Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Clin Pharmacokinet ; 62(4): 541-558, 2023 04.
Article in English | MEDLINE | ID: mdl-36928983

ABSTRACT

Cardiovascular diseases are the leading cause of death worldwide. Although there have been substantial advances over the last decades, recurrent adverse cardiovascular events after myocardial infarction are still frequent, particularly during the first year of the index event. For decades, high-density lipoprotein (HDL) has been among the therapeutic targets for long-term prevention after an ischemic event. However, early trials focusing on increasing HDL circulating levels showed no improvement in clinical outcomes. Recently, the paradigm has shifted to increasing the functionality of HDL rather than its circulating plasma levels. For this purpose, apolipoprotein-AI-based infusion therapies have been developed, including reconstituted HDL, such as CSL112. During the last decade, CSL112 has been extensively studied in Phase 1 and 2 trials and has shown promising results. In particular, CSL112 has been studied in the Phase 2b AEGIS trial exhibiting good safety and tolerability profiles, which has led to the ongoing large-scale Phase 3 AEGIS-II trial. This systematic overview will provide a comprehensive summary of the CSL112 drug development program focusing on its pharmacodynamic, pharmacokinetic, and safety profiles.


Subject(s)
Lipoproteins, HDL , Myocardial Infarction , Humans , Lipoproteins, HDL/pharmacology , Lipoproteins, HDL/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/chemically induced , Apolipoprotein A-I/pharmacokinetics , Apolipoprotein A-I/therapeutic use , Drug Development
2.
Clin Transl Sci ; 15(10): 2331-2341, 2022 10.
Article in English | MEDLINE | ID: mdl-35933730

ABSTRACT

CSL112 (apolipoprotein A-I [apoA-I, human]) is a novel drug in development to reduce the risk of recurrent cardiovascular events following acute myocardial infarction by increasing cholesterol efflux capacity (CEC). This phase I study aimed to compare the pharmacokinetics (PKs), pharmacodynamics (PDs), and safety of CSL112 in Japanese and White subjects. A total of 34 Japanese subjects were randomized to receive a single infusion of CSL112 (2, 4, or 6 g) or placebo and 18 White subjects were randomized to receive a single dose of 6 g CSL112 or placebo, followed by PK/PD assessment and adverse events monitoring. In addition, PK/PD parameters were compared across the CSL112 clinical development program. Plasma exposure of apoA-I increased in a dose-dependent but nonlinear manner in Japanese subjects receiving a single dose of CSL112. Mean baseline-corrected area under the curve from 0 to 72 h (AUC0-72 ) increased from 840 to 6490 mg h/dl, in the 2 and 6 g cohorts, respectively, followed by dose-dependent increase of CEC. The plasma PK profile of apoA-I and increases in total and ATP binding cassette transporter A1 dependent CEC were comparable in Japanese and White subjects. The geometric mean ratio (Japanese:White) for plasma apoA-I AUC0-72 and maximum plasma concentration (Cmax ) was 1.08 and 0.945, respectively. Cross-study comparison analysis demonstrated similar CSL112 exposure and CEC enhancement in Japanese and non-Japanese subjects (including patients with cardiovascular disease) and further confirmed consistent PKs/PDs of CSL112. This study suggests CSL112 acutely enhances CEC and is well-tolerated with no differences between Japanese and White subjects.


Subject(s)
Apolipoprotein A-I , Lipoproteins, HDL , Humans , Apolipoprotein A-I/pharmacokinetics , Biological Transport , Cholesterol , Double-Blind Method
3.
Arterioscler Thromb Vasc Biol ; 38(4): 953-963, 2018 04.
Article in English | MEDLINE | ID: mdl-29437574

ABSTRACT

OBJECTIVE: CSL112 (apolipoprotein A-I [apoA-I; human]) is a novel formulation of apoA-I in development for reduction of early recurrent cardiovascular events after acute myocardial infarction. Cholesterol efflux capacity (CEC) is a marker of high-density lipoprotein (HDL) function that is strongly correlated with incident cardiovascular disease. Impaired CEC has been observed in patients with coronary heart disease. Here, we determined whether infused apoA-I improves CEC when administered to patients with stable atherosclerotic disease versus healthy volunteers. APPROACH AND RESULTS: Measurements of apoA-I, HDL unesterified cholesterol, HDL esterified cholesterol, pre-ß1-HDL, and CEC were determined in samples from patients with stable atherosclerotic disease before and after intravenous administration of CSL112. These measures were compared with 2 prior studies in healthy volunteers for differences in CEC at baseline and after CSL112 infusion. Patients with stable atherosclerotic disease exhibited significantly lower ATP-binding cassette transporter 1-mediated CEC at baseline (P<0.0001) despite slightly higher apoA-I levels when compared with healthy individuals (2 phase 1 studies pooled; P≤0.05), suggesting impaired HDL function. However, no differences were observed in apoA-I pharmacokinetics or in pre-ß1-HDL (P=0.5) or CEC (P=0.1) after infusion of CSL112. Similar elevation in CEC was observed in patients with low or high baseline HDL function (based on tertiles of apoA-I-normalized CEC; P=0.1242). These observations were extended and confirmed using cholesterol esterification as an additional measure. CONCLUSIONS: CSL112 shows comparable, strong, and immediate effects on CEC despite underlying cardiovascular disease. CSL112 is, therefore, a promising novel therapy for lowering the burden of atherosclerosis and reducing the risk of recurrent cardiovascular events.


Subject(s)
Anticholesteremic Agents/therapeutic use , Apolipoprotein A-I/therapeutic use , Atherosclerosis/drug therapy , Cholesterol/blood , Lipid Metabolism/drug effects , Lipoproteins, HDL/therapeutic use , Adolescent , Adult , Aged , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacokinetics , Apolipoprotein A-I/blood , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/blood , Atherosclerosis/diagnosis , Biomarkers/blood , Cholesterol, HDL/blood , Female , Healthy Volunteers , High-Density Lipoproteins, Pre-beta/blood , Humans , Lipoproteins, HDL/blood , Lipoproteins, HDL/pharmacokinetics , Male , Middle Aged , Queensland , South Australia , Treatment Outcome , United States , Young Adult
4.
J Am Heart Assoc ; 6(9)2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28939717

ABSTRACT

BACKGROUND: Subcutaneously injected lipid-free apoA-I (apolipoprotein A-I) reduces accumulation of lipid and immune cells within the aortic root of hypercholesterolemic mice without increasing high-density lipoprotein-cholesterol concentrations. Lymphatic vessels are now recognized as prerequisite players in the modulation of cholesterol removal from the artery wall in experimental conditions of plaque regression, and particular attention has been brought to the role of the collecting lymphatic vessels in early atherosclerosis-related lymphatic dysfunction. In the present study, we address whether and how preservation of collecting lymphatic function contributes to the protective effect of apoA-I. METHODS AND RESULTS: Atherosclerotic Ldlr-/- mice treated with low-dose lipid-free apoA-I showed enhanced lymphatic transport and abrogated collecting lymphatic vessel permeability in atherosclerotic Ldlr-/- mice when compared with albumin-control mice. Treatment of human lymphatic endothelial cells with apoA-I increased the adhesion of human platelets on lymphatic endothelial cells, in a bridge-like manner, a mechanism that could strengthen endothelial cell-cell junctions and limit atherosclerosis-associated collecting lymphatic vessel dysfunction. Experiments performed with blood platelets isolated from apoA-I-treated Ldlr-/- mice revealed that apoA-I decreased ex vivo platelet aggregation. This suggests that in vivo apoA-I treatment limits platelet thrombotic potential in blood while maintaining the platelet activity needed to sustain adequate lymphatic function. CONCLUSIONS: Altogether, we bring forward a new pleiotropic role for apoA-I in lymphatic function and unveil new potential therapeutic targets for the prevention and treatment of atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Apolipoprotein A-I/administration & dosage , Atherosclerosis/prevention & control , Lymphatic Vessels/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Genetic Predisposition to Disease , Humans , Injections, Subcutaneous , Lipid Metabolism/drug effects , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Mice, Inbred C57BL , Mice, Knockout , Permeability , Phenotype , Plaque, Atherosclerotic , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-3/metabolism
5.
Clin Transl Sci ; 10(6): 455-469, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28795506

ABSTRACT

A single dose of the apolipoprotein (apo)A-I mimetic peptide D-4F rendered high-density lipoprotein (HDL) less inflammatory, motivating the first multiple-dose study. We aimed to assess safety/tolerability, pharmacokinetics, and pharmacodynamics of daily, orally administered D-4F. High-risk coronary heart disease (CHD) subjects added double-blinded placebo or D-4F to statin for 13 days, randomly assigned 1:3 to ascending cohorts of 100, 300, then 500 mg (n = 62; 46 men/16 women). D-4F was safe and well-tolerated. Mean ± SD plasma D-4F area under the curve (AUC, 0-8h) was 6.9 ± 5.7 ng/mL*h (100 mg), 22.7 ± 19.6 ng/mL*h (300 mg), and 104.0 ± 60.9 ng/mL*h (500 mg) among men, higher among women. Whereas placebo dropped HDL inflammatory index (HII) 28% 8 h postdose (range, 1.25-0.86), 300-500 mg D-4F effectively halved HII: 1.35-0.57 and 1.22-0.63, respectively (P < 0.03 vs. placebo). Oral D-4F peptide dose predicted HII suppression, whereas plasma D-4F exposure was dissociated, suggesting plasma penetration is unnecessary. In conclusion, oral D-4F dosing rendered HDL less inflammatory, affirming oral D-4F as a potential therapy to improve HDL function.


Subject(s)
Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/therapeutic use , Inflammation/drug therapy , Lipoproteins, HDL/metabolism , Administration, Oral , Adult , Aged , Apolipoprotein A-I/adverse effects , Apolipoprotein A-I/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Risk Factors , Time Factors
6.
Arterioscler Thromb Vasc Biol ; 37(7): 1301-1306, 2017 07.
Article in English | MEDLINE | ID: mdl-28522696

ABSTRACT

OBJECTIVE: Apolipoprotein A-I (apoA-I) mimetic peptides have antiatherogenic properties of high-density lipoprotein in vitro and have been shown to inhibit atherosclerosis in vivo. It is unclear, however, if each in vitro antiatherogenic property of these peptides translates to a corresponding activity in vivo, and if so, which of these contributes most to reduce atherosclerosis. APPROACH AND RESULTS: The effect of 7 apoA-I mimetic peptides, which were developed to selectively reproduce a specific component of the antiatherogenic properties of apoA-I, on the development of atherosclerosis was investigated in apolipoprotein E-deficient mice fed a high-fat diet for 4 or 12 weeks. The peptides include those that selectively upregulate cholesterol efflux, or are anti-inflammatory, or have antioxidation properties. All the peptides studied effectively inhibited the in vivo development of atherosclerosis in this model to the same extent. However, none of the peptides had the same selective effect in vivo as they had exhibited in vitro. None of the tested peptides affected plasma lipoprotein profile; capacity of plasma to support cholesterol efflux was increased modestly and similarly for all peptides. CONCLUSIONS: There is a discordance between the selective in vitro and in vivo functional properties of apoA-I mimetic peptides, and the in vivo antiatherosclerotic effect of apoA-I-mimetic peptides is independent of their in vitro functional profile. Comparing the properties of apoA-I mimetic peptides in plasma rather than in the lipid-free state is better for predicting their in vivo effects on atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Apolipoprotein A-I/pharmacology , Atherosclerosis/prevention & control , Hypolipidemic Agents/pharmacology , Macrophages/drug effects , Peptide Fragments/pharmacology , Animals , Aortic Diseases/blood , Aortic Diseases/pathology , Apolipoprotein A-I/pharmacokinetics , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Biological Mimicry , Biomarkers/blood , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Genetic Predisposition to Disease , Lipoproteins/blood , Macrophages/metabolism , Male , Mice , Mice, Knockout , Peptide Fragments/pharmacokinetics , Phenotype , Plaque, Atherosclerotic , RAW 264.7 Cells , Tissue Distribution
7.
Protein Expr Purif ; 134: 18-24, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28336201

ABSTRACT

Apolipoprotein (apo) A-I is the major protein component of high-density lipoprotein (HDL) and plays key roles in the Reverse Cholesterol Transport pathway. In the past decade, reconstituted HDL (rHDL) has been employed as a therapeutic agent for treatment of atherosclerosis. The ability of rHDL to promote cholesterol efflux from peripheral cells has been documented to reduce the size of atherosclerotic plaque lesions. However, development of apoA-I rHDL-based therapeutics for human use requires a cost effective process to generate an apoA-I product that meets "Good Manufacturing Practice" standards. Methods available for production and isolation of unmodified recombinant human apoA-I at scale are cumbersome, laborious and complex. To overcome this obstacle, a streamlined two-step procedure has been devised for isolation of recombinant untagged human apoA-I from E. coli that takes advantage of its ability to re-fold to a native conformation following denaturation. Heat treatment of a sonicated E. coli supernatant fraction induced precipitation of a large proportion of host cell proteins (HCP), yielding apoA-I as the major soluble protein. Reversed-phase HPLC of this material permitted recovery of apoA-I largely free of HCP and endotoxin. Purified apoA-I possessed α-helix secondary structure, formed rHDL upon incubation with phospholipid and efficiently promoted cholesterol efflux from cholesterol loaded J774 macrophages.


Subject(s)
Apolipoprotein A-I , Escherichia coli/metabolism , Protein Refolding , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/isolation & purification , Apolipoprotein A-I/pharmacokinetics , Biological Transport, Active/drug effects , Cell Line , Cholesterol/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Humans , Macrophages/metabolism , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
8.
J Lipid Res ; 58(1): 124-136, 2017 01.
Article in English | MEDLINE | ID: mdl-27881716

ABSTRACT

apoA-I, apoA-I mimetic peptides, and their lipid complexes or reconstituted high-density lipoprotein (HDL) have been studied as treatments for various pathologies. However, consensus is lacking about the best method for administration, by intravenous (IV) or intraperitoneal (IP) routes, and formulation, as an HDL particle or in a lipid-free form. The objective of this study was to systematically examine peptide plasma levels, cholesterol mobilization, and lipoprotein remodeling in vivo following administration of lipid-free apoA-I peptide (22A) or phospholipid reconstituted 22A-sHDL by IV and IP routes. The mean circulation half-life was longer for 22A-sHDL (T1/2 = 6.27 h) than for free 22A (T1/2 = 3.81 h). The percentage of 22A absorbed by the vascular compartment after the IP dosing was ∼50% for both 22A and 22A-sHDL. The strongest pharmacologic response came from IV injection of 22A-sHDL, specifically a 5.3-fold transient increase in plasma-free cholesterol (FC) level compared with 1.3- and 1.8-fold FC increases for 22A-IV and 22A-sHDL-IP groups. Addition of either 22A or 22A-sHDL to rat plasma caused lipoprotein remodeling and appearance of a lipid-poor apoA-I. Hence, both the route of administration and the formulation of apoA-I peptide significantly affect its pharmacokinetics and pharmacodynamics.


Subject(s)
Apolipoprotein A-I/administration & dosage , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Peptides/administration & dosage , Administration, Intravenous , Animals , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacokinetics , Humans , Injections, Intraperitoneal , Peptides/metabolism , Peptides/pharmacokinetics , Rats
9.
Pharmacol Res ; 111: 86-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27155060

ABSTRACT

Epidemiological studies support an inverse correlation between HDL-C and cardiovascular disease. However, low HDL-C levels do not always segregate with premature disease. These include, LCAT deficiency and the apolipoproteinA-IMilano (AIM) variant. AIM has a cysteine for arginine at position 173 in the otherwise cysteine free protein permitting AIM homodimerization and apoA-II heterodimerization. We relate the biochemical characteristics of low HDL-C phenotype AIM carriers to lipoprotein changes in humans administered recombinant dimeric AIM/palmitoyl-oleoyl phosphatidyl choline (ETC-216). Pharmacokinetic analysis of infused ETC-216 suggest a slow distribution of AIM into peripheral tissue and an extremely long terminal half-life in plasma. Following ETC-216 administration to normal human volunteers, an initial dose-dependent HDL-C elevation was observed. Thereafter, subjects transiently acquired a lipoprotein profile similar to that of AIM carriers, including reduced HDL-C and mild hypertriglyceridemia. The time-dependent changes in plasma lipids/lipoproteins may support an increased tissue cholesterol removing capacity of ETC-216. These findings provide mechanistic insight into the rapid removal of atheromatous plaques observed in humans, possibly linked to enhanced cholesterol removal capacity of ETC-216.


Subject(s)
Anticholesteremic Agents/administration & dosage , Apolipoprotein A-I/administration & dosage , Heterozygote , Phosphatidylcholines/administration & dosage , Adult , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacokinetics , Apolipoprotein A-I/adverse effects , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Apolipoprotein A-I/pharmacokinetics , Biomarkers/blood , Cholesterol, HDL/blood , Double-Blind Method , Female , Genotype , Half-Life , Healthy Volunteers , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/genetics , Infusions, Intravenous , Male , Middle Aged , Models, Biological , Models, Statistical , Phenotype , Phosphatidylcholines/adverse effects , Phosphatidylcholines/blood , Phosphatidylcholines/pharmacokinetics , Tissue Distribution , Triglycerides/blood , Young Adult
10.
ACS Nano ; 10(3): 3015-41, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26889958

ABSTRACT

High-density lipoproteins (HDL) are endogenous nanoparticles involved in the transport and metabolism of cholesterol, phospholipids, and triglycerides. HDL is well-known as the "good" cholesterol because it not only removes excess cholesterol from atherosclerotic plaques but also has anti-inflammatory and antioxidative properties, which protect the cardiovascular system. Circulating HDL also transports endogenous proteins, vitamins, hormones, and microRNA to various organs. Compared with other synthetic nanocarriers, such as liposomes, micelles, and inorganic and polymeric nanoparticles, HDL has unique features that allow them to deliver cargo to specific targets more efficiently. These attributes include their ultrasmall size (8-12 nm in diameter), high tolerability in humans (up to 8 g of protein per infusion), long circulating half-life (12-24 h), and intrinsic targeting properties to different recipient cells. Various recombinant ApoA proteins and ApoA mimetic peptides have been recently developed for the preparation of reconstituted HDL that exhibits properties similar to those of endogenous HDL and has a potential for industrial scale-up. In this review, we will summarize (a) clinical pharmacokinetics and safety of reconstituted HDL products, (b) comparison of HDL with inorganic and other organic nanoparticles,


Subject(s)
Drug Carriers/chemistry , Lipoproteins, HDL/chemistry , Nanoparticles/chemistry , Animals , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacokinetics , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/pharmacokinetics , Nanomedicine/methods , Nanoparticles/analysis , Nanoparticles/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics
11.
Toxicol Sci ; 150(2): 378-89, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26781512

ABSTRACT

High density lipoprotein (HDL)-targeted therapies, which promote cholesterol efflux from cells, are currently in development for reducing cardiovascular events in acute coronary syndrome. Human apolipoprotein A-I (apoA-I), the major HDL protein, was fused to the trimerization domain of tetranectin (TN) and complexed with phospholipids to generate a HDL mimetic (lipidated TN-ApoA-I) with reduced renal clearance and enhanced efficacy. Cynomolgus monkeys received 24-h intravenous infusions of control, 100 mg/kg or 400 mg/kg lipidated TN-ApoA-I every 4 days for 3 weeks, followed by a 6-week recovery period. After multiple infusions of lipidated TN-ApoA-I, clinical condition deteriorated and was accompanied by changes indicative of a progressive inflammatory response; increased levels of cytokines, C-reactive protein and vascular/perivascular infiltrates in multiple tissues. Rapid formation of antidrug antibodies occurred in all animals receiving lipidated TN-ApoA-I. Enhanced drug clearance corresponding to a relative lack of high molecular weight immune complexes in blood, suggestive of preferred removal/clearance, was observed in some animals. Expected dose-dependent increases in serum lipids were accompanied by vacuolated monocytes/macrophages in multiple organs, which in the glomeruli were shown to be CD68-positive, contain lipid and co-localized with granular IgG deposits. Lipid accumulation may have been a direct result of a high drug load, possibly enhanced by immune complex formation, inflammation, and altered lipid metabolism. Noteworthy was the inter- individual inconsistency in the severity of clinical and histopathologic findings, drug clearance and inflammatory markers. In conclusion, multiple infusions of lipidated TN-ApoA-I resulted in high immunogenicity, lipid accumulation and were not well tolerated in nonhuman primates.


Subject(s)
Antigen-Antibody Complex/blood , Apolipoprotein A-I/toxicity , Lectins, C-Type/administration & dosage , Lipids/blood , Recombinant Fusion Proteins/toxicity , Animals , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/immunology , Apolipoprotein A-I/pharmacokinetics , C-Reactive Protein/analysis , Cytokines/blood , Dose-Response Relationship, Drug , Fibrin Fibrinogen Degradation Products/analysis , Fibrinogen/analysis , Inflammation/blood , Inflammation/chemically induced , Infusions, Intravenous , Lectins, C-Type/immunology , Lipids/immunology , Macaca fascicularis , Male , Metabolic Clearance Rate , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics
12.
Biochemistry ; 54(4): 1123-31, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25564321

ABSTRACT

It is thought that apolipoprotein A-I (apoA-I) spontaneously exchanges between high-density lipoprotein (HDL)-bound and lipid-free states, which is relevant to the occurrence of preß-HDL particles in plasma. To improve our understanding of the mechanistic basis for this phenomenon, we performed kinetic and thermodynamic analyses for apoA-I exchange between discoidal HDL-bound and lipid-free forms using fluorescence-labeled apoA-I variants. Gel filtration experiments demonstrated that addition of excess lipid-free apoA-I to discoidal HDL particles promotes exchange of apoA-I between HDL-associated and lipid-free pools without alteration of the steady-state HDL particle size. Kinetic analysis of time-dependent changes in NBD fluorescence upon the transition of NBD-labeled apoA-I from HDL-bound to lipid-free state indicates that the exchange kinetics are independent of the collision frequency between HDL-bound and lipid-free apoA-I, in which the lipid binding ability of apoA-I affects the rate of association of lipid-free apoA-I with the HDL particles and not the rate of dissociation of HDL-bound apoA-I. Thus, C-terminal truncations or mutations that reduce the lipid binding affinity of apoA-I strongly impair the transition of lipid-free apoA-I to the HDL-bound state. Thermodynamic analysis of the exchange kinetics demonstrated that the apoA-I exchange process is enthalpically unfavorable but entropically favorable. These results explain the thermodynamic basis of the spontaneous exchange reaction of apoA-I associated with HDL particles. The altered exchangeability of dysfunctional apoA-I would affect HDL particle rearrangement, leading to perturbed HDL metabolism.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Thermodynamics , Apolipoprotein A-I/pharmacokinetics , Kinetics , Lipoproteins, HDL/pharmacokinetics , Protein Binding/physiology , Protein Engineering/methods
13.
J Am Heart Assoc ; 3(6): e001156, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25392541

ABSTRACT

BACKGROUND: Brain lipoprotein metabolism is dependent on lipoprotein particles that resemble plasma high-density lipoproteins but that contain apolipoprotein (apo) E rather than apoA-I as their primary protein component. Astrocytes and microglia secrete apoE but not apoA-I; however, apoA-I is detectable in both cerebrospinal fluid and brain tissue lysates. The route by which plasma apoA-I enters the central nervous system is unknown. METHODS AND RESULTS: Steady-state levels of murine apoA-I in cerebrospinal fluid and interstitial fluid are 0.664 and 0.120 µg/mL, respectively, whereas brain tissue apoA-I is ≈10% to 15% of its levels in liver. Recombinant, fluorescently tagged human apoA-I injected intravenously into mice localizes to the choroid plexus within 30 minutes and accumulates in a saturable, dose-dependent manner in the brain. Recombinant, fluorescently tagged human apoA-I accumulates in the brain for 2 hours, after which it is eliminated with a half-life of 10.3 hours. In vitro, human apoA-I is specifically bound, internalized, and transported across confluent monolayers of primary human choroid plexus epithelial cells and brain microvascular endothelial cells. CONCLUSIONS: Following intravenous injection, recombinant human apoA-I rapidly localizes predominantly to the choroid plexus. Because apoA-I mRNA is undetectable in murine brain, our results suggest that plasma apoA-I, which is secreted from the liver and intestine, gains access to the central nervous system primarily by crossing the blood-cerebrospinal fluid barrier via specific cellular mediated transport, although transport across the blood-brain barrier may also contribute to a lesser extent.


Subject(s)
Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/pharmacokinetics , Blood-Brain Barrier/metabolism , Choroid Plexus/metabolism , Animals , Apolipoprotein A-I/blood , Apolipoprotein A-I/cerebrospinal fluid , Apolipoprotein A-I/genetics , Biological Transport , Capillary Permeability , Cells, Cultured , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Half-Life , Humans , Injections, Intravenous , Metabolic Clearance Rate , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Tissue Distribution
14.
J Control Release ; 191: 15-23, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25016969

ABSTRACT

L4F, an alpha helical peptide inspired by the lipid-binding domain of the ApoA1 protein, has potential applications in the reduction of inflammation involved with cardiovascular disease as well as an antioxidant effect that inhibits liver fibrosis. In addition to its biological activity, amphipathic peptides such as L4F are likely candidates to direct the molecular assembly of peptide nanostructures. Here we describe the stabilization of the amphipathic L4F peptide through fusion to a high molecular weight protein polymer. Comprised of multiple pentameric repeats, elastin-like polypeptides (ELPs) are biodegradable protein polymers inspired from the human gene for tropoelastin. Dynamic light scattering confirmed that the fusion peptide forms nanoparticles with a hydrodynamic radius of approximately 50nm, which is unexpectedly above that observed for the free ELP (~5.1nm). To further investigate their morphology, conventional and cryogenic transmission electron microscopy were used to reveal that they are unilamellar vesicles. On average, these vesicles are 49nm in radius with lamellae 8nm in thickness. To evaluate their therapeutic potential, the L4F nanoparticles were incubated with hepatic stellate cells. Stellate cell activation leads to hepatic fibrosis; furthermore, their activation is suppressed by anti-oxidant activity of ApoA1 mimetic peptides. Consistent with this observation, L4F nanoparticles were found to suppress hepatic stellate cell activation in vitro. To evaluate the in vivo potential for these nanostructures, their plasma pharmacokinetics were evaluated in rats. Despite the assembly of nanostructures, both free L4F and L4F nanoparticles exhibited similar half-lives of approximately 1h in plasma. This is the first study reporting the stabilization of peptide-based vesicles using ApoA1 mimetic peptides fused to a protein polymer; furthermore, this platform for peptide-vesicle assembly may have utility in the design of biodegradable nanostructures.


Subject(s)
Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Apolipoprotein A-I/chemistry , Drug Carriers , Peptide Fragments/chemistry , Tropoelastin/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/pharmacokinetics , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Apolipoprotein A-I/pharmacokinetics , Cells, Cultured , Chemistry, Pharmaceutical , Genetic Engineering , Half-Life , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Injections, Intravenous , Mice , Nanoparticles , Nanotechnology , Particle Size , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Protein Stability , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/chemistry , Technology, Pharmaceutical/methods , Tropoelastin/genetics , Tropoelastin/metabolism
15.
Mol Nutr Food Res ; 58(7): 1537-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24863413

ABSTRACT

SCOPE: It is well known that trans-fatty acids have proatherogenic properties while HDL has antiatherogenic activities in plasma. However, there has been no report on the effects of trans-fat on the functional and structural properties of HDL. METHODS AND RESULTS: To compare physiological properties, we synthesized reconstituted HDL (rHDL) containing stearic acid (18:0), oleic acid (18:1, cis), or elaidic acid (EA, 18:1, trans). An rHDL containing EA (EA-rHDL) showed loss of antioxidant ability and induced the highest uptake of oxidized LDL into human macrophages. EA-rHDL caused the strongest cellular senescence in human dermal fibroblast cells along with the highest production of inflammatory species in macrophages co-treated with fructose. Injection of EA-rHDL into zebrafish embryos resulted in acute embryonic toxicity with the lowest survivability. Consumption of trans-fat for 20 weeks resulted in remarkable hyperlipidemia, elevation of serum cholesteryl ester transfer protein activity, hepatic inflammation, and fatty liver changes. CONCLUSION: Incorporation of EA impaired the beneficial effects of rHDL against atherogenesis. In zebrafish, EA-rHDL resulted in acute embryonic toxicity, and consumption of EA caused remarkable hyperlipidemia, inflammation, and fatty liver changes.


Subject(s)
Fatty Liver/blood , Hyperlipidemias/blood , Lipoproteins, HDL/blood , Oleic Acid/administration & dosage , Oleic Acid/adverse effects , Animals , Antioxidants/pharmacology , Apolipoprotein A-I/blood , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/blood , Cells, Cultured , Cholesterol Ester Transfer Proteins/blood , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Fatty Liver/etiology , Fatty Liver/pathology , Humans , Hyperlipidemias/etiology , Hyperlipidemias/pathology , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Macrophages/metabolism , Oleic Acids , Reactive Oxygen Species , Stearic Acids/administration & dosage , Stearic Acids/adverse effects , Toxicity Tests, Acute , Trans Fatty Acids/administration & dosage , Trans Fatty Acids/adverse effects , Triglycerides/blood , Zebrafish/embryology
16.
Arterioscler Thromb Vasc Biol ; 33(9): 2202-11, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868939

ABSTRACT

OBJECTIVE: The ability of high-density lipoprotein (HDL) to remove cholesterol from atherosclerotic plaque is thought to underlie its inverse correlation with cardiovascular risk. Our objective was to produce and characterize a human apolipoprotein AI (apoA-I) product optimized to treat clinical atherosclerotic disease. APPROACH AND RESULTS: A new formulation of full length, plasma-derived human apoA-I termed CSL112 was designed to maximize the cholesterol efflux from cells and exhibit favorable pharmacological properties. CSL112 is a disc-shaped particle that strongly elevates cholesterol esterification and shows good pharmacokinetics in rabbits. Infusion of CSL112 into rabbits caused a strong and immediate increase in the ATP binding cassette transporter A1 (ABCA1)-dependent efflux capacity of plasma, an increase in plasma unesterified cholesterol and rapid subsequent cholesterol esterification. In the presence of human plasma, CSL112 was significantly more potent than native HDL at enhancing cholesterol efflux from macrophages, and the efflux elevation was predominantly via the ABCA1 transporter. Consistent with this observation, addition of CSL112 to plasma led to generation of high levels of HDL-VS, a favorable substrate for ABCA1. The lipid profile of plasma did not affect these behaviors. In studies with whole human blood, CSL112 reduced expression of intercellular adhesion molecule 1 and cytokine secretion, and as with cholesterol efflux, these activities were substantially greater than those of native HDL assayed in parallel. CONCLUSIONS: CSL112 has favorable pharmacological properties and strongly elevates the ability of plasma to withdraw cholesterol from cells. Preferential elevation of ABCA1-dependent efflux may target atherosclerotic plaque for cholesterol removal and this property makes CSL112 a promising candidate therapy for acute coronary syndrome.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Anticholesteremic Agents/pharmacology , Apolipoprotein A-I/pharmacology , Cholesterol, HDL/blood , Cholesterol/blood , Lipoproteins, HDL/pharmacology , Macrophages/drug effects , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/blood , Animals , Anti-Inflammatory Agents/pharmacology , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacokinetics , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/blood , Apolipoprotein A-I/pharmacokinetics , Biological Transport , Cell Line , Cholesterol Esters/blood , Cytokines/blood , Female , Humans , Inflammation Mediators/blood , Infusions, Intravenous , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/blood , Lipoproteins, HDL/pharmacokinetics , Macrophages/immunology , Macrophages/metabolism , Mice , Particle Size , Rabbits , Up-Regulation
17.
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
18.
J Drug Target ; 21(4): 367-74, 2013 May.
Article in English | MEDLINE | ID: mdl-23600747

ABSTRACT

High-density lipoprotein (HDL) particles can deliver cholesterol from peripheral tissues to the liver through apolipoprotein A1 (Apo A1), which specifically binds to the scavenger receptor class B type 1 (SR-B1) receptor on the surface of hepatocytes. Therefore, ApoA1 can be potentially used to target drugs to the liver. In this study, we successfully loaded doxorubicin hydrochloride (Dox or Dox-HCl), which is a hydrophilic drug used in a wide variety of clinical applications, into the core of reconstituted HDL (rHDL prepared by apoAI and egg phospholipids) to form a doxorubicin-HDL complex (rHDL-Dox). The MTT assays showed that rHDL-Dox particles also had higher cytotoxicity against several cells lines compared to free drug or Dox encapsulated into liposomes. A cellular uptake assay demonstrated that rHDL-Dox had higher absorption in SR-BI receptor positive liver cells. Importantly, in vivo experiments showed that rHDL-Dox can reduce tumor growth more effectively than liposomes. In addition, an in vitro hemolysis assay showed that rHDL-Dox caused only limited hemolysis in the case of high doses. Taken together, our findings indicate that rHDL is a safe and effective drug delivery system for targeting liver.


Subject(s)
Apolipoprotein A-I/administration & dosage , Carrier Proteins/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Liver/drug effects , Liver/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/pharmacokinetics , Carrier Proteins/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Hep G2 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/pharmacokinetics , Liposomes/administration & dosage , Liposomes/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude
19.
Int J Nanomedicine ; 7: 2805-16, 2012.
Article in English | MEDLINE | ID: mdl-22745543

ABSTRACT

BACKGROUND: Nanoparticles undergoing physicochemical changes to release enclosed drugs at acidic pH conditions are promising vehicles for antitumor drug delivery. Among the various drug carriers, high-density lipoprotein (HDL)-like nanoparticles have been shown to be beneficial for cancer chemotherapy, but have not yet been designed to be pH-responsive. METHODS AND RESULTS: In this study, we developed a pH-responsive HDL-like nanoparticle that selectively releases paclitaxel, a model antitumor drug, at acidic pH. While the well known HDL-like nanoparticle containing phospholipids, phosphatidylcholine, and apolipoprotein A-I, as well as paclitaxel (PTX-PL-NP) was structurally robust at a wide range of pH values (3.8-10.0), the paclitaxel nanoparticle that only contained paclitaxel and apoA-I selectively released paclitaxel into the medium at low pH. The paclitaxel nanoparticle was stable at physiological and basic pH values, and over a wide range of temperatures, which is a required feature for efficient cancer chemotherapy. The homogeneous assembly enabled high paclitaxel loading per nanoparticle, which was 62.2% (w/w). The molar ratio of apolipoprotein A-I and paclitaxel was 1:55, suggesting that a single nanoparticle contained approximately 110 paclitaxel particles in a spherical structure with a 9.2 nm diameter. Among the several reconstitution methods applied, simple dilution following sonication enhanced the reconstitution yield of soluble paclitaxel nanoparticles, which was 0.66. As a result of the pH responsiveness, the anticancer effect of paclitaxel nanoparticles was much more potent than free paclitaxel or PTX-PL-NP. CONCLUSION: The anticancer efficacy of both paclitaxel nanoparticles and PTX-PL-NP was dependent on the expression of scavenger receptor class B type I, while the killing efficacy of free paclitaxel was independent of this receptor. We speculate that the pH responsiveness of paclitaxel nanoparticles enabled efficient endosomal escape of paclitaxel before lysosomal break down. This is the first report on pH-responsive nanoparticles that do not contain any synthetic polymer.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Paclitaxel/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/pharmacokinetics , Apolipoprotein A-I/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Stability , Humans , Hydrogen-Ion Concentration , Molecular Conformation , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Particle Size , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
20.
Hepatology ; 53(6): 1864-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21425312

ABSTRACT

UNLABELLED: Interferon alpha (IFNα) is widely used for the treatment of viral hepatitis but substantial toxicity hampers its clinical use. In this work, we aimed at improving the efficacy of IFNα therapy by increasing the IFNα half-life and providing liver tropism. We selected apolipoprotein A-I (ApoA-I) as the stabilizing and targeting moiety. We generated plasmids encoding IFNα, albumin bound to IFNα (ALF), or IFNα linked to ApoA-I (IA) and mice were treated either by hydrodynamic administration of the plasmids or by injection of the corresponding recombinant proteins or high-density lipoproteins containing IA. The plasma half-life of IA was intermediate between IFNα and ALF. IA was targeted to the liver and induced higher hepatic expression of interferon-stimulated genes than IFNα or even ALF. IA exhibits stronger in vivo antiviral activity than IFNα and the hematologic cytopenic effects of IA are milder than those observed when using IFNα or ALF. In contrast to IFNα, IA does not cause activation-dependent cell death of lymphocytes in vitro. Accordingly, in vivo studies showed that IA boosts T-cell immune responses more efficiently than IFNα or ALF. The difference in immunostimulatory activity between IFNα and IA disappears in scavenger receptor class B type I (SR-BI) knockout mice, suggesting that crosstalk between SR-BI and IFNα receptor is essential for enhanced induction of cytotoxic T cells by IA. CONCLUSION: Anchoring IFNα to ApoA-I prolongs the half-life of IFNα and promotes targeting to the liver. Importantly, the fusion protein shows increased immunostimulatory properties and lower hematological toxicity.


Subject(s)
Apolipoprotein A-I/pharmacokinetics , Hepatitis C/immunology , Immunization , Interferon-alpha/pharmacokinetics , Liver/metabolism , Liver/virology , Recombinant Fusion Proteins/pharmacokinetics , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Apolipoprotein A-I/pharmacology , Apolipoprotein A-I/therapeutic use , Apoptosis/drug effects , CD36 Antigens/genetics , CD36 Antigens/physiology , Cell Line , Cell Line, Tumor , Disease Models, Animal , Female , Half-Life , Hepacivirus/drug effects , Hepacivirus/isolation & purification , Hepatitis C/drug therapy , Hepatitis C/physiopathology , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Lymphocytes/drug effects , Lymphocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/physiology , Receptor, Interferon alpha-beta/physiology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...