1
|
Nguyen PH, Cui S, Kozarich AM, Rautio A, Roberts AG, Xiong MP. Utilizing surface plasmon resonance as a novel method for monitoring in-vitro P-glycoprotein efflux. FRONTIERS IN BIOPHYSICS 2024; 2:1367511. [PMID: 38645731 PMCID: PMC11027885 DOI: 10.3389/frbis.2024.1367511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
P-glycoprotein (Pgp) is known for its dichotomous roles as both a safeguarding efflux transporter against xenobiotics and as a catalyst for multidrug resistance. Given the susceptibility of numerous therapeutic compounds to Pgp-mediated resistance, compliance with Food and Drug Administration (FDA) guidelines mandates an in-depth in vitro transport assay during drug development. This study introduces an innovative transport assay that aligns with these regulatory imperatives but also addresses limitations in the currently established techniques. Using Pgp-reconstituted liposomes and employing surface plasmon resonance (SPR), this study developed a distinct method of measuring the relative transport rates of Pgp substrates in a controlled microenvironment. The Pgp substrates selected for this study-quinidine, methadone, and desipramine-resulted in transport ratios that corroborate with trends previously observed. To assess the kinetics of Pgp-mediated transport, the results were analyzed by fitting the data to both currently proposed Pgp substrate translocation models-the vacuum cleaner and flippase models. While the resulting kinetic analysis in this study lends support predominantly to the vacuum cleaner model, this study most notably developed a novel method of assessing Pgp-mediated transport rates and real-time kinetics using surface plasmon resonance.
Collapse
Affiliation(s)
- Phuong H. Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Shuolin Cui
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Amanda M. Kozarich
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Alex Rautio
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Arthur G. Roberts
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - May P. Xiong
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
2
|
Dudas B, Miteva MA. Computational and artificial intelligence-based approaches for drug metabolism and transport prediction. Trends Pharmacol Sci 2024; 45:39-55. [PMID: 38072723 DOI: 10.1016/j.tips.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 01/07/2024]
Abstract
Drug metabolism and transport, orchestrated by drug-metabolizing enzymes (DMEs) and drug transporters (DTs), are implicated in drug-drug interactions (DDIs) and adverse drug reactions (ADRs). Reliable and precise predictions of DDIs and ADRs are critical in the early stages of drug development to reduce the rate of drug candidate failure. A variety of experimental and computational technologies have been developed to predict DDIs and ADRs. Recent artificial intelligence (AI) approaches offer new opportunities for better predicting and understanding the complex processes related to drug metabolism and transport. We summarize the role of major DMEs and DTs, and provide an overview of current progress in computational approaches for the prediction of drug metabolism, transport, and DDIs, with an emphasis on AI including machine learning (ML) and deep learning (DL) modeling.
Collapse
Affiliation(s)
- Balint Dudas
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France
| | - Maria A Miteva
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France.
| |
Collapse
|
3
|
Komura H, Watanabe R, Mizuguchi K. The Trends and Future Prospective of In Silico Models from the Viewpoint of ADME Evaluation in Drug Discovery. Pharmaceutics 2023; 15:2619. [PMID: 38004597 PMCID: PMC10675155 DOI: 10.3390/pharmaceutics15112619] [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: 10/09/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Drug discovery and development are aimed at identifying new chemical molecular entities (NCEs) with desirable pharmacokinetic profiles for high therapeutic efficacy. The plasma concentrations of NCEs are a biomarker of their efficacy and are governed by pharmacokinetic processes such as absorption, distribution, metabolism, and excretion (ADME). Poor ADME properties of NCEs are a major cause of attrition in drug development. ADME screening is used to identify and optimize lead compounds in the drug discovery process. Computational models predicting ADME properties have been developed with evolving model-building technologies from a simplified relationship between ADME endpoints and physicochemical properties to machine learning, including support vector machines, random forests, and convolution neural networks. Recently, in the field of in silico ADME research, there has been a shift toward evaluating the in vivo parameters or plasma concentrations of NCEs instead of using predictive results to guide chemical structure design. Another research hotspot is the establishment of a computational prediction platform to strengthen academic drug discovery. Bioinformatics projects have produced a series of in silico ADME models using free software and open-access databases. In this review, we introduce prediction models for various ADME parameters and discuss the currently available academic drug discovery platforms.
Collapse
Affiliation(s)
- Hiroshi Komura
- University Research Administration Center, Osaka Metropolitan University, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Osaka, Japan
| | - Reiko Watanabe
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Osaka, Japan; (R.W.); (K.M.)
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), 3-17 Senrioka-shinmachi, Settu 566-0002, Osaka, Japan
| | - Kenji Mizuguchi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Osaka, Japan; (R.W.); (K.M.)
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), 3-17 Senrioka-shinmachi, Settu 566-0002, Osaka, Japan
| |
Collapse
|
4
|
Zhang Y, Sharma S, Jonnalagadda S, Kumari S, Queen A, Esfahani SH, Archie SR, Nozohouri S, Patel D, Trippier PC, Karamyan VT, Abbruscato TJ. Discovery of the Next Generation of Non-peptidomimetic Neurolysin Activators with High Blood-Brain Barrier Permeability: a Pharmacokinetics Study in Healthy and Stroke Animals. Pharm Res 2023; 40:2747-2758. [PMID: 37833570 DOI: 10.1007/s11095-023-03619-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
PURPOSE There is growing interest in seeking pharmacological activation of neurolysin (Nln) for stroke treatment. Discovery of central nervous system drugs remains challenging due to the protection of the blood-brain barrier (BBB). The previously reported peptidomimetic Nln activators display unsatisfactory BBB penetration. Herein, we investigate the next generation of non-peptidomimetic Nln activators with high BBB permeability. METHODS A BBB-mimicking model was used to evaluate their in vitro BBB permeability. Protein binding, metabolic stability, and efflux assays were performed to determine their unbound fraction, half-lives in plasma and brains, and dependence of BBB transporter P-glycoprotein (P-gp). The in vivo pharmacokinetic profiles were elucidated in healthy and stroke mice. RESULTS Compounds KS52 and KS73 out of this generation exhibit improved peptidase activity and BBB permeability compared to the endogenous activator and previous peptidomimetic activators. They show reasonable plasma and brain protein binding, improved metabolic stability, and independence of P-gp-mediated efflux. In healthy animals, they rapidly distribute into brains and reach peak levels of 18.69% and 12.10% injected dose (ID)/ml at 10 min. After 4 h, their total brain concentrations remain 7.78 and 12.34 times higher than their A50(minimal concentration required for enhancing 50% peptidase activity). Moreover, the ipsilateral hemispheres of stroke animals show comparable uptake to the corresponding contralateral hemispheres and healthy brains. CONCLUSIONS This study provides essential details about the pharmacokinetic properties of a new generation of potent non-peptidomimetic Nln activators with high BBB permeability and warrants the future development of these agents as potential neuroprotective pharmaceutics for stroke treatment.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Shirisha Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Shikha Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Aarfa Queen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Shiva Hadi Esfahani
- Department of Foundational Medical Studies, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
- Laboratory for Neurodegenerative Disease & Drug Discovery, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Dhavalkumar Patel
- Office of Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Vardan T Karamyan
- Department of Foundational Medical Studies, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
- Laboratory for Neurodegenerative Disease & Drug Discovery, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
5
|
Mamada H, Takahashi M, Ogino M, Nomura Y, Uesawa Y. Predictive Models Based on Molecular Images and Molecular Descriptors for Drug Screening. ACS OMEGA 2023; 8:37186-37195. [PMID: 37841172 PMCID: PMC10568689 DOI: 10.1021/acsomega.3c04073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023]
Abstract
Various toxicity and pharmacokinetic evaluations as screening experiments are needed at the drug discovery stage. Currently, to reduce the use of animal experiments and developmental expenses, the development of high-performance predictive models based on quantitative structure-activity relationship analysis is desired. From these evaluation targets, we selected 50% lethal dose (LD50), blood-brain barrier penetration (BBBP), and the clearance (CL) pathway for this investigation and constructed predictive models for each target using 636-11,886 compounds. First, we constructed predictive models using the DeepSnap-deep learning (DL) method and images of compounds as features. The calculated area under the curve (AUC) and balanced accuracy (BAC) were, respectively, 0.887 and 0.818 for LD50, 0.893 and 0.824 for BBBP, and 0.883 and 0.763 for the CL pathway. Next, molecular descriptors (MDs) of compounds were calculated using Molecular Operating Environment, alvaDesc, and ADMET Predictor to construct predictive models using the MD-based method. Using these MDs, we constructed predictive models using DataRobot. The calculated AUC and BAC were, respectively, 0.931 and 0.805 for LD50, 0.919 and 0.849 for BBBP, and 0.900 and 0.807 for the CL pathway. In this investigation, we constructed predictive models combining the DeepSnap-DL and MD-based methods. In ensemble models using the mean predictive probability of the DeepSnap-DL and MD-based methods, the calculated AUC and BAC were, respectively, 0.942 and 0.842 for LD50, 0.936 and 0.853 for BBBP, and 0.908 and 0.832 for the CL pathway, with improved predictive performance observed for all variables compared with either single method alone. Moreover, in consensus models that adopted only compounds for which the results of the two methods agreed, the calculated BAC for LD50, BBBP, and the CL pathway were 0.916, 0.918, and 0.847, respectively, indicating higher predictive performance than the ensemble models for all three variables. The predictive models combining the DeepSnap-DL and MD-based methods displayed high predictive performance for LD50, BBBP, and the CL pathway. Therefore, the application of this approach to prediction targets in various drug discovery screenings is expected to accelerate drug discovery.
Collapse
Affiliation(s)
- Hideaki Mamada
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Mari Takahashi
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Mizuki Ogino
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yukihiro Nomura
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yoshihiro Uesawa
- Department
of Medical Molecular Informatics, Meiji
Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-858, Japan
| |
Collapse
|
6
|
Jeong SH, Jang JH, Lee YB. P-glycoprotein mechanical functional analysis using in silico molecular modeling: Pharmacokinetic variability according to ABCB1 c.2677G > T/A genetic polymorphisms. Int J Biol Macromol 2023; 249:126777. [PMID: 37683742 DOI: 10.1016/j.ijbiomac.2023.126777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/24/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
P-glycoprotein (P-gp) is a widely membrane-expressed multi-drug transporter. It is unclear whether the pharmacokinetic diversity of P-gp substrates is highly dependent on ABCB1 polymorphisms encoding P-gp. The purpose of this study is to analyze the mechanistic function of P-gp through in silico molecular modeling and to approach the resolution of controversy over pharmacokinetic differences according to ABCB1 polymorphisms. P-gp conformations of apo, ligand-docked, and outward-facing states can be modeled based on structural information of human P-gp. And polymorphic P-gp structures were constructed through homology modeling. ABCB1 c.2677G > T/A (Ala893Ser/Thr), did not correspond to P-gp's nucleotide-binding-domain (NBD) or drug-binding-pocket (DBP) or involve mechanical conformational changes. Although amino acid substitution by ABCB1 c.2677G > T/A caused a 30 % increased strain in an α-helix hinge between the NBD and DBP in P-gp's internal tunnel, there were no overall structural changes compared to wild-type. ABCB1 c.2677G > T/A may increase torsional energy, impacting conformational change rate, but this does not significantly affect P-gp's general functioning. Fexofenadine docking into P-gp's DBP explained the substrate interaction, but no effect by ABCB1 c.2677G > T/A was confirmed. Our findings provide additional insights useful in resolving the debate about the influence of ABCB1 polymorphisms on the interindividual pharmacokinetic variability of P-gp substrates.
Collapse
Affiliation(s)
- Seung-Hyun Jeong
- Department of Pharmacy, College of Pharmacy, Sunchon National University, Suncheon-si 57922, Republic of Korea; College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon-si 57922, Republic of Korea
| | - Ji-Hun Jang
- Department of Pharmacy, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong-Bok Lee
- Department of Pharmacy, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
7
|
Adachi A, Yamashita T, Kanaya S, Kosugi Y. Ensemble Machine Learning Approaches Based on Molecular Descriptors and Graph Convolutional Networks for Predicting the Efflux Activities of MDR1 and BCRP Transporters. AAPS J 2023; 25:88. [PMID: 37700207 DOI: 10.1208/s12248-023-00853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/19/2023] [Indexed: 09/14/2023] Open
Abstract
Multidrug resistance (MDR1) and breast cancer resistance protein (BCRP) play important roles in drug absorption and distribution. Computational prediction of substrates for both transporters can help reduce time in drug discovery. This study aimed to predict the efflux activity of MDR1 and BCRP using multiple machine learning approaches with molecular descriptors and graph convolutional networks (GCNs). In vitro efflux activity was determined using MDR1- and BCRP-expressing cells. Predictive performance was assessed using an in-house dataset with a chronological split and an external dataset. CatBoost and support vector regression showed the best predictive performance for MDR1 and BCRP efflux activities, respectively, of the 25 descriptor-based machine learning methods based on the coefficient of determination (R2). The single-task GCN showed a slightly lower performance than descriptor-based prediction in the in-house dataset. In both approaches, the percentage of compounds predicted within twofold of the observed values in the external dataset was lower than that in the in-house dataset. Multi-task GCN did not show any improvements, whereas multimodal GCN increased the predictive performance of BCRP efflux activity compared with single-task GCN. Furthermore, the ensemble approach of descriptor-based machine learning and GCN achieved the highest predictive performance with R2 values of 0.706 and 0.587 in MDR1 and BCRP, respectively, in time-split test sets. This result suggests that two different approaches to represent molecular structures complement each other in terms of molecular characteristics. Our study demonstrated that predictive models using advanced machine learning approaches are beneficial for identifying potential substrate liability of both MDR1 and BCRP.
Collapse
Affiliation(s)
- Asahi Adachi
- Global DMPK, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
- Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayamacho, Ikoma, Nara, 630-0101, Japan
| | - Tomoki Yamashita
- Global DMPK, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Shigehiko Kanaya
- Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayamacho, Ikoma, Nara, 630-0101, Japan
| | - Yohei Kosugi
- Global DMPK, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
8
|
Santiago-Silva KMD, Camargo P, Felix da Silva Gomes G, Sotero AP, Orsato A, Perez CC, Nakazato G, da Silva Lima CH, Bispo M. In silico approach identified benzoylguanidines as SARS-CoV-2 main protease (M pro) potential inhibitors. J Biomol Struct Dyn 2023; 41:7686-7699. [PMID: 36124832 DOI: 10.1080/07391102.2022.2123396] [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: 03/01/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
The coronavirus disease-2019 (COVID-19) pandemic, caused by the novel coronavirus severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), became the highest public health crisis nowadays. Although the use of approved vaccines for emergency immunization and the reuse of FDA-approved drugs remains at the forefront, the search for new, more selective, and potent drug candidates from synthetic compounds is also a viable alternative to combat this viral disease. In this context, the present study employed a computational virtual screening approach based on molecular docking and molecular dynamics (MD) simulation to identify possible inhibitors for SARS-CoV-2 Mpro (main protease), an important molecular target required for the maturation of the various polyproteins involved in viral replication. The virtual screening approach selected four potential inhibitors against SARS-CoV-2 Mpro. In addition, MD simulation studies revealed changes in the positions of the ligands during the simulations compared to the complex obtained in the molecular docking studies, showing the benzoylguanidines LMed-110 and LMed-136 have a higher affinity for the active site compared to the other structures that tended to leave the active site. Besides, there was a better understanding of the formation and stability of the existing H-bonds in the formed complexes and the energetic contributions to the stability of the target-ligand molecular complexes. Finally, the in silico prediction of the ADME profile suggested that LMed-136 has drug-like characteristics and good pharmacokinetic properties. Therefore, from the present study, it can be suggested that these structures can inhibit SARS-CoV-2 Mpro. Nevertheless, further studies are needed in vitro assays to investigate the antiviral properties of these structures against SARS-CoV-2.
Collapse
Affiliation(s)
- Kaio Maciel de Santiago-Silva
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Priscila Camargo
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Gabriel Felix da Silva Gomes
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Ana Paula Sotero
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Orsato
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Carla Cristina Perez
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Gerson Nakazato
- Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Camilo Henrique da Silva Lima
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelle Bispo
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
9
|
Sharma S, Zhang Y, Akter KA, Nozohouri S, Archie SR, Patel D, Villalba H, Abbruscato T. Permeability of Metformin across an In Vitro Blood-Brain Barrier Model during Normoxia and Oxygen-Glucose Deprivation Conditions: Role of Organic Cation Transporters (Octs). Pharmaceutics 2023; 15:pharmaceutics15051357. [PMID: 37242599 DOI: 10.3390/pharmaceutics15051357] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Our lab previously established that metformin, a first-line type two diabetes treatment, activates the Nrf2 pathway and improves post-stroke recovery. Metformin's brain permeability value and potential interaction with blood-brain barrier (BBB) uptake and efflux transporters are currently unknown. Metformin has been shown to be a substrate of organic cationic transporters (Octs) in the liver and kidneys. Brain endothelial cells at the BBB have been shown to express Octs; thus, we hypothesize that metformin uses Octs for its transport across the BBB. We used a co-culture model of brain endothelial cells and primary astrocytes as an in vitro BBB model to conduct permeability studies during normoxia and hypoxia using oxygen-glucose deprivation (OGD) conditions. Metformin was quantified using a highly sensitive LC-MS/MS method. We further checked Octs protein expression using Western blot analysis. Lastly, we completed a plasma glycoprotein (P-GP) efflux assay. Our results showed that metformin is a highly permeable molecule, uses Oct1 for its transport, and does not interact with P-GP. During OGD, we found alterations in Oct1 expression and increased permeability for metformin. Additionally, we showed that selective transport is a key determinant of metformin's permeability during OGD, thus, providing a novel target for improving ischemic drug delivery.
Collapse
Affiliation(s)
- Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Yong Zhang
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Khondker Ayesha Akter
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Heidi Villalba
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
10
|
Nukaga T, Takemura A, Endo Y, Uesawa Y, Ito K. Estimating drug-induced liver injury risk by in vitro molecular initiation response and pharmacokinetic parameters for during early drug development. Toxicol Res (Camb) 2023; 12:86-94. [PMID: 36866207 PMCID: PMC9972805 DOI: 10.1093/toxres/tfac083] [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: 06/12/2022] [Revised: 11/02/2022] [Accepted: 12/02/2022] [Indexed: 01/10/2023] Open
Abstract
Drug-induced liver injury (DILI) is a major factor influencing new drug withdrawal; therefore, an appropriate toxicity assessment at the preclinical stage is required. Previous in silico models have been established using compound information listed in large data sources, thereby limiting the DILI risk prediction for new drugs. Herein, we first constructed a model to predict DILI risk based on a molecular initiating event (MIE) predicted by quantitative structure-activity relationships, admetSAR parameters (e.g. cytochrome P450 reactivity, plasma protein binding, and water-solubility), and clinical information (maximum daily dose [MDD] and reactive metabolite [RM]) for 186 compounds. The accuracy of the models using MIE, MDD, RM, and admetSAR alone were 43.2%, 47.3%, 77.0%, and 68.9%, while the "predicted MIE + admetSAR + MDD + RM" model's accuracy was 75.7%. The contribution of MIE to the overall prediction accuracy was little effect or rather worsening it. However, it was considered that MIE was a valuable parameter and that it contributed to detect high DILI risk compounds in the early development stage. We next examined the effect of stepwise changes in MDD on altering the DILI risk and estimating the maximum safety dose (MSD) for clinical use based on structural information, admetSAR, and MIE parameters because it is important to estimate the dose that could prevent the DILI onset in clinical conditions. Low-MSD compounds might increase the DILI risk, as these compounds were classified as "most-DILI concern" at low doses. In conclusion, MIE parameters were especially useful to check the DILI concern compounds and to prevent the underestimation of DILI risk in the early stage of drug development.
Collapse
Affiliation(s)
- Takumi Nukaga
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Akinori Takemura
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Yuka Endo
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
11
|
Mora Lagares L, Novič M. Recent Advances on P-Glycoprotein (ABCB1) Transporter Modelling with In Silico Methods. Int J Mol Sci 2022; 23:ijms232314804. [PMID: 36499131 PMCID: PMC9740644 DOI: 10.3390/ijms232314804] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
ABC transporters play a critical role in both drug bioavailability and toxicity, and with the discovery of the P-glycoprotein (P-gp), this became even more evident, as it plays an important role in preventing intracellular accumulation of toxic compounds. Over the past 30 years, intensive studies have been conducted to find new therapeutic molecules to reverse the phenomenon of multidrug resistance (MDR) ), that research has found is often associated with overexpression of P-gp, the most extensively studied drug efflux transporter; in MDR, therapeutic drugs are prevented from reaching their targets due to active efflux from the cell. The development of P-gp inhibitors is recognized as a good way to reverse this type of MDR, which has been the subject of extensive studies over the past few decades. Despite the progress made, no effective P-gp inhibitors to reverse multidrug resistance are yet on the market, mainly because of their toxic effects. Computational studies can accelerate this process, and in silico models such as QSAR models that predict the activity of compounds associated with P-gp (or analogous transporters) are of great value in the early stages of drug development, along with molecular modelling methods, which provide a way to explain how these molecules interact with the ABC transporter. This review highlights recent advances in computational P-gp research, spanning the last five years to 2022. Particular attention is given to the use of machine-learning approaches, drug-transporter interactions, and recent discoveries of potential P-gp inhibitors that could act as modulators of multidrug resistance.
Collapse
Affiliation(s)
- Liadys Mora Lagares
- Correspondence: (L.M.L.); (M.N.); Tel.: +386-1-4760-438 (L.M.L.); +386-1-4760-253 (M.N.)
| | - Marjana Novič
- Correspondence: (L.M.L.); (M.N.); Tel.: +386-1-4760-438 (L.M.L.); +386-1-4760-253 (M.N.)
| |
Collapse
|
12
|
Kuroda M, Watanabe R, Esaki T, Kawashima H, Ohashi R, Sato T, Honma T, Komura H, Mizuguchi K. Utilizing public and private sector data to build better machine learning models for the prediction of pharmacokinetic parameters. Drug Discov Today 2022; 27:103339. [PMID: 35973660 DOI: 10.1016/j.drudis.2022.103339] [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: 04/23/2022] [Revised: 07/11/2022] [Accepted: 08/11/2022] [Indexed: 11/20/2022]
Abstract
One solution to compensate for the shortage of publicly available data is to collect more quality-controlled data from the private sector through public-private partnerships. However, several issues must be resolved before implementing such a system. Here, we review the technical aspects of public-private partnerships using our initiative in Japan as an example. In particular, we focus on the procedure for collecting data from multiple private sector companies and building prediction models and discuss how merging public and private sector datasets will help to improve the chemical space coverage and prediction performance. Teaser: Japan's first public-private consortium in pharmacokinetics has incorporated data from multiple pharmaceutical companies to create useful predictive models.
Collapse
Affiliation(s)
- Masataka Kuroda
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Reiko Watanabe
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - Tsuyoshi Esaki
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; The Centre for Data Science Education and Research, Shiga University, 1-1-1, Banba, Hikone, Shiga 522-8522, Japan
| | - Hitoshi Kawashima
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Rikiya Ohashi
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Tomohiro Sato
- RIKEN Center for Biosystems Dynamics Research, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Teruki Honma
- RIKEN Center for Biosystems Dynamics Research, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Komura
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; University Research Administration Centre, Osaka Metropolitan University, 1-2-7, Asahi, Abeno-ku, Osaka 545-0051, Japan
| | - Kenji Mizuguchi
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan.
| |
Collapse
|
13
|
Ma BB, Montgomery AP, Chen B, Kassiou M, Danon JJ. Strategies for targeting the P2Y 12 receptor in the central nervous system. Bioorg Med Chem Lett 2022; 71:128837. [PMID: 35640763 DOI: 10.1016/j.bmcl.2022.128837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 11/28/2022]
Abstract
The purinergic 2Y type 12 receptor (P2Y12R) is a well-known biological target for anti-thrombotic drugs due to its role in platelet aggregation and blood clotting. While the importance of the P2Y12R in the periphery has been known for decades, much less is known about its expression and roles in the central nervous system (CNS), where it is expressed exclusively on microglia - the first responders to brain insults and neurodegeneration. Several seminal studies have shown that P2Y12 is a robust, translatable biomarker for anti-inflammatory and neuroprotective microglial phenotypes in models of degenerative diseases such as multiple sclerosis and Alzheimer's disease. An enduring problem for studying this receptor in vivo, however, is the lack of selective, high-affinity small molecule ligands that can bypass the blood-brain barrier and accumulate in the CNS. In this Digest, we discuss previous attempts by researchers to target the P2Y12R in the CNS and opine on strategies that may be employed to design and assess the suitability of novel P2Y12 ligands for this purpose going forward.
Collapse
Affiliation(s)
- Ben B Ma
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Biling Chen
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan J Danon
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
14
|
Jackson IM, Webb EW, Scott PJ, James ML. In Silico Approaches for Addressing Challenges in CNS Radiopharmaceutical Design. ACS Chem Neurosci 2022; 13:1675-1683. [PMID: 35606334 PMCID: PMC9945852 DOI: 10.1021/acschemneuro.2c00269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Positron emission tomography (PET) is a highly sensitive and versatile molecular imaging modality that leverages radiolabeled molecules, known as radiotracers, to interrogate biochemical processes such as metabolism, enzymatic activity, and receptor expression. The ability to probe specific molecular and cellular events longitudinally in a noninvasive manner makes PET imaging a particularly powerful technique for studying the central nervous system (CNS) in both health and disease. Unfortunately, developing and translating a single CNS PET tracer for clinical use is typically an extremely resource-intensive endeavor, often requiring synthesis and evaluation of numerous candidate molecules. While existing in vitro methods are beginning to address the challenge of derisking molecules prior to costly in vivo PET studies, most require a significant investment of resources and possess substantial limitations. In the context of CNS drug development, significant time and resources have been invested into the development and optimization of computational methods, particularly involving machine learning, to streamline the design of better CNS therapeutics. However, analogous efforts developed and validated for CNS radiotracer design are conspicuously limited. In this Perspective, we overview the requirements and challenges of CNS PET tracer design, survey the most promising computational methods for in silico CNS drug design, and bridge these two areas by discussing the potential applications and impact of computational design tools in CNS radiotracer design.
Collapse
Affiliation(s)
- Isaac M. Jackson
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - E. William Webb
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109
| | - Peter J.H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109;,Corresponding Authors: Peter J. H. Scott − Department of Radiology, University of Michigan, Ann Arbor, MI 48109, United States; , Michelle L. James − Departments of Radiology, and Neurology & Neurological Sciences, 1201 Welch Rd., P-206, Stanford, CA 94305-5484, United States;
| | - Michelle L. James
- Department of Radiology, Stanford University, Stanford, CA 94305;,Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94304.,Corresponding Authors: Peter J. H. Scott − Department of Radiology, University of Michigan, Ann Arbor, MI 48109, United States; , Michelle L. James − Departments of Radiology, and Neurology & Neurological Sciences, 1201 Welch Rd., P-206, Stanford, CA 94305-5484, United States;
| |
Collapse
|
15
|
Mamada H, Nomura Y, Uesawa Y. Novel QSAR Approach for a Regression Model of Clearance That Combines DeepSnap-Deep Learning and Conventional Machine Learning. ACS OMEGA 2022; 7:17055-17062. [PMID: 35647436 PMCID: PMC9134387 DOI: 10.1021/acsomega.2c00261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/29/2022] [Indexed: 05/03/2023]
Abstract
The toxicity, absorption, distribution, metabolism, and excretion properties of some targets are difficult to predict by quantitative structure-activity relationship analysis. Therefore, there is a need for a new prediction method that performs well for these targets. The aim of this study was to develop a new regression model of rat clearance (CL). We constructed a regression model using 1545 in-house compounds for which we had rat CL data. Molecular descriptors were calculated using molecular operating environment, alvaDesc, and ADMET Predictor software. The classification model of DeepSnap and Deep Learning (DeepSnap-DL) with images of the three-dimensional chemical structures of compounds as features was constructed, and the prediction probabilities for each compound were calculated. For molecular descriptor-based methods that use molecular descriptors and conventional machine learning algorithms selected by DataRobot, the correlation coefficient (R 2) and root mean square error (RMSE) were 0.625-0.669 and 0.295-0.318, respectively. We combined molecular descriptors and prediction probability of DeepSnap-DL as features and developed a novel regression method we called the combination model. In the combination model with these two types of features and conventional algorithms selected by DataRobot, R 2 and RMSE were 0.710-0.769 and 0.247-0.278, respectively. This finding shows that the combination model performed better than molecular descriptor-based methods. Our combination model will contribute to the design of more rational compounds for drug discovery. This method may be applicable not only to rat CL but also to other pharmacokinetic and pharmacological activity and toxicity parameters; therefore, applying it to other parameters may help to accelerate drug discovery.
Collapse
Affiliation(s)
- Hideaki Mamada
- Department
of Medical Molecular Informatics, Meiji
Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yukihiro Nomura
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical
Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yoshihiro Uesawa
- Department
of Medical Molecular Informatics, Meiji
Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan
- . Phone: +81-42-495-8983. Fax: +81-42-495-8983
| |
Collapse
|
16
|
Dziadziuszko R, Hung T, Wang K, Choeurng V, Drilon A, Doebele RC, Barlesi F, Wu C, Dennis L, Skoletsky J, Woodhouse R, Li M, Chang C, Simmons B, Riehl T, Wilson TR. Pre- and post-treatment blood-based genomic landscape of patients with ROS1 or NTRK fusion-positive solid tumours treated with entrectinib. Mol Oncol 2022; 16:2000-2014. [PMID: 35338679 PMCID: PMC9120896 DOI: 10.1002/1878-0261.13214] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/25/2022] [Accepted: 03/24/2022] [Indexed: 11/08/2022] Open
Abstract
Genomic tumour profiling informs targeted treatment options. Entrectinib is a tyrosine kinase inhibitor with efficacy in NTRK fusion-positive (-fp) solid tumours and ROS1-fp non-small cell lung cancer. FoundationOne® Liquid CDx (F1L CDx), a non-invasive in vitro next-generation sequencing (NGS)-based diagnostic, detects genomic alterations in plasma circulating tumour DNA (ctDNA). We evaluated the clinical validity of F1L CDx as an aid in identifying patients with NTRK-fp or ROS1-fp tumours and assessed the genomic landscape pre- and post-entrectinib treatment. Among evaluable pre-treatment clinical samples (N = 85), positive percentage agreements between F1L CDx and clinical trial assays (CTAs) were 47.4% (NTRK fusions) and 64.5% (ROS1 fusions); positive predictive value was 100% for both. The objective response rate for CTA+ F1L CDx+ patients was 72.2% in both cohorts. The median duration of response significantly differed between F1L CDx+ and F1L CDx- samples in ROS1-fp (5.6 vs. 17.3 months) but not NTRK-fp (9.2 vs. 12.9 months) patients. Fifteen acquired resistance mutations were detected. We conclude that F1L CDx is a clinically valid complement to tissue-based testing to identify patients who may benefit from entrectinib and those with acquired resistance mutations associated with disease progression.
Collapse
Affiliation(s)
- Rafal Dziadziuszko
- Department of Oncology and RadiotherapyMedical University of GdańskGdańskPoland
| | - Tiffany Hung
- Oncology Biomarker DepartmentGenentech, Inc.South San FranciscoCAUSA
| | - Kun Wang
- BiostatisticsFoundation Medicine Inc.CambridgeMAUSA
| | - Voleak Choeurng
- Oncology BiostatisticsGenentech, Inc.South San FranciscoCAUSA
| | - Alexander Drilon
- Early Drug Development ServiceMemorial Sloan Kettering Cancer Center, and Weill Cornell Medical CollegeNew YorkNYUSA
| | | | - Fabrice Barlesi
- The National Centre for Scientific Research (CNRS)The National Institute of Health and Medical Research (INSERM)Aix Marseille UniversityMarseilleFrance
- Medical OncologyGustave RoussyVillejuifFrance
| | - Charlie Wu
- Oncology Biomarker DepartmentGenentech, Inc.South San FranciscoCAUSA
| | - Lucas Dennis
- Franchise DevelopmentFoundation Medicine Inc.CambridgeMAUSA
| | - Joel Skoletsky
- Companion Diagnostics DevelopmentFoundation Medicine Inc.CambridgeMAUSA
| | - Ryan Woodhouse
- Regulatory AffairsFoundation Medicine Inc.CambridgeMAUSA
| | - Meijuan Li
- Biometrics and BiomarkersFoundation Medicine Inc.CambridgeMAUSA
| | - Ching‐Wei Chang
- Oncology BiostatisticsGenentech, Inc.South San FranciscoCAUSA
| | - Brian Simmons
- Product Development OncologyGenentech, Inc.South San FranciscoCAUSA
| | - Todd Riehl
- Product Development OncologyGenentech, Inc.South San FranciscoCAUSA
| | - Timothy R. Wilson
- Oncology Biomarker DepartmentGenentech, Inc.South San FranciscoCAUSA
| |
Collapse
|
17
|
In-Vivo and Ex-Vivo Brain Uptake Studies of Peptidomimetic Neurolysin Activators in Healthy and Stroke Animals. Pharm Res 2022; 39:1587-1598. [PMID: 35239135 DOI: 10.1007/s11095-022-03218-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/25/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Neurolysin (Nln) is a peptidase that functions to preserve the brain following ischemic stroke by hydrolyzing various neuropeptides. Nln activation has emerged as an attractive drug discovery target for treatment of ischemic stroke. Among first-in-class peptidomimetic Nln activators, we selected three lead compounds (9d, 10c, 11a) for quantitative pharmacokinetic analysis to provide valuable information for subsequent preclinical development. METHODS Pharmacokinetic profile of these compounds was studied in healthy and ischemic stroke-induced mice after bolus intravenous administration. Brain concentration and brain uptake clearance (Kin) was calculated from single time point analysis. The inter-relationship between LogP with in-vitro and in-vivo permeability was studied to determine CNS penetration. Brain slice uptake method was used to study tissue binding, whereas P-gp-mediated transport was evaluated to understand the potential brain efflux of these compounds. RESULTS According to calculated parameters, all three compounds showed a detectable amount in the brain after intravenous administration at 4 mg/kg; however, 11a had the highest brain concentration and brain uptake clearance. A strong correlation was documented between in-vitro and in-vivo permeability data. The efflux ratio of 10c was ~6-fold higher compared to 11a and correlated well with its lower Kin value. In experimental stroke animals, the Kin of 11a was significantly higher in ischemic vs. contralateral and intact hemispheres, though it remained below its A50 value required to activate Nln. CONCLUSIONS Collectively, these preclinical pharmacokinetic studies reveal promising BBB permeability of 11a and indicate that it can serve as an excellent lead for developing improved drug-like Nln activators.
Collapse
|
18
|
Rathod S, Desai H, Patil R, Sarolia J. Non-ionic Surfactants as a P-Glycoprotein(P-gp) Efflux Inhibitor for Optimal Drug Delivery-A Concise Outlook. AAPS PharmSciTech 2022; 23:55. [PMID: 35043278 DOI: 10.1208/s12249-022-02211-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Significant research efforts have been devoted to unraveling the mystery of P-glycoprotein(P-gp) in drug delivery applications. The efflux membrane transporter P-gp is widely distributed in the body and accountable for restricting drug absorption and bioavailability. For these reasons, it is the primary cause of developing multidrug resistance (MDR) in most drug delivery applications. Therefore, P-gp inhibitors must be explored to address MDR and the low bioavailability of therapeutic substrates. Several experimental models in kinetics and dynamic studies identified the sensitivity of drug molecules and excipients as a P-gp inhibitor. In this review, we aimed to emphasize nonionic surface-active agents for effective reversal of P-gp inhibition. As it is inert, non-toxic, noncharged, and quickly reaching the cytosolic lipid membrane (the point of contact with P-gp efflux protein) enables it to be more efficient as P-gp inhibitors. Moreover, nonionic surfactant improves drug absorption and bioavailability through the various mechanism, involving (i) association of drug with surfactant improves solubilization, facilitating its cell penetration and absorption; (ii) weakening the lateral membrane packing density, facilitating the passive drug influx; and (iii) inhibition of the ATP binding cassette of transporter P-glycoprotein. The application of nonionic surfactant as P-gp inhibitors is well established and supported by various experiments. Altogether, herein, we have primarily focused on various nonionic surfactants and their development strategies to conquer the MDR-causing effects of P-gp efflux protein in drug delivery. Graphical Abstract.
Collapse
|
19
|
Parvizpour S, Masoudi-Sobhanzadeh Y, Pourseif MM, Barzegari A, Razmara J, Omidi Y. Pharmacoinformatics-based phytochemical screening for anticancer impacts of yellow sweet clover, Melilotus officinalis (Linn.) Pall. Comput Biol Med 2021; 138:104921. [PMID: 34656871 DOI: 10.1016/j.compbiomed.2021.104921] [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: 07/11/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022]
Abstract
To date, much attention has been paid to phytochemicals because of their diverse pharmacological effects on a variety of diseases such as cancer. In this regard, computer-aided drug design, as a cost- and time-effective approach, is primarily applied to investigate the drug candidates before their further costly in vitro and in vivo experimental evaluations. Accordingly, different signaling pathways and proteins can be targeted using such strategies. As a key protein for the initiation of eukaryotic DNA replication, mini-chromosome maintenance complex component 7 (MCM7) overexpression is related to the initiation and progression of aggressive malignancies. The current study was conducted to identify new potential natural compounds from the yellow sweet clover, Melilotus officinalis (Linn.) Pall, by examining the potential of 40 isolated phytochemicals against MCM7 protein. A structure-based pharmacophore model to the protein active site cavity was generated and followed by virtual screening and molecular docking. Overall, four compounds were selected for further evaluation based on their binding affinities. Our analyses revealed that two novel compounds, namely rosmarinic acid (PubChem CID:5281792) and melilotigenin (PubChem CID:14059499) might be druggable and offer safe usage in human. The stability of these two protein-ligand complex structures was confirmed through molecular dynamics simulation. The findings of this study reveal the potential of these two phytochemicals to serve as anticancer agents, while further pharmacological experiments are required to confirm their effectiveness against human cancers.
Collapse
Affiliation(s)
- Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yosef Masoudi-Sobhanzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Razmara
- Department of Computer Science, Faculty of Mathematics, Statistics, and Computer Science, University of Tabriz, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, United States.
| |
Collapse
|
20
|
Xiong B, Wang Y, Chen Y, Xing S, Liao Q, Chen Y, Li Q, Li W, Sun H. Strategies for Structural Modification of Small Molecules to Improve Blood-Brain Barrier Penetration: A Recent Perspective. J Med Chem 2021; 64:13152-13173. [PMID: 34505508 DOI: 10.1021/acs.jmedchem.1c00910] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the development of central nervous system (CNS) drugs, the blood-brain barrier (BBB) restricts many drugs from entering the brain to exert therapeutic effects. Although many novel delivery methods of large molecule drugs have been designed to assist transport, small molecule drugs account for the vast majority of the CNS drugs used clinically. From this perspective, we review studies from the past five years that have sought to modify small molecules to increase brain exposure. Medicinal chemists make it easier for small molecules to cross the BBB by improving diffusion, reducing efflux, and activating carrier transporters. On the basis of their excellent work, we summarize strategies for structural modification of small molecules to improve BBB penetration. These strategies are expected to provide a reference for the future development of small molecule CNS drugs.
Collapse
Affiliation(s)
- Baichen Xiong
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuanyuan Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinghong Liao
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Qi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.,School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Wei Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
21
|
Mamada H, Nomura Y, Uesawa Y. Prediction Model of Clearance by a Novel Quantitative Structure-Activity Relationship Approach, Combination DeepSnap-Deep Learning and Conventional Machine Learning. ACS OMEGA 2021; 6:23570-23577. [PMID: 34549154 PMCID: PMC8444299 DOI: 10.1021/acsomega.1c03689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/23/2021] [Indexed: 05/19/2023]
Abstract
Some targets predicted by machine learning (ML) in drug discovery remain a challenge because of poor prediction. In this study, a new prediction model was developed and rat clearance (CL) was selected as a target because it is difficult to predict. A classification model was constructed using 1545 in-house compounds with rat CL data. The molecular descriptors calculated by Molecular Operating Environment (MOE), alvaDesc, and ADMET Predictor software were used to construct the prediction model. In conventional ML using 100 descriptors and random forest selected by DataRobot, the area under the curve (AUC) and accuracy (ACC) were 0.883 and 0.825, respectively. Conversely, the prediction model using DeepSnap and Deep Learning (DeepSnap-DL) with compound features as images had AUC and ACC of 0.905 and 0.832, respectively. We combined the two models (conventional ML and DeepSnap-DL) to develop a novel prediction model. Using the ensemble model with the mean of the predicted probabilities from each model improved the evaluation metrics (AUC = 0.943 and ACC = 0.874). In addition, a consensus model using the results of the agreement between classifications had an increased ACC (0.959). These combination models with a high level of predictive performance can be applied to rat CL as well as other pharmacokinetic parameters, pharmacological activity, and toxicity prediction. Therefore, these models will aid in the design of more rational compounds for the development of drugs.
Collapse
Affiliation(s)
- Hideaki Mamada
- Department
of Medical Molecular Informatics, Meiji
Pharmaceutical University, 2-522-1, Noshio, Kiyose-shi, Tokyo 204-858, Japan
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1, Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yukihiro Nomura
- Drug
Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1, Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yoshihiro Uesawa
- Department
of Medical Molecular Informatics, Meiji
Pharmaceutical University, 2-522-1, Noshio, Kiyose-shi, Tokyo 204-858, Japan
- . Tel.: +81-42-495-8983. Fax: +81-42-495-8983
| |
Collapse
|
22
|
Hanafy AS, Dietrich D, Fricker G, Lamprecht A. Blood-brain barrier models: Rationale for selection. Adv Drug Deliv Rev 2021; 176:113859. [PMID: 34246710 DOI: 10.1016/j.addr.2021.113859] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023]
Abstract
Brain delivery is a broad research area, the outcomes of which are far hindered by the limited permeability of the blood-brain barrier (BBB). Over the last century, research has been revealing the BBB complexity and the crosstalk between its cellular and molecular components. Pathologically, BBB alterations may precede as well as be concomitant or lead to brain diseases. To simulate the BBB and investigate options for drug delivery, several in vitro, in vivo, ex vivo, in situ and in silico models are used. Hundreds of drug delivery vehicles successfully pass preclinical trials but fail in clinical settings. Inadequate selection of BBB models is believed to remarkably impact the data reliability leading to unsatisfactory results in clinical trials. In this review, we suggest a rationale for BBB model selection with respect to the addressed research question and downstream applications. The essential considerations of an optimal BBB model are discussed.
Collapse
Affiliation(s)
- Amira Sayed Hanafy
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Dirk Dietrich
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany.
| |
Collapse
|
23
|
Multi-Target Actions of Acridones from Atalantia monophylla towards Alzheimer's Pathogenesis and Their Pharmacokinetic Properties. Pharmaceuticals (Basel) 2021; 14:ph14090888. [PMID: 34577588 PMCID: PMC8470973 DOI: 10.3390/ph14090888] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Ten acridones isolated from Atalantia monophylla were evaluated for effects on Alzheimer’s disease pathogenesis including antioxidant effects, acetylcholinesterase (AChE) inhibition, prevention of beta-amyloid (Aβ) aggregation and neuroprotection. To understand the mechanism, the type of AChE inhibition was investigated in vitro and binding interactions between acridones and AChE or Aβ were explored in silico. Drug-likeness and ADMET parameters were predicted in silico using SwissADME and pKCSM programs, respectively. All acridones showed favorable drug-likeness and possessed multifunctional activities targeting AChE function, Aβ aggregation and oxidation. All acridones inhibited AChE in a mixed-type manner and bound AChE at both catalytic anionic and peripheral anionic sites. In silico analysis showed that acridones interfered with Aβ aggregation by interacting at the central hydrophobic core, C-terminal hydrophobic region, and the key residues 41 and 42. Citrusinine II showed potent multifunctional action with the best ADMET profile and could alleviate neuronal cell damage induced by hydrogen peroxide and Aβ1-42 toxicity.
Collapse
|
24
|
Santana R, Onieva E, Zuluaga R, Duardo-Sánchez A, Gañán P. The Role of Machine Learning in Centralized Authorization Process of Nanomedicines in European Union. Curr Top Med Chem 2021; 21:828-838. [PMID: 33745436 DOI: 10.2174/1568026621666210319101847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/12/2020] [Accepted: 12/31/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Machine Learning (ML) has experienced an increasing use, given the possibilities to expand the scientific knowledge of different disciplines, such as nanotechnology. This has allowed the creation of Cheminformatic models capable of predicting biological activity and physicochemical characteristics of new components with high success rates in training and test partitions. Given the current gaps of scientific knowledge and the need for efficient application of medicines products law, this paper analyzes the position of regulators for marketing medicinal nanoproducts in the European Union and the role of ML in the authorization process. METHODS In terms of methodology, a dogmatic study of the European regulation and the guidance of the European Medicine Agency on the use of predictive models for nanomaterials was carried out. The study has, as the framework of reference, the European Regulation 726/2004 and has focused on the analysis of how ML processes are contemplated in the regulations. RESULTS As a result, we present a discussion of the information that must be provided for every case for simulation methods. The results show a favorable and flexible position for the development of the use of predictive models to complement the applicant's information. CONCLUSION It is concluded that Machine Learning has the capacity to help improve the application of nanotechnology medicine products regulation. Future regulations should promote this kind of information given the advanced state of the art in terms of algorithms that are able to build accurate predictive models. This especially applies to methods, such as Perturbation Theory Machine Learning (PTML), given that it is aligned with principles promoted by the standards of Organization for Economic Co-operation and Development (OECD), European Union regulations, and European Authority Medicine. To our best knowledge, this is the first study focused on nanotechnology medicine products and machine learning used to support technical European public assessment reports (EPAR) for complementary information.
Collapse
Affiliation(s)
- Ricardo Santana
- DeustoTech-Fundacion Deusto, Avda. Universidades, 24,48007 Bilbao, Spain
| | - Enrique Onieva
- DeustoTech-Fundacion Deusto, Avda. Universidades, 24,48007 Bilbao, Spain
| | - Robin Zuluaga
- Facultad de Ingeniería Agroindustrial, Universidad Pontificia Bolivariana UPB050031, Medellin, Colombia
| | - Aliuska Duardo-Sánchez
- Department of Public Law, Law and the Human Genome Research Group, University of the Basque Country UPV/EHU 48940, Leioa, Biscay, Spain
| | - Piedad Gañán
- Facultad de Ingenieria Quimica, Universidad Pontificia Bolivariana UPB050031, Medellin, Colombia
| |
Collapse
|
25
|
The effect of hypergravity in intestinal permeability of nanoformulations and molecules. Eur J Pharm Biopharm 2021; 163:38-48. [PMID: 33785416 DOI: 10.1016/j.ejpb.2021.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 11/22/2022]
Abstract
The oral administration of drugs remains a challenge due to rapid enzymatic degradation and minimal absorption in the gastrointestinal tract. Mechanical forces, namely hypergravity, can interfere with cellular integrity and drug absorption, and there is no study describing its influence in the intestinal permeability. In this work, it was studied the effect of hypergravity on intestinal Caco-2 cells and its influence in the intestinal permeability of different nanoformulations and molecules. It was shown that the cellular metabolic activity and integrity were maintained after exposure to different gravity-levels (g-levels). Expression of important drug transporters and tight junctions' proteins was evaluated and, most proteins demonstrated a switch of behavior in their expression. Furthermore, paracellular transport of FITC-Dextran showed to significantly increase with hypergravity, which agrees with the decrease of transepithelial electrical resistance and the increase of claudin-2 at higher g-levels. The diffusion of camptothecin released from polymeric micelles revealed a significant decrease, which agrees with the increased expression of the P-gp observed with the increase in g-levels, responsible for pumping this drug out. The neonatal Fc receptor-mediated transport of albumin-functionalized nanoparticles loaded with insulin showed no significant changes when increasing the g-levels. Thus, this study supports the effect of hypergravity on intestinal permeability is dependent on the molecule studied and the mechanism by which it is absorbed in the intestine.
Collapse
|
26
|
Watanabe R, Esaki T, Ohashi R, Kuroda M, Kawashima H, Komura H, Natsume-Kitatani Y, Mizuguchi K. Development of an In Silico Prediction Model for P-glycoprotein Efflux Potential in Brain Capillary Endothelial Cells toward the Prediction of Brain Penetration. J Med Chem 2021; 64:2725-2738. [PMID: 33619967 DOI: 10.1021/acs.jmedchem.0c02011] [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: 01/15/2023]
Abstract
Developing in silico models to predict the brain penetration of drugs remains a challenge owing to the intricate involvement of multiple transport systems in the blood brain barrier, and the necessity to consider a combination of multiple pharmacokinetic parameters. P-glycoprotein (P-gp) is one of the most important transporters affecting the brain penetration of drugs. Here, we developed an in silico prediction model for P-gp efflux potential in brain capillary endothelial cells (BCEC). Using the representative values of P-gp net efflux ratio in BCEC, we proposed a novel prediction system for brain-to-plasma concentration ratio (Kp,brain) and unbound brain-to-plasma concentration ratio (Kp,uu,brain) of P-gp substrates. We validated the proposed prediction system using newly acquired experimental brain penetration data of 28 P-gp substrates. Our system improved the predictive accuracy of brain penetration of drugs using only chemical structure information compared with that of previous studies.
Collapse
Affiliation(s)
- Reiko Watanabe
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Tsuyoshi Esaki
- The Center for Data Science Education and Research, Shiga University, Hikone, Shiga 522-8522, Japan
| | - Rikiya Ohashi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Masataka Kuroda
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Hitoshi Kawashima
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Hiroshi Komura
- URA Center, Osaka City University, Osaka 545-0051, Japan
| | - Yayoi Natsume-Kitatani
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of In-Silico Drug Design, Center of Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| |
Collapse
|
27
|
Mughal H, Wang H, Zimmerman M, Paradis MD, Freundlich JS. Random Forest Model Prediction of Compound Oral Exposure in the Mouse. ACS Pharmacol Transl Sci 2021; 4:338-343. [PMID: 33615183 DOI: 10.1021/acsptsci.0c00197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Indexed: 11/29/2022]
Abstract
An early hurdle in the optimization of small-molecule chemical probes and drug discovery entities is the attainment of sufficient exposure in the mouse via oral administration of the compound. While computational approaches have attempted to predict molecular properties related to the mouse pharmacokinetic (PK) profile, we present herein a machine learning approach to specifically predict the oral exposure of a compound as measured in the mouse snapshot PK assay. A random forest workflow was found to produce the best cross-validation and external test set statistics after processing of the input data set and optimization of model features. The modeling approach should be useful to the chemical biology and drug discovery communities to predict this key molecular property and afford chemical entities of translational significance.
Collapse
Affiliation(s)
- Haseeb Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Han Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Marc D Paradis
- Holdings & Ventures, Northwell Health, Manhasset, New York 11030, United States
| | - Joel S Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States.,Division of Infectious Disease, Department of Medicine and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States
| |
Collapse
|
28
|
Predicting blood-to-plasma concentration ratios of drugs from chemical structures and volumes of distribution in humans. Mol Divers 2021; 25:1261-1270. [PMID: 33569705 PMCID: PMC8342319 DOI: 10.1007/s11030-021-10186-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/18/2021] [Indexed: 11/05/2022]
Abstract
Abstract Despite their importance in determining the dosing regimen of drugs in the clinic, only a few studies have investigated methods for predicting blood-to-plasma concentration ratios (Rb). This study established an Rb prediction model incorporating typical human pharmacokinetics (PK) parameters. Experimental Rb values were compiled for 289 compounds, offering reliable predictions by expanding the applicability domain. Notably, it is the largest list of Rb values reported so far. Subsequently, human PK parameters calculated from plasma drug concentrations, including the volume of distribution (Vd), clearance, mean residence time, and plasma protein binding rate, as well as 2702 kinds of molecular descriptors, were used to construct quantitative structure–PK relationship models for Rb. Among the evaluated PK parameters, logVd correlated best with Rb (correlation coefficient of 0.47). Thus, in addition to molecular descriptors selected by XGBoost, logVd was employed to construct the prediction models. Among the analyzed algorithms, artificial neural networks gave the best results. Following optimization using six molecular descriptors and logVd, the model exhibited a correlation coefficient of 0.64 and a root-mean-square error of 0.205, which were superior to those previously reported for other Rb prediction methods. Since Vd values and chemical structures are known for most medications, the Rb prediction model described herein is expected to be valuable in clinical settings. Graphical abstract ![]()
Supplementary informations The online version of this article (10.1007/s11030-021-10186-7) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Fischer H, Senn C, Ullah M, Cantrill C, Schuler F, Yu L. Calculation of an Apical Efflux Ratio from P-Glycoprotein (P-gp) In Vitro Transport Experiments Shows an Improved Correlation with In Vivo Cerebrospinal Fluid Measurements in Rats: Impact on P-gp Screening and Compound Optimization. J Pharmacol Exp Ther 2020; 376:322-329. [PMID: 33288523 DOI: 10.1124/jpet.120.000158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/02/2020] [Indexed: 11/22/2022] Open
Abstract
P-glycoprotein (P-gp) is a major blood-brain barrier (BBB) efflux transporter. In vitro approaches, including bidirectional efflux ratio (ER), are used to measure P-gp-mediated transport, but findings can be inconsistent across models. We propose a novel, more physiologically relevant, in vitro model: unidirectional apical efflux ratio (AP-ER)-a ratio of permeability rates at the apical side of the BBB with and without P-gp inhibitor. To test our approach, ER and AP-ER were calculated for 3227 structurally diverse compounds in porcine kidney epithelial cells (LLC-PK1) overexpressing human or mouse P-gp and classified based on their passive transcellular P-gp permeability or charged properties. In vivo rat infusion studies were performed for selected compounds with high ER but low AP-ER. One-third of the 3227 compounds had bidirectional ER that was much higher than AP-ER; very few had AP-ER higher than ER. Compounds with a large difference between AP-ER and ER were typically basic compounds with low-to-medium passive permeability and high lipophilicity and/or amphiphilicity, leading to strong membrane binding. Outcomes in the human model were similar to those in mice, suggesting AP-ER/ER ratios may be conserved for at least two species. AP-ER predicted measured cerebrospinal fluid (CSF) concentration better than ER for the five compounds tested in our in vivo rat infusion studies. We report superior estimations of the CSF concentrations of the compounds when based on less resource-intensive AP-ER versus classic ER. Better understanding of the properties leading to high P-gp-mediated efflux in vivo could support more efficient brain-penetrant compound screening and optimization. SIGNIFICANCE STATEMENT: To address inconsistencies associated with the historical, bidirectional efflux ratio (ER) calculation of P-glycoprotein-mediated transport, we propose to use the novel, more physiologically relevant, unidirectional apical efflux ratio (AP-ER) model. In vitro experiments suggested that compounds with strong membrane binding showed the largest difference between AP-ER and ER, and in vivo infusion studies showed that AP-ER predicted cerebrospinal fluid concentrations of compounds better than ER; outcomes in the human model were similar to those in mice.
Collapse
Affiliation(s)
- Holger Fischer
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Claudia Senn
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Mohammed Ullah
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Carina Cantrill
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Franz Schuler
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Li Yu
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| |
Collapse
|
30
|
Esposito C, Wang S, Lange UEW, Oellien F, Riniker S. Combining Machine Learning and Molecular Dynamics to Predict P-Glycoprotein Substrates. J Chem Inf Model 2020; 60:4730-4749. [DOI: 10.1021/acs.jcim.0c00525] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Carmen Esposito
- Laboratory of Physical Chemistry, ETH Zurich, Vladimir-Prelog-Weg 2, 8093 Zurich, Switzerland
| | - Shuzhe Wang
- Laboratory of Physical Chemistry, ETH Zurich, Vladimir-Prelog-Weg 2, 8093 Zurich, Switzerland
| | - Udo E. W. Lange
- Neuroscience Discovery, Medicinal Chemistry, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Frank Oellien
- Neuroscience Discovery, Medicinal Chemistry, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Sereina Riniker
- Laboratory of Physical Chemistry, ETH Zurich, Vladimir-Prelog-Weg 2, 8093 Zurich, Switzerland
| |
Collapse
|
31
|
Fischer H, Ullah M, de la Cruz CC, Hunsaker T, Senn C, Wirz T, Wagner B, Draganov D, Vazvaei F, Donzelli M, Paehler A, Merchant M, Yu L. Entrectinib, a TRK/ROS1 inhibitor with anti-CNS tumor activity: differentiation from other inhibitors in its class due to weak interaction with P-glycoprotein. Neuro Oncol 2020; 22:819-829. [PMID: 32383735 PMCID: PMC7283026 DOI: 10.1093/neuonc/noaa052] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Studies evaluating the CNS penetration of a novel tyrosine kinase inhibitor, entrectinib, proved challenging, particularly due to discrepancies across earlier experiments regarding P-glycoprotein (P-gp) interaction and brain distribution. To address this question, we used a novel "apical efflux ratio" (AP-ER) model to assess P-gp interaction with entrectinib, crizotinib, and larotrectinib, and compared their brain-penetration properties. METHODS AP-ER was designed to calculate P-gp interaction with the 3 drugs in vitro using P-gp-overexpressing cells. Brain penetration was studied in rat plasma, brain, and cerebrospinal fluid (CSF) samples after intravenous drug infusion. Unbound brain concentrations were estimated through kinetic lipid membrane binding assays and ex vivo experiments, while the antitumor activity of entrectinib was evaluated in a clinically relevant setting using an intracranial tumor mouse model. RESULTS Entrectinib showed lower AP-ER (1.1-1.15) than crizotinib and larotrectinib (≥2.8). Despite not reaching steady-state brain exposures in rats after 6 hours, entrectinib presented a more favorable CSF-to-unbound concentration in plasma (CSF/Cu,p) ratio (>0.2) than crizotinib and larotrectinib at steady state (both: CSF/Cu,p ~0.03). In vivo experiments validated the AP-ER approach. Entrectinib treatment resulted in strong tumor inhibition and full survival benefit in the intracranial tumor model at clinically relevant systemic exposures. CONCLUSIONS Entrectinib, unlike crizotinib and larotrectinib, is a weak P-gp substrate that can sustain CNS exposure based on our novel in vitro and in vivo experiments. This is consistent with the observed preclinical and clinical efficacy of entrectinib in neurotrophic tropomyosin receptor kinase (NTRK) and ROS1 fusion-positive CNS tumors and secondary CNS metastases.
Collapse
Affiliation(s)
- Holger Fischer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Mohammed Ullah
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | | | | | - Claudia Senn
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Thomas Wirz
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Björn Wagner
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Dragomir Draganov
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Faye Vazvaei
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., NJ, USA
| | - Massimiliano Donzelli
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | - Axel Paehler
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Switzerland
| | | | - Li Yu
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., NJ, USA
| |
Collapse
|
32
|
Wei S, Gao J, Zhang M, Dou Z, Li W, Zhao L. Dual delivery nanoscale device for miR-451 and adriamycin co-delivery to combat multidrug resistant in bladder cancer. Biomed Pharmacother 2019; 122:109473. [PMID: 31918263 DOI: 10.1016/j.biopha.2019.109473] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
The outcome of current cancer therapy is usually impeded by complicated extracellular and intracellular barriers. Most importantly, untargeted distribution and multidrug resistance (MDR) are considered as two important difficulties responsible for the poor performance of many currently available drug delivery systems (DDS). As a result, in our study, we developed a cancer cell membrane (CM) coated calcium carbonate (CC) nanoparticles to co-delivery miR-451 with adriamycin (Adr) to address the dilemma occurred in the therapy of bladder cancer (MCC/R-A). The homologous CCM from MDR bladder cancer cells (BIU-87/Adr) was employed to increase targeted retention of DDS within the tumor tissue and to bypass the extracellular barriers. Moreover, the MDR of cancer cells was conquered through downregulation of P-gp expression using miR-451 since it was confirmed by previous reports that miR-451 could significantly downregulate the level of P-gp in MDR cells, which in turn elevated the cellular drug retention in BIU-87/Adr. Our in vitro and in vivo experiments have revealed that MCC/R-A showed a greatly enhanced therapeutic effect on BIU-87/Adr, which was superior than applying miR-451 or Adr alone. The preferable effect of MCC/R-A on conquering the MDR in bladder cancer provides a novel alternative for effective chemotherapy of MDR cancers.
Collapse
Affiliation(s)
- Shuguang Wei
- Department of Urology Surgery, the First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, Henan Province, China
| | - Jiannan Gao
- Yidu Central Hospital of Weifang, Weifang, 262500, Shandong Province, China
| | - Maopeng Zhang
- Department of Pharmacy, Jiyang District People's Hospital of Jinan City, 251400, Shandong province, China
| | - Zhongling Dou
- Department of Urology Surgery, the First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, Henan Province, China
| | - Wensheng Li
- Department of Urology Surgery, the First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, Henan Province, China
| | - Leizuo Zhao
- Attending physician, Department of Urology, Dongying People's hospital, Jinan, 257091, Shandong Province, China.
| |
Collapse
|
33
|
Gatarić B, Parojčić J. An Investigation into the Factors Governing Drug Absorption and Food Effect Prediction Based on Data Mining Methodology. AAPS JOURNAL 2019; 22:11. [PMID: 31823145 DOI: 10.1208/s12248-019-0394-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Drug absorption is a complex process governed by a number of interrelated physicochemical, biopharmaceutical, and pharmacokinetic factors. In order to explore complex relationships among these factors, multivariate exploratory analysis was performed on the dataset of drugs with diverse bioperformance. The investigated dataset included subset of drugs for which bioequivalence between solid dosage form and oral solution has been reported, and subset of drugs described in the literature as low solubility/low permeability compounds. Discriminatory power of hierarchical clustering on principal components was somewhat higher when applied on the data subsets of drugs with similar bioperformance, while analysis of the integrated dataset indicated existence of two groups of drugs with the boundaries reflected in Peff value of approximately 2 × 10-4 cm/s and Fa and Fm values higher than 85% and 50%, respectively. Majority of the investigated drugs within the integrated dataset were grouped within their initial subset indicating that overall drug bioperformance is closely related to its physicochemical, biopharmaceutical and pharmacokinetic properties. Classification models constructed using the random forest (RF) and support vector machine with polynomial kernel function were able to predict food effect based on drug dose/solubility ratio (D/S), effective permeability (Peff), percent of dose metabolized (Fm), and elimination half-life (τ1/2). Although both models performed well during training and testing, only RF kept satisfying performance when applied on the external dataset (kappa value > 0.4). The results obtained indicate that data mining can be employed as useful tool in biopharmaceutical drug characterization which merits further investigation.
Collapse
Affiliation(s)
- Biljana Gatarić
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000, Banja Luka, Bosnia and Herzegovina.
| | - Jelena Parojčić
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, 11221, Serbia
| |
Collapse
|
34
|
He J, Gong C, Qin J, Li M, Huang S. Cancer Cell Membrane Decorated Silica Nanoparticle Loaded with miR495 and Doxorubicin to Overcome Drug Resistance for Effective Lung Cancer Therapy. NANOSCALE RESEARCH LETTERS 2019; 14:339. [PMID: 31705398 PMCID: PMC6841775 DOI: 10.1186/s11671-019-3143-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/30/2019] [Indexed: 05/03/2023]
Abstract
Current cancer therapy usually succumbs to many extracellular and intracellular barriers, among which untargeted distribution and multidrug resistance (MDR) are two important difficulties responsible for poor outcome of many drug delivery systems (DDS). Here, in our study, the dilemma was addressed by developing a cancer cell membrane (CCM)-coated silica (SLI) nanoparticles to co-deliver miR495 with doxorubicin (DOX) for effective therapy of lung cancer (CCM/SLI/R-D). The homologous CCM from MDR lung cancer cells (A549/DOX) was supposed to increase the tumor-homing property of the DDS to bypass the extracellular barriers. Moreover, the MDR of cancer cells were conquered through downregulation of P-glycoprotein (P-gp) expression using miR495. It was proved that miR495 could significantly decrease the expression of P-gp which elevated intracellular drug accumulation in A549/DOX. The in vitro and in vivo results exhibited that CCM/SLI/R-D showed a greatly enhanced therapeutic effect on A549/DOX, which was superior than applying miR495 or DOX alone. The preferable effect of CCM/SLI/R-D on conquering the MDR in lung cancer provides a novel alternative for effective chemotherapy of MDR cancers.
Collapse
Affiliation(s)
- Jinyuan He
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Chulian Gong
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Jie Qin
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Mingan Li
- Department of Interventional Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Shaohong Huang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| |
Collapse
|
35
|
Kadioglu O, Efferth T. A Machine Learning-Based Prediction Platform for P-Glycoprotein Modulators and Its Validation by Molecular Docking. Cells 2019; 8:E1286. [PMID: 31640190 PMCID: PMC6829872 DOI: 10.3390/cells8101286] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (P-gp) is an important determinant of multidrug resistance (MDR) because its overexpression is associated with increased efflux of various established chemotherapy drugs in many clinically resistant and refractory tumors. This leads to insufficient therapeutic targeting of tumor populations, representing a major drawback of cancer chemotherapy. Therefore, P-gp is a target for pharmacological inhibitors to overcome MDR. In the present study, we utilized machine learning strategies to establish a model for P-gp modulators to predict whether a given compound would behave as substrate or inhibitor of P-gp. Random forest feature selection algorithm-based leave-one-out random sampling was used. Testing the model with an external validation set revealed high performance scores. A P-gp modulator list of compounds from the ChEMBL database was used to test the performance, and predictions from both substrate and inhibitor classes were selected for the last step of validation with molecular docking. Predicted substrates revealed similar docking poses than that of doxorubicin, and predicted inhibitors revealed similar docking poses than that of the known P-gp inhibitor elacridar, implying the validity of the predictions. We conclude that the machine-learning approach introduced in this investigation may serve as a tool for the rapid detection of P-gp substrates and inhibitors in large chemical libraries.
Collapse
Affiliation(s)
- Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128 Mainz, Germany.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128 Mainz, Germany.
| |
Collapse
|