1
|
Peng Y, Zhang AH, Wei L, Welsh WJ. Preclinical Evaluation of Sigma 1 Receptor Antagonists as a Novel Treatment for Painful Diabetic Neuropathy. ACS Pharmacol Transl Sci 2024; 7:2358-2368. [PMID: 39144554 PMCID: PMC11320727 DOI: 10.1021/acsptsci.4c00186] [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: 04/01/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
The global prevalence of diabetes is steadily rising, with an estimated 537 million adults affected by diabetes in 2021, projected to reach 783 million by 2045. A severe consequence of diabetes is the development of painful diabetic neuropathy (PDN), afflicting approximately one in every three diabetic patients and significantly compromising their quality of life. Current pharmacotherapies for PDN provide inadequate pain relief for many patients, underscoring the need for novel treatments that are both safe and effective. The Sigma 1 Receptor (S1R) is a ligand-operated chaperone protein that resides at the mitochondria-associated membrane of the endoplasmic reticulum. The S1R has been shown to play crucial roles in regulating cellular processes implicated in pain modulation. This study explores the potential of PW507, a novel S1R antagonist, as a therapeutic candidate for PDN. PW507 exhibited promising in vitro and in vivo properties in terms of ADME, toxicity, pharmacokinetics, and safety. In preclinical rat models of Streptozotocin-induced diabetic neuropathy, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration, without inducing tolerance and visual evidence of toxicity. To the best of our knowledge, this is the first report to evaluate an S1R antagonist in STZ-induced diabetic rats following both acute and 2-week chronic administration, offering compelling preclinical evidence for the potential use of PW507 as a promising therapeutic option for PDN.
Collapse
Affiliation(s)
- Youyi Peng
- Biomedical
Informatics Shared Resource, Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903, United States
| | - Allen H. Zhang
- Department
of Biology, Emory College of Arts and Sciences, Atlanta, Georgia 30322, United States
| | - Liping Wei
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - William J. Welsh
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
2
|
Kashyap D, Koirala S, Saini V, Bagde PH, Samanta S, Kar P, Jha HC. Prediction of Rab5B inhibitors through integrative in silico techniques. Mol Divers 2024; 28:2547-2562. [PMID: 37505376 DOI: 10.1007/s11030-023-10693-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/03/2023] [Indexed: 07/29/2023]
Abstract
Rab5B is a small monomeric G protein that regulates early endocytosis and controls signaling pathways related to cell growth, survival, and apoptosis. Dysregulation of Rab5B protein expression has been linked to the development of several cancers such as leukemia, lymphoma, kidney, prostate, ovarian, breast cancer, etc. Our research shows the first attempt to identify inhibitors that can target Rab5B GTPase. In this study, we performed molecular docking using Autodock Vina 1.5.6 and identified eight molecules with docking scores ranging from -9.8 to -10.6 kcal/mol. Thereafter, we examined the pharmacological characteristics of these compounds, and selected compounds were further analyzed for their conformational dynamics and thermodynamic stability using molecular dynamics simulations and molecular mechanics Poisson-Boltzmann surface area (MM-PBSA)-based free energy calculations. Notably, our findings revealed that strychnine had the highest binding affinity to Rab5B followed by anonaine, helioxanthin, and taiwanin E, with a ΔGbind value of -21.43, -17.11, -15.11, and -14.09 kcal/mol respectively. The binding free energy calculations showed that Van der Waals interactions are the primary contributor to the binding between Rab5B and the inhibitor. The interaction between the inhibitor and Rab5B was shown to be controlled by certain hot spot residues, including Phe45, Tyr48, Ala64, and Ala30. Overall, we believe that these findings could facilitate the exploration and development of potential hits against Rab5B, subject to optimization and further research.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Suman Koirala
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Pranit Hemant Bagde
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Sunanda Samanta
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India.
- Lab No. POD 1B 502, Computational Biophysics Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India.
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India.
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India.
| |
Collapse
|
3
|
Knippler CM, Arnst JL, Robinson IE, Matsuk V, Khatib TO, Harvey RD, Shanmugam M, Mouw JK, Fu H, Ganesh T, Marcus AI. Bisbiguanide analogs induce mitochondrial stress to inhibit lung cancer cell invasion. iScience 2024; 27:109591. [PMID: 38632988 PMCID: PMC11022046 DOI: 10.1016/j.isci.2024.109591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
Targeting cancer metabolism to limit cellular energy and metabolite production is an attractive therapeutic approach. Here, we developed analogs of the bisbiguanide, alexidine, to target lung cancer cell metabolism and assess a structure-activity relationship (SAR). The SAR led to the identification of two analogs, AX-4 and AX-7, that limit cell growth via G1/G0 cell-cycle arrest and are tolerated in vivo with favorable pharmacokinetics. Mechanistic evaluation revealed that AX-4 and AX-7 induce potent mitochondrial defects; mitochondrial cristae were deformed and the mitochondrial membrane potential was depolarized. Additionally, cell metabolism was rewired, as indicated by reduced oxygen consumption and mitochondrial ATP production, with an increase in extracellular lactate. Importantly, AX-4 and AX-7 impacted overall cell behavior, as these compounds reduced collective cell invasion. Taken together, our study establishes a class of bisbiguanides as effective mitochondria and cell invasion disrupters, and proposes bisbiguanides as promising approaches to limiting cancer metastasis.
Collapse
Affiliation(s)
- Christina M. Knippler
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Jamie L. Arnst
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA 30322, USA
| | - Isaac E. Robinson
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
| | - Veronika Matsuk
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, USA
| | - Tala O. Khatib
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | - R. Donald Harvey
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Janna K. Mouw
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Haian Fu
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Thota Ganesh
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Adam I. Marcus
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
4
|
Chen J, Yuan Z, Tu Y, Hu W, Xie C, Ye L. Experimental and computational models to investigate intestinal drug permeability and metabolism. Xenobiotica 2023; 53:25-45. [PMID: 36779684 DOI: 10.1080/00498254.2023.2180454] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Oral administration is the preferred route for drug administration that leads to better therapy compliance. The intestine plays a key role in the absorption and metabolism of oral drugs, therefore, new intestinal models are being continuously proposed, which contribute to the study of intestinal physiology, drug screening, drug side effects, and drug-drug interactions.Advances in pharmaceutical processes have produced more drug formulations, causing challenges for intestinal models. To adapt to the rapid evolution of pharmaceuticals, more intestinal models have been created. However, because of the complexity of the intestine, few models can take all aspects of the intestine into account, and some functions must be sacrificed to investigate other areas. Therefore, investigators need to choose appropriate models according to the experimental stage and other requirements to obtain the desired results.To help researchers achieve this goal, this review summarised the advantages and disadvantages of current commonly used intestinal models and discusses possible future directions, providing a better understanding of intestinal models.
Collapse
Affiliation(s)
- Jinyuan Chen
- Institute of Scientific Research, Southern Medical University, Guangzhou, P.R. China.,TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ziyun Yuan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Yifan Tu
- Boehringer-Ingelheim, Connecticut, P.R. USA
| | - Wanyu Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Cong Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ling Ye
- TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
5
|
Ali HSHM, Altayb HN, Firoz A, Bayoumi AAM, El Omri A, Chaieb K. Inhibitory activity of marine sponge metabolites on SARS-CoV-2 RNA dependent polymerase: virtual screening and molecular dynamics simulation. J Biomol Struct Dyn 2022; 40:10191-10202. [PMID: 34151745 DOI: 10.1080/07391102.2021.1940283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Marine species are known as rich sources of metabolites involved mainly in the pharmaceutical industry. This study aimed to evaluate the effect of biologically active compounds in the marine sponge on the SARS-CoV-2 RNA-dependent-RNA polymerase protein (RdRp) using the in-silico method. A total of 51 marine compounds were checked for their possible interaction with SARS-CoV-2 RdRp using Maestro interface for molecular docking, molecular dynamic (MD) simulation, and MM/GBSA method to estimate compounds binding affinities. Among the 51 compounds screened in this study, two (mycalamide A, and nakinadine B) exhibited the lowest docking energy and best interaction. Among these compounds, mycalamide A was identified as a potent inhibitor of SARS-CoV-2 RdRp that showed the best and stable interaction during molecular dynamic simulation, with residues (Asp760 and Asp761) found in the catalytic domain of RdRp. The analysis through MM/GBSA for molecular dynamic simulation results revealed binding energy -59.7 ± 7.18 for Mycalamide A and -56 ± 10.55 for Nakinadine B. These results elucidate the possible use of mycalamide A for treating coronavirus disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hani S H Mohammed Ali
- Faculty of Science, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hisham N Altayb
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Firoz
- Faculty of Science, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Abdelfatteh El Omri
- Center of Excellence in Bio-nanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Science, Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kamel Chaieb
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Laboratory of Analysis, Treatment, and valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, Monastir University, Monastir, Tunisia
| |
Collapse
|
6
|
Pagare P, Obeng S, Huang B, Marcus MM, Nicholson KL, Townsend AE, Banks ML, Zhang Y. Preclinical Characterization and Development on NAQ as a Mu Opioid Receptor Partial Agonist for Opioid Use Disorder Treatment. ACS Pharmacol Transl Sci 2022; 5:1197-1209. [PMID: 36407950 PMCID: PMC9667545 DOI: 10.1021/acsptsci.2c00178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Indexed: 11/06/2022]
Abstract
Mu opioid receptor (MOR) selective antagonists and partial agonists have clinical utility for the treatment of opioid use disorders (OUDs). However, the development of many has suffered due to their poor pharmacokinetic properties and/or rapid metabolism. Our recent efforts to identify MOR modulators have provided 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ), a low-efficacy partial agonist, that showed sub-nanomolar binding affinity to the MOR (K i 0.6 nM) with selectivity over the delta opioid receptor (δ/μ 241) and the kappa opioid receptor (κ/μ 48). Its potent inhibition of the analgesic effect of morphine (AD50 0.46 mg/kg) and precipitation of significantly less withdrawal symptoms even at 100-fold greater dose than naloxone represents a promising molecule for further development as a novel OUD therapeutic agent. Therefore, further in vitro and in vivo characterization of its pharmacokinetics and pharmacodynamics properties was conducted to fully understand its pharmaceutical profile. NAQ showed favorable in vitro ADMET properties and no off-target binding to several classes of GPCRs, enzymes, and ion channels. Following intravenous administration, 1 mg/kg dose of NAQ showed a similar in vivo pharmacokinetic profile to naloxone; however, orally administered 10 mg/kg NAQ demonstrated significantly improved oral bioavailability over both naloxone and naltrexone. Abuse liability assessment of NAQ in rats demonstrated that NAQ functioned as a less potent reinforcer than heroin. Chronic 5 day NAQ pretreatment decreased heroin self-administration in a heroin-vs-food choice procedure similar to the clinically used MOR partial agonist buprenorphine. Taken together, these studies provide evidence supporting NAQ as a promising lead to develop novel OUD therapeutics.
Collapse
Affiliation(s)
- Piyusha
P. Pagare
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Samuel Obeng
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Boshi Huang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Madison M. Marcus
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Katherine L. Nicholson
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Andrew E. Townsend
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Matthew L. Banks
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Yan Zhang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| |
Collapse
|
7
|
Sk MF, Kar P. Finding inhibitors and deciphering inhibitor-induced conformational plasticity in the Janus kinase via multiscale simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2022; 33:833-859. [PMID: 36398489 DOI: 10.1080/1062936x.2022.2145352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
The Janus kinase (JAK) is a master regulator of the JAK/STAT pathway. Dysregulation of this signalling cascade causes neuroinflammation and autoimmune disorders. Therefore, JAKs have been characterized as an attractive target for developing anti-inflammatory drugs. Nowadays, designing efficient, effective, and specific targeted therapeutics without being cytotoxic has gained interest. We performed the virtual screening of natural products in combination with pharmacological analyses. Subsequently, we performed molecular dynamics simulations to study the stability of the ligand-bound complexes and ligand-induced inactive conformations. Notably, inactive kinases display remarkable conformational plasticity; however, ligand-induced molecular mechanisms of these conformations are still poorly understood. Herein, we performed a free energy landscape analysis to explore the conformational plasticity of the JAK1 kinase. Leonurine, STOCK1N-68642, STOCK1N-82656, and STOCK1N-85809 bound JAK1 exhibited a smooth transition from an active (αC-in) to a completely inactive conformation (αC-out). Ligand binding induces disorders in the αC-helix. Molecular mechanics Poisson Boltzmann surface area (MM/PBSA) calculation suggested three phytochemicals, namely STOCK1N-68642, Epicatechin, and STOCK1N-98615, have higher binding affinity compared to other ligand molecules. The ligand-induced conformational plasticity revealed by our simulations differs significantly from the available crystal structures, which might help in designing allosteric drugs.
Collapse
Affiliation(s)
- M F Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, India
| | - P Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, India
| |
Collapse
|
8
|
Davie RL, Edwards HJ, Evans DM, Hodgson ST, Stocks MJ, Smith AJ, Rushbrooke LJ, Pethen SJ, Roe MB, Clark DE, McEwan PA, Hampton SL. Sebetralstat (KVD900): A Potent and Selective Small Molecule Plasma Kallikrein Inhibitor Featuring a Novel P1 Group as a Potential Oral On-Demand Treatment for Hereditary Angioedema. J Med Chem 2022; 65:13629-13644. [PMID: 36251573 PMCID: PMC9620001 DOI: 10.1021/acs.jmedchem.2c00921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Indexed: 11/29/2022]
Abstract
Hereditary angioedema (HAE) is a rare genetic disorder in which patients experience sudden onset of swelling in various locations of the body. HAE is associated with uncontrolled plasma kallikrein (PKa) enzyme activity and generation of the potent inflammatory mediator, bradykinin, resulting in episodic attacks of angioedema. Herein, we disclose the discovery and optimization of novel small molecule PKa inhibitors. Starting from molecules containing highly basic P1 groups, which typically bind to an aspartic acid residue (Asp189) in the serine protease S1 pocket, we identified novel P1 binding groups likely to have greater potential for oral-drug-like properties. The optimization of P4 and the central core together with the particularly favorable properties of 3-fluoro-4-methoxypyridine P1 led to the development of sebetralstat, a potent, selective, orally bioavailable PKa inhibitor in phase 3 for on-demand treatment of HAE attacks.
Collapse
Affiliation(s)
- Rebecca L. Davie
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - Hannah J. Edwards
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - D. Michael Evans
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - Simon T. Hodgson
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - Michael J. Stocks
- School
of Pharmacy, University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K.
| | - Alun J. Smith
- Sygnature
Discovery, Biocity, Pennyfoot Street, Nottingham, NG1 1GR, U.K.
| | | | - Stephen J. Pethen
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - Michael B. Roe
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| | - David E. Clark
- Charles
River Early Discovery, 6-9 Spire Green Centre, Harlow, Essex CM19 5TR, U.K.
| | - Paul A. McEwan
- Evotec, 114 Innovation Drive Milton Science
Park, Abingdon, OX14 4RZ, U.K.
| | - Sally L. Hampton
- KalVista
Pharmaceuticals Limited, Porton Science Park, Salisbury, SP4 0BF, U.K.
| |
Collapse
|
9
|
Mazumder A, Dobyns BM, Howard MP, Beckingham BS. Theoretical and Experimental Considerations for Investigating Multicomponent Diffusion in Hydrated, Dense Polymer Membranes. MEMBRANES 2022; 12:942. [PMID: 36295701 PMCID: PMC9610993 DOI: 10.3390/membranes12100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
In many applications of hydrated, dense polymer membranes-including fuel cells, desalination, molecular separations, electrolyzers, and solar fuels devices-the membrane is challenged with aqueous streams that contain multiple solutes. The presence of multiple solutes presents a complex process because each solute can have different interactions with the polymer membrane and with other solutes, which collectively determine the transport behavior and separation performance that is observed. It is critical to understand the theoretical framework behind and experimental considerations for understanding how the presence of multiple solutes impacts diffusion, and thereby, the design of membranes. Here, we review models for multicomponent diffusion in the context of the solution-diffusion framework and the associated experiments for characterizing multicomponent transport using diffusion cells. Notably, multicomponent effects are typically not considered when discussing or investigating transport in dense, hydrated polymer membranes, however recent research has shown that these effects can be large and important for understanding the transport behavior.
Collapse
Affiliation(s)
- Antara Mazumder
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Breanna M. Dobyns
- Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Michael P. Howard
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Bryan S. Beckingham
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
10
|
Cai X, Patel S, Huang C, Paiva A, Sun Y, Barker G, Weller H, Shou W. Comprehensive characterization and optimization of Caco-2 cells enabled the development of a miniaturized 96-well permeability assay. Xenobiotica 2022; 52:742-750. [PMID: 36217915 DOI: 10.1080/00498254.2022.2133648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Assessment of compound permeability through a Caco-2 cell monolayer is a well-accepted model to evaluate its in-vivo permeability potential and transporter interaction. While this assay has commonly been conducted using a 24-well assay plate format, a miniaturised 96-well assay format is highly desirable to achieve greater capacity and higher efficiency.Previous attempts to convert this assay from 24-well to 96-well format at our lab, however, had met with varied efflux capacities and unacceptable efflux ratios for digoxin, a substrate of P-glycoprotein (Pgp), which indicated inadequate Pgp transporter expression in the 96-well format.These challenges in converting the assays were attributed to the heterogeneous and unstable nature of the Caco-2 cells. To overcome the challenges, single-cell sorting of Caco-2 cells was conducted by flow cytometry to obtain a more homogeneous and stable cell population. The sorted cells were then seeded to 96-well transwell plates and the Pgp expression under various cell culture conditions was monitored by a LC-MS/MS-based targeted proteomics method.Through cell sorting and direct Pgp expression measurement, Caco-2 cells with adequate and sustained Pgp expression in a 96-well format were obtained, which led to the successful development and implementation of a 96-well Caco-2 assay with significant efficiency gain and faster turnaround time than the historical 24-well assay.
Collapse
Affiliation(s)
- Xianmei Cai
- Discovery Chemistry, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | - Shivani Patel
- Discovery Chemistry, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | | | - Anthony Paiva
- Discovery Chemistry, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | - Yongnian Sun
- Lead Discovery and Optimization, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | - Gregory Barker
- Lead Discovery and Optimization, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | - Harold Weller
- Discovery Chemistry, Bristol Myers Squibb. Co, Princeton, NJ, USA
| | - Wilson Shou
- Discovery Chemistry, Bristol Myers Squibb. Co, Princeton, NJ, USA
| |
Collapse
|
11
|
Leśniak RK, Nichols RJ, Schonemann M, Zhao J, Gajera CR, Lam G, Nguyen KC, Langston JW, Smith M, Montine TJ. Discovery of 1 H-Pyrazole Biaryl Sulfonamides as Novel G2019S-LRRK2 Kinase Inhibitors. ACS Med Chem Lett 2022; 13:981-988. [DOI: 10.1021/acsmedchemlett.2c00116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Robert K. Leśniak
- Medicinal Chemistry Knowledge Center, Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - R. Jeremy Nichols
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Marcus Schonemann
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Jing Zhao
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Chandresh R. Gajera
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Grace Lam
- Departments of Medicine and Microbiology and Immunology, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Khanh C. Nguyen
- Departments of Medicine and Microbiology and Immunology, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - J. William Langston
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
- Department of Neurology and Neuroscience, Stanford University, 300 Pasteur Drive, Stanford, California 94304, United States
| | - Mark Smith
- Medicinal Chemistry Knowledge Center, Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Thomas J. Montine
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| |
Collapse
|
12
|
Huđek Turković A, Gunjača M, Marjanović M, Lovrić M, Butorac A, Rašić D, Peraica M, Vujčić Bok V, Šola I, Rusak G, Durgo K. Proteome changes in human bladder T24 cells induced by hydroquinone derived from Arctostaphylos uva-ursi herbal preparation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115092. [PMID: 35143933 DOI: 10.1016/j.jep.2022.115092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Arctostaphylos uva-ursi (L.) Spreng. (bearberry) is a well-known traditional herbal plant used as a urinary tract disinfectant. Its antiseptic and diuretic properties can be attributed to hydroquinone, obtained by hydrolysis of arbutin. AIM OF THE STUDY This study aimed to determine the toxic profile of free hydroquinone on urinary bladder cells (T24) as a target of therapeutic action. MATERIALS AND METHODS Quantitative and qualitative analysis of the extract and the digestive stability and bioavailability of arbutin and hydroquinone were performed by HPLC assay and simulated in vitro digestion, respectively. Cytotoxic effect, reactive oxygen species induction and proteome changes in T24 cells after hydroquinone treatment were determined using Neutral red assay, 2',7'-dichlorofluorescein-diacetate (DCFH-DA) assay and mass spectrometry, respectively. RESULTS Through in vitro digestion, arbutin was stable, but hydroquinone increased after pepsin treatment (109.6%) and then decreased after the small intestine phase (65.38%). The recommended doses of Uva-ursi had a cytotoxic effect on T24 cells only when all hydroquinone conjugates were converted to free hydroquinone (320 and 900 μg/mL) and the toxic effect was enhanced by recovery. One cup of the therapeutic dose had a prooxidative effect after 4 h of incubation. Shorter time of cell exposure (2 h) to hydroquinone did not have any impact on reactive oxygen species induction. Proteomic analysis found 17 significantly up-regulated proteins compared to control. Hydroquinone activated proteins related to oxidative stress response, stress-adaptive signalling, heat shock response and initiation of translation. CONCLUSIONS Despite the therapeutic properties of bearberry, up-regulated T24 cell proteins are evidence that plant compounds, although from a natural source, may exhibit negative properties.
Collapse
Affiliation(s)
- Ana Huđek Turković
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000, Zagreb, Croatia.
| | - Marija Gunjača
- BICRO BIOCentre, Ltd., Central Laboratory, Borongajska cesta 83H, 10000, Zagreb, Croatia.
| | - Marko Marjanović
- BICRO BIOCentre, Ltd., Central Laboratory, Borongajska cesta 83H, 10000, Zagreb, Croatia.
| | - Marija Lovrić
- BICRO BIOCentre, Ltd., Central Laboratory, Borongajska cesta 83H, 10000, Zagreb, Croatia.
| | - Ana Butorac
- BICRO BIOCentre, Ltd., Central Laboratory, Borongajska cesta 83H, 10000, Zagreb, Croatia.
| | - Dubravka Rašić
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia.
| | - Maja Peraica
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia.
| | - Valerija Vujčić Bok
- Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia.
| | - Ivana Šola
- Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia.
| | - Gordana Rusak
- Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia.
| | - Ksenija Durgo
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000, Zagreb, Croatia.
| |
Collapse
|
13
|
Unadkat V, Rohit S, Parikh P, Patel K, Sanna V, Singh S. Identification of 1,2,4-Oxadiazoles-Based Novel EGFR Inhibitors: Molecular Dynamics Simulation-Guided Identification and in vitro ADME Studies. Onco Targets Ther 2022; 15:479-495. [PMID: 35535170 PMCID: PMC9077134 DOI: 10.2147/ott.s357765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022] Open
Abstract
Background In this work, we have identified heterocyclic derivatives with 1,2,4 oxadiazole scaffold mimicking the functions of tyrosine kinase inhibitors. Fourteen molecules that displayed the best fit were picked from the library of compounds and studied under in-silico and in-vitro conditions. Four compounds were selected for further cytotoxicity and ADME (Absorption, Distribution, Metabolism, Elimination) profiling showing IC50 (from 8–13 µM) values against EGFR positive cancer cell line (MCF7). Methods A molecular dynamics simulation study was performed to understand the correlation of non-covalent binding energies with biological activity. The drug-like properties of the selected four compounds (7a, 7b, 7e, and 7m) were evaluated by in-vitro ADME studies. Compounds 7a, 7b, and 7m were the active compounds in the molecular dynamics simulations study. Further, EGFR binding activity was confirmed with EGFRWT and EGFRT790M kinase assay using a luminescence-based method. Results These compounds (7a, 7b, and 7m) showed activity against EGFRWT and mutant EGFRT790M, exhibiting IC50 values of <10 and <50 micromolar, respectively. These compounds also possess moderate aqueous solubility in 40–70 µg/mL at pH 7.4 and 30–100 µg/mL at pH 4.0. Further, 7a, 7b, and 7m showed balanced lipophilicity with Log D values ranging from 1–3. They demonstrated a good correlation in Caco-2 permeability with Apparent permeability (Papp) 1 to 5 × 10−6 cm/s in comparison with 7e, which was found to be highly lipophilic (Log D >5) and showed high permeability (Papp 17 × 10−6 cm/s). Lastly, all these compounds were moderately stable in liver microsomes at alkaline pH with a half-life of 30–60 min, while at a highly acidic pH (2.0), the compounds were stable up to 15–20 min. Conclusion Overall, in-vitro ADME results of these molecules showed good drug-like properties, which are well correlated with the in-silico ADME data, making them ideal for developing an oral drug delivery formulation.
Collapse
Affiliation(s)
- Vishal Unadkat
- Kashiv Biosciences Pvt Ltd, Ahmedabad, 382210, Gujarat, India
- Division of Biological & Life Sciences (Formerly Institute of Life Sciences), School of Arts & Sciences, Ahmedabad University, Ahmedabad, 380009, Gujarat, India
| | - Shishir Rohit
- Kashiv Biosciences Pvt Ltd, Ahmedabad, 382210, Gujarat, India
| | - Paranjay Parikh
- Department of Advanced Organic Chemistry, P.D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Gujarat, 388421, India
| | - Kaushal Patel
- Department of Advanced Organic Chemistry, P.D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Gujarat, 388421, India
| | - Vinod Sanna
- Piramal Pharma Solutions, Ahmedabad, 382213, Gujarat, India
| | - Sanjay Singh
- Division of Biological & Life Sciences (Formerly Institute of Life Sciences), School of Arts & Sciences, Ahmedabad University, Ahmedabad, 380009, Gujarat, India
- National Institute of Animal Biotechnology, Hyderabad, 500032, Telangana, India
- Correspondence: Sanjay Singh, Division of Biological & Life Sciences (Formerly Institute of Life Sciences), School of Arts & Sciences, Ahmedabad University, Navaragnpura, Ahmedabad, 380009, Gujarat, India, Email
| |
Collapse
|
14
|
Basu S, Middya S, Banerjee M, Ghosh R, Pryde DC, Yadav DB, Shrivastava R, Surya A. The discovery of potent small molecule cyclic urea activators of STING. Eur J Med Chem 2022; 229:114087. [PMID: 34998056 DOI: 10.1016/j.ejmech.2021.114087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/25/2021] [Accepted: 12/26/2021] [Indexed: 01/08/2023]
Abstract
STING mediates innate immune responses that are triggered by the presence of cytosolic DNA. Activation of STING to boost antigen recognition is a therapeutic modality that is currently being tested in cancer patients using nucleic-acid based macrocyclic STING ligands. We describe here the discovery of 3,4-dihydroquinazolin-2(1H)-one based 6,6-bicyclic heterocyclic agonists of human STING that activate all known human variants of STING with high potency.
Collapse
Affiliation(s)
- Sourav Basu
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - Sandip Middya
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - Monali Banerjee
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - Rajib Ghosh
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - David C Pryde
- Curadev Pharma Ltd., Innovation House, Discovery Park, Ramsgate Road, Sandwich, Kent, CT13 9ND, UK
| | - Dharmendra B Yadav
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - Ritesh Shrivastava
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India
| | - Arjun Surya
- Curadev Pharma Pvt. Ltd., B-87, Sector 83, Noida, 201305, Uttar Pradesh, India.
| |
Collapse
|
15
|
Gomes TL, de Oliveira-Marques V, Hampson RJ, Jacinto A, de Moraes LV, Martinho RG. theLiTE™: A Screening Platform to Identify Compounds that Reinforce Tight Junctions. Front Pharmacol 2022; 12:752787. [PMID: 35069190 PMCID: PMC8771259 DOI: 10.3389/fphar.2021.752787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/24/2021] [Indexed: 11/13/2022] Open
Abstract
Tight junctions (TJ) are formed by transmembrane and intracellular proteins that seal the intercellular space and control selective permeability of epithelia. Integrity of the epithelial barrier is central to tissue homeostasis and barrier dysfunction has been linked to many pathological conditions. TJ support the maintenance of cell polarity through interactions with the Par complex (Cdc42-Par-6-Par-3-aPKC) in which Par-6 is an adaptor and links the proteins of the complex together. Studies have shown that Par-6 overexpression delays the assembly of TJ proteins suggesting that Par-6 negatively regulates TJ assembly. Because restoring barrier integrity is of key therapeutic and prophylactic value, we focus on finding compounds that have epithelial barrier reinforcement properties; we developed a screening platform (theLiTE™) to identify compounds that modulate Par-6 expression in follicular epithelial cells from Par-6-GFP Drosophila melanogaster egg chambers. Hits identified were then tested whether they improve epithelial barrier function, using measurements of transepithelial electrical resistance (TEER) or dye efflux to evaluate paracellular permeability. We tested 2,400 compounds, found in total 10 hits. Here we present data on six of them: the first four hits allowed us to sequentially build confidence in theLiTE™ and two compounds that were shortlisted for further development (myricetin and quercetin). We selected quercetin due to its clinical and scientific validation as a compound that regulates TJ; food supplement formulated on the basis of this discovery is currently undergoing clinical evaluation in gastroesophageal reflux disease (GERD) sufferers.
Collapse
Affiliation(s)
- Teresa Lopes Gomes
- Thelial Technologies SA, Parque Tecnológico de Cantanhede, Cantanhede, Portugal
| | | | - Richard John Hampson
- Thelial Technologies SA, Parque Tecnológico de Cantanhede, Cantanhede, Portugal
- Thelial BV (Epinutra), Wageningen, Netherlands
| | - António Jacinto
- iNOVA4Health, Chronic Diseases Research Centre - CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Luciana Vieira de Moraes
- Thelial Technologies SA, Parque Tecnológico de Cantanhede, Cantanhede, Portugal
- Thelial BV (Epinutra), Wageningen, Netherlands
| | - Rui Gonçalo Martinho
- Thelial Technologies SA, Parque Tecnológico de Cantanhede, Cantanhede, Portugal
- Departamento de Ciências Biomédicas e Medicina, and Center for Biomedical Research, Universidade do Algarve, Faro, Portugal
- Department of Medical Sciences and Institute for Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| |
Collapse
|
16
|
Pechenov S, Revell J, Will S, Naylor J, Tyagi P, Patel C, Liang L, Tseng L, Huang Y, Rosenbaum AI, Balic K, Konkar A, Grimsby J, Subramony JA. Development of an orally delivered GLP-1 receptor agonist through peptide engineering and drug delivery to treat chronic disease. Sci Rep 2021; 11:22521. [PMID: 34795324 PMCID: PMC8602401 DOI: 10.1038/s41598-021-01750-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/01/2021] [Indexed: 01/13/2023] Open
Abstract
Peptide therapeutics are increasingly used in the treatment of disease, but their administration by injection reduces patient compliance and convenience, especially for chronic diseases. Thus, oral administration of a peptide therapeutic represents a significant advance in medicine, but is challenged by gastrointestinal instability and ineffective uptake into the circulation. Here, we have used glucagon-like peptide-1 (GLP-1) as a model peptide therapeutic for treating obesity-linked type 2 diabetes, a common chronic disease. We describe a comprehensive multidisciplinary approach leading to the development of MEDI7219, a GLP-1 receptor agonist (GLP-1RA) specifically engineered for oral delivery. Sites of protease/peptidase vulnerabilities in GLP-1 were removed by amino acid substitution and the peptide backbone was bis-lipidated to promote MEDI7219 reversible plasma protein binding without affecting potency. A combination of sodium chenodeoxycholate and propyl gallate was used to enhance bioavailability of MEDI7219 at the site of maximal gastrointestinal absorption, targeted by enteric-coated tablets. This synergistic approach resulted in MEDI7219 bioavailability of ~ 6% in dogs receiving oral tablets. In a dog model of obesity and insulin resistance, MEDI7219 oral tablets significantly decreased food intake, body weight and glucose excursions, validating the approach. This novel approach to the development of MEDI7219 provides a template for the development of other oral peptide therapeutics.
Collapse
Affiliation(s)
- Sergei Pechenov
- Drug Delivery, Dosage Form Design and Development, AstraZeneca, Gaithersburg, MD, USA
| | | | - Sarah Will
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jacqueline Naylor
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Puneet Tyagi
- Drug Delivery, Dosage Form Design and Development, AstraZeneca, Gaithersburg, MD, USA
| | - Chandresh Patel
- Drug Delivery, Dosage Form Design and Development, AstraZeneca, Gaithersburg, MD, USA
| | - Lihuan Liang
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Leo Tseng
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Yue Huang
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Kemal Balic
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Anish Konkar
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Joseph Grimsby
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | |
Collapse
|
17
|
Zhao Y, Lou J, Zhang H, Sun H, Zhang M, Wang S, Sha X, Zhan Z, Wang Y, Ma C, Li WJ. Measurement methods of single cell drug response. Talanta 2021; 239:123035. [PMID: 34839926 DOI: 10.1016/j.talanta.2021.123035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 11/25/2022]
Abstract
In the last decades, a wide multitude of research activity has been focused on the development of new drugs, and devoted to overcome the challenges of high cost and low efficiency in drug evaluation. The measurement of drug response at the single cell level is a quicker, more direct and more accurate way to reflect drug efficacy, which can shorten the drug development period and reduce research costs. Therefore, the single cell drug response (SCDR) measurement technology has aroused extensive attention from researchers, and has become a hot topic in the fields of drug research and cell biology. Recent years have seen the emergence of various SCDR measurement technologies that feature different working principles and different levels of measurement performance. To better examine, compare and summarize the characteristics and functions of these technologies, we select signal-to-noise ratio, throughput, content, invasion, and device complexity as the criteria to evaluate them from the drug efficacy perspective. This review aims to highlight sixteen kinds of SCDR measurement technologies, including patch-clamp technique, live-cell interferometry, capillary electrophoresis, secondary ion mass spectrometry, and more, and report widespread representative examples of SCDR measurement the recent approaches for over the past forty years. Based on their reaction principles, these technologies are classified into four categories: electrical, optical, electrochemical, and mass spectrometry, and a detailed comparison is made between them. After in-depth understanding of these technologies, it is expected to improve or integrate these technologies to propose better SCDR measurement strategies, and explore methods in new drug development and screening, as well as disease diagnosis and treatment.
Collapse
Affiliation(s)
- Yuliang Zhao
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Jiazhi Lou
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Hongyu Zhang
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Hui Sun
- Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Menglin Zhang
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Shuyu Wang
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Xiaopeng Sha
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, 066004, China
| | - Zhikun Zhan
- School of Electrical Engineering, Yanshan University at Qinhuangdao, Qinhuangdao, 066004, China.
| | - Ying Wang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Cuihua Ma
- Department of Clinical Laboratory, First Hospital of Qinhuangdao, Qinhuangdao, 066004, China.
| | - Wen Jung Li
- Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China.
| |
Collapse
|
18
|
Han A, Olsen O, D'Souza C, Shan J, Zhao F, Yanolatos J, Hovhannisyan Z, Haxhinasto S, Delfino F, Olson W. Development of Novel Glucocorticoids for Use in Antibody-Drug Conjugates for the Treatment of Inflammatory Diseases. J Med Chem 2021; 64:11958-11971. [PMID: 34378927 DOI: 10.1021/acs.jmedchem.1c00541] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glucocorticoids (GCs) are widely used to treat a variety of autoimmune and inflammatory diseases; however, systemic delivery of GCs is associated with side effects that affect essentially every organ system, reflecting the nearly ubiquitous expression of the glucocorticoid receptor (GR). Targeted delivery of GCs to diseased tissues using antibody-glucocorticoid conjugates (GC-ADCs) offers a therapeutic alternative to overcome these adverse effects. Herein, we describe novel classes of GCs that exhibited greater potency than dexamethasone and budesonide, a 100-fold selectivity toward the GR over other nuclear receptors, and no in vitro safety liability in pharmacology assays (hERG, AMES) and that demonstrated a substantial reduction in tumor necrosis factor-α (TNF-α) release in mice challenged with lipopolysaccharide (LPS). The site-specific conjugated GC-ADCs via cathepsin-cleavable linkers were highly stable in plasma and specifically released GCs in antigen-positive cells, suggesting that these novel GCs can serve as ADC payloads to treat autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Amy Han
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Olav Olsen
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | | | - Jing Shan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Feng Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Jean Yanolatos
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | | | - Sokol Haxhinasto
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Frank Delfino
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - William Olson
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| |
Collapse
|
19
|
Pokorný J, Olejníková D, Frydrych I, Lišková B, Gurská S, Benická S, Šarek J, Kotulová J, Hajdúch M, Džubák P, Urban M. Substituted dienes prepared from betulinic acid - Synthesis, cytotoxicity, mechanism of action, and pharmacological parameters. Eur J Med Chem 2021; 224:113706. [PMID: 34311159 DOI: 10.1016/j.ejmech.2021.113706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
A set of new substituted dienes were synthesized from betulinic acid by its oxidation to 30-oxobetulinic acid followed by the Wittig reaction. Cytotoxicity of all compounds was tested in vitro in eight cancer cell lines and two noncancer fibroblasts. Almost all dienes were more cytotoxic than betulinic acid. Compounds 4.22, 4.30, 4.33, 4.39 had IC50 below 5 μmol/L; 4.22 and 4.39 were selected for studies of the mechanism of action. Cell cycle analysis revealed an increase in the number of apoptotic cells at 5 × IC50 concentration, where activation of irreversible changes leading to cell death can be expected. Both 4.22 and 4.39 led to the accumulation of cells in the G0/G1 phase with partial inhibition of DNA/RNA synthesis at 1 × IC50 and almost complete inhibition at 5 × IC50. Interestingly, compound 4.39 at 5 × IC50 caused the accumulation of cells in the S phase. Higher concentrations of tested drugs probably inhibit more off-targets than lower concentrations. Mechanisms disrupting cellular metabolism can induce the accumulation of cells in the S phase. Both compounds 4.22 and 4.39 trigger selective apoptosis in cancer cells via intrinsic pathway, which we have demonstrated by changes in the expression of the crucial apoptosis-related protein. Pharmacological parameters of derivative 4.22 were superior to 4.39, therefore 4.22 was the finally selected candidate for the development of anticancer drug.
Collapse
Affiliation(s)
- Jan Pokorný
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Denisa Olejníková
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Ivo Frydrych
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Barbora Lišková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Soňa Gurská
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Sandra Benická
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Jan Šarek
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Jana Kotulová
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic
| | - Milan Urban
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 779 00, Olomouc, Czech Republic.
| |
Collapse
|
20
|
Celik S, Demirag AD, E. Ozel A, Akyuz S. Molecular Structure, Molecular Docking and Absorption, Distribution, Metabolism, Excretion and Toxicity study of cellulose II. J CHIN CHEM SOC-TAIP 2021. [DOI: 10.1002/jccs.202000515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sefa Celik
- Faculty of Science, Department of Physics Istanbul University Istanbul Turkey
| | - Aliye Demet Demirag
- Department of Physics, Institute of Graduate Studies in Sciences Istanbul University Istanbul Turkey
| | - Aysen E. Ozel
- Faculty of Science, Department of Physics Istanbul University Istanbul Turkey
| | - Sevim Akyuz
- Faculty of Science and Letters, Department of Physics Istanbul Kultur University Istanbul Turkey
| |
Collapse
|
21
|
Roy R, Sk MF, Jonniya NA, Poddar S, Kar P. Finding potent inhibitors against SARS-CoV-2 main protease through virtual screening, ADMET, and molecular dynamics simulation studies. J Biomol Struct Dyn 2021; 40:6556-6568. [DOI: 10.1080/07391102.2021.1897680] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Rajarshi Roy
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, MP, India
| | - Md Fulbabu Sk
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, MP, India
| | - Nisha Amarnath Jonniya
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, MP, India
| | - Sayan Poddar
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, MP, India
| | - Parimal Kar
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, MP, India
| |
Collapse
|
22
|
Lukin A, Bakholdina A, Zhurilo N, Onopchenko O, Zhuravel E, Zozulya S, Gureev M, Safrygin A, Krasavin M. Exploration of the nitrogen heterocyclic periphery around the core of the advanced FFA1 agonist fasiglifam (TAK-875). Arch Pharm (Weinheim) 2020; 354:e2000275. [PMID: 33270252 DOI: 10.1002/ardp.202000275] [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: 07/30/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 11/06/2022]
Abstract
Three types of heterocyclic moieties-piperidines fused to a heteroaromatic moiety-were explored as potential periphery motifs for the pharmacophoric core of fasiglifam (TAK-875), with fasiglifam being the most advanced agonist of free fatty acid receptor 1, a promising target for therapeutic intervention in type 2 diabetes. Several observed structure-activity relationship trends were corroborated by in silico docking results. Balanced selection based on potency and Caco-2 permeability advanced six compounds to cellular efficacy tests (glucose-stimulated insulin secretion in rat insulinoma INS1E cells). This led to the nomination of compound 16a (LK1408, 3-[4-({4-[(3-{[(2-fluorobenzyl)oxy]methyl}-1-methyl-1,4,6,7-tetrahydro-5H-pyrazolo[4,3-c]pyridin-5-yl)methyl]benzyl}oxy)phenyl]propanoic acid hydrochloride) as the lead for further development.
Collapse
Affiliation(s)
- Alexey Lukin
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Moscow, Russian Federation
| | - Anna Bakholdina
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Moscow, Russian Federation
| | - Nikolay Zhurilo
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Moscow, Russian Federation
| | | | | | - Sergey Zozulya
- Enamine Ltd., Kyiv, Ukraine.,Taras Shevchenko National University, Kyiv, Ukraine
| | - Maxim Gureev
- I. M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander Safrygin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Mikhail Krasavin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg, Russian Federation
| |
Collapse
|
23
|
VZHE-039, a novel antisickling agent that prevents erythrocyte sickling under both hypoxic and anoxic conditions. Sci Rep 2020; 10:20277. [PMID: 33219275 PMCID: PMC7679387 DOI: 10.1038/s41598-020-77171-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
Sickle cell disease (SCD) results from a hemoglobin (Hb) mutation βGlu6 → βVal6 that changes normal Hb (HbA) into sickle Hb (HbS). Under hypoxia, HbS polymerizes into rigid fibers, causing red blood cells (RBCs) to sickle; leading to numerous adverse pathological effects. The RBC sickling is made worse by the low oxygen (O2) affinity of HbS, due to elevated intra-RBC concentrations of the natural Hb effector, 2,3-diphosphoglycerate. This has prompted the development of Hb modifiers, such as aromatic aldehydes, with the intent of increasing Hb affinity for O2 with subsequent prevention of RBC sickling. One such molecule, Voxelotor was recently approved by U.S. FDA to treat SCD. Here we report results of a novel aromatic aldehyde, VZHE-039, that mimics both the O2-dependent and O2-independent antisickling properties of fetal hemoglobin. The latter mechanism of action—as elucidated through crystallographic and biological studies—is likely due to disruption of key intermolecular contacts necessary for stable HbS polymer formation. This dual antisickling mechanism, in addition to VZHE-039 metabolic stability, has translated into significantly enhanced and sustained pharmacologic activities. Finally, VZHE-039 showed no significant inhibition of several CYPs, demonstrated efficient RBC partitioning and high membrane permeability, and is not an efflux transporter (P-gp) substrate.
Collapse
|
24
|
Dobyns BM, Kim JM, Beckingham BS. Multicomponent transport of methanol and sodium acetate in poly(ethylene glycol) diacrylate membranes of varied fractional free volume. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109809] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Bhowmik D, Jagadeesan R, Rai P, Nandi R, Gugan K, Kumar D. Evaluation of potential drugs against leishmaniasis targeting catalytic subunit of Leishmania donovani nuclear DNA primase using ligand based virtual screening, docking and molecular dynamics approaches. J Biomol Struct Dyn 2020; 39:1838-1852. [PMID: 32141397 DOI: 10.1080/07391102.2020.1739557] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Leishmania donovani, causes leishmaniasis, a global health trouble with around 89 different countries and its population under its risk. Replication initiation events have been instrumental in regulating the DNA duplication and as the small subunit of L. donovani nuclear DNA primase (Ld-PriS) inherits the catalytic site, it plays a vital role in DNA replication. In this study we have aimed Ld-PriS for the first time as a prospective target for the application of drug against Leishmania parasite. 3-D structures of Ld-PriS were built and ligand-based virtual screening was performed using hybrid similarity recognition techniques. Ligands from the ZINC database were used for the screening purposes based on known DNA primase inhibitor Sphingosine as a query. Top 150 ligands were taken into consideration for molecular docking against the query protein (Ld-PriS) using PyRx and iGEMDOCK softwares. Top five compounds with the best docking score were selected for pharmacokinetic investigation and molecular dynamic simulation. These top five screened inhibitors showed very poor binding affinity toward the catalytic subunit of human primase indicating their safety toward the host normal replication mechanism. The top five compounds showed good pharmacokinetic profiles and ADMET predictions revealed good absorption, solubility, permeability, uniform distribution, proper metabolism, minimal toxicity and good bioavailability. Simulation studies upto 50 ns revealed the three leads ZINC000009219046, ZINC000025998119 and ZINC000004677901 bind with Ld-PriS throughout the simulation and there were no huge variations in their backbone suggesting that these three may play as potential lead compounds for developing new drug against leishmaniasis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Deep Bhowmik
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Rahul Jagadeesan
- CAS in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai, India
| | - Praveen Rai
- Department of Biotechnology, Central University of Rajasthan, Bandarsindri, India
| | - Rajat Nandi
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Kothandan Gugan
- CAS in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, Assam, India
| |
Collapse
|
26
|
Li X, Payne DT, Ampolu B, Bland N, Brown JT, Dutton MJ, Fitton CA, Gulliver A, Hale L, Hamza D, Jones G, Lane R, Leach AG, Male L, Merisor EG, Morton MJ, Quy AS, Roberts R, Scarll R, Schulz-Utermoehl T, Stankovic T, Stevenson B, Fossey JS, Agathanggelou A. Derivatisation of parthenolide to address chemoresistant chronic lymphocytic leukaemia. MEDCHEMCOMM 2019; 10:1379-1390. [PMID: 32952998 PMCID: PMC7478165 DOI: 10.1039/c9md00297a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
Parthenolide is a natural product that exhibits anti-leukaemic activity, however, its clinical use is limited by its poor bioavailability. It may be extracted from feverfew and protocols for growing, extracting and derivatising it are reported. A novel parthenolide derivative with good bioavailability and pharmacological properties was identified through a screening cascade based on in vitro anti-leukaemic activity and calculated "drug-likeness" properties, in vitro and in vivo pharmacokinetics studies and hERG liability testing. In vitro studies showed the most promising derivative to have comparable anti-leukaemic activity to DMAPT, a previously described parthenolide derivative. The newly identified compound was shown to have pro-oxidant activity and in silico molecular docking studies indicate a prodrug mode of action. A synthesis scheme is presented for the production of amine 7 used in the generation of 5f.
Collapse
Affiliation(s)
- Xingjian Li
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Daniel T Payne
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Badarinath Ampolu
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Nicholas Bland
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Jane T Brown
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Mark J Dutton
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Catherine A Fitton
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Abigail Gulliver
- Winterbourne Botanic Garden, University of Birmingham, 58 Edgbaston Park Road, Edgbaston, Birmingham, West Midlands B15 2RT, UK
| | - Lee Hale
- Winterbourne Botanic Garden, University of Birmingham, 58 Edgbaston Park Road, Edgbaston, Birmingham, West Midlands B15 2RT, UK
| | - Daniel Hamza
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Geraint Jones
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Rebecca Lane
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Andrew G Leach
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Louise Male
- X-Ray Crystallography Facility, School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK
| | - Elena G Merisor
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Michael J Morton
- ApconiX Ltd, Alderly Park, Nether Alderly, Cheshire, SK10 4TG, UK
| | - Alex S Quy
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Ruth Roberts
- ApconiX Ltd, Alderly Park, Nether Alderly, Cheshire, SK10 4TG, UK
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK
| | - Rosanna Scarll
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | | | - Tatjana Stankovic
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Brett Stevenson
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Angelo Agathanggelou
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| |
Collapse
|
27
|
Lemieux MR, Siricilla S, Mitachi K, Eslamimehr S, Wang Y, Yang D, Pressly JD, Kong Y, Park F, Franzblau SG, Kurosu M. An antimycobacterial pleuromutilin analogue effective against dormant bacilli. Bioorg Med Chem 2018; 26:4787-4796. [PMID: 30145051 PMCID: PMC6154393 DOI: 10.1016/j.bmc.2018.07.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/06/2018] [Accepted: 07/19/2018] [Indexed: 11/17/2022]
Abstract
Pleuromutilin is a promising pharmacophore to design new antibacterial agents for Gram-positive bacteria. However, there are limited studies on the development of pleuromutilin analogues that inhibit growth of Mycobacterium tuberculosis (Mtb). In screening of our library of pleuromutilin derivatives, UT-800 (1) was identified to kill replicating- and non-replicating Mtb with the MIC values of 0.83 and 1.20 μg/mL, respectively. UT-800 also kills intracellular Mtb faster than rifampicin at 2× MIC concentrations. Pharmacokinetic studies indicate that 1 has an oral bioavailability with an average F-value of 27.6%. Pleuromutilin may have the potential to be developed into an orally administered anti-TB drug.
Collapse
Affiliation(s)
- Maddie R Lemieux
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Shajila Siricilla
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Katsuhiko Mitachi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Shakiba Eslamimehr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL 60612, United States
| | - Dong Yang
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, 858 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Jeffrey D Pressly
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Ying Kong
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, 858 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL 60612, United States
| | - Michio Kurosu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163-0001, United States.
| |
Collapse
|
28
|
Zheng Y, Feng G, Sun Y, Liu S, Pi Z, Song F, Liu Z. Study on the compatibility interactions of formula Ding-Zhi-Xiao-Wan based on their main components transport characteristics across Caco-2 monolayers model. J Pharm Biomed Anal 2018; 159:179-185. [DOI: 10.1016/j.jpba.2018.06.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/25/2018] [Accepted: 06/30/2018] [Indexed: 01/04/2023]
|
29
|
Liu H, Ye H, Sun C, Xi H, Ma J, Lai F, Wu H. Antioxidant activity in HepG2 cells, immunomodulatory effects in RAW 264.7 cells and absorption characteristics in Caco-2 cells of the peptide fraction isolated from Dendrobium aphyllum. Int J Food Sci Technol 2018. [DOI: 10.1111/ijfs.13783] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Huifan Liu
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Hongji Ye
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Chongzhen Sun
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Hongru Xi
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Juanjuan Ma
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Furao Lai
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| | - Hui Wu
- College of Food Science and Engineering; South China University of Technology; Guangzhou, Guangdong 510640 China
| |
Collapse
|
30
|
Ayehunie S, Landry T, Stevens Z, Armento A, Hayden P, Klausner M. Human Primary Cell-Based Organotypic Microtissues for Modeling Small Intestinal Drug Absorption. Pharm Res 2018; 35:72. [PMID: 29476278 PMCID: PMC6599640 DOI: 10.1007/s11095-018-2362-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE The study evaluates the use of new in vitro primary human cell-based organotypic small intestinal (SMI) microtissues for predicting intestinal drug absorption and drug-drug interaction. METHODS The SMI microtissues were reconstructed using human intestinal fibroblasts and enterocytes cultured on a permeable support. To evaluate the suitability of the intestinal microtissues to model drug absorption, the permeability coefficients across the microtissues were determined for a panel of 11 benchmark drugs with known human absorption and Caco-2 permeability data. Drug-drug interactions were examined using efflux transporter substrates and inhibitors. RESULTS The 3D-intestinal microtissues recapitulate the structural features and physiological barrier properties of the human small intestine. The microtissues also expressed drug transporters and metabolizing enzymes found on the intestinal wall. Functionally, the SMI microtissues were able to discriminate between low and high permeability drugs and correlated better with human absorption data (r2 = 0.91) compared to Caco-2 cells (r2 = 0.71). Finally, the functionality of efflux transporters was confirmed using efflux substrates and inhibitors which resulted in efflux ratios of >2.0 fold and by a decrease in efflux ratios following the addition of inhibitors. CONCLUSION The SMI microtissues appear to be a useful pre-clinical tool for predicting drug bioavailability of orally administered drugs.
Collapse
Affiliation(s)
- Seyoum Ayehunie
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA.
| | - Tim Landry
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Zachary Stevens
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Alex Armento
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Patrick Hayden
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | | |
Collapse
|
31
|
Uba AI, Yelekçi K. Identification of potential isoform-selective histone deacetylase inhibitors for cancer therapy: a combined approach of structure-based virtual screening, ADMET prediction and molecular dynamics simulation assay. J Biomol Struct Dyn 2017; 36:3231-3245. [PMID: 28938863 DOI: 10.1080/07391102.2017.1384402] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Histone deacetylases (HDACs) have gained increased attention as targets for anticancer drug design and development. HDAC inhibitors have proven to be effective for reversing the malignant phenotype in HDAC-dependent cancer cases. However, lack of selectivity of the many HDAC inhibitors in clinical use and trials contributes to toxicities to healthy cells. It is believed that, the continued identification of isoform-selective inhibitors will eliminate these undesirable adverse effects - a task that remains a major challenge to HDAC inhibitor designs. Here, in an attempt to identify isoform-selective inhibitors, a large compound library containing 2,703,000 compounds retrieved from Otava database was screened against class I HDACs by exhaustive approach of structure-based virtual screening using rDOCK and Autodock Vina. A total of 41 compounds were found to show high-isoform selectivity and were further redocked into their respective targets using Autodock4. Thirty-six compounds showed remarkable isoform selectivity and passed drug-likeness and absorption, distribution, metabolism, elimination and toxicity prediction tests using ADMET Predictor™ and admetSAR. Furthermore, to study the stability of ligand binding modes, 10 ns-molecular dynamics (MD) simulations of the free HDAC isoforms and their complexes with respective best-ranked ligands were performed using nanoscale MD software. The inhibitors remained bound to their respective targets over time of the simulation and the overall potential energy, root-mean-square deviation, root-mean-square fluctuation profiles suggested that the detected compounds may be potential isoform-selective HDAC inhibitors or serve as promising scaffolds for further optimization towards the design of selective inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Abdullahi Ibrahim Uba
- a Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences , Kadir Has University , Fatih, Istanbul 34083 , Turkey.,b Center for Biotechnology Research , Bayero University , P.M.B. 3011, B.U.K. Road, Kano , Nigeria
| | - Kemal Yelekçi
- a Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences , Kadir Has University , Fatih, Istanbul 34083 , Turkey
| |
Collapse
|
32
|
Wang Q, Kuang Y, Song W, Qian Y, Qiao X, Guo DA, Ye M. Permeability through the Caco-2 cell monolayer of 42 bioactive compounds in the TCM formula Gegen-Qinlian Decoction by liquid chromatography tandem mass spectrometry analysis. J Pharm Biomed Anal 2017; 146:206-213. [PMID: 28886521 DOI: 10.1016/j.jpba.2017.08.042] [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: 06/06/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 10/19/2022]
Abstract
Caco-2 cell monolayer model was used to evaluate the intestinal permeability of 42 bioactive compounds in the famous traditional Chinese medicine (TCM) formula Gegen-Qinlian Decoction (GQD). These compounds include alkaloids, flavonoids and glycosides, triterpenoid saponins, and coumarins. Their transportations across the cell monolayers in the forms of herb extract and formula extract were monitored by liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) analysis. Most alkaloids from Huang-Lian; flavonoid C-glycosides from Ge-Gen and Huang-Qin; O-glycosides from Ge-Gen, Huang-Qin and Gan-Cao; O-glucuronides from Huang-Qin; and coumarins from Gan-Cao exhibited favorable permeability. Their PAB values were >1.05×10-5cm/s, and efflux ratios (ER, PBA/PAB) were≤1.0. In contrast, triterpenoid saponins showed poor permeability (PAB≤1.50×10-6cm/s, ER≤1.5), indicating a paracellular diffusion mechanism. Furthermore, GQD could remarkably improve the intestinal transport of alkaloids in Huang-Lian, flavonoid C-glycosides in Ge-Gen, as well as coumarins and flavonoid O-glycosides in Gan-Cao. These results indicate herb-herb interactions in GQD.
Collapse
Affiliation(s)
- Qi Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Baojian Road 157, Nangang District, Harbin 150081, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Yi Kuang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Wei Song
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Yi Qian
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Xue Qiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| | - De-An Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| |
Collapse
|
33
|
Mathur S, Park JD, Kim DH, Hartmann RW. A Method for Screening Enzyme Inhibitors Using Size Exclusion Chromatography and ESI-LC-MS/MS. ACTA ACUST UNITED AC 2016; 10:30-5. [PMID: 15695341 DOI: 10.1177/1087057104270270] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A pilot study was performed for the development of a method to screen compound libraries using an electrospray mass spectrometer interfaced with liquid chromatography (LC). The mixture of compounds was obtained by combining low-molecular weight inhibitors of carboxypeptidase A (CPA), a representative zinc-containing proteolytic enzyme. After the incubation of the mixture with CPA, the enzyme-bound compounds were separated by size exclusion chromatography (SEC) from unbound compounds. The separation of compounds was affected by LC. Three compounds were identified, which represent the tight binding inhibitors of the library. These compounds were quantitated using an automatic switching valve to avoid the interference of buffer salts with the detection of analytes. The quantitated amounts of the compounds were found to be in good accordance with the Ki values. ( Journal of Biomolecular Screening 2005:30-35)
Collapse
Affiliation(s)
- Sonal Mathur
- FR 8.5, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbruecken, Germany
| | | | | | | |
Collapse
|
34
|
Júnior MADR, de Faria ACM, Velozo EDS, Dalla Costa T, de Andrade FP, de Castro WV. Determination of fexofenadine in Hank's balanced salt solution by high-performance liquid chromatography with ultraviolet detection: application to Caco-2 cell permeability studies. Biomed Chromatogr 2014; 29:537-44. [DOI: 10.1002/bmc.3310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/26/2014] [Accepted: 07/22/2014] [Indexed: 01/18/2023]
|
35
|
Defaux J, Antoine M, Le Borgne M, Schuster T, Seipelt I, Aicher B, Teifel M, Günther E, Gerlach M, Marchand P. Discovery of 7-Aryl-Substituted (1,5-Naphthyridin-4-yl)ureas as Aurora Kinase Inhibitors. ChemMedChem 2013; 9:217-32. [DOI: 10.1002/cmdc.201300384] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/04/2013] [Indexed: 11/09/2022]
|
36
|
Smith AJ, Kavuru P, Arora KK, Kesani S, Tan J, Zaworotko MJ, Shytle RD. Crystal engineering of green tea epigallocatechin-3-gallate (EGCg) cocrystals and pharmacokinetic modulation in rats. Mol Pharm 2013; 10:2948-61. [PMID: 23730870 PMCID: PMC3795472 DOI: 10.1021/mp4000794] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The
most abundant polyphenol in green tea, epigallocatechin-3-gallate
(EGCg), has recently received considerable attention due to the discovery
of numerous health-promoting bioactivities. Despite reports of its
poor oral bioavailability, EGCg has been included in many dietary
supplement formulations. Conventional preformulation methods have
been employed to improve the bioavailability of EGCg. However, these
methods have limitations that hinder the development of EGCg as an
effective therapeutic agent. In this study, we have utilized the basic
concepts of crystal engineering and several crystallization techniques
to screen for various solid crystalline forms of EGCg and evaluated
the efficacy of crystal engineering for modulating the pharmacokinetics
of EGCg. We synthesized and characterized seven previously undescribed
crystal forms of EGCg including the pure crystal structure of EGCg.
The aqueous solubility profiles of four new EGCg cocrystals were determined.
These cocrystals were subsequently dosed at 100 mg EGCg per kg body
weight in rats, and the plasma levels were monitored over the course
of eight hours following the single oral dose. Two of the EGCg cocrystals
were found to exhibit modest improvements in relative bioavailability.
Further, cocrystallization resulted in marked effects on pharmacokinetic
parameters including Cmax, Tmax, area under curve, relative bioavailability, and apparent
terminal half-life. Our findings suggest that modulation of the pharmacokinetic
profile of EGCg is possible using cocrystallization and that it offers
certain opportunities that could be useful during its development
as a therapeutic agent.
Collapse
Affiliation(s)
- Adam J Smith
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Health College of Medicine, University of South Florida , Tampa, Florida 33612, United States
| | | | | | | | | | | | | |
Collapse
|
37
|
Ketola RA, Mauriala T. Mass spectrometric tools for cell and tissue studies. Eur J Pharm Sci 2012; 46:293-314. [DOI: 10.1016/j.ejps.2012.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/14/2012] [Accepted: 03/23/2012] [Indexed: 10/28/2022]
|
38
|
Pratt J, Venkatraman N, Brinker A, Xiao Y, Blasberg J, Thompson DC, Bourner M. Use of Zinc Finger Nuclease Technology to Knock Out Efflux Transporters in C2BBe1 Cells. ACTA ACUST UNITED AC 2012; Chapter 23:Unit 23.2.. [DOI: 10.1002/0471140856.tx2302s52] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | | | | | - Jim Blasberg
- Life Sciences R&D, Sigma‐Aldrich St. Louis Missouri
| | | | | |
Collapse
|
39
|
Smith AJ, Kavuru P, Wojtas L, Zaworotko MJ, Shytle RD. Cocrystals of quercetin with improved solubility and oral bioavailability. Mol Pharm 2011; 8:1867-76. [PMID: 21846121 DOI: 10.1021/mp200209j] [Citation(s) in RCA: 219] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Flavonoids have been studied extensively due to the observation that diets rich in these compounds are associated with lower incidences of many diseases. One of the most studied flavonoids, quercetin, is also the most abundant of these compounds in the plant kingdom. Numerous therapeutic bioactivities have been identified in vitro. However, its in vivo efficacy in pure form is limited by poor bioavailability, primarily due to its low solubility and consequent low absorption in the gut. Cocrystallization has gained attention recently as a means for improving the physicochemical characteristics of a compound. Here, we synthesized and evaluated four new cocrystals of quercetin (QUE): quercetin:caffeine (QUECAF), quercetin:caffeine:methanol (QUECAF·MeOH), quercetin:isonicotinamide (QUEINM), and quercetin:theobromine dihydrate (QUETBR · 2H(2)O). Each of these cocrystals exhibited pharmacokinetic properties that are vastly superior to those of quercetin alone. Cocrystallization was able to overcome the water insolubility of quercetin, with all four cocrystals exhibiting some degree of solubility. The QUECAF and QUECAF·MeOH cocrystals increased the solubility of QUE by 14- and 8-fold when compared to QUE dihydrate. We hypothesized that this improved solubility would translate into enhanced systemic absorption of QUE. This hypothesis was supported in our pharmacokinetic study. The cocrystals outperformed QUE dihydrate with increases in bioavailability up to nearly 10-fold.
Collapse
Affiliation(s)
- Adam J Smith
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Health College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | | | | | | |
Collapse
|
40
|
Yeon JH, Park JK. Drug permeability assay using microhole-trapped cells in a microfluidic device. Anal Chem 2010; 81:1944-51. [PMID: 19203200 DOI: 10.1021/ac802351w] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As orally administered drugs must be absorbed from the intestine into the blood circulation, permeability assays of drug candidates have been widely used in the early screening stages of drug discovery. In this study, a microfluidic device was developed for the drug permeability assay, considering the in vivo delivery path of drugs in humans. A microhole array for cell trapping was fabricated using the poly(dimethylsiloxane) (PDMS) molding technique by mimicking the intestinal epithelial cell membrane. On the basis of mathematical simulations, the configuration of the microfluidic device, including a microhole array and a mixing channel, was optimized to trap cells firmly in each microhole. At the flow rate under optimal conditions, cells were effectively trapped in a microhole array without cell damage. We measured the permeability of 10 drugs, including those with high and low permeability in microchannels, and compared the results with the reported values of permeability in the human and rat intestine. Most drugs had a high p value (p > 0.4), and only a few drugs had a low p value less than 0.05 by t test. Though their measured permeabilities are not the same as those in vivo human intestine, it shows that in vivo permeabilities in the human and rat intestine are highly correlated with those measured by the microfluidic device (R(2) = 0.9013 and R(2) = 0.8765, respectively). Also, the fraction of the dose absorbed in the human intestine (F(a)) indicated that the drug permeability measured using this device was significantly correlated (R(2) = 0.9641) with those in human subjects. As the microfluidic assay system is dependent on cells trapped inside a microhole array, it is a valuable tool in drug discovery as well as an alternative to animal testing.
Collapse
Affiliation(s)
- Ju Hun Yeon
- Department of Bio and Brain Engineering, College of Life Science and Bioengineering, KAIST, 335 Gwahangno, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | | |
Collapse
|
41
|
Smith A, Giunta B, Bickford PC, Fountain M, Tan J, Shytle RD. Nanolipidic particles improve the bioavailability and α-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer's disease. Int J Pharm 2010. [DOI: 78495111110.1016/j.ijpharm.2010.01.012' target='_blank'>'"<>78495111110.1016/j.ijpharm.2010.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [78495111110.1016/j.ijpharm.2010.01.012','', '10.1002/(sici)1096-9888(200001)35:1<71::aid-jms915>3.0.co;2-5')">Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
78495111110.1016/j.ijpharm.2010.01.012" />
|
42
|
Smith A, Giunta B, Bickford PC, Fountain M, Tan J, Shytle RD. Nanolipidic particles improve the bioavailability and alpha-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer's disease. Int J Pharm 2010; 389:207-12. [PMID: 20083179 DOI: 10.1016/j.ijpharm.2010.01.012] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 01/05/2010] [Accepted: 01/10/2010] [Indexed: 01/27/2023]
Abstract
Prevention of amyloidogenic processing of amyloid precursor protein with the use of natural phytochemicals capable of enhancing alpha-secretase activity may be a therapeutic approach for treatment of neurodegenerative diseases including Alzheimer's disease (AD) and HIV-associated dementia (HAD). We have recently shown promising preclinical results with the use of green tea polyphenol, (-)-epigallocatechin-3-gallate (EGCG) in mouse models of both diseases, however the translation into clinical use has been problematic primarily as a result of poor bioavailability and inefficient delivery to the central nervous system (CNS). While the antioxidant properties of EGCG are well known, we have shown that it is able to promote non-amyloidogenic processing of amyloid precursor protein (APP) by upregulating alpha-secretase, thus preventing brain beta amyloid plaque formation, a hallmark of AD pathology and common finding in HIV infection. In this preliminary study, we investigated the ability of one preformulation method to improve the oral bioavailability of EGCG. We found that forming nanolipidic EGCG particles improves the neuronal (SweAPP N2a cells) alpha-secretase enhancing ability in vitro by up to 91% (P<001) and it's oral bioavailability in vivo by more than two-fold over free EGCG.
Collapse
Affiliation(s)
- Adam Smith
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | |
Collapse
|
43
|
Nomeir AA, Morrison R, Prelusky D, Korfmacher W, Broske L, Hesk D, McNamara P, Mei H. Estimation of the extent of oral absorption in animals from oral and intravenous pharmacokinetic data in drug discovery. J Pharm Sci 2009; 98:4027-38. [DOI: 10.1002/jps.21705] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
44
|
Wessling ST, Ross BP, Koda Y, Blanchfield JT, Toth I. Caco-2 cell permeability and stability of two d-glucopyranuronamide conjugates of thyrotropin-releasing hormone. Bioorg Med Chem 2007; 15:4946-50. [PMID: 17498958 DOI: 10.1016/j.bmc.2007.04.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/28/2007] [Accepted: 04/25/2007] [Indexed: 10/23/2022]
Abstract
Caco-2 cell permeability and stability assays were used as an in vitro model to study the intestinal epithelial transport and stability of two analogues of thyrotropin-releasing hormone (TRH; Pyr-His-Pro-NH2). Peptide 1 (Pyr-His-Pro-D-glucopyranuronamide) was more permeable across the Caco-2 cell monolayer compared with the permeability of the parent TRH peptide (Papp=5.10+/-1.89x10(-6) cm/s c.f. Papp=0.147+/-0.0474x10(-6) cm/s respectively). The permeability of peptide 1 was improved threefold by attaching a 2-aminooctanoic acid moiety to the N-terminus to form peptide 2 (2-aminooctanoic acid-Gln-His-Pro-D-glucopyranuronamide) (Papp=16.3+/-2.47x10(-6) cm/s). The half-life for both peptide 1 and peptide 2 was approximately 20 min in a homogenate of Caco-2 cells compared with the half-life of TRH which is approximately 3 min. It was concluded that the permeability of peptides 1 and 2 was enhanced because of their increased stability, while the higher permeability of peptide 2 compared with peptide 1 may be attributed to its increased lipophilicity which results in enhanced passive diffusion.
Collapse
Affiliation(s)
- Susanne T Wessling
- School of Molecular and Microbial Sciences, The University of Queensland, Brisbane, Qld. 4072, Australia
| | | | | | | | | |
Collapse
|
45
|
Cheng KC, Korfmacher WA, White RE, Njoroge FG. Lead Optimization in Discovery Drug Metabolism and Pharmacokinetics/Case study: The Hepatitis C Virus (HCV) Protease Inhibitor SCH 503034. PERSPECTIVES IN MEDICINAL CHEMISTRY 2007. [DOI: 10.1177/1177391x0700100001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Lead optimization using drug metabolism and pharmacokinetics (DMPK) parameters has become one of the primary focuses of research organizations involved in drug discovery in the last decade. Using a combination of rapid in vivo and in vitro DMPK screening procedures on a large array of compounds during the lead optimization process has resulted in development of compounds that have acceptable DMPK properties. In this review, we present a general screening paradigm that is currently being used as part of drug discovery at Schering-Plough and we describe a case study using the Hepatitis C Virus (HCV) protease inhibitor program as an example. By using the DMPK optimization tools, a potent HCV protease inhibitor, SCH 503034, was selected for development as a candidate drug.
Collapse
Affiliation(s)
- K.-C. Cheng
- Department of Drug Metabolism and Pharmacokinetics
| | | | | | - F. George Njoroge
- Chemistry Department Schering-Plough Research Institute, 2015 Galloping Hill Road, Kenilworth, NJ 07033 U.S.A
| |
Collapse
|
46
|
Arnold NJ, Arnold R, Beer D, Bhalay G, Collingwood SP, Craig S, Devereux N, Dodds M, Dunstan AR, Fairhurst RA, Farr D, Fullerton JD, Glen A, Gomez S, Haberthuer S, Hatto JDI, Howes C, Jones D, Keller TH, Leuenberger B, Moser HE, Muller I, Naef R, Nicklin PA, Sandham DA, Turner KL, Tweed MF, Watson SJ, Zurini M. Potent and selective xanthine-based inhibitors of phosphodiesterase 5. Bioorg Med Chem Lett 2007; 17:2376-9. [PMID: 17337182 DOI: 10.1016/j.bmcl.2006.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 11/04/2006] [Accepted: 11/06/2006] [Indexed: 11/30/2022]
Abstract
Inhibitors of PDE5 are useful therapeutic agents for treatment of erectile dysfunction. A series of novel xanthine derivatives has been identified as potent inhibitors of PDE5, with good levels of selectivity against other PDE isoforms, including PDE6. Studies in the dog indicate excellent oral bioavailability for compound 21.
Collapse
Affiliation(s)
- Nichola J Arnold
- Novartis Institutes of Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Mensch J, Noppe M, Adriaensen J, Melis A, Mackie C, Augustijns P, Brewster ME. Novel generic UPLC/MS/MS method for high throughput analysis applied to permeability assessment in early Drug Discovery. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 847:182-7. [PMID: 17095304 DOI: 10.1016/j.jchromb.2006.10.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Revised: 10/02/2006] [Accepted: 10/05/2006] [Indexed: 10/23/2022]
Abstract
A novel generic ultra performance liquid chromatography-tandem mass spectrometric (UPLC/MS/MS) method for the high throughput quantification of samples generated during permeability assessment (PAMPA) has been developed and validated. The novel UPLC/MS/MS methodology consists of two stages. Firstly, running a 1.5min isocratic method, compound-specific multiple reaction monitoring (MRM) methods were automatically prepared. In a second stage, samples were analyzed by a 1.5min generic gradient UPLC method on a BEH C18 column (50mmx2.1mm). Compounds were detected with a Waters Micromass Quattro Premier mass spectrometer operating in positive electrospray ionization using the compound-specific MRM methods. The linearity for the validation compounds (caffeine, propranolol, ampicillin, atenolol, griseofulvin and carbamazepine) typically ranges from 3.05nM to 12,500nM and the limits of detection for all generically developed methods are in the range between 0.61nM and 12nM in an aqueous buffer. The novel generic methodology was successfully introduced within early Drug Discovery and resulted in a four-fold increase of throughput as well as a significant increase in sensitivity compared to other in-house generic LC/MS methods.
Collapse
Affiliation(s)
- Jurgen Mensch
- Pharmaceutical Sciences, Johnson & Johnson Pharmaceutical Research & Development, A Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, 2340 Beerse, Belgium.
| | | | | | | | | | | | | |
Collapse
|
49
|
Yu P, Liu B, Kodadek T. A convenient, high-throughput assay for measuring the relative cell permeability of synthetic compounds. Nat Protoc 2007; 2:23-30. [PMID: 17401333 DOI: 10.1038/nprot.2007.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We describe here a convenient procedure for assessing the relative cell permeability of chemical compounds. The assay can be used in a high-throughput mode and is particularly applicable for the evaluation of the relative permeability of compounds in a combinatorial library. The compound of interest is conjugated to a dexamethasone derivative. The entry of the conjugate into living mammalian cells triggers the nuclear transport of a Gal4 DNA binding domain-glucocorticoid receptor ligand binding domain-VP16 activation domain fusion protein and, consequently, the activation of a Gal4-responsive luciferase reporter gene. The relative cell permeability is thus described quantitatively by the level of luciferase expression. The experiments take only a few days once chemical synthesis and conjugation are finished.
Collapse
Affiliation(s)
- Peng Yu
- Division of Translational Research, Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9185, USA
| | | | | |
Collapse
|
50
|
Chang C, Ekins S, Bahadduri P, Swaan PW. Pharmacophore-based discovery of ligands for drug transporters. Adv Drug Deliv Rev 2006; 58:1431-50. [PMID: 17097188 PMCID: PMC1773055 DOI: 10.1016/j.addr.2006.09.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Accepted: 09/04/2006] [Indexed: 11/24/2022]
Abstract
The ability to identify ligands for drug transporters is an important step in drug discovery and development. It can both improve accurate profiling of lead pharmacokinetic properties and assist in the discovery of new chemical entities targeting transporters. In silico approaches, especially pharmacophore-based database screening methods have great potential in improving the throughput of current transporter ligand identification assays, leading to a higher hit rate by focusing in vitro testing to the most promising hits. In this review, the potential of different in silico methods in transporter ligand identification studies are compared and summarized with an emphasis on pharmacophore modeling. Various implementations of pharmacophore model generation, database compilation and flexible screening algorithms are also introduced. Recent successful utilization of database searching with pharmacophores to identify novel ligands for the pharmaceutically significant transporters hPepT1, P-gp, BCRP, MRP1 and DAT are reviewed and the challenges encountered with current approaches are discussed.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Pharmaceutical Sciences, School of Pharmacy,
University of Maryland, Baltimore, MD 21201 and
| | - Sean Ekins
- Department of Pharmaceutical Sciences, School of Pharmacy,
University of Maryland, Baltimore, MD 21201 and
- ACT LLC, 1 Penn Plaza-36th Floor, New York, NY 10119
| | - Praveen Bahadduri
- Department of Pharmaceutical Sciences, School of Pharmacy,
University of Maryland, Baltimore, MD 21201 and
| | - Peter W. Swaan
- Department of Pharmaceutical Sciences, School of Pharmacy,
University of Maryland, Baltimore, MD 21201 and
- Author for correspondence: Peter W. Swaan, Ph.D., Department of
Pharmaceutical Sciences, 20 Penn Street, HSF2-621, University of Maryland,
Baltimore, Baltimore, MD 21201, Tel: 410-706 –0130, Fax:
410-706-5017,
| |
Collapse
|