201
|
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]
|
202
|
Toward an international consensus—Integrating lipoprotein apheresis and new lipid-lowering drugs. J Clin Lipidol 2017; 11:858-871.e3. [DOI: 10.1016/j.jacl.2017.04.114] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/30/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
|
203
|
Leiter LA, Zamorano JL, Bujas‐Bobanovic M, Louie MJ, Lecorps G, Cannon CP, Handelsman Y. Lipid-lowering efficacy and safety of alirocumab in patients with or without diabetes: A sub-analysis of ODYSSEY COMBO II. Diabetes Obes Metab 2017; 19:989-996. [PMID: 28206704 PMCID: PMC5485164 DOI: 10.1111/dom.12909] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 12/15/2022]
Abstract
AIM This sub-analysis of the ODYSSEY COMBO II study compared the effects of alirocumab, a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor, in high cardiovascular risk patients with or without diabetes mellitus (DM) receiving maximally tolerated statin therapy. METHODS COMBO II was a 104-week, double-blind study (n = 720) enrolling patients with documented atherosclerotic cardiovascular disease (ASCVD) and baseline LDL-C ≥70 mg/dL (1.8 mmol/L), and patients without documented ASCVD at high cardiovascular risk with LDL-C ≥100 mg/dL (2.6 mmol/L). Patients receiving maximally tolerated statin therapy were randomized (2:1) to alirocumab 75 mg every 2 weeks (Q2W; 1 mL subcutaneous injection) or oral ezetimibe 10 mg daily. Alirocumab dose was increased to 150 mg Q2W (also 1 mL) at Week 12 if Week 8 LDL-C was ≥70 mg/dL. RESULTS History of DM was reported in 31% (n = 148) of patients on alirocumab and 32% (n = 77) of patients on ezetimibe. At Week 24, alirocumab consistently reduced LDL-C from baseline in patients with (-49.1%) or without DM (-51.2%) to a significantly greater extent than ezetimibe (-18.4% and -21.8%, respectively). Occurrence of treatment-emergent adverse events was similar between groups. Efficacy results at 104 weeks were similar to those at 24 weeks. CONCLUSIONS Over a 104-week double-blind study period, alirocumab provided consistently greater LDL-C reductions than ezetimibe, with similar LDL-C results in patients with or without DM. Safety of alirocumab was similar regardless of baseline DM status.
Collapse
Affiliation(s)
- Lawrence A. Leiter
- Li Ka Shing Knowledge Institute and Keenan Research Center for Biomedical ScienceSt. Michael's Hospital, University of TorontoTorontoCanada
| | - José Luis Zamorano
- Hospital Universitario Ramon y Cajal, University Alcala de HenaresMadridSpain
| | | | | | | | | | | |
Collapse
|
204
|
Lipid Metabolism and Emerging Targets for Lipid-Lowering Therapy. Can J Cardiol 2017; 33:872-882. [DOI: 10.1016/j.cjca.2016.12.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/12/2016] [Accepted: 12/26/2016] [Indexed: 12/25/2022] Open
|
205
|
Wills-Sanín B, Rincón A, Montenegro AC, Buitrago-Sandoval AF. Inhibidores de la paraproteína convertasa subtilisina/kexina tipo 9: una alternativa de manejo para las dislipidemias. REVISTA COLOMBIANA DE CARDIOLOGÍA 2017. [DOI: 10.1016/j.rccar.2016.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
206
|
Abstract
PURPOSE OF REVIEW Monoclonal antibodies that inhibit proprotein convertase subtilisin/kexin type 9 (PCSK9) have emerged as a novel approach to low-density lipoprotein cholesterol (LDL-C) lowering. The potential role of PCSK9 inhibitors in clinical practice will be reviewed. RECENT FINDINGS Clinical trials have demonstrated that PCSK9 inhibitors produce robust LDL-C lowering when administered either as monotherapy or in combination with statins. This provides the opportunity to achieve effective lipid lowering in familial hypercholesterolemia, patients with either established atherosclerotic cardiovascular disease or high risk primary prevention and an important opportunity to treat patients with statin intolerance. The findings from plaque imaging and patients with established atherosclerotic cardiovascular disease suggest that PCSK9 inhibition has favorable outcomes beyond improving lipid profiles, which has the opportunity to expand their use. PCSK9 inhibitors represent a new approach to achieving effective cardiovascular risk reduction in a broader number of patients. How these agents will be taken up in clinical practice remains to be determined.
Collapse
Affiliation(s)
- Peta King
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA, 5001, Australia
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA, 5001, Australia.
| |
Collapse
|
207
|
Ming JE, Abrams RE, Bartlett DW, Tao M, Nguyen T, Surks H, Kudrycki K, Kadambi A, Friedrich CM, Djebli N, Goebel B, Koszycki A, Varshnaya M, Elassal J, Banerjee P, Sasiela WJ, Reed MJ, Barrett JS, Azer K. A Quantitative Systems Pharmacology Platform to Investigate the Impact of Alirocumab and Cholesterol-Lowering Therapies on Lipid Profiles and Plaque Characteristics. GENE REGULATION AND SYSTEMS BIOLOGY 2017; 11:1177625017710941. [PMID: 28804243 PMCID: PMC5484552 DOI: 10.1177/1177625017710941] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 04/17/2017] [Indexed: 12/20/2022]
Abstract
Reduction in low-density lipoprotein cholesterol (LDL-C) is associated with decreased risk for cardiovascular disease. Alirocumab, an antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), significantly reduces LDL-C. Here, we report development of a quantitative systems pharmacology (QSP) model integrating peripheral and liver cholesterol metabolism, as well as PCSK9 function, to examine the mechanisms of action of alirocumab and other lipid-lowering therapies, including statins. The model predicts changes in LDL-C and other lipids that are consistent with effects observed in clinical trials of single or combined treatments of alirocumab and other treatments. An exploratory model to examine the effects of lipid levels on plaque dynamics was also developed. The QSP platform, on further development and qualification, may support dose optimization and clinical trial design for PCSK9 inhibitors and lipid-modulating drugs. It may also improve our understanding of factors affecting therapeutic responses in different phenotypes of dyslipidemia and cardiovascular disease.
Collapse
Affiliation(s)
- Jeffrey E Ming
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Ruth E Abrams
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | | | - Mengdi Tao
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Tu Nguyen
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Howard Surks
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | | | | | | | - Nassim Djebli
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Britta Goebel
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Alex Koszycki
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Meera Varshnaya
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | | | | | | | | | - Jeffrey S Barrett
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| | - Karim Azer
- Sanofi, Bridgewater, NJ, USA; Frankfurt Am Main, Germany, and Montpellier, France
| |
Collapse
|
208
|
Teramoto T, Kondo A, Kiyosue A, Harada-Shiba M, Ishigaki Y, Tobita K, Kawabata Y, Ozaki A, Baccara-Dinet MT, Sata M. Efficacy and safety of alirocumab in patients with hypercholesterolemia not adequately controlled with non-statin lipid-lowering therapy or the lowest strength of statin: ODYSSEY NIPPON study design and rationale. Lipids Health Dis 2017. [PMID: 28623954 PMCID: PMC5474052 DOI: 10.1186/s12944-017-0513-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Statins are generally well-tolerated and serious side effects are infrequent, but some patients experience adverse events and reduce their statin dose or discontinue treatment altogether. Alirocumab is a highly specific, fully human monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), which can produce substantial and sustained reductions of low-density lipoprotein cholesterol (LDL-C). Methods The randomized, double-blind, placebo-controlled, parallel-group, phase 3 ODYSSEY NIPPON study will explore alirocumab 150 mg every 4 weeks (Q4W) in 163 Japanese patients with hypercholesterolemia who are on the lowest-strength dose of atorvastatin (5 mg/day) or are receiving a non-statin lipid-lowering therapy (LLT) (fenofibrate, bezafibrate, ezetimibe, or diet therapy alone). Hypercholesterolemia is defined as LDL-C ≥ 100 mg/dL (2.6 mmol/L) in patients with heterozygous familial hypercholesterolemia or non-familial hypercholesterolemia with a history of documented coronary heart disease, or ≥120 mg/dL (3.1 mmol/L) in patients with non-familial hypercholesterolemia classified as primary prevention category III (i.e. high-risk patients). During the 12-week double-blind treatment period, patients will be randomized (1:1:1) to receive alirocumab subcutaneously (SC) 150 mg Q4W alternating with placebo for alirocumab Q4W, or alirocumab 150 mg SC every 2 weeks (Q2W), or SC placebo Q2W. The primary efficacy endpoint is the percentage change in calculated LDL-C from baseline to week 12. The long-term safety and tolerability of alirocumab will also be investigated. Discussion The ODYSSEY NIPPON study will provide insights into the efficacy and safety of alirocumab 150 mg Q4W or 150 mg Q2W among Japanese patients with hypercholesterolemia who are on the lowest-strength dose of atorvastatin, or are receiving a non-statin LLT (including diet therapy alone). Trial registration ClinicalTrials.gov number: NCT02584504
Collapse
Affiliation(s)
- Tamio Teramoto
- Teikyo Academic Research Center, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Akira Kondo
- Asia Pacific Development, R&D, Sanofi, Tokyo, Japan
| | - Arihiro Kiyosue
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Yasushi Ishigaki
- Division of Diabetes and Metabolism, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | | | | | | | | | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
209
|
Schiele F, Ecarnot F, Chopard R. Coronary artery disease: Risk stratification and patient selection for more aggressive secondary prevention. Eur J Prev Cardiol 2017; 24:88-100. [DOI: 10.1177/2047487317706586] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- François Schiele
- Department of Cardiology, University Hospital Besançon and EA3920, University of Burgundy Franche-Comté, Besançon, France
| | - Fiona Ecarnot
- Department of Cardiology, University Hospital Besançon and EA3920, University of Burgundy Franche-Comté, Besançon, France
| | - Romain Chopard
- Department of Cardiology, University Hospital Besançon and EA3920, University of Burgundy Franche-Comté, Besançon, France
| |
Collapse
|
210
|
Pucci G, Cicero AF, Borghi C, Schillaci G. Emerging biologic therapies for hypercholesterolaemia. Expert Opin Biol Ther 2017; 17:1077-1087. [DOI: 10.1080/14712598.2017.1341485] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Giacomo Pucci
- Dipartimento di Medicina, Università di Perugia, Perugia, Italy
- Struttura Complessa di Medicina Interna, Azienda Ospedaliera “S. Maria” di Terni, Terni, Italy
| | - Arrigo F Cicero
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Claudio Borghi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Giuseppe Schillaci
- Dipartimento di Medicina, Università di Perugia, Perugia, Italy
- Struttura Complessa di Medicina Interna, Azienda Ospedaliera “S. Maria” di Terni, Terni, Italy
| |
Collapse
|
211
|
Welsh P, Grassia G, Botha S, Sattar N, Maffia P. Targeting inflammation to reduce cardiovascular disease risk: a realistic clinical prospect? Br J Pharmacol 2017; 174:3898-3913. [PMID: 28409825 PMCID: PMC5660005 DOI: 10.1111/bph.13818] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 12/16/2022] Open
Abstract
Data from basic science experiments is overwhelmingly supportive of the causal role of immune-inflammatory response(s) at the core of atherosclerosis, and therefore, the theoretical potential to manipulate the inflammatory response to prevent cardiovascular events. However, extrapolation to humans requires care and we still lack definitive evidence to show that interfering in immune-inflammatory processes may safely lessen clinical atherosclerosis. In this review, we discuss key therapeutic targets in the treatment of vascular inflammation, placing basic research in a wider clinical perspective, as well as identifying outstanding questions. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Paul Welsh
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gianluca Grassia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Shani Botha
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom campus, South Africa
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
212
|
Abstract
In recent years, biochemical and genetic studies have identified proprotein convertase subtilisin/kexin type 9 (PCSK9) as a major mediator of low-density lipoprotein cholesterol (LDL-c) levels and thereby a potential novel target for reducing risk of coronary heart disease (CHD). These observations led to the development of PCSK9 inhibitors, which lower LDL-c levels more than any other non-invasive lipid-lowering therapy presently available. The PCSK9 inhibitors furthest along in clinical trials are subcutaneously injected monoclonal antibodies. These PCSK9 inhibitors have demonstrated LDL-c-lowering efficacy with acceptable safety in phase III clinical trials and may offer a useful therapy in addition to maximally tolerated HMG-CoA reductase inhibitors (statins) in certain patient groups. Longer-term data are required to ensure sustained efficacy and safety of this new class of medications. This review provides an overview of the biology, genetics, development, and clinical trials of monoclonal antibodies designed to inhibit PCSK9.
Collapse
|
213
|
AlHajri L, AlHadhrami A, AlMheiri S, AlMutawa Y, AlHashimi Z. The efficacy of evolocumab in the management of hyperlipidemia: a systematic review. Ther Adv Cardiovasc Dis 2017; 11:155-169. [PMID: 28488460 DOI: 10.1177/1753944717698925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Hyperlipidemia or dyslipidemia has been a concern for a long time, with various guidelines emphasizing the importance of managing the lipid profile to prevent cardiac incidences. Although statins have been found to be highly effective, resistance and intolerability to side effects will continue to be a stumbling block for certain patients. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors tackle lipid profile via a novel mechanism and therefore provide an additional effective option for managing lipid profile. The overarching aim of this systematic review was to evaluate the efficacy of evolocumab among various populations with hypercholesterolemia. METHODS A comprehensive search was conducted in ProQuest Health & Medical Complete, Google Scholar, ScienceDirect, and PubMed to identify potential records; then titles, abstracts, and full texts were screened using the inclusion criteria to filter out irrelevant studies. Data extraction and quality assessment were undertaken using standardized tools and the results were narratively synthesized and presented in tables. RESULTS Eight studies were included in this systematic review after screening 1191 records. All studies demonstrated a statistically significant reduction in low-density lipoprotein cholesterol (LDL-C) values in the groups that received evolocumab compared with the comparator groups ( p < 0.05). The decline in LDL-C levels from baseline in the majority of studies ranged from 40% to 80%, whether used alone or in combination with other agents. Also, high-density lipoprotein cholesterol, lipoprotein (a) and apolipoprotein B were improved with the use of evolocumab. CONCLUSIONS This study helped to collate evidence from studies that tested the effectiveness of evolocumab in the management of hyperlipidemia. Evolocumab seems to be highly effective in reducing LDL-C and other lipid parameters. Hence, it provides an excellent alternative for patients with refractory disease or patients who develop intolerable side effects, therefore helping to overcome the stumbling block to achieving optimal lipid management.
Collapse
Affiliation(s)
- Lamia AlHajri
- Department of Health Sciences, Higher Colleges of Technology, PO Box 16062, Dubai, UAE Department of Health Research, Lancaster University, UK
| | - Asma AlHadhrami
- Department of Health Sciences, Higher Colleges of Technology, Dubai, UAE
| | - Shama AlMheiri
- Department of Health Sciences, Higher Colleges of Technology, Dubai, UAE
| | - Yalwah AlMutawa
- Department of Health Sciences, Higher Colleges of Technology, Dubai, UAE
| | - Zainab AlHashimi
- Department of Health Sciences, Higher Colleges of Technology, Dubai, UAE
| |
Collapse
|
214
|
Effect of alirocumab dose increase on LDL-lowering and lipid goal attainment. Coron Artery Dis 2017; 28:186-187. [DOI: 10.1097/mca.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
215
|
Schmidt AF, Pearce LS, Wilkins JT, Overington JP, Hingorani AD, Casas JP. PCSK9 monoclonal antibodies for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2017; 4:CD011748. [PMID: 28453187 PMCID: PMC6478267 DOI: 10.1002/14651858.cd011748.pub2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Despite the availability of effective drug therapies that reduce low-density lipoprotein (LDL)-cholesterol (LDL-C), cardiovascular disease (CVD) remains an important cause of mortality and morbidity. Therefore, additional LDL-C reduction may be warranted, especially for patients who are unresponsive to, or unable to take, existing LDL-C-reducing therapies. By inhibiting the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme, monoclonal antibodies (PCSK9 inhibitors) may further reduce LDL-C, potentially reducing CVD risk as well. OBJECTIVES Primary To quantify short-term (24 weeks), medium-term (one year), and long-term (five years) effects of PCSK9 inhibitors on lipid parameters and on the incidence of CVD. Secondary To quantify the safety of PCSK9 inhibitors, with specific focus on the incidence of type 2 diabetes, cognitive function, and cancer. Additionally, to determine if specific patient subgroups were more or less likely to benefit from the use of PCSK9 inhibitors. SEARCH METHODS We identified studies by systematically searching the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and Web of Science. We also searched Clinicaltrials.gov and the International Clinical Trials Registry Platform and screened the reference lists of included studies. We identified the studies included in this review through electronic literature searches conducted up to May 2016, and added three large trials published in March 2017. SELECTION CRITERIA All parallel-group and factorial randomised controlled trials (RCTs) with a follow-up time of at least 24 weeks were eligible. DATA COLLECTION AND ANALYSIS Two review authors independently reviewed and extracted data. When data were available, we calculated pooled effect estimates. MAIN RESULTS We included 20 studies with data on 67,237 participants (median age 61 years; range 52 to 64 years). Twelve trials randomised participants to alirocumab, three trials to bococizumab, one to RG7652, and four to evolocumab. Owing to the small number of trials using agents other than alirocumab, we did not differentiate between types of PCSK9 inhibitors used. We compared PCSK9 inhibitors with placebo (thirteen RCTs), ezetimibe (two RCTs) or ezetimibe and statins (five RCTs).Compared with placebo, PCSK9 inhibitors decreased LDL-C by 53.86% (95% confidence interval (CI) 58.64 to 49.08; eight studies; 4782 participants; GRADE: moderate) at 24 weeks; compared with ezetimibe, PCSK9 inhibitors decreased LDL-C by 30.20% (95% CI 34.18 to 26.23; two studies; 823 participants; GRADE: moderate), and compared with ezetimibe and statins, PCSK9 inhibitors decreased LDL-C by 39.20% (95% CI 56.15 to 22.26; five studies; 5376 participants; GRADE: moderate).Compared with placebo, PCSK9 inhibitors decreased the risk of CVD events, with a risk difference (RD) of 0.91% (odds ratio (OR) of 0.86, 95% CI 0.80 to 0.92; eight studies; 59,294 participants; GRADE: moderate). Compared with ezetimibe and statins, PCSK9 inhibitors appeared to have a stronger protective effect on CVD risk, although with considerable uncertainty (RD 1.06%, OR 0.45, 95% CI 0.27 to 0.75; three studies; 4770 participants; GRADE: very low). No data were available for the ezetimibe only comparison. Compared with placebo, PCSK9 probably had little or no effect on mortality (RD 0.03%, OR 1.02, 95% CI 0.91 to 1.14; 12 studies; 60,684 participants; GRADE: moderate). Compared with placebo, PCSK9 inhibitors increased the risk of any adverse events (RD 1.54%, OR 1.08, 95% CI 1.04 to 1.12; 13 studies; 54,204 participants; GRADE: low). Similar effects were observed for the comparison of ezetimibe and statins: RD 3.70%, OR 1.18, 95% CI 1.05 to 1.34; four studies; 5376 participants; GRADE: low. Clinical event data were unavailable for the ezetimibe only comparison. AUTHORS' CONCLUSIONS Over short-term to medium-term follow-up, PCSK9 inhibitors reduced LDL-C. Studies with medium-term follow-up time (longest median follow-up recorded was 26 months) reported that PCSK9 inhibitors (compared with placebo) decreased CVD risk but may have increased the risk of any adverse events (driven by SPIRE-1 and -2 trials). Available evidence suggests that PCSK9 inhibitor use probably leads to little or no difference in mortality. Evidence on relative efficacy and safety when PCSK9 inhibitors were compared with active treatments was of low to very low quality (GRADE); follow-up times were short and events were few. Large trials with longer follow-up are needed to evaluate PCSK9 inhibitors versus active treatments as well as placebo. Owing to the predominant inclusion of high-risk patients in these studies, applicability of results to primary prevention is limited. Finally, estimated risk differences indicate that PCSK9 inhibitors only modestly change absolute risks (often to less than 1%).
Collapse
Affiliation(s)
- Amand F Schmidt
- University College LondonInstitute of Cardiovascular Science222 Euston Road, Room 206LondonUKNW1 2DA
| | - Lucy S Pearce
- London School of Hygiene & Tropical MedicineDepartment of Non‐communicable Disease EpidemiologyKeppel StreetLondonUKWC1 E7HT
| | - John T Wilkins
- Northwestern University Feinberg School of MedicineThe Department of Medicine (Cardiology) and the Department of Preventive MedicineSuite 1400 680 N. Lakeshore DriveChicagoUSA60611
| | | | - Aroon D Hingorani
- University College LondonInstitute of Cardiovascular Science222 Euston Road, Room 206LondonUKNW1 2DA
| | - Juan P Casas
- University College LondonFarr Institute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| |
Collapse
|
216
|
Della Pepa G, Bozzetto L, Annuzzi G, Rivellese AA. Alirocumab for the treatment of hypercholesterolaemia. Expert Rev Clin Pharmacol 2017; 10:571-582. [PMID: 28395555 DOI: 10.1080/17512433.2017.1318063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Prescription of statins for low-density lipoprotein cholesterol (LDL-C) reduction is the standard of care in primary and secondary prevention of cardiovascular disease; nevertheless, a large number of patients treated with statins are unable to reach the recommended LDL-C targets. Therefore, there is need for safe and effective novel therapies for the pharmacological management of hypercholesterolaemia, in addition or as alternative to lipid-lowering therapies (LLT) currently in use. Areas covered: In 2015, the Food and Drug Administration and the European Medicines Agency approved alirocumab (Praluent®; Sanofi), a fully human monoclonal antibody against proprotein convertase subtilisin/kexin type 9 (PCSK9), for the treatment of hypercholesterolaemic patients unable to meet LDL-C targets, as an adjunct to diet in addition/alternative to LLT. The authors review the pharmacological features, clinical efficacy, and safety of alirocumab in lowering LDL-C, and discuss its therapeutic perspectives based on the most recent clinical trials. Expert commentary: Alirocumab causes a marked reduction in LDL-C, presents good safety and tolerability, and represents a promising approach for LDL-C lowering, particularly in patients with intolerance to statin or elevated LDL-C despite maximal statin therapy; nevertheless, further long-term data on safety and efficacy are necessary, such as data on the improvement of cardiovascular outcomes.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- a Department of Clinical Medicine and Surgery , Federico II University , Naples , Italy
| | - Lutgarda Bozzetto
- a Department of Clinical Medicine and Surgery , Federico II University , Naples , Italy
| | - Giovanni Annuzzi
- a Department of Clinical Medicine and Surgery , Federico II University , Naples , Italy
| | | |
Collapse
|
217
|
Abstract
The use of low-density lipoprotein cholesterol (LDL-C)-lowering medications has led to a significant reduction of cardiovascular risk in both primary and secondary prevention. Statin therapy, one of the cornerstones for the prevention and treatment of cardiovascular disease (CVD), has been demonstrated to be effective in lowering LDL-C levels and in reducing the risk for CVD and is generally well-tolerated. However, compliance with statins remains suboptimal. One of the main reasons is limitations by adverse events, notably myopathies, which can lead to non-compliance with the prescribed statin regimen. Reducing the burden of elevated LDL-C levels is critical in patients with CVD as well as in patients with very high baseline levels of LDL-C (e.g. patients with familial hypercholesterolaemia), as statin therapy is insufficient for optimally reducing LDL-C below target values. In this review, we discuss alternative treatment options after maximally tolerated doses of statin therapy, including ezetimibe, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, and cholesteryl ester transfer protein (CETP) inhibitors. Difficult-to-treat patients may benefit from combination therapy with ezetimibe or a PCSK9 inhibitor (evolocumab or alirocumab, which are now available). Updates of treatment guidelines are needed to guide the management of patients who will best benefit from these new treatments.
Collapse
Affiliation(s)
- Stephan Krähenbühl
- Division of Clinical Pharmacology and Toxicology, University Hospital, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | | | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zürich, Zurich, Switzerland
- Zürich Center for Integrative Human Physiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
218
|
Efficacy and safety of alirocumab in insulin-treated patients with type 1 or type 2 diabetes and high cardiovascular risk: Rationale and design of the ODYSSEY DM-INSULIN trial. DIABETES & METABOLISM 2017; 43:453-459. [PMID: 28347654 DOI: 10.1016/j.diabet.2017.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/01/2017] [Indexed: 01/07/2023]
Abstract
AIMS The coadministration of alirocumab, a PCSK9 inhibitor for treatment of hypercholesterolaemia, and insulin in diabetes mellitus (DM) requires further study. Described here is the rationale behind a phase-IIIb study designed to characterize the efficacy and safety of alirocumab in insulin-treated patients with type 1 (T1) or type 2 (T2) DM with hypercholesterolaemia and high cardiovascular (CV) risk. METHODS ODYSSEY DM-INSULIN (NCT02585778) is a randomized, double-blind, placebo-controlled, multicentre study that planned to enrol around 400 T2 and up to 100 T1 insulin-treated DM patients. Participants had low-density lipoprotein cholesterol (LDL-C) levels at screening≥70mg/dL (1.81mmol/L) with stable maximum tolerated statin therapy or were statin-intolerant, and taking (or not) other lipid-lowering therapy; they also had established CV disease or at least one additional CV risk factor. Eligible patients were randomized 2:1 to 24weeks of alirocumab 75mg every 2weeks (Q2W) or a placebo. Alirocumab-treated patients with LDL-C≥70mg/dL at week 8 underwent a blinded dose increase to 150mg Q2W at week 12. Primary endpoints were the difference between treatment arms in percentage change of calculated LDL-C from baseline to week 24, and alirocumab safety. RESULTS This is an ongoing clinical trial, with 76 T1 and 441 T2 DM patients enrolled; results are expected in mid-2017. CONCLUSION The ODYSSEY DM-INSULIN study will provide information on the efficacy and safety of alirocumab in insulin-treated individuals with T1 or T2 DM who are at high CV risk and have hypercholesterolaemia not adequately controlled by the maximum tolerated statin therapy.
Collapse
|
219
|
Therapeutic efficacy and safety of PCSK9-monoclonal antibodies on familial hypercholesterolemia and statin-intolerant patients: A meta-analysis of 15 randomized controlled trials. Sci Rep 2017; 7:238. [PMID: 28331223 PMCID: PMC5428249 DOI: 10.1038/s41598-017-00316-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Proprotein convertase subtilisin/kexin9 monoclonal antibodies (PCSK9-mAb) have been studied intensively to identify their effect in lowering levels of low density lipoprotein cholesterol (LDL-C). However, the applicable target of PCSK9-mAbs remains inconclusive so far. Therefore, this first meta-analysis was carried out to clarify the therapeutic efficacy and safety of PCSK9-mAbs on the potential patients: familial hypercholesterolemia and statin-intolerant patients. All randomized controlled trials that met the search terms were retrieved in multiple databases. Efficacy outcomes included parameter changes from baseline in LDL-C and other lipid levels. Therapeutic safety were evaluated by rates of common adverse events. A total of 15 studies encompassing 4,288 patients with at least 8 weeks duration were selected. Overall, the therapeutic efficacy was achieved with significant reduction in LDL-C, TC, TG, Lp(a), Apo-B versus placebo. The decline in familial hypercholesterolemia patients (-53.28%, 95% CI: -59.88 to -46.68%) was even more obvious than that in statin-intolerant patients (-34.95%, 95% CI: -41.46 to -28.45%). No obvious safety difference was found out in the rates of common and serious adverse events. To conclude, PCSK9-mAb contributes to the decreased level of LDL-C and other lipids in familial hypercholesterolemia and statin-intolerant patients with satisfactory safety and tolerability.
Collapse
|
220
|
Tomlinson B, Hu M, Zhang Y, Chan P, Liu ZM. Alirocumab for the treatment of hypercholesterolemia. Expert Opin Biol Ther 2017; 17:633-643. [PMID: 28277798 DOI: 10.1080/14712598.2017.1305354] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Alirocumab is a human monoclonal antibody inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9) that is administered by subcutaneous injection every 2 weeks. Area covered: Herein, the authors discuss the background to inhibition of PCSK9 and the pharmacodynamics, pharmacokinetics and clinical trials with alirocumab. Alirocumab produces substantial reductions in low density lipoprotein cholesterol (LDL-C) in patients with and without background statin treatment. The safety profile appears very promising from the relatively short term studies that have been completed but there are some remaining concerns about long term risks of neurocognitive events and developing diabetes. Expert opinion: The profound reduction in LDL-C with alirocumab is most likely to translate into cardiovascular benefits in the ODYSSEY OUTCOMES trial and is unlikely in itself to result in any major adverse effects. The high cost and the current lack of long-term safety and efficacy data will restrict the use of alirocumab to patients who have high cardiovascular risk from established atherosclerotic cardiovascular disease or heterozygous familial hypercholesterolemia and who are unable to achieve LDL-C targets with maximally tolerated dose of statins with or without other lipid-lowering drugs. When further data become available, these indications are likely to be expanded.
Collapse
Affiliation(s)
- Brian Tomlinson
- a Research Center for Translational Medicine , Shanghai East Hospital Affiliated to Tongji University School of Medicine , Shanghai , China.,b Department of Medicine & Therapeutics , The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Miao Hu
- b Department of Medicine & Therapeutics , The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Yuzhen Zhang
- a Research Center for Translational Medicine , Shanghai East Hospital Affiliated to Tongji University School of Medicine , Shanghai , China
| | - Paul Chan
- c Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| | - Zhong-Min Liu
- d Department of Cardiac Surgery, Shanghai East Hospital , Tongji University , Shanghai , China
| |
Collapse
|
221
|
Rashid H, Meredith IT, Nasis A. PCSK9 Monoclonal Antibodies in 2016: Current Status and Future Challenges. Heart Lung Circ 2017; 26:786-798. [PMID: 28286091 DOI: 10.1016/j.hlc.2016.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 11/02/2016] [Accepted: 12/13/2016] [Indexed: 11/28/2022]
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in developed nations, with elevated low-density lipoprotein-cholesterol (LDL-C) levels being a major modifiable risk factor for coronary atherosclerosis. While lipid-lowering therapies such as statins are effective in lowering LDL-C, a proportion of patients do not achieve target LDL-C goals with statins or are intolerant to statins necessitating other treatment options. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are a new class of agents that reduce LDL-C beyond the maximum achievable LDL-C reductions with statins, and have been well studied in different patient groups. However, there are concerns regarding their potential adverse effects and cost, given that morbidity and mortality benefits have not yet been demonstrated. This state-of-the art review provides an overview of the development of PCSK9 inhibitors, the evidence regarding their clinical efficacy in specific target populations, and highlights future trials and challenges that need to be addressed before PCSK9 inhibitors are widely adopted into contemporary clinical practice.
Collapse
Affiliation(s)
- Hashrul Rashid
- Monash Cardiovascular Research Centre, MonashHEART, Monash Health and Monash University Department of Medicine (MMC), Melbourne, Vic, Australia
| | - Ian T Meredith
- Monash Cardiovascular Research Centre, MonashHEART, Monash Health and Monash University Department of Medicine (MMC), Melbourne, Vic, Australia
| | - Arthur Nasis
- Monash Cardiovascular Research Centre, MonashHEART, Monash Health and Monash University Department of Medicine (MMC), Melbourne, Vic, Australia.
| |
Collapse
|
222
|
Tatlock S, Arbuckle R, Sanchez R, Grant L, Khan I, Manvelian G, Spertus JA. Psychometric Evaluation of a Treatment Acceptance Measure for Use in Patients Receiving Treatment via Subcutaneous Injection. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2017; 20:430-440. [PMID: 28292488 DOI: 10.1016/j.jval.2016.09.2410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 08/15/2016] [Accepted: 09/28/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Alirocumab, a proprotein convertase subtilisin/kexin type 9 inhibitor, significantly reduces low-density lipoprotein cholesterol, but requires subcutaneous injections rather than oral pills. To measure patients' acceptance of this treatment modality, a new patient-reported outcome, the Injection-Treatment Acceptance Questionnaire (I-TAQ), was developed. OBJECTIVES To psychometrically evaluate the I-TAQ with patients at high risk of cardiovascular events receiving alirocumab. METHODS The 22-item, 5-domain I-TAQ was administered cross-sectionally to 151 patients enrolled in alirocumab clinical trials. Item response distributions, factor and multitrait analyses, interitem correlations, correlations with an existing measure of acceptance (convergent validity), and comparison of known-groups were performed to assess the I-TAQ's psychometric properties. RESULTS Completion rates were high, with no patients missing more than two items and 91.4% missing no data. All items displayed high ceiling effects (>30%) because of high treatment acceptance. Factor analysis supported the a priori hypothesized item-domain structure with good fit indices (root mean square error approximation = 0.070; comparative fit index = 0.988) and high factor loadings. All items demonstrated item convergent validity (item-scale correlation ≥0.40), except for the side effects domain, which was limited by small numbers (n = 46). Almost all items correlated most highly with the domain to which they were assigned (item discriminant validity). Internal reliability was acceptable for all domains (Cronbach α range 0.72-0.88) and convergent validity was supported by a logical pattern of correlations with the Chronic Treatment Acceptance Questionnaire. CONCLUSIONS These findings provide initial evidence of validity and reliability for the I-TAQ in patients treated with subcutaneous alirocumab. The I-TAQ could prove to be a valuable patient-reported outcome for therapies requiring subcutaneous injection.
Collapse
Affiliation(s)
| | | | | | - Laura Grant
- Adelphi Values Ltd., Bollington, Cheshire, UK
| | | | | | - John A Spertus
- Mid America Heart Institute of Saint Luke's Hospital and the University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
223
|
Efficacy and Safety of Alirocumab in Patients with Heterozygous Familial Hypercholesterolemia and LDL-C of 160 mg/dl or Higher. Cardiovasc Drugs Ther 2017; 30:473-483. [PMID: 27618825 PMCID: PMC5055560 DOI: 10.1007/s10557-016-6685-y] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Even with statins and other lipid-lowering therapy (LLT), many patients with heterozygous familial hypercholesterolemia (heFH) continue to have elevated low-density lipoprotein cholesterol (LDL-C) levels. ODYSSEY HIGH FH (NCT01617655) assessed the efficacy and safety of alirocumab, a proprotein convertase subtilisin/kexin type 9 monoclonal antibody, versus placebo in patients with heFH and LDL-C ≥ 160 mg/dl despite maximally tolerated statin ± other LLT. METHODS Patients were randomized to subcutaneous alirocumab 150 mg or placebo every 2 weeks (Q2W) for 78 weeks. The primary endpoint was percent change in LDL-C from baseline to week 24. RESULTS Mean baseline LDL-C levels were 196.3 mg/dl in the alirocumab (n = 71) and 201.0 mg/dl in the placebo groups (n = 35). Significant mean (standard error [SE]) reductions in LDL-C from baseline to week 24 were observed with alirocumab (-45.7 [3.5] %) versus placebo (-6.6 [4.9] %), a difference of -39.1 (6.0) % (P < 0.0001). Absolute mean (SE) LDL-C levels were reduced from baseline by 90.8 (6.7) mg/dl with alirocumab at week 24, with reductions maintained to week 78. Treatment-emergent adverse events were generally comparable between groups. Injection-site reactions were more frequent in the alirocumab group (8.3 %) versus placebo (5.7 %); most were mild in severity and did not result in study medication discontinuation. CONCLUSIONS In patients with heFH and very high LDL-C baseline levels despite maximally tolerated statin ± other LLT, alirocumab 150 mg Q2W demonstrated significant reductions in LDL-C levels with 41 % of patients achieving predefined LDL-C goals. Alirocumab was generally well tolerated.
Collapse
|
224
|
Abstract
Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors are novel agents indicated for the treatment of hyperlipidemia. Inhibition of PCSK9 produces an increase in surface low-density lipoprotein (LDL) receptors and increases removal of LDL from the circulation. Alirocumab (Praluent; Sanofi/Regeneron, Bridgewater, NJ) and evolocumab (Repatha; Amgen, Thousand Oaks, CA) are currently available and approved for use in patients with heterozygous familial hypercholesterolemia, homozygous familial hypercholesterolemia, and clinical atherosclerotic cardiovascular disease. Bococizumab (RN316; Pfizer, New York, NY) is currently being studied in similar indications, with an estimated approval date in late 2016. The pharmacodynamic effects of PCSK9 inhibitors have been extensively studied in various patient populations. They have been shown to produce significant reductions in LDL and are well tolerated in clinical studies, but they are very costly when compared with statins, the current mainstay of hyperlipidemia treatment. Clinical outcome studies are underway, but not yet available; however, meta-analyses have pointed to a reduction in cardiovascular death and cardiovascular events with the use of PCSK9 inhibitors. This review will discuss the novel mechanism of action of PCSK9 inhibitors, the results of clinical studies, and the clinical considerations of these agents in current therapy.
Collapse
|
225
|
Abstract
PURPOSE Chronic kidney disease (CKD) is accompanied by a number of secondary metabolic dysregulations, such as lipid abnormalities, presenting with unique characteristics. Proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors have been introduced as the new era in the management of dyslipidemia with promising results in groups with refractory lipid abnormalities. Increasing number of studies investigate the possible association of PCSK9 levels with kidney function, especially with nephrotic range proteinuria, as well as its role as a prognostic cardiovascular risk marker in CKD. In this review, we discuss the existing evidence for PCSK9 levels in patient groups with nephrotic syndrome, non-dialysis CKD, end-stage renal disease and kidney transplantation. METHODS Online research was conducted in MEDLINE database to identify articles investigating PCSK9 in all different aspects of CKD. References from relevant studies were screened for supplementary articles. RESULTS Four cross-sectional studies, one secondary analysis, one publication from two independent cohort studies and one multicentre prospective cohort study assessed PCSK9 plasma levels in different subgroups of CKD patients. PCSK9 levels increase in nephrotic syndrome and have a positive correlation with proteinuria. In CKD patients, no correlation was found between PCSK9 levels and estimated GFR. Peritoneal dialysis patients have higher PCSK9 levels compared with hemodialysis and renal transplant patients as well as general population. CONCLUSION Accumulative evidence focuses on the possible association of PCSK9 levels with kidney function. No data are available for the administration of PCSK9 inhibitors in CKD patients. Further research will optimize knowledge on the role of PCSK9 levels and PCSK9 inhibitors in CKD.
Collapse
|
226
|
Androulakis E, Zacharia E, Papageorgiou N, Lioudaki E, Bertsias D, Charakida M, Siasos G, Tousoulis D. High-density Lipoprotein and Low-density Lipoprotein Therapeutic Approaches in Acute Coronary Syndromes. Curr Cardiol Rev 2017; 13:168-182. [PMID: 28190386 PMCID: PMC5633711 DOI: 10.2174/1573403x13666170209145622] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/26/2017] [Accepted: 02/03/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL), and especially its oxidized form, renders the atherosclerotic plaque vulnerable to rupture in acute coronary syndromes (ACS). On the other hand, high-density lipoprotein (HDL) is considered an anti-atherogenic molecule. The more recent HDL-targeted drugs may prove to be superior to those used before. Indeed, delipidated HDL and HDL mimetics are efficient in increasing HDL levels, while the apoA-I upregulation with RVX-208 appears to offer a clinical benefit which is beyond the HDL related effects. HDL treatment however has not shown a significant improvement in the outcomes of patients with ACS so far, studies have therefore focused again on LDL. In addition to statins and ezetimibe, novel drugs such as PSCK9 inhibitors and apolipoprotein B inhibitors appear to be both effective and safe for patients with hyperlipidemia. CONCLUSION Data suggest these could potentially improve the cardiovascular outcomes of patient with ACS. Yet, there is still research to be done, in order to confirm whether ACS patients would benefit from LDL- or HDL-targeted therapies or a combination of both.
Collapse
Affiliation(s)
| | - Effimia Zacharia
- 1 Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | | | | | - Dimitris Bertsias
- 1 Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | | | - Gerasimos Siasos
- 1 Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- 1 Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| |
Collapse
|
227
|
Ito MK, Santos RD. PCSK9 Inhibition With Monoclonal Antibodies: Modern Management of Hypercholesterolemia. J Clin Pharmacol 2017; 57:7-32. [PMID: 27195910 PMCID: PMC5215586 DOI: 10.1002/jcph.766] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 01/31/2023]
Abstract
Current guidelines for hypercholesterolemia treatment emphasize lifestyle modification and lipid-modifying therapy to reduce the risk for cardiovascular disease. Statins are the primary class of agents used for the treatment of hypercholesterolemia. Although statins are effective for many patients, they fail to achieve optimal reduction in lipids for some patients, including those who have or are at high risk for cardiovascular disease. The PCSK9 gene was identified in the past decade as a potential therapeutic target for the management of patients with hypercholesterolemia. Pharmacologic interventions to decrease PCSK9 levels are in development, with the most promising approach using monoclonal antibodies that bind to PCSK9 in the plasma. Two monoclonal antibodies, alirocumab and evolocumab, have recently been approved for the treatment of hypercholesterolemia, and a third one, bococizumab, is in phase 3 clinical development. All 3 agents achieve significant reductions in levels of low-density lipoprotein cholesterol, as well as reductions in non-high-density lipoprotein cholesterol, apolipoprotein B, and lipoprotein(a). Long-term outcome trials are under way to determine the sustained efficacy, safety, and tolerability of PCSK9 inhibitors and whether this novel class of agents decreases the risk for major cardiovascular events in patients on lipid-modifying therapy. Available data suggest that PCSK9 inhibitors provide a robust reduction in atherogenic cholesterol levels with a good safety profile, especially for patients who fail to obtain an optimal clinical response to statin therapy, those who are statin intolerant or have contraindications to statin therapy, and those with familial hypercholesterolemia.
Collapse
Affiliation(s)
- Matthew K. Ito
- Sanofi USBridgewaterNJUSA
- Oregon State University/Oregon Health and Science UniversityCollege of PharmacyPortlandOR, USA (during initial development of this article)
| | - Raul D. Santos
- Lipid Clinic, Heart Institute (InCor), University of Sao Paulo Medical School Hospital, and Preventive Medicine Center and Cardiology ProgramHospital Israelita Albert EinsteinSao PauloBrazil
| |
Collapse
|
228
|
Alberto Z. Reducing cholesterol levels to decrease cardiovascular events. J Cardiovasc Med (Hagerstown) 2017; 18 Suppl 1:e184-e186. [DOI: 10.2459/jcm.0000000000000468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
229
|
Kastelein JJ, Hovingh GK, Langslet G, Baccara-Dinet MT, Gipe DA, Chaudhari U, Zhao J, Minini P, Farnier M. Efficacy and safety of the proprotein convertase subtilisin/kexin type 9 monoclonal antibody alirocumab vs placebo in patients with heterozygous familial hypercholesterolemia. J Clin Lipidol 2017; 11:195-203.e4. [DOI: 10.1016/j.jacl.2016.12.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/11/2016] [Indexed: 01/30/2023]
|
230
|
Khan AR, Bavishi C, Riaz H, Farid TA, Khan S, Atlas M, Hirsch G, Ikram S, Bolli R. Increased Risk of Adverse Neurocognitive Outcomes With Proprotein Convertase Subtilisin-Kexin Type 9 Inhibitors. Circ Cardiovasc Qual Outcomes 2017; 10:CIRCOUTCOMES.116.003153. [DOI: 10.1161/circoutcomes.116.003153] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/26/2016] [Indexed: 01/16/2023]
Abstract
Background—
There is encouraging evidence of the efficacy of proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors; however, their long-term safety remains unclear. We performed a meta-analysis of studies to evaluate the long-term safety of PCSK9 inhibitors.
Methods and Results—
Our search strategy yielded 11 studies (9 smaller early-phase and 2 larger outcome trials). The outcomes assessed were cumulative serious adverse events, musculoskeletal adverse events, neurocognitive adverse events, and stroke. Odds ratio (OR) was calculated using the Mantel–Haenszel method. Subgroup analysis was done to assess the difference in safety between the smaller early-phase studies and the larger outcome studies. Our meta-analysis suggested no difference in the incidence of serious adverse events (OR, 1.00; 95% confidence interval [CI], 0.88−1.15), musculoskeletal adverse events (OR, 1.01; 95% CI, 0.87−1.13), neurocognitive adverse events (OR, 1.29; 95% CI, 0.64−2.59), or stroke (OR, 1.44; 95% CI, 0.57−3.65) with the use of PCSK9 inhibitors. Subgroup analysis of the 2 large outcome studies did suggest an increased incidence of neurocognitive adverse events (OR, 2.85; 95% CI, 1.34−6.06) with the use of PCSK9 inhibitors. However, the overall incidence of neurocognitive adverse events and stroke was <1%, whereas the cumulative incidence of serious adverse events and musculoskeletal events was >10% in both the groups.
Conclusions—
Our analysis suggests that PCSK9 inhibitors are not associated with an increased risk of cumulative severe adverse effects, musculoskeletal effects, or stroke. There is a signal toward adverse neurocognitive effects, seen in the outcome studies with a larger sample size and longer follow-up. There should be close monitoring, for the increased risk of neurocognitive events in the ongoing outcome studies and post-marketing surveillance.
Collapse
Affiliation(s)
- Abdur Rahman Khan
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Chirag Bavishi
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Haris Riaz
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Talha A. Farid
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Sobia Khan
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Michel Atlas
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Glenn Hirsch
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Sohail Ikram
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| | - Roberto Bolli
- From the Division of Cardiovascular Medicine (A.R.K., T.A.F., S.K., G.H., S.I., R.B.), Institute of Molecular Cardiology (A.R.K., T.A.F., R.B.), and Kornhauser Health Sciences Library (M.A.), University of Louisville, KY; Division of Cardiovascular Medicine, St Lukes Roosevelt Hospital, New York, NY (C.B.); and Department of Internal Medicine, Cleveland Clinic, OH (H.R.)
| |
Collapse
|
231
|
Gaudet D, Watts GF, Robinson JG, Minini P, Sasiela WJ, Edelberg J, Louie MJ, Raal FJ. Effect of Alirocumab on Lipoprotein(a) Over ≥1.5 Years (from the Phase 3 ODYSSEY Program). Am J Cardiol 2017; 119:40-46. [PMID: 27793396 DOI: 10.1016/j.amjcard.2016.09.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 10/20/2022]
Abstract
Elevated lipoprotein(a) [Lp(a)] is independently associated with increased cardiovascular risk. However, treatment options for elevated Lp(a) are limited. Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9, reduced low-density lipoprotein cholesterol (LDL-C) by up to 62% from baseline in phase 3 studies, with adverse event rates similar between alirocumab and controls. We evaluated the effect of alirocumab on serum Lp(a) using pooled data from the phase 3 ODYSSEY program: 4,915 patients with hypercholesterolemia from 10 phase 3 studies were included. Eight studies evaluated alirocumab 75 mg every 2 weeks (Q2W), with possible increase to 150 mg Q2W at week 12 depending on LDL-C at week 8 (75/150 mg Q2W); the other 2 studies evaluated alirocumab 150-mg Q2W from the outset. Comparators were placebo or ezetimibe. Eight studies were conducted on a background of statins, and 2 studies were carried out with no statins. Alirocumab was associated with significant reductions in Lp(a), regardless of starting dose and use of concomitant statins. At week 24, reductions from baseline were 23% to 27% with alirocumab 75/150-mg Q2W and 29% with alirocumab 150-mg Q2W (all comparisons p <0.0001 vs controls). Reductions were sustained over 78 to 104 weeks. Lp(a) reductions with alirocumab were independent of race, gender, presence of familial hypercholesterolemia, baseline Lp(a), and LDL-C concentrations, or use of statins. In conclusion, in addition to marked reduction in LDL-C, alirocumab leads to a significant and sustained lowering of Lp(a).
Collapse
|
232
|
Reyes-Soffer G, Pavlyha M, Ngai C, Thomas T, Holleran S, Ramakrishnan R, Karmally W, Nandakumar R, Fontanez N, Obunike J, Marcovina SM, Lichtenstein AH, Matthan NR, Matta J, Maroccia M, Becue F, Poitiers F, Swanson B, Cowan L, Sasiela WJ, Surks HK, Ginsberg HN. Effects of PCSK9 Inhibition With Alirocumab on Lipoprotein Metabolism in Healthy Humans. Circulation 2016; 135:352-362. [PMID: 27986651 PMCID: PMC5262523 DOI: 10.1161/circulationaha.116.025253] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/07/2016] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), lowers plasma low-density lipoprotein (LDL) cholesterol and apolipoprotein B100 (apoB). Although studies in mice and cells have identified increased hepatic LDL receptors as the basis for LDL lowering by PCSK9 inhibitors, there have been no human studies characterizing the effects of PCSK9 inhibitors on lipoprotein metabolism. In particular, it is not known whether inhibition of PCSK9 has any effects on very low-density lipoprotein or intermediate-density lipoprotein (IDL) metabolism. Inhibition of PCSK9 also results in reductions of plasma lipoprotein (a) levels. The regulation of plasma Lp(a) levels, including the role of LDL receptors in the clearance of Lp(a), is poorly defined, and no mechanistic studies of the Lp(a) lowering by alirocumab in humans have been published to date. Methods: Eighteen (10 F, 8 mol/L) participants completed a placebo-controlled, 2-period study. They received 2 doses of placebo, 2 weeks apart, followed by 5 doses of 150 mg of alirocumab, 2 weeks apart. At the end of each period, fractional clearance rates (FCRs) and production rates (PRs) of apoB and apo(a) were determined. In 10 participants, postprandial triglycerides and apoB48 levels were measured. Results: Alirocumab reduced ultracentrifugally isolated LDL-C by 55.1%, LDL-apoB by 56.3%, and plasma Lp(a) by 18.7%. The fall in LDL-apoB was caused by an 80.4% increase in LDL-apoB FCR and a 23.9% reduction in LDL-apoB PR. The latter was due to a 46.1% increase in IDL-apoB FCR coupled with a 27.2% decrease in conversion of IDL to LDL. The FCR of apo(a) tended to increase (24.6%) without any change in apo(a) PR. Alirocumab had no effects on FCRs or PRs of very low-density lipoproteins-apoB and very low-density lipoproteins triglycerides or on postprandial plasma triglycerides or apoB48 concentrations. Conclusions: Alirocumab decreased LDL-C and LDL-apoB by increasing IDL- and LDL-apoB FCRs and decreasing LDL-apoB PR. These results are consistent with increases in LDL receptors available to clear IDL and LDL from blood during PCSK9 inhibition. The increase in apo(a) FCR during alirocumab treatment suggests that increased LDL receptors may also play a role in the reduction of plasma Lp(a). Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01959971.
Collapse
Affiliation(s)
- Gissette Reyes-Soffer
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.).
| | - Marianna Pavlyha
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Colleen Ngai
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Tiffany Thomas
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Stephen Holleran
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Rajasekhar Ramakrishnan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Wahida Karmally
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Renu Nandakumar
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Nelson Fontanez
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Joseph Obunike
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Santica M Marcovina
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Alice H Lichtenstein
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Nirupa R Matthan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - James Matta
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Magali Maroccia
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Frederic Becue
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Franck Poitiers
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Brian Swanson
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Lisa Cowan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - William J Sasiela
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Howard K Surks
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Henry N Ginsberg
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.).
| |
Collapse
|
233
|
Jones PH, Bays HE, Chaudhari U, Pordy R, Lorenzato C, Miller K, Robinson JG. Safety of Alirocumab (A PCSK9 Monoclonal Antibody) from 14 Randomized Trials. Am J Cardiol 2016; 118:1805-1811. [PMID: 27729106 DOI: 10.1016/j.amjcard.2016.08.072] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 10/21/2022]
Abstract
Previous individual trials of alirocumab (a PCSK9 monoclonal antibody) showed significant low-density lipoprotein cholesterol reductions with overall treatment-emergent adverse event (TEAE) rates comparable with controls. This analysis evaluated safety data from 14 trials (4 phase 2 and 10 phase 3, 8 to 104 weeks; n = 5,234), in 2 pools according to control (placebo/ezetimibe). Overall, 3,340 patients received alirocumab (4,029 patient-years' exposure), 1,276 received placebo, and 618 received ezetimibe. Incidence of deaths, serious TEAEs, discontinuations because of TEAEs, and overall TEAEs were similar between alirocumab and control groups. Alirocumab was associated with a higher incidence of local injection site reactions (7.4% vs 5.3% with placebo; 3.1% vs 2.3% with ezetimibe), pruritus (1.3% vs 0.4% placebo; 0.9% vs 0.5% ezetimibe), and upper respiratory tract infection signs and symptoms (2.1% vs 1.1% placebo; 1.3% vs 0.8% ezetimibe). Incidence of musculoskeletal, neurologic, neurocognitive, ophthalmologic, hepatic events, and TEAEs related to diabetes/diabetes complications was similar between alirocumab and control groups. In a prespecified analysis of phase 3 studies, adjudicated major adverse cardiovascular events (coronary heart disease death, nonfatal myocardial infarction, ischemic stroke, and unstable angina requiring hospitalization) occurred in 1.8% alirocumab versus 2.6% placebo patients (hazard ratio 0.69, 95% confidence interval 0.43 to 1.11) and 2.8% alirocumab versus 1.5% ezetimibe patients (hazard ratio 1.4, 95% confidence interval 0.65 to 3.02). In conclusion, pooled safety data from 14 trials demonstrate that alirocumab is generally well tolerated with a favorable safety profile.
Collapse
|
234
|
Ference BA, Robinson JG, Brook RD, Catapano AL, Chapman MJ, Neff DR, Voros S, Giugliano RP, Davey Smith G, Fazio S, Sabatine MS. Variation in PCSK9 and HMGCR and Risk of Cardiovascular Disease and Diabetes. N Engl J Med 2016; 375:2144-2153. [PMID: 27959767 DOI: 10.1056/nejmoa1604304] [Citation(s) in RCA: 560] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pharmacologic inhibitors of proprotein convertase subtilisin-kexin type 9 (PCSK9) are being evaluated in clinical trials for the treatment of cardiovascular disease. The effect of lowering low-density lipoprotein (LDL) cholesterol levels by inhibiting PCSK9 on the risk of cardiovascular events or diabetes is unknown. METHODS We used genetic scores consisting of independently inherited variants in the genes encoding PCSK9 and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR; the target of statins) as instruments to randomly assign 112,772 participants from 14 studies, with 14,120 cardiovascular events and 10,635 cases of diabetes, to groups according to the number of LDL cholesterol-lowering alleles that they had inherited. We compared the effects of lower LDL cholesterol levels that were mediated by variants in PCSK9, HMGCR, or both on the risk of cardiovascular events and the risk of diabetes. RESULTS Variants in PCSK9 and HMGCR were associated with nearly identical protective effects on the risk of cardiovascular events per decrease of 10 mg per deciliter (0.26 mmol per liter) in the LDL cholesterol level: odds ratio for cardiovascular events, 0.81 (95% confidence interval [CI], 0.74 to 0.89) for PCSK9 and 0.81 (95% CI, 0.72 to 0.90) for HMGCR. Variants in these two genes were also associated with very similar effects on the risk of diabetes: odds ratio for each 10 mg per deciliter decrease in LDL cholesterol, 1.11 (95% CI, 1.04 to 1.19) for PCSK9 and 1.13 (95% CI, 1.06 to 1.20) for HMGCR. The increased risk of diabetes was limited to persons with impaired fasting glucose levels for both scores and was lower in magnitude than the protective effect against cardiovascular events. When present together, PCSK9 and HMGCR variants had additive effects on the risk of both cardiovascular events and diabetes. CONCLUSIONS In this study, variants in PCSK9 had approximately the same effect as variants in HMGCR on the risk of cardiovascular events and diabetes per unit decrease in the LDL cholesterol level. The effects of these variants were independent and additive. (Funded by the Medical Research Council and the National Heart, Lung, and Blood Institute.).
Collapse
Affiliation(s)
- Brian A Ference
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Jennifer G Robinson
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Robert D Brook
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Alberico L Catapano
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - M John Chapman
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - David R Neff
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Szilard Voros
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Robert P Giugliano
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - George Davey Smith
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Sergio Fazio
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| | - Marc S Sabatine
- From the Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit (B.A.F.), the Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor (R.D.B.), and Michigan State University, East Lansing (D.R.N.) - all in Michigan; the Departments of Epidemiology and Medicine, College of Public Health, University of Iowa, Iowa City (J.G.R.); the Department of Pharmacological and Biomolecular Sciences, University of Milan and MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan (A.L.C.); INSERM, Pitié-Salpêtrière University Hospital, Paris (M.J.C.); the Global Genomics Group, Richmond, VA (S.V.); the Thrombolysis in Myocardial Infarction Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston (R.P.G., M.S.S.); the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (G.D.S.); and the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, Portland (S.F.)
| |
Collapse
|
235
|
Meah F, Basit A, Mazhari A, Emanuele MA, Emanuele N. New lipid therapies: PCSK9 inhibitors. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2016. [DOI: 10.1016/j.jecr.2016.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
236
|
Fanton-Aita F, Matteau A, Iliza AC, Mitchell D, Guertin JR, Dubois A, Dubé MP, Tardif JC, LeLorier J. Maximal expected benefits from lowering cholesterol in primary prevention for a high-risk population. Curr Med Res Opin 2016; 32:1955-1958. [PMID: 27648984 DOI: 10.1080/03007995.2016.1222514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AIMS The objective of this study was to estimate the maximal clinical benefit that could be reasonably expected from a cholesterol-lowering intervention. MATERIALS AND METHODS We used a hypothetical population at high risk of cardiovascular disease events from three risk assessment models including the Framingham risk function, the Score Canada and the Pooled Cohort Risk Assessment Equations. Our source population were all 55-year-old smoking men with diabetes, hypertension and low HDL. From this population, we identified two different subpopulations named "high" and "low", referring to their cholesterol levels which were set at 8.60 and 4.14 mmol/L respectively. Both subpopulations were identified for each risk assessment model in order to estimate the maximal impact of lowering cholesterol on cardiovascular disease events. RESULTS Our extrapolations estimated that the maximal theoretical efficacy of a cholesterol-lowering intervention corresponds to a risk ratio ranging between 0.46 and 0.66 over a 10-year period. The number of events prevented during this period were between 21 and 29 per 100 patients which corresponds to a number needed to treat varying from 3.47 to 4.76. CONCLUSIONS Our estimation showed the maximal clinical benefit that could be reasonably expected by an intervention that would lower total cholesterol in high-risk patients.
Collapse
Affiliation(s)
- Fiorella Fanton-Aita
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- b Centre de Recherche du Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
| | - Alexis Matteau
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- b Centre de Recherche du Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
- c Cardiology Division , Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
| | - Ange Christelle Iliza
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- b Centre de Recherche du Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
| | - Dominic Mitchell
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- b Centre de Recherche du Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
| | - Jason R Guertin
- d McMaster University , Department of Clinical Epidemiology and Biostatistics , Hamilton , ON , Canada
- e Programs for Assessment of Technology in Health, St. Joseph's Healthcare Hamilton , Hamilton , ON , Canada
| | - Anick Dubois
- f Beaulieu-Saucier Pharmacogenomics Centre, Montreal Heart Institute, Université de Montréal , Montreal , QC , Canada
| | - Marie-Pierre Dubé
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- f Beaulieu-Saucier Pharmacogenomics Centre, Montreal Heart Institute, Université de Montréal , Montreal , QC , Canada
| | - Jean-Claude Tardif
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- f Beaulieu-Saucier Pharmacogenomics Centre, Montreal Heart Institute, Université de Montréal , Montreal , QC , Canada
| | - Jacques LeLorier
- a Faculté de Médecine , Université de Montréal , Montreal , QC , Canada
- b Centre de Recherche du Centre Hospitalier de l'Université de Montréal , Montreal , QC , Canada
| |
Collapse
|
237
|
Gouni‐Berthold I, Descamps OS, Fraass U, Hartfield E, Allcott K, Dent R, März W. Systematic review of published Phase 3 data on anti-PCSK9 monoclonal antibodies in patients with hypercholesterolaemia. Br J Clin Pharmacol 2016; 82:1412-1443. [PMID: 27478094 PMCID: PMC5099564 DOI: 10.1111/bcp.13066] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/14/2016] [Accepted: 07/04/2016] [Indexed: 01/06/2023] Open
Abstract
AIMS Two anti-proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibodies, alirocumab and evolocumab, have been approved for the treatment of hypercholesterolaemia in certain patients. We reviewed data from Phase 3 studies to evaluate the efficacy and safety of these antibodies. METHODS We systematically reviewed Phase 3 English-language studies in patients with hypercholesterolaemia, published between 1 January 2005 and 20 October 2015. Congress proceedings from 16 November 2012 to 16 November 2015 were also reviewed. RESULTS We identified 12 studies of alirocumab and nine of evolocumab, including over 10 000 patients overall. Most studies enrolled patients with hypercholesterolaemia and used anti-PCSK9 antibodies with statins. The ODYSSEY FH I, FH II and HIGH FH alirocumab studies and the RUTHERFORD-2 evolocumab study exclusively recruited patients with heterozygous familial hypercholesterolaemia. Two evolocumab studies focused mainly on homozygous familial hypercholesterolaemia (HoFH): TESLA Part B and TAUSSIG (a TESLA sub-study); only those data for HoFH are reported here. All comparator studies demonstrated a reduction in LDL cholesterol (LDL-C) with the anti-PCSK9 antibodies. No head-to-head studies were conducted between alirocumab and evolocumab. Up to 87% of patients receiving alirocumab and up to 98% receiving evolocumab reached LDL-C goals. Both antibodies were effective and well tolerated across a broad population of patients and in specific subgroups, such as those with type 2 diabetes. CONCLUSIONS Using anti-PCSK9 antibodies as add-on therapy to other lipid-lowering treatments or as monotherapy for patients unable to tolerate statins may help patients with high cardiovascular risk to achieve their LDL-C goals.
Collapse
Affiliation(s)
- Ioanna Gouni‐Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine (ZEDP)University of CologneCologneGermany
| | - Olivier S. Descamps
- Department of Internal MedicineCentres Hospitaliers JolimontHaine Saint‐PaulBelgium
| | | | | | | | | | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University GrazGrazAustria
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty MannheimUniversity of HeidelbergGermany
- Synlab AcademySynlab Holding Deutschland GmbHMannheim and AugsburgGermany
| |
Collapse
|
238
|
Gupta S. Development of proprotein convertase subtilisin/kexin type 9 inhibitors and the clinical potential of monoclonal antibodies in the management of lipid disorders. Vasc Health Risk Manag 2016; 12:421-433. [PMID: 27877050 PMCID: PMC5108562 DOI: 10.2147/vhrm.s83719] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The aim of this manuscript is to review available data to evaluate the present status of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors in the treatment of hypercholesterolemia. Relevant literature since 2003 is reviewed. The effectiveness of PCSK9 inhibitors in lowering low-density lipoprotein cholesterol and other atherogenic lipid fractions was studied in various Phase 2 and Phase 3 trials of Alirocumab, Evolocumab, and Bococizumab. The results of published long-term ODYSSEY and OSLER studies are summarized. There have been three excellent meta-analysis studies on PCSK9 inhibitors which are outlined. The complex problem of cost-effectiveness was carefully evaluated by the Institute for Clinical and Economic Review (ICER). The draft report (ICER-2015) is summarized herewith. The cardiovascular outcome trials with Evolocumab (FOURIER), Alirocumab (ODYSSEY OUTCOME) and Bococizumab (SPIRE-1 and SPIRE-2) are the ongoing clinical trials, and their results are expected in 2017-2018. The search for new cost-effective analogs of PCSK9 inhibitors is ongoing.
Collapse
Affiliation(s)
- Sanjiv Gupta
- Department of Interventional Cardiology, Santokba Durlabhji Memorial Hospital Cum Medical Research Institute, Jaipur, India
| |
Collapse
|
239
|
Population Pharmacokinetics (PK) and Pharmacodynamics (PD) Analysis of LY3015014, a Monoclonal Antibody to Protein Convertase Subtilisin/Kexin Type 9 (PCSK9) in Healthy Subjects and Hypercholesterolemia Patients. Pharm Res 2016; 34:185-192. [DOI: 10.1007/s11095-016-2054-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/17/2016] [Indexed: 10/20/2022]
|
240
|
Farnier M, Gaudet D, Valcheva V, Minini P, Miller K, Cariou B. Efficacy of alirocumab in high cardiovascular risk populations with or without heterozygous familial hypercholesterolemia: Pooled analysis of eight ODYSSEY Phase 3 clinical program trials. Int J Cardiol 2016; 223:750-757. [DOI: 10.1016/j.ijcard.2016.08.273] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/16/2016] [Indexed: 12/14/2022]
|
241
|
McCormack T, Dent R, Blagden M. Very low LDL-C levels may safely provide additional clinical cardiovascular benefit: the evidence to date. Int J Clin Pract 2016; 70:886-897. [PMID: 27739167 PMCID: PMC5215677 DOI: 10.1111/ijcp.12881] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of death in Europe and increased low-density lipoprotein cholesterol (LDL-C) is a major contributor to CVD risk. Extensive evidence from clinical studies of statins has demonstrated a linear relationship between LDL-C levels and CVD risk. It has been proposed that lower LDL-C levels than those currently recommended may provide additional clinical benefit to patients. AIM This review summarises the genetic and clinical evidence on the efficacy and safety of achieving very low LDL-C levels. METHODS Relevant epidemiological and clinical studies were identified using PubMed and by searching abstracts published at major congresses. RESULTS Genetic evidence demonstrates that individuals with naturally very low LDL-C levels are healthy and have a low risk of CVD. Clinical evidence has shown that those patients who achieve very low LDL-C levels through using lipid-lowering therapies (LLTs), such as statins, have reduced CVD risk compared with patients who only just achieve recommended target LDL-C levels. These data show that the incidence of adverse events in patients achieving very low LDL-C levels using LLT is comparable to those reaching the recommended LDL-C targets. CONCLUSIONS Genetic and clinical evidence supports the concept that reduction in LDL-C levels below current recommended targets may provide additional clinical benefit to patients without adversely impacting patient safety. Statin add-on therapies, such as ezetimibe and the recently approved proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors alirocumab and evolocumab, allow patients to achieve very low LDL-C levels and are likely to impact on future treatment paradigms.
Collapse
Affiliation(s)
- Terry McCormack
- Hull York Medical SchoolWhitby Group PracticeSpring Vale Medical CentreWhitbyUK
| | | | | |
Collapse
|
242
|
Ray KK, Ginsberg HN, Davidson MH, Pordy R, Bessac L, Minini P, Eckel RH, Cannon CP. Reductions in Atherogenic Lipids and Major Cardiovascular Events: A Pooled Analysis of 10 ODYSSEY Trials Comparing Alirocumab With Control. Circulation 2016; 134:1931-1943. [PMID: 27777279 PMCID: PMC5147039 DOI: 10.1161/circulationaha.116.024604] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/06/2016] [Indexed: 01/14/2023]
Abstract
Supplemental Digital Content is available in the text. Background: A continuous relationship between reductions in low-density lipoprotein cholesterol (LDL-C) and major adverse cardiovascular events (MACE) has been observed in statin and ezetimibe outcomes trials down to achieved levels of 54 mg/dL. However, it is uncertain whether this relationship extends to LDL-C levels <50 mg/dL. We assessed the relationship between additional LDL-C, non–high-density lipoprotein cholesterol, and apolipoprotein B100 reductions and MACE among patients within the ODYSSEY trials that compared alirocumab with controls (placebo/ezetimibe), mainly as add-on therapy to maximally tolerated statin. Methods: Data were pooled from 10 double-blind trials (6699 patient-years of follow-up). Randomization was to alirocumab 75/150 mg every 2 weeks or control for 24 to 104 weeks, added to background statin therapy in 8 trials. This analysis included 4974 patients (3182 taking alirocumab, 1174 taking placebo, 618 taking ezetimibe). In a post hoc analysis, the relationship between average on-treatment lipid levels and percent reductions in lipids from baseline were correlated with MACE (coronary heart disease death, nonfatal myocardial infarction, ischemic stroke, or unstable angina requiring hospitalization) in multivariable analyses. Results: Overall, 33.1% of the pooled cohort achieved average LDL-C <50 mg/dL (44.7%–52.6% allocated to alirocumab, 6.5% allocated to ezetimibe, and 0% allocated to placebo). In total, 104 patients experienced MACE (median time to event, 36 weeks). For every 39 mg/dL lower achieved LDL-C, the risk of MACE appeared to be 24% lower (adjusted hazard ratio, 0.76; 95% confidence interval, 0.63–0.91; P=0.0025). Percent reductions in LDL-C from baseline were inversely correlated with MACE rates (hazard ratio, 0.71; 95% confidence interval, 0.57–0.89 per additional 50% reduction from baseline; P=0.003). Strengths of association materially similar to those described for LDL-C were observed with achieved non–high-density lipoprotein cholesterol and apolipoprotein B100 levels or percentage reductions. Conclusions: In a post hoc analysis from 10 ODYSSEY trials, greater percentage reductions in LDL-C and lower on-treatment LDL-C were associated with a lower incidence of MACE, including very low levels of LDL-C (<50 mg/dL). These findings require further validation in the ongoing prospective ODYSSEY OUTCOMES trial. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifiers: NCT01507831, NCT01623115, NCT01709500, NCT01617655, NCT01644175, NCT01644188, NCT01644474, NCT01730040, NCT01730053, and NCT01709513.
Collapse
Affiliation(s)
- Kausik K Ray
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.).
| | - Henry N Ginsberg
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Michael H Davidson
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Robert Pordy
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Laurence Bessac
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Pascal Minini
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Robert H Eckel
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| | - Christopher P Cannon
- From Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK (K.K.R.); Columbia University, New York, NY (H.N.G.); Department of Medicine, University of Chicago Medicine, Chicago, IL (M.H.D.); Regeneron Pharmaceuticals, Inc Tarrytown, NY (R.P.); Sanofi, Paris, France (L.B.); Biostatistics and Programming, Sanofi, Chilly-Mazarin, France (P.M.); University of Colorado, Anschutz Medical Campus, Aurora (R.H.E.); and Harvard Clinical Research Institute, Boston, MA (C.P.C.)
| |
Collapse
|
243
|
Adamski P, Adamska U, Ostrowska M, Koziński M, Kubica J. New directions for pharmacotherapy in the treatment of acute coronary syndrome. Expert Opin Pharmacother 2016; 17:2291-2306. [PMID: 27677394 DOI: 10.1080/14656566.2016.1241234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Acute coronary syndromes (ACS) are one of the leading causes of death worldwide. Several landmark trials, followed by a widespread introduction of new agents, have significantly improved ACS outcomes in recent years. However, despite the use of contemporary therapy, a substantial number of ACS patients continue to suffer from cardiovascular events. Areas covered: The aim of this review was to summarize available data on innovative drugs and pharmacological strategies that have potential to amend the current ACS therapy. We present the results of recent large clinical trials, as well as insights from ongoing phase III and phase IV studies, exploring the value of new strategies for the improvement of outcomes in ACS. Expert opinion: More potent platelet inhibition, more profound lipid reduction and possibly anti-inflammatory action are considered to have potential to further reduce the rates of adverse cardiovascular and thrombotic events in ACS patients. 'Hit fast, hit hard' approach regarding novel antiplatelet and lipid-lowering therapy seems attractive, but it has to be considered that these strategies may be associated with increased adverse events rate. Introduction of cangrelor and ezetimibe, and potentially future recognition of proprotein convertase subtilisin/kexin type 9 antibodies, are likely to alter the landscape of ACS pharmacotherapy.
Collapse
Affiliation(s)
- Piotr Adamski
- a Department of Principles of Clinical Medicine, Collegium Medicum , Nicolaus Copernicus University , Bydgoszcz , Poland
| | - Urszula Adamska
- b Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Collegium Medicum , Nicolaus Copernicus University , Bydgoszcz , Poland
| | - Małgorzata Ostrowska
- a Department of Principles of Clinical Medicine, Collegium Medicum , Nicolaus Copernicus University , Bydgoszcz , Poland
| | - Marek Koziński
- a Department of Principles of Clinical Medicine, Collegium Medicum , Nicolaus Copernicus University , Bydgoszcz , Poland
| | - Jacek Kubica
- c Department of Cardiology and Internal Medicine, Collegium Medicum , Nicolaus Copernicus University , Bydgoszcz , Poland
| |
Collapse
|
244
|
Yadav K, Sharma M, Ferdinand KC. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors: Present perspectives and future horizons. Nutr Metab Cardiovasc Dis 2016; 26:853-862. [PMID: 27352986 DOI: 10.1016/j.numecd.2016.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/04/2016] [Accepted: 05/17/2016] [Indexed: 12/31/2022]
Abstract
AIMS Our comprehensive review highlights the drug development and pharmacogenomics leading to the recent approval of PCSK9 inhibitors. We also review the anticipated future advances into the uses of PCSK9 inhibition. BACKGROUND Despite the present advances in pharmacotherapy, atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Low density lipoprotein-cholesterol (LDL-C) lowering is the primary target for ASCVD risk reduction, showing demonstrable benefits in mortality. However, 70% of events occur even in the presence of statins. This residual risk may be approached with additional LDL-C reduction. Statin intolerance is a common clinical concern affecting adherence and the benefit with statins. There is also significant variation of individual lipid-lowering. Following rapid development, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have progressed from genetic observations, to mechanistic studies, to closer realization of the goal of CVD risk reduction. This review discusses the science behind PCSK9 inhibition, evidence of trials involving efficacy and safety, and reflections of its present and future role in clinical care, especially in high-risk patients with ASCVD, persons with suboptimal responses to statins and familial hyperlipidemia. Monoclonal antibodies have demonstrated LDL-C lowering of up to 57% as monotherapy and up to 73% when added to statins. Statins have limited efficacy in reduction of LDL-C due to an increased number of LDL-receptors. Elevated lipoprotein (a) levels may also be significantly lowered by PCSK9i. The journey from discovery to PSCK9 target validation took less than five years, and development and approval of therapeutic modalities for PCSK9 inhibitors happened over the next seven. This review highlights the drug development and pharmacogenomics leading to the recent approval of two agents, alirocumab and evolocumab, with a third bococizumab, and other novel approaches to the pathway pending. DATA SYNTHESIS We searched MEDLINE database via Pubmed for reviews, research publications and relevant trials available on PCSK9 inhibition. CONCLUSION Despite decades of medical advances, ASCVD remains one of the major causes of morbidity and mortality worldwide. Statin use has multiplied since the validation of LDL hypothesis, however, it is undeniable a more effective and well-tolerated agent is needed in significant number or patients. With the arrival of the era of unprecedented CV protection with PCSK9 inhibition, this exciting new therapy holds a pivotal promise as the future of lipid management. The data available already indicate safety, tolerability and superb efficacy of these agents, which are already changing contemporary cholesterol management. The rapid translation of innovative basic science research into drug development may lead to CV outcomes reduction and confirm that this pathway will become prominently utilized.
Collapse
Affiliation(s)
- K Yadav
- Tulane Heart and Vascular Institute, Tulane University School of Medicine, 1430 Tulane Avenue, #8548, New Orleans, LA, 70112, USA
| | - M Sharma
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - K C Ferdinand
- Tulane Heart and Vascular Institute, Tulane University School of Medicine, 1430 Tulane Avenue, #8548, New Orleans, LA, 70112, USA.
| |
Collapse
|
245
|
Abstract
Decreasing low-density lipoprotein cholesterol (LDL-C) is one of the few established and proven principles for the prevention and treatment of atherosclerosis. The higher the individual cardiovascular risk, the higher the benefit of lipid-lowering pharmacotherapy. Therefore, treatment options are chosen based on a patient's total cardiovascular risk. The latter depends not only on the levels of LDL-C but also on the presence of cardiovascular disease (CVD) and on the number and severity of other risk factors. Current guidelines recommend the lowering of LDL-C to 115 mg/dl (3 mmol/l) in patients with low and moderate risk. The LDL-C treatment target is <100 mg/dl (2.6 mmol/l) for patients at high risk and <70 mg/dl (1.8 mmol/l) for patients at very high risk. Although lifestyle measures remain a fundamental part of treatment, many patients require drug therapy to achieve their LDL-C targets. Statins are the drugs of choice, with other options including ezetimibe and the newly available monoclonal antibodies against PCSK9 (proprotein convertase subtilisin/kexin type 9). In some cases, bile acid-binding sequestrants and fibrates can also be considered. Nicotinic acid is no longer available in Germany. PCSK9 antibodies decrease LDL-C about 50-60 % and are well tolerated. Their effects on clinical endpoints are being investigated in large randomized trials. The aim of the present review is to summarize the current guidelines and treatment options for hypercholesterolemia. Moreover, we provide an appraisal of PCSK9 antibodies and propose their use in selected patient populations, particularly in those at very high cardiovascular risk whose LDL-C levels under maximally tolerated lipid-lowering therapy are significantly over their treatment target.
Collapse
|
246
|
Abstract
Objective: To evaluate the safety and efficacy of 2 human monoclonal antibodies, alirocumab and evolocumab, on reduction of low-density lipoprotein cholesterol (LDL-C), cardiovascular benefits, and their place in current practice. Data Sources: A search of MEDLINE and Scopus databases (1966 to May 2016) with search terms “alirocumab,” “evolocumab,” “LDL,” and “PCSK9.” Study Selection and Data Extraction: The search identified phase 3 randomized control trials in English language in the past 10 years that studied LDL-C reduction of alirocumab or evolocumab. The studies were assessed for all efficacy and safety endpoints. Data Synthesis: Twelve total studies were identified evaluating alirocumab or evolocumab. These monoclonal antibodies have been shown to significantly decrease LDL-C as monotherapy and in combination with statins in phase 3 clinical trials in patients with primary hypercholesterolemia as well as familial hypercholesterolemia by inhibiting PCSK9. Alirocumab significantly reduced LDL-C by up to 61%, while evolocumab significantly reduced LDL-C by up to 66%. Adverse effects of these medications have been low and overall well tolerated. Conclusion: Although these monoclonal antibodies have shown to significantly reduce LDL-C, their effect on cardiovascular outcomes has not yet been determined. Further studies are being conducted to assess the cardiovascular benefit of both alirocumab and evolocumab. Until these studies demonstrate a reduction in atherosclerotic cardiovascular disease risk, statins should remain first-line therapy for most patients. However, alirocumab and evolocumab can be used as an effective adjunctive therapy option to lower LDL-C or in patients who are statin intolerant.
Collapse
|
247
|
Banach M, Stulc T, Dent R, Toth PP. Statin non-adherence and residual cardiovascular risk: There is need for substantial improvement. Int J Cardiol 2016; 225:184-196. [PMID: 27728862 DOI: 10.1016/j.ijcard.2016.09.075] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022]
Abstract
Although statin therapy has proven to be the cornerstone for prevention and treatment of cardiovascular disease (CVD), there are many patients for whom long-term therapy remains suboptimal. The aims of this article are to review the current complex issues associated with statin use and to explore when novel treatment approaches should be considered. Statin discontinuation as well as adherence to statin therapy remain two of the greatest challenges for lipidologists. Evidence suggests that between 40 and 75% of patients discontinue their statin therapy within one year after initiation. Furthermore, whilst the reasons for persistence with statin therapy are complex, evidence shows that low-adherence to statins negatively impacts clinical outcomes and residual CV risk remains a major concern. Non-adherence or lack of persistence with long-term statin therapy in real-life may be the main cause of inadequate low density lipoprotein cholesterol lowering with statins. There is a large need for the improvement of the use of statins, which have good safety profiles and are inexpensive. On the other hand, in a non-cost-constrained environment, proprotein convertase subtilisin/kexin type 9 inhibitors should arguably be used more often in those patients in whom treatment with statins remains unsatisfactory.
Collapse
Affiliation(s)
- Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland; Healthy Aging Research Centre, Medical University of Lodz, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute, Lodz, Poland.
| | - Tomas Stulc
- 3rd Department of Internal Medicine, 1st University of Medicine, Prague, Czech Republic
| | | | - Peter P Toth
- CGH Medical Center, Sterling, IL, USA; Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
248
|
Abstract
Although many clinical trials and meta-analyses have demonstrated that lower serum low-density lipoprotein cholesterol (LDL-C) levels are associated with proportionately greater reductions in the risk of cardiovascular disease events, not all patients with hypercholesterolemia are able to attain risk-stratified LDL-C goals with statin monotherapy. Elucidation of the pathophysiology of genetic disorders of lipid metabolism (e.g., familial hypercholesterolemia) has led to the development of several novel lipid-lowering strategies, including blocking the degradation of hepatic LDL-C receptors that are important in LDL-C clearance, or the inhibition of apoprotein synthesis and lipidation. Mipomersen and lomitapide are highly efficacious new agents available for the treatment of patients with homozygous familial hypercholesterolemia. The recent introduction of PCSK9 inhibitors (alirocumab and evolocumab) have made it possible for many patients to achieve very low LDL-C concentrations (e.g., <40 mg/dl) that are usually not attainable with statin monotherapy. Ongoing clinical trials are examining the impact of very low LDL-C levels on cardiovascular disease event rates and the long-term safety of this approach.
Collapse
Affiliation(s)
- Peter P Toth
- Community General Hospital Medical Center, Sterling, Illinois; University of Illinois School of Medicine, Peoria, Illinois; Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
249
|
Stroes E, Guyton JR, Lepor N, Civeira F, Gaudet D, Watts GF, Baccara-Dinet MT, Lecorps G, Manvelian G, Farnier M. Efficacy and Safety of Alirocumab 150 mg Every 4 Weeks in Patients With Hypercholesterolemia Not on Statin Therapy: The ODYSSEY CHOICE II Study. J Am Heart Assoc 2016; 5:JAHA.116.003421. [PMID: 27625344 PMCID: PMC5079013 DOI: 10.1161/jaha.116.003421] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background The PCSK9 antibody alirocumab (75 mg every 2 weeks; Q2W) as monotherapy reduced low‐density lipoprotein‐cholesterol (LDL‐C) levels by 47%. Because the option of a monthly dosing regimen is convenient, ODYSSEY CHOICE II evaluated alirocumab 150 mg Q4W in patients with inadequately controlled hypercholesterolemia and not on statin (majority with statin‐associated muscle symptoms), receiving treatment with fenofibrate, ezetimibe, or diet alone. Methods and Results Patients were randomly assigned to placebo, alirocumab 150 mg Q4W or 75 mg Q2W (calibrator arm), with dose adjustment to 150 mg Q2W at week (W) 12 if W8 predefined LDL‐C target levels were not met. The primary efficacy endpoint was LDL‐C percentage change from baseline to W24. Mean baseline LDL‐C levels were 163.9 mg/dL (alirocumab 150 mg Q4W, n=59), 154.5 mg/dL (alirocumab 75 mg Q2W, n=116), and 158.5 mg/dL (placebo, n=58). In the alirocumab 150 mg Q4W and 75 mg Q2W groups (49.1% and 36.0% of patients received dose adjustment, respectively), least‐squares mean LDL‐C changes from baseline to W24 were −51.7% and −53.5%, respectively (placebo [+4.7%]; both groups P<0.0001 versus placebo). In total, 63.9% and 70.3% of alirocumab‐treated patients achieved their LDL‐C targets at W24. Treatment‐emergent adverse events occurred in 77.6% (alirocumab 150 mg Q4W), 73.0% (alirocumab 75 mg Q2W), and 63.8% (placebo) of patients, with injection‐site reactions among the most common treatment‐emergent adverse events. Conclusions Alirocumab 150 mg Q4W can be considered in patients not on statin with inadequately controlled hypercholesterolemia as a convenient option for lowering LDL‐C. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT02023879.
Collapse
Affiliation(s)
- Erik Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Norman Lepor
- Westside Medical Associates of Los Angeles Cedars-Sinai Heart Institute, Beverly Hills, CA
| | - Fernando Civeira
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, Zaragoza, Spain
| | - Daniel Gaudet
- ECOGENE-21 Clinical and Translational Research Center and Department of Medicine, Université de Montréal, Chicoutimi, Quebec, Canada
| | - Gerald F Watts
- Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | | | | | | | | |
Collapse
|
250
|
Parizo J, Sarraju A, Knowles JW. Novel Therapies for Familial Hypercholesterolemia. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2016; 18:64. [PMID: 27620638 DOI: 10.1007/s11936-016-0486-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Both HeFH and HoFH require dietary and lifestyle modification. Pharmacotherapy of adult HeFH patients is largely driven by the American Heart Association (AHA) algorithm. A high-potency statin is started initially with a goal low-density lipoprotein cholesterol (LDL-C) reduction of >50 %. The LDL-C target is adjusted to <100 or <70 mg/dL in subjects with coronary artery disease (CAD) with ezetimibe being second line. If necessary, a third adjunctive therapy, such as a PSCK9 inhibitor (not yet approved in children) or bile acid-binding resin, can be added. Finally, LDL-C apheresis can be considered in patients with LDL-C >300 mg/dL (or >200 mg/dL with significant CAD, although now approved for LDL-C as low as 160 mg/dL with CAD). Due to the early, severe LDL-C elevation in HoFH patients, concerning natural history, rarity of the condition, and nuances of treatment, all HoFH patients should be treated at a pediatric or adult center with HoFH experience. LDL-C apheresis should be considered as early as 5 years of age. However, apheresis availability and tolerability is limited and pharmacotherapy is required. Generally, the AHA algorithm with reference to the European Atherosclerosis Society Consensus Panel recommendations is reasonable with all patients initiated on high-dose, high-potency statin, ezetimibe, and bile acid-binding resins. In most, additional LDL-C lowering is required with PCSK9 inhibitors and/or lomitapide or mipomersen. Liver transplantation can also be considered at experienced centers as a last resort.
Collapse
Affiliation(s)
- Justin Parizo
- Stanford University Medical Center, 300 Pasteur Ave, Stanford, CA, 94305, USA
| | - Ashish Sarraju
- Stanford University Medical Center, 300 Pasteur Ave, Stanford, CA, 94305, USA
| | - Joshua W Knowles
- Stanford University School of Medicine and Cardiovascular Institute, Falk CVRC, 300 Pasteur Drive, MC 5406, Stanford, CA, 94305, USA.
| |
Collapse
|