Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Pharm Res ; 39(2): 213-222, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35112229

ABSTRACT

The Free Drug Hypothesis is a well-established concept within the scientific lexicon pervading many areas of Drug Discovery and Development, and yet it is poorly defined by virtue of many variations appearing in the literature. Clearly, unbound drug is in dynamic equilibrium with respect to absorption, distribution, metabolism, elimination, and indeed, interaction with the desired pharmacological target. Binding interactions be they specific (e.g. high affinity) or nonspecific (e.g. lower affinity/higher capacity) are governed by the same fundamental physicochemical tenets including Hill-Langmuir Isotherms, the Law of Mass Action and Drug Receptor Theory. With this in mind, it is time to recognise a more coherent version and consider it the Free Drug Theory and a hypothesis no longer. Today, we have the experimental and modelling capabilities, pharmacological knowledge, and an improved understanding of unbound drug distribution (e.g. Kpuu) to raise the bar on our understanding and analysis of experimental data. The burden of proof should be to rule out mechanistic possibilities and/or experimental error before jumping to the conclusion that any observations contradict these fundamentals.


Subject(s)
Drug Development , Drug Discovery , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Humans , Molecular Targeted Therapy , Network Pharmacology , Pharmaceutical Preparations/blood , Protein Binding , Signal Transduction
2.
Crit Care ; 21(1): 234, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28877748

ABSTRACT

BACKGROUND: Renin-angiotensin system (RAS) signaling and angiotensin-converting enzyme 2 (ACE2) have been implicated in the pathogenesis of acute respiratory distress syndrome (ARDS). We postulated that repleting ACE2 using GSK2586881, a recombinant form of human angiotensin-converting enzyme 2 (rhACE2), could attenuate acute lung injury. METHODS: We conducted a two-part phase II trial comprising an open-label intrapatient dose escalation and a randomized, double-blind, placebo-controlled phase in ten intensive care units in North America. Patients were between the ages of 18 and 80 years, had an American-European Consensus Criteria consensus diagnosis of ARDS, and had been mechanically ventilated for less than 72 h. In part A, open-label GSK2586881 was administered at doses from 0.1 mg/kg to 0.8 mg/kg to assess safety, pharmacokinetics, and pharmacodynamics. Following review of data from part A, a randomized, double-blind, placebo-controlled investigation of twice-daily doses of GSK2586881 (0.4 mg/kg) for 3 days was conducted (part B). Biomarkers, physiological assessments, and clinical endpoints were collected over the dosing period and during follow-up. RESULTS: Dose escalation in part A was well-tolerated without clinically significant hemodynamic changes. Part B was terminated after 39 of the planned 60 patients following a planned futility analysis. Angiotensin II levels decreased rapidly following infusion of GSK2586881, whereas angiotensin-(1-7) and angiotensin-(1-5) levels increased and remained elevated for 48 h. Surfactant protein D concentrations were increased, whereas there was a trend for a decrease in interleukin-6 concentrations in rhACE2-treated subjects compared with placebo. No significant differences were noted in ratio of partial pressure of arterial oxygen to fraction of inspired oxygen, oxygenation index, or Sequential Organ Failure Assessment score. CONCLUSIONS: GSK2586881 was well-tolerated in patients with ARDS, and the rapid modulation of RAS peptides suggests target engagement, although the study was not powered to detect changes in acute physiology or clinical outcomes. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01597635 . Registered on 26 January 2012.


Subject(s)
Peptidyl-Dipeptidase A/pharmacology , Respiratory Distress Syndrome/drug therapy , Adult , Aged , Angiotensin-Converting Enzyme 2 , Blood Gas Analysis/statistics & numerical data , Double-Blind Method , Female , Humans , Intensive Care Units/organization & administration , Male , Middle Aged , North America , Peptidyl-Dipeptidase A/therapeutic use , Pilot Projects , Placebos
3.
Clin Transl Sci ; 15(3): 588-600, 2022 03.
Article in English | MEDLINE | ID: mdl-34716976

ABSTRACT

Translational model-based approaches have played a role in increasing success in the development of novel anticancer treatments. However, despite this, significant translational uncertainty remains from animal models to patients. Optimization of dose and scheduling (regimen) of drugs to maximize the therapeutic utility (maximize efficacy while avoiding limiting toxicities) is still predominately driven by clinical investigations. Here, we argue that utilizing pragmatic mechanism-based translational modeling of nonclinical data can further inform this optimization. Consequently, a prototype model is demonstrated that addresses the required fundamental mechanisms.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/therapeutic use , Humans , Medical Oncology , Neoplasms/chemically induced , Neoplasms/drug therapy
4.
Circ Res ; 93(3): e26-32, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12869391

ABSTRACT

Trimetazidine acts as an effective antianginal clinical agent by modulating cardiac energy metabolism. Recent published data support the hypothesis that trimetazidine selectively inhibits long-chain 3-ketoacyl CoA thiolase (LC 3-KAT), thereby reducing fatty acid oxidation resulting in clinical benefit. The aim of this study was to assess whether trimetazidine and ranolazine, which may also act as a metabolic modulator, are specific inhibitors of LC 3-KAT. We have demonstrated that trimetazidine and ranolazine do not inhibit crude and purified rat heart or recombinant human LC 3-KAT by methods that both assess the ability of LC 3-KAT to turnover specific substrate, and LC 3-KAT activity as a functional component of intact cellular beta-oxidation. Furthermore, we have demonstrated that trimetazidine does not inhibit any component of beta-oxidation in an isolated human cardiomyocyte cell line. Ranolazine, however, did demonstrate a partial inhibition of beta-oxidation in a dose-dependent manner (12% at 100 micromol/L and 30% at 300 micromol/L). Both trimetazidine (10 micromol/L) and ranolazine (20 micromol/L) improved the recovery of cardiac function after a period of no flow ischemia in the isolated working rat heart perfused with a buffer containing a relatively high concentration (1.2 mmol/L) of free fatty acid. In summary, both trimetazidine and ranolazine were able to improve ischemic cardiac function but inhibition of LC 3-KAT is not part of their mechanism of action. The full text of this article is available online at http://www.circresaha.org.


Subject(s)
Acetyl-CoA C-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mitochondria, Heart/enzymology , Trimetazidine/pharmacology , Vasodilator Agents/pharmacology , Acetanilides , Acetyl-CoA C-Acyltransferase/isolation & purification , Acetyl-CoA C-Acyltransferase/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Heart/drug effects , Heart/physiopathology , Humans , In Vitro Techniques , Male , Mitochondria, Heart/chemistry , Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , Myocardium/enzymology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Oxidation-Reduction/drug effects , Piperazines/pharmacology , Ranolazine , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
5.
J Med Chem ; 54(22): 7797-814, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21888439

ABSTRACT

This paper describes the identification and optimization of a novel series of DFG-out binding p38 inhibitors as inhaled agents for the treatment of chronic obstructive pulmonary disease. Structure based drug design and "inhalation by design" principles have been applied to the optimization of the lead series exemplied by compound 1a. Analogues have been designed to be potent and selective for p38, with an emphasis on slow enzyme dissociation kinetics to deliver prolonged lung p38 inhibition. Pharmacokinetic properties were tuned with high intrinsic clearance and low oral bioavailability in mind, to minimize systemic exposure and reduce systemically driven adverse events. High CYP mediated clearance and glucuronidation were targeted to achieve high intrinsic clearance coupled with multiple routes of clearance to minimize drug-drug interactions. Furthermore, pharmaceutical properties such as stability, crystallinity, and solubility were considered to ensure compatibility with a dry powder inhaler. 1ab (PF-03715455) was subsequently identified as a clinical candidate from this series with efficacy and safety profiles confirming its potential as an inhaled agent for the treatment of COPD.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Azabicyclo Compounds/chemical synthesis , Methylurea Compounds/chemical synthesis , Pulmonary Disease, Chronic Obstructive/drug therapy , Pyrazoles/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Administration, Inhalation , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azabicyclo Compounds/pharmacokinetics , Azabicyclo Compounds/pharmacology , Binding Sites , Cell Membrane Permeability , Crystallography, X-Ray , Dogs , Drug Stability , Humans , In Vitro Techniques , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Methylurea Compounds/pharmacokinetics , Methylurea Compounds/pharmacology , Models, Molecular , Protein Binding , Protein Conformation , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats , Solubility , Surface Plasmon Resonance , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/chemistry
6.
J Med Chem ; 54(19): 6888-904, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21870878

ABSTRACT

A novel tertiary amine series of potent muscarinic M(3) receptor antagonists are described that exhibit potential as inhaled long-acting bronchodilators for the treatment of chronic obstructive pulmonary disease. Geminal dimethyl functionality present in this series of compounds confers very long dissociative half-life (slow off-rate) from the M(3) receptor that mediates very long-lasting smooth muscle relaxation in guinea pig tracheal strips. Optimization of pharmacokinetic properties was achieved by combining rapid oxidative clearance with targeted introduction of a phenolic moiety to secure rapid glucuronidation. Together, these attributes minimize systemic exposure following inhalation, mitigate potential drug-drug interactions, and reduce systemically mediated adverse events. Compound 47 (PF-3635659) is identified as a Phase II clinical candidate from this series with in vivo duration of action studies confirming its potential for once-daily use in humans.


Subject(s)
Azetidines/chemical synthesis , Bronchodilator Agents/chemical synthesis , Diphenylacetic Acids/chemical synthesis , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptor, Muscarinic M3/antagonists & inhibitors , Administration, Inhalation , Animals , Azetidines/chemistry , Azetidines/pharmacology , Bronchodilator Agents/chemistry , Bronchodilator Agents/pharmacology , CHO Cells , Cell Line , Cell Membrane Permeability , Cricetinae , Cricetulus , Diphenylacetic Acids/chemistry , Diphenylacetic Acids/pharmacology , Dogs , Female , Guinea Pigs , Hepatocytes/metabolism , Humans , In Vitro Techniques , Kinetics , Male , Microsomes, Liver/metabolism , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Radioligand Assay , Rats , Receptor, Muscarinic M3/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship , Trachea/drug effects , Trachea/physiology
SELECTION OF CITATIONS
SEARCH DETAIL