Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Pharm Sci ; 110(7): 2728-2732, 2021 07.
Article in English | MEDLINE | ID: mdl-33631170

ABSTRACT

In Drug Discovery, pharmacology studies often require benign formulation compositions for safe administration in animal models. Here, we applied Adaptive Focused Acoustics™ (AFA) to a molecular scaffold with challenging physicochemical properties for intraperitoneal administration. Nanosuspensions can be prepared at small scales and provide broad applicability. Our results show that nanosuspension formulations prepared by AFA have improved PK performance relative to a DMSO solution formulation that is prone to precipitation in-vivo.


Subject(s)
Nanoparticles , Animals , Particle Size , Solubility , Suspensions
2.
ACS Med Chem Lett ; 8(2): 261-265, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28197323

ABSTRACT

JNJ-63623872 (2) is a first-in-class, orally bioavailable compound that offers significant potential for the treatment of pandemic and seasonal influenza. Early lead optimization efforts in our 7-azaindole series focused on 1,3-diaminocyclohexyl amide and urea substitutions on the pyrimidine-7-azaindole motif. In this work, we explored two strategies to eliminate observed aldehyde oxidase (AO)-mediated metabolism at the 2-position of these 7-azaindole analogues. Substitution at the 2-position of the azaindole ring generated somewhat less potent analogues, but reduced AO-mediated metabolism. Incorporation of a ring nitrogen generated 7-azaindazole analogues that were equipotent to the parent 2-H-7-azaindole, but surprisingly, did not appear to improve AO-mediated metabolism. Overall, we identified multiple 2-substituted 7-azaindole analogues with enhanced AO stability and we present data for one such compound (12) that demonstrate a favorable oral pharmacokinetic profile in rodents. These analogues have the potential to be further developed as anti-influenza agents for the treatment of influenza.

3.
ACS Med Chem Lett ; 8(2): 256-260, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28197322

ABSTRACT

In our efforts to develop novel small-molecule inhibitors for the treatment of influenza, we utilized molecular modeling and the X-ray crystal structure of the PB2 subunit of the influenza polymerase to optimize a series of acyclic ß-amino acid inhibitors, highlighted by compound 4. Compound 4 showed good oral exposure in both rat and mouse. More importantly, it showed strong potency versus multiple influenza-A strains, including pandemic 2009 H1N1 and avian H5N1 strains and showed a strong efficacy profile in a mouse influenza model even when treatment was initiated 48 h after infection. Compound 4 offers good oral bioavailability with great potential for the treatment of both pandemic and seasonal influenza.

4.
Antimicrob Agents Chemother ; 59(10): 6007-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26169418

ABSTRACT

Through antigenic drift and shifts, influenza virus infections continue to be an annual cause of morbidity in healthy populations and of death among elderly and at-risk patients. The emergence of highly pathogenic avian influenza viruses such as H5N1 and H7N9 and the rapid spread of the swine-origin H1N1 influenza virus in 2009 demonstrate the continued need for effective therapeutic agents for influenza. While several neuraminidase inhibitors have been developed for the treatment of influenza virus infections, these have shown a limited window for treatment initiation, and resistant variants have been noted in the population. In addition, an older class of antiviral drugs for influenza, the adamantanes, are no longer recommended for treatment due to widespread resistance. There remains a need for new influenza therapeutic agents with improved efficacy as well as an expanded window for the initiation of treatment. Azaindole compounds targeting the influenza A virus PB2 protein and demonstrating excellent in vitro and in vivo properties have been identified. To evaluate the in vivo efficacy of these PB2 inhibitors, we utilized a mouse influenza A virus infection model. In addition to traditional endpoints, i.e., death, morbidity, and body weight loss, we measured lung function using whole-body plethysmography, and we used these data to develop a composite efficacy score that takes compound exposure into account. This model allowed the rapid identification and ranking of molecules relative to each other and to oseltamivir. The ability to identify compounds with enhanced preclinical properties provides an opportunity to develop more-effective treatments for influenza in patients.


Subject(s)
Antiviral Agents/pharmacology , Aza Compounds/pharmacology , Indoles/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Research Design , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Aza Compounds/chemical synthesis , Aza Compounds/pharmacokinetics , Drug Evaluation, Preclinical , Drug Resistance, Viral , Gene Expression , Indoles/chemical synthesis , Indoles/pharmacokinetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology , Respiratory Function Tests , Survival Analysis , Viral Proteins/genetics , Viral Proteins/metabolism
5.
J Med Chem ; 57(15): 6668-78, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25019388

ABSTRACT

In our effort to develop agents for the treatment of influenza, a phenotypic screening approach utilizing a cell protection assay identified a series of azaindole based inhibitors of the cap-snatching function of the PB2 subunit of the influenza A viral polymerase complex. Using a bDNA viral replication assay (Wagaman, P. C., Leong, M. A., and Simmen, K. A. Development of a novel influenza A antiviral assay. J. Virol. Methods 2002, 105, 105-114) in cells as a direct measure of antiviral activity, we discovered a set of cyclohexyl carboxylic acid analogues, highlighted by VX-787 (2). Compound 2 shows strong potency versus multiple influenza A strains, including pandemic 2009 H1N1 and avian H5N1 flu strains, and shows an efficacy profile in a mouse influenza model even when treatment was administered 48 h after infection. Compound 2 represents a first-in-class, orally bioavailable, novel compound that offers potential for the treatment of both pandemic and seasonal influenza and has a distinct advantage over the current standard of care treatments including potency, efficacy, and extended treatment window.


Subject(s)
Antiviral Agents/chemistry , Aza Compounds/chemistry , Indoles/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Aza Compounds/chemical synthesis , Aza Compounds/pharmacology , Biological Availability , Dogs , Drug Resistance, Viral , Indoles/chemical synthesis , Indoles/pharmacology , Influenza A virus/drug effects , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Models, Molecular , Molecular Structure , Orthomyxoviridae Infections/drug therapy , Rats , Species Specificity , Stereoisomerism , Structure-Activity Relationship , Virus Replication/drug effects
6.
J Pharm Sci ; 101(9): 3134-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22374830

ABSTRACT

The purpose of this work was to study the permeability of two relatively lipophilic sulfenamide prodrugs of linezolid (clogP 0.85), N-(phenylthio)linezolid (1, clogP 2.77) and N-[(2-ethoxycarbonyl)ethylthio]linezolid (2, clogP 1.43), across Caco-2 cell monolayers. Both prodrugs were found to convert to linezolid in the donor compartment presumably from the reaction with free thiol groups on proteins on the surface of the Caco-2 cells, as no conversion was seen in the donor compartment media per se. Neither of the prodrugs could be detected in the receptor phase from either apical (AP) to basolateral (BL) or BL to AP studies. However, the appearance of linezolid in the receptor phase was biphasic with an initial rapid phase suggesting that the prodrugs were indeed more permeable, and for a short period, some prodrug was able to permeate in competition with conversion to linezolid on the donor phase surface. It appears that the prodrug was able to permeate was rapidly converted to linezolid prior to acceptor phase appearance. The second slower phase was due to the permeability of the donor-phase-formed linezolid, with the slopes similar to those from control experiments with linezolid. The limitations and possible utility of oral sulfenamide prodrugs are discussed.


Subject(s)
Acetamides/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Oxazolidinones/metabolism , Prodrugs/metabolism , Sulfamerazine/metabolism , Acetamides/chemistry , Caco-2 Cells , Humans , Kinetics , Linezolid , Models, Biological , Oxazolidinones/chemistry , Permeability , Prodrugs/chemistry , Sulfamerazine/chemistry
7.
J Pharm Sci ; 100(7): 3023-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21547913

ABSTRACT

The purpose of this work was to study the reaction kinetics between two model sulfenamide prodrugs of linezolid, N-(phenylthio)linezolid and N-[(2-ethoxycarbonyl)ethylthio]linezolid, with free thiol-containing proteins; commercial human serum albumin (HSA); a constitutively active mutant of the protein tyrosine phosphatase PRL-1 (PRL-1-C170S-C171S), a model protein; and diluted fresh human plasma. The reaction was followed by high-performance liquid chromatography, both for the loss of prodrug and appearance of linezolid, and at different pH values with molar excess of the proteins relative to the prodrugs. Pseudo first-order kinetics was observed. Consistent with earlier findings for the reaction between similar sulfenamides and small-molecule thiols, the reaction kinetics appeared to be consistent with thiolate attack at the sulfenamide bond to release the parent drug. The proteins reacted significantly slower on a molar basis than their small-molecule counterparts. It appears that proteins such as HSA may play a role in the in vivo conversion of sulfenamide prodrugs to their parent drug.


Subject(s)
Acetamides/chemistry , Cell Cycle Proteins/chemistry , Membrane Proteins/chemistry , Oxazolidinones/chemistry , Prodrugs/chemistry , Protein Tyrosine Phosphatases/chemistry , Serum Albumin/chemistry , Sulfamerazine/chemistry , Sulfhydryl Compounds/chemistry , Cell Cycle Proteins/genetics , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Drug Stability , Humans , Hydrogen-Ion Concentration , Kinetics , Linezolid , Membrane Proteins/genetics , Mutation , Plasma/chemistry , Protein Tyrosine Phosphatases/genetics , Technology, Pharmaceutical/methods
8.
Bioorg Med Chem Lett ; 21(9): 2780-3, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21513867

ABSTRACT

The objective of this Letter is both to report the permeability results of a linezolid-based sulfenamide prodrug in an MDCK cell model (enterocyte surrogate system) and to discuss the strategic implications of these results for considering sulfenamide prodrugs to enhance the oral delivery of weakly acidic NH-acids (e.g., amides, ureas, etc.). The two main findings from this study are that the sulfenamide prodrug does not appear to survive intracellular transport due to conversion to linezolid and that there appears to be an apically-oriented surface conversion pathway that can additionally serve to convert the sulfenamide prodrug to linezolid upon approach of the apical membrane. It is hoped that these findings, along with the discussion of the strategic implications, will facilitate a greater awareness of the potential strengths and weaknesses inherent in the sulfenamide prodrug approach for enhancing the oral delivery of weakly acidic NH-acid drugs.

9.
J Pharm Sci ; 100(3): 1001-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20957747

ABSTRACT

Sulfenamide prodrugs of amide and urea functional group containing drugs have recently been proposed as a means of altering the physical and bioproperties of problematic drug molecules containing these two functionalities. Sulfenamides have been shown to revert to the parent drug via reaction with thiols. Explored here is the mechanism for this reaction. The stoichiometry and pH dependency of the in vitro reversion of two model prodrugs of the oxazolidinone, linezolid, and a sulfenamide of phthalimide were studied at 25 °C in the presence of thiols, including cysteine and glutathione, of varying basicity. High-performance/pressure liquid chromatography and liquid chromatography-mass spectrometry results showed the near quantitative reversion of the sulfenamides to the parent drug with simultaneous formation of a mixed disulfide. The pH and the dependency of the reaction on the basicity of the thiol strongly supported the role of the thiolate species in the conversion. The reaction is consistent with an S(N)2 type mechanism seen in the reaction of some thiols with disulfides.


Subject(s)
Prodrugs/chemistry , Sulfamerazine/chemistry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Cysteine/chemistry , Disulfides/analysis , Disulfides/chemistry , Drug Stability , Glutathione/chemistry , Hydrogen-Ion Concentration , Mass Spectrometry , Prodrugs/pharmacokinetics , Sulfamerazine/pharmacokinetics , Sulfhydryl Compounds/chemistry
10.
Bioorg Med Chem Lett ; 21(1): 172-5, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21126873

ABSTRACT

The objective of this Letter is both to report the permeability results of a linezolid-based sulfenamide prodrug in an MDCK cell model (enterocyte surrogate system) and to discuss the strategic implications of these results for considering sulfenamide prodrugs to enhance the oral delivery of weakly acidic NH-acids (e.g., amides, ureas, etc.). The two main findings from this study are that the sulfenamide prodrug does not appear to survive intracellular transport due to conversion to linezolid and that there appears to be an apically-oriented surface conversion pathway that can additionally serve to convert the sulfenamide prodrug to linezolid upon approach of the apical membrane. It is hoped that these findings, along with the discussion of the strategic implications, will facilitate a greater awareness of the potential strengths and weaknesses inherent in the sulfenamide prodrug approach for enhancing the oral delivery of weakly acidic NH-acid drugs.


Subject(s)
Prodrugs/chemistry , Prodrugs/metabolism , Sulfamerazine/chemistry , Sulfamerazine/metabolism , Acids/administration & dosage , Administration, Oral , Animals , Cell Line , Cell Membrane Permeability , Dogs , Prodrugs/chemical synthesis , Sulfamerazine/chemical synthesis
11.
Bioorg Med Chem Lett ; 17(23): 6629-32, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17928225

ABSTRACT

Improved synthetic methods are reported for the preparation of sulfenamide derivatives of carbamazepine (CBZ) for evaluation as prodrugs. These sulfenamide prodrugs were designed to rapidly release CBZ in vivo by cleavage of the sulfenamide bond by chemical reaction with glutathione and other sulfhydryl compounds. Physicochemical characterization and in vivo conversion of a new prodrug of CBZ was evaluated to further establish the proof of concept of the sulfenamide prodrug approach.


Subject(s)
Carbamazepine/chemical synthesis , Carbamazepine/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism , Sulfamerazine/chemical synthesis , Sulfamerazine/metabolism , Water/chemistry , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/chemical synthesis , Anticonvulsants/metabolism , Carbamazepine/administration & dosage , Glutathione/metabolism , Models, Chemical , Prodrugs/administration & dosage , Rats , Solubility , Sulfamerazine/administration & dosage
12.
Adv Drug Deliv Rev ; 59(7): 677-94, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17628203

ABSTRACT

Drug design in recent years has attempted to explore new chemical spaces resulting in more complex, larger molecular weight molecules, often with limited water solubility. To deliver molecules with these properties, pharmaceutical scientists have explored many different techniques. An older but time-tested strategy is the design of bioreversible, more water-soluble derivatives of the problematic molecule, or prodrugs. This review explores the use of prodrugs to effect improved oral and parenteral delivery of poorly water-soluble problematic drugs, using both marketed as well as investigational prodrugs as examples. Prodrug interventions should be considered early in the drug discovery paradigm rather than as a technique of last resort. Their importance is supported by the increasing percentage of approved new drug entities that are, in fact, prodrugs.


Subject(s)
Drug Design , Prodrugs/chemistry , Chemistry, Pharmaceutical , Drug Administration Routes , Prodrugs/administration & dosage , Solubility , Structure-Activity Relationship , Water
SELECTION OF CITATIONS
SEARCH DETAIL
...