Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Clin Pharmacol Drug Dev ; 10(2): 131-143, 2021 02.
Article in English | MEDLINE | ID: mdl-32255549

ABSTRACT

MW01-6-189WH (MW189) is a novel central nervous system-penetrant small-molecule drug candidate that selectively attenuates stressor-induced proinflammatory cytokine overproduction and is efficacious in intracerebral hemorrhage and traumatic brain injury animal models. We report first-in-human, randomized, double-blind, placebo-controlled phase 1 studies to evaluate the safety, tolerability, and pharmacokinetics (PK) of single and multiple ascending intravenous doses of MW189 in healthy adult volunteers. MW189 was safe and well tolerated in single and multiple doses up to 0.25 mg/kg, with no clinically significant concerns. The most common drug-related treatment-emergent adverse event was infusion-site reactions, likely related to drug solution acidity. No clinically concerning changes were seen in vital signs, electrocardiograms, physical or neurological examinations, or safety laboratory results. PK analysis showed dose-proportional increases in plasma concentrations of MW189 after single or multiple doses, with approximately linear kinetics and no significant drug accumulation. Steady state was achieved by dose 3 for all dosing cohorts. A pilot pharmacodynamic study administering low-dose endotoxin to induce a systemic inflammatory response was done to evaluate the effects of a single intravenous dose of MW189 on plasma cytokine levels. MW189 treatment resulted in lower levels of the proinflammatory cytokine TNF-α and higher levels of the anti-inflammatory cytokine IL-10 compared with placebo treatment. The outcomes are consistent with the pharmacological mechanism of MW189. Overall, the safety profile, PK properties, and pharmacodynamic effect support further development of MW189 for patients with acute brain injury.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Inflammation/drug therapy , Piperazines/administration & dosage , Pyridazines/administration & dosage , Pyridines/administration & dosage , Adult , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Pilot Projects , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyridazines/adverse effects , Pyridazines/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Young Adult
2.
Inflammation ; 44(1): 270-277, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32886268

ABSTRACT

Plasma gelsolin (pGSN) is the secreted isoform of an intracellular actin remodeling protein found in high concentrations in human plasma. Clinical studies demonstrate reduced pGSN concentrations in several disease states, including severe trauma, burns, and sepsis. Markedly decreased pGSN concentrations in these conditions precede and predict adverse clinical outcomes. In this study, we measured pGSN in patients with chronic granulomatous disease (CGD), a primary immunodeficiency characterized by recurrent infections and dysregulated inflammation. pGSN was quantified using a sandwich ELISA in plasma from healthy volunteers, clinically stable CGD patients, and X-linked CGD carriers and in sera from 12 CGD patients undergoing bone marrow transplantation. pGSN was also quantified in healthy volunteers challenged with intravenous endotoxin. pGSN concentrations were lower in CGD patients without active infection or systemic inflammation compared with healthy control subjects. In CGD patients undergoing bone marrow transplantation, pGSN concentrations increased significantly following successful transplant. X-linked carriers of CGD had normal pGSN. Despite reduction of pGSN in CGD patients, we did not detect significant changes in pGSN over 24 h following challenge of healthy volunteers with intravenous endotoxin (4 ng/kg) that elicited a febrile response. We describe, for the first time, significantly lower pGSN in clinically stable patients with CGD compared with age- and sex-matched healthy volunteers. Low pGSN levels in CGD patients significantly increased following bone marrow transplantation. X-linked carriers of CGD had normal pGSN. In healthy volunteers challenged with intravenous endotoxin, pGSN is not an acute phase reactant.


Subject(s)
Gelsolin/blood , Granulomatous Disease, Chronic/blood , Granulomatous Disease, Chronic/diagnosis , Adolescent , Adult , Biomarkers/blood , Bone Marrow Transplantation/methods , Cohort Studies , Endotoxins/toxicity , Female , Fever/blood , Fever/chemically induced , Fever/therapy , Humans , Male , Middle Aged , Young Adult
5.
J Clin Pharmacol ; 59(5): 717-730, 2019 05.
Article in English | MEDLINE | ID: mdl-30703258

ABSTRACT

Nitroxyl (HNO) is a reactive nitrogen molecule that has potential therapeutic benefits for patients with acute heart failure. The results of the first-in-human study for BMS-986231, a novel HNO donor, are reported. The aim of this sequential cohort study was to evaluate the safety, tolerability, and pharmacokinetic profile of BMS-986231 after 24- and 48-hour intravenous infusions in healthy volunteers. Eighty subjects were randomized and dosed. Seven cohorts (stratum A) received BMS-986231 0.1, 0.33, 1, 3, 5, 10, and 15 µg/kg/min or placebo, infused over 24 hours. An additional cohort (stratum B) received 10 µg/kg/min or placebo, infused over 48 hours. Adverse events (AEs) were reported for 30 days after completion of infusion. Blood/urine samples were collected at regular intervals; other parameters (blood pressure, heart rate/rhythm, cardiac index) were also assessed. Headaches were the most commonly reported drug-related AE (48%) in those who received BMS-986231, although their severity was reduced by hydration. No other significant drug-related AEs were noted. BMS-986231 was associated with dose-dependent and well-tolerated reductions in systolic and diastolic blood pressure versus baseline; cardiac index, as measured noninvasively, was increased. BMS-986231 had no clinically significant effect on heart rate/rhythm or laboratory parameters. Its mean elimination half-life was 0.7-2.5 hours. BMS-986231 was safe and well-tolerated for up to 24 hours (15 µg/kg/min) or 48 hours (10 µg/kg/min), with a favorable hemodynamic profile observed. Ongoing studies continue to evaluate the potential benefit of BMS-986231 in patients with acute heart failure.


Subject(s)
Nitric Oxide Donors/pharmacokinetics , Nitrogen Oxides/pharmacokinetics , Adult , Blood Pressure/drug effects , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Heart Failure/drug therapy , Heart Rate/drug effects , Hemodynamics , Humans , Infusions, Intravenous , Male , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/blood , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/adverse effects , Nitrogen Oxides/blood , Nitrogen Oxides/pharmacology , Young Adult
6.
Clin Immunol ; 191: 10-20, 2018 06.
Article in English | MEDLINE | ID: mdl-29518577

ABSTRACT

This study evaluated the short-term effects of tofacitinib treatment on peripheral blood leukocyte phenotype and function, and the reversibility of any such effects following treatment withdrawal in healthy volunteers. Cytomegalovirus (CMV)-seropositive subjects received oral tofacitinib 10 mg twice daily for 4 weeks and were followed for 4 weeks after drug withdrawal. There were slight increases in total lymphocyte and total T-cell counts during tofacitinib treatment, and B-cell counts increased by up to 26%. There were no significant changes in granulocyte or monocyte counts, or granulocyte function. Naïve and central memory T-cell counts increased during treatment, while all subsets of activated T cells were decreased by up to 69%. T-cell subsets other than effector memory cluster of differentiation (CD)4+, activated naïve CD4+ and effector CD8+ T-cell counts and B-cell counts, normalized 4 weeks after withdrawal. Following ex vivo activation, measures of CMV-specific T-cell responses, and antigen non-specific T-cell-mediated cytotoxicity and interferon (IFN)-γ production, decreased slightly. These T-cell functional changes were most pronounced at Day 15, partially normalized while still on tofacitinib and returned to baseline after drug withdrawal. Total natural killer (NK)-cell counts decreased by 33%, returning towards baseline after drug withdrawal. NK-cell function decreased during tofacitinib treatment, but without a consistent time course across measured parameters. However, markers of NK-cell-mediated cytotoxicity, antibody-dependent cellular cytotoxicity and IFN-γ production were decreased up to 42% 1 month after drug withdrawal. CMV DNA was not detectable in whole blood, and there were no cases of herpes zoster reactivation. No new safety concerns arose. In conclusion, the effect of short-term tofacitinib treatment on leukocyte composition and function in healthy CMV+ volunteers is modest and largely reversible 4 weeks after withdrawal.


Subject(s)
Janus Kinase Inhibitors/pharmacology , Leukocytes/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Adult , Aged , Arthritis, Rheumatoid/drug therapy , Female , Healthy Volunteers , Humans , Leukocytes/immunology , Lymphocyte Count , Male , Middle Aged , Phenotype , Piperidines/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
7.
Clin Pharmacol Ther ; 103(5): 888-898, 2018 05.
Article in English | MEDLINE | ID: mdl-28857147

ABSTRACT

We hypothesized that concomitant pharmacological inhibition of the endothelin and adenosine pathway is safe and improves exercise performance in hypoxic humans, via a mechanism that does not involve augmentation of blood oxygenation. To test this hypothesis, we established safety and drug interactions for aminophylline (500 mg) plus ambrisentan (5 mg) in normoxic volunteers. Subsequently, a placebo-controlled study was employed to test the combination in healthy resting and exercising volunteers at simulated altitude (4,267 m). No serious adverse events occurred. Drug interaction was minimal or absent. Aminophylline alleviated hypoxia-induced headaches. Aminophylline, ambrisentan, and their combination all significantly (P < 0.05 vs. placebo) improved submaximal hypoxic exercise performance (19.5, 20.6, and 19.1% >placebo). Single-dose ambrisentan increased blood oxygenation in resting, hypoxic subjects. We conclude that combined aminophylline and ambrisentan offer promise to safely increase exercise capacity in hypoxemic humans without relying on increasing blood oxygen availability.


Subject(s)
Aminophylline/adverse effects , Aminophylline/therapeutic use , Endothelins/drug effects , Exercise/physiology , Hypoxia/drug therapy , Phenylpropionates/adverse effects , Phenylpropionates/therapeutic use , Pyridazines/adverse effects , Pyridazines/therapeutic use , Adenosine/metabolism , Adolescent , Adult , Altitude , Double-Blind Method , Drug Therapy, Combination/adverse effects , Endothelins/metabolism , Female , Humans , Hypoxia/metabolism , Male , Middle Aged , Signal Transduction/drug effects , Young Adult
8.
Can J Physiol Pharmacol ; 95(9): 1009-1018, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28467859

ABSTRACT

Hypoxemia can be life-threatening, both acutely and chronically. Because hypoxemia causes vascular dysregulation that further restricts oxygen availability to tissue, it can be pharmacologically addressed. We hypothesized that theophylline can be safely combined with the ß2-adrenergic vasodilator bambuterol to improve oxygen availability in hypoxemic patients. Ergogenicity and hemodynamic effects of bambuterol and theophylline were measured in rats under hypobaric and normobaric hypoxia (12% O2). Feasibility in humans was assessed using randomized, double-blind testing of the influence of combined slow-release theophylline (300 mg) and bambuterol (20 mg) on adverse events (AEs), plasma K+, pulse, blood pressure, and drug interaction. Both drugs and their combination significantly improved hypoxic endurance in rats. In humans, common AEs were low K+ (<3.5 mmol/L; bambuterol: 12, theophylline: 4, combination: 13 episodes) and tremors (10, 0, 14 episodes). No exacerbation or serious AE occurred when drugs were combined. A drop in plasma K+ coincided with peak bambuterol plasma concentrations. Bambuterol increased heart rate by approximately 13 bpm. Drug interaction was present but small. We report promise, feasibility, and relative safety of combined theophylline and bambuterol as a treatment of hypoxemia in humans. Cardiac safety and blood K+ will be important safety endpoints when testing these drugs in hypoxemic subjects.


Subject(s)
Hypoxia/drug therapy , Terbutaline/analogs & derivatives , Theophylline/pharmacology , Adult , Animals , Biological Availability , Drug Interactions , Female , Half-Life , Hemodynamics/drug effects , Humans , Hypoxia/blood , Hypoxia/physiopathology , Male , Physical Conditioning, Animal , Rats , Safety , Terbutaline/adverse effects , Terbutaline/pharmacokinetics , Terbutaline/pharmacology , Terbutaline/therapeutic use , Theophylline/adverse effects , Theophylline/pharmacokinetics , Theophylline/therapeutic use , Treatment Outcome , Young Adult
9.
J Clin Pharmacol ; 57(6): 770-776, 2017 06.
Article in English | MEDLINE | ID: mdl-27990643

ABSTRACT

Spontaneous intracranial hemorrhage (ICH) remains a devastating stroke subtype, affecting as many as 80,000 people annually in the United States and associated with extremely high mortality. In the absence of any pharmacological interventions demonstrated to improve outcome, care for patients with ICH remains largely supportive. Thus, despite advances in the understanding of ICH and brain injury, there remains an unmet need for interventions that improve neurologic recovery and outcomes. Recent research suggesting inflammation and APOE genotype play a role in modifying neurologic outcome after brain injury has led to the development of an APOE-derived peptide agent (CN-105). Preclinical studies have demonstrated that CN-105 effectively downregulates the inflammatory response in acute brain injury, including ICH. Following Investigational New Drug (IND) enabling studies in murine models, this first-in-human single escalating dose and multiple dose placebo-controlled clinical trial was performed to define the safety and pharmacokinetics (PK) of CN-105. A total of 48 subjects (12 control, 36 active) were randomized in this study; all subjects completed the study. No significant safety issues were identified with both dosing regimens, and PK analysis revealed linearity without significant drug accumulation. The median half-life in the terminal elimination phase of CN-105 following a single or repeated dosing regimen did not change (approximately 3.6 hours). With the PK and preliminary safety of CN-105 established, the drug is now poised to begin first-in-disease phase 2 clinical trials in patients with ICH who urgently need new therapeutic options.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Adult , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/urine , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Neuroprotective Agents/adverse effects , Neuroprotective Agents/blood , Neuroprotective Agents/urine
10.
Thromb Haemost ; 117(2): 238-245, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27853809

ABSTRACT

Of the new direct oral anticoagulants, direct factor Xa inhibitors are limited by the absence of a proven reversal agent. We assessed the safety, tolerability and impact on anticoagulation reversal of ciraparantag (PER977) alone and following a 60 mg dose of the FXa inhibitor edoxaban. Escalating, single IV doses of ciraparantag were administered alone and following a 60 mg oral dose of edoxaban in a double-blind, placebo-controlled fashion to healthy subjects. Serial assessments of the pharmacokinetics and pharmacodynamic effects of ciraparantag were performed. Eighty male subjects completed the study. Following edoxaban (60 mg), a single IV dose of ciraparantag (100 to 300 mg) demonstrated full reversal of anticoagulation within 10 minutes and sustained for 24 hours. Fibrin diameter within clots was restored to normal 30 minutes after a single dose of 100 to 300 mg ciraparantag as determined by scanning electron microscopy and change in fibrin diameter quantified by automated image analysis. Potentially related adverse events were periorbital and facial flushing and cool sensation following IV injection of ciraparantag. Renal excretion of ciraparantag metabolite was the main elimination route. There was no evidence of procoagulant activity following ciraparantag as assessed by D-dimer, prothrombin fragments 1.2, and tissue factor pathway inhibitor levels. In conclusion, ciraparantag in healthy subjects is safe and well tolerated with minor, non-dose limiting adverse events. Baseline haemostasis was restored from the anticoagulated state with doses of 100 to 300 mg ciraparantag within 10-30 minutes of administration and sustained for at least 24 hours.


Subject(s)
Arginine/analogs & derivatives , Blood Coagulation/drug effects , Factor Xa Inhibitors/administration & dosage , Piperazines/administration & dosage , Pyridines/administration & dosage , Thiazoles/administration & dosage , Administration, Oral , Adult , Arginine/administration & dosage , Arginine/adverse effects , Arginine/pharmacokinetics , Double-Blind Method , Drug Monitoring/methods , Factor Xa Inhibitors/adverse effects , Humans , Injections, Intravenous , Male , North Carolina , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyridines/adverse effects , Thiazoles/adverse effects , Whole Blood Coagulation Time
11.
12.
J Nucl Med ; 56(11): 1793-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26315828

ABSTRACT

UNLABELLED: Intraarterial microdosing (IAM) is a novel drug development approach combining intraarterial drug delivery and microdosing. We aimed to demonstrate that IAM leads to target exposure similar to that of systemic full-dose administration but with minimal systemic exposure. IAM could enable the safe, inexpensive, and early study of novel drugs at the first-in-human stage and the study of established drugs in vulnerable populations. METHODS: Insulin was administered intraarterially (ipsilateral femoral artery) or systemically to 8 CD IGS rats just before blood sampling or 60-min (18)F-FDG uptake PET imaging of ipsilateral and contralateral leg muscles (lateral gastrocnemius) and systemic muscles (spinotrapezius). The (18)F-FDG uptake slope analysis was used to compare the interventions. Plasma levels of insulin and glucose were compared using area under the curve calculated by the linear trapezoidal method. A physiologically based computational pharmacokinetics/pharmacodynamics model was constructed to simulate the relationship between the administered dose and response over time. RESULTS: (18)F-FDG slope analysis found no difference between IAM and systemic full-dose slopes (0.0066 and 0.0061, respectively; 95% confidence interval [CI], -0.024 to 0.029; P = 0.7895), but IAM slope was statistically significantly greater than systemic microdose (0.0018; 95% CI, -0.045 to -0.007; P = 0.0147) and sham intervention (-0.0015; 95% CI, 0.023-0.058; P = 0.0052). The pharmacokinetics/pharmacodynamics data were used to identify model parameters that describe membrane insulin binding and glucose-insulin dynamics. CONCLUSION: Target exposure after IAM was similar to systemic full dose administration but with minimal systemic effects. The computational pharmacokinetics/pharmacodynamics model can be generalized to predict whole-body response. Findings should be validated in larger, controlled studies in animals and humans using a range of targets and classes of drugs.


Subject(s)
Fluorodeoxyglucose F18/administration & dosage , Positron-Emission Tomography/methods , Radiopharmaceuticals/administration & dosage , Algorithms , Animals , Blood Glucose/metabolism , Computer Simulation , Drug Delivery Systems , Fluorodeoxyglucose F18/adverse effects , Fluorodeoxyglucose F18/pharmacokinetics , Hypoglycemic Agents/pharmacology , Image Interpretation, Computer-Assisted , Injections, Intra-Arterial , Insulin/blood , Insulin/pharmacology , Male , Models, Statistical , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Rats
14.
J Am Heart Assoc ; 3(4)2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25012289

ABSTRACT

BACKGROUND: C-reactive protein (CRP) binds to damaged cells, activates the classical complement pathway, is elevated in multiple inflammatory conditions, and provides prognostic information on risk of future atherosclerotic events. It is controversial, however, as to whether inhibiting CRP synthesis would have any direct anti-inflammatory effects in humans. METHODS AND RESULTS: A placebo-controlled study was used to evaluate the effects of ISIS 329993 (ISIS-CRPR x) on the acute-phase response after endotoxin challenge in 30 evaluable subjects. Healthy adult males were randomly allocated to receive 6 injections over a 22-day period of placebo or active therapy with ISIS 329993 at 400- or 600-mg doses. Eligible subjects were subsequently challenged with a bolus of endotoxin (2 ng/kg). Inflammatory and hematological biomarkers were measured before and serially after the challenge. ISIS-CRPR x was well tolerated with no serious adverse events. Median CRP levels increased more than 50-fold from baseline 24 hours after endotoxin challenge in the placebo group. In contrast, the median increase in CRP levels was attenuated by 37% (400 mg) and 69% (600 mg) in subjects pretreated with ISIS-CRPR x (P<0.05 vs. placebo). All other aspects of the acute inflammatory response were similar between treatment groups. CONCLUSION: Pretreatment of subjects with ISIS-CRPR x selectively reduced the endotoxin-induced increase in CRP levels in a dose-dependent manner, without affecting other components of the acute-phase response. These data demonstrate the specificity of antisense oligonucleotides and provide an investigative tool to further define the role of CRP in human pathological conditions.


Subject(s)
Acute-Phase Reaction , C-Reactive Protein/drug effects , Endotoxins/pharmacology , Oligonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Adolescent , Adult , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/biosynthesis , Chemokine CCL2/blood , Chemokine CCL2/drug effects , E-Selectin/blood , E-Selectin/drug effects , Fibrin Fibrinogen Degradation Products/drug effects , Healthy Volunteers , Humans , Interleukin-6/blood , Male , Peptide Fragments/blood , Peptide Fragments/drug effects , Prothrombin/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/drug effects , Young Adult
15.
PLoS One ; 9(6): e99309, 2014.
Article in English | MEDLINE | ID: mdl-24960187

ABSTRACT

Rapid ascent to high altitude causes illness and fatigue, and there is a demand for effective acute treatments to alleviate such effects. We hypothesized that increased oxygen delivery to the tissue using a combination of a hypertensive agent and an endothelin receptor A antagonist drugs would limit exercise-induced fatigue at simulated high altitude. Our data showed that the combination of 0.1 mg/kg ambrisentan with either 20 mg/kg ephedrine or 10 mg/kg methylphenidate significantly improved exercise duration in rats at simulated altitude of 4,267 m, whereas the individual compounds did not. In normoxic, anesthetized rats, ephedrine alone and in combination with ambrisentan increased heart rate, peripheral blood flow, carotid and pulmonary arterial pressures, breathing rate, and vastus lateralis muscle oxygenation, but under inspired hypoxia, only the combination treatment significantly enhanced muscle oxygenation. Our results suggest that sympathomimetic agents combined with endothelin-A receptor blockers offset altitude-induced fatigue in rats by synergistically increasing the delivery rate of oxygen to hypoxic muscle by concomitantly augmenting perfusion pressure and improving capillary conductance in the skeletal muscle. Our findings might therefore serve as a basis to develop an effective treatment to prevent high-altitude illness and fatigue in humans.


Subject(s)
Altitude Sickness/drug therapy , Endothelin A Receptor Antagonists/administration & dosage , Ephedrine/administration & dosage , Fatigue/drug therapy , Methylphenidate/administration & dosage , Phenylpropionates/administration & dosage , Pyridazines/administration & dosage , Sympathomimetics/administration & dosage , Acclimatization , Altitude , Altitude Sickness/physiopathology , Animals , Cell Hypoxia/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Fatigue/physiopathology , Injections, Intraperitoneal , Rats
16.
Drug Des Devel Ther ; 7: 619-25, 2013.
Article in English | MEDLINE | ID: mdl-23926420

ABSTRACT

OBJECTIVE: MAP0004 is an investigational product which delivers dihydroergotamine (DHE) through the lung via a breath-synchronized metered dose inhaler. The objective of this study was to compare the acute effects of orally inhaled and intravenous (IV) DHE to placebo on maximum change and area under the curve for pulmonary arterial systolic pressure (PASP). RESEARCH DESIGN AND METHODS: A randomized, double-blind, placebo-controlled, 3-period, crossover study of 24 health adults. Trial registration NCT01089062. Study assessments included pharmacokinetics, electrocardiograms (ECG), and validated echocardiographic (Doppler)-derived measures of PASP by echocardiogram. The primary endpoint was the absolute change in calculated PASP using area under the curve, 0 to 2 hours (AUC(0-2h)). RESULTS: The change in PASP with IV DHE was significantly different than MAP0004 and placebo (AUC(0-2h)2857, 2624, and 2453 mmHg*min, respectively). After a second dose of MAP0004, AUC(0-4h) remained lower with MAP0004 than with a single dose of IV DHE. Adverse events were more common with IV DHE than with MAP0004 or placebo. None of the treatments produced clinically significant changes in PASP or other cardiac parameters. Changes in PASP were significantly smaller with MAP0004 compared with IV DHE. CONCLUSION: These results indicate the effects 1 mg of orally inhaled DHE on the cardiovascular system are less than with 1 mg of IV DHE, and that serial echocardiography can be a useful noninvasive means of assessing acute systemic effects.


Subject(s)
Dihydroergotamine/administration & dosage , Echocardiography , Administration, Inhalation , Administration, Intravenous , Adolescent , Adult , Cross-Over Studies , Dihydroergotamine/adverse effects , Dihydroergotamine/pharmacokinetics , Double-Blind Method , Female , Humans , Male , Metered Dose Inhalers , Middle Aged , Migraine Disorders/drug therapy , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Systole/drug effects
17.
Am J Cardiovasc Drugs ; 13(5): 331-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23784266

ABSTRACT

BACKGROUND: Edoxaban, an oral direct factor Xa inhibitor, is in development for thromboprophylaxis, including prevention of stroke and systemic embolism in patients with atrial fibrillation (AF). P-glycoprotein (P-gp), an efflux transporter, modulates absorption and excretion of xenobiotics. Edoxaban is a P-gp substrate, and several cardiovascular (CV) drugs have the potential to inhibit P-gp and increase drug exposure. OBJECTIVE: To assess the potential pharmacokinetic interactions of edoxaban and 6 cardiovascular drugs used in the management of AF and known P-gp substrates/inhibitors. METHODS: Drug-drug interaction studies with edoxaban and CV drugs with known P-gp substrate/inhibitor potential were conducted in healthy subjects. In 4 crossover, 2-period, 2-treatment studies, subjects received edoxaban 60 mg alone and coadministered with quinidine 300 mg (n = 42), verapamil 240 mg (n = 34), atorvastatin 80 mg (n = 32), or dronedarone 400 mg (n = 34). Additionally, edoxaban 60 mg alone and coadministered with amiodarone 400 mg (n = 30) or digoxin 0.25 mg (n = 48) was evaluated in a single-sequence study and 2-cohort study, respectively. RESULTS: Edoxaban exposure measured as area under the curve increased for concomitant administration of edoxaban with quinidine (76.7 %), verapamil (52.7 %), amiodarone (39.8 %), and dronedarone (84.5 %), and exposure measured as 24-h concentrations for quinidine (11.8 %), verapamil (29.1 %), and dronedarone (157.6 %) also increased. Administration of edoxaban with amiodarone decreased the 24-h concentration for edoxaban by 25.7 %. Concomitant administration with digoxin or atorvastatin had minimal effects on edoxaban exposure. CONCLUSION: Coadministration of the P-gp inhibitors quinidine, verapamil, and dronedarone increased edoxaban exposure. Modest/minimal effects were observed for amiodarone, atorvastatin, and digoxin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Anticoagulants/pharmacokinetics , Cardiovascular Agents/pharmacology , Pyridines/pharmacokinetics , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Area Under Curve , Cohort Studies , Cross-Over Studies , Drug Interactions , Factor Xa Inhibitors , Female , Humans , Male , Young Adult
18.
Expert Opin Drug Metab Toxicol ; 9(7): 817-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23550938

ABSTRACT

INTRODUCTION: Microdosing is an approach to early drug development where exploratory pharmacokinetic data are acquired in humans using inherently safe sub-pharmacologic doses of drug. The first publication of microdose data was 10 years ago and this review comprehensively explores the microdose concept from conception, over the past decade, up until the current date. AREAS COVERED: The authors define and distinguish the concept of microdosing from similar approaches. The authors review the ability of microdosing to provide exploratory pharmacokinetics (concentration-time data) but exclude microdosing using positron emission tomography. The article provides a comprehensive review of data within the peer-reviewed literature as well as the latest applications and a look into the future, towards where microdosing may be headed. EXPERT OPINION: Evidence so far suggests that microdosing may be a better predictive tool of human pharmacokinetics than alternative methods and combination with physiologically based modelling may lead to much more reliable predictions in the future. The concept has also been applied to drug-drug interactions, polymorphism and assessing drug concentrations over time at its site of action. Microdosing may yet have more to offer in unanticipated directions and provide benefits that have not been fully realised to date.


Subject(s)
Clinical Trials, Phase I as Topic , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Administration, Intravenous/trends , Administration, Oral , Animals , Databases, Factual , Dose-Response Relationship, Drug , Drug Interactions , Guidelines as Topic , Humans , Positron-Emission Tomography
19.
Br J Clin Pharmacol ; 75(4): 966-78, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22924409

ABSTRACT

AIMS: To evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of edoxaban, an oral direct factor Xa inhibitor, in healthy subjects switching from warfarin. METHODS: Seventy-two subjects were randomized to edoxaban 60 mg once daily (n = 48) or matching placebo (n = 24) for 5 days at 24 h after the last dose of warfarin treatment (INR 2.0 to 3.0). Safety/tolerability was the primary outcome measure. Pharmacokinetics, INR, aPTT, anti-FXa, thrombin generation and other coagulation assays were assessed. RESULTS: Seventy-two subjects were randomized and 64 subjects received at least one dose of edoxaban (n = 43) or placebo (n = 21) after achieving a target INR of 2.0 to 3.0 on warfarin treatment. Edoxaban 60 mg administered 24 h post-warfarin appeared to be safe and well tolerated. Adverse events were similar across treatments. For bleeding-related adverse events, eight subjects tested positive for faecal occult blood, five subjects during warfarin treatment and three subjects during edoxaban treatment. The mean (SD) baseline (post-dose of warfarin) INR was 2.31 (0.193) which increased to 3.84 (0.744) over 2 h during the edoxaban treatment (P < 0.0001 vs. placebo), returning to post-warfarin baseline within 12 h. A similar time course of effects for the other coagulation assays was observed in accordance with the drugs' mechanisms of action. CONCLUSION: In this study of healthy subjects, edoxaban administered 24 h after the last dose of warfarin was safe and well tolerated with transient increases across the various coagulation assays above post-warfarin baseline levels.


Subject(s)
Anticoagulants , Factor Xa Inhibitors , Pyridines , Thiazoles , Warfarin/administration & dosage , Administration, Oral , Adolescent , Adult , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Blood Coagulation Tests , Female , Humans , Male , Middle Aged , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/pharmacology , Thiazoles/administration & dosage , Thiazoles/adverse effects , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
20.
J Cardiovasc Pharmacol ; 60(4): 335-41, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23064240

ABSTRACT

The oral anticoagulant edoxaban, a factor Xa inhibitor, will likely be coadministered with digoxin in some patients with atrial fibrillation. Both drugs are substrates for P-glycoprotein. The objective of this phase 1, parallel study was to assess the effects of coadministration of both drugs on their respective pharmacokinetics (PK) and pharmacodynamics (PD). Forty-eight subjects, aged 18 to 45 years, received either edoxaban 60 mg once daily × 7 days (n = 24) or digoxin 0.25 mg twice daily × 2 days and once daily × 5 days (n = 24) and then concomitantly for 7 days. Serial blood and urine samples were collected for digoxin and edoxaban concentrations on days 7 and 14. Serial coagulation assays were measured for edoxaban on days 7 and 14. Edoxaban PK parameters demonstrated mild increases in area under the curve and peak concentrations of 9.5% and 15.6%, respectively, when coadministered with digoxin. Although digoxin PK parameters demonstrated increased area under the curve and peak concentrations of 8.3% and 28%, respectively, plasma concentrations were within the established therapeutic range. Edoxaban PD were consistent with PK. Both drugs were well tolerated alone or in combination. No clinically significant changes in PK, PD, or renal elimination were observed with concomitant administration of edoxaban and digoxin.


Subject(s)
Digoxin/pharmacokinetics , Factor Xa Inhibitors , Pyridines/pharmacokinetics , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Area Under Curve , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/pharmacology , Cross-Over Studies , Digoxin/adverse effects , Digoxin/pharmacology , Drug Interactions , Female , Humans , Male , Middle Aged , Pyridines/adverse effects , Pyridines/pharmacology , Thiazoles/adverse effects , Thiazoles/pharmacology , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...