51
|
Dagenais S, Russo L, Madsen A, Webster J, Becnel L. Use of Real-World Evidence to Drive Drug Development Strategy and Inform Clinical Trial Design. Clin Pharmacol Ther 2022; 111:77-89. [PMID: 34839524 PMCID: PMC9299990 DOI: 10.1002/cpt.2480] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 10/30/2021] [Indexed: 12/28/2022]
Abstract
Interest in real-world data (RWD) and real-world evidence (RWE) to expedite and enrich the development of new biopharmaceutical products has proliferated in recent years, spurred by the 21st Century Cures Act in the United States and similar policy efforts in other countries, willingness by regulators to consider RWE in their decisions, demands from third-party payers, and growing concerns about the limitations of traditional clinical trials. Although much of the recent literature on RWE has focused on potential regulatory uses (e.g., product approvals in oncology or rare diseases based on single-arm trials with external control arms), this article reviews how biopharmaceutical companies can leverage RWE to inform internal decisions made throughout the product development process. Specifically, this article will review use of RWD to guide pipeline and portfolio strategy; use of novel sources of RWD to inform product development, use of RWD to inform clinical development, use of advanced analytics to harness "big" RWD, and considerations when using RWD to inform internal decisions. Topics discussed will include the use of molecular, clinicogenomic, medical imaging, radiomic, and patient-derived xenograft data to augment traditional sources of RWE, the use of RWD to inform clinical trial eligibility criteria, enrich trial population based on predicted response, select endpoints, estimate sample size, understand disease progression, and enhance diversity of participants, the growing use of data tokenization and advanced analytical techniques based on artificial intelligence in RWE, as well as the importance of data quality and methodological transparency in RWE.
Collapse
Affiliation(s)
| | - Leo Russo
- Global Medical Epidemiology, Worldwide Medical and SafetyPfizer IncCollegevillePennsylvaniaUSA
| | - Ann Madsen
- Global Medical Epidemiology, Worldwide Medical and SafetyPfizer IncNew YorkNew YorkUSA
| | - Jen Webster
- Real World EvidencePfizer IncNew YorkNew YorkUSA
| | | |
Collapse
|
52
|
Ulloa-Aguirre A, Zariñán T, Gutiérrez-Sagal R, Tao YX. Targeting trafficking as a therapeutic avenue for misfolded GPCRs leading to endocrine diseases. Front Endocrinol (Lausanne) 2022; 13:934685. [PMID: 36093106 PMCID: PMC9452723 DOI: 10.3389/fendo.2022.934685] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/13/2022] [Indexed: 02/05/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are plasma membrane proteins associated with an array of functions. Mutations in these receptors lead to a number of genetic diseases, including diseases involving the endocrine system. A particular subset of loss-of-function mutant GPCRs are misfolded receptors unable to traffic to their site of function (i.e. the cell surface plasma membrane). Endocrine disorders in humans caused by GPCR misfolding include, among others, hypo- and hyper-gonadotropic hypogonadism, morbid obesity, familial hypocalciuric hypercalcemia and neonatal severe hyperparathyroidism, X-linked nephrogenic diabetes insipidus, congenital hypothyroidism, and familial glucocorticoid resistance. Several in vitro and in vivo experimental approaches have been employed to restore function of some misfolded GPCRs linked to endocrine disfunction. The most promising approach is by employing pharmacological chaperones or pharmacoperones, which assist abnormally and incompletely folded proteins to refold correctly and adopt a more stable configuration to pass the scrutiny of the cell's quality control system, thereby correcting misrouting. This review covers the most important aspects that regulate folding and traffic of newly synthesized proteins, as well as the experimental approaches targeted to overcome protein misfolding, with special focus on GPCRs involved in endocrine diseases.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre,
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology & Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| |
Collapse
|
53
|
Brooks-Warburton J, Ashton J, Dhar A, Tham T, Allen PB, Hoque S, Lovat LB, Sebastian S. Artificial intelligence and inflammatory bowel disease: practicalities and future prospects. Frontline Gastroenterol 2021; 13:325-331. [PMID: 35722596 PMCID: PMC9186028 DOI: 10.1136/flgastro-2021-102003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/16/2021] [Indexed: 02/04/2023] Open
Abstract
Artificial intelligence (AI) is an emerging technology predicted to have significant applications in healthcare. This review highlights AI applications that impact the patient journey in inflammatory bowel disease (IBD), from genomics to endoscopic applications in disease classification, stratification and self-monitoring to risk stratification for personalised management. We discuss the practical AI applications currently in use while giving a balanced view of concerns and pitfalls and look to the future with the potential of where AI can provide significant value to the care of the patient with IBD.
Collapse
Affiliation(s)
- Johanne Brooks-Warburton
- Department of Clinical Pharmacology and Biological Sciences, University of Hertfordshire, Hatfield, UK,Gastroenterology Department, Lister Hospital, Stevenage, UK
| | - James Ashton
- Paediatric Gastroenterology, Southampton University Hospitals NHS Trust, Southampton, UK
| | - Anjan Dhar
- Gastroenterology, County Durham & Darlington NHS Foundation Trust, Bishop Auckland, UK
| | - Tony Tham
- Department of Gastroenterology, Ulster Hospital, Dundonald, UK
| | - Patrick B Allen
- Department of Gastroenterology, Ulster Hospital, Dundonald, UK
| | - Sami Hoque
- Department of Gastroenterology, Barts Health NHS Trust, London, UK
| | - Laurence B Lovat
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Shaji Sebastian
- Department of Gastroenterology, Hull University Teaching Hospitals NHS Trust, Hull, UK,Hull York Medical School, Hull, UK
| |
Collapse
|
54
|
Thomas M, Boardman A, Garcia-Ortegon M, Yang H, de Graaf C, Bender A. Applications of Artificial Intelligence in Drug Design: Opportunities and Challenges. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2390:1-59. [PMID: 34731463 DOI: 10.1007/978-1-0716-1787-8_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Artificial intelligence (AI) has undergone rapid development in recent years and has been successfully applied to real-world problems such as drug design. In this chapter, we review recent applications of AI to problems in drug design including virtual screening, computer-aided synthesis planning, and de novo molecule generation, with a focus on the limitations of the application of AI therein and opportunities for improvement. Furthermore, we discuss the broader challenges imposed by AI in translating theoretical practice to real-world drug design; including quantifying prediction uncertainty and explaining model behavior.
Collapse
Affiliation(s)
- Morgan Thomas
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Andrew Boardman
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Miguel Garcia-Ortegon
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK.,Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, UK
| | - Hongbin Yang
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
55
|
Artificial intelligence for the discovery of novel antimicrobial agents for emerging infectious diseases. Drug Discov Today 2021; 27:1099-1107. [PMID: 34748992 PMCID: PMC8570449 DOI: 10.1016/j.drudis.2021.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 01/08/2023]
Abstract
The search for effective drugs to treat new and existing diseases is a laborious one requiring a large investment of capital, resources, and time. The coronavirus 2019 (COVID-19) pandemic has been a painful reminder of the lack of development of new antimicrobial agents to treat emerging infectious diseases. Artificial intelligence (AI) and other in silico techniques can drive a more efficient, cost-friendly approach to drug discovery by helping move potential candidates with better clinical tolerance forward in the pipeline. Several research teams have developed successful AI platforms for hit identification, lead generation, and lead optimization. In this review, we investigate the technologies at the forefront of spearheading an AI revolution in drug discovery and pharmaceutical sciences.
Collapse
|
56
|
Zagidullin B, Wang Z, Guan Y, Pitkänen E, Tang J. Comparative analysis of molecular fingerprints in prediction of drug combination effects. Brief Bioinform 2021; 22:bbab291. [PMID: 34401895 PMCID: PMC8574997 DOI: 10.1093/bib/bbab291] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/01/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Application of machine and deep learning methods in drug discovery and cancer research has gained a considerable amount of attention in the past years. As the field grows, it becomes crucial to systematically evaluate the performance of novel computational solutions in relation to established techniques. To this end, we compare rule-based and data-driven molecular representations in prediction of drug combination sensitivity and drug synergy scores using standardized results of 14 high-throughput screening studies, comprising 64 200 unique combinations of 4153 molecules tested in 112 cancer cell lines. We evaluate the clustering performance of molecular representations and quantify their similarity by adapting the Centered Kernel Alignment metric. Our work demonstrates that to identify an optimal molecular representation type, it is necessary to supplement quantitative benchmark results with qualitative considerations, such as model interpretability and robustness, which may vary between and throughout preclinical drug development projects.
Collapse
Affiliation(s)
- B Zagidullin
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
| | - Z Wang
- Department of Electrical Engineering & Computer Science, University of Michigan, Ann Arbor, USA
| | - Y Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, USA
| | - E Pitkänen
- Institute for Molecular Medicine Finland (FIMM) & Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Finland
| | - J Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
| |
Collapse
|
57
|
Deng J, Yang Z, Ojima I, Samaras D, Wang F. Artificial intelligence in drug discovery: applications and techniques. Brief Bioinform 2021; 23:6420092. [PMID: 34734228 DOI: 10.1093/bib/bbab430] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/02/2021] [Accepted: 09/18/2021] [Indexed: 12/23/2022] Open
Abstract
Artificial intelligence (AI) has been transforming the practice of drug discovery in the past decade. Various AI techniques have been used in many drug discovery applications, such as virtual screening and drug design. In this survey, we first give an overview on drug discovery and discuss related applications, which can be reduced to two major tasks, i.e. molecular property prediction and molecule generation. We then present common data resources, molecule representations and benchmark platforms. As a major part of the survey, AI techniques are dissected into model architectures and learning paradigms. To reflect the technical development of AI in drug discovery over the years, the surveyed works are organized chronologically. We expect that this survey provides a comprehensive review on AI in drug discovery. We also provide a GitHub repository with a collection of papers (and codes, if applicable) as a learning resource, which is regularly updated.
Collapse
Affiliation(s)
- Jianyuan Deng
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11790, USA
| | - Zhibo Yang
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11790, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11790, USA
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11790, USA
| | - Fusheng Wang
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11790, USA.,Department of Computer Science, Stony Brook University, Stony Brook, NY 11790, USA
| |
Collapse
|
58
|
Zhou Y, Zhang Y, Lian X, Li F, Wang C, Zhu F, Qiu Y, Chen Y. Therapeutic target database update 2022: facilitating drug discovery with enriched comparative data of targeted agents. Nucleic Acids Res 2021; 50:D1398-D1407. [PMID: 34718717 PMCID: PMC8728281 DOI: 10.1093/nar/gkab953] [Citation(s) in RCA: 319] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/14/2022] Open
Abstract
Drug discovery relies on the knowledge of not only drugs and targets, but also the comparative agents and targets. These include poor binders and non-binders for developing discovery tools, prodrugs for improved therapeutics, co-targets of therapeutic targets for multi-target strategies and off-target investigations, and the collective structure-activity and drug-likeness landscapes of enhanced drug feature. However, such valuable data are inadequately covered by the available databases. In this study, a major update of the Therapeutic Target Database, previously featured in NAR, was therefore introduced. This update includes (a) 34 861 poor binders and 12 683 non-binders of 1308 targets; (b) 534 prodrug-drug pairs for 121 targets; (c) 1127 co-targets of 672 targets regulated by 642 approved and 624 clinical trial drugs; (d) the collective structure-activity landscapes of 427 262 active agents of 1565 targets; (e) the profiles of drug-like properties of 33 598 agents of 1102 targets. Moreover, a variety of additional data and function are provided, which include the cross-links to the target structure in PDB and AlphaFold, 159 and 1658 newly emerged targets and drugs, and the advanced search function for multi-entry target sequences or drug structures. The database is accessible without login requirement at: https://idrblab.org/ttd/.
Collapse
Affiliation(s)
- Ying Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Yintao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaoxin Wang
- Department of Computer Science, Kansas State University, Manhattan 66506, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.,Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| |
Collapse
|
59
|
Mak KK, Balijepalli MK, Pichika MR. Success stories of AI in drug discovery - where do things stand? Expert Opin Drug Discov 2021; 17:79-92. [PMID: 34553659 DOI: 10.1080/17460441.2022.1985108] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Artificial intelligence (AI) in drug discovery and development (DDD) has gained more traction in the past few years. Many scientific reviews have already been made available in this area. Thus, in this review, the authors have focused on the success stories of AI-driven drug candidates and the scientometric analysis of the literature in this field. AREA COVERED The authors explore the literature to compile the success stories of AI-driven drug candidates that are currently being assessed in clinical trials or have investigational new drug (IND) status. The authors also provide the reader with their expert perspectives for future developments and their opinions on the field. EXPERT OPINION Partnerships between AI companies and the pharma industry are booming. The early signs of the impact of AI on DDD are encouraging, and the pharma industry is hoping for breakthroughs. AI can be a promising technology to unveil the greatest successes, but it has yet to be proven as AI is still at the embryonic stage.
Collapse
Affiliation(s)
- Kit-Kay Mak
- School of Postgraduate Studies and Research, International Medical University, Bukit Jalil, Malaysia.,Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| | | | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| |
Collapse
|
60
|
Patrick Walters W. Comparing classification models-a practical tutorial. J Comput Aided Mol Des 2021; 36:381-389. [PMID: 34549368 DOI: 10.1007/s10822-021-00417-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023]
Abstract
While machine learning models have become a mainstay in Cheminformatics, the field has yet to agree on standards for model evaluation and comparison. In many cases, authors compare methods by performing multiple folds of cross-validation and reporting the mean value for an evaluation metric such as the area under the receiver operating characteristic. These comparisons of mean values often lack statistical rigor and can lead to inaccurate conclusions. In the interest of encouraging best practices, this tutorial provides an example of how multiple methods can be compared in a statistically rigorous fashion.
Collapse
|
61
|
Moreira-Filho JT, Silva AC, Dantas RF, Gomes BF, Souza Neto LR, Brandao-Neto J, Owens RJ, Furnham N, Neves BJ, Silva-Junior FP, Andrade CH. Schistosomiasis Drug Discovery in the Era of Automation and Artificial Intelligence. Front Immunol 2021; 12:642383. [PMID: 34135888 PMCID: PMC8203334 DOI: 10.3389/fimmu.2021.642383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/30/2021] [Indexed: 12/20/2022] Open
Abstract
Schistosomiasis is a parasitic disease caused by trematode worms of the genus Schistosoma and affects over 200 million people worldwide. The control and treatment of this neglected tropical disease is based on a single drug, praziquantel, which raises concerns about the development of drug resistance. This, and the lack of efficacy of praziquantel against juvenile worms, highlights the urgency for new antischistosomal therapies. In this review we focus on innovative approaches to the identification of antischistosomal drug candidates, including the use of automated assays, fragment-based screening, computer-aided and artificial intelligence-based computational methods. We highlight the current developments that may contribute to optimizing research outputs and lead to more effective drugs for this highly prevalent disease, in a more cost-effective drug discovery endeavor.
Collapse
Affiliation(s)
- José T. Moreira-Filho
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás – UFG, Goiânia, Brazil
| | - Arthur C. Silva
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás – UFG, Goiânia, Brazil
| | - Rafael F. Dantas
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Barbara F. Gomes
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Lauro R. Souza Neto
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jose Brandao-Neto
- Diamond Light Source Ltd., Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | - Raymond J. Owens
- The Rosalind Franklin Institute, Harwell, United Kingdom
- Division of Structural Biology, The Wellcome Centre for Human Genetic, University of Oxford, Oxford, United Kingdom
| | - Nicholas Furnham
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Bruno J. Neves
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás – UFG, Goiânia, Brazil
| | - Floriano P. Silva-Junior
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Carolina H. Andrade
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás – UFG, Goiânia, Brazil
| |
Collapse
|
62
|
Kimber TB, Chen Y, Volkamer A. Deep Learning in Virtual Screening: Recent Applications and Developments. Int J Mol Sci 2021; 22:4435. [PMID: 33922714 PMCID: PMC8123040 DOI: 10.3390/ijms22094435] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 01/03/2023] Open
Abstract
Drug discovery is a cost and time-intensive process that is often assisted by computational methods, such as virtual screening, to speed up and guide the design of new compounds. For many years, machine learning methods have been successfully applied in the context of computer-aided drug discovery. Recently, thanks to the rise of novel technologies as well as the increasing amount of available chemical and bioactivity data, deep learning has gained a tremendous impact in rational active compound discovery. Herein, recent applications and developments of machine learning, with a focus on deep learning, in virtual screening for active compound design are reviewed. This includes introducing different compound and protein encodings, deep learning techniques as well as frequently used bioactivity and benchmark data sets for model training and testing. Finally, the present state-of-the-art, including the current challenges and emerging problems, are examined and discussed.
Collapse
Affiliation(s)
| | | | - Andrea Volkamer
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (T.B.K.); (Y.C.)
| |
Collapse
|