1
|
Kavey REW. Myopathy in Statin-Treated Children and Adolescents: A Practical Approach. Curr Atheroscler Rep 2024; 26:683-692. [PMID: 39316353 DOI: 10.1007/s11883-024-01239-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE OF REVIEW This paper reviews the existing literature on statin-related myopathy in children and adolescents, to inform development of a practical management approach. RECENT FINDINGS Reports of statin treatment in the pediatric population revealed no evidence of muscle pathology, with asymptomatic elevation of creatine kinase(CK) levels and symptoms of muscle pain without CK elevation seen equally in subjects and controls in RCTs. By contrast, rare cases of rhabdomyolysis have now been documented in statin-treated children; this serious problem had never been previously reported. Statin-induced myopathy is rare in childhood so routine monitoring of CK levels is unnecessary in asymptomatic patients, reserved for those with muscle pain. Rare case reports of rhabdomyolysis in statin-treated children and adolescents suggest that parent and patient education on symptoms of adverse statin effects should include immediate physician contact with the appearance of dark urine, with or without muscle pain.
Collapse
Affiliation(s)
- Rae-Ellen W Kavey
- University of Rochester Medical Center, 1475 East Avenue, Rochester, NY, 14610, USA.
| |
Collapse
|
2
|
Gougeon A, Aribi I, Guernouche S, Lega JC, Wright JM, Verstuyft C, Lajoinie A, Gueyffier F, Grenet G. Publication bias in pharmacogenetics of statin-associated muscle symptoms: A meta-epidemiological study. Atherosclerosis 2024:118624. [PMID: 39488449 DOI: 10.1016/j.atherosclerosis.2024.118624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND AND AIMS Statin-associated muscle symptoms (SAMS) are a major cause of treatment discontinuation. Clinical Pharmacogenetics Implementation Consortium (CPIC) recommend dose adjustment for statin treatment according to known SLCO1B1 genotype to reduce SAMS. We hypothesized that the association between SLCO1B1 genotype and SAMS is misestimated because of publication bias. METHODS We searched published systematic reviews on the association between SLCO1B1 genotype and SAMS. To assessed publication bias, we used funnel plot visual inspection, Egger's test, and the Bayes Factor (BFPublication-bias) from Robust Bayesian Meta-Analysis (RoBMA). We compared the odds ratios (ORUncorrected) from meta-analyses before and after correcting for publication bias using trim-and-fill (ORTrim&Fill) and RoBMA (ORRoBMA) methods. RESULTS We included 8 cohort and 11 case-control studies, totaling 62 OR of three SLCO1B1 genotypes and six statin drugs. In the primary analysis, the funnel plot was suggestive of publication bias, confirmed by Egger's test (p=0.001) and RoBMA (BFPublication-bias = 18). Correcting the estimate for publication bias resulted in loss of the association, from a significant ORUncorrected (1.31 95%CI [1.13-1.53]) to corrected ORs suggesting no difference: ORTrim&Fill (1.07 95%CI [0.89-1.30]) and ORRoBMA (1.02 95%CI [1.00-1.33]). This suggested that publication bias overestimated the association by 18 % and 23 %, respectively. Similar results were found for genotype rs4149056, simvastatin and atorvastatin. CONCLUSIONS The effect of the SLCO1B1 genotype on the risk of developing SAMS is overestimated in the published literature, especially rs4149056. This could lead prescribers to incorrectly decreasing statin doses or even avoiding statin use, leading to a loss of the potential cardiovascular benefit of statins.
Collapse
Affiliation(s)
- A Gougeon
- Laboratoire de Biométrie et Biologie Evolutive UMR CNRS 5558, Université Lyon 1, Université de Lyon, Villeurbanne, France; RCTs, Lyon, France.
| | - I Aribi
- Laboratoire de Biométrie et Biologie Evolutive UMR CNRS 5558, Université Lyon 1, Université de Lyon, Villeurbanne, France
| | - S Guernouche
- Service de Neurochirurgie Pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| | - J C Lega
- Laboratoire de Biométrie et Biologie Evolutive UMR CNRS 5558, Université Lyon 1, Université de Lyon, Villeurbanne, France; Service de Rhumatologie, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France; Service Hospitalo-Universitaire de Pharmacotoxicologie, Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - J M Wright
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - C Verstuyft
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | | | - F Gueyffier
- Laboratoire de Biométrie et Biologie Evolutive UMR CNRS 5558, Université Lyon 1, Université de Lyon, Villeurbanne, France
| | - G Grenet
- Laboratoire de Biométrie et Biologie Evolutive UMR CNRS 5558, Université Lyon 1, Université de Lyon, Villeurbanne, France; Service Hospitalo-Universitaire de Pharmacotoxicologie, Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
3
|
Rocca B, Bigagli E, Cerbai E. Ticagrelor and Statins: Dangerous Liaisons? Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07624-7. [PMID: 39348077 DOI: 10.1007/s10557-024-07624-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/01/2024]
Abstract
Polypharmacy is often necessary in complex, chronic, comorbid and cardiovascular patients and is a known risk factor for potential drug-drug interaction (DDI) that can cause adverse reactions (toxicity or therapeutic failure). Anti-thrombotic drugs (largely low-dose aspirin and a platelet P2Y12 receptor inhibitor) and statins are among the most co-administered drugs in cardiovascular patients. Ticagrelor is a selective antagonist of the platelet P2Y12-receptor, highly effective in inhibiting platelet aggregation and bio-transformed by the CYP3A4 and substrate of transporters, such as the breast cancer resistance protein (BCRP). Statins have different pharmacokinetic profiles; some undergo CYP3A4-mediated metabolism; rosuvastatin is primarily metabolized by the CYP2C9; and they have different affinities for drug transporters. Rhabdomyolysis is a very rare but severe adverse event, which is specific for statins which can be triggered by DDIs that increase statin's concentrations through blockade of their biotransformation and/or elimination. Large pharmacovigilance and small observational studies reported increased rhabdomyolysis in patients treated with some statins and ticagrelor but not aspirin, clopidogrel or prasugrel. Recent studies in vitro, pharmacokinetic trials and in silico drug modelling identified and validated the BCRP inhibition by ticagrelor, as a mechanism contributing to the DDI with statins, as 'victim' drugs, leading to increased rhabdomyolysis. While the clinical impact of this DDI deserves further investigation, a careful evaluation should be advised when ticagrelor is co-prescribed with some statins.
Collapse
Affiliation(s)
- Bianca Rocca
- Department of Safety and Bioethics, Catholic University, Largo F. Vito 1, Rome, Italy
- Department of Medicine and Surgery, LUM University, SS 100, km 18, Casamassima, Bari, Italy
| | - Elisabetta Bigagli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini 6, Florence, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini 6, Florence, Italy.
| |
Collapse
|
4
|
Németh S, Kriegshäuser G, Hayrapetyan H, Oberkanins C, Sarkisian T. Allelic frequencies of polymorphism c.521T>C (rs4149056) favor preemptive SLCO1B1 genotyping in Armenia. Drug Metab Pers Ther 2024; 39:159-161. [PMID: 38997114 DOI: 10.1515/dmpt-2024-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024]
Abstract
OBJECTIVES Statins represent an important pharmacological factor for the prevention of cardiovascular diseases but may also cause severe cases of myotoxicity. Numerous studies have described the association of the SLCO1B1 gene variant c.521C with statin-induced myopathy across different populations. This study aimed at evaluating the usefulness of preemptive SLCO1B1 genotyping in Armenia. METHODS A total of 202 Armenian patients referred to the Center of Medical Genetics and Primary Health Care in Yerevan for upper respiratory tract infection between January and May 2022 were included in this study. Genotyping for SLCO1B1 c.521T>C (rs4149056) was performed using a commercially available real-time PCR assay (RealFast™). RESULTS In total, 3/202 (1.5 %) samples were C/C homozygotes and 52/202 (25.7 %) were T/C heterozygotes, associated with a high and increased risk for statin-induced myopathy, respectively. The SLCO1B1 c.521C allelic frequency was 14.4 %. CONCLUSIONS The observed allele frequency of 14.4 % for the c.521C variant is slightly lower than frequencies reported from Europe, but relatively high compared to Asian populations, suggesting that preemptive SLCO1B1 genotyping could be a useful approach for the reduction of statin-induced adverse effects in Armenia.
Collapse
Affiliation(s)
| | - Gernot Kriegshäuser
- Department of Medical Genetics, Yerevan State Medical University, Yerevan, Armenia
| | - Hasmik Hayrapetyan
- Department of Medical Genetics, Yerevan State Medical University, Yerevan, Armenia
- Center of Medical Genetics and Primary Health Care, Yerevan, Armenia
| | | | - Tamara Sarkisian
- Department of Medical Genetics, Yerevan State Medical University, Yerevan, Armenia
- Center of Medical Genetics and Primary Health Care, Yerevan, Armenia
| |
Collapse
|
5
|
Zhang P, Munier JJ, Wiese CB, Vergnes L, Link JC, Abbasi F, Ronquillo E, Scheker K, Muñoz A, Kuang YL, Theusch E, Lu M, Sanchez G, Oni-Orisan A, Iribarren C, McPhaul MJ, Nomura DK, Knowles JW, Krauss RM, Medina MW, Reue K. X chromosome dosage drives statin-induced dysglycemia and mitochondrial dysfunction. Nat Commun 2024; 15:5571. [PMID: 38956041 PMCID: PMC11219728 DOI: 10.1038/s41467-024-49764-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
Statin drugs lower blood cholesterol levels for cardiovascular disease prevention. Women are more likely than men to experience adverse statin effects, particularly new-onset diabetes (NOD) and muscle weakness. Here we find that impaired glucose homeostasis and muscle weakness in statin-treated female mice are associated with reduced levels of the omega-3 fatty acid, docosahexaenoic acid (DHA), impaired redox tone, and reduced mitochondrial respiration. Statin adverse effects are prevented in females by administering fish oil as a source of DHA, by reducing dosage of the X chromosome or the Kdm5c gene, which escapes X chromosome inactivation and is normally expressed at higher levels in females than males. As seen in female mice, we find that women experience more severe reductions than men in DHA levels after statin administration, and that DHA levels are inversely correlated with glucose levels. Furthermore, induced pluripotent stem cells from women who developed NOD exhibit impaired mitochondrial function when treated with statin, whereas cells from men do not. These studies identify X chromosome dosage as a genetic risk factor for statin adverse effects and suggest DHA supplementation as a preventive co-therapy.
Collapse
Affiliation(s)
- Peixiang Zhang
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Joseph J Munier
- Molecular, Cellular & Integrative Physiology, University of California, Los Angeles, CA, USA
| | - Carrie B Wiese
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Laurent Vergnes
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jenny C Link
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
- Department of Biology, Whittier College, Whittier, CA, USA
| | - Fahim Abbasi
- Division of Cardiovascular Medicine and Cardiovascular Institute, Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Emilio Ronquillo
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Katherine Scheker
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Antonio Muñoz
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Yu-Lin Kuang
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Elizabeth Theusch
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Meng Lu
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | | | - Akinyemi Oni-Orisan
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | | | - Michael J McPhaul
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA, 92675, USA
| | - Daniel K Nomura
- Nutritional Sciences and Toxicology, and Novartis-Berkeley Center of Proteomics and Chemistry Technologies, University of California, Berkeley, Berkeley, CA, USA
| | - Joshua W Knowles
- Division of Cardiovascular Medicine and Cardiovascular Institute, Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald M Krauss
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Marisa W Medina
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Karen Reue
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Dagli-Hernandez C, Ferreira GM, Freitas RCCD, Borges JB, Oliveira VFD, Gonçalves RM, Faludi AA, Marçal EDSR, Bastos GM, Bortolin RH, Hirata MH, Hirata RDC. Predicted deleterious variants in ABCA1, LPL, LPA and KIF6 are associated with statin response and adverse events in patients with familial hypercholesterolemia and disturb protein structure and stability. Pharmacogenet Genomics 2024; 34:91-104. [PMID: 38682317 DOI: 10.1097/fpc.0000000000000524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
OBJECTIVES This study explored the association of deleterious variants in pharmacodynamics (PD) genes with statin response and adverse effects in patients with familial hypercholesterolemia (FH) and analyzed their potential effects on protein structure and stability. METHODS Clinical and laboratory data were obtained from 144 adult FH patients treated with statins. A panel of 32 PD genes was analyzed by exon-targeted gene sequencing. Deleterious variants were identified using prediction algorithms and their structural effects were analyzed by molecular modeling studies. RESULTS A total of 102 variants were predicted as deleterious (83 missense, 8 stop-gain, 4 frameshift, 1 indel, 6 splicing). The variants ABCA1 rs769705621 (indel), LPA rs41267807 (p.Tyr2023Cys) and KIF6 rs20455 (p.Trp719Arg) were associated with reduced low-density lipoprotein cholesterol (LDLc) response to statins, and the LPL rs1801177 (p.Asp36Asn) with increased LDLc response (P < 0.05). LPA rs3124784 (p.Arg2016Cys) was predicted to increase statin response (P = 0.022), and ABCA1 rs769705621 to increase the risk of statin-related adverse events (SRAE) (P = 0.027). LPA p.Arg2016Cys and LPL p.Asn36Asp maintained interactions with solvent, LPA p.Tyr2023Cys reduced intramolecular interaction with Gln1987, and KIF6 p.Trp719Arg did not affect intramolecular interactions. DDMut analysis showed that LPA p.Arg2016Cys and p.Tyr2023Cys and LPL p.Asp36Asn caused energetically favorable changes, and KIF6 p.Trp719Arg resulted in unfavorable energetic changes, affecting protein stability. CONCLUSION Deleterious variants in ABCA1, LPA, LPL and KIF6 are associated with variability in LDLc response to statins, and ABCA1 rs769705621 is associated with SRAE risk in FH patients. Molecular modeling studies suggest that LPA p.Tyr2023Cys and KIF6 p.Trp719Arg disturb protein conformational structure and stability.
Collapse
Affiliation(s)
- Carolina Dagli-Hernandez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Glaucio Monteiro Ferreira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Renata Caroline Costa de Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Victor Fernandes de Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Pham N, Benhammou JN. Statins in Chronic Liver Disease: Review of the Literature and Future Role. Semin Liver Dis 2024; 44:191-208. [PMID: 38701856 DOI: 10.1055/a-2319-0694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Chronic liver disease (CLD) is a major contributor to global mortality, morbidity, and healthcare burden. Progress in pharmacotherapeutic for CLD management is lagging given its impact on the global population. While statins are indicated for the management of dyslipidemia and cardiovascular disease, their role in CLD prevention and treatment is emerging. Beyond their lipid-lowering effects, their liver-related mechanisms of action are multifactorial and include anti-inflammatory, antiproliferative, and immune-protective effects. In this review, we highlight what is known about the clinical benefits of statins in viral and nonviral etiologies of CLD and hepatocellular carcinoma (HCC), and explore key mechanisms and pathways targeted by statins. While their benefits may span the spectrum of CLD and potentially HCC treatment, their role in CLD chemoprevention is likely to have the largest impact. As emerging data suggest that genetic variants may impact their benefits, the role of statins in precision hepatology will need to be further explored.
Collapse
Affiliation(s)
- Nguyen Pham
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jihane N Benhammou
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
- Veterans Affairs Greater Los Angeles, Los Angeles, California
- Comprehensive Liver Research Center at University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
8
|
Pan Y, Wang Y, Zheng Y, Chen J, Li J. A disproportionality analysis of FDA adverse event reporting system (FAERS) events for ticagrelor. Front Pharmacol 2024; 15:1251961. [PMID: 38655177 PMCID: PMC11035729 DOI: 10.3389/fphar.2024.1251961] [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: 07/03/2023] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
Background Ticagrelor is a commonly used antiplatelet agent, but due to the stringent criteria for trial population inclusion and the limited sample size, its safety profile has not been fully elucidated. Method We utilized OpenVigil 2.1 to query the FDA Adverse Event Reporting System database and retrieved reports by the generic name "ticagrelor" published between 1 October 2010 and 31 March 2023. Adverse drug events (ADEs) were classified and described according to the preferred terms and system organ classes in the Medical Dictionary of Regulatory Activity. Proportional reporting ratio (PRR), reporting odds ratio (ROR) and Bayesian Confidence Propagation Neural Network (BCPNN) were used to detect signals. Results The number of ADE reports with ticagrelor as the primary suspect drug was 12,909. The top three ADEs were dyspnea [1824 reports, ROR 7.34, PRR 6.45, information component (IC) 2.68], chest pain (458 reports, ROR 5.43, PRR 5.27, IC 2.39), and vascular stent thrombosis (406 reports, ROR 409.53, PRR 396.68, IC 8.02). The highest ROR, 630.24, was found for "vascular stent occlusion". Cardiac arrest (137 reports, ROR 3.41, PRR 3.39, IC 1.75), atrial fibrillation (99 reports, ROR 2.05, PRR 2.04, IC 1.03), asphyxia (101 reports, ROR 23.60, PRR 23.43, IC 4.51), and rhabdomyolysis (57 reports, ROR 2.75, PRR 2.75, IC 1.45) were suspected new adverse events of ticagrelor. Conclusion The FAERS database produced potential signals associated with ticagrelor that have not been recorded in the package inserts, such as cardiac arrest, atrial fibrillation, asphyxia, and rhabdomyolysis. Further clinical surveillance is needed to quantify and validate potential hazards associated with ticagrelor-related adverse events.
Collapse
Affiliation(s)
- Yunyan Pan
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yu Wang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yifan Zheng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Clinical Pharmacy Translational Science, University of Michigan College of Pharmacy, Ann Arbor, MI, United States
| | - Jie Chen
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia Li
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Lauwers C, De Bruyn L, Langouche L. Impact of critical illness on cholesterol and fatty acids: insights into pathophysiology and therapeutic targets. Intensive Care Med Exp 2023; 11:84. [PMID: 38015312 PMCID: PMC10684846 DOI: 10.1186/s40635-023-00570-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
Critical illness is characterized by a hypercatabolic response encompassing endocrine and metabolic alterations. Not only the uptake, synthesis and metabolism of glucose and amino acids is majorly affected, but also the homeostasis of lipids and cholesterol is altered during acute and prolonged critical illness. Patients who suffer from critically ill conditions such as sepsis, major trauma, surgery or burn wounds display an immediate and sustained reduction in low plasma LDL-, HDL- and total cholesterol concentrations, together with a, less pronounced, increase in plasma free fatty acids. The severity of these alterations is associated with severity of illness, but the underlying pathophysiological mechanisms are multifactorial and only partly clarified. This narrative review aims to provide an overview of the current knowledge of how lipid and cholesterol uptake, synthesis and metabolism is affected during critical illness. Reduced nutritional uptake, increased scavenging of lipoproteins as well as an increased conversion to cortisol or other cholesterol-derived metabolites might all play a role in the decrease in plasma cholesterol. The acute stress response to critical illness creates a lipolytic cocktail, which might explain the increase in plasma free fatty acids, although reduced uptake and oxidation, but also increased lipogenesis, especially in prolonged critical illness, will also affect the circulating levels. Whether a disturbed lipid homeostasis warrants intervention or should primarily be interpreted as a signal of severity of illness requires further research.
Collapse
Affiliation(s)
- Caroline Lauwers
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lauren De Bruyn
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium.
| |
Collapse
|
10
|
Vell MS, Loomba R, Krishnan A, Wangensteen KJ, Trebicka J, Creasy KT, Trautwein C, Scorletti E, Seeling KS, Hehl L, Rendel MD, Zandvakili I, Li T, Chen J, Vujkovic M, Alqahtani S, Rader DJ, Schneider KM, Schneider CV. Association of Statin Use With Risk of Liver Disease, Hepatocellular Carcinoma, and Liver-Related Mortality. JAMA Netw Open 2023; 6:e2320222. [PMID: 37358849 PMCID: PMC10293910 DOI: 10.1001/jamanetworkopen.2023.20222] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/11/2023] [Indexed: 06/27/2023] Open
Abstract
Importance Given the burden of chronic liver disease on the health care system, more information on the hepatoprotective association of statins in the general population is needed. Objective To examine whether regular statin use is associated with a reduction in liver disease, particularly hepatocellular carcinoma (HCC) and liver-related deaths, in the general population. Design, Setting, and Participants This cohort study used data from the UK Biobank (UKB) (individuals aged 37-73 years) collected from baseline (2006-2010) to the end of follow-up in May 2021, from the TriNetX cohort (individuals aged 18-90 years) enrolled from baseline (2011-2020) until end of follow-up in September 2022, and from the Penn Medicine Biobank (PMBB) (individuals aged 18-102 years) with ongoing enrollment starting in 2013 to the end of follow-up in December 2020. Individuals were matched using propensity score matching according to the following criteria: age, sex, body mass index, ethnicity, diabetes with or without insulin or biguanide use, hypertension, ischemic heart disease, dyslipidemia, aspirin use, and number of medications taken (UKB only). Data analysis was performed from April 2021 to April 2023. Exposure Regular statin use. Main Outcomes and Measures Primary outcomes were liver disease and HCC development as well as liver-associated death. Results A total of 1 785 491 individuals were evaluated after matching (aged 55 to 61 years on average, up to 56% men, and up to 49% women). A total of 581 cases of liver-associated death, 472 cases of incident HCC, and 98 497 new liver diseases were registered during the follow-up period. Individuals were aged 55-61 years on average, with a slightly higher proportion of men (up to 56%). In UKB individuals (n = 205 057) without previously diagnosed liver disease, statin users (n = 56 109) had a 15% lower hazard ratio (HR) for the association of developing a new liver disease (HR, 0.85; 95% CI, 0.78-0.92; P < .001). In addition, statin users demonstrated a 28% lower HR for the association with liver-related death (HR, 0.72; 95% CI, 0.59-0.88; P = .001) and a 42% lower HR for the development of HCC (HR, 0.58; 95% CI, 0.35-0.96; P = .04). In TriNetX individuals (n = 1 568 794), the HR for the association of HCC was reduced even further for statin users (HR, 0.26; 95% CI, 0.22-0.31; P = .003). The hepatoprotective association of statins was time and dose dependent, with a significant association in PMBB individuals (n = 11 640) for incident liver diseases after 1 year of statin use (HR, 0.76; 95% CI, 0.59-0.98; P = .03). Taking statins was particularly beneficial in men, individuals with diabetes, and individuals with a high Fibrosis-4 index at baseline. Carriers of the heterozygous minor allele of PNPLA3 rs738409 benefited from statin use and had a 69% lower HR for the association with HCC (UKB HR, 0.31; 95% CI, 0.11-0.85; P = .02). Conclusions and Relevance This cohort study indicates substantial preventive associations of statins against liver disease, with an association with duration and dose of intake.
Collapse
Affiliation(s)
- Mara Sophie Vell
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Rohit Loomba
- Division of Gastroenterology, University of California, San Diego, La Jolla
| | - Arunkumar Krishnan
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown
| | - Kirk J. Wangensteen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jonel Trebicka
- Medical Clinic B, Gastroenterology, Hepatology, Endocrinology, Clinical Infectiology, University Hospital Münster, Münster, Germany
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia
| | - Christian Trautwein
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Eleonora Scorletti
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Katharina Sophie Seeling
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Leonida Hehl
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Miriam Daphne Rendel
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Inuk Zandvakili
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Tang Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jinbo Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marijana Vujkovic
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Saleh Alqahtani
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Daniel James Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kai Markus Schneider
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Carolin Victoria Schneider
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
11
|
Roule V, Alexandre J, Lemaitre A, Chrétien B, Sassier M, Fedrizzi S, Beygui F, Dolladille C. Rhabdomyolysis with Co-Administration of Statins and Antiplatelet Therapies-Analysis of the WHO Pharmacovigilance Database. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07459-8. [PMID: 37115431 DOI: 10.1007/s10557-023-07459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE While statins and antiplatelet therapies are largely prescribed together worldwide, limited information is available on the safety of their association regarding rhabdomyolysis occurrence. We aimed to assess the reporting of rhabdomyolysis in patients treated with a combination of statin and antiplatelet therapy, compared to statin alone. METHODS We used the World Health Organization pharmacovigilance database (VigiBase®) to compare the rhabdomyolysis reporting between statin (atorvastatin, fluvastatin, pravastatin, rosuvastatin, and simvastatin) plus antiplatelet therapy (acetylsalicylic acid, clopidogrel, prasugrel and ticagrelor) groups versus statin alone groups, for each statin and antiplatelet therapy. Study setting was restricted to patients aged 45 or older, including reports up until 1st September, 2021. We computed reporting Odds-Ratio (ROR) and their 95% confidence interval (CI) to quantify the disproportionality between groups, adjusted on age and sex. RESULTS Among the 11,431,708 reports of adverse reactions, we extracted 9,489 cases of rhabdomyolysis in patients treated with statins, of whom 2,464 (26%) were also treated with antiplatelet therapy. The reporting of rhabdomyolysis was increased when ticagrelor was associated with atorvastatin (ROR 1.30 [1.02-1.65]) or rosuvastatin (ROR 1.90 [1.42-2.54]) compared to the respective statin alone but did not change when aspirin, clopidogrel or prasugrel were considered. CONCLUSION Rhabdomyolysis reporting was increased when ticagrelor -but not other antiplatelet agents- was notified with the most prescribed statins in practice. This finding needs to be considered by physicians especially in high-risk patients.
Collapse
Affiliation(s)
- Vincent Roule
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France.
- UMRS 1237, INSERM, GIP Cyceron, 14000, Caen, France.
- Cardiology department, Caen University Hospital, Avenue Cote de Nacre, 14033, Caen, France.
| | - Joachim Alexandre
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Adrien Lemaitre
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France
| | - Basile Chrétien
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Marion Sassier
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Sophie Fedrizzi
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Farzin Beygui
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France
- UMRS 1237, INSERM, GIP Cyceron, 14000, Caen, France
- ACTION Study Group, Pitié-Salpêtrière University Hospital, Cardiology Department, Paris, France
| | - Charles Dolladille
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| |
Collapse
|
12
|
Tariq S, Goriparthi L, Ismail D, Kankeu Tonpouwo G, Thapa M, Khalid K, Cooper AC, Jean-Charles G. Correlates of Myopathy in Diabetic Patients Taking Statins. Cureus 2023; 15:e37708. [PMID: 37206522 PMCID: PMC10191392 DOI: 10.7759/cureus.37708] [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] [Accepted: 04/16/2023] [Indexed: 05/21/2023] Open
Abstract
Diabetes is one of the most common chronic ailments; its incidence has reached epidemic proportions in the 21st century. Diabetes significantly increases micro and macrovascular complications, which are effectively managed with statins. Therefore, statins' pharmacokinetics, pharmacodynamics, and pharmacogenetics have been extensively studied. Although statins act as a keystone in preventing cardiovascular complications, at the same time, they pose a threat to the quality of life of diabetics due to the resulting muscular side effects. This article summarizes the prevalence, clinical manifestations, pathophysiology, and risk factors of statin-induced myopathy in diabetic patients. Among the diverse predisposing risk factors, the primary variables identified for causing myopathy in diabetic patients include age, gender, ethnicity, duration and severity of illness, comorbid conditions, level of physical activity, alcohol use, cholecalciferol (vitamin D3) levels, type and dose of statins, and anti-diabetic drugs or other drugs used concomitantly. In addition, cardiovascular risk quotients also potentially impact diabetic patients making them more vulnerable to developing myopathy from statins. Therefore, this study highlights the importance of managing statin-associated myopathic side effects by providing consensus guidelines on diagnostic, monitoring, and treatment strategies. We also discussed statins' prognostic value in reducing cardiovascular events in diabetic individuals.
Collapse
Affiliation(s)
- Sara Tariq
- Internal Medicine, Mayo Hospital, Lahore, PAK
- Internal Medicine, JC (Jean-Charles) Medical Center, Orlando, USA
| | - Lakshmi Goriparthi
- General Surgery, Osmania Medical College, Hyderabad, IND
- Internal Medicine, JC (Jean-Charles) Medical Center, Orlando, USA
| | - Dina Ismail
- Internal Medicine, JC (Jean-Charles) Medical Center, Orlando, USA
- Family Medicine, University Hassan II of Casablanca Faculty of Medicine and Pharmacy, Casablanca, MAR
| | - Gauvain Kankeu Tonpouwo
- Internal Medicine, Faculty of Medicine, University of Lubumbashi, Plaine Tshombé, Lubumbashi, COD
| | - Milan Thapa
- Internal Medicine, Monmouth Medical Center, Long Branch, USA
| | - Khizer Khalid
- Internal Medicine, JC (Jean-Charles) Medical Center, Orlando, USA
| | | | - Gutteridge Jean-Charles
- Internal Medicine, AdventHealth Orlando Hospital, Orlando, USA
- Internal Medicine, JC (Jean-Charles) Medical Center, Orlando, USA
| |
Collapse
|
13
|
Obeng AO, Scott SA, Kaszemacher T, Ellis SB, Mejia A, Gomez A, Nadukuru R, Abul-Husn NS, Vega A, Waite E, Gottesman O, Cho J, Bottinger EP. Prescriber Adoption of SLCO1B1 Genotype-Guided Simvastatin Clinical Decision Support in a Clinical Pharmacogenetics Program. Clin Pharmacol Ther 2023; 113:321-327. [PMID: 36372942 DOI: 10.1002/cpt.2773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacogenetic implementation programs are increasingly feasible due to the availability of clinical guidelines for implementation research. The utilization of these resources has been reported with selected drug-gene pairs; however, little is known about how prescribers respond to pharmacogenetic recommendations for statin therapy. We prospectively assessed prescriber interaction with point-of-care clinical decision support (CDS) to guide simvastatin therapy for a diverse cohort of primary care patients enrolled in a clinical pharmacogenetics program. Of the 1,639 preemptively genotyped patients, 298 (18.2%) had an intermediate function (IF) OATP1B1 phenotype and 25 (1.53%) had a poor function (PF) phenotype, predicted by a common single nucleotide variant in the SLCO1B1 gene (c.521T>C; rs4149056). Clinicians were presented with CDS when simvastatin was prescribed for patients with IF or PF through the electronic health record. Importantly, 64.2% of the CDS deployed at the point-of-care was accepted by the prescribers and resulted in prescription changes. Statin intensity was found to significantly influence prescriber adoption of the pharmacogenetic-guided CDS, whereas patient gender or race, prescriber type, or pharmacogenetic training status did not significantly influence adoption. This study demonstrates that primary care providers readily adopt pharmacogenetic information to guide statin therapy for the majority of patients with preemptive genotype data.
Collapse
Affiliation(s)
- Aniwaa Owusu Obeng
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Pharmacy Department, The Mount Sinai Hospital, New York, New York, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stuart A Scott
- Department of Pathology, Stanford University, Stanford, California, USA.,Clinical Genomics Laboratory, Stanford Health Care, Palo Alto, California, USA
| | - Tom Kaszemacher
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen B Ellis
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ana Mejia
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alanna Gomez
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rajiv Nadukuru
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Noura S Abul-Husn
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,23andMe Inc., Sunnyvale, California, USA
| | - Aida Vega
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Mount Sinai Faculty Practice Associates, Primary Care Program, The Mount Sinai Health system, New York, New York, USA
| | - Eva Waite
- Mount Sinai Faculty Practice Associates, Primary Care Program, The Mount Sinai Health system, New York, New York, USA
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Empirico Inc., San Diego, California, USA
| | - Judy Cho
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erwin P Bottinger
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
| |
Collapse
|
14
|
Lloyd-Jones DM, Morris PB, Ballantyne CM, Birtcher KK, Covington AM, DePalma SM, Minissian MB, Orringer CE, Smith SC, Waring AA, Wilkins JT. 2022 ACC Expert Consensus Decision Pathway on the Role of Nonstatin Therapies for LDL-Cholesterol Lowering in the Management of Atherosclerotic Cardiovascular Disease Risk: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2022; 80:1366-1418. [PMID: 36031461 DOI: 10.1016/j.jacc.2022.07.006] [Citation(s) in RCA: 188] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
15
|
Munier JJ, Pank JT, Severino A, Wang H, Zhang P, Vergnes L, Reue K. Simultaneous monitoring of mouse grip strength, force profile, and cumulative force profile distinguishes muscle physiology following surgical, pharmacologic and diet interventions. Sci Rep 2022; 12:16428. [PMID: 36180720 PMCID: PMC9525296 DOI: 10.1038/s41598-022-20665-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/16/2022] [Indexed: 01/04/2023] Open
Abstract
Grip strength is a valuable preclinical assay to study muscle physiology in disease and aging by directly determining changes in muscle force generation in active laboratory mice. Existing methods to statistically evaluate grip strength, however, have limitations in the power and scope of the physiological features that are assessed. We therefore designed a microcontroller whose serial measure of resistance-based force enables the simultaneous readout of (1) peak grip strength, (2) force profile (the non-linear progress of force exerted throughout a standard grip strength trial), and (3) cumulative force profile (the integral of force with respect to time of a single grip strength trial). We hypothesized that muscle pathologies of different etiologies have distinct effects on these parameters. To test this, we used our apparatus to assess the three muscle parameters in mice with impaired muscle function resulting from surgically induced peripheral pain, genetic peripheral neuropathy, adverse muscle effects induced by statin drug, and metabolic alterations induced by a high-fat diet. Both surgically induced peripheral nerve injury and statin-associated muscle damage diminished grip strength and force profile, without affecting cumulative force profile. Conversely, genetic peripheral neuropathy resulting from lipin 1 deficiency led to a marked reduction to all three parameters. A chronic high-fat diet led to reduced grip strength and force profile when normalized to body weight. In high-fat fed mice that were exerted aerobically and allowed to recover for 30 min, male mice exhibited impaired force profile parameters, which female mice were more resilient. Thus, simultaneous analysis of peak grip strength, force profile and cumulative force profile distinguishes the muscle impairments that result from distinct perturbations and may reflect distinct motor unit recruitment strategies.
Collapse
Affiliation(s)
- Joseph J Munier
- Department of Molecular, Cellular, and Integrative Physiology, University of California, Los Angeles, CA, 90034, USA
| | - Justin T Pank
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Amie Severino
- Department of Psychiatry and Biobehavioral Disease, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Huan Wang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Peixiang Zhang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
16
|
Liu C, Yan W, Shi J, Wang S, Peng A, Chen Y, Huang K. Biological Actions, Implications, and Cautions of Statins Therapy in COVID-19. Front Nutr 2022; 9:927092. [PMID: 35811982 PMCID: PMC9257176 DOI: 10.3389/fnut.2022.927092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) showed worse prognosis and higher mortality in individuals with obesity. Dyslipidemia is a major link between obesity and COVID-19 severity. Statins as the most common lipid regulating drugs have shown favorable effects in various pathophysiological states. Importantly, accumulating observational studies have suggested that statin use is associated with reduced risk of progressing to severe illness and in-hospital death in COVID-19 patients. Possible explanations underlie these protective impacts include their abilities of reducing cholesterol, suppressing viral entry and replication, anti-inflammation and immunomodulatory effects, as well as anti-thrombosis and anti-oxidative properties. Despite these benefits, statin therapies have side effects that should be considered, such as elevated creatinine kinase, liver enzyme and serum glucose levels, which are already elevated in severe COVID-19. Concerns are also raised whether statins interfere with the efficacy of COVID-19 vaccines. Randomized controlled trials are being conducted worldwide to confirm the values of statin use for COVID-19 treatment. Generally, the results suggest no necessity to discontinue statin use, and no evidence suggesting interference between statins and COVID-19 vaccines. However, concomitant administration of statins and COVID-19 antiviral drug Paxlovid may increase statin exposure and the risk of adverse effects, because most statins are metabolized mainly through CYP3A4 which is potently inhibited by ritonavir, a major component of Paxlovid. Therefore, more clinical/preclinical studies are still warranted to understand the benefits, harms and mechanisms of statin use in the context of COVID-19.
Collapse
Affiliation(s)
- Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyao Yan
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China
| | - Jiajian Shi
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Wang
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anlin Peng
- Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Quirós-Fernández R, López-Plaza B, Bermejo LM, Palma Milla S, Zangara A, Candela CG. Oral Supplement Containing Hydroxytyrosol and Punicalagin Improves Dyslipidemia in an Adult Population without Co-Adjuvant Treatment: A Randomized, Double-Blind, Controlled and Crossover Trial. Nutrients 2022; 14:nu14091879. [PMID: 35565844 PMCID: PMC9103949 DOI: 10.3390/nu14091879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
Hydroxytyrosol (HT) and punicalagin (PC) exert cardioprotective and antiatherosclerotic effects. This study evaluated the effect of an oral supplement containing HT and PC (SAx) on dyslipidemia in an adult population. A randomized, double-blind, controlled, crossover trial was conducted over a 20-week period. SAx significantly reduced the plasma levels of triglycerides (TG) in subjects with hypertriglyceridemia (≥150 mg/dL) (from 200.67 ± 51.38 to 155.33 ± 42.44 mg/dL; p < 0.05), while no such effects were observed in these subjects after the placebo. SAx also significantly decreased the plasma levels of low-density lipoprotein cholesterol (LDL-C) in subjects with high plasma levels of LDL-C (≥160 mg/dL) (from 179.13 ± 16.18 to 162.93 ± 27.05 mg/dL; p < 0.01), while no such positive effect was observed with the placebo. In addition, the placebo significantly reduced the plasma levels of high-density lipoprotein cholesterol (HDL-C) in the total population (from 64.49 ± 12.65 to 62.55 ± 11.57 mg/dL; p < 0.05), while SAx significantly increased the plasma levels of HDL-C in subjects with low plasma levels of HDL-C (<50 mg/dL) (from 44.25 ± 3.99 to 48.00 ± 7.27 mg/dL; p < 0.05). In conclusion, the supplement containing HT and PC exerted antiatherosclerotic and cardio-protective effects by considerably improving dyslipidemia in an adult population, without co-adjuvant treatment or adverse effects.
Collapse
Affiliation(s)
- Rebeca Quirós-Fernández
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
- Correspondence: (R.Q.-F.); (B.P.-L.)
| | - Bricia López-Plaza
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
- Correspondence: (R.Q.-F.); (B.P.-L.)
| | - Laura M. Bermejo
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
| | - Samara Palma Milla
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain; (S.P.M.); (C.G.C.)
| | - Andrea Zangara
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122, Australia;
- Euromed S.A., C/Rec de Dalt, 21-23, Pol. Ind. Can Magarola, 08100 Mollet del Valles, Spain
| | - Carmen Gómez Candela
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain; (S.P.M.); (C.G.C.)
| |
Collapse
|
18
|
Xiong A, Hu Z, Zhou S, Qiang Y, Song Z, Chen H, Xiang Q, Zhang Y, Cao Y, Cui H, Luo J, Wang Y, Yang Y, Cui B, Li M, Shuai S. Cardiovascular events in adult polymyositis and dermatomyositis: a meta-analysis of observational studies. Rheumatology (Oxford) 2021; 61:2728-2739. [PMID: 34791063 DOI: 10.1093/rheumatology/keab851] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/11/2021] [Accepted: 11/06/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES We aimed to review whether polymyositis (PM) and dermatomyositis (DM) patients have an increased cardiovascular (CV) risk, including ischaemic heart disease (IHD), cerebrovascular accidents (CVA), and venous thromboembolism (VTE). METHODS We searched PubMed, Embase, and the Cochrane database for relevant studies from inception to February 2021. RESULTS Twenty-two studies comprising 25,433 patients were included. With PM/DM versus general populations, the risk was significantly increased for CV events (relative risk (RR)=2.37, 95%CI:1.86-3.02). The RR of CV events for males with PM/DM was higher than for females (RR = 1.43; 95%CI:1.17-1.74). PM/DM patients followed for one to five years had a significantly higher CV risk than those followed for five to ten years (RR = 3.51, 95%CI:1.95-6.32). The risk was increased for North Americans (RR = 4.28, 95%CI:2.57-7.11), Europeans (RR = 2.29, 95%CI:1.58-3.31), and Asians (RR = 2.03, 95%CI:1.41-2.90). Our meta-analysis found that the elevated CV event risk was related to PM (RR = 2.35, 95%CI:1.51-3.66) and DM (RR = 2.55, 95%CI:1.66-3.93). Subgroup analyses showed that the risk was significantly increased for IHD (RR = 1.76, 95%CI:1.40-2.21), CVA morbidity (RR = 1.31, 95%CI:1.03-1.67), and ischaemic stroke (IS) (RR = 1.47, 95%CI:1.26-1.73), with no statistically significant increased risk of haemorrhagic stroke mortality (RR = 1.43, 95%CI:0.92-2.21). The CV event risk was increased for VTE (RR = 4.60, 95%CI:3.17-6.66), deep venous thrombosis (DVT) (RR = 5.53, 95%CI:3.25-9.39), and pulmonary embolism (PE) (RR = 5.26, 95%CI:2.62-10.55). CONCLUSION This meta-analysis found that PM/DM patients had a ∼2.37 times increased CV risk, particularly males diagnosed in the previous five years. PM/DM may be an independent risk factor for developing IHD, IS, DVT, and PE.
Collapse
Affiliation(s)
- Anji Xiong
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China.,Inflammation and Immunology Key Laboratory of Nanchong City, Sichuan, China
| | - Ziyi Hu
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Shifeng Zhou
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Yiying Qiang
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhuoyao Song
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Huini Chen
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Qilang Xiang
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Yan Zhang
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuzi Cao
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Hongxu Cui
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Jie Luo
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Ye Wang
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuan Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Beibei Cui
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Menglan Li
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China.,Inflammation and Immunology Key Laboratory of Nanchong City, Sichuan, China
| | - Shiquan Shuai
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China.,Inflammation and Immunology Key Laboratory of Nanchong City, Sichuan, China
| |
Collapse
|
19
|
Zhu Y, Chiang C, Wang L, Brock G, Milks MW, Cao W, Zhang P, Zeng D, Donneyong M, Li L. A multistate transition model for statin-induced myopathy and statin discontinuation. CPT Pharmacometrics Syst Pharmacol 2021; 10:1236-1244. [PMID: 34562311 PMCID: PMC8520747 DOI: 10.1002/psp4.12691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/10/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
The overarching goal of this study was to simultaneously model the dynamic relationships among statin exposure, statin discontinuation, and potentially statin-related myopathic outcomes. We extracted data from the Indiana Network of Patient Care for 134,815 patients who received statin therapy between January 4, 2004, and December 31, 2008. All individuals began statin treatment, some discontinued statin use, and some experienced myopathy and/or rhabdomyolysis while taking the drug or after discontinuation. We developed a militate model to characterize 12 transition probabilities among six different states defined by use or discontinuation of statin and its associated myopathy or rhabdomyolysis. We found that discontinuation of statin therapy was common and frequently early, with 44.4% of patients discontinuing therapy after 1 month, and discontinuation is a strong indicator for statin-induced myopathy (risk ratio, 10.8; p < 0.05). Women more likely than men (p < 0.05) and patients aged 65 years and older had a higher risk than those aged younger than 65 years to discontinue statin use or experience myopathy. In conclusion, we introduce an innovative multistate model that allows clear depiction of the relationship between statin discontinuation and statin-induced myopathy. For the first time, we have successfully demonstrated and quantified the relative risk of myopathy between patients who continued and discontinued statin therapy. Age and sex were two strong risk factors for both statin discontinuation and incident myopathy.
Collapse
Affiliation(s)
- Yuxi Zhu
- Division of BiostatisticsCollege of Public HealthThe Ohio State UniversityColumbusOhioUSA
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Chien‐Wei Chiang
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Lei Wang
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Guy Brock
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - M. Wesley Milks
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Weidan Cao
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Pengyue Zhang
- BiostatisticsSchool of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Donglin Zeng
- Department of BiostatisticsUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Macarius Donneyong
- Division of Pharmacy Practice and ScienceCollege of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Lang Li
- Department of Biomedical InformaticsCollege of MedicineThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
20
|
Lee SA, Kim W, Hong TJ, Ahn Y, Kim MH, Hong SJ, Kim BS, Kim SY, Chae IH, Kim BJ, Rhee MY, Shin JH, Kang TS, Cho JM, Kim JS, Lee CW. Effects of Fixed-dose Combination of Low-intensity Rosuvastatin and Ezetimibe Versus Moderate-intensity Rosuvastatin Monotherapy on Lipid Profiles in Patients With Hypercholesterolemia: A Randomized, Double-blind, Multicenter, Phase III Study. Clin Ther 2021; 43:1573-1589. [PMID: 34429197 DOI: 10.1016/j.clinthera.2021.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/25/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE We investigated whether the combination therapy of low-intensity rosuvastatin and ezetimibe is an useful alternative to moderate-intensity rosuvastatin monotherapy in patients requiring cholesterol-lowering therapy. METHODS This was a multicenter randomized, double-blind study to investigate the safety and efficacy of a fixed-dose combination of rosuvastatin 2.5 mg and ezetimibe 10 mg (R2.5+E10) compared to those of ezetimibe 10 mg monotherapy (E10), rosuvastatin 2.5 mg (R2.5), and rosuvastatin 5 mg monotherapy (R5) in patients with hypercholesterolemia. A total of 348 patients at 15 centers in Korea were screened, and 279 patients were randomized to different groups in the study. Clinical and laboratory examinations were performed at baseline and 4 and 8 weeks after intervention. The primary endpoint was the percentage change of low-density lipoprotein (LDL) cholesterol levels at the 8-week follow-up. FINDINGS Baseline characteristics were similar among the four groups. There were significant changes in lipid profiles at the 8-week follow-up. A greater decrease in the LDL cholesterol levels (primary endpoint) were found in the R2.5+E10 group (-45.7±18.6%) than in the E10 group (-16.7±14.7%, p<0.0001), R2.5 group (-32.6±15.1%, p<0.0001), and R5 group (-38.9±13.9%, p=0.0003). Similar outcomes were observed regarding the decrease in total cholesterol, non-high-density lipoprotein (HDL) cholesterol, and apolipoprotein B protein. In addition, changes in the triglyceride and HDL levels in the R2.5+E10 group were significantly different compared with those in the E10 group; however, the changes were similar to those in the other treatment groups. In patients with low and moderate risk, all patients achieved the target LDL cholesterol levels in the R2.5+E10 group (100%) compared to 13.0% in the E10 group, 47.6% in the R2.5 group, and 65.2% in the R5 group. Adverse effects were rare and similar in the four groups. IMPLICATIONS Fixed-dose combination of low-intensity rosuvastatin and ezetimibe was more effective in lowering LDL cholesterol and achieving LDL cholesterol goals than moderate-intensity rosuvastatin monotherapy. These findings suggest that the combination therapy of low-intensity rosuvastatin and ezetimibe is an useful alternative to moderate-intensity rosuvastatin monotherapy for cholesterol management, particularly in patients with low and moderate risk. ClinicalTrials.gov identifier: NCT04652349.
Collapse
Affiliation(s)
- Seung-Ah Lee
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Weon Kim
- Department of Cardiovascular Medicine, Kyung Hee University Hospital, Seoul, Republic of Korea
| | - Taek Jong Hong
- Division of Cardiology, Department of Internal Medicine, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Youngkeun Ahn
- Division of Cardiology, Department of Internal Medicine, Cardiovascular Center, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Moo Hyun Kim
- Division of Cardiology, Department of Internal Medicine, Dong-A University Medical Center, Busan, Republic of Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Bong Sik Kim
- Department of Internal Medicine, Hyundae General Hospital, Gyeonggi-do, Republic of Korea
| | - Seok Yeon Kim
- Department of Cardiovascular Center, Seoul Medical Center, Seoul, Republic of Korea
| | - In-Ho Chae
- Cardiovascular Center, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Byung Jin Kim
- Division of Cardiology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Moo-Yong Rhee
- Cardiovascular Center, Dongguk University Ilsan Hospital, Gyeonggi-do, Republic of Korea
| | - Joon Han Shin
- Division of Cardiology, Ajou University Medical Center, Gyeonggi-do, Republic of Korea
| | - Tae Soo Kang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Dankook University Hospital, Dankook University School of Medicine, Chungcheongnam-do, Republic of Korea
| | - Jin Man Cho
- Cardiovascular Center, Kyunghee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Jung-Sun Kim
- Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Cheol Whan Lee
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Steinmeyer J, Flechtenmacher J. Drug-induced Myopathies. ZEITSCHRIFT FUR ORTHOPADIE UND UNFALLCHIRURGIE 2021; 161:175-181. [PMID: 34320661 DOI: 10.1055/a-1488-6912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Differential diagnosis of muscle pain and weakness is extensive, including neurological, vertebral, arthrogenic, vascular, traumatic, immunological, endocrine, genetic and infectious aetiologies, as well as medication or toxin-related causes. Muscles are highly sensitive to a large number of drugs, especially with high doses. Although many drug classes can cause toxic myopathy, a significant number of cases are caused by lipid-lowering drugs, long-term use of corticosteroids, and, most often, alcohol misuse. Some drug interactions, e.g. those that are metabolised via the enzyme CYP3A4, can increase the serum levels of the drugs and drug-induced toxicity. A careful history of patient's drug and alcohol consumption is therefore vital. Clinical symptoms depend on the drug, dosage and patient's sensitivity. They can vary from asymptomatic increase in serum levels of creatine kinase, mild myalgia and cramps to muscle weakness, rhabdomyolysis, kidney failure and even death. The pathogenesis is often only partially known and multifactorial. Toxic myopathy is often reversible once the drug is discontinued, alternative drug therapy is started or a different dosage regimen is chosen. Complications such as acute kidney failure must be avoided, and analgesic therapy may be indicated.
Collapse
Affiliation(s)
- Jürgen Steinmeyer
- Laboratory for Experimental Orthopaedics, Dept. of Orthopaedics, Justus Liebig University Giessen, Giessen, Germany
| | - Johannes Flechtenmacher
- Professional Association for Orthopaedic and Trauma Surgery (BVOU), Berlin, Germany.,Ortho-Zentrum, Orthopaedic Group Practice at Ludwigsplatz, Karlsruhe, Germany
| |
Collapse
|
22
|
Casula M, Gazzotti M, Bonaiti F, OImastroni E, Arca M, Averna M, Zambon A, Catapano AL. Reported muscle symptoms during statin treatment amongst Italian dyslipidaemic patients in the real-life setting: the PROSISA Study. J Intern Med 2021; 290:116-128. [PMID: 33259671 PMCID: PMC8359216 DOI: 10.1111/joim.13219] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/14/2020] [Accepted: 11/05/2020] [Indexed: 12/31/2022]
Abstract
AIM Statin-associated muscle symptoms (SAMS) are a major determinant of poor treatment adherence and/or discontinuation, but a definitive diagnosis of SAMS is challenging. The PROSISA study was an observational retrospective study aimed to assess the prevalence of reported SAMS in a cohort of dyslipidaemic patients. METHODS Demographic/anamnestic data, biochemical values and occurrence of SAMS were collected by 23 Italian Lipid Clinics. Adjusted logistic regression was performed to estimate odds ratio (OR) and 95% confidence intervals for association between probability of reporting SAMS and several factors. RESULTS Analyses were carried out on 16 717 statin-treated patients (mean ± SD, age 60.5 ± 12.0 years; 52.1% men). During statin therapy, 9.6% (N = 1599) of patients reported SAMS. Women and physically active subjects were more likely to report SAMS (OR 1.23 [1.10-1.37] and OR 1.35 [1.14-1.60], respectively), whist age ≥ 65 (OR 0.79 [0.70-0.89]), presence of type 2 diabetes mellitus (OR 0.62 [0.51-0.74]), use of concomitant nonstatin lipid-lowering drugs (OR 0.87 [0.76-0.99]), use of high-intensity statins (OR 0.79 [0.69-0.90]) and use of potential interacting drugs (OR 0.63 [0.48-0.84]) were associated with lower probability of reporting SAMS. Amongst patients reporting SAMS, 82.2% underwent dechallenge (treatment interruption) and/or rechallenge (change or restart of statin therapy), with reappearance of muscular symptoms in 38.4% (3.01% of the whole cohort). CONCLUSIONS The reported prevalence of SAMS was 9.6% of the whole PROSISA cohort, but only a third of patients still reported SAMS after dechallenge/rechallenge. These results emphasize the need for a better management of SAMS to implement a more accurate diagnosis and treatment re-evaluation.
Collapse
Affiliation(s)
- M Casula
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,IRCCS MultiMedica, Milan, Italy
| | - M Gazzotti
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - F Bonaiti
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - E OImastroni
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - M Arca
- Department of Translational and Precision Medicine, Unit of Internal Medicine and Metabolic Diseases, Sapienza University, Rome, Italy
| | - M Averna
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - A Zambon
- IRCCS MultiMedica, Milan, Italy.,Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - A L Catapano
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,IRCCS MultiMedica, Milan, Italy
| | | |
Collapse
|
23
|
Watanabe LM, Navarro AM, Seale LA. Intersection between Obesity, Dietary Selenium, and Statin Therapy in Brazil. Nutrients 2021; 13:2027. [PMID: 34204631 PMCID: PMC8231251 DOI: 10.3390/nu13062027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 01/14/2023] Open
Abstract
Obesity is among the most alarming health concerns, impacting public health and causing a socioeconomic challenge, especially in developing countries like Brazil, where approximately one quart of the population presents obesity. As an established risk factor for numerous comorbidities with a multifactorial etiology, obesity is a consequence of energy-dense overfeeding, however with significant undernourishment, leading to excessive adipose tissue accumulation and dysfunction, dyslipidemia, and micronutrient deficiencies. About 60% of patients with obesity take statins, a cholesterol-lowering medication, to curb dyslipidemia, with ~10% of these patients presenting various myopathies as side effects. Statins act upon the rate-limiting enzyme of cholesterol biosynthesis in the liver, which is a pathway providing intermediates to the synthesis of selenoproteins, i.e., enzymes containing the micronutrient selenium. Statins have been postulated to negatively impact selenoprotein synthesis, particularly in conditions of selenium deficiency, and potentially implicated in the myopathies occurring as side effects of statins. The Brazilian population is prone to selenium deficiency, hence could be considered more susceptible to statin side effects. This review examines the specific consequences to the Brazilian population of the harmful intersection between obesity development and concomitant micronutrient deficiencies, particularly selenium, combined with statin treatment in the context of nutrition in Brazil.
Collapse
Affiliation(s)
- Ligia M. Watanabe
- Department of Health Sciences, Division of Nutrition and Metabolism, Ribeirão Preto Medical School, University of São Paulo—FMRP/USP, Ribeirão Preto 14040-900, SP, Brazil; (L.M.W.); (A.M.N.)
| | - Anderson M. Navarro
- Department of Health Sciences, Division of Nutrition and Metabolism, Ribeirão Preto Medical School, University of São Paulo—FMRP/USP, Ribeirão Preto 14040-900, SP, Brazil; (L.M.W.); (A.M.N.)
| | - Lucia A. Seale
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
24
|
SACRED: Effect of simvastatin on hepatic decompensation and death in subjects with high-risk compensated cirrhosis: Statins and Cirrhosis: Reducing Events of Decompensation. Contemp Clin Trials 2021; 104:106367. [PMID: 33771685 PMCID: PMC8422958 DOI: 10.1016/j.cct.2021.106367] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/21/2021] [Accepted: 03/20/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIMS The development of decompensation in cirrhosis demarcates a marked change in the natural history of chronic liver disease. HMG-CoA reductase inhibitors (statins) exert pleiotropic effects that reduce inflammation and fibrosis as well as improve vascular reactivity. Retrospective studies uniformly have associated statin utilization with improved outcomes for patients with cirrhosis. Prospective human studies have shown that statins reduce portal hypertension and reduce death in patients with decompensated cirrhosis after variceal hemorrhage when added to standard therapy with an acceptable safety profile. This proposal aims to extend these findings to demonstrate that simvastatin reduces incident hepatic decompensation events among cirrhotic patients at high risk for hepatic decompensation. METHODS We will perform the SACRED Trial (NCT03654053), a phase III, prospective, multi-center, double-blind, randomized clinical trial at 11 VA Medical Centers. Patients with compensated cirrhosis with clinically significant portal hypertension will be stratified based upon the concomitant use of nonselective beta-blockers and randomized to simvastatin 40 mg/day versus placebo for up to 24 months. Patients will be observed for the development of hepatic decompensation (variceal hemorrhage, ascites, encephalopathy), hepatocellular carcinoma, liver-related death, death from any cause, and/or complications of statin therapy. Ancillary studies will evaluate patient-reported outcomes and pharmacogenetic corollaries of safety and/or efficacy. CONCLUSION Statins have a long track-record of safety and tolerability. This class of medications is generic and inexpensive, and thus, if the hypothesis is proven, there will be few barriers to widespread acceptance of the role of statins to prevent decompensation in patients with compensated cirrhosis. ClinicalTrials.gov Identifier: NCT03654053.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Statins are highly effective therapies for reducing low-density lipoprotein cholesterol and preventing cardiovascular events. However, many patients taking statins experience statin-associated muscle symptoms. In the current manuscript, we review algorithms to define statin intolerance and approaches to optimize cardiovascular risk reduction and reduce the nocebo effect among individuals reporting statin-associated muscle pain. RECENT FINDINGS Patients with statin intolerance have a higher cardiovascular event risk. These data underscore the need to apply clinical strategies that improve treatment utilization and adherence of patients experiencing statin-related side effects. Recent data have shown that the nocebo effect is frequent with statin therapy. This may be explained by the high frequency of muscle symptoms in the general population and media misinformation. When statins even at a low dosage are not tolerated other therapies can be used such as fibrate, ezetimibe nutraceuticals and antiPCSK9 antibodies. Recent data have identified other alternative therapeutic strategies such as bempedoic acid. SUMMARY There are multiple strategies for the management of statin-intolerance, both pharmacological and nonpharmacological. Patient involvement in the justification of statin treatment indication and therapeutic choice is the first step to overcome misbelief and reduce nocebo effect.
Collapse
Affiliation(s)
- Antonio Gallo
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | | | | |
Collapse
|
26
|
Mauvais-Jarvis F, Berthold HK, Campesi I, Carrero JJ, Dakal S, Franconi F, Gouni-Berthold I, Heiman ML, Kautzky-Willer A, Klein SL, Murphy A, Regitz-Zagrosek V, Reue K, Rubin JB. Sex- and Gender-Based Pharmacological Response to Drugs. Pharmacol Rev 2021; 73:730-762. [PMID: 33653873 PMCID: PMC7938661 DOI: 10.1124/pharmrev.120.000206] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In humans, the combination of all sex-specific genetic, epigenetic, and hormonal influences of biologic sex produces different in vivo environments for male and female cells. We dissect how these influences of sex modify the pharmacokinetics and pharmacodynamics of multiple drugs and provide examples for common drugs acting on specific organ systems. We also discuss how gender of physicians and patients may influence the therapeutic response to drugs. We aim to highlight sex as a genetic modifier of the pharmacological response to drugs, which should be considered as a necessary step toward precision medicine that will benefit men and women. SIGNIFICANCE STATEMENT: This study discusses the influences of biologic sex on the pharmacokinetics and pharmacodynamics of drugs and provides examples for common drugs acting on specific organ systems. This study also discusses how gender of physicians and patients influence the therapeutic response to drugs.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Heiner K Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ilaria Campesi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Juan-Jesus Carrero
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Santosh Dakal
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Flavia Franconi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ioanna Gouni-Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Mark L Heiman
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Alexandra Kautzky-Willer
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Sabra L Klein
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Anne Murphy
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Vera Regitz-Zagrosek
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Karen Reue
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Joshua B Rubin
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| |
Collapse
|
27
|
Risk Factors Associated with Statin-Associated Muscle Symptoms in Patients Attending a Specialized Regional Lipid Clinic. J Lipids 2021; 2021:8882706. [PMID: 33791130 PMCID: PMC7997746 DOI: 10.1155/2021/8882706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Background Statin-associated muscle symptoms (SAMS) are the major side effects reported for statins. Data from previous studies suggest that 7-29% of patients on statin had associated muscle symptoms. In the UK, there is a lack of corresponding data on SAMS and factors associated with the development of SAMS. Objective This analysis is aimed at establishing the prevalence of SAMS and identifying major contributory risk factors in patients attending a lipid clinic. Methods Clinical records of 535 consecutive patients, who visited the lipid clinic in the University Hospitals of Leicester, were studied retrospectively between 2009 and 2012. SAMS were defined by the presence of muscle symptoms with two or more different statins. Patients who reported muscle symptoms to statin with one or no rechallenge were excluded. The association of SAMS with clinical characteristics such as age and BMI, sex, smoking, excess alcohol, comorbidities, and medications was tested for statistical significance. A binomial logistic regression model was applied to adjust for risk factors significantly associated with SAMS. Results The prevalence of SAMS was found to be 11%. On unadjusted analysis, the mean age of patients who had SAMS was significantly higher than those without SAMS (59.4 ± 10.5 years vs. 50.3 ± 13.4 years, respectively, P < 0.001). Nonsmokers were more likely to develop SAMS in comparison to active smokers (P = 0.037). Patients taking antihypertensive medications were more likely to develop SAMS (P = 0.010). In binomial logistic regression analysis, only age was positively and significantly associated with SAMS after adjusting for other risk factors (β = 0.054, P = 0.001). Conclusion To the best of our knowledge, this study is the largest cohort of patients with SAMS in the United Kingdom. Our data suggest that the prevalence of SAMS is 11% and increased age is a risk factor associated with the development of SAMS in our cohort of patients.
Collapse
|
28
|
Oh M, Kim H, Shin EW, Sung C, Kim DH, Moon DH, Lee JS, Lee PH, Lee SW, Lee CW. Statin/ezetimibe combination therapy vs statin monotherapy for carotid atherosclerotic plaque inflammation. Medicine (Baltimore) 2021; 100:e25114. [PMID: 33725908 PMCID: PMC7969286 DOI: 10.1097/md.0000000000025114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/17/2021] [Indexed: 01/05/2023] Open
Abstract
It remains uncertain whether statin/ezetimibe combination therapy serves as a useful and equivalent alternative to statin monotherapy for reducing atherosclerotic plaque inflammation. The aim of the present study was to compare the effects of statin/ezetimibe combination therapy and statin monotherapy on carotid atherosclerotic plaque inflammation using 18F-fluorodeoxyglucose (18FDG) positron emission tomography (PET)/computed tomography (CT) imaging. Data were pooled from 2 clinical trials that used serial 18FDG PET/CT examination to investigate the effects of cholesterol-lowering therapy on carotid atherosclerotic plaque inflammation. The primary outcome was the percent change in the target-to-background ratio (TBR) of the index vessel in the most diseased segment (MDS) at 6-month follow-up. Baseline characteristics were largely similar between the 2 groups. At the 6-month follow-up, the MDS TBR of the index vessel significantly decreased in both groups. The percent change in the MDS TBR of the index vessel (primary outcome) did not differ significantly between the 2 groups (-8.41 ± 15.9% vs -8.08 ± 17.0%, respectively, P = .936). Likewise, the percent change in the whole vessel TBR of the index vessel did not differ significantly between the 2 groups. There were significant decreases in total and LDL cholesterol levels in both groups at follow-up (P < .001). There were no significant correlations between the percent changes in MDS TBR of the index vessel, changes in the lipid, and high-sensitive C-reactive protein levels. The reduction in carotid atherosclerotic plaque inflammation by statin/ezetimibe combination therapy was equivalent to that by the statin monotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ji Sung Lee
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
29
|
Mizus MC, Tiniakou E. Lipid-lowering Therapies in Myositis. Curr Rheumatol Rep 2020; 22:70. [PMID: 32845379 PMCID: PMC7986053 DOI: 10.1007/s11926-020-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW The use of lipid-lowering therapies in patients with idiopathic inflammatory myopathies (IIM) is complicated and there are no guidelines for diagnosing, monitoring, or treating atherosclerotic cardiovascular disease (ASCVD) in this group of patients. RECENT FINDINGS The use of lipid-lowering therapies, especially statins, is recommended in patients with increased risk for ASCVD, which includes patients with inflammatory diseases, based on recent American College of Cardiology/American Heart Association (ACC/AHA) guidelines for ASCVD management. There is accumulating evidence that patients with IIM are at increased risk for ASCVD, similar to other inflammatory diseases. Lipid-lowering therapies have side effects that may be pronounced or confounding in myositis patients, potentially limiting their use. Statins are specifically contraindicated in patients with anti 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) antibodies. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have been shown to be safe and potentially beneficial in patients with IIM. Here, we propose a framework for (1) ASCVD risk assessment and treatment based on ACC/AHA ASCVD primary prevention guidelines; (2) myositis disease monitoring while undergoing lipid-lowering therapy; and (3) management of statin intolerance, including, indications for the use of PCSK9 inhibitors.
Collapse
Affiliation(s)
- Marisa C Mizus
- Department of Medicine, Division of Rheumatology, Mason Lord, Center Tower, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Baltimore, MD, 21224, USA.
| | - Eleni Tiniakou
- Department of Medicine, Division of Rheumatology, Mason Lord, Center Tower, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Baltimore, MD, 21224, USA.
| |
Collapse
|
30
|
|
31
|
Streicher C, Daulange A, Madranges N, Vayre L. Severe rhabdomyolysis induced by possible drug-drug interaction between Ribociclib and Simvastatin. J Oncol Pharm Pract 2020; 27:722-726. [PMID: 32727321 DOI: 10.1177/1078155220945365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Drug-drug interactions with cyclin-dependent kinases inhibitors 4 and 6 (CDK4/6) are known and should be taken into account. CASE REPORT A 68-year-old woman, on prior Simvastatin therapy, developed severe rhabdomyolysis after three weeks of Ribociclib initiation. She showed general weakness with mobility problems and was admitted to our hospital. MANAGEMENT AND OUTCOME Ribociclib and Simvastatin were discontinued and the patient received intensive intravenous hydration. She finally recovered her mobility after two weeks. DISCUSSION We hypothesize that Simvastatin induced rhabdomyolysis by possible interaction with Ribociclib. Ribociclib is a strong inhibitor of CYP 3A4 and a potential inhibitor of OATP1B1 membrane transporter. Simvastatin plasma concentration may reach toxic levels due to Ribociclib inhibition. To assess the relevance of our hypothesis, we used the Drug Interaction Scale. With a total score of 7, the interaction is considered as "probable." Because of the high risk of severe rhabdomyolysis, the concomitant use of Simvastatin with Ribociclib should be avoided or otherwise careful monitoring of creatine kinase is warranted.
Collapse
Affiliation(s)
- Caroline Streicher
- Unité de préparation et de contrôle pharmaceutique, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France.,Service d'oncologie, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France
| | - Annick Daulange
- Unité de préparation et de contrôle pharmaceutique, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France.,Service d'oncologie, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France
| | - Nicolas Madranges
- Service d'oncologie, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France
| | - Laure Vayre
- Service d'oncologie, Centre Hospitalier de Brive La Gaillarde, Brive La Gaillarde Cedex, France
| |
Collapse
|
32
|
Tan A, Fraser C, Khoo P, Watson S, Ooi K. Statins in Neuro-ophthalmology. Neuroophthalmology 2020; 45:219-237. [PMID: 34366510 PMCID: PMC8312600 DOI: 10.1080/01658107.2020.1755872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/05/2020] [Accepted: 04/11/2020] [Indexed: 10/24/2022] Open
Abstract
Statins are effective and well-tolerated hypolipidaemic agents which have been increasingly studied for their pleiotropic immunomodulatory and anti-inflammatory effects. Statins have potential therapeutic benefit in a range of neuro-ophthalmological conditions but may also induce or exacerbate certain neurological disorders. This literature review examines evidence from clinical and in vitro studies assessing the effects of statins in myasthenia gravis, myopathy, multiple sclerosis, neuromyelitis optica, idiopathic intracranial hypertension (pseudotumour cerebri), migraine, giant cell arteritis, Bell's palsy, ocular ischaemia, stroke, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Alvin Tan
- Discipline of Ophthalmology, Faculty of Health and Medicine, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Clare Fraser
- Discipline of Ophthalmology, Faculty of Health and Medicine, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Pauline Khoo
- Discipline of Ophthalmology, Faculty of Health and Medicine, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Stephanie Watson
- Discipline of Ophthalmology, Faculty of Health and Medicine, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Kenneth Ooi
- Discipline of Ophthalmology, Faculty of Health and Medicine, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Shahid A, Akbar MA, Ariff M. Features of Rhabdomyolysis Secondary to Immobility and Statin-Induced Myopathy in a 70-Year-Old Female. Cureus 2020; 12:e8330. [PMID: 32617207 PMCID: PMC7325407 DOI: 10.7759/cureus.8330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Treatment with statins requires close monitoring of serum creatine kinase levels to prevent myopathy, which is a rare but potentially serious dose-dependent adverse effect of these drugs. Statins are cholesterol-lowering drugs that are among the most prescribed drugs worldwide and are considered effective in reducing the risk of major cardiovascular events. Although statins are generally well-tolerated, myopathies are a rare but known adverse event, ranging from muscle pain to very rare cases of life-threatening rhabdomyolysis. In this report, we aim to highlight the features of rhabdomyolysis secondary to immobility and statin-induced myopathy.
Collapse
Affiliation(s)
- Arzoo Shahid
- Internal Medicine, University of Alberta Hospital Edmonton, Alberta, CAN
| | - Mobassir A Akbar
- Family Medicine, University of Alberta Hospital Edmonton, Alberta, CAN.,Ears, Nose and Throat, Jinnah Post Graduate Medical Center, Karachi, PAK
| | - Madiha Ariff
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| |
Collapse
|
34
|
Comparison of High-Dose Rosuvastatin Versus Low-Dose Rosuvastatin Plus Ezetimibe on Carotid Atherosclerotic Plaque Inflammation in Patients with Acute Coronary Syndrome. J Cardiovasc Transl Res 2020; 13:900-907. [DOI: 10.1007/s12265-020-10009-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/17/2020] [Indexed: 02/03/2023]
|
35
|
The impact of statins on physical activity and exercise capacity: an overview of the evidence, mechanisms, and recommendations. Eur J Appl Physiol 2020; 120:1205-1225. [PMID: 32248287 DOI: 10.1007/s00421-020-04360-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Statins are among the most widely prescribed medications worldwide. Considered the 'gold-standard' treatment for cardiovascular disease (CVD), statins inhibit HMG-CoA reductase to ultimately reduce serum LDL-cholesterol levels. Unfortunately, the main adverse event of statin use is the development of muscle-associated problems, referred to as SAMS (statin-associated muscle symptoms). While regular moderate physical activity also decreases CVD risk, there is apprehension that physical activity may induce and/or exacerbate SAMS. While much work has gone into identifying the epidemiology of SAMS, only recent research has focused on the extent to which these muscle symptoms are accompanied by functional declines. The purpose of this review is to provide an overview of possible mechanisms underlying SAMS and summarize current evidence regarding the relationship between statin treatment, physical activity, exercise capacity, and SAMS development. METHODS PubMed and Google Scholar databases were used to search the most relevant and up-to-date peer-reviewed research on the topic. RESULTS The mechanism(s) behind SAMS, including altered mitochondrial metabolism, reduced coenzyme Q10 levels, reduced vitamin D levels, impaired calcium homeostasis, elevated extracellular glutamate, and genetic polymorphisms, still lack consensus and remain up for debate. Our summation of the evidence leads us to suggest that the etiology of SAMS development is likely multifactorial. Our review also demonstrates that there is limited evidence for statins impairing exercise adaptations or reducing exercise capacity for the majority of the investigated populations. CONCLUSION The available evidence indicates that the benefits of engaging in physical activity while on statin medication largely outweigh the risks.
Collapse
|
36
|
|
37
|
Abstract
OBJECTIVE Statins are a class of drugs that competitively bind to the active site of HMG-CoA reductase enzyme, thereby inhibiting the initial steps in cholesterol synthesis. Originally approved for use in lowering serum cholesterol, a risk factor for developing atherosclerosis and coronary heart disease, statins have subsequently been noted to have myriad extrahepatic effects, including potential effects on cognition, diabetes, breast cancer, bone, and muscle. This narrative review assesses the current state of the science regarding the risks and benefits of statin therapy in women to identify areas where additional research is needed. METHODS Basic and clinical studies were identified by searching PubMed with particular attention to inclusion of female animals, women, randomized controlled trials, and sex-specific analyses. RESULTS Statin therapy is generally recommended to reduce the risk of cardiovascular disease. None of the current clinical guidelines, however, offer sex-specific recommendations for women due to lack of understanding of sex differences and underlying mechanisms of disease processes. In addition, conclusions regarding efficacy of treatments do not consider lipid solubility for the drug, dosing, duration of treatment, interactions with estrogen, or comorbidities. Pleiotropic effects of statins are often derived from secondary analysis of studies with cardiovascular events as primary outcomes. CONCLUSIONS Many of the trials that have established the efficacy and safety of statins were conducted predominantly or entirely in men, with results extrapolated to women. Additional research is needed to guide clinical recommendations specific to women. : Video Summary:http://links.lww.com/MENO/A462.
Collapse
Affiliation(s)
- Stephanie S. Faubion
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Ekta Kapoor
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - Ann M. Moyer
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Howard N. Hodis
- Atherosclerosis Research Unit, Departments of Medicine and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Virginia M. Miller
- Departments of Surgery and Physiology & Biomedical Engineering, Women’s Health Research Center, Mayo Clinic, Rochester, MN
| |
Collapse
|
38
|
Santos TOD, Nascimento MMGD, Nascimento YA, Oliveira GCBD, Martins UCDM, Silva DFD, Oliveira DRD. Drug interactions among older adults followed up in a comprehensive medication management service at Primary Care. EINSTEIN-SAO PAULO 2019; 17:eAO4725. [PMID: 31460618 PMCID: PMC6706225 DOI: 10.31744/einstein_journal/2019ao4725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/27/2019] [Indexed: 11/25/2022] Open
Abstract
Objective: To estimate the prevalence of drug interactions and associated factors among older adults followed up in a Comprehensive Medication Management Service at Primary Care. Methods: Firstly, the Beers criteria 2015 was used to define drug interactions; later, drug interactions proposed by Dumbreck for patients with diabetes, depression, and heart failure were evaluated. The associated factors were assessed by univariate (Pearson's χ2) and multivariate analyses (logistic regression). The significance level of 5% was set for all analyses. Results: The mean age of the studied population was 70.2±7.8 years; 52.2% were between 60 and 69 years, and 61.3% were female. Among the older adults, 94.5% used two or more drugs (condition for the occurrence of drug-drug interaction). The prevalence of drug interaction according to the Beers criteria was 4.9%. After multivariate analysis, diseases of the central nervous system, arrhythmia, number of medications, and female sex were positively associated with drug interaction. The prevalence of drug interaction according to Dumbreck was 27.2%. After multivariate analysis, the number of medications, the presence of heart failure, and Charlson comorbidity index greater than 1 were conditions positively associated with drug interactions. Conclusion: The holistic and individualized approach used in comprehensive medication management services for older patients is important, considering the prevalence of drug interactions and the need to minimize adverse events.
Collapse
Affiliation(s)
- Tayane Oliveira Dos Santos
- Centro de Estudos em Atenção Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Yone Almeida Nascimento
- Centro de Estudos em Atenção Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Danielle Fernandes da Silva
- Centro de Estudos em Atenção Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Djenane Ramalho de Oliveira
- Centro de Estudos em Atenção Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
39
|
Abu Mellal A, Hussain N, Said AS. The clinical significance of statins-macrolides interaction: comprehensive review of in vivo studies, case reports, and population studies. Ther Clin Risk Manag 2019; 15:921-936. [PMID: 31413581 PMCID: PMC6661989 DOI: 10.2147/tcrm.s214938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
The objectives of this article were to review the mechanism and clinical significance of statins-macrolides interaction, determine which combination has the highest risk for the interaction, and identify key patients' risk factors for the interaction in relation to the development of muscle toxicity. A literature review was conducted in PubMed and Embase (1946 to December 2018) using combined terms: statins - as group and individual agents, macrolides - as group and individual agents, drug interaction, muscle toxicity, rhabdomyolysis, CYP3A4 inhibitors, and OAT1B inhibitors, with forward and backward citation tracking. Relevant English language in vivo studies in healthy volunteers, case reports, and population studies were included. The interaction between statins and macrolides depends on the type of statin and macrolide used. The mechanism of the interaction is due to macrolides' inhibition of CYP3A4 isoenzyme and OAT1B transporter causing increased exposure to statins. The correlation of this increased statin's exposure to the development of muscle toxicity could not be established, unless the patient had other risk factors such as advanced age, cardiovascular diseases, renal impairment, diabetes, and the concomitant use of other CYP3A4 inhibitors. Simvastatin, lovastatin, and to lesser extent atorvastatin are the statins most affected by this interaction. Rosuvastatin, fluvastatin, and pravastatin are not significantly affected by this interaction. Telithromycin, clarithromycin, and erythromycin are the most "offending" macrolides, while azithromycin appears to be safe to use with statins. This review presented a clear description of the clinical significance of this interaction in real practice. Also, it provided health care professionals with clear suggestions and recommendations on how to overcome this interaction. In conclusion, understanding the different characteristics of each statin and macrolide, as well as key patients' risk factors, will enable health care providers to utilize both groups effectively without compromising patient safety.
Collapse
Affiliation(s)
- Abdallah Abu Mellal
- College of Health and Human Sciences, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Nadia Hussain
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, UAE
| | - Amira Sa Said
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, UAE
| |
Collapse
|
40
|
Bogman K, Brumm J, Hofmann C, Giraudon M, Niggli M, Sturm-Pellanda C, Sauter A, Sturm S, Mangold B, Schmitt C. Assessment of Drug–Drug Interactions between Taspoglutide, a Glucagon-Like Peptide-1 Agonist, and Drugs Commonly Used in Type 2 Diabetes Mellitus: Results of Five Phase I Trials. Clin Pharmacokinet 2019; 58:1205-1214. [DOI: 10.1007/s40262-019-00757-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Long B, Koyfman A, Gottlieb M. An evidence-based narrative review of the emergency department evaluation and management of rhabdomyolysis. Am J Emerg Med 2019; 37:518-523. [DOI: 10.1016/j.ajem.2018.12.061] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/20/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
|