Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Pharmacokinet ; 58(1): 77-88, 2019 01.
Article in English | MEDLINE | ID: mdl-29777529

ABSTRACT

BACKGROUND: Preclinical and clinical studies suggest amantadine immediate-release (IR) may reduce dyskinesia in Parkinson's disease (PD), although higher doses are associated with increased CNS adverse events (AEs). ADS-5102 is an extended release amantadine capsule formulation, designed for once-daily dosing at bedtime (qhs) to provide high concentrations upon waking and throughout the day, with lower concentrations in the evening. The pharmacokinetics (PK) of ADS-5102 were assessed in two phase I studies in healthy subjects, and a blinded, randomized phase II/III dose-finding study in PD patients. METHODS: The first phase I study assessed single ADS-5102 doses (68.5, 137, and 274 mg) in a crossover design, whereas the second phase I study evaluated ADS-5102 137 mg for 7 days followed by amantadine IR 81 mg twice daily (or reverse order). In the phase II/III double-blind study, PD patients with dyskinesia were randomized to ADS-5102 (210, 274, or 338 mg) or placebo for 8 weeks. RESULTS: Single ADS-5102 doses resulted in a slow initial rise in amantadine plasma concentration, with delayed time to maximum concentration (12-16 h). Amantadine plasma concentrations were higher in PD patients versus healthy volunteers. The steady-state profile of once-daily ADS-5102 was significantly different from that of twice-daily amantadine IR, such that the two formulations are not bioequivalent. PK modeling suggested the recommended daily ADS-5102 dosage (274 mg qhs) resulted in 1.4- to 2.0-fold higher amantadine plasma concentrations during the day versus amantadine IR. CONCLUSIONS: ADS-5102 can be administered once-daily qhs to achieve high amantadine plasma concentrations in the morning and throughout the day, when symptoms of dyskinesia occur.


Subject(s)
Amantadine/administration & dosage , Amantadine/pharmacokinetics , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Dyskinesias/metabolism , Parkinson Disease/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Double-Blind Method , Drug Administration Schedule , Dyskinesias/drug therapy , Female , Humans , Male , Middle Aged , Parkinson Disease/drug therapy , Young Adult
2.
Sci Transl Med ; 9(387)2017 04 26.
Article in English | MEDLINE | ID: mdl-28446688

ABSTRACT

Conventional cytotoxic chemotherapy is highly effective in certain cancers but causes dose-limiting damage to normal proliferating cells, especially hematopoietic stem and progenitor cells (HSPCs). Serial exposure to cytotoxics causes a long-term hematopoietic compromise ("exhaustion"), which limits the use of chemotherapy and success of cancer therapy. We show that the coadministration of G1T28 (trilaciclib), which is a small-molecule inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), contemporaneously with cytotoxic chemotherapy protects murine hematopoietic stem cells (HSCs) from chemotherapy-induced exhaustion in a serial 5-fluorouracil treatment model. Consistent with a cell-intrinsic effect, we show directly preserved HSC function resulting in a more rapid recovery of peripheral blood counts, enhanced serial transplantation capacity, and reduced myeloid skewing. When administered to healthy human volunteers, G1T28 demonstrated excellent in vivo pharmacology and transiently inhibited bone marrow (BM) HSPC proliferation. These findings suggest that the combination of CDK4/6 inhibitors with cytotoxic chemotherapy should provide a means to attenuate therapy-induced BM exhaustion in patients with cancer.


Subject(s)
Hematopoietic Stem Cells/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Female , Fluorouracil/pharmacology , Healthy Volunteers , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL
3.
Am J Hematol ; 87(11): 1017-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22887019

ABSTRACT

Therapeutics which reduce the pathology in sickle cell syndromes are needed, particularly noncytotoxic therapeutics. Fetal hemoglobin (HbF, α(2) γ(2) ) is established as a major regulator of disease severity; increased HbF levels correlate with milder clinical courses and improved survival. Accordingly, sodium dimethylbutyrate (HQK-1001), an orally-bioavailable, promoter-targeted fetal globin gene-inducing agent, was evaluated in a randomized, blinded, dose-ranging Phase I/II trial in 24 adult patients with HbSS or S/ß thalassemia, to determine safety and tolerability of three escalating dose levels. The study therapeutic was administered once daily for two 6-week cycles, with a two-week interim dose holiday. Twenty-one patients completed the study. Five patients received study drug at 10 or 20 mg/kg doses, seven patients received study drug at 30 mg/kg/dose, and 4 patients received placebo. HQK-1001 was well-tolerated with no unexpected drug-related adverse events; a dose-limiting toxicity was not identified. Plasma drug levels were sustained above targeted levels for 24 hr. Increases in HbF above baseline were observed particularly with 30 mg/kg/day doses; in five of seven treated patients, a mean absolute increase in HbF of 0.2 g/dl and a mean increase in total hemoglobin (Hgb) of 0.83 g/dl above baseline were observed, whereas no increases occurred in placebo-treated controls. These findings of favorable PK profiles, tolerability, early rises in HbF, and total Hgb indicate that trials of longer duration appear warranted to more definitively evaluate the therapeutic potential of HQK-1001 in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/drug therapy , Butyrates/adverse effects , Butyrates/pharmacokinetics , Fetal Hemoglobin/biosynthesis , Hematinics/adverse effects , Hematinics/pharmacokinetics , Administration, Oral , Adolescent , Adult , Anemia, Sickle Cell/blood , Biological Availability , Butyrates/therapeutic use , Chromatography, High Pressure Liquid , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Fetal Hemoglobin/analysis , Hematinics/therapeutic use , Humans , Male , Middle Aged , Young Adult
4.
Eur Heart J ; 32(19): 2412-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21724623

ABSTRACT

AIMS: Establishing factor IX inhibition in patients with acute coronary syndrome/non-ST-elevation myocardial infarction (ACS/NSTEMI), a setting characterized by increased factor IX activity, is critical to investigate the REG1 system in this target population. The REG1 system (Regado Biosciences, Basking Ridge, NJ) consists of pegnivacogin (RB006), an RNA aptamer that directly inhibits factor IXa, and anivamersen (RB007), its complementary control agent. METHODS AND RESULTS: RADAR is a Phase 2b study investigating the use of pegnivacogin in patients (n = 800) with ACS undergoing planned early cardiac catheterization. To validate dose selection and stability of anticoagulation throughout the time of cardiac catheterization at an early stage of the clinical trial, 33 patients, 22 of whom had not received recent prior heparin, underwent thorough pharmacokinetic and pharmacodynamic assessment. Fold prolongation of activated partial thromboplastin time (aPTT) was used to impute factor IX inhibition. Pegnivacogin 1 mg/kg rapidly achieved a high pegnivacogin plasma concentration (26.1 ± 4.6 µg/mL), prolonged the aPTT (mean aPTT 93.0 ± 9.5 s), and approached near complete factor IX inhibition (mean fold increase from baseline 2.9 ± 0.3). These levels remained stable from the time of drug administration through completion of the catheterization. CONCLUSION: Pegnivacogin administered at a weight-adjusted dose of 1 mg/kg consistently achieves a high level of factor IX activity inhibition among patients with ACS and provides stable anticoagulation during cardiac catheterization. These findings support the dose of pegnivacogin selected for the RADAR study.


Subject(s)
Acute Coronary Syndrome/drug therapy , Anticoagulants/pharmacology , Aptamers, Nucleotide/pharmacology , Factor IXa/antagonists & inhibitors , Myocardial Infarction/drug therapy , Acute Coronary Syndrome/blood , Aged , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/pharmacokinetics , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Myocardial Infarction/blood , Partial Thromboplastin Time , Treatment Outcome
5.
Blood Cells Mol Dis ; 47(2): 107-16, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21641240

ABSTRACT

The ß-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the ß-globin chain of hemoglobin A (α(2)ß(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and ß-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.


Subject(s)
Anemia/drug therapy , Biological Products/administration & dosage , Drug Design , Erythroid Precursor Cells/drug effects , Fetal Hemoglobin/biosynthesis , Small Molecule Libraries/administration & dosage , beta-Thalassemia/drug therapy , gamma-Globins/biosynthesis , Administration, Oral , Anemia/genetics , Anemia/metabolism , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Cells, Cultured , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/genetics , Gene Expression , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Models, Molecular , Papio , Phlebotomy , Polymerase Chain Reaction , RNA, Messenger/analysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , beta-Globins/deficiency , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , gamma-Globins/genetics
6.
J Clin Pharmacol ; 51(8): 1186-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21422239

ABSTRACT

Pharmacologic induction of fetal globin synthesis is an accepted therapeutic strategy for treatment of the beta hemoglobinopathies and thalassemias, as even small increases in hemoglobin F (HbF) levels reduce clinical severity in sickle cell disease (SCD) and reduce anemia in beta thalassemia. Prior generation short chain fatty acid therapeutics, arginine butyrate (AB), and phenylbutyrate, increased fetal and total hemoglobin levels in patients, but were limited by high doses or intravenous (IV) infusion. A fetal globin-inducing therapeutic with convenient oral dosing would be an advance for these classic molecular diseases. Healthy adult human subjects were treated with a novel short chain fatty acids (SCFA) derivative, sodium 2,2 dimethylbutyrate (SDMB), or placebo, with 1 of 4 single dose levels (2, 5, 10, and 20 mg/kg) or daily doses (5, 10, or 15 mg/kg) over 14 days, and monitored for adverse clinical and laboratory events, drug levels, reticulocytes, and HbF assays. SDMB was well-tolerated with no clinically significant adverse events related to study medication. The terminal half-life ranged from 9 to 15 hours. Increases in mean absolute reticulocytes were observed at all dose levels in the 14-day study. The favorable pharmacokinetics (PK) profiles and safety findings indicate that SDMB warrants further investigation for treatment of anemic subjects with beta hemoglobinopathies.


Subject(s)
Butyrates/adverse effects , Butyrates/pharmacokinetics , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Hematinics/adverse effects , Hematinics/pharmacokinetics , Administration, Oral , Adolescent , Adult , Biological Availability , Blood Cell Count , Butyrates/administration & dosage , Butyrates/analysis , Dose-Response Relationship, Drug , Double-Blind Method , Drugs, Investigational/administration & dosage , Drugs, Investigational/analysis , Female , Fetal Hemoglobin/analysis , Food-Drug Interactions , Half-Life , Hematinics/administration & dosage , Hematinics/analysis , Humans , Male , Metabolic Clearance Rate , Plasma/chemistry , Reticulocytes/drug effects , Urine/chemistry , Young Adult
7.
Am Heart J ; 161(2): 261-268.e1-2, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21315207

ABSTRACT

Anticoagulants are the cornerstone of current acute coronary syndrome (ACS) therapy; however, anticoagulation regimens that aggressively reduce ischemic events are almost uniformly associated with more bleeding. REG1, an anticoagulation system, consists of RB006 (pegnivacogin), an RNA oligonucleotide factor IXa inhibitor, and RB007 (anivamersen), its complementary controlling agent. Phase I and IIa studies defined predictable relationships between doses of RB006, RB007, and degree of antifactor IX activity. The efficacy and safety of REG1 for the treatment of patients with ACS managed invasively and the safety of reversing RB006 with RB007 after cardiac catheterization are unknown. Randomized, partially-blinded, multicenter, active-controlled, dose-ranging study assessing the safety, efficacy, and pharmacodynamics of the REG1 anticoagulation system compared to unfractionated heparin or low molecular heparin in subjects with acute coronary syndrome (RADAR) is designed to assess both the efficacy of the anticoagulant RB006 and the safety of a range of levels of RB006 reversal with RB007. The objectives of RADAR are (1) to determine the safety of a range of levels of RB006 reversal with RB007 after catheterization, (2) to confirm whether a dose of 1 mg/kg RB006 results in near-complete inhibition of factor IXa in patients with ACS, and (3) to assess the efficacy of RB006 as an anticoagulant in patients with ACS undergoing percutaneous coronary intervention.


Subject(s)
Acute Coronary Syndrome/drug therapy , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/pharmacokinetics , Clinical Trials, Phase II as Topic , Multicenter Studies as Topic , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacokinetics , Randomized Controlled Trials as Topic , Clinical Trials, Phase II as Topic/methods , Humans , Randomized Controlled Trials as Topic/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...