Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Pharm Anal ; 8(5): 302-306, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30345143

ABSTRACT

Freeze-thaw cycles impact the amount of aggregation observed in antibodies and Fc-fusion proteins. Various formulation strategies are used to mitigate the amount of aggregation that occurs upon putting a protein solution through a freeze-thaw cycle. Additionally, low pH solutions cause native antibodies to unfold, which are prone to aggregate upon pH neutralization. There is great interest in the mechanism that causes therapeutic proteins to aggregate since aggregate species can cause unwanted immunogenicity in patients. Herein, an increase in aggregation is reported when the pH is adjusted from pH 3 up to a pH ranging from pH 4 to pH 7 during the thaw process of a frozen antibody solution. Raising the pH during the thaw process caused a significant increase in the percent aggregation observed. Two antibodies and one Fc-fusion protein were evaluated during the pH jump thaw process and similar effects were observed. The results provide a new tool to study the kinetics of therapeutic protein aggregation upon pH increase.

2.
ChemMedChem ; 12(23): 1994-2005, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29105342

ABSTRACT

Kinetoplastid-based infections are neglected diseases that represent a significant human health issue. Chemotherapeutic options are limited due to toxicity, parasite susceptibility, and poor patient compliance. In response, we studied a molecular-target-directed approach involving intervention of hexokinase activity-a pivotal enzyme in parasite metabolism. A benzamidobenzoic acid hit with modest biochemical inhibition of Trypanosoma brucei hexokinase 1 (TbHK1, IC50 =9.1 µm), low mammalian cytotoxicity (IMR90 cells, EC50 >25 µm), and no appreciable activity on whole bloodstream-form (BSF) parasites was optimized to afford a probe with improved TbHK1 potency and, significantly, efficacy against whole BSF parasites (TbHK1, IC50 =0.28 µm; BSF, ED50 =1.9 µm). Compounds in this series also inhibited the hexokinase enzyme from Leishmania major (LmHK1), albeit with less potency than toward TbHK1, suggesting that inhibition of the glycolytic pathway may be a promising opportunity to target multiple disease-causing trypanosomatid protozoa.


Subject(s)
Antiparasitic Agents/pharmacology , Benzamides/pharmacology , Benzoates/pharmacology , Enzyme Inhibitors/pharmacology , Hexokinase/antagonists & inhibitors , Antiparasitic Agents/chemical synthesis , Antiparasitic Agents/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Benzoates/chemical synthesis , Benzoates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hexokinase/metabolism , Leishmania major/drug effects , Leishmania major/enzymology , Leishmania major/growth & development , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/growth & development
3.
SLAS Discov ; 22(1): 21-31, 2017 01.
Article in English | MEDLINE | ID: mdl-27628691

ABSTRACT

Although there has been substantial success in the development of specific inhibitors for protein kinases, little progress has been made in the identification of specific inhibitors for their protein phosphatase counterparts. Inhibitors of PP1 and PP5 are desired as probes for research and to test their potential for drug development. We developed and miniaturized (1536-well plate format) nearly identical homogeneous, fluorescence intensity (FLINT) enzymatic assays to detect inhibitors of PP1 or PP5. The assays were used in an ultra-high-throughput screening (uHTS) campaign, testing >315,000 small-molecule compounds. Both assays demonstrated robust performance, with a Z' of 0.92 ± 0.03 and 0.95 ± 0.01 for the PP1 and PP5 assays, respectively. Screening the same library with both assays aided the identification of class inhibitors and assay artifacts. Confirmation screening and hit prioritization assays used [32P/33P]-radiolabel protein substrates, revealing excellent agreement between the FLINT and radiolabel assays. This screening campaign led to the discovery of four novel unrelated small-molecule inhibitors of PP1 and ~30 related small-molecule inhibitors of PP5. The results suggest that this uHTS approach is suitable for identifying selective chemical probes that inhibit PP1 or PP5 activity, and it is likely that similar assays can be developed for other PPP-family phosphatases.


Subject(s)
Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Protein Phosphatase 1/antagonists & inhibitors , Catalysis , Enzyme Assays , Enzyme Inhibitors/chemistry , Humans , Miniaturization , Phosphoproteins/metabolism , Protein Phosphatase 1/metabolism , Radiopharmaceuticals/chemistry , Reproducibility of Results , Substrate Specificity
4.
Antimicrob Agents Chemother ; 60(8): 4552-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27185801

ABSTRACT

Viral emergence and reemergence underscore the importance of developing efficacious, broad-spectrum antivirals. Here, we report the discovery of tetrahydrobenzothiazole-based compound 1, a novel, broad-spectrum antiviral lead that was optimized from a hit compound derived from a cytopathic effect (CPE)-based antiviral screen using Venezuelan equine encephalitis virus. Compound 1 showed antiviral activity against a broad range of RNA viruses, including alphaviruses, flaviviruses, influenza virus, and ebolavirus. Mechanism-of-action studies with metabolomics and molecular approaches revealed that the compound inhibits host pyrimidine synthesis and establishes an antiviral state by inducing a variety of interferon-stimulated genes (ISGs). Notably, the induction of the ISGs by compound 1 was independent of the production of type 1 interferons. The antiviral activity of compound 1 was cell type dependent with a robust effect observed in human cell lines and no observed antiviral effect in mouse cell lines. Herein, we disclose tetrahydrobenzothiazole compound 1 as a novel lead for the development of a broad-spectrum, antiviral therapeutic and as a molecular probe to study the mechanism of the induction of ISGs that are independent of type 1 interferons.


Subject(s)
Antiviral Agents/pharmacology , Interferon Type I/metabolism , Pyrimidines/biosynthesis , Cell Line , HIV-1/drug effects , Humans , Real-Time Polymerase Chain Reaction , Virus Replication/drug effects
5.
Org Biomol Chem ; 14(16): 3950-5, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27050086

ABSTRACT

The efficient generation of novel, N-linked benzamidines resulting from a regiospecific rearrangement of quinazolinones is described. This methodology study explored reaction parameters including the effect of changing solvent and temperature, as well as varying electronic substituents on the structural core. The transformation was extensively optimized in terms of reaction conditions and scope, resulting in a protocol that consistently affords diversely functionalized amidines in high yield. The process permits regional structural derivatization that was previously inaccessible, and the multistep process was also reduced to a telescoped, five-step sequence that efficiently affords pharmacologically unique (E)-benzamidoamidines from N-BOC protected γ- and δ-amino acids.


Subject(s)
Amino Acids/chemistry , Benzamidines/chemistry , Quinazolinones/chemistry
6.
Biochem Pharmacol ; 109: 14-26, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27002182

ABSTRACT

Cantharidin is a natural toxin and an active constituent in a traditional Chinese medicine used to treat tumors. Cantharidin acts as a semi-selective inhibitor of PPP-family ser/thr protein phosphatases. Despite sharing a common catalytic mechanism and marked structural similarity with PP1C, PP2AC and PP5C, human PP4C was found to be insensitive to the inhibitory activity of cantharidin. To explore the molecular basis for this selectivity, we synthesized and tested novel C5/C6-derivatives designed from quantum-based modeling of the interactions revealed in the co-crystal structures of PP5C in complex with cantharidin. Structure-activity relationship studies and analysis of high-resolution (1.25Å) PP5C-inhibitor co-crystal structures reveal close contacts between the inhibitor bridgehead oxygen and both a catalytic metal ion and a non-catalytic phenylalanine residue, the latter of which is substituted by tryptophan in PP4C. Quantum chemistry calculations predicted that steric clashes with the bulkier tryptophan side chain in PP4C would force all cantharidin-based inhibitors into an unfavorable binding mode, disrupting the strong coordination of active site metal ions observed in the PP5C co-crystal structures, thereby rendering PP4C insensitive to the inhibitors. This prediction was confirmed by inhibition studies employing native human PP4C. Mutation of PP5C (F446W) and PP1C (F257W), to mimic the PP4C active site, resulted in markedly suppressed sensitivity to cantharidin. These observations provide insight into the structural basis for the natural selectivity of cantharidin and provide an avenue for PP4C deselection. The novel crystal structures also provide insight into interactions that provide increased selectivity of the C5/C6 modifications for PP5C versus other PPP-family phosphatases.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cantharidin/chemistry , Enzyme Inhibitors/chemistry , Nuclear Proteins/chemistry , Phosphoprotein Phosphatases/chemistry , Protein Phosphatase 1/chemistry , Amino Acid Sequence , Binding Sites , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Domains , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Structure-Activity Relationship
7.
PLoS One ; 10(8): e0134317, 2015.
Article in English | MEDLINE | ID: mdl-26247207

ABSTRACT

Overactive GTPases have often been linked to human diseases. The available inhibitors are limited and have not progressed far in clinical trials. We report here a first-in-class small molecule pan-GTPase inhibitor discovered from a high throughput screening campaign. The compound CID1067700 inhibits multiple GTPases in biochemical, cellular protein and protein interaction, as well as cellular functional assays. In the biochemical and protein interaction assays, representative GTPases from Rho, Ras, and Rab, the three most generic subfamilies of the GTPases, were probed, while in the functional assays, physiological processes regulated by each of the three subfamilies of the GTPases were examined. The chemical functionalities essential for the activity of the compound were identified through structural derivatization. The compound is validated as a useful molecular probe upon which GTPase-targeting inhibitors with drug potentials might be developed.


Subject(s)
Enzyme Inhibitors/chemistry , GTP Phosphohydrolases/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/chemistry , Molecular Probes/chemistry , Thiourea/analogs & derivatives , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , HeLa Cells , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Integrins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Probes/metabolism , Molecular Probes/pharmacology , Phosphorylation/drug effects , Protein Binding , Signal Transduction/drug effects , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/chemistry , Thiourea/pharmacology , U937 Cells , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , ras Proteins/metabolism
8.
J Med Chem ; 57(24): 10314-28, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25399509

ABSTRACT

A quinazolinedione-derived screening hit 2 was discovered with cellular antiviral activity against respiratory syncytial virus (CPE EC50 = 2.1 µM), moderate efficacy in reducing viral progeny (4.2 log at 10 µM), and marginal cytotoxic liability (selectivity index, SI ∼ 24). Scaffold optimization delivered analogs with improved potency and selectivity profiles. Most notable were compounds 15 and 19 (EC50 = 300-500 nM, CC50 > 50 µM, SI > 100), which significantly reduced viral titer (>400,000-fold), and several analogs were shown to block the activity of the RNA-dependent RNA-polymerase complex of RSV.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Quinazolinones/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Virus Replication/drug effects , Antiviral Agents/chemical synthesis , Benzamides/chemical synthesis , Drug Design , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/virology , Structure-Activity Relationship
9.
J Med Chem ; 57(20): 8608-21, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25244572

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an emerging pathogenic alphavirus that can cause significant disease in humans. Given the absence of therapeutic options available and the significance of VEEV as a weaponized agent, an optimization effort was initiated around a quinazolinone screening hit 1 with promising cellular antiviral activity (EC50 = 0.8 µM), limited cytotoxic liability (CC50 > 50 µM), and modest in vitro efficacy in reducing viral progeny (63-fold at 5 µM). Scaffold optimization revealed a novel rearrangement affording amidines, specifically compound 45, which was found to potently inhibit several VEEV strains in the low nanomolar range without cytotoxicity (EC50 = 0.02-0.04 µM, CC50 > 50 µM) while limiting in vitro viral replication (EC90 = 0.17 µM). Brain exposure was observed in mice with 45. Significant protection was observed in VEEV-infected mice at 5 mg kg(-1) day(-1) and viral replication appeared to be inhibited through interference of viral nonstructural proteins.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzamides/pharmacology , Encephalitis Virus, Venezuelan Equine/drug effects , Piperazines/pharmacology , Animals , Benzamides/chemistry , Drug Evaluation, Preclinical/methods , Encephalomyelitis, Venezuelan Equine/drug therapy , Heterocyclic Compounds, 2-Ring/chemistry , Mice, Inbred C3H , Mice, Inbred C57BL , Piperazines/chemistry , Quinazolinones/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects
10.
ACS Chem Biol ; 9(10): 2393-403, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25126694

ABSTRACT

The hepatitis C virus (HCV) nonstructural protein 3 (NS3) is both a protease, which cleaves viral and host proteins, and a helicase that separates nucleic acid strands, using ATP hydrolysis to fuel the reaction. Many antiviral drugs, and compounds in clinical trials, target the NS3 protease, but few helicase inhibitors that function as antivirals have been reported. This study focuses on the analysis of the mechanism by which ebselen (2-phenyl-1,2-benzisoselenazol-3-one), a compound previously shown to be a HCV antiviral agent, inhibits the NS3 helicase. Ebselen inhibited the abilities of NS3 to unwind nucleic acids, to bind nucleic acids, and to hydrolyze ATP, and about 1 µM ebselen was sufficient to inhibit each of these activities by 50%. However, ebselen had no effect on the activity of the NS3 protease, even at 100 times higher ebselen concentrations. At concentrations below 10 µM, the ability of ebselen to inhibit HCV helicase was reversible, but prolonged incubation of HCV helicase with higher ebselen concentrations led to irreversible inhibition and the formation of covalent adducts between ebselen and all 14 cysteines present in HCV helicase. Ebselen analogues with sulfur replacing the selenium were just as potent HCV helicase inhibitors as ebselen, but the length of the linker between the phenyl and benzisoselenazol rings was critical. Modifications of the phenyl ring also affected compound potency over 30-fold, and ebselen was a far more potent helicase inhibitor than other, structurally unrelated, thiol-modifying agents. Ebselen analogues were also more effective antiviral agents, and they were less toxic to hepatocytes than ebselen. Although the above structure-activity relationship studies suggest that ebselen targets a specific site on NS3, we were unable to confirm binding to either the NS3 ATP binding site or nucleic acid binding cleft by examining the effects of ebselen on NS3 proteins lacking key cysteines.


Subject(s)
Antiviral Agents/pharmacology , Azoles/pharmacology , Hepatitis C/virology , Nucleic Acids/metabolism , Organoselenium Compounds/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Antioxidants/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Proliferation , Electrophoretic Mobility Shift Assay , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/metabolism , Humans , Hydrolysis , Isoindoles , Kinetics , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Models, Molecular , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Tumor Cells, Cultured
11.
PLoS Pathog ; 10(6): e1004213, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967809

ABSTRACT

Alphaviruses present serious health threats as emerging and re-emerging viruses. Venezuelan equine encephalitis virus (VEEV), a New World alphavirus, can cause encephalitis in humans and horses, but there are no therapeutics for treatment. To date, compounds reported as anti-VEEV or anti-alphavirus inhibitors have shown moderate activity. To discover new classes of anti-VEEV inhibitors with novel viral targets, we used a high-throughput screen based on the measurement of cell protection from live VEEV TC-83-induced cytopathic effect to screen a 340,000 compound library. Of those, we identified five novel anti-VEEV compounds and chose a quinazolinone compound, CID15997213 (IC50 = 0.84 µM), for further characterization. The antiviral effect of CID15997213 was alphavirus-specific, inhibiting VEEV and Western equine encephalitis virus, but not Eastern equine encephalitis virus. In vitro assays confirmed inhibition of viral RNA, protein, and progeny synthesis. No antiviral activity was detected against a select group of RNA viruses. We found mutations conferring the resistance to the compound in the N-terminal domain of nsP2 and confirmed the target residues using a reverse genetic approach. Time of addition studies showed that the compound inhibits the middle stage of replication when viral genome replication is most active. In mice, the compound showed complete protection from lethal VEEV disease at 50 mg/kg/day. Collectively, these results reveal a potent anti-VEEV compound that uniquely targets the viral nsP2 N-terminal domain. While the function of nsP2 has yet to be characterized, our studies suggest that the protein might play a critical role in viral replication, and further, may represent an innovative opportunity to develop therapeutic interventions for alphavirus infection.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalomyelitis, Venezuelan Equine/drug therapy , Quinazolinones/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Drug Resistance, Viral/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/virology , High-Throughput Screening Assays , Mice , Mice, Inbred C3H , Species Specificity , Structure-Activity Relationship , Vero Cells , Viral Plaque Assay , Virus Replication/drug effects
12.
Antimicrob Agents Chemother ; 57(8): 3731-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716053

ABSTRACT

Parasites in the genus Plasmodium cause disease throughout the tropic and subtropical regions of the world. P. falciparum, one of the deadliest species of the parasite, relies on glycolysis for the generation of ATP while it inhabits the mammalian red blood cell. The first step in glycolysis is catalyzed by hexokinase (HK). While the 55.3-kDa P. falciparum HK (PfHK) shares several biochemical characteristics with mammalian HKs, including being inhibited by its products, it has limited amino acid identity (~26%) to the human HKs, suggesting that enzyme-specific therapeutics could be generated. To that end, interrogation of a selected small-molecule library of HK inhibitors has identified a class of PfHK inhibitors, isobenzothiazolinones, some of which have 50% inhibitory concentrations (IC50s) of <1 µM. Inhibition was reversible by dilution but not by treatment with a reducing agent, suggesting that the basis for enzyme inactivation was not covalent association with the inhibitor. Lastly, six of these compounds and the related molecule ebselen inhibited P. falciparum growth in vitro (50% effective concentration [EC50] of ≥ 0.6 and <6.8 µM). These findings suggest that the chemotypes identified here could represent leads for future development of therapeutics against P. falciparum.


Subject(s)
Antimalarials/pharmacology , Benzothiazoles/pharmacology , Hexokinase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Azoles/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glycolysis , Inhibitory Concentration 50 , Isoindoles , Molecular Sequence Data , Organoselenium Compounds/pharmacology , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Recombinant Proteins/metabolism , Structure-Activity Relationship
13.
J Biol Chem ; 288(12): 8531-8543, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23382385

ABSTRACT

Cdc42 plays important roles in cytoskeleton organization, cell cycle progression, signal transduction, and vesicle trafficking. Overactive Cdc42 has been implicated in the pathology of cancers, immune diseases, and neuronal disorders. Therefore, Cdc42 inhibitors would be useful in probing molecular pathways and could have therapeutic potential. Previous inhibitors have lacked selectivity and trended toward toxicity. We report here the characterization of a Cdc42-selective guanine nucleotide binding lead inhibitor that was identified by high throughput screening. A second active analog was identified via structure-activity relationship studies. The compounds demonstrated excellent selectivity with no inhibition toward Rho and Rac in the same GTPase family. Biochemical characterization showed that the compounds act as noncompetitive allosteric inhibitors. When tested in cellular assays, the lead compound inhibited Cdc42-related filopodia formation and cell migration. The lead compound was also used to clarify the involvement of Cdc42 in the Sin Nombre virus internalization and the signaling pathway of integrin VLA-4. Together, these data present the characterization of a novel Cdc42-selective allosteric inhibitor and a related analog, the use of which will facilitate drug development targeting Cdc42-related diseases and molecular pathway studies that involve GTPases.


Subject(s)
Enzyme Inhibitors/pharmacology , Molecular Probes/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , 3T3 Cells , Allosteric Regulation , Animals , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Integrin alpha4beta1/antagonists & inhibitors , Integrin alpha4beta1/physiology , Mice , Oligopeptides/metabolism , Phenylurea Compounds/metabolism , Protein Binding , Pseudopodia/drug effects , Sin Nombre virus/physiology , Structure-Activity Relationship , Virus Internalization/drug effects , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
14.
J Biomol Screen ; 18(1): 26-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22923785

ABSTRACT

Chemotherapeutics tumor resistance is a principal reason for treatment failure, and clinical and experimental data indicate that multidrug transporters such as ATP-binding cassette (ABC) B1 and ABCG2 play a leading role by preventing cytotoxic intracellular drug concentrations. Functional efflux inhibition of existing chemotherapeutics by these pumps continues to present a promising approach for treatment. A contributing factor to the failure of existing inhibitors in clinical applications is limited understanding of specific substrate/inhibitor/pump interactions. We have identified selective efflux inhibitors by profiling multiple ABC transporters against a library of small molecules to find molecular probes to further explore such interactions. In our primary screening protocol using JC-1 as a dual-pump fluorescent reporter substrate, we identified a piperazine-substituted pyrazolo[1,5-a]pyrimidine substructure with promise for selective efflux inhibition. As a result of a focused structure-activity relationship (SAR)-driven chemistry effort, we describe compound 1 (CID44640177), an efflux inhibitor with selectivity toward ABCG2 over ABCB1. Compound 1 is also shown to potentiate the activity of mitoxantrone in vitro as well as preliminarily in vivo in an ABCG2-overexpressing tumor model. At least two analogues significantly reduce tumor size in combination with the chemotherapeutic topotecan. To our knowledge, low nanomolar chemoreversal activity coupled with direct evidence of efflux inhibition for ABCG2 is unprecedented.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line , Drug Resistance, Neoplasm , Flow Cytometry , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Small Molecule Libraries , Structure-Activity Relationship , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
ACS Chem Biol ; 7(6): 1095-108, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22486388

ABSTRACT

Mapping the functionality of GTPases through small molecule inhibitors represents an underexplored area in large part due to the lack of suitable compounds. Here we report on the small chemical molecule 2-(benzoylcarbamothioylamino)-5,5-dimethyl-4,7-dihydrothieno[2,3-c]pyran-3-carboxylic acid (PubChem CID 1067700) as an inhibitor of nucleotide binding by Ras-related GTPases. The mechanism of action of this pan-GTPase inhibitor was characterized in the context of the Rab7 GTPase as there are no known inhibitors of Rab GTPases. Bead-based flow cytometry established that CID 1067700 has significant inhibitory potency on Rab7 nucleotide binding with nanomolar inhibitor (K(i)) values and an inhibitory response of ≥97% for BODIPY-GTP and BODIPY-GDP binding. Other tested GTPases exhibited significantly lower responses. The compound behaves as a competitive inhibitor of Rab7 nucleotide binding based on both equilibrium binding and dissociation assays. Molecular docking analyses are compatible with CID 1067700 fitting into the nucleotide binding pocket of the GTP-conformer of Rab7. On the GDP-conformer, the molecule has greater solvent exposure and significantly less protein interaction relative to GDP, offering a molecular rationale for the experimental results. Structural features pertinent to CID 1067700 inhibitory activity have been identified through initial structure-activity analyses and identified a molecular scaffold that may serve in the generation of more selective probes for Rab7 and other GTPases. Taken together, our study has identified the first competitive GTPase inhibitor and demonstrated the potential utility of the compound for dissecting the enzymology of the Rab7 GTPase, as well as serving as a model for other small molecular weight GTPase inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nucleotides/metabolism , rab GTP-Binding Proteins/antagonists & inhibitors , Binding, Competitive , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Protein Binding , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...