15 results on '"Luettgen J"'
Search Results
2. First-in-human study to assess the safety, pharmacokinetics and pharmacodynamics of BMS-986177/JNJ-70033093, a direct, reversible, small molecule Factor XIa inhibitor in healthy volunteers
- Author
-
Perera, V, primary, Wang, Z, additional, Luettgen, J, additional, Wang, J, additional, Li, D, additional, Desouza, M, additional, Cerra, M, additional, and Seiffert, D, additional
- Published
- 2020
- Full Text
- View/download PDF
3. Apixaban, an oral, direct and highly selective factor Xa inhibitor: in vitro, antithrombotic and antihemostatic studies
- Author
-
WONG, P. C., CRAIN, E. J., XIN, B., WEXLER, R. R., LAM, P. Y. S., PINTO, D. J., LUETTGEN, J. M., and KNABB, R. M.
- Published
- 2008
4. Discovery of 1-(3-Aminobenzisoxazol-5-yl)-3-trifluoromethyl-N-[2-fluoro-4- [(2-dimethylaminomethyl)imidazol-1-yl]phenyl]-1H-pyrazole-5-carboxyamide Hydrochloride (Razaxaban), a Highly Potent, Selective, and Orally Bioavailable Factor Xa Inhibitor
- Author
-
Quan, M. L., Lam, P. Y. S., Han, Q., Pinto, D. J. P., He, M. Y., Li, R., Ellis, C. D., Clark, C. G., Teleha, C. A., Sun, J.-H., Alexander, R. S., Bai, S., Luettgen, J. M., Knabb, R. M., Wong, P. C., and Wexler, R. R.
- Abstract
Modification of a series of pyrazole factor Xa inhibitors to incorporate an aminobenzisoxazole as the P
1 ligand resulted in compounds with improved selectivity for factor Xa relative to trypsin and plasma kallikrein. Further optimization of the P4 moiety led to compounds with enhanced permeability and reduced protein binding. The SAR and pharmacokinetic profile of this series of compounds is described herein. These efforts culminated in 1-(3-aminobenzisoxazol-5-yl)-3-trifluoromethyl-N-[2-fluoro-4-[(2-dimethylaminomethyl)imidazol-1-yl]phenyl]-1H-pyrazole-5-carboxyamide (11d ), a potent, selective, and orally bioavailable inhibitor of factor Xa. On the basis of its excellent in vitro potency and selectivity profile, high free fraction in human plasma, good oral bioavailability, and in vivo efficacy in antithrombotic models, the HCl salt of this compound was selected for clinical development as razaxaban (DPC 906, BMS-561389).- Published
- 2005
5. Discovery of 1-(2-Aminomethylphenyl)-3-trifluoromethyl-N- [3-fluoro-2-(aminosulfonyl)[1,1-biphenyl)]-4-yl]-1H-pyrazole-5-carboxyamide (DPC602), a Potent, Selective, and Orally Bioavailable Factor Xa Inhibitor<SUP>1</SUP>
- Author
-
Pruitt, J. R., Pinto, D. J. P., Galemmo, R. A., Jr., Alexander, R. S., Rossi, K. A., Wells, B. L., Drummond, S., Bostrom, L. L., Burdick, D., Bruckner, R., Chen, H., Smallwood, A., Wong, P. C., Wright, M. R., Bai, S., Luettgen, J. M., Knabb, R. M., Lam, P. Y. S., and Wexler, R. R.
- Abstract
Factor Xa, a serine protease, is at the critical juncture between the intrinsic and extrinsic pathways of the coagulation cascade. Inhibition of factor Xa has the potential to provide effective treatment for both venous and arterial thrombosis. We recently described a series of meta-substituted phenylpyrazoles that are highly potent, selective, and orally bioavailable factor Xa inhibitors. In this paper we report our efforts to further optimize the selectivity profile of our factor Xa inhibitors with a series of ortho- and/or para-substituted phenylpyrazole derivatives. The most potent compounds display sub-nanomolar inhibition constants for factor Xa and show greater than 1000-fold selectivity against other serine proteases. These compounds are also effective in a rabbit model of arteriovenous shunt thrombosis. Optimization of this series led to the preclinical development of DPC602, a 2-(aminomethyl)phenylpyrazole analogue, as a highly potent, selective, and orally bioavailable factor Xa inhibitor.
- Published
- 2003
6. Structure-Based Design of Novel Guanidine/Benzamidine Mimics: Potent and Orally Bioavailable Factor Xa Inhibitors as Novel Anticoagulants
- Author
-
Lam, P. Y. S., Clark, C. G., Li, R., Pinto, D. J. P., Orwat, M. J., Galemmo, R. A., Fevig, J. M., Teleha, C. A., Alexander, R. S., Smallwood, A. M., Rossi, K. A., Wright, M. R., Bai, S. A., He, K., Luettgen, J. M., Wong, P. C., Knabb, R. M., and Wexler, R. R.
- Abstract
As part of an ongoing effort to prepare orally active factor Xa inhibitors using structure-based drug design techniques and molecular recognition principles, a systematic study has been performed on the pharmacokinetic profile resulting from replacing the benzamidine in the P1 position with less basic benzamidine mimics or neutral residues. It is demonstrated that lowering the pK
a of the P1 ligand resulted in compounds (3-benzylamine,15a ; 1-aminoisoquinoline,24a ; 3-aminobenzisoxazole,23a ; 3-phenylcarboxamide,22b ; and 4-methoxyphenyl,22a ) with improved pharmacokinetic features mainly as a result of decreased clearance, increased volume of distribution, and enhanced oral absorption. This work resulted in a series of potent and orally bioavailable factor Xa inhibitors that ultimately led to the discovery of SQ311,24a . SQ311, which utilizes a 1-aminoisoquinoline as the P1 ligand, inhibits factor Xa with a Ki of 0.33 nM and demonstrates both good in vivo antithrombotic efficacy and oral bioavailability.- Published
- 2003
7. Discovery of 1-[3-(Aminomethyl)phenyl]-N-[3-fluoro-2-(methylsulfonyl)- [1,1-biphenyl]-4-yl]-3-(trifluoromethyl)-1H-pyrazole-5-carboxamide (DPC423), a Highly Potent, Selective, and Orally Bioavailable Inhibitor of Blood Coagulation Factor Xa<SUP>1</SUP><BBR RID="jm000409zb00001">
- Author
-
Pinto, D. J. P., Orwat, M. J., Wang, S., Fevig, J. M., Quan, M. L., Amparo, E., Cacciola, J., Rossi, K. A., Alexander, R. S., Smallwood, A. M., Luettgen, J. M., Liang, L., Aungst, B. J., Wright, M. R., Knabb, R. M., Wong, P. C., Wexler, R. R., and Lam, P. Y. S.
- Abstract
Factor Xa (fXa) plays a critical role in the coagulation cascade, serving as the point of convergence of the intrinsic and extrinsic pathways. Together with nonenzymatic cofactor Va and Ca2+ on the phospholipid surface of platelets or endothelial cells, factor Xa forms the prothrombinase complex, which is responsible for the proteolysis of prothrombin to catalytically active thrombin. Thrombin, in turn, catalyzes the cleavage of fibrinogen to fibrin, thus initiating a process that ultimately leads to clot formation. Recently, we reported on a series of isoxazoline and isoxazole monobasic noncovalent inhibitors of factor Xa which show good potency in animal models of thrombosis. In this paper, we wish to report on the optimization of the heterocyclic core, which ultimately led to the discovery of a novel pyrazole SN429 (
2b ; fXa Ki = 13 pM). We also report on our efforts to improve the oral bioavailability and pharmacokinetic profile of this series while maintaining subnanomolar potency and in vitro selectivity. This was achieved by replacing the highly basic benzamidine P1 with a less basic benzylamine moiety. Further optimization of the pyrazole core substitution and the biphenyl P4 culminated in the discovery of DPC423 (17h ), a highly potent, selective, and orally active factor Xa inhibitor which was chosen for clinical development.- Published
- 2001
8. Design, Synthesis, and Biological Evaluation of Potent and Selective Amidino Bicyclic Factor Xa Inhibitors
- Author
-
Han, Q., Dominguez, C., Stouten, P. F. W., Park, J. M., Duffy, D. E., Galemmo, R. A., Jr., Rossi, K. A., Alexander, R. S., Smallwood, A. M., Wong, P. C., Wright, M. M., Luettgen, J. M., Knabb, R. M., and Wexler, R. R.
- Abstract
Thrombotic diseases are a major cause of death and morbidity. Factor Xa (fXa) plays a vital role in the regulation of normal homeostasis and abnormal intravascular thrombus development in the blood coagulation cascade. A novel series of fXa inhibitors incorporating an amidino 6,5-fused bicyclic moiety at the P1 position has been designed and synthesized based on molecular modeling studies. Structure−activity relationship (SAR) studies have led to selective subnanomolar fXa inhibitors. The most potent fXa inhibitor in this series (
72 , SE170) has a potent inhibition constant (Ki = 0.3 nM), is 350-fold selective for fXa over trypsin, and also shows good in vivo efficacy in a rabbit arterio-venous thrombosis model (ID50 = 0.14 μmol/kg/h). An X-ray crystal structure of72 complexed to bovine trypsin was completed, and a binding mode of72 with fXa has been proposed based on modeling with human des-Gla-fXa.- Published
- 2000
9. Design and synthesis of potent and selective 5,6-fused heterocyclic thrombin inhibitors
- Author
-
Dominguez, C., Duffy, D. E., Han, Q., Alexander, R. S., Galemmo, R. A., Park, J. M., Wong, P. C., Amparo, E. C., Knabb, R. M., and Luettgen, J.
- Published
- 1999
- Full Text
- View/download PDF
10. Preventing Violent Re-offending in Not Criminally Responsible Patients: An Evaluation of a Continuity of Treatment Program
- Author
-
Luettgen, J., Chrapko, W. E., and Reddon, J. R.
- Published
- 1998
- Full Text
- View/download PDF
11. Safety, tolerability, pharmacokinetics and pharmacodynamics of milvexian with aspirin and/or clopidogrel in healthy participants.
- Author
-
Perera V, Abelian G, Luettgen J, Aronson R, Li D, Wang Z, Zhang L, Lubin S, Merali S, and Murthy B
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Cross-Over Studies, Young Adult, Clopidogrel pharmacokinetics, Clopidogrel administration & dosage, Clopidogrel adverse effects, Aspirin pharmacokinetics, Aspirin administration & dosage, Healthy Volunteers, Platelet Aggregation Inhibitors pharmacokinetics, Platelet Aggregation Inhibitors pharmacology, Platelet Aggregation Inhibitors adverse effects, Platelet Aggregation Inhibitors administration & dosage, Drug Interactions
- Abstract
Milvexian, an oral activated Factor XI (FXIa) inhibitor, is in clinical studies where it may be combined with antiplatelet agents, including aspirin and/or clopidogrel, to prevent thromboembolic diseases. This phase I trial assessed safety, pharmacokinetics, and pharmacodynamics of milvexian coadministration with aspirin and/or clopidogrel in healthy participants through 3 drug-drug interaction studies using a 3-period, 3-treatment, crossover design. A total of 113 participants were randomized to receive milvexian (200 mg; twice daily for 5 days) or matched placebo coadministered with once-daily aspirin (325 mg for 5 days) and/or clopidogrel (Day 1: 300 mg; Days 2-5: 75 mg). Milvexian was safe and well tolerated, with and without aspirin and/or clopidogrel. Eight mild bleeding adverse events (AEs) were reported in 5 of 113 participants across various treatment arms. Peak and total exposures of milvexian were similar with or without clopidogrel and/or aspirin. Exposure-dependent prolongation of activated partial thromboplastin time and reduction of FXI clotting activity by milvexian were similar with coadministration of aspirin and/or clopidogrel. Milvexian, with or without coadministration of aspirin and/or clopidogrel, did not affect bleeding time or platelet aggregation. Administration of milvexian alone or with aspirin and/or clopidogrel was safe and well tolerated without increased incidence of AEs, including bleeding. Pharmacokinetic and pharmacodynamic effects of milvexian, including bleeding time, were similar with or without aspirin and/or clopidogrel.ClinicalTrials.gov Identifier: NCT03698513., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. First-in-human study of milvexian, an oral, direct, small molecule factor XIa inhibitor.
- Author
-
Perera V, Wang Z, Luettgen J, Li D, DeSouza M, Cerra M, and Seiffert D
- Subjects
- Administration, Oral, Adult, Dose-Response Relationship, Drug, Double-Blind Method, Female, Food-Drug Interactions, Humans, Male, Middle Aged, Partial Thromboplastin Time, Pyrimidines pharmacokinetics, Pyrimidines pharmacology, Triazoles pharmacokinetics, Triazoles pharmacology, Factor XIa drug effects, Pyrimidines administration & dosage, Triazoles administration & dosage
- Abstract
Milvexian (BMS-986177/JNJ-70033093) is a small molecule, active-site inhibitor of factor XIa (FXIa) being developed to prevent and treat thrombotic events. The safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of milvexian were assessed in a two-part, double-blind, placebo-controlled, sequential single ascending dose (SAD) and multiple ascending dose (MAD) study in healthy adults. Participants in SAD panels (6 panels of 8 participants; n = 48) were randomized (3:1) to receive milvexian (4, 20, 60, 200, 300, or 500 mg) or placebo. The 200- and 500-mg panels investigated the pharmacokinetic impact of a high-fat meal. Participants in MAD panels (7 panels of 8 participants; n = 56) were randomized (3:1) to receive milvexian (once- or twice-daily) or placebo for 14 days. All milvexian dosing regimens were safe and well-tolerated, with only mild treatment-emergent adverse events and no clinically significant bleeding events. In SAD panels, maximum milvexian plasma concentration occurred 3 h postdose in all fasted panels. The terminal half-life (T
1/2 ) ranged from 8.3 to 13.8 h. In fasted panels from 20 to 200 mg, absorption was dose-proportional; results at higher doses (300 and 500 mg) were consistent with saturable absorption. Food increased milvexian bioavailability in a dose-dependent fashion. In MAD panels, steady-state milvexian plasma concentration was reached within 3 and 6 dosing days with once- and twice-daily dosing, respectively. Renal excretion was less than 20% in all panels. Prolongation of activated partial thromboplastin time was observed and was directly related to drug exposure. These results suggest that the safety, tolerability, PK, and PD properties of milvexian are suitable for further clinical development., (© 2021 Bristol Myers Squibb. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of the American Society for Clinical Pharmacology and Therapeutics.)- Published
- 2022
- Full Text
- View/download PDF
13. Plasma biomarkers associated with adverse outcomes in patients with calcific aortic stenosis.
- Author
-
Vidula MK, Orlenko A, Zhao L, Salvador L, Small AM, Horton E, Cohen JB, Adusumalli S, Denduluri S, Kobayashi T, Hyman M, Fiorilli P, Magro C, Singh B, Pourmussa B, Greczylo C, Basso M, Ebert C, Yarde M, Li Z, Cvijic ME, Wang Z, Walsh A, Maranville J, Kick E, Luettgen J, Adam L, Schafer P, Ramirez-Valle F, Seiffert D, Moore JH, Gordon D, and Chirinos JA
- Subjects
- Biomarkers, Humans, Natriuretic Peptide, Brain, Peptide Fragments, Prognosis, Aortic Valve Stenosis, Calcinosis, Heart Failure
- Abstract
Aims: Enhanced risk stratification of patients with aortic stenosis (AS) is necessary to identify patients at high risk for adverse outcomes, and may allow for better management of patient subgroups at high risk of myocardial damage. The objective of this study was to identify plasma biomarkers and multimarker profiles associated with adverse outcomes in AS., Methods and Results: We studied 708 patients with calcific AS and measured 49 biomarkers using a Luminex platform. We studied the correlation between biomarkers and the risk of (i) death and (ii) death or heart failure-related hospital admission (DHFA). We also utilized machine-learning methods (a tree-based pipeline optimizer platform) to develop multimarker models associated with the risk of death and DHFA. In this cohort with a median follow-up of 2.8 years, multiple biomarkers were significantly predictive of death in analyses adjusted for clinical confounders, including tumour necrosis factor (TNF)-α [hazard ratio (HR) 1.28, P < 0.0001], TNF receptor 1 (TNFRSF1A; HR 1.38, P < 0.0001), fibroblast growth factor (FGF)-23 (HR 1.22, P < 0.0001), N-terminal pro B-type natriuretic peptide (NT-proBNP) (HR 1.58, P < 0.0001), matrix metalloproteinase-7 (HR 1.24, P = 0.0002), syndecan-1 (HR 1.27, P = 0.0002), suppression of tumorigenicity-2 (ST2) (IL1RL1; HR 1.22, P = 0.0002), interleukin (IL)-8 (CXCL8; HR 1.22, P = 0.0005), pentraxin (PTX)-3 (HR 1.17, P = 0.001), neutrophil gelatinase-associated lipocalin (LCN2; HR 1.18, P < 0.0001), osteoprotegerin (OPG) (TNFRSF11B; HR 1.26, P = 0.0002), and endostatin (COL18A1; HR 1.28, P = 0.0012). Several biomarkers were also significantly predictive of DHFA in adjusted analyses including FGF-23 (HR 1.36, P < 0.0001), TNF-α (HR 1.26, P < 0.0001), TNFR1 (HR 1.34, P < 0.0001), angiopoietin-2 (HR 1.26, P < 0.0001), syndecan-1 (HR 1.23, P = 0.0006), ST2 (HR 1.27, P < 0.0001), IL-8 (HR 1.18, P = 0.0009), PTX-3 (HR 1.18, P = 0.0002), OPG (HR 1.20, P = 0.0013), and NT-proBNP (HR 1.63, P < 0.0001). Machine-learning multimarker models were strongly associated with adverse outcomes (mean 1-year probability of death of 0%, 2%, and 60%; mean 1-year probability of DHFA of 0%, 4%, 97%; P < 0.0001). In these models, IL-6 (a biomarker of inflammation) and FGF-23 (a biomarker of calcification) emerged as the biomarkers of highest importance., Conclusions: Plasma biomarkers are strongly associated with the risk of adverse outcomes in patients with AS. Biomarkers of inflammation and calcification were most strongly related to prognosis., (© 2021 European Society of Cardiology.)
- Published
- 2021
- Full Text
- View/download PDF
14. Antithrombotic Effects of Combined PAR (Protease-Activated Receptor)-4 Antagonism and Factor Xa Inhibition.
- Author
-
Meah MN, Raftis J, Wilson SJ, Perera V, Garonzik SM, Murthy B, Everlof JG, Aronson R, Luettgen J, and Newby DE
- Subjects
- Adult, Blood Platelets metabolism, Double-Blind Method, Drug Therapy, Combination, Factor Xa Inhibitors pharmacokinetics, Female, Fibrinolytic Agents pharmacokinetics, Humans, Male, Pyrazoles pharmacokinetics, Pyridones pharmacokinetics, Receptors, Thrombin blood, Signal Transduction, Thrombosis blood, Young Adult, Blood Platelets drug effects, Factor Xa Inhibitors administration & dosage, Fibrinolytic Agents administration & dosage, Platelet Aggregation drug effects, Pyrazoles administration & dosage, Pyridones administration & dosage, Receptors, Thrombin antagonists & inhibitors, Thrombosis prevention & control
- Abstract
Objective: PAR (protease-activated receptor)-4 antagonism has antiplatelet effects under conditions of high shear stress. We aimed to establish whether PAR4 antagonism had additive antithrombotic activity in the presence of factor Xa inhibition in an ex vivo model of acute arterial injury. Approach and Results: Fifteen healthy volunteers (29±6 years, 7 women) completed a phase zero double-blind randomized controlled crossover trial. Ex vivo platelet activation, platelet aggregation, and thrombus formation were measured following blood perfusion of low shear and high shear stress chambers. Upstream of the chambers, extracorporeal blood was admixed with (1) vehicle, (2) low-dose apixaban (20 ng/mL), (3) high-dose apixaban (80 ng/mL), (4) BMS-986141 (400 ng/mL), (5) BMS-968141 and low-dose apixaban, or (6) BMS-968141 and high-dose apixaban in 6 sequential studies performed in random order. Compared with vehicle, BMS-986141 demonstrated selective inhibition of PAR4-AP (agonist peptide)-stimulated platelet aggregation, platelet-monocyte aggregates, and P-selectin expression ( P ≤0.01 for all). Total thrombus area was reduced under both low shear and high shear stress conditions for all drug infusions ( P <0.0001 for all versus vehicle). BMS-968141 reduced total (≤44.4%) and platelet-rich (≤39.3%) thrombus area, whereas apixaban reduced total (≤42.9%) and fibrin-rich (≤31.6%) thrombus area. Combination of BMS-986141 with apixaban caused a further modest reduction in total thrombus area (9.6%-12.4%), especially under conditions of high shear stress ( P ≤0.027)., Conclusions: In the presence of factor Xa inhibition, PAR4 antagonism with BMS-986141 further reduces thrombus formation, especially under conditions of high shear stress. This suggests the potential for additive efficacy of combination PAR4 antagonism and factor Xa inhibition in the prevention of atherothrombotic events.
- Published
- 2020
- Full Text
- View/download PDF
15. Inhibitory effect of apixaban compared with rivaroxaban and dabigatran on thrombin generation assay.
- Author
-
Wong PC, White A, and Luettgen J
- Subjects
- Anticoagulants pharmacology, Anticoagulants therapeutic use, Antithrombins pharmacology, Antithrombins therapeutic use, Benzimidazoles therapeutic use, Dabigatran, Humans, Morpholines therapeutic use, Pyrazoles therapeutic use, Pyridones therapeutic use, Rivaroxaban, Thiophenes therapeutic use, Thrombin analysis, Thrombin Time, beta-Alanine pharmacology, beta-Alanine therapeutic use, Benzimidazoles pharmacology, Morpholines pharmacology, Pyrazoles pharmacology, Pyridones pharmacology, Thiophenes pharmacology, Thrombin biosynthesis, beta-Alanine analogs & derivatives
- Abstract
The effect of the oral direct activated factor X (factor Xa) inhibitor apixaban on tissue factor-induced thrombin generation in human plasma was investigated in vitro using the calibrated automated thrombogram (CAT) method and compared with the oral direct factor Xa inhibitor rivaroxaban and the direct thrombin inhibitor dabigatran. Pooled citrated, anticoagulated, platelet-poor human plasma was spiked with apixaban, rivaroxaban, or dabigatran at concentrations of 0.01 to 10 μM. The inhibitory potencies of the compounds were quantified by 5 CAT parameters: the control thrombin lag time (LT) and time to thrombin peak (TTP) for the doubling of inhibitor concentration (IC2x); and the control endogenous thrombin potential (ETP), thrombin peak, and maximum rate of thrombin generation (Vmax) for the inhibitor concentration, which inhibited 50% (IC50). The inhibitors modified CAT concentration dependently. Their inhibitory potencies, expressed as IC2x LT, IC2x TTP, IC50 ETP, IC50 peak thrombin, and IC50 Vmax, were as follows: 0.10 ± 0.01, 0.19 ± 0.02, 0.65 ± 0.11, 0.089 ± 0.019, and 0.049 ± 0.007 μM for apixaban; 0.049 ± 0.007, 0.070 ± 0.009, 0.43 ± 0.07, 0.048 ± 0.008, and 0.022 ± 0.005 μM for rivaroxaban; and 0.063 ± 0.019, 0.18 ± 0.06, 0.50 ± 0.08, 0.55 ± 0.06, and 0.57 ± 0.27 μM for dabigatran. In summary, apixaban, rivaroxaban, and dabigatran have similar potencies in the prolongation of LT and TTP. The CAT parameters that are related to the rate of thrombin generation during the propagation phase (ie, peak thrombin and Vmax) are more sensitive to activities of apixaban and rivaroxaban than dabigatran. The ETP is the least sensitive parameter for measuring the activities of these inhibitors. Recombinant activated factor VII at 5 and 50 μg/mL reversed the anticoagulant effects of apixaban more at 0.2 μM than at 2 μM. Our study suggests that the CAT method is a sensitive assay to monitor the pharmacodynamic and pharmacokinetic properties of apixaban, rivaroxaban, and dabigatran, and may provide insight into the mechanism of action of these inhibitors. Recombinant activated factor VII may have some potential to reverse the anticoagulant effects of apixaban in vitro.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.