301
|
Garip S, Yapici E, Ozek NS, Severcan M, Severcan F. Evaluation and discrimination of simvastatin-induced structural alterations in proteins of different rat tissues by FTIR spectroscopy and neural network analysis. Analyst 2010; 135:3233-41. [DOI: 10.1039/c0an00540a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
302
|
Macchiarulo A, Thornton JM, Nobeli I. Mapping human metabolic pathways in the small molecule chemical space. J Chem Inf Model 2009; 49:2272-89. [PMID: 19795883 DOI: 10.1021/ci900196u] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The work presented here is a study of human metabolic pathways, as projected in the chemical space of the small molecules they comprise, and it is composed of three parts: a) a study of the extent of clustering and overlap of these pathways in chemical space, b) the development and assessment of a statistical model for estimating the proximity to a given pathway of any small molecule, and c) the use of the above model in estimating the proximity of marketed drugs to human metabolic pathways. The distribution, overlap, and relationships of human metabolic pathways in this space are revealed using both visual and quantitative approaches. A set of selected physicochemical and topological descriptors is used to build a classifier, whose aim is to predict metabolic class and pathway membership of any small molecule. The classifier performs well for tightly clustered, isolated pathways but is, naturally, much less accurate for strongly overlapping pathways. Finally, the extent of overlap of a set of known drugs with the human metabolome is examined, and the classifier is used to predict likely cross-interactions between drugs and the major metabolic pathways in humans.
Collapse
Affiliation(s)
- Antonio Macchiarulo
- Dip. Chimica e Tecnologia del Farmaco, Faculty of Pharmacy, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy.
| | | | | |
Collapse
|
303
|
Pharmacologic inhibition of squalene synthase and other downstream enzymes of the cholesterol synthesis pathway: a new therapeutic approach to treatment of hypercholesterolemia. Cardiol Rev 2009; 17:70-6. [PMID: 19367148 DOI: 10.1097/crd.0b013e3181885905] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hypercholesterolemia is a major risk factor for the development of atherosclerotic vascular diseases. The most popular agents for cholesterol reduction are the statin drugs, which are competitive inhibitors of hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase, the primary rate-limiting enzyme in the hepatic biosynthesis of cholesterol. Although relatively safe and effective, the available statins can cause elevations in liver enzymes and myopathy. Squalene synthase is another enzyme that is downstream to HMG-CoA reductase in the cholesterol synthesis pathway and modulates the first committed step of hepatic cholesterol biosynthesis at the final branch point of the cholesterol biosynthetic pathway. Squalene epoxidase and oxidosqualene cyclase are other enzymes that act distally to squalene synthase. Pharmacologic inhibitors of these downstream enzymes have been developed, which may reduce low-density lipoprotein cholesterol and reduce the myopathy side effect seen with upstream inhibition of HMG-CoA. At this juncture, one squalene synthase inhibitor, lapaquistat (TAK-475) is in active clinical trials as a monotherapy, but there have been suggestions of increased hepatotoxicity with the drug.
Collapse
|
304
|
Mancuso M, Orsucci D, Calsolaro V, Choub A, Siciliano G. Coenzyme Q10 and Neurological Diseases. Pharmaceuticals (Basel) 2009; 2:134-149. [PMID: 27713230 PMCID: PMC3978538 DOI: 10.3390/ph203134] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 11/26/2009] [Accepted: 11/30/2009] [Indexed: 12/13/2022] Open
Abstract
Coenzyme Q10 (CoQ10, or ubiquinone) is a small electron carrier of the mitochondrial respiratory chain with antioxidant properties. CoQ10 supplementation has been widely used for mitochondrial disorders. The rationale for using CoQ10 is very powerful when this compound is primary decreased because of defective synthesis. Primary CoQ10 deficiency is a treatable condition, so heightened "clinical awareness" about this diagnosis is essential. CoQ10 and its analogue, idebenone, have also been widely used in the treatment of other neurodegenerative disorders. These compounds could potentially play a therapeutic role in Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, and other conditions which have been linked to mitochondrial dysfunction. This article reviews the physiological roles of CoQ10, as well as the rationale and the role in clinical practice of CoQ10 supplementation in different neurological diseases, from primary CoQ10 deficiency to neurodegenerative disorders.
Collapse
Affiliation(s)
- Michelangelo Mancuso
- Department of Neuroscience, Neurological Clinic, University of Pisa, Tuscany, Italy.
| | - Daniele Orsucci
- Department of Neuroscience, Neurological Clinic, University of Pisa, Tuscany, Italy
| | - Valeria Calsolaro
- Department of Neuroscience, Neurological Clinic, University of Pisa, Tuscany, Italy
| | - Anna Choub
- Department of Neuroscience, Neurological Clinic, University of Pisa, Tuscany, Italy
| | - Gabriele Siciliano
- Department of Neuroscience, Neurological Clinic, University of Pisa, Tuscany, Italy
| |
Collapse
|
305
|
Ghatak A, Faheem O, Thompson PD. The genetics of statin-induced myopathy. Atherosclerosis 2009; 210:337-43. [PMID: 20042189 DOI: 10.1016/j.atherosclerosis.2009.11.033] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 11/20/2009] [Accepted: 11/20/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Our goal was to use genetic variants to identify factors contributing to the muscular side effects of statins. BACKGROUND Statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) are usually well tolerated medications, but muscle symptoms, ranging from mild myalgia to clinically important rhabdomyolysis are an important side effect of these drugs and a leading cause of noncompliance. Recent results suggest that genetic factors increase the risk of statin-related muscle complaints. We performed a systematic review of the medical literature to determine genetic factors associated with statin myopathy. METHODS We identified English language articles relating statin myopathy and genetic diseases and gene variants via a PubMed search. Articles pertinent to the topic were reviewed in detail. RESULTS/CONCLUSIONS Our review suggests that some patients are susceptible to statin myopathy because of pre-existing subclinical inherited muscular disorders, or genetic variation in statin uptake proteins encoded by SLCO1B1 or the cytochrome P enzyme system. Variations in genes affecting pain perception and polymorphism in vascular receptors may also contribute to statin myopathy. None of the variants identified in this review suggested novel metabolic mechanisms leading to statin myopathy.
Collapse
Affiliation(s)
- Abhijit Ghatak
- Department of Internal Medicine, University of Connecticut, Farmington, CT 06032, USA
| | | | | |
Collapse
|
306
|
Knauer MJ, Urquhart BL, Meyer zu Schwabedissen HE, Schwarz UI, Lemke CJ, Leake BF, Kim RB, Tirona RG. Human skeletal muscle drug transporters determine local exposure and toxicity of statins. Circ Res 2009; 106:297-306. [PMID: 19940267 DOI: 10.1161/circresaha.109.203596] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, or statins, are important drugs used in the treatment and prevention of cardiovascular disease. Although statins are well tolerated, many patients develop myopathy manifesting as muscle aches and pain. Rhabdomyolysis is a rare but severe toxicity of statins. Interindividual differences in the activities of hepatic membrane drug transporters and metabolic enzymes are known to influence statin plasma pharmacokinetics and risk for myopathy. Interestingly, little is known regarding the molecular determinants of statin distribution into skeletal muscle and its relevance to toxicity. OBJECTIVE We sought to identify statin transporters in human skeletal muscle and determine their impact on statin toxicity in vitro. METHODS AND RESULTS We demonstrate that the uptake transporter OATP2B1 (human organic anion transporting polypeptide 2B1) and the efflux transporters, multidrug resistance-associated protein (MRP)1, MRP4, and MRP5 are expressed on the sarcolemmal membrane of human skeletal muscle fibers and that atorvastatin and rosuvastatin are substrates of these transporters when assessed using a heterologous expression system. In an in vitro model of differentiated, primary human skeletal muscle myoblast cells, we demonstrate basal membrane expression and drug efflux activity of MRP1, which contributes to reducing intracellular statin accumulation. Furthermore, we show that expression of human OATP2B1 in human skeletal muscle myoblast cells by adenoviral vectors increases intracellular accumulation and toxicity of statins and such effects were abrogated when cells overexpressed MRP1. CONCLUSIONS These results identify key membrane transporters as modulators of skeletal muscle statin exposure and toxicity.
Collapse
Affiliation(s)
- Michael J Knauer
- Department of Physiology & Pharmacology, Department of Medicine, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | |
Collapse
|
307
|
Orsucci D, Filosto M, Siciliano G, Mancuso M. Electron transfer mediators and other metabolites and cofactors in the treatment of mitochondrial dysfunction. Nutr Rev 2009; 67:427-38. [PMID: 19674340 DOI: 10.1111/j.1753-4887.2009.00221.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial disorders (MDs) are caused by impairment of the mitochondrial electron transport chain (ETC). The ETC is needed for oxidative phosphorylation, which provides the cell with the most efficient energy outcome in terms of ATP production. One of the pathogenic mechanisms of MDs is the accumulation of reactive oxygen species. Mitochondrial dysfunction and oxidative stress appear to also have a strong impact on the pathogenesis of neurodegenerative diseases and cancer. The treatment of MDs is still inadequate. Therapies that have been attempted include ETC cofactors, other metabolites secondarily decreased in MDs, antioxidants, and agents acting on lactic acidosis. However, the role of these dietary supplements in the treatment of the majority of MDs remains unclear. This article reviews the rationale for their use and their role in clinical practice in the context of MDs and other disorders involving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Daniele Orsucci
- Department of Neuroscience, Neurological Clinic, University of Pisa, Italy
| | | | | | | |
Collapse
|
308
|
Abstract
The fundamental role of coenzyme Q(10) (CoQ(10)) in mitochondrial bioenergetics and its well-acknowledged antioxidant properties constitute the basis for its clinical applications, although some of its effects may be related to a gene induction mechanism. Cardiovascular disease is still the main field of study and the latest findings confirm a role of CoQ(10) in improving endothelial function. The possible relation between CoQ(10) deficiency and statin side effects is highly debated, particularly the key issue of whether CoQ(10) supplementation counteracts statin myalgias. Furthermore, in cardiac patients, plasma CoQ(10) was found to be an independent predictor of mortality. Studies on CoQ(10) and physical exercise have confirmed its effect in improving subjective fatigue sensation and physical performance and in opposing exercise-related damage. In the field of mitochondrial myopathies, primary CoQ(10) deficiencies have been identified, involving different genes of the CoQ(10) biosynthetic pathway; some of these conditions were found to be highly responsive to CoQ(10) administration. The initial observations of CoQ(10) effects in Parkinson's and Huntington's diseases have been extended to Friedreich's ataxia, where CoQ(10) and other quinones have been tested. CoQ(10) is presently being used in a large phase III trial in Parkinson's disease. CoQ(10) has been found to improve sperm count and motility on asthenozoospermia. Moreover, for the first time CoQ(10) was found to decrease the incidence of preeclampsia in pregnancy. The ability of CoQ(10) to mitigate headache symptoms in adults was also verified in pediatric and adolescent populations.
Collapse
|
309
|
Callegari S, McKinnon RA, Andrews S, de Barros Lopes MA. Atorvastatin-induced cell toxicity in yeast is linked to disruption of protein isoprenylation. FEMS Yeast Res 2009; 10:188-98. [PMID: 20002195 DOI: 10.1111/j.1567-1364.2009.00593.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Statins, used to treat hypercholesterolemia, are one of the most frequently prescribed drug classes in the developed world. However, a significant proportion of users suffer symptoms of myotoxicity, and currently, the molecular mechanisms underlying myotoxicity remain ambiguous. In this study, Saccharomyces cerevisiae was exploited as a model system to gain further insight into the molecular mechanisms of atorvastatin toxicity. Atorvastatin-treated yeast cells display marked morphological deformities, have reduced cell viability and are highly vulnerable to perturbed mitochondrial function. Supplementation assays of atorvastatin-treated cells reveal that both loss of viability and mitochondrial dysfunction occur as a consequence of perturbation of the sterol synthesis pathway. This was further investigated by supplementing statin-treated cells with various metabolites of the sterol synthesis pathway that are believed to be essential for cell function. Ergosterol, coenzyme Q and a heme precursor were all ineffective in the prevention of statin-induced mitochondrial disruption and cell death. However, the addition of geranylgeranyl pyrophosphate and farnesyl pyrophosphate significantly restored cell viability, although these did not overcome petite induction. This highlights the pleiotropic nature of statin toxicity, but has established protein prenylation disruption as one of the principal mechanisms underlying statin-induced cell death in yeast.
Collapse
Affiliation(s)
- Sylvie Callegari
- Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
310
|
Viljoen A, Wierzbicki AS. Towards companion diagnostics for the management of statin therapy. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2009; 3:659-671. [PMID: 23496050 DOI: 10.1517/17530050903222254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
BACKGROUND Statins are the most commonly prescribed drugs in the world and are established first-line therapy for cardiovascular disease. Statin toxicity is related to dose, age, gender, ethnicity, body mass, renal and endocrine function and also to concomitant medications - particularly those that inhibit cytochrome P450 3A4. OBJECTIVE/METHOD This review describes the tests used before initiation of statin therapy, to establish their efficacy and to monitor their principal side effects. Lipids and apolipoproteins are used to measure efficacy and compliance, whereas transaminases and creatine kinase are used to measure toxicity. Guidelines agree in general, but differ in the details of measurement of baseline levels, action limits and management strategies for statin toxicity. Genetic factors are relevant to both the efficacy and the toxicity of statin therapy, with efficacy being associated with polymorphisms in lipid-related genes, whereas a function-related polymorphism in the organic anion transporting polypeptide 1B1 (OATP1B1; SLCO1B1) is associated with 60% of the cases of myopathy with high-dose simvastatin. CONCLUSIONS Although basic efficacy and safety panels for the initiation and monitoring of statin therapy are well established, controversy remains about the need for ancillary diagnostics in patients and to which patient groups these should be applied.
Collapse
Affiliation(s)
- Adie Viljoen
- Lister Hospital, Department of Clinical Biochemistry, Stevenage SG1 4AB, UK +44 1438 314 333 ext 5972 ; +44 1438 781 147 ;
| | | |
Collapse
|
311
|
Sumien N, Heinrich KR, Shetty RA, Sohal RS, Forster MJ. Prolonged intake of coenzyme Q10 impairs cognitive functions in mice. J Nutr 2009; 139:1926-32. [PMID: 19710165 PMCID: PMC2744613 DOI: 10.3945/jn.109.110437] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Coenzyme Q(10) (CoQ(10)) is widely consumed as a dietary supplement to enhance bioenergetic capacity and to ameliorate the debilitative effects of the aging process or certain pathological conditions. Our main purpose in this study was to determine whether CoQ(10) intake does indeed attenuate the age-associated losses in motor, sensory, and cognitive functions or decrease the rate of mortality in mice. Mice were fed a control nonpurified diet or that diet containing 0.68 mg/g (low dosage) or 2.6 mg/g (high dosage) CoQ(10), starting at 4 mo of age, and were tested for sensory, motor, and cognitive function at 7, 15, and 25 mo of age. Amounts of the ubiquinols CoQ(9)H(2) and CoQ(10)H(2) measured in a parallel study were augmented in the cerebral cortex but not in any other region of the brain. Intake of the low-CoQ(10) diet did not affect age-associated decrements in muscle strength, balance, coordinated running, or learning/memory, whereas intake at the higher amount increased spontaneous activity, worsened the age-related losses in acuity to auditory and shock stimuli, and impaired the spatial learning/memory of old mice. The CoQ(10) diets did not affect survivorship of mice through 25 mo of age. Our results suggest that prolonged intake of CoQ(10) in low amounts has no discernable impact on cognitive and motor functions whereas intake at higher amounts exacerbates cognitive and sensory impairments encountered in old mice. These findings do not support the notion that CoQ(10) is a fitness-enhancing or an "antiaging" substance under normal physiological conditions.
Collapse
Affiliation(s)
- Nathalie Sumien
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; and Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| | - Kevin R. Heinrich
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; and Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| | - Ritu A. Shetty
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; and Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| | - Rajindar S. Sohal
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; and Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| | - Michael J. Forster
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; and Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
312
|
Bentov Y, Esfandiari N, Burstein E, Casper RF. The use of mitochondrial nutrients to improve the outcome of infertility treatment in older patients. Fertil Steril 2009; 93:272-5. [PMID: 19732879 DOI: 10.1016/j.fertnstert.2009.07.988] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/22/2009] [Accepted: 07/14/2009] [Indexed: 11/27/2022]
Abstract
We present a hypothesis emphasizing the role of mitochondrial dysfunction in reproductive senescence and suggesting the use of mitochondrial nutrients as an adjuvant treatment in older patients with infertility.
Collapse
Affiliation(s)
- Yaakov Bentov
- Toronto Centre for Advanced Reproductive Technology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
313
|
|
314
|
Haas D, Niklowitz P, Hörster F, Baumgartner ER, Prasad C, Rodenburg RJ, Hoffmann GF, Menke T, Okun JG. Coenzyme Q(10) is decreased in fibroblasts of patients with methylmalonic aciduria but not in mevalonic aciduria. J Inherit Metab Dis 2009; 32:570-5. [PMID: 19504350 DOI: 10.1007/s10545-009-1150-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/20/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
Abstract
The content of coenzyme Q(10) (CoQ(10)) was examined in skin fibroblasts of 10 patients with mevalonic aciduria (MVA) and of 22 patients with methylmalonic aciduria (MMA). Patients with these inborn errors of metabolism are thought to be at risk for CoQ(10) depletion either by direct inhibition of the proximal pathway of CoQ(10) synthesis (MVA) or indirectly by inhibition of mitochondrial energy metabolism (MMA). We demonstrated that CoQ(10) concentrations were not significantly different from controls in MVA patients, suggesting that there may be upregulatory effects. On the other hand the CoQ(10) content in fibroblasts of patients with MMA was significantly reduced.
Collapse
Affiliation(s)
- D Haas
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children's Hospital, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Smiley WH, Khan BV, Sperling LS. Management of the statin-intolerant patient. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2009; 11:263-71. [DOI: 10.1007/s11936-009-0027-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
316
|
Allison GW, Perla RJ, Belliveau PP, Angelis SM. Elevated creatine phosphokinase levels associated with linezolid therapy. Am J Health Syst Pharm 2009; 66:1097-100. [DOI: 10.2146/ajhp080228] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Glenn W. Allison
- Hospital Medicine Program, MetroWest Medical Center, Framingham, MA
| | - Rocco J. Perla
- Institute for Healthcare Improvement, Cambridge, MA, and Microbiology Section Head and Epidemiologist, HealthAlliance Hospital, Leominster, MA
| | - Paul P. Belliveau
- Department of Pharmacy Practice, Massachusetts College of Pharmacy and Health Sciences, Worcester, MA
| | | |
Collapse
|
317
|
Cao P, Hanai JI, Tanksale P, Imamura S, Sukhatme VP, Lecker SH. Statin-induced muscle damage and atrogin-1 induction is the result of a geranylgeranylation defect. FASEB J 2009; 23:2844-54. [PMID: 19406843 DOI: 10.1096/fj.08-128843] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Statins are widely used to treat hypercholesterolemia but can lead to a number of side effects in muscle, including rhabdomyolysis. Our recent findings implicated the induction of atrogin-1, a gene required for the development of muscle atrophy, in statin-induced muscle damage. Since statins inhibit many biochemical reactions besides cholesterol synthesis, we sought to define the statin-inhibited pathways responsible for atrogin-1 expression and muscle damage. We report here that lovastatin-induced atrogin-1 expression and muscle damage in cultured mouse myotubes and zebrafish can be prevented in the presence of geranylgeranol but not farnesol. Further, inhibitors of the transfer of geranylgeranyl isoprene units to protein targets cause statin muscle damage and atrogin-1 induction in cultured cells and in fish. These findings support the concept that dysfunction of small GTP-binding proteins lead to statin-induced muscle damage since these molecules require modification by geranylgeranyl moieties for their cellular localization and activity. Collectively, our animal and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be regulated by novel signaling pathways.
Collapse
Affiliation(s)
- Peirang Cao
- Renal Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | | | | | |
Collapse
|
318
|
Suspected statin-induced respiratory muscle myopathy during long-term inspiratory muscle training in a patient with diaphragmatic paralysis. Phys Ther 2009; 89:257-66. [PMID: 19147707 DOI: 10.2522/ptj.20080155] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND PURPOSE Abnormal lipids are associated with the development of coronary heart disease; for this reason, lipid-lowering agents have become a standard of care. The purposes of this case report are: (1) to highlight the association of impaired inspiratory muscle performance (IMP) with statin therapy and (2) to describe potentially useful methods of examining and treating people with known or suspected statin-induced skeletal myopathies (SISMs). CASE DESCRIPTION The patient had breathlessness on exertion and a restrictive lung disorder from a right hemidiaphragmatic paralysis, for which he was prescribed high-intensity inspiratory muscle training (IMT). He had a secondary diagnosis of hyperlipidemia, which was treated with 40 mg of simvastatin after 5(1/2) months of IMT. OUTCOMES The improvements in IMP, symptoms, and functional status obtained from almost 6 months of high-intensity IMT were lost after the commencement of simvastatin. However, 3 weeks after termination of simvastatin combined with high-intensity IMT, the patient's IMP, symptoms, and functional status exceeded pre-statin levels. DISCUSSION This case report suggests that high-intensity IMT can be used effectively in a patient with impaired diaphragmatic function and during recovery from a respiratory SISM. The marked reduction in IMP and inability to perform IMT resolved with the cessation of statin therapy. The case report also highlights the potential effects of SISMs in all skeletal muscle groups. The clinical implications of this case report include the potential role of physical therapy in monitoring and possibly facilitating the spontaneous recovery of an SISM, as well as the need to investigate the IMP of a person with dyspnea and fatigue who is taking a statin.
Collapse
|
319
|
Abstract
Approximately 10% of patients treated with statins experience some form of muscle-related side effects in clinical practice. These can range from asymptomatic creatine kinase (CK) elevation, to muscle pain, weakness, and its most severe form, rhabdomyolysis. Higher risk patients for statin myopathy are those older than 80, with a small body frame, on higher statin doses, on other medications, or with other systemic diseases including hepatic or renal diseases, diabetes mellitus, or hypothyroidism. The cause of statin myopathy is presumed to be the same for its variable presentation but has not been defined. In patients with myopathic symptoms, their symptoms and CK levels determine whether statin therapy can be continued or must be stopped.
Collapse
Affiliation(s)
- Carmelo V Venero
- The Henry Low Heart Center, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, USA
| | | |
Collapse
|
320
|
Stein EA. Other therapies for reducing low-density lipoprotein cholesterol: medications in development. Endocrinol Metab Clin North Am 2009; 38:99-119. [PMID: 19217514 DOI: 10.1016/j.ecl.2008.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although the past 30 years have been fruitful and productive in lipid research, from basic science to drug development to demonstration of clinical benefit, cardiovascular disease remains the major cause of mortality and morbidity in industrialized societies. With the rapid industrialization of countries, such as India and China, cardiovascular disease rapidly is becoming the leading cause of global death and disability. Although most of the effective lipid-lowering drugs, the statins, have become generic and inexpensive, there remains a need for effective and safe agents. Hopefully, some of those discussed in this article will fill that need.
Collapse
Affiliation(s)
- Evan A Stein
- Metabolic and Atherosclerosis Research Center, 4685 Forest Avenue, Cincinnati, OH, USA.
| |
Collapse
|
321
|
Statins: mechanisms of neuroprotection. Prog Neurobiol 2009; 88:64-75. [PMID: 19428962 DOI: 10.1016/j.pneurobio.2009.02.002] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 01/09/2009] [Accepted: 02/10/2009] [Indexed: 12/17/2022]
Abstract
Clinical trials report that the class of drugs known as statins may be neuroprotective in Alzheimer's and Parkinson's disease, and further trials are currently underway to test whether these drugs are also beneficial in multiple sclerosis and acute stroke treatment. Since statins are well tolerated and have relatively few side effects, they may be considered as viable drugs to ameliorate neurodegenerative diseases. However, the mechanism of their neuroprotective effects is only partly understood. In this article, we review the current data on the neuroprotective effects of statins and their underlying mechanisms. In the first section, we detail the mechanisms by which statins affect cellular signalling. The primary action of statins is to inhibit cellular cholesterol synthesis. However, the cholesterol synthesis pathway also has several by-products, the non-sterol isoprenoids that are also important in cellular functioning. Furthermore, reduced cholesterol levels may deplete the cholesterol-rich membrane domains known as lipid rafts, which in turn could affect cellular signalling. In the second section, we summarize how the effects on signalling translate into general neuroprotective effects through peripheral systems. Statins improve blood-flow, reduce coagulation, modulate the immune system and reduce oxidative damage. The final section deals with the effects of statins on the central nervous system, particularly during Alzheimer's and Parkinson's disease, stroke and multiple sclerosis.
Collapse
|
322
|
Affiliation(s)
- Tatjána Ábel
- 1 Állami Egészségügyi Központ Szakrendelő Intézet Budapest
| | - János Fehér
- 2 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest
| |
Collapse
|
323
|
Vaklavas C, Chatzizisis YS, Ziakas A, Zamboulis C, Giannoglou GD. Molecular basis of statin-associated myopathy. Atherosclerosis 2009; 202:18-28. [DOI: 10.1016/j.atherosclerosis.2008.05.021] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2007] [Revised: 05/13/2008] [Accepted: 05/13/2008] [Indexed: 12/18/2022]
|
324
|
Itagaki M, Takaguri A, Kano S, Kaneta S, Ichihara K, Satoh K. Possible Mechanisms Underlying Statin-Induced Skeletal Muscle Toxicity in L6 Fibroblasts and in Rats. J Pharmacol Sci 2009; 109:94-101. [DOI: 10.1254/jphs.08238fp] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
325
|
Clearfield M. Clinical trials report. Curr Atheroscler Rep 2008. [DOI: 10.1007/s11883-009-0001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
326
|
|
327
|
|
328
|
Pearson GJ, Thompson AE, Semchuk W. Guidelines for the Management of Dyslipidemia and Prevention of Cardiovascular Disease by Pharmacists. Can Pharm J (Ott) 2008. [DOI: 10.3821/1913-701x-141.sp2.s11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Glen J. Pearson
- From the Division of Cardiology, University of Alberta (Pearson) and Capital Health Regional Pharmacy Services (Thompson), Edmonton, Alberta; and the Regina Qu'Appelle Health Region (Semchuk). Contact
| | - Ann E. Thompson
- From the Division of Cardiology, University of Alberta (Pearson) and Capital Health Regional Pharmacy Services (Thompson), Edmonton, Alberta; and the Regina Qu'Appelle Health Region (Semchuk). Contact
| | - William Semchuk
- From the Division of Cardiology, University of Alberta (Pearson) and Capital Health Regional Pharmacy Services (Thompson), Edmonton, Alberta; and the Regina Qu'Appelle Health Region (Semchuk). Contact
| |
Collapse
|
329
|
Current world literature. Curr Opin Rheumatol 2008; 20:729-35. [PMID: 18946335 DOI: 10.1097/bor.0b013e328317a234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
330
|
|
331
|
Ausili A, Torrecillas A, Aranda F, de Godos A, Sánchez-Bautista S, Corbalán-García S, Gómez-Fernández JC. Redox State of Coenzyme Q10 Determines Its Membrane Localization. J Phys Chem B 2008; 112:12696-702. [DOI: 10.1021/jp802215s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alessio Ausili
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Alejandro Torrecillas
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Francisco Aranda
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Ana de Godos
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Sonia Sánchez-Bautista
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Senena Corbalán-García
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | - Juan C. Gómez-Fernández
- Departamento de Bioquímica y Biología Molecular (A), Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| |
Collapse
|
332
|
Current world literature. Curr Opin Neurol 2008; 21:615-24. [PMID: 18769258 DOI: 10.1097/wco.0b013e32830fb782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
333
|
Buettner C, Lecker SH. Molecular basis for statin-induced muscle toxicity: implications and possibilities. Pharmacogenomics 2008; 9:1133-42. [DOI: 10.2217/14622416.9.8.1133] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Statins are widely used to treat hypercholesterolemia, a known risk factor for atherosclerosis. These drugs can lead to a number of side effects in muscle, including rhabdomyolysis; however, the mechanism of muscle injury is poorly defined. We review the clinical characteristics of this diverse syndrome, as well as the biochemical mechanisms that might provide an explanation for the toxicity of these agents. New findings implicating atrogin-1, a gene required for muscle atrophy, in the pathophysiology of statin-induced muscle injury are discussed, as well as implications of these novel discoveries.
Collapse
Affiliation(s)
- Catherine Buettner
- General Medicine and Primary Care, Atrium Suite 1, Shapiro 1, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue Boston, MA, 02215 USA
| | - Stewart H Lecker
- Divisions of Nephrology, Renal Unit, DA517, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215 USA
| |
Collapse
|
334
|
Pacanowski MA, Frye RF, Enogieru O, Schofield RS, Zineh I. Plasma Coenzyme Q10 Predicts Lipid-lowering Response to High-Dose Atorvastatin. J Clin Lipidol 2008; 2:289-97. [PMID: 19649137 PMCID: PMC2598393 DOI: 10.1016/j.jacl.2008.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Coenzyme Q10 (CoQ10) is a provitamin synthesized via the HMG-CoA reductase pathway, and thus may serve as a potential marker of intrinsic HMG-CoA reductase activity. HMG-CoA reductase inhibitors (statins) decrease CoQ10, although it is unclear whether this is due to reductions in lipoproteins, which transport CoQ10. OBJECTIVES We evaluated whether baseline plasma CoQ10 concentrations predict the lipid-lowering response to high-dose atorvastatin, and to what extent CoQ10 changes following atorvastatin therapy depend on lipoprotein changes. METHODS Individuals without dyslipidemia or known cardiovascular disease (n=84) received atorvastatin 80 mg daily for 16 weeks. Blood samples collected at baseline and after 4, 8, and 16 weeks of treatment were assayed for CoQ10. RESULTS Individuals with higher baseline CoQ10:LDL-C ratios displayed diminished absolute and percent LDL-C reductions at 8 and 16 weeks of atorvastatin treatment (P<0.001 to 0.01). After 16 weeks of atorvastatin, plasma CoQ10 decreased 45% from 762+/-301 ng/ml to 374+/-150 ng/ml (P<0.001). CoQ10 changes were correlated with LDL-C and apolipoprotein B changes (r=0.27-0.38, P=0.001-0.02), but remained significant when normalized to all lipoproteins. CoQ10 changes were not associated with adverse drug reactions. CONCLUSION Baseline CoQ10:LDL-C ratio was associated with the degree of LDL-C response to atorvastatin. Atorvastatin decreased CoQ10 concentrations in a manner that was not completely dependent on lipoprotein changes. The utility of CoQ10 as a predictor of atorvastatin response should be further explored in patients with dyslipidemia.
Collapse
Affiliation(s)
- Michael A. Pacanowski
- University of Florida College of Pharmacy, Department of Pharmacy Practice and Center for Pharmacogenomics; Gainesville, FL, USA
| | - Reginald F. Frye
- University of Florida College of Pharmacy, Department of Pharmacy Practice and Center for Pharmacogenomics; Gainesville, FL, USA
| | - Osatohanmen Enogieru
- University of Florida College of Pharmacy, Department of Pharmacy Practice and Center for Pharmacogenomics; Gainesville, FL, USA
| | - Richard S. Schofield
- University of Florida College of Medicine, Division of Cardiovascular Medicine; Gainesville, FL, USA
- Department of Veterans Affairs Medical Center; Gainesville, FL, USA
| | - Issam Zineh
- University of Florida College of Pharmacy, Department of Pharmacy Practice and Center for Pharmacogenomics; Gainesville, FL, USA
- University of Florida College of Medicine, Division of Cardiovascular Medicine; Gainesville, FL, USA
| |
Collapse
|
335
|
Soran H, Durrington P. Rosuvastatin: efficacy, safety and clinical effectiveness. Expert Opin Pharmacother 2008; 9:2145-60. [DOI: 10.1517/14656566.9.12.2145] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
336
|
Jacobson TA. Toward "pain-free" statin prescribing: clinical algorithm for diagnosis and management of myalgia. Mayo Clin Proc 2008; 83:687-700. [PMID: 18533086 DOI: 10.4065/83.6.687] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Myalgia, which often manifests as pain or soreness in skeletal muscles, is among the most salient adverse events associated with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins). Clinical issues related to statin-associated myotoxicity include (1) incidence in randomized controlled trials and occurrence in postmarketing surveillance databases; (2) potential differences between statins in their associations with such adverse events; and (3) diagnostic and treatment strategies to prevent, recognize, and manage these events. Data from systematic reviews, meta-analyses, clinical and observational trials, and post-marketing surveillance indicate that statin-associated myalgia typically affects approximately 5.0% of patients, as myopathy in 0.1% and as rhabdomyolysis in 0.01%. However, studies also suggest that myalgia is among the leading reasons patients discontinue statins (particularly high-dose statin monotherapy) and that treatment with certain statins (eg, fluvastatin) is unlikely to result in such adverse events. This review presents a clinical algorithm for monitoring and managing statin-associated myotoxicity. The algorithm highlights risk factors for muscle toxicity and provides recommendations for (1) creatine kinase measurements and monitoring; (2) statin dosage reduction, discontinuation, and rechallenge; and (3) treatment alternatives, such as extended-release fluvastatin with or without ezetimibe, low-dose or alternate-day rosuvastatin, or ezetimibe with or without colesevelam. The algorithm should help to inform and enhance patient care and reduce the risk of myalgia and other potentially treatment-limiting muscle effects that might undermine patient adherence and compromise the overall cardioprotective benefits of statins.
Collapse
Affiliation(s)
- Terry A Jacobson
- Office of Health Promotion and Disease Prevention, Emory University, Faculty Office Building, 49 Jessie Hill Jr Dr SE, Atlanta, GA 30303, USA.
| |
Collapse
|
337
|
|
338
|
Abstract
Although hyperlipidemia clearly plays a role in developing cardiovascular disease, the 10% of the population with the highest LDL levels account for only 20% of cardiovascular disease events. Thus, in order to significantly reduce the burden of cardiovascular disease, subjects with only modest dyslipidemia need to be treated with lipid-lowering therapy. Current guidelines have recommended aggressive lipid-lowering therapy in subjects at high risk of cardiovascular disease. The rationale behind this and treatment options to achieve target lipid goals are discussed.
Collapse
Affiliation(s)
- Lisa R Tannock
- Division of Endocrinology and Molecular Medicine, University of Kentucky, and, Veterans Affairs Medical Center, Lexington, Kentucky 40502, USA.
| |
Collapse
|
339
|
|
340
|
Chatzizisis YS, Vaklavas C, Giannoglou GD. Coenzyme Q10 depletion: etiopathogenic or predisposing factor in statin associated myopathy? Am J Cardiol 2008; 101:1071. [PMID: 18359340 DOI: 10.1016/j.amjcard.2007.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 12/01/2007] [Accepted: 12/09/2007] [Indexed: 11/16/2022]
|
341
|
|
342
|
|
343
|
Hydrophobic statins induce autophagy in cultured human rhabdomyosarcoma cells. Biochem Biophys Res Commun 2008; 367:462-7. [PMID: 18178158 DOI: 10.1016/j.bbrc.2007.12.166] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2007] [Accepted: 12/27/2007] [Indexed: 11/20/2022]
Abstract
Statins are widely used to treat hypercholesterolemia, but they are associated with muscle-related adverse events, by as yet, inadequately resolved mechanisms. In this study, we report that statins induced autophagy in cultured human rhabdomyosarcoma A204 cells. Potency differed widely among the statins: cerivastatin induced autophagy at 0.1muM, simvastatin at 10muM but none was induced by pravastatin. Addition of mevalonate, but not cholesterol, blocked induction of autophagy by cerivastatin, suggesting that this induction is dependent on modulation of isoprenoid metabolic pathways. The statin-induced autophagy was not observed in other types of cells, such as human hepatoma HepG2 or embryonic kidney HEK293 cells. Muscle-specific abortive induction of autophagy by hydrophobic statins is a possible mechanism for statin-induced muscle-related side effects.
Collapse
|
344
|
Edwards IR. The Author’s Reply. Drug Saf 2008. [DOI: 10.2165/00002018-200831020-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
345
|
Liebson PR. ILLUMINATE-Part 2 and CORONA. PREVENTIVE CARDIOLOGY 2008; 11:123-126. [PMID: 18401242 DOI: 10.1111/j.1751-7141.2008.07765.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
346
|
|
347
|
Kjekshus J, Apetrei E, Barrios V, Böhm M, Cleland JGF, Cornel JH, Dunselman P, Fonseca C, Goudev A, Grande P, Gullestad L, Hjalmarson A, Hradec J, Jánosi A, Kamenský G, Komajda M, Korewicki J, Kuusi T, Mach F, Mareev V, McMurray JJV, Ranjith N, Schaufelberger M, Vanhaecke J, van Veldhuisen DJ, Waagstein F, Wedel H, Wikstrand J. Rosuvastatin in older patients with systolic heart failure. N Engl J Med 2007; 357:2248-61. [PMID: 17984166 DOI: 10.1056/nejmoa0706201] [Citation(s) in RCA: 1137] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Patients with systolic heart failure have generally been excluded from statin trials. Acute coronary events are uncommon in this population, and statins have theoretical risks in these patients. METHODS A total of 5011 patients at least 60 years of age with New York Heart Association class II, III, or IV ischemic, systolic heart failure were randomly assigned to receive 10 mg of rosuvastatin or placebo per day. The primary composite outcome was death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke. Secondary outcomes included death from any cause, any coronary event, death from cardiovascular causes, and the number of hospitalizations. RESULTS As compared with the placebo group, patients in the rosuvastatin group had decreased levels of low-density lipoprotein cholesterol (difference between groups, 45.0%; P<0.001) and of high-sensitivity C-reactive protein (difference between groups, 37.1%; P<0.001). During a median follow-up of 32.8 months, the primary outcome occurred in 692 patients in the rosuvastatin group and 732 in the placebo group (hazard ratio, 0.92; 95% confidence interval [CI], 0.83 to 1.02; P=0.12), and 728 patients and 759 patients, respectively, died (hazard ratio, 0.95; 95% CI, 0.86 to 1.05; P=0.31). There were no significant differences between the two groups in the coronary outcome or death from cardiovascular causes. In a prespecified secondary analysis, there were fewer hospitalizations for cardiovascular causes in the rosuvastatin group (2193) than in the placebo group (2564) (P<0.001). No excessive episodes of muscle-related or other adverse events occurred in the rosuvastatin group. CONCLUSIONS Rosuvastatin did not reduce the primary outcome or the number of deaths from any cause in older patients with systolic heart failure, although the drug did reduce the number of cardiovascular hospitalizations. The drug did not cause safety problems. (ClinicalTrials.gov number, NCT00206310.)
Collapse
Affiliation(s)
- John Kjekshus
- Department of Cardiology, University of Oslo, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Velavan P, Huan Loh P, Clark A, Cleland JGF. The Cholesterol Paradox in Heart Failure. ACTA ACUST UNITED AC 2007; 13:336-41. [DOI: 10.1111/j.1527-5299.2007.07211.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
349
|
Marcoff L, Thompson PD. Reply. J Am Coll Cardiol 2007. [DOI: 10.1016/j.jacc.2007.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
350
|
Wolinsky H. Coenzyme Q10 in Statin-Associated Myopathy. J Am Coll Cardiol 2007; 50:1911; author reply 1911. [DOI: 10.1016/j.jacc.2007.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 07/18/2007] [Indexed: 10/22/2022]
|