1
|
Rabinovich-Nikitin I, Liu S, Kirshenbaum LA. Sex-specific considerations in cardiovascular drug therapy. Can J Physiol Pharmacol 2024; 102:523-529. [PMID: 38781601 DOI: 10.1139/cjpp-2024-0040] [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] [Indexed: 05/25/2024]
Abstract
Despite major advances in cardiac research over the past three decades, cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality in women and men worldwide. However, a major challenge for health care providers is that the current guidelines for cardiovascular drug therapies do not consider the impact of sex in the development of treatment plan for optimizing therapies for women. Clinical research in recent years suggests significant pharmacological and pharmacokinetic differences between females and males, which have been attributed in part to differences in body composition, plasma protein binding capacity, drug metabolism, and excretion. Herein, we provide a comprehensive review regarding sex-specific differences and drugs commonly used for CVDs in women and men. Understanding how sex-related differences influence drug efficacy and CVD outcomes is crucial for not only optimizing treatment strategies for women and men but also to encourage the implementation of specific guidelines that address sex difference as a consideration for the treatment of CVDs.
Collapse
Affiliation(s)
- Inna Rabinovich-Nikitin
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Shuangbo Liu
- Section of Cardiology, Department of Medicine, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lorrie A Kirshenbaum
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba , Winnipeg, MB, Canada
| |
Collapse
|
2
|
Feng Z, Li X, Tong WK, He Q, Zhu X, Xiang X, Tang Z. Real-world safety of PCSK9 inhibitors: A pharmacovigilance study based on spontaneous reports in FAERS. Front Pharmacol 2022; 13:894685. [PMID: 36506552 PMCID: PMC9729267 DOI: 10.3389/fphar.2022.894685] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Objective: We aimed to evaluate alirocumab- and evolocumab-related adverse events (AEs) in real-world compared with all other drugs, overall and by gender and age subgroups; we also aimed to compare their risks of cognitive impairment, musculoskeletal disorders and diabetes with various statins and ezetimibe. Methods: We retrospectively extracted AE reports from the FDA Adverse Event Reporting System (FAERS) database during July 2015-June 2021. Disproportionality analyses were performed using reporting odds ratios (RORs) to detect AE signals of alirocumab and evolocumab in the overall population and in different age and gender subgroups, respectively. Results: Compared with all other drugs, both alirocumab and evolocumab had a significant signal in "musculoskeletal and connective tissue disorders" (ROR1 = 2.626, 95% CI 2.552-2.702; ROR2 = 2.575, 95% CI 2.538-2.613). The highest ROR value of 2.311 (95% CI 2.272-2.351) was for "injury, poisoning and procedural complications" and was found in patients aged ≥65 years on evolocumab. The most frequent AEs were "general disorders and administration site conditions" and "musculoskeletal and connective tissue disorders" for all subpopulations. At the preferred term level, the most frequent AE signal was myalgia for alirocumab and injection site pain for evolocumab, overall and by subgroups. Compared with statins/ezetimibe, PCSK9 inhibitors exhibited lower ROR values for adverse events associated with SOC "nervous system disorders", "psychiatric disorders" and "metabolism and nutrition disorders" (all RORs < 1), but mixed results for musculoskeletal disorders. Compared with all other drugs, undocumented AEs, such as acute cardiac event (ROR = 30.0, 95% CI 9.4-95.3) and xanthoma (ROR = 9.3, 95% CI 3.4-25.5), were also reported. Conclusion: Real-world evidence showed that PCSK9 inhibitors were associated with an increased risk of musculoskeletal and connective tissue disorders and general disorders and administration site conditions, overall and by subgroups. Muscle toxicity, injection site reactions, and influenza-like illness were significant AE signals. Compared with various statins and ezetimibe, PCSK9 inhibitors have shown a favorable safety profile in muscle-related events, cognitive impairment and diabetes. Some undocumented AE signals were also reported. Due to the limitations of spontaneous reporting databases, further studies are still needed to establish causality and validate our results.
Collapse
Affiliation(s)
- Zhen Feng
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoye Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wai Kei Tong
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhijia Tang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China,*Correspondence: Zhijia Tang,
| |
Collapse
|
3
|
Abstract
IMPORTANCE Statins are the drug class most commonly used to treat hyperlipidemia. Recently, they have been used during pregnancy for the prevention or treatment of preeclampsia. However, the safety of statin use during pregnancy has been questioned, and the sample sizes of most previous studies have been small. OBJECTIVE To examine the perinatal outcomes among offspring associated with maternal use of statins during pregnancy. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study included 1 443 657 pregnant women 18 years of age or older with their first infant born during the period from January 1, 2004, to December 31, 2014. Data for this study were taken from the Taiwan National Health Insurance Research Database. Statistical analysis was performed from April 7, 2020, to July 31, 2021. EXPOSURES Maternal statin use during pregnancy. MAIN OUTCOMES AND MEASURES Women who have received a diagnosis of hyperlipidemia before pregnancy and who were receiving prescription statins during pregnancy were the statin-exposed group. Data on congenital anomalies, birth weight, gestational age, preterm birth, low birth weight, very low birth weight, fetal distress, and Apgar score were compared between participants with and partcipants without statin exposure during pregnancy. Risk ratios (RRs) were calculated by multivariable analyses using Poisson regression models to adjust for potential confounders. Subgroup analysis was performed to compare offspring of women who used statins for more than 3 months prior to pregnancy and maintained or stopped statin use after pregnancy. RESULTS A total of 469 women (mean [SD] age, 32.6 [5.4] years; mean [SD] gestational age, 38.4 [1.6] weeks) who used statins during pregnancy and 4690 age-matched controls (mean [SD] age, 32.0 [4.9] years; mean [SD] gestational age, 37.3 [2.4] weeks) with no statin exposure during pregnancy were enrolled. After controlling for maternal comorbidities and age, low birth weight was more common among offspring in the statin-exposed group (RR, 1.51 [95% CI, 1.05-2.16]), with a greater chance of preterm birth (RR, 1.99 [95% CI, 1.46-2.71]), and a lower 1-minute Apgar score (RR, 1.83 [95% CI, 1.04-3.20]). Congenital anomalies were not associated with statin exposure during pregnancy. In addition, multivariable analysis showed that there was no association between statin use for periconceptual hyperlipidemia and adverse perinatal outcomes among women who had used statins prior to pregnancy. CONCLUSIONS AND RELEVANCE This study suggests that statins may be safe when used during pregnancy because there was no association with congenital anomalies, but caution is needed because of an increased risk of low birth weight and preterm labor. The data also suggest that statins could be safely used during pregnancy for women with long-term use of statins before pregnancy.
Collapse
Affiliation(s)
- Jui-Chun Chang
- Department of Obstetrics and Gynecology and Women’s Health, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Ju Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wei-Szu Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Ming Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan
- Department of Public Health, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Institute of Public Health and Community Medicine Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Zongo A, Simpson S, Johnson JA, Eurich DT. Effect of a pharmacy comprehensive chronic diseases care plan on use of lipid-lowering drugs among patients with hypertension. J Manag Care Spec Pharm 2021. [DOI: 10.18553/jmcp.2021.27.4.426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Arsène Zongo
- School of Public Health, University of Alberta, Edmonton, Canada; Faculty of Pharmacy, Université Laval, Quebec City, Canada; and Population Health and Optimal Health Practices Research Unit, CHU de Québec, Université Laval Research Centre, Quebec City, Canada
| | - Scot Simpson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | | - Dean T Eurich
- School of Public Health, University of Alberta, Edmonton, Canada
| |
Collapse
|
5
|
Orringer CE, Tokgozoglu L, Maki KC, Ray KK, Saseen JJ, Catapano AL. Transatlantic Lipid Guideline Divergence: Same Data But Different Interpretations. J Am Heart Assoc 2020; 9:e018189. [PMID: 33092440 PMCID: PMC7763403 DOI: 10.1161/jaha.120.018189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite consensus that excessive circulating concentrations of apoB‐lipoproteins is a key driver for the atherosclerotic process and that treatments that low‐density lipoprotein cholesterol lowering by up‐regulation of low‐density lipoprotein cholesterol receptor expression reduces that risk, divergent viewpoints on interpretation of study data have resulted in substantial differences in European and American lipid guideline recommendations. This article explores those differences and highlights the importance of understanding guideline‐based lipid management to improve patient care and reduce the risk of clinical atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Carl E Orringer
- Department of Medicine Cardiovascular Division University of Miami Miller School of Medicine Miami FL
| | | | - Kevin C Maki
- Department of Applied Health Science Indiana University School of Public Health Bloomington IN.,Midwest Biomedical Research Addison IL
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention Department of Primary Care and Public Health Imperial College London United Kingdom
| | - Joseph J Saseen
- Departments of Clinical Pharmacy and Family Medicine Schools of Pharmacy and Medicine University of Colorado Anschutz Medical Campus Aurora CO
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences University of Milan Italy.,IRCCS MultiMedica Sesto S. Giovanni Milan Italy
| |
Collapse
|
6
|
Liu Y, Shen Y, Guo T, Parnell LD, Westerman KE, Smith CE, Ordovas JM, Lai CQ. Statin Use Associates With Risk of Type 2 Diabetes via Epigenetic Patterns at ABCG1. Front Genet 2020; 11:622. [PMID: 32612641 PMCID: PMC7308584 DOI: 10.3389/fgene.2020.00622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 11/13/2022] Open
Abstract
Statin is the medication most widely prescribed to reduce plasma cholesterol levels. Yet, how the medication contributes to diabetes risk and impaired glucose metabolism is not clear. This study aims to examine the epigenetic mechanisms of ABCG1 through which statin use associates with risk of type 2 diabetes. We determined the association between the statin use, DNA methylation at ABCG1 and type 2 diabetes/glycemic traits in the Framingham Heart Study Offspring (FHS, n = 2741), with validation in the Women’s Health Initiative Study (WHI, n = 2020). The causal effect of statin use on the risk of type 2 diabetes was examined using a two-step Mendelian randomization approach. Next, based on transcriptome analysis, we determined the links between the medication-associated epigenetic status of ABCG1 and biological pathways on the pathogenesis of type 2 diabetes. Our results showed that DNA methylation levels at cg06500161 of ABCG1 were positively associated with the use of statin, type 2 diabetes and related traits (fasting glucose and insulin) in FHS and WHI. Two-step Mendelian randomization suggested a causal effect of statin use on type 2 diabetes and related traits through epigenetic mechanisms, specifically, DNA methylation at cg06500161. Our results highlighted that gene expression of ABCG1, ABCA1 and ACSL3, involved in both cholesterol metabolism and glycemic pathways, was inversely associated with statin use, CpG methylation, and diabetic signatures. We concluded that DNA methylation site cg06500161 at ABCG1 is a mediator of the association between statins and risk of type 2 diabetes.
Collapse
Affiliation(s)
- Yuwei Liu
- School of Public Health, Fudan University, Shanghai, China.,Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Yu Shen
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Tao Guo
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Laurence D Parnell
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Kenneth E Westerman
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Chao-Qiang Lai
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| |
Collapse
|
7
|
Abstract
The causal relation between elevated levels of LDL-C and cardiovascular disease has been largely established by experimental and clinical studies. Thus, the reduction of LDL-C levels is a major target for the prevention of cardiovascular disease. In the last decades, statins have been used as the main therapeutic approach to lower plasma cholesterol levels; however, the presence of residual lipid-related cardiovascular risk despite maximal statin therapy raised the need to develop additional lipid-lowering drugs to be used in combination with or in alternative to statins in patients intolerant to the treatment. Several new drugs have been approved which have mechanisms of action different from statins or impact on different lipoprotein classes.
Collapse
|
8
|
Effect of Statins (Zocor) on Oxygen-Dependent Processes in Muscle Tissue and Erythrocytes in Animals with Hypercholesterolemia. ACTA BIOMEDICA SCIENTIFICA 2019. [DOI: 10.29413/abs.2019-4.3.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background. Statin therapy may be accompanied by a toxic effect on skeletal muscle and liver cells, the molecular mechanisms of which have not yet been fully understood.Aim.To clarify the peculiarities of changes in the oxygen transport function of erythrocytes and antioxidant mechanisms in erythrocytes and muscles of animals with prolonged administration of statins (simvastatin).Materials and methods.The study was conducted on outbred male rats, which during the experiment were randomly assigned to following groups: the control group contained animals on a common ration of the vivarium, the group of comparison – animals with induced hypercholesterolemia, the main group – animals with induced hypercholesterolemia treated with simvastatin. In erythrocytes and muscle tissue of animals, indicators characterizing the state of antioxidant defense and carbohydrate metabolism were determined.The results.The experiment showed that the administration of simvastatin to animals was characterized by aggravation of hypoxia due to hypercholesterolemia, as indicated by a sharp increase in the concentration of 2,3-BPG and lactate in erythrocytes of animals, as well as a significant decrease in the activity of G6PD. In the muscle tissue of animals, there was a decrease in the concentration of pyruvic acid and lactate relative to the comparison group, which indicates their intensive participation in metabolic processes.Conclusion.Based on the data obtained, it can be assumed that the peculiarity of the action of statins in erythrocytes is the increased oxygen delivery to the tissues, which in muscles was accompanied by a decrease in the level of oxidized products. At the same time, against the background of the use of statins, despite the positive direction of adaptive reactions, signs of oxidative stress remain, which is documented by the imbalance of the SOD-catalase system and a decrease in the activity of glutathione-dependent reactions
Collapse
|
9
|
Carreras A, Pane LS, Nitsch R, Madeyski-Bengtson K, Porritt M, Akcakaya P, Taheri-Ghahfarokhi A, Ericson E, Bjursell M, Perez-Alcazar M, Seeliger F, Althage M, Knöll R, Hicks R, Mayr LM, Perkins R, Lindén D, Borén J, Bohlooly-Y M, Maresca M. In vivo genome and base editing of a human PCSK9 knock-in hypercholesterolemic mouse model. BMC Biol 2019; 17:4. [PMID: 30646909 PMCID: PMC6334452 DOI: 10.1186/s12915-018-0624-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
Abstract
Background Plasma concentration of low-density lipoprotein (LDL) cholesterol is a well-established risk factor for cardiovascular disease. Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9), which regulates cholesterol homeostasis, has recently emerged as an approach to reduce cholesterol levels. The development of humanized animal models is an important step to validate and study human drug targets, and use of genome and base editing has been proposed as a mean to target disease alleles. Results To address the lack of validated models to test the safety and efficacy of techniques to target human PCSK9, we generated a liver-specific human PCSK9 knock-in mouse model (hPCSK9-KI). We showed that plasma concentrations of total cholesterol were higher in hPCSK9-KI than in wildtype mice and increased with age. Treatment with evolocumab, a monoclonal antibody that targets human PCSK9, reduced cholesterol levels in hPCSK9-KI but not in wildtype mice, showing that the hypercholesterolemic phenotype was driven by overexpression of human PCSK9. CRISPR-Cas9-mediated genome editing of human PCSK9 reduced plasma levels of human and not mouse PCSK9, and in parallel reduced plasma concentrations of total cholesterol; genome editing of mouse Pcsk9 did not reduce cholesterol levels. Base editing using a guide RNA that targeted human and mouse PCSK9 reduced plasma levels of human and mouse PCSK9 and total cholesterol. In our mouse model, base editing was more precise than genome editing, and no off-target editing nor chromosomal translocations were identified. Conclusions Here, we describe a humanized mouse model with liver-specific expression of human PCSK9 and a human-like hypercholesterolemia phenotype, and demonstrate that this mouse can be used to evaluate antibody and gene editing-based (genome and base editing) therapies to modulate the expression of human PCSK9 and reduce cholesterol levels. We predict that this mouse model will be used in the future to understand the efficacy and safety of novel therapeutic approaches for hypercholesterolemia. Electronic supplementary material The online version of this article (10.1186/s12915-018-0624-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alba Carreras
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.,Present Address: Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Luna Simona Pane
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Roberto Nitsch
- Advanced Medicines Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Katja Madeyski-Bengtson
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Michelle Porritt
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Pinar Akcakaya
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Amir Taheri-Ghahfarokhi
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Elke Ericson
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Mikael Bjursell
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Marta Perez-Alcazar
- Pathology Science, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Frank Seeliger
- Pathology Science, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Magnus Althage
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ralph Knöll
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lorenz M Mayr
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.,Present Address: GE Healthcare Life Sciences, The Grove Centre, White Lion Road, Amersham, UK
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Daniel Lindén
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mohammad Bohlooly-Y
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.
| | - Marcello Maresca
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.
| |
Collapse
|
10
|
Carreras A, Pane LS, Nitsch R, Madeyski-Bengtson K, Porritt M, Akcakaya P, Taheri-Ghahfarokhi A, Ericson E, Bjursell M, Perez-Alcazar M, Seeliger F, Althage M, Knöll R, Hicks R, Mayr LM, Perkins R, Lindén D, Borén J, Bohlooly-Y M, Maresca M. In vivo genome and base editing of a human PCSK9 knock-in hypercholesterolemic mouse model. BMC Biol 2019. [PMID: 30646909 DOI: 10.1186/s12915-018-0624-2.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasma concentration of low-density lipoprotein (LDL) cholesterol is a well-established risk factor for cardiovascular disease. Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9), which regulates cholesterol homeostasis, has recently emerged as an approach to reduce cholesterol levels. The development of humanized animal models is an important step to validate and study human drug targets, and use of genome and base editing has been proposed as a mean to target disease alleles. RESULTS To address the lack of validated models to test the safety and efficacy of techniques to target human PCSK9, we generated a liver-specific human PCSK9 knock-in mouse model (hPCSK9-KI). We showed that plasma concentrations of total cholesterol were higher in hPCSK9-KI than in wildtype mice and increased with age. Treatment with evolocumab, a monoclonal antibody that targets human PCSK9, reduced cholesterol levels in hPCSK9-KI but not in wildtype mice, showing that the hypercholesterolemic phenotype was driven by overexpression of human PCSK9. CRISPR-Cas9-mediated genome editing of human PCSK9 reduced plasma levels of human and not mouse PCSK9, and in parallel reduced plasma concentrations of total cholesterol; genome editing of mouse Pcsk9 did not reduce cholesterol levels. Base editing using a guide RNA that targeted human and mouse PCSK9 reduced plasma levels of human and mouse PCSK9 and total cholesterol. In our mouse model, base editing was more precise than genome editing, and no off-target editing nor chromosomal translocations were identified. CONCLUSIONS Here, we describe a humanized mouse model with liver-specific expression of human PCSK9 and a human-like hypercholesterolemia phenotype, and demonstrate that this mouse can be used to evaluate antibody and gene editing-based (genome and base editing) therapies to modulate the expression of human PCSK9 and reduce cholesterol levels. We predict that this mouse model will be used in the future to understand the efficacy and safety of novel therapeutic approaches for hypercholesterolemia.
Collapse
Affiliation(s)
- Alba Carreras
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.,Present Address: Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Luna Simona Pane
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Roberto Nitsch
- Advanced Medicines Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Katja Madeyski-Bengtson
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Michelle Porritt
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Pinar Akcakaya
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Amir Taheri-Ghahfarokhi
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Elke Ericson
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Mikael Bjursell
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden
| | - Marta Perez-Alcazar
- Pathology Science, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Frank Seeliger
- Pathology Science, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Magnus Althage
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ralph Knöll
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lorenz M Mayr
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.,Present Address: GE Healthcare Life Sciences, The Grove Centre, White Lion Road, Amersham, UK
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Daniel Lindén
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mohammad Bohlooly-Y
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.
| | - Marcello Maresca
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 43 183, Gothenburg, Sweden.
| |
Collapse
|
11
|
Pasipoularides A. Implementing genome-driven personalized cardiology in clinical practice. J Mol Cell Cardiol 2018; 115:142-157. [PMID: 29343412 PMCID: PMC5820118 DOI: 10.1016/j.yjmcc.2018.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/04/2018] [Accepted: 01/12/2018] [Indexed: 12/18/2022]
Abstract
Genomics designates the coordinated investigation of a large number of genes in the context of a biological process or disease. It may be long before we attain comprehensive understanding of the genomics of common complex cardiovascular diseases (CVDs) such as inherited cardiomyopathies, valvular diseases, primary arrhythmogenic conditions, congenital heart syndromes, hypercholesterolemia and atherosclerotic heart disease, hypertensive syndromes, and heart failure with preserved/reduced ejection fraction. Nonetheless, as genomics is evolving rapidly, it is constructive to survey now pertinent concepts and breakthroughs. Today, clinical multimodal electronic medical/health records (EMRs/EHRs) incorporating genomic information establish a continuously-learning, vast knowledge-network with seamless cycling between clinical application and research. It can inform insights into specific pathogenetic pathways, guide biomarker-assisted precise diagnoses and individualized treatments, and stratify prognoses. Complex CVDs blend multiple interacting genomic variants, epigenetics, and environmental risk-factors, engendering progressions of multifaceted disease-manifestations, including clinical symptoms and signs. There is no straight-line linkage between genetic cause(s) or causal gene-variant(s) and disease phenotype(s). Because of interactions involving modifier-gene influences, (micro)-environmental, and epigenetic effects, the same variant may actually produce dissimilar abnormalities in different individuals. Implementing genome-driven personalized cardiology in clinical practice reveals that the study of CVDs at the level of molecules and cells can yield crucial clinical benefits. Complementing evidence-based medicine guidelines from large ("one-size fits all") randomized controlled trials, genomics-based personalized or precision cardiology is a most-creditable paradigm: It provides customizable approaches to prevent, diagnose, and manage CVDs with treatments directly/precisely aimed at causal defects identified by high-throughput genomic technologies. They encompass stem cell and gene therapies exploiting CRISPR-Cas9-gene-editing, and metabolomic-pharmacogenomic therapeutic modalities, precisely fine-tuned for the individual patient. Following the Human Genome Project, many expected genomics technology to provide imminent solutions to intractable medical problems, including CVDs. This eagerness has reaped some disappointment that advances have not yet materialized to the degree anticipated. Undoubtedly, personalized genetic/genomics testing is an emergent technology that should not be applied without supplementary phenotypic/clinical information: Genotype≠Phenotype. However, forthcoming advances in genomics will naturally build on prior attainments and, combined with insights into relevant epigenetics and environmental factors, can plausibly eradicate intractable CVDs, improving human health and well-being.
Collapse
Affiliation(s)
- Ares Pasipoularides
- Consulting Professor of Surgery, Emeritus Faculty of Surgery and of Biomedical Engineering, Duke University School of Medicine and Graduate School, Durham, NC 27710, USA.
| |
Collapse
|
12
|
Ward NC, Pang J, Ryan JDM, Watts GF. Nutraceuticals in the management of patients with statin-associated muscle symptoms, with a note on real-world experience. Clin Cardiol 2018; 41:159-165. [PMID: 29363784 DOI: 10.1002/clc.22862] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
There is considerable evidence for the role of low-density lipoprotein cholesterol (LDL-C) in the development of atherosclerotic cardiovascular disease. Although statin therapy remains the most frequency prescribed medication to reduce LDL-C and lower risk of cardiovascular disease, a considerable number of patients develop muscle-related side affects. This review summarizes recent literature supporting the role of nutraceuticals as LDL-C-lowering therapy in statin-intolerant patients, with evidence from our own clinical practices.
Collapse
Affiliation(s)
- Natalie C Ward
- School of Medicine, Faculty of Health & Medical Sciences, University of Western Australia, Perth.,School of Biomedical Sciences and Curtin Health Innovation Research Institute, Perth, Australia
| | - Jing Pang
- School of Medicine, Faculty of Health & Medical Sciences, University of Western Australia, Perth.,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Jacqueline D M Ryan
- School of Medicine, Faculty of Health & Medical Sciences, University of Western Australia, Perth.,Perth Lipid Clinic, Primary Care, Perth, Australia
| | - Gerald F Watts
- School of Medicine, Faculty of Health & Medical Sciences, University of Western Australia, Perth.,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
13
|
Melendez QM, Wooten CJ, Lopez D. Atorvastatin and lovastatin, but not pravastatin, increased cellular complex formation between PCSK9 and the LDL receptor in human hepatocyte-like C3A cells. Biochem Biophys Res Commun 2017; 492:103-108. [PMID: 28802576 DOI: 10.1016/j.bbrc.2017.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
Statins are the first-line treatment for hypercholesterolemic patients. Herein, the effects of three statins on complex formation between proprotein convertase subtilisin-kexin 9 (PCSK9) and the low density lipoprotein receptor (LDLR), a critical step for the PCSK9-dependent degradation of LDLR in the lysosome, were examined. Human hepatocyte-like C3A cells grown in control (containing 10% fetal bovine serum) or MITO+ (supplemented with BD™ MITO + serum extender) medium were also treated with atorvastatin (Atorv), lovastatin (Lov), or pravastatin (Prav) for 24 h. RNA and protein expression studies and determinations of PCSK9/LDLR complex formation were performed. As expected, the statins increased the expression of PCSK9 and LDLR independently of the medium employed. Interestingly, Atov and Lov caused increases in PCSK9/LDLR complex formation, whereas Prav decreased complex formation when compared to cells treated without drugs. These results may explain why Prav works better for statin intolerant patients than other statins such as Atorv and Lov.
Collapse
Affiliation(s)
- Quantil M Melendez
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Catherine J Wooten
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Dayami Lopez
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, NC, 27707, USA.
| |
Collapse
|
14
|
Effectiveness of adherence to lipid lowering therapy on LDL-cholesterol in patients with very high cardiovascular risk: A real-world evidence study in primary care. Atherosclerosis 2017; 263:36-41. [DOI: 10.1016/j.atherosclerosis.2017.05.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/18/2017] [Accepted: 05/17/2017] [Indexed: 11/21/2022]
|
15
|
Boccara F, Dent R, Ruilope L, Valensi P. Practical Considerations for the Use of Subcutaneous Treatment in the Management of Dyslipidaemia. Adv Ther 2017; 34:1876-1896. [PMID: 28717862 PMCID: PMC5565663 DOI: 10.1007/s12325-017-0586-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Indexed: 02/06/2023]
Abstract
Suboptimal drug adherence represents a major challenge to effective primary and secondary prevention of cardiovascular disease. While adherence is influenced by multiple considerations, polypharmacy and dosing frequency appear to be rate-limiting factors in patient satisfaction and subsequent adherence. The cardiovascular and metabolic therapeutic areas have recently benefited from a number of advances in drug therapy, in particular protease proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and incretin-based therapies, respectively. These drugs are administered subcutaneously and offer efficacious treatment options with reduced dosing frequency. Whilst patients with diabetes and diabetologists are well initiated to injectable therapies, the cardiovascular therapeutic arena has traditionally been dominated by oral agents. It is therefore important to examine the practical aspects of treating patients with these new lipid-lowering agents, to ensure they are optimally deployed in everyday clinical practice.
Collapse
Affiliation(s)
- Franck Boccara
- Cardiology Unit, Hôpital Saint-Antoine, AP-HP, Hôpitaux de l'Est Parisien, Paris, France.
- INSERM, UMR_S 938, Faculty of Medicine, Sorbonne Universities, UPMC University Paris 06, Paris, France.
| | - Ricardo Dent
- Amgen (Europe) GmbH, Zug, Switzerland
- Esperion Therapeutics Inc, Ann Arbor, MI, USA
| | - Luis Ruilope
- Institute of Research, Hypertension Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Paul Valensi
- Department of Endocrinology-Diabetology-Nutrition, Jean Verdier Hospital, APHP, CRNH-IdF, CINFO, Paris Nord University, Bondy, France
| |
Collapse
|
16
|
Farnier M, Colhoun HM, Sasiela WJ, Edelberg JM, Asset G, Robinson JG. Long-term treatment adherence to the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab in 6 ODYSSEY Phase III clinical studies with treatment duration of 1 to 2 years. J Clin Lipidol 2017; 11:986-997. [DOI: 10.1016/j.jacl.2017.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022]
|
17
|
Catapano AL, Lee LV, Louie MJ, Thompson D, Bergeron J, Krempf M. Efficacy of alirocumab according to background statin type and dose: pooled analysis of 8 ODYSSEY Phase 3 clinical trials. Sci Rep 2017; 7:45788. [PMID: 28374849 PMCID: PMC5379546 DOI: 10.1038/srep45788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) reductions with the PCSK9 monoclonal antibody alirocumab may be affected by background statin dose due to increased PCSK9 levels with higher statin doses. Data from 8 Phase 3 trials conducted with background statin (n = 4629) were pooled by alirocumab dose (75 or 150 mg every 2 weeks) and control (placebo/ezetimibe), and analyzed by background statin type/dose. Overall, 58.4% received high-dose statins (atorvastatin 40-80 mg, rosuvastatin 20-40 mg, simvastatin 80 mg), 28.6% moderate-dose statins (atorvastatin 20-<40 mg, rosuvastatin 10-<20 mg, simvastatin 40-<80 mg), and 12.9% low-dose statins (atorvastatin <20 mg, rosuvastatin <10 mg, simvastatin <40 mg). Mean baseline PCSK9 levels were higher with high versus moderate and low statin doses (318.5 vs 280.6 ng/mL). Baseline LDL-C levels were similar across pools, regardless of statin intensity. No associations were observed between statin type/dose and LDL-C % change from baseline or % of patients achieving LDL-C goals at Week 24 for alirocumab versus control (interaction P-values non-significant). Incidence of adverse events was similar for alirocumab versus control, except for a higher rate of injection-site reactions with alirocumab. In summary, alirocumab provided consistent LDL-C reductions and was generally well tolerated independent of background statin type/dose.
Collapse
Affiliation(s)
- Alberico L. Catapano
- Department of Pharmacological and Biomolecular Sciences, Università di Milano and IRCCS Multimedica, Milan, Italy
| | | | | | | | - Jean Bergeron
- Clinique des Maladies Lipidiques, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Michel Krempf
- CHU de Nantes – Hôpital Nord Laennec, Saint-Herblain, France
| |
Collapse
|
18
|
Cangemi R, Romiti GF, Campolongo G, Ruscio E, Sciomer S, Gianfrilli D, Raparelli V. Gender related differences in treatment and response to statins in primary and secondary cardiovascular prevention: The never-ending debate. Pharmacol Res 2017; 117:148-155. [DOI: 10.1016/j.phrs.2016.12.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/21/2016] [Accepted: 12/20/2016] [Indexed: 02/08/2023]
|
19
|
Reply to: “Statins probably do not cause cataracts”. Atherosclerosis 2016; 254:311-312. [DOI: 10.1016/j.atherosclerosis.2016.09.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/15/2016] [Indexed: 11/18/2022]
|
20
|
Mikashinovich ZI, Belousova ES. Biochemical Changes in Erythrocytes as a Molecular Marker of Cell Damage during Long-Term Simvastatin Treatment. Bull Exp Biol Med 2016; 161:600-3. [PMID: 27591878 DOI: 10.1007/s10517-016-3467-2] [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: 10/07/2015] [Indexed: 11/29/2022]
Abstract
Long-term administration of simvastatin to rats, irrespective of the baseline cholesterol levels, induced biochemical changes in erythrocytes attesting to hypoxic damage (accumulation of lactate and 2,3-diphosphoglycerate), disturbances in ATP-dependent mechanisms of ion homeostasis regulation (decrease in total ATPase and Ca(2+)-ATPase activities), and antioxidant enzymes system imbalance. These changes can be considered as a sensitive indicator and molecular basis of cell damage during long-term administration of statins.
Collapse
Affiliation(s)
- Z I Mikashinovich
- Department of General and Clinical Biochemistry No. 1, Rostov State Medical University, Ministry of Health of the Russian Federation, Rostov-on-Don, Russia
| | - E S Belousova
- Department of Pharmaceutical Chemistry and Pharmacology, Rostov State Medical University, Ministry of Health of the Russian Federation, Rostov-on-Don, Russia.
| |
Collapse
|
21
|
Mancini GJ, Baker S, Bergeron J, Fitchett D, Frohlich J, Genest J, Gupta M, Hegele RA, Ng D, Pearson GJ, Pope J, Tashakkor AY. Diagnosis, Prevention, and Management of Statin Adverse Effects and Intolerance: Canadian Consensus Working Group Update (2016). Can J Cardiol 2016; 32:S35-65. [DOI: 10.1016/j.cjca.2016.01.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 12/24/2022] Open
|
22
|
Ren H, Lv H. Neuromuscular Effects of Rocuronium Bromide in Patients in Statin Therapy for at least Three Months. Basic Clin Pharmacol Toxicol 2016; 119:582-587. [PMID: 27234897 DOI: 10.1111/bcpt.12628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/23/2016] [Indexed: 11/28/2022]
Abstract
Statins cause skeletal muscle myopathy. However, the neuromuscular effects of non-depolarizing neuromuscular-blocking agent in patients in long-term statin therapy remain unclear. Hence, we investigated the neuromuscular effects of rocuronium and muscle injury in patients in long-term statin therapy. Eighteen statin users using statins for at least 3 months were included in the statin group and 18 non-statin users were included in the non-statin group. General anaesthesia was induced with intravenous midazolam, etomidate, sufentanil and rocuronium 0.9 mg/kg (3ED95 ) for intubation. Anaesthesia was maintained with 1% propofol and remifentanil. The onset time and duration 10% T1 and 25% T1 of rocuronium were recorded. Blood samples were obtained before induction and 5 min., 1 hr, 2 hr, 4 hr, 12 hr and 24 hr after rocuronium administration to measure creatine kinase (CK), myoglobin and potassium. Myalgia was determined at 2 and 24 hr after surgery. There were no significant differences in the basic clinical characteristics between the two groups. The onset time of the statin group was significantly shorter than that of the non-statin group (p = 0.02), while the duration 10% T1 and duration 25% T1 of the statin group were significantly longer than those of the non-statin group (p = 0.006; p = 0.045). The myoglobin and CK concentrations increased after rocuronium administration as compared to baseline in both groups. CK concentration in the statin group was significantly higher than in the non-statin group just at 24 hr (p = 0.000003). However, myoglobin showed no significant difference between the two groups. The onset time of rocuronium decreases and its duration time increases in patients in long-term statin therapy.
Collapse
Affiliation(s)
- Hongwei Ren
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Huangwei Lv
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
[PCSK9 - "missing link" in familial hypercholesterolemia : New therapeutic options in hypercholesterolemia and coronary artery disease]. Herz 2016; 41:281-9. [PMID: 27215417 DOI: 10.1007/s00059-016-4435-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Lowering plasma low-density lipoprotein cholesterol (LDL-C) levels to individual therapeutic goals is one of the most effective measures for the prevention of cardiovascular disease. Besides dietary measures, this can be achieved pharmaceutically by inhibition of hepatic cholesterol synthesis with statins or inhibition of intestinal cholesterol absorption (e.g., ezetimibe and bile acid sequestrants). Decisive for lowering LDL is an increased hepatic uptake of circulating LDL via an increase in LDL receptors (LDLR) in hepatic cell membranes. The formation of new LDLR and recirculation of existing LDLR play a decisive role in this process. An important modulator of LDLR is proprotein convertase subtilisin/kexin type 9 (PCSK9). In the last years genetic studies have identified several mutations in the PCSK9 gene leading to a gain of function and carriers of these mutations suffer from autosomal dominant hypercholesterolemia. In contrast, carriers of PCSK9 loss of function mutations show very low plasma LDL-C concentrations and a markedly reduced risk for coronary artery disease. These fundamental discoveries have sparked the development of a completely novel therapeutic approach to treating hypercholesterolemia. At present, inhibition of PCSK9 by monoclonal antibodies presents the most promising therapeutic approach. First human antibodies were recently approved as the first immunotherapeutic agents for the treatment of severe hypercholesterolemia and in patients with statin intolerance. An additional PCSK9 antibody is presently being studied in phase III clinical trials.
Collapse
|
24
|
Abstract
Muscle problems and other adverse symptoms associated with statin use are frequent reasons for non-adherence and discontinuation of statin therapy, which results in inadequate control of hyperlipidemia and increased cardiovascular risk. However, most patients who experience adverse symptoms during statin use are able to tolerate at least some degree of statin therapy. Given the profound cardiovascular benefits derived from statins, an adequate practical approach to statin intolerance is, therefore, of great clinical importance. Statin intolerance can be defined as the occurrence of myalgia or other adverse symptoms that are attributed to statin therapy and that lead to its discontinuation. In reality, these symptoms are actually unrelated to statin use in many patients, especially in those with atypical presentations following long periods of treatment. Thus, the first step in approaching patients with adverse symptoms during the course of statin therapy is identification of those patients for whom true statin intolerance is unlikely, since most of these patients would probably be capable of tolerating adequate statin therapy. In patients with statin intolerance, an altered dosing regimen of very low doses of statins should be attempted and, if tolerated, should gradually be increased to achieve the highest tolerable doses. In addition, other lipid-lowering drugs may be needed, either in combination with statins, or alone, if statins are not tolerated at all. Stringent control of other risk factors can aid in reducing cardiovascular risk if attaining lipid treatment goals proves difficult.
Collapse
Affiliation(s)
- Tomáš Stulc
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, U Nemocnice 1, CZ 128 21, Praha 2 / Prague, Czech Republic.
| | - Richard Ceška
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, U Nemocnice 1, CZ 128 21, Praha 2 / Prague, Czech Republic
| | - Antonio M Gotto
- Weill Cornell Medical College, New York, 1305 York Avenue, Y-807, New York, NY, 10021, USA
| |
Collapse
|
25
|
Lupin protein exerts cholesterol-lowering effects targeting PCSK9: From clinical evidences to elucidation of the in vitro molecular mechanism using HepG2 cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.02.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
26
|
Patel S. Functional food red yeast rice (RYR) for metabolic syndrome amelioration: a review on pros and cons. World J Microbiol Biotechnol 2016; 32:87. [PMID: 27038957 DOI: 10.1007/s11274-016-2035-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/19/2016] [Indexed: 12/14/2022]
Abstract
Red yeast rice (RYR), the fermentation product of mold Monascus purpureus has been an integral part of Oriental food and traditional Chinese medicine, long before the discovery of their medicinal roles. With the identification of bioactive components as polyketide pigments (statins), and unsaturated fatty acids, RYR has gained a nutraceutical status. Hypercholesterolemic effect of this fermented compound has been validated and monacolin K has been recognized as the pivotal component in cholesterol alleviation. Functional similarity with commercial drug lovastatin sans the side effects has catapulted its popularity in other parts of the world as well. Apart from the hypotensive role, ameliorative benefits of RYR as anti-inflammatory, antidiabetic, anticancer and osteogenic agent have emerged, fueling intense research on it. Mechanistic studies have revealed their interaction with functional agents like coenzyme Q10, astaxanthin, vitamin D, folic acid, policosanol, and berberine. On the other hand, concurrence of mycotoxin citrinin and variable content of statin has marred its integration in mainstream medication. In this disputable scenario, evaluation of the scopes and lacunae to overcome seems to contribute to an eminent area of healthcare. Red yeast rice (RYR), the rice-based fermentation product of mold Monascus purpureus is a functional food. Its bioactive component monacolin K acts like synthetic drug lovastatin, without the severe side effects of the latter. RYR has been validated to lower cholesterol, control high blood pressure; confer anti-flammation, hypoglycaemic, anticancer and osteogenic properties. However, dose inconsistency and co-occurrence of toxin citrinin hampers its dietary supplementation prospect. Further research might facilitate development of RYR as a nutraceutical.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, 5500 Campanile Dr, San Diego, CA, 92182, USA.
| |
Collapse
|
27
|
Trentman TL, Avey SG, Ramakrishna H. Current and emerging treatments for hypercholesterolemia: A focus on statins and proprotein convertase subtilisin/kexin Type 9 inhibitors for perioperative clinicians. J Anaesthesiol Clin Pharmacol 2016; 32:440-445. [PMID: 28096572 PMCID: PMC5187606 DOI: 10.4103/0970-9185.194773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Statins are a mainstay of hyperlipidemia treatment. These drugs inhibit the enzyme 3-hydroxy-3-methylglutaryl coenzyme A reductase and have beneficial effects on atherosclerosis including plaque stabilization, reduction of platelet activation, and reduction of plaque proliferation and inflammation. Statins also have a benefit beyond atherosclerotic plaque, including anticoagulation, vasodilatation, antioxidant effects, and reduction of mediators of inflammation. In the perioperative period, statins appear to contribute to improved outcomes via these mechanisms. Both vascular and nonvascular surgery patients have been shown in prospective studies to have lower risk of adverse cardiac outcomes when initiated on statins preoperatively. However, not all patients can tolerate statins; the search for novel lipid-lowering therapies led to the discovery of the proprotein convertase subtilisin/kexin Type 9 (PCSK9) inhibitors. These drugs are fully-humanized, injectable monoclonal antibodies. With lower PCSK9 activity, low-density lipoprotein cholesterol (LDL-C) receptors are more likely to be recycled to the hepatocyte surface, where they serve to clear plasma LDL-C. Evidence from several prospective studies shows that these new agents can significantly lower LDL-C levels. While PCSK9 inhibitors offer hope of effective therapy for patients with familial hyperlipidemia or intolerance of statins, several important questions remain, including the results of long term cardiovascular outcome studies. The perioperative effects of new LDL-C-lowering drugs are unknown at present but are likely to be similar to the older agents.
Collapse
Affiliation(s)
| | - Steven G Avey
- MedImpact Healthcare Systems, Inc, San Diego CA, USA
| | | |
Collapse
|
28
|
Marcuzzi A, Piscianz E, Loganes C, Vecchi Brumatti L, Knowles A, Bilel S, Tommasini A, Bortul R, Zweyer M. Innovative Target Therapies Are Able to Block the Inflammation Associated with Dysfunction of the Cholesterol Biosynthesis Pathway. Int J Mol Sci 2015; 17:47. [PMID: 26729102 PMCID: PMC4730292 DOI: 10.3390/ijms17010047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 01/24/2023] Open
Abstract
The cholesterol pathway is an essential biochemical process aimed at the synthesis of bioactive molecules involved in multiple crucial cellular functions. The end products of this pathway are sterols, such as cholesterol, which are essential components of cell membranes, precursors of steroid hormones, bile acids and other molecules such as ubiquinone. Several diseases are caused by defects in this metabolic pathway: the most severe forms of which cause neurological involvement (psychomotor retardation and cerebellar ataxia) as a result of a variety of cellular impairments, including mitochondrial dysfunction. These pathologies are induced by convergent mechanisms in which the mitochondrial unit plays a pivotal role contributing to defective apoptosis, autophagy and mitophagy processes. Unraveling these mechanisms would contribute to the development of effective drug treatments for these disorders. In addition, the development of biochemical models could have a substantial impact on the understanding of the mechanism of action of drugs that act on this pathway in multifactor disorders. In this review we will focus in particular on inhibitors of cholesterol synthesis, mitochondria-targeted drugs and inhibitors of the inflammasome.
Collapse
Affiliation(s)
- Annalisa Marcuzzi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Elisa Piscianz
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Claudia Loganes
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Liza Vecchi Brumatti
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Alessandra Knowles
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Sabrine Bilel
- Cluster in Biomedicine (CBM scrl), Trieste 34128, Italy.
| | - Alberto Tommasini
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Roberta Bortul
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Marina Zweyer
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| |
Collapse
|