Statins

Reviewed on July 22, 2024

Introduction

Based on extensive evidence, statins are recommended as first-line pharmacologic therapy for atherosclerotic cardiovascular disease (ASCVD) risk reduction by the 2018 multi-society cholesterol guidelines, as well as all other guidelines. Consistent evidence from over 24 trials shows that statins reduce the risk of ASCVD in proportion to the magnitude of low-density lipoprotein cholesterol (LDL-C) lowering across a range of ASCVD risk, as long as heart failure (NYHA Class II-IV) or end-stage renal disease/hemodialysis are not present. Each 39 mg/dL reduction in LDL-C reduces the risk of major ASCVD events by about 21% (Figure 18-1). The full relative risk reduction occurs within 2 years of initiating statin therapy.Statins reduce the risk of major ASCVD by a similar magnitude across all patient subgroups, risk factor levels, LDL-C levels and other characteristics, with the exception that there is some evidence of a slightly greater reduction in major ASCVD events in men…

Introduction

Based on extensive evidence, statins are recommended as first-line pharmacologic therapy for atherosclerotic cardiovascular disease (ASCVD) risk reduction by the 2018 multi-society cholesterol guidelines, as well as all other guidelines. Consistent evidence from over 24 trials shows that statins reduce the risk of ASCVD in proportion to the magnitude of low-density lipoprotein cholesterol (LDL-C) lowering across a range of ASCVD risk, as long as heart failure (NYHA Class II-IV) or end-stage renal disease/hemodialysis are not present. Each 39 mg/dL reduction in LDL-C reduces the risk of major ASCVD events by about 21% (Figure 18-1). The full relative risk reduction occurs within 2 years of initiating statin therapy. Statins reduce the risk of major ASCVD by a similar magnitude across all patient subgroups, risk factor levels, LDL-C levels and other characteristics, with the exception that there is some evidence of a slightly greater reduction in major ASCVD events in men than in women (Figures 18-2 and 18-3). Statins have also been shown to be safe and well-tolerated in properly selected patients, similar to those who participated in a broad range of primary and secondary prevention trials.

Clinical Highlight I

  • Statins are first-line therapy for reducing ASCVD risk.
  • Use the appropriate intensity of statin in patients most likely to benefit.
  • Statins are very well tolerated in the large majority of patients who might benefit.
Enlarge  Figure 18-1: Relationship Between the Reduction in LDL-C and Relative Reduction in ASCVD Events and the Relative Risk Reduction in ASCVD Events Observed by Year. Source: Cholesterol Treatment Trialists. Lancet. 2005;366:1267-1278.
Figure 18-1: Relationship Between the Reduction in LDL-C and Relative Reduction in ASCVD Events and the Relative Risk Reduction in ASCVD Events Observed by Year. Source: Cholesterol Treatment Trialists. Lancet. 2005;366:1267-1278.
Enlarge  Figure 18-2: Statins Reduce the Relative Risk of Major CVD Across All Patient Subgroups and Characteristics. Rate ratios (RR) are plotted for each comparison of first event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. Missing data are not plotted. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.
Figure 18-2: Statins Reduce the Relative Risk of Major CVD Across All Patient Subgroups and Characteristics. Rate ratios (RR) are plotted for each comparison of first event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. Missing data are not plotted. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.
Enlarge  Figure 18-3: Statins Reduce Major CVD Events Across Baseline LDL-C Levels. Key: Rate ratios (RRs) are plotted for each comparison of first event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. Analyses were done with trial-specific and subgroup-specific LDL weights for each baseline LDL cholesterol category. Missing data are not plotted. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.
Figure 18-3: Statins Reduce Major CVD Events Across Baseline LDL-C Levels. Key: Rate ratios (RRs) are plotted for each comparison of first event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. Analyses were done with trial-specific and subgroup-specific LDL weights for each baseline LDL cholesterol category. Missing data are not plotted. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.

Appropriate Uses

The 2018 multi-society cholesterol guideline strongly recommends statin therapy for four groups of patients most likely to experience a net ASCVD risk reduction benefit.

  • Statins are recommended (Class of Recommendation [COR] I) for high-risk patients, such as those with:
  • Clinical ASCVD (Figure 8-2)
  • Primary prevention when LDL-C is ≥190 mg/dL (Figure 8-3)
  • Diabetes in patients 40-75 years of age (Figure 8-3)
  • Primary prevention in patients 40-75 years of age and LDL-C of 70-189 mg/dL with a high (≥20%) estimated 10-year ASCVD risk (Figure 8-3).

Statins can also be considered for primary prevention following a clinician-patient discussion about the risk estimate and risk enhancers (see Figure 8-3), for:

  • Patients 40 to 75 years of age with intermediate (≥7.5% to 20%) 10-year ASCVD risk (COR I)
  • Patients 40 to 75 years of age with borderline (≥5% to 7.5%) 10-year ASCVD risk (COR IIb).

High-intensity statins (Table 18-1) are recommended for high-risk patients without safety concerns and can be considered for those with intermediate 10-year ASCVD risk. Moderate-intensity statins are otherwise recommended. In patients unable to tolerate moderate- or high-intensity statin therapy, the maximally tolerated intensity should be used.

Enlarge  Figure 8-2: 2018 ACC/AHA Cholesterol Guideline Algorithm for Secondary Prevention of ASCVD. Clinical history of ASCVD consists of acute coronary syndrome, history of myocardial infarction, stable or unstable angina, or coronary other arterial revascularization, stroke, transient ischemic attack, or peripheral artery disease including aortic aneurysm, all of atherosclerotic origin. Very high-risk includes a history of multiple major ASCVD events or one major ASCVD event and multiple high-risk conditions (Table 8.1). Source:  Adapted from Grundy SM, et al. <em>J Am Coll Cardiol</em>. 2019;73(24):3168-3209
Figure 8-2: 2018 ACC/AHA Cholesterol Guideline Algorithm for Secondary Prevention of ASCVD. Clinical history of ASCVD consists of acute coronary syndrome, history of myocardial infarction, stable or unstable angina, or coronary other arterial revascularization, stroke, transient ischemic attack, or peripheral artery disease including aortic aneurysm, all of atherosclerotic origin. Very high-risk includes a history of multiple major ASCVD events or one major ASCVD event and multiple high-risk conditions (Table 8.1). Source: Adapted from Grundy SM, et al. J Am Coll Cardiol. 2019;73(24):3168-3209
Enlarge  Figure 8-3: 2018 Multi-Society Cholesterol Guideline Algorithm for Primary Prevention of ASCVD. apoB, apolipoprotein B; CAC, coronary artery calcium; HIV, human immunodeficiency virus; hs-CRP, high-sensitivity C-reactive protein; Lp(a), lipoprotein (a). Source: Adapted from Grundy SM, et al. <em>J Am Coll Cardiol</em>. 2019;73(24):3168-3209.
Figure 8-3: 2018 Multi-Society Cholesterol Guideline Algorithm for Primary Prevention of ASCVD. apoB, apolipoprotein B; CAC, coronary artery calcium; HIV, human immunodeficiency virus; hs-CRP, high-sensitivity C-reactive protein; Lp(a), lipoprotein (a). Source: Adapted from Grundy SM, et al. J Am Coll Cardiol. 2019;73(24):3168-3209.

Clinical Highlight II

  • Use high-intensity statins to reduce ASCVD risk in high-risk patients (and consider them for intermediate-risk patients) unless there are safety concerns.
  • Use moderate-intensity statins to reduce ASCVD risk if there are safety concerns with high-intensity statins and in intermediate- and borderline-risk patients.
  • Use the maximally tolerated statin dose if unable to tolerate the recommended statin intensity.

Mechanism of Action

Statins block the rate-limiting enzyme in cholesterol synthesis 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase. This enzyme catalyzes the conversion of HMG-CoA to mevalonic acid, which through a series of subsequent steps is converted to cholesterol. Inhibiting HMG-CoA reductase reduces intracellular and plasma cholesterol levels, resulting in upregulated expression of low-density lipoprotein (LDL) receptors in the liver and extrahepatic tissues. The increased number of LDL receptors can then remove more of the cholesterol-rich apolipoproteins: LDL, very low density lipoprotein (VLDL) and VLDL remnants from plasma. Reduced intracellular cholesterol levels also increase PCSK9 levels, which attenuates statin efficacy at higher doses.

There are seven statins available in the United States (Figure 18-4). In 2023, all seven are available as generics.

Enlarge  Figure 18-4: Structural Formulas of Available Statins
Figure 18-4: Structural Formulas of Available Statins

Pleiotropic Effects of Statins

Statins have multiple “pleiotropic” or non–LDL-C effects including lowering high-sensitivity C-reative protein (hs-CRP). However, these effects do not appear to result in additional ASCVD risk reduction compared with other treatments that primarily lower LDL-C, including partial ileal bypass surgery, bile acid sequestrants, diet, or ezetimibe (Figure 18-5).

Enlarge  Figure 18-5: Estimated Change in the 5-Year Relative Risk of Nonfatal MI or CAD Death Associated With Mean LDL-C Reduction.  Key: 4S, Scandinavian Simvastatin Survival Study; AF/TexCAPS, Air Force/Texas Coronary Atherosclerosis Prevention Study; ALERT, Assessment of LEscol in Renal Transplantation; ASCOT-LLA, Anglo-Scandinavian Cardiac Outcomes Trial–Lipid Lowering Arm; CARDS, Collaborative Atorvastatin Diabetes Study; CARE, Cholesterol and Recurrent Events study; CAD, coronary artery disease; HPS, Heart Protection Study; LDL-C, low-density lipoprotein cholesterol; LIPID, Long-Term Intervention with Pravastatin in Ischemic Disease; LRC, Lipid Research Clinics; MRC, Medical Research Council; NHLBI, National Heart, Lung, and Blood Institute; POSCH, Program on the Surgical Control of the Hyperlipidemias; PROSPER, PROspective Study of Pravastatin in the Elderly at Risk; WOSCOPS, West of Scotland Coronary Prevention Study. Estimated change in the 5-year relative risk of nonfatal MI or CHD death associated with mean LDL-C reduction for the diet, bile-acid sequestrant, surgery, and statin trials (dashed line) along with the 95% probability interval (dotted line). The solid line has a slope = 1. The crude risk estimates from the individual studies are plotted along with their associated 95% confidence intervals. The Sydney trial is not shown but was included in the analysis. Statin trials are designated by the boldface symbols.  Source 1) Robinson JG, et al. J Am Coll Cardiol. 2005;46:1855-1862. 2) Cannon CP, et al. N Engl J Med. 2015;372:2387-2397.
Figure 18-5: Estimated Change in the 5-Year Relative Risk of Nonfatal MI or CAD Death Associated With Mean LDL-C Reduction. Key: 4S, Scandinavian Simvastatin Survival Study; AF/TexCAPS, Air Force/Texas Coronary Atherosclerosis Prevention Study; ALERT, Assessment of LEscol in Renal Transplantation; ASCOT-LLA, Anglo-Scandinavian Cardiac Outcomes Trial–Lipid Lowering Arm; CARDS, Collaborative Atorvastatin Diabetes Study; CARE, Cholesterol and Recurrent Events study; CAD, coronary artery disease; HPS, Heart Protection Study; LDL-C, low-density lipoprotein cholesterol; LIPID, Long-Term Intervention with Pravastatin in Ischemic Disease; LRC, Lipid Research Clinics; MRC, Medical Research Council; NHLBI, National Heart, Lung, and Blood Institute; POSCH, Program on the Surgical Control of the Hyperlipidemias; PROSPER, PROspective Study of Pravastatin in the Elderly at Risk; WOSCOPS, West of Scotland Coronary Prevention Study. Estimated change in the 5-year relative risk of nonfatal MI or CHD death associated with mean LDL-C reduction for the diet, bile-acid sequestrant, surgery, and statin trials (dashed line) along with the 95% probability interval (dotted line). The solid line has a slope = 1. The crude risk estimates from the individual studies are plotted along with their associated 95% confidence intervals. The Sydney trial is not shown but was included in the analysis. Statin trials are designated by the boldface symbols. Source 1) Robinson JG, et al. J Am Coll Cardiol. 2005;46:1855-1862. 2) Cannon CP, et al. N Engl J Med. 2015;372:2387-2397.

LDL-C Efficacy

The 2018 multi-society cholesterol guideline grouped statin regimens as high-, moderate-, or low-intensity (Table 8-1) according to their use in randomized trials. High-intensity statins include the highest doses of atorvastatin (40-80 mg) and rosuvastatin (20-40 mg), or a statin used in combination with another LDL-C lowering agent such as ezetimibe.

High-intensity statin regimens reduce LDL-C by ≥50%, which is desirable for high-risk patients. High-intensity statins reduced ASCVD events more than moderate-intensity statins in three randomized trials (TNT, IDEAL, PROVE-IT). Moderate-intensity statin regimens lower LDL-C by approximately 30% to <50%. Starting doses of statins generally achieve this degree of LDL-C lowering (pitavastatin 2 mg, atorvastatin 10 mg, lovastatin or pravastatin 40 mg, rosuvastatin 10 mg, simvastatin 40 mg and fluvastatin 80 mg).

Simvastatin 80 mg failed to reduce ASCVD events more than simvastatin 20 mg in two randomized trials in high-risk patients (SEARCH, A-to-Z), and also reduces LDL-C by <50% on average, so it is grouped with the moderate-intensity statins. Note, however, the FDA does not recommend initiating simvastatin 80 mg due to concerns about muscle safety; patients tolerating simvastatin 80 mg may remain on it.

Only patients who are intolerant to all available moderate-intensity statins should receive a low-intensity dose of statin or less than daily doses.

Each doubling of the statin dose will result in an additional 6% reduction in LDL-C and non– high-density lipoprotein cholesterol (HDL-C) (“rule of sixes”).

Effects on Other Lipids

Statins reduce non–HDL-C to a similar degree as LDL-C. Statins and fibrates reduce coronary artery disease (CAD) risk in proportion to the magnitude of non–HDL-C lowering (Figure 18-6).

Moderate-intensity statins lower triglycerides by about 15% to 20%, while high-intensity statins can lower triglycerides by up to 30%. Statins modestly raise HDL-C (by 2% to 10%), regardless of intensity. Although HDL-C and triglyceride levels predict cardiovascular risk, the increases in HDL-C or decreases in triglyceride from statin or other pharmacologic therapy apparently do not contribute to further cardiovascular risk reduction in addition to that obtained from LDL-C lowering.

Enlarge  Figure 18-6: Estimated Change in the Relative Risk  of a CAD Event (Nonfatal MI or CAD Death) Associated With Non–HDL-C Reduction With Statins at a Mean Follow-Up of 4.5 Years.  Key: Estimated change in the relative risk of a CAD event (nonfatal MI or CAD death) associated with non-HDL-C reduction with statins at a mean follow-up of 4.5 years (dashed line) along with the 95% Bayesian CI (dashed boundary lines). The solid line indicates a 1:1 relationship. The crude risk estimates from the individual studies are plotted along with their associated 95% CI. Statin trials are in black; fibrate trials are in red; niacin trials are in blue (UCSF-SCOR was not plotted); and POSCH, Oslo and LRC are in green. The RRs from the three trials POSCH, Oslo, and LRC were plotted but they are not included in the modeling. Source: Robinson JG, et al. J Am Coll Cardiol. 2009;53:316-322.
Figure 18-6: Estimated Change in the Relative Risk of a CAD Event (Nonfatal MI or CAD Death) Associated With Non–HDL-C Reduction With Statins at a Mean Follow-Up of 4.5 Years. Key: Estimated change in the relative risk of a CAD event (nonfatal MI or CAD death) associated with non-HDL-C reduction with statins at a mean follow-up of 4.5 years (dashed line) along with the 95% Bayesian CI (dashed boundary lines). The solid line indicates a 1:1 relationship. The crude risk estimates from the individual studies are plotted along with their associated 95% CI. Statin trials are in black; fibrate trials are in red; niacin trials are in blue (UCSF-SCOR was not plotted); and POSCH, Oslo and LRC are in green. The RRs from the three trials POSCH, Oslo, and LRC were plotted but they are not included in the modeling. Source: Robinson JG, et al. J Am Coll Cardiol. 2009;53:316-322.

Clinical Highlight III

  • High-intensity statins reduce LDL-C by 50% or more on average.
  • Moderate-intensity statins reduce LDL-C by 30-<50% on average.
  • The magnitude of LDL-C lowering predicts the ASCVD risk reduction benefit from statin therapy.

Patient Selection to Enhance Safety

Statins were well tolerated in the randomized ASCVD outcomes trials. The participants in these trials were generally healthy and had no history of statin associated side effects. Women of childbearing potential or who were pregnant or breastfeeding were excluded from the statin trials due to the statin contraindication in pregnancy.

A number of characteristics are associated with an increased risk of statin myopathy, many of which may have precluded or reduced the likelihood of participation in the statin trials (Table 18-2). These characteristics may alter statin metabolism, and the risk of myopathy and rhabdomyolysis is often related to circulating drug levels (see Managing Adverse Effects During Statin Therapy for more information on the management of statin adverse effects).

Notably, moderate-intensity statins were generally well tolerated in the four trials that did include individuals with serious comorbidities (NYHA Class II-IV heart failure or undergoing maintenance hemodialysis: CORONA, GISSI-HF, AURORA, GDDS). However, very old patients, frail patients, or patients with other serious comorbidities were excluded from statin trials. Therefore, the potential for both adverse effects and an ASCVD risk reduction benefit is not known for groups of patients systematically excluded from the randomized trials.

Clinical Highlight IV

  • Individuals who do not have serious comorbidities or a history of statin-associated side effects are unlikely to have adverse effects related to statin therapy.

Metabolism

Statins are primarily metabolized in the liver by the hepatic cytochrome P450 enzyme (CYP) system, glucuronidation, or both (Table 18-2). Exposure to other drugs that are metabolized by these pathways may increase statin blood levels and thereby increase the potential for adverse effects (see below).

Renal Function

Some statins have relatively greater renal excretion: lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin. Dose adjustment may be considered for patients with markedly impaired renal excretion.

All statins should be used with caution in patients with a glomerular filtration rate <30 mL/min/1.73m2 since substantially impaired renal excretion is also a marker for other patient characteristics that may increase the potential for adverse muscle effects, including advanced age, frailty and polypharmacy.

Contraindications

Contraindications to statin therapy are few:

  • Active liver disease, which may include unexplained persistent elevations in hepatic transaminase levels
  • Women who are pregnant or may become pregnant
  • Nursing mothers
  • Hypersensitivity to any component of the medication.

Clinical Highlight V

  • A woman of childbearing potential who is likely to experience a net benefit from statin therapy, and her partner, should be carefully counselled to practice effective contraception during statin therapy.

Potential Adverse Effects

Although patients taking statins commonly report adverse effects, few of these are attributable to statin therapy. In the randomized blinded statin trials, adverse effect rates were similar in the statin and placebo groups, with the exception of a slight excess of overt diabetes in statin-treated patients.

Muscle Symptoms

Rates of muscle symptoms and myopathy were similar in statin and control groups in the randomized trials. Serious myopathy and rhabdomyolysis occurred at a rate of about 0.01 excess case per 100 patients treated with a statin for 1 year. While 10%-15% of patients taking a statin report muscle symptoms, in the majority of patients they are unrelated to statin use.

Hepatic Transaminase Elevations

In the randomized trials, statins did not increase hepatic transaminases and did not cause serious hepatotoxicity or liver failure. As a result, the FDA and the 2018 multi-society cholesterol guideline do not recommend routine monitoring liver function tests.

Hemorrhagic Stroke

Hemorrhagic stroke was very uncommon in the statin trials with a rate of about 0.01 excess cases per 100 patients treated for 1 year. The risk of hemorrhagic stroke may be higher in patients with a history of hemorrhagic stroke receiving a high-intensity statin and in Asian patients. The reduction in ischemic stroke with statins outweighs any excess risk of hemorrhagic stroke, resulting in a reduction in total stroke.

Diabetes

Statins are associated with a modest excess risk of diabetes, which is related to intensity. Moderate-intensity statins increase the excess risk of diabetes by 0.1 excess case of diabetes per 100 patients treated for 1 year; high-intensity statins increase the excess risk to 0.3 excess case per 100 patients per year. This rate occurs in addition to the background risk of diabetes, which is often relatively high in candidates for statin therapy on the basis of their ASCVD risk. Statins are associated with increased risk of diabetes only in patients who have risk factors for diabetes. Diabetes diagnosis occurs 2 to 4 months earlier in statin-treated patients than in those not receiving a statin, making it unlikely to have any sequelae.

Cognitive Function

The statin trials have not reported an excess of cognitive impairment. In those trials that formally assessed cognitive function, moderate intensity statins had no effect on cognitive function in the trials overall (HPS, PROSPER), with some suggestion of a benefit in clinical trial participants >70 years of age (HPS). In patients with dementia, statins had a neutral effect on cognition in randomized trials.

However, postmarketing reports have associated cognitive impairment (e.g., memory loss, forgetfulness, amnesia, memory impairment, confusion) with statin use. Symptoms were generally mild and reversible upon statin discontinuation, with variable times to symptom onset (1 day to years) and symptom resolution (median of 3 weeks). As a result, the FDA has issued a warning that statins could cause ill-defined memory loss or impairment, which is reversible upon statin discontinuation.

The general approach to statin withdrawal/rechallenge used for the management of muscle symptoms during statin therapy may help clarify the relationship of the cognitive symptoms to statin use.

Mood

A meta-analysis of randomized trials found no negative effects from statins on psychological outcomes and evidence of mood-related benefits.

Neuropathy

Rates of neuropathy and other neurologic conditions are similar in statin and control groups in randomized trials. Rarely, idiosyncratic peripheral neuropathy may develop that resolves with statin discontinuation.

Renal

Statins have no adverse renal effects and may prevent decline in renal function with time.

Cancer

Cancer rates are similar in statin and control groups in individual and meta-analyses of randomized trials. Cancer rates have remained similar in the two treatment groups in long-term follow-up of several statin trials. In large population studies, statin use is associated with lower cancer mortality. Figure 18-7 shows the incidence of cancer in statin trials.

Enlarge  Figure 18-7: Cancer Incidence in Statin Trials. Rate ratios (RRs) are plotted for each comparison of first-event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Analyses are of first cancers subdivided by site: gastrointestinal, genitourinary, respiratory, female breast, hematological, melanoma, other/unknown site. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.
Figure 18-7: Cancer Incidence in Statin Trials. Rate ratios (RRs) are plotted for each comparison of first-event rates between treatment groups and are weighted per 1.0 mmol/L LDL cholesterol (LDL-C) difference at 1 year. RRs are shown with horizontal lines denoting 99% CIs or with open diamonds showing 95% CIs. Analyses are of first cancers subdivided by site: gastrointestinal, genitourinary, respiratory, female breast, hematological, melanoma, other/unknown site. Source: Cholesterol Treatment Trialists Collaboration. Lancet. 2010;376:1670-1681.

Drug-Drug Interactions

Statins have interactions with various drugs depending on their primary pathway for metabolism. Healthcare professionals should refer to the drug labels for the latest recommendations.

Metabolized by CYP 3A4

The three statins that are metabolized by hepatic cytochrome P450 enzyme (CYP) 3A4 have the greatest potential for drug interactions—atorvastatin, lovastatin and simvastatin (remember as “A, L, S”) (Table 18-3).

Avoid concomitant use of these agents with potent inhibitors of CYP3A4, including azole antifungals, macrolide antibiotics, rifampin and protease inhibitors (Table 18-4).

Lower doses of simvastatin and lovastatin are recommended for patients receiving weaker CYP3A4 inhibitors such as amiodarone and the calcium channel blockers diltiazem and verapamil (alternatives include amlodipine and nifedipine). Interactions with some antidepressants have also been reported; paroxetine and venlafaxine may be alternatives for some patients.

Not Metabolized by CYP3A4

Alternatives to CYP3A4 metabolized statins include:

  • Rosuvastatin, which is minimally metabolized
  • Pravastatin, which has no cytochrome P450 metabolism
  • Fluvastatin, which is metabolized by the 2C9 pathway
  • Pitavastatin, which has no significant CYP3A4, 2C9, or 2C8 metabolism.

Glucuronidation

All statins are glucuronidated, increasing the potential for interaction with gemfibrozil. Fenofibrate has minimal effects on glucuronidation.

Cyclosporine

Cyclosporine raises blood levels of virtually all statins by both cytochrome P450 and other pathways, and low doses of statins (preferably other than atorvastatin, lovastatin and simvastatin) should be titrated carefully if needed. See Special Clinical Populations for further discussion of other immunosuppressant drugs.

Clinical Highlight VI

  • The vast majority of patients tolerate statins well.
  • Rarely, idiosyncratic adverse events may occur; establish statin causality by withdrawing and rechallenging with the statin or statins.
  • Atorvastatin and simvastatin are appropriate for most patients; avoid concomitant use with CYP3A4 inhibitors.
  • Rosuvastatin, pitavastatin and fluvastatin are alternatives with fewer drug-drug interactions.
  • Avoid concomitant use of gemfibrozil and statins.

 

References

  • Robinson JG. Clinical Lipid Management, 2nd ed. Professional Communications Inc. 2023
  • Alsheikh-Ali AA, Trikalinos TA, Kent DM, Karas RH. Statins, low-density lipoprotein cholesterol, and risk of cancer. J Am Coll Cardiol. 2008;52:1141-1147.
  • Altoprev (lovastatin extended-release) [package insert]. Zug, Switzerland; Covis Pharma. 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021316s036lbl.pdf. Accessed January 15, 2023.
  • Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Statins can improve proteinuria and glomerular filtration rate loss in chronic kidney disease patients, further reducing cardiovascular risk. Fact or fiction? Expert Opin Pharmacother. 2015;16:1449-1461.
  • Baigent C, Keech A, Kearney PM, et al; Cholesterol Treatment Trialists’ (CTT) Collaborators. Efficacy and safety of cholesterol lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366:1267-1278.
  • Brass LM, Alberts MJ, Sparks L; National Lipid Association Statin Safety Task Force Neurology Expert Panel. An assessment of statin safety by neurologists. Am J Cardiol. 2006;97:86C-88C.
  • Briel M, Ferreira-Gonzalez I, You JJ, et al. Association between change in high density lipoprotein cholesterol and cardiovascular disease morbidity and mortality: systematic review and meta- regression analysis. BMJ. 2009;338:b92.
  • Cannon CP, Braunwald E, McCabe CH et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med. 2004;350:1495-1504.
  • Chatterjee S, Krishnamoorthy P, Ranjan P, et al. Statins and cognitive function: an updated review. Curr Cardiol Rep. 2015;17:4.
  • Cholesterol Treatment Trialists’ (CTT) Collaboration, Baigent C, Blackwell L, Emberson J, et al. 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;376:1670-1681.
  • Cholesterol Treatment Trialists’ (CTT) Collaborators, Mihaylova B, Emberson J, Blackwell L, et al. 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.
  • Crestor (rosuvstatin calcium). [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2022. https://den8dhaj6zs0e.cloudfront.net/50fd68b9-106b-4550-b5d0-12b045f8b184/fd6c9085-0722-46d7-b166-bd68f09c43d5/fd6c9085-0722-46d7-b166-bd68f09c43d5_viewable_rendition__v.pdf. Accessed January 15, 2023
  • Davidson MH, Robinson JG. Lipid-lowering effects of statins: a comparative review. Exp Opin Pharmacother. 2006;7:1701-1714.
  • de Lemos JA, Blazing MA, Wiviott SD, et al; Investigators. Early intensive vs a delayed conservative simvastatin strategy in patients with acute coronary syndromes: phase Z of the A to Z trial. JAMA. 2004;292:1307-1316.
  • Desai CS, Martin SS, Blumenthal RS. Non-cardiovascular effects associated with statins. BMJ. 2014;349:g3743.
  • Fellström BC, Jardine AG, Schmieder RE, et al; AURORA Study Group. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med. 2009;360:1395-1407.
  • Finegold JA, Manisty CH, Goldacre B, Barron AJ, Francis DP. What proportion of symptomatic side effects in patients taking statins are genuinely caused by the drug? Systematic review of randomized placebo-controlled trials to aid individual patient choice. Eur J Prev Cardiol. 2014;21:464-474.
  • Gissi-HF Investigators, Tavazzi L, Maggioni AP, Marchioli R, et al. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo- controlled trial. Lancet. 2008;372:1231-1239.
  • Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1082-e1143.
  • Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360:7-22.
  • Kasiske BL, Wanner C, O’Neill WC; National Lipid Association Statin Safety Task Force Kidney Expert Panel. An assessment of statin safety by nephrologists. Am J Cardiol. 2006;97:82C-85C.
  • Kjekshus J, Apetrei E, Barrios V, et al; CORONA Group. Rosuvastatin in older patients with systolic heart failure. N Engl J Med. 2007;357:2248-2261.
  • LaRosa JC, Grundy SM, Waters DD, et al; Treating to New Targets (TNT) Investigators. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med. 2005;352:1425-1435.
  • Lescol XL (fluvastatin sodium). [package insert]. East Hanover, NJ: Novartis Pharmaceutical Corporation; 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021192s031lbl.pdf. Accessed January 15, 2023.
  • Lipitor (atorvastatin calcium.) [package insert]. New York, NY: Pfizer-Parke-Davis; 2015. http://labeling.pfizer.com/ ShowLabeling.aspx?id=587. Accessed January 15, 2023.
  • Livalo (pitivastatin). [package insert]. Montgomery, AL: Kowa Pharmaceuticals America, Inc.; 2013. http://www. kowapharma.com/documents/LIVALO_PI_CURRENT.pdf. Accessed January 15, 2023.
  • Marelli C, Gunnarsson C, Ross S, et al. Statins and risk of cancer: a retrospective cohort analysis of 45,857 matched pairs from an electronic medical records database of 11 million adult Americans. J Am Coll Cardiol. 2011;58:530-537.
  • Naci H, Brugts J, Ades T. Comparative tolerability and harms of individual statins: a study-level network meta-analysis of 246 955 participants from 135 randomized, controlled trials. Circ Cardiovasc Qual Outcomes. 2013;6:390-399.
  • Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid-lowering drugs: mechanisms and clinical relevance. Clin Pharmacol Ther. 2006;80:565-581.
  • Nielsen SF, Nordestgaard BG, Bojesen SE. Statin use and reduced cancer-related mortality. N Engl J Med. 2012;367:1792-1802.
  • O’Neil A, Sanna L, Redlich C, et al. The impact of statins on psychological wellbeing: a systematic review and meta-analysis. BMC Med. 2012;10:154.
  • Pedersen TR, Faergeman O, Kastelein JJ, et al; Incremental Decrease in End Points Through Aggressive Lipid Lowering (IDEAL) Study Group. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial infarction: the IDEAL study: a randomized controlled trial. JAMA. 2005;294:2437-2445.
  • Pravachol (pravastatin sodium). [package insert]. Princeton, NJ: Bristol Myers Squibb Company.; 2020. http://packageinserts.bms.com/pi/pi_pravachol.pdf. Accessed January 15, 2023.
  • 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;305:2556-2564.
  • 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.
  • Robinson J, Davidson M. Combination therapy with ezetimibe and simvastatin to acheive aggressive LDL reduction. Expert Rev Cardiovasc Ther. 2006;4:461-476.
  • Robinson J, Stone N. The 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular disease risk: a new paradigm supported by more evidence. Eur Heart J. 2015;36:2110-2118.
  • Robinson JG, Bakris G, Torner J, Stone NJ, Wallace R. Is it time for a cardiovascular primary prevention trial in the elderly? Stroke. 2007;38:441-450.
  • 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;46:1855-1862.
  • 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;53:316-322.
  • Robinson JG. Statins and diabetes risk: how real is it and what are the mechanisms? Curr Opin Lipidol. 2015;26:228-235.
  • Rojas-Fernandez CH, Cameron JC. Is statin-associated cognitive impairment clinically relevant? A narrative review and clinical recommendations. Ann Pharmacother. 2012;46:549-557.
  • Rosenson RS, Baker SK, Jacobson TA, Kopecky SL, Parker BA, The National Lipid Association’s Muscle Safety Expert Panel. An assessment by the Statin Muscle Safety Task Force: 2014 update. J Clin Lipidol. 2014;8(3 Suppl):S58-S71.
  • Russo MW, Hoofnagle JH, Gu J, et al. Spectrum of statin hepatotoxicity: experience of the drug-induced liver injury network. Hepatology. 2014;60:679-686.
  • Sørensen HT, Riis AH, Lash TL, Pedersen L. Statin use and risk of amyotrophic lateral sclerosis and other motor neuron disorders. Circ Cardiovasc Qual Outcomes. 2010;3:413-417.
  • Study of the Effectiveness of Additional Reductions in Cholesterol and Homocysteine (SEARCH) Collaborative Group, Armitage J, Bowman L, Wallendszus K, et al. Intensive lowering of LDL cholesterol with 80 mg versus 20 mg simvastatin daily in 12,064 survivors of myocardial infarction: a double-blind randomised trial. Lancet. 2010;376:1658-1669.
  • Trompet S, van Vliet P, de Craen AJ, et al. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol. 2010;257:85-90.
  • US Food and Drug Administration. FDA Drug Safety Communication: Important safety label changes to cholesterol-lowering statin drugs. FDA Drug Safety Communication. 2016; https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-important-safety-label-changes-cholesterol-lowering-statin-drugs. Accessed January 15, 2023.
  • Wanner C, Krane V, März W, et al; German Diabetes and Dialysis Study Investigators. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med. 2005;353:238-248.
  • Zocor (simvastatin). [package insert]. Jersey City, NJ: Organon, LLC; 2022. https://www.organon.com/product/usa/pi_circulars/z/zocor/zocor_pi.pdf. Accessed January 15, 2023.