1
|
El-Wakil MH, Ghazala RA, El-Dershaby HA, Drozdowska D, Wróbel-Tałałaj A, Parzych C, Ratkiewicz A, Kolesińska B, Abd El-Razik HA, Soliman FSG. Rational design, synthesis, and molecular modelling insights of dual DNA binders/DHFR inhibitors bearing arylidene-hydrazinyl-1,3-thiazole scaffold with apoptotic and anti-migratory potential in breast MCF-7 cancer cells. J Enzyme Inhib Med Chem 2025; 40:2468353. [PMID: 40035286 PMCID: PMC11881662 DOI: 10.1080/14756366.2025.2468353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
In light of searching for new breast cancer therapies, DNA-targeted small molecules were rationally designed to simultaneously bind DNA and inhibit human dihydrofolate reductase (hDHFR). Fourteen new arylidene-hydrazinyl-1,3-thiazoles (5-18) were synthesised and their dual DNA groove binding potential and in vitro hDHFR inhibition were performed. Two compounds, 5 and 11, proved their dual efficacy. Molecular docking and molecular dynamics simulations were performed for those active derivatives to explore their mode of binding and stability of interactions inside DHFR active site. Anti-breast cancer activity was assessed for 5 and 11 on MCF-7 cells using MTX as reference. IC50 measurements revealed that both compounds were more potent and selective than MTX. Cytotoxicity was examined against normal skin fibroblasts to examine safety and selectivity Moreover, mechanistic studies including apoptosis induction and wound healing were performed. Further in silico ADMET assessment was conducted to determine their eligibility as drug leads suitable for future optimisation and development.
Collapse
Affiliation(s)
- Marwa H. El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rasha A. Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hadeel A. El-Dershaby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Danuta Drozdowska
- Department of Organic Chemistry, Medical University of Bialystok, Bialystok, Poland
| | | | - Cezary Parzych
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Artur Ratkiewicz
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Beata Kolesińska
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Heba A. Abd El-Razik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Farid S. G. Soliman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Ermis M, Kir F, Sahin S. Development and validation of a RP-HPLC method for simultaneous determination of cimetidine, metoprolol tartrate and phenol red for intestinal perfusion studies. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1252:124449. [PMID: 39787726 DOI: 10.1016/j.jchromb.2025.124449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
A new reversed phase high-performance liquid chromatography (RP-HPLC) method, with a short analysis time and easy to apply, was developed for the simultaneous detection of cimetidine (CIM), metoprolol tartrate (MT) and phenol red (PR) for use in intestinal perfusion studies. The analysis was performed with phosphate buffer (pH 5.0, 12.5 mM)-acetonitrile mixture as mobile phase and C18 column (Inertsil ODS-3; 5 µm, 4.6 × 250 mm) as stationary phase. Gradient analysis conditions were used and the acetonitrile ratio in the mobile phase varied from 10 to 50% in 10 min. Total run time for analysis was 10 min and the injection volume was 20 µL. Detection of compounds was performed at 207 nm. Under optimum HPLC conditions, retention times were 4.03 min for CIM, 6.99 min for MT and 8.49 min for PR. The method was validated according to ICH Q2 (R1) guideline for specificity, linearity, sensitivity, precision, accuracy, stability and robustness. Developed method was linear and determination coefficients of the calibration curves were 0.9993, 0.9991 and 1.0 for CIM, MT and PR, respectively. The limits of quantification were 6.20, 2.78 and 0.45 μg/mL for CIM, MT and PR, respectively. The precision and accuracy values of the developed analytical method met the ICH Q2 (R1) limits. The applicability of the method was demonstrated by preliminary in-situ intestinal perfusion studies. In conclusion, samples obtained from in-situ intestinal perfusion studies performed to examine the absorption/permeability of CIM, MT, and PR can be analyzed with the developed HPLC method.
Collapse
Affiliation(s)
- Meryem Ermis
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara 06100 Türkiye; Ankara Medipol University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara 06570 Türkiye
| | - Fatma Kir
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara 06100 Türkiye
| | - Selma Sahin
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara 06100 Türkiye.
| |
Collapse
|
3
|
Pulido-Saavedra A, Oliva HNP, Prudente TP, Kitaneh R, Nunes EJ, Fogg C, Funaro MC, Weleff J, Nia AB, Angarita GA. Effects of psychedelics on opioid use disorder: a scoping review of preclinical studies. Cell Mol Life Sci 2025; 82:49. [PMID: 39833376 PMCID: PMC11747050 DOI: 10.1007/s00018-024-05519-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/01/2024] [Accepted: 11/19/2024] [Indexed: 01/22/2025]
Abstract
The current opioid crisis has had an unprecedented public health impact. Approved medications for opioid use disorder (OUD) exist, yet their limitations indicate a need for innovative treatments. Limited preliminary clinical studies suggest specific psychedelics might aid OUD treatment, though most clinical evidence remains observational, with few controlled trials. This review aims to bridge the gap between preclinical findings and potential clinical applications, following PRISMA-ScR guidelines. Searches included MEDLINE, Embase, Scopus, and Web of Science, focusing on preclinical in vivo studies involving opioids and psychedelics in animals, excluding pain studies and those lacking control groups. Forty studies met criteria, covering both classic and non-classic psychedelics. Most studies showed that 18-methoxycoronaridine (18-MC), ibogaine, noribogaine, and ketamine could reduce opioid self-administration, alleviate withdrawal symptoms, and change conditioned place preference. However, seven studies (two on 2,5-dimethoxy-4-methylamphetamine (DOM), three on ibogaine, one on 18-MC, and one on ketamine) showed no improvement over controls. A methodological quality assessment rated most of the studies as having unclear quality. Interestingly, most preclinical studies are limited to iboga derivatives, which were effective, but these agents may have higher cardiovascular risk than other psychedelics under-explored to date. This review strengthens support for translational studies testing psychedelics as potential innovative targets for OUD. It also suggests clinical studies need to include a broader range of agents beyond iboga derivatives but can also explore several ongoing questions in the field, such as the mechanism of action behind the potential therapeutic effect, safety profiles, doses, and frequency of administrations needed.
Collapse
Affiliation(s)
- Alejandra Pulido-Saavedra
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Room 359, 34 Park Street, New Haven, CT, 06519, USA
| | - Henrique Nunes Pereira Oliva
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Room 359, 34 Park Street, New Haven, CT, 06519, USA
| | - Tiago Paiva Prudente
- Faculdade de Medicina, Universidade Federal de Goiás, 235 Street, Goiânia, Brasil
| | - Razi Kitaneh
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Room 359, 34 Park Street, New Haven, CT, 06519, USA
| | - Eric J Nunes
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Yale Tobacco Center of Regulatory Science, Yale University School of Medicine, New Haven, CT, USA
| | - Colleen Fogg
- Pharmacy Department, Yale-New Haven Health, Connecticut Mental Health Center, 34 Park Street, New Haven, CT, 06515, USA
| | - Melissa C Funaro
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Jeremy Weleff
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Anahita Bassir Nia
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Room 359, 34 Park Street, New Haven, CT, 06519, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Room 359, 34 Park Street, New Haven, CT, 06519, USA.
| |
Collapse
|
4
|
Vashchenko OV, Ye Brodskii R, Davydova IO, Vashchenko PV, Ivaniuk OI, Ruban OA. Biopharmaceutical studies of a novel sedative sublingual lozenge based on glycine and tryptophan: A rationale for mucoadhesive agent selection. Eur J Pharm Biopharm 2024; 203:114469. [PMID: 39186958 DOI: 10.1016/j.ejpb.2024.114469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Effective sedative drugs are in great demand due to increasing incidence of nervous disorders. The present work was aimed to develop a novel sublingual sedative drug based on glycine and L-tryptophan amino acids. Carbopol and different hydroxypropyl methylcellulose species were alternatively tested as mucoadhesive agents intended to prolong tryptophan sublingual release time. A model lipid medium of fully hydrated L-α-dimyristoylphosphatidylcholine was used for optimal mucoadhesive agents selection. Simultaneous processes of drug release and diffusion in lipid medium were first investigated involving both experimental and theoretical approaches. Individual substances, their selected combinations as well as different drug formulations were consecutively examined. Application of kinetic differential scanning calorimetry method allowed us to reveal a number of specific drug-excipient effects. Lactose was found to essentially facilitate tryptophan release and provide its ability to get into the bloodstream simultaneously with glycine, which is necessary to achieve glycine-tryptophan synergism. Introduction of a mucoadhesive agent into the formulation was shown to change kinetics of drug-membrane interactions variously depending on viscosity grade. Among the mucoadhesive agents, hydroxypropyl methylcellulose species K4M and E4M were shown to further accelerate drug release, therefore they were selected as optimal. Thus, effectiveness of the novel sedative drug was provided by including some excipients, such as lactose and the selected mucoadhesive agent species. A dynamic mathematical model was developed properly describing release and diffusion in lipid medium of various drug substances. Our study clearly showed applicability of a lipid medium to meet challenges such as drug-excipient interactions and optimization of drug formulations.
Collapse
Affiliation(s)
- O V Vashchenko
- Institute for Scintillation Materials, National Academy of Science of Ukraine, 60 Nauky Ave., 61072 Kharkov, Ukraine.
| | - R Ye Brodskii
- Institute for Single Crystals, National Academy of Science of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine
| | - I O Davydova
- National University of Pharmacy, 53 H. Skovorody Str., 61002 Kharkiv, Ukraine
| | - P V Vashchenko
- Institute for Scintillation Materials, National Academy of Science of Ukraine, 60 Nauky Ave., 61072 Kharkov, Ukraine
| | - O I Ivaniuk
- National University of Pharmacy, 53 H. Skovorody Str., 61002 Kharkiv, Ukraine
| | - O A Ruban
- National University of Pharmacy, 53 H. Skovorody Str., 61002 Kharkiv, Ukraine
| |
Collapse
|
5
|
El-Wakil MH, El-Dershaby HA, Ghazallah RA, El-Yazbi AF, Abd El-Razik HA, Soliman FSG. Identification of new 5-(2,6-dichlorophenyl)-3-oxo-2,3-dihydro-5H-thiazolo[3,2-a]pyrimidine-7-carboxylic acids as p38α MAPK inhibitors: Design, synthesis, antitumor evaluation, molecular docking and in silico studies. Bioorg Chem 2024; 145:107226. [PMID: 38377818 DOI: 10.1016/j.bioorg.2024.107226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024]
Abstract
In pursuit of discovering novel scaffolds that demonstrate potential inhibitory activity against p38α MAPK and possess strong antitumor effects, we herein report the design and synthesis of new series of 17 final target 5-(2,6-dichlorophenyl)-3-oxo-2,3-dihydro-5H-thiazolo[3,2-a]pyrimidine-7-carboxylic acids (4-20). Chemical characterization of the compounds was performed using FT-IR, NMR, elemental analyses and mass spectra of some representative examples. With many compounds showing potential inhibitory activity against p38α MAPK, two derivatives, 8 and 9, demonstrated the highest activity (>70 % inhibition) among the series. Derivative 9 displayed IC50 value nearly 2.5 folds more potent than 8. As anticipated, they both showed explicit interactions inside the kinase active site with the key binding amino acid residues. Screening both compounds for cytotoxic effects, they exhibited strong antitumor activities against lung (A549), breast (MCF-7 and MDA MB-231), colon (HCT-116) and liver (Hep-G2) cancers more potent than reference 5-FU. Their noticeable strong antitumor activity pointed out to the possibility of an augmented DNA binding mechanism of antitumor action besides their kinase inhibition. Both 8 and 9 exhibited strong ctDNA damaging effects in nanomolar range. Further mechanistic antitumor studies revealed ability of compounds 8 and 9 to arrest cell cycle in MCF-7 cells at S phase, while in HCT-116 treated cells at G0-G1 and G2/M phases. They also displayed apoptotic induction effects in both MCF-7 and HCT-116 with total cell deaths more than control untreated cells in reference to 5-FU. Finally, the compounds were tested for their anti-migratory potential utilizing wound healing assay. They induced a significant decrease in wound closure percentage after 24 h treatment in the examined cancer cells when compared to untreated control MCF-7 and HCT-116 cells better than 5-FU. In silico computation of physicochemical parameters revealed the drug-like properties of 8 and 9 with no violation to Lipinski's rule of five as well as their tolerable ADMET parameters, thus suggesting their utilization as potential future drug leads amenable for further optimization and development.
Collapse
Affiliation(s)
- Marwa H El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Hadeel A El-Dershaby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Rasha A Ghazallah
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria 21521, Egypt
| | - Amira F El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Heba A Abd El-Razik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Farid S G Soliman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
6
|
Luo X, Zhang Z, Mu R, Hu G, Liu L, Liu X. Simultaneously Predicting the Pharmacokinetics of CES1-Metabolized Drugs and Their Metabolites Using Physiologically Based Pharmacokinetic Model in Cirrhosis Subjects. Pharmaceutics 2024; 16:234. [PMID: 38399287 PMCID: PMC10893190 DOI: 10.3390/pharmaceutics16020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Hepatic carboxylesterase 1 (CES1) metabolizes numerous prodrugs into active ingredients or direct-acting drugs into inactive metabolites. We aimed to develop a semi-physiologically based pharmacokinetic (semi-PBPK) model to simultaneously predict the pharmacokinetics of CES1 substrates and their active metabolites in liver cirrhosis (LC) patients. Six prodrugs (enalapril, benazepril, cilazapril, temocapril, perindopril and oseltamivir) and three direct-acting drugs (flumazenil, pethidine and remimazolam) were selected. Parameters such as organ blood flows, plasma-binding protein concentrations, functional liver volume, hepatic enzymatic activity, glomerular filtration rate (GFR) and gastrointestinal transit rate were integrated into the simulation. The pharmacokinetic profiles of these drugs and their active metabolites were simulated for 1000 virtual individuals. The developed semi-PBPK model, after validation in healthy individuals, was extrapolated to LC patients. Most of the observations fell within the 5th and 95th percentiles of simulations from 1000 virtual patients. The estimated AUC and Cmax were within 0.5-2-fold of the observed values. The sensitivity analysis showed that the decreased plasma exposure of active metabolites due to the decreased CES1 was partly attenuated by the decreased GFR. Conclusion: The developed PBPK model successfully predicted the pharmacokinetics of CES1 substrates and their metabolites in healthy individuals and LC patients, facilitating tailored dosing of CES1 substrates in LC patients.
Collapse
Affiliation(s)
| | | | | | | | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (Z.Z.); (R.M.); (G.H.)
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (Z.Z.); (R.M.); (G.H.)
| |
Collapse
|
7
|
Mtetwa LM, Salifu EY, Omolo CA, Soliman ME, Faya M. Halting aberrant DNA methylation via in silico Identification of potent inhibitors of DNMT3B enzyme: Atomistic insights. Comput Biol Chem 2023; 105:107909. [PMID: 37418952 DOI: 10.1016/j.compbiolchem.2023.107909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 07/09/2023]
Abstract
To date, Cancer remains a global threat due to its impact on growing life expectancy. With the many efforts and methods of combating the disease, complete success remains a challenge owing to several limitations including cancer cells developing resistance through mutations, off-target effect of some cancer drugs resulting in toxicities, among many others. Aberrant DNA methylation is understood to be the primary reason for improper gene silence, which can result in neoplastic transformation, carcinogenesis, and tumour progression. DNA methyltransferase B (DNMT3B) enzyme is considered a potential target for the treatment of several cancers due to its important role in DNA methylation. However, only a few DNMT3B inhibitors have been reported to date. Herein, in silico molecular recognition techniques such as Molecular docking, Pharmacophore-based virtual screen and MD simulation were employed to identify potential inhibitors of DNMT3B that can halt aberrancy in DNA methylation. Findings initially identified 878 hit compounds based on a designed pharmacophore model from the reference compound Hypericin. Molecular docking was used to rank the hits by testing their efficiency when bound to the target enzyme and the top three (3) selected. All three (3) of the top hits showed excellent pharmacokinetic properties but two (2) (Zinc33330198 and Zinc77235130) were identified to be non-toxic. Molecular dynamic simulation of the final two hits showed good stability, flexibility, and structural rigidity of the compounds on DNMT3B. Finally, thermodynamic energy estimations show both compounds had favourable free energies comprising - 26.04 kcal/mol for Zinc77235130 and - 15.73 kcal/mol for Zinc33330198. Amongst the final two hits, Zinc77235130 showed consistency in favourable results across all the tested parameters and was thus selected as the lead compound for further experimental validation. The identification of this lead compound will form important basis for the inhibition of aberrant DNA methylation in cancer therapy.
Collapse
Affiliation(s)
- Lusanda M Mtetwa
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Elliasu Y Salifu
- West African Centre for Computational Analysis, Accra, Ghana; Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; School of Pharmacy and Health Sciences, United States International University of Africa, Nairobi, Kenya
| | - Mahmoud E Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Mbuso Faya
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
8
|
Ieritano C, Hopkins WS. The hitchhiker's guide to dynamic ion-solvent clustering: applications in differential ion mobility spectrometry. Phys Chem Chem Phys 2022; 24:20594-20615. [PMID: 36000315 DOI: 10.1039/d2cp02540j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This article highlights the fundamentals of ion-solvent clustering processes that are pertinent to understanding an ion's behaviour during differential mobility spectrometry (DMS) experiments. We contrast DMS with static-field ion mobility, where separation is affected by mobility differences under the high-field and low-field conditions of an asymmetric oscillating electric field. Although commonly used in mass spectrometric (MS) workflows to enhance signal-to-noise ratios and remove isobaric contaminants, the chemistry and physics that underpins the phenomenon of differential mobility has yet to be fully fleshed out. Moreover, we are just now making progress towards understanding how the DMS separation waveform creates a dynamic clustering environment when the carrier gas is seeded with the vapour of a volatile solvent molecule (e.g., methanol). Interestingly, one can correlate the dynamic clustering behaviour observed in DMS experiments with gas-phase and solution-phase molecular properties such as hydrophobicity, acidity, and solubility. However, to create a generalized, global model for property determination using DMS data one must employ machine learning. In this article, we provide a first-principles description of differential ion mobility in a dynamic clustering environment. We then discuss the correlation between dynamic clustering propensity and analyte physicochemical properties and demonstrate that analytes exhibiting similar ion-solvent interactions (e.g., charge-dipole) follow well-defined trends with respect to DMS clustering behaviour. Finally, we describe how supervised machine learning can be used to create predictive models of molecular properties using DMS data. We additionally highlight open questions in the field and provide our perspective on future directions that can be explored.
Collapse
Affiliation(s)
- Christian Ieritano
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada. .,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.,Watermine Innovation, Waterloo, Ontario, N0B 2T0, Canada
| | - W Scott Hopkins
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada. .,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.,Watermine Innovation, Waterloo, Ontario, N0B 2T0, Canada.,Centre for Eye and Vision Research, 17W Hong Kong Science Park, New Territories, 999077, Hong Kong
| |
Collapse
|
9
|
Yue P, Zhu Y, Brotherton-Pleiss C, Fu W, Verma N, Chen J, Nakamura K, Chen W, Chen Y, Alonso-Valenteen F, Mikhael S, Medina-Kauwe L, Kershaw KM, Celeridad M, Pan S, Limpert AS, Sheffler DJ, Cosford NDP, Shiao SL, Tius MA, Lopez-Tapia F, Turkson J. Novel potent azetidine-based compounds irreversibly inhibit Stat3 activation and induce antitumor response against human breast tumor growth in vivo. Cancer Lett 2022; 534:215613. [PMID: 35276290 PMCID: PMC9867837 DOI: 10.1016/j.canlet.2022.215613] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/27/2022] [Indexed: 01/26/2023]
Abstract
Signal transducer and activator of transcription (Stat)3 is a valid anticancer therapeutic target. We have discovered a highly potent chemotype that amplifies the Stat3-inhibitory activity of lead compounds to levels previously unseen. The azetidine-based compounds, including H172 (9f) and H182, irreversibly bind to Stat3 and selectively inhibit Stat3 activity (IC50 0.38-0.98 μM) over Stat1 or Stat5 (IC50 > 15.8 μM) in vitro. Mass spectrometry detected the Stat3 cysteine peptides covalently bound to the azetidine compounds, and the key residues, Cys426 and Cys468, essential for the high potency inhibition, were confirmed by site-directed mutagenesis. In triple-negative breast cancer (TNBC) models, treatment with the azetidine compounds inhibited constitutive and ligand-induced Stat3 signaling, and induced loss of viable cells and tumor cell death, compared to no effect on the induction of Janus kinase (JAK)2, Src, epidermal growth factor receptor (EGFR), and other proteins, or weak effects on cells that do not harbor aberrantly-active Stat3. H120 (8e) and H182 as a single agent inhibited growth of TNBC xenografts, and H278 (hydrochloric acid salt of H182) in combination with radiation completely blocked mouse TNBC growth and improved survival in syngeneic models. We identify potent azetidine-based, selective, irreversible Stat3 inhibitors that inhibit TNBC growth in vivo.
Collapse
Affiliation(s)
- Peibin Yue
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA,Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Yinsong Zhu
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA,Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Christine Brotherton-Pleiss
- Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, HI, 96813, USA,Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI, 96825, USA
| | - Wenzhen Fu
- Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, HI, 96813, USA,Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI, 96825, USA
| | - Nagendra Verma
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA,Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Jasmine Chen
- Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, HI, 96813, USA
| | - Kayo Nakamura
- Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI, 96825, USA
| | - Weiliang Chen
- Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI, 96825, USA
| | - Yue Chen
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA,Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Felix Alonso-Valenteen
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Simoun Mikhael
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Lali Medina-Kauwe
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Kathleen M. Kershaw
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Department of Radiation Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Maria Celeridad
- Cell and Molecular Biology of Cancer Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Songqin Pan
- W. M. Keck Proteomics Laboratory, University of California, Riverside, CA, 92521, USA
| | - Allison S. Limpert
- Cell and Molecular Biology of Cancer Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Douglas J. Sheffler
- Cell and Molecular Biology of Cancer Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Nicholas D. P. Cosford
- Cell and Molecular Biology of Cancer Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Stephen L. Shiao
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Department of Radiation Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Marcus A. Tius
- Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, HI, 96813, USA,Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI, 96825, USA
| | - Francisco Lopez-Tapia
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA,Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA,Corresponding author. Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA. (J. Turkson)
| | - James Turkson
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angenes, CA, 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.
| |
Collapse
|
10
|
Liu H, Iketani S, Zask A, Khanizeman N, Bednarova E, Forouhar F, Fowler B, Hong SJ, Mohri H, Nair MS, Huang Y, Tay NES, Lee S, Karan C, Resnick SJ, Quinn C, Li W, Shion H, Xia X, Daniels JD, Bartolo-Cruz M, Farina M, Rajbhandari P, Jurtschenko C, Lauber MA, McDonald T, Stokes ME, Hurst BL, Rovis T, Chavez A, Ho DD, Stockwell BR. Development of optimized drug-like small molecule inhibitors of the SARS-CoV-2 3CL protease for treatment of COVID-19. Nat Commun 2022; 13:1891. [PMID: 35393402 PMCID: PMC8989888 DOI: 10.1038/s41467-022-29413-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
The SARS-CoV-2 3CL protease is a critical drug target for small molecule COVID-19 therapy, given its likely druggability and essentiality in the viral maturation and replication cycle. Based on the conservation of 3CL protease substrate binding pockets across coronaviruses and using screening, we identified four structurally distinct lead compounds that inhibit SARS-CoV-2 3CL protease. After evaluation of their binding specificity, cellular antiviral potency, metabolic stability, and water solubility, we prioritized the GC376 scaffold as being optimal for optimization. We identified multiple drug-like compounds with <10 nM potency for inhibiting SARS-CoV-2 3CL and the ability to block SARS-CoV-2 replication in human cells, obtained co-crystal structures of the 3CL protease in complex with these compounds, and determined that they have pan-coronavirus activity. We selected one compound, termed coronastat, as an optimized lead and characterized it in pharmacokinetic and safety studies in vivo. Coronastat represents a new candidate for a small molecule protease inhibitor for the treatment of SARS-CoV-2 infection for eliminating pandemics involving coronaviruses. Small molecule drugs promise to remain a valuable tool in controlling the ongoing COVID-19 pandemic. Here the authors describe optimized drug-like small molecule inhibitors of the SARS-CoV-2 3CL protease for potential treatment of COVID-19.
Collapse
Affiliation(s)
- Hengrui Liu
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Arie Zask
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Nisha Khanizeman
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Eva Bednarova
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Farhad Forouhar
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Brandon Fowler
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Seo Jung Hong
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hiroshi Mohri
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Nicholas E S Tay
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Sumin Lee
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Charles Karan
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, 10032, USA
| | - Samuel J Resnick
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Colette Quinn
- Waters Corporation, 34 Maple Street, Milford, MA, 01757, USA
| | - Wenjing Li
- Waters Corporation, 34 Maple Street, Milford, MA, 01757, USA
| | - Henry Shion
- Waters Corporation, 34 Maple Street, Milford, MA, 01757, USA
| | - Xin Xia
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Jacob D Daniels
- Department of Pharmacology and Molecular Therapeutics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Marcelo Farina
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.,Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Presha Rajbhandari
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | | | | | - Thomas McDonald
- Waters Corporation, 34 Maple Street, Milford, MA, 01757, USA
| | - Michael E Stokes
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Brett L Hurst
- Institute for Antiviral Research, Utah State University, Logan, UT, 84322, USA
| | - Tomislav Rovis
- Department of Chemistry, Columbia University, New York, NY, 10027, USA.
| | - Alejandro Chavez
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, 10027, USA. .,Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
11
|
Williams J, Siramshetty V, Nguyễn ÐT, Padilha EC, Kabir M, Yu KR, Wang AQ, Zhao T, Itkin M, Shinn P, Mathé EA, Xu X, Shah P. Using in vitro ADME data for lead compound selection: An emphasis on PAMPA pH 5 permeability and oral bioavailability. Bioorg Med Chem 2022; 56:116588. [PMID: 35030421 PMCID: PMC9843724 DOI: 10.1016/j.bmc.2021.116588] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 01/19/2023]
Abstract
Membrane permeability plays an important role in oral drug absorption. Caco-2 and Madin-Darby Canine Kidney (MDCK) cell culture systems have been widely used for assessing intestinal permeability. Since most drugs are absorbed passively, Parallel Artificial Membrane Permeability Assay (PAMPA) has gained popularity as a low-cost and high-throughput method in early drug discovery when compared to high-cost, labor intensive cell-based assays. At the National Center for Advancing Translational Sciences (NCATS), PAMPA pH 5 is employed as one of the Tier I absorption, distribution, metabolism, and elimination (ADME) assays. In this study, we have developed a quantitative structure activity relationship (QSAR) model using our ∼6500 compound PAMPA pH 5 permeability dataset. Along with ensemble decision tree-based methods such as Random Forest and eXtreme Gradient Boosting, we employed deep neural network and a graph convolutional neural network to model PAMPA pH 5 permeability. The classification models trained on a balanced training set provided accuracies ranging from 71% to 78% on the external set. Of the four classifiers, the graph convolutional neural network that directly operates on molecular graphs offered the best classification performance. Additionally, an ∼85% correlation was obtained between PAMPA pH 5 permeability and in vivo oral bioavailability in mice and rats. These results suggest that data from this assay (experimental or predicted) can be used to rank-order compounds for preclinical in vivo testing with a high degree of confidence, reducing cost and attrition as well as accelerating the drug discovery process. Additionally, experimental data for 486 compounds (PubChem AID: 1645871) and the best models have been made publicly available (https://opendata.ncats.nih.gov/adme/).
Collapse
Affiliation(s)
- Jordan Williams
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Vishal Siramshetty
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ðắc-Trung Nguyễn
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Elias Carvalho Padilha
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Md Kabir
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States,The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, United States
| | - Kyeong-Ri Yu
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States,Department of Surgery, Virginia Commonwealth University Health Systems, 1200 E Broad St, Richmond, Virginia 23298, United States
| | - Amy Q. Wang
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Tongan Zhao
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Misha Itkin
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ewy A. Mathé
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Xin Xu
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Pranav Shah
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, Maryland 20850, United States,Corresponding Author: Pranav Shah,
| |
Collapse
|
12
|
Hoshijima H, Hunt M, Nagasaka H, Yaksh T. Systematic Review of Systemic and Neuraxial Effects of Acetaminophen in Preclinical Models of Nociceptive Processing. J Pain Res 2021; 14:3521-3552. [PMID: 34795520 PMCID: PMC8594782 DOI: 10.2147/jpr.s308028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/11/2021] [Indexed: 12/29/2022] Open
Abstract
Acetaminophen (APAP) in humans has robust effects with a high therapeutic index in altering postoperative and inflammatory pain states in clinical and experimental pain paradigms with no known abuse potential. This review considers the literature reflecting the preclinical actions of acetaminophen in a variety of pain models. Significant observations arising from this review are as follows: 1) acetaminophen has little effect upon acute nociceptive thresholds; 2) acetaminophen robustly reduces facilitated states as generated by mechanical and thermal hyperalgesic end points in mouse and rat models of carrageenan and complete Freund’s adjuvant evoked inflammation; 3) an antihyperalgesic effect is observed in models of facilitated processing with minimal inflammation (eg, phase II intraplantar formalin); and 4) potent anti-hyperpathic effects on the thermal hyperalgesia, mechanical and cold allodynia, allodynic thresholds in rat and mouse models of polyneuropathy and mononeuropathies and bone cancer pain. These results reflect a surprisingly robust drug effect upon a variety of facilitated states that clearly translate into a wide range of efficacy in preclinical models and to important end points in human therapy. The specific systems upon which acetaminophen may act based on targeted delivery suggest both a spinal and a supraspinal action. Review of current targets for this molecule excludes a role of cyclooxygenase inhibitor but includes effects that may be mediated through metabolites acting on the TRPV1 channel, or by effect upon cannabinoid and serotonin signaling. These findings suggest that the mode of action of acetaminophen, a drug with a long therapeutic history of utilization, has surprisingly robust effects on a variety of pain states in clinical patients and in preclinical models with a good therapeutic index, but in spite of its extensive use, its mechanisms of action are yet poorly understood.
Collapse
Affiliation(s)
- Hiroshi Hoshijima
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Matthew Hunt
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| | - Hiroshi Nagasaka
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Tony Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| |
Collapse
|
13
|
Sakolish C, Luo YS, Valdiviezo A, Vernetti LA, Rusyn I, Chiu WA. Prediction of hepatic drug clearance with a human microfluidic four-cell liver acinus microphysiology system. Toxicology 2021; 463:152954. [PMID: 34543702 PMCID: PMC8585690 DOI: 10.1016/j.tox.2021.152954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022]
Abstract
Predicting human hepatic clearance remains a fundamental challenge in both pharmaceutical drug development and toxicological assessments of environmental chemicals, with concerns about both accuracy and precision of in vitro-derived estimates. Suggested sources of these issues have included differences in experimental protocols, differences in cell sourcing, and use of a single cell type, liver parenchymal cells (hepatocytes). Here we investigate the ability of human microfluidic four-cell liver acinus microphysiology system (LAMPS) to make predictions as to hepatic clearance for seven representative compounds: Caffeine, Pioglitazone, Rosiglitazone, Terfenadine, Tolcapone, Troglitazone, and Trovafloxacin. The model, whose reproducibility was recently confirmed in an inter-lab comparison, was constructed using primary human hepatocytes or human induced pluripotent stem cell (iPSC)-derived hepatocytes and 3 human cell lines for the endothelial, Kupffer and stellate cells. We calculated hepatic clearance estimates derived from experiments using LAMPS or traditional 2D cultures and compared the outcomes with both in vivo human clinical study-derived and in vitro human hepatocyte suspension culture-derived values reported in the literature. We found that, compared to in vivo clinically-derived values, the LAMPS model with iPSC-derived hepatocytes had higher precision as compared to primary cells in suspension or 2D culture, but, consistent with previous studies in other microphysiological systems, tended to underestimate in vivo clearance. Overall, these results suggest that use of LAMPS and iPSC-derived hepatocytes together with an empirical scaling factor warrants additional study with a larger set of compounds, as it has the potential to provide more accurate and precise estimates of hepatic clearance.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; Institute of Food Safety and Health, National Taiwan University, Taipei 10617, Taiwan(1)
| | - Alan Valdiviezo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Lawrence A Vernetti
- Drug Discovery Institute and Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
14
|
Omoboyowa DA, Balogun TA, Omomule OM, Saibu OA. Identification of Terpenoids From Abrus precatorius Against Parkinson's Disease Proteins Using In Silico Approach. Bioinform Biol Insights 2021; 15:11779322211050757. [PMID: 34707350 PMCID: PMC8544761 DOI: 10.1177/11779322211050757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD) is the second major neuro-degenrative disorder that causes morbidity and mortality among older populations. Terpenoids were reported as potential neuro-protective agents. Therefore, this study seeks to unlock the inhibitory potential of terpenoids from Abrus precatorius seeds against proteins involve in PD pathogenesis. In this study, in silico molecular docking of 5 terpenoids derived from high-performance liquid chromatography (HPLC) analysis of A. precatorius seeds against α-synuclein, catechol-o-methyltransferase, and monoamine oxidase B which are markers of PD was performed using Autodock vina. The absorption, distribution, metabolism, excretion, and toxicity (ADME/Tox) of the hits were done using Swiss ADME predictor and molecular dynamic (MD) simulation of the hit-protein complex was performed using Desmond Schrodinger software. Five out of 6 compounds satisfied the ADME/Tox parameters and showed varying degrees of binding affinities with selected proteins. Drimenin-α-synuclein complex showed the lowest binding energy of −9.1 kcal/mol followed by interaction with key amino acid residues necessary for α-synuclein inhibition. The selection of this complex was justified by its stability in MD simulation conducted for 10 ns and exhibited stable interaction in terms of root mean square deviation (RMSD) and root mean square deviation error fluctuation (RMSF) values.
Collapse
Affiliation(s)
| | | | | | - Oluwatosin A Saibu
- Department of Environmental Toxicology, Universitat Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
15
|
Computer-aided anticancer drug design: In vitro and in silico studies of new iminocoumarin derivative. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
16
|
Toyama K, Furuie H, Kuroda K, Ishizuka T, Okuda Y, Shimizu T, Kato M, Igawa Y, Nishikawa Y, Ishizuka H. Effects of Repeated Oral Administration of Esaxerenone on the Pharmacokinetics of Midazolam in Healthy Japanese Males. Eur J Drug Metab Pharmacokinet 2021; 46:685-694. [PMID: 34383278 PMCID: PMC8397627 DOI: 10.1007/s13318-021-00701-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Esaxerenone showed the potential to inhibit and induce activity against cytochrome P450 (CYP) 3A in in vitro studies. We investigated whether repeated administration of 5 mg/day esaxerenone for 14 days influences the pharmacokinetics of midazolam, a sensitive CYP3A substrate, in healthy Japanese males. METHODS This single-centre, open-label, single-sequence study had two administration periods: period 1: single oral dose of 2 mg midazolam (day 0); period 2: repeated oral doses of 5 mg/day esaxerenone for 14 days, with a single oral dose of 2 mg midazolam on day 14. Full pharmacokinetic profiles of midazolam and 1-hydroxymidazolam on days 0 and 14 and safety data were obtained. Primary pharmacokinetic endpoints for midazolam were area under the plasma concentration-time curve (AUC) from zero to time of the last measurable concentration (AUClast), AUC from zero to infinity (AUCinf), and peak plasma concentration (Cmax). RESULTS The study included 28 male subjects. One subject was withdrawn because of a mild adverse event (increased hepatic enzyme levels) that resolved without intervention. Repeated administration of esaxerenone increased midazolam AUClast, AUCinf, and Cmax by about 1.2-fold (1.201, 1.201, and 1.224, respectively) compared with administration of midazolam alone. However, repeated administration of esaxerenone did not affect the elimination half-life of midazolam (2.86 versus 2.63 h with and without esaxerenone). There were no safety concerns associated with concomitant administration of esaxerenone and midazolam. CONCLUSIONS Esaxerenone 5 mg/day had no clinically significant effect on midazolam pharmacokinetics and was not associated with any safety issues. Esaxerenone can be concomitantly administered with drugs of CYP3A substrates without dose adjustments. CLINICAL TRIAL REGISTRATION JapiCTI-152832.
Collapse
|
17
|
Wall BJ, Will MF, Yawson GK, Bothwell PJ, Platt DC, Apuzzo CF, Jones MA, Ferrence GM, Webb MI. Importance of Hydrogen Bonding: Structure-Activity Relationships of Ruthenium(III) Complexes with Pyridine-Based Ligands for Alzheimer's Disease Therapy. J Med Chem 2021; 64:10124-10138. [PMID: 34197109 DOI: 10.1021/acs.jmedchem.1c00360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, where one of the pathological hallmarks of AD is extracellular protein deposits, the primary component of which is the peptide amyloid-β (Aβ). Recently, the soluble form of Aβ has been recognized as the primary neurotoxic species, making it an important target for therapeutic development. Metal-based drugs are promising candidates to target Aβ, as the interactions with the peptide can be tuned by ligand design. In the current study, 11 ruthenium complexes containing pyridine-based ligands were prepared, where the functional groups at the para position on the coordinated pyridine ligand were varied to determine structure-activity relationships. Overall, the complexes with terminal primary amines had the greatest impact on modulating the aggregation of Aβ and diminishing its cytotoxicity. These results identify the importance of specific intermolecular interactions and are critical in the advancement of metal-based drugs for AD therapy.
Collapse
Affiliation(s)
- Brendan J Wall
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Mark F Will
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gideon K Yawson
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois 60115, United States
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - C Fiore Apuzzo
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gregory M Ferrence
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| |
Collapse
|
18
|
Brotherton-Pleiss C, Yue P, Zhu Y, Nakamura K, Chen W, Fu W, Kubota C, Chen J, Alonso-Valenteen F, Mikhael S, Medina-Kauwe L, Tius MA, Lopez-Tapia F, Turkson J. Discovery of Novel Azetidine Amides as Potent Small-Molecule STAT3 Inhibitors. J Med Chem 2021; 64:695-710. [PMID: 33352047 PMCID: PMC7816766 DOI: 10.1021/acs.jmedchem.0c01705] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Indexed: 02/07/2023]
Abstract
We optimized our previously reported proline-based STAT3 inhibitors into an exciting new series of (R)-azetidine-2-carboxamide analogues that have sub-micromolar potencies. 5a, 5o, and 8i have STAT3-inhibitory potencies (IC50) of 0.55, 0.38, and 0.34 μM, respectively, compared to potencies greater than 18 μM against STAT1 or STAT5 activity. Further modifications derived analogues, including 7e, 7f, 7g, and 9k, that addressed cell membrane permeability and other physicochemical issues. Isothermal titration calorimetry analysis confirmed high-affinity binding to STAT3, with KD of 880 nM (7g) and 960 nM (9k). 7g and 9k inhibited constitutive STAT3 phosphorylation and DNA-binding activity in human breast cancer, MDA-MB-231 or MDA-MB-468 cells. Furthermore, treatment of breast cancer cells with 7e, 7f, 7g, or 9k inhibited viable cells, with an EC50 of 0.9-1.9 μM, cell growth, and colony survival, and induced apoptosis while having relatively weaker effects on normal breast epithelial, MCF-10A or breast cancer, MCF-7 cells that do not harbor constitutively active STAT3.
Collapse
Affiliation(s)
- Christine Brotherton-Pleiss
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - Peibin Yue
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| | - Yinsong Zhu
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| | - Kayo Nakamura
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Weiliang Chen
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Wenzhen Fu
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Casie Kubota
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
| | - Jasmine Chen
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
| | - Felix Alonso-Valenteen
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Simoun Mikhael
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Lali Medina-Kauwe
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Marcus A. Tius
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - Francisco Lopez-Tapia
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - James Turkson
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| |
Collapse
|
19
|
Farid A, Hesham M, El-Dewak M, Amin A. The hidden hazardous effects of stevia and sucralose consumption in male and female albino mice in comparison to sucrose. Saudi Pharm J 2020; 28:1290-1300. [PMID: 33132722 PMCID: PMC7584803 DOI: 10.1016/j.jsps.2020.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
Replacing sucrose with non-caloric sweeteners is an approach to avoid overweight and diabetes development. Non-caloric sweeteners are classified into either artificial as sucralose or natural as stevia. Both of them have been approved by FDA, but the effects of their chronic consumption are controversial. The present study aimed to evaluate the effects of these two sweeteners, in male and female albino mice, on different blood biochemical parameters, enzymes activities and immunological parameters after 8 and 16 weeks of sweeteners administration. 40.5 mg/ml of sucrose, 5.2 mg/ml of sucralose and 4.2 mg/ml of stevia were dissolved individually in distilled water. Mice were administrated by sweetener's solution for 5 h daily. Male and female mice showed a preference for water consumption with sucralose or stevia. Both of the two sweeteners significantly reduced the hemoglobin level, HCT%, RBCs and WBCs count. After 18 weeks, significant elevations in liver and kidney function enzymes were observed in male and female mice administrated with both non-caloric sweeteners. Histopathological examination in sucralose and stevia administrated groups confirmed the biochemical results; where it revealed a severe damage in liver and kidney sections. While, sucrose administration elevated, only, the levels of ALT, AST and cholesterol in male mice. A vigorous elevation in levels of different immunoglobulin (IgG, IgE and IgA) and pro-inflammatory cytokines (IL-6 and -8), that was accompanied by a significant reduction in level of anti-inflammatory cytokine IL-10, was observed in male and female mice groups administrated with sucralose or stevia. On the other hand, sucrose administration led to an elevation in IgA and reduction in IL-10 levels.
Collapse
Affiliation(s)
- Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Egypt
| | - Marim Hesham
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Egypt
| | - Mohamed El-Dewak
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Egypt
| | - Ayman Amin
- Department of Plant Physiology, Faculty of Agriculture, Cairo University, Egypt
| |
Collapse
|
20
|
Arjmand MH, Zahedi-Avval F, Barneh F, Mousavi SH, Asgharzadeh F, Hashemzehi M, Soleimani A, Avan A, Fakhraie M, Nasiri SN, Mehraban S, Ferns GA, Ryzhikov M, Jafari M, Khazaei M, Hassanian SM. Intraperitoneal Administration of Telmisartan Prevents Postsurgical Adhesion Band Formation. J Surg Res 2020; 248:171-181. [PMID: 31923833 DOI: 10.1016/j.jss.2019.10.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Angiotensin II receptor blockers (ARBs) have a potential role in reducing inflammation and fibrosis. We have integrated systems and molecular biology approaches to investigate the therapeutic potential of ARBs in preventing postsurgical adhesion band formation. MATERIAL AND METHODS we have followed the ARRIVE guidelines point by point during experimental studies. Telmisartan (1 and 9 mg/kg), valsartan (1 and 9 mg/kg), and losartan (1 and 10 mg/kg) were administered intraperitoneally in different groups of male albino Wistar rat. After 7 d of treatment, macroscopic evidence and score of fibrotic bands based on scaling methods was performed. Moreover, the anti-inflammatory and antifibrosis effects of telmisartan on reduction of fibrotic bands were investigated by using histopathology, ELISA, and real-time polymerase chain reaction methods. RESULTS Telmisartan, but not losartan or valsartan, prevented the frequency as well as the stability of adhesion bands. Telmisartan appears to elicit anti-inflammatory responses by attenuating submucosal edema, suppressing proinflammatory cytokines, decreasing proinflammatory cell infiltration, and inhibiting oxidative stress at the site of peritoneal surgery. We also showed that telmisartan prevents fibrotic adhesion band formation by reducing excessive collagen deposition and suppression of profibrotic genes expression at the peritoneum adhesion tissues. CONCLUSIONS These results support the potential application of telmisartan in preventing postsurgical adhesion band formation by inhibiting key pathologic responses of inflammation and fibrosis in postsurgery patients.
Collapse
Affiliation(s)
- Mohammad-Hassan Arjmand
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Zahedi-Avval
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Barneh
- Genetics Research Center, University of Social Welfare and Rehabilitation, Tehran, Iran
| | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Hashemzehi
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee and Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Maryam Fakhraie
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Najibeh Nasiri
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeedeh Mehraban
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, Sussex, UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, Missouri
| | - Mohieddin Jafari
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Determining molecular properties with differential mobility spectrometry and machine learning. Nat Commun 2018; 9:5096. [PMID: 30504922 PMCID: PMC6269546 DOI: 10.1038/s41467-018-07616-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 11/06/2018] [Indexed: 01/30/2023] Open
Abstract
The fast and accurate determination of molecular properties is highly desirable for many facets of chemical research, particularly in drug discovery where pre-clinical assays play an important role in paring down large sets of drug candidates. Here, we present the use of supervised machine learning to treat differential mobility spectrometry - mass spectrometry data for ten topological classes of drug candidates. We demonstrate that the gas-phase clustering behavior probed in our experiments can be used to predict the candidates' condensed phase molecular properties, such as cell permeability, solubility, polar surface area, and water/octanol distribution coefficient. All of these measurements are performed in minutes and require mere nanograms of each drug examined. Moreover, by tuning gas temperature within the differential mobility spectrometer, one can fine tune the extent of ion-solvent clustering to separate subtly different molecular geometries and to discriminate molecules of very similar physicochemical properties.
Collapse
|
22
|
Nair A, Morsy MA, Jacob S. Dose translation between laboratory animals and human in preclinical and clinical phases of drug development. Drug Dev Res 2018; 79:373-382. [DOI: 10.1002/ddr.21461] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Anroop Nair
- Department of Pharmaceutical SciencesCollege of Clinical Pharmacy, King Faisal University Al‐Ahsa Saudi Arabia
| | - Mohamed Aly Morsy
- Department of Pharmaceutical SciencesCollege of Clinical Pharmacy, King Faisal University Al‐Ahsa Saudi Arabia
- Department of Pharmacology, Faculty of MedicineMinia University El‐Minia Egypt
| | - Shery Jacob
- Department of Pharmaceutical SciencesCollege of Pharmacy, Gulf Medical University Ajman UAE
| |
Collapse
|
23
|
In Silico study of the active site of Helicobacter pylori urease and its inhibition by hydroxamic acids. J Mol Graph Model 2018; 83:64-73. [PMID: 29775804 DOI: 10.1016/j.jmgm.2018.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 11/21/2022]
Abstract
Hydroxamic acids have emerged as the most promising candidates from among the different classes of inhibitors of urease. In order to understand the mechanism of their action, we have studied in detail using quantum mechanics the active site of Helicobacter pylori urease complexed with acetohydroxamic acid. A diverse library of ligands having the hydroxamate moiety has been prepared and docked into the active site of urease using the QM/MM methodology. It is found that hydroxamic acids with hydrophobic groups attached to them are more potent inhibitors of urease because they can easily penetrate the hydrophobic environment surrounding the active site. The -CONHO- moiety of the hydroxamic acid is also found to be absolutely necessary for chelation and inhibition of urease. In order to determine the roles of residues His 221 and Ala 365, which are not part of the active site, but are nevertheless involved in hydrogen bonding with the ligand, we have performed Molecular Dynamics simulations, both on the wild urease and also on its mutated counterpart, with the two residues substituted, respectively, by alanine and glycine.
Collapse
|
24
|
Cabrera-Pérez MÁ, Pham-The H. Computational modeling of human oral bioavailability: what will be next? Expert Opin Drug Discov 2018; 13:509-521. [PMID: 29663836 DOI: 10.1080/17460441.2018.1463988] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The oral route is the most convenient way of administrating drugs. Therefore, accurate determination of oral bioavailability is paramount during drug discovery and development. Quantitative structure-property relationship (QSPR), rule-of-thumb (RoT) and physiologically based-pharmacokinetic (PBPK) approaches are promising alternatives to the early oral bioavailability prediction. Areas covered: The authors give insight into the factors affecting bioavailability, the fundamental theoretical framework and the practical aspects of computational methods for predicting this property. They also give their perspectives on future computational models for estimating oral bioavailability. Expert opinion: Oral bioavailability is a multi-factorial pharmacokinetic property with its accurate prediction challenging. For RoT and QSPR modeling, the reliability of datasets, the significance of molecular descriptor families and the diversity of chemometric tools used are important factors that define model predictability and interpretability. Likewise, for PBPK modeling the integrity of the pharmacokinetic data, the number of input parameters, the complexity of statistical analysis and the software packages used are relevant factors in bioavailability prediction. Although these approaches have been utilized independently, the tendency to use hybrid QSPR-PBPK approaches together with the exploration of ensemble and deep-learning systems for QSPR modeling of oral bioavailability has opened new avenues for development promising tools for oral bioavailability prediction.
Collapse
Affiliation(s)
- Miguel Ángel Cabrera-Pérez
- a Unit of Modeling and Experimental Biopharmaceutics , Chemical Bioactive Center, Central University of Las Villas , Santa Clara , Cuba.,b Department of Pharmacy and Pharmaceutical Technology , University of Valencia , Burjassot , Spain.,c Department of Engineering, Area of Pharmacy and Pharmaceutical Technology , Miguel Hernández University , Alicante , Spain
| | - Hai Pham-The
- d Department of Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| |
Collapse
|
25
|
Malikanti R, Vadija R, Veeravarapu H, Mustyala KK, Malkhed V, Vuruputuri U. Identification of small molecular ligands as potent inhibitors of fatty acid metabolism in Mycobacterium tuberculosis. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.08.090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
26
|
Patel RD, Kumar SP, Patel CN, Shankar SS, Pandya HA, Solanki HA. Parallel screening of drug-like natural compounds using Caco-2 cell permeability QSAR model with applicability domain, lipophilic ligand efficiency index and shape property: A case study of HIV-1 reverse transcriptase inhibitors. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Wong SSW, Rasid O, Laskaris P, Fekkar A, Cavaillon JM, Steinbach WJ, Ibrahim-Granet O. Treatment of Cyclosporin A retains host defense against invasive pulmonary aspergillosis in a non-immunosuppressive murine model by preserving the myeloid cell population. Virulence 2017; 8:1744-1752. [PMID: 28594271 DOI: 10.1080/21505594.2017.1339007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Cyclosporin A (CsA) is widely used as an immunosuppressive agent for organ transplant recipients. CsA inhibits calcineurin, which is highly conserved in mammals and fungi, and thus affects both types of organism. In mammals, the immunosuppressive effect of CsA is via hampering T cell activation. In fungi, the growth inhibitory effect of CsA is via interference with hyphal growth. The aim of this study was to determine whether CsA renders mice susceptible to invasive pulmonary aspergillosis (IPA) and whether it can protect immunosuppressed mice from infection. We therefore examined both the antifungal and the immunosuppressive activity of CsA in immunosuppressed and in immunocompetent mice infected with Aspergillus fumigatus to model IPA. We found that daily injections of CsA could not produce an antifungal effect sufficient to rescue immunosuppressed mice from lethal IPA. However, a 100% survival rate was obtained in non-immunosuppressed mice receiving daily CsA, indicating that CsA did not render the mice vulnerable to IPA. The lymphocyte subset was significantly suppressed by CsA, while the myeloid subset was not. Therefore, we speculate that CsA does not impair the host defense against IPA since the myeloid cells are preserved.
Collapse
Affiliation(s)
| | - Orhan Rasid
- b Unité Cytokines & Inflammation , Institut Pasteur , Paris , France
| | - Paris Laskaris
- b Unité Cytokines & Inflammation , Institut Pasteur , Paris , France
| | - Arnaud Fekkar
- c AP-HP , Groupe hospitalier La Pitié-Salpêtrière, Service de Parasitologie Mycologie , Paris , France.,d Centre d'Immunologie et des Maladies Infectieuses , CIMI-Paris , Paris , France.,e Sorbonne Universités , UPMC Univ Paris 06 , Paris , France
| | | | - William J Steinbach
- f Department of Pediatrics , Division of Pediatric Infectious Diseases, Duke University , NC , USA
| | | |
Collapse
|
28
|
High yield synthesis of cyclic analogues of antibacterial peptides P-113 by Sortase A-mediated ligation and their conformation studies. CHINESE CHEM LETT 2017. [DOI: 10.1016/j.cclet.2016.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Liu C, Le Blanc JCY, Schneider BB, Shields J, Federico JJ, Zhang H, Stroh JG, Kauffman GW, Kung DW, Ieritano C, Shepherdson E, Verbuyst M, Melo L, Hasan M, Naser D, Janiszewski JS, Hopkins WS, Campbell JL. Assessing Physicochemical Properties of Drug Molecules via Microsolvation Measurements with Differential Mobility Spectrometry. ACS CENTRAL SCIENCE 2017; 3:101-109. [PMID: 28280776 PMCID: PMC5324087 DOI: 10.1021/acscentsci.6b00297] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Indexed: 05/18/2023]
Abstract
The microsolvated state of a molecule, represented by its interactions with only a small number of solvent molecules, can play a key role in determining the observable bulk properties of the molecule. This is especially true in cases where strong local hydrogen bonding exists between the molecule and the solvent. One method that can probe the microsolvated states of charged molecules is differential mobility spectrometry (DMS), which rapidly interrogates an ion's transitions between a solvated and desolvated state in the gas phase (i.e., few solvent molecules present). However, can the results of DMS analyses of a class of molecules reveal information about the bulk physicochemical properties of those species? Our findings presented here show that DMS behaviors correlate strongly with the measured solution phase pKa and pKb values, and cell permeabilities of a set of structurally related drug molecules, even yielding high-resolution discrimination between isomeric forms of these drugs. This is due to DMS's ability to separate species based upon only subtle (yet predictable) changes in structure: the same subtle changes that can influence isomers' different bulk properties. Using 2-methylquinolin-8-ol as the core structure, we demonstrate how DMS shows promise for rapidly and sensitively probing the physicochemical properties of molecules, with particular attention paid to drug candidates at the early stage of drug development. This study serves as a foundation upon which future drug molecules of different structural classes could be examined.
Collapse
Affiliation(s)
- Chang Liu
- SCIEX, 71 Four Valley Drive, Concord, Ontario, L4K 4V8, Canada
| | | | | | - Jefry Shields
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - James J. Federico
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Hui Zhang
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Justin G. Stroh
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory W. Kauffman
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Daniel W. Kung
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Christian Ieritano
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Evan Shepherdson
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Mitch Verbuyst
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Luke Melo
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Moaraj Hasan
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Dalia Naser
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - John S. Janiszewski
- Pfizer
Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
- E-mail:
| | - W. Scott Hopkins
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
- E-mail:
| | - J. Larry Campbell
- SCIEX, 71 Four Valley Drive, Concord, Ontario, L4K 4V8, Canada
- Department
of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
- E-mail:
| |
Collapse
|
30
|
Cheng Li, Tongtong Liu, Xiaoming Cui, Uss AS, Cheng KC. Development of In Vitro Pharmacokinetic Screens Using Caco-2, Human Hepatocyte, and Caco-2/Human Hepatocyte Hybrid Systems for the Prediction of Oral Bioavailability in Humans. ACTA ACUST UNITED AC 2016; 12:1084-91. [DOI: 10.1177/1087057107308892] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, in vitro systems were used to build 2 pharmacokinetic models that predict human oral bioavailability: the Caco-2/hepatocyte combination model and the Caco-2/hepatocyte hybrid model. Data obtained in vitro on Caco-2 cell permeability and hepatocyte clearance are routinely used to predict the fraction of absorption after oral administration and the extent of first-pass metabolism, respectively. In the Caco-2/hepatocyte combination model, results from a Caco-2 cell permeability assay and a hepatocyte clearance assay were combined to project oral bioavailability. Comparison of oral bioavailabilities predicted by the combination model and reported oral bioavailabilities in humans for 30 marketed compounds resulted in a modest correlation ( r2 = 0.66). The Caco-2/hepatocyte hybrid model, as previously reported, joins the Caco-2 and hepatocyte clearance systems into 1 assay. Improvements to the previous model were made by incorporating an elimination phase into the Caco-2/hepatocyte hybrid model. In the new hybrid model, the compound was added to a Caco-2-containing donor compartment and allowed to permeate for 2 h to a hepatocyte-containing receiver compartment. Subsequently, to mimic an elimination phase, the donor compartment was removed, and permeated compound was incubated with hepatocytes alone for an additional 3 h. The area under the concentration versus time curve (AUC) was determined for each of the same 30 marketed compounds assessed by the combination model. A linear regression analysis comparing the in vitro AUCs and reported oral bioavailabilities in humans showed a reasonable correlation ( r 2 = 0.73). This study demonstrates that the Caco-2/hepatocyte hybrid model is more favorable and further proves the potential and feasibility of using in vitro screenings for the prediction of in vivo pharmacokinetics in humans. ( Journal of Biomolecular Screening 2007:1084-1091)
Collapse
Affiliation(s)
- Cheng Li
- Department of Exploratory Drug Metabolism, Schering-Plough Research Institute, Kenilworth, New Jersey
| | - Tongtong Liu
- Department of Exploratory Drug Metabolism, Schering-Plough Research Institute, Kenilworth, New Jersey
| | - Xiaoming Cui
- Department of Exploratory Drug Metabolism, Schering-Plough Research Institute, Kenilworth, New Jersey
| | - Annette S. Uss
- Department of Exploratory Drug Metabolism, Schering-Plough Research Institute, Kenilworth, New Jersey
| | - K.-C. Cheng
- Department of Exploratory Drug Metabolism, Schering-Plough Research Institute, Kenilworth, New Jersey,
| |
Collapse
|
31
|
Abstract
Understanding the concept of extrapolation of dose between species is important for pharmaceutical researchers when initiating new animal or human experiments. Interspecies allometric scaling for dose conversion from animal to human studies is one of the most controversial areas in clinical pharmacology. Allometric approach considers the differences in body surface area, which is associated with animal weight while extrapolating the doses of therapeutic agents among the species. This review provides basic information about translation of doses between species and estimation of starting dose for clinical trials using allometric scaling. The method of calculation of injection volume for parenteral formulation based on human equivalent dose is also briefed.
Collapse
Affiliation(s)
- Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Shery Jacob
- Department of Pharmaceutics, College of Pharmacy, Gulf Medical University, Ajman, UAE
| |
Collapse
|
32
|
New IVIVE method for the prediction of total human clearance and relative elimination pathway contributions from in vitro hepatocyte and microsome data. Eur J Pharm Sci 2016; 86:96-102. [PMID: 26948853 DOI: 10.1016/j.ejps.2016.02.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/08/2016] [Accepted: 02/29/2016] [Indexed: 11/20/2022]
Abstract
Total human clearance is a key determinant for the pharmacokinetic behavior of drug candidates. Our group recently introduced the Extended Clearance Model (ECM) as an accurate in vitro-in vivo extrapolation (IVIVE) method for the prediction of hepatic clearance. Yet, knowledge about relative elimination pathway contributions is needed in order to predict the total human clearance of drug candidates. In the present work, a training set of 18 drug compounds was used to describe the affiliations between in vitro sinusoidal uptake clearance and the fractional contributions of hepatic (metabolic and biliary) or renal clearance to overall in vivo elimination. By means of these quantitative relationships and using a validation set of 10 diverse drug molecules covering different (sub)classes of the Extended Clearance Concept Classification System (ECCCS), the relative contributions of elimination pathways were calculated and demonstrated to well correlate with human reference data. Likewise, ECM- and pathway-based predictions of total clearances from both data sets demonstrated a strong correlation with the observed clinical values with 26 out of 28 compounds within a three-fold deviation. Hence, total human clearance and relative contributions of elimination pathways were successfully predicted by the presented method using solely hepatocyte and microsome in vitro data.
Collapse
|
33
|
Lavanya P, Ramaiah S, Anbarasu A. Computational analysis reveal inhibitory action of nimbin against dengue viral envelope protein. Virusdisease 2015; 26:243-54. [PMID: 26645034 DOI: 10.1007/s13337-015-0280-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 09/16/2015] [Indexed: 11/29/2022] Open
Abstract
Dengue has emerged to be global health problem worldwide. Hence there is an immediate need to adopt new strategies in the development of effective anti-dengue drugs. Extracts from the leaves of Azadirachta indica has been traditionally used in folk medicine for viral infections. In the present study we report the anti-viral potency of nimbin, the active compound from the neem leaf extract against the envelope protein of dengue virus. Progression of viral entry into the host cell is facilitated by the envelope protein of dengue virus, suggesting; it as an effective anti-viral target. Nimbin is found to be effective against the envelope protein of all four types of dengue virus (dengue 1-4), which is evident from our in silico analysis. Our findings suggest the clinical importance of nimbin, which can serve as effective lead compound for further analysis.
Collapse
Affiliation(s)
- P Lavanya
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu 632014 India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu 632014 India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu 632014 India
| |
Collapse
|
34
|
The identification of lobeglitazone metabolites in rat liver microsomes and the kinetics of the in vivo formation of the major metabolite M1 in rats. J Pharm Biomed Anal 2015; 115:375-82. [DOI: 10.1016/j.jpba.2015.07.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 07/19/2015] [Accepted: 07/26/2015] [Indexed: 11/22/2022]
|
35
|
Sahasrabudhe V, Zhu T, Vaz A, Tse S. Drug Metabolism and Drug Interactions: Potential Application to Antituberculosis Drugs. J Infect Dis 2015; 211 Suppl 3:S107-14. [DOI: 10.1093/infdis/jiv009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
36
|
Lee JH, Son JH, Chae YJ, Ahn SH, Lee W, Kim DD, Chung SJ. Gender differences in the hepatic elimination and pharmacokinetics of lobeglitazone in rats. Biopharm Drug Dispos 2015; 36:410-415. [DOI: 10.1002/bdd.1954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Jong-Hwa Lee
- College of Pharmacy; Seoul National University; 1 Gwanak-ro, Gwanak-ro, Gwanak-gu Seoul 151-742 Republic of Korea
- Korea Institute of Toxicology; 141 Gajeong-ro, Yuseong-gu Daejeon 305-343 Republic of Korea
| | - Jun-Hyeng Son
- College of Pharmacy; Seoul National University; 1 Gwanak-ro, Gwanak-ro, Gwanak-gu Seoul 151-742 Republic of Korea
| | - Yoon-Jee Chae
- Chong Kun Dang Research Institute; Dongbaekjukjeon-daero, Giheung-gu Yongin-si Gyeonggi-do 446-916 Republic of Korea
| | - Sung Hoon Ahn
- College of Pharmacy; Kangwon National University; 1 Kangwondaehak-ro Chuncheon Kangwon-do 200-701 Republic of Korea
| | - Wooin Lee
- College of Pharmacy; Seoul National University; 1 Gwanak-ro, Gwanak-ro, Gwanak-gu Seoul 151-742 Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy; Seoul National University; 1 Gwanak-ro, Gwanak-ro, Gwanak-gu Seoul 151-742 Republic of Korea
| | - Suk-Jae Chung
- College of Pharmacy; Seoul National University; 1 Gwanak-ro, Gwanak-ro, Gwanak-gu Seoul 151-742 Republic of Korea
| |
Collapse
|
37
|
Kleeb S, Pang L, Mayer K, Eris D, Sigl A, Preston RC, Zihlmann P, Sharpe T, Jakob RP, Abgottspon D, Hutter AS, Scharenberg M, Jiang X, Navarra G, Rabbani S, Smiesko M, Lüdin N, Bezençon J, Schwardt O, Maier T, Ernst B. FimH antagonists: bioisosteres to improve the in vitro and in vivo PK/PD profile. J Med Chem 2015; 58:2221-39. [PMID: 25666045 DOI: 10.1021/jm501524q] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Urinary tract infections (UTIs), predominantly caused by uropathogenic Escherichia coli (UPEC), belong to the most prevalent infectious diseases worldwide. The attachment of UPEC to host cells is mediated by FimH, a mannose-binding adhesin at the tip of bacterial type 1 pili. To date, UTIs are mainly treated with antibiotics, leading to the ubiquitous problem of increasing resistance against most of the currently available antimicrobials. Therefore, new treatment strategies are urgently needed. Here, we describe the development of an orally available FimH antagonist. Starting from the carboxylate substituted biphenyl α-d-mannoside 9, affinity and the relevant pharmacokinetic parameters (solubility, permeability, renal excretion) were substantially improved by a bioisosteric approach. With 3'-chloro-4'-(α-d-mannopyranosyloxy)biphenyl-4-carbonitrile (10j) a FimH antagonist with an optimal in vitro PK/PD profile was identified. Orally applied, 10j was effective in a mouse model of UTI by reducing the bacterial load in the bladder by about 1000-fold.
Collapse
Affiliation(s)
- Simon Kleeb
- Institute of Molecular Pharmacy, Pharmacenter, University of Basel , Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
In vitro metabolism, disposition, preclinical pharmacokinetics and prediction of human pharmacokinetics of DNDI-VL-2098, a potential oral treatment for Visceral Leishmaniasis. Eur J Pharm Sci 2014; 65:147-55. [DOI: 10.1016/j.ejps.2014.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 08/08/2014] [Accepted: 09/05/2014] [Indexed: 11/22/2022]
|
39
|
Peng Y, Yadava P, Heikkinen AT, Parrott N, Railkar A. Applications of a 7-day Caco-2 cell model in drug discovery and development. Eur J Pharm Sci 2014; 56:120-30. [DOI: 10.1016/j.ejps.2014.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 01/30/2014] [Accepted: 02/13/2014] [Indexed: 11/26/2022]
|
40
|
Novel 3-nitro-1H-1,2,4-triazole-based compounds as potential anti-Chagasic drugs: in vivo studies. Future Med Chem 2014; 5:1763-76. [PMID: 24144412 DOI: 10.4155/fmc.13.108] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chagas disease is caused by the parasite Trypanosoma cruzi, is endemic in Latin America and leads to an estimated 14,000 deaths per year and around 100 million people at risk of infection. Drugs currently used in the treatment of Chagas are old, partially effective and have numerous side effects. METHODOLOGY We have previously reported that 3-nitro-1H-1,2,4-triazole-based compounds demonstrate significant and selective activity against T. cruzi amastigotes in infected L6 cells via activation of a type I nitroreductase, specific to trypanosomatids. In the present work we evaluated in vivo 13 of these compounds based on their high in vitro potency against T. cruzi (IC50 < 1 µM) and selectivity (SI: toxicity to L6 cells/toxicity against T. cruzi amastigotes > 200). Representative compounds of different chemical classes were included. A fast luminescence assay with transgenic parasites that express luciferase, and live imaging techniques were used. A total of 11 out of 13 compounds demonstrated significant antichagasic activity when administered intraperitoneally for 5-10 days at relatively small doses. The best in vivo activity was demonstrated by amides and sulfonamide derivatives. ADMET studies were performed for specific compounds. CONCLUSION At least three compounds were identified as effective, non-toxic antichagasic agents suitable for further development.
Collapse
|
41
|
Foster JR, Lund G, Sapelnikova S, Tyrrell DL, Kneteman NM. Chimeric rodents with humanized liver: bridging the preclinical/clinical trial gap in ADME/toxicity studies. Xenobiotica 2013; 44:109-22. [DOI: 10.3109/00498254.2013.867553] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Kimber JA, Kazarian SG, Štěpánek F. DEM simulation of drug release from structurally heterogeneous swelling tablets. POWDER TECHNOL 2013. [DOI: 10.1016/j.powtec.2012.12.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Péry ARR, Brochot C, Zeman FA, Mombelli E, Desmots S, Pavan M, Fioravanzo E, Zaldívar JM. Prediction of dose-hepatotoxic response in humans based on toxicokinetic/toxicodynamic modeling with or without in vivo data: a case study with acetaminophen. Toxicol Lett 2013; 220:26-34. [PMID: 23566899 DOI: 10.1016/j.toxlet.2013.03.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/23/2013] [Accepted: 03/25/2013] [Indexed: 02/04/2023]
Abstract
In the present legislations, the use of methods alternative to animal testing is explicitly encouraged, to use animal testing only 'as a last resort' or to ban it. The use of alternative methods to replace kinetics or repeated dose in vivo tests is a challenging issue. We propose here a strategy based on in vitro tests and QSAR (Quantitative Structure Activity Relationship) models to calibrate a dose-response model predicting hepatotoxicity. The dose response consists in calibrating and coupling a PBPK (physiologically-based pharmacokinetic) model with a toxicodynamic model for cell viability. We applied our strategy to acetaminophen and compared three different ways to calibrate the PBPK model: only with in vitro and in silico methods, using rat data or using all available data including data on humans. Some estimates of kinetic parameters differed substantially among the three calibration processes, but, at the end, the three models were quite comparable in terms of liver toxicity predictions and close to the usual range of human overdose. For the model based on alternative methods, the good adequation with the two other models resulted from an overestimated renal elimination rate which compensated for the underestimation of the metabolism rate. Our study points out that toxicokinetics/toxicodynamics approaches, based on alternative methods and modelling only, can predict in vivo liver toxicity with accuracy comparable to in vivo methods.
Collapse
Affiliation(s)
- Alexandre R R Péry
- Unité « Modèles pour l'écotoxicologie et la toxicologie » (METO), INERIS, Parc Alata BP2, 60550 Verneuil-en-Halatte, France.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Li YF, Zhang XQ, Hu WY, Li Z, Liu PX, Zhang ZQ. Rapid screening of drug-protein binding using high-performance affinity chromatography with columns containing immobilized human serum albumin. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2013; 2013:439039. [PMID: 23607050 PMCID: PMC3625562 DOI: 10.1155/2013/439039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 02/28/2013] [Indexed: 06/02/2023]
Abstract
For drug candidates, a plasma protein binding (PPB) more than 90% is more meaningful and deserves further investigation in development. In the study, a high-performance liquid chromatography method employing column containing immobilized human serum albumin (HSA) to screen in vitro PPB of leading compounds was established and successfully applied to tested compounds. Good correlation (a coefficient correlation of 0.96) was attained between the reciprocal values (X) of experimentally obtained retention time of reference compounds eluted through HSA column and the reported PPB values (Y) with a correlation equation of Y = 92.03 - 97.01X. The method was successfully applied to six test compounds, and the result was confirmed by the conventional ultrafiltration technique, and both yielded equal results. However, due to the particular protein immobilized to column, the method cannot be applied for all compounds and should be exploited judiciously based on the value of the logarithmic measure of the acid dissociation constant (pKa) as per the requirement. If α1-acid glycoprotein and other plasma proteins could be immobilized like HSA with their actual ratio in plasma to column simultaneously, the result attained using immobilized column may be more accurate, and the method could be applied to more compounds without pKa limitation.
Collapse
Affiliation(s)
- Ying-Fei Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | | | | | | | | | | |
Collapse
|
45
|
Gheysens O, Akurathi V, Chekol R, Dresselaers T, Celen S, Koole M, Dauwe D, Cleynhens BJ, Claus P, Janssens S, Verbruggen AM, Nuyts J, Himmelreich U, Bormans GM. Preclinical evaluation of carbon-11 and fluorine-18 sulfonamide derivatives for in vivo radiolabeling of erythrocytes. EJNMMI Res 2013; 3:4. [PMID: 23316861 PMCID: PMC3561128 DOI: 10.1186/2191-219x-3-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/02/2013] [Indexed: 11/25/2022] Open
Abstract
UNLABELLED BACKGROUND To date, few PET tracers for in vivo labeling of red blood cells (RBCs) are available. In this study, we report the radiosynthesis and in vitro and in vivo evaluation of 11C and 18F sulfonamide derivatives targeting carbonic anhydrase II (CA II), a metallo-enzyme expressed in RBCs, as potential blood pool tracers. A proof-of-concept in vivo imaging study was performed to demonstrate the feasibility to assess cardiac function and volumes using electrocardiogram (ECG)-gated positron emission tomography (PET) acquisition in comparison with cine magnetic resonance imaging (cMRI) in rats and a pig model of myocardial infarction. METHODS The inhibition constants (Ki) of CA II were determined in vitro for the different compounds by assaying CA-catalyzed CO2 hydration activity. Binding to human RBCs was estimated after in vitro incubation of the compounds with whole blood. Biodistribution studies were performed to evaluate tracer kinetics in NMRI mice. ECG-gated PET acquisition was performed in Wistar rats at rest and during pharmacological stress by infusing dobutamine at 10 μg/kg/min and in a pig model of myocardial infarction. Left ventricular ejection fraction (LVEF) and volumes were compared with values from cMRI. RESULTS The Ki of the investigated compounds for human CA II was found to be in the range of 8 to 422 nM. The fraction of radioactivity associated with RBCs was found to be ≥90% at 10- and 60-min incubation of tracers with heparinized human blood at room temperature for all tracers studied. Biodistribution studies in mice indicated that 30% to 67% of the injected dose was retained in the blood pool at 60 min post injection. A rapid and sustained tracer uptake in the heart region with an average standardized uptake value of 2.5 was observed from micro-PET images. The LVEF values obtained after pharmacological stress in rats closely matched between the cMRI and micro-PET values, whereas at rest, a larger variation between LVEF values obtained by both techniques was observed. In the pig model, a good agreement was observed between PET and MRI for quantification of left ventricular volumes and ejection fraction. CONCLUSIONS The 11C and 18F sulfonamide derivatives can be used for efficient in vivo radiolabeling of RBCs, and proof-of-concept in vivo imaging studies have shown the feasibility and potential of these novel tracers to assess cardiac function.
Collapse
Affiliation(s)
- Olivier Gheysens
- Nuclear Medicine, University Hospital Leuven, Herestraat 49, Leuven BE-3000, Belgium
- Department of Imaging and Pathology, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Vamsidhar Akurathi
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| | - Rufael Chekol
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| | - Tom Dresselaers
- Biomedical NMR Unit, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Leuven, BE-3000, Belgium
| | - Sofie Celen
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| | - Michel Koole
- Nuclear Medicine, University Hospital Leuven, Herestraat 49, Leuven BE-3000, Belgium
- Department of Imaging and Pathology, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Dieter Dauwe
- Cardiovascular Diseases, University Hospital Leuven, Herestraat 49, Leuven, BE-3000, Belgium
- Division of Cardiology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Bernard J Cleynhens
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| | - Piet Claus
- Division of Imaging and Cardiovascular Dynamics, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Stefan Janssens
- Cardiovascular Diseases, University Hospital Leuven, Herestraat 49, Leuven, BE-3000, Belgium
- Division of Cardiology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Alfons M Verbruggen
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| | - Johan Nuyts
- Nuclear Medicine, University Hospital Leuven, Herestraat 49, Leuven BE-3000, Belgium
- Department of Imaging and Pathology, Katholieke Universiteit Leuven, Herestraat 49, Leuven, BE-3000, Belgium
| | - Uwe Himmelreich
- Biomedical NMR Unit, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Leuven, BE-3000, Belgium
| | - Guy M Bormans
- Laboratory of Radiopharmacy, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Box 821, Leuven, BE-3000, Belgium
| |
Collapse
|
46
|
Towards the virtual screening of BIK inhibitors with the homology-modeled protein structure. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0105-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
47
|
Sherer EC, Verras A, Madeira M, Hagmann WK, Sheridan RP, Roberts D, Bleasby K, Cornell WD. QSAR Prediction of Passive Permeability in the LLC-PK1 Cell Line: Trends in Molecular Properties and Cross-Prediction of Caco-2 Permeabilities. Mol Inform 2012; 31:231-45. [DOI: 10.1002/minf.201100157] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/06/2012] [Indexed: 01/16/2023]
|
48
|
A ligand-based approach for enhancing the pharmacokinetic profile of highly charged antibacterial agents. Med Chem Res 2012. [DOI: 10.1007/s00044-010-9538-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
49
|
Padovan J, Ralić J, Letfus V, Milić A, Bencetić Mihaljević V. Investigating the barriers to bioavailability of macrolide antibiotics in the rat. Eur J Drug Metab Pharmacokinet 2011; 37:163-71. [DOI: 10.1007/s13318-011-0074-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/09/2011] [Indexed: 11/30/2022]
|
50
|
Xiao X, Wu J, Trigili C, Chen H, Chu JW, Zhao Y, Lu P, Sheng L, Li Y, Sharom FJ, Barasoain I, Diaz JF, Fang WS. Effects of C7 substitutions in a high affinity microtubule-binding taxane on antitumor activity and drug transport. Bioorg Med Chem Lett 2011; 21:4852-6. [DOI: 10.1016/j.bmcl.2011.06.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 06/08/2011] [Accepted: 06/10/2011] [Indexed: 10/18/2022]
|