Vol 82, No 5 (2024)
Review paper
Published online: 2024-05-02

open access

Page views 425
Article views/downloads 299
Get Citation

Connect on Social Media

Connect on Social Media

REVIEW

Overcoming patient reluctance to statin intolerance

Basant E Katamesh1Alicia A Mickow2Linda Huang3Brian M Dougan4Basem M Ratrout4Sanjeev Nanda4Ann Vincent4
1Research Fellow in the Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
2Division of Preventive Cardiology, Mayo Clinic, Rochester, Minnesota, United States
3Pharmacy Services, Mayo Clinic, Rochester, Minnesota, United States
4Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States

Correspondence to:

Ann Vincent, MD,

Division of General Internal Medicine,

Mayo Clinic,

200 First St SW, Rochester, MN 55905,

phone: 507 284 25 11,

e-mail: Vincent.Ann@mayo.edu

Copyright by the Author(s), 2024

DOI: 10.33963/v.phj.100526

Received: March 21, 2024

Accepted: May 2, 2024

Early publication date: May 2, 2024

ABSTRACT
Statin therapy is a cornerstone in the management of dyslipidemia, both in primary and secondary prevention of cardiovascular events. Despite strong guidelines supporting statin use, concerns regarding side effects, particularly musculoskeletal symptoms, contribute to statin intolerance and patient reluctance. While statin intolerance is reported in 5% to 30% of patients, its true prevalence may be overestimated due to the influence of the nocebo effect. Factors associated with higher incidence of statin intolerance include older age, female sex, comorbidities such as diabetes and chronic kidney disease, and concurrent use of medications such as antiarrhythmic agents or calcium channel blockers. Clinical characterization of statin intolerance requires thorough evaluation and exclusion of alternative causes of musculoskeletal symptoms. Strategies to address statin intolerance include reassessing cardiovascular risk, engaging in shared decision-making, statin rechallenge after appropriate washout periods, dosage titration for tolerability, and consideration of alternative therapies when low-density lipoprotein goals cannot be achieved with statins. This review provides an overview of the spectrum of statin intolerance, its clinical assessment, and a systematic approach to caring for a patient with statin intolerance.
Key words: cholesterol, dyslipidemia, statins

The American College of Cardiology (ACC) 2022 guidelines recommend statin therapy as the primary prevention for patients diagnosed with familial hypercholesterolemia (FH), patients aged 20 to 39 years with a family history or premature atherosclerotic cardiovascular disease (ASCVD) and low-density lipoprotein cholesterol (LDL-C) ≥160 mg/dl, patients aged 40 to 75 years with diabetes mellitus regardless of LDL-C level, and patients of any age with an LDL-C ≥190 mg/dl [1]. Statins are also the standard of care for secondary prevention in patients with clinical ASCVD, including acute coronary syndrome, stable angina, arterial calcification, stroke, transient ischemic attack, and peripheral arterial disease [1]. Recommended LDL-C targets are 70100 mg/dl for primary prevention and 5570 mg/dl for secondary prevention [1]. European guidelines for lipid-lowering therapy recommend a comprehensive risk assessment utilizing the SCORE2 tool [2, 3]. This tool estimates the 10-year cardiovascular risk of developing fatal and non-fatal cardiovascular diseases by factoring in variables such as age, sex, smoking status, lipid levels, diabetes, and blood pressure [2, 3]. Patients are classified into low risk (SCORE2 risk <1%), moderate (SCORE2 risk 1%–5%), high (SCORE2 risk 5%–10%), and very high risk (SCORE2 risk ≥10%) [3]. The European Society of Cardiology (ESC) recommends statin therapy for individuals classified as high-risk (goal LDL-C <70 mg/dl and ≥50% baseline reduction) or very high-risk (goal LDL-C <55 mg/dl ≥50% baseline reduction) in the SCORE-2 model in addition to all patients with post-acute myocardial infarction [2, 3]. They also recommend statin therapy for patients with moderate risk on the SCORE2 model if lifestyle strategies do not reduce LDL-C <100 mg/dl [2, 3]. In addition, the Polish Cardiac Society and the International Lipid Expert Panel have defined criteria for patients with extremely high-risk ASCVD and recommend a lower LDL-C target of <40 mg/dl for this patient group [4, 5]. Extremely high-risk patients include those with multivessel coronary artery disease, polyvascular disease, heterozygous FH, and diabetes mellitus [4, 5]. This category also takes into account other risk factors such as high-sensitivity C-reactive protein ≥3 mg/l, chronic kidney disease with estimated glomerular filtration rate <60 ml/min/1.73 m2, lipoprotein a >50 mg/dl, and patients with a history of vascular events within the last 2 years who did not achieve a target LDL-C <55 mg/dl [4, 5]. Statins may also benefit patients with atherosclerotic vascular aneurysms [6]. Despite all of these recommendations, some patients are unwilling to consider statin therapy.

The literature cites multiple reasons why patients are reluctant to consider statin therapy (Figure 1).

Figure 1. Reasons for patient reluctance to statin therapy

In a survey study conducted on an online health forum posted by over 2000 patients utilizing statins, 90% reported discontinuing statins due to musculoskeletal side effects [7]. Similarly, in a lipid managing registry evaluating lipid management practices, 55% of patients reported discontinuing statins due to musculoskeletal side effects [8]. Among patients who were reluctant to consider statins (10%), the most common reason was worry regarding side effects, concerns for liver disease, memory loss, and musculoskeletal issues [8]. Other reasons included patients wanting to try diet or exercise, the belief that statins were unnecessary, not wanting to take medications, preferring natural remedies, and cost/insurance reasons [8]. In another survey study, the Statin Adverse Treatment Experience survey, reporting the experience of 1500 statin patients, 22.1% of patients reported musculoskeletal symptoms of muscle pain, muscle cramps, stiffness, joint pain, muscle weakness, and other symptoms such as easy fatigability [9]. Additionally, the impact of symptoms on performance, productivity, ability to be physically and socially active, and getting restorative sleep were important patient concerns [9]. Patient-reported symptoms do not necessarily correlate with the dosage of statins. This was demonstrated in an observational study of 1000 statin users, 78% of whom were on low-intensity statins (5 mg rosuvastatin, 10 mg atorvastatin, or 20 mg simvastatin) [10]. In this study, approximately 10% of patients reported musculoskeletal symptoms, demonstrating the occurrence of symptoms even with low-intensity statin therapy [10].

Statin intolerance, reported in 5% to 30% of patients, is defined by the National Lipid Association as adverse symptoms that occur with statin pharmacotherapy and resolve either with discontinuation, dose reduction, or spacing of statin dosage [11–13]. The most common adverse symptoms are musculoskeletal symptoms; others include liver toxicity and dysglycemia [14]. Statin intolerance may be partial or complete; partial intolerance is intolerance at specific doses, while patients with complete intolerance cannot tolerate any dose [11]. Patients with partial intolerance can typically tolerate smaller statin doses or alternative statins, but such treatments are usually insufficient for achieving the desired LDL-C reduction [11]. According to the National Lipid Association, a patient must be trialed on a minimum of two statins with at least one at the lowest approved daily dose before a diagnosis of statin intolerance can be established. There are several factors reported to be associated with a higher incidence of statin intolerance. These include older age, female sex, Asian or Black ethnicity, alcohol use, obesity, diabetes mellitus, hypothyroidism, chronic liver disease, kidney disease, and use of certain medications (e.g., antiarrhythmic agents or calcium channel blockers) [15]. Since the presentation of statin intolerance can mimic symptom presentation in other conditions such as vitamin D deficiency, polymyalgia rheumatica, poly­myo­sitis, hypothyroidism, and medication interactions, it is important to exclude these conditions before establishing the presence of statin intolerance [16]. Statin intolerance has also been reported as part of the spectrum in patients with multiple drug intolerances [17]. Although the etiology of multiple drug intolerance is unknown, factors reported to be associated with this phenomenon included female sex, long-standing hypertension, comorbidities involving respiratory, gastrointestinal, rheumatological, and endocrine systems, and chronic use of medications such as analgesics and beta-blockers [17].

The true number of patients with statin intolerance may be overestimated by clinicians as patient-reported symptoms can be factually true or perceived to be true (nocebo effect) [15]. In a meta-analysis of over 4 million patients, Bytyci et al. [15] evaluated statin intolerance across 176 cohort studies and randomized controlled trials. Using statin intolerance research criteria established by international lipid societies, the authors estimated that the prevalence of statin intolerance in randomized blinded controlled trials was significantly lower when compared to cohort studies (4.9% vs. 17%), supporting the concept of overestimation [15]. These estimates which are lower than those reported in observational studies suggest that a fraction of patient-reported symptoms may reflect the nocebo effect [18, 19]. Nocebo effects reported by patients varied from exaggeration of existing symptoms to reporting new symptoms, or exaggerated fear of medication side effects, all of which may result in patients losing trust in their healthcare providers or intentionally non-adhering or discontinuing treatment [20, 21]. Factors that influence nocebo effects include female gender, cultural and contextual elements, misinformation from friends, family members, and media, and patient expectations influenced by information from healthcare providers, pharmacists, and medication package inserts [22–24].

Clinical characterization of statin intolerance in practice can be a challenge. In the absence of abnormalities in biomarkers related to muscle function, clinicians must rely on patient reports supplemented with a thorough workup to rule out other causes of musculoskeletal symptoms. Details regarding all prior statin use including statin type, dosage at which adverse effects occurred, resolution of adverse effects with discontinuation of statins, alternative statin used, and length of washout period before statin retrial will help establish a diagnosis. Although a good history may be sufficient to establish the presence of statin intolerance, there are tools that clinicians can avail to assess statin intolerance and guide medication choices. One such tool is the ACC Statin Intolerance Tool [25]. The tool has 3 components: 1) evaluation, 2) follow-up, and 3) drug comparison. The first component aids in evaluating the patient’s likelihood of statin intolerance taking into account the prescribed statin and specific dosage, secondary factors, and medical comorbidities that may contribute to musculoskeletal symptoms. It also evaluates whether medication interactions may be playing a role in musculoskeletal symptoms. The second component, follow-up, guides appropriate laboratory workup, washout period, and provides options for rechallenging with statins. Additionally, it includes an algorithm to guide evaluation of musculoskeletal symptoms that may occur with a rechallenge. The third component offers dosage guidelines for statins and evaluates potential medication interactions [25]. A second tool, Statin Choice Decision Aid, although not specific to statin intolerance, can be used for shared decision-making with the patient regarding atherosclerotic cardiovascular risk, need for statin therapy, and the pros and cons of standard dose versus high dose versus no statin [26].

Several strategies have been recommended to address statin intolerance and patient concerns related to intolerance. A systematic review by Meza-Contreras et al. [16] identified 26 articles that described clinical strategies to address statin intolerance. Most of the studies recommended early exclusion of other causes of musculoskeletal symptoms such as vitamin D deficiency, polymyositis, hypothyroidism, or drug-drug interactions. Almost all studies recommended stopping the current statin and recalculating ASCVD risk, and all studies recommended rechallenging with a statin after a washout period of 26 weeks. Regarding the selection of type and dosage of statin, recommendations included re-assessing the current statin dose, using a lower dose, switching to another statin, altering the statin dosing schedule, adding a non-statin drug to the best-tolerated statin therapy, or switching to non-statin. Ezetimibe, while lacking pleiotropic benefits, was the most recommended first-line non-statin therapy and the most frequently used when transitioning patients from statins [27–29]. Other non-statin therapies included proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, nutraceuticals, bempedoic acid, and ion exchange resins [16].

Based on recommendations in the literature and our clinical experience in the cardiovascular lipid clinic, we suggest the following approaches to address statin intolerance and patient reluctance toward statins:

Evaluate for statin intolerance and address other etiologies for musculoskeletal symptoms

Evaluate statin intolerance using a detailed history of current or previous statin use, symptoms and their chronological association with statin initiation, biomarker abnormalities at the time of undesirable effects, and details regarding symptom resolution once statin was discontinued. As part of this evaluation, a thorough workup to assess for other causes of musculoskeletal symptoms such as myopathies and polymyositis, exclusion of conditions such as vitamin D deficiency, hypothyroidism, and medication interactions should be considered [16]. In rare instances, if indicated, workup could include testing for gene variants and antibodies (solute carrier organic anion transporter family member 1B1 [SLCO1B1] and human menopausal gonadotropin CoA reductase antibodies) in patients who may be genetic carriers and therefore at higher risk of statin myopathy [30–32].

Reassess ASCVD risk and benefits and risks of statin therapy through shared decision-making

  • Once the existence of statin-associated symptoms has been verified, we recommend reassessing ASCVD risk and re-establishing the need for statin therapy.
  • This can occur in shared decision-making in a clinical encounter and if needed can be assisted by electronic clinical decision aids [33].
  • ASCVD risk can be calculated by online tools such as the ASCVD [34], MESA [35], or the Astro-CHARM ASCVD risk calculator [36] in the United States, and SCORE2-based online tool in Europe [3].
  • Another tool that incorporates both ASCVD risks and benefits and risks of statin therapy for individual patients is the Statin Choice Decision Aid developed by the Knowledge and Evaluation Research Unit of the Mayo Clinic [26]. This tool is currently embedded in electronic medical records and systems such as EPIC [37] and can help assess the patient’s present and future risk of coronary disease using either the ACC and American Heart Association ASCVD, Framingham risk score, or Reynolds risk score calculators depending on the type of patient data available [26]. Using this tool, clinicians can input patient data, calculate current risk, and predict future risk scenarios in various conditions (without a statin, with a low dose, or with a high-dose statin) to determine the most appropriate statin dose based on the patient’s current risk [26].
  • During shared decision-making, other considerations that may assist with enhancing compliance to therapy include inquiries into patient preferences regarding therapy, education regarding possible side effects, their management, the potential for nocebo effect, and options to navigate this.
  • Additionally, discussion regarding additional lipid-lowering benefits of cardiovascular lifestyle activities (healthy diet, moderate exercise, weight loss, and cessation of smoking) may be encouraged. Discussion regarding the use of dietary supplements such as CoQ10 to mitigate muscle symptoms may be considered [38–41]. Although occasionally patients may gravitate to natural products such as fermented red yeast rice, fish oil, cinnamon, garlic, and turmeric, there is insufficient credibility and/or evidence to justify using these products as lipid-lowering therapies [42, 43].

Consider alternative dosing strategies and washout periods before retrials

Once a choice of statin has been narrowed, we recommend initiation of a statin starting with a low dose and gradually titrating it to obtain the optimal dosage. In a patient with a history of statin-associated symptoms and currently not on a statin, we recommend using a lower dose of a previously used statin or an alternative statin after an appropriate washout period of a minimum of 4 weeks. Considerations include starting the patient on a lower statin than recommended i.e., 25%-50% of the lowest strength of that statin either daily or on alternate days or two times a week, and dosage or frequency can be titrated. If, at any time during this titration, patients report symptoms, dosage or frequency can be decreased to previously tolerable doses/frequency. Consider a short washout period before retrials. If the patient is currently on a statin and is experiencing symptoms, we recommend a minimum of a 4-week washout period before retrying the statin. In such cases, the previously mentioned considerations should be implemented [16].

Establish statin intolerance

To meet the criteria for statin intolerance, patients should have failed trials with a minimum of two different statins, at least one of which is trialed at the lowest approved daily dose. Statin intolerance may be either a complete inability to tolerate any statin or partial intolerance with higher doses necessitating adjunctive treatments [11, 44, 45].

Approaches for patients with statin intolerance

  • If target LDL-C cannot be achieved by low-dose statins, then non-statins such as ezetimibe, bempedoic acid alone or in combination can be utilized for modest LDL-C reduction, or PCSK9 monoclonal antibodies (evolocumab and alirocumab) can be considered for more aggressive LDL-C reduction [1, 46–48]. These medications have lower reported musculoskeletal side effects, however, they may require insurance justification to rationalize the need for nonstatin; this could be a high ASCVD score, imaging evidence of ASCVD, and/or documentation of statin intolerance [49].
  • In patients with complete statin intolerance, non-statin therapies can be utilized as alternative therapy. The choice of non-stains in these situations depends on clinical scenarios including ASCVD risk, presence of established coronary artery disease, and other medical comorbidities such as diabetes and chronic kidney disease. Inclisiran, which acts through PCSK9 inhibition and is FDA-approved for clinical ASCVD or familial hyperlipidemia, is another consideration [11]. An overview of these treatment options is presented in Table 1.
Table 1. Comparative overview of statin and non-statin therapies

Medication

Dose and route

Mean LDL-C reduction

Side effects

Monitoring

Atorvastatin

10–80 mg/day, once orally

Up to 50%

  • Musculoskeletal side effects
  • Elevation in transaminases
  • Lipid monitoring within 2–4 weeks of initiation/titration
  • Liver enzymes monitoring with dosage initiation/titration

Rosuvastatin

5–40 mg/day, once orally

Simvastatin

5–40 mg/day, once orally,

Pravastatin

10–80 mg/day, once orally

Less than 30%

Bempedoic acid

180 mg/day, once orally, may be combined with 10 mg ezetimibe

17%–18%

  • Upper respiratory tract infection
  • Muscle spasm
  • Hyperuricemia
  • Back pain
  • Abdominal pain
  • Bronchitis
  • Pain in extremity
  • Anemia
  • Elevated liver enzyme
  • Uric acid
  • Liver enzymes

Ezetimibe

10 mg/day, orally

18% (alone)

25% (combined with statin)

Monotherapy:

  • Upper respiratory tract infection
  • Diarrhea
  • Arthralgia
  • Sinusitis
  • Pain in extremity

Combined with statins:

  • Nasopharyngitis
  • Myalgia
  • Upper respiratory tract infection
  • Arthralgia
  • Diarrhea
  • Aspartate transaminase
  • Alanine transaminase

PCSK9 mAb

Alirocumab:

  • 75 mg, s.c. every 2 weeks, may increase to 150 mg every two weeks (ASCVD, or primary hyperlipidemia)
  • 300 mg, s.c. every 4 weeks (ASCVD, or primary hyperlipidemia)
  • 150 mg, s.c. every 2 weeks (HeFH undergoing LDL apheresis, or HoFH)

Evolocumab:

  • 140 mg, s.c. every 2 weeks (ASCVD, or HeFH)
  • 420 mg, s.c. once monthly, if needed can be increased to every 2 weeks (HoFH)
  • 420 mg, s.c. every 2 weeks (HoFH on LDL apheresis)

Alirocumab: 45%–58%

Evolocumab: 58%–64%

Primary hyperlipidemia:

  • Nasopharyngitis
  • Injection site reaction
  • Influenza
  • Urinary tract infection
  • Back pain

ASCVD:

  • Non -cardiac chest pain
  • Nasopharyngitis
  • Myalgia
  • Diabetes
  • Urinary tract infection

Repeat lipid panel before 5th dose and annually

Inclisiran

284 mg s.c. on day 1, 90, then every 6 months in an Infusion Center

48%–52%

  • Injection site reaction
  • Arthralgia
  • Urinary tract infection
  • Bronchitis
  • Diarrhea
  • Pain in extremity
  • Dyspnea

Recheck before the 2nd dose and annually

Special scenarios

Statins are the mainstay treatment for FH patients [50]. For patients who are intolerant to statins or do not achieve target LDL-C reduction, the ESC recommends adding ezetimibe [50, 51]. The ESC considers statins combined with ezetimibe as the cornerstone for treatment of FH patients [50]. If patients do not achieve the target LDL-C level with the maximally tolerated statin dose and ezetimibe, guidelines recommend adding PCSK9 inhibitors to therapy [50].

Article information

Acknowledgment: The Scientific Publications staff at Mayo Clinic provided copyediting support.

Conflict of interest: None declared.

Funding: None.

Open access: This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, which allows downloading and sharing articles with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially. For commercial use, please contact the journal office at polishheartjournal@ptkardio.pl

REFERENCES

  1. Lloyd-Jones DM, Morris PB, Ballantyne CM, et al. 2022 ACC expert consensus decision pathway on the role of nonstatin therapies for ldl-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2022; 80(14): 13661418, doi: 10.1016/j.jacc.2022.07.006, indexed in Pubmed: 36031461.
  2. Mach F, Baigent C, Catapano AL, et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk: The Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS). Eur Heart J. 2020; 41(1): 111188, doi: 10.1093/eurheartj/ehz455, indexed in Pubmed: 31504418.
  3. SCORE2 working group, ESC Cardiovascular risk collaboration. SCORE2 risk prediction algorithms: new models to estimate 10-year risk of cardiovascular disease in Europe. Eur Heart J. 2021; 42(25): 243954, doi: 10.1093/eurheartj/ehab309, indexed in Pubmed: 34120177.
  4. Mitkowski P, Witkowski A, Stępińska J, et al. Position of the Polish Cardiac Society on therapeutic targets for LDL cholesterol concentrations in secondary prevention of myocardial infarctions. Kardiol Pol. 2023; 81(7-8): 818823, doi: 10.33963/KP.a2023.0162, indexed in Pubmed: 37489830.
  5. Rallidis LS, Tsamoulis D, Leventis I, et al. Extremely high-risk patients with acute coronary syndrome: How „extreme” should lipid-lowering therapy be if the LDL-C target <40 mg/dl is considered? Kardiol Pol. 2023; 81(10): 10121014, doi: 10.33963/KP.a2023.0147, indexed in Pubmed: 37401581.
  6. Stein LH, Berger J, Tranquilli M, et al. Effect of statin drugs on thoracic aortic aneurysms. Am J Cardiol. 2013; 112(8): 12401245, doi: 10.1016/j.amjcard.2013.05.081, indexed in Pubmed: 24079445.
  7. Golder Su, Weissenbacher D, O’Connor K, et al. Patient-reported reasons for switching or discontinuing statin therapy: A mixed methods study using social media. Drug Saf. 2022; 45(9): 971981, doi: 10.1007/s40264-022-01212-0, indexed in Pubmed: 35933649.
  8. Bradley CK, Wang TY, Li S, et al. Patient-Reported reasons for declining or discontinuing statin therapy: Insights from the PALM registry. J Am Heart Assoc. 2019; 8(7): e011765, doi: 10.1161/JAHA.118.011765, indexed in Pubmed: 30913959.
  9. Jacobson TA, Cheeley MK, Jones PH, et al. The statin adverse treatment experience survey: Experience of patients reporting side effects of statin therapy. J Clin Lipidol. 2019; 13(3): 415424, doi: 10.1016/j.jacl.2019.04.011, indexed in Pubmed: 31113745.
  10. Rosenbaum D, Dallongeville J, Sabouret P, et al. Discontinuation of statin therapy due to muscular side effects: a survey in real life. Nutr Metab Cardiovasc Dis. 2013; 23(9): 871875, doi: 10.1016/j.numecd.2012.04.012, indexed in Pubmed: 22748604.
  11. Cheeley MK, Saseen JJ, Agarwala A, et al. NLA scientific statement on statin intolerance: a new definition and key considerations for ASCVD risk reduction in the statin intolerant patient. J Clin Lipidol. 2022; 16(4): 361375, doi: 10.1016/j.jacl.2022.05.068, indexed in Pubmed: 35718660.
  12. Fitchett DH, Hegele RA, Verma S. Cardiology patient page. Statin intolerance. Circulation. 2015; 131(13): e389e391, doi: 10.1161/CIRCULATIONAHA.114.013189, indexed in Pubmed: 25825402.
  13. Gaine SP, Kulkarni A, Dixon DL, Patel J. NLA 2022 Definition of Statin Intolerance. 2022.
  14. Newman CB, Preiss D, Tobert JA, et al. Statin safety and associated adverse events: A scientific statement from the American Heart Association. Arterioscler Thromb Vasc Biol. 2019; 39(2): e38e81, doi: 10.1161/ATV.0000000000000073, indexed in Pubmed: 30580575.
  15. Bytyçi I, Penson PE, Mikhailidis DP, et al. Prevalence of statin intolerance: A meta-analysis. Eur Heart J. 2022; 43(34): 32133223, doi: 10.1093/eurheartj/ehac015, indexed in Pubmed: 35169843.
  16. Meza-Contreras A, Wenczenovicz C, Ruiz-Arellanos K, et al. Statin intolerance management: A systematic review. Endocrine. 2023; 79(3): 430436, doi: 10.1007/s12020-022-03263-w, indexed in Pubmed: 36459335.
  17. Polaczyk M, Olszanecka A, Wojciechowska W, et al. Multiple drug intolerance in patients with arterial hypertension: prevalence and determining factors. Pol Arch Intern Med. 2023; 133(3), doi: 10.20452/pamw.16399, indexed in Pubmed: 36602061.
  18. Krishnamurthy A, Bradley C, Ascunce R, et al. SAMSON and the nocebo effect: Management of statin intolerance. Curr Cardiol Rep. 2022; 24(9): 11011108, doi: 10.1007/s11886-022-01729-x, indexed in Pubmed: 35759168.
  19. Moon J, Cohen Sedgh R, Jackevicius CA. Examining the nocebo effect of statins through statin adverse events reported in the Food and Drug Administration adverse event reporting system. Circ Cardiovasc Qual Outcomes. 2021; 14(1): e007480, doi: 10.1161/CIRCOUTCOMES.120.007480, indexed in Pubmed: 33161769.
  20. Blasini M, Corsi N, Klinger R, et al. Nocebo and pain: An overview of the psychoneurobiological mechanisms. Pain Rep. 2017; 2(2), doi: 10.1097/PR9.0000000000000585, indexed in Pubmed: 28971165.
  21. Wartolowska K. The nocebo effect as a source of bias in the assessment of treatment effects. F1000Res. 2019; 8: 5, doi: 10.12688/f1000research.17611.2, indexed in Pubmed: 31354941.
  22. Penson PE, Banach M. Nocebo/drucebo effect in statin-intolerant patients: an attempt at recommendations. Eur Heart J. 2021; 42(47): 47874788, doi: 10.1093/eurheartj/ehab358, indexed in Pubmed: 34151941.
  23. Nissen SE. Statin denial: An internet-driven cult with deadly consequences. Ann Intern Med. 2017; 167(4): 281282, doi: 10.7326/M17-1566, indexed in Pubmed: 28738422.
  24. Penson PE, Mancini GB, Toth PP, et al. Introducing the ‚Drucebo’ effect in statin therapy: a systematic review of studies comparing reported rates of statin-associated muscle symptoms, under blinded and open-label conditions. J Cachexia Sarcopenia Muscle. 2018; 9(6): 10231033, doi: 10.1002/jcsm.12344, indexed in Pubmed: 30311434.
  25. Cardiology ACo. American College of Cardiology Statin Intolerance Tool. https://tools.acc.org/StatinIntolerance/?_ga=2.222956114.890717625.1704825019-1126116753.1704041760#!/ (accessed: March 20, 2024).
  26. Mayo Clinic. Statin Choice Decision Aid. https://statindecisionaid.mayoclinic.org/ (accessed: March 20, 2024).
  27. Choudhary A, Rawat U, Kumar P, et al. Pleotropic effects of statins: the dilemma of wider utilization of statin. Egypt Heart J. 2023; 75(1): 1, doi: 10.1186/s43044-023-00327-8, indexed in Pubmed: 36602642.
  28. Oesterle A, Laufs U, Liao JK. Pleiotropic effects of statins on the cardiovascular system. Circ Res. 2017; 120(1): 229243, doi: 10.1161/CIRCRESAHA.116.308537, indexed in Pubmed: 28057795.
  29. Wasim R, Ansari TM, Ahsan F, et al. Pleiotropic benefits of statins in cardiovascular diseases. Drug Res (Stuttg). 2022; 72(9): 477486, doi: 10.1055/a-1873-1978, indexed in Pubmed: 35868336.
  30. Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-induced myopathy a genomewide study. N Engl J Med. 2008; 359(8): 789799, doi: 10.1056/NEJMoa0801936, indexed in Pubmed: 18650507.
  31. Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle symptoms: impact on statin therapy European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J. 2015; 36(17): 10121022, doi: 10.1093/eurheartj/ehv043, indexed in Pubmed: 25694464.
  32. Vassy JL, Gaziano JM, Green RC, et al. Effect of pharmacogenetic testing for statin myopathy risk vs usual care on blood cholesterol: A randomized clinical trial. JAMA Netw Open. 2020; 3(12): e2027092, doi: 10.1001/jamanetworkopen.2020.27092, indexed in Pubmed: 33270123.
  33. Orringer CE, Grant JK, Tokgozoglu L. A review of statin intolerance: A focus on statin-attributed muscle symptoms. Curr Atheroscler Rep. 2022; 24(11): 839847, doi: 10.1007/s11883-022-01059-x, indexed in Pubmed: 36001213.
  34. ASCVD Risk Estimator. https://tools.acc.org/ascvd-risk-estimator/default.aspx (accessed: March 20, 2024).
  35. MESA Risk Score and Coronary Age Calculator. https://internal.mesa-nhlbi.org/about/procedures/tools/mesa-score-risk-calculator (accessed: March 20, 2024).
  36. Astro-CHARM. https://astrocharm.org/calculator-working/ (accessed: March 20, 2024).
  37. EPIC. https://www.epic.com/ (accessed: March 20, 2024).
  38. Gepner AD, Piper ME, Johnson HM, et al. Effects of smoking and smoking cessation on lipids and lipoproteins: outcomes from a randomized clinical trial. Am Heart J. 2011; 161(1): 145151, doi: 10.1016/j.ahj.2010.09.023, indexed in Pubmed: 21167347.
  39. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019; 140(11): e596e646, doi: 10.1161/CIR.0000000000000678, indexed in Pubmed: 30879355.
  40. Liu Z, Tian Z, Zhao D, et al. Effects of coenzyme Q10 supplementation on lipid profiles in adults: A meta-analysis of randomized controlled trials. J Clin Endocrinol Metab. 2022; 108(1): 232249, doi: 10.1210/clinem/dgac585, indexed in Pubmed: 36337001.
  41. Ghodeshwar GK, Dube A, Khobragade D. Impact of lifestyle modifications on cardiovascular health: A narrative review. Cureus. 2023; 15(7): e42616, doi: 10.7759/cureus.42616, indexed in Pubmed: 37641769.
  42. Ma ZY, Yang SP, Li Y, et al. Associations between the use of red yeast rice preparations and adverse health outcomes: An umbrella review of meta-analyses of randomized controlled trials. J Integr Med. 2024; 22(2): 126136, doi: 10.1016/j.joim.2024.01.008, indexed in Pubmed: 38413255.
  43. Laffin LJ, Bruemmer D, Garcia M, et al. Comparative effects of low-dose rosuvastatin, placebo, and dietary supplements on lipids and inflammatory biomarkers. J Am Coll Cardiol. 2023; 81(1): 112, doi: 10.1016/j.jacc.2022.10.013, indexed in Pubmed: 36351465.
  44. Reiner Z. Resistance and intolerance to statins. Nutr Metab Cardiovasc Dis. 2014; 24(10): 10571066, doi: 10.1016/j.numecd.2014.05.009, indexed in Pubmed: 24996502.
  45. Banach M, Rizzo M, Toth PP, et al. Statin intolerance an attempt at a unified definition. Position paper from an International Lipid Expert Panel. Arch Med Sci. 2015; 11(1): 123, doi: 10.5114/aoms.2015.49807, indexed in Pubmed: 25861286.
  46. Nicholls SJ. PCSK9 inhibitors and reduction in cardiovascular events: Current evidence and future perspectives. Kardiol Pol. 2023; 81(2): 115122, doi: 10.33963/KP.a2023.0030, indexed in Pubmed: 36739653.
  47. Nissen SE, Menon V, Nicholls SJ, et al. Bempedoic acid and cardiovascular outcomes in statin-intolerant patients. N Engl J Med. 2023; 388(15): 13531364, doi: 10.1056/NEJMoa2215024, indexed in Pubmed: 36876740.
  48. Raschi E, Casula M, Cicero AFG, et al. Beyond statins: New pharmacological targets to decrease LDL-cholesterol and cardiovascular events. Pharmacol Ther. 2023; 250: 108507, doi: 10.1016/j.pharmthera.2023.108507, indexed in Pubmed: 37567512.
  49. Singh A, Cho LS. Nonstatin therapy to reduce low-density lipoprotein cholesterol and improve cardiovascular outcomes. Cleve Clin J Med. 2024; 91(1): 5363, doi: 10.3949/ccjm.91a.23058, indexed in Pubmed: 38167398.
  50. Anagnostis P, Antza C, Florentin M, et al. Familial hypercholesterolemia and its manifestations: Practical considerations for general practitioners. Kardiol Pol. 2023; 81(11): 10811088, doi: 10.33963/v.kp.97845, indexed in Pubmed: 37937357.
  51. Morrone D, Weintraub WS, Toth PP, et al. Lipid-altering efficacy of ezetimibe plus statin and statin monotherapy and identification of factors associated with treatment response: a pooled analysis of over 21,000 subjects from 27 clinical trials. Atherosclerosis. 2012; 223(2): 251261, doi: 10.1016/j.atherosclerosis.2012.02.016, indexed in Pubmed: 22410123.