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.
Invest New Drugs ; 35(3): 298-306, 2017 06.
Article in English | MEDLINE | ID: mdl-28050790

ABSTRACT

Background DR5 is a transmembrane receptor that transduces extracellular ligand-binding to activate apoptosis signaling cascades. This phase 1 study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics of a new monoclonal antibody potent DR5 agonist, DS-8273a, in subjects with advanced solid tumors. Methods The study comprised dose escalation and dose expansion cohorts. The dose escalation cohorts intended to determine the safety and to identify the maximum tolerated dose (MTD) or maximum administered dose (MAD) and to characterize the pharmacokinetics and pharmacodynamics by a conventional 3 + 3 design (starting at 2 mg/kg and escalating through 8, 16 and 24 mg/kg once every 3 weeks). In the dose expansion cohort, additional subjects were treated at the MAD for further evaluation of PK and safety. Results Thirty two subjects were enrolled and treated, 16 in the dose escalation cohorts and 16 in the dose expansion cohort. No subjects experienced a dose limiting toxicity (DLT). Treatment emergent adverse events were observed in 29 (91%) subjects, 14 (44%) of which were attributed to study-drug; all drug-related events were grade 1 and 2 in severity, and were mainly fatigue, nausea, vomiting and diarrhea. Measures of plasma exposure increased dose-proportionally and the mean terminal elimination half-life was 11 days. Blood samples available from a subset of patients treated at 24 mg/kg revealed declines in myeloid derived suppressor cells (MDSC) at 2 weeks. No objective responses were observed in any subjects. Conclusions DS-8273a was well tolerated and demonstrated linear pharmacokinetics. Decreases in MDSC were temporally associated with DS-8273a exposure. This agent could be studied further in combination with other agents, pending further proof-of-target-engagement.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Cell Count , Diarrhea/chemically induced , Fatigue/chemically induced , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Myeloid-Derived Suppressor Cells/drug effects , Nausea/chemically induced , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Vomiting/chemically induced
2.
Mol Imaging Biol ; 18(3): 446-53, 2016 06.
Article in English | MEDLINE | ID: mdl-26567113

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the safety, dosimetry, and apparent receptor occupancy (RO) of [(64)Cu]DOTA-patritumab, a radiolabeled monoclonal antibody directed against HER3/ERBB3 in subjects with advanced solid tumors. PROCEDURES: Dosimetry subjects (n = 5) received [(64)Cu]DOTA-patritumab and underwent positron emission tomography (PET)/X-ray computed tomography (CT) at 3, 24, and 48 h. Evaluable RO subjects (n = 3 out of 6) received [(64)Cu]DOTA-patritumab at day 1 and day 8 (after 9.0 mg/kg patritumab) followed by PET/CT at 24 h post-injection. Endpoints included safety, tumor uptake, and efficacy. RESULTS: The tumor SUVmax (± SD) was 5.6 ± 4.5, 3.3 ± 1.7, and 3.0 ± 1.1 at 3, 24, and 48 h in dosimetry subjects. The effective dose and critical organ dose (liver) averaged 0.044 ± 0.008 mSv/MBq and 0.46 ± 0.086 mGy/MBq, respectively. In RO subjects, tumor-to-blood ratio decreased from 1.00 ± 0.32 at baseline to 0.57 ± 0.17 after stable patritumab, corresponding to a RO of 42.1 ± 3. CONCLUSIONS: [(64)Cu]DOTA-patritumab was safe. These limited results suggest that this PET-based method can be used to determine tumor-apparent RO.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Organometallic Compounds/adverse effects , Organometallic Compounds/therapeutic use , Radiometry , Adult , Aged , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Organometallic Compounds/pharmacokinetics , Positron-Emission Tomography , Receptors, Cell Surface , Tomography, X-Ray Computed , Treatment Outcome
3.
Clin Drug Investig ; 35(7): 447-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26068927

ABSTRACT

BACKGROUND AND OBJECTIVE: The oral direct factor Xa inhibitor edoxaban is a P-glycoprotein (P-gp) substrate metabolized via carboxylesterase-1 and cytochrome P450 (CYP) 3A4/5. The effect of rifampin-induced induction of P-gp and CYP3A4/5 on transport and metabolism of edoxaban through the CYP3A4/5 pathway was investigated in a single-dose edoxaban study. METHODS: This was a phase 1, open-label, two-treatment, two-period, single-sequence drug interaction study in healthy adults. All subjects received a single oral 60 mg edoxaban dose in period 1, and 7 days of 600 mg rifampin (2 × 300 mg capsules once daily) with a single oral edoxaban 60 mg dose administered concomitantly on day 7 in period 2. A 6-day washout period separated the treatments. Plasma concentrations of edoxaban and its metabolites M4 and M6 were measured, and limited assessments of pharmacodynamic markers of coagulation were performed. RESULTS: In total, 34 healthy subjects were enrolled; 32 completed the study. Coadministration of rifampin with edoxaban decreased edoxaban exposure but increased active metabolite exposure. Rifampin increased apparent oral clearance of edoxaban by 33 % and decreased its half-life by 50 %. Anticoagulant effects based on the prothrombin time (PT) and the activated partial thromboplastin time (aPTT) with and without rifampin at early time points were maintained to a greater-than-expected degree than with edoxaban exposure alone, presumably because of an increased contribution from the active metabolites. Edoxaban was well tolerated in this healthy adult population. CONCLUSIONS: Rifampin reduced exposure to edoxaban while increasing exposure to its active metabolites M4 and M6. PT and aPTT at early time points did not change appreciably; however, the data should be interpreted with caution.


Subject(s)
Factor Xa Inhibitors/pharmacokinetics , Pyridines/pharmacokinetics , Rifampin/pharmacology , Thiazoles/pharmacokinetics , Adult , Drug Interactions , Female , Humans , Male , Prothrombin Time
4.
Circulation ; 131(1): 82-90, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25403645

ABSTRACT

BACKGROUND: The oral factor Xa inhibitor edoxaban has demonstrated safety and efficacy in stroke prevention in patients with atrial fibrillation and in the treatment and secondary prevention of venous thromboembolism. This study investigated the reversal of edoxaban's effects on bleeding measures and biomarkers by using a 4-factor prothrombin complex concentrate (4F-PCC). METHODS AND RESULTS: This was a phase 1 study conducted at a single site. This was a double-blind, randomized, placebo-controlled, 2-way crossover study to determine the reversal effect of descending doses of 4F-PCC on bleeding duration and bleeding volume following edoxaban treatment. A total of 110 subjects (17 in part 1, 93 in part 2) were treated. Intravenous administration of 4F-PCC 50, 25, or 10 IU/kg following administration of edoxaban (60 mg) dose-dependently reversed edoxaban's effects on bleeding duration and endogenous thrombin potential, with complete reversal at 50 IU/kg. Effects on prothrombin time were partially reversed at 50 IU/kg. A similar trend was seen for bleeding volume. CONCLUSIONS: The 4F-PCC dose-dependently reversed the effects of edoxaban (60 mg), with complete reversal of bleeding duration and endogenous thrombin potential and partial reversal of prothrombin time following 50 IU/kg. Edoxaban alone and in combination with 4F-PCC was safe and well tolerated in these healthy subjects. A dose of 50 IU/kg 4F-PCC may be suitable for reversing edoxaban anticoagulation. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02047565.


Subject(s)
Biopsy, Needle/adverse effects , Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , Factor Xa Inhibitors/therapeutic use , Hemorrhage/prevention & control , Pyridines/therapeutic use , Thiazoles/therapeutic use , Administration, Intravenous , Adolescent , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/adverse effects , Female , Humans , Male , Middle Aged , Prothrombin Time , Pyridines/administration & dosage , Pyridines/adverse effects , Thiazoles/administration & dosage , Thiazoles/adverse effects , Time Factors , Young Adult
5.
Biochem Biophys Res Commun ; 349(1): 221-8, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16930533

ABSTRACT

In a high-throughput screen of four million compounds from combinatorial libraries for small-molecule modulators of the chemokine receptor CXCR3, two classes of receptor agonists, based on tetrahydroisoquinoline and piperidinyl diazepanone templates, were identified. Several of these compounds stimulated calcium flux in HEK293 cells expressing the recombinant human CXCR3 receptor with efficacies and kinetics similar to those of native ligand CXCL11/I-TAC and stimulated chemotaxis of activated human T-cells. The agonist small molecules also inhibited binding of another CXCR3 ligand, CXCL10/IP-10, to the receptor. The response to small-molecule agonists was inhibited by a CXCR3-specific small-molecule antagonist previously identified within the same combinatorial compound collection but structurally unrelated to the agonists. Remarkably, while other, non-amino acid substituents were present in the majority of the library compounds screened, the agonists from both classes contained a positively charged amino acid component, with preference for Arg>Lys, as well as a hydrophobic component.


Subject(s)
Receptors, Chemokine/agonists , Biochemistry/methods , Chemokine CXCL10 , Chemokine CXCL11 , Chemokines, CXC/chemistry , Chemokines, CXC/metabolism , Chemotaxis , Combinatorial Chemistry Techniques , Dose-Response Relationship, Drug , Humans , Kinetics , Ligands , Models, Chemical , Protein Binding , Receptors, CXCR3 , T-Lymphocytes/metabolism , Tetrahydroisoquinolines/pharmacology
6.
J Pharmacol Exp Ther ; 318(2): 495-502, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16702443

ABSTRACT

The p38 mitogen-activated protein (MAP) kinases are a family of serine/threonine protein kinases that play important roles in cellular responses to inflammation and external stress. Inhibitors of the p38 MAP kinase have shown promise for potential treatment of inflammatory disorders such as rheumatoid arthritis, acute coronary syndrome, psoriasis, and Crohn's disease. We identified a novel class of p38 inhibitors via high-throughput screening. PS200981 [3-(4-(1,4-diazepan-1-yl)-6-(((1S,2R,5S)-6,6-dimethylbicyclo[3.1.1]heptan-2-yl)methylamino)-1,3,5-triazin-2-ylamino)-4-methylbenzamide], a representative compound identified from screening a collection of combinatorial libraries, amounting to 2.1 million compounds, inhibits p38alpha kinase and the lipopolysaccharide (LPS)-induced increase in tumor necrosis factor (TNF) alpha levels in cell media of human monocytes with IC50 values of 1 microM. The screening data revealed a preferred synthon, 3-amino-4-methyl benzamide, which is critical for the activity against p38. This synthon appeared almost exclusively in screening hits including PS200981, and slight variations of this synthon including 3-amino benzamide and 2-amino-4-methyl benzamide also contained in the library were inactive. PS200981 is equally potent against the alpha and beta forms of p38 but did not inhibit p38 gamma and is >25-fold selective versus a panel of other kinases. PS200981 inhibited the LPS-induced increase in TNFalpha levels when administered at 30 mg/kg to mice. Selectivity and in vivo activity of this class of p38 inhibitors was further demonstrated by PS166276 [(R)-3-(4-(isobutyl(methyl)-amino)-6-(pyrrolidin-3-ylamino)-1,3,5-triazin-2-ylamino)-4-methylbenzamide], a highly structurally related but more potent and less cytotoxic inhibitor, in several intracellular signaling assays, and in LPS-challenged mice. Overall, this novel class of p38 inhibitors is potent, active in vitro and in vivo, and is highly selective.


Subject(s)
Aniline Compounds/chemical synthesis , Aniline Compounds/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Triazines/chemical synthesis , Triazines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/antagonists & inhibitors , Animals , Antigen-Antibody Complex/metabolism , Binding, Competitive/drug effects , Blotting, Western , Cell Line , Cell Survival/drug effects , Drug Design , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Med Chem ; 47(25): 6283-91, 2004 Dec 02.
Article in English | MEDLINE | ID: mdl-15566298

ABSTRACT

A new structural class of triaminotriazine aniline amides possessing potent p38 enzyme activity has been discovered. The initial hit (compound 1a) was identified through screening the Pharmacopeia ECLiPS compound collection. SAR modification led to the identification of a short acting triaminotriazine aniline methoxyamide (compound 1m) possessing in vitro and in vivo oral activity in animal models of acute and chronic inflammatory disease. An X-ray crystal structure of compound 1m in this class, cocrystallized with unactivated p38 alpha protein, indicates that these compounds bind to the ATP binding pocket and possess key H-bonding interactions within a deeper cleft. Hydrogen bonding between one of the triazine nitrogens and the backbone NH of the Met109 residue occurs through a water molecule. The methoxyamide NH and carbonyl oxygen are within H-bonding distance of Glu71 and Asp168.


Subject(s)
Amides/chemical synthesis , Aniline Compounds/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Benzamides/chemical synthesis , Triazines/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Administration, Oral , Amides/chemistry , Amides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Benzamides/chemistry , Benzamides/pharmacology , Crystallography, X-Ray , Female , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Monocytes/drug effects , Monocytes/metabolism , Rats , Rats, Inbred Lew , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...