statin-treated patients from the trial over the next decade or two (158,159). Long-term follow-up of the ongoing CVD outcomes trials of new agents will also help characterize the legacy effect of plaque stabilization and regression in high risk patients. It may be that very aggressive LDL cholesterol lowering for three to four years may stabilize plaque in most patients, and subsequent maintenance on maximal statin therapy could be adequate to suppress new plaque formation. Thus the cost of expensive new drugs could amortized over a longer time period
Animal data suggest dramatic LDL cholesterol reduction early in the course of atherosclerosis can completely regress atherosclerosis and normalize arterial function (160). Together, these data suggest that early, aggressive LDL cholesterol lowering can reset the vascular aging clock, and intermittent retreatment every decade or so might, in essence, “cure” atherosclerosis (132).
Targets or no targets?
Re-opening the debate on the role of target lipid levels in treatment guidelines is beyond the scope of this review. Employing an evidence base that relied on drugs and doses from RCTs, the 2013 AHA/ACC guidelines eliminated lipid targets, instead advising treatment decisions based on CVD risk (91). Other jurisdictions have retained targets for now, in part due to local values and preferences among community practitioners in favor of targets (125,126). Validity of targets versus no targets was evaluated in the offspring and third-generation cohorts of the FHS based on Framingham risk factors, LDL thresholds based on the updated ATP III guidelines (5) and the 2013 ACC/AHA pooled cohort calculator (91,161). Incident CVD was determined at a median 9.4 year follow-up (162). Statin-eligible participants by the 2013 ACC/AHA guidelines had increased hazard ratios for incident CVD compared with those eligible by ATP III guidelines: 6.8 (95%CI, 3.8–11.9) vs 3.1 (95%CI, 1.9–5.0), respectively (P<0.001). Thus, compared to LDL cholesterol thresholds in ATP III, the ACC/AHA guidelines seemed to more accurately identify increased risk of incident CVD and subclinical CHD, particularly in intermediate-risk subjects. However, the availability PCSK9 inhibitors and pending results of large CVD outcomes studies using those drugs will likely initiate re-evaluation of the concept of LDL cholesterol targets in clinical practice.
Conclusion
The biological, genetic, epidemiological and clinical trial evidence supporting a direct causal role for LDL cholesterol in atherogenesis and resulting major cardiovascular events is compelling. In the clinic, whether the physician or patient believes that statin benefits derive primarily from LDL cholesterol reduction or from other pleiotropic effects is practically irrelevant. Committing to treatment is based on evidence of CVD risk reduction from RCTs. Statins have the greatest body of RCT evidence supporting benefit in CVD risk reduction, but until recently, the relative importance of LDL cholesterol versus other pleiotropic effects of statins in driving these benefits has been disputed. The issue is important because non-statin therapies lower LDL cholesterol without statins’ other effects, and these agents may play an increasingly important role in CVD prevention. Recent RCT evidence, specifically significant CVD end point reductions seen with ezetimibe in IMPROVE-IT in patients with acute coronary syndromes and with both evolocumab and alirocumab over 52–78 weeks complement earlier RCT evidence of CVD event reduction with such non-statin therapies such as diet, intestinal bypass, and monotherapy with cholestyramine, niacin and fibrates. The common link between the CVD benefits of statins and the large number of non-statin agents is LDL cholesterol reduction, often via upregulation of the LDL receptor.
At some point, it becomes unwieldy for even the most passionate LDL skeptic to invoke individual non-LDL-related pleiotropic effects, given the wide range of different mechanisms of action of non-statins. Non-statins have a place for patients who are absolutely statin intolerant whose dyslipidemia requires management. In the near future, larger, longer term RCT results of PCSK9 mAbs may provide definitive support to this growing body of evidence. While diet and statins represent the cornerstones of management of dyslipidemia, our review suggests that non-statin treatments will play an increasingly important role. Rather than stoking the debate over LDL cholesterol targets, going forward we now have data from IMPROVE-IT on when to add ezetimibe to a statin. Moreover, algorithms for clinical action with PCSK9 inhibitors used either as monotherapy or added to statin drugs to reduce CVD will soon be clear. In the future, we anticipate that there will be an increasing focus on the optimal timing of initiating treatment, so that event rate reductions predicted from Mendelian randomization studies can be achieved for the general population.
Acknowledgments
We are grateful to Paolo Hunter for excellent help with manuscript preparation.
Sources of Funding
RAH is supported by the Jacob J. Wolfe Distinguished Medical Research Chair, the Martha G. Blackburn Chair in Cardiovascular Research, and operating grants from the Canadian Institutes for Health Research (MOP-13430, MOP-79523), the Heart and Stroke Foundation of Ontario (NA-6059, T-000353) and Genome Canada through Genome Quebec (award 4530). FKW is supported by NIH grants P50 HL083813, RO1 DE018184, RO1 HL089945 and RO1 HL117933.
Footnotes
Disclosures
RAH: Aegerion, Amgen, Lilly, Pfizer, Sanofi, Valeant
SSG: None
HNG: Amgen, Kowa, Merck, Sanofi, Sanofi Regereron
RM: Aegerion, Amgen, Lilly, Pfizer, Sanofi, Valeant
FJR: Amgen, Astra Zeneca, Pfizer, Sanofi Regeneron,
DJR: Aegerion, Alnylam, Sanofi
JR: Amarin, Amgen, Astra-Zeneca, Eli-Lilly, Esai, Glaxo-Smith-Kline, Merck, Pfizer, Sanofi Regeneron, Takeda
FKW: None
References
- 1.Endo A. A gift from nature: the birth of the statins. Nat Med. 2008;14:1050–2. doi: 10.1038/nm1008-1050. [DOI] [PubMed] [Google Scholar]
- 2.Cholesterol Treatment Trialists’ (CTT) Collaboration. Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet. 2010;376:1670–81. doi: 10.1016/S0140-6736(10)61350-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Davignon J, Laaksonen R. Low-density lipoprotein-independent effects of statins. Curr Opin Lipidol. 1999;10:543–59. doi: 10.1097/00041433-199912000-00010. [DOI] [PubMed] [Google Scholar]
- 4.Robinson JG, Smith B, Maheshwari N, Schrott H. Pleiotropic effects of statins: benefit beyond cholesterol reduction? A meta-regression analysis. J Am Coll Cardiol. 2005;46:1855–62. doi: 10.1016/j.jacc.2005.05.085. [DOI] [PubMed] [Google Scholar]
- 5.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. Circulation. 2002;106:3143. [PubMed] [Google Scholar]
- 6.Castelli WP, Anderson K. A population at risk. Prevalence of high cholesterol levels in hypertensive patients in the Framingham study. Am J Med. 1986;80:23–32. doi: 10.1016/0002-9343(86)90157-9. [DOI] [PubMed] [Google Scholar]
- 7.Wilson PWF, D’Agostino RB, Levy D, Belanger AM, Silbershatz H, Kannel WB. Prediction of coronary heart disease using risk factor categories. Circulation. 1998;97:1837–47. doi: 10.1161/01.cir.97.18.1837. [DOI] [PubMed] [Google Scholar]
- 8.Stamler J, Wentworth D, Neaton JD the MRFIT Research Group. Is relationship between serum cholesterol and risk of premature death from coronary heart disease continuous and graded? Findings in 356 222 primary screenees of the Multiple Risk Factor Intervention Trial (MRFIT) JAMA. 1986;256:2823–8. [PubMed] [Google Scholar]
- 9.Law MR, Wald NJ. An ecological study of serum cholesterol and ischaemic heart disease between 1950 and 1990. Eur J Clin Nutr. 1994;48:305–25. [PubMed] [Google Scholar]
- 10.Berliner JA, Navab M, Fogelman AM, Frank JS, Demer LL, Edwards PA, Watson AD, Lusis AJ. Atherosclerosis: basic mechanisms. Oxidation, inflammation, and genetics. Circulation. 1995;91:2488–96. doi: 10.1161/01.cir.91.9.2488. [DOI] [PubMed] [Google Scholar]
- 11.Lusis AJ. Atherosclerosis. Nature. 2000;407:233–41. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–22. doi: 10.1038/nm.2538. [DOI] [PubMed] [Google Scholar]
- 13.Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. 2011;128 (Suppl 5):S213–256. doi: 10.1542/peds.2009-2107C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gidding SS, McMahan CA, McGill HC, Colangelo LA, Schreiner PJ, Williams OD, Liu K. Prediction of coronary artery calcium in young adults using the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) risk score: The CARDIA study. Arch Intern Med. 2006;166:2341–2347. doi: 10.1001/archinte.166.21.2341. [DOI] [PubMed] [Google Scholar]
- 15.Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa heart study. N Engl J Med. 1998;338:1650–1656. doi: 10.1056/NEJM199806043382302. [DOI] [PubMed] [Google Scholar]
- 16.Mahoney LT, Burns TL, Stanford W, Thompson BH, Witt JD, Rost CA, Lauer RM. Coronary risk factors measured in childhood and young adult life are associated with coronary artery calcification in young adults: the Muscatine study. J Am Coll Cardiol. 1996;27:277–284. doi: 10.1016/0735-1097(95)00461-0. [DOI] [PubMed] [Google Scholar]
- 17.Raitakari OT, Juonala M, Kahonen M, Taittonen L, Laitinen T, Maki-Torkko N, Jarvisalo MJ, Uhari M, Jokinen E, Ronnemaa T, Akerblom HK, Viikari JS. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: the cardiovascular risk in young Finns study. JAMA. 2003;290:2277–2283. doi: 10.1001/jama.290.17.2277. [DOI] [PubMed] [Google Scholar]
- 18.Li S, Chen W, Srinivasan SR, Bond MG, Tang R, Urbina EM, Berenson GS. Childhood cardiovascular risk factors and carotid vascular changes in adulthood: the Bogalusa heart study. JAMA. 2003;290:2271–2276. doi: 10.1001/jama.290.17.2271. [DOI] [PubMed] [Google Scholar]
- 19.Nordestgaard BG, Chapman MJ, Humphries SE, et al. European Atherosclerosis Society Consensus Panel. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J. 2013;34:3478–90. doi: 10.1093/eurheartj/eht273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Evans DM, Davey SG. Mendelian randomization: new applications in the coming age of hypothesis-free causality. Annu Rev Genomics Hum Genet. 2015 Apr 22; doi: 10.1146/annurev-genom-090314-050016. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 21.Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354:1264–1272. doi: 10.1056/NEJMoa054013. [DOI] [PubMed] [Google Scholar]
- 22.Stitziel NO, Won HH, Morrison AC, et al. Inactivating mutations in NPC1L1 and protection from coronary heart disease. N Engl J Med. 2014;371:2072–2082. doi: 10.1056/NEJMoa1405386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ference BA, Majeed F, Penumetcha R, Flack JM, Brook RD. Effect of naturally random allocation to lower low-density lipoprotein cholesterol on the risk of coronary heart disease mediated by polymorphisms in NPC1L1, HMGCR, or both: a 2 × 2 factorial Mendelian randomization study. J Am Coll Cardiol. 2015;65:1552–61. doi: 10.1016/j.jacc.2015.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stamler J. The coronary drug project--findings with regard to estrogen, dextrothyroxine, clofibrate and niacin. Adv Exp Med Biol. 1977;82:52–75. doi: 10.1007/978-1-4613-4220-5_6. [DOI] [PubMed] [Google Scholar]
- 25.Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, Faas FH, Linares E, Schaefer EJ, Schectman G, Wilt TJ, Wittes J. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans affairs high-density lipoprotein cholesterol intervention trial study group. N Engl J Med. 1999;341:410–418. doi: 10.1056/NEJM199908053410604. [DOI] [PubMed] [Google Scholar]
- 26.Canner PL, Berge KG, Wenger NK, Stamler J, Friedman L, Prineas RJ, Friedewald W. Fifteen year mortality in coronary drug project patients: long-term benefit with niacin. J Am Coll Cardiol. 1986;8:1245–1255. doi: 10.1016/s0735-1097(86)80293-5. [DOI] [PubMed] [Google Scholar]
- 27.Ginsberg HN, Elam MB, Lovato LC, et al. ACCORD Study Group. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med. 2010;362:1563–1574. doi: 10.1056/NEJMoa1001282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.The HPS2-THRIVE Collaborative Group. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371:203–12. doi: 10.1056/NEJMoa1300955. [DOI] [PubMed] [Google Scholar]
- 29.Hooper L, Martin N, Abdelhamid A, Davey SG. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev. 2015;6:CD011737. doi: 10.1002/14651858.CD011737. [DOI] [PubMed] [Google Scholar]
- 30.Lipid Research Clinics Program. The Lipid Research clinics coronary primary prevention trial results, 1: reduction in the incidence of coronary artery disease. JAMA. 1984;251:351–64. doi: 10.1001/jama.1984.03340270029025. [DOI] [PubMed] [Google Scholar]
- 31.Buchwald H, Varco RL, Matts JP, et al. the POSCH Group. Effect of partial ileal bypass surgery on mortality and morbidity from coronary heart disease in patients with hypercholesterolemia. Report of the Program on the Surgical Control of the Hyperlipidemias (POSCH) N Engl J Med. 1990;323:946–955. doi: 10.1056/NEJM199010043231404. [DOI] [PubMed] [Google Scholar]
- 32.Cannon CP, Blazing MA, Giugliano RP, et al. IMPROVE-IT Investigators. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–2397. doi: 10.1056/NEJMoa1410489. [DOI] [PubMed] [Google Scholar]
- 33.Sachais BS, Katz J, Ross J, Rader DJ. Long-term effects of LDL apheresis in patients with severe hypercholesterolemia. J Clin Apher. 2005;20:252–5. doi: 10.1002/jca.20036. [DOI] [PubMed] [Google Scholar]
- 34.Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S) Lancet. 1994;344:1383–1389. [PubMed] [Google Scholar]
- 35.Shepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR, MacFarlane PW, McKillop JH, Packard CJ. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland coronary prevention study group. N Engl J Med. 1995;333:1301–1307. doi: 10.1056/NEJM199511163332001. [DOI] [PubMed] [Google Scholar]
- 36.Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ JUPITER Study Group. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195–2207. doi: 10.1056/NEJMoa0807646. [DOI] [PubMed] [Google Scholar]
- 37.Lipid Research Clinics Program. The Lipid Research Clinics coronary primary prevention trial results, II: the relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA. 1984;251:365–74. [PubMed] [Google Scholar]
- 38.Mihaylova B, Emberson J, Blackwell L, Keech A, Simes J, Barnes EH, Voysey M, Gray A, Collins R, Baigent C. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet. 2012;380:581–590. doi: 10.1016/S0140-6736(12)60367-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.McPherson R, Hegele RA. Ezetimibe: rescued by randomization (clinical and Mendelian) Arterioscler Thromb Vasc Biol. 2015;35:e13–5. doi: 10.1161/ATVBAHA.114.305012. [DOI] [PubMed] [Google Scholar]
- 40.Baigent C, Landray MJ, Reith C, et al. SHARP Investigators. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (study of heart and renal protection): a randomised placebo-controlled trial. Lancet. 2011;377:2181–92. doi: 10.1016/S0140-6736(11)60739-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tsujita K, Sugiyama S, Sumida H, et al. PRECISE–IVUS Investigators. Impact of dual lipid-lowering strategy with ezetimibe and atorvastatin on coronary plaque regression in patients with percutaneous coronary intervention. J Am Coll Cardiol. 2015;66:495–507. doi: 10.1016/j.jacc.2015.05.065. [DOI] [PubMed] [Google Scholar]
- 42.Grundy SM, Ahrens EH, Salen G. Interruption of the enterohepatic circulation of bile acids in man: comparative effects of cholestyramine and ileal exclusion on cholesterol metabolism. J Lab Clin Med. 1971;178:94–121. [PubMed] [Google Scholar]
- 43.Black DM. Gut-acting drugs for lowering cholesterol. Curr Atheroscler Rep. 2002;4:71–75. doi: 10.1007/s11883-002-0065-8. [DOI] [PubMed] [Google Scholar]
- 44.Knopp RH. Drug treatment of lipid disorders. N Engl J Med. 1999;341:498–511. doi: 10.1056/NEJM199908123410707. [DOI] [PubMed] [Google Scholar]
- 45.Knapp HH, Schrott H, Ma P, Knopp R, Chin B, Gaziano JM, Donovan JM, Burke SK, Davidson MH. Efficacy and safety of combination simvastatin and colesevelam in patients with primary hypercholesterolemia. Am J Med. 2001;110:352–60. doi: 10.1016/s0002-9343(01)00638-6. [DOI] [PubMed] [Google Scholar]
- 46.Fonseca VA, Handelsman Y, Staels B. Colesevelam lowers glucose and lipid levels in type 2 diabetes: the clinical evidence. Diabetes Obes Metab. 2010;12:384–92. doi: 10.1111/j.1463-1326.2009.01181.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.West RJ, Lloyd JK, Leonard JV. Long-term follow-up of children with familial hypercholesterolemia treated with cholestyramine. Lancet. 1980;2:873–5. doi: 10.1016/s0140-6736(80)92044-9. [DOI] [PubMed] [Google Scholar]
- 48.Creider JC, Hegele RA, Joy TR. Niacin: another look at an underutilized lipid-lowering medication. Nat Rev Endocrinol. 2012;8:517–28. doi: 10.1038/nrendo.2012.22. [DOI] [PubMed] [Google Scholar]
- 49.Carlson LA, Hamsten A, Asplund A. Pronounced lowering of serum levels of lipoprotein Lp(a) in hyperlipidaemic subjects treated with nicotinic acid. J Intern Med. 1989;226:271–6. doi: 10.1111/j.1365-2796.1989.tb01393.x. [DOI] [PubMed] [Google Scholar]
- 50.Coronary Drug Project Research Group. Clofibrate and niacin in coronary heart disease. JAMA. 1975;231:360–381. [PubMed] [Google Scholar]
- 51.Taylor AJ, Villines TC, Stanek EJ, Devine PJ, Griffen L, Miller M, Weissman NJ, Turco M. Extended-release niacin or ezetimibe and carotid intima-media thickness. N Engl J Med. 2009;361:2113–22. doi: 10.1056/NEJMoa0907569. [DOI] [PubMed] [Google Scholar]
- 52.The AIM-HIGH Investigators. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365:2255–67. doi: 10.1056/NEJMoa1107579. [DOI] [PubMed] [Google Scholar]
- 53.Lloyd-Jones DM. Niacin and HDL cholesterol –time to face facts. N Engl J Med. 2014;371:271–273. doi: 10.1056/NEJMe1406410. [DOI] [PubMed] [Google Scholar]
- 54.Guigliano RP. Niacin at 56 years of age –time for an early retirement? N Engl J Med. 2011;365:2318–2320. doi: 10.1056/NEJMe1112346. [DOI] [PubMed] [Google Scholar]
- 55.Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr. 2009;4:113–119. doi: 10.1111/j.1559-4572.2008.00044.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mente A, Yusuf S, Islam S, McQueen MJ, Tanomsup S, Onen CL, Rangarajan S, Gerstein HC, Anand SS INTERHEART Investigators. Metabolic syndrome and risk of acute myocardial infarction a case-control study of 26,903 subjects from 52 countries. J Am Coll Cardiol. 2010;55:2390–2398. doi: 10.1016/j.jacc.2009.12.053. [DOI] [PubMed] [Google Scholar]
- 57.Deedwania P, Barter P, Carmena R for the Treating to New Targets Investigators. Reduction of low-density lipoprotein cholesterol in patients with coronary heart disease and metabolic syndrome: Analysis of the Treating to New Targets Study. Lancet. 2006;368:919–928. doi: 10.1016/S0140-6736(06)69292-1. [DOI] [PubMed] [Google Scholar]
- 58.Colhoun HM, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Livingstone SJ, Thomason MJ, Mackness MI, Charlton-Menys V, Fuller JH 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;364:685–696. doi: 10.1016/S0140-6736(04)16895-5. [DOI] [PubMed] [Google Scholar]
- 59.Cholesterol Treatment Trialists’ (CTT) Collaborators. Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C. Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet. 2008;371:117–25. doi: 10.1016/S0140-6736(08)60104-X. [DOI] [PubMed] [Google Scholar]
- 60.Chapman MJ, Ginsberg HN, Amarenco P, et al. European Atherosclerosis Society Consensus Panel. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J. 2011;32:1345–61. doi: 10.1093/eurheartj/ehr112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sacks FM, Tonkin AM, Craven T, Pfeffer MA, Shepherd J, Keech A, Furberg CD, Braunwald E. Coronary heart disease in patients with low LDL-cholesterol: benefit of pravastatin in diabetic and enhanced role for HDL-cholesterol and triglycerides as risk factors. Circulation. 2002;105:1424–1428. doi: 10.1161/01.cir.0000012918.84068.43. [DOI] [PubMed] [Google Scholar]
- 62.Faergeman O, Holme I, Fayyad R, Bhatia S, Grundy SM, Kastelein JJ, LaRosa JC, Larsen ML, Lindahl C, Olsson AG, Tikkanen MJ, Waters DD, Pedersen TR Steering Committees of IDEAL and TNT Trials. Plasma triglycerides and cardiovascular events in the treating to new targets and incremental decrease in endpoints through aggressive lipid lowering trials of statins in patients with coronary artery disease. Am J Cardiol. 2009;104:459–463. doi: 10.1016/j.amjcard.2009.04.008. [DOI] [PubMed] [Google Scholar]
- 63.Barter P, Gotto AM, LaRosa JC, Maroni J, Szarek M, Grundy SM, Kastelein JJ, Bittner V, Fruchart JC Treating to New Targets Investigators, Treating to New Targets Investigators. HDL cholesterol, very low levels of LDL cholesterol, and cardiovascular events. N Engl J Med. 2007;357:1301–1310. doi: 10.1056/NEJMoa064278. [DOI] [PubMed] [Google Scholar]
- 64.Frick MH, Elo O, Haapa K, et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med. 1987;317:1237–45. doi: 10.1056/NEJM198711123172001. [DOI] [PubMed] [Google Scholar]
- 65.Primary prevention of ischaemic heart disease: WHO coordinated cooperative trial. A summary report. Bull World Health Organ. 1979;57:801–5. [PMC free article] [PubMed] [Google Scholar]
- 66.Bezafibrate Infarction Prevention (BIP) study. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation. 2000;102:21–7. doi: 10.1161/01.cir.102.1.21. [DOI] [PubMed] [Google Scholar]
- 67.Keech A, Simes RJ, Barter P, et al. FIELD Study Investigators. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005;366:1849–61. doi: 10.1016/S0140-6736(05)67667-2. [DOI] [PubMed] [Google Scholar]
- 68.Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet. 2010;375:1875–84. doi: 10.1016/S0140-6736(10)60656-3. [DOI] [PubMed] [Google Scholar]
- 69.Mychaleckyj JC, Craven T, Nayak U, Buse J, Crouse JR, Elam M, Kirchner K, Lorber D, Marcovina S, Sivitz W, Sperl-Hillen J, Bonds DE, Ginsberg H. Reversibility of fenofibrate therapy-induced renal function impairment in ACCORD type 2 diabetes participants. Diabetes Care. 2012;35:1008–14. doi: 10.2337/dc11-1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kusters DM, Caceres M, Coll M, Cuffie C, Gagne C, Jacobson MS, Kwiterovich PO, Lee R, Lowe RS, Massaad R, McCrindle BW, Musliner TA, Triscari J, Kastelein JJ. Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia. J Pediatr. 2015;166:1377–1384. e1373. doi: 10.1016/j.jpeds.2015.02.043. [DOI] [PubMed] [Google Scholar]
- 71.van der Graaf A, Cuffie-Jackson C, Vissers MN, Trip MD, Gagne C, Shi G, Veltri E, Avis HJ, Kastelein JJ. Efficacy and safety of coadministration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008;52:1421–1429. doi: 10.1016/j.jacc.2008.09.002. [DOI] [PubMed] [Google Scholar]
- 72.Stein EA, Marais AD, Szamosi T, Raal FJ, Schurr D, Urbina EM, Hopkins PN, Karki S, Xu J, Misir S, Melino M. Colesevelam hydrochloride: efficacy and safety in pediatric subjects with heterozygous familial hypercholesterolemia. J Pediatr. 2010;156:231–236. e231–233. doi: 10.1016/j.jpeds.2009.08.037. [DOI] [PubMed] [Google Scholar]
- 73.Abifadel M, Varret M, Rabès J, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34:154–6. doi: 10.1038/ng1161. [DOI] [PubMed] [Google Scholar]
- 74.Benn M, Nordestgaard BG, Grande P, Schnohr P, Tybjærg-Hansen A. PCSK9 R46L, Low-density lipoprotein cholesterol levels, and risk of ischemic heart disease: 3 independent studies and meta-analyses. J Am Coll Cardiol. 2010;55:2833–42. doi: 10.1016/j.jacc.2010.02.044. [DOI] [PubMed] [Google Scholar]
- 75.Urban D, Pöss J, Böhm M, Laufs U. Targeting the proprotein convertase subtilisin/kexin type 9 for the treatment of dyslipidemia and atherosclerosis. J Am Coll Cardiol. 2013;62:1401–8. doi: 10.1016/j.jacc.2013.07.056. [DOI] [PubMed] [Google Scholar]
- 76.Catapano AL, Papadopoulos N. The safety of therapeutic monoclonal antibodies: implications for cardiovascular disease and targeting the PCSK9 pathway. Atherosclerosis. 2013;228:18–28. doi: 10.1016/j.atherosclerosis.2013.01.044. [DOI] [PubMed] [Google Scholar]
- 77.Robinson JG, Nedergaard BS, Rogers WJ, Fialkow J, Neutel JM, Ramstad D, Somaratne R, Legg JC, Nelson P, Scott R, Wasserman SM, Weiss R LAPLACE-2 Investigators. Effect of evolocumab or ezetimibe added to moderate- or high-intensity statin therapy on ldl-c lowering in patients with hypercholesterolemia: The LAPLACE-2 randomized clinical trial. JAMA. 2014;311:1870–82. doi: 10.1001/jama.2014.4030. [DOI] [PubMed] [Google Scholar]
- 78.Robinson JG, Farnier M, Krempf M, et al. ODYSSEY LONG TERM Investigators. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med. 2015;372:1489–99. doi: 10.1056/NEJMoa1501031. [DOI] [PubMed] [Google Scholar]
- 79.Blom DJ, Hala T, Bolognese M, et al. DESCARTES Investigators. A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N Engl J Med. 2014;370:1809–19. doi: 10.1056/NEJMoa1316222. [DOI] [PubMed] [Google Scholar]
- 80.Raal FJ, Stein EA, Dufour R, et al. RUTHERFORD-2 Investigators. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD-2): a randomised, double-blind, placebo-controlled trial. The Lancet. 2015;385(9965):331–40. doi: 10.1016/S0140-6736(14)61399-4. [DOI] [PubMed] [Google Scholar]
- 81.Kastelein JJP, Ginsberg H, Langslet G, Hovingh, Ceska GKR, Dufour R, Blom D, Civeira F, Krempf M, Farnier M. Efficacy and safety of alirocumab in patients with heterozygous familial hypercholesterolemia (heFH) not adequately controlled with current lipid lowering therapy: Results of ODYSSEY FH I and FH II studies. American Heart Association Scientific Sessions; November 2014; Chicago, IL. 2014. [Google Scholar]
- 82.Raal FJ, Honarpour N, Blom DJ, Hovingh GK, Xu F, Scott R, Wasserman SM, Stein EA TESLA Investigators. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA Part B): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;385:341–50. doi: 10.1016/S0140-6736(14)61374-X. [DOI] [PubMed] [Google Scholar]
- 83.Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, Ballantyne CM, Somaratne R, Legg J, Wasserman SM, Scott R, Koren MJ, Stein EA Open-Label Study of Long-Term Evaluation against LDL Cholesterol (OSLER) Investigators. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med. 2015;372:1500–9. doi: 10.1056/NEJMoa1500858. [DOI] [PubMed] [Google Scholar]
- 84.Stroes E, Colquhoun D, Sullivan D, Civeira F, Rosenson RS, Watts GF, Bruckert E, Cho L, Dent R, Knusel B, Xue A, Scott R, Wasserman SM, Rocco M GAUSS-2 Investigators. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63:2541–8. doi: 10.1016/j.jacc.2014.03.019. [DOI] [PubMed] [Google Scholar]
- 85.Moriarty PM, Thompson PD, Cannon C, Guyton JR, Bergeron J, Zieve FJ, Bruckert E, Jacobson TA, Baccara-Dinet MT, Zhao J, Pordy R, Gipe D. ODYSSEY ALTERNATIVE: Efficacy and safety of the proprotein convertase subtilisin/kexin type 9 monoclonal antibody, alirocumab, versus ezetimibe, in patients with statin intolerance as defined by a placebo run-in and statin rechallenge arm [abstract] Circulation. 2014;130:2105–26. [Google Scholar]
- 86.Navarese EP, Kołodziejczak M, Schulze V, Gurbel PA, Tantry U, Lin Y, Brockmeyer M, Kandzari DE, Kubica JM, D’Agostino RB, Sr, Kubica J, Volpe M, Agewall S, Kereiakes DJ, Kelm M. Effects of proprotein convertase subtilisin/kexin type 9 antibodies in adults with hypercholesterolemia. Ann of Intern Med. 2015;163(1):40–51. doi: 10.7326/M14-2957. [DOI] [PubMed] [Google Scholar]
- 87.Amgen. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. Further cardiovascular outcomes research with PCSK9 inhibition in subjects with elevated risk (FOURIER) [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01764633 NLM Identifier: NCT01764633. [Google Scholar]
- 88.Sanofi/Regeneron. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. ODYSSEY outcomes: evaluation of cardiovascular outcomes after an acute coronary syndrome during treatment with alirocumab SAR236553 (REGN727) [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01663402 NLM Identifier: NCT01663402. [Google Scholar]
- 89.Pfizer. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. The evaluation of bococizumab (PF-04950615; RN316) in reducing the occurrence of major cardiovascular events in high risk subjects (SPIRE-2) [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01975389 NLM Identifier: NCT01975389. [Google Scholar]
- 90.Pfizer. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. The evaluation of bococizumab (PF-04950615;RN316) in reducing the occurrence of major cardiovascular events in high risk subjects (SPIRE-1) [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01975376 NLM Identifier: NCT01975376. [Google Scholar]
- 91.Stone NJ, Robinson JG, Lichtenstein AH, et al. American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2013 ACC/AHA Guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2889–934. doi: 10.1016/j.jacc.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 92.Bell DA, Hooper AJ, Watts GF, Burnett JR. Mipomersen and other therapies for the treatment of severe familial hypercholesterolemia. Vasc Health Risk Manag. 2012;8:651–9. doi: 10.2147/VHRM.S28581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Stein EA, Raal FJ. New therapies for reducing low-density lipoprotein cholesterol. Endocrinol Metab Clin N Am. 2014;43:1007–33. doi: 10.1016/j.ecl.2014.08.008. [DOI] [PubMed] [Google Scholar]
- 94.Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, Lachmann RH, Gaudet D, Tan JL, Chasan-Taber S, Tribble DL, Flaim JD, Crooke ST. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375:998–1006. doi: 10.1016/S0140-6736(10)60284-X. [DOI] [PubMed] [Google Scholar]
- 95.Rader DJ, Kastelein JJP. Lomitapide and mipomersen: two first-in-class drugs for reducing low-density lipoprotein cholesterol in patients with homozygous familial hypercholesterolemia. Circulation. 2014;129:1022–32. doi: 10.1161/CIRCULATIONAHA.113.001292. [DOI] [PubMed] [Google Scholar]
- 96.Wetterau JR, Lin MC, Jamil H. Microsomal triglyceride transfer protein. Biochim Biophys Acta. 1997;1345:136–50. doi: 10.1016/s0005-2760(96)00168-3. [DOI] [PubMed] [Google Scholar]
- 97.Cuchel M, Bloedon LT, Szapary PO, Kolansky DM, Wolfe ML, Sarkis A, Millar JS, Ikewaki K, Siegelman ES, Gregg RE, Rader DJ. Inhibition of microsomal triglyceride transfer protein in familial hypercholesterolemia. N Engl J Med. 2007;352(2):148–56. doi: 10.1056/NEJMoa061189. [DOI] [PubMed] [Google Scholar]
- 98.Cuchel M, Meagher EA, du Toit Theron H, et al. Phase 3 HoFH Lomitapide Study investigators. Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolemia: a single-arm, open-label, phase 3 study. Lancet. 2013;381:40–60. doi: 10.1016/S0140-6736(12)61731-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sacks FM, Stanesa M, Hegele RA. Severe hypertriglyceridemia with pancreatitis. Thirteen years’ treatment with lomitapide. JAMA Intern Med. 2014;174:443–7. doi: 10.1001/jamainternmed.2013.13309. [DOI] [PubMed] [Google Scholar]
- 100.Barter PJ, Caulfield M, Eriksson M, et al. ILLUMINATE Investigators. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357:2109–22. doi: 10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
- 101.Schwartz GG, Olsson AG, Abt M, et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367:2089–99. doi: 10.1056/NEJMoa1206797. [DOI] [PubMed] [Google Scholar]
- 102.Cannon CP, Shah S, Dansky HM, Davidson M, Brinton EA, Gotto AM, Stepanavage M, Liu SX, Gibbons P, Ashraf TB, Zafarino J, Mitchel Y, Barter P Determining the Efficacy and Tolerability Investigators. Safety of anacetrapib in patients with or at high risk for coronary heart disease. N Engl J Med. 2010;363:2406–15. doi: 10.1056/NEJMoa1009744. [DOI] [PubMed] [Google Scholar]
- 103.Nicholls SJ, Brewer HB, Kastelein JJ, Krueger KA, Wang MD, Shao M, Hu B, McErlean E, Nissen SE. Effects of the CETP inhibitor evacetrapib administered as monotherapy or in combination with statins on HDL and LDL cholesterol: a randomized controlled trial. JAMA. 2011;306:2099–109. doi: 10.1001/jama.2011.1649. [DOI] [PubMed] [Google Scholar]
- 104.Krishna R, Anderson MS, Bergman AJ, Jin B, Fallon M, Cote J, Rosko K, Chavez-Eng C, Lutz R, Bloomfield DM, et al. Effect of the cholesteryl ester transfer protein inhibitor, anacetrapib, on lipoproteins in patients with dyslipidaemia and on 24-h ambulatory blood pressure in healthy individuals: two double-blind, randomised placebo-controlled phase I studies. Lancet. 2007;370:1907–1914. doi: 10.1016/S0140-6736(07)61813-3. [DOI] [PubMed] [Google Scholar]
- 105.Eli Lilly and Company. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. A study of evacetrapib in high-risk vascular disease (ACCELERATE) [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01687998 NLM Identifier: NCT01687998. [Google Scholar]
- 106.University of Oxford, Merck Sharp & Dohme Corp. ClinicalTrials.gov [Internet] Bethesda (MD): National Library of Medicine (US); 2000. REVEAL: Randomized EValuation of the Effects of Anacetrapib through Lipid-modification. [cited 2015 Apr 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT01252953 NLM Identifier: NCT01252953. [Google Scholar]
- 107.Thompson PD, Rubino J, Janik MJ, MacDougall DE, McBride SJ, Margulies JR, Newton RS. Use of ETC-1002 to treat hypercholesterolemia in patients with statin intolerance. J Clin Lipidol. 2015;9:295–304. doi: 10.1016/j.jacl.2015.03.003. [DOI] [PubMed] [Google Scholar]
- 108.Ballantyne CM, Davidson MH, MacDougall DE, Bays HE, Dicarlo LA, Rosenberg NL, Margulies J, Newton RS. Efficacy and safety of a novel dual modulator of adenosine triphosphate-citrate lyase and adenosine monophosphate-activated protein kinase in patients with hypercholesterolemia: results of a multicenter, randomized, double-blind, placebo-controlled, parallel-group trial. J Am Coll Cardiol. 2013;62:1154–62. doi: 10.1016/j.jacc.2013.05.050. [DOI] [PubMed] [Google Scholar]
- 109.Gutierrez MJ, Rosenberg NL, MacDougall DE, Hanselman JC, Margulies JR, Strange P, Milad MA, McBride SJ, Newton RS. Efficacy and safety of ETC-1002, a novel investigational low-density lipoprotein-cholesterol-lowering therapy for the treatment of patients with hypercholesterolemia and type 2 diabetes mellitus. Arterioscler Thromb Vasc Biol. 2014;34:676–83. doi: 10.1161/ATVBAHA.113.302677. [DOI] [PubMed] [Google Scholar]
- 110.Sniderman AD, Lamarche B, Contois JH, de Graaf J. Discordance analysis and the Gordian Knot of LDL and non-HDL cholesterol versus apoB. Curr Opin Lipidol. 2014;25:461–7. doi: 10.1097/MOL.0000000000000127. [DOI] [PubMed] [Google Scholar]
- 111.Fredrickson DS, Levy RI, Lees RS. Fat transport in lipoproteins—an integrated approach to mechanisms and disorders. N Engl J Med. 1967;276:148–156. doi: 10.1056/NEJM196701192760305. [DOI] [PubMed] [Google Scholar]
- 112.Havel RJ, Kane JP. Introduction: structure and metabolism of plasma lipoproteins. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Bases of Inherited Disease. 7. New York, NY: McGraw-Hill; 1995. pp. 1841–51. [Google Scholar]
- 113.Barter PJ, Ballantyne CM, Carmena R, et al. Apo B versus cholesterol in estimating cardiovascular risk and in guiding therapy: report of the thirty-person/ten country panel. J Intern Med. 2006;259:247–258. doi: 10.1111/j.1365-2796.2006.01616.x. [DOI] [PubMed] [Google Scholar]
- 114.McQueen MJ, Hawken S, Wang X, Ounpuu S, Sniderman A, Probstfield J, Steyn K, Sanderson JE, Hasani M, Volkova E, Kazmi K, Yusuf S INTERHEART study investigators. Lipids, lipoproteins, and apolipoproteins as risk markers of myocardial infarction in 52 countries (the INTERHEART study): a case-control study. Lancet. 2008;372:224–33. doi: 10.1016/S0140-6736(08)61076-4. [DOI] [PubMed] [Google Scholar]
- 115.Pischon T, Girman CJ, Sacks FM, Rifai N, Stampfer MJ, Rimm EB. Non-high-density lipoprotein cholesterol and apolipoprotein B in the prediction of coronary heart disease in men. Circulation. 2005;112:3375–3383. doi: 10.1161/CIRCULATIONAHA.104.532499. [DOI] [PubMed] [Google Scholar]
- 116.Ingelsson E, Schaefer EJ, Contois JH, McNamara JR, Sullivan L, Keyes MJ, Pencina MJ, Schoonmaker C, Wilson PW, D’Agostino RB, Vasan RS. Clinical utility of different lipid measures for prediction of coronary heart disease in men and women. JAMA. 2007;298(7):776–785. doi: 10.1001/jama.298.7.776. [DOI] [PubMed] [Google Scholar]
- 117.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):1363–1368. doi: 10.1016/j.amjcard.2006.06.032. [DOI] [PubMed] [Google Scholar]
- 118.Ridker P, Rifai N, Cook N, Bradwin G, Buring JE. Non-HDL cholesterol, apolipoproteins A-I and B100, standard lipid measures, lipid ratios, and CRP as risk factors for cardiovascular disease in women. JAMA. 2005;294:326–33. doi: 10.1001/jama.294.3.326. [DOI] [PubMed] [Google Scholar]
- 119.Emerging Risk Factors Collaboration. Major lipids, apolipoproteins, and risk of vascular disease. JAMA. 2009;302:1993–2000. doi: 10.1001/jama.2009.1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kastelein JJ, van der Steeg WA, Holme I, et al. IDEAL Study Group. Lipids, apolipoproteins, and their ratios in relation to cardiovascular events with statin treatment. Circulation. 2008;117:3002–9. doi: 10.1161/CIRCULATIONAHA.107.713438. [DOI] [PubMed] [Google Scholar]
- 121.Charlton-Menys V, Betteridge DJ, Colhoun H, Fuller J, France M, Hitman GA, Livingstone SJ, Neil HA, Newman CB, Szarek M, DeMicco DA, Durrington PN. Targets of statin therapy: LDL cholesterol, non-HDL cholesterol, and apolipoprotein B in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS) Clin Chem. 2009;55:473–80. doi: 10.1373/clinchem.2008.111401. [DOI] [PubMed] [Google Scholar]
- 122.Sniderman AD, Williams K, Contois JH, Monroe HM, McQueen MJ, de Graff J, Furberg CD. A meta-analysis of low-density lipoprotein cholesterol, non-high-density lipoprotein cholesterol, and apolipoprotein B as markers of cardiovascular risk. Circ Cardiovasc Qual Outcomes. 2011;4:337–45. doi: 10.1161/CIRCOUTCOMES.110.959247. [DOI] [PubMed] [Google Scholar]
- 123.Boekholdt SM, Arsenault BJ, Mora S, et al. Association of LDL cholesterol, non-HDL cholesterol, and apolipoprotein B levels with risk of cardiovascular events among patients treated with statins: a meta-analysis. JAMA. 2012;307:1302–9. doi: 10.1001/jama.2012.366. [DOI] [PubMed] [Google Scholar]
- 124.Robinson JG, Wang S, Smith BJ, Jacobson TA. Meta-analysis of the relationship between non-high-density lipoprotein cholesterol reduction and coronary heart disease risk. J Am Coll Cardiol. 2009;53:316–22. doi: 10.1016/j.jacc.2008.10.024. [DOI] [PubMed] [Google Scholar]
- 125.European Association for Cardiovascular Prevention & Rehabilitation. Reiner Z, Catapano AL, et al., editors. ESC Committee for Practice Guidelines (CPG) 2008–2010 and 2010–2012 Committees. ESC/EAS Guidelines for the management of dyslipidaemias: the Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and the European Atherosclerosis Society (EAS) Eur Heart J. 2011;32:1769–818. doi: 10.1093/eurheartj/ehr158. [DOI] [PubMed] [Google Scholar]
- 126.Anderson TJ, Grégoire J, Hegele RA, et al. Update of the Canadian Cardiovascular Society guidelines for the diagnosis and treatment of dyslipidemia for the prevention of cardiovascular disease in the adult. Can J Cardiol. 2013;29:151–67. doi: 10.1016/j.cjca.2012.11.032. [DOI] [PubMed] [Google Scholar]
- 127.Wiegman A, Gidding SS, Watts GF, et al. European Atherosclerosis Society Consensus Panel. Familial hypercholesterolaemia in children and adolescents: gaining decades of life by optimizing detection and treatment. Eur Heart J. 2015 doi: 10.1093/eurheartj/ehv157. pii:ehv157 Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Canas JA, Ross JL, Taboada MV, Sikes KM, Damaso LC, Hossain J, Caulfield MP, Gidding SS, Mauras N. A randomized, double blind, placebo-controlled pilot trail of the safety and efficacy of atorvastatin in children with elevated low-density lipoprotein cholesterol (LDL-C) and type 1 diabetes. Pediatr Diabetes. 2015;2:79–89. doi: 10.1111/pedi.12245. [DOI] [PubMed] [Google Scholar]
- 129.Schanberg LE, Sandborg C, Barnhart HX, et al. Atherosclerosis Prevention in Pediatric Lupus Erythematosus Investigators. Use of atorvastatin in systemic lupus erythematosus in children and adolescents. Arthritis Rheum. 2012;64:285–96. doi: 10.1002/art.30645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Duan C, Du ZD, Wang Y, Jia LQ. Effect of pravastatin on endothelial dysfunction in children with medium to giant coronary aneurysms due to Kawasaki disease. World J Pediatric. 2014;10:232–7. doi: 10.1007/s12519-014-0498-5. [DOI] [PubMed] [Google Scholar]
- 131.McGill HC, Jr, McMahan CA, Gidding SS. Preventing heart disease in the 21st century: implications of the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) study. Circulation. 2008;117:1216–27. doi: 10.1161/CIRCULATIONAHA.107.717033. [DOI] [PubMed] [Google Scholar]
- 132.Robinson JG, Gidding SS. Curing atherosclerosis should be the next major cardiovascular prevention goal. J Am Coll Cardiol. 2014;63:2779–85. doi: 10.1016/j.jacc.2014.04.009. [DOI] [PubMed] [Google Scholar]
- 133.Cuchel M, Bruckert E, Ginsberg HN, et al. European Atherosclerosis Society Consensus Panel on Familial Hypercholesterolaemia. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the consensus panel on familial hypercholesterolaemia of the European Atherosclerosis Society. Eur Heart J. 2014;35:2146–57. doi: 10.1093/eurheartj/ehu274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Raal FJ, Pilcher GJ, Panz VR, van Deventer HE, Brice BC, Blom DJ, Marais AD. Reduction in mortality in subjects with homozygous familial hypercholesterolemia associated with advances in lipid-lowering therapy. Circulation. 2011;124:2202–7. doi: 10.1161/CIRCULATIONAHA.111.042523. [DOI] [PubMed] [Google Scholar]
- 135.Gagne C, Gaudet D, Bruckert E. Efficacy and safety of ezetimibe coadministered with atorvastatin or simvastatin in patients with homozygous familial hypercholesterolemia. Circulation. 2002;105:2469–75. doi: 10.1161/01.cir.0000018744.58460.62. [DOI] [PubMed] [Google Scholar]
- 136.Pletcher MJ, Sibley CT, Pignone M, Vittinghoff E, Greenland P. Interpretation of the coronary artery calcium score in combination with conventional cardiovascular risk factors: The Multi-Ethnic Study of Atherosclerosis (MESA) Circulation. 2013;128:1076–84. doi: 10.1161/CIRCULATIONAHA.113.002598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hlatky MA, Greenland P, Arnett DK, et al. American Heart Association Expert Panel on Subclinical Atherosclerotic Diseases and Emerging Risk Factors and the Stroke Council. Criteria for evaluation of novel markers of cardiovascular risk. A scientific statement from the American Heart Association. Circulation. 2009;119:2408–16. doi: 10.1161/CIRCULATIONAHA.109.192278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Miname MH, Ribeiro MS, 2nd, Parga Filho J, Avila LF, Bortolotto LA, Martinez LR, Rochitte CE, Santos RD. Evaluation of subclinical atherosclerosis by computed tomography coronary angiography and its association with risk factors in familial hypercholesterolemia. Atherosclerosis. 2010;213:486–491. doi: 10.1016/j.atherosclerosis.2010.10.001. [DOI] [PubMed] [Google Scholar]
- 139.Puri R, Nissen SE, Nicholls SJ. Statin-induced coronary artery disease regression rates differ in men and women. Curr Opin Lipidol. 2015;26:276–81. doi: 10.1097/MOL.0000000000000195. [DOI] [PubMed] [Google Scholar]
- 140.van Wijk DF, Sjouke B, Figueroa A, et al. Nonpharmacological lipoprotein apheresis reduces arterial inflammation in familial hypercholesterolemia. J Am Coll Cardiol. 2014;64:1418–26. doi: 10.1016/j.jacc.2014.01.088. [DOI] [PubMed] [Google Scholar]
- 141.Della Corte C, Fintini D, Giordano U, Cappa M, Brufani C, Majo F, Mennini C, Nobili V. Fatty liver and insulin resistance in children with hypobetalipoproteinemia: the importance of aetiology. Clin Endocrinol. 2013;79:49–54. doi: 10.1111/j.1365-2265.2012.04498.x. [DOI] [PubMed] [Google Scholar]
- 142.Tarugi P, Lonardo A. Heterozygous familial hypobetalipoproteinemia associated with fatty liver. Am J Gastroenterol. 1997;92:1400–1402. [PubMed] [Google Scholar]
- 143.Bonnefont-Rousselot D, Condat B, Sassolas A, Chebel S, Bittar R, Federspiel MC, Cazals-Hatem D, Bruckert E. Cryptogenic cirrhosis in a patient with familial hypocholesterolemia due to a new truncated form of apolipoprotein B. Eur J Gastroenterol Hepatol. 2009;21:104–108. doi: 10.1097/MEG.0b013e3282ffd9f8. [DOI] [PubMed] [Google Scholar]
- 144.Lonardo A, Tarugi P, Ballarini G, Bagni A. Familial heterozygous hypobetalipoproteinemia, extrahepatic primary malignancy, and hepatocellular carcinoma. Dig Dis Sc. 1998;43:2489–2492. doi: 10.1023/a:1026646618643. [DOI] [PubMed] [Google Scholar]
- 145.Cefalù AB, Pirruccello JP, Noto D, Gabriel S, Valenti V, Gupta N, Spina R, Tarugi P, Kathiresan S, Averna MR. A novel apoB mutation identified by exome sequencing cosegregates with steatosis, liver cancer and hypocholesterolemia. Arterioscler Thromb Vasc Biol. 2013;33:2021–5. doi: 10.1161/ATVBAHA.112.301101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Noto D, Cefalù AB, Valenti V, Fayer F, Pinotti E, Ditta M, Spina R, Vigna G, Yue P, Kathiresan S, Tarugi P, Averna MR. Prevalence of ANGPTL3 and APOB gene mutations in subjects with combined hypolipidemia. Arterioscler Thromb Vasc Biol. 2012;32:805–809. doi: 10.1161/ATVBAHA.111.238766. [DOI] [PubMed] [Google Scholar]
- 147.Swiger KJ, Martin SS. PCSK9 inhibitors and neurocognitive adverse events: exploring the FDA directive and a proposal for N-of-1 Trials. Drug Saf. 2015;38:519–526. doi: 10.1007/s40264-015-0296-6. [DOI] [PubMed] [Google Scholar]
- 148.Everett BM, Mora S, Glynn RJ, MacFadyen J, Ridker PM. Safety profile of subjects treated to very low low-density lipoprotein cholesterol levels (<30 mg/dL) with rosuvastatin 20 mg daily (from JUPITER) Am J Cardiol. 2014;114:1682–1689. doi: 10.1016/j.amjcard.2014.08.041. [DOI] [PubMed] [Google Scholar]
- 149.Ridker PM, Pradhan A, MacFadyen JG, Libby P, Glynn RJ. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: an analysis from the JUPITER trial. Lancet. 2012;380:565–571. doi: 10.1016/S0140-6736(12)61190-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Sattar N, Preiss D, Murray HM, et al. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet. 2010;375:735–42. doi: 10.1016/S0140-6736(09)61965-6. [DOI] [PubMed] [Google Scholar]
- 151.Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA. 2011;305:2556–64. doi: 10.1001/jama.2011.860. [DOI] [PubMed] [Google Scholar]
- 152.Waters DD, Ho JE, DeMicco DA, Breazna A, Arsenault BJ, Wun CC, Kastelein JJ, Colhoun H, Barter P. Predictors of new-onset diabetes in patients treated with atorvastatin: results from 3 large randomized clinical trials. J Am Coll Cardiol. 2011;57:1535–45. doi: 10.1016/j.jacc.2010.10.047. [DOI] [PubMed] [Google Scholar]
- 153.Brault M, Ray J, Gomez Y-H, Mantzoros CS, Daskalopoulou SS. Statin treatment and new-onset diabetes: A review of proposed mechanisms. Metabolism. 2014;63:735–745. doi: 10.1016/j.metabol.2014.02.014. [DOI] [PubMed] [Google Scholar]
- 154.Swerdlow DI, Preiss D, Kuchenbaecker KB, et al. HMG-coenzyme A reductase inhibition, type 2 diabetes, and bodyweight: evidence from genetic analysis and randomised trials. Lancet. 2015;385:351–61. doi: 10.1016/S0140-6736(14)61183-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Besseling J, Kastelein JJ, Defesche JC, Hutten BA, Hovingh GK. Association between familial hypercholesterolemia and prevalence of type 2 diabetes mellitus. JAMA. 2015;313:1029–36. doi: 10.1001/jama.2015.1206. [DOI] [PubMed] [Google Scholar]
- 156.Boekholdt SM, Hovingh GK, Mora S, et al. Very low levels of atherogenic lipoproteins and the risk for cardiovascular events: a meta-analysis of statin trials. J Am Coll Cardiol. 2014;64:485–94. doi: 10.1016/j.jacc.2014.02.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Puri R, Nissen SE, Shao M, Uno K, Kataoka Y, Kapadia SR, Tuzcu EM, Nicholls SJ. Impact of baseline lipoprotein and C-reactive protein levels on coronary atheroma regression following high-intensity statin therapy. Am J Cardiol. 2014;114:1465–72. doi: 10.1016/j.amjcard.2014.08.009. [DOI] [PubMed] [Google Scholar]
- 158.Heart Protection Study Collaborative Group. Effects on 11-year mortality and morbidity of lowering LDL cholesterol with simvastatin for about 5 years in 20_536 high-risk individuals: a randomised controlled trial. Lancet. 2011;378:2013–20. doi: 10.1016/S0140-6736(11)61125-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Packard C, Ford I, Murray HM, McCowan C. Lifetime clinical and economic benefits of statin-based LDL lowering in the 20-year follow-up of the west of Scotland coronary prevention study. Circulation. 2014;130:2105–26. [Google Scholar]
- 160.Williams KJ, Feig JE, Fisher EA. Rapid regression of atherosclerosis: insights from the clinical and experiemental literature. Nat Clin Pract Cardiovasc Med. 2008;5:91–102. doi: 10.1038/ncpcardio1086. [DOI] [PubMed] [Google Scholar]
- 161.Goff DC, Jr, Lloyd-Jones DM, Bennett G, et al. American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2935–2959. doi: 10.1016/j.jacc.2013.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Pursnani A, Massaro JM, D’Agostino RB, O’Donnell CJ, Hoffmann U. Guideline-based statin eligibility, coronary artery calcification and cardiovascular events. JAMA. 2015;314:134–141. doi: 10.1001/jama.2015.7515. [DOI] [PMC free article] [PubMed] [Google Scholar]