Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Bioorg Med Chem Lett ; 49: 128318, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34391892

ABSTRACT

Lipophilicity is one of the principal QSAR parameters which influences among others the pharmacodynamics and pharmacokinetic properties of a drug candidates. In this paper, the lipophilicity of 14 amide derivatives of 1,3-dimethyl-2,6-dioxopurin-7-yl-alkylcarboxylic acids as multifunctional TRPA1 channel antagonists and phosphodiesterase 4/7 inhibitors with analgesic activity were investigated, using reversed-phase thin-layer chromatography method. It was observed that the retention behavior of the analyzed compounds was dependent on their structural features i.e. an aliphatic linker length, a kind of substituent at 8 position of purine-2,6-dione scaffold as well as on a substitution in a phenyl group. The experimental parameters (RM0) were compared with computationally calculated partition coefficient values by Principal Component Analysis (PCA). To verify the influence of lipophilic parameter of the investigated compounds on their biological activity the Kruskal-Wallis test was performed. The lowest lipophilicity was observed for the compounds with weak PDE4/7 inhibitory potency. The differences between the lipophilicity of potent inhibitors and inactive compounds were statistically significant. It was found that the presence of more lipophilic propoxy- or butoxy- substituents as well as the elongation of the aliphatic chain to propylene one between the purine-2,6-dione core and amide group were preferable for desired multifunctional activity.


Subject(s)
Analgesics/chemistry , Benzeneacetamides/chemistry , Phosphodiesterase 4 Inhibitors/chemistry , TRPA1 Cation Channel/antagonists & inhibitors , Xanthines/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Phenylbutyrates/chemistry , Principal Component Analysis , Quantitative Structure-Activity Relationship
2.
J Am Heart Assoc ; 10(12): e019091, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34056915

ABSTRACT

Background Pulmonary hypertension (PH) is a deadly disease characterized by vascular stiffness and altered cellular metabolism. Current treatments focus on vasodilation and not other root causes of pathogenesis. Previously, it was demonstrated that glutamine metabolism, as catalyzed by GLS1 (glutaminase 1) activity, is mechanoactivated by matrix stiffening and the transcriptional coactivators YAP1 (yes-associated protein 1) and transcriptional coactivator with PDZ-binding motif (TAZ), resulting in pulmonary vascular proliferation and PH. Pharmacologic inhibition of YAP1 (by verteporfin) or glutaminase (by CB-839) improved PH in vivo. However, systemic delivery of these agents, particularly YAP1 inhibitors, may have adverse chronic effects. Furthermore, simultaneous use of pharmacologic blockers may offer additive or synergistic benefits. Therefore, a strategy that delivers these drugs in combination to local lung tissue, thus avoiding systemic toxicity and driving more robust improvement, was investigated. Methods and Results We used poly(lactic-co-glycolic) acid polymer-based microparticles for delivery of verteporfin and CB-839 simultaneously to the lungs of rats suffering from monocrotaline-induced PH. Microparticles released these drugs in a sustained fashion and delivered their payload in the lungs for 7 days. When given orotracheally to the rats weekly for 3 weeks, microparticles carrying this drug combination improved hemodynamic (right ventricular systolic pressure and right ventricle/left ventricle+septum mass ratio), histologic (vascular remodeling), and molecular markers (vascular proliferation and stiffening) of PH. Importantly, only the combination of drug delivery, but neither verteporfin nor CB-839 alone, displayed significant improvement across all indexes of PH. Conclusions Simultaneous, lung-specific, and controlled release of drugs targeting YAP1 and GLS1 improved PH in rats, addressing unmet needs for the treatment of this deadly disease.


Subject(s)
Benzeneacetamides/administration & dosage , Drug Carriers , Enzyme Inhibitors/administration & dosage , Glutaminase/antagonists & inhibitors , Hypertension, Pulmonary/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Thiadiazoles/administration & dosage , Verteporfin/administration & dosage , Administration, Inhalation , Animals , Benzeneacetamides/chemistry , Cells, Cultured , Delayed-Action Preparations , Disease Models, Animal , Drug Combinations , Drug Compounding , Enzyme Inhibitors/chemistry , Glutaminase/metabolism , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Lung/physiopathology , Male , Mechanotransduction, Cellular , Monocrotaline , Particle Size , Rats, Sprague-Dawley , Thiadiazoles/chemistry , Time Factors , Vascular Remodeling/drug effects , Ventricular Function, Right/drug effects , Verteporfin/chemistry , YAP-Signaling Proteins
3.
J Chem Inf Model ; 60(12): 5803-5814, 2020 12 28.
Article in English | MEDLINE | ID: mdl-33174415

ABSTRACT

The main protease (Mpro) of the SARS-CoV-2 virus is one focus of drug development efforts for COVID-19. Here, we show that interactive molecular dynamics in virtual reality (iMD-VR) is a useful and effective tool for creating Mpro complexes. We make these tools and models freely available. iMD-VR provides an immersive environment in which users can interact with MD simulations and so build protein complexes in a physically rigorous and flexible way. Recently, we have demonstrated that iMD-VR is an effective method for interactive, flexible docking of small molecule drugs into their protein targets (Deeks et al. PLoS One 2020, 15, e0228461). Here, we apply this approach to both an Mpro inhibitor and an oligopeptide substrate, using experimentally determined crystal structures. For the oligopeptide, we test against a crystallographic structure of the original SARS Mpro. Docking with iMD-VR gives models in agreement with experimentally observed (crystal) structures. The docked structures are also tested in MD simulations and found to be stable. Different protocols for iMD-VR docking are explored, e.g., with and without restraints on protein backbone, and we provide recommendations for its use. We find that it is important for the user to focus on forming binding interactions, such as hydrogen bonds, and not to rely on using simple metrics (such as RMSD), in order to create realistic, stable complexes. We also test the use of apo (uncomplexed) crystal structures for docking and find that they can give good results. This is because of the flexibility and dynamic response allowed by the physically rigorous, atomically detailed simulation approach of iMD-VR. We make our models (and interactive simulations) freely available. The software framework that we use, Narupa, is open source, and uses commodity VR hardware, so these tools are readily accessible to the wider research community working on Mpro (and other COVID-19 targets). These should be widely useful in drug development, in education applications, e.g., on viral enzyme structure and function, and in scientific communication more generally.


Subject(s)
Antiviral Agents/chemistry , Benzeneacetamides/chemistry , COVID-19/metabolism , Coronavirus 3C Proteases/metabolism , Imidazoles/chemistry , SARS-CoV-2/enzymology , Viral Protease Inhibitors/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Benzeneacetamides/pharmacokinetics , Benzeneacetamides/pharmacology , Coronavirus 3C Proteases/genetics , Crystallization , Cyclohexylamines , Drug Design , Humans , Hydrogen Bonding , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Oligopeptides/chemistry , Oligopeptides/metabolism , Protein Conformation , Pyridines , Structure-Activity Relationship , Viral Protease Inhibitors/pharmacokinetics , Viral Protease Inhibitors/pharmacology
4.
J Genet Genomics ; 47(7): 389-395, 2020 07 20.
Article in English | MEDLINE | ID: mdl-33004309

ABSTRACT

Many cancer types reprogram their metabolism to become addicted to glutamine. One of the critical enzymes in the utilization of glutamine in these cells is glutaminase. CB-839 (telaglenastat) is a drug that targets glutaminase that is currently being evaluated in many clinical trials for efficacy in various cancer types that are known to be driven by glutamine metabolism. Despite its use, there are limited assays available for testing the pharmacodynamic on-target effects of CB-839 on the limited, small-volume patient samples that are obtained in early-phase clinical trials. Thus, we developed an assay based on the cellular thermal shift assay technique using AlphaLISA technology to show that CB-839 specifically engages glutaminase in colon cancer cell lines in vitro and in minute quantities of mouse xenograft tumors. Notably, we show that this assay detects CB-839 binding to glutaminase in platelets of patients collected while receiving CB-839 on a clinical trial. This assay may be used to study the pharmacodynamic profile of CB-839 in very small tissue samples obtained from patients on a clinical trial and may be useful in future studies designed to screen other inhibitors of glutaminase.


Subject(s)
Colonic Neoplasms/genetics , Glutaminase/genetics , Glutamine/metabolism , Animals , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacology , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Glutaminase/antagonists & inhibitors , Glutaminase/chemistry , Heterografts , Humans , Mice , Thiadiazoles/chemistry , Thiadiazoles/pharmacology
5.
Folia Med (Plovdiv) ; 62(3): 532-538, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33009766

ABSTRACT

INTRODUCTION: Examination of the potential possibilities of 2-chloro-N-(1-(3,4-dimethoxyphenyl)propan-2-yl)-2-phenylacetamide (IQP) to affect bioelectrogenesis and the contractile activity of isolated smooth muscles (SM) from stomach. AIM: Having in mind the structural similarities between the molecules of papaverine and IQP, the aim of the present study was to examine such features of the newly synthesized molecule that may potentially affect the muscle tonus, spontaneous bioelectrical and contractile activities of smooth muscles isolated from the stomach, basing on specific mechanisms of papaverine. MATERIALS AND METHODS: The synthesis of IQP is based on the initially formed aziridine ring by principles of Gilbert's reaction. Impact of IQP on the bioelectrogenesis and the contractile activity of isolated smooth muscles from male Wistar rats was measured by the single sucrose-gap method and isometrically recorded. RESULTS: IQP (1×10-5 - 2.5×10-4 mol/l) causes muscle relaxation, producing changes in two processes that have influence on the mechanical activity of smooth muscles:1.    Blocked Ca2+ influx through the potential-dependent membrane Ca2+ channels, followed in turn by lowering the Ca2+ intracellular levels. This effect is proved by the changes in the frequency and amplitude of spike-potentials in sucrose-bridge experiments when IQP is applied.2.    Activation of a cAMP-dependent signal cascade. The relaxing effect of IQP was significantly reduced in the presence of KT5720(5×10-6 mol/l), an inhibitor of protein kinase A. CONCLUSION: We assume that there might be interconnections between these two IQP-dependent processes, because PKA-dependent phosphorylation of the L-type Ca2+ channels in smooth muscles provokes a reaction of inactivation.


Subject(s)
Benzeneacetamides , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacology , Biomechanical Phenomena/drug effects , Calcium/metabolism , Male , Papaverine , Rats , Rats, Wistar
6.
Int J Mol Sci ; 21(13)2020 Jun 28.
Article in English | MEDLINE | ID: mdl-32605171

ABSTRACT

The aim of this study was to identify novel antimelanogenic drugs from an epigenetic screening library containing various modulators targeting DNA methyltransferases, histone deacetylases, and other related enzymes/proteins. Of 141 drugs tested, K8 (4-((hydroxyamino)carbonyl)-N-(2-hydroxyethyl)-N-phenyl-benzeneacetamide; HPOB) was found to effectively inhibit the α-melanocyte-stimulating hormone (α-MSH)-induced melanin synthesis in B16-F10 murine melanoma cells without accompanying cytotoxicity. Additional experiments showed that K8 did not significantly reduce the mRNA and protein level of tyrosinase (TYR) or microphthalmia-associated transcription factor (MITF) in cells, but it potently inhibited the catalytic activity TYR in vitro (IC50, 1.1-1.5 µM) as compared to ß-arbutin (IC50, 500-700 µM) or kojic acid (IC50, 63 µM). K8 showed copper chelating activity similar to kojic acid. Therefore, these data suggest that K8 inhibits cellular melanin synthesis not by downregulation of TYR protein expression through an epigenetic mechanism, but by direct inhibition of TYR catalytic activity through copper chelation. Metal chelating activity of K8 is not surprising because it is known to inhibit histone deacetylase (HDAC) 6 through zinc chelation. This study identified K8 as a potent inhibitor of cellular melanin synthesis, which may be useful for the treatment of hyperpigmentation disorders.


Subject(s)
Antineoplastic Agents/pharmacology , Benzeneacetamides/chemistry , Epigenesis, Genetic , Melanins/metabolism , Melanoma, Experimental/drug therapy , Monophenol Monooxygenase/antagonists & inhibitors , Pharmaceutical Preparations/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Pharmaceutical Preparations/isolation & purification , Tumor Cells, Cultured
7.
J Chromatogr Sci ; 58(6): 562-568, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32303742

ABSTRACT

A simple and accurate chiral liquid chromatographic method was developed for enantiomeric resolution and determination of 2-(5-fluoro-2-hydroxyphenyl)-2-(1-oxo-2,3-dihydro-1H-isoindol-2-yl)-N-(1,3-thiazol-2-yl)acetamide (EAI045). The enantiomers of EAI045 were baseline resolved on a Chiralpak AD-H (250 mm × 4.6 mm, 5 µm) column using a mobile phase system containing n-hexane: 2-propanol (75: 25 v/v) at a flow rate of 1 mL min-1 at 30°C. The eluted analytes were subsequently detected with an ultraviolet detector at 254 nm. The effects of organic modifiers and temperature on the enantioselectivity and resolution of the enantiomers were evaluated. The calibration curves were plotted within the concentration range between 2 and 600 µg mL-1 (n = 11), and recoveries between 98.74% and 101.52% were obtained, with relative standard deviation < 1.4%. The limit of detection and limit of quantitation for R-enantiomer were 0.94 and 3.07 µg mL-1 and for S-enantiomer were 0.86 and 2.84 µg mL-1, respectively. The validated method was found to be suitable for enantiomeric separation and sufficiently accurate for the determination of enantiomeric purity of EAI045 in bulk drugs.


Subject(s)
Benzeneacetamides , Chromatography, Liquid/methods , Thiazoles , Amylose/analogs & derivatives , Amylose/chemistry , Animals , Benzeneacetamides/blood , Benzeneacetamides/chemistry , Benzeneacetamides/isolation & purification , Benzeneacetamides/pharmacokinetics , Limit of Detection , Linear Models , Mice , Phenylcarbamates/chemistry , Reproducibility of Results , Stereoisomerism , Thiazoles/blood , Thiazoles/chemistry , Thiazoles/isolation & purification , Thiazoles/pharmacokinetics
8.
Photochem Photobiol ; 95(2): 563-571, 2019 03.
Article in English | MEDLINE | ID: mdl-30059157

ABSTRACT

A wide variety of marine bioluminescent organisms emit light via the excited-state coelenteramide, which is produced from the coelenterazine oxidation via a series of complicated chemical reactions in protein. Photoluminescence of coelenteramide is a simple way to produce light without experiencing the intricate reactions starting from coelenterazine. To extend the color range of light emission, many coelenterazine analogues were synthesized, but mostly only produce blue and cyan fluorescence. Based on the 42 synthesized coelenterazine analogues, we theoretically studied the absorption and fluorescence properties of the corresponding coelenteramide analogues. The electronic effect, steric effect, conjugated effect and solvated effect were considered. The results indicated that conjugated effect has great influence on the strength and wavelength of fluorescence and large electron transfer is beneficial to redshift. Based on the regularities, we theoretically designed six coelenteramide analogues, and together with the original coelenteramide, the seven-ones emit the seven colors of rainbow via their photoluminescences. This study expands the coelenteramide fluorescence to the whole visible light region and could inspire new application.


Subject(s)
Benzeneacetamides/chemistry , Luminescent Measurements , Pyrazines/chemistry , Fluorescence , Spectrometry, Fluorescence
9.
J Biomol Struct Dyn ; 37(16): 4384-4394, 2019 10.
Article in English | MEDLINE | ID: mdl-30499387

ABSTRACT

Lung cancer is the leading cause of cancer death, and epidermal growth factor receptor (EGFR) kinase domain mutations are a common cause of non-small-cell lung cancer (NSCLC), a major subtype of lung cancers. Patients harboring most of these mutations respond well to the EGFR inhibitors Gefitinib and Erlotinib initially, but soon develop resistance to them due to the emergence of the gatekeeper mutation T790M. The new-generation inhibitors such as AZD9291, HM61713, CO-1686 and WZ4002 can overcome T790M through covalent binding to Cys 797, but ultimately lose their efficacy upon the emergence of the C797S mutation that abolishes the covalent bonding. Allosteric inhibitors EAI001 and EAI045 are a new type of EGFR inhibitors that bind to EGFR away from the ATP-binding site and not relying on Cys 797. In this study, molecular dynamics simulations and free energy calculations were carried out on EAI001 and EAI045 in complex with EGFR, revealing the detailed inhibitory mechanism of EAI001 and EAI045 as EGFR allosteric inhibitor, which was expected to provide a basis for rational drug design of the EGFR allosteric inhibitors. Communicated by Ramaswamy H. Sarma.


Subject(s)
Benzeneacetamides/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Protein Kinase Inhibitors/chemistry , Thiazoles/chemistry , Acrylamides/adverse effects , Allosteric Regulation/drug effects , Aniline Compounds/adverse effects , Benzeneacetamides/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride/adverse effects , Gefitinib/adverse effects , Humans , Molecular Dynamics Simulation , Mutation , Protein Binding/drug effects , Pyrimidines/adverse effects , Thiazoles/therapeutic use
10.
J Pharm Sci ; 107(12): 3089-3097, 2018 12.
Article in English | MEDLINE | ID: mdl-30170009

ABSTRACT

Nepafenac is a water-insoluble nonsteroidal antiinflammatory drug that is available as an ophthalmic suspension (Nevanac®). Suspensions are undesirable for 2 reasons: they tend to cause foreign body sensation and lacrimation, which could limit residence time and drug bioavailability. This decreases the amount of time the drug has to reach the site of action, the cornea. Previously, we improved the solubility and ocular permeability of nepafenac by complexing the drug with hydroxypropyl-ß-cyclodextrin. In this study, we used the complex to formulate an ion-activated in situ gel system using sodium alginate, Protanal PH 1033, to increase the residence time and to reduce repeat eye drop instillation. Rheological properties of the formulations revealed that the viscosity of the optimized formulation was increased 30-fold when exposed to the simulated tear fluid (35°C). Permeation studies showed that the drug concentration of the in situ formulations were approximately 10 times higher than the commercial product, Nevanac® (p < 0.001). In addition, the in situ gel formulations had 5-fold higher concentrations of nepafenac retained in the cornea when compared to Nevanac® (p <0.001). Finally, ex vivo drug distribution studies in the porcine eye perfusion model revealed a higher drug retention in various ocular tissues such as cornea, sclera, retina, as compared to Nevanac®.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Benzeneacetamides/administration & dosage , Benzeneacetamides/pharmacokinetics , Drug Carriers/chemistry , Eye/metabolism , Gels/chemistry , Phenylacetates/administration & dosage , Phenylacetates/pharmacokinetics , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , Administration, Ophthalmic , Alginates/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzeneacetamides/chemistry , Biological Availability , Cornea/metabolism , Ocular Absorption , Permeability , Phenylacetates/chemistry , Solubility , Swine , Viscosity
11.
Cancer Biother Radiopharm ; 33(6): 221-232, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29894210

ABSTRACT

BACKGROUND: Recently, the direct intratumoral (i.t.) injection of anticancer agents has been investigated. A newly synthesized Antineoplaston A10 analog 3-(4-methoxybenzoylamino)-2,6-piperidinedione (MPD) showed an antitumor activity in human breast cancer cell line. Unfortunately, MPD suffered from poor water solubility. MATERIALS AND METHODS: Pseudoternary phase diagram of oil (isopropyl myristate), surfactant (Tween 80), cosurfactant (ethanol), and water was plotted. MPD microemulsion (MPDME) was developed and characterized for particle size (PS), polydispersity index (PDI), zeta potential (ZP), and morphology (transmission electron microscopy). MPDME and MPD solution (MPDS) were radiolabeled with technetium 99m (99mTc) using stannous chloride dihydrate (SnCl2.2H2O). Molecular docking of MPD and 99mTc-MPD was performed to study the interaction with DNA. RESULTS: The impacts of intravenous (i.v.) and i.t. injections of 99mTc-MPDME and 99mTc-MPDS on biodistribution were studied. The developed MPDME showed spherical droplets with mean PS (74.00 ± 5.69 nm), PDI (0.25 ± 0.03), and ZP (33.90 ± 0.90 mV). Labeling yield of 99mTc-MPDME and 99mTc-MPDS was 97.00% ± 0.60% and 92.02% ± 0.45%, respectively. MPD and 99mTc-MPD showed almost same binding affinity with DNA binding site. Biodistribution results showed that i.t. injection of 99mTc-MPDME significantly enhanced tumor retention compared to i.v. route. CONCLUSIONS: Herein, the authors concluded that microemulsion could be used as i.t. injectable delivery vehicle to improve targeting and tumor retention of MPD.


Subject(s)
Antineoplastic Agents/pharmacology , Benzeneacetamides/pharmacology , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Piperidones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzeneacetamides/chemistry , Benzeneacetamides/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Emulsions , Female , Humans , Injections, Intralesional , Injections, Intravenous , Mice , Molecular Docking Simulation , Particle Size , Piperidones/chemistry , Piperidones/therapeutic use , Technetium/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
12.
AAPS PharmSciTech ; 19(6): 2554-2563, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948988

ABSTRACT

Nepafenac is a nonsteroidal anti-inflammatory drug (NSAID), currently only available as 0.1% ophthalmic suspension (Nevanac®). This study utilized hydroxypropyl-ß-cyclodextrin (HPBCD) to increase the water solubility and trans-corneal permeation of nepafenac. The nepafenac-HPBCD complexation in the liquid and solid states were confirmed by phase solubility, differential scanning calorimetry (DSC), X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), and nuclear magnetic resonance spectroscopy (NMR) analyses. Nepafenac 0.1% ophthalmic solution was formulated using HPBCD (same pH and osmolality as that of Nevanac®) and pig eye trans-corneal permeation was studied versus Nevanac®. Furthermore, nepafenac content in cornea, sclera, iris, lens, aqueous humor, choroid, ciliary body, retina, and vitreous humor was studied in a continuous isolated pig eye perfusion model in comparison to the suspension and Nevanac®. Permeation studies using porcine corneas revealed that the solution formulation had a permeation rate 18 times higher than Nevanac®. Furthermore, the solution had 11 times higher corneal retention than Nevanac®. Drug distribution studies using porcine eyes revealed that the solution formulation enables detectable levels in various ocular tissues while the drug was undetectable by Nevanac®. The ocular solution formulation had a significantly higher drug concentration in the cornea compared to the suspension or Nevanac®.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzeneacetamides/chemistry , Eye/metabolism , Phenylacetates/chemistry , beta-Cyclodextrins/chemistry , Animals , Benzeneacetamides/pharmacokinetics , Ophthalmic Solutions , Permeability , Phenylacetates/pharmacokinetics , Solubility , Swine
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 204: 260-266, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-29936223

ABSTRACT

The first spectrofluorimetric report investigating the fluorimetric behavior of the antihistaminic drug, azelastine (AZEL), and the non-steroidal anti-inflammatory drug, nepafenac (NEP), either in bulk or in their dosage forms, eye drops and ophthalmic suspension. After a full investigation of the factors that may influence their spectrofluorimetric behavior: pH, different organized media and organic solvents, the optimum factors were set in order to enable the analysis of each drug with maximum sensitivity. The AZEL spectrofluorimetric analysis was set at 286/364 (λex/λem) in distilled water while for NEP, the analysis was set at 228/303 (λex/λem) in methanol. The linearity range for AZEL was from 0.1 to 1.5 µg/mL while that of NEP was from 0.2 to 1.5 µg/mL. The linearity yielded good regression parameters with low LOD (0.022 and 0.032 µg/mL for AZEL and NEP, respectively) and LOQ (0.073 and 1.08 µg/mL for AZEL and NEP, respectively) when compared with those obtained from many previous spectroscopic and chromatographic reports in literature. The method was ICH validated and was applied to the analysis of AZEL and NEP with good selectivity regarding the inactive ingredients.


Subject(s)
Benzeneacetamides/analysis , Ophthalmic Solutions/analysis , Phenylacetates/analysis , Phthalazines/analysis , Spectrometry, Fluorescence/methods , Benzeneacetamides/chemistry , Drug Stability , Limit of Detection , Linear Models , Ophthalmic Solutions/chemistry , Phenylacetates/chemistry , Phthalazines/chemistry , Reproducibility of Results
14.
Biochem Biophys Res Commun ; 502(3): 332-337, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29802850

ABSTRACT

Lung cancer is the leading cause of cancer deaths. Epidermal growth factor receptor (EGFR) kinase domain mutations are a common cause of non-small cell lung cancers (NSCLCs), a major subtype of lung cancers. Patients harboring most of these mutations respond well to the anti-EGFR tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib initially, but soon develop resistance to them in about half of the cases due to the emergence of the gatekeeper mutation T790M. The third-generation TKIs such as AZD9291, HM61713, CO-1686 and WZ4002 can overcome T790M through covalent binding to the EGFR kinase through Cys 797, but ultimately lose their efficacy upon emergence of the C797S mutation that abolishes the covalent bonding. Therefore to develop new TKIs to overcome EGFR drug-resistant mutants harboring T790M/C797S is urgently demanded. EAI001 and EAI045 are a new type of EGFR TKIs that bind to EGFR reversibly and not relying on Cys 797. EAI045 in combination with cetuximab is effective in mouse models of lung cancer driven by EGFR L858R/T790M and L858R/T790M/C797S. Here we report the crystal structure of EGFR T790M/C797S/V948R in complex with EAI045, and compare it to EGFR T790M/V948R in complex with EAI001. The complex structure reveals why EAI045 binds tighter to EGFR than does EAI001, and why EAI001 and EAI045 prefer binding to EGFR T790M. The knowledge may facilitate future drug development studies targeting this very important cancer target.


Subject(s)
Benzeneacetamides/chemistry , ErbB Receptors/chemistry , ErbB Receptors/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Kinase Inhibitors/chemistry , Thiazoles/chemistry , Amino Acid Substitution , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzeneacetamides/administration & dosage , Benzeneacetamides/pharmacology , Binding Sites , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cetuximab/administration & dosage , Crystallography, X-Ray , Drug Design , ErbB Receptors/antagonists & inhibitors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Mutation, Missense , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thiazoles/administration & dosage , Thiazoles/pharmacology
15.
Int J Pharm ; 545(1-2): 240-253, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29733973

ABSTRACT

A10, (3-phenylacetylamino-2,6-piperidinedione), is a natural peptide with broad antineoplastic activity. Recently, in vitro antitumor effect of a new A10 analog [3-(4-methoxybenzoylamino)-2,6-piperidinedione] (MPD) has been verified. However, poor aqueous solubility represents an obstacle towards intravenous formulation of MPD and impedes successful in vivo antitumor activity. To surmount such limitation, MPD microemulsion (MPDME) was developed. A 3122 full factorial design using Design-Expert® software was adopted to study the influence of different parameters and select the optimum formulation (MPDME1). Transmission electron microscopy (TEM) displayed spherical droplets of MPDME1. The cytotoxicity of MPDME1 in Michigan Cancer Foundation 7 (MCF-7) breast cancer cell line exceeded that of MPD solution (MPDS) and tamoxifen. Compatibility with injectable diluents, in vitro hemolytic studies and in vivo histopathological examination confirmed the safety of parenteral application of MPDME1. Molecular docking results showed almost same binding affinity of A10, MPD and 125I-MPD with histone deacetylase 8 (HDAC8) receptor. Accordingly, radioiodination of MPDME1 and MPDS was done via direct electrophilic substitution reaction. Biodistribution of 125I-MPDME1 and 125I-MPDS in normal and tumor (ascites and solid) bearing mice showed high accumulation of 125I-MPDME1 in tumor tissues. Overall, the results proved that MPDME represents promising parenteral delivery system capable of improving antineoplastic activity of MPD.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzeneacetamides/administration & dosage , Breast Neoplasms/drug therapy , Carcinoma, Ehrlich Tumor/drug therapy , Molecular Docking Simulation , Piperidones/administration & dosage , Technology, Pharmaceutical/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Drug Compounding , Emulsions , Ethanol/chemistry , Female , Histone Deacetylases/metabolism , Humans , Injections, Intravenous , Iodine Radioisotopes , MCF-7 Cells , Male , Microscopy, Electron, Transmission , Piperidones/chemistry , Piperidones/pharmacokinetics , Polysorbates/chemistry , Rabbits , Repressor Proteins/metabolism , Tissue Distribution
16.
Comput Biol Chem ; 74: 167-189, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29627693

ABSTRACT

The tyrosine kinase inhibitors (TKI) against epidermal growth factor receptor (EGFR) are generally utilized as a part of patients with non-small cell lung carcinoma (NSCLC). However, EGFR T790M mutation results in resistance to most clinically available EGFR TKIs. Third-generation EGFR TKIs against the T790M mutation has been in active clinical development to triumph the resistance problem; they covalently bind with conserved Cys797 inside the EGFR active site, offering both potency and kinase-selectivity. Third generation drugs target C797, which makes the C797S resistance mutation more subtle. EGFR C797S mutation was accounted to be a main mechanism of resistance to the third-generation inhibitors. The C797S mutation gives off an impression of being an ideal target for conquering the acquired resistance to the third generation inhibitors. We have performed structure based-virtual screening strategies for binding of glucokinase activator to EGFR C797S, which can overcome EGFR resistance impediment with mutant-selective allosteric inhibition towards all kinds of mutant EGFR (T790M, L858R, TMLR) and WT EGFR. The final filter of Lipinski's Rule of Five, Jargan's Rule of Three and in silico ADME predictions gave 23 hits, which conform to Lipinski's rule and Jorgensen's rule and all their pharmacokinetic parameters are inside the appropriate range characterized for human use, in this manner demonstrating their potential as a drug-like molecule.


Subject(s)
Benzeneacetamides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Glucokinase/metabolism , Lung Neoplasms/drug therapy , Molecular Docking Simulation , Mutation , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Allosteric Regulation/drug effects , Benzeneacetamides/chemistry , Binding Sites/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Thiazoles/chemistry
17.
Int J Biol Macromol ; 110: 336-345, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29408555

ABSTRACT

This research was aimed to develop and evaluate nepafenac loaded silica nanoparticles dispersed in-situ gel system for the improved treatment of ocular diseases. The blank silica nanoparticles prepared by stober's process showed the particle size of 151 nm to 285 nm with the zeta potential of -19.6 to -31.9 mV. The nepafenac loaded silica nanoparticles were spherical in shape with smooth outer surface. The nepafenac loaded silica nanoparticles dispersed in poloxamer - chitosan in-situ gelling system showed gelling temperature of 32 °C with sustained release of nepafenac and higher permeation (58.79 µg) across the goat cornea than poloxamer - poloxamer (21.18 µg) in-situ gelling system. Hence the developed in-situ gelling system containing nepafenac loaded silica nanoparticle could be a promising tool for the topical delivery of drugs to the eye.


Subject(s)
Benzeneacetamides , Drug Carriers , Nanoparticles/chemistry , Phenylacetates , Silicon Dioxide , Animals , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacokinetics , Benzeneacetamides/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Gels , Mice , Phenylacetates/chemistry , Phenylacetates/pharmacokinetics , Phenylacetates/pharmacology , RAW 264.7 Cells , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology
18.
Int J Biol Macromol ; 112: 163-168, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29382580

ABSTRACT

Aequorin as an old small calcium-sensitive photoprotein is a blue fluorescence protein which converts coelenterazine (a substrate) to coelenteramide with a flash type emission. The decay kinetics and emission properties of this protein can be changed using directed mutagenesis of crucial amino acid residue. In this work, we prepared three double mutants: Y82F/W86F, Y82F/D153G, and W86F/D153G. According to our results, it seems that presence of Y82F mutation results in shift of emission to longer wavelengths while the W86F mutation shifts the emission to shorter wavelengths. Furthermore, comparison of the variants for light half-life indicated decreased t1/2 for the two variants of Y82F/D153G and W86F/D153G. But in compared to wild type aequorin, the Y82F/W86F variant displayed a 2-fold increase of light half-life. On the other hand, the thermostability properties of double mutants confirmed that only Y82F/D153G variant of apoaequorin is higher stability than others. Also, the single W86F mutant reached the highest stability against thermal shock. Our data suggest that replacement of single or few point mutations in the binding pocket or active site of aequorin affects its bioluminescence and kinetic properties and so could be used for new reporter production of this photoprotein with the feasibility and limited substitutions.


Subject(s)
Aequorin/chemistry , Amino Acids/chemistry , Hydrozoa/chemistry , Mutagenesis , Amino Acids/genetics , Animals , Benzeneacetamides/chemistry , Calcium/chemistry , Imidazoles/chemistry , Kinetics , Luminescent Measurements , Mutation/genetics , Proteolysis , Pyrazines/chemistry
19.
J Biol Chem ; 293(10): 3535-3545, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29317493

ABSTRACT

Altered glycolytic flux in cancer cells (the "Warburg effect") causes their proliferation to rely upon elevated glutamine metabolism ("glutamine addiction"). This requirement is met by the overexpression of glutaminase C (GAC), which catalyzes the first step in glutamine metabolism and therefore represents a potential therapeutic target. The small molecule CB-839 was reported to be more potent than other allosteric GAC inhibitors, including the parent compound bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl (BPTES), and is in clinical trials. Recently, we described the synthesis of BPTES analogs having distinct saturated heterocyclic cores as a replacement for the flexible chain moiety, with improved microsomal stability relative to CB-839 and BPTES. Here, we show that one of these new compounds, UPGL00004, like CB-839, more potently inhibits the enzymatic activity of GAC, compared with BPTES. We also compare the abilities of UPGL00004, CB-839, and BPTES to directly bind to recombinant GAC and demonstrate that UPGL00004 has a similar binding affinity as CB-839 for GAC. We also show that UPGL00004 potently inhibits the growth of triple-negative breast cancer cells, as well as tumor growth when combined with the anti-vascular endothelial growth factor antibody bevacizumab. Finally, we compare the X-ray crystal structures for UPGL00004 and CB-839 bound to GAC, verifying that UPGL00004 occupies the same binding site as CB-839 or BPTES and that all three inhibitors regulate the enzymatic activity of GAC via a similar allosteric mechanism. These results provide insights regarding the potency of these inhibitors that will be useful in designing novel small-molecules that target a key enzyme in cancer cell metabolism.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glutaminase/antagonists & inhibitors , Models, Molecular , Neoplasm Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Allosteric Site/drug effects , Amino Acid Substitution , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Benzeneacetamides/chemistry , Benzeneacetamides/metabolism , Benzeneacetamides/pharmacology , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glutaminase/chemistry , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/antagonists & inhibitors , Glutamine/chemistry , Glutamine/metabolism , Humans , Hydrogen Bonding , Molecular Conformation , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sulfides/chemistry , Sulfides/metabolism , Sulfides/pharmacology , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Thiadiazoles/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
20.
Bioorg Med Chem Lett ; 28(4): 694-699, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29366652

ABSTRACT

Mutations in the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) are commonly found in gliomas. AGI-5198, a potent and selective inhibitor of the mutant IDH1 enzyme, was radiolabeled with radioiodine and fluorine-18. These radiotracers were evaluated as potential probes for imaging mutant IDH1 expression in tumors with positron emission tomography (PET). Radioiodination of AGI-5198 was achieved using a tin precursor in 79 ±â€¯6% yield (n = 9), and 18F-labeling was accomplished by the Ugi reaction in a decay-corrected radiochemical yield of 2.6 ±â€¯1.6% (n = 5). The inhibitory potency of the analogous nonradioactive compounds against mutant IDH1 (IDH1-R132H) was determined in enzymatic assays. Cell uptake studies using radiolabeled AGI-5198 analogues revealed somewhat higher uptake in IDH1-mutated cells than that in wild-type IDH1 cells. The radiolabeled compounds displayed favorable tissue distribution characteristics in vivo, and good initial uptake in IDH1-mutated tumor xenografts; however, tumor uptake decreased with time. Radioiodinated AGI-5198 exhibited higher tumor-to-background ratios compared with 18F-labeled AGI-5198; unfortunately, similar results were observed in wild-type IDH1 tumor xenografts as well, indicating lack of selectivity for mutant IDH1 for this tracer. These results suggest that AGI-5198 analogues are not a promising platform for radiotracer development. Nonetheless, insights gained from this study may help in design and optimization of novel chemical scaffolds for developing radiotracers for imaging the mutant IDH1 enzyme.


Subject(s)
Benzeneacetamides/pharmacology , Glioma/metabolism , Imidazoles/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Radiopharmaceuticals/pharmacology , Amino Acid Substitution , Animals , Benzeneacetamides/chemical synthesis , Benzeneacetamides/chemistry , Cell Line, Tumor , Fluorine Radioisotopes , Halogenation , Heterografts , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Iodine Radioisotopes , Isocitrate Dehydrogenase/genetics , Mice, Nude , Muscles/metabolism , Mutation , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...