1
|
Snyder SH, Vignaux PA, Ozalp MK, Gerlach J, Puhl AC, Lane TR, Corbett J, Urbina F, Ekins S. The Goldilocks paradigm: comparing classical machine learning, large language models, and few-shot learning for drug discovery applications. Commun Chem 2024; 7:134. [PMID: 38866916 PMCID: PMC11169557 DOI: 10.1038/s42004-024-01220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
Recent advances in machine learning (ML) have led to newer model architectures including transformers (large language models, LLMs) showing state of the art results in text generation and image analysis as well as few-shot learning (FSLC) models which offer predictive power with extremely small datasets. These new architectures may offer promise, yet the 'no-free lunch' theorem suggests that no single model algorithm can outperform at all possible tasks. Here, we explore the capabilities of classical (SVR), FSLC, and transformer models (MolBART) over a range of dataset tasks and show a 'goldilocks zone' for each model type, in which dataset size and feature distribution (i.e. dataset "diversity") determines the optimal algorithm strategy. When datasets are small ( < 50 molecules), FSLC tend to outperform both classical ML and transformers. When datasets are small-to-medium sized (50-240 molecules) and diverse, transformers outperform both classical models and few-shot learning. Finally, when datasets are of larger and of sufficient size, classical models then perform the best, suggesting that the optimal model to choose likely depends on the dataset available, its size and diversity. These findings may help to answer the perennial question of which ML algorithm is to be used when faced with a new dataset.
Collapse
Affiliation(s)
- Scott H Snyder
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Patricia A Vignaux
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Mustafa Kemal Ozalp
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - John Corbett
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| |
Collapse
|
2
|
Yang JJ, Goff A, Wild DJ, Ding Y, Annis A, Kerber R, Foote B, Passi A, Duerksen JL, London S, Puhl AC, Lane TR, Braunstein M, Waddell SJ, Ekins S. Computational drug repositioning identifies niclosamide and tribromsalan as inhibitors of Mycobacterium tuberculosis and Mycobacterium abscessus. Tuberculosis (Edinb) 2024; 146:102500. [PMID: 38432118 PMCID: PMC10978224 DOI: 10.1016/j.tube.2024.102500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/20/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Tuberculosis (TB) is still a major global health challenge, killing over 1.5 million people each year, and hence, there is a need to identify and develop novel treatments for Mycobacterium tuberculosis (M. tuberculosis). The prevalence of infections caused by nontuberculous mycobacteria (NTM) is also increasing and has overtaken TB cases in the United States and much of the developed world. Mycobacterium abscessus (M. abscessus) is one of the most frequently encountered NTM and is difficult to treat. We describe the use of drug-disease association using a semantic knowledge graph approach combined with machine learning models that has enabled the identification of several molecules for testing anti-mycobacterial activity. We established that niclosamide (M. tuberculosis IC90 2.95 μM; M. abscessus IC90 59.1 μM) and tribromsalan (M. tuberculosis IC90 76.92 μM; M. abscessus IC90 147.4 μM) inhibit M. tuberculosis and M. abscessus in vitro. To investigate the mode of action, we determined the transcriptional response of M. tuberculosis and M. abscessus to both compounds in axenic log phase, demonstrating a broad effect on gene expression that differed from known M. tuberculosis inhibitors. Both compounds elicited transcriptional responses indicative of respiratory pathway stress and the dysregulation of fatty acid metabolism.
Collapse
Affiliation(s)
- Jeremy J Yang
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA; Data2Discovery, Inc., Bloomington, IN, USA; Department of Internal Medicine Translational Informatics Division, University of New Mexico, Albuquerque, NM, USA
| | - Aaron Goff
- Department of Global Health and Infection, Brighton & Sussex Medical School, University of Sussex, UK
| | - David J Wild
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA; Data2Discovery, Inc., Bloomington, IN, USA
| | - Ying Ding
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA; Data2Discovery, Inc., Bloomington, IN, USA; School of Information, Dell Medical School, University of Texas, Austin, TX, USA
| | - Ayano Annis
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599, USA
| | | | | | - Anurag Passi
- Department of Pediatrics, UC San Diego, San Diego, CA, USA
| | | | | | - Ana C Puhl
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Thomas R Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Miriam Braunstein
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Simon J Waddell
- Department of Global Health and Infection, Brighton & Sussex Medical School, University of Sussex, UK
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| |
Collapse
|
3
|
Puhl AC, Raman R, Havener TM, Minerali E, Hickey AJ, Ekins S. Identification of New Modulators and Inhibitors of Palmitoyl-Protein Thioesterase 1 for CLN1 Batten Disease and Cancer. ACS OMEGA 2024; 9:11870-11882. [PMID: 38496939 PMCID: PMC10938339 DOI: 10.1021/acsomega.3c09607] [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: 12/01/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 03/19/2024]
Abstract
Palmitoyl-protein thioesterase 1 (PPT1) is an understudied enzyme that is gaining attention due to its role in the depalmitoylation of several proteins involved in neurodegenerative diseases and cancer. PPT1 is overexpressed in several cancers, specifically cholangiocarcinoma and esophageal cancers. Inhibitors of PPT1 lead to cell death and have been shown to enhance the killing of tumor cells alongside known chemotherapeutics. PPT1 is hence a viable target for anticancer drug development. Furthermore, mutations in PPT1 cause a lysosomal storage disorder called infantile neuronal ceroid lipofuscinosis (CLN1 disease). Molecules that can inhibit, stabilize, or modulate the activity of this target are needed to address these diseases. We used PPT1 enzymatic assays to identify molecules that were subsequently tested by using differential scanning fluorimetry and microscale thermophoresis. Selected compounds were also tested in neuroblastoma cell lines. The resulting PPT1 screening data was used for building machine learning models to help select additional compounds for testing. We discovered two of the most potent PPT1 inhibitors reported to date, orlistat (IC50 178.8 nM) and palmostatin B (IC50 11.8 nM). When tested in HepG2 cells, it was found that these molecules had decreased activity, indicating that they were likely not penetrating the cells. The combination of in vitro enzymatic and biophysical assays enabled the identification of several molecules that can bind or inhibit PPT1 and may aid in the discovery of modulators or chaperones. The molecules identified could be used as a starting point for further optimization as treatments for other potential therapeutic applications outside CLN1 disease, such as cancer and neurological diseases.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Renuka Raman
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Tammy M. Havener
- UNC
Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eni Minerali
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Anthony J. Hickey
- UNC
Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- RTI
International, Research Triangle
Park, North Carolina 27709, United States
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
4
|
Linciano P, Quotadamo A, Luciani R, Santucci M, Zorn KM, Foil DH, Lane TR, Cordeiro da Silva A, Santarem N, B Moraes C, Freitas-Junior L, Wittig U, Mueller W, Tonelli M, Ferrari S, Venturelli A, Gul S, Kuzikov M, Ellinger B, Reinshagen J, Ekins S, Costi MP. High-Throughput Phenotypic Screening and Machine Learning Methods Enabled the Selection of Broad-Spectrum Low-Toxicity Antitrypanosomatidic Agents. J Med Chem 2023; 66:15230-15255. [PMID: 37921561 PMCID: PMC10683024 DOI: 10.1021/acs.jmedchem.3c01322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Broad-spectrum anti-infective chemotherapy agents with activity against Trypanosomes, Leishmania, and Mycobacterium tuberculosis species were identified from a high-throughput phenotypic screening program of the 456 compounds belonging to the Ty-Box, an in-house industry database. Compound characterization using machine learning approaches enabled the identification and synthesis of 44 compounds with broad-spectrum antiparasitic activity and minimal toxicity against Trypanosoma brucei, Leishmania Infantum, and Trypanosoma cruzi. In vitro studies confirmed the predictive models identified in compound 40 which emerged as a new lead, featured by an innovative N-(5-pyrimidinyl)benzenesulfonamide scaffold and promising low micromolar activity against two parasites and low toxicity. Given the volume and complexity of data generated by the diverse high-throughput screening assays performed on the compounds of the Ty-Box library, the chemoinformatic and machine learning tools enabled the selection of compounds eligible for further evaluation of their biological and toxicological activities and aided in the decision-making process toward the design and optimization of the identified lead.
Collapse
Affiliation(s)
- Pasquale Linciano
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rosaria Luciani
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Matteo Santucci
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Kimberley M. Zorn
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H. Foil
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R. Lane
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Anabela Cordeiro da Silva
- Institute
for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto
de Investigaçao e Inovaçao em Saúde, Universidade do Porto and Institute for Molecular
and Cell Biology, 4150-180 Porto, Portugal
| | - Nuno Santarem
- Institute
for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto
de Investigaçao e Inovaçao em Saúde, Universidade do Porto and Institute for Molecular
and Cell Biology, 4150-180 Porto, Portugal
| | - Carolina B Moraes
- Brazilian
Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil
| | - Lucio Freitas-Junior
- Brazilian
Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil
| | - Ulrike Wittig
- Scientific
Databases and Visualization Group and Molecular and Cellular Modelling
Group, Heidelberg Institute for Theoretical
Studies (HITS), D-69118 Heidelberg, Germany
| | - Wolfgang Mueller
- Scientific
Databases and Visualization Group and Molecular and Cellular Modelling
Group, Heidelberg Institute for Theoretical
Studies (HITS), D-69118 Heidelberg, Germany
| | - Michele Tonelli
- Department
of Pharmacy, University of Genoa, Viale Benedetto XV n.3, 16132 Genoa, Italy
| | - Stefania Ferrari
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Alberto Venturelli
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- TYDOCK
PHARMA S.r.l., Strada
Gherbella 294/b, 41126 Modena, Italy
| | - Sheraz Gul
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Maria Paola Costi
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
5
|
Wang Y, Wang Z, Liu Y, Yu Q, Liu Y, Luo C, Wang S, Liu H, Liu M, Zhang G, Fan Y, Li K, Huang L, Duan M, Zhou F. Reconstructing the cytokine view for the multi-view prediction of COVID-19 mortality. BMC Infect Dis 2023; 23:622. [PMID: 37735372 PMCID: PMC10514938 DOI: 10.1186/s12879-023-08291-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 04/28/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a rapidly developing and sometimes lethal pulmonary disease. Accurately predicting COVID-19 mortality will facilitate optimal patient treatment and medical resource deployment, but the clinical practice still needs to address it. Both complete blood counts and cytokine levels were observed to be modified by COVID-19 infection. This study aimed to use inexpensive and easily accessible complete blood counts to build an accurate COVID-19 mortality prediction model. The cytokine fluctuations reflect the inflammatory storm induced by COVID-19, but their levels are not as commonly accessible as complete blood counts. Therefore, this study explored the possibility of predicting cytokine levels based on complete blood counts. METHODS We used complete blood counts to predict cytokine levels. The predictive model includes an autoencoder, principal component analysis, and linear regression models. We used classifiers such as support vector machine and feature selection models such as adaptive boost to predict the mortality of COVID-19 patients. RESULTS Complete blood counts and original cytokine levels reached the COVID-19 mortality classification area under the curve (AUC) values of 0.9678 and 0.9111, respectively, and the cytokine levels predicted by the feature set alone reached the classification AUC value of 0.9844. The predicted cytokine levels were more significantly associated with COVID-19 mortality than the original values. CONCLUSIONS Integrating the predicted cytokine levels and complete blood counts improved a COVID-19 mortality prediction model using complete blood counts only. Both the cytokine level prediction models and the COVID-19 mortality prediction models are publicly available at http://www.healthinformaticslab.org/supp/resources.php .
Collapse
Affiliation(s)
- Yueying Wang
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, 130012, Changchun, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 130021, Changchun, Jilin Province, China
| | - Zhao Wang
- College of Software, Jilin University, 130012, Changchun, China
| | - Yaqing Liu
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 130021, Changchun, Jilin Province, China
| | - Yujia Liu
- College of Software, Jilin University, 130012, Changchun, China
| | - Changfan Luo
- College of Software, Jilin University, 130012, Changchun, China
| | - Siyang Wang
- College of Software, Jilin University, 130012, Changchun, China
| | - Hongmei Liu
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, 130012, Changchun, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Mingyou Liu
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Gongyou Zhang
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Yusi Fan
- College of Software, Jilin University, 130012, Changchun, China
| | - Kewei Li
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Lan Huang
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Meiyu Duan
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China.
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, 130012, Changchun, China.
| | - Fengfeng Zhou
- College of Computer Science and Technology, Jilin University, 130012, Changchun, China.
- School of Biology and Engineering, Guizhou Medical University, 550025, Guiyang, Guizhou, China.
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, 130012, Changchun, China.
| |
Collapse
|
6
|
Lane T, Makarov V, Nelson JAE, Meeker RB, Sanna G, Riabova O, Kazakova E, Monakhova N, Tsedilin A, Urbina F, Jones T, Suchy A, Ekins S. N-Phenyl-1-(phenylsulfonyl)-1 H-1,2,4-triazol-3-amine as a New Class of HIV-1 Non-nucleoside Reverse Transcriptase Inhibitor. J Med Chem 2023; 66:6193-6217. [PMID: 37130343 PMCID: PMC10269403 DOI: 10.1021/acs.jmedchem.2c02055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Highly active antiretroviral therapy (HAART) has revolutionized human immunodeficiency virus (HIV) healthcare, turning it from a terminal to a potentially chronic disease, although some patients can develop severe comorbidities. These include neurological complications, such as HIV-associated neurocognitive disorders (HAND), which result in cognitive and/or motor function symptoms. We now describe the discovery, synthesis, and evaluation of a new class of N-phenyl-1-(phenylsulfonyl)-1H-1,2,4-triazol-3-amine HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTI) aimed at avoiding HAND. The most promising molecule, 12126065, exhibited antiviral activity against wild-type HIV-1 in TZM cells (EC50 = 0.24 nM) with low in vitro cytotoxicity (CC50 = 4.8 μM) as well as retained activity against clinically relevant HIV mutants. 12126065 also demonstrated no in vivo acute or subacute toxicity, good in vivo brain penetration, and minimal neurotoxicity in mouse neurons up to 10 μM, with a 50% toxicity concentration (TC50) of >100 μM, well below its EC50.
Collapse
Affiliation(s)
- Thomas Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Julie A. E. Nelson
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Rick B. Meeker
- Department of Neurology, University of North Carolina, NC 27514, USA
| | - Giuseppina Sanna
- Department of Biomedical Science, University of Cagliari, Monserrato, 09042, Italy
| | - Olga Riabova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Elena Kazakova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Natalia Monakhova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Andrey Tsedilin
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Fabio Urbina
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Thane Jones
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Ashley Suchy
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| |
Collapse
|
7
|
Novak J, Pathak P, Grishina MA, Potemkin VA. The design of compounds with desirable properties - The anti-HIV case study. J Comput Chem 2023; 44:1016-1030. [PMID: 36533526 DOI: 10.1002/jcc.27061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/14/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Efficacy and safety are among the most desirable characteristics of an ideal drug. The tremendous increase in computing power and the entry of artificial intelligence into the field of computational drug design are accelerating the process of identifying, developing, and optimizing potential drugs. Here, we present novel approach to design new molecules with desired properties. We combined various neural networks and linear regression algorithms to build models for cytotoxicity and anti-HIV activity based on Continual Molecular Interior analysis (CoMIn) and Cinderella's Shoe (CiS) derived molecular descriptors. After validating the reliability of the models, a genetic algorithm was coupled with the Des-Pot Grid algorithm to generate new molecules from a predefined pool of molecular fragments and predict their bioactivity and cytotoxicity. This combination led to the proposal of 16 hit molecules with high anti-HIV activity and low cytotoxicity. The anti-SARS-CoV-2 activity of the hits was predicted.
Collapse
Affiliation(s)
- Jurica Novak
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Center for Artificial Intelligence and Cybersecurity, University of Rijeka, Rijeka, Croatia
- Scientific and Educational Center "Biomedical Technologies", Higher Medical & Biological School, South Ural State University, Chelyabinsk, Russia
| | - Prateek Pathak
- Laboratory of Computational Modelling of Drugs, Higher Medical & Biological School, South Ural State University, Chelyabinsk, Russia
| | - Maria A Grishina
- Laboratory of Computational Modelling of Drugs, Higher Medical & Biological School, South Ural State University, Chelyabinsk, Russia
| | - Vladimir A Potemkin
- Laboratory of Computational Modelling of Drugs, Higher Medical & Biological School, South Ural State University, Chelyabinsk, Russia
| |
Collapse
|
8
|
Lane TR, Harris J, Urbina F, Ekins S. Comparing LD 50/LC 50 Machine Learning Models for Multiple Species. ACS CHEMICAL HEALTH & SAFETY 2023. [DOI: 10.1021/acs.chas.2c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Joshua Harris
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
9
|
Urbina F, Ekins S. The Commoditization of AI for Molecule Design. ARTIFICIAL INTELLIGENCE IN THE LIFE SCIENCES 2022; 2:100031. [PMID: 36211981 PMCID: PMC9541920 DOI: 10.1016/j.ailsci.2022.100031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Anyone involved in designing or finding molecules in the life sciences over the past few years has witnessed a dramatic change in how we now work due to the COVID-19 pandemic. Computational technologies like artificial intelligence (AI) seemed to become ubiquitous in 2020 and have been increasingly applied as scientists worked from home and were separated from the laboratory and their colleagues. This shift may be more permanent as the future of molecule design across different industries will increasingly require machine learning models for design and optimization of molecules as they become "designed by AI". AI and machine learning has essentially become a commodity within the pharmaceutical industry. This perspective will briefly describe our personal opinions of how machine learning has evolved and is being applied to model different molecule properties that crosses industries in their utility and ultimately suggests the potential for tight integration of AI into equipment and automated experimental pipelines. It will also describe how many groups have implemented generative models covering different architectures, for de novo design of molecules. We also highlight some of the companies at the forefront of using AI to demonstrate how machine learning has impacted and influenced our work. Finally, we will peer into the future and suggest some of the areas that represent the most interesting technologies that may shape the future of molecule design, highlighting how we can help increase the efficiency of the design-make-test cycle which is currently a major focus across industries.
Collapse
Affiliation(s)
- Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
10
|
Machine learning outperformed logistic regression classification even with limit sample size: A model to predict pediatric HIV mortality and clinical progression to AIDS. PLoS One 2022; 17:e0276116. [PMID: 36240212 PMCID: PMC9565414 DOI: 10.1371/journal.pone.0276116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 09/29/2022] [Indexed: 12/01/2022] Open
Abstract
Logistic regression (LR) is the most common prediction model in medicine. In recent years, supervised machine learning (ML) methods have gained popularity. However, there are many concerns about ML utility for small sample sizes. In this study, we aim to compare the performance of 7 algorithms in the prediction of 1-year mortality and clinical progression to AIDS in a small cohort of infants living with HIV from South Africa and Mozambique. The data set (n = 100) was randomly split into 70% training and 30% validation set. Seven algorithms (LR, Random Forest (RF), Support Vector Machine (SVM), K-Nearest Neighbor (KNN), Naïve Bayes (NB), Artificial Neural Network (ANN), and Elastic Net) were compared. The variables included as predictors were the same across the models including sociodemographic, virologic, immunologic, and maternal status features. For each of the models, a parameter tuning was performed to select the best-performing hyperparameters using 5 times repeated 10-fold cross-validation. A confusion-matrix was built to assess their accuracy, sensitivity, and specificity. RF ranked as the best algorithm in terms of accuracy (82,8%), sensitivity (78%), and AUC (0,73). Regarding specificity and sensitivity, RF showed better performance than the other algorithms in the external validation and the highest AUC. LR showed lower performance compared with RF, SVM, or KNN. The outcome of children living with perinatally acquired HIV can be predicted with considerable accuracy using ML algorithms. Better models would benefit less specialized staff in limited resources countries to improve prompt referral in case of high-risk clinical progression.
Collapse
|
11
|
Puhl AC, Gao ZG, Jacobson KA, Ekins S. Machine Learning for Discovery of New ADORA Modulators. Front Pharmacol 2022; 13:920643. [PMID: 35814244 PMCID: PMC9257522 DOI: 10.3389/fphar.2022.920643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 01/12/2023] Open
Abstract
Adenosine (ADO) is an extracellular signaling molecule generated locally under conditions that produce ischemia, hypoxia, or inflammation. It is involved in modulating a range of physiological functions throughout the brain and periphery through the membrane-bound G protein-coupled receptors, called adenosine receptors (ARs) A1AR, A2AAR, A2BAR, and A3AR. These are therefore important targets for neurological, cardiovascular, inflammatory, and autoimmune diseases and are the subject of drug development directed toward the cyclic adenosine monophosphate and other signaling pathways. Initially using public data for A1AR agonists we generated and validated a Bayesian machine learning model (Receiver Operator Characteristic of 0.87) that we used to identify molecules for testing. Three selected molecules, crisaborole, febuxostat and paroxetine, showed initial activity in vitro using the HEK293 A1AR Nomad cell line. However, radioligand binding, β-arrestin assay and calcium influx assay did not confirm this A1AR activity. Nevertheless, several other AR activities were identified. Febuxostat and paroxetine both inhibited orthosteric radioligand binding in the µM range for A2AAR and A3AR. In HEK293 cells expressing the human A2AAR, stimulation of cAMP was observed for crisaborole (EC50 2.8 µM) and paroxetine (EC50 14 µM), but not for febuxostat. Crisaborole also increased cAMP accumulation in A2BAR-expressing HEK293 cells, but it was weaker than at the A2AAR. At the human A3AR, paroxetine did not show any agonist activity at 100 µM, although it displayed binding with a Ki value of 14.5 µM, suggesting antagonist activity. We have now identified novel modulators of A2AAR, A2BAR and A3AR subtypes that are clinically used for other therapeutic indications, and which are structurally distinct from previously reported tool compounds or drugs.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States,*Correspondence: Ana C. Puhl, ; Sean Ekins,
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States,*Correspondence: Ana C. Puhl, ; Sean Ekins,
| |
Collapse
|
12
|
Chalcones from Angelica keiskei (ashitaba) inhibit key Zika virus replication proteins. Bioorg Chem 2022; 120:105649. [PMID: 35124513 PMCID: PMC9187613 DOI: 10.1016/j.bioorg.2022.105649] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 12/25/2022]
Abstract
Zika virus (ZIKV) is a dangerous human pathogen and no antiviral drugs have been approved to date. The chalcones are a group of small molecules that are found in a number of different plants, including Angelica keiskei Koidzumi, also known as ashitaba. To examine chalcone anti-ZIKV activity, three chalcones, 4-hydroxyderricin (4HD), xanthoangelol (XA), and xanthoangelol-E (XA-E), were purified from a methanol-ethyl acetate extract from A. keiskei. Molecular and ensemble docking predicted that these chalcones would establish multiple interactions with residues in the catalytic and allosteric sites of ZIKV NS2B-NS3 protease, and in the allosteric site of the NS5 RNA-dependent RNA-polymerase (RdRp). Machine learning models also predicted 4HD, XA and XA-E as potential anti-ZIKV inhibitors. Enzymatic and kinetic assays confirmed chalcone inhibition of the ZIKV NS2B-NS3 protease allosteric site with IC50s from 18 to 50 µM. Activity assays also revealed that XA, but not 4HD or XA-E, inhibited the allosteric site of the RdRp, with an IC50 of 6.9 µM. Finally, we tested these chalcones for their anti-viral activity in vitro with Vero cells. 4HD and XA-E displayed anti-ZIKV activity with EC50 values of 6.6 and 22.0 µM, respectively, while XA displayed relatively weak anti-ZIKV activity with whole cells. With their simple structures and relative ease of modification, the chalcones represent attractive candidates for hit-to-lead optimization in the search of new anti-ZIKV therapeutics.
Collapse
|
13
|
Lane TR, Urbina F, Rank L, Gerlach J, Riabova O, Lepioshkin A, Kazakova E, Vocat A, Tkachenko V, Cole S, Makarov V, Ekins S. Machine Learning Models for Mycobacterium tuberculosisIn Vitro Activity: Prediction and Target Visualization. Mol Pharm 2022; 19:674-689. [PMID: 34964633 PMCID: PMC9121329 DOI: 10.1021/acs.molpharmaceut.1c00791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tuberculosis (TB) is a major global health challenge, with approximately 1.4 million deaths per year. There is still a need to develop novel treatments for patients infected with Mycobacterium tuberculosis (Mtb). There have been many large-scale phenotypic screens that have led to the identification of thousands of new compounds. Yet, there is very limited investment in TB drug discovery which points to the need for new methods to increase the efficiency of drug discovery against Mtb. We have used machine learning approaches to learn from the public Mtb data, resulting in many data sets and models with robust enrichment and hit rates leading to the discovery of new active compounds. Recently, we have curated predominantly small-molecule Mtb data and developed new machine learning classification models with 18 886 molecules at different activity cutoffs. We now describe the further validation of these Bayesian models using a library of over 1000 molecules synthesized as part of EU-funded New Medicines for TB and More Medicines for TB programs. We highlight molecular features which are enriched in these active compounds. In addition, we provide new regression and classification models that can be used for scoring compound libraries or used to design new molecules. We have also visualized these molecules in the context of known molecular targets and identified clusters in chemical property space, which may aid in future target identification efforts. Finally, we are also making these data sets publicly available, representing a significant increase to the available Mtb inhibition data in the public domain.
Collapse
Affiliation(s)
- Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Laura Rank
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Olga Riabova
- Research Center of Biotechnology RAS, 119071 Moscow, Russia
| | | | - Elena Kazakova
- Research Center of Biotechnology RAS, 119071 Moscow, Russia
| | - Anthony Vocat
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Valery Tkachenko
- Science Data Experts, 14909 Forest Landing Cir, Rockville, MD 20850
| | | | - Vadim Makarov
- Research Center of Biotechnology RAS, 119071 Moscow, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| |
Collapse
|
14
|
Schmalstig AA, Zorn KM, Murci S, Robinson A, Savina S, Komarova E, Makarov V, Braunstein M, Ekins S. Mycobacterium abscessus drug discovery using machine learning. Tuberculosis (Edinb) 2022; 132:102168. [PMID: 35077930 PMCID: PMC8855326 DOI: 10.1016/j.tube.2022.102168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/30/2021] [Accepted: 01/14/2022] [Indexed: 01/22/2023]
Abstract
The prevalence of infections by nontuberculous mycobacteria is increasing, having surpassed tuberculosis in the United States and much of the developed world. Nontuberculous mycobacteria occur naturally in the environment and are a significant problem for patients with underlying lung diseases such as bronchiectasis, chronic obstructive pulmonary disease, and cystic fibrosis. Current treatment regimens are lengthy, complicated, toxic and they are often unsuccessful as seen by disease recurrence. Mycobacterium abscessus is one of the most commonly encountered organisms in nontuberculous mycobacteria disease and it is the most difficult to eradicate. There is currently no systematically proven regimen that is effective for treating M. abscessus infections. Our approach to drug discovery integrates machine learning, medicinal chemistry and in vitro testing and has been previously applied to Mycobacterium tuberculosis. We have now identified several novel 1-(phenylsulfonyl)-1H-benzimidazol-2-amines that have weak activity on M. abscessus in vitro but may represent a starting point for future further medicinal chemistry optimization. We also address limitations still to be overcome with the machine learning approach for M. abscessus.
Collapse
Affiliation(s)
- Alan A. Schmalstig
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
| | - Kimberley M. Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive Lab 3510, Raleigh, North Carolina, 27606, USA
| | - Sebastian Murci
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
| | - Andrew Robinson
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
| | - Svetlana Savina
- Research Center of Biotechnology RAS, Moscow, 119071, Russia
| | - Elena Komarova
- Research Center of Biotechnology RAS, Moscow, 119071, Russia
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Moscow, 119071, Russia
| | - Miriam Braunstein
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive Lab 3510, Raleigh, North Carolina, 27606, USA.,Corresponding author: Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive Lab 3510, Raleigh, North Carolina, 27606, USA.
| |
Collapse
|
15
|
Urbina F, Batra K, Luebke KJ, White JD, Matsiev D, Olson LL, Malerich JP, Hupcey MAZ, Madrid PB, Ekins S. UV-adVISor: Attention-Based Recurrent Neural Networks to Predict UV-Vis Spectra. Anal Chem 2021; 93:16076-16085. [PMID: 34812602 DOI: 10.1021/acs.analchem.1c03741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ultraviolet-visible (UV-Vis) absorption spectra are routinely collected as part of high-performance liquid chromatography (HPLC) analysis systems and can be used to identify chemical reaction products by comparison to the reference spectra. Here, we present UV-adVISor as a new computational tool for predicting the UV-Vis spectra from a molecule's structure alone. UV-Vis prediction was approached as a sequence-to-sequence problem. We utilized Long-Short Term Memory and attention-based neural networks with Extended Connectivity Fingerprint Diameter 6 or molecule SMILES to generate predictive models for the UV spectra. We have produced two spectrum datasets (dataset I, N = 949, and dataset II, N = 2222) using different compound collections and spectrum acquisition methods to train, validate, and test our models. We evaluated the prediction accuracy of the complete spectra by the correspondence of wavelengths of absorbance maxima and with a series of statistical measures (the best test set median model parameters are in parentheses for model II), including RMSE (0.064), R2 (0.71), and dynamic time warping (DTW, 0.194) of the entire spectrum curve. Scrambling molecule structures with the experimental spectra during training resulted in a degraded R2, confirming the utility of the approaches for prediction. UV-adVISor is able to provide fast and accurate predictions for libraries of compounds.
Collapse
Affiliation(s)
- Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kushal Batra
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States.,Computer Science, NC State University, Raleigh, North Carolina 27606, United States
| | - Kevin J Luebke
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Jason D White
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Daniel Matsiev
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Lori L Olson
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Jeremiah P Malerich
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Maggie A Z Hupcey
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Peter B Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
16
|
Calcium channels and iron metabolism: A redox catastrophe in Parkinson's disease and an innovative path to novel therapies? Redox Biol 2021; 47:102136. [PMID: 34653841 PMCID: PMC8517601 DOI: 10.1016/j.redox.2021.102136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 01/09/2023] Open
Abstract
Autonomously spiking dopaminergic neurons of the substantia nigra pars compacta (SNpc) are exquisitely specialized and suffer toxic iron-loading in Parkinson's disease (PD). However, the molecular mechanism involved remains unclear and critical to decipher for designing new PD therapeutics. The long-lasting (L-type) CaV1.3 voltage-gated calcium channel is expressed at high levels amongst nigral neurons of the SNpc, and due to its role in calcium and iron influx, could play a role in the pathogenesis of PD. Neuronal iron uptake via this route could be unregulated under the pathological setting of PD and potentiate cellular stress due to its redox activity. This Commentary will focus on the role of the CaV1.3 channels in calcium and iron uptake in the context of pharmacological targeting. Prospectively, the audacious use of artificial intelligence to design innovative CaV1.3 channel inhibitors could lead to breakthrough pharmaceuticals that attenuate calcium and iron entry to ameliorate PD pathology.
Collapse
|
17
|
Gawriljuk VO, Zin PPK, Puhl AC, Zorn KM, Foil DH, Lane TR, Hurst B, Tavella TA, Costa FTM, Lakshmanane P, Bernatchez J, Godoy AS, Oliva G, Siqueira-Neto JL, Madrid PB, Ekins S. Machine Learning Models Identify Inhibitors of SARS-CoV-2. J Chem Inf Model 2021; 61:4224-4235. [PMID: 34387990 DOI: 10.1021/acs.jcim.1c00683] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
With the rapidly evolving SARS-CoV-2 variants of concern, there is an urgent need for the discovery of further treatments for the coronavirus disease (COVID-19). Drug repurposing is one of the most rapid strategies for addressing this need, and numerous compounds have already been selected for in vitro testing by several groups. These have led to a growing database of molecules with in vitro activity against the virus. Machine learning models can assist drug discovery through prediction of the best compounds based on previously published data. Herein, we have implemented several machine learning methods to develop predictive models from recent SARS-CoV-2 in vitro inhibition data and used them to prioritize additional FDA-approved compounds for in vitro testing selected from our in-house compound library. From the compounds predicted with a Bayesian machine learning model, lumefantrine, an antimalarial was selected for testing and showed limited antiviral activity in cell-based assays while demonstrating binding (Kd 259 nM) to the spike protein using microscale thermophoresis. Several other compounds which we prioritized have since been tested by others and were also found to be active in vitro. This combined machine learning and in vitro testing approach can be expanded to virtually screen available molecules with predicted activity against SARS-CoV-2 reference WIV04 strain and circulating variants of concern. In the process of this work, we have created multiple iterations of machine learning models that can be used as a prioritization tool for SARS-CoV-2 antiviral drug discovery programs. The very latest model for SARS-CoV-2 with over 500 compounds is now freely available at www.assaycentral.org.
Collapse
Affiliation(s)
- Victor O Gawriljuk
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100-Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Phyo Phyo Kyaw Zin
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Brett Hurst
- Institute for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States.,Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah 84322-4815, United States
| | - Tatyana Almeida Tavella
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Premkumar Lakshmanane
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill North Carolina 27599, United States
| | - Jean Bernatchez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, California 92093, United States
| | - Andre S Godoy
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100-Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Glaucius Oliva
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100-Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, California 92093, United States
| | - Peter B Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
18
|
Gawriljuk VO, Foil DH, Puhl AC, Zorn KM, Lane TR, Riabova O, Makarov V, Godoy AS, Oliva G, Ekins S. Development of Machine Learning Models and the Discovery of a New Antiviral Compound against Yellow Fever Virus. J Chem Inf Model 2021; 61:3804-3813. [PMID: 34286575 DOI: 10.1021/acs.jcim.1c00460] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Yellow fever (YF) is an acute viral hemorrhagic disease transmitted by infected mosquitoes. Large epidemics of YF occur when the virus is introduced into heavily populated areas with high mosquito density and low vaccination coverage. The lack of a specific small molecule drug treatment against YF as well as for homologous infections, such as zika and dengue, highlights the importance of these flaviviruses as a public health concern. With the advancement in computer hardware and bioactivity data availability, new tools based on machine learning methods have been introduced into drug discovery, as a means to utilize the growing high throughput screening (HTS) data generated to reduce costs and increase the speed of drug development. The use of predictive machine learning models using previously published data from HTS campaigns or data available in public databases, can enable the selection of compounds with desirable bioactivity and absorption, distribution, metabolism, and excretion profiles. In this study, we have collated cell-based assay data for yellow fever virus from the literature and public databases. The data were used to build predictive models with several machine learning methods that could prioritize compounds for in vitro testing. Five molecules were prioritized and tested in vitro from which we have identified a new pyrazolesulfonamide derivative with EC50 3.2 μM and CC50 24 μM, which represents a new scaffold suitable for hit-to-lead optimization that can expand the available drug discovery candidates for YF.
Collapse
Affiliation(s)
- Victor O Gawriljuk
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100 - Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071 Moscow, Russia
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071 Moscow, Russia
| | - Andre S Godoy
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100 - Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Glaucius Oliva
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100 - Santa Angelina, São Carlos, São Paulo 13563-120, Brazil
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
19
|
Urbina F, Zorn KM, Brunner D, Ekins S. Comparing the Pfizer Central Nervous System Multiparameter Optimization Calculator and a BBB Machine Learning Model. ACS Chem Neurosci 2021; 12:2247-2253. [PMID: 34028255 DOI: 10.1021/acschemneuro.1c00265] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ability to calculate whether small molecules will cross the blood-brain barrier (BBB) is an important task for companies working in neuroscience drug discovery. For a decade, scientists have relied on relatively simplistic rules such as Pfizer's central nervous system multiparameter optimization models (CNS-MPO) for guidance during the drug selection process. In parallel, there has been a continued development of more sophisticated machine learning models that utilize different molecular descriptors and algorithms; however, these models represent a "black box" and are generally less interpretable. In both cases, these methods predict the ability of small molecules to cross the BBB using the molecular structure information on its own without in vitro or in vivo data. We describe here the implementation of two versions of Pfizer's algorithm (Pf-MPO.v1 and Pf-MPO.v2) and compare it with a Bayesian machine learning model of BBB penetration trained on a data set of 2296 active and inactive compounds using extended connectivity fingerprint descriptors. The predictive ability of these approaches was compared with 40 known CNS active drugs initially used by Pfizer as their positive set for validation of the Pf-MPO.v1 score. 37/40 (92.5%) compounds were predicted as active by the Bayesian model, while only 30/40 (75%) received a desirable Pf-MPO.v1 score ≥4 and 33/40 (82.5%) received a desirable Pf-MPO.v2 score ≥4, suggesting the Bayesian model is more accurate than MPO algorithms. This also indicates machine learning models are more flexible and have better predictive power for BBB penetration than simple rule sets that require multiple, accurate descriptor calculations. Our machine learning model statistics are comparable to recent published studies. We describe the implications of these findings and how machine learning may have a role alongside more interpretable methods.
Collapse
Affiliation(s)
- Fabio Urbina
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, United States
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kimberley M. Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniela Brunner
- PsychoGenics, 215 College Road, Paramus, New Jersey 07652, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
20
|
Donlin MJ, Lane TR, Riabova O, Lepioshkin A, Xu E, Lin J, Makarov V, Ekins S. Discovery of 5-Nitro-6-thiocyanatopyrimidines as Inhibitors of Cryptococcus neoformans and Cryptococcus gattii. ACS Med Chem Lett 2021; 12:774-781. [PMID: 34055225 DOI: 10.1021/acsmedchemlett.1c00038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/31/2021] [Indexed: 12/27/2022] Open
Abstract
Opportunistic infections from pathogenic fungi present a major challenge to healthcare because of a very limited arsenal of antifungal drugs, an increasing population of immunosuppressed patients, and increased prevalence of resistant clinical strains due to overuse of the few available antifungals. Cryptococcal meningitis is a life-threatening opportunistic fungal infection caused by one of two species in the Cryptococcus genus, Cryptococcus neoformans and Cryptococcus gattii. Eighty percent of cryptococcosis diseases are caused by C. neoformans that is endemic in the environment. The standard of care is limited to old antifungals, and under a high standard of care, mortality remains between 10 and 30%. We have identified a series of 5-nitro-6-thiocyanatopyrimidine antifungal drug candidates using in vitro and computational machine learning approaches. These compounds can inhibit C. neoformans growth at submicromolar levels, are effective against fluconazole-resistant C. neoformans and a clinical strain of C. gattii, and are not antagonistic with currently approved antifungals.
Collapse
Affiliation(s)
- Maureen J. Donlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Institute for Drug and Biotherapeutic Development, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Department of Biology, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Alexander Lepioshkin
- Department of Biology, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Evan Xu
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Jeffrey Lin
- Department of Biology, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Vadim Makarov
- Research Center of Biotechnology RAS, 119071 Moscow, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| |
Collapse
|
21
|
Tarasova O, Poroikov V. Machine Learning in Discovery of New Antivirals and Optimization of Viral Infections Therapy. Curr Med Chem 2021; 28:7840-7861. [PMID: 33949929 DOI: 10.2174/0929867328666210504114351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 11/22/2022]
Abstract
Nowadays, computational approaches play an important role in the design of new drug-like compounds and optimization of pharmacotherapeutic treatment of diseases. The emerging growth of viral infections, including those caused by the Human Immunodeficiency Virus (HIV), Ebola virus, recently detected coronavirus, and some others, leads to many newly infected people with a high risk of death or severe complications. A huge amount of chemical, biological, clinical data is at the disposal of the researchers. Therefore, there are many opportunities to find the relationships between the particular features of chemical data and the antiviral activity of biologically active compounds based on machine learning approaches. Biological and clinical data can also be used for building models to predict relationships between viral genotype and drug resistance, which might help determine the clinical outcome of treatment. In the current study, we consider machine-learning approaches in the antiviral research carried out during the past decade. We overview in detail the application of machine-learning methods for the design of new potential antiviral agents and vaccines, drug resistance prediction, and analysis of virus-host interactions. Our review also covers the perspectives of using the machine-learning approaches for antiviral research, including Dengue, Ebola viruses, Influenza A, Human Immunodeficiency Virus, coronaviruses, and some others.
Collapse
Affiliation(s)
- Olga Tarasova
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow. Russian Federation
| | - Vladimir Poroikov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow. Russian Federation
| |
Collapse
|
22
|
Winkler DA. Use of Artificial Intelligence and Machine Learning for Discovery of Drugs for Neglected Tropical Diseases. Front Chem 2021; 9:614073. [PMID: 33791277 PMCID: PMC8005575 DOI: 10.3389/fchem.2021.614073] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Neglected tropical diseases continue to create high levels of morbidity and mortality in a sizeable fraction of the world’s population, despite ongoing research into new treatments. Some of the most important technological developments that have accelerated drug discovery for diseases of affluent countries have not flowed down to neglected tropical disease drug discovery. Pharmaceutical development business models, cost of developing new drug treatments and subsequent costs to patients, and accessibility of technologies to scientists in most of the affected countries are some of the reasons for this low uptake and slow development relative to that for common diseases in developed countries. Computational methods are starting to make significant inroads into discovery of drugs for neglected tropical diseases due to the increasing availability of large databases that can be used to train ML models, increasing accuracy of these methods, lower entry barrier for researchers, and widespread availability of public domain machine learning codes. Here, the application of artificial intelligence, largely the subset called machine learning, to modelling and prediction of biological activities and discovery of new drugs for neglected tropical diseases is summarized. The pathways for the development of machine learning methods in the short to medium term and the use of other artificial intelligence methods for drug discovery is discussed. The current roadblocks to, and likely impacts of, synergistic new technological developments on the use of ML methods for neglected tropical disease drug discovery in the future are also discussed.
Collapse
Affiliation(s)
- David A Winkler
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Latrobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia.,School of Pharmacy, University of Nottingham, Nottingham, United Kingdom.,CSIRO Data61, Pullenvale, QLD, Australia
| |
Collapse
|
23
|
Lima CS, Mottin M, de Assis LR, Mesquita NCDMR, Sousa BKDP, Coimbra LD, Santos KBD, Zorn KM, Guido RVC, Ekins S, Marques RE, Proença-Modena JL, Oliva G, Andrade CH, Regasini LO. Flavonoids from Pterogyne nitens as Zika virus NS2B-NS3 protease inhibitors. Bioorg Chem 2021; 109:104719. [PMID: 33636437 PMCID: PMC8227833 DOI: 10.1016/j.bioorg.2021.104719] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022]
Abstract
Although the widespread epidemic of Zika virus (ZIKV) and its neurological complications are well-known there are still no approved drugs available to treat this arboviral disease or vaccine to prevent the infection. Flavonoids from Pterogyne nitens have already demonstrated anti-flavivirus activity, although their target is unknown. In this study, we virtually screened an in-house database of 150 natural and semi-synthetic compounds against ZIKV NS2B-NS3 protease (NS2B-NS3p) using docking-based virtual screening, as part of the OpenZika project. As a result, we prioritized three flavonoids from P. nitens, quercetin, rutin and pedalitin, for experimental evaluation. We also used machine learning models, built with Assay Central® software, for predicting the activity and toxicity of these flavonoids. Biophysical and enzymatic assays generally agreed with the in silico predictions, confirming that the flavonoids inhibited ZIKV protease. The most promising hit, pedalitin, inhibited ZIKV NS2B-NS3p with an IC50 of 5 μM. In cell-based assays, pedalitin displayed significant activity at 250 and 500 µM, with slight toxicity in Vero cells. The results presented here demonstrate the potential of pedalitin as a candidate for hit-to-lead (H2L) optimization studies towards the discovery of antiviral drug candidates to treat ZIKV infections.
Collapse
Affiliation(s)
- Caroline Sprengel Lima
- Laboratory of Antibiotics and Chemotherapeutics (LAQ), Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), São José do Rio Preto, SP, Brazil
| | - Melina Mottin
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Leticia Ribeiro de Assis
- Laboratory of Antibiotics and Chemotherapeutics (LAQ), Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), São José do Rio Preto, SP, Brazil
| | | | - Bruna Katiele de Paula Sousa
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Lais Durco Coimbra
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - Karina Bispo-Dos- Santos
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States
| | - Rafael V C Guido
- Institute of Physics of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - José Luiz Proença-Modena
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Glaucius Oliva
- Institute of Physics of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Luis Octavio Regasini
- Laboratory of Antibiotics and Chemotherapeutics (LAQ), Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), São José do Rio Preto, SP, Brazil.
| |
Collapse
|
24
|
Goodswen SJ, Barratt JLN, Kennedy PJ, Kaufer A, Calarco L, Ellis JT. Machine learning and applications in microbiology. FEMS Microbiol Rev 2021; 45:6174022. [PMID: 33724378 PMCID: PMC8498514 DOI: 10.1093/femsre/fuab015] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 02/28/2021] [Indexed: 12/15/2022] Open
Abstract
To understand the intricacies of microorganisms at the molecular level requires making sense of copious volumes of data such that it may now be humanly impossible to detect insightful data patterns without an artificial intelligence application called machine learning. Applying machine learning to address biological problems is expected to grow at an unprecedented rate, yet it is perceived by the uninitiated as a mysterious and daunting entity entrusted to the domain of mathematicians and computer scientists. The aim of this review is to identify key points required to start the journey of becoming an effective machine learning practitioner. These key points are further reinforced with an evaluation of how machine learning has been applied so far in a broad scope of real-life microbiology examples. This includes predicting drug targets or vaccine candidates, diagnosing microorganisms causing infectious diseases, classifying drug resistance against antimicrobial medicines, predicting disease outbreaks and exploring microbial interactions. Our hope is to inspire microbiologists and other related researchers to join the emerging machine learning revolution.
Collapse
Affiliation(s)
- Stephen J Goodswen
- School of Life Sciences, University of Technology Sydney (UTS), Ultimo, NSW, Australia
| | - Joel L N Barratt
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney (UTS), Ultimo, NSW, Australia
| | - Alexa Kaufer
- School of Life Sciences, University of Technology Sydney (UTS), Ultimo, NSW, Australia
| | - Larissa Calarco
- School of Life Sciences, University of Technology Sydney (UTS), Ultimo, NSW, Australia
| | - John T Ellis
- School of Life Sciences, University of Technology Sydney (UTS), Ultimo, NSW, Australia
| |
Collapse
|
25
|
Zorn KM, Sun S, McConnon CL, Ma K, Chen EK, Foil DH, Lane TR, Liu LJ, El-Sakkary N, Skinner DE, Ekins S, Caffrey CR. A Machine Learning Strategy for Drug Discovery Identifies Anti-Schistosomal Small Molecules. ACS Infect Dis 2021; 7:406-420. [PMID: 33434015 PMCID: PMC7887754 DOI: 10.1021/acsinfecdis.0c00754] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Schistosomiasis is a chronic and
painful disease of poverty caused
by the flatworm parasite Schistosoma. Drug discovery
for antischistosomal compounds predominantly employs in vitro whole organism (phenotypic) screens against two developmental stages
of Schistosoma mansoni, post-infective larvae (somules)
and adults. We generated two rule books and associated scoring systems
to normalize 3898 phenotypic data points to enable machine learning.
The data were used to generate eight Bayesian machine learning models
with the Assay Central software according to parasite’s developmental
stage and experimental time point (≤24, 48, 72, and >72
h).
The models helped predict 56 active and nonactive compounds from commercial
compound libraries for testing. When these were screened against S. mansoni in vitro, the prediction accuracy for active
and inactives was 61% and 56% for somules and adults, respectively;
also, hit rates were 48% and 34%, respectively, far exceeding the
typical 1–2% hit rate for traditional high throughput screens.
Collapse
Affiliation(s)
- Kimberley M. Zorn
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Shengxi Sun
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Cecelia L. McConnon
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Kelley Ma
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Eric K. Chen
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Daniel H. Foil
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Lawrence J. Liu
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Nelly El-Sakkary
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Danielle E. Skinner
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0021, United States
| |
Collapse
|
26
|
Klein J, Baker NC, Foil DH, Zorn KM, Urbina F, Puhl AC, Ekins S. Using Bibliometric Analysis and Machine Learning to Identify Compounds Binding to Sialidase-1. ACS OMEGA 2021; 6:3186-3193. [PMID: 33553934 PMCID: PMC7860073 DOI: 10.1021/acsomega.0c05591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/05/2021] [Indexed: 05/20/2023]
Abstract
Rare diseases impact hundreds of millions of individuals worldwide. However, few therapies exist to treat the rare disease population because financial resources are limited, the number of patients affected is low, bioactivity data is often nonexistent, and very few animal models exist to support preclinical development efforts. Sialidosis is an ultrarare lysosomal storage disorder in which mutations in the NEU1 gene result in the deficiency of the lysosomal enzyme sialidase-1. This enzyme catalyzes the removal of sialic acid moieties from glycoproteins and glycolipids. Therefore, the defective or deficient protein leads to the buildup of sialylated glycoproteins as well as several characteristic symptoms of sialidosis including visual impairment, ataxia, hepatomegaly, dysostosis multiplex, and developmental delay. In this study, we used a bibliometric tool to generate links between lysosomal storage disease (LSD) targets and existing bioactivity data that could be curated in order to build machine learning models and screen compounds in silico. We focused on sialidase as an example, and we used the data curated from the literature to build a Bayesian model which was then used to score compound libraries and rank these molecules for in vitro testing. Two compounds were identified from in vitro testing using microscale thermophoresis, namely sulfameter (K d 2.15 ± 1.02 μM) and mexenone (K d 8.88 ± 4.02 μM), which validated our approach to identifying new molecules binding to this protein, which could represent possible drug candidates that can be evaluated further as potential chaperones for this ultrarare lysosomal disease for which there is currently no treatment. Combining bibliometric and machine learning approaches has the ability to assist in curating small molecule data and model building, respectively, for rare disease drug discovery. This approach also has the capability to identify new compounds that are potential drug candidates.
Collapse
Affiliation(s)
- Jennifer
J. Klein
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Nancy C. Baker
- ParlezChem, 123 W Union Street, Hillsborough, North Carolina 27278, United States
| | - Daniel H. Foil
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kimberley M. Zorn
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C. Puhl
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
27
|
Vignaux PA, Minerali E, Lane TR, Foil DH, Madrid PB, Puhl AC, Ekins S. The Antiviral Drug Tilorone Is a Potent and Selective Inhibitor of Acetylcholinesterase. Chem Res Toxicol 2021; 34:1296-1307. [PMID: 33400519 DOI: 10.1021/acs.chemrestox.0c00466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acetylcholinesterase (AChE) is an important drug target in neurological disorders like Alzheimer's disease, Lewy body dementia, and Parkinson's disease dementia as well as for other conditions like myasthenia gravis and anticholinergic poisoning. In this study, we have used a combination of high-throughput screening, machine learning, and docking to identify new inhibitors of this enzyme. Bayesian machine learning models were generated with literature data from ChEMBL for eel and human AChE inhibitors as well as butyrylcholinesterase inhibitors (BuChE) and compared with other machine learning methods. High-throughput screens for the eel AChE inhibitor model identified several molecules including tilorone, an antiviral drug that is well-established outside of the United States, as a newly identified nanomolar AChE inhibitor. We have described how tilorone inhibits both eel and human AChE with IC50's of 14.4 nM and 64.4 nM, respectively, but does not inhibit the closely related BuChE IC50 > 50 μM. We have docked tilorone into the human AChE crystal structure and shown that this selectivity is likely due to the reliance on a specific interaction with a hydrophobic residue in the peripheral anionic site of AChE that is absent in BuChE. We also conducted a pharmacological safety profile (SafetyScreen44) and kinase selectivity screen (SelectScreen) that showed tilorone (1 μM) only inhibited AChE out of 44 toxicology target proteins evaluated and did not appreciably inhibit any of the 485 kinases tested. This study suggests there may be a potential role for repurposing tilorone or its derivatives in conditions that benefit from AChE inhibition.
Collapse
Affiliation(s)
- Patricia A Vignaux
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Eni Minerali
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Peter B Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
28
|
Lane TR, Foil DH, Minerali E, Urbina F, Zorn KM, Ekins S. Bioactivity Comparison across Multiple Machine Learning Algorithms Using over 5000 Datasets for Drug Discovery. Mol Pharm 2020; 18:403-415. [PMID: 33325717 DOI: 10.1021/acs.molpharmaceut.0c01013] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Machine learning methods are attracting considerable attention from the pharmaceutical industry for use in drug discovery and applications beyond. In recent studies, we and others have applied multiple machine learning algorithms and modeling metrics and, in some cases, compared molecular descriptors to build models for individual targets or properties on a relatively small scale. Several research groups have used large numbers of datasets from public databases such as ChEMBL in order to evaluate machine learning methods of interest to them. The largest of these types of studies used on the order of 1400 datasets. We have now extracted well over 5000 datasets from CHEMBL for use with the ECFP6 fingerprint and in comparison of our proprietary software Assay Central with random forest, k-nearest neighbors, support vector classification, naïve Bayesian, AdaBoosted decision trees, and deep neural networks (three layers). Model performance was assessed using an array of fivefold cross-validation metrics including area-under-the-curve, F1 score, Cohen's kappa, and Matthews correlation coefficient. Based on ranked normalized scores for the metrics or datasets, all methods appeared comparable, while the distance from the top indicated that Assay Central and support vector classification were comparable. Unlike prior studies which have placed considerable emphasis on deep neural networks (deep learning), no advantage was seen in this case. If anything, Assay Central may have been at a slight advantage as the activity cutoff for each of the over 5000 datasets representing over 570,000 unique compounds was based on Assay Central performance, although support vector classification seems to be a strong competitor. We also applied Assay Central to perform prospective predictions for the toxicity targets PXR and hERG to further validate these models. This work appears to be the largest scale comparison of these machine learning algorithms to date. Future studies will likely evaluate additional databases, descriptors, and machine learning algorithms and further refine the methods for evaluating and comparing such models.
Collapse
Affiliation(s)
- Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Eni Minerali
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, United States
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
29
|
Zorn KM, Foil DH, Lane TR, Hillwalker W, Feifarek DJ, Jones F, Klaren WD, Brinkman AM, Ekins S. Comparing Machine Learning Models for Aromatase (P450 19A1). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:15546-15555. [PMID: 33207874 PMCID: PMC8194505 DOI: 10.1021/acs.est.0c05771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Aromatase, or cytochrome P450 19A1, catalyzes the aromatization of androgens to estrogens within the body. Changes in the activity of this enzyme can produce hormonal imbalances that can be detrimental to sexual and skeletal development. Inhibition of this enzyme can occur with drugs and natural products as well as environmental chemicals. Therefore, predicting potential endocrine disruption via exogenous chemicals requires that aromatase inhibition be considered in addition to androgen and estrogen pathway interference. Bayesian machine learning methods can be used for prospective prediction from the molecular structure without the need for experimental data. Herein, the generation and evaluation of multiple machine learning models utilizing different sources of aromatase inhibition data are described. These models are applied to two test sets for external validation with molecules relevant to drug discovery from the public domain. In addition, the performance of multiple machine learning algorithms was evaluated by comparing internal five-fold cross-validation statistics of the training data. These methods to predict aromatase inhibition from molecular structure, when used in concert with estrogen and androgen machine learning models, allow for a more holistic assessment of endocrine-disrupting potential of chemicals with limited empirical data and enable the reduction of the use of hazardous substances.
Collapse
Affiliation(s)
- Kimberley M. Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Daniel H. Foil
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Wendy Hillwalker
- Global Product Safety, SC Johnson and Son, Inc., Racine, WI, USA
| | | | - Frank Jones
- Global Product Safety, SC Johnson and Son, Inc., Racine, WI, USA
| | | | | | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| |
Collapse
|
30
|
Miller SR, Zhang X, Hau RK, Jilek JL, Jennings EQ, Galligan JJ, Foil DH, Zorn KM, Ekins S, Wright SH, Cherrington NJ. Predicting Drug Interactions with Human Equilibrative Nucleoside Transporters 1 and 2 Using Functional Knockout Cell Lines and Bayesian Modeling. Mol Pharmacol 2020; 99:147-162. [PMID: 33262250 DOI: 10.1124/molpharm.120.000169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Equilibrative nucleoside transporters (ENTs) 1 and 2 facilitate nucleoside transport across the blood-testis barrier (BTB). Improving drug entry into the testes with drugs that use endogenous transport pathways may lead to more effective treatments for diseases within the reproductive tract. In this study, CRISPR/CRISPR-associated protein 9 was used to generate HeLa cell lines in which ENT expression was limited to ENT1 or ENT2. We characterized uridine transport in these cell lines and generated Bayesian models to predict interactions with the ENTs. Quantification of [3H]uridine uptake in the presence of the ENT-specific inhibitor S-(4-nitrobenzyl)-6-thioinosine (NBMPR) demonstrated functional loss of each transporter. Nine nucleoside reverse-transcriptase inhibitors and 37 nucleoside/heterocycle analogs were evaluated to identify ENT interactions. Twenty-one compounds inhibited uridine uptake and abacavir, nevirapine, ticagrelor, and uridine triacetate had different IC50 values for ENT1 and ENT2. Total accumulation of four identified inhibitors was measured with and without NBMPR to determine whether there was ENT-mediated transport. Clofarabine and cladribine were ENT1 and ENT2 substrates, whereas nevirapine and lexibulin were ENT1 and ENT2 nontransported inhibitors. Bayesian models generated using Assay Central machine learning software yielded reasonably high internal validation performance (receiver operator characteristic > 0.7). ENT1 IC50-based models were generated from ChEMBL; subvalidations using this training data set correctly predicted 58% of inhibitors when analyzing activity by percent uptake and 63% when using estimated-IC50 values. Determining drug interactions with these transporters can be useful in identifying and predicting compounds that are ENT1 and ENT2 substrates and can thereby circumvent the BTB through this transepithelial transport pathway in Sertoli cells. SIGNIFICANCE STATEMENT: This study is the first to predict drug interactions with equilibrative nucleoside transporter (ENT) 1 and ENT2 using Bayesian modeling. Novel CRISPR/CRISPR-associated protein 9 functional knockouts of ENT1 and ENT2 in HeLa S3 cells were generated and characterized. Determining drug interactions with these transporters can be useful in identifying and predicting compounds that are ENT1 and ENT2 substrates and can circumvent the blood-testis barrier through this transepithelial transport pathway in Sertoli cells.
Collapse
Affiliation(s)
- Siennah R Miller
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Xiaohong Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Raymond K Hau
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Joseph L Jilek
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Erin Q Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Daniel H Foil
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Kimberley M Zorn
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Sean Ekins
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Stephen H Wright
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| |
Collapse
|
31
|
Egorova A, Bogner E, Novoselova E, Zorn KM, Ekins S, Makarov V. Dispirotripiperazine-core compounds, their biological activity with a focus on broad antiviral property, and perspectives in drug design (mini-review). Eur J Med Chem 2020; 211:113014. [PMID: 33218683 PMCID: PMC7658596 DOI: 10.1016/j.ejmech.2020.113014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022]
Abstract
Viruses are obligate intracellular parasites and have evolved to enter the host cell. To gain access they come into contact with the host cell through an initial adhesion, and some viruses from different genus may use heparan sulfate proteoglycans for it. The successful inhibition of this early event of the infection by synthetic molecules has always been an attractive target for medicinal chemists. Numerous reports have yielded insights into the function of compounds based on the dispirotripiperazine scaffold. Analysis suggests that this is a structural requirement for inhibiting the interactions between viruses and cell-surface heparan sulfate proteoglycans, thus preventing virus entry and replication. This review summarizes our current knowledge about the early history of development, synthesis, structure-activity relationships and antiviral evaluation of dispirotripiperazine-based compounds and where they are going in the future.
Collapse
Affiliation(s)
- Anna Egorova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia
| | - Elke Bogner
- Institute of Virology, Charité Universitätsmedizin Berlin, Charité Campus Mitte, Chariteplatz 1, 10117, Berlin, Germany
| | - Elena Novoselova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia.
| |
Collapse
|
32
|
Zorn KM, Foil DH, Lane TR, Hillwalker W, Feifarek DJ, Jones F, Klaren WD, Brinkman AM, Ekins S. Comparison of Machine Learning Models for the Androgen Receptor. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:13690-13700. [PMID: 33085465 PMCID: PMC8243727 DOI: 10.1021/acs.est.0c03984] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The androgen receptor (AR) is a target of interest for endocrine disruption research, as altered signaling can affect normal reproductive and neurological development for generations. In an effort to prioritize compounds with alternative methodologies, the U.S. Environmental Protection Agency (EPA) used in vitro data from 11 assays to construct models of AR agonist and antagonist signaling pathways. While these EPA ToxCast AR models require in vitro data to assign a bioactivity score, Bayesian machine learning methods can be used for prospective prediction from molecule structure alone. This approach was applied to multiple types of data corresponding to the EPA's AR signaling pathway with proprietary software, Assay Central. The training performance of all machine learning models, including six other algorithms, was evaluated by internal 5-fold cross-validation statistics. Bayesian machine learning models were also evaluated with external predictions of reference chemicals to compare prediction accuracies to published results from the EPA. The machine learning model group selected for further studies of endocrine disruption consisted of continuous AC50 data from the February 2019 release of ToxCast/Tox21. These efforts demonstrate how machine learning can be used to predict AR-mediated bioactivity and can also be applied to other targets of endocrine disruption.
Collapse
Affiliation(s)
- Kimberley M. Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Daniel H. Foil
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Wendy Hillwalker
- Global Product Safety, SC Johnson and Son, Inc., Racine, WI, USA
| | | | - Frank Jones
- Global Product Safety, SC Johnson and Son, Inc., Racine, WI, USA
| | | | | | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| |
Collapse
|
33
|
Vignaux P, Minerali E, Foil DH, Puhl AC, Ekins S. Machine Learning for Discovery of GSK3β Inhibitors. ACS OMEGA 2020; 5:26551-26561. [PMID: 33110983 PMCID: PMC7581251 DOI: 10.1021/acsomega.0c03302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/25/2020] [Indexed: 05/08/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting approximately 35 million people worldwide. The current treatment options for people with AD consist of drugs designed to slow the rate of decline in memory and cognition, but these treatments are not curative, and patients eventually suffer complete cognitive injury. With the substantial amounts of published data on targets for this disease, we proposed that machine learning software could be used to find novel small-molecule treatments that can supplement the AD drugs currently on the market. In order to do this, we used publicly available data in ChEMBL to build and validate Bayesian machine learning models for AD target proteins. The first AD target that we have addressed with this method is the serine-threonine kinase glycogen synthase kinase 3 beta (GSK3β), which is a proline-directed serine-threonine kinase that phosphorylates the microtubule-stabilizing protein tau. This phosphorylation prompts tau to dissociate from the microtubule and form insoluble oligomers called paired helical filaments, which are one of the components of the neurofibrillary tangles found in AD brains. Using our Bayesian machine learning model for GSK3β consisting of 2368 molecules, this model produced a five-fold cross validation ROC of 0.905. This model was also used for virtual screening of large libraries of FDA-approved drugs and clinical candidates. Subsequent testing of selected compounds revealed a selective small-molecule inhibitor, ruboxistaurin, with activity against GSK3β (avg IC50 = 97.3 nM) and GSK3α (IC50 = 695.9 nM). Several other structurally diverse inhibitors were also identified. We are now applying this machine learning approach to additional AD targets to identify approved drugs or clinical trial candidates that can be repurposed as AD therapeutics. This represents a viable approach to accelerate drug discovery and do so at a fraction of the cost of traditional high throughput screening.
Collapse
Affiliation(s)
- Patricia
A. Vignaux
- Collaborations Pharmaceuticals,
Inc., 840 Main Campus
Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Eni Minerali
- Collaborations Pharmaceuticals,
Inc., 840 Main Campus
Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H. Foil
- Collaborations Pharmaceuticals,
Inc., 840 Main Campus
Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C. Puhl
- Collaborations Pharmaceuticals,
Inc., 840 Main Campus
Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals,
Inc., 840 Main Campus
Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
34
|
Russo DP, Yan X, Shende S, Huang H, Yan B, Zhu H. Virtual Molecular Projections and Convolutional Neural Networks for the End-to-End Modeling of Nanoparticle Activities and Properties. Anal Chem 2020; 92:13971-13979. [PMID: 32970421 DOI: 10.1021/acs.analchem.0c02878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Digitalizing complex nanostructures into data structures suitable for machine learning modeling without losing nanostructure information has been a major challenge. Deep learning frameworks, particularly convolutional neural networks (CNNs), are especially adept at handling multidimensional and complex inputs. In this study, CNNs were applied for the modeling of nanoparticle activities exclusively from nanostructures. The nanostructures were represented by virtual molecular projections, a multidimensional digitalization of nanostructures, and used as input data to train CNNs. To this end, 77 nanoparticles with various activities and/or physicochemical property results were used for modeling. The resulting CNN model predictions show high correlations with the experimental results. An analysis of a trained CNN quantitatively showed that neurons were able to recognize distinct nanostructure features critical to activities and physicochemical properties. This "end-to-end" deep learning approach is well suited to digitalize complex nanostructures for data-driven machine learning modeling and can be broadly applied to rationally design nanoparticles with desired activities.
Collapse
Affiliation(s)
- Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States
| | - Xiliang Yan
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Institute of Environmental Research at Greater Bay, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Sunil Shende
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Department of Computer Science, Rutgers University, 227 Penn Street, Camden, New Jersey 08102, United States
| | | | - Bing Yan
- Institute of Environmental Research at Greater Bay, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.,School of Environmental Science and Engineering, Shandong University, Jinan 250100, China
| | - Hao Zhu
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Department of Chemistry, Rutgers University, 315 Penn Street, Camden, New Jersey 08102, United States
| |
Collapse
|
35
|
Zorn KM, Foil DH, Lane TR, Russo DP, Hillwalker W, Feifarek DJ, Jones F, Klaren WD, Brinkman AM, Ekins S. Machine Learning Models for Estrogen Receptor Bioactivity and Endocrine Disruption Prediction. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:12202-12213. [PMID: 32857505 PMCID: PMC8194504 DOI: 10.1021/acs.est.0c03982] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The U.S. Environmental Protection Agency (EPA) periodically releases in vitro data across a variety of targets, including the estrogen receptor (ER). In 2015, the EPA used these data to construct mathematical models of ER agonist and antagonist pathways to prioritize chemicals for endocrine disruption testing. However, mathematical models require in vitro data prior to predicting estrogenic activity, but machine learning methods are capable of prospective prediction from the molecular structure alone. The current study describes the generation and evaluation of Bayesian machine learning models grouped by the EPA's ER agonist pathway model using multiple data types with proprietary software, Assay Central. External predictions with three test sets of in vitro and in vivo reference chemicals with agonist activity classifications were compared to previous mathematical model publications. Training data sets were subjected to additional machine learning algorithms and compared with rank normalized scores of internal five-fold cross-validation statistics. External predictions were found to be comparable or superior to previous studies published by the EPA. When assessing six additional algorithms for the training data sets, Assay Central performed similarly at a reduced computational cost. This study demonstrates that machine learning can prioritize chemicals for future in vitro and in vivo testing of ER agonism.
Collapse
Affiliation(s)
- Kimberley M Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University, Camden, New Jersey 08102, United States
| | - Wendy Hillwalker
- Global Product Safety, SC Johnson and Son, Inc., Racine, Wisconsin 53404, United States
| | - David J Feifarek
- Global Product Safety, SC Johnson and Son, Inc., Racine, Wisconsin 53404, United States
| | - Frank Jones
- Global Product Safety, SC Johnson and Son, Inc., Racine, Wisconsin 53404, United States
| | - William D Klaren
- Global Product Safety, SC Johnson and Son, Inc., Racine, Wisconsin 53404, United States
| | - Ashley M Brinkman
- Global Product Safety, SC Johnson and Son, Inc., Racine, Wisconsin 53404, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
36
|
Lane TR, Dyall J, Mercer L, Goodin C, Foil DH, Zhou H, Postnikova E, Liang JY, Holbrook MR, Madrid PB, Ekins S. Repurposing Pyramax®, quinacrine and tilorone as treatments for Ebola virus disease. Antiviral Res 2020; 182:104908. [PMID: 32798602 PMCID: PMC7425680 DOI: 10.1016/j.antiviral.2020.104908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/03/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022]
Abstract
We have recently identified three molecules (tilorone, quinacrine and pyronaridine tetraphosphate) which all demonstrated efficacy in the mouse model of infection with mouse-adapted Ebola virus (EBOV) model of disease and had similar in vitro inhibition of an Ebola pseudovirus (VSV-EBOV-GP), suggesting they interfere with viral entry. Using a machine learning model to predict lysosomotropism these compounds were evaluated for their ability to possess a lysosomotropic mechanism in vitro. We now demonstrate in vitro that pyronaridine tetraphosphate is an inhibitor of Lysotracker accumulation in lysosomes (IC50 = 0.56 μM). Further, we evaluated antiviral synergy between pyronaridine and artesunate (Pyramax®), which are used in combination to treat malaria. Artesunate was not found to have lysosomotropic activity in vitro and the combination effect on EBOV inhibition was shown to be additive. Pyramax® may represent a unique example of the repurposing of a combination product for another disease.
Collapse
Affiliation(s)
- Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Luke Mercer
- Cambrex, 3501 Tricenter Blvd, Suite C, Durham, NC, 27713, USA
| | - Caleb Goodin
- Cambrex, 3501 Tricenter Blvd, Suite C, Durham, NC, 27713, USA
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Huanying Zhou
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | | | - Janie Y Liang
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Michael R Holbrook
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Peter B Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, CA, 94025, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| |
Collapse
|
37
|
Anderson E, Havener TM, Zorn KM, Foil DH, Lane TR, Capuzzi SJ, Morris D, Hickey AJ, Drewry DH, Ekins S. Synergistic drug combinations and machine learning for drug repurposing in chordoma. Sci Rep 2020; 10:12982. [PMID: 32737414 PMCID: PMC7395084 DOI: 10.1038/s41598-020-70026-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/20/2020] [Indexed: 12/18/2022] Open
Abstract
Chordoma is a devastating rare cancer that affects one in a million people. With a mean-survival of just 6 years and no approved medicines, the primary treatments are surgery and radiation. In order to speed new medicines to chordoma patients, a drug repurposing strategy represents an attractive approach. Drugs that have already advanced through human clinical safety trials have the potential to be approved more quickly than de novo discovered medicines on new targets. We have taken two strategies to enable this: (1) generated and validated machine learning models of chordoma inhibition and screened compounds of interest in vitro. (2) Tested combinations of approved kinase inhibitors already being individually evaluated for chordoma. Several published studies of compounds screened against chordoma cell lines were used to generate Bayesian Machine learning models which were then used to score compounds selected from the NIH NCATS industry-provided assets. Out of these compounds, the mTOR inhibitor AZD2014, was the most potent against chordoma cell lines (IC50 0.35 µM U-CH1 and 0.61 µM U-CH2). Several studies have shown the importance of the mTOR signaling pathway in chordoma and suggest it as a promising avenue for targeted therapy. Additionally, two currently FDA approved drugs, afatinib and palbociclib (EGFR and CDK4/6 inhibitors, respectively) demonstrated synergy in vitro (CI50 = 0.43) while AZD2014 and afatanib also showed synergy (CI50 = 0.41) against a chordoma cell in vitro. These findings may be of interest clinically, and this in vitro- and in silico approach could also be applied to other rare cancers.
Collapse
Affiliation(s)
- Edward Anderson
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tammy M Havener
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA
| | - Stephen J Capuzzi
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dave Morris
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony J Hickey
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- RTI International, Research Triangle Park, NC, USA
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sean Ekins
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, USA.
| |
Collapse
|
38
|
Minerali E, Foil DH, Zorn KM, Lane TR, Ekins S. Comparing Machine Learning Algorithms for Predicting Drug-Induced Liver Injury (DILI). Mol Pharm 2020; 17:2628-2637. [PMID: 32422053 DOI: 10.1021/acs.molpharmaceut.0c00326] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug-induced liver injury (DILI) is one the most unpredictable adverse reactions to xenobiotics in humans and the leading cause of postmarketing withdrawals of approved drugs. To date, these drugs have been collated by the FDA to form the DILIRank database, which classifies DILI severity and potential. These classifications have been used by various research groups in generating computational predictions for this type of liver injury. Recently, groups from Pfizer and AstraZeneca have collated DILI in vitro data and physicochemical properties for compounds that can be used along with data from the FDA to build machine learning models for DILI. In this study, we have used these data sets, as well as the Biopharmaceutics Drug Disposition Classification System data set, to generate Bayesian machine learning models with our in-house software, Assay Central. The performance of all machine learning models was assessed through both the internal 5-fold cross-validation metrics and prediction accuracy of an external test set of compounds with known hepatotoxicity. The best-performing Bayesian model was based on the DILI-concern category from the DILIRank database with an ROC of 0.814, a sensitivity of 0.741, a specificity of 0.755, and an accuracy of 0.746. A comparison of alternative machine learning algorithms, such as k-nearest neighbors, support vector classification, AdaBoosted decision trees, and deep learning methods, produced similar statistics to those generated with the Bayesian algorithm in Assay Central. This study demonstrates machine learning models grouped in a tool called MegaTox that can be used to predict early-stage clinical compounds, as well as recent FDA-approved drugs, to identify potential DILI.
Collapse
Affiliation(s)
- Eni Minerali
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
39
|
Ekins S, Mottin M, Ramos PRPS, Sousa BKP, Neves BJ, Foil DH, Zorn KM, Braga RC, Coffee M, Southan C, Puhl AC, Andrade CH. Déjà vu: Stimulating open drug discovery for SARS-CoV-2. Drug Discov Today 2020; 25:928-941. [PMID: 32320852 PMCID: PMC7167229 DOI: 10.1016/j.drudis.2020.03.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
In the past decade we have seen two major Ebola virus outbreaks in Africa, the Zika virus in Brazil and the Americas and the current pandemic of coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There is a strong sense of déjà vu because there are still no effective treatments. In the COVID-19 pandemic, despite being a new virus, there are already drugs suggested as active in in vitro assays that are being repurposed in clinical trials. Promising SARS-CoV-2 viral targets and computational approaches are described and discussed. Here, we propose, based on open antiviral drug discovery approaches for previous outbreaks, that there could still be gaps in our approach to drug discovery.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA.
| | - Melina Mottin
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Paulo R P S Ramos
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Bruna K P Sousa
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Bruno Junior Neves
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Daniel H Foil
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | | | - Megan Coffee
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University, NY, USA; Department of Population and Family Health, Mailman School of Public Health, Columbia University, NY, USA
| | | | - Ana C Puhl
- Collaborations Pharmaceuticals, 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Carolina Horta Andrade
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil; Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-864, Brazil.
| |
Collapse
|
40
|
Jackson SS, Sumner LE, Finnegan MA, Billings EA, Huffman DL, Rush MA. A 35-Year Review of Pre-Clinical HIV Therapeutics Research Reported by NIH ChemDB: Influences of Target Discoveries, Drug Approvals and Research Funding. JOURNAL OF AIDS & CLINICAL RESEARCH 2020; 11:11. [PMID: 33364074 PMCID: PMC7757624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We present a retrospective analysis of trends in human immunodeficiency virus (HIV) small molecule drug development over the last thirty-five years based on data captured by ChemDB, a United States (US) National Institutes of Health (NIH) database of chemical and biological HIV testing data. These data are analyzed alongside NIH funding levels, US Food and Drug Administration (FDA) drug approvals, and new target identifications to explore the influences of these factors on anti-HIV drug discovery research. The NIH's ChemDB database collects chemical and biological testing data describing published and patented pre-clinical compounds in development as potential HIV therapeutics. These data were used as a proxy for estimating overall levels of HIV therapeutics research activities in order to assess research trends. Data extracted from ChemDB were compared with records of drug approvals from the FDA, NIH funding levels, and drug target discoveries to elucidate the influences that these factors have on levels of HIV therapeutics research activities. Despite the increasingly wide suite of HIV therapeutic options that have accumulated during decades of research, interest in HIV therapeutics research activities remains strong. While decreases in research activity levels have followed cuts in research funding, FDA-approved HIV therapeutics have continued to accumulate. The comparisons presented here indicate that HIV drug research activity levels have historically been more responsive to changes in funding levels and the identification of new drug targets, than they have been to drug approvals. Continued interest in HIV therapeutics research may reflect that fact that of the 55 drugs approved for HIV treatment as of 2018, only seven inhibitory targets are represented. Moreover, drug resistance presents substantial clinical challenges. Sustained research interest despite drug approvals and fluctuations in available funding likely reflects the clinical need for safer, more palatable and more efficacious therapeutics; robust attention to both novel therapeutics and inhibitory targets is necessary given the speed of development of drug-resistant HIV strains. Only with such continued interest will we reduce the burden of acquired immunodeficiency syndrome (AIDS) disease and control the AIDS epidemic.
Collapse
Affiliation(s)
| | | | | | | | | | - Margaret A. Rush
- Gryphon Scientific, LLC, Takoma Park, MD, USA,Address for Correspondence: Margaret A. Rush, Gryphon Scientific, LLC, 6930 Carroll Avenue, Suite 900, Takoma Park, MD 20912, USA, Tel: +1+301-270-0647;
| |
Collapse
|
41
|
Fan Y, Zhang Y, Hua Y, Wang Y, Zhu L, Zhao J, Yang Y, Chen X, Lu S, Lu T, Chen Y, Liu H. Investigation of Machine Intelligence in Compound Cell Activity Classification. Mol Pharm 2019; 16:4472-4484. [PMID: 31580683 DOI: 10.1021/acs.molpharmaceut.9b00558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Machine intelligence has been greatly developed in the past decades and has been widely used in many fields. In the recent years, many reports have shown its satisfactory effect in drug discovery. In this study, machine intelligence methods were explored to assist the cell activity prediction. Multiple machine intelligence methods including support vector machine, decision tree, random forest, extra trees, gradient boosting machine, convolutional neural network, long short-term memory network, and gated recurrent unit network were employed to separate compounds based on their cell activity. Different from some reported classification models, compounds were expressed as a string by the simplified molecular input line entry system and directly used as input rather than any chemical descriptors, which mimicked natural language processing. Both the single cell strain and whole data set under the balanced and imbalanced data distributions were discussed, respectively. Different activity cutoffs were set for the single (Z-score = 3) and the whole (Z-score = 5 and 6) data set. Nine metrics were used to evaluate the models including accuracy, precision, recall, f1-score, area under the receiver operating characteristic curve score, Cohen's κ, Brier score, Matthews correlation coefficient, and balanced accuracy. The results show that the gradient boosting machine is competent at balanced data distribution, and convolutional neural network is qualified for the imbalanced one. The results demonstrate that both classic machine learning methods and deep learning methods have potential in classification of compound cell activity.
Collapse
Affiliation(s)
- Yuanrong Fan
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yanmin Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yi Hua
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yuchen Wang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Lu Zhu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Junnan Zhao
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yan Yang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Xingye Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Shuai Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China.,State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Haichun Liu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| |
Collapse
|
42
|
Vásquez-Domínguez E, Armijos-Jaramillo VD, Tejera E, González-Díaz H. Multioutput Perturbation-Theory Machine Learning (PTML) Model of ChEMBL Data for Antiretroviral Compounds. Mol Pharm 2019; 16:4200-4212. [PMID: 31426639 DOI: 10.1021/acs.molpharmaceut.9b00538] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Retroviral infections, such as HIV, are, until now, diseases with no cure. Medicine and pharmaceutical chemistry need and consider it a huge goal to define target proteins of new antiretroviral compounds. ChEMBL manages Big Data features with a complex data set, which is hard to organize. This makes information difficult to analyze due to a big number of characteristics described in order to predict new drug candidates for retroviral infections. For this reason, we propose to develop a new predictive model combining perturbation theory (PT) bases and machine learning (ML) modeling to create a new tool that can take advantage of all the available information. The PTML model proposed in this work for the ChEMBL data set preclinical experimental assays for antiretroviral compounds consists of a linear equation with four variables. The PT operators used are founded on multicondition moving averages, combining different features and simplifying the difficulty to manage all data. More than 140 000 preclinical assays for 56 105 compounds with different characteristics or experimental conditions have been carried out and can be found in ChEMBL database, covering combinations with 359 biological activity parameters (c0), 55 protein accessions (c1), 83 cell lines (c2), 64 organisms of assay (c3), and 773 subtypes or strains. We have included 150 148 preclinical experimental assays for HIV virus, 1188 for HTLV virus, 84 for simian immunodeficiency virus, 370 for murine leukemia virus, 119 for Rous sarcoma virus, 1581 for MMTV, etc. We also included 5277 assays for hepatitis B virus. The developed PTML model reached considerable values in sensibility (73.05% for training and 73.10% for validation), specificity (86.61% for training and 87.17% for validation), and accuracy (75.84% for training and 75.98% for validation). We also compared alternative PTML models with different PT operators such as covariance, moments, and exponential terms. Finally, we made a comparison between literature ML models with our PTML model and also artificial neural network (ANN) nonlinear models. We conclude that this PTML model is the first one to consider multiple characteristics of preclinical experimental antiretroviral assays combined, generating a simple, useful, and adaptable instrument, which could reduce time and costs in antiretroviral drugs research.
Collapse
Affiliation(s)
- Emilia Vásquez-Domínguez
- Department of Organic Chemistry II , University of Basque Country UPV/EHU , 48940 Leioa , Spain.,Faculty of Engineering and Applied Sciences-Biotechnology , Universidad de Las Américas (UDLA) , 170125 Quito , Ecuador
| | - Vinicio Danilo Armijos-Jaramillo
- Faculty of Engineering and Applied Sciences-Biotechnology , Universidad de Las Américas (UDLA) , 170125 Quito , Ecuador.,Bio-chemioinformatics group , Universidad de Las Américas (UDLA) , 170125 Quito , Ecuador
| | - Eduardo Tejera
- Faculty of Engineering and Applied Sciences-Biotechnology , Universidad de Las Américas (UDLA) , 170125 Quito , Ecuador.,Bio-chemioinformatics group , Universidad de Las Américas (UDLA) , 170125 Quito , Ecuador
| | - 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
|
43
|
Ekins S, Gerlach J, Zorn KM, Antonio BM, Lin Z, Gerlach A. Repurposing Approved Drugs as Inhibitors of K v7.1 and Na v1.8 to Treat Pitt Hopkins Syndrome. Pharm Res 2019; 36:137. [PMID: 31332533 DOI: 10.1007/s11095-019-2671-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 07/10/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Pitt Hopkins Syndrome (PTHS) is a rare genetic disorder caused by mutations of a specific gene, transcription factor 4 (TCF4), located on chromosome 18. PTHS results in individuals that have moderate to severe intellectual disability, with most exhibiting psychomotor delay. PTHS also exhibits features of autistic spectrum disorders, which are characterized by the impaired ability to communicate and socialize. PTHS is comorbid with a higher prevalence of epileptic seizures which can be present from birth or which commonly develop in childhood. Attenuated or absent TCF4 expression results in increased translation of peripheral ion channels Kv7.1 and Nav1.8 which triggers an increase in after-hyperpolarization and altered firing properties. METHODS We now describe a high throughput screen (HTS) of 1280 approved drugs and machine learning models developed from this data. The ion channels were expressed in either CHO (KV7.1) or HEK293 (Nav1.8) cells and the HTS used either 86Rb+ efflux (KV7.1) or a FLIPR assay (Nav1.8). RESULTS The HTS delivered 55 inhibitors of Kv7.1 (4.2% hit rate) and 93 inhibitors of Nav1.8 (7.2% hit rate) at a screening concentration of 10 μM. These datasets also enabled us to generate and validate Bayesian machine learning models for these ion channels. We also describe a structure activity relationship for several dihydropyridine compounds as inhibitors of Nav1.8. CONCLUSIONS This work could lead to the potential repurposing of nicardipine or other dihydropyridine calcium channel antagonists as potential treatments for PTHS acting via Nav1.8, as there are currently no approved treatments for this rare disorder.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina, 27606, USA.
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina, 27606, USA
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina, 27606, USA
| | - Brett M Antonio
- Icagen, Inc., 4222 Emperor Blvd, Durham, North Carolina, 27703, USA
| | - Zhixin Lin
- Icagen, Inc., 4222 Emperor Blvd, Durham, North Carolina, 27703, USA
| | - Aaron Gerlach
- Icagen, Inc., 4222 Emperor Blvd, Durham, North Carolina, 27703, USA
| |
Collapse
|