106 results on '"Hongmao Sun"'
Search Results
2. Virtual Screening for the Discovery of Microbiome β-Glucuronidase Inhibitors to Alleviate Cancer Drug Toxicity.
- Author
-
Anup P. Challa, Xin Hu, Ya-Qin Zhang, Jeffrey Hymes, Bret D. Wallace, Surendra Karavadhi, Hongmao Sun, Samarjit Patnaik, Matthew D. Hall, and Min Shen
- Published
- 2022
- Full Text
- View/download PDF
3. Discovery of Small-Molecule VapC1 Nuclease Inhibitors by Virtual Screening and Scaffold Hopping from an Atomic Structure Revealing Protein-Protein Interactions with a Native VapB1 Inhibitor.
- Author
-
Hongmao Sun, Nathan P. Coussens, Carina Danchik, Leah M. Wachsmuth, Mark J. Henderson, Samarjit Patnaik, Matthew D. Hall, Ashley L. Molinaro, Dayle A. Daines, and Min Shen
- Published
- 2022
- Full Text
- View/download PDF
4. Improving Molecular Property Prediction on Limited Data with Deep Multi-Label Learning.
- Author
-
Hehuan Ma, Chaochao Yan, Yuzhi Guo, Sheng Wang 0001, Yuhong Wang, Hongmao Sun, and Junzhou Huang
- Published
- 2020
- Full Text
- View/download PDF
5. SMILES-BERT: Large Scale Unsupervised Pre-Training for Molecular Property Prediction.
- Author
-
Sheng Wang 0001, Yuzhi Guo, Yuhong Wang, Hongmao Sun, and Junzhou Huang
- Published
- 2019
- Full Text
- View/download PDF
6. Chemoprotective antimalarials identified through quantitative high-throughput screening of Plasmodium blood and liver stage parasites
- Author
-
Dorjbal Dorjsuren, Richard T. Eastman, Kathryn J. Wicht, Daniel Jansen, Daniel C. Talley, Benjamin A. Sigmon, Alexey V. Zakharov, Norma Roncal, Andrew T. Girvin, Yevgeniya Antonova-Koch, Paul M. Will, Pranav Shah, Hongmao Sun, Carleen Klumpp-Thomas, Sachel Mok, Tomas Yeo, Stephan Meister, Juan Jose Marugan, Leila S. Ross, Xin Xu, David J. Maloney, Ajit Jadhav, Bryan T. Mott, Richard J. Sciotti, Elizabeth A. Winzeler, Norman C. Waters, Robert F. Campbell, Wenwei Huang, Anton Simeonov, and David A. Fidock
- Subjects
Medicine ,Science - Abstract
Abstract The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration–response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.
- Published
- 2021
- Full Text
- View/download PDF
7. A Comprehensive Phenotypic Screening Strategy to Identify Modulators of Cargo Translocation by the Bacterial Type IVB Secretion System
- Author
-
Eric Cheng, Dorjbal Dorjsuren, Stephanie Lehman, Charles L. Larson, Steven A. Titus, Hongmao Sun, Alexey Zakharov, Ganesha Rai, Robert A. Heinzen, Anton Simeonov, and Matthias P. Machner
- Subjects
effector protein ,small molecule library ,high throughput screen ,beta-lactamase reporter ,Dot/Icm secretion system ,Microbiology ,QR1-502 - Abstract
ABSTRACT Bacterial type IV secretion systems (T4SSs) are macromolecular machines that translocate effector proteins across multiple membranes into infected host cells. Loss of function mutations in genes encoding protein components of the T4SS render bacteria avirulent, highlighting the attractiveness of T4SSs as drug targets. Here, we designed an automated high-throughput screening approach for the identification of compounds that interfere with the delivery of a reporter-effector fusion protein from Legionella pneumophila into RAW264.7 mouse macrophages. Using a fluorescence resonance energy transfer (FRET)-based detection assay in a bacteria/macrophage coculture format, we screened a library of over 18,000 compounds and, upon vetting compound candidates in a variety of in vitro and cell-based secondary screens, isolated several hits that efficiently interfered with biological processes that depend on a functional T4SS, such as intracellular bacterial proliferation or lysosomal avoidance, but had no detectable effect on L. pneumophila growth in culture medium, conditions under which the T4SS is dispensable. Notably, the same hit compounds also attenuated, to varying degrees, effector delivery by the closely related T4SS from Coxiella burnetii, notably without impacting growth of this organism within synthetic media. Together, these results support the idea that interference with T4SS function is a possible therapeutic intervention strategy, and the emerging compounds provide tools to interrogate at a molecular level the regulation and dynamics of these virulence-critical translocation machines. IMPORTANCE Multi-drug-resistant pathogens are an emerging threat to human health. Because conventional antibiotics target not only the pathogen but also eradicate the beneficial microbiota, they often cause additional clinical complications. Thus, there is an urgent need for the development of “smarter” therapeutics that selectively target pathogens without affecting beneficial commensals. The bacterial type IV secretion system (T4SS) is essential for the virulence of a variety of pathogens but dispensable for bacterial viability in general and can, thus, be considered a pathogen’s Achilles heel. By identifying small molecules that interfere with cargo delivery by the T4SS from two important human pathogens, Legionella pneumophila and Coxiella burnetii, our study represents the first step in our pursuit toward precision medicine by developing pathogen-selective therapeutics capable of treating the infections without causing harm to commensal bacteria.
- Published
- 2022
- Full Text
- View/download PDF
8. Discovery of Novel Small-Molecule Scaffolds for the Inhibition and Activation of WIP1 Phosphatase from a RapidFire Mass Spectrometry High-Throughput Screen
- Author
-
Victor Clausse, Yuhong Fang, Dingyin Tao, Harichandra D. Tagad, Hongmao Sun, Yuhong Wang, Surendra Karavadhi, Kelly Lane, Zhen-Dan Shi, Olga Vasalatiy, Christopher A. LeClair, Rebecca Eells, Min Shen, Samarjit Patnaik, Ettore Appella, Nathan P. Coussens, Matthew D. Hall, and Daniel H. Appella
- Subjects
Pharmacology ,Pharmacology (medical) - Abstract
Wild-type P53-induced phosphatase 1 (WIP1), also known as
- Published
- 2022
- Full Text
- View/download PDF
9. Structure Based Model for the Prediction of Phospholipidosis Induction Potential of Small Molecules.
- Author
-
Hongmao Sun, Sampada Shahane, Menghang Xia, Christopher P. Austin, and Ruili Huang
- Published
- 2012
- Full Text
- View/download PDF
10. Exploring Polypharmacology Using a ROCS-Based Target Fishing Approach.
- Author
-
Mohamed Diwan M. AbdulHameed, Sidhartha Chaudhury, Narender Singh, Hongmao Sun, Anders Wallqvist, and Gregory J. Tawa
- Published
- 2012
- Full Text
- View/download PDF
11. An 8 Kb/s low-delay CELP speech coder.
- Author
-
Hongmao Sun, Chunyan Wang 0004, M. Omair Ahmad, and M. N. S. Swamy
- Published
- 2000
- Full Text
- View/download PDF
12. Predictive Models for Cytochrome P450 Isozymes Based on Quantitative High Throughput Screening Data.
- Author
-
Hongmao Sun, Henrike Veith, Menghang Xia, Christopher P. Austin, and Ruili Huang
- Published
- 2011
- Full Text
- View/download PDF
13. Structure-Based Ligand Design I
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
14. Introduction to the Book
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
15. Homology Modeling and Ligand-Based Molecule Design
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
16. Quantitative Structure–Property Relationships Models for Lipophilicity and Aqueous Solubility
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
17. Structures, Limitations, and Pitfalls
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
18. Quantitative Structure–Activity Relationships
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
19. In Silico ADMET Profiling
- Author
-
Hongmao, Sun, primary
- Published
- 2016
- Full Text
- View/download PDF
20. A High-Throughput Screen Identifies 2,9-Diazaspiro[5.5]Undecanes as Inducers of the Endoplasmic Reticulum Stress Response with Cytotoxic Activity in 3D Glioma Cell Models.
- Author
-
Natalia J Martinez, Ganesha Rai, Adam Yasgar, Wendy A Lea, Hongmao Sun, Yuhong Wang, Diane K Luci, Shyh-Ming Yang, Kana Nishihara, Shunichi Takeda, Mohiuddin Sagor, Irina Earnshaw, Tetsuya Okada, Kazutoshi Mori, Kelli Wilson, Gregory J Riggins, Menghang Xia, Maurizio Grimaldi, Ajit Jadhav, David J Maloney, and Anton Simeonov
- Subjects
Medicine ,Science - Abstract
The endoplasmic reticulum (ER) is involved in Ca2+ signaling and protein folding. ER Ca2+ depletion and accumulation of unfolded proteins activate the molecular chaperone GRP78 (glucose-regulated protein 78) which in turn triggers the ER stress response (ERSR) pathway aimed to restore ER homeostasis. Failure to adapt to stress, however, results in apoptosis. We and others have shown that malignant cells are more susceptible to ERSR-induced apoptosis than their normal counterparts, implicating the ERSR as a potential target for cancer therapeutics. Predicated on these findings, we developed an assay that uses a GRP78 biosensor to identify small molecule activators of ERSR in glioma cells. We performed a quantitative high-throughput screen (qHTS) against a collection of ~425,000 compounds and a comprehensive panel of orthogonal secondary assays was formulated for stringent compound validation. We identified novel activators of ERSR, including a compound with a 2,9-diazaspiro[5.5]undecane core, which depletes intracellular Ca2+ stores and induces apoptosis-mediated cell death in several cancer cell lines, including patient-derived and 3D cultures of glioma cells. This study demonstrates that our screening platform enables the identification and profiling of ERSR inducers with cytotoxic activity and advocates for characterization of these compound in in vivo models.
- Published
- 2016
- Full Text
- View/download PDF
21. Correction: A High-Throughput Screen Identifies 2,9-Diazaspiro[5.5]Undecanes as Inducers of the Endoplasmic Reticulum Stress Response with Cytotoxic Activity in 3D Glioma Cell Models.
- Author
-
Natalia J Martinez, Ganesha Rai, Adam Yasgar, Wendy A Lea, Hongmao Sun, Yuhong Wang, Diane K Luci, Shyh-Ming Yang, Kana Nishihara, Shunichi Takeda, Mohiuddin, Irina Earnshaw, Tetsuya Okada, Kazutoshi Mori, Kelli Wilson, Gregory J Riggins, Menghang Xia, Maurizio Grimaldi, Ajit Jadhav, David J Maloney, and Anton Simeonov
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0161486.].
- Published
- 2016
- Full Text
- View/download PDF
22. Discovery of VapC1 small molecule nuclease inhibitors by virtual screening and scaffold hopping from an atomic structure revealing protein-protein interactions with native VapB1 inhibitor
- Author
-
Hongmao Sun, Nathan P. Coussens, Carina Danchik, Leah M. Wachsmuth, Mark J. Henderson, Samarjit Patnaik, Matthew D. Hall, Ashley L. Molinaro, Dayle A. Daines, and Min Shen
- Subjects
Ribonucleases ,Bacterial Proteins ,General Chemical Engineering ,Bacterial Toxins ,Humans ,General Chemistry ,Antitoxins ,Library and Information Sciences ,Haemophilus influenzae ,Article ,Computer Science Applications - Abstract
Nontypeable Haemophilus influenzae (NTHi) are clinically important Gram-negative bacteria that are responsible for various human mucosal diseases, including otitis media (OM). Recurrent OM caused by NTHi is common and infections which recur less than two weeks following antimicrobial therapy are largely attributable to recurrence of the same strain of bacteria. Toxin-antitoxin (TA) modules encoded by bacteria enable rapid responses to environmental stresses and are thought to facilitate growth arrest, persistence, and tolerance to antibiotics. The vapBC-1 locus of NTHi encodes a type II TA system, comprising the ribonuclease toxin VapC1 and its cognate antitoxin VapB1. The activity of VapC1 has been linked to the survival of NTHi during antibiotic treatment both in vivo and ex vivo. Therefore, inhibitors of VapC1 might serve as adjuvants to antibiotics, preventing NTHi from entering growth arrest and surviving; however, none have been reported to date. A truncated VapB1 peptide from a crystal structure of the VapBC-1 complex was used to generate pharmacophore queries to facilitate a scaffold hopping approach for the identification of small molecule VapC1 inhibitors. The National Center for Advancing Translational Sciences small molecule library was virtually screened using the shape-based method Rapid Overlay of Chemical Structures (ROCS) and the top-ranking hits were docked into the VapB1 binding pocket of VapC1. Two hundred virtual screening hits with the best docking scores were selected and tested in a biochemical VapC1 activity assay, which confirmed eight compounds as VapC1 inhibitors. An additional sixty compounds were selected with structural similarities to the confirmed VapC1 inhibitors, of which twenty inhibited VapC1 activity. Intracellular target engagement of five inhibitors was indicated by the destabilization of VapC1 within bacterial cells from a cellular thermal shift assay; however, no impact on bacterial growth was observed. Thus, this virtual screening and scaffold hopping approach enabled the discovery of VapC1 ribonuclease inhibitors that might serve as starting points for preclinical development.
- Published
- 2022
23. Automated Pharmacophore Query Optimization with Genetic AlgorithmsA Case Study Using the MC4R System.
- Author
-
Lei Jia, Jinming Zou, Sung-Sau So, and Hongmao Sun
- Published
- 2007
- Full Text
- View/download PDF
24. Enhancing Specificity and Sensitivity of Pharmacophore-Based Virtual Screening by Incorporating Chemical and Shape Features-A Case Study of HIV Protease Inhibitors.
- Author
-
Deepangi Pandit, Sung-Sau So, and Hongmao Sun
- Published
- 2006
- Full Text
- View/download PDF
25. A Universal Molecular Descriptor System for Prediction of LogP, LogS, LogBB, and Absorption.
- Author
-
Hongmao Sun
- Published
- 2004
- Full Text
- View/download PDF
26. Prediction of Chemical Carcinogenicity from Molecular Structure.
- Author
-
Hongmao Sun
- Published
- 2004
- Full Text
- View/download PDF
27. Correction to: Thymine DNA glycosylase as a novel target for melanoma
- Author
-
Pietro Mancuso, Rossella Tricarico, Vikram Bhattacharjee, Laura Cosentino, Yuwaraj Kadariya, Jaroslav Jelinek, Emmanuelle Nicolas, Margret Einarson, Neil Beeharry, Karthik Devarajan, Richard A. Katz, Dorjbal G. Dorjsuren, Hongmao Sun, Anton Simeonov, Antonio Giordano, Joseph R. Testa, Guillaume Davidson, Irwin Davidson, Lionel Larue, Robert W. Sobol, Timothy J. Yen, and Alfonso Bellacosa
- Subjects
Cancer Research ,Genetics ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
28. Development of Quantitative Structure-Property Relationship Models for Early ADME Evaluation in Drug Discovery. 2. Blood-Brain Barrier Penetration.
- Author
-
Ruifeng Liu, Hongmao Sun, and Sung-Sau So
- Published
- 2001
- Full Text
- View/download PDF
29. Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens
- Author
-
Sharlyn J. Mazur, Olga Vasalatiy, Dingyin Tao, Daniel H. Appella, Subrata Debnath, Yuhong Wang, Rebecca Eells, Harichandra D. Tagad, Mark J. Henderson, Victor Clausse, Kelly Lane, Nathan P. Coussens, Christopher A. LeClair, Zhen-Dan Shi, Martin R. Webb, Min Shen, Yuhong Fang, Lynn K. Baker, Matthew D. Hall, Hongmao Sun, and Ettore Appella
- Subjects
0301 basic medicine ,Phosphopeptides ,Phosphatase ,Wip1 ,Enzyme Activators ,Peptide ,Biochemistry & Proteomics ,Biochemistry ,phosphatase ,Substrate Specificity ,Serine ,Small Molecule Libraries ,03 medical and health sciences ,high-throughput screening (HTS) ,oncogene ,enzyme kinetics ,therapeutics ,cancer ,Humans ,mass spectrometry (MS) ,Enzyme kinetics ,Surface plasmon resonance ,Molecular Biology ,chemistry.chemical_classification ,030102 biochemistry & molecular biology ,phosphorylation ,Methods and Resources ,Cell Biology ,assay ,Small molecule ,High-Throughput Screening Assays ,Protein Phosphatase 2C ,030104 developmental biology ,Enzyme ,chemistry ,RapidFire ,kinetics ,Phosphorylation ,cancer therapy ,fluorescence ,Tumor Suppressor Protein p53 ,Structural Biology & Biophysics - Abstract
Wildtype P53-induced phosphatase 1 (WIP1) is a member of the magnesium-dependent serine/threonine protein phosphatase (PPM) family and is induced by P53 in response to DNA damage. In several human cancers, the WIP1 protein is overexpressed, which is generally associated with a worse prognosis. Although WIP1 is an attractive therapeutic target, no potent, selective, and bioactive small-molecule modulator with favorable pharmacokinetics has been reported. Phosphatase enzymes are among the most challenging targets for small molecules because of the difficulty of achieving both modulator selectivity and bioavailability. Another major obstacle has been the availability of robust and physiologically relevant phosphatase assays that are suitable for high-throughput screening. Here, we describe orthogonal biochemical WIP1 activity assays that utilize phosphopeptides from native WIP1 substrates. We optimized an MS assay to quantify the enzymatically dephosphorylated peptide reaction product in a 384-well format. Additionally, a red-shifted fluorescence assay was optimized in a 1,536-well format to enable real-time WIP1 activity measurements through the detection of the orthogonal reaction product, inorganic phosphate. We validated these two optimized assays by quantitative high-throughput screening against the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection and used secondary assays to confirm and evaluate inhibitors identified in the primary screen. Five inhibitors were further tested with an orthogonal WIP1 activity assay and surface plasmon resonance binding studies. Our results validate the application of miniaturized physiologically relevant and orthogonal WIP1 activity assays to discover small-molecule modulators from high-throughput screens.
- Published
- 2019
30. Retro Drug Design: From Target Properties to Molecular Structures
- Author
-
Yuhong Wang, Sam Michael, Shyh-Ming Yang, Ruili Huang, Kennie Cruz-Gutierrez, Yaqing Zhang, Jinghua Zhao, Menghang Xia, Paul Shinn, and Hongmao Sun
- Subjects
Drug ,AI drug design ,Coronavirus disease 2019 (COVID-19) ,Computer science ,media_common.quotation_subject ,General Chemical Engineering ,Drug target ,Library and Information Sciences ,Article ,Machine Learning ,Artificial Intelligence ,Molecular descriptor ,Drug Discovery ,Humans ,Pharmaceutical sciences ,media_common ,μ opioid receptor (MOR) ,retro drug design ,Molecular Structure ,Drug discovery ,Biomolecules (q-bio.BM) ,General Chemistry ,Small molecule ,Computer Science Applications ,Quantitative Biology - Biomolecules ,atom typing ,Drug Design ,FOS: Biological sciences ,Biological system ,Bbb permeability - Abstract
To generate drug molecules of desired properties with computational methods is the holy grail in pharmaceutical research. Here we describe an AI strategy, retro drug design, or RDD, to generate novel small molecule drugs from scratch to meet predefined requirements, including but not limited to biological activity against a drug target, and optimal range of physicochemical and ADMET properties. Traditional predictive models were first trained over experimental data for the target properties, using an atom typing based molecular descriptor system, ATP. Monte Carlo sampling algorithm was then utilized to find the solutions in the ATP space defined by the target properties, and the deep learning model of Seq2Seq was employed to decode molecular structures from the solutions. To test feasibility of the algorithm, we challenged RDD to generate novel drugs that can activate {\mu} opioid receptor (MOR) and penetrate blood brain barrier (BBB). Starting from vectors of random numbers, RDD generated 180,000 chemical structures, of which 78% were chemically valid. About 42,000 (31%) of the valid structures fell into the property space defined by MOR activity and BBB permeability. Out of the 42,000 structures, only 267 chemicals were commercially available, indicating a high extent of novelty of the AI-generated compounds. We purchased and assayed 96 compounds, and 25 of which were found to be MOR agonists. These compounds also have excellent BBB scores. The results presented in this paper illustrate that RDD has potential to revolutionize the current drug discovery process and create novel structures with multiple desired properties, including biological functions and ADMET properties. Availability of an AI-enabled fast track in drug discovery is essential to cope with emergent public health threat, such as pandemic of COVID-19., Comment: 27 pages, 6 figures
- Published
- 2021
31. Deep Graph Learning with Property Augmentation for Predicting Drug-Induced Liver Injury
- Author
-
Ruili Huang, Hehuan Ma, Junzhou Huang, Weizhi An, Yuhong Wang, and Hongmao Sun
- Subjects
Computer science ,Model parameters ,010501 environmental sciences ,Toxicology ,Machine learning ,computer.software_genre ,01 natural sciences ,Article ,03 medical and health sciences ,Deep Learning ,Humans ,030304 developmental biology ,0105 earth and related environmental sciences ,0303 health sciences ,Training set ,Molecular Structure ,business.industry ,Deep learning ,General Medicine ,Models, Chemical ,Pharmaceutical Preparations ,Graph (abstract data type) ,Artificial intelligence ,Chemical and Drug Induced Liver Injury ,business ,computer - Abstract
Drug-induced liver injury (DILI) is a crucial factor in determining the qualification of potential drugs. However, the DILI property is excessively difficult to obtain due to the complex testing process. Consequently, an in silico screening in the early stage of drug discovery would help to reduce the total development cost by filtering those drug candidates with a high risk to cause DILI. To serve the screening goal, we apply several computational techniques to predict the DILI property, including traditional machine learning methods and graph-based deep learning techniques. While deep learning models require large training data to tune huge model parameters, the DILI data set only contains a few hundred annotated molecules. To alleviate the data scarcity problem, we propose a property augmentation strategy to include massive training data with other property information. Extensive experiments demonstrate that our proposed method significantly outperforms all existing baselines on the DILI data set by obtaining a 81.4% accuracy using cross-validation with random splitting, 78.7% using leave-one-out cross-validation, and 76.5% using cross-validation with scaffold splitting.
- Published
- 2020
32. Improving Molecular Property Prediction on Limited Data with Deep Multi-Label Learning
- Author
-
Chaochao Yan, Junzhou Huang, Hehuan Ma, Yuhong Wang, Yuzhi Guo, Sheng Wang, and Hongmao Sun
- Subjects
0303 health sciences ,Property (programming) ,Computer science ,business.industry ,Multi label learning ,Machine learning ,computer.software_genre ,Molecular Fingerprint ,Data modeling ,030507 speech-language pathology & audiology ,03 medical and health sciences ,Sample size determination ,Molecular property ,Labeled data ,Artificial intelligence ,0305 other medical science ,business ,Encoder ,computer ,030304 developmental biology - Abstract
Acquiring labeled data has been widely recognized as a major challenge in molecular property prediction. Since it generally requires a series of specialized biochemical experiments which are time-consuming, costly, as well as labor-intensive. The deficiency of labeled property data makes it difficult to learn a good prediction model. Here, we propose an RNN-based multi-label molecular property prediction method to alleviate the data scarcity issue in two stages: 1) utilize the abundant unlabeled SMILES data to pre-train a seq2seq model whose encoder learns to generate molecular fingerprint based on the given SMILES; and 2) finetune the pre-trained model on the labeled molecular property data. Since labeled data is limited, we train those properties with limited sample size jointly with other properties which contain relatively sufficient samples. This approach brings in the idea of multi-label training, which is able to pre-train and fine-tune the encoder network, as well as train the prediction network with a data augmentation strategy. Extensive experiments on molecular property prediction demonstrate that our proposed method has achieved superior performance compared with the state-of-the-art approaches on properties with limited sample size.
- Published
- 2020
- Full Text
- View/download PDF
33. Chemoprotective antimalarials identified through quantitative high-throughput screening of Plasmodium blood and liver stage parasites
- Author
-
Paul M Will, Wenwei Huang, Stephan Meister, Dorjbal Dorjsuren, Anton Simeonov, Bryan T. Mott, Andrew T Girvin, Pranav Shah, David A. Fidock, Daniel C. Talley, Benjamin A Sigmon, Richard T. Eastman, Xin Xu, Sachel Mok, Leila S. Ross, Tomas Yeo, Daniel J. Jansen, Robert F. Campbell, Alexey V. Zakharov, Richard J. Sciotti, Carleen Klumpp-Thomas, Yevgeniya Antonova-Koch, Norman C. Waters, Juan J. Marugan, Norma Roncal, Elizabeth A. Winzeler, David J. Maloney, Kathryn J. Wicht, Ajit Jadhav, and Hongmao Sun
- Subjects
0301 basic medicine ,Parasitic infection ,Phenotypic screening ,Plasmodium berghei ,High-throughput screening ,Science ,030106 microbiology ,Plasmodium falciparum ,Drug Evaluation, Preclinical ,Pharmacology ,Protective Agents ,Plasmodium ,Article ,03 medical and health sciences ,Antimalarials ,Structure-Activity Relationship ,Parasitic Sensitivity Tests ,parasitic diseases ,Potency ,Humans ,Malaria, Falciparum ,Liver stage ,Multidisciplinary ,biology ,Chemotype ,Molecular Structure ,Thiadiazines ,Drug discovery ,Reproducibility of Results ,Hep G2 Cells ,biology.organism_classification ,High-Throughput Screening Assays ,030104 developmental biology ,Liver ,Chemoprotective ,Medicine - Abstract
The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration–response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.
- Published
- 2020
34. The AKT modulator A-443654 reduces α-synuclein expression and normalizes ER stress and autophagy
- Author
-
Stefan M. Pulst, Ganesha Rai, Warunee Dansithong, Alexey V. Zakharov, Daniel R. Scoles, Duong P. Huynh, Mandi Gandelman, Erika Aoyama, Stephen C. Kales, Anton Simeonov, Sharan Paul, Mark J. Henderson, Gentrie Maag, Hongmao Sun, Thomas S. Dexheimer, Brooke M. Whitehill, and Ajit Jadhav
- Subjects
alpha-synuclein (α-synuclein) ,autophagy ,DMEM, Dulbecco's modified Eagle medium ,iPSC, induced pluripotent stem cell ,Indazoles ,Indoles ,dCas9, deactivated Cas9 ,GF-AFC, Gly-Phe-7-amino-4-trifluoromethylcoumarin ,Cell ,CHOP ,Biochemistry ,ER, endoplasmic reticulum ,A-443654 ,UPR, unfolded protein response ,high-throughput screening (HTS) ,medicine ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,PD, Parkinson’s disease ,DCLK1, doublecortin like kinase 1 ,Kinase ,Chemistry ,AKT ,Endoplasmic reticulum ,Autophagy ,Parkinson Disease ,Cell Biology ,Endoplasmic Reticulum Stress ,nervous system diseases ,CRISPRi, CRISPR interference ,staufen1 ,HEK293 Cells ,medicine.anatomical_structure ,Gene Expression Regulation ,STAU1 ,alpha-Synuclein ,Unfolded protein response ,Cancer research ,endoplasmic reticulum stress (ER stress) ,mTOR, molecular target of rapamycin ,SNCA ,Proto-Oncogene Proteins c-akt ,Research Article - Abstract
Accumulation of α-synuclein is a main underlying pathological feature of Parkinson’s disease and α-synucleinopathies, for which lowering expression of the α-synuclein gene (SNCA) is a potential therapeutic avenue. Using a cell-based luciferase reporter of SNCA expression we performed a quantitative high-throughput screen of 155,885 compounds and identified A-443654, an inhibitor of the multiple functional kinase AKT, as a potent inhibitor of SNCA. HEK-293 cells with CAG repeat expanded ATXN2 (ATXN2-Q58 cells) have increased levels of α-synuclein. We found that A-443654 normalized levels of both SNCA mRNA and α-synuclein monomers and oligomers in ATXN2-Q58 cells. A-443654 also normalized levels of α-synuclein in fibroblasts and iPSC-derived dopaminergic neurons from a patient carrying a triplication of the SNCA gene. Analysis of autophagy and endoplasmic reticulum stress markers showed that A-443654 successfully prevented α-synuclein toxicity and restored cell function in ATXN2-Q58 cells, normalizing the levels of mTOR, LC3-II, p62, STAU1, BiP, and CHOP. A-443654 also decreased the expression of DCLK1, an inhibitor of α-synuclein lysosomal degradation. Our study identifies A-443654 and AKT inhibition as a potential strategy for reducing SNCA expression and treating Parkinson’s disease pathology.
- Published
- 2021
- Full Text
- View/download PDF
35. Human GPR17 missense variants identified in metabolic disease patients have distinct downstream signaling profiles
- Author
-
Kristin L. Ayers, Hongxia Ren, Matthew D. Hall, Min Shen, Hu Zhu, Jason M. Conley, Rong Chen, and Hongmao Sun
- Subjects
0301 basic medicine ,Nonsynonymous substitution ,Cell signaling ,Mutation, Missense ,PBS, phosphate-buffered saline ,human genetics ,FOXO1 ,Biology ,Biochemistry ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,POMC, proopiomelanocortin ,LDL, low-density lipoprotein ,Cyclic AMP ,Humans ,Missense mutation ,Molecular Biology ,beta-Arrestins ,GPCR, G-protein-coupled receptor ,G protein-coupled receptor ,Metabolic Syndrome ,Genetics ,Regulation of gene expression ,cyclic AMP (cAMP) ,calcium ,AgRP, agouti-related peptide ,030102 biochemistry & molecular biology ,arrestin ,Cell Biology ,metabolic disease ,SIR, severe insulin resistance ,G-protein-coupled receptor (GPCR) ,Human genetics ,Protein Transport ,HEK293 Cells ,FoxO1, Forkhead box protein O1 ,030104 developmental biology ,Signal transduction ,gene regulation ,signaling ,metabolism ,signal transduction ,Research Article ,HA, hemagglutinin - Abstract
GPR17 is a G-protein-coupled receptor (GPCR) implicated in the regulation of glucose metabolism and energy homeostasis. Such evidence is primarily drawn from mouse knockout studies and suggests GPR17 as a potential novel therapeutic target for the treatment of metabolic diseases. However, links between human GPR17 genetic variants, downstream cellular signaling, and metabolic diseases have yet to be reported. Here, we analyzed GPR17 coding sequences from control and disease cohorts consisting of individuals with adverse clinical metabolic deficits including severe insulin resistance, hypercholesterolemia, and obesity. We identified 18 nonsynonymous GPR17 variants, including eight variants that were exclusive to the disease cohort. We characterized the protein expression levels, membrane localization, and downstream signaling profiles of nine GPR17 variants (F43L, V96M, V103M, D105N, A131T, G136S, R248Q, R301H, and G354V). These nine GPR17 variants had similar protein expression and subcellular localization as wild-type GPR17; however, they showed diverse downstream signaling profiles. GPR17-G136S lost the capacity for agonist-mediated cAMP, Ca2+, and β-arrestin signaling. GPR17-V96M retained cAMP inhibition similar to GPR17-WT, but showed impaired Ca2+ and β-arrestin signaling. GPR17-D105N displayed impaired cAMP and Ca2+ signaling, but unaffected agonist-stimulated β-arrestin recruitment. The identification and functional profiling of naturally occurring human GPR17 variants from individuals with metabolic diseases revealed receptor variants with diverse signaling profiles, including differential signaling perturbations that resulted in GPCR signaling bias. Our findings provide a framework for structure–function relationship studies of GPR17 signaling and metabolic disease.
- Published
- 2021
- Full Text
- View/download PDF
36. Highly predictive and interpretable models for PAMPA permeability
- Author
-
Pranav Shah, Kyeong Ri Yu, Hongmao Sun, Zhengyin Yan, Edward H. Kerns, Kimloan Nguyen, Xin Xu, and Ajit Jadhav
- Subjects
0301 basic medicine ,Cell Membrane Permeability ,Support Vector Machine ,Cell membrane permeability ,In silico ,Clinical Biochemistry ,Pharmaceutical Science ,Bioinformatics ,Models, Biological ,Biochemistry ,Article ,03 medical and health sciences ,Search engine ,0302 clinical medicine ,Artificial Intelligence ,Molecular descriptor ,Drug Discovery ,Humans ,Organic Chemicals ,Molecular Biology ,Receiver operating characteristic ,Chemistry ,Drug discovery ,Organic Chemistry ,Support vector machine ,Permeability (earth sciences) ,030104 developmental biology ,030220 oncology & carcinogenesis ,Regression Analysis ,Molecular Medicine ,Caco-2 Cells ,Biological system - Abstract
Cell membrane permeability is an important determinant for oral absorption and bioavailability of a drug molecule. An in-silico model predicting drug permeability is described, which is built based on a large permeability dataset of 7488 compound entries or 5,435 structurally unique molecules measured by the same lab using parallel artificial membrane permeability assay (PAMPA). On the basis of customized molecular descriptors, the support vector regression (SVR) model trained with 4,071 compounds with quantitative data is able to predict the remaining 1,364 compounds with the qualitative data with an area under the curve of receiver operating characteristic (AUC-ROC) of 0.90. The support vector classification (SVC) model trained with half of the whole dataset comprised of both the quantitative and the qualitative data produced accurate predictions to the remaining data with the AUC-ROC of 0.88. The results suggest that the developed SVR model is highly predictive and provides medicinal chemists a useful in-silico tool to facilitate design and synthesis of novel compounds with optimal drug-like properties, and thus accelerate the lead optimization in drug discovery.
- Published
- 2017
- Full Text
- View/download PDF
37. SMILES-BERT
- Author
-
Hongmao Sun, Yuhong Wang, Yuzhi Guo, Junzhou Huang, and Sheng Wang
- Subjects
0301 basic medicine ,Computer science ,business.industry ,Deep learning ,0206 medical engineering ,Mechanism based ,02 engineering and technology ,Semi-supervised learning ,Machine learning ,computer.software_genre ,03 medical and health sciences ,030104 developmental biology ,Labeled data ,Artificial intelligence ,business ,computer ,020602 bioinformatics ,Pace - Abstract
With the rapid progress of AI in both academia and industry, Deep Learning has been widely introduced into various areas in drug discovery to accelerate its pace and cut R&D costs. Among all the problems in drug discovery, molecular property prediction has been one of the most important problems. Unlike general Deep Learning applications, the scale of labeled data is limited in molecular property prediction. To better solve this problem, Deep Learning methods have started focusing on how to utilize tremendous unlabeled data to improve the prediction performance on small-scale labeled data. In this paper, we propose a semi-supervised model named SMILES-BERT, which consists of attention mechanism based Transformer Layer. A large-scale unlabeled data has been used to pre-train the model through a Masked SMILES Recovery task. Then the pre-trained model could easily be generalized into different molecular property prediction tasks via fine-tuning. In the experiments, the proposed SMILES-BERT outperforms the state-of-the-art methods on all three datasets, showing the effectiveness of our unsupervised pre-training and great generalization capability of the pre-trained model.
- Published
- 2019
- Full Text
- View/download PDF
38. Predictive models of aqueous solubility of organic compounds built on A large dataset of high integrity
- Author
-
Hongmao Sun, Pranav Shah, Kimloan Nguyen, Ed Kerns, Kyeong Ri Yu, Kabir, Yuhong Wang, and Xin Xu
- Subjects
010405 organic chemistry ,Drug discovery ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,High integrity ,Biological activity ,01 natural sciences ,Biochemistry ,Article ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Pharmacokinetics ,Pharmaceutical Preparations ,Solubility ,Computational chemistry ,Test set ,Aqueous solubility ,Drug Discovery ,Molecular Medicine ,Organic Chemicals ,Molecular Biology - Abstract
Aqueous solubility is one of the most important properties in drug discovery, as it has profound impact on various drug properties, including biological activity, pharmacokinetics (PK), toxicity, and in vivo efficacy. Both kinetic and thermodynamic solubilities are determined during different stages of drug discovery and development. Since kinetic solubility is more relevant in preclinical drug discovery research, especially during the structure optimization process, we have developed predictive models for kinetic solubility with in-house data generated from 11,780 compounds collected from over 200 NCATS intramural research projects. This represents one of the largest kinetic solubility datasets of high quality and integrity. Based on the customized atom type descriptors, the support vector classification (SVC) models were trained on 80% of the whole dataset, and exhibited high predictive performance for estimating the solubility of the remaining 20% compounds within the test set. The values of the area under the receiver operating characteristic curve (AUC-ROC) for the compounds in the test sets reached 0.93 and 0.91, when the threshold for insoluble compounds was set to 10 and 50 μg/mL respectively. The predictive models of aqueous solubility can be used to identify insoluble compounds in drug discovery pipeline, provide design ideas for improving solubility by analyzing the atom types associated with poor solubility and prioritize compound libraries to be purchased or synthesized.
- Published
- 2019
39. Thymine DNA glycosylase as a novel target for melanoma
- Author
-
Dorjbal Dorjsuren, Guillaume Davidson, Karthik Devarajan, Jaroslav Jelinek, Irwin Davidson, Rossella Tricarico, Timothy J. Yen, Anton Simeonov, Margret B. Einarson, Neil Beeharry, Joseph R. Testa, Vikram Bhattacharjee, Antonio Giordano, Laura Cosentino, Robert W. Sobol, Richard A. Katz, Yuwaraj Kadariya, Pietro Mancuso, Lionel Larue, Hongmao Sun, Emmanuelle Nicolas, Alfonso Bellacosa, Jadavpur University, MD Anderson Cancer Center [Houston], The University of Texas Health Science Center at Houston (UTHealth), Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Fox Chase Cancer Center, Institut de génétique et biologie moléculaire et cellulaire (IGBMC), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Louis Pasteur - Strasbourg I, Signalisation normale et pathologique de l'embryon aux thérapies innovantes des cancers, Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université Louis Pasteur - Strasbourg I-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Signalisation normale et pathologique de l'embryon aux thérapies innovante des cancers, and Institut Curie-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Cancer Research ,DNA repair ,Melanoma, Experimental ,Mice, Transgenic ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mice, SCID ,Biology ,Article ,03 medical and health sciences ,Cytosine ,0302 clinical medicine ,1234567890() ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Enzyme Inhibitors ,Molecular Biology ,Melanoma ,ComputingMilieux_MISCELLANEOUS ,Cell Proliferation ,Mice, Knockout ,Cell Cycle ,Base excision repair ,Cell cycle ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,Thymine DNA Glycosylase ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,DNA demethylation ,030220 oncology & carcinogenesis ,Cancer cell ,DNA methylation ,Cancer research ,Female ,Thymine-DNA glycosylase - Abstract
Melanoma is an aggressive neoplasm with increasing incidence that is classified by the NCI as a recalcitrant cancer, i.e., a cancer with poor prognosis, lacking progress in diagnosis and treatment. In addition to conventional therapy, melanoma treatment is currently based on targeting the BRAF/MEK/ERK signaling pathway and immune checkpoints. As drug resistance remains a major obstacle to treatment success, advanced therapeutic approaches based on novel targets are still urgently needed. We reasoned that the base excision repair enzyme thymine DNA glycosylase (TDG) could be such a target for its dual role in safeguarding the genome and the epigenome, by performing the last of the multiple steps in DNA demethylation. Here we show that TDG knockdown in melanoma cell lines causes cell cycle arrest, senescence, and death by mitotic alterations; alters the transcriptome and methylome; and impairs xenograft tumor formation. Importantly, untransformed melanocytes are minimally affected by TDG knockdown, and adult mice with conditional knockout of Tdg are viable. Candidate TDG inhibitors, identified through a high-throughput fluorescence-based screen, reduced viability and clonogenic capacity of melanoma cell lines and increased cellular levels of 5-carboxylcytosine, the last intermediate in DNA demethylation, indicating successful on-target activity. These findings suggest that TDG may provide critical functions specific to cancer cells that make it a highly suitable anti-melanoma drug target. By potentially disrupting both DNA repair and the epigenetic state, targeting TDG may represent a completely new approach to melanoma therapy.
- Published
- 2019
- Full Text
- View/download PDF
40. Identification of SARS-CoV-2 viral entry inhibitors using machine learning and cell-based pseudotyped particle assay
- Author
-
Catherine Z. Chen, Miao Xu, Wei Zheng, Min Shen, Hui Guo, Yuhong Wang, Misha Itkin, and Hongmao Sun
- Subjects
Support Vector Machine ,Clinical Biochemistry ,Consensus prediction ,Pharmaceutical Science ,Microbial Sensitivity Tests ,Computational biology ,Antiviral Agents ,01 natural sciences ,Biochemistry ,Article ,Small Molecule Libraries ,Viral entry ,Molecular descriptor ,Drug Discovery ,Humans ,Pseudotyped particles assay ,Molecular Biology ,Repurposing ,ComputingMethodologies_COMPUTERGRAPHICS ,010405 organic chemistry ,Chemistry ,Drug discovery ,SARS-CoV-2 ,Organic Chemistry ,Drug Repositioning ,COVID-19 ,Virus Internalization ,0104 chemical sciences ,Support vector machine ,010404 medicinal & biomolecular chemistry ,Drug repositioning ,HEK293 Cells ,ROC Curve ,Drug development ,Area Under Curve ,Molecular Medicine ,Support vector machine (SVM) ,Databases, Chemical - Abstract
Graphical abstract, In response to the pandemic caused by SARS-CoV-2, we constructed a hybrid support vector machine (SVM) classification model using a set of publicly posted SARS-CoV-2 pseudotyped particle (PP) entry assay repurposing screen data to identify novel potent compounds as a starting point for drug development to treat COVID-19 patients. Two different molecular descriptor systems, atom typing descriptors and 3D fingerprints (FPs), were employed to construct the SVM classification models. Both models achieved reasonable performance, with the area under the curve of receiver operating characteristic (AUC-ROC) of 0.84 and 0.82, respectively. The consensus prediction outperformed the two individual models with significantly improved AUC-ROC of 0.91, where the compounds with inconsistent classifications were excluded. The consensus model was then used to screen the 173,898 compounds in the NCATS annotated and diverse chemical libraries. Of the 255 compounds selected for experimental confirmation, 116 compounds exhibited inhibitory activities in the SARS-CoV-2 PP entry assay with IC50 values ranged between 0.17 µM and 62.2 µM, representing an enrichment factor of 3.2. These 116 active compounds with diverse and novel structures could potentially serve as starting points for chemistry optimization for COVID-19 drug discovery.
- Published
- 2021
41. A physicochemical descriptor-based scoring scheme for effective and rapid filtering of kinase-like chemical space.
- Author
-
Narender Singh, Hongmao Sun, Sidhartha Chaudhury, Mohamed AbdulHameed, Anders Wallqvist, and Gregory J. Tawa
- Published
- 2012
- Full Text
- View/download PDF
42. Human GPR17 missense variants identified in metabolic disease patients have distinct downstream signaling profiles.
- Author
-
Conley, Jason M., Hongmao Sun, Ayers, Kristin L., Hu Zhu, Rong Chen, Min Shen, Hall, Matthew D., and Hongxia Ren
- Subjects
- *
MISSENSE mutation , *METABOLIC disorders , *GENETIC variation , *INSULIN resistance , *METABOLIC regulation , *ARRESTINS - Abstract
GPR17 is a G-protein-coupled receptor (GPCR) implicated in the regulation of glucose metabolism and energy homeostasis. Such evidence is primarily drawn from mouse knockout studies and suggests GPR17 as a potential novel therapeutic target for the treatment of metabolic diseases. However, links between human GPR17 genetic variants, downstream cellular signaling, and metabolic diseases have yet to be reported. Here, we analyzed GPR17 coding sequences from control and disease cohorts consisting of individuals with adverse clinical metabolic deficits including severe insulin resistance, hypercholesterolemia, and obesity. We identified 18 nonsynonymous GPR17 variants, including eight variants that were exclusive to the disease cohort. We characterized the protein expression levels, membrane localization, and downstream signaling profiles of nine GPR17 variants (F43L, V96M, V103M, D105N, A131T, G136S, R248Q, R301H, and G354V). These nine GPR17 variants had similar protein expression and subcellular localization as wild-type GPR17; however, they showed diverse downstream signaling profiles. GPR17-G136S lost the capacity for agonist-mediated cAMP, Ca2+, and ß-arrestin signaling. GPR17-V96M retained cAMP inhibition similar to GPR17-WT, but showed impaired Ca2+ and ß-arrestin signaling. GPR17-D105N displayed impaired cAMP and Ca2+ signaling, but unaffected agonist-stimulated ß-arrestin recruitment. The identification and functional profiling of naturally occurring human GPR17 variants from individuals with metabolic diseases revealed receptor variants with diverse signaling profiles, including differential signaling perturbations that resulted in GPCR signaling bias. Our findings provide a framework for structure-function relationship studies of GPR17 signaling and metabolic disease. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
43. Predictive models for estimating cytotoxicity on the basis of chemical structures
- Author
-
Yuhong Wang, Matthew D. Hall, Dorian M. Cheff, Min Shen, and Hongmao Sun
- Subjects
Models, Molecular ,Support Vector Machine ,Cell Survival ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Computational biology ,01 natural sciences ,Biochemistry ,Article ,Cell Line ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Cytotoxicity ,Molecular Biology ,Cell Proliferation ,ADME ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Drug discovery ,Chemistry ,Organic Chemistry ,HEK 293 cells ,Small molecule ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,HaCaT ,Mechanism of action ,Molecular Medicine ,Drug Screening Assays, Antitumor ,medicine.symptom - Abstract
Cytotoxicity is a critical property in determining the fate of a small molecule in the drug discovery pipeline. Cytotoxic compounds are identified and triaged in both target-based and cell-based phenotypic approaches due to their off-target toxicity or on-target and on-mechanism toxicity for oncology and neurodegenerative targets. It is critical that chemical-induced cytotoxicity be reliably predicted before drug candidates advance to the late stage of development, or more ideally, before compounds are synthesized. In this study, we assessed the cell-based cytotoxicity of nearly 10,000 compounds in NCATS annotated libraries against four 'normal' cell lines (HEK 293, NIH 3T3, CRL-7250 and HaCat) using CellTiter-Glo (CTG) technology and constructed highly predictive models to estimate cytotoxicity from chemical structures. There are 5,241 non-redundant compounds having unambiguous activities in the four different cell lines, among which 11.8% compounds exhibited cytotoxicity in two or more cell lines and are thus labelled cytotoxic. The support vector classification (SVC) models trained with 80% randomly selected molecules achieved the area under the receiver operating characteristic curve (AUC-ROC) of 0.88 on average for the remaining 20% compounds in the test sets in 10 repeating experiments. Application of under-sampling rebalancing method further improved the averaged AUC-ROC to 0.90. Analysis of structural features shared by cytotoxic compounds may offer medicinal chemists heuristic design ideas to eliminate undesirable cytotoxicity. The profiling of cytotoxicity of drug-like molecules with annotated primary mechanism of action (MOA) will inform on the roles played by different targets or pathways in cellular viability. The predictive models for cytotoxicity (accessible at https://tripod.nih.gov/web_adme/cytotox.html) provide the scientific community a fast yet reliable way to prioritize molecules with little or no cytotoxicity for downstream development.
- Published
- 2020
- Full Text
- View/download PDF
44. A Practical Guide to Rational Drug Design
- Author
-
Hongmao, Sun, Hongmao, Sun, Hongmao, Sun, and Hongmao, Sun
- Abstract
This book is not going to be an exhaustive survey covering all aspects of rational drug design. Instead, it is going to provide critical know-how through real-world examples. Relevant case studies will be presented and analyzed to illustrate the following: how to optimize a lead compound whether one has high or low levels of structural information; how to derive hits from competitors' active compounds or from natural ligands of the targets; how to springboard from competitors' SAR knowledge in lead optimization; how to design a ligand to interfere with protein-protein interactions by correctly examining the PPI interface; how to circumvent IP blockage using data mining; how to construct and fully utilize a knowledge-based molecular descriptor system; how to build a reliable QSAR model by focusing on data quality and proper selection of molecular descriptors and statistical approaches. A Practical Guide to Rational Drug Design focuses on computational drug design, with only basic coverage of biology and chemistry issues, such as assay design, target validation and synthetic routes.Discusses various tactics applicable to daily drug design Readers can download the materials used in the book, including structures, scripts, raw data, protocols, and codes, making this book suitable resource for short courses or workshopsOffers a unique viewpoint on drug discovery research due to the author's cross-discipline education background Explores the author's rich experiences in both pharmaceutical and academic settings
- Published
- 2015
45. Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing
- Author
-
Eugene N. Muratov, Rosita R. Asawa, Ajit Jadhav, Anton Simeonov, Alexander Tropsha, Elizabeth Cunningham, William P. Janzen, Hongmao Sun, Stephen J. Capuzzi, Sherif Farag, Min Shen, Julie Blatt, Yaqin Zhang, and Natalia J. Martinez
- Subjects
0301 basic medicine ,drug repurposing ,3D cultures ,business.industry ,Phenotypic screening ,Pharmacy ,Computational biology ,Pediatric cancer ,Antiparasitic agent ,pediatric cancer ,03 medical and health sciences ,Drug repositioning ,quantitative high-throughput screening ,030104 developmental biology ,Oncology ,Medicine ,Viability assay ,Dosing ,business ,Repurposing ,Research Paper - Abstract
Drug repurposing approaches have the potential advantage of facilitating rapid and cost-effective development of new therapies. Particularly, the repurposing of drugs with known safety profiles in children could bypass or streamline toxicity studies. We employed a phenotypic screening paradigm on a panel of well-characterized cell lines derived from pediatric solid tumors against a collection of ∼3,800 compounds spanning approved drugs and investigational agents. Specifically, we employed titration-based screening where compounds were tested at multiple concentrations for their effect on cell viability. Molecular and cellular target enrichment analysis indicated that numerous agents across different therapeutic categories and modes of action had an antiproliferative effect, notably antiparasitic/protozoal drugs with non-classic antineoplastic activity. Focusing on active compounds with dosing and safety information in children according to the Children's Pharmacy Collaborative database, we identified compounds with therapeutic potential through further validation using 3D tumor spheroid models. Moreover, we show that antiparasitic agents induce cell death via apoptosis induction. This study demonstrates that our screening platform enables the identification of chemical agents with cytotoxic activity in pediatric cancer cell lines of which many have known safety/toxicity profiles in children. These agents constitute attractive candidates for efficacy studies in pre-clinical models of pediatric solid tumors.
- Published
- 2017
46. Synthesis and Structure–Activity Relationship Studies of N-Benzyl-2-phenylpyrimidin-4-amine Derivatives as Potent USP1/UAF1 Deubiquitinase Inhibitors with Anticancer Activity against Nonsmall Cell Lung Cancer
- Author
-
Junjun Chen, Mark A. Villamil, Anton Simeonov, Qin Liang, Zhihao Zhuang, Hongmao Sun, David J. Maloney, Thomas S. Dexheimer, Edward H. Kerns, Diane K. Luci, Andrew S. Rosenthal, and Ajit Jadhav
- Subjects
Lung Neoplasms ,DNA damage ,Druggability ,Antineoplastic Agents ,Article ,Deubiquitinating enzyme ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Ubiquitin ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Proliferating Cell Nuclear Antigen ,Drug Discovery ,medicine ,Structure–activity relationship ,Humans ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Arabidopsis Proteins ,Ubiquitination ,Cancer ,Nuclear Proteins ,medicine.disease ,Small molecule ,3. Good health ,Pyrimidines ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,Ubiquitin-Specific Proteases - Abstract
Deregulation of ubiquitin conjugation or deconjugation has been implicated in the pathogenesis of many human diseases including cancer. The deubiquitinating enzyme USP1 (ubiquitin-specific protease 1), in association with UAF1 (USP1-associated factor 1), is a known regulator of DNA damage response and has been shown as a promising anticancer target. To further evaluate USP1/UAF1 as a therapeutic target, we conducted a quantitative high throughput screen of >400000 compounds and subsequent medicinal chemistry optimization of small molecules that inhibit the deubiquitinating activity of USP1/UAF1. Ultimately, these efforts led to the identification of ML323 (70) and related N-benzyl-2-phenylpyrimidin-4-amine derivatives, which possess nanomolar USP1/UAF1 inhibitory potency. Moreover, we demonstrate a strong correlation between compound IC50 values for USP1/UAF1 inhibition and activity in nonsmall cell lung cancer cells, specifically increased monoubiquitinated PCNA (Ub-PCNA) levels and decreased cell survival. Our results establish the druggability of the USP1/UAF1 deubiquitinase complex and its potential as a molecular target for anticancer therapies.
- Published
- 2014
47. Development of a multitask deep learning QSAR model using data from individual cytochrome P450 isozymes
- Author
-
Eric Gonzalez, Pranav Shah, Alexey V. Zakharov, Dac-Trung guyen, Cornelis E. C. A. Hop, Hongmao Sun, Xin Xu, R. Scott Obach, and Anton Simeonov
- Subjects
Pharmacology ,0209 industrial biotechnology ,Quantitative structure–activity relationship ,biology ,Computer science ,business.industry ,Deep learning ,Pharmaceutical Science ,Cytochrome P450 ,02 engineering and technology ,Computational biology ,Isozyme ,020901 industrial engineering & automation ,0202 electrical engineering, electronic engineering, information engineering ,biology.protein ,020201 artificial intelligence & image processing ,Pharmacology (medical) ,Artificial intelligence ,business - Published
- 2018
- Full Text
- View/download PDF
48. Pyrido[2,3-d]pyrimidines: Discovery and preliminary SAR of a novel series of DYRK1B and DYRK1A inhibitors
- Author
-
Peter Michael Wovkulich, Cheryl Janson, Hongmao Sun, Yang He, Rossman Pamela Loreen, Qing Xiang, Xiaolei Zhang, Zhi Chen, Qi Qiao, Romyr Dominique, Christine Lukacs, Kin-Chun Luk, Yi Chen, Kevin W. Anderson, Aruna Railkar, Kelli Glenn, Ann Polonskaia, and Masha Vilenchik
- Subjects
DYRK1B ,DYRK1A ,Clinical Biochemistry ,Cancer therapy ,Pharmaceutical Science ,Molecular Dynamics Simulation ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,Downregulation and upregulation ,Drug Discovery ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Molecular Biology ,Binding Sites ,Chemistry ,Organic Chemistry ,Protein-Tyrosine Kinases ,Protein Structure, Tertiary ,Rats ,Enzyme Activation ,Pyrimidines ,Mechanism of action ,Cancer cell ,Molecular Medicine ,A kinase ,medicine.symptom ,Half-Life - Abstract
DYRK1B is a kinase over-expressed in certain cancer cells (including colon, ovarian, pancreatic, etc.). Recent publications have demonstrated inhibition of DYRK1B could be an attractive target for cancer therapy. From a data-mining effort, the team has discovered analogues of pyrido[2,3-d]pyrimidines as potent enantio-selective inhibitors of DYRK1B. Cells treated with a tool compound from this series showed the same cellular effects as down regulation of DYRK1B with siRNA. Such effects are consistent with the proposed mechanism of action. Progress of the SAR study is presented.
- Published
- 2013
- Full Text
- View/download PDF
49. Identification of potent Yes1 kinase inhibitors using a library screening approach
- Author
-
Mindy I. Davis, Paresma R. Patel, Samuel Q. Li, Javed Khan, Min Shen, Craig J. Thomas, and Hongmao Sun
- Subjects
Kinase ,YES1 ,Inhibitor ,Cell Survival ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Molecular Docking Simulation ,Article ,Cell Line ,Small Molecule Libraries ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Rhabdomyosarcoma ,Drug Discovery ,Humans ,Structure–activity relationship ,Protein Kinase Inhibitors ,Molecular Biology ,030304 developmental biology ,Proto-Oncogene Proteins c-yes ,0303 health sciences ,Binding Sites ,Chemistry ,Organic Chemistry ,Assay ,Yes1 ,Hydrogen Bonding ,Small molecule ,Protein Structure, Tertiary ,3. Good health ,Docking (molecular) ,Drug Design ,030220 oncology & carcinogenesis ,Screening ,Cancer research ,Molecular Medicine ,HTS - Abstract
Yes1 kinase has been implicated as a potential therapeutic target in a number of cancers including melanomas, breast cancers, and rhabdomyosarcomas. Described here is the development of a robust and miniaturized biochemical assay for Yes1 kinase that was applied in a high throughput screen (HTS) of kinase-focused small molecule libraries. The HTS provided 144 (17% hit rate) small molecule compounds with IC50 values in the sub-micromolar range. Three of the most potent Yes1 inhibitors were then examined in a cell-based assay for inhibition of cell survival in rhabdomyosarcoma cell lines. Homology models of Yes1 were generated in active and inactive conformations, and docking of inhibitors supports binding to the active conformation (DFG-in) of Yes1. This is the first report of a large high throughput enzymatic activity screen for identification of Yes1 kinase inhibitors, thereby elucidating the polypharmacology of a variety of small molecules and clinical candidates.
- Published
- 2013
- Full Text
- View/download PDF
50. Genomic and functional characterizations of phosphodiesterase subtype 4D in human cancers
- Author
-
Ngan B. Doan, De-Chen Lin, Nicole Urban, Paresma R. Patel, Li Zhen Liu, Michèl Schummer, Patrick Tan, Hongmao Sun, Craig J. Thomas, Liang Xu, Beth Y. Karlan, H. Phillip Koeffler, Jonathan W. Said, Jay Vadgama, Dong Yin, Arjun Sharma, Martin J. Walsh, Danny Chan, Henry Yang, Ling-Wen Ding, and Jenny Lester
- Subjects
Apoptosis ,Biology ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Regulation of gene expression ,Microphthalmia-Associated Transcription Factor ,Multidisciplinary ,Cell Death ,Gene Expression Profiling ,Melanoma ,Genomics ,Biological Sciences ,Microphthalmia-associated transcription factor ,medicine.disease ,Immunohistochemistry ,Molecular biology ,Primary tumor ,Cyclic Nucleotide Phosphodiesterases, Type 3 ,Cyclic Nucleotide Phosphodiesterases, Type 4 ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,Proto-Oncogene Proteins c-bcl-2 ,Cell culture ,Cancer cell ,Cancer research ,Ectopic expression ,Gene Deletion - Abstract
Discovery of cancer genes through interrogation of genomic dosage is one of the major approaches in cancer research. In this study, we report that phosphodiesterase subtype 4D (PDE4D) gene was homozygously deleted in 198 cases of 5,569 primary solid tumors (3.56%), with most being internal microdeletions. Unexpectedly, the microdeletions did not result in loss of their gene products. Screening PDE4D expression in 11 different types of primary tumor samples ( n = 165) with immunohistochemistry staining revealed that its protein levels were up-regulated compared with corresponding nontransformed tissues. Importantly, depletion of endogenous PDE4D with three independent shRNAs caused apoptosis and growth inhibition in multiple types of cancer cells, including breast, lung, ovary, endometrium, gastric, and melanoma, which could be rescued by reexpression of PDE4D. We further showed that antitumor events triggered by PDE4D suppression were lineage-dependently associated with Bcl-2 interacting mediator of cell death (BIM) induction and microphthalmia-associated transcription factor (MITF) down-regulation. Furthermore, ectopic expression of the PDE4D short isoform, PDE4D2, enhanced the proliferation of cancer cells both in vitro and in vivo. Moreover, treatment of cancer cells with a unique specific PDE4D inhibitor, 26B, triggered massive cell death and growth retardation. Notably, these antineoplastic effects induced by either shRNAs or small molecule occurred preferentially in cancer cells but not in nonmalignant epithelial cells. These results suggest that although targeted by genomic homozygous microdeletions, PDE4D functions as a tumor-promoting factor and represents a unique targetable enzyme of cancer cells.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.