1
|
Sun Z, Huo D, Guo J, Yan A. Modeling and Interpretability Study of the Structure-Activity Relationship for Multigeneration EGFR Inhibitors. ACS OMEGA 2025; 10:11176-11187. [PMID: 40160792 PMCID: PMC11947818 DOI: 10.1021/acsomega.4c10464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
The fourth-generation EGFR inhibitors targeting L858R/T790M/C797S mutations are in clinical trials mostly, and it is necessary to develop new inhibitors. In this study, an internal data set containing 2302 multitarget EGFR inhibitors targeting the wild type (83%) and the L858R (92%), L858R/T790M (96%), and L858R/T790M/C797S (60%) mutations was collected. We established a structure-activity relationship model for predicting the bioactivities of multigeneration EGFR inhibitors by a multitask deep neural network (MT-DNN). We also constructed four single-task models on 1384 L858R/T790M/C797S (60%) mutation inhibitors by support vector machine (SVM), random forest (RF), XGBoost (XGB), and single-target neural network (ST-DNN), respectively. The MT-DNN model significantly outperformed single-task models on the external data set of 304 fourth-generation EGFR inhibitors. Furthermore, the combined application of MT-DNN and SHAP/delta-SHAP value interpretability analysis offers rigorous structural information from a global perspective. With SHAP/delta-SHAP methods, the MT-DNN model can mine the core scaffold and important fragments of multigeneration EGFR inhibitors and provide valuable information from a structure-activity relationship perspective to address the resistant mutation problem.
Collapse
Affiliation(s)
- Zhiqi Sun
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Donghui Huo
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Jiangyu Guo
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Aixia Yan
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| |
Collapse
|
2
|
Priya MGR, Manisha J, Lazar LPM, Rathore SS, Solomon VR. Computer-aided Drug Discovery Approaches in the Identification of Anticancer Drugs from Natural Products: A Review. Curr Comput Aided Drug Des 2025; 21:1-14. [PMID: 38698753 DOI: 10.2174/0115734099283410240406064042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 05/05/2024]
Abstract
Natural plant sources are essential in the development of several anticancer drugs, such as vincristine, vinblastine, vinorelbine, docetaxel, paclitaxel, camptothecin, etoposide, and teniposide. However, various chemotherapies fail due to adverse reactions, drug resistance, and target specificity. Researchers are now focusing on developing drugs that use natural compounds to overcome these issues. These drugs can affect multiple targets, have reduced adverse effects, and are effective against several cancer types. Developing a new drug is a highly complex, expensive, and time-consuming process. Traditional drug discovery methods take up to 15 years for a new medicine to enter the market and cost more than one billion USD. However, recent Computer Aided Drug Discovery (CADD) advancements have changed this situation. This paper aims to comprehensively describe the different CADD approaches in identifying anticancer drugs from natural products. Data from various sources, including Science Direct, Elsevier, NCBI, and Web of Science, are used in this review. In-silico techniques and optimization algorithms can provide versatile solutions in drug discovery ventures. The structure-based drug design technique is widely used to understand chemical constituents' molecular-level interactions and identify hit leads. This review will discuss the concept of CADD, in-silico tools, virtual screening in drug discovery, and the concept of natural products as anticancer therapies. Representative examples of molecules identified will also be provided.
Collapse
Affiliation(s)
- Muthiah Gnana Ruba Priya
- College of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Jessica Manisha
- Department of Pharmacology, Sridevi College of Pharmacy, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | | | - Seema Singh Rathore
- College of Pharmaceutical Sciences, Department of Pharmaceutics, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Viswas Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Telangana, India
| |
Collapse
|
3
|
Selvam S, Balaji PD, Sohn H, Madhavan T. AISMPred: A Machine Learning Approach for Predicting Anti-Inflammatory Small Molecules. Pharmaceuticals (Basel) 2024; 17:1693. [PMID: 39770535 PMCID: PMC11676721 DOI: 10.3390/ph17121693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Inflammation serves as a vital response to diverse harmful stimuli like infections, toxins, or tissue injuries, aiding in the elimination of pathogens and tissue repair. However, persistent inflammation can lead to chronic diseases. Peptide therapeutics have gained attention for their specificity in targeting cells, yet their development remains costly and time-consuming. Therefore, small molecules, with their stability, low immunogenicity, and oral bioavailability, have become a focal point for predicting anti-inflammatory small molecules (AISMs). Methods: In this study, we introduce a computational method called AISMPred, designed to classify AISMs and non-AISMs. To develop this approach, we constructed a dataset comprising 1750 AISMs and non-AISMs, each annotated with IC50 values sourced from the PubChem BioAssay database. We computed two distinct types of molecular descriptors using PaDEL and Mordred tools. Subsequently, these descriptors were concatenated to form a hybrid feature set. The SVC-L1 regularization method was implemented for the optimum feature selection to develop robust Machine learning (ML) models. Five different conventional ML classifiers were employed, such as RF, ET, KNN, LR, and Ensemble methods. Results: A total of 15 ML models were developed using 2D, FP, and Hybrid feature sets, with the ET model with hybrid features achieving the highest accuracy of 92% and an AUC of 0.97 on the independent test dataset. Conclusions: This study provides an effective method for screening AISMs, potentially impacting drug discovery and design.
Collapse
Affiliation(s)
- Subathra Selvam
- Computational Biology Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu 603203, Tamil Nadu, India; (S.S.); (P.D.B.)
| | - Priya Dharshini Balaji
- Computational Biology Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu 603203, Tamil Nadu, India; (S.S.); (P.D.B.)
| | - Honglae Sohn
- Department of Chemistry, Chosun University, Gwangju 501-759, Republic of Korea
| | - Thirumurthy Madhavan
- Computational Biology Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu 603203, Tamil Nadu, India; (S.S.); (P.D.B.)
| |
Collapse
|
4
|
Fatima I, Rehman A, Ding Y, Wang P, Meng Y, Rehman HU, Warraich DA, Wang Z, Feng L, Liao M. Breakthroughs in AI and multi-omics for cancer drug discovery: A review. Eur J Med Chem 2024; 280:116925. [PMID: 39378826 DOI: 10.1016/j.ejmech.2024.116925] [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: 06/28/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Abstract
Cancer is one of the biggest medical challenges we face today. It is characterized by abnormal, uncontrolled growth of cells that can spread to different parts of the body. Cancer is extremely complex, with genetic variations and the ability to adapt and evolve. This means we must continuously pursue innovative approaches to developing new cancer drugs. While traditional drug discovery methods have led to important breakthroughs, they also have significant limitations that make it difficult to efficiently create new, cost-effective cancer therapies. Integrating computational tools into the cancer drug discovery process is a major step forward. By harnessing computing power, we can overcome some of the inherent barriers of traditional methods. This review examines the range of computational techniques now being used, such as molecular docking, QSAR models, virtual screening, and pharmacophore modeling. It looks at recent advances in areas like machine learning and molecular simulations. The review also discusses the current challenges with these technologies and envisions future directions, underscoring how transformative these computational tools can be for creating targeted, new cancer treatments.
Collapse
Affiliation(s)
- Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| | - Yanheng Ding
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuxuan Meng
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Hafeez Ur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Dawood Ahmad Warraich
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhibo Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Lijun Feng
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
5
|
Moussaoui M, Baammi S, Soufi H, Baassi M, Salah M, Allali AEL, Mohammed BE, Daoud R, Belaaouad S. Design and Optimization of Quinazoline Derivatives as Potent EGFR Inhibitors for Lung Cancer Treatment: A Comprehensive QSAR, ADMET, and Molecular Modeling Investigation. ACS OMEGA 2024; 9:45842-45857. [PMID: 39583714 PMCID: PMC11579736 DOI: 10.1021/acsomega.4c04639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 11/26/2024]
Abstract
The epidermal growth factor receptor (EGFR) is part of a protein family that controls cell growth and development. Due to its importance, EGFR has been identified as a suitable target for creating novel drugs. For this research, we conducted a 2D-QSAR analysis on a set of 31 molecules derived from quinazoline, which exhibited inhibitory activity against human lung cancer. This investigation incorporated principal component analysis (PCA) and multiple linear regression (MLR), leading to the development of QSAR models with strong predictive capabilities (R 2 = 0.745, R 2_adj = 0.723, MSE = 0.061, R 2_test = 0.941, and Q 2_cv = 0.669). The reliability of these models was confirmed through internal, external, Y-randomization, and applicability domain validations. Leveraging the predictions from the QSAR model, we designed 18 new molecules based on the modifications at the N-3 and C-6 positions of the quinazoline ring, with electronegative substituents at these positions fostering optimal polar interactions and hydrophobic contacts within the ATP-binding site of EGFR, significantly enhancing the inhibitory activity against the lung cancer cell line. Subsequently, ADMET predictions were conducted for these 18 compounds, revealing outstanding ADMET profiles. Molecular docking analyses were performed to investigate the interactions between the newly designed molecules-Pred15, Pred17, Pred20, Pred21-and the EGFR protein, indicating high affinity of these proposed compounds to EGFR. Furthermore, molecular dynamics (MD) simulations were utilized to assess the stability and binding modes of compounds Pred17, Pred20, and Pred21, reinforcing their potential as novel inhibitors against human lung cancer. Overall, our findings suggest that these investigated compounds can serve as effective inhibitors, showcasing the utility of our analytical and design approach in the identification of promising therapeutic agents.
Collapse
Affiliation(s)
- Mohamed Moussaoui
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Soukayna Baammi
- Bioinformatics
Laboratory, College of Computing, Mohammed
VI Polytechnic University, Ben Guerir 20670, Morocco
| | - Hatim Soufi
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Mouna Baassi
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Mohammed Salah
- Team
of Chemoinformatics Research and Spectroscopy and Quantum Chemistry,
Department of Chemistry, Faculty of Science, University Chouaib Doukkali, B. P. 20, El Jadida 2300, Morocco
| | - Achraf EL Allali
- Bioinformatics
Laboratory, College of Computing, Mohammed
VI Polytechnic University, Ben Guerir 20670, Morocco
| | - Belghiti Elalaoui Mohammed
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
- Laboratory
of Nernest Technology, 163 Willington Street, Sherbrook, Quebec J1H5C7, Canada
| | - Rachid Daoud
- Chemical
and Biochemical Sciences-Green Processing Engineering, Mohammed VI Polytechnic University, Ben Guerir 43150, Morocco
| | - Said Belaaouad
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| |
Collapse
|
6
|
Gomatam A, Hirlekar BU, Singh KD, Murty US, Dixit VA. Improved QSAR models for PARP-1 inhibition using data balancing, interpretable machine learning, and matched molecular pair analysis. Mol Divers 2024; 28:2135-2152. [PMID: 38374474 DOI: 10.1007/s11030-024-10809-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/07/2024] [Indexed: 02/21/2024]
Abstract
The poly (ADP-ribose) polymerase-1 (PARP-1) enzyme is an important target in the treatment of breast cancer. Currently, treatment options include the drugs Olaparib, Niraparib, Rucaparib, and Talazoparib; however, these drugs can cause severe side effects including hematological toxicity and cardiotoxicity. Although in silico models for the prediction of PARP-1 activity have been developed, the drawbacks of these models include low specificity, a narrow applicability domain, and a lack of interpretability. To address these issues, a comprehensive machine learning (ML)-based quantitative structure-activity relationship (QSAR) approach for the informed prediction of PARP-1 activity is presented. Classification models built using the Synthetic Minority Oversampling Technique (SMOTE) for data balancing gave robust and predictive models based on the K-nearest neighbor algorithm (accuracy 0.86, sensitivity 0.88, specificity 0.80). Regression models were built on structurally congeneric datasets, with the models for the phthalazinone class and fused cyclic compounds giving the best performance. In accordance with the Organization for Economic Cooperation and Development (OECD) guidelines, a mechanistic interpretation is proposed using the Shapley Additive Explanations (SHAP) to identify the important topological features to differentiate between PARP-1 actives and inactives. Moreover, an analysis of the PARP-1 dataset revealed the prevalence of activity cliffs, which possibly negatively impacts the model's predictive performance. Finally, a set of chemical transformation rules were extracted using the matched molecular pair analysis (MMPA) which provided mechanistic insights and can guide medicinal chemists in the design of novel PARP-1 inhibitors.
Collapse
Affiliation(s)
- Anish Gomatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, (NIPER Guwahati), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), Dist: Kamrup, P.O.: Changsari, Guwahati, Assam, 781101, India
| | - Bhakti Umesh Hirlekar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, (NIPER Guwahati), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), Dist: Kamrup, P.O.: Changsari, Guwahati, Assam, 781101, India
| | - Krishan Dev Singh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, (NIPER Guwahati), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), Dist: Kamrup, P.O.: Changsari, Guwahati, Assam, 781101, India
| | - Upadhyayula Suryanarayana Murty
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, (NIPER Guwahati), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), Dist: Kamrup, P.O.: Changsari, Guwahati, Assam, 781101, India
| | - Vaibhav A Dixit
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, (NIPER Guwahati), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), Dist: Kamrup, P.O.: Changsari, Guwahati, Assam, 781101, India.
| |
Collapse
|
7
|
Sengupta A, Singh SK, Kumar R. Support Vector Machine-Based Prediction Models for Drug Repurposing and Designing Novel Drugs for Colorectal Cancer. ACS OMEGA 2024; 9:18584-18592. [PMID: 38680332 PMCID: PMC11044175 DOI: 10.1021/acsomega.4c01195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024]
Abstract
Colorectal cancer (CRC) has witnessed a concerning increase in incidence and poses a significant therapeutic challenge due to its poor prognosis. There is a pressing demand to identify novel drug therapies to combat CRC. In this study, we addressed this need by utilizing the pharmacological profiles of anticancer drugs from the Genomics of Drug Sensitivity in Cancer (GDSC) database and developed QSAR models using the Support Vector Machine (SVM) algorithm for prediction of alternative and promiscuous anticancer compounds for CRC treatment. Our QSAR models demonstrated their robustness by achieving a high correlation of determination (R2) after 10-fold cross-validation. For 12 CRC cell lines, R2 ranged from 0.609 to 0.827. The highest performance was achieved for SW1417 and GP5d cell lines with R2 values of 0.827 and 0.786, respectively. Further, we listed the most common chemical descriptors in the drug profiles of the CRC cell lines and we also further reported the correlation of these descriptors with drug activity. The KRFP314 fingerprint was the predominantly occurring descriptor, with the KRFPC314 fingerprint following closely in prevalence within the drug profiles of the CRC cell lines. Beyond predictive modeling, we also confirmed the applicability of our developed QSAR models via in silico methods by conducting descriptor-drug analyses and recapitulating drug-to-oncogene relationships. We also identified two potential anti-CRC FDA-approved drugs, viomycin and diamorphine, using QSAR models. To ensure the easy accessibility and utility of our research findings, we have incorporated these models into a user-friendly prediction Web server named "ColoRecPred", available at https://project.iith.ac.in/cgntlab/colorecpred. We anticipate that this Web server can be used for screening of chemical libraries to identify potential anti-CRC drugs.
Collapse
Affiliation(s)
- Avik Sengupta
- Department
of Biotechnology, Indian Institute of Technology
Hyderabad, Kandi, Telangana 502284, India
| | - Saurabh Kumar Singh
- Department
of Chemistry, Indian Institute of Technology
Hyderabad, Kandi, Telangana 502284, India
| | - Rahul Kumar
- Department
of Biotechnology, Indian Institute of Technology
Hyderabad, Kandi, Telangana 502284, India
| |
Collapse
|
8
|
Iwata H, Nakai T, Koyama T, Matsumoto S, Kojima R, Okuno Y. VGAE-MCTS: A New Molecular Generative Model Combining the Variational Graph Auto-Encoder and Monte Carlo Tree Search. J Chem Inf Model 2023; 63:7392-7400. [PMID: 37993764 PMCID: PMC10716893 DOI: 10.1021/acs.jcim.3c01220] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023]
Abstract
Molecular generation is crucial for advancing drug discovery, materials science, and chemical exploration. It expedites the search for new drug candidates, facilitates tailored material creation, and enhances our understanding of molecular diversity. By employing artificial intelligence techniques such as molecular generative models based on molecular graphs, researchers have tackled the challenge of identifying efficient molecules with desired properties. Here, we propose a new molecular generative model combining a graph-based deep neural network and a reinforcement learning technique. We evaluated the validity, novelty, and optimized physicochemical properties of the generated molecules. Importantly, the model explored uncharted regions of chemical space, allowing for the efficient discovery and design of new molecules. This innovative approach has considerable potential to revolutionize drug discovery, materials science, and chemical research for accelerating scientific innovation. By leveraging advanced techniques and exploring previously unexplored chemical spaces, this study offers promising prospects for the efficient discovery and design of new molecules in the field of drug development.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
| | - Taichi Nakai
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
| | - Takuto Koyama
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
| | - Shigeyuki Matsumoto
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
| | - Ryosuke Kojima
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
| | - Yasushi Okuno
- Graduate
School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto-shi, Kyoto 606-8507, Japan
- HPC-
and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe-shi, Hyogo 650-0047, Japan
| |
Collapse
|
9
|
Kleandrova VV, Cordeiro MNDS, Speck-Planche A. Optimizing drug discovery using multitasking models for quantitative structure-biological effect relationships: an update of the literature. Expert Opin Drug Discov 2023; 18:1231-1243. [PMID: 37639708 DOI: 10.1080/17460441.2023.2251385] [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: 06/08/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Drug discovery has provided modern societies with the means to fight against many diseases. In this sense, computational methods have been at the forefront, playing an important role in rationalizing the search for novel drugs. Yet, tackling phenomena such as the multi-genic nature of diseases and drug resistance are limitations of the current computational methods. Multi-tasking models for quantitative structure-biological effect relationships (mtk-QSBER) have emerged to overcome such limitations. AREAS COVERED The present review describes an update on the fundamentals and applications of the mtk-QSBER models as tools to accelerate multiple stages/substages of the drug discovery process. EXPERT OPINION Computational approaches are extremely important for the rationalization of the search for novel and efficacious therapeutic agents. However, they need to focus more on the multi-target drug discovery paradigm. In this sense, mtk-QSBER models are particularly suited for multi-target drug discovery, offering encouraging opportunities across multiple therapeutic areas and scientific disciplines associated with drug discovery.
Collapse
Affiliation(s)
- Valeria V Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Russian Biotechnological University, Moscow, Russian Federation
| | - M Natália D S Cordeiro
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Alejandro Speck-Planche
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Li J, Wang C, Yue L, Chen F, Cao X, Wang Z. Nano-QSAR modeling for predicting the cytotoxicity of metallic and metal oxide nanoparticles: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113955. [PMID: 35961199 DOI: 10.1016/j.ecoenv.2022.113955] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/11/2022] [Accepted: 08/03/2022] [Indexed: 06/15/2023]
Abstract
Given the rapid development of nanotechnology, it is crucial to understand the effects of nanoparticles on living organisms. However, it is laborious to perform toxicological tests on a case-by-case basis. Quantitative structure-activity relationship (QSAR) is an effective computational technique because it saves time, costs, and animal sacrifice. Therefore, this review presents general procedures for the construction and application of nano-QSAR models of metal-based and metal-oxide nanoparticles (MBNPs and MONPs). We also provide an overview of available databases and common algorithms. The molecular descriptors and their roles in the toxicological interpretation of MBNPs and MONPs are systematically reviewed and the future of nano-QSAR is discussed. Finally, we address the growing demand for novel nano-specific descriptors, new computational strategies to address the data shortage, in situ data for regulatory concerns, a better understanding of the physicochemical properties of NPs with bioactivity, and, most importantly, the design of nano-QSAR for real-life environmental predictions rather than laboratory simulations.
Collapse
Affiliation(s)
- Jing Li
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chuanxi Wang
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Le Yue
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Feiran Chen
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xuesong Cao
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhenyu Wang
- Institute of Environmental Processes and Pollution Control, and School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
11
|
Artificial intelligence and machine-learning approaches in structure and ligand-based discovery of drugs affecting central nervous system. Mol Divers 2022; 27:959-985. [PMID: 35819579 DOI: 10.1007/s11030-022-10489-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 12/11/2022]
Abstract
CNS disorders are indications with a very high unmet medical needs, relatively smaller number of available drugs, and a subpar satisfaction level among patients and caregiver. Discovery of CNS drugs is extremely expensive affair with its own unique challenges leading to extremely high attrition rates and low efficiency. With explosion of data in information age, there is hardly any aspect of life that has not been touched by data driven technologies such as artificial intelligence (AI) and machine learning (ML). Drug discovery is no exception, emergence of big data via genomic, proteomic, biological, and chemical technologies has driven pharmaceutical giants to collaborate with AI oriented companies to revolutionise drug discovery, with the goal of increasing the efficiency of the process. In recent years many examples of innovative applications of AI and ML techniques in CNS drug discovery has been reported. Research on therapeutics for diseases such as schizophrenia, Alzheimer's and Parkinsonism has been provided with a new direction and thrust from these developments. AI and ML has been applied to both ligand-based and structure-based drug discovery and design of CNS therapeutics. In this review, we have summarised the general aspects of AI and ML from the perspective of drug discovery followed by a comprehensive coverage of the recent developments in the applications of AI/ML techniques in CNS drug discovery.
Collapse
|
12
|
Speck-Planche A, Kleandrova VV, Scotti MT. In Silico Drug Repurposing for Anti-Inflammatory Therapy: Virtual Search for Dual Inhibitors of Caspase-1 and TNF-Alpha. Biomolecules 2021; 11:biom11121832. [PMID: 34944476 PMCID: PMC8699067 DOI: 10.3390/biom11121832] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/15/2021] [Accepted: 12/02/2021] [Indexed: 12/27/2022] Open
Abstract
Inflammation involves a complex biological response of the body tissues to damaging stimuli. When dysregulated, inflammation led by biomolecular mediators such as caspase-1 and tumor necrosis factor-alpha (TNF-alpha) can play a detrimental role in the progression of different medical conditions such as cancer, neurological disorders, autoimmune diseases, and cytokine storms caused by viral infections such as COVID-19. Computational approaches can accelerate the search for dual-target drugs able to simultaneously inhibit the aforementioned proteins, enabling the discovery of wide-spectrum anti-inflammatory agents. This work reports the first multicondition model based on quantitative structure–activity relationships and a multilayer perceptron neural network (mtc-QSAR-MLP) for the virtual screening of agency-regulated chemicals as versatile anti-inflammatory therapeutics. The mtc-QSAR-MLP model displayed accuracy higher than 88%, and was interpreted from a physicochemical and structural point of view. When using the mtc-QSAR-MLP model as a virtual screening tool, we could identify several agency-regulated chemicals as dual inhibitors of caspase-1 and TNF-alpha, and the experimental information later retrieved from the scientific literature converged with our computational results. This study supports the capabilities of our mtc-QSAR-MLP model in anti-inflammatory therapy with direct applications to current health issues such as the COVID-19 pandemic.
Collapse
Affiliation(s)
- Alejandro Speck-Planche
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Correspondence:
| | - Valeria V. Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production, Volokolamskoe shosse 11, 125080 Moscow, Russia;
| | - Marcus T. Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| |
Collapse
|
13
|
Chen Z, Lin L, Wu C, Li C, Xu R, Sun Y. Artificial intelligence for assisting cancer diagnosis and treatment in the era of precision medicine. Cancer Commun (Lond) 2021; 41:1100-1115. [PMID: 34613667 PMCID: PMC8626610 DOI: 10.1002/cac2.12215] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/10/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past decade, artificial intelligence (AI) has contributed substantially to the resolution of various medical problems, including cancer. Deep learning (DL), a subfield of AI, is characterized by its ability to perform automated feature extraction and has great power in the assimilation and evaluation of large amounts of complicated data. On the basis of a large quantity of medical data and novel computational technologies, AI, especially DL, has been applied in various aspects of oncology research and has the potential to enhance cancer diagnosis and treatment. These applications range from early cancer detection, diagnosis, classification and grading, molecular characterization of tumors, prediction of patient outcomes and treatment responses, personalized treatment, automatic radiotherapy workflows, novel anti-cancer drug discovery, and clinical trials. In this review, we introduced the general principle of AI, summarized major areas of its application for cancer diagnosis and treatment, and discussed its future directions and remaining challenges. As the adoption of AI in clinical use is increasing, we anticipate the arrival of AI-powered cancer care.
Collapse
Affiliation(s)
- Zi‐Hang Chen
- Department of Radiation OncologyState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080P. R. China
| | - Li Lin
- Department of Radiation OncologyState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Chen‐Fei Wu
- Department of Radiation OncologyState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Chao‐Feng Li
- Artificial Intelligence LaboratoryState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Rui‐Hua Xu
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Ying Sun
- Department of Radiation OncologyState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| |
Collapse
|
14
|
Begum S, Shareef MZ, Bharathi K. Part-II- in silico drug design: application and success. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2018-0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
In silico tools have indeed reframed the steps involved in traditional drug discovery and development process and the term in silico has become a familiar term in pharmaceutical sector like the terms in vitro and in vivo. The successful design of HIV protease inhibitors, Saquinavir, Indinavir and other important medicinal agents, initiated interest of researchers in structure based drug design approaches (SBDD). The interactions between biomolecules and a ligand, binding energy, free energy and stability of biomolecule-ligand complex can be envisioned and predicted by applying molecular docking studies. Protein-ligand, protein-protein, DNA-ligand interactions etc. aid in elucidating molecular level mechanisms of drug molecules. In the Ligand based drug design (LBDD) approaches, QSAR studies have tremendously contributed to the development of antimicrobial, anticancer, antimalarial agents. In the recent years, multiQSAR (mt-QSAR) approaches have been successfully employed for designing drugs against multifactorial diseases. Output of a research in several instances is rewarding when both SBDD and LBDD approaches are combined. Application of in silico studies for prediction of pharmacokinetics was once a real challenge but one can see unlimited number publications comprising tools, data bases which can accurately predict almost all the pharmacokinetic parameters. Absorption, distribution, metabolism, transporters, blood brain barrier permeability, hERG toxicity, P-gp affinity and several toxicological end points can be accurately predicted for a candidate molecule before its synthesis. In silico approaches are greatly encouraged a result of growing limitations and new legislations related to the animal use for research. The combined use of in vitro data and in silico tools will definitely decrease the use of animal testing in the future.In this chapter, in silico approaches and their applications are reviewed and discussed giving suitable examples.
Collapse
Affiliation(s)
- Shaheen Begum
- Institute of Pharmaceutical Technology , Sri Padmavati Mahila Visvavidyalayam , 517501 Tirupati , Andhra Pradesh , India
| | - Mohammad Zubair Shareef
- Institute of Pharmaceutical Technology , Sri Padmavati Mahila Visvavidyalayam , 517501 Tirupati , Andhra Pradesh , India
| | - Koganti Bharathi
- Institute of Pharmaceutical Technology , Sri Padmavati Mahila Visvavidyalayam , 517501 Tirupati , Andhra Pradesh , India
| |
Collapse
|
15
|
Pathania S, Singh PK, Narang RK, Rawal RK. Structure based designing of thiazolidinone-pyrimidine derivatives as ERK2 inhibitors: Synthesis and in vitro evaluation. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2021; 32:793-816. [PMID: 34583590 DOI: 10.1080/1062936x.2021.1973094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
Breast cancer has been associated with an overexpression of various molecular targets; accordingly, various target-specific chemotherapeutic agents have been developed. Inhibition of ERK2, a member of MAPK pathway, is an important target involved in the treatment of both oestrogen receptor-positive and triple-negative breast cancer. Thus, in continuation of our previous work on the ERK2 target, we here report novel inhibitors of this kinase. Out of three lead molecules reported in our previous study, we selected the thiazolidinone-pyrimidine scaffold for further development of small molecule inhibitors of ERK2. Analogues of the lead molecule were docked in the target kinase, followed by molecular dynamic simulations and MM-GBSA calculations. Analogues maintaining key interactions with amino acid residues in the ATP-binding domain of ERK2 were selected and duly synthesized. In vitro biochemical evaluation of these molecules against ERK2 kinase disclosed that two molecules possess significant kinase inhibitory potential with IC50 values ≤ 0.5 µM.
Collapse
Affiliation(s)
- S Pathania
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - P K Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - R K Narang
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - R K Rawal
- Department of Chemistry, Maharishi Markandeshwar (Deemed to Be University), Ambala, India
- CSIR-North East Institute of Science and Technology, Jorhat, India
| |
Collapse
|
16
|
Kleandrova VV, Speck-Planche A. The QSAR Paradigm in Fragment-Based Drug Discovery: From the Virtual Generation of Target Inhibitors to Multi-Scale Modeling. Mini Rev Med Chem 2021; 20:1357-1374. [PMID: 32013845 DOI: 10.2174/1389557520666200204123156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022]
Abstract
Fragment-Based Drug Design (FBDD) has established itself as a promising approach in modern drug discovery, accelerating and improving lead optimization, while playing a crucial role in diminishing the high attrition rates at all stages in the drug development process. On the other hand, FBDD has benefited from the application of computational methodologies, where the models derived from the Quantitative Structure-Activity Relationships (QSAR) have become consolidated tools. This mini-review focuses on the evolution and main applications of the QSAR paradigm in the context of FBDD in the last five years. This report places particular emphasis on the QSAR models derived from fragment-based topological approaches to extract physicochemical and/or structural information, allowing to design potentially novel mono- or multi-target inhibitors from relatively large and heterogeneous databases. Here, we also discuss the need to apply multi-scale modeling, to exemplify how different datasets based on target inhibition can be simultaneously integrated and predicted together with other relevant endpoints such as the biological activity against non-biomolecular targets, as well as in vitro and in vivo toxicity and pharmacokinetic properties. In this context, seminal papers are briefly analyzed. As huge amounts of data continue to accumulate in the domains of the chemical, biological and biomedical sciences, it has become clear that drug discovery must be viewed as a multi-scale optimization process. An ideal multi-scale approach should integrate diverse chemical and biological data and also serve as a knowledge generator, enabling the design of potentially optimal chemicals that may become therapeutic agents.
Collapse
Affiliation(s)
- Valeria V Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production, Volokolamskoe Shosse 11, 125080, Moscow, Russian Federation
| | - Alejandro Speck-Planche
- Department of Chemistry, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Trubetskaya Str., 8, b. 2, 119992, Moscow, Russian Federation
| |
Collapse
|
17
|
Rajagopal K, Sri VB, Byran G, Gomathi S. Pyrazole Substituted 9-Anilinoacridines as HER2 Inhibitors Targeting Breast Cancer - An In-Silico Approach. Curr Drug Res Rev 2021; 14:61-72. [PMID: 34139975 DOI: 10.2174/2589977513666210617160302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/06/2021] [Accepted: 03/05/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer is one of the malignant tumours which mainly affect the female population. Total 20% of the cases of breast cancer are due to overexpression of Human epidermal growth factor receptor-2 (HER2), which is the dominant tyrosine kinase receptor. In general, 9-anilinoacridine derivatives play an important role as antitumor agents due to their DNA-intercalating properties. OBJECTIVE Some novel 9-anilinoacridines substituted with pyrazole moiety(1a-z) were designed, and their HER2enzyme (PDB id-3PP0) inhibition activity was evaluated by molecular docking studies using the Glide module of Schrodinger suite 2019-4. METHODS Glide module of the Schrodinger suite was used to perform docking studies, qikprop module was used for in-silico ADMET screening, and the Prime-MM-GBSA module was used for free binding energy calculations. Using GLIDE scoring functions, we can determine the binding affinity of ligands (1a-z) towards HER2. RESULTS The inhibitory activity of ligands against HER2 was mainly due to the strong hydrophobic and hydrogen bonding interactions. Almost all the compounds 1a-z have a good binding affinity with Glide scores in the range of -4.9 to -9.75 compared to the standard drugs CK0403(-4.105) and Tamoxifen (-3.78). From the results of in-silico ADMET properties, most of the compounds fall within the recommended values. MM-GBSA binding calculations of the most potent inhibitors are more favourable. CONCLUSION The results of in-silico studies provide strong evidence for the consideration of valuable ligands in pyrazole substituted 9-anilinoacridines as potential HER2 inhibitors, and the compounds, 1v,s,r,d, a,o with significant Glide scores may produce significant anti-breast cancer activity for further development.
Collapse
Affiliation(s)
- Kalirajan Rajagopal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty 643001, [JSS Academy of Higher Education & Research-(Deemed to be University)], The Nilgiris (Tamilnadu), India
| | - Vulsi Bodhya Sri
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty 643001, [JSS Academy of Higher Education & Research-(Deemed to be University)], The Nilgiris (Tamilnadu), India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty 643001, [JSS Academy of Higher Education & Research-(Deemed to be University)], The Nilgiris (Tamilnadu), India
| | - Swaminathan Gomathi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty 643001, [JSS Academy of Higher Education & Research-(Deemed to be University)], The Nilgiris (Tamilnadu), India
| |
Collapse
|
18
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
19
|
AKT Inhibitors: The Road Ahead to Computational Modeling-Guided Discovery. Int J Mol Sci 2021; 22:ijms22083944. [PMID: 33920446 PMCID: PMC8070654 DOI: 10.3390/ijms22083944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/26/2022] Open
Abstract
AKT, is a serine/threonine protein kinase comprising three isoforms-namely: AKT1, AKT2 and AKT3, whose inhibitors have been recognized as promising therapeutic targets for various human disorders, especially cancer. In this work, we report a systematic evaluation of multi-target Quantitative Structure-Activity Relationship (mt-QSAR) models to probe AKT' inhibitory activity, based on different feature selection algorithms and machine learning tools. The best predictive linear and non-linear mt-QSAR models were found by the genetic algorithm-based linear discriminant analysis (GA-LDA) and gradient boosting (Xgboost) techniques, respectively, using a dataset containing 5523 inhibitors of the AKT isoforms assayed under various experimental conditions. The linear model highlighted the key structural attributes responsible for higher inhibitory activity whereas the non-linear model displayed an overall accuracy higher than 90%. Both these predictive models, generated through internal and external validation methods, were then used for screening the Asinex kinase inhibitor library to identify the most potential virtual hits as pan-AKT inhibitors. The virtual hits identified were then filtered by stepwise analyses based on reverse pharmacophore-mapping based prediction. Finally, results of molecular dynamics simulations were used to estimate the theoretical binding affinity of the selected virtual hits towards the three isoforms of enzyme AKT. Our computational findings thus provide important guidelines to facilitate the discovery of novel AKT inhibitors.
Collapse
|
20
|
Kleandrova VV, Scotti L, Bezerra Mendonça Junior FJ, Muratov E, Scotti MT, Speck-Planche A. QSAR Modeling for Multi-Target Drug Discovery: Designing Simultaneous Inhibitors of Proteins in Diverse Pathogenic Parasites. Front Chem 2021; 9:634663. [PMID: 33777898 PMCID: PMC7987820 DOI: 10.3389/fchem.2021.634663] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/22/2021] [Indexed: 11/21/2022] Open
Abstract
Parasitic diseases remain as unresolved health issues worldwide. While for some parasites the treatments involve drug combinations with serious side effects, for others, chemical therapies are inefficient due to the emergence of drug resistance. This urges the search for novel antiparasitic agents able to act through multiple mechanisms of action. Here, we report the first multi-target model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mt-QSAR-MLP) to virtually design and predict versatile inhibitors of proteins involved in the survival and/or infectivity of different pathogenic parasites. The mt-QSAR-MLP model exhibited high accuracy (>80%) in both training and test sets for the classification/prediction of protein inhibitors. Several fragments were directly extracted from the physicochemical and structural interpretations of the molecular descriptors in the mt-QSAR-MLP model. Such interpretations enabled the generation of four molecules that were predicted as multi-target inhibitors against at least three of the five parasitic proteins reported here with two of the molecules being predicted to inhibit all the proteins. Docking calculations converged with the mt-QSAR-MLP model regarding the multi-target profile of the designed molecules. The designed molecules exhibited drug-like properties, complying with Lipinski’s rule of five, as well as Ghose’s filter and Veber’s guidelines.
Collapse
Affiliation(s)
- Valeria V Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production, Moscow, Russian Federation
| | - Luciana Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Eugene Muratov
- Laboratory for Molecular Modeling, The UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Marcus T Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Alejandro Speck-Planche
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, Brazil
| |
Collapse
|
21
|
Sampaio-Dias IE, Rodríguez-Borges JE, Yáñez-Pérez V, Arrasate S, Llorente J, Brea JM, Bediaga H, Viña D, Loza MI, Caamaño O, García-Mera X, González-Díaz H. Synthesis, Pharmacological, and Biological Evaluation of 2-Furoyl-Based MIF-1 Peptidomimetics and the Development of a General-Purpose Model for Allosteric Modulators (ALLOPTML). ACS Chem Neurosci 2021; 12:203-215. [PMID: 33347281 DOI: 10.1021/acschemneuro.0c00687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
This work describes the synthesis and pharmacological evaluation of 2-furoyl-based Melanostatin (MIF-1) peptidomimetics as dopamine D2 modulating agents. Eight novel peptidomimetics were tested for their ability to enhance the maximal effect of tritiated N-propylapomorphine ([3H]-NPA) at D2 receptors (D2R). In this series, 2-furoyl-l-leucylglycinamide (6a) produced a statistically significant increase in the maximal [3H]-NPA response at 10 pM (11 ± 1%), comparable to the effect of MIF-1 (18 ± 9%) at the same concentration. This result supports previous evidence that the replacement of proline residue by heteroaromatic scaffolds are tolerated at the allosteric binding site of MIF-1. Biological assays performed for peptidomimetic 6a using cortex neurons from 19-day-old Wistar-Kyoto rat embryos suggest that 6a displays no neurotoxicity up to 100 μM. Overall, the pharmacological and toxicological profile and the structural simplicity of 6a makes this peptidomimetic a potential lead compound for further development and optimization, paving the way for the development of novel modulating agents of D2R suitable for the treatment of CNS-related diseases. Additionally, the pharmacological and biological data herein reported, along with >20 000 outcomes of preclinical assays, was used to seek a general model to predict the allosteric modulatory potential of molecular candidates for a myriad of target receptors, organisms, cell lines, and biological activity parameters based on perturbation theory (PT) ideas and machine learning (ML) techniques, abbreviated as ALLOPTML. By doing so, ALLOPTML shows high specificity Sp = 89.2/89.4%, sensitivity Sn = 71.3/72.2%, and accuracy Ac = 86.1%/86.4% in training/validation series, respectively. To the best of our knowledge, ALLOPTML is the first general-purpose chemoinformatic tool using a PTML-based model for the multioutput and multicondition prediction of allosteric compounds, which is expected to save both time and resources during the early drug discovery of allosteric modulators.
Collapse
Affiliation(s)
- Ivo E. Sampaio-Dias
- LAQV/REQUIMTE, Dept. of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - José E. Rodríguez-Borges
- LAQV/REQUIMTE, Dept. of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Víctor Yáñez-Pérez
- Dept. of Organic Chemistry II, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
| | - Sonia Arrasate
- Dept. of Pharmacology, Faculty of Medicine and Nursing, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
| | - Javier Llorente
- Dept. of Pharmacology, Faculty of Medicine and Nursing, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
- Dept. of Pharmacology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José M. Brea
- Innopharma Screening Platform, Biofarma Research group, Centre of Research in Molecular Medicine and Chronic Diseases CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Harbil Bediaga
- Dept. of Organic Chemistry II, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
- Dept. of Physical Chemistry, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
| | - Dolores Viña
- Dept. of Pharmacology, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Centre of Research in Molecular Medicine and Chronic Diseases CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María Isabel Loza
- Innopharma Screening Platform, Biofarma Research group, Centre of Research in Molecular Medicine and Chronic Diseases CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Olga Caamaño
- Dept. of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Xerardo García-Mera
- Dept. of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Humberto González-Díaz
- Dept. of Organic Chemistry II, University of Basque Country (UPV-EHU), 48940 Leioa, Spain
- Basque Center for Biophysics (CSIC UPV/EHU), University of Basque Country (UPV-EHU), 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
22
|
Chemoinformatics and QSAR. Adv Bioinformatics 2021. [DOI: 10.1007/978-981-33-6191-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
23
|
Cabrera-Andrade A, López-Cortés A, Jaramillo-Koupermann G, González-Díaz H, Pazos A, Munteanu CR, Pérez-Castillo Y, Tejera E. A Multi-Objective Approach for Anti-Osteosarcoma Cancer Agents Discovery through Drug Repurposing. Pharmaceuticals (Basel) 2020; 13:ph13110409. [PMID: 33266378 PMCID: PMC7700154 DOI: 10.3390/ph13110409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor. Although nowadays 5-year survival rates can reach up to 60–70%, acute complications and late effects of osteosarcoma therapy are two of the limiting factors in treatments. We developed a multi-objective algorithm for the repurposing of new anti-osteosarcoma drugs, based on the modeling of molecules with described activity for HOS, MG63, SAOS2, and U2OS cell lines in the ChEMBL database. Several predictive models were obtained for each cell line and those with accuracy greater than 0.8 were integrated into a desirability function for the final multi-objective model. An exhaustive exploration of model combinations was carried out to obtain the best multi-objective model in virtual screening. For the top 1% of the screened list, the final model showed a BEDROC = 0.562, EF = 27.6, and AUC = 0.653. The repositioning was performed on 2218 molecules described in DrugBank. Within the top-ranked drugs, we found: temsirolimus, paclitaxel, sirolimus, everolimus, and cabazitaxel, which are antineoplastic drugs described in clinical trials for cancer in general. Interestingly, we found several broad-spectrum antibiotics and antiretroviral agents. This powerful model predicts several drugs that should be studied in depth to find new chemotherapy regimens and to propose new strategies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Correspondence: (A.C.-A.); (E.T.)
| | - Andrés López-Cortés
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), 28029 Madrid, Spain
| | - Gabriela Jaramillo-Koupermann
- Laboratorio de Biología Molecular, Subproceso de Anatomía Patológica, Hospital de Especialidades Eugenio Espejo, Quito 170403, Ecuador;
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, and Basque Center for Biophysics CSIC-UPV/EHU, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Alejandro Pazos
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Cristian R. Munteanu
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito 170125, Ecuador
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de Las Américas, Quito 170125, Ecuador
- Correspondence: (A.C.-A.); (E.T.)
| |
Collapse
|
24
|
Ortega-Tenezaca B, Quevedo-Tumailli V, Bediaga H, Collados J, Arrasate S, Madariaga G, Munteanu CR, Cordeiro MND, González-Díaz H. PTML Multi-Label Algorithms: Models, Software, and Applications. Curr Top Med Chem 2020; 20:2326-2337. [DOI: 10.2174/1568026620666200916122616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
By combining Machine Learning (ML) methods with Perturbation Theory (PT), it is possible
to develop predictive models for a variety of response targets. Such combination often known as
Perturbation Theory Machine Learning (PTML) modeling comprises a set of techniques that can handle
various physical, and chemical properties of different organisms, complex biological or material
systems under multiple input conditions. In so doing, these techniques effectively integrate a manifold
of diverse chemical and biological data into a single computational framework that can then be applied
for screening lead chemicals as well as to find clues for improving the targeted response(s).
PTML models have thus been extremely helpful in drug or material design efforts and found to be
predictive and applicable across a broad space of systems. After a brief outline of the applied methodology,
this work reviews the different uses of PTML in Medicinal Chemistry, as well as in other
applications. Finally, we cover the development of software available nowadays for setting up PTML
models from large datasets.
Collapse
Affiliation(s)
| | | | - Harbil Bediaga
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Jon Collados
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Sonia Arrasate
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Gotzon Madariaga
- Department of Condensed Matter Physics, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Cristian R Munteanu
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, 15071 A Coruna, Spain
| | - M. Natália D.S. Cordeiro
- LAQV@REQUIMTE, Department of Chemistry and Biochemistry, University of Porto, 4169-007 Porto, Portugal
| | - Humbert González-Díaz
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| |
Collapse
|
25
|
Kleandrova VV, Scotti MT, Scotti L, Nayarisseri A, Speck-Planche A. Cell-based multi-target QSAR model for design of virtual versatile inhibitors of liver cancer cell lines. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2020; 31:815-836. [PMID: 32967475 DOI: 10.1080/1062936x.2020.1818617] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
Liver cancers are one of the leading fatal diseases among malignant neoplasms. Current chemotherapeutic treatments used to fight these illnesses have become less efficient in terms of both efficacy and safety. Therefore, there is a great need of search for new anti-liver cancer agents and this can be accelerated by using computer-aided drug discovery approaches. In this work, we report the development of the first cell-based multi-target model based on quantitative structure-activity relationships (CBMT-QSAR) for the design and prediction of chemicals as anticancer agents against 17 liver cancer cell lines. While having a good quality and predictive power (accuracy higher than 80%) in the training and test sets, respectively, the CBMT-QSAR model was employed as a tool to directly extract suitable fragments from the physicochemical and structural interpretations of the molecular descriptors. Some of these desirable fragments were assembled, leading to the virtual design of eight molecules with drug-like properties, with six of them being predicted as versatile anticancer agents against the 17 liver cancer cell lines reported here.
Collapse
Affiliation(s)
- V V Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production , Moscow, Russian Federation
| | - M T Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba , João Pessoa, Brazil
| | - L Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba , João Pessoa, Brazil
| | - A Nayarisseri
- In Silico Research Laboratory, Eminent Biosciences , Indore, Madhya Pradesh, India
| | - A Speck-Planche
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba , João Pessoa, Brazil
| |
Collapse
|
26
|
Cabrera-Andrade A, López-Cortés A, Munteanu CR, Pazos A, Pérez-Castillo Y, Tejera E, Arrasate S, González-Díaz H. Perturbation-Theory Machine Learning (PTML) Multilabel Model of the ChEMBL Dataset of Preclinical Assays for Antisarcoma Compounds. ACS OMEGA 2020; 5:27211-27220. [PMID: 33134682 PMCID: PMC7594149 DOI: 10.1021/acsomega.0c03356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Sarcomas are a group of malignant neoplasms of connective tissue with a different etiology than carcinomas. The efforts to discover new drugs with antisarcoma activity have generated large datasets of multiple preclinical assays with different experimental conditions. For instance, the ChEMBL database contains outcomes of 37,919 different antisarcoma assays with 34,955 different chemical compounds. Furthermore, the experimental conditions reported in this dataset include 157 types of biological activity parameters, 36 drug targets, 43 cell lines, and 17 assay organisms. Considering this information, we propose combining perturbation theory (PT) principles with machine learning (ML) to develop a PTML model to predict antisarcoma compounds. PTML models use one function of reference that measures the probability of a drug being active under certain conditions (protein, cell line, organism, etc.). In this paper, we used a linear discriminant analysis and neural network to train and compare PT and non-PT models. All the explored models have an accuracy of 89.19-95.25% for training and 89.22-95.46% in validation sets. PTML-based strategies have similar accuracy but generate simplest models. Therefore, they may become a versatile tool for predicting antisarcoma compounds.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Carrera
de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
| | - Andrés López-Cortés
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Centro
de Investigación Genética y Genómica, Facultad
de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, Quito 170129, Ecuador
| | - Cristian R. Munteanu
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Biomedical
Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
- Centro de
Investigación en Tecnologías de la Información
y las Comunicaciones (CITIC), Campus de
Elviña s/n, A Coruña 15071, Spain
| | - Alejandro Pazos
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Biomedical
Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Yunierkis Pérez-Castillo
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Escuela
de Ciencias Físicas y Matemáticas, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
| | - Eduardo Tejera
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Facultad
de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
| | - Sonia Arrasate
- Department
of Organic Chemistry II and Basque Center for Biophysics, University of Basque Country UPV/EHU, Leioa 48940, Biscay, Spain
| | - Humbert González-Díaz
- Department
of Organic Chemistry II and Basque Center for Biophysics, University of Basque Country UPV/EHU, Leioa 48940, Biscay, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48011, Biscay, Spain
| |
Collapse
|
27
|
Kleandrova VV, Speck-Planche A. PTML Modeling for Alzheimer’s Disease: Design and Prediction of Virtual Multi-Target Inhibitors of GSK3B, HDAC1, and HDAC6. Curr Top Med Chem 2020; 20:1661-1676. [DOI: 10.2174/1568026620666200607190951] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/12/2019] [Accepted: 01/05/2020] [Indexed: 01/23/2023]
Abstract
Background:
Alzheimer’s disease is characterized by a progressive pattern of cognitive and
functional impairment, which ultimately leads to death. Computational approaches have played an important
role in the context of drug discovery for anti-Alzheimer's therapies. However, most of the computational
models reported to date have been focused on only one protein associated with Alzheimer's,
while relying on small datasets of structurally related molecules.
Objective:
We introduce the first model combining perturbation theory and machine learning based on
artificial neural networks (PTML-ANN) for simultaneous prediction and design of inhibitors of three
Alzheimer’s disease-related proteins, namely glycogen synthase kinase 3 beta (GSK3B), histone deacetylase
1 (HDAC1), and histone deacetylase 6 (HDAC6).
Methods:
The PTML-ANN model was obtained from a dataset retrieved from ChEMBL, and it relied on
a classification approach to predict chemicals as active or inactive.
Results:
The PTML-ANN model displayed sensitivity and specificity higher than 85% in both training
and test sets. The physicochemical and structural interpretation of the molecular descriptors in the model
permitted the direct extraction of fragments suggested to favorably contribute to enhancing the multitarget
inhibitory activity. Based on this information, we assembled ten molecules from several fragments
with positive contributions. Seven of these molecules were predicted as triple target inhibitors while the
remaining three were predicted as dual-target inhibitors. The estimated physicochemical properties of
the designed molecules complied with Lipinski’s rule of five and its variants.
Conclusion:
This work opens new horizons toward the design of multi-target inhibitors for anti- Alzheimer's
therapies.
Collapse
Affiliation(s)
- Valeria V. Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production, Volokolamskoe Shosse 11, 125080, Moscow, Russian Federation
| | - Alejandro Speck-Planche
- Programa Institucional de Fomento a la Investigacion, Desarrollo e Innovacion, Universidad Tecnologica Metropolitana, Ignacio Valdivieso 2409, P.O. Box 8940577, San Joaquin, Santiago, Chile
| |
Collapse
|
28
|
Mansbach RA, Leus IV, Mehla J, Lopez CA, Walker JK, Rybenkov VV, Hengartner NW, Zgurskaya HI, Gnanakaran S. Machine Learning Algorithm Identifies an Antibiotic Vocabulary for Permeating Gram-Negative Bacteria. J Chem Inf Model 2020; 60:2838-2847. [PMID: 32453589 DOI: 10.1021/acs.jcim.0c00352] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Drug discovery faces a crisis. The industry has used up the "obvious" space in which to find novel drugs for biomedical applications, and productivity is declining. One strategy to combat this is rational approaches to expand the search space without relying on chemical intuition, to avoid rediscovery of similar spaces. In this work, we present proof of concept of an approach to rationally identify a "chemical vocabulary" related to a specific drug activity of interest without employing known rules. We focus on the pressing concern of multidrug resistance in Pseudomonas aeruginosa by searching for submolecules that promote compound entry into this bacterium. By synergizing theory, computation, and experiment, we validate our approach, explain the molecular mechanism behind identified fragments promoting compound entry, and select candidate compounds from an external library that display good permeation ability.
Collapse
Affiliation(s)
- Rachael A Mansbach
- Department of Theoretical Biology and Biophysics, Los Alamos National Lab, MS-K710, P.O. Box 1663, Los Alamos, New Mexico 87545-0001, United States
| | - Inga V Leus
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, SLSRC, Rm 1000, Norman, Oklahoma 73019-5251, United States
| | - Jitender Mehla
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, SLSRC, Rm 1000, Norman, Oklahoma 73019-5251, United States
| | - Cesar A Lopez
- Department of Theoretical Biology and Biophysics, Los Alamos National Lab, MS-K710, P.O. Box 1663, Los Alamos, New Mexico 87545-0001, United States
| | - John K Walker
- Pharmacology and Physiological Science, School of Medicine, Saint Louis University, Schwitalla Hall, Room M362, St. Louis, Missouri 63104, United States
| | - Valentin V Rybenkov
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, SLSRC, Rm 1000, Norman, Oklahoma 73019-5251, United States
| | - Nicolas W Hengartner
- Department of Theoretical Biology and Biophysics, Los Alamos National Lab, MS-K710, P.O. Box 1663, Los Alamos, New Mexico 87545-0001, United States
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, SLSRC, Rm 1000, Norman, Oklahoma 73019-5251, United States
| | - S Gnanakaran
- Department of Theoretical Biology and Biophysics, Los Alamos National Lab, MS-K710, P.O. Box 1663, Los Alamos, New Mexico 87545-0001, United States
| |
Collapse
|
29
|
Halder AK, Melo A, Cordeiro MNDS. A unified in silico model based on perturbation theory for assessing the genotoxicity of metal oxide nanoparticles. CHEMOSPHERE 2020; 244:125489. [PMID: 31812055 DOI: 10.1016/j.chemosphere.2019.125489] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
Nanomaterials (NMs) are an ever-increasing field of interest, due to their wide range of applications in science and technology. However, despite providing solutions to many societal problems and challenges, NMs are associated with adverse effects with potential severe damages towards biological species and their ecosystems. Particularly, it has been confirmed that NMs may induce serious genotoxic effects on various biological targets. Given the difficulties of experimental assays for estimating the genotoxicity of many NMs on diverse biological targets, development of alternative methodologies is crucial to establish their level of safety. In silico modelling approaches, such as Quantitative Structure-Toxicity Relationships (QSTR), are now considered a promising solution for such purpose. In this work, a perturbation theory machine learning (PTML) based QSTR approach is proposed for predicting the genotoxicity of metal oxide NMs under various experimental assay conditions. The application of such perturbation approach to 6084 NM-NM pair cases, set up from 78 unique NMs, afforded a final PTML-QSTR model with an accuracy better than 96% for both training and test sets. This model was then used to predict the genotoxicity of some NMs not included in the modelling dataset. The results for this independent data set were in excellent agreement with the experimental ones. Overall, that thus suggests that the derived PTML-QSTR model is a reliable in silico tool to rapidly and cost-efficiently assess the genotoxicity of metal oxide NMs. Finally, and most importantly, the model provides important insights regarding the mechanism of the genotoxicity triggered by these NMs.
Collapse
Affiliation(s)
- Amit Kumar Halder
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, University of Porto, 4169-007, Porto, Portugal.
| | - André Melo
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, University of Porto, 4169-007, Porto, Portugal
| | - M Natália D S Cordeiro
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, University of Porto, 4169-007, Porto, Portugal.
| |
Collapse
|
30
|
Lin X, Li X, Lin X. A Review on Applications of Computational Methods in Drug Screening and Design. Molecules 2020; 25:E1375. [PMID: 32197324 PMCID: PMC7144386 DOI: 10.3390/molecules25061375] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/27/2022] Open
Abstract
Drug development is one of the most significant processes in the pharmaceutical industry. Various computational methods have dramatically reduced the time and cost of drug discovery. In this review, we firstly discussed roles of multiscale biomolecular simulations in identifying drug binding sites on the target macromolecule and elucidating drug action mechanisms. Then, virtual screening methods (e.g., molecular docking, pharmacophore modeling, and QSAR) as well as structure- and ligand-based classical/de novo drug design were introduced and discussed. Last, we explored the development of machine learning methods and their applications in aforementioned computational methods to speed up the drug discovery process. Also, several application examples of combining various methods was discussed. A combination of different methods to jointly solve the tough problem at different scales and dimensions will be an inevitable trend in drug screening and design.
Collapse
Affiliation(s)
- Xiaoqian Lin
- Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China;
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiu Li
- School of Chemistry and Material Science, Shanxi Normal University, Linfen 041004, China;
| | - Xubo Lin
- Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China;
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
31
|
Yu JW, Yuan HW, Bao LD, Si LG. Interaction between piperine and genes associated with sciatica and its mechanism based on molecular docking technology and network pharmacology. Mol Divers 2020; 25:233-248. [PMID: 32130644 PMCID: PMC7870775 DOI: 10.1007/s11030-020-10055-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022]
Abstract
Abstract Piperine is the main active component of Piper longum L., which is also the main component of anti-sciatica Mongolian medicine Naru Sanwei pill. It has many pharmacological activities such as anti-inflammatory and immune regulation.
This paper aims to preliminarily explore the potential mechanism of piperine in the treatment of sciatica through network pharmacology and molecular docking. TCMSP, ETCM database and literature mining were used to collect the active compounds of Piper longum L. Swiss TargetPrediction and SuperPred server were used to find the targets of compounds. At the same time, CTD database was used to collect the targets of sciatica. Then the above targets were compared and analyzed to select the targets of anti-sciatica in Piper longum L. The Go (gene ontology) annotation and KEGG pathway of the targets were enriched and analyzed by Metascape database platform. The molecular docking between the effective components and the targets was verified by Autodock. After that, the sciatica model of rats was established and treated with piperine. The expression level of inflammatory factors and proteins in the serum and tissues of rat sciatic nerve were detected by ELISA and Western blot. HE staining and immunohistochemistry were carried out on the sciatica tissues of rats. The results showed that Piper longum L. can regulate the development of sciatica and affect the expressions of PPARG and NF-kB1 through its active ingredient piperine, and there is endogenous interaction between PPARG and NF-kB1. Graphic abstract ![]()
Collapse
Affiliation(s)
- Jiu-Wang Yu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, Inner Mongolia, People's Republic of China
| | - Hong-Wei Yuan
- Department of Pathology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, Inner Mongolia, People's Republic of China
| | - Li-Dao Bao
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, Inner Mongolia, People's Republic of China.
| | - Leng-Ge Si
- Mongolia Medical School, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, People's Republic of China
| |
Collapse
|
32
|
Wang Y, Wang LF, Zhang LL, Sun HB, Zhao J. Molecular mechanism of inhibitor bindings to bromodomain-containing protein 9 explored based on molecular dynamics simulations and calculations of binding free energies. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2020; 31:149-170. [PMID: 31851834 DOI: 10.1080/1062936x.2019.1701075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/02/2019] [Indexed: 06/10/2023]
Abstract
Recently, bromodomain-containing protein 9 (BRD9) has been a prospective therapeutic target for anticancer drug design. Molecular dynamics (MD) simulations combined with molecular mechanics generalized Born surface area (MM-GBSA) method were adopted to explore binding modes of three inhibitors (5SW, 5U2, and 5U6) to BRD9 and identify the hot spot of the inhibitor-BRD9 binding. The results indicate that the inhibitor 5SW has the strongest binding ability to BRD9 among the current three inhibitors. Furthermore, the rank of the binding free energies predicted by MM-GBSA approach agrees with that determined by the experimental values. In addition, inhibitor-residue interactions were computed by using residue-based free-energy decomposition method and the results suggest that residue His42 produces the CH-H interactions, residues Asn100, Ile53 and Val49 produce the CH-[Formula: see text] interactions with three inhibitors and Tyr106, Phe45 and Phe44 generate the π-π interactions with inhibitors. Notably, the residue Asn140 forms hydrogen bonding interactions with three inhibitors. This research is expected to provide useful molecular basis and dynamics information at atomic levels for the design of potent inhibitors inhibiting the activity of BRD9.
Collapse
Affiliation(s)
- Y Wang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - L F Wang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - L L Zhang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - H B Sun
- School of Science, Shandong Jiaotong University, Jinan, China
| | - J Zhao
- School of Science, Shandong Jiaotong University, Jinan, China
| |
Collapse
|
33
|
Carracedo-Reboredo P, Corona R, Martinez-Nunes M, Fernandez-Lozano C, Tsiliki G, Sarimveis H, Aranzamendi E, Arrasate S, Sotomayor N, Lete E, Munteanu CR, González-Díaz H. MCDCalc: Markov Chain Molecular Descriptors Calculator for Medicinal Chemistry. Curr Top Med Chem 2019; 20:305-317. [PMID: 31878856 DOI: 10.2174/1568026620666191226092431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 11/22/2022]
Abstract
AIMS Cheminformatics models are able to predict different outputs (activity, property, chemical reactivity) in single molecules or complex molecular systems (catalyzed organic synthesis, metabolic reactions, nanoparticles, etc.). BACKGROUND Cheminformatics models are able to predict different outputs (activity, property, chemical reactivity) in single molecules or complex molecular systems (catalyzed organic synthesis, metabolic reactions, nanoparticles, etc.). OBJECTIVE Cheminformatics prediction of complex catalytic enantioselective reactions is a major goal in organic synthesis research and chemical industry. Markov Chain Molecular Descriptors (MCDs) have been largely used to solve Cheminformatics problems. There are different types of Markov chain descriptors such as Markov-Shannon entropies (Shk), Markov Means (Mk), Markov Moments (πk), etc. However, there are other possible MCDs that have not been used before. In addition, the calculation of MCDs is done very often using specific software not always available for general users and there is not an R library public available for the calculation of MCDs. This fact, limits the availability of MCMDbased Cheminformatics procedures. METHODS We studied the enantiomeric excess ee(%)[Rcat] for 324 α-amidoalkylation reactions. These reactions have a complex mechanism depending on various factors. The model includes MCDs of the substrate, solvent, chiral catalyst, product along with values of time of reaction, temperature, load of catalyst, etc. We tested several Machine Learning regression algorithms. The Random Forest regression model has R2 > 0.90 in training and test. Secondly, the biological activity of 5644 compounds against colorectal cancer was studied. RESULTS We developed very interesting model able to predict with Specificity and Sensitivity 70-82% the cases of preclinical assays in both training and validation series. CONCLUSION The work shows the potential of the new tool for computational studies in organic and medicinal chemistry.
Collapse
Affiliation(s)
- Paula Carracedo-Reboredo
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071, A Coruña, Spain.,Group of Artificial Neural Networks and Adaptative Systems, Medical Imaging, and Diagnostic Radiology (RNASA-IMEDIR), Institute of Biomedical Research of Coruna (INIBIC), Hospital Complex of University of A Coruna (CHUAC), Sergas, University of Coruna (UDC), Xubias de arriba 84, 15006, A Coruna, Spain.,Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Ramiro Corona
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Mikel Martinez-Nunes
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Carlos Fernandez-Lozano
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071, A Coruña, Spain.,Group of Artificial Neural Networks and Adaptative Systems, Medical Imaging, and Diagnostic Radiology (RNASA-IMEDIR), Institute of Biomedical Research of Coruna (INIBIC), Hospital Complex of University of A Coruna (CHUAC), Sergas, University of Coruna (UDC), Xubias de arriba 84, 15006, A Coruna, Spain
| | - Georgia Tsiliki
- Institute for the Management of Information Systems, ATHENA Research and Innovation Centre, 15125, Athens, Greece
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, Zografou, Campus, 15780, Athens, Greece.,Pharma-Informatics Unit, ATHENA Research and Innovation Centre, 15125, Athens, Greece
| | - Eider Aranzamendi
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Sonia Arrasate
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Nuria Sotomayor
- Group of Artificial Neural Networks and Adaptative Systems, Medical Imaging, and Diagnostic Radiology (RNASA-IMEDIR), Institute of Biomedical Research of Coruna (INIBIC), Hospital Complex of University of A Coruna (CHUAC), Sergas, University of Coruna (UDC), Xubias de arriba 84, 15006, A Coruna, Spain
| | - Esther Lete
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain
| | - Cristian Robert Munteanu
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071, A Coruña, Spain.,Group of Artificial Neural Networks and Adaptative Systems, Medical Imaging, and Diagnostic Radiology (RNASA-IMEDIR), Institute of Biomedical Research of Coruna (INIBIC), Hospital Complex of University of A Coruna (CHUAC), Sergas, University of Coruna (UDC), Xubias de arriba 84, 15006, A Coruna, Spain
| | - Humbert González-Díaz
- Basque Center for Biophysics, University of the Basque Country UPV/EHU, 48940, Leioa, Bilbao, Spain.,IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| |
Collapse
|
34
|
Construction of Quantitative Structure Activity Relationship (QSAR) Models to Predict Potency of Structurally Diversed Janus Kinase 2 Inhibitors. Molecules 2019; 24:molecules24234393. [PMID: 31805692 PMCID: PMC6930640 DOI: 10.3390/molecules24234393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/24/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022] Open
Abstract
Janus kinase 2 (JAK2) inhibitors represent a promising therapeutic class of anticancer agents against many myeloproliferative disorders. Bioactivity data on pIC50 of 2229 JAK2 inhibitors were employed in the construction of quantitative structure-activity relationship (QSAR) models. The models were built from 100 data splits using decision tree (DT), support vector machine (SVM), deep neural network (DNN) and random forest (RF). The predictive power of RF models were assessed via 10-fold cross validation, which afforded excellent predictive performance with R2 and RMSE of 0.74 ± 0.05 and 0.63 ± 0.05, respectively. Moreover, test set has excellent performance of R2 (0.75 ± 0.03) and RMSE (0.62 ± 0.04). In addition, Y-scrambling was utilized to evaluate the possibility of chance correlation of the predictive model. A thorough analysis of the substructure fingerprint count was conducted to provide insights on the inhibitory properties of JAK2 inhibitors. Molecular cluster analysis revealed that pyrazine scaffolds have nanomolar potency against JAK2.
Collapse
|
35
|
Dutt R, Garg V, Khatri N, Madan AK. Phytochemicals in Anticancer Drug Development. Anticancer Agents Med Chem 2019; 19:172-183. [PMID: 30398123 DOI: 10.2174/1871520618666181106115802] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/19/2017] [Accepted: 03/21/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND In spite of major technological advances in conventional therapies, cancer continues to remain the leading cause of mortality worldwide. Phytochemicals are gradually emerging as a rich source of effective but safer agents against many life-threatening diseases. METHODS Various phytochemicals with reported anticancer activity have been simply categorized into major phytoconstituents- alkaloids, polyphenols, saponins, tannins and terpenoids. RESULTS The adverse effects associated with currently available anticancer medications may be overcome by using plant-derived compounds either alone or in combination. Exploration of plant kingdom may provide new leads for the accelerated development of new anticancer agents. CONCLUSION Although numerous potent synthetic drugs have been introduced for cancer chemotherapy, yet their serious toxicity concerns to normal cells apart from drug resistance have emerged as the major obstacles for their clinical utility over a prolonged duration of time. Current status and potential of phytochemicals and their derivatives in cancer therapy have been briefly reviewed in the present manuscript.
Collapse
Affiliation(s)
- Rohit Dutt
- Department of Pharmacy, G.D. Goenka University, Gurgaon-122103, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, M. D. University, Rohtak-124001, India
| | - Naveen Khatri
- Faculty of Pharmaceutical Sciences, Pt. B. D. Sharma University of Health Sciences Rohtak- 124001, India
| | - Anil K Madan
- Faculty of Pharmaceutical Sciences, Pt. B. D. Sharma University of Health Sciences Rohtak- 124001, India
| |
Collapse
|
36
|
Speck-Planche A. Multiple Perspectives in Anti-cancer Drug Discovery: From old Targets and Natural Products to Innovative Computational Approaches. Anticancer Agents Med Chem 2019; 19:146-147. [PMID: 31298144 DOI: 10.2174/187152061902190418105054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Alejandro Speck-Planche
- Research Program on Biomedical Informatics (GRIB) Hospital del Mar Medical Research Institute (IMIM) Barcelona, Spain
| |
Collapse
|
37
|
Halder AK, Giri AK, Cordeiro MNDS. Multi-Target Chemometric Modelling, Fragment Analysis and Virtual Screening with ERK Inhibitors as Potential Anticancer Agents. Molecules 2019; 24:molecules24213909. [PMID: 31671605 PMCID: PMC6864583 DOI: 10.3390/molecules24213909] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/21/2019] [Accepted: 10/25/2019] [Indexed: 02/07/2023] Open
Abstract
Two isoforms of extracellular regulated kinase (ERK), namely ERK-1 and ERK-2, are associated with several cellular processes, the aberration of which leads to cancer. The ERK-1/2 inhibitors are thus considered as potential agents for cancer therapy. Multitarget quantitative structure–activity relationship (mt-QSAR) models based on the Box–Jenkins approach were developed with a dataset containing 6400 ERK inhibitors assayed under different experimental conditions. The first mt-QSAR linear model was built with linear discriminant analysis (LDA) and provided information regarding the structural requirements for better activity. This linear model was also utilised for a fragment analysis to estimate the contributions of ring fragments towards ERK inhibition. Then, the random forest (RF) technique was employed to produce highly predictive non-linear mt-QSAR models, which were used for screening the Asinex kinase library and identify the most potential virtual hits. The fragment analysis results justified the selection of the hits retrieved through such virtual screening. The latter were subsequently subjected to molecular docking and molecular dynamics simulations to understand their possible interactions with ERK enzymes. The present work, which utilises in-silico techniques such as multitarget chemometric modelling, fragment analysis, virtual screening, molecular docking and dynamics, may provide important guidelines to facilitate the discovery of novel ERK inhibitors.
Collapse
Affiliation(s)
- Amit Kumar Halder
- Department of Chemistry and Biochemistry, University of Porto, 4169-007 Porto, Portugal.
| | - Amal Kanta Giri
- Department of Chemistry and Biochemistry, University of Porto, 4169-007 Porto, Portugal.
| | | |
Collapse
|
38
|
Halder AK, Cordeiro MNDS. Development of Multi-Target Chemometric Models for the Inhibition of Class I PI3K Enzyme Isoforms: A Case Study Using QSAR-Co Tool. Int J Mol Sci 2019; 20:ijms20174191. [PMID: 31461863 PMCID: PMC6747073 DOI: 10.3390/ijms20174191] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
The present work aims at establishing multi-target chemometric models using the recently launched quantitative structure–activity relationship (QSAR)-Co tool for predicting the activity of inhibitor compounds against different isoforms of phosphoinositide 3-kinase (PI3K) under various experimental conditions. The inhibitors of class I phosphoinositide 3-kinase (PI3K) isoforms have emerged as potential therapeutic agents for the treatment of various disorders, especially cancer. The cell-based enzyme inhibition assay results of PI3K inhibitors were curated from the CHEMBL database. Factors such as the nature and mutation of cell lines that may significantly alter the assay outcomes were considered as important experimental elements for mt-QSAR model development. The models, in turn, were developed using two machine learning techniques as implemented in QSAR-Co: linear discriminant analysis (LDA) and random forest (RF). Both techniques led to models with high accuracy (ca. 90%). Several molecular fragments were extracted from the current dataset, and their quantitative contributions to the inhibitory activity against all the proteins and experimental conditions under study were calculated. This case study also demonstrates the utility of QSAR-Co tool in solving multi-factorial and complex chemometric problems. Additionally, the combination of different in silico methods employed in this work can serve as a valuable guideline to speed up early discovery of PI3K inhibitors.
Collapse
Affiliation(s)
- Amit Kumar Halder
- Department of Chemistry and Biochemistry, University of Porto, 4169-007 Porto, Portugal
| | | |
Collapse
|
39
|
Yang X, Wang Y, Byrne R, Schneider G, Yang S. Concepts of Artificial Intelligence for Computer-Assisted Drug Discovery. Chem Rev 2019; 119:10520-10594. [PMID: 31294972 DOI: 10.1021/acs.chemrev.8b00728] [Citation(s) in RCA: 422] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Artificial intelligence (AI), and, in particular, deep learning as a subcategory of AI, provides opportunities for the discovery and development of innovative drugs. Various machine learning approaches have recently (re)emerged, some of which may be considered instances of domain-specific AI which have been successfully employed for drug discovery and design. This review provides a comprehensive portrayal of these machine learning techniques and of their applications in medicinal chemistry. After introducing the basic principles, alongside some application notes, of the various machine learning algorithms, the current state-of-the art of AI-assisted pharmaceutical discovery is discussed, including applications in structure- and ligand-based virtual screening, de novo drug design, physicochemical and pharmacokinetic property prediction, drug repurposing, and related aspects. Finally, several challenges and limitations of the current methods are summarized, with a view to potential future directions for AI-assisted drug discovery and design.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Yifei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Ryan Byrne
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Gisbert Schneider
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| |
Collapse
|
40
|
Ambure P, Halder AK, González Díaz H, Cordeiro MNDS. QSAR-Co: An Open Source Software for Developing Robust Multitasking or Multitarget Classification-Based QSAR Models. J Chem Inf Model 2019; 59:2538-2544. [PMID: 31083984 DOI: 10.1021/acs.jcim.9b00295] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Quantitative structure-activity relationships (QSAR) modeling is a well-known computational technique with wide applications in fields such as drug design, toxicity predictions, nanomaterials, etc. However, QSAR researchers still face certain problems to develop robust classification-based QSAR models, especially while handling response data pertaining to diverse experimental and/or theoretical conditions. In the present work, we have developed an open source standalone software "QSAR-Co" (available to download at https://sites.google.com/view/qsar-co ) to setup classification-based QSAR models that allow mining the response data coming from multiple conditions. The software comprises two modules: (1) the Model development module and (2) the Screen/Predict module. This user-friendly software provides several functionalities required for developing a robust multitasking or multitarget classification-based QSAR model using linear discriminant analysis or random forest techniques, with appropriate validation, following the principles set by the Organisation for Economic Co-operation and Development (OECD) for applying QSAR models in regulatory assessments.
Collapse
Affiliation(s)
- Pravin Ambure
- LAQV@REQUIMTE, Department of Chemistry and Biochemistry , University of Porto , 4169-007 Porto , Portugal
| | - Amit Kumar Halder
- LAQV@REQUIMTE, Department of Chemistry and Biochemistry , University of Porto , 4169-007 Porto , Portugal
| | - Humbert González Díaz
- Department of Organic Chemistry II , University of Basque Country UPV/EHU , 48940 Leioa , Spain
| | - M Natália D S Cordeiro
- LAQV@REQUIMTE, Department of Chemistry and Biochemistry , University of Porto , 4169-007 Porto , Portugal
| |
Collapse
|
41
|
Kalirajan R, Pandiselvi A, Gowramma B, Balachandran P. In-silico Design, ADMET Screening, MM-GBSA Binding Free Energy of Some Novel Isoxazole Substituted 9-Anilinoacridines as HER2 Inhibitors Targeting Breast Cancer. Curr Drug Res Rev 2019; 11:118-128. [PMID: 31513003 DOI: 10.2174/2589977511666190912154817] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Human Epidermal development factor Receptor-2 (HER2) is a membrane tyrosine kinase which is overexpressed and gene amplified in human breast cancers. HER2 amplification and overexpression have been linked to important tumor cell proliferation and survival pathways for 20% of instances of breast cancer. 9-aminoacridines are significant DNA-intercalating agents because of their antiproliferative properties. OBJECTIVE Some novel isoxazole substituted 9-anilinoacridines(1a-z) were designed by in-silico technique for their HER2 inhibitory activity. Docking investigations of compounds 1a-z are performed against HER2 (PDB id-3PP0) by using Schrodinger suit 2016-2. METHODS Molecular docking study for the designed molecules 1a-z are performed by Glide module, in-silico ADMET screening by QikProp module and binding free energy by Prime-MMGBSA module of Schrodinger suit. The binding affinity of designed molecules 1a-z towards HER2 was chosen based on GLIDE score. RESULTS Many compounds showed good hydrophobic communications and hydrogen bonding associations to hinder HER2. The compounds 1a-z, aside from 1z have significant Glide scores in the scope of - 4.91 to - 10.59 when compared with the standard Ethacridine (- 4.23) and Tamoxifen (- 3.78). The in-silico ADMET properties are inside the suggested about drug likeness. MM-GBSA binding of the most intense inhibitor is positive. CONCLUSION The outcomes reveal that this study provides evidence for the consideration of isoxazole substituted 9-aminoacridine derivatives as potential HER2 inhibitors. The compounds, 1s,x,v,a,j,r with significant Glide scores may produce significant anti breast cancer activity and further in vitro and in vivo investigations may prove their therapeutic potential.
Collapse
Affiliation(s)
- Rajagopal Kalirajan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, A Constituent College of JSS Academy of Higher Education & Research-(Deemed to be University), Udhagamandalam - 643001 (Tamilnadu), India
| | - Arumugasamy Pandiselvi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, A Constituent College of JSS Academy of Higher Education & Research-(Deemed to be University), Udhagamandalam - 643001 (Tamilnadu), India
| | - Byran Gowramma
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, A Constituent College of JSS Academy of Higher Education & Research-(Deemed to be University), Udhagamandalam - 643001 (Tamilnadu), India
| | | |
Collapse
|
42
|
Speck-Planche A. Combining Ensemble Learning with a Fragment-Based Topological Approach To Generate New Molecular Diversity in Drug Discovery: In Silico Design of Hsp90 Inhibitors. ACS OMEGA 2018; 3:14704-14716. [PMID: 30555986 PMCID: PMC6289491 DOI: 10.1021/acsomega.8b02419] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/23/2018] [Indexed: 05/05/2023]
Abstract
Machine learning methods have revolutionized modern science, providing fast and accurate solutions to multiple problems. However, they are commonly treated as "black boxes". Therefore, in important scientific fields such as medicinal chemistry and drug discovery, machine learning methods are restricted almost exclusively to the task of performing predictions of large and heterogeneous data sets of chemicals. The lack of interpretability prevents the full exploitation of the machine learning models as generators of new chemical knowledge. This work focuses on the development of an ensemble learning model for the prediction and design of potent dual heat shock protein 90 (Hsp90) inhibitors. The model displays accuracy higher than 80% in both training and test sets. To use the ensemble model as a generator of new chemical knowledge, three steps were followed. First, a physicochemical and/or structural interpretation was provided for each molecular descriptor present in the ensemble learning model. Second, the term "pseudolinear equation" was introduced within the context of machine learning to calculate the relative quantitative contributions of different molecular fragments to the inhibitory activity against the two Hsp90 isoforms studied here. Finally, by assembling the fragments with positive contributions, new molecules were designed, being predicted as potent Hsp90 inhibitors. According to Lipinski's rule of five, the designed molecules were found to exhibit potentially good oral bioavailability, a primordial property that chemicals must have to pass early stages in drug discovery. The present approach based on the combination of ensemble learning and fragment-based topological design holds great promise in drug discovery, and it can be adapted and applied to many different scientific disciplines.
Collapse
|
43
|
Bediaga H, Arrasate S, González-Díaz H. PTML Combinatorial Model of ChEMBL Compounds Assays for Multiple Types of Cancer. ACS COMBINATORIAL SCIENCE 2018; 20:621-632. [PMID: 30240186 DOI: 10.1021/acscombsci.8b00090] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Determining the target proteins of new anticancer compounds is a very important task in Medicinal Chemistry. In this sense, chemists carry out preclinical assays with a high number of combinations of experimental conditions (c j). In fact, ChEMBL database contains outcomes of 65 534 different anticancer activity preclinical assays for 35 565 different chemical compounds (1.84 assays per compound). These assays cover different combinations of c j formed from >70 different biological activity parameters ( c0), >300 different drug targets ( c1), >230 cell lines ( c2), and 5 organisms of assay ( c3) or organisms of the target ( c4). It include a total of 45 833 assays in leukemia, 6227 assays in breast cancer, 2499 assays in ovarian cancer, 3499 in colon cancer, 3159 in lung cancer, 2750 in prostate cancer, 601 in melanoma, etc. This is a very complex data set with multiple Big Data features. This data is hard to be rationalized by researchers to extract useful relationships and predict new compounds. In this context, we propose to combine perturbation theory (PT) ideas and machine learning (ML) modeling to solve this combinatorial-like problem. In this work, we report a PTML (PT + ML) model for ChEMBL data set of preclinical assays of anticancer compounds. This is a simple linear model with only three variables. The model presented values of area under receiver operating curve = AUROC = 0.872, specificity = Sp(%) = 90.2, sensitivity = Sn(%) = 70.6, and overall accuracy = Ac(%) = 87.7 in training series. The model also have Sp(%) = 90.1, Sn(%) = 71.4, and Ac(%) = 87.8 in external validation series. The model use PT operators based on multicondition moving averages to capture all the complexity of the data set. We also compared the model with nonlinear artificial neural network (ANN) models obtaining similar results. This confirms the hypothesis of a linear relationship between the PT operators and the classification as anticancer compounds in different combinations of assay conditions. Last, we compared the model with other PTML models reported in the literature concluding that this is the only one PTML model able to predict activity against multiple types of cancer. This model is a simple but versatile tool for the prediction of the targets of anticancer compounds taking into consideration multiple combinations of experimental conditions in preclinical assays.
Collapse
Affiliation(s)
- Harbil Bediaga
- Department of Organic Chemistry II, University of Basque Country UPV/EHU, 48940, Leioa, Spain
| | - Sonia Arrasate
- Department of Organic Chemistry II, University of Basque Country UPV/EHU, 48940, Leioa, Spain
| | - Humbert González-Díaz
- Department of Organic Chemistry II, University of Basque Country UPV/EHU, 48940, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| |
Collapse
|
44
|
BET bromodomain inhibitors: fragment-based in silico design using multi-target QSAR models. Mol Divers 2018; 23:555-572. [PMID: 30421269 DOI: 10.1007/s11030-018-9890-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/30/2018] [Indexed: 12/17/2022]
Abstract
Epigenetics has become a focus of interest in drug discovery. In this sense, bromodomain-containing proteins have emerged as potential epigenetic targets in cancer research and other therapeutic areas. Several computational approaches have been applied to the prediction of bromodomain inhibitors. Nevertheless, such approaches have several drawbacks such as the fact that they predict activity against only one bromodomain-containing protein, using structurally related compounds. Also, there are no reports focused on meaningfully analyzing the physicochemical/structural features that are necessary for the design of a bromodomain inhibitor. This work describes the development of two different multi-target models based on quantitative structure-activity relationships (mt-QSAR) for the prediction and in silico design of multi-target bromodomain inhibitors against the proteins BRD2, BRD3, and BRD4. The first model relied on linear discriminant analysis (LDA) while the second focused on artificial neural networks. Both models exhibited accuracies higher than 85% in the dataset. Several molecular fragments were extracted, and their contributions to the inhibitory activity against the three BET proteins were calculated by the LDA model. Six molecules were designed by assembling the fragments with positive contributions, and they were predicted as multi-target BET bromodomain inhibitors by the two mt-QSAR models. Molecular docking calculations converged with the predictions performed by the mt-QSAR models, suggesting that the designed molecules can exhibit potent activity against the three BET proteins. These molecules complied with the Lipinski's rule of five.
Collapse
|
45
|
Recent advances in fragment-based computational drug design: tackling simultaneous targets/biological effects. Future Med Chem 2018; 10:2021-2024. [DOI: 10.4155/fmc-2018-0213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
46
|
Bian Y, Xie XQS. Computational Fragment-Based Drug Design: Current Trends, Strategies, and Applications. AAPS JOURNAL 2018; 20:59. [PMID: 29633051 DOI: 10.1208/s12248-018-0216-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/08/2018] [Indexed: 01/08/2023]
Abstract
Fragment-based drug design (FBDD) has become an effective methodology for drug development for decades. Successful applications of this strategy brought both opportunities and challenges to the field of Pharmaceutical Science. Recent progress in the computational fragment-based drug design provide an additional approach for future research in a time- and labor-efficient manner. Combining multiple in silico methodologies, computational FBDD possesses flexibilities on fragment library selection, protein model generation, and fragments/compounds docking mode prediction. These characteristics provide computational FBDD superiority in designing novel and potential compounds for a certain target. The purpose of this review is to discuss the latest advances, ranging from commonly used strategies to novel concepts and technologies in computational fragment-based drug design. Particularly, in this review, specifications and advantages are compared between experimental and computational FBDD, and additionally, limitations and future prospective are discussed and emphasized.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,NIH National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Xiang-Qun Sean Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,NIH National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Department of Computational Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.
| |
Collapse
|
47
|
Chen L, Zhuang C, Lu J, Jiang Y, Sheng C. Discovery of Novel KRAS-PDEδ Inhibitors by Fragment-Based Drug Design. J Med Chem 2018; 61:2604-2610. [PMID: 29510040 DOI: 10.1021/acs.jmedchem.8b00057] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Targeting KRAS-PDEδ protein-protein interactions with small molecules represents a promising opportunity for developing novel antitumor agents. However, current KRAS-PDEδ inhibitors are limited by poor cellular antitumor potency and the druggability of the target remains to be validated by new inhibitors. To tackle these challenges, herein, novel, highly potent KRAS-PDEδ inhibitors were identified by fragment-based drug design, providing promising lead compounds or chemical probes for investigating the biological functions and druggability of KRAS-PDEδ interaction.
Collapse
Affiliation(s)
- Long Chen
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , China
| | - Chunlin Zhuang
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , China
| | - Junjie Lu
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , China
| | - Yan Jiang
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , China
| | - Chunquan Sheng
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , China
| |
Collapse
|
48
|
Kang D, Pang X, Lian W, Xu L, Wang J, Jia H, Zhang B, Liu AL, Du GH. Discovery of VEGFR2 inhibitors by integrating naïve Bayesian classification, molecular docking and drug screening approaches. RSC Adv 2018; 8:5286-5297. [PMID: 35542432 PMCID: PMC9078101 DOI: 10.1039/c7ra12259d] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/27/2017] [Indexed: 02/06/2023] Open
Abstract
The high morbidity and mortality of cancer make it one of the leading causes of global death, thus it is an urgent need to develop effective drugs for cancer therapy.
Collapse
Affiliation(s)
- De Kang
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Xiaocong Pang
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Wenwen Lian
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Lvjie Xu
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Jinhua Wang
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Hao Jia
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Baoyue Zhang
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Ai-Lin Liu
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| | - Guan-Hua Du
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
- China
| |
Collapse
|
49
|
Antanasijević D, Antanasijević J, Trišović N, Ušćumlić G, Pocajt V. From Classification to Regression Multitasking QSAR Modeling Using a Novel Modular Neural Network: Simultaneous Prediction of Anticonvulsant Activity and Neurotoxicity of Succinimides. Mol Pharm 2017; 14:4476-4484. [PMID: 29130688 DOI: 10.1021/acs.molpharmaceut.7b00582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Succinimides, which contain a pharmacophore responsible for anticonvulsant activity, are frequently used antiepileptic drugs and the synthesis of their new derivatives with improved efficacy and tolerability presents an important task. Nowadays, multitarget/tasking methodologies focused on quantitative-structure activity relationships (mt-QSAR/mtk-QSAR) have an important role in the rational design of drugs since they enable simultaneous prediction of several standard measures of biological activities at diverse experimental conditions and against different biological targets. Relating to this very topic, the mt-QSAR/mtk-QSAR methodology can give only binary classification models, and as such, in this study a regression mtk-QSAR (rmtk-QSAR) model based on a novel modular neural network (MNN) has been proposed. The MNN uses standard classification mtk-QSAR models as input modules, while the regression is performed by the output module. The rmtk-QSAR model has been successfully developed for the simultaneous prediction of anticonvulsant activity and neurotoxicity of succinimides, with a satisfactory accuracy in testing (R2 = 0.87). Thus, the proposed mtk-QSAR regression method can be regarded as a viable alternative to the standard QSAR methodology.
Collapse
Affiliation(s)
- Davor Antanasijević
- Innovation Center of the Faculty of Technology and Metallurgy and ‡Faculty of Technology and Metallurgy, University of Belgrade , Karnegijeva 4, Belgrade 11120, Serbia
| | - Jelena Antanasijević
- Innovation Center of the Faculty of Technology and Metallurgy and ‡Faculty of Technology and Metallurgy, University of Belgrade , Karnegijeva 4, Belgrade 11120, Serbia
| | - Nemanja Trišović
- Innovation Center of the Faculty of Technology and Metallurgy and ‡Faculty of Technology and Metallurgy, University of Belgrade , Karnegijeva 4, Belgrade 11120, Serbia
| | - Gordana Ušćumlić
- Innovation Center of the Faculty of Technology and Metallurgy and ‡Faculty of Technology and Metallurgy, University of Belgrade , Karnegijeva 4, Belgrade 11120, Serbia
| | - Viktor Pocajt
- Innovation Center of the Faculty of Technology and Metallurgy and ‡Faculty of Technology and Metallurgy, University of Belgrade , Karnegijeva 4, Belgrade 11120, Serbia
| |
Collapse
|
50
|
Abstract
Inverse quantitative structure-activity relationship (QSAR) modeling encompasses the generation of compound structures from values of descriptors corresponding to high activity predicted with a given QSAR model. Structure generation proceeds from descriptor coordinates optimized for activity prediction. Herein, we concentrate on the first phase of the inverse QSAR process and introduce a new methodology for coordinate optimization, termed differential evolution (DE), that originated from computer science and engineering. Using simulation and compound activity data, we demonstrate that DE in combination with support vector regression (SVR) yields effective and robust predictions of optimized coordinates satisfying model constraints and requirements. For different compound activity classes, optimized coordinates are obtained that exclusively map to regions of high activity in feature space, represent novel positions for structure generation, and are chemically meaningful.
Collapse
Affiliation(s)
- Tomoyuki Miyao
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan.,Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Bonn, D-53113, Germany
| | - Kimito Funatsu
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Bonn, D-53113, Germany
| |
Collapse
|