1
|
Sun X, Bai S, Wu H, Wang T, Du R. Administration of Evolocumab in Patients with STEMI After Emergency PCI: A Real-World Cohort Study. Am J Cardiovasc Drugs 2025:10.1007/s40256-025-00722-3. [PMID: 39992584 DOI: 10.1007/s40256-025-00722-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND AND OBJECTIVE Evolocumab can reduce low-density lipoprotein cholesterol (LDL-C) levels and improve cardiovascular (CV) outcomes. While its benefits are well established in broader populations, its potential impact on patients with ST-segment elevation myocardial infarction (STEMI) undergoing emergency percutaneous coronary intervention (PCI) remains underexplored, particularly in real-world settings. This study aimed to evaluate its efficacy and safety in this specific patient group on the basis of real-world clinical experience. METHODS A total of 384 patients with STEMI who underwent emergency PCI at Hebei General Hospital between 1 July 2021 and 23 September 2022 were enrolled in this retrospective, single-center study. Of these, 85 patients received evolocumab (140 mg every 2 weeks) plus standard of care (SOC), while 299 received SOC alone. Patients were monitored for CV events and lipid levels during follow-up. Propensity score matching (PSM) and inverse probability treatment weighting (IPTW) were used to balance covariates. RESULTS The experimental group had a lower cumulative incidence of the primary composite endpoint over 18 months in the unadjusted analysis (hazard ratio [HR] = 0.353; 95% confidence interval [CI] 0.180-0.693; P = 0.002), as well as after adjustment for PSM (HR = 0.341; 95% CI 0.165-0.706; P = 0.004) and IPTW (HR = 0.461; 95% CI 0.241-0.881; P = 0.019). The 18-month cumulative incidence was 10 (12%) for evolocumab + SOC and 95 (32%) for SOC. LDL-C levels in the evolocumab + SOC group showed significant reductions across different cohorts, compared with the SOC group. No significant differences in adverse events were observed between the two groups. CONCLUSIONS Evolocumab plus SOC significantly reduced postoperative CV events and LDL-C levels in patients with STEMI after emergency PCI.
Collapse
Affiliation(s)
- Xuefeng Sun
- Graduate School of Hebei Medical University, Shijiazhuang, China
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Shiru Bai
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Haibo Wu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Tingting Wang
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Rongpin Du
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China.
| |
Collapse
|
2
|
Brobst M, Chapet N, Benchalkha D, Bourgeois E, Herman F, Molinari N, Leclercq F, Pasquié JL, Breuker C, Sultan A, Roubille F. Impact of obesity on tolerance and persistence of statins in patients within 3months following an acute myocardial infarction: A real-world study. Arch Cardiovasc Dis 2025; 118:85-92. [PMID: 39732563 DOI: 10.1016/j.acvd.2024.10.332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Recommended treatment after acute coronary syndrome (ACS) involves high-intensity statin therapy to achieve the low-density lipoprotein (LDL-C) target of<1.4mmol/L (European guidelines), but many patients discontinue statins because of real or perceived side-effects. Whether body mass index (BMI) influences statin intolerance remains unclear. AIM To assess statin tolerance 3months after initiation, and to identify factors determining tolerance and persistence. METHODS STATIC was a single-centre cohort study (November 2021 to April 2023) of patients admitted to cardiac intensive care units for ACS. The study had three stages: T0 (admission); W6 (6 weeks after ACS: statin efficiency); and M3 (3months after ACS: statin tolerance and persistence). SAMS score was used to evaluate imputability in patients reporting muscular side-effects. Multivariable analysis identified factors influencing tolerance; statin persistence was assessed using pharmacy dispensing data. RESULTS Overall, 289 patients were included (77.9% men; mean age 64.2years; 22.7% with BMI≥30kg/m2). At T0, 38.1% had hypertension, 28.5% dyslipidaemia and 15.9% diabetes. At discharge, 269 patients received statins: 97.0% had a high-intensity statin; 43.5% had a statin/ezetimibe combination. At W6, mean LDL-C was 1.58mmol/L, with 45.5% at the LDL-C target. At M3, 6.0% reported side-effects (3.6% muscular, 1.2% liver, 1.2% gastrointestinal). Mean SAMS score was 5.67. No significant differences in muscular or hepatic side-effects were found between patients with BMI≥30 versus<30 kg/m2. Persistence was 98.4% at M3 follow-up. The proportion of patients on a high-intensity statin or a statin/ezetimibe did not change from discharge to M3 (P=0.45 and P=1.00, respectively). CONCLUSIONS Statins are effective, but not always enough to reach LDL-C target. Tolerance and persistence were good, with muscular side-effects as expected, but without any guarantee of statin imputability. BMI did not influence statin tolerance in this study.
Collapse
Affiliation(s)
- Morgane Brobst
- Department of Pharmacy, CHU Montpellier, 34295 Montpellier, France.
| | - Nicolas Chapet
- Department of Pharmacy, CHU Montpellier, 34295 Montpellier, France
| | | | - Elise Bourgeois
- Department of Endocrinology, CHU Montpellier, 34295 Montpellier, France
| | - Fanchon Herman
- Department of Medical Information, CHU Montpellier, 34295 Montpellier, France
| | - Nicolas Molinari
- Department of Medical Information, CHU Montpellier, 34295 Montpellier, France
| | - Florence Leclercq
- Department of Cardiology, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Luc Pasquié
- Department of Cardiology, CHU Montpellier, 34295 Montpellier, France
| | - Cyril Breuker
- Department of Pharmacy, CHU Montpellier, 34295 Montpellier, France
| | - Ariane Sultan
- Department of Endocrinology, CHU Montpellier, 34295 Montpellier, France
| | - François Roubille
- Department of Cardiology, CHU Montpellier, 34295 Montpellier, France
| |
Collapse
|
3
|
Ward NC, Reid CM, Watts GF. Low-density lipoprotein-cholesterol lowering effect of a nutraceutical regimen with or without ezetimibe in hypercholesterolaemic patients with statin intolerance. Front Cardiovasc Med 2022; 9:1060252. [PMID: 36505352 PMCID: PMC9732015 DOI: 10.3389/fcvm.2022.1060252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
Abstract
Background Statins are the most widely prescribed medication to lower low-density lipoprotein cholesterol (LDL-c). However, a significant portion of patients are unable to tolerate them due to side effects, most commonly muscle related. Nutraceuticals, natural plant derivatives with lipid-lowering properties, may provide an alternative to lower LDL-c in these patients. Aims To investigate whether a nutraceutical regimen, either alone or in combination with ezetimibe, can lower LDL-c in patients with hypercholesterolemia who are intolerant to statins. Methods Participants were recruited into a double-blind, randomized, placebo-controlled intervention study. Treatments were (i) placebo, (ii) nutraceutical (500 mg berberine, 200 mg red yeast rice (RYR), 2 g plant sterols)/daily, (iii) ezetimibe (10 mg)/daily, or (iv) the combination of nutraceutical and ezetimibe/daily. At baseline and week 8, all participants provide a fasting blood sample for assessment of lipid profile and safety bloods. Results Fifty participants were randomized, with 44 completing the treatment period. Following adjustment for baseline levels and compared with placebo, LDL-c was significantly reduced (all p < 0.0001) with ezetimibe (-1.02 mmol/L), nutraceutical (-1.15 mmol/L) and the nutraceutical and ezetimibe combination (-1.92 mmol/L). Non-HDL cholesterol was significantly reduced (all p < 0.0001) with ezetimibe (-1.29 mmol/L), nutraceutical (-1.37 mmol/L) and the nutraceutical and ezetimibe combination (-2.18 mmol/L). Remnant cholesterol and triglycerides was significantly reduced with the nutraceutical and ezetimibe combination (p = 0.018). Conclusion A nutraceutical regimen (berberine, RYR and plant sterols) and ezetimibe independently and additively lower LDL-c in patients with hypercholesterolemia who are intolerant to statins.
Collapse
Affiliation(s)
- Natalie C. Ward
- Dobney Hypertension Centre, Medical School, University of Western Australia, Perth, WA, Australia,*Correspondence: Natalie C. Ward,
| | | | - Gerald F. Watts
- Medical School, University of Western Australia, Perth, WA, Australia,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, WA, Australia
| |
Collapse
|
4
|
Abstract
This article reviews the safety of statins and non-statin medications for management of dyslipidemia. Statins have uncommon serious adverse effects: myopathy/ rhabdomyolysis, which resolve with statin discontinuation, and diabetes, usually in people with risk factors for diabetes. The CVD benefit of statins far exceeds the risk of diabetes. Statin myalgia, without CK elevation, is likely caused by muscle symptoms with another etiology, or the nocebo effect. Notable adverse effects of non-statin medicines include injection site reactions (alirocumab, evolocumab, inclisiran), increased uric acid and gout (bempedoic acid), atrial fibrillation/flutter (omega-3-fatty acids), and myopathy in combination with a statin (gemfibrozil).
Collapse
Affiliation(s)
- Connie B Newman
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, 435 East 30th street, Sixth floor, New York, NY 10016, USA.
| |
Collapse
|
5
|
Wiggins BS, Backes JM, Hilleman D. Statin-associated muscle symptoms-A review: Individualizing the approach to optimize care. Pharmacotherapy 2022; 42:428-438. [PMID: 35388918 DOI: 10.1002/phar.2681] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, also known as "statins" are considered first-line pharmacologic therapy for reducing low-density lipoprotein cholesterol (LDL-C). They have been demonstrated efficacy in a variety of patients populations to reduce atherosclerotic cardiovascular disease (ASCVD) risk. Like any pharmacologic therapy, however, they are not without possible adverse effects that can lead to discontinuation, thus leading to a loss of benefit. The most common side effect related to statin therapy impacting compliance is musculoskeletal related, commonly referred to as statin-associated muscle systems (SAMS). While the overall incidence is relatively low, the consequences of nonadherence to statin therapy can have a negative impact on patient care. Therefore, it is important for healthcare providers to understand risk factors, how to diagnose, and how to manage this unfortunate adverse effect in order to optimize care.
Collapse
Affiliation(s)
- Barbara S Wiggins
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - James M Backes
- University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| | - Daniel Hilleman
- Creighton University School of Pharmacy and Health Professions, Omaha, Nebraska, USA
| |
Collapse
|
6
|
Laufs U, Ballantyne CM, Banach M, Bays H, Catapano AL, Duell PB, Goldberg AC, Gotto AM, Leiter LA, Ray KK, Bloedon LT, MacDougall D, Zhang Y, Mancini GBJ. Efficacy and safety of bempedoic acid in patients not receiving statins in phase 3 clinical trials. J Clin Lipidol 2022; 16:286-297. [DOI: 10.1016/j.jacl.2022.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 02/03/2023]
|
7
|
Padidela R, Whyte MP, Glorieux FH, Munns CF, Ward LM, Nilsson O, Portale AA, Simmons JH, Namba N, Cheong HI, Pitukcheewanont P, Sochett E, Högler W, Muroya K, Tanaka H, Gottesman GS, Biggin A, Perwad F, Williams A, Nixon A, Sun W, Chen A, Skrinar A, Imel EA. Patient-Reported Outcomes from a Randomized, Active-Controlled, Open-Label, Phase 3 Trial of Burosumab Versus Conventional Therapy in Children with X-Linked Hypophosphatemia. Calcif Tissue Int 2021; 108:622-633. [PMID: 33484279 PMCID: PMC8064984 DOI: 10.1007/s00223-020-00797-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
Changing to burosumab, a monoclonal antibody targeting fibroblast growth factor 23, significantly improved phosphorus homeostasis, rickets, lower-extremity deformities, mobility, and growth versus continuing oral phosphate and active vitamin D (conventional therapy) in a randomized, open-label, phase 3 trial involving children aged 1-12 years with X-linked hypophosphatemia. Patients were randomized (1:1) to subcutaneous burosumab or to continue conventional therapy. We present patient-reported outcomes (PROs) from this trial for children aged ≥ 5 years at screening (n = 35), using a Patient-Reported Outcomes Measurement Information System (PROMIS) questionnaire and SF-10 Health Survey for Children. PROMIS pain interference, physical function mobility, and fatigue scores improved from baseline with burosumab at weeks 40 and 64, but changed little with continued conventional therapy. Pain interference scores differed significantly between groups at week 40 (- 5.02, 95% CI - 9.29 to - 0.75; p = 0.0212) but not at week 64. Between-group differences were not significant at either week for physical function mobility or fatigue. Reductions in PROMIS pain interference and fatigue scores from baseline were clinically meaningful with burosumab at weeks 40 and 64 but not with conventional therapy. SF-10 physical health scores (PHS-10) improved significantly with burosumab at week 40 (least-squares mean [standard error] + 5.98 [1.79]; p = 0.0008) and week 64 (+ 5.93 [1.88]; p = 0.0016) but not with conventional therapy (between-treatment differences were nonsignificant). In conclusion, changing to burosumab improved PRO measures, with statistically significant differences in PROMIS pain interference at week 40 versus continuing with conventional therapy and in PHS-10 at weeks 40 and 64 versus baseline.Trial registration: ClinicalTrials.gov NCT02915705.
Collapse
Affiliation(s)
- Raja Padidela
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, UK.
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Michael P Whyte
- Shriners Hospitals for Children -Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Francis H Glorieux
- Shriners Hospital for Children - Canada, McGill University, Montreal, QC, Canada
| | - Craig F Munns
- The University of Sydney Children's Hospital Westmead Clinical School, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Department of Endocrinology, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Leanne M Ward
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Ola Nilsson
- Division of Pediatric Endocrinology & Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Anthony A Portale
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jill H Simmons
- Departments of Pediatrics, Division of Endocrinology and Diabetes, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Noriyuki Namba
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hae Il Cheong
- Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Pisit Pitukcheewanont
- Center of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Etienne Sochett
- Department of Paediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Koji Muroya
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroyuki Tanaka
- Okayama Saiseikai General Hospital Outpatient Center, Okayama, Japan
| | | | - Andrew Biggin
- The University of Sydney Children's Hospital Westmead Clinical School, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Farzana Perwad
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Wei Sun
- Kyowa Kirin Pharmaceutical Development, Princeton, NJ, USA
| | - Angel Chen
- Ultragenyx Pharmaceutical, Novato, CA, USA
| | | | - Erik A Imel
- Department of Medicine and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
8
|
Ödesjö H, Björck S, Franzén S, Hjerpe P, Manhem K, Rosengren A, Thorn J, Adamsson Eryd S. Adherence to lipid-lowering guidelines for secondary prevention and potential reduction in CVD events in Swedish primary care: a cross-sectional study. BMJ Open 2020; 10:e036920. [PMID: 33039993 PMCID: PMC7549446 DOI: 10.1136/bmjopen-2020-036920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The protective effect of lipid-lowering treatment for secondary prevention after coronary heart disease (CHD) has been well documented. Current guidelines recommend a target level for low-density lipoprotein cholesterol (LDL-C) of ≤1.8 mmol/L. The aim was to describe lipid-lowering treatment patterns and to provide an estimate of the potential reductions in cardiovascular disease (CVD) events with improved adherence to guidelines. DESIGN Cross-sectional. SETTING Primary care in a large Swedish region. PARTICIPANTS 37 120 patients with CHD in a Swedish regional primary care quality register (QregPV), by 31 December 2015. PRIMARY AND SECONDARY OUTCOME MEASURES Proportion of patients on statin treatment and proportion of patients achieving LDL-C ≤1.8 mmol/L. Estimated number of CVD events calculated for (1) current treatment, (2) improved treatment and (3) lowered LDL-C, based on applying rate reductions from meta-analyses of randomised trials to the potentially undertreated population. Risk estimation modelling was based on 52 042 patients in the same register on January 2011 followed for 5 years. RESULTS Of 37 120 patients, 18% reached LDL-C ≤1.8 mmol/L and 32% were not on statin treatment. Based on individual risks, the estimated number of CVD events in the study group over 5 years was 9209/37 120. If all patients without a statin or with less potent statin treatment were given atorvastatin 80 mg, an estimated reduction of CVD events by 14% (7901 vs 9209) was seen. If all patients achieved LDL-C ≤1.8 mmol/L, the number of events was estimated to be reduced by 18% (7577 vs 9209). CONCLUSION One-third of patients with CHD in primary care were not on lipid-lowering treatment. Based on the assumption that included patients would react to statin therapy the same way as the patients in randomised trials, improved adherence to treatment guidelines could lead to a substantial reduction in new CVD events.
Collapse
Affiliation(s)
- Helena Ödesjö
- Primary Health Care, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Per Hjerpe
- Primary Health Care, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Manhem
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jörgen Thorn
- Primary Health Care, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Samuel Adamsson Eryd
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
De Luca M, Iacono O, Lucci R, Guardasole V, Bosso G, Cittadini A, Oliviero U. Atorvastatin-linked rhabdomyolysis caused by the simultaneous intake of amoxicillin clavulanic acid. J Basic Clin Physiol Pharmacol 2020; 32:/j/jbcpp.ahead-of-print/jbcpp-2020-0108/jbcpp-2020-0108.xml. [PMID: 32903207 DOI: 10.1515/jbcpp-2020-0108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/12/2020] [Indexed: 11/15/2022]
Abstract
Objectives Rhabdomyolysis is a rare syndrome in which a serious muscle damage suddenly appears, with the possible occurrence of severe complications such as kidney failure, electrolyte imbalances and death, and represents the most severe form of statin-induced muscle injury. Case presentation Here we present the case of a 55-year-old woman who started therapy with amoxicillin clavulanic acid on a background of atorvastatin therapy, resulting in rhabdomyolysis. Conclusions This case highlights the importance of evaluating potential drug interactions in patients taking statin and the need of monitoring clinical and laboratory findings suggestive of rhabdomyolysis.
Collapse
Affiliation(s)
- Mariarosaria De Luca
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| | - Olimpia Iacono
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| | - Rosa Lucci
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| | - Vincenzo Guardasole
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| | - Giorgio Bosso
- Santa Maria delle Grazie Hospital, Via Domitiana, Pozzuoli, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| | - Ugo Oliviero
- Department of Translational Medical Sciences, University Federico II, Via Sergio Pansini 5, Naples, Italy
| |
Collapse
|
10
|
Lipid-Modifying Agents, From Statins to PCSK9 Inhibitors. J Am Coll Cardiol 2020; 75:1945-1955. [DOI: 10.1016/j.jacc.2019.11.072] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
|
11
|
Pergolizzi JV, Coluzzi F, Colucci RD, Olsson H, LeQuang JA, Al-Saadi J, Magnusson P. Statins and muscle pain. Expert Rev Clin Pharmacol 2020; 13:299-310. [PMID: 32089020 DOI: 10.1080/17512433.2020.1734451] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Statins remain among the most frequently prescribed drugs and constitute a cornerstone in the prevention of cardiovascular disease. However, muscle symptoms are often reported from patients on statins. Muscle symptoms are frequently reported as adverse events associated with statin therapy.Areas covered: In the present narrative review, statin-associated muscle pain is discussed. It elucidates potential mechanisms and possible targets for management.Expert opinion: In general, the evidence in support of muscle pain caused by statins is in some cases equivocal and not particularly strong. Reported symptoms are difficult to quantify. Rarely is it possible to establish a causal link between statins and muscle pain. In randomized controlled trials, statins are well tolerated, and muscle-pain related side-effects is similar to placebo. There are also nocebo effects of statins. Exchange of statin may be beneficial although all statins have been associated with muscle pain. In some patients reduction of dose is worth trying, especially in primary prevention Although the benefits of statins outweigh potential risks in the vast majority of cases, careful clinical judgment may be necessary in certain cases to manage potential side effects on an individual basis.
Collapse
Affiliation(s)
| | - Flaminia Coluzzi
- Department of Medical and Surgical Sciences and Biotechnologies, Unit of Anaesthesia, Intensive Care and Pain Medicine, Sapienza University of Rome, Rome, Italy
| | - Robert D Colucci
- NEMA Research, Inc., Naples, FL, USA.,Colucci & Associates, LLC, Newtown, Connecticut, USA
| | - Hanna Olsson
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden
| | | | - Jonathan Al-Saadi
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden
| | - Peter Magnusson
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden.,Cardiology Research Unit, Institution of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Khattib A, Atrahimovich D, Dahli L, Vaya J, Khatib S. Lyso-diacylglyceryltrimethylhomoserine (lyso-DGTS) isolated from Nannochloropsis microalgae improves high-density lipoprotein (HDL) functions. Biofactors 2020; 46:146-157. [PMID: 31660677 DOI: 10.1002/biof.1580] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Many population studies have shown that blood concentrations of high-density lipoprotein (HDL) cholesterol are inversely correlated with risk of cardiovascular disease (CVD). However, in recent studies, increasing blood HDL cholesterol concentrations failed to reduce CVD events. On the other hand, studies suggest that improving HDL quality can be a more efficient tool for assessing atherosclerotic risk than simply measuring blood HDL cholesterol concentration. Thus, improving HDL activity using natural substances might be a useful therapeutic approach to reducing CVD risk. We previously isolated a novel active compound from Nannochloropsis microalgae termed lyso-diacylglyceryltrimethylhomoserine (lyso-DGTS), which increased activity of paraoxonase 1, the main antioxidant enzyme associated with HDL. Here we examined the effect of lyso-DGTS on HDL quality and function. Tryptophan-fluorescence-quenching assay showed that lyso-DGTS interacts spontaneously with the entire HDL lipoprotein and with apolipoprotein A1 (ApoA1), the major structural and functional HDL protein, with high affinity (Ka = 2.17 × 104 M-1 at 37°C). Lyso-DGTS added to HDL and to ApoA1 increased cholesterol efflux from macrophage cells, the main antiatherogenic function of HDL, dose-dependently, and significantly increased HDL's ability to induce nitric oxide production from endothelial cells. In-vivo supplementation of lyso-DGTS to the circulation of mice fed a high-fat diet via osmotic mini-pumps implanted subcutaneously enhanced HDL anti-inflammatory effect significantly as compared to controls. Our findings suggest that lyso-DGTS may have a beneficial effect in decreasing atherosclerosis risk by interacting with HDL particles and improving their quality and antiatherogenic functions.
Collapse
Affiliation(s)
- Ali Khattib
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Tel-Hai College, Upper Galilee, Israel
| | - Dana Atrahimovich
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | - Loureen Dahli
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Tel-Hai College, Upper Galilee, Israel
| | - Jacob Vaya
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Tel-Hai College, Upper Galilee, Israel
| | - Soliman Khatib
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Tel-Hai College, Upper Galilee, Israel
| |
Collapse
|
13
|
Newman CB, Preiss D, Tobert JA, Jacobson TA, Page RL, Goldstein LB, Chin C, Tannock LR, Miller M, Raghuveer G, Duell PB, Brinton EA, Pollak A, Braun LT, Welty FK. Statin Safety and Associated Adverse Events: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2019; 39:e38-e81. [PMID: 30580575 DOI: 10.1161/atv.0000000000000073] [Citation(s) in RCA: 440] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One in 4 Americans >40 years of age takes a statin to reduce the risk of myocardial infarction, ischemic stroke, and other complications of atherosclerotic disease. The most effective statins produce a mean reduction in low-density lipoprotein cholesterol of 55% to 60% at the maximum dosage, and 6 of the 7 marketed statins are available in generic form, which makes them affordable for most patients. Primarily using data from randomized controlled trials, supplemented with observational data where necessary, this scientific statement provides a comprehensive review of statin safety and tolerability. The review covers the general patient population, as well as demographic subgroups, including the elderly, children, pregnant women, East Asians, and patients with specific conditions such as chronic disease of the kidney and liver, human immunodeficiency viral infection, and organ transplants. The risk of statin-induced serious muscle injury, including rhabdomyolysis, is <0.1%, and the risk of serious hepatotoxicity is ≈0.001%. The risk of statin-induced newly diagnosed diabetes mellitus is ≈0.2% per year of treatment, depending on the underlying risk of diabetes mellitus in the population studied. In patients with cerebrovascular disease, statins possibly increase the risk of hemorrhagic stroke; however, they clearly produce a greater reduction in the risk of atherothrombotic stroke and thus total stroke, as well as other cardiovascular events. There is no convincing evidence for a causal relationship between statins and cancer, cataracts, cognitive dysfunction, peripheral neuropathy, erectile dysfunction, or tendonitis. In US clinical practices, roughly 10% of patients stop taking a statin because of subjective complaints, most commonly muscle symptoms without raised creatine kinase. In contrast, in randomized clinical trials, the difference in the incidence of muscle symptoms without significantly raised creatinine kinase in statin-treated compared with placebo-treated participants is <1%, and it is even smaller (0.1%) for patients who discontinued treatment because of such muscle symptoms. This suggests that muscle symptoms are usually not caused by pharmacological effects of the statin. Restarting statin therapy in these patients can be challenging, but it is important, especially in patients at high risk of cardiovascular events, for whom prevention of these events is a priority. Overall, in patients for whom statin treatment is recommended by current guidelines, the benefits greatly outweigh the risks.
Collapse
|
14
|
Abstract
There is now overwhelming evidence to support lowering LDL-c (low-density lipoprotein cholesterol) to reduce cardiovascular morbidity and mortality. Statins are a class of drugs frequently prescribed to lower cholesterol. However, in spite of their wide-spread use, discontinuation and nonadherence remains a major gap in both the primary and secondary prevention of atherosclerotic cardiovascular disease. The major reason for statin discontinuation is because of the development of statin-associated muscle symptoms, but a range of other statin-induced side effects also exist. Although the mechanisms behind these side effects have not been fully elucidated, there is an urgent need to identify those at increased risk of developing side effects as well as provide alternative treatment strategies. In this article, we review the mechanisms and clinical importance of statin toxicity and focus on the evaluation and management of statin-associated muscle symptoms.
Collapse
Affiliation(s)
- Natalie C Ward
- From the School of Public Health, Curtin University, Perth, Western Australia, Australia (N.C.W.).,School of Medicine, University of Western Australia, Perth, Australia (N.C.W., G.F.W.)
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia (N.C.W., G.F.W.).,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Western Australia, Australia (G.F.W.)
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora (R.H.E.)
| |
Collapse
|
15
|
Daniel H, Christian W, Robin H, Lars S, Thomas M. Statin treatment after acute coronary syndrome: Adherence and reasons for non-adherence in a randomized controlled intervention trial. Sci Rep 2019; 9:12079. [PMID: 31427637 PMCID: PMC6700346 DOI: 10.1038/s41598-019-48540-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Studies of secondary prevention for cardiovascular disease show low fulfilment of guideline-recommended targets. This study explored whether nurse-led follow-up could increase adherence to statins over time and reasons for discontinuation. All patients admitted for acute coronary syndrome at Östersund hospital between 2010-2014 were screened for the randomized controlled NAILED-ACS trial. The trial comprises two groups, one with nurse-led annual follow-up and medical titration by telephone to reach set intervention targets and one with usual care. All discontinuations of statins were recorded prospectively for at least 36 months and categorized as avoidable or unavoidable. Kaplan-Meier estimates were conducted for first and permanent discontinuations. Predictors for discontinuation were analysed using multivariate Cox regression, statin type and mean LDL-C at end of follow-up. Female gender was a predictor for discontinuation. Allocation in the intervention group predicted increased risk for a first but decreased risk for permanent discontinuation. A nurse-led telemedical secondary prevention programme in a relatively unselected ACS cohort leads to increased adherence to statins over time, greater percentage on potent treatment and lower LDL-C compared to usual care. An initially increased tendency toward early discontinuation in the intervention group stresses the importance of a longer duration of structured follow-up.
Collapse
Affiliation(s)
- Huber Daniel
- Department of Public Health and Clinical Medicine, Unit of Research, Education and Development - Östersund, Umeå University, Östersund, Sweden.
| | - Wikén Christian
- Department of Public Health and Clinical Medicine, Unit of Research, Education and Development - Östersund, Umeå University, Östersund, Sweden
| | - Henriksson Robin
- Department of Public Health and Clinical Medicine, Unit of Research, Education and Development - Östersund, Umeå University, Östersund, Sweden
| | - Söderström Lars
- Unit of Research, Development and Education, Region Jämtland Härjedalen, Östersund Hospital, Östersund, Sweden
| | - Mooe Thomas
- Department of Public Health and Clinical Medicine, Unit of Research, Education and Development - Östersund, Umeå University, Östersund, Sweden
| |
Collapse
|
16
|
Vonbank A, Drexel H, Agewall S, Lewis BS, Dopheide JF, Kjeldsen K, Ceconi C, Savarese G, Rosano G, Wassmann S, Niessner A, Schmidt TA, Saely CH, Baumgartner I, Tamargo J. Reasons for disparity in statin adherence rates between clinical trials and real-world observations: a review. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2019; 4:230-236. [PMID: 30099530 DOI: 10.1093/ehjcvp/pvy028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/06/2018] [Indexed: 11/14/2022]
Abstract
With statins, the reported rate of adverse events differs widely between randomized clinical trials (RCTs) and observations in clinical practice, the rates being 1-2% in RCTs vs. 10-20% in the so-called real world. One possible explanation is the claim that RCTs mostly use a run-in period with a statin. This would exclude intolerant patients from remaining in the trial and therefore favour a bias towards lower rates of intolerance. We here review data from RCTs with more than 1000 participants with and without a run-in period, which were included in the Cholesterol Treatment Trialists Collaboration. Two major conclusions arise: (i) the majority of RCTs did not have a test dose of a statin in the run-in phase. (ii) A test dose in the run-in phase was not associated with a significantly improved adherence rate within that trial when compared to trials without a test dose. Taken together, the RCTs of statins reviewed here do not suggest a bias towards an artificially higher adherence rate because of a run-in period with a test dose of the statin. Other possible explanations for the apparent disparity between RCTs and real-world observations are also included in this review albeit mostly not supported by scientific data.
Collapse
Affiliation(s)
- Alexander Vonbank
- Department of Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Liechtenstein.,Drexel University College of Medicine, Philadelphia, PA, USA.,Division of Angiology, Swiss Cardiovascular Center, University Hospital Bern, Switzerland
| | - Stefan Agewall
- Department of Cardiology, Ullevål, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Sciences, Søsterhjemmet, University of Oslo, Oslo, Norway
| | - Basil S Lewis
- Lady Davis Carmel Medical Center, Haifa, Israel.,Technion-Israel Institute of Technology, Ruth and Bruce Rappaport School of Medicine, Haifa, Israel
| | - Joern F Dopheide
- Division of Angiology, Swiss Cardiovascular Center, University Hospital Bern, Switzerland
| | - Keld Kjeldsen
- Division of Cardiology, Department of Medicine, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Claudio Ceconi
- Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Gianluigi Savarese
- Department of Medicine, Cardiology Division, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Giuseppe Rosano
- Department of Medical Sciences, Irccs San Raffaele Hospital, Rome, Italy
| | - Sven Wassmann
- Cardiology Pasing, Munich, Germany and University of the Saarland, Homburg, Saar, Germany
| | - Alexander Niessner
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Thomas Andersen Schmidt
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Department of Emergency Medicine, Holbaek Hospital, University of Copenhagen, Denmark
| | - Christoph H Saely
- Department of Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Liechtenstein.,Division of Angiology, Swiss Cardiovascular Center, University Hospital Bern, Switzerland
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, University Hospital Bern, Switzerland
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Ciudad Universitaria, Madrid, Spain
| |
Collapse
|
17
|
Dahli L, Atrahimovich D, Vaya J, Khatib S. Lyso-DGTS lipid isolated from microalgae enhances PON1 activities in vitro and in vivo, increases PON1 penetration into macrophages and decreases cellular lipid accumulation. Biofactors 2018; 44:299-310. [PMID: 29659105 DOI: 10.1002/biof.1427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
High-density lipoprotein (HDL) plays an important role in preventing atherosclerosis. The antioxidant effect of HDL is mostly associated with paraoxonase 1 (PON1) activity. Increasing PON1 activity using nutrients might improve HDL function and quality and thus, decrease atherosclerotic risk. We previously isolated and identified a novel active compound, lyso-DGTS (C20:5,0) from Nannochloropsis sp. ethanol extract. In the present study, its effect on PON1 activities was examined and the mechanism by which the compound affects PON1 activity was explored. Lyso-DGTS elevated recombinant PON1 (rePON1) lactonase and esterase activities in a dose- and time-responsive manner, and further stabilized and preserved rePON1 lactonase activity. Incubation of lyso-DGTS with human serum for 4 h at 37 °C also increased PON1 lactonase activity in a dose-responsive manner. Using tryptophan-fluorescence-quenching assay, lyso-DGTS was found to interact with rePON1 spontaneously with negative free energy (ΔG = -22.87 kJ mol-1 at 25 °C). Thermodynamic parameters and molecular modeling calculations showed that the main interaction of lyso-DGTS with the enzyme is through a hydrogen bond with supporting van der Waals interactions. Furthermore, lyso-DGTS significantly increased rePON1 influx into macrophages and prevented lipid accumulation in macrophages stimulated with oxidized low-density lipid dose-dependently. In vivo supplementation of lyso-DGTS to the circulation of mice fed a high-fat diet via osmotic mini-pumps implanted subcutaneously significantly increased serum PON1 lactonase activity and decreased serum glucose concentrations to the level of mice fed a normal diet. Our findings suggest a beneficial effect of lyso-DGTS on increasing PON1 activity and thus, improving HDL quality and atherosclerotic risk factors. © 2018 BioFactors, 44(3):299-310, 2018.
Collapse
Affiliation(s)
- Loureen Dahli
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel
| | - Dana Atrahimovich
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona 11016, Israel
| | - Jacob Vaya
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel
| | - Soliman Khatib
- Department of Oxidative Stress and Human Diseases, MIGAL-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel
| |
Collapse
|
18
|
Irwin JC, Khalesi S, Fenning AS, Vella RK. The effect of lipophilicity and dose on the frequency of statin-associated muscle symptoms: A systematic review and meta-analysis. Pharmacol Res 2018; 128:264-273. [DOI: 10.1016/j.phrs.2017.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
|
19
|
Jacobson TA, Khan A, Maki KC, Brinton EA, Cohen JD. Provider recommendations for patient-reported muscle symptoms on statin therapy: Insights from the Understanding Statin Use in America and Gaps in Patient Education survey. J Clin Lipidol 2018; 12:78-88. [DOI: 10.1016/j.jacl.2017.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/26/2017] [Accepted: 09/19/2017] [Indexed: 01/24/2023]
|
20
|
Melendez QM, Wooten CJ, Lopez D. Atorvastatin and lovastatin, but not pravastatin, increased cellular complex formation between PCSK9 and the LDL receptor in human hepatocyte-like C3A cells. Biochem Biophys Res Commun 2017; 492:103-108. [PMID: 28802576 DOI: 10.1016/j.bbrc.2017.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
Statins are the first-line treatment for hypercholesterolemic patients. Herein, the effects of three statins on complex formation between proprotein convertase subtilisin-kexin 9 (PCSK9) and the low density lipoprotein receptor (LDLR), a critical step for the PCSK9-dependent degradation of LDLR in the lysosome, were examined. Human hepatocyte-like C3A cells grown in control (containing 10% fetal bovine serum) or MITO+ (supplemented with BD™ MITO + serum extender) medium were also treated with atorvastatin (Atorv), lovastatin (Lov), or pravastatin (Prav) for 24 h. RNA and protein expression studies and determinations of PCSK9/LDLR complex formation were performed. As expected, the statins increased the expression of PCSK9 and LDLR independently of the medium employed. Interestingly, Atov and Lov caused increases in PCSK9/LDLR complex formation, whereas Prav decreased complex formation when compared to cells treated without drugs. These results may explain why Prav works better for statin intolerant patients than other statins such as Atorv and Lov.
Collapse
Affiliation(s)
- Quantil M Melendez
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Catherine J Wooten
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Dayami Lopez
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA.
| |
Collapse
|
21
|
Siddiqui MK, Veluchamy A, Maroteau C, Tavendale R, Carr F, Pearson E, Colhoun H, Morris AD, George J, Doney A, Pirmohamed M, Alfirevic A, Wadelius M, Maitland van der Zee AH, Ridker PM, Chasman DI, Palmer CNA. CKM Glu83Gly Is Associated With Blunted Creatine Kinase Variation, but Not With Myalgia. CIRCULATION. CARDIOVASCULAR GENETICS 2017; 10:e001737. [PMID: 28790154 DOI: 10.1161/circgenetics.117.001737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/23/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND To test the association of a recently reported variant in the creatine kinase (CK) muscle gene, CKM Glu83Gly (rs11559024) with constitutive creatine phosphokinase (CK) levels, CK variation, and inducibility. Given the diagnostic importance of CK in determining muscle damage, we tested the association of the variant with myalgia. METHODS AND RESULTS Meta-analysis between longitudinal cohort GoDARTS (Genetics of Diabetes Audit and Research, Tayside Scotland), minor allele frequency (=0.02), and randomized clinical trial (JUPITER [Justification for the Use of Statins in Primary Prevention: An Intervention Trial Evaluating Rosuvastatin], minor allele frequency=0.018) was used to replicate the association with baseline CK measures. GoDARTS was used to study the relationship with CK variability. Myalgia was studied in JUPITER trial participants. Baseline and SDs of CK were on average 18% (P value=6×10-63) and 24% (P value=2×10-5) lower for carriers of the variant, respectively. The variant was not associated with myalgia (odds ratio, 0.84; 95% confidence interval, 0.52-1.38). CONCLUSIONS This study highlights that a genetic factor known to be associated with constitutive CK levels is also associated with CK variability and inducibility. This is discussed in the context of evidence to suggest that the variant has an impact on inducibility of CK by trauma through a previously reported case of a homozygous carrier. However, the lack of association between the variant and myalgia suggests that it cannot reliably be used as a biomarker for muscle symptoms.
Collapse
Affiliation(s)
- Moneeza Kalhan Siddiqui
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Abirami Veluchamy
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Cyrielle Maroteau
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Roger Tavendale
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Fiona Carr
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Ewan Pearson
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Helen Colhoun
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Andrew D Morris
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Jacob George
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Alexander Doney
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Munir Pirmohamed
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Ana Alfirevic
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Mia Wadelius
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Anke H Maitland van der Zee
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Paul M Ridker
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Daniel I Chasman
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.)
| | - Colin N A Palmer
- From the Pat McPherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (M.K.S., A.V., C.M., R.T., F.C., E.P., J.G., A.D., C.N.A.P.); Centre for Genomic and Experimental Medicine (H.C.) and Usher Institute of Population Health Sciences and Informatics (A.D.M.), University of Edinburgh, United Kingdom; Institute of Translational Medicine, University of Liverpool, United Kingdom (M.P., A.A.); Department of Medical Sciences, Clinical Pharmacology and Science of Life Laboratory, Uppsala University, Sweden (M.W.); Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, The Netherlands (A.H.M.v.d.Z.); Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.H.M.v.d.Z.); Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.M.R., D.I.C.).
| |
Collapse
|
22
|
Hypercholesterolemia: The role of PCSK9. Arch Biochem Biophys 2017; 625-626:39-53. [DOI: 10.1016/j.abb.2017.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 01/06/2023]
|
23
|
Statins for primary prevention in people with a 10% 10-year cardiovascular risk: too much medicine too soon? Br J Gen Pract 2016; 67:40-41. [PMID: 28034950 DOI: 10.3399/bjgp17x688789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/19/2016] [Indexed: 10/31/2022] Open
|
24
|
Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L, Blumenthal R, Danesh J, Smith GD, DeMets D, Evans S, Law M, MacMahon S, Martin S, Neal B, Poulter N, Preiss D, Ridker P, Roberts I, Rodgers A, Sandercock P, Schulz K, Sever P, Simes J, Smeeth L, Wald N, Yusuf S, Peto R. Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 2016; 388:2532-2561. [PMID: 27616593 DOI: 10.1016/s0140-6736(16)31357-5] [Citation(s) in RCA: 1228] [Impact Index Per Article: 136.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023]
Abstract
This Review is intended to help clinicians, patients, and the public make informed decisions about statin therapy for the prevention of heart attacks and strokes. It explains how the evidence that is available from randomised controlled trials yields reliable information about both the efficacy and safety of statin therapy. In addition, it discusses how claims that statins commonly cause adverse effects reflect a failure to recognise the limitations of other sources of evidence about the effects of treatment. Large-scale evidence from randomised trials shows that statin therapy reduces the risk of major vascular events (ie, coronary deaths or myocardial infarctions, strokes, and coronary revascularisation procedures) by about one-quarter for each mmol/L reduction in LDL cholesterol during each year (after the first) that it continues to be taken. The absolute benefits of statin therapy depend on an individual's absolute risk of occlusive vascular events and the absolute reduction in LDL cholesterol that is achieved. For example, lowering LDL cholesterol by 2 mmol/L (77 mg/dL) with an effective low-cost statin regimen (eg, atorvastatin 40 mg daily, costing about £2 per month) for 5 years in 10 000 patients would typically prevent major vascular events from occurring in about 1000 patients (ie, 10% absolute benefit) with pre-existing occlusive vascular disease (secondary prevention) and in 500 patients (ie, 5% absolute benefit) who are at increased risk but have not yet had a vascular event (primary prevention). Statin therapy has been shown to reduce vascular disease risk during each year it continues to be taken, so larger absolute benefits would accrue with more prolonged therapy, and these benefits persist long term. The only serious adverse events that have been shown to be caused by long-term statin therapy-ie, adverse effects of the statin-are myopathy (defined as muscle pain or weakness combined with large increases in blood concentrations of creatine kinase), new-onset diabetes mellitus, and, probably, haemorrhagic stroke. Typically, treatment of 10 000 patients for 5 years with an effective regimen (eg, atorvastatin 40 mg daily) would cause about 5 cases of myopathy (one of which might progress, if the statin therapy is not stopped, to the more severe condition of rhabdomyolysis), 50-100 new cases of diabetes, and 5-10 haemorrhagic strokes. However, any adverse impact of these side-effects on major vascular events has already been taken into account in the estimates of the absolute benefits. Statin therapy may cause symptomatic adverse events (eg, muscle pain or weakness) in up to about 50-100 patients (ie, 0·5-1·0% absolute harm) per 10 000 treated for 5 years. However, placebo-controlled randomised trials have shown definitively that almost all of the symptomatic adverse events that are attributed to statin therapy in routine practice are not actually caused by it (ie, they represent misattribution). The large-scale evidence available from randomised trials also indicates that it is unlikely that large absolute excesses in other serious adverse events still await discovery. Consequently, any further findings that emerge about the effects of statin therapy would not be expected to alter materially the balance of benefits and harms. It is, therefore, of concern that exaggerated claims about side-effect rates with statin therapy may be responsible for its under-use among individuals at increased risk of cardiovascular events. For, whereas the rare cases of myopathy and any muscle-related symptoms that are attributed to statin therapy generally resolve rapidly when treatment is stopped, the heart attacks or strokes that may occur if statin therapy is stopped unnecessarily can be devastating.
Collapse
Affiliation(s)
- Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Christina Reith
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan Emberson
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jane Armitage
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Colin Baigent
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Lisa Blackwell
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Roger Blumenthal
- Ciccarone Center for the Prevention of Heart Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - David DeMets
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Stephen Evans
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Malcolm Law
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Stephen MacMahon
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Seth Martin
- Ciccarone Center for the Prevention of Heart Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Neal
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Neil Poulter
- International Centre for Circulatory Health & Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - David Preiss
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Paul Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian Roberts
- Clinical Trials Unit, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Anthony Rodgers
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Peter Sandercock
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenneth Schulz
- FHI 360, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Peter Sever
- International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London, London, UK
| | - John Simes
- National Health and Medical Research Council Clinical Trial Centre, University of Sydney, Sydney, Australia
| | - Liam Smeeth
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Nicholas Wald
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Masana L, Lennep JRV. Dose wisely! How lipid-lowering undertreatment can lead to overtreatment. Atherosclerosis 2016; 255:126-127. [PMID: 27793333 DOI: 10.1016/j.atherosclerosis.2016.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/07/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Luis Masana
- Research Unit of Lipids and Atherosclerosis, University Rovira i Virgili, C. Sant Llorenç 21, 43201, Reus, Spain.
| | | |
Collapse
|
26
|
Mancini GJ, Baker S, Bergeron J, Fitchett D, Frohlich J, Genest J, Gupta M, Hegele RA, Ng D, Pearson GJ, Pope J, Tashakkor AY. Diagnosis, Prevention, and Management of Statin Adverse Effects and Intolerance: Canadian Consensus Working Group Update (2016). Can J Cardiol 2016; 32:S35-65. [DOI: 10.1016/j.cjca.2016.01.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 12/24/2022] Open
|
27
|
Tobert JA, Newman CB. The nocebo effect in the context of statin intolerance. J Clin Lipidol 2016; 10:739-747. [DOI: 10.1016/j.jacl.2016.05.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/03/2016] [Accepted: 05/07/2016] [Indexed: 02/08/2023]
|
28
|
Reith C, Blackwell L, Emberson J, Mihaylova B, Armitage J, Fulcher J, Keech A, Simes J, Baigent C, Collins R. Protocol for analyses of adverse event data from randomized controlled trials of statin therapy. Am Heart J 2016; 176:63-9. [PMID: 27264221 PMCID: PMC4906243 DOI: 10.1016/j.ahj.2016.01.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/16/2016] [Indexed: 01/14/2023]
Abstract
The Cholesterol Treatment Trialists' (CTT) Collaboration was originally established to conduct individual participant data meta-analyses of major vascular events, cause-specific mortality, and site-specific cancers in large, long-term, randomized trials of statin therapy (and other cholesterol-modifying treatments). The results of the trials of statin therapy and their associated meta-analyses have shown that statins significantly reduce the risk of major vascular events without any increase in the risk of nonvascular causes of death or of site-specific cancer, but do produce small increases in the incidence of myopathy, diabetes, and, probably, hemorrhagic stroke. The CTT Collaboration has not previously sought data on other outcomes, and so a comprehensive meta-analysis of all adverse events recorded in each of the eligible trials has not been conducted. This protocol prospectively describes plans to extend the CTT meta-analysis data set so as to provide a more complete understanding of the nature and magnitude of any other effects of statin therapy.
Collapse
|