51
|
Morton J, Bao S, Vanags LZ, Tsatralis T, Ridiandries A, Siu CW, Ng KM, Tan JTM, Celermajer DS, Ng MKC, Bursill CA. Strikingly Different Atheroprotective Effects of Apolipoprotein A-I in Early- Versus Late-Stage Atherosclerosis. ACTA ACUST UNITED AC 2018; 3:187-199. [PMID: 30062204 PMCID: PMC6059906 DOI: 10.1016/j.jacbts.2017.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 01/23/2023]
Abstract
The atheroprotective effects of apoA-I are dependent on the plaque stage from which apoA-I is infused. The atheroprotective effects of apoA-I infusions are also impaired in older mice with a greater disease milieu. Ex vivo studies with mouse HDL found an impairment in HDL functionality with increasing disease/age of the mice as well as a reduced ability of apoA-I infusions to improve the atheroprotective functions of HDL. Our study provides understanding regarding the disparity between the very positive results of HDL/apoA-I raising in preclinical studies, largely performed in younger animals with early-stage disease, and the large-scale HDL-raising clinical trials in more elderly patients with established plaque that have failed to show benefit.
Preclinical studies have shown benefit of apolipoprotein A-I (apoA-I)/high-density lipoprotein (HDL) raising in atherosclerosis; however, this has not yet translated into a successful clinical therapy. Our studies demonstrate that apoA-I raising is more effective at reducing early-stage atherosclerosis than late-stage disease, indicating that the timing of HDL raising is a critical factor in its atheroprotective effects. To date, HDL-raising clinical trials have only been performed in aged patients with advanced atherosclerotic disease. Our findings therefore provide insight, related to important temporal aspects of HDL raising, as to why the clinical trials have thus far been largely neutral.
Collapse
Key Words
- Bcl-xL, B-cell lymphoma-extra large
- HCAEC, human coronary artery endothelial cell
- HDL, high-density lipoprotein
- HFD, high-fat diet
- LDL, low-density lipoprotein
- LVApoAI, lentivirus overexpressing apolipoprotein A-I
- LVGFP, lentivirus overexpressing green fluorescence protein
- MCP, monocyte chemoattractant protein
- SAA, serum amyloid amylase
- SMC, smooth muscle cell
- SNP, single-nucleotide polymorphism
- TNF, tumor necrosis factor
- VCAM, vascular cell adhesion molecule
- apoA-I, apolipoprotein A-I
- apoE−/−, apolipoprotein E deficient
- atherosclerosis
- cholesterol
- high-density lipoproteins
- micro-CT, micro-computed tomography
- rHDL, reconstituted high-density lipoprotein
Collapse
Affiliation(s)
- Jamie Morton
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Shisan Bao
- Discipline of Pathology, University of Sydney, Sydney, Australia
| | - Laura Z Vanags
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Tania Tsatralis
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
| | - Anisyah Ridiandries
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Chung-Wah Siu
- Division of Cardiology, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Kwong-Man Ng
- Division of Cardiology, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Joanne T M Tan
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia
| | - David S Celermajer
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Martin K C Ng
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Christina A Bursill
- Immunobiology Group, The Heart Research Institute, Sydney, Australia.,Department of Medicine, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
52
|
Ghosh GC, Bhadra R, Ghosh RK, Banerjee K, Gupta A. RVX 208: A novel BET protein inhibitor, role as an inducer of apo A-I/HDL and beyond. Cardiovasc Ther 2018; 35. [PMID: 28423226 DOI: 10.1111/1755-5922.12265] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 12/17/2016] [Accepted: 04/13/2017] [Indexed: 12/28/2022] Open
Abstract
Low-density cholesterol (LDL) has been the prime target of currently available lipid-lowering therapies although current research is expanding the focus beyond LDL lowering and has included high-density cholesterol (HDL) also as the target. Bromo and extra-terminal (BET) proteins are implicated in the regulation of transcription of several regulatory genes and regulation of proinflammatory pathways. As atherosclerosis is an inflammatory pathway and studies showed that BET inhibition has a role in inhibiting inflammation, the concept of BET inhibition came in the field of atherosclerosis. RVX 208 is a novel, orally active, BET protein inhibitor and the only BET inhibitor currently available in the field of atherosclerosis. RVX 208 acts primarily by increasing apo A-I (apolipoprotein A-I) and HDL levels. RVX 208 has a novel action of increasing larger, more cardio-protective HDL particles. Post hoc analysis of Phase II trials also showed that RVX 208 reduced major adverse cardiovascular events (MACE) in treated patients, over and above that of apo A-I/HDL increasing action. This MACE reducing actions of RVX 208 were largely due to its novel anti-inflammatory actions. Currently, a phase III trial, BETonMACE, is recruiting patients to look for the effects of RVX 208 in patients with increased risk of atherosclerotic cardiovascular disease. So BET inhibitors act in multiple ways to inhibit and modulate atherosclerosis and would be an emerging and potential option in the management of multifactorial disease like coronary artery disease by inhibiting a single substrate. But we need long-term phase III trial data's to look for effects on real-world patients.
Collapse
Affiliation(s)
- Gopal C Ghosh
- Department of Cardiology, Christian Medical College, Vellore, India
| | - Rajarshi Bhadra
- Department of Medicine, St. Vincent Charity Medical Center, A Teaching Hospital of Case Western Reserve University, Cleveland, OH, USA
| | - Raktim K Ghosh
- Department of Cardiovascular Medicine, St. Vincent Charity Medical Center, A Teaching Hospital of Case Western Reserve University, Cleveland, OH, USA
| | | | - Anjan Gupta
- Department of Cardiovascular Medicine, St. Vincent Charity Medical Center, A Teaching Hospital of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
53
|
Wasiak S, Tsujikawa LM, Halliday C, Stotz SC, Gilham D, Jahagirdar R, Kalantar-Zadeh K, Robson R, Sweeney M, Johansson JO, Wong NC, Kulikowski E. Benefit of Apabetalone on Plasma Proteins in Renal Disease. Kidney Int Rep 2018; 3:711-721. [PMID: 29854980 PMCID: PMC5976837 DOI: 10.1016/j.ekir.2017.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Apabetalone, a small molecule inhibitor, targets epigenetic readers termed BET proteins that contribute to gene dysregulation in human disorders. Apabetalone has in vitro and in vivo anti-inflammatory and antiatherosclerotic properties. In phase 2 clinical trials, this drug reduced the incidence of major adverse cardiac events in patients with cardiovascular disease. Chronic kidney disease is associated with a progressive loss of renal function and a high risk of cardiovascular disease. We studied the impact of apabetalone on the plasma proteome in patients with impaired kidney function. METHODS Subjects with stage 4 or 5 chronic kidney disease and matched controls received a single dose of apabetalone. Plasma was collected for pharmacokinetic analysis and for proteomics profiling using the SOMAscan 1.3k platform. Proteomics data were analyzed with Ingenuity Pathway Analysis to identify dysregulated pathways in diseased patients, which were targeted by apabetalone. RESULTS At baseline, 169 plasma proteins (adjusted P value <0.05) were differentially enriched in renally impaired patients versus control subjects, including cystatin C and β2 microglobulin, which correlate with renal function. Bioinformatics analysis of the plasma proteome revealed a significant activation of 42 pathways that control immunity and inflammation, oxidative stress, endothelial dysfunction, vascular calcification, and coagulation. At 12 hours postdose, apabetalone countered the activation of pathways associated with renal disease and reduced the abundance of disease markers, including interleukin-6, plasminogen activator inhibitor-1, and osteopontin. CONCLUSION These data demonstrated plasma proteome dysregulation in renally impaired patients and the beneficial impact of apabetalone on pathways linked to chronic kidney disease and its cardiovascular complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Richard Robson
- Christchurch Clinical Studies Trust, Christchurch, New Zealand
| | | | | | | | | |
Collapse
|
54
|
Nicholls SJ, Ray KK, Johansson JO, Gordon A, Sweeney M, Halliday C, Kulikowski E, Wong N, Kim SW, Schwartz GG. Selective BET Protein Inhibition with Apabetalone and Cardiovascular Events: A Pooled Analysis of Trials in Patients with Coronary Artery Disease. Am J Cardiovasc Drugs 2018; 18:109-115. [PMID: 29027131 DOI: 10.1007/s40256-017-0250-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Inhibition of bromodomain and extra-terminal (BET) proteins can modulate lipoprotein and inflammatory factors that mediate atherosclerosis. The impact of the BET inhibitor, apabetalone, on cardiovascular events is unknown. OBJECTIVE Our objective was to investigate the impact of apabetalone on cardiovascular event rates in a pooled analysis of clinical studies in patients with established coronary artery disease. METHODS We conducted a pooled analysis of patients (n = 798) with coronary artery disease who participated in clinical trials (ASSERT, ASSURE, SUSTAIN) that evaluated the impact of 3-6 months of treatment with apabetalone on lipid parameters and coronary atherosclerosis. The incidence of major adverse cardiovascular events (death, myocardial infarction, coronary revascularization, hospitalization for cardiovascular causes) in the treatment groups was evaluated. RESULTS At baseline, patients treated with apabetalone were more likely to be Caucasian, have a history of dyslipidemia, and be undertreated with ß-blocker and anti-platelet agents. Treatment with apabetalone produced the following dose-dependent changes compared with placebo: increases in apolipoprotein A-I (apoA-I) of up to 6.7% (P < 0.001), increases in high-density lipoprotein cholesterol (HDL-C) of up to 6.5% (P < 0.001), increases in large HDL particles of up to 23.3% (P < 0.001), and decreases in high-sensitivity C-reactive protein (hsCRP) of - 21.1% (P = 0.04). Apabetalone treatment did not affect atherogenic lipoproteins compared with placebo. Patients treated with apabetalone experienced fewer major adverse cardiovascular events than those treated with placebo (5.9 vs. 10.4%; P = 0.02), a finding that was more prominent in patients with diabetes (5.4 vs. 12.7%; P = 0.02), with baseline HDL-C < 39 mg/dl (5.5 vs. 12.8%; P = 0.01), or with elevated hsCRP levels (5.4 vs. 14.2%; P = 0.02). CONCLUSION Pooled analysis of short-term studies demonstrated fewer cardiovascular events among patients treated with the BET protein inhibitor, apabetalone, than among those treated with placebo. BET protein inhibition warrants further investigation as a novel approach to cardiovascular risk reduction.
Collapse
Affiliation(s)
- Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA, 5001, Australia.
| | - Kausik K Ray
- School of Public Health, Imperial College London, London, UK
| | | | | | | | | | | | | | - Susan W Kim
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA, 5001, Australia
| | | |
Collapse
|
55
|
Runcie AC, Zengerle M, Chan KH, Testa A, van Beurden L, Baud MGJ, Epemolu O, Ellis LCJ, Read KD, Coulthard V, Brien A, Ciulli A. Optimization of a "bump-and-hole" approach to allele-selective BET bromodomain inhibition. Chem Sci 2018; 9:2452-2468. [PMID: 29732121 PMCID: PMC5909127 DOI: 10.1039/c7sc02536j] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 01/23/2018] [Indexed: 12/27/2022] Open
Abstract
Allele-specific chemical genetics enables selective inhibition within families of highly-conserved proteins.
Allele-specific chemical genetics enables selective inhibition within families of highly-conserved proteins. The four BET (bromodomain & extra-terminal domain) proteins – BRD2, BRD3, BRD4 and BRDT bind acetylated chromatin via their bromodomains and regulate processes such as cell proliferation and inflammation. BET bromodomains are of particular interest, as they are attractive therapeutic targets but existing inhibitors are pan-selective. We previously established a bump-&-hole system for the BET bromodomains, pairing a leucine/alanine mutation with an ethyl-derived analogue of an established benzodiazepine scaffold. Here we optimize upon this system with the introduction of a more conservative and less disruptive leucine/valine mutation. Extensive structure–activity-relationships of diverse benzodiazepine analogues guided the development of potent, mutant-selective inhibitors with desirable physiochemical properties. The active enantiomer of our best compound – 9-ME-1 – shows ∼200 nM potency, >100-fold selectivity for the L/V mutant over wild-type and excellent DMPK properties. Through a variety of in vitro and cellular assays we validate the capabilities of our optimized system, and then utilize it to compare the relative importance of the first and second bromodomains to chromatin binding. These experiments confirm the primacy of the first bromodomain in all BET proteins, but also significant variation in the importance of the second bromodomain. We also show that, despite having a minor role in chromatin recognition, BRD4 BD2 is still essential for gene expression, likely through the recruitment of non-histone proteins. The disclosed inhibitor:mutant pair provides a powerful tool for future cellular and in vivo target validation studies.
Collapse
Affiliation(s)
- A C Runcie
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - M Zengerle
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - K-H Chan
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - A Testa
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - L van Beurden
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - M G J Baud
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - O Epemolu
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - L C J Ellis
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - K D Read
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| | - V Coulthard
- Reach Separations Ltd , BioCity Nottingham , Nottingham , UK
| | - A Brien
- Reach Separations Ltd , BioCity Nottingham , Nottingham , UK
| | - A Ciulli
- Division of Biological Chemistry and Drug Discovery , School of Life Sciences , University of Dundee , Dundee , Scotland , UK .
| |
Collapse
|
56
|
Lu P, Shen Y, Yang H, Wang Y, Jiang Z, Yang X, Zhong Y, Pan H, Xu J, Lu H, Zhu H. BET inhibitors RVX-208 and PFI-1 reactivate HIV-1 from latency. Sci Rep 2017; 7:16646. [PMID: 29192216 PMCID: PMC5709369 DOI: 10.1038/s41598-017-16816-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/16/2017] [Indexed: 02/02/2023] Open
Abstract
Persistent latent reservoir in resting CD4+ T cells is a major obstacle in curing HIV-1 infection. Effective strategies for eradication of the HIV-1 reservoir are urgently needed. We report here for the first time that two BET inhibitors, RVX-208, which has entered phase II clinical trials for diverse cardiovascular disorders, and PFI-1, which has been widely studied in oncology, can reactivate HIV-1 from latency. RVX-208 and PFI-1 treatment alone or in combination with other latency reversing agents efficiently reactivated HIV-1 transcription through an up-regulation of P-TEFb by increasing CDK9 Thr-186 phosphorylation in latently infected Jurkat T cells in vitro. The two BET inhibitors also reactivated HIV-1 transcription in cART treated patient-derived resting CD4+ T cells ex vivo, without influence on global immune cell activation. Our findings, in combination with previous reports, further confirm that BET inhibitors are a group of leading compounds for combating HIV-1 latency for viral eradication.
Collapse
Affiliation(s)
- Panpan Lu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yinzhong Shen
- Department of Infectious Diseases, and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200433, China
| | - He Yang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yanan Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zhengtao Jiang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xinyi Yang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yangcheng Zhong
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Hanyu Pan
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jianqing Xu
- Department of Infectious Diseases, and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200433, China
| | - Hongzhou Lu
- Department of Infectious Diseases, and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200433, China
| | - Huanzhang Zhu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
57
|
Weber C, Badimon L, Mach F, van der Vorst EPC. Therapeutic strategies for atherosclerosis and atherothrombosis: Past, present and future. Thromb Haemost 2017; 117:1258-1264. [DOI: 10.1160/th16-10-0814] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/10/2016] [Indexed: 12/14/2022]
Abstract
SummaryEven two centuries after they were first described, atherosclerosis and atherothrombosis remain among the leading causes of death worldwide. Over the last decades it has become clear that atherosclerosis is not only a lipid-driven disease but also a multifactorial process largely driven by inflammatory mediators, an insight that has instigated additional research and drug development focussing on anti-inflammatory therapies. In this review, we will provide a brief historical overview, followed by a more general synopsis of the range of currently available state-of-the-art therapies for atherosclerosis and atherothrombosis. Finally, we will highlight some of the promising therapeutic strategies that are currently under intense investigation. We believe that the next years will witness highly interesting developments and clinical trials investigating yet more novel therapies, and at the same time looking into potential combinations of all available therapies. This prospect closes in on the ultimate goal, which is to reduce the residual risk that still persists despite present therapeutic options.
Collapse
|
58
|
Xu Y, Vakoc CR. Targeting Cancer Cells with BET Bromodomain Inhibitors. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026674. [PMID: 28213432 DOI: 10.1101/cshperspect.a026674] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer cells are often hypersensitive to the targeting of transcriptional regulators, which may reflect the deregulated gene expression programs that underlie malignant transformation. One of the most prominent transcriptional vulnerabilities in human cancer to emerge in recent years is the bromodomain and extraterminal (BET) family of proteins, which are coactivators that link acetylated transcription factors and histones to the activation of RNA polymerase II. Despite unclear mechanisms underlying the gene specificity of BET protein function, small molecules targeting these regulators preferentially suppress the transcription of cancer-promoting genes. As a consequence, BET inhibitors elicit anticancer activity in numerous malignant contexts at doses that can be tolerated by normal tissues, a finding supported by animal studies and by phase I clinical trials in human cancer patients. In this review, we will discuss the remarkable, and often perplexing, therapeutic effects of BET bromodomain inhibition in cancer.
Collapse
Affiliation(s)
- Yali Xu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | | |
Collapse
|
59
|
Smolders L, Plat J, Mensink RP. Dietary Strategies and Novel Pharmaceutical Approaches Targeting Serum ApoA-I Metabolism: A Systematic Overview. J Nutr Metab 2017; 2017:5415921. [PMID: 28695008 PMCID: PMC5485365 DOI: 10.1155/2017/5415921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/16/2017] [Indexed: 12/19/2022] Open
Abstract
The incidence of CHD is still increasing, which underscores the need for new preventive and therapeutic approaches to decrease CHD risk. In this respect, increasing apoA-I concentrations may be a promising approach, especially through increasing apoA-I synthesis. This review first provides insight into current knowledge on apoA-I production, clearance, and degradation, followed by a systematic review of dietary and novel pharmacological approaches to target apoA-I metabolism. For this, a systematic search was performed to identify randomized controlled intervention studies that examined effects of whole foods and (non)nutrients on apoA-I metabolism. In addition, novel pharmacological approaches were searched for, which were specifically developed to target apoA-I metabolism. We conclude that both dietary components and pharmacological approaches can be used to increase apoA-I concentrations or functionality. For the dietary components in particular, more knowledge about the underlying mechanisms is necessary, as increasing apoA-I per se does not necessarily translate into a reduced CHD risk.
Collapse
Affiliation(s)
- Lotte Smolders
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| | - Jogchum Plat
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| | - Ronald P. Mensink
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| |
Collapse
|
60
|
Downregulation of the Complement Cascade In Vitro, in Mice and in Patients with Cardiovascular Disease by the BET Protein Inhibitor Apabetalone (RVX-208). J Cardiovasc Transl Res 2017; 10:337-347. [PMID: 28567671 PMCID: PMC5585290 DOI: 10.1007/s12265-017-9755-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/17/2017] [Indexed: 12/11/2022]
Abstract
Apabetalone (RVX-208) is an epigenetic regulator developed to treat cardiovascular disease (CVD) that targets BET proteins. Through transcriptional regulation RVX-208 modulates pathways that underlie CVD including reverse cholesterol transport, vascular inflammation, coagulation, and complement. Using transcriptomics and proteomics we show that complement is one of the top pathways downregulated by RVX-208 in primary human hepatocytes (PHH) and in plasma from CVD patients. RVX-208 reduces basal and cytokine-driven expression of complement factors in PHH and in chimeric mice with humanized livers. Plasma proteomics of CVD patients shows that RVX-208 decreases complement proteins and regulators, including complement activators SAP and CRP. Circulating activated fragments C5a, C3b, and C5b-C6 are reduced by 51, 32, and 10%, respectively, indicating decreased activity of complement in patients. As complement components are linked to CVD and metabolic syndrome, including major acute cardiac events, modulating their levels and activity by RVX-208 may alleviate risks associated with these diseases.
Collapse
|
61
|
Rahman MS, Murphy AJ, Woollard KJ. Effects of dyslipidaemia on monocyte production and function in cardiovascular disease. Nat Rev Cardiol 2017; 14:387-400. [PMID: 28300081 DOI: 10.1038/nrcardio.2017.34] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Monocytes are heterogeneous effector cells involved in the maintenance and restoration of tissue integrity. Monocytes and macrophages are involved in cardiovascular disease progression, and are associated with the development of unstable atherosclerotic plaques. Hyperlipidaemia can accelerate cardiovascular disease progression. However, monocyte responses to hyperlipidaemia are poorly understood. In the past decade, accumulating data describe the relationship between the dynamic blood lipid environment and the heterogeneous circulating monocyte pool, which might have profound consequences for cardiovascular disease. In this Review, we explore the updated view of monocytes in cardiovascular disease and their relationship with macrophages in promoting the homeostatic and inflammatory responses related to atherosclerosis. We describe the different definitions of dyslipidaemia, highlight current theories on the ontogeny of monocyte heterogeneity, discuss how dyslipidaemia might alter monocyte production, and explore the mechanistic interface linking dyslipidaemia with monocyte effector functions, such as migration and the inflammatory response. Finally, we discuss the role of dietary and endogenous lipid species in mediating dyslipidaemic responses, and the role of these lipids in promoting the risk of cardiovascular disease through modulation of monocyte behaviour.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology Lab, Baker IDI Heart &Diabetes Research Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Department of Immunology, Monash University, 89 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Kevin J Woollard
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
62
|
Choi HY, Hafiane A, Schwertani A, Genest J. High-Density Lipoproteins: Biology, Epidemiology, and Clinical Management. Can J Cardiol 2017; 33:325-333. [DOI: 10.1016/j.cjca.2016.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 01/29/2023] Open
|
63
|
Yeh TC, O'Connor G, Petteruti P, Dulak A, Hattersley M, Barrett JC, Chen H. Identification of CCR2 and CD180 as Robust Pharmacodynamic Tumor and Blood Biomarkers for Clinical Use with BRD4/BET Inhibitors. Clin Cancer Res 2017; 23:1025-1035. [DOI: 10.1158/1078-0432.ccr-16-1658] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/22/2016] [Indexed: 11/16/2022]
|
64
|
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
|
65
|
Di Bartolo BA, Scherer DJ, Nicholls SJ. Inducing apolipoprotein A-I synthesis to reduce cardiovascular risk: from ASSERT to SUSTAIN and beyond. Arch Med Sci 2016; 12:1302-1307. [PMID: 27904522 PMCID: PMC5108390 DOI: 10.5114/aoms.2016.62906] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/17/2015] [Indexed: 12/29/2022] Open
Abstract
Increasing attention has focused on efforts to promote the biological activities of high-density lipoproteins (HDL) in order to reduce cardiovascular risk. Targeting apolipoprotein A-I (apoA-I), the major protein carried on HDL particles, represents an attractive approach to promoting HDL by virtue of its ability to increase endogenous synthesis of functional HDL particles. A number of pharmacological strategies that target apoA-I, including upregulation of its production with the bromodomain and extraterminal (BET) protein inhibitor RVX-208, development of short peptide sequences that mimic its action, and administration as a component of reconstituted HDL particles, have undergone clinical development. The impact of these approaches on cardiovascular biomarkers will be reviewed.
Collapse
Affiliation(s)
- Belinda A Di Bartolo
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Daniel J Scherer
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
66
|
Andrews J, Puri R, Kataoka Y, Nicholls SJ, Psaltis PJ. Therapeutic modulation of the natural history of coronary atherosclerosis: lessons learned from serial imaging studies. Cardiovasc Diagn Ther 2016; 6:282-303. [PMID: 27500089 DOI: 10.21037/cdt.2015.10.02] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite advances in risk prediction, preventive and therapeutic strategies, atherosclerotic cardiovascular disease remains a major public health challenge worldwide, carrying considerable morbidity, mortality and health economic burden. There continues to be a need to better understand the natural history of this disease to guide the development of more effective treatment, integral to which is the rapidly evolving field of coronary artery imaging. Various imaging modalities have been refined to enable detailed visualization of the pathological substrate of atherosclerosis, providing accurate and reproducible measures of coronary plaque burden and composition, including the presence of high-risk characteristics. The serial application of such techniques, including coronary computed tomography angiography (CTA), intravascular ultrasound (IVUS) and optical coherence tomography (OCT) have uncovered important insights into the progression of coronary plaque over time in patients with stable and unstable coronary artery disease (CAD), and its responsiveness to therapeutic interventions. Here we review the use of different imaging modalities for the surveillance of coronary atherosclerosis and the lessons they have provided about the modulation of CAD by both traditional and experimental therapies.
Collapse
Affiliation(s)
- Jordan Andrews
- Vascular Research Centre, Heart Health Theme, South Australian Health and Medical Research Institute & School of Medicine, University of Adelaide, Adelaide, Australia
| | - Rishi Puri
- Québec Heart & Lung Institute (IUCPQ), Hospital Laval, Québec (Québec), Canada; ; Cleveland Clinic Coordinating Center for Clinical Research (C5R), Cleveland, Ohio, USA
| | - Yu Kataoka
- National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Stephen J Nicholls
- Vascular Research Centre, Heart Health Theme, South Australian Health and Medical Research Institute & School of Medicine, University of Adelaide, Adelaide, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart Health Theme, South Australian Health and Medical Research Institute & School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
67
|
Abstract
PURPOSE OF REVIEW Low HDL-cholesterol (HDL-C) levels are predictive of incident atherosclerotic cardiovascular disease events. However, the use of medication to raise HDL-C levels has not consistently shown clinical benefit. As a result, studies have shifted toward HDL function, specifically cholesterol efflux, which has been inversely associated with prevalent subclinical atherosclerosis as well as subsequent atherosclerotic cardiovascular disease events. The purpose of this review is to summarize the effects of current medications and interventions on cholesterol efflux capacity. RECENT FINDINGS Medications for cardiovascular health, including statins, fibrates, niacin, and novel therapeutics, are reviewed for their effect on cholesterol efflux. Differences in population studied and assay used are addressed appropriately. Lifestyle interventions, including diet and exercise, are also included in the review. SUMMARY The modification of cholesterol efflux capacity (CEC) by current medications and interventions has been investigated in both large randomized control trials and smaller observational cohorts. This review serves to compile the results of these studies and evaluate CEC modulation by commonly used medications. Altering CEC could be a novel therapeutic approach to improving cardiovascular risk profiles.
Collapse
Affiliation(s)
- Nicholas Brownell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
68
|
Okopień B, Bułdak Ł, Bołdys A. Current and future trends in the lipid lowering therapy. Pharmacol Rep 2016; 68:737-47. [DOI: 10.1016/j.pharep.2016.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/25/2016] [Accepted: 03/25/2016] [Indexed: 12/31/2022]
|
69
|
Bhatt A, Rohatgi A. HDL Cholesterol Efflux Capacity: Cardiovascular Risk Factor and Potential Therapeutic Target. Curr Atheroscler Rep 2016; 18:2. [PMID: 26710794 DOI: 10.1007/s11883-015-0554-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Low high-density lipoprotein cholesterol (HDL-C) levels are associated with incident cardiovascular events; however, many therapies targeting increases in HDL-C have failed to show consistent clinical benefit. Thus, focus has recently shifted toward measuring high-density lipoprotein (HDL) function. HDL is the key mediator of reverse cholesterol transport, the process of cholesterol extraction from foam cells, and eventual excretion into the biliary system. Cholesterol efflux from peripheral macrophages to HDL particles has been associated with atherosclerosis in both animals and humans. We review the mechanism of cholesterol efflux and the emerging evidence on the association between cholesterol efflux capacity and cardiovascular disease in human studies. We also focus on the completed and ongoing trials of novel therapies targeting different aspects of HDL cholesterol efflux.
Collapse
Affiliation(s)
- Anish Bhatt
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8830, USA.
| | - Anand Rohatgi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8830, USA.
| |
Collapse
|
70
|
Siebel AL, Trinh SK, Formosa MF, Mundra PA, Natoli AK, Reddy-Luthmoodoo M, Huynh K, Khan AA, Carey AL, van Hall G, Cobelli C, Dalla-Man C, Otvos JD, Rye KA, Johansson J, Gordon A, Wong NCW, Sviridov D, Barter P, Duffy SJ, Meikle PJ, Kingwell BA. Effects of the BET-inhibitor, RVX-208 on the HDL lipidome and glucose metabolism in individuals with prediabetes: A randomized controlled trial. Metabolism 2016; 65:904-14. [PMID: 27173469 DOI: 10.1016/j.metabol.2016.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/18/2016] [Accepted: 03/03/2016] [Indexed: 12/14/2022]
Abstract
AIMS High-density lipoprotein (HDL) and apolipoprotein A-I (apoA-I) can modulate glucose metabolism through multiple mechanisms. This study determined the effects of a novel bromodomain and extra-terminal (BET) inhibitor (RVX-208) and putative apoA-I inducer on lipid species contained within HDL (HDL lipidome) and glucose metabolism. MATERIALS AND METHODS Twenty unmedicated males with prediabetes received 100mg b.i.d. RVX-208 and placebo for 29-33days separated by a wash-out period in a randomized, cross-over design trial. Plasma HDL-cholesterol and apoA-I were assessed as well as lipoprotein particle size and distribution using NMR spectroscopy. An oral glucose tolerance test (OGTT) protocol with oral and infused stable isotope tracers was employed to assess postprandial plasma glucose, indices of insulin secretion and insulin sensitivity, glucose kinetics and lipolysis. Whole plasma and HDL lipid profiles were measured using mass spectrometry. RESULTS RVX-208 treatment for 4weeks increased 6 sphingolipid and 4 phospholipid classes in the HDL lipidome (p≤0.05 versus placebo), but did not change conventional clinical lipid measures. The concentration of medium-sized HDL particles increased by 11% (P=0.01) and small-sized HDL particles decreased by 10% (P=0.04) after RVX-208 treatment. In response to a glucose load, after RVX-208 treatment, plasma glucose peaked at a similar level to placebo, but 30min later with a more sustained elevation (treatment effect, P=0.003). There was a reduction and delay in total (P=0.001) and oral (P=0.003) glucose rates of appearance in plasma and suppression of endogenous glucose production (P=0.014) after RVX-208 treatment. The rate of glucose disappearance was also lower following RVX-208 (P=0.016), with no effect on glucose oxidation or total glucose disposal. CONCLUSIONS RVX-208 increased 10 lipid classes in the plasma HDL fraction, without altering the concentrations of either apoA-I or HDL-cholesterol (HDL-C). RVX-208 delayed and reduced oral glucose absorption and endogenous glucose production, with plasma glucose maintained via reduced peripheral glucose disposal. If sustained, these effects may protect against the development of type 2 diabetes.
Collapse
Affiliation(s)
- Andrew L Siebel
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Si Khiang Trinh
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | | | - Alaina K Natoli
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | - Kevin Huynh
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Anmar A Khan
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew L Carey
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Gerrit van Hall
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Chiara Dalla-Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Kerry-Anne Rye
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | | | | | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Philip Barter
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW A low level of plasma high density lipoprotein cholesterol (HDL-C) is a strong and independent risk factor for atherosclerotic cardiovascular disease (ASCVD). However, several large studies recently revealed that pharmacologic interventions that increase HDL-C concentration have not improved cardiovascular outcomes when added to standard therapy. In addition, specific genetic variants that raise HDL-C levels are not clearly associated with reduced risk of coronary heart disease. These observations have challenged the 'HDL hypothesis' that HDL-C is causally related to ASCVD and that intervention to raise HDL-C will reduce ASCVD events. This article will present the current data on the HDL hypothesis and provide a revised paradigm of considering HDL in the atherosclerotic pathway. RECENT FINDINGS Recent evidence has shed light on the complex nature of HDL-C metabolism and function. There are compelling data that the ability of HDL to promote cholesterol efflux from macrophages, the first step in the 'reverse cholesterol transport' (RCT) pathway, is inversely associated with risk for ASCVD even after controlling for HDL-C. This has led to the 'HDL flux hypothesis' that therapeutic intervention that targets macrophage cholesterol efflux and RCT may reduce risk. Preclinical studies of such interventions show promise and early phase clinical studies, though small, are encouraging. SUMMARY The role of HDL-C in modulating atherosclerotic disease is as yet uncertain. However, new findings and therapies targeting HDL-C show early promise and may provide an important intervention in attenuating the burden of ASCVD in the future.
Collapse
|
72
|
Nikolic D, Rizzo M, Mikhailidis DP, Wong NC, Banach M. An evaluation of RVX-208 for the treatment of atherosclerosis. Expert Opin Investig Drugs 2016; 24:1389-98. [PMID: 26364508 DOI: 10.1517/13543784.2015.1083010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION RVX-208 is a first-in-class, orally active, novel small molecule in development by Resverlogix Corporation (Calgary, AB, Canada). It acts through an epigenetic mechanism by inhibiting the bromodomain and extraterminal (BET) family of proteins, increasing apolipoprotein A-I (apoA-I) and targeting high-density lipoprotein (HDL) metabolism, including generating of nascent HDL and increased larger HDL particles, resulting in the stimulation of reverse cholesterol transport. RVX-208 also has a beneficial effect on inflammatory factors known to be involved in atherosclerosis and plaque stability. New therapeutic strategies are needed for patients with atherosclerosis. AREAS COVERED In this review, the authors evaluate the use of RVX-208 as an agent for the treatment of atherosclerosis. The article is based on a literature search considering both animal and human studies available on PubMed as well as Media Releases from the Resverlogix Corporation. EXPERT OPINION The current evidence suggests promising beneficial effects of this novel drug in the prevention and treatment of atherosclerosis and other metabolic disorders. Its unique mechanism of action is encouraging; it affects several pathways and has a modest effect on HDL levels. There is also a shift in particle size to larger HDL particles, which may have potent atheroprotective effects. Future clinical development is needed, including safety assessment.
Collapse
Affiliation(s)
- Dragana Nikolic
- a 1 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Palermo, Italy
| | - Manfredi Rizzo
- a 1 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Palermo, Italy.,b 2 Euro-Mediterranean Institute of Science and Technology , Italy
| | - Dimitri P Mikhailidis
- c 3 University College London (UCL), University College London Medical School, Department of Clinical Biochemistry , Royal Free Campus, London, UK
| | - Norman C Wong
- d 4 University of Calgary, Department of Medicine, Biochemistry and Molecular Biology , Calgary, Alberta, Canada
| | - Maciej Banach
- e 5 University of Lodz, Medical University of Lodz, WAM University Hospital in Lodz, Department of Hypertension , Zeromskiego 113; 90-549 Lodz, Poland +48 4 2639 3771 ; +48 4 2639 3771 ;
| |
Collapse
|
73
|
Abstract
Several recent reports have raised doubts about the atheroprotective role of high-density lipoprotein cholesterol (HDL-C). Nevertheless, a substantial body of work supports the validity of pharmacological interventions able to enhance HDL function, as opposed to raising HDL-C levels per se. In this article, we briefly review the development of pharmacological interventions that target apoA-I and HDL function as a means of reducing atherosclerotic risk: small molecule pharmaceuticals, small HDL mimetic peptides, and infusion of apoA-I-containing particles.
Collapse
|
74
|
Effect of the BET Protein Inhibitor, RVX-208, on Progression of Coronary Atherosclerosis: Results of the Phase 2b, Randomized, Double-Blind, Multicenter, ASSURE Trial. Am J Cardiovasc Drugs 2016; 16:55-65. [PMID: 26385396 DOI: 10.1007/s40256-015-0146-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Bromodomain and extra-terminal (BET) proteins regulate transcription of lipoprotein and inflammatory factors implicated in atherosclerosis. The impact of BET inhibition on atherosclerosis progression is unknown. METHODS ASSURE was a double-blind, randomized, multicenter trial in which 323 patients with angiographic coronary disease and low high-density lipoprotein cholesterol (HDL-C) levels were randomized in a 3:1 fashion to treatment with the BET protein inhibitor RVX-208 200 mg or placebo for 26 weeks. Plaque progression was measured with serial intravascular ultrasound imaging. Lipid levels, safety, and tolerability were also assessed. RESULTS During treatment, apolipoprotein (apo)A-I increased by 10.6% with placebo (P < 0.001 compared with baseline) and 12.8% with RVX-208 (P < 0.001 compared with baseline), between groups P = 0.18. HDL-C increased by 9.1% with placebo (P < 0.001 compared with baseline) and 11.1% with RVX-208 (P < 0.001 compared with baseline), between groups P = 0.24. Low-density lipoprotein cholesterol (LDL-C) decreased by 17.9% with placebo (P < 0.001 compared with baseline) and 15.8% with RVX-208 (P < 0.001 compared with baseline), between groups P = 0.55. The primary endpoint, the change in percent atheroma volume, decreased 0.30% in placebo-treated patients (P = 0.23 compared with baseline) and 0.40% in the RVX-208 group (P = 0.08 compared with baseline), between groups P = 0.81. Total atheroma volume decreased 3.8 mm(3) in the placebo group (P = 0.01 compared with baseline) and 4.2 mm(3) in the RVX-208 group (P < 0.001 compared with baseline), P = 0.86 between groups. A greater incidence of elevated liver enzymes was observed in RVX-208-treated patients (7.1 vs. 0%, P = 0.009). CONCLUSION Administration of the BET protein inhibitor RVX-208 showed no greater increase in apoA-I or HDL-C or incremental regression of atherosclerosis than administration of placebo. TRIAL REGISTRATION ClinicalTrials.gov identifier-NCT01067820.
Collapse
|
75
|
Gilham D, Wasiak S, Tsujikawa LM, Halliday C, Norek K, Patel RG, Kulikowski E, Johansson J, Sweeney M, Wong NCW, Gordon A, McLure K, Young P. RVX-208, a BET-inhibitor for treating atherosclerotic cardiovascular disease, raises ApoA-I/HDL and represses pathways that contribute to cardiovascular disease. Atherosclerosis 2016; 247:48-57. [PMID: 26868508 DOI: 10.1016/j.atherosclerosis.2016.01.036] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/18/2022]
Abstract
High density lipoproteins (HDL), through activity of the main protein component apolipoprotein A-I (ApoA-I), can reduce the risk of cardiovascular disease (CVD) by removing excess cholesterol from atherosclerotic plaque. In this study, we demonstrate that the bromodomain and extraterminal domain (BET) inhibitor RVX-208 increases ApoA-I gene transcription and protein production in human and primate primary hepatocytes. Accordingly, RVX-208 also significantly increases levels of ApoA-I, HDL-associated cholesterol, and HDL particle number in patients who received the compound in recently completed phase 2b trials SUSTAIN and ASSURE. Moreover, a post-hoc analysis showed lower instances of major adverse cardiac events in patients receiving RVX-208. To understand the effects of RVX-208 on biological processes underlying cardiovascular risk, we performed microarray analyses of human primary hepatocytes and whole blood treated ex vivo. Overall, data showed that RVX-208 raises ApoA-I/HDL and represses pro-inflammatory, pro-atherosclerotic and pro-thrombotic pathways that can contribute to CVD risk.
Collapse
|
76
|
Abstract
The armamentarium for the treatment of dyslipidemia today comprises six different modes of action with overall around 24 different drugs. The treatment of lipid disorders was revolutionized with the introduction of statins which have become the most important therapeutic option available today to reduce and prevent atherosclerosis and its detrimental consequences like cardiovascular diseases and stroke. With and optimized reduction of elevated LDL levels with statins, the risk for cardiovascular diseases (CVD) can be reduced by 30%, indicating a residual remaining risk of 70% for the development and progression of CVD notifying still a high medical need for more effective antilipidemic drugs. Consequently, the search for novel lipid-modifying drugs is still one of the most active areas in research and development in the pharmaceutical industry. Major focus lies on approaches to LDL-lowering drugs superior to statins with regard to efficacy, safety, and patient compliance and on approaches modifying plasma levels and functionality of HDL particles based on the clinically validated inverse relationship between high-plasma HDL levels and the risk for CVD. The available drugs today for the treatment of dyslipidemia are small organic molecules or nonabsorbable polymers for binding of bile acids to be applied orally. Besides small molecules for novel targets, biological drugs such as monoclonal antibodies, antisense or gene-silencing oligonucleotides, peptidomimetics, reconstituted synthetic HDL particles and therapeutic proteins are novel approaches in clinical development are which have to be applied by injection or infusion. The promising clinical results of several novel drug candidates, particularly for LDL cholesterol lowering with monoclonal antibodies raised against PCSK9, may indicate more than a decade after the statins, the entrance of new breakthrough therapies to treat lipid disorders.
Collapse
Affiliation(s)
- Werner Kramer
- Institute of Biochemistry, Biocenter, Goethe-Universität Frankfurt, Max-von-Laue-Str. 9, Frankfurt, Germany.
| |
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW Increasing interest has focused on the strategies that target the atheroprotective properties of HDL in order to reduce cardiovascular risk. The potential impact of strategies to acutely promote HDL functionality will be reviewed. RECENT FINDINGS Population and animal studies suggest that HDLs have a protective impact on atherosclerotic plaque. However, the failure of recent clinical trials of HDL cholesterol-raising agents has raised concerns that this may not be a viable strategy to reduce cardiovascular risk. Increasing attention has highlighted the importance of the functional quality, as opposed to quantity, of HDL with evidence of impaired HDL function in the setting of acute coronary syndromes (ACSs). The finding that infusing HDL in patients with recent acute ischemic events promotes the rapid regression of coronary atherosclerosis suggests a potentially useful strategy for ACS patients, although this remains to be fully established in large clinical outcome trials. SUMMARY Infusing HDL has favorable effects on coronary atherosclerosis in ACS patients, suggesting a potentially beneficial therapeutic strategy to acutely promote HDL functionality.
Collapse
Affiliation(s)
- MyNgan Duong
- aSouth Australian Health and Medical Research InstitutebDepartment of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | | |
Collapse
|
78
|
Gadkar K, Lu J, Sahasranaman S, Davis J, Mazer NA, Ramanujan S. Evaluation of HDL-modulating interventions for cardiovascular risk reduction using a systems pharmacology approach. J Lipid Res 2015; 57:46-55. [PMID: 26522778 PMCID: PMC4689335 DOI: 10.1194/jlr.m057943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Indexed: 11/20/2022] Open
Abstract
The recent failures of cholesteryl ester transport protein inhibitor drugs to decrease CVD risk, despite raising HDL cholesterol (HDL-C) levels, suggest that pharmacologic increases in HDL-C may not always reflect elevations in reverse cholesterol transport (RCT), the process by which HDL is believed to exert its beneficial effects. HDL-modulating therapies can affect HDL properties beyond total HDL-C, including particle numbers, size, and composition, and may contribute differently to RCT and CVD risk. The lack of validated easily measurable pharmacodynamic markers to link drug effects to RCT, and ultimately to CVD risk, complicates target and compound selection and evaluation. In this work, we use a systems pharmacology model to contextualize the roles of different HDL targets in cholesterol metabolism and provide quantitative links between HDL-related measurements and the associated changes in RCT rate to support target and compound evaluation in drug development. By quantifying the amount of cholesterol removed from the periphery over the short-term, our simulations show the potential for infused HDL to treat acute CVD. For the primary prevention of CVD, our analysis suggests that the induction of ApoA-I synthesis may be a more viable approach, due to the long-term increase in RCT rate.
Collapse
Affiliation(s)
- Kapil Gadkar
- Genentech Research and Early Development, South San Francisco, CA
| | - James Lu
- Roche Pharma Research and Early Development, Clinical Pharmacology, Disease Modeling Group, Roche Innovation Center Basel, Basel, Switzerland
| | | | - John Davis
- Genentech Research and Early Development, South San Francisco, CA
| | - Norman A Mazer
- Roche Pharma Research and Early Development, Clinical Pharmacology, Disease Modeling Group, Roche Innovation Center Basel, Basel, Switzerland
| | - Saroja Ramanujan
- Genentech Research and Early Development, South San Francisco, CA
| |
Collapse
|
79
|
Guo Y, Fan Y, Zhang J, Lomberk GA, Zhou Z, Sun L, Mathison AJ, Garcia-Barrio MT, Zhang J, Zeng L, Li L, Pennathur S, Willer CJ, Rader DJ, Urrutia R, Chen YE. Perhexiline activates KLF14 and reduces atherosclerosis by modulating ApoA-I production. J Clin Invest 2015; 125:3819-30. [PMID: 26368306 DOI: 10.1172/jci79048] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 08/07/2015] [Indexed: 12/20/2022] Open
Abstract
Recent genome-wide association studies have revealed that variations near the gene locus encoding the transcription factor Krüppel-like factor 14 (KLF14) are strongly associated with HDL cholesterol (HDL-C) levels, metabolic syndrome, and coronary heart disease. However, the precise mechanisms by which KLF14 regulates lipid metabolism and affects atherosclerosis remain largely unexplored. Here, we report that KLF14 is dysregulated in the liver of 2 dyslipidemia mouse models. We evaluated the effects of both KLF14 overexpression and genetic inactivation and determined that KLF14 regulates plasma HDL-C levels and cholesterol efflux capacity by modulating hepatic ApoA-I production. Hepatic-specific Klf14 deletion in mice resulted in decreased circulating HDL-C levels. In an attempt to pharmacologically target KLF14 as an experimental therapeutic approach, we identified perhexiline, an approved therapeutic small molecule presently in clinical use to treat angina and heart failure, as a KLF14 activator. Indeed, in WT mice, treatment with perhexiline increased HDL-C levels and cholesterol efflux capacity via KLF14-mediated upregulation of ApoA-I expression. Moreover, perhexiline administration reduced atherosclerotic lesion development in apolipoprotein E-deficient mice. Together, these results provide comprehensive insight into the KLF14-dependent regulation of HDL-C and subsequent atherosclerosis and indicate that interventions that target the KLF14 pathway should be further explored for the treatment of atherosclerosis.
Collapse
|
80
|
Tricoci P, D'Andrea DM, Gurbel PA, Yao Z, Cuchel M, Winston B, Schott R, Weiss R, Blazing MA, Cannon L, Bailey A, Angiolillo DJ, Gille A, Shear CL, Wright SD, Alexander JH. Infusion of Reconstituted High-Density Lipoprotein, CSL112, in Patients With Atherosclerosis: Safety and Pharmacokinetic Results From a Phase 2a Randomized Clinical Trial. J Am Heart Assoc 2015; 4:e002171. [PMID: 26307570 PMCID: PMC4599471 DOI: 10.1161/jaha.115.002171] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background CSL112 is a new formulation of human apolipoprotein A-I (apoA-I) being developed to reduce cardiovascular events following acute coronary syndrome. This phase 2a, randomized, double-blind, multicenter, dose-ranging trial represents the first clinical investigation to assess the safety and pharmacokinetics/pharmacodynamics of a CSL112 infusion among patients with stable atherosclerotic disease. Methods and Results Patients were randomized to single ascending doses of CSL112 (1.7, 3.4, or 6.8 g) or placebo, administered over a 2-hour period. Primary safety assessments consisted of alanine aminotransferase or aspartate aminotransferase elevations >3× upper limits of normal and study drug–related adverse events. Pharmacokinetic/pharmacodynamic assessments included apoA-I plasma concentration and measures of the ability of serum to promote cholesterol efflux from cells ex vivo. Of 45 patients randomized, 7, 12, and 14 received 1.7-, 3.4-, and 6.8-g CSL112, respectively, and 11 received placebo. There were no clinically significant elevations (>3× upper limit of normal) in alanine aminotransferase or aspartate aminotransferase. Adverse events were nonserious and mild and occurred in 5 (71%), 5 (41%), and 6 (43%) patients in the CSL112 1.7-, 3.4-, and 6.8-g groups, respectively, compared with 3 (27%) placebo patients. The imbalance in adverse events was attributable to vessel puncture/infusion-site bruising. CSL112 resulted in rapid (Tmax≈2 hours) and dose-dependent increases in apoA-I (145% increase in the 6.8-g group) and total cholesterol efflux (up to 3.1-fold higher than placebo) (P<0.001). Conclusions CSL112 infusion was well tolerated in patients with stable atherosclerotic disease. CSL112 immediately raised apoA-I levels and caused a rapid and marked increase in the capacity of serum to efflux cholesterol. This potential novel approach for the treatment of atherosclerosis warrants further investigation. Clinical Trial Registration URL: http://www.ClinicalTrials.gov. Unique identifier: NCT01499420.
Collapse
Affiliation(s)
| | | | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore and Johns Hopkins University School of Medicine, Baltimore, MD (P.A.G.)
| | - Zhenling Yao
- CSL Behring, King of Prussia, PA (D.M.A., Z.Y., C.L.S., S.D.W.)
| | | | - Brion Winston
- Black Hills Cardiovascular Research, Rapid City, SD (B.W.)
| | | | | | | | - Louis Cannon
- Cardiac and Vascular Research Center of Northern Michigan, Petoskey, MI (L.C.)
| | - Alison Bailey
- Gill Heart Institute, University of Kentucky, Lexington, KY (A.B.)
| | | | | | - Charles L Shear
- CSL Behring, King of Prussia, PA (D.M.A., Z.Y., C.L.S., S.D.W.)
| | - Samuel D Wright
- CSL Behring, King of Prussia, PA (D.M.A., Z.Y., C.L.S., S.D.W.)
| | - John H Alexander
- Duke Clinical Research Institute, Durham, NC (P.T., M.A.B., J.H.A.)
| |
Collapse
|
81
|
Ladeiras-Lopes R, Agewall S, Tawakol A, Staels B, Stein E, Mentz RJ, Leite-Moreira A, Zannad F, Koenig W. Atherosclerosis: Recent trials, new targets and future directions. Int J Cardiol 2015; 192:72-81. [DOI: 10.1016/j.ijcard.2015.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/09/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
|
82
|
Abstract
The pathogenesis and progression of atherosclerosis are integrally connected to the concentration and function of lipoproteins in various classes. This review examines existing and emerging approaches to modify low-density lipoprotein and lipoprotein (a), triglyceride-rich lipoproteins, and high-density lipoproteins, emphasizing approaches that have progressed to clinical evaluation. Targeting of nuclear receptors and phospholipases is also discussed.
Collapse
Affiliation(s)
- Rose Q Do
- VA Medical Center, University of Colorado School of Medicine, Denver, CO, USA
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute and University of Adelaide, Adelaide, SA, Australia
| | - Gregory G Schwartz
- VA Medical Center, University of Colorado School of Medicine, Denver, CO, USA
| |
Collapse
|
83
|
Du Y, Wang L, Hong B. High-density lipoprotein-based drug discovery for treatment of atherosclerosis. Expert Opin Drug Discov 2015; 10:841-55. [PMID: 26022101 DOI: 10.1517/17460441.2015.1051963] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Although there has been great progress achieved by the use of intensive statin therapy, the burden of atherosclerotic cardiovascular disease (CVD) remains high. This has initiated the search for novel high-density lipoprotein (HDL)-based therapeutics. Recent years have witnessed a shift from traditional raising HDL-C levels to enhancing HDL functionality, in which the process of reverse cholesterol transport (RCT) has acquired much attention. AREAS COVERED In this review, the authors describe the key factors involved in RCT process for potential drug targets to reduce the CVD risk. Furthermore, the review provides a summary of the effective screening methods that have been developed to target RCT and their applications. This review also introduces some new strategies currently being clinically developed, which have the potential to improve HDL function in the RCT process. EXPERT OPINION It is rational that the functionality of HDL is more important than the plasma HDL-C level in the evaluation of pharmacological treatment in atherosclerosis. HDL-based strategies designed to promote macrophage RCT are a major area of current drug discovery and development for atherosclerotic diseases. A better understanding of the functionality of HDL and its relationship with atherosclerosis will expand our knowledge of the role of HDL in lipid metabolism, holding promise for a future successful HDL-based therapy.
Collapse
Affiliation(s)
- Yu Du
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Tiantan Xili, Beijing 100050 , China
| | | | | |
Collapse
|
84
|
|
85
|
Underappreciated Opportunities for High-Density Lipoprotein Particles in Risk Stratification and Potential Targets of Therapy. Cardiovasc Drugs Ther 2015; 29:41-50. [DOI: 10.1007/s10557-014-6567-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
86
|
Brand M, Measures AM, Wilson BG, Cortopassi WA, Alexander R, Höss M, Hewings DS, Rooney TPC, Paton RS, Conway SJ. Small molecule inhibitors of bromodomain-acetyl-lysine interactions. ACS Chem Biol 2015; 10:22-39. [PMID: 25549280 DOI: 10.1021/cb500996u] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bromodomains are protein modules that bind to acetylated lysine residues. Their interaction with histone proteins suggests that they function as "readers" of histone lysine acetylation, a component of the proposed "histone code". Bromodomain-containing proteins are often found as components of larger protein complexes with roles in fundamental cellular process including transcription. The publication of two potent ligands for the BET bromodomains in 2010 demonstrated that small molecules can inhibit the bromodomain-acetyl-lysine protein-protein interaction. These molecules display strong phenotypic effects in a number of cell lines and affect a range of cancers in vivo. This work stimulated intense interest in developing further ligands for the BET bromodomains and the design of ligands for non-BET bromodomains. Here we review the recent progress in the field with particular attention paid to ligand design, the assays employed in early ligand discovery, and the use of computational approaches to inform ligand design.
Collapse
Affiliation(s)
- Michael Brand
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Angelina M. Measures
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Brian G. Wilson
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Wilian A. Cortopassi
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
- Department
of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K
| | | | | | - David S. Hewings
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Timothy P. C. Rooney
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Robert S. Paton
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
- Department
of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K
| | - Stuart J. Conway
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| |
Collapse
|
87
|
Santos-Gallego CG, Badimon JJ, Rosenson RS. Beginning to understand high-density lipoproteins. Endocrinol Metab Clin North Am 2014; 43:913-47. [PMID: 25432389 DOI: 10.1016/j.ecl.2014.08.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This article reconciles the classic view of high-density lipoproteins (HDL) associated with low risk for cardiovascular disease (CVD) with recent data (genetics studies and randomized clinical trials) casting doubt over the widely accepted beneficial role of HDL regarding CVD risk. Although HDL cholesterol has been used as a surrogate measure to investigate HDL function, the cholesterol content in HDL particles is not an indicator of the atheroprotective properties of HDL. Thus, more precise measures of HDL metabolism are needed to reflect and account for the beneficial effects of HDL particles. Current and emerging therapies targeting HDL are discussed.
Collapse
Affiliation(s)
- Carlos G Santos-Gallego
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1030, New York, NY 10029, USA
| | - Juan J Badimon
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1030, New York, NY 10029, USA
| | - Robert S Rosenson
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1030, New York, NY 10029, USA.
| |
Collapse
|
88
|
Tough DF, Lewis HD, Rioja I, Lindon MJ, Prinjha RK. Epigenetic pathway targets for the treatment of disease: accelerating progress in the development of pharmacological tools: IUPHAR Review 11. Br J Pharmacol 2014; 171:4981-5010. [PMID: 25060293 PMCID: PMC4253452 DOI: 10.1111/bph.12848] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/22/2014] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
The properties of a cell are determined both genetically by the DNA sequence of its genes and epigenetically through processes that regulate the pattern, timing and magnitude of expression of its genes. While the genetic basis of disease has been a topic of intense study for decades, recent years have seen a dramatic increase in the understanding of epigenetic regulatory mechanisms and a growing appreciation that epigenetic misregulation makes a significant contribution to human disease. Several large protein families have been identified that act in different ways to control the expression of genes through epigenetic mechanisms. Many of these protein families are finally proving tractable for the development of small molecules that modulate their function and represent new target classes for drug discovery. Here, we provide an overview of some of the key epigenetic regulatory proteins and discuss progress towards the development of pharmacological tools for use in research and therapy.
Collapse
Affiliation(s)
- David F Tough
- Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Epinova DPU, Stevenage, UK
| | | | | | | | | |
Collapse
|
89
|
Phenotypic screening and fragment-based approaches to the discovery of small-molecule bromodomain ligands. Future Med Chem 2014; 6:179-204. [PMID: 24467243 DOI: 10.4155/fmc.13.197] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bromodomains are protein modules that bind to acetylated lysine residues and hence facilitate protein-protein interactions. These bromodomain-mediated interactions often play key roles in transcriptional regulation and their dysfunction is implicated in a large number of diseases. The discovery of potent and selective small-molecule bromodomain and extra C-terminal domain bromodomain ligands, which show promising results for the treatment of cancers and atherosclerosis, has promoted intense interest in this area. Here we describe the progress that has been made to date in the discovery of small-molecule bromodomain ligands, with particular emphasis on the roles played by phenotypic screening and fragment-based approaches. In considering the future of the field we discuss the prospects for development of molecular probes and drugs for the non-bromodomain and extra C-terminal domain bromodomains.
Collapse
|
90
|
|
91
|
Abstract
High-density lipoproteins (HDL) are a target for drug development because of their proposed anti-atherogenic properties. In this review, we will briefly discuss the currently established drugs for increasing HDL-C, namely niacin and fibrates, and some of their limitations. Next, we will focus on novel alternative therapies that are currently being developed for raising HDL-C, such as CETP inhibitors. Finally, we will conclude with a review of novel drugs that are being developed for modulating the function of HDL based on HDL mimetics. Gaps in our knowledge and the challenges that will have to be overcome for these new HDL based therapies will also be discussed.
Collapse
Affiliation(s)
- Alan T Remaley
- National Heart, Lung and Blood Institute, NIH, 10 Center Drive, Bldg. 10, Rm. 2C-433, Bethesda, MD, USA
| | - Giuseppe D Norata
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy The Blizard Institute, Centre for Diabetes, Barts and The London School of Medicine & Dentistry, Queen Mary University, London, UK
| | - Alberico L Catapano
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
92
|
BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc Natl Acad Sci U S A 2014; 111:E2721-30. [PMID: 24979794 DOI: 10.1073/pnas.1406722111] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The bromodomain and extraterminal (BET) domain family of proteins binds to acetylated lysines on histones and regulates gene transcription. Recently, BET inhibitors (BETi) have been developed that show promise as potent anticancer drugs against various solid and hematological malignancies. Here we show that the structurally novel and orally bioavailable BET inhibitor RVX2135 inhibits proliferation and induces apoptosis of lymphoma cells arising in Myc-transgenic mice in vitro and in vivo. We find that BET inhibition exhibits broad transcriptional effects in Myc-transgenic lymphoma cells affecting many transcription factor networks. By examining the genes induced by BETi, which have largely been ignored to date, we discovered that these were similar to those induced by histone deacetylase inhibitors (HDACi). HDACi also induced cell-cycle arrest and cell death of Myc-induced murine lymphoma cells and synergized with BETi. Our data suggest that BETi sensitize Myc-overexpressing lymphoma cells partly by inducing HDAC-silenced genes, and suggest synergistic and therapeutic combinations by targeting the genetic link between BETi and HDACi.
Collapse
|
93
|
Barylski M, Toth PP, Nikolic D, Banach M, Rizzo M, Montalto G. Emerging therapies for raising high-density lipoprotein cholesterol (HDL-C) and augmenting HDL particle functionality. Best Pract Res Clin Endocrinol Metab 2014; 28:453-61. [PMID: 24840270 DOI: 10.1016/j.beem.2013.11.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
High-density lipoprotein (HDL) particles are highly complex polymolecular aggregates capable of performing a remarkable range of atheroprotective functions. Considerable research is being performed throughout the world to develop novel pharmacologic approaches to: (1) promote apoprotein A-I and HDL particle biosynthesis; (2) augment capacity for reverse cholesterol transport so as to reduce risk for the development and progression of atherosclerotic disease; and (3) modulate the functionality of HDL particles in order to increase their capacity to antagonize oxidation, inflammation, thrombosis, endothelial dysfunction, insulin resistance, and other processes that participate in arterial wall injury. HDL metabolism and the molecular constitution of HDL particles are highly complex and can change in response to both acute and chronic alterations in the metabolic milieu. To date, some of these interventions have been shown to positively impact rates of coronary artery disease progression. However, none of them have as yet been shown to significantly reduce risk for cardiovascular events. In the next 3-5 years a variety of pharmacologic interventions for modulating HDL metabolism and functionality will be tested in large, randomized, prospective outcomes trials. It is hoped that one or more of these therapeutic approaches will result in the ability to further reduce risk for cardiovascular events once low-density lipoprotein cholesterol and non-HDL-cholesterol targets have been attained.
Collapse
Affiliation(s)
- Marcin Barylski
- Department of Internal Medicine and Cardiological Rehabilitation, Medical University of Lodz, Lodz, Poland.
| | - Peter P Toth
- CGH Medical Center, Sterling, IL 61081, USA; University of Illinois School of Medicine, Peoria, IL, USA.
| | - Dragana Nikolic
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Maciej Banach
- Nephrology and Hypertension, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Manfredi Rizzo
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy.
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| |
Collapse
|
94
|
Kingwell BA, Chapman MJ, Kontush A, Miller NE. HDL-targeted therapies: progress, failures and future. Nat Rev Drug Discov 2014; 13:445-64. [DOI: 10.1038/nrd4279] [Citation(s) in RCA: 256] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
95
|
Subedi BH, Joshi PH, Jones SR, Martin SS, Blaha MJ, Michos ED. Current guidelines for high-density lipoprotein cholesterol in therapy and future directions. Vasc Health Risk Manag 2014; 10:205-16. [PMID: 24748800 PMCID: PMC3986285 DOI: 10.2147/vhrm.s45648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many studies have suggested that a significant risk factor for atherosclerotic cardiovascular disease (ASCVD) is low high-density lipoprotein cholesterol (HDL-C). Therefore, increasing HDL-C with therapeutic agents has been considered an attractive strategy. In the prestatin era, fibrates and niacin monotherapy, which cause modest increases in HDL-C, reduced ASCVD events. Since their introduction, statins have become the cornerstone of lipoprotein therapy, the benefits of which are primarily attributed to decrease in low-density lipoprotein cholesterol. Findings from several randomized trials involving niacin or cholesteryl ester transfer protein inhibitors have challenged the concept that a quantitative elevation of plasma HDL-C will uniformly translate into ASCVD benefits. Consequently, the HDL, or more correctly, HDL-C hypothesis has become more controversial. There are no clear guidelines thus far for targeting HDL-C or HDL due to lack of solid outcomes data for HDL specific therapies. HDL-C levels are only one marker of HDL out of its several structural or functional properties. Novel approaches are ongoing in developing and assessing agents that closely mimic the structure of natural HDL or replicate its various functions, for example, reverse cholesterol transport, vasodilation, anti-inflammation, or inhibition of platelet aggregation. Potential new approaches like HDL infusions, delipidated HDL, liver X receptor agonists, Apo A-I upregulators, Apo A mimetics, and gene therapy are in early phase trials. This review will outline current therapies and describe future directions for HDL therapeutics.
Collapse
Affiliation(s)
- Bishnu H Subedi
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA ; Greater Baltimore Medical Center, Baltimore, MD, USA
| | - Parag H Joshi
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Steven R Jones
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Seth S Martin
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Erin D Michos
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| |
Collapse
|
96
|
Nicholls SJ, Pisaniello AD, Kataoka Y, Puri R. Lipid pharmacotherapy for treatment of atherosclerosis. Expert Opin Pharmacother 2014; 15:1119-25. [DOI: 10.1517/14656566.2014.904287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
97
|
Hajhosseiny R, Sabir I, Khavandi K, Wierzbicki AS. The ebbs and flows in the development of cholesterol-lowering drugs: prospects for the future. Clin Pharmacol Ther 2014; 96:64-73. [PMID: 24699033 DOI: 10.1038/clpt.2014.76] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/27/2014] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, and its prevalence is increasing worldwide. Statins are the mainstay of treatment but do not address all aspects of CVD risk. Other lipid-lowering therapies are available but are less effective than statins. New therapies to lower low-density-lipoprotein cholesterol (LDL-C) by as much as statins, to reduce triglycerides (TGs), and to modify the metabolism of high-density lipoproteins (HDLs) are in development.
Collapse
Affiliation(s)
- R Hajhosseiny
- Department of Cardiology, St Thomas' Hospital, King's College Academic Health Partners, British Heart Foundation Centre of Cardiovascular Excellence, London, UK
| | - I Sabir
- Department of Cardiology, St Thomas' Hospital, King's College Academic Health Partners, British Heart Foundation Centre of Cardiovascular Excellence, London, UK
| | - K Khavandi
- Department of Cardiology, St Thomas' Hospital, King's College Academic Health Partners, British Heart Foundation Centre of Cardiovascular Excellence, London, UK
| | - A S Wierzbicki
- Department of Metabolic Medicine and Chemical Pathology, Guy's & St Thomas' Hospitals, London, UK
| |
Collapse
|
98
|
Abstract
PURPOSE OF REVIEW To examine the current and future therapeutic option of HDL-based therapies. RECENT FINDINGS The inverse association between plasma level of high-density lipoprotein cholesterol (HDL-C) is strong and coherent across the population studied. In-vitro and in-vivo studies show the strong biological plausibility for HDL as a therapeutic target. Mendelian randomization does not support HDL-C as a causal (protective) cardiovascular risk factor, and clinical data does not support the concept that raising HDL-cholesterol mass alters the outcomes. Better biomarkers of HDL function are being examined in the clinical trials. These include cellular cholesterol efflux, antioxidant and anti-inflammatory effects, effects on vascular endothelial cells (inflammation and nitric oxide release) and endothelial progenitor cells. Novel therapeutic agents that alter HDL function are in advanced phase 3 trials and in early preclinical trials. These include inhibitors of cholesteryl ester transfer protein, reconstituted proteoliposomes, apolipoprotein A-I and HDL mimetic peptides and small molecules that increase apo A-I production rate. SUMMARY Targeting HDL-C has, to date, not led to changes in the cardiovascular outcomes. Novel therapeutic advances target the HDL function. In keeping with the recent 2013 American College of Cardiology/American Heart Association Guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults, the major focus of prevention lies with LDL-cholesterol reduction.
Collapse
Affiliation(s)
- Anouar Hafiane
- McGill University Health Center/Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
99
|
Karavia EA, Zvintzou E, Petropoulou PI, Xepapadaki E, Constantinou C, Kypreos KE. HDL quality and functionality: what can proteins and genes predict? Expert Rev Cardiovasc Ther 2014; 12:521-32. [DOI: 10.1586/14779072.2014.896741] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
100
|
van Capelleveen JC, Brewer HB, Kastelein JJP, Hovingh GK. Novel therapies focused on the high-density lipoprotein particle. Circ Res 2014; 114:193-204. [PMID: 24385512 DOI: 10.1161/circresaha.114.301804] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiovascular disease (CVD) remains a major burden for morbidity and mortality in the general population, despite current efficacious low-density lipoprotein-cholesterol-lowering therapies. Consequently, novel therapies are required to reduce this residual risk. Prospective epidemiological studies have shown that high-density lipoprotein-cholesterol (HDL-C) levels are inversely correlated with cardiovascular disease risk, and this initiated the quest for HDL-C-increasing therapies. Consequently, several different targets in HDL metabolism have been identified. Initial studies addressing the effect of cholesteryl ester transfer protein inhibition on cardiovascular disease outcome have been discontinued for reasons of futility or increased mortality. As of yet, 2 cholesteryl ester transfer protein inhibitors are still in phase III studies. Other HDL-based interventions, such as apolipoprotein A1-based compounds, ABC-transporter upregulators, selective peroxisome proliferator-activated receptor modulators and lecithin-cholesterol acyltransferase-based therapy, hold great promise for the future. The aim of this review is to provide a comprehensive overview of HDL-targeted pharmaceutical strategies in humans, both in early development as well as in late stage clinical trials.
Collapse
Affiliation(s)
- Julian C van Capelleveen
- From the Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands (J.C.v.C., J.J.P.K., G.K.H.); and MedStar Research Institute, Washington Hospital Center, Washington, DC (H.B.B.)
| | | | | | | |
Collapse
|