Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Thromb Haemost ; 122(5): 808-817, 2022 May.
Article in English | MEDLINE | ID: mdl-34555861

ABSTRACT

The objective of this study was to assess the relationship between factor XI (FXI) deficiency and the risks of bleeding and cardiovascular (CV) events. We conducted a retrospective cohort study using data from Maccabi Healthcare Services (MHS). We identified adults with FXI deficiency (severe: <15%, partial: 15 to <50%, any deficiency: <50%) that had been tested for FXI between 2007 and 2018 and matched to patients from the general MHS population. We estimated 10-year risks of outcomes using the Kaplan-Meier approach. Using Cox proportional hazards regression, we compared outcomes among patients with versus without FXI deficiency. Less than 10% of patients tested for FXI activity had activity levels <50% (mean age: 39 years; 72.2% females). Compared with the general population, patients with any FXI deficiency were at higher risk of severe bleeding (adjusted hazard ratio [aHR]: 2.56, 95% confidence interval [CI]: 1.13-5.81; 10-year risk: 1.90%, 95% CI: 0.50-3.20% vs. 0.90%, 95% CI: 0.50-1.30%) and clinically relevant nonsevere bleeding (CRNSB) (aHR: 1.45, 95% CI: 1.08-1.97; 10-year risk: 11.60%, 95% CI: 8.30-14.80% vs. 9.20%, 95% CI: 8.00-10.40%). Severe FXI deficiency was associated with a greater risk of CRNSB. While few CV events (N = 2) and venous thromboembolisms (VTE) (N = 1) were observed in the FXI overall deficient group, there was a nonsignificant negative association between any FXI deficiency and CV events (aHR: 0.55; 95% CI: 0.13-2.36) and VTEs (aHR: 0.45; 95% CI: 0.06-3.47). Overall FXI deficiency was associated with an increased risk of severe bleeding and CRNSB. Further research is warranted to explore the lower risk of CV and VTE among patients with FXI deficiency compared with the general population.


Subject(s)
Factor XI Deficiency , Venous Thromboembolism , Venous Thrombosis , Adult , Factor XI , Factor XI Deficiency/complications , Female , Hemorrhage/complications , Hemorrhage/epidemiology , Humans , Male , Retrospective Studies , Venous Thromboembolism/complications , Venous Thromboembolism/diagnosis , Venous Thromboembolism/epidemiology , Venous Thrombosis/complications
2.
Nat Immunol ; 22(2): 128-139, 2021 02.
Article in English | MEDLINE | ID: mdl-33398182

ABSTRACT

Complement hyperactivation, angiopathic thrombosis and protein-losing enteropathy (CHAPLE disease) is a lethal disease caused by genetic loss of the complement regulatory protein CD55, leading to overactivation of complement and innate immunity together with immunodeficiency due to immunoglobulin wasting in the intestine. We report in vivo human data accumulated using the complement C5 inhibitor eculizumab for the medical treatment of patients with CHAPLE disease. We observed cessation of gastrointestinal pathology together with restoration of normal immunity and metabolism. We found that patients rapidly renormalized immunoglobulin concentrations and other serum proteins as revealed by aptamer profiling, re-established a healthy gut microbiome, discontinued immunoglobulin replacement and other treatments and exhibited catch-up growth. Thus, we show that blockade of C5 by eculizumab effectively re-establishes regulation of the innate immune complement system to substantially reduce the pathophysiological manifestations of CD55 deficiency in humans.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/therapeutic use , Energy Metabolism/drug effects , Hypoproteinemia/drug therapy , Immunity, Innate/drug effects , Protein-Losing Enteropathies/drug therapy , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Biomarkers/blood , CD55 Antigens/deficiency , CD55 Antigens/genetics , Complement C5/metabolism , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Genetic Predisposition to Disease , Humans , Hypoproteinemia/genetics , Hypoproteinemia/immunology , Hypoproteinemia/metabolism , Mutation , Phenotype , Protein-Losing Enteropathies/genetics , Protein-Losing Enteropathies/immunology , Protein-Losing Enteropathies/metabolism , Treatment Outcome
3.
Pharm Stat ; 17(3): 278-289, 2018 05.
Article in English | MEDLINE | ID: mdl-29624854

ABSTRACT

The trimmed mean is a method of dealing with patient dropout in clinical trials that considers early discontinuation of treatment a bad outcome rather than leading to missing data. The present investigation is the first comprehensive assessment of the approach across a broad set of simulated clinical trial scenarios. In the trimmed mean approach, all patients who discontinue treatment prior to the primary endpoint are excluded from analysis by trimming an equal percentage of bad outcomes from each treatment arm. The untrimmed values are used to calculated means or mean changes. An explicit intent of trimming is to favor the group with lower dropout because having more completers is a beneficial effect of the drug, or conversely, higher dropout is a bad effect. In the simulation study, difference between treatments estimated from trimmed means was greater than the corresponding effects estimated from untrimmed means when dropout favored the experimental group, and vice versa. The trimmed mean estimates a unique estimand. Therefore, comparisons with other methods are difficult to interpret and the utility of the trimmed mean hinges on the reasonableness of its assumptions: dropout is an equally bad outcome in all patients, and adherence decisions in the trial are sufficiently similar to clinical practice in order to generalize the results. Trimming might be applicable to other inter-current events such as switching to or adding rescue medicine. Given the well-known biases in some methods that estimate effectiveness, such as baseline observation carried forward and non-responder imputation, the trimmed mean may be a useful alternative when its assumptions are justifiable.


Subject(s)
Clinical Trials, Phase III as Topic/statistics & numerical data , Models, Statistical , Patient Dropouts/statistics & numerical data , Clinical Trials, Phase III as Topic/methods , Humans , Treatment Outcome
4.
Pharmacotherapy ; 36(7): 749-56, 2016 07.
Article in English | MEDLINE | ID: mdl-27284735

ABSTRACT

STUDY OBJECTIVE: To examine the effect of increased gastric pH on exposure to evacetrapib, a cholesteryl ester transfer protein inhibitor evaluated for the treatment of atherosclerotic heart disease. DESIGN: Open-label, two-treatment, two-period, fixed-sequence crossover study. SETTING: Clinical research unit. SUBJECTS: Thirty-four healthy subjects. INTERVENTION: In period 1, subjects received a single oral dose of evacetrapib 130 mg on day 1, followed by 7 days of analysis for evacetrapib plasma concentrations. In period 2, subjects received a once/day oral dose of omeprazole 40 mg on days 8-20, with a single oral dose of evacetrapib 130 mg administered 2 hours after the omeprazole dose on day 14, followed by 7 days of pharmacokinetic sampling. Subjects were discharged on day 21 and returned for a follow-up visit at least 14 days after the last dose of evacetrapib in period 2. Gastric pH was measured before subjects received each evacetrapib dose. MEASUREMENTS AND MAIN RESULTS: Noncompartmental pharmacokinetic parameters were estimated from plasma concentration-time data and compared between periods 1 and 2. Geometric mean ratios with 90% confidence intervals (CIs) were reported. Safety and tolerability were also assessed. The mean age of the 34 subjects was 40.9 years; mean body mass index was 27.2 kg/m(2) . Omeprazole treatment increased mean gastric pH across all subjects by 2.80 and increased evacetrapib area under the concentration versus time curve from time zero extrapolated to infinity (AUC0-∞ ) and maximum observed drug concentration (Cmax ) by 15% (90% CI -2 to 35) and 30% (90% CI 3-63), respectively. For both parameters, the upper bound of the 90% CI of the ratio of geometric least-squares means exceeded 1.25 but was less than 2, indicating a weak interaction. To assess the effect of gastric pH on subjects who responded best to omeprazole treatment, the analyses were repeated to include only the 22 subjects whose predose gastric pH was 3.0 or lower in period 1 and 4.0 or higher in period 2. In this subpopulation, mean gastric pH increased by 4.15 during omeprazole treatment, and evacetrapib AUC0-∞ and Cmax increased by 22% (90% CI 4-42) and 35% (90% CI 1-80), respectively. Despite the small mathematical differences between the analyses, the overall effect in both was a minimal increase in evacetrapib exposure. Of 35 adverse events reported during the study, 4 (11.4%) were considered to be treatment-related, and most were mild in severity. CONCLUSION: The impact of increased gastric pH on evacetrapib pharmacokinetics would not be expected to be clinically relevant. The magnitude of change in pH did not affect the degree of the interaction.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Benzodiazepines/pharmacokinetics , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Gastric Acidity Determination , Adult , Benzodiazepines/adverse effects , Cross-Over Studies , Female , Humans , Male , Middle Aged
5.
J Clin Lipidol ; 10(3): 519-527.e4, 2016.
Article in English | MEDLINE | ID: mdl-27206939

ABSTRACT

BACKGROUND: Potent CETP inhibitors reduce plasma concentrations of atherogenic lipoprotein biomarkers of cardiovascular risk. OBJECTIVES: To evaluate the effects of the cholesteryl ester transfer protein (CETP) inhibitor evacetrapib, as monotherapy or with statins, on atherogenic apolipoprotein B (apoB)-containing lipoproteins in mildly hypercholesterolemic patients. METHODS: VLDL and LDL particle concentrations and sizes (using nuclear magnetic resonance spectroscopy) and lipoprotein(a) concentration (using nephelometry) were measured at baseline and week 12 in a placebo-controlled trial of 393 patients treated with evacetrapib as monotherapy (30 mg/d, 100 mg/d, or 500 mg/d) or in combination with statins (100 mg plus simvastatin 40 mg/d, atorvastatin 20 mg/d, or rosuvastatin 10 mg/d; Clinicaltrials.gov Identifier: NCT01105975). RESULTS: Evacetrapib monotherapy resulted in significant placebo-adjusted dose-dependent decreases from baseline in Lp(a) (up to -40% with evacetrapib 500 mg), total LDL particle (LDL-P) (up to -54%), and small LDL particle (sLDL) (up to -95%) concentrations. Compared to statin alone, coadministration of evacetrapib and statins also resulted in significant reduction from baseline in Lp(a) (-31%), LDL-P (-22%), and sLDL (-60%) concentrations. The percentage of patients with concentrations above optimal concentrations for LDL-P (>1000 nmol/L) and sLDL (>600 nmol/L) decreased from 88% and 55% at baseline, respectively, to 20% and 12% at week 12, for patients treated with evacetrapib plus statins. Evacetrapib, alone or with statins, significantly increased LDL-P size. CONCLUSIONS: Evacetrapib, as monotherapy or with statins, significantly reduces the concentrations of atherogenic apoB-containing lipoproteins, including Lp(a), LDL-P, and sLDL.


Subject(s)
Benzodiazepines/pharmacology , Cholesterol, LDL/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Lipoprotein(a)/blood , Particle Size , Benzodiazepines/therapeutic use , Cholesterol, LDL/chemistry , Drug Interactions , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Middle Aged
6.
Eur Heart J ; 37(17): 1360-9, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26757788

ABSTRACT

AIMS: The objective of this study was to evaluate the efficacy, safety, and tolerability of LY3015014 (LY), a neutralizing antibody of proprotein convertase subtilisin/kexin type 9 (PCSK9), administered every 4 or 8 weeks in patients with primary hypercholesterolaemia, when added to a background of standard-of-care lipid-lowering therapy, including statins. METHODS AND RESULTS: Double-blind, placebo-controlled trial randomized 527 patients with primary hypercholesterolaemia from June 2013 to January 2014 at 61 community and academic centres in North America, Europe, and Japan. Patients were randomized to subcutaneous injections of LY 20, 120, or 300 mg every 4 weeks (Q4W); 100 or 300 mg every 8 weeks (Q8W) alternating with placebo Q4W; or placebo Q4W. The primary endpoint was percentage change from baseline in low-density lipoprotein cholesterol (LDL-C) by beta quantification at Week 16. The mean baseline LDL-C by beta quantification was 136.3 (SD, 45.0)mg/dL. LY3015014 dose-dependently decreased LDL-C, with a maximal reduction of 50.5% with 300 mg LY Q4W and 37.1% with 300 mg LY Q8W compared with a 7.6% increase with placebo maintained at the end of the dosing interval. There were no treatment-related serious adverse events (AEs). The most common AE terms (>10% of any treatment group) reported more frequently with LY compared with placebo were injection site (IS) pain and IS erythema. No liver or muscle safety issues emerged. CONCLUSIONS: LY3015014 dosed every 4 or 8 weeks, resulted in robust and durable reductions in LDL-C. No clinically relevant safety issues emerged with the administration of LY. The long-term effects on cardiovascular outcomes require further investigation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/adverse effects , Anticholesteremic Agents , Cholesterol, LDL , Double-Blind Method , Europe , Humans , Japan , Proprotein Convertase 9 , Proprotein Convertases , Serine Endopeptidases , Subtilisins , Treatment Outcome
7.
J Am Coll Cardiol ; 66(20): 2201-2210, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26564598

ABSTRACT

BACKGROUND: Potent cholesteryl ester transfer protein (CETP) inhibitors have been shown to substantially increase high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I levels as monotherapy and combined with statins. However, data on the effects of this class of drugs on macrophage cholesterol efflux capacity (CEC), a functional assay that characterizes a key step in the process of reverse cholesterol transport, are limited. OBJECTIVES: This study assessed the impact of evacetrapib, statins, or combination therapy on CEC. METHODS: We analyzed samples from 377 subjects with elevated low-density lipoprotein cholesterol (LDL-C) or low HDL-C levels who were enrolled in a phase 2 trial of evacetrapib. Percent changes from baseline in CEC (total, non-ABCA1-, and ABCA1-specific) and HDL subpopulations were evaluated after 12 weeks of treatment with placebo, statin monotherapy, evacetrapib monotherapy, or evacetrapib combined with statins. Pre-beta-1 HDL levels were quantified by immunofixation and nondenaturing 2-dimensional gel electrophoresis (2DGE). RESULTS: Relative to placebo, evacetrapib monotherapy increased dose-dependent total and non-ABCA1-specific CEC up to 34% and 47%, respectively. Evacetrapib monotherapy also increased ABCA1-specific CEC up to 26%. Relative to statin monotherapy, evacetrapib with statins also increased total, non-ABCA1-, and ABCA1-specific CEC by 21%, 27%, and 15%, respectively. In contrast, rosuvastatin and simvastatin significantly reduced total and ABCA1-specific CEC, whereas atorvastatin had no significant effect. Consistent with ABCA1-specific CEC, evacetrapib monotherapy and evacetrapib combined with statins significantly increased pre-beta-1 HDL levels as measured by either method. CONCLUSIONS: Evacetrapib, as monotherapy and combined with statins, not only increased total CEC, but also increased ABCA1-specific CEC and pre-beta-1 HDL. The mechanisms by which potent CETP inhibition increases ABCA1-specific CEC and pre-beta-1 HDL require further study. (A Study of LY2484595 in Patients With High LDL-C or Low HDL-C; NCT01105975).


Subject(s)
Anticholesteremic Agents/therapeutic use , Benzodiazepines/therapeutic use , Cholesterol/blood , Dyslipidemias/drug therapy , High-Density Lipoproteins, Pre-beta/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , ATP Binding Cassette Transporter 1/metabolism , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Double-Blind Method , Drug Therapy, Combination , Dyslipidemias/blood , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Middle Aged
8.
Pharmacol Res Perspect ; 3(5): e00179, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26516590

ABSTRACT

Evacetrapib is an investigational cholesteryl ester transfer protein inhibitor (CETPi) for reduction of risk of major adverse cardiovascular events in patients with high-risk vascular disease. Understanding evacetrapib disposition, metabolism, and the potential for drug-drug interactions (DDI) may help guide prescribing recommendations. In vitro, evacetrapib metabolism was investigated with a panel of human recombinant cytochromes P450 (CYP). The disposition, metabolism, and excretion of evacetrapib following a single 100-mg oral dose of (14)C-evacetrapib were determined in healthy subjects, and the pharmacokinetics of evacetrapib were evaluated in the presence of strong CYP3A or CYP2C8 inhibitors. In vitro, CYP3A was responsible for about 90% of evacetrapib's CYP-associated clearance, while CYP2C8 accounted for about 10%. In the clinical disposition study, only evacetrapib and two minor metabolites circulated in plasma. Evacetrapib metabolism was extensive. A mean of 93.1% and 2.30% of the dose was excreted in feces and urine, respectively. In clinical DDI studies, the ratios of geometric least squares means for evacetrapib with/without the CYP3A inhibitor ketoconazole were 2.37 for area under the curve (AUC)(0-∞) and 1.94 for C max. There was no significant difference in evacetrapib AUC(0-τ) or C max with/without the CYP2C8 inhibitor gemfibrozil, with ratios of 0.996 and 1.02, respectively. Although in vitro results indicated that both CYP3A and CYP2C8 metabolized evacetrapib, clinical studies confirmed that evacetrapib is primarily metabolized by CYP3A. However, given the modest increase in evacetrapib exposure and robust clinical safety profile to date, there is a low likelihood of clinically relevant DDI with concomitant use of strong CYP3A or CYP2C8 inhibitors.

9.
Br J Clin Pharmacol ; 80(6): 1388-98, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26264702

ABSTRACT

AIMS: Evacetrapib is a cholesteryl ester transfer protein (CETP) inhibitor under development for reducing cardiovascular events in patients with high risk vascular disease. CETP inhibitors are likely to be utilized as 'add-on' therapy to statins in patients receiving concomitant medications, so the potential for evacetrapib to cause clinically important drug-drug interactions (DDIs) with cytochromes P450 (CYP) was evaluated. METHODS: The DDI potential of evacetrapib was investigated in vitro, followed by predictions to determine clinical relevance. Potential DDIs with possible clinical implications were then investigated in the clinic. RESULTS: In vitro, evacetrapib inhibited all of the major CYPs, with inhibition constants (K(i)) ranging from 0.57 µM (CYP2C9) to 7.6 µM (CYP2C19). Evacetrapib was a time-dependent inhibitor and inducer of CYP3A. The effects of evacetrapib on CYP3A and CYP2C9 were assessed in a phase 1 study using midazolam and tolbutamide as probe substrates, respectively. After 14 days of daily dosing with evacetrapib (100 or 300 mg), midazolam exposures (AUC) changed by factors (95% CI) of 1.19 (1.06, 1.33) and 1.44 (1.28, 1.62), respectively. Tolbutamide exposures (AUC) changed by factors of 0.85 (0.77, 0.94) and 1.06 (0.95, 1.18), respectively. In a phase 2 study, evacetrapib 100 mg had minimal impact on AUC of co-administered simvastatin vs. simvastatin alone with a ratio of 1.25 (1.03, 1.53) at steady-state, with no differences in reported hepatic or muscular adverse events. CONCLUSIONS: Taken together, the extent of CYP-mediated DDI with the potential clinical dose of evacetrapib is weak and clinically important DDIs are not expected to occur in patients taking concomitant medications.


Subject(s)
Anticholesteremic Agents/pharmacology , Benzodiazepines/pharmacology , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cells, Cultured , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Double-Blind Method , Drug Interactions , Humans , Microsomes, Liver/enzymology , Midazolam/pharmacokinetics , Simvastatin/pharmacokinetics , Tolbutamide/pharmacokinetics
10.
Eur Heart J ; 36(43): 3020-2, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26112886

ABSTRACT

AIMS: Fibrate medications weakly stimulate the nuclear receptor peroxisome proliferator-activated receptor-α (PPAR-α) and are currently employed clinically in patients with dyslipidaemia. The potent and selective agonist of PPAR-α LY518674 is known to substantially increase apolipoprotein A-I (apoA-I) turnover without major impact on steady-state levels of apoA-I or high-density lipoprotein-cholesterol (HDL-C). We sought to determine whether therapy with a PPAR-α agonist impacts cholesterol efflux capacity, a marker of HDL function. METHODS AND RESULTS: Cholesterol efflux capacity was measured at baseline and after 8 weeks of therapy in a randomized, placebo-controlled trial involving participants with metabolic syndrome treated with either LY518674 100 µg daily (n = 13) or placebo (n = 15). Efflux capacity assessment was quantified using a previously validated ex vivo assay that measures the ability of apolipoprotein-B depleted plasma to mobilize cholesterol from macrophages. LY518674 led to a 15.7% increase from baseline (95% CI 3.3-28.1%; P = 0.02, P vs. placebo = 0.01) in efflux capacity. The change in apoA-I production rate in the active treatment arm was strongly linked to change in cholesterol efflux capacity (r = 0.67, P = 0.01). CONCLUSIONS: Potent stimulation of PPAR-α leads to accelerated turnover of apoA-I and an increase in cholesterol efflux capacity in metabolic syndrome patients despite no change in HDL-C or apoA-I levels. This finding reinforces the notion that changes in HDL-C levels may poorly predict impact on functionality and thus has implications for ongoing pharmacologic efforts to enhance apoA-I metabolism.


Subject(s)
Cholesterol, HDL/metabolism , Metabolic Syndrome/drug therapy , PPAR alpha/antagonists & inhibitors , Propionates/therapeutic use , Triazoles/therapeutic use , Apolipoprotein A-I/metabolism , Apolipoproteins B/metabolism , Female , Humans , Male , Middle Aged
11.
J Pharm Pharmacol ; 66(11): 1576-85, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24961753

ABSTRACT

OBJECTIVES: We investigated the safety, tolerability, pharmacokinetics and pharmacodynamics of evacetrapib. METHODS: Healthy volunteers received multiple daily doses of evacetrapib (10-600 mg) administered for up to 15 days in a placebo-controlled study. KEY FINDINGS: Mean peak plasma concentrations of evacetrapib occurred at 4-6 h and terminal half-life ranged 24-44 h. Steady state was achieved at approximately 10 days; all subjects had undetectable levels of evacetrapib 3 weeks after their last dose. The trough inhibition of cholesteryl ester transfer protein (CETP) activity was 65 and 84% at 100 and 300 mg, respectively. At the highest dose (600 mg), evacetrapib significantly inhibited CETP activity (91%), increased HDL-C (87%) and apo AI (42%), and decreased LDL-C (29%) and apo B (26%) relative to placebo. For the highest dose tested, levels of evacetrapib, CETP activity, CETP mass, HDL-C and LDL-C returned to levels at or near baseline after a 2-week washout period. Evacetrapib at the highest dose tested did not produce any significant effect on 24-h ambulatory systolic or diastolic blood pressure. CONCLUSIONS: Multiple doses of evacetrapib potently inhibited CETP activity, leading to substantial elevations in HDL-C and lowering of LDL-C. Evacetrapib was devoid of clinically relevant effects on blood pressure and mineralocorticoid levels.


Subject(s)
Apolipoprotein A-I/blood , Apolipoproteins B/blood , Benzodiazepines/pharmacology , Blood Pressure/drug effects , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Adult , Anticholesteremic Agents/pharmacology , Blood Pressure Monitoring, Ambulatory , Double-Blind Method , Female , Humans , Lipoproteins/blood , Male , Middle Aged , Reference Values , Young Adult
12.
JAMA ; 306(19): 2099-109, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22089718

ABSTRACT

CONTEXT: Interest remains high in cholesteryl ester transfer protein (CETP) inhibitors as cardioprotective agents. Few studies have documented the efficacy and safety of CETP inhibitors in combination with commonly used statins. OBJECTIVE: To examine the biochemical effects, safety, and tolerability of evacetrapib, as monotherapy and in combination with statins, in patients with dyslipidemia. DESIGN, SETTING, AND PARTICIPANTS: Randomized controlled trial conducted among 398 patients with elevated low-density lipoprotein cholesterol (LDL-C) or low high-density lipoprotein cholesterol (HDL-C) levels from April 2010 to January 2011 at community and academic centers in the United States and Europe. INTERVENTIONS: Following dietary lead-in, patients were randomly assigned to receive placebo (n = 38); evacetrapib monotherapy, 30 mg/d (n = 40), 100 mg/d (n = 39), or 500 mg/d (n = 42); or statin therapy (n = 239) (simvastatin, 40 mg/d; atorvastatin, 20 mg/d; or rosuvastatin, 10 mg/d) with or without evacetrapib, 100 mg/d, for 12 weeks. MAIN OUTCOME MEASURES: The co-primary end points were percentage changes from baseline in HDL-C and LDL-C after 12 weeks of treatment. RESULTS: The mean baseline HDL-C level was 55.1 (SD, 15.3) mg/dL and the mean baseline LDL-C level was 144.3 (SD, 26.6) mg/dL. As monotherapy, evacetrapib produced dose-dependent increases in HDL-C of 30.0 to 66.0 mg/dL (53.6% to 128.8%) compared with a decrease with placebo of -0.7 mg/dL (-3.0%; P < .001 for all compared with placebo) and decreases in LDL-C of -20.5 to -51.4 mg/dL (-13.6% to -35.9%) compared with an increase with placebo of 7.2 mg/dL (3.9%; P < .001 for all compared with placebo). In combination with statin therapy, evacetrapib, 100 mg/d, produced increases in HDL-C of 42.1 to 50.5 mg/dL (78.5% to 88.5%; P < .001 for all compared with statin monotherapy) and decreases in LDL-C of -67.1 to -75.8 mg/dL (-11.2% to -13.9%; P < .001 for all compared with statin monotherapy). Compared with evacetrapib monotherapy, the combination of statins and evacetrapib resulted in greater reductions in LDL-C (P <.001) but no greater increase in HDL-C (P =.39). Although the study was underpowered, no adverse effects were observed. CONCLUSIONS: Compared with placebo or statin monotherapy, evacetrapib as monotherapy or in combination with statins increased HDL-C levels and decreased LDL-C levels. The effects on cardiovascular outcomes require further investigation. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01105975.


Subject(s)
Benzodiazepines/administration & dosage , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol, HDL/drug effects , Cholesterol, LDL/drug effects , Dyslipidemias/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Aged , Atorvastatin , Benzodiazepines/pharmacology , Cardiovascular Diseases/prevention & control , Double-Blind Method , Drug Therapy, Combination , Female , Fluorobenzenes/administration & dosage , Heptanoic Acids/administration & dosage , Humans , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Rosuvastatin Calcium , Simvastatin/administration & dosage , Sulfonamides/administration & dosage , Treatment Outcome
13.
Arterioscler Thromb Vasc Biol ; 29(1): 140-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18988892

ABSTRACT

OBJECTIVE: The study of PPAR-alpha activation on apoA-I production in humans has been limited to fibrates, relatively weak PPAR-alpha agonists that may have other molecular effects. We sought to determine the effect of a potent and highly specific PPAR-alpha agonist, LY518674, on apoA-I, apoA-II, and apoB-100 kinetics in humans with metabolic syndrome and low levels of HDL cholesterol (C). METHODS AND RESULTS: Subjects were randomized to receive LY518674 (100 microg) once daily (n=13) or placebo (n=15) for 8 weeks. Subjects underwent a kinetic study using a deuterated leucine tracer to measure apolipoprotein production and fractional catabolic rates (FCR) at baseline and after treatment. LY518674 significantly reduced VLDL-C (-38%, P=0.002) and triglyceride (-23%, P=0.002) levels whereas LDL-C and HDL-C levels were unchanged. LY518674 significantly reduced VLDL apoB-100 (-12%, P=0.01) levels, attributable to an increased VLDL apoB-100 FCR with no change in VLDL apoB-100 production. IDL and LDL apoB-100 kinetics were unchanged. LY518674 significantly increased the apoA-I production rate by 31% (P<0.0001), but this was accompanied by a 33% increase in the apoA-I FCR (P=0.002), resulting in no change in plasma apoA-I. There was a 71% increase in the apoA-II production rate (P<0.0001) accompanied by a 25% increase in the FCR (P<0.0001), resulting in a significant increase in plasma apoA-II. CONCLUSIONS: Activation of PPAR-alpha with LY518674 (100 microg) in subjects with metabolic syndrome and low HDL-C increased the VLDL apoB-100 FCR consistent with enhanced lipolysis of plasma triglyceride. Significant increases in the apoA-I and apoA-II production rates were accompanied by increased FCRs resulting in no change in HDL-C levels. These data indicate a major effect of LY518674 on the production and clearance of apoA-I and HDL despite no change in the plasma concentration. The effect of these changes on reverse cholesterol transport remains to be determined.


Subject(s)
Apolipoprotein A-I/blood , Metabolic Syndrome/blood , PPAR alpha/agonists , Propionates/pharmacology , Triazoles/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Apolipoprotein A-I/genetics , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Deuterium , Double-Blind Method , Female , Humans , Kinetics , Male , Middle Aged , Placebos , Triglycerides/blood , Young Adult
14.
Biom J ; 49(3): 365-77, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17623342

ABSTRACT

We review a Bayesian predictive approach for interim data monitoring and propose its application to interim sample size reestimation for clinical trials. Based on interim data, this approach predicts how the sample size of a clinical trial needs to be adjusted so as to claim a success at the conclusion of the trial with an expected probability. The method is compared with predictive power and conditional power approaches using clinical trial data. Advantages of this approach over the others are discussed.


Subject(s)
Clinical Trials as Topic/statistics & numerical data , Bayes Theorem , Clinical Trials, Phase II as Topic/statistics & numerical data , Data Interpretation, Statistical , Sample Size
15.
JAMA ; 297(12): 1362-73, 2007 Mar 28.
Article in English | MEDLINE | ID: mdl-17384435

ABSTRACT

CONTEXT: Fibrates are weak agonists of peroxisome proliferator-activated receptor alpha (PPAR-alpha). No trials have reported effects of more potent and selective agents. OBJECTIVES: To examine the safety and efficacy of LY518674, a PPAR-alpha agonist. DESIGN, SETTING, AND PARTICIPANTS: Two multicenter, randomized, double-blind, placebo-controlled trials: 1 in patients with elevated triglycerides and low HDL-C (atherogenic dyslipidemia), the other in patients with elevated LDL-C (hypercholesterolemia). Between August 2005 and August 2006, the dyslipidemia study randomized 309 patients at US centers; the hypercholesterolemia study, 304 patients. INTERVENTIONS: Dyslipidemia study: placebo, fenofibrate (200 mg), or LY518674 (10, 25, 50, or 100 microg) for 12 weeks. Hypercholesterolemia study: placebo or atorvastatin (10 or 40 mg) for 4 weeks, then placebo or LY518674 (10 or 50 microg) for 12 more weeks. MAIN OUTCOME MEASURES: Dyslipidemia study: percentage change in levels of HDL-C and triglycerides. Hypercholesterolemia study: percentage change in levels of LDL-C. RESULTS: Dyslipidemia study: LY518674 (25 mug) and fenofibrate increased HDL-C by 5.9 and 5.5 mg/dL (15.8% and 14.4%) (both P< or =.001 vs placebo, P = .79 between treatments). Higher LY518674 doses yielded smaller increases. LY518674 decreased triglycerides by 97.3 to 114.5 mg/dL (34.9% to 41.7%) but was similar to fenofibrate. LY518674 produced a dose-dependent increase in LDL-C, reaching 20.4 mg/dL (19.5%) for the 100-mug dose vs 0.3 mg/dL (2.3%) for fenofibrate (P< or =.01). Fenofibrate and LY518674 (50 microg and 100 microg) increased serum creatinine (P< or =.001 vs placebo), with 38% and 37.3% of patients exceeding the normal range. Fenofibrate, but not LY518674, increased creatine phosphokinase (P = .004 vs placebo). Hypercholesterolemia study: LY518674 (10 mug or 50 microg) decreased LDL-C by 21.4 to 26.0 mg/dL (13.2%-15.8%) and triglycerides approximately 37% for both doses, and increased HDL-C by 6.3 to 6.7 mg/dL (12.5%-15.0%). When added to atorvastatin, LY518674 changed HDL-C by -0.7 to 6.2 mg/dL (-0.6% to 11.9%) and significantly decreased triglycerides but had no additional effect on LDL-C. CONCLUSIONS: In patients with dyslipidemia, LY518674 and fenofibrate decreased triglycerides and increased HDL-C but also increased serum creatinine. LY518674, but not fenofibrate, increased LDL-C. In those with hypercholesterolemia, LY518674 reduced triglycerides and increased HDL-C, but did not further reduce LDL-C in combination with atorvastatin. Fenofibrate and LY518674 both raised safety concerns. TRIAL REGISTRATION: clinicaltrials.gov Identifiers: NCT00133380 and NCT00116519


Subject(s)
Dyslipidemias/drug therapy , Hypercholesterolemia/drug therapy , Hypolipidemic Agents/therapeutic use , PPAR alpha/agonists , Propionates/therapeutic use , Triazoles/therapeutic use , Atorvastatin , Double-Blind Method , Dyslipidemias/blood , Female , Fenofibrate/therapeutic use , Heptanoic Acids/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Lipids/blood , Male , Middle Aged , Pyrroles/therapeutic use
16.
Stat Med ; 25(13): 2178-95, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16007570

ABSTRACT

This paper reviews Bayesian strategies for monitoring clinical trial data. It focuses on a Bayesian stochastic curtailment method based on the predictive probability of observing a clinically significant outcome at the scheduled end of the study given the observed data. The proposed method is applied to derive efficacy and futility stopping rules in clinical trials with continuous, normally distributed and binary endpoints. The sensitivity of the resulting stopping rules to the choice of prior distributions is examined and guidelines for choosing a prior distribution of the treatment effect are discussed. The Bayesian predictive approach is compared to the frequentist (conditional power) and mixed Bayesian-frequentist (predictive power) approaches. The interim monitoring strategies discussed in the paper are illustrated using examples from a small proof-of-concept study and a large mortality trial.


Subject(s)
Bayes Theorem , Clinical Trials as Topic/methods , Data Interpretation, Statistical , Anti-Anxiety Agents/therapeutic use , Anti-Bacterial Agents/therapeutic use , Anxiety Disorders/drug therapy , Humans , Sepsis/drug therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...