Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS One ; 19(2): e0296052, 2024.
Article in English | MEDLINE | ID: mdl-38408107

ABSTRACT

HDL-apolipoprotein A-I exchange (HAE) measures a functional property associated with HDL's ability to mediate reverse cholesterol transport. HAE has been used to examine HDL function in case-control studies but not in studies of therapeutics that alter HDL particle composition. This study investigates whether niacin and omega-3 fatty acids induce measurable changes in HAE using a cohort of fifty-six subjects with metabolic syndrome (MetS) who were previously recruited to a double-blind trial where they were randomized to 16 weeks of treatment with dual placebo, extended-release niacin (ERN, 2g/day), prescription omega-3 ethyl esters (P-OM3, 4g/day), or the combination. HAE was assessed at the beginning and end of the study. Compared to placebo, ERN and P-OM3 alone significantly increased HAE by 15.1% [8.2, 22.0] (P<0.0001) and 11.1% [4.5, 17.7] (P<0.0005), respectively, while in combination they increased HAE by 10.0% [2.5, 15.8] (P = 0.005). When HAE was evaluated per unit mass of apoA-I ERN increased apoA-I specific exchange activity by 20% (2, 41 CI, P = 0.02) and P-OM3 by 28% (9.6, 48 CI, P<0.0006). However the combination had no statistically significant effect, 10% (-9, 31 CI, P = 0.39). With regard to P-OM3 therapy in particular, the HAE assay detected an increase in this property in the absence of a concomitant rise in HDL-C and apoA-I levels, suggesting that the assay can detect functional changes in HDL that occur in the absence of traditional biomarkers.


Subject(s)
Fatty Acids, Omega-3 , Metabolic Syndrome , Niacin , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Niacin/therapeutic use , Apolipoprotein A-I/therapeutic use , Metabolic Syndrome/drug therapy , Cholesterol, HDL , Double-Blind Method
2.
Clin Drug Investig ; 37(5): 483-491, 2017 May.
Article in English | MEDLINE | ID: mdl-28213743

ABSTRACT

BACKGROUND: CER-001 comprises recombinant human apolipoprotein A-I complexed with phospholipids that mimics natural, nascent, pre-ß high-density lipoprotein (HDL). We present animal model data showing dose-dependent increases in cholesterol efflux with CER-001 and its subsequent elimination by reverse lipid transport, together with inhibition of atherosclerotic plaque progression. We report the first phase I study results with CER-001 in humans, starting at 0.25 mg/kg, which is 1/80th of the safe dose (20 mg/kg) established in 4-week multiple-dose animal studies dosed every second day. METHODS: Healthy volunteers, 18-55 years old with a low-density lipoprotein-cholesterol:HDL-cholesterol ratio greater than 3.0, received single intravenous escalating doses of CER-001 (0.25-45.0 mg/kg) and placebo in a double-blind randomised cross-over fashion. Subjects were followed up for 3 weeks post-dose. Assessments included adverse event monitoring, blood sampling, and clinical laboratory measurements. RESULTS: Thirty-two subjects were enrolled. All CER-001 doses (0.25-45 mg/kg) were safe and well tolerated, with an adverse event profile similar to placebo. Effects on clinical chemistry, haematology and coagulation parameters were comparable to placebo. No adverse effects of CER-001 on electrocardiograms were observed. No antibodies to apolipoprotein A-I were detected following single-dose administration of CER-001. Plasma apolipoprotein A-I levels increased in a dose-related manner and returned to baseline by 24 h post-dose for doses up to 10 mg/kg but remained in circulation for >72 h post-dose for doses >10 mg/kg. CER-001 caused elevations in plasma cholesterol and total and unesterified cholesterol in the HDL fraction. Mobilisation of unesterified cholesterol in the HDL fraction was seen with CER-001 at doses as low as 2 mg/kg. CONCLUSION: CER-001 is well tolerated when administered to humans as single doses up to 45 mg/kg and mobilises and eliminates cholesterol via reverse lipid transport.


Subject(s)
Apolipoprotein A-I/blood , Apolipoprotein A-I/therapeutic use , Cholesterol/blood , Phospholipids/therapeutic use , Recombinant Proteins/therapeutic use , Adult , Animals , Apolipoprotein A-I/pharmacology , Cholesterol, HDL/antagonists & inhibitors , Cholesterol, HDL/blood , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Evaluation, Preclinical/methods , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Phospholipids/pharmacology , Rabbits , Recombinant Proteins/pharmacology
4.
Arterioscler Thromb Vasc Biol ; 34(3): 543-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357062

ABSTRACT

OBJECTIVE: This study questions whether high-density lipoproteins (HDLs) and apolipoprotein A-I inhibit joint inflammation in streptococcal cell wall peptidoglycan-polysaccharide (PG-PS)-induced arthritis in female Lewis rats. APPROACH AND RESULTS: Administration of PG-PS to female Lewis rats caused acute joint inflammation after 4 days, followed by remission by day 8. The animals subsequently developed chronic joint inflammation that persisted until euthanasia at day 21. Treatment with apolipoprotein A-I 24 hours before and 24 hours after PG-PS administration reduced the acute and chronic joint inflammation. Treatment with apolipoprotein A-I at days 7, 9, and 11 after PG-PS administration reduced the chronic joint inflammation. Treatment with apolipoprotein A-I or reconstituted HDLs consisting of apolipoprotein A-I complexed with phosphatidylcholine 24 hours before and at days 1, 7, 9, and 11 after PG-PS administration reduced acute and chronic joint inflammation. Treatment with apolipoprotein A-I also reduced the inflammatory white blood cell count, synovial fluid proinflammatory cytokine levels, synovial tissue macrophage accumulation, as well as toll-like receptor 2, and inflammatory cytokine expression. At the molecular level, preincubation of human monocyte-derived macrophages with apolipoprotein A-I or reconstituted HDLs before PG-PS stimulation inhibited the PG-PS-induced increase in toll-like receptor 2 and myeloid differentiation primary response gene (88) mRNA levels, nuclear factor-κB activation, and proinflammatory cytokine production. The effects of apolipoprotein A-I and reconstituted HDLs were abolished by transfecting the human monocyte-derived macrophages with ATP-binding cassette transporter A1 or G1 siRNA. CONCLUSIONS: Apolipoprotein A-I and reconstituted HDLs attenuate PG-PS-induced arthritis in the rat. Studies in human monocyte-derived macrophages indicate that this benefit may be because of the inhibition of toll-like receptor 2 expression and decreased nuclear factor-κB activation in macrophages.


Subject(s)
Apolipoprotein A-I/therapeutic use , Arthritis, Experimental/drug therapy , Cholesterol, HDL/therapeutic use , Lipoproteins, HDL/therapeutic use , Phosphatidylcholines/therapeutic use , ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/physiology , Animals , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/antagonists & inhibitors , Apolipoprotein A-I/genetics , Apolipoprotein A-I/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Chemotaxis, Leukocyte/drug effects , Cholesterol, HDL/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , Humans , Leukocytes/pathology , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/pharmacology , Macrophages/metabolism , Myeloid Cells/pathology , Myeloid Differentiation Factor 88/biosynthesis , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Peptidoglycan/toxicity , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/pharmacology , Polysaccharides, Bacterial/toxicity , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred Lew , Synovial Membrane/metabolism , Synovial Membrane/pathology , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/genetics , Transfection
5.
Circ Res ; 113(1): e1-e9, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23613182

ABSTRACT

RATIONALE: Infusions of apolipoprotein AI (apoAI), mimetic peptides, or high-density lipoprotein (HDL) remain a promising approach for the treatment of atherosclerotic coronary disease. However, rapid clearance leads to a requirement for repeated administration of large amounts of material and limits effective plasma concentrations. OBJECTIVE: Because pegylation of purified proteins is commonly used as a method to increase their half-life in the circulation, we determined whether pegylation of apoAI or HDL would increase its plasma half-life and in turn its antiatherogenic potential. METHODS AND RESULTS: Initial pegylation attempts using lipid-poor apoAI showed a marked tendency to form multi-pegylated (PEG) species with reduced ability to promote cholesterol efflux from macrophage foam cells. However, pegylation of human holo-HDL or reconstituted phospholipid/apoAI particles (rHDL) led to selective N-terminal monopegylation of apoAI with full preservation of cholesterol efflux activity. The plasma clearance of PEG-rHDL was estimated after injection into hypercholesterolemic Apoe-/- mice; the half-life of pegylated PEG-apoAI after injection of PEG-rHDL was increased ≈7-fold compared with apoAI in nonpegylated rHDL. In comparison with nonpegylated rHDL, infusion of PEG-rHDL (40 mg/kg) into hypercholesterolemic Apoe-/- mice led to more pronounced suppression of bone marrow myeloid progenitor cell proliferation and monocytosis, as well as reduced atherosclerosis and a stable plaque phenotype. CONCLUSIONS: We describe a novel method for effective monopegylation of apoAI in HDL particles, in which lipid binding seems to protect against pegylation of key functional residues. Pegylation of apoAI in rHDL markedly increases its plasma half-life and enhances antiatherogenic properties in vivo.


Subject(s)
Aortic Diseases/prevention & control , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/prevention & control , Cholesterol/metabolism , Hypercholesterolemia/drug therapy , Lipoproteins, HDL/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Animals , Aortic Diseases/etiology , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/blood , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Cell Line/drug effects , Cell Line/metabolism , Drug Evaluation, Preclinical , Foam Cells/drug effects , Foam Cells/metabolism , Half-Life , Hematopoietic Stem Cells/metabolism , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Infusions, Intravenous , Injections, Intravenous , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/blood , Lipoproteins, HDL/therapeutic use , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control , Polyethylene Glycols/administration & dosage
6.
J Cardiovasc Pharmacol ; 57(3): 325-33, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21164355

ABSTRACT

Apolipoprotein A-I (ApoA-I)/high-density lipoprotein (HDL)-raising treatments are effective antiatherosclerotic strategies. We have compared the antiatherogenic effects of human ApoA-I (hApoA-I) overexpression by intraportal and intramuscular gene transfer in atherosclerotic ApoE-knockout mice. Atherosclerotic lesions were induced by atherogenic diet. After atherosclerosis induction, a group of animals was killed and served as atherosclerosis baseline-control group. The remaining animals were randomized into the following groups: (1) atherosclerosis-progression-control, (2) intraportal/vector administration, and (3) intramuscular/vector administration. Aortas and hearts were processed for atherosclerotic quantification by en face Sudan IV and Oil Red-O, respectively. Liver and muscle specimens were processed for protein/gene expression analysis. A sustained increase in hApoA-I/HDL plasma levels was observed in both transduced groups. hApoA-I overexpression abolished plaque progression versus progression-control group. hApoA-I overexpression significantly reduced lesion macrophage, feature indicative of plaque stabilization. Scavenger receptor class-B type I (SR-BI), but not ATP-binding cassette, sub-family A (ABCA), member 1 (ABCA-1), was significantly upregulated in treated groups versus progression-controls. The results of this study show a similar effect of hApoA-I/HDL overexpression on plaque progression/stabilization by 2 different routes of administration. Our results showing similar effects using either intramuscular administration and intraportal route of administration may have significant clinical implications, given the reduced medical risk to patient and cost of intramuscular injections.


Subject(s)
Aorta/drug effects , Apolipoprotein A-I/genetics , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Dependovirus/metabolism , Liver/drug effects , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , Animals , Aorta/pathology , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/blood , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol, HDL/blood , Dependovirus/genetics , Diet, Atherogenic , Disease Progression , Drug Evaluation, Preclinical , Genetic Vectors , Humans , Injections, Intramuscular , Injections, Intravenous , Liver/anatomy & histology , Liver/physiopathology , Mice , Mice, Knockout , Molecular Targeted Therapy , Scavenger Receptors, Class B/analysis , Scavenger Receptors, Class B/genetics , Time Factors , Transduction, Genetic
7.
Proc Natl Acad Sci U S A ; 107(46): 19997-20002, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21041624

ABSTRACT

We examined whether reduced levels of Apolipoprotein A-I (apoA-I) in ovarian cancer patients are causal in ovarian cancer in a mouse model. Mice expressing a human apoA-I transgene had (i) increased survival (P < 0.0001) and (ii) decreased tumor development (P < 0.01), when compared with littermates, following injection of mouse ovarian epithelial papillary serous adenocarcinoma cells (ID-8 cells). ApoA-I mimetic peptides reduced viability and proliferation of ID8 cells and cis-platinum-resistant human ovarian cancer cells, and decreased ID-8 cell-mediated tumor burden in C57BL/6J mice when administered subcutaneously or orally. Serum levels of lysophosphatidic acid, a well-characterized modulator of tumor cell proliferation, were significantly reduced (>50% compared with control mice, P < 0.05) in mice that received apoA-I mimetic peptides (administered either subcutaneously or orally), suggesting that binding and removal of lysophosphatidic acid is a potential mechanism for the inhibition of tumor development by apoA-I mimetic peptides, which may serve as a previously unexplored class of anticancer agents.


Subject(s)
Apolipoprotein A-I/therapeutic use , Ovarian Neoplasms/drug therapy , Peptides/therapeutic use , Precancerous Conditions/drug therapy , Animals , Apolipoprotein A-I/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drinking Behavior/drug effects , Female , Humans , Injections , Lysophospholipids/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation/pathology , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Peptides/pharmacology , Precancerous Conditions/pathology , Survival Analysis , Tumor Burden , Water
8.
Curr Opin Investig Drugs ; 11(9): 989-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20730693

ABSTRACT

Although statin treatment leads consistently to a reduction in major adverse coronary events and death in clinical trials, approximately 60 to 70% residual risk of these outcomes still remains. One frontier of investigational drug research is treatment to increase HDL, the 'good cholesterol' that is associated with a reduced risk of coronary artery disease. HDL and its major protein apolipoprotein A-I (apoAI) are protective against atherosclerosis through several mechanisms, including the ability to mediate reverse cholesterol transport. This review focuses on the preclinical and clinical findings for two types of therapies for the treatment of atherosclerosis: apoAI-containing compounds and apoAI mimetic peptides. Both of these therapies have excellent potential to be useful clinically to promote atherosclerosis regression and stabilize existing plaques, but significant hurdles must be overcome in order to develop these approaches into safe and effective therapies.


Subject(s)
Apolipoprotein A-I/therapeutic use , Atherosclerosis/drug therapy , Lipoproteins, HDL/blood , Peptides/therapeutic use , Animals , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacology , Cholesterol, HDL/metabolism , Clinical Trials as Topic , Coronary Artery Disease/drug therapy , Coronary Artery Disease/prevention & control , Drug Evaluation, Preclinical , Female , Humans , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/pharmacology , Lipoproteins, HDL/therapeutic use , Risk Reduction Behavior
9.
Expert Opin Investig Drugs ; 15(3): 227-41, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16503760

ABSTRACT

High-density lipoprotein (HDL) therapy is a novel and emerging area of therapeutic development in the cardiovascular field. It attempts to supplement and improve the vascular benefit exerted by other agents that are active on lipid metabolism, for example, hypolipidaemic drugs. Furthermore, it takes advantage of the novel techniques of coronary evaluation. A number of reports have examined the potential therapeutic properties of the synthetic HDLs prepared by complexing recombinant apolipoprotein (apo) A-I(Milano), a variant form of native apoA-I, with phospholipids. The availability of synthetic HDL complexes containing recombinant apoA-I(Milano) has opened up a new era of therapeutic management for coronary disease. HDL formulations of recombinant apoA-I(Milano)-phospholipid complexes have clearly shown rapid regression of a focal carotid atheroma as well as powerful protection from myocardial infarction in a rabbit model. In a pilot study, ETC-216 showed a significant reduction in coronary plaque burden after five weekly treatments, assessed by intravascular ultrasound in patients with acute coronary syndrome. Other therapeutic options of HDL therapy have recently became available.


Subject(s)
Lipoproteins, HDL/therapeutic use , Peptides/therapeutic use , Animals , Apolipoprotein A-I/therapeutic use , Arteriosclerosis/drug therapy , Arteriosclerosis/pathology , Coronary Disease/drug therapy , Coronary Disease/pathology , Humans , Phosphatidylcholines/therapeutic use
11.
Circulation ; 109(25): 3215-20, 2004 Jun 29.
Article in English | MEDLINE | ID: mdl-15197147

ABSTRACT

BACKGROUND: These studies were designed to determine the mechanism of action of an oral apolipoprotein (apo) A-I mimetic peptide, D-4F, which previously was shown to dramatically reduce atherosclerosis in mice. METHODS AND RESULTS: Twenty minutes after 500 microg of D-4F was given orally to apoE-null mice, small cholesterol-containing particles (CCPs) of 7 to 8 nm with pre-beta mobility and enriched in apoA-I and paraoxonase activity were found in plasma. Before D-4F, both mature HDL and the fast protein liquid chromatography fractions containing the CCPs were proinflammatory. Twenty minutes after oral D-4F, HDL and CCPs became antiinflammatory, and there was an increase in HDL-mediated cholesterol efflux from macrophages in vitro. Oral D-4F also promoted reverse cholesterol transport from intraperitoneally injected cholesterol-loaded macrophages in vivo. In addition, oral D-4F significantly reduced lipoprotein lipid hydroperoxides (LOOH), except for pre-beta HDL fractions, in which LOOH increased. CONCLUSIONS: The mechanism of action of oral D-4F in apoE-null mice involves rapid formation of CCPs, with pre-beta mobility enriched in apoA-I and paraoxonase activity. As a result, lipoprotein LOOH are reduced, HDL becomes antiinflammatory, and HDL-mediated cholesterol efflux and reverse cholesterol transport from macrophages are stimulated.


Subject(s)
Apolipoprotein A-I/pharmacology , Apolipoproteins E/deficiency , Arteriosclerosis/genetics , Cholesterol/metabolism , Hyperlipoproteinemia Type II/genetics , Lipoproteins, HDL/biosynthesis , Macrophages, Peritoneal/drug effects , Administration, Oral , Amino Acid Sequence , Animals , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/genetics , Arteriosclerosis/blood , Aryldialkylphosphatase/blood , Biological Transport/drug effects , Cells, Cultured , Chemotaxis/drug effects , Coculture Techniques , Drug Evaluation, Preclinical , Female , High-Density Lipoproteins, Pre-beta , Humans , Hyperlipoproteinemia Type II/blood , Inflammation , Lipid Peroxidation/drug effects , Lipoproteins, HDL/blood , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL