86 results on '"Richard T, Eastman"'
Search Results
2. Rapid-response RNA-fluorescence in situ hybridization (FISH) assay platform for coronavirus antiviral high-throughput screening
- Author
-
Ryan Chan, Christian Shema Mugisha, Vorada Chuenchob, Stephanie A. Moquin, Ujjini H. Manjunatha, Nadine Jarrousse, Vineet D. Menachery, Xuping Xie, Erika L. Flannery, and Richard T. Eastman
- Subjects
Coronavirus ,RNA-FISH ,High-content assay ,High-throughput screening ,OC43 ,229E ,Medicine (General) ,R5-920 ,Biotechnology ,TP248.13-248.65 - Abstract
Over the past 25 years, the global community has faced challenges posed by three distinct outbreaks of coronaviruses including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The identification of a novel alphacoronavirus canine CoV (CCoV-HuPn2018) in human patients in Malaysia underscores the potential for crossover infections to humans. The threat of the ever-evolving nature of viral infections as well as the lingering health and socioeconomic effects of the recent SARS-CoV-2 pandemic emphasize the urgent need for advanced antiviral drug screening tools that can be quickly implemented to strengthen preparedness and preventive measures against future outbreaks. Here, we present the development and validation of a novel RNA-fluorescence in situ hybridization (FISH) imaging assay as a 384-well, high-throughput rapid response platform for antiviral drug discovery. RNA-FISH is a powerful tool to visualize specific mRNA in cultured cells using a high-content imaging platform. The flexibility of RNA-FISH probe sets allows for the rapid design of viral genome-specific probes, enabling in vitro assay development to test for inhibition of viral replication by either biologic or small molecule inhibitors. Screening of 170 antiviral compounds in concentration-response demonstrates a strong correlation between the RNA-FISH assay and an immunofluorescence assay (IFA) for both human coronaviruses HCoV-OC43 and HCoV-229E. Additionally, we successfully applied this methodology in the context of CCoV strain 1–71, proving rapid development and deployment, opening new avenues for the evaluation of antiviral drugs to potential future emerging threats.
- Published
- 2024
- Full Text
- View/download PDF
3. 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
4. Remdesivir: A Review of Its Discovery and Development Leading to Emergency Use Authorization for Treatment of COVID-19
- Author
-
Richard T. Eastman, Jacob S. Roth, Kyle R. Brimacombe, Anton Simeonov, Min Shen, Samarjit Patnaik, and Matthew D. Hall
- Subjects
Chemistry ,QD1-999 - Published
- 2020
- Full Text
- View/download PDF
5. A platform of assays for the discovery of anti-Zika small-molecules with activity in a 3D-bioprinted outer-blood-retina model.
- Author
-
Dorjbal Dorjsuren, Richard T Eastman, Min Jae Song, Adam Yasgar, Yuchi Chen, Kapil Bharti, Alexey V Zakharov, Ajit Jadhav, Marc Ferrer, Pei-Yong Shi, and Anton Simeonov
- Subjects
Medicine ,Science - Abstract
The global health emergency posed by the outbreak of Zika virus (ZIKV), an arthropod-borne flavivirus causing severe neonatal neurological conditions, has subsided, but there continues to be transmission of ZIKV in endemic regions. As such, there is still a medical need for discovering and developing therapeutical interventions against ZIKV. To identify small-molecule compounds that inhibit ZIKV disease and transmission, we screened multiple small-molecule collections, mostly derived from natural products, for their ability to inhibit wild-type ZIKV. As a primary high-throughput screen, we used a viral cytopathic effect (CPE) inhibition assay conducted in Vero cells that was optimized and miniaturized to a 1536-well format. Suitably active compounds identified from the primary screen were tested in a panel of orthogonal assays using recombinant Zika viruses, including a ZIKV Renilla luciferase reporter assay and a ZIKV mCherry reporter system. Compounds that were active in the wild-type ZIKV inhibition and ZIKV reporter assays were further evaluated for their inhibitory effects against other flaviviruses. Lastly, we demonstrated that wild-type ZIKV is able to infect a 3D-bioprinted outer-blood-retina barrier tissue model and disrupt its barrier function, as measured by electrical resistance. One of the identified compounds (3-Acetyl-13-deoxyphomenone, NCGC00380955) was able to prevent the pathological effects of the viral infection on this clinically relevant ZIKV infection model.
- Published
- 2022
- Full Text
- View/download PDF
6. A humanized nanobody phage display library yields potent binders of SARS CoV-2 spike.
- Author
-
Ying Fu, Juliana da Fonseca Rezende E Mello, Bryan D Fleming, Alex Renn, Catherine Z Chen, Xin Hu, Miao Xu, Kirill Gorshkov, Quinlin Hanson, Wei Zheng, Emily M Lee, Lalith Perera, Robert Petrovich, Manisha Pradhan, Richard T Eastman, Zina Itkin, Thomas B Stanley, Allen Hsu, Venkata Dandey, Kedar Sharma, William Gillette, Troy Taylor, Nitya Ramakrishnan, Shelley Perkins, Dominic Esposito, Eunkeu Oh, Kimihiro Susumu, Mason Wolak, Marc Ferrer, Matthew D Hall, Mario J Borgnia, and Anton Simeonov
- Subjects
Medicine ,Science - Abstract
Neutralizing antibodies targeting the SARS-CoV-2 spike protein have shown a great preventative/therapeutic potential. Here, we report a rapid and efficient strategy for the development and design of SARS-CoV-2 neutralizing humanized nanobody constructs with sub-nanomolar affinities and nanomolar potencies. CryoEM-based structural analysis of the nanobodies in complex with spike revealed two distinct binding modes. The most potent nanobody, RBD-1-2G(NCATS-BL8125), tolerates the N501Y RBD mutation and remains capable of neutralizing the B.1.1.7 (Alpha) variant. Molecular dynamics simulations provide a structural basis for understanding the neutralization process of nanobodies exclusively focused on the spike-ACE2 interface with and without the N501Y mutation on RBD. A primary human airway air-lung interface (ALI) ex vivo model showed that RBD-1-2G-Fc antibody treatment was effective at reducing viral burden following WA1 and B.1.1.7 SARS-CoV-2 infections. Therefore, this presented strategy will serve as a tool to mitigate the threat of emerging SARS-CoV-2 variants.
- Published
- 2022
- Full Text
- View/download PDF
7. Plasmodium vivax chloroquine resistance links to pvcrt transcription in a genetic cross
- Author
-
Juliana M. Sá, Sarah R. Kaslow, Roberto R. Moraes Barros, Nicholas F. Brazeau, Christian M. Parobek, Dingyin Tao, Rebecca E. Salzman, Tyler J. Gibson, Soundarapandian Velmurugan, Michael A. Krause, Viviana Melendez-Muniz, Whitney A. Kite, Paul K. Han, Richard T. Eastman, Adam Kim, Evan G. Kessler, Yonas Abebe, Eric R. James, Sumana Chakravarty, Sachy Orr-Gonzalez, Lynn E. Lambert, Theresa Engels, Marvin L. Thomas, Pius S. Fasinu, David Serre, Robert W. Gwadz, Larry Walker, Derrick K. DeConti, Jianbing Mu, Jeffrey A. Bailey, B. Kim Lee Sim, Stephen L. Hoffman, Michael P. Fay, Rhoel R. Dinglasan, Jonathan J. Juliano, and Thomas E. Wellems
- Subjects
Science - Abstract
Here, a cross of Plasmodium vivax malaria parasites links a chloroquine resistance (CQR) phenotype to a 76 kb region of chromosome 1 and greater expression of pvcrt, an ortholog of the Plasmodium falciparum CQR transporter gene.
- Published
- 2019
- Full Text
- View/download PDF
8. Quantitative Bioactivity Signatures of Dietary Supplements and Natural Products
- Author
-
Adam Yasgar, Danielle Bougie, Richard T. Eastman, Ruili Huang, Misha Itkin, Jennifer Kouznetsova, Caitlin Lynch, Crystal McKnight, Mitch Miller, Deborah K. Ngan, Tyler Peryea, Pranav Shah, Paul Shinn, Menghang Xia, Xin Xu, Alexey V. Zakharov, and Anton Simeonov
- Subjects
Pharmacology ,Pharmacology (medical) - Published
- 2023
- Full Text
- View/download PDF
9. Drug Repurposing Screen for Compounds Inhibiting the Cytopathic Effect of SARS-CoV-2
- Author
-
Catherine Z. Chen, Paul Shinn, Zina Itkin, Richard T. Eastman, Robert Bostwick, Lynn Rasmussen, Ruili Huang, Min Shen, Xin Hu, Kelli M. Wilson, Brianna M. Brooks, Hui Guo, Tongan Zhao, Carleen Klump-Thomas, Anton Simeonov, Samuel G. Michael, Donald C. Lo, Matthew D. Hall, and Wei Zheng
- Subjects
COVID-19 ,cytopathic effect ,drug repurposing and discovery ,HTS ,SARS-CoV-2 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Drug repurposing is a rapid approach to identify therapeutics for the treatment of emerging infectious diseases such as COVID-19. To address the urgent need for treatment options, we carried out a quantitative high-throughput screen using a SARS-CoV-2 cytopathic assay with a compound collection of 8,810 approved and investigational drugs, mechanism-based bioactive compounds, and natural products. Three hundred and nineteen compounds with anti-SARS-CoV-2 activities were identified and confirmed, including 91 approved drugs and 49 investigational drugs. The anti-SARS-CoV-2 activities of 230 of these confirmed compounds, of which 38 are approved drugs, have not been previously reported. Chlorprothixene, methotrimeprazine, and piperacetazine were the three most potent FDA-approved drugs with anti-SARS-CoV-2 activities. These three compounds have not been previously reported to have anti-SARS-CoV-2 activities, although their antiviral activities against SARS-CoV and Ebola virus have been reported. These results demonstrate that this comprehensive data set is a useful resource for drug repurposing efforts, including design of new drug combinations for clinical trials for SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
10. Canvass: A Crowd-Sourced, Natural-Product Screening Library for Exploring Biological Space
- Author
-
Sara E. Kearney, Gergely Zahoránszky-Kőhalmi, Kyle R. Brimacombe, Mark J. Henderson, Caitlin Lynch, Tongan Zhao, Kanny K. Wan, Zina Itkin, Christopher Dillon, Min Shen, Dorian M. Cheff, Tobie D. Lee, Danielle Bougie, Ken Cheng, Nathan P. Coussens, Dorjbal Dorjsuren, Richard T. Eastman, Ruili Huang, Michael J. Iannotti, Surendra Karavadhi, Carleen Klumpp-Thomas, Jacob S. Roth, Srilatha Sakamuru, Wei Sun, Steven A. Titus, Adam Yasgar, Ya-Qin Zhang, Jinghua Zhao, Rodrigo B. Andrade, M. Kevin Brown, Noah Z. Burns, Jin K. Cha, Emily E. Mevers, Jon Clardy, Jason A. Clement, Peter A. Crooks, Gregory D. Cuny, Jake Ganor, Jesus Moreno, Lucas A. Morrill, Elias Picazo, Robert B. Susick, Neil K. Garg, Brian C. Goess, Robert B. Grossman, Chambers C. Hughes, Jeffrey N. Johnston, Madeleine M. Joullie, A. Douglas Kinghorn, David G.I. Kingston, Michael J. Krische, Ohyun Kwon, Thomas J. Maimone, Susruta Majumdar, Katherine N. Maloney, Enas Mohamed, Brian T. Murphy, Pavel Nagorny, David E. Olson, Larry E. Overman, Lauren E. Brown, John K. Snyder, John A. Porco, Fatima Rivas, Samir A. Ross, Richmond Sarpong, Indrajeet Sharma, Jared T. Shaw, Zhengren Xu, Ben Shen, Wei Shi, Corey R.J. Stephenson, Alyssa L. Verano, Derek S. Tan, Yi Tang, Richard E. Taylor, Regan J. Thomson, David A. Vosburg, Jimmy Wu, William M. Wuest, Armen Zakarian, Yufeng Zhang, Tianjing Ren, Zhong Zuo, James Inglese, Sam Michael, Anton Simeonov, Wei Zheng, Paul Shinn, Ajit Jadhav, Matthew B. Boxer, Matthew D. Hall, Menghang Xia, Rajarshi Guha, and Jason M. Rohde
- Subjects
Chemistry ,QD1-999 - Published
- 2018
- Full Text
- View/download PDF
11. A systematic and prospectively validated approach for identifying synergistic drug combinations against malaria
- Author
-
Yasaman KalantarMotamedi, Richard T. Eastman, Rajarshi Guha, and Andreas Bender
- Subjects
Synergy prediction ,Malaria ,Machine learning ,Compound combination modelling ,Transcriptional drug repositioning ,Synergistic anti-malaria compound combinations ,Arctic medicine. Tropical medicine ,RC955-962 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Nearly half of the world’s population (3.2 billion people) were at risk of malaria in 2015, and resistance to current therapies is a major concern. While the standard of care includes drug combinations, there is a pressing need to identify new combinations that can bypass current resistance mechanisms. In the work presented here, a combined transcriptional drug repositioning/discovery and machine learning approach is proposed. Methods The integrated approach utilizes gene expression data from patient-derived samples, in combination with large-scale anti-malarial combination screening data, to predict synergistic compound combinations for three Plasmodium falciparum strains (3D7, DD2 and HB3). Both single compounds and combinations predicted to be active were prospectively tested in experiment. Results One of the predicted single agents, apicidin, was active with the AC50 values of 74.9, 84.1 and 74.9 nM in 3D7, DD2 and HB3 P. falciparum strains while its maximal safe plasma concentration in human is 547.6 ± 136.6 nM. Apicidin at the safe dose of 500 nM kills on average 97% of the parasite. The synergy prediction algorithm exhibited overall precision and recall of 83.5 and 65.1% for mild-to-strong, 48.8 and 75.5% for moderate-to-strong and 12.0 and 62.7% for strong synergies. Some of the prospectively predicted combinations, such as tacrolimus-hydroxyzine and raloxifene-thioridazine, exhibited significant synergy across the three P. falciparum strains included in the study. Conclusions Systematic approaches can play an important role in accelerating discovering novel combinational therapies for malaria as it enables selecting novel synergistic compound pairs in a more informed and cost-effective manner.
- Published
- 2018
- Full Text
- View/download PDF
12. A Plasmodium yoelii HECT-like E3 ubiquitin ligase regulates parasite growth and virulence
- Author
-
Sethu C. Nair, Ruixue Xu, Sittiporn Pattaradilokrat, Jian Wu, Yanwei Qi, Martine Zilversmit, Sundar Ganesan, Vijayaraj Nagarajan, Richard T. Eastman, Marlene S. Orandle, John C. Tan, Timothy G. Myers, Shengfa Liu, Carole A. Long, Jian Li, and Xin-zhuan Su
- Subjects
Science - Abstract
Many strains of Plasmodium differ in virulence, but factors that control these distinctions are not known. Here the authors comparatively map virulence loci using the offspring from a P. yoelii YM and N67 genetic cross, and identify a putative HECT E3 ubiquitin ligase that may explain the variance.
- Published
- 2017
- Full Text
- View/download PDF
13. Deep learning identifies synergistic drug combinations for treating COVID-19.
- Author
-
Wengong Jin, Jonathan M. Stokes, Richard T. Eastman, Zina Itkin, Alexey V. Zakharov, James J. Collins, Tommi S. Jaakkola, and Regina Barzilay
- Published
- 2021
- Full Text
- View/download PDF
14. Novel Transmission-Blocking Antimalarials Identified by High-Throughput Screening of Plasmodium berghei Ookluc
- Author
-
Juliana Calit, Jessica E. Araújo, Bingbing Deng, Kazutoyo Miura, Xiomara A. Gaitán, Maisa da Silva Araújo, Jansen F. Medeiros, Carole A. Long, Anton Simeonov, Richard T. Eastman, and Daniel Y. Bargieri
- Subjects
Pharmacology ,Infectious Diseases ,Pharmacology (medical) - Abstract
Safe and effective malaria transmission-blocking chemotherapeutics would allow a community-level approach to malaria control and eradication efforts by targeting the mosquito sexual stage of the parasite life cycle. However, only a single drug, primaquine, is currently approved for use in reducing transmission, and drug toxicity limits its widespread implementation.
- Published
- 2023
- Full Text
- View/download PDF
15. A Plasmodium falciparum RING Finger E3 Ubiquitin Ligase Modifies the Roles of PfMDR1 and PfCRT in Parasite Drug Responses
- Author
-
Brajesh K. Singh, Cui Zhang, Jian Wu, Yu-Chih Peng, Xiao He, Keyla C. Tumas, Juliana M. Sá, Kristin D. Lane, Richard T. Eastman, David L. Narum, Thomas E. Wellems, and Xin-zhuan Su
- Subjects
Pharmacology ,Infectious Diseases ,Pharmacology (medical) - Abstract
Protein ubiquitination is an important posttranslational regulation mechanism that mediates Plasmodium development and modifies parasite responses to antimalarial drugs. Although mutations in several parasite ubiquitination enzymes have been linked to increased drug tolerance, the molecular mechanisms by which ubiquitination pathways mediate these parasite responses remain largely unknown.
- Published
- 2023
- Full Text
- View/download PDF
16. Correction to Remdesivir: A Review of Its Discovery and Development Leading to Human Clinical Trials for Treatment of COVID-19
- Author
-
Richard T. Eastman, Jacob S. Roth, Kyle R. Brimacombe, Anton Simeonov, Min Shen, Samarjit Patnaik, and Matthew D. Hall
- Subjects
Chemistry ,QD1-999 - Published
- 2020
- Full Text
- View/download PDF
17. Using Machine Learning to Predict Synergistic Antimalarial Compound Combinations With Novel Structures
- Author
-
Daniel J. Mason, Richard T. Eastman, Richard P. I. Lewis, Ian P. Stott, Rajarshi Guha, and Andreas Bender
- Subjects
synergy ,combinations ,malaria ,plasmodium falciparum ,artificial intelligence ,modeling ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The parasite Plasmodium falciparum is the most lethal species of Plasmodium to cause serious malaria infection in humans, and with resistance developing rapidly novel treatment modalities are currently being sought, one of which being combinations of existing compounds. The discovery of combinations of antimalarial drugs that act synergistically with one another is hence of great importance; however an exhaustive experimental screen of large drug space in a pairwise manner is not an option. In this study we apply our machine learning approach, Combination Synergy Estimation (CoSynE), which can predict novel synergistic drug interactions using only prior experimental combination screening data and knowledge of compound molecular structures, to a dataset of 1,540 antimalarial drug combinations in which 22.2% were synergistic. Cross validation of our model showed that synergistic CoSynE predictions are enriched 2.74 × compared to random selection when both compounds in a predicted combination are known from other combinations among the training data, 2.36 × when only one compound is known from the training data, and 1.5 × for entirely novel combinations. We prospectively validated our model by making predictions for 185 combinations of 23 entirely novel compounds. CoSynE predicted 20 combinations to be synergistic, which was experimentally validated for nine of them (45%), corresponding to an enrichment of 1.70 × compared to random selection from this prospective data set. Such enrichment corresponds to a 41% reduction in experimental effort. Interestingly, we found that pairwise screening of the compounds CoSynE individually predicted to be synergistic would result in an enrichment of 1.36 × compared to random selection, indicating that synergy among compound combinations is not a random event. The nine novel and correctly predicted synergistic compound combinations mainly (where sufficient bioactivity information is available) consist of efflux or transporter inhibitors (such as hydroxyzine), combined with compounds exhibiting antimalarial activity alone (such as sorafenib, apicidin, or dihydroergotamine). However, not all compound synergies could be rationalized easily in this way. Overall, this study highlights the potential for predictive modeling to expedite the discovery of novel drug combinations in fight against antimalarial resistance, while the underlying approach is also generally applicable.
- Published
- 2018
- Full Text
- View/download PDF
18. Quantitative bioactivity signatures of dietary supplements and natural products
- Author
-
Adam Yasgar, Danielle Bougie, Richard T. Eastman, Ruili Huang, Misha Itkin, Jennifer Kouznetsova, Caitlin Lynch, Crystal McKnight, Mitch Miller, Deborah K. Ngan, Tyler Peryea, Pranav Shah, Paul Shinn, Menghang Xia, Alexey V. Zakharov, and Anton Simeonov
- Abstract
Dietary Supplements and Natural Products have minor oversight of their safety and efficacy. We assembled a collection of Dietary Supplements and Natural Products (DSNP) as well as Traditional Chinese Medicinal (TCM) Plant extracts, which were screened against an in vitro panel of assays, including a liver cytochrome p450 enzyme panel, CAR/PXR signaling pathways, and P-gp transporter assays, to assess their activity. This pipeline facilitated the interrogation of Natural Product-Drug Interaction (NaPDI) through prominent metabolizing pathways. In addition, we compared the activity profiles of the DSNP/TCM substances with those of an approved drug collection. Many of the approved drugs have well-annotated mechanisms of action (MOA) while the MOAs for most of the DSNP and TCM samples remain unknown. Based on the premise that compounds with similar activity profiles tend to share similar targets or MOA, we clustered the library activity profiles to identify overlap with the NCATS Pharmaceutical Collection to predict the MOAs of the DSNP/TCM substances. Overall, we highlight four significant bioactivity profiles (measured by p-values) as examples of this prediction. These results can be used as a starting point for further exploration on the toxicity potential and clinical relevance of these substances.
- Published
- 2022
- Full Text
- View/download PDF
19. Small Molecule Antiviral Compound Collection (SMACC): a database to support the discovery of broad-spectrum antiviral drug molecules
- Author
-
Holli-Joi Martin, Cleber C. Melo-Filho, Daniel Korn, Richard T. Eastman, Ganesha Rai, Anton Simeonov, Alexey V. Zakharov, Eugene Muratov, and Alexander Tropsha
- Subjects
Article - Abstract
Diseases caused by new viruses costs thousands if not millions of human lives and trillions of dollars in damage to the global economy. Despite the rapid development of vaccines for SARS-CoV-2, the lack of small molecule antiviral drugs that work against multiple viral families (broad-spectrum antivirals; BSAs) has left the entire world’s human population vulnerable to the infection between the beginning of the outbreak and the widespread availability of vaccines. Developing BSAs is an attractive, yet challenging, approach that could prevent the next, inevitable, viral outbreak from becoming a global catastrophe. To explore whether historical medicinal chemistry efforts suggest the possibility of discovering novel BSAs, we (i) identified, collected, curated, and integrated all chemical bioactivity data available in ChEMBL for molecules tested in respective assays for 13 emerging viruses that, based on published literature, hold the greatest potential threat to global human health; (ii) identified and solved the challenges related to data annotation accuracy including assay description ambiguity, missing cell or target information, and incorrect BioAssay Ontology (BAO) annotations; (iii) developed a highly curated and thoroughly annotated database of compounds tested in both phenotypic (21,392 entries) and target-based (11,123 entries) assays for these viruses; and (iv) identified a subset of compounds showing BSA activity. For the latter task, we eliminated inconclusive and annotated duplicative entries by checking the concordance between multiple assay results and identified eight compounds active against 3-4 viruses from the phenotypic data, 16 compounds active against two viruses from the target-based data, and 35 compounds active in at least one phenotypic and one target-based assay. The pilot version of our SMACC (Small Molecule Antiviral Compound Collection) database contains over 32,500 entries for 13 viruses. Our analysis indicates that previous research yielded very small number of BSA compounds. We posit that focused and coordinated efforts strategically targeting the discovery of such agents must be established and maintained going forward. The SMACC database publicly available at https://smacc.mml.unc.edu may serve as a reference for virologists and medicinal chemists working on the development of novel BSA agents in preparation for future viral outbreaks.
- Published
- 2022
20. Small molecule antiviral compound collection (SMACC): A comprehensive, highly curated database to support the discovery of broad-spectrum antiviral drug molecules
- Author
-
Holli-Joi Martin, Cleber C. Melo-Filho, Daniel Korn, Richard T. Eastman, Ganesha Rai, Anton Simeonov, Alexey V. Zakharov, Eugene Muratov, and Alexander Tropsha
- Subjects
Pharmacology ,Virology - Published
- 2023
- Full Text
- View/download PDF
21. Identification and Profiling of a Novel Diazaspiro[3.4]octane Chemical Series Active against Multiple Stages of the Human Malaria Parasite Plasmodium falciparum and Optimization Efforts
- Author
-
Theresa L. Coetzer, Lizette L. Koekemoer, Elizabeth A. Winzeler, Gregory S. Basarab, Richard T. Eastman, Jean Dam, Gurminder Kaur, André Horatscheck, Sonja B Lauterbach, Sabine Ottilie, Hui Guo, Michael J. Delves, Luisa Nardini, Jacek W. Zawada, Dennis A. Smith, James Duffy, Leslie J. Street, Tanya Paquet, Belinda C. Bezuidenhout, Claire Le Manach, Lyn-Marie Birkholtz, Dorjbal Dorjsuren, Liezl Gibhard, Dalu Mancama, Anton Simeonov, David A. Fidock, Christel Brunschwig, Sachel Mok, Daniel C. Talley, Nelius Venter, Mariëtte van der Watt, Grant A. Boyle, John G Woodland, Sergio Wittlin, Ayesha Aswat, Janette Reader, Nina Lawrence, Dale Taylor, Mathew Njoroge, Kelly Chibale, Tomas Yeo, Anjo Theron, Lutete Peguy Khonde, Erica Erlank, Thomas W. von Geldern, and Kathryn J. Wicht
- Subjects
Multiple stages ,0303 health sciences ,biology ,Plasmodium falciparum ,Computational biology ,biology.organism_classification ,medicine.disease ,01 natural sciences ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,High-Throughput Screening Assays ,Gametocyte ,medicine ,Molecular Medicine ,Structure–activity relationship ,Parasite hosting ,Malaria ,030304 developmental biology ,Octane - Abstract
A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (
- Published
- 2021
- Full Text
- View/download PDF
22. Establishment of human post-vaccination SARS-CoV-2 standard reference sera
- Author
-
Jinhua Xiang, Louis Katz, Patricia L. Winokur, Ashok Chaudhary, Rebecca Bradford, Sujatha Rashid, Sudakshina Ghosh, Angela Robertson, Joseph Menetski, Taylor Lee, Brittany Poelaert, Richard T. Eastman, Matthew D. Hall, and Jack T. Stapleton
- Abstract
As SARS-CoV-2 variants emerge, there is a critical need to understand the effectiveness of serum elicited by different SARS-CoV-2 vaccines. A reference reagent comprised of post-vaccination sera from recipients of different vaccines allows evaluation of in vitro variant neutralization, and provides a reference for comparing assay results across laboratories. We prepared and pooled >1 L serum from donors who received the SARS-CoV-2 mRNA vaccines (BNT162b2, Pfizer and mRNA-1273, Moderna), a replication-incompetent adenovirus type 26 vaccine (Ad26.COV2.S, Johnson and Johnson), or recombinant spike protein expressed by baculovirus incorporated into a nanoparticle vaccine plus Matrix-M adjuvant (NVX-CoV2373, Novavax). Twice frozen sera were aliquoted and are available for distribution to the research community (BEI Resources). The calculated WHO titer of pooled sera to spike protein was 1,312, 1,447, 1,936, and 587 and the reciprocal RBD binding to ACE-2 IC90-titers were 60, 64, 118, and 46 for BNT162b2, mRNA1273, Ad26.CoV2373, and NVX-CoV2373 sera, respectively.
- Published
- 2022
- Full Text
- View/download PDF
23. A humanized nanobody phage display library yields potent binders of SARS CoV-2 spike
- Author
-
Ying Fu, Juliana da Fonseca Rezende e Mello, Bryan D. Fleming, Alex Renn, Catherine Z. Chen, Xin Hu, Miao Xu, Kirill Gorshkov, Quinlin Hanson, Wei Zheng, Emily M. Lee, Lalith Perera, Robert Petrovich, Manisha Pradhan, Richard T. Eastman, Zina Itkin, Thomas B. Stanley, Allen Hsu, Venkata Dandey, Kedar Sharma, William Gillette, Troy Taylor, Nitya Ramakrishnan, Shelley Perkins, Dominic Esposito, Eunkeu Oh, Kimihiro Susumu, Mason Wolak, Marc Ferrer, Matthew D. Hall, Mario J. Borgnia, and Anton Simeonov
- Subjects
Mutation ,Multidisciplinary ,biology ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,Spike Protein ,Human airway ,Single-Domain Antibodies ,Antibodies, Viral ,medicine.disease_cause ,Antibodies, Neutralizing ,Virology ,Article ,Neutralization ,Spike Glycoprotein, Coronavirus ,medicine ,biology.protein ,Humans ,Bacteriophages ,Spike (software development) ,Antibody ,Ex vivo ,Protein Binding - Abstract
Neutralizing antibodies targeting the SARS-CoV-2 spike protein have shown a great preventative/therapeutic potential. Here, we report a rapid and efficient strategy for the development and design of SARS-CoV-2 neutralizing humanized nanobody constructs with sub-nanomolar affinities and nanomolar potencies. CryoEM-based structural analysis of the nanobodies in complex with spike revealed two distinct binding modes. The most potent nanobody, RBD-1-2G(NCATS-BL8125), tolerates the N501Y RBD mutation and remains capable of neutralizing the B.1.1.7 (Alpha) variant. Molecular dynamics simulations provide a structural basis for understanding the neutralization process of nanobodies exclusively focused on the spike-ACE2 interface with and without the N501Y mutation on RBD. A primary human airway air-lung interface (ALI) ex vivo model showed that RBD-1-2G-Fc antibody treatment was effective at reducing viral burden following WA1 and B.1.1.7 SARS-CoV-2 infections. Therefore, this presented strategy will serve as a tool to mitigate the threat of emerging SARS-CoV-2 variants. ONE-SENTENCE SUMMARY: A cost-effective, high-throughput, adaptable pipeline capable of identifying effective humanized nanobodies against SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
24. Regulation of Plasmodium yoelii Oocyst Development by Strain- and Stage-Specific Small-Subunit rRNA
- Author
-
Yanwei Qi, Feng Zhu, Richard T. Eastman, Young Fu, Martine Zilversmit, Sittiporn Pattaradilokrat, Lingxian Hong, Shengfa Liu, Thomas F. McCutchan, Weiqing Pan, Wenyue Xu, Jian Li, Fusheng Huang, and Xin-zhuan Su
- Subjects
Microbiology ,QR1-502 - Abstract
ABSTRACT One unique feature of malaria parasites is the differential transcription of structurally distinct rRNA (rRNA) genes at different developmental stages: the A-type genes are transcribed mainly in asexual stages, whereas the S-type genes are expressed mostly in sexual or mosquito stages. Conclusive functional evidence of different rRNAs in regulating stage-specific parasite development, however, is still absent. Here we performed genetic crosses of Plasmodium yoelii parasites with one parent having an oocyst development defect (ODD) phenotype and another producing normal oocysts to identify the gene(s) contributing to the ODD. The parent with ODD—characterized as having small oocysts and lacking infective sporozoites—was obtained after introduction of a plasmid with a green fluorescent protein gene into the parasite genome and subsequent passages in mice. Quantitative trait locus analysis of genome-wide microsatellite genotypes of 48 progeny from the crosses linked an ~200-kb segment on chromosome 6 containing one of the S-type genes (D-type small subunit rRNA gene [D-ssu]) to the ODD. Fine mapping of the plasmid integration site, gene expression pattern, and gene knockout experiments demonstrated that disruption of the D-ssu gene caused the ODD phenotype. Interestingly, introduction of the D-ssu gene into the same parasite strain (self), but not into a different subspecies, significantly affected or completely ablated oocyst development, suggesting a stage- and subspecies (strain)-specific regulation of oocyst development by D-ssu. This study demonstrates that P. yoelii D-ssu is essential for normal oocyst and sporozoite development and that variation in the D-ssu sequence can have dramatic effects on parasite development. IMPORTANCE Malaria parasites are the only known organisms that express structurally distinct rRNA genes at different developmental stages. The differential expression of these genes suggests that they play unique roles during the complex life cycle of the parasites. Conclusive functional proof of different rRNAs in regulating parasite development, however, is still absent or controversial. Here we functionally demonstrate for the first time that a stage-specifically expressed D-type small-subunit rRNA gene (D-ssu) is essential for oocyst development of the malaria parasite Plasmodium yoelii in the mosquito. This study also shows that variations in D-ssu sequence and/or the timing of transcription may have profound effects on parasite oocyst development. The results show that in addition to protein translation, rRNAs of malaria parasites also regulate parasite development and differentiation in a strain-specific manner, which can be explored for controlling parasite transmission.
- Published
- 2015
- Full Text
- View/download PDF
25. Thematic review series: Lipid Posttranslational Modifications. Fighting parasitic disease by blocking protein farnesylation
- Author
-
Richard T. Eastman, Frederick S. Buckner, Kohei Yokoyama, Michael H. Gelb, and Wesley C. Van Voorhis
- Subjects
antiprotozoal drugs ,Plasmodium ,Trypanosoma ,Toxoplasma ,Giardia ,Entamoeba ,Biochemistry ,QD415-436 - Abstract
Protein farnesylation is a form of posttranslational modification that occurs in most, if not all, eukaryotic cells. Inhibitors of protein farnesyltransferase (PFTIs) have been developed as anticancer chemotherapeutic agents. Using the knowledge gained from the development of PFTIs for the treatment of cancer, researchers are currently investigating the use of PFTIs for the treatment of eukaryotic pathogens. This “piggy-back” approach not only accelerates the development of a chemotherapeutic agent for protozoan pathogens but is also a means of mitigating the costs associated with de novo drug design. PFTIs have already been shown to be efficacious in the treatment of eukaryotic pathogens in animal models, including both Trypanosoma brucei, the causative agent of African sleeping sickness, and Plasmodium falciparum, one of the causative agents of malaria. Here, current evidence and progress are summarized that support the targeting of protein farnesyltransferase for the treatment of parasitic diseases.
- Published
- 2006
- Full Text
- View/download PDF
26. Deep learning identifies synergistic drug combinations for treating COVID-19
- Author
-
Tommi S. Jaakkola, Regina Barzilay, Richard T. Eastman, Zina Itkin, Wengong Jin, Jonathan M. Stokes, Alexey V. Zakharov, and James J. Collins
- Subjects
Drug ,drug synergy ,Coronavirus disease 2019 (COVID-19) ,Cell Survival ,Computer science ,media_common.quotation_subject ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Computational biology ,Antiviral Agents ,drug discovery ,Humans ,Drug Interactions ,media_common ,Pharmacology ,Alanine ,Multidisciplinary ,Training set ,Computer Sciences ,SARS-CoV-2 ,Drug discovery ,business.industry ,Deep learning ,deep learning ,Drug Synergism ,Biological Sciences ,Adenosine Monophosphate ,COVID-19 Drug Treatment ,Drug Combinations ,Synergy ,Reduced toxicity ,Physical Sciences ,Artificial intelligence ,business - Abstract
Significance COVID-19 has caused more than 2.5 million deaths worldwide. It is imperative that we develop therapies that can mitigate the effect of the disease. While searching for individual drugs for this purpose has been met with difficulties, synergistic drug combinations offer a promising alternative. However, the lack of high-quality training data pertaining to drug combinations makes it challenging to use existing machine learning methods for effective novel combination prediction tasks. Our proposed approach addresses this challenge by leveraging additional readily available data, such as drug−target interactions, thus enabling an effective in silico search for synergistic combinations against SARS-CoV-2., Effective treatments for COVID-19 are urgently needed. However, discovering single-agent therapies with activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been challenging. Combination therapies play an important role in antiviral therapies, due to their improved efficacy and reduced toxicity. Recent approaches have applied deep learning to identify synergistic drug combinations for diseases with vast preexisting datasets, but these are not applicable to new diseases with limited combination data, such as COVID-19. Given that drug synergy often occurs through inhibition of discrete biological targets, here we propose a neural network architecture that jointly learns drug−target interaction and drug−drug synergy. The model consists of two parts: a drug−target interaction module and a target−disease association module. This design enables the model to utilize drug−target interaction data and single-agent antiviral activity data, in addition to available drug−drug combination datasets, which may be small in nature. By incorporating additional biological information, our model performs significantly better in synergy prediction accuracy than previous methods with limited drug combination training data. We empirically validated our model predictions and discovered two drug combinations, remdesivir and reserpine as well as remdesivir and IQ-1S, which display strong antiviral SARS-CoV-2 synergy in vitro. Our approach, which was applied here to address the urgent threat of COVID-19, can be readily extended to other diseases for which a dearth of chemical−chemical combination data exists.
- Published
- 2021
- Full Text
- View/download PDF
27. Identification and Profiling of a Novel Diazaspiro[3.4]octane Chemical Series Active against Multiple Stages of the Human Malaria Parasite
- Author
-
Claire, Le Manach, Jean, Dam, John G, Woodland, Gurminder, Kaur, Lutete P, Khonde, Christel, Brunschwig, Mathew, Njoroge, Kathryn J, Wicht, André, Horatscheck, Tanya, Paquet, Grant A, Boyle, Liezl, Gibhard, Dale, Taylor, Nina, Lawrence, Tomas, Yeo, Sachel, Mok, Richard T, Eastman, Dorjbal, Dorjsuren, Daniel C, Talley, Hui, Guo, Anton, Simeonov, Janette, Reader, Mariëtte, van der Watt, Erica, Erlank, Nelius, Venter, Jacek W, Zawada, Ayesha, Aswat, Luisa, Nardini, Theresa L, Coetzer, Sonja B, Lauterbach, Belinda C, Bezuidenhout, Anjo, Theron, Dalu, Mancama, Lizette L, Koekemoer, Lyn-Marie, Birkholtz, Sergio, Wittlin, Michael, Delves, Sabine, Ottilie, Elizabeth A, Winzeler, Thomas W, von Geldern, Dennis, Smith, David A, Fidock, Leslie J, Street, Gregory S, Basarab, James, Duffy, and Kelly, Chibale
- Subjects
Male ,Molecular Structure ,Plasmodium falciparum ,High-Throughput Screening Assays ,Rats ,Antimalarials ,Mice ,Structure-Activity Relationship ,Germ Cells ,Parasitic Sensitivity Tests ,Anopheles ,Microsomes, Liver ,Animals ,Humans ,Female ,Spiro Compounds - Abstract
A novel diazaspiro[3.4]octane series was identified from a
- Published
- 2021
28. Synergistic and Antagonistic Drug Combinations against SARS-CoV-2
- Author
-
Wei Zheng, Paul Shinn, Lu Chen, Matthew D. Hall, Catherine Z. Chen, Richard T. Eastman, Anton Simeonov, Sam Michael, Hui Guo, Eugene N. Muratov, Zina Itkin, Tesia Bobrowski, and Alexey V. Zakharov
- Subjects
Drug ,Combination therapy ,medicine.drug_class ,media_common.quotation_subject ,Amodiaquine ,Pharmacology ,Antiviral Agents ,Article ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Genetics ,Humans ,Medicine ,Molecular Biology ,030304 developmental biology ,media_common ,0303 health sciences ,Alanine ,SARS-CoV-2 ,business.industry ,Drug Synergism ,Nitazoxanide ,Umifenovir ,Adenosine Monophosphate ,COVID-19 Drug Treatment ,Drug Combinations ,Drug repositioning ,Synergy ,030220 oncology & carcinogenesis ,Molecular Medicine ,Antiviral drug ,business ,Hydroxychloroquine ,medicine.drug - Abstract
COVID-19 is undoubtedly the most impactful viral disease of the current century, afflicting millions worldwide. As yet, there is not an approved vaccine, as well as limited options from existing drugs for treating this disease. We hypothesized that combining drugs with independent mechanisms of action could result in synergy against SARS-CoV-2. Using in silico approaches, we prioritized 73 combinations of 32 drugs with potential activity against SARS-CoV-2 and then tested them in vitro . Overall, we identified 16 synergistic and 8 antagonistic combinations, 4 of which were both synergistic and antagonistic in a dose-dependent manner. Among the 16 synergistic cases, combinations of nitazoxanide with three other compounds (remdesivir, amodiaquine and umifenovir) were the most notable, all exhibiting significant synergy against SARS-CoV-2. The combination of nitazoxanide, an FDA-approved drug, and remdesivir, FDA emergency use authorization for the treatment of COVID-19, demonstrate a strong synergistic interaction. Notably, the combination of remdesivir and hydroxychloroquine demonstrated strong antagonism. Overall, our results emphasize the importance of both drug repurposing and preclinical testing of drug combinations for potential therapeutic use against SARS-CoV-2 infections.
- Published
- 2021
- Full Text
- View/download PDF
29. Drug Repurposing Screen for Compounds Inhibiting the Cytopathic Effect of SARS-CoV-2
- Author
-
Hui Guo, Brianna M. Brooks, Paul Shinn, Zina Itkin, Kelli M. Wilson, Catherine Z. Chen, Richard T. Eastman, Wei Zheng, Donald C. Lo, Min Shen, Robert Bostwick, Ruili Huang, Tongan Zhao, Matthew D. Hall, Carleen Klump-Thomas, Lynn Rasmussen, Xin Hu, Samuel G. Michael, and Anton Simeonov
- Subjects
Drug ,media_common.quotation_subject ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,drug repurposing and discovery ,Pharmacology ,medicine.disease_cause ,Approved drug ,cytopathic effect ,medicine ,Pharmacology (medical) ,skin and connective tissue diseases ,media_common ,Cytopathic effect ,Original Research ,Ebola virus ,business.industry ,SARS-CoV-2 ,lcsh:RM1-950 ,fungi ,virus diseases ,COVID-19 ,Piperacetazine ,Methotrimeprazine ,Clinical trial ,body regions ,Drug repositioning ,lcsh:Therapeutics. Pharmacology ,HTS ,business ,medicine.drug - Abstract
Drug repurposing is a rapid approach to identifying therapeutics for the treatment of emerging infectious diseases such as COVID-19. To address the urgent need for treatment options, we carried out a quantitative high-throughput screen using a SARS-CoV-2 cytopathic assay with a compound collection of 8,810 approved and investigational drugs, mechanism-based bioactive compounds, and natural products. Three hundred and nineteen compounds with anti-SARS-CoV-2 activities were identified and confirmed, including 91 approved drug and 49 investigational drugs. Among these confirmed compounds, the anti-SARS-CoV-2 activities of 230 compounds, including 38 approved drugs, have not been previously reported. Chlorprothixene, methotrimeprazine, and piperacetazine were the three most potent FDA approved drugs with anti-SARS-CoV-2 activities. These three compounds have not been previously reported to have anti-SARS-CoV-2 activities, although their antiviral activities against SARS-CoV and Ebola virus have been reported. These results demonstrate that this comprehensive data set of drug repurposing screen for SARS-CoV-2 is useful for drug repurposing efforts including design of new drug combinations for clinical trials.
- Published
- 2021
30. Modulation of Triple Artemisinin-Based Combination Therapy Pharmacodynamics by Plasmodium falciparum Genotype
- Author
-
Chanaki Amaratunga, Seila Suon, Anton Simeonov, Lu Chen, Rob W. van der Pluijm, Tyler Peryea, Gergely Zahoranszky-Kohalmi, Juliana M. Sá, Charlotte V. Hobbs, Zina Itkin, Richard T. Eastman, Megan R. Ansbro, Thomas E. Wellems, Olivo Miotto, Arjen M. Dondorp, Intensive Care Medicine, Graduate School, AII - Infectious diseases, APH - Aging & Later Life, APH - Global Health, and APH - Quality of Care
- Subjects
Drug ,Plasmodium ,Combination therapy ,media_common.quotation_subject ,malaria ,Drug resistance ,Pharmacology ,Piperaquine ,parasitic diseases ,medicine ,pharmacodynamics ,Pharmacology (medical) ,Artemisinin ,media_common ,pharmacogenetics ,Pyronaridine ,antimalarial ,biology ,Plasmodium falciparum ,biology.organism_classification ,medicine.disease ,Pharmacodynamics ,Malaria ,triple-drug combination ,medicine.drug - Abstract
The first-line treatments for uncomplicated Plasmodium falciparum malaria are artemisinin-based combination therapies (ACTs), consisting of an artemisinin derivative combined with a longer acting partner drug. However, the spread of P. falciparum with decreased susceptibility to artemisinin and partner drugs presents a significant challenge to malaria control efforts. To stem the spread of drug resistant parasites, novel chemotherapeutic strategies are being evaluated, including the implementation of triple artemisinin-based combination therapies (TACTs). Currently, there is limited knowledge on the pharmacodynamics and pharmacogenetic interactions of proposed TACT drug combinations. To evaluate these interactions, we established an in vitro high-throughput process for measuring the drug dose-response to three distinct antimalarial drugs present in a TACT. Sixteen different TACT combinations were screened against fifteen parasite lines from Cambodia, with a focus on parasites with differential susceptibilities to piperaquine and artemisinins. Analysis revealed drug-drug interactions unique to specific genetic backgrounds, including antagonism between piperaquine and pyronaridine associated with gene amplification of plasmepsin II/III, two aspartic proteases that localize to the parasite digestive vacuole. From this initial study, we identified parasite genotypes with decreased susceptibility to specific TACTs, as well as potential TACTs that display antagonism in a genotype-dependent manner. Our assay and analysis platform can be further leveraged to inform drug implementation decisions and evaluate next-generation TACTs.One Sentence SummaryIn vitro process to evaluate triple-drug combinations for prioritizing antimalarial combinations for in vivo evaluation.
- Published
- 2020
31. Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors
- Author
-
Avindra Nath, Alexey V. Zakharov, Anuradha Balasubramanian, Tadahisa Teramoto, Rachel P. M. Abrams, Adam Yasgar, Ajit Jadhav, Joseph P. Steiner, Kyle R. Brimacombe, Radhakrishnan Padmanabhan, Muzna Bachani, Richard T. Eastman, Dorjbal Dorjsuren, Myoung-Hwa Lee, Anton Simeonov, Wenxue Li, Matthew D. Hall, and Nasir Malik
- Subjects
Drug ,Tetracycline ,medicine.drug_class ,media_common.quotation_subject ,medicine.medical_treatment ,viruses ,Antibiotics ,Drug Evaluation, Preclinical ,Quantitative Structure-Activity Relationship ,Antiviral Agents ,Zika virus ,Small Molecule Libraries ,Inhibitory Concentration 50 ,Artificial Intelligence ,Chlorocebus aethiops ,medicine ,Animals ,Protease Inhibitors ,Vero Cells ,Methacycline ,media_common ,Serine protease ,Multidisciplinary ,Protease ,biology ,business.industry ,Zika Virus Infection ,Zika Virus ,Biological Sciences ,medicine.disease ,biology.organism_classification ,Virology ,High-Throughput Screening Assays ,Mice, Inbred C57BL ,Flavivirus ,Disease Models, Animal ,biology.protein ,business ,Immunocompetence ,Encephalitis ,medicine.drug - Abstract
When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.
- Published
- 2020
32. 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
33. Targeting ACE2-RBD Interaction as a Platform for COVID-19 Therapeutics: Development and Drug-Repurposing Screen of an AlphaLISA Proximity Assay
- Author
-
Paul Shinn, Min Shen, Zina Itkin, Richard T. Eastman, Matthew D. Hall, Quinlin M Hanson, and Kelli M. Wilson
- Subjects
Pharmacology ,assay development ,Coronavirus disease 2019 (COVID-19) ,drug repurposing ,business.industry ,COVID19 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,High-throughput screening ,Spike Protein ,ACE2 ,Computational biology ,Small molecule ,high-throughput screening ,Article ,Drug repositioning ,Host cell invasion ,Medicine ,Pharmacology (medical) ,business ,Rapid response ,AlphaLISA - Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, is a pressing public health emergency garnering a rapid response from scientists across the globe. Host cell invasion is initiated through direct binding of the viral spike protein to the host receptor angiotensin-converting enzyme 2 (ACE2). Disrupting the spike protein-ACE2 interaction is a potential therapeutic target for treating COVID-19. We have developed a proximity-based AlphaLISA assay to measure the binding of SARS-CoV-2 spike protein receptor binding domain (RBD) to ACE2. Utilizing this assay platform, a drug-repurposing screen against 3384 small-molecule drugs and preclinical compounds was carried out, yielding 25 high-quality primary hits, of which only corilagin was validated in cherry-picking. This established AlphaLISA RBD-ACE2 platform can facilitate evaluation of biologics or small molecules that can perturb this essential viral-host interaction to further the development of interventions to address the global health pandemic.
- Published
- 2020
34. Modulation of Triple Artemisinin-Based Combination Therapy Pharmacodynamics by
- Author
-
Megan R, Ansbro, Zina, Itkin, Lu, Chen, Gergely, Zahoranszky-Kohalmi, Chanaki, Amaratunga, Olivo, Miotto, Tyler, Peryea, Charlotte V, Hobbs, Seila, Suon, Juliana M, Sá, Arjen M, Dondorp, Rob W, van der Pluijm, Thomas E, Wellems, Anton, Simeonov, and Richard T, Eastman
- Subjects
parasitic diseases - Abstract
[Image: see text] The first-line treatments for uncomplicated Plasmodium falciparum malaria are artemisinin-based combination therapies (ACTs), consisting of an artemisinin derivative combined with a longer acting partner drug. However, the spread of P. falciparum with decreased susceptibility to artemisinin and partner drugs presents a significant challenge to malaria control efforts. To stem the spread of drug resistant parasites, novel chemotherapeutic strategies are being evaluated, including the implementation of triple artemisinin-based combination therapies (TACTs). Currently, there is limited knowledge on the pharmacodynamic and pharmacogenetic interactions of proposed TACT drug combinations. To evaluate these interactions, we established an in vitro high-throughput process for measuring the drug concentration–response to three distinct antimalarial drugs present in a TACT. Sixteen different TACT combinations were screened against 15 parasite lines from Cambodia, with a focus on parasites with differential susceptibilities to piperaquine and artemisinins. Analysis revealed drug–drug interactions unique to specific genetic backgrounds, including antagonism between piperaquine and pyronaridine associated with gene amplification of plasmepsin II/III, two aspartic proteases that localize to the parasite digestive vacuole. From this initial study, we identified parasite genotypes with decreased susceptibility to specific TACTs, as well as potential TACTs that display antagonism in a genotype-dependent manner. Our assay and analysis platform can be further leveraged to inform drug implementation decisions and evaluate next-generation TACTs.
- Published
- 2020
35. Discovery of Synergistic and Antagonistic Drug Combinations against SARS-CoV-2 In Vitro
- Author
-
Hui Guo, Zina Itkin, Alexey V. Zakharov, Anton Simeonov, Tesia Bobrowski, Catherine Chen, Sam Michael, Wei Zheng, Richard T. Eastman, Eugene N. Muratov, Lu Chen, Matthew D. Hall, and Paul Shinn
- Subjects
Drug ,drug combinations ,Emergency Use Authorization ,drug synergy ,in silico design ,media_common.quotation_subject ,In silico ,Amodiaquine ,Pharmacology ,combination therapy ,medicine ,CPE assay ,knowledge mining ,media_common ,drug repurposing ,SARS-CoV-2 ,business.industry ,COVID-19 ,Nitazoxanide ,Hydroxychloroquine ,Umifenovir ,nitazoxanide remdesivir combo ,Drug repositioning ,Original Article ,business ,medicine.drug - Abstract
Antiviral drug development for coronavirus disease 2019 (COVID-19) is occurring at an unprecedented pace, yet there are still limited therapeutic options for treating this disease. We hypothesized that combining drugs with independent mechanisms of action could result in synergy against SARS-CoV-2, thus generating better antiviral efficacy. Using in silico approaches, we prioritized 73 combinations of 32 drugs with potential activity against SARS-CoV-2 and then tested them in vitro. Sixteen synergistic and eight antagonistic combinations were identified; among 16 synergistic cases, combinations of the US Food and Drug Administration (FDA)-approved drug nitazoxanide with remdesivir, amodiaquine, or umifenovir were most notable, all exhibiting significant synergy against SARS-CoV-2 in a cell model. However, the combination of remdesivir and lysosomotropic drugs, such as hydroxychloroquine, demonstrated strong antagonism. Overall, these results highlight the utility of drug repurposing and preclinical testing of drug combinations for discovering potential therapies to treat COVID-19., Graphical Abstract, Sixteen synergistic drug combinations against SARS-CoV-2 were identified in silico and confirmed experimentally. Combinations of nitazoxanide with remdesivir, amodiaquine, or umifenovir exhibited significant synergy against SARS-CoV-2. Remdesivir combined with hydroxychloroquine or other lysosomotropic drugs resulted in strong antagonism.
- Published
- 2020
- Full Text
- View/download PDF
36. Targeting ACE2-RBD interaction as a platform for COVID19 therapeutics: Development and drug repurposing screen of an AlphaLISA proximity assay
- Author
-
Matthew D. Hall, Kelli M. Wilson, Zina Itkin, Paul Shinn, Richard T. Eastman, Quinlin M Hanson, and Min Shen
- Subjects
Drug repositioning ,Coronavirus disease 2019 (COVID-19) ,Host cell invasion ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Direct binding ,Spike Protein ,Computational biology ,Biology ,Small molecule ,Rapid response - Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, is a pressing public health emergency garnering rapid response from scientists across the globe. Host cell invasion is initiated through direct binding of the viral spike protein to the host receptor angiotensin-converting enzyme 2 (ACE2). Disrupting the spike-ACE2 interaction is a potential therapeutic target for treating COVID-19. We have developed a proximity-based AlphaLISA assay to measure binding of SARS-CoV-2 spike protein Receptor Binding Domain (RBD) to ACE2. Utilizing this assay platform, a drug-repurposing screen against 3,384 small molecule drugs and pre-clinical compounds was performed, yielding 25 high-quality, small-molecule hits that can be evaluated in cell-based models. This established AlphaLISA RBD-ACE2 platform can facilitate evaluation of biologics or small molecules that can perturb this essential viral-host interaction to further the development of interventions to address the global health pandemic.
- Published
- 2020
- Full Text
- View/download PDF
37. Correction to Remdesivir: A Review of Its Discovery and Development Leading to Human Clinical Trials for Treatment of COVID-19
- Author
-
Jacob S. Roth, Anton Simeonov, Richard T. Eastman, Matthew Hall, Samarjit Patnaik, Kyle R. Brimacombe, and Min Shen
- Subjects
medicine.medical_specialty ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,General Chemical Engineering ,MEDLINE ,General Chemistry ,Addition/Correction ,Clinical trial ,Chemistry ,medicine ,Intensive care medicine ,business ,QD1-999 - Abstract
The global pandemic of SARS-CoV-2, the causative viral pathogen of COVID-19, has driven the biomedical community to action-to uncover and develop antiviral interventions. One potential therapeutic approach currently being evaluated in numerous clinical trials is the agent remdesivir, which has endured a long and winding developmental path. Remdesivir is a nucleotide analogue prodrug that perturbs viral replication, originally evaluated in clinical trials to thwart the Ebola outbreak in 2014. Subsequent evaluation by numerous virology laboratories demonstrated the ability of remdesivir to inhibit coronavirus replication, including SARS-CoV-2. Here, we provide an overview of remdesivir's discovery, mechanism of action, and the current studies exploring its clinical effectiveness.
- Published
- 2020
38. An OpenData portal to share COVID-19 drug repurposing data in real time
- Author
-
Manisha Pradhan, Amit Viraktamath, Stephen C. Kales, Alexey V. Zakharov, Alex Renn, Tongan Zhao, Miao Xu, Bolormaa Baljinnyam, Catherine Z. Chen, Ke Wang, Sam Michael, Mark Backus, Zina Itkin, Donald C. Lo, Anton Simeonov, Kyle R. Brimacombe, Tara Eicher, Quinlin M Hanson, Carleen Klumpp-Thomas, Wei Zhu, Ying Fu, Tim Mierzwa, Kirill Gorshkov, Min Shen, Emily M. Lee, Xin Hu, Wei Zheng, Lu Chen, Ewy Mathé, Matthew D. Hall, Paul Shinn, Kelli M. Wilson, Richard T. Eastman, Andrew Patt, Hui Guo, Jonathan H. Shrimp, and Marc Ferrer
- Subjects
World Wide Web ,Drug repositioning ,Open data ,Coronavirus disease 2019 (COVID-19) ,Computer science ,Small Molecule Libraries ,Translational science ,Approved drug ,Article ,Repurposing - Abstract
The National Center for Advancing Translational Sciences (NCATS) has developed an online open science data portal for its COVID-19 drug repurposing campaign – named OpenData – with the goal of making data across a range of SARS-CoV-2 related assays available in real-time. The assays developed cover a wide spectrum of the SARS-CoV-2 life cycle, including both viral and human (host) targets. In total, over 10,000 compounds are being tested in full concentration-response ranges from across multiple annotated small molecule libraries, including approved drug, repurposing candidates and experimental therapeutics designed to modulate a wide range of cellular targets. The goal is to support research scientists, clinical investigators and public health officials through open data sharing and analysis tools to expedite the development of SARS-CoV-2 interventions, and to prioritize promising compounds and repurposed drugs for further development in treating COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
39. Discovery of Synergistic and Antagonistic Drug Combinations Against SARS-CoV-2 In Vitro
- Author
-
Tesia Bobrowski, Lu Chen, Richard T. Eastman, Zina Itkin, Paul Shinn, Catherine Z. Chen, Hui Guo, Wei Zheng, Sam Michael, Anton Simeonov, Matthew Hall, Alexey V. Zakharov, and Eugene Muratov
- Published
- 2020
- Full Text
- View/download PDF
40. Canvass: A Crowd-Sourced, Natural-Product Screening Library for Exploring Biological Space
- Author
-
Rajarshi Guha, Anton Simeonov, Jesus Moreno, Min Shen, Yi Tang, James Inglese, Jared T. Shaw, Jimmy Ming-Tai Wu, Samir A. Ross, Tobie D. Lee, Surendra Karavadhi, Dorian M. Cheff, Susruta Majumdar, Adam Yasgar, Richard E. Taylor, Sam Michael, Zhengren Xu, Wei Sun, Lucas A. Morrill, David A. Vosburg, Wei Zheng, Corey R. J. Stephenson, Noah Z. Burns, Michael J. Iannotti, Carleen Klumpp-Thomas, Richard T. Eastman, Ohyun Kwon, Armen Zakarian, Yufeng Zhang, Matthew D. Hall, Sara E. Kearney, Ya Qin Zhang, Tongan Zhao, Ken Cheng, Indrajeet Sharma, Nathan P. Coussens, Ruili Huang, Derek S. Tan, Paul Shinn, Michael J. Krische, Caitlin Lynch, Jon Clardy, Larry E. Overman, Kanny K. Wan, Chambers C. Hughes, Madeleine M. Joullié, Thomas J. Maimone, Matthew B. Boxer, Jason A. Clement, A. Douglas Kinghorn, Peter A. Crooks, Brian C. Goess, Robert B. Susick, Gergely Zahoránszky-Kőhalmi, Alyssa L. Verano, Danielle Bougie, Jacob S. Roth, Ben Shen, Dorjbal Dorjsuren, Elias Picazo, Jinghua Zhao, John K. Snyder, Rodrigo B. Andrade, Wei Shi, Jin K. Cha, Brian T. Murphy, Fatima Rivas, William M. Wuest, Jake Ganor, Robert B. Grossman, Pavel Nagorny, Emily Mevers, Enas I. Mohamed, David E. Olson, John A. Porco, Neil K. Garg, Srilatha Sakamuru, Jason M. Rohde, M. Kevin Brown, Menghang Xia, Tianjing Ren, Steve Titus, Christopher Dillon, Mark J. Henderson, Zhong Zuo, Regan J. Thomson, David G. I. Kingston, Lauren E. Brown, Jeffrey N. Johnston, Katherine N. Maloney, Richmond Sarpong, Zina Itkin, Gregory D. Cuny, Ajit Jadhav, and Kyle R. Brimacombe
- Subjects
0301 basic medicine ,Natural product ,General Chemical Engineering ,General Chemistry ,Biological potential ,Space (commercial competition) ,Data science ,Chemical library ,Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Workflow ,chemistry ,Identification (biology) ,Cluster analysis ,QD1-999 ,Limited resources ,Research Article - Abstract
Natural products and their derivatives continue to be wellsprings of nascent therapeutic potential. However, many laboratories have limited resources for biological evaluation, leaving their previously isolated or synthesized compounds largely or completely untested. To address this issue, the Canvass library of natural products was assembled, in collaboration with academic and industry researchers, for quantitative high-throughput screening (qHTS) across a diverse set of cell-based and biochemical assays. Characterization of the library in terms of physicochemical properties, structural diversity, and similarity to compounds in publicly available libraries indicates that the Canvass library contains many structural elements in common with approved drugs. The assay data generated were analyzed using a variety of quality control metrics, and the resultant assay profiles were explored using statistical methods, such as clustering and compound promiscuity analyses. Individual compounds were then sorted by structural class and activity profiles. Differential behavior based on these classifications, as well as noteworthy activities, are outlined herein. One such highlight is the activity of (−)-2(S)-cathafoline, which was found to stabilize calcium levels in the endoplasmic reticulum. The workflow described here illustrates a pilot effort to broadly survey the biological potential of natural products by utilizing the power of automation and high-throughput screening., Canvass, a crowd-sourced library of natural products, was evaluated through HTS in a spectrum of disease-relevant assays to survey the broad biological potential of natural products in drug discovery.
- Published
- 2018
41. Artemisinin resistance phenotypes and K13 inheritance in a Plasmodium falciparum cross and Aotus model
- Author
-
Chanaki Amaratunga, John H. Adams, Carole A. Long, Kenneth O. Udenze, Sarah R. Kaslow, Juliana M. Sá, Rebecca E. Salzman, Robert W. Gwadz, Paul K. J. Han, Kazutoyo Miura, J. Patrick Mershon, Jianbing Mu, Rick M. Fairhurst, Christine E. Figan, Richard T. Eastman, Marvin L. Thomas, Xin-zhuan Su, Kimberly F Breglio, Ramoncito L. Caleon, Suzanne Li, Ababacar Diouf, Sumana Chakravarty, Nina F. Gnädig, Min Zhang, Eric R. James, Stacy M. Ricklefs, Michael P. Fay, Viviana A. Melendez-Muniz, Stephen L. Hoffman, Whitney A. Kite, David A. Fidock, Bingbing Deng, Roberto R. Moraes Barros, Gregory Tullo, Anna Liu, B. Kim Lee Sim, Daniel E. Sturdevant, Thomas E. Wellems, Tyler J. Gibson, Michael Krause, Rifat S. Rahman, Soundarapandian Velmurugan, Erika P. Nishiguchi, Stephen F. Porcella, Judith Straimer, and Theresa Engels
- Subjects
0301 basic medicine ,Multidisciplinary ,Combination therapy ,medicine.medical_treatment ,Dihydroartemisinin ,Plasmodium falciparum ,Biology ,biology.organism_classification ,medicine.disease ,Virology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Artesunate ,medicine ,Parasite hosting ,Artemisinin ,Genotyping ,Malaria ,medicine.drug - Abstract
Concerns about malaria parasite resistance to treatment with artemisinin drugs (ARTs) have grown with findings of prolonged parasite clearance t1/2s (>5 h) and their association with mutations in Plasmodium falciparum Kelch-propeller protein K13. Here, we describe a P. falciparum laboratory cross of K13 C580Y mutant with C580 wild-type parasites to investigate ART response phenotypes in vitro and in vivo. After genotyping >400 isolated progeny, we evaluated 20 recombinants in vitro: IC50 measurements of dihydroartemisinin were at similar low nanomolar levels for C580Y- and C580-type progeny (mean ratio, 1.00; 95% CI, 0.62–1.61), whereas, in a ring-stage survival assay, the C580Y-type progeny had 19.6-fold (95% CI, 9.76–39.2) higher average counts. In splenectomized Aotus monkeys treated with three daily doses of i.v. artesunate, t1/2 calculations by three different methods yielded mean differences of 0.01 h (95% CI, −3.66 to 3.67), 0.80 h (95% CI, −0.92 to 2.53), and 2.07 h (95% CI, 0.77–3.36) between C580Y and C580 infections. Incidences of recrudescence were 57% in C580Y (4 of 7) versus 70% in C580 (7 of 10) infections (−13% difference; 95% CI, −58% to 35%). Allelic substitution of C580 in a C580Y-containing progeny clone (76H10) yielded a transformant (76H10C580Rev) that, in an infected monkey, recrudesced regularly 13 times over 500 d. Frequent recrudescences of ART-treated P. falciparum infections occur with or without K13 mutations and emphasize the need for improved partner drugs to effectively eliminate the parasites that persist through the ART component of combination therapy.
- Published
- 2018
- Full Text
- View/download PDF
42. A single nucleotide polymorphism in the Plasmodium falciparum atg18 gene associates with artemisinin resistance and confers enhanced parasite survival under nutrient deprivation
- Author
-
Roberto Amato, Rajarshi Guha, David W. Dorward, Craig J. Thomas, Rick M. Fairhurst, Kimberly F Breglio, Juliana M. Sá, Crystal McKnight, Sundar Ganesan, Carleen Klumpp-Thomas, Anna Katharina Simon, Richard T. Eastman, Pharath Lim, and David L. Roberts
- Subjects
0301 basic medicine ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Plasmodium falciparum ,030106 microbiology ,Mutant ,Protozoan Proteins ,Autophagy-Related Proteins ,Single-nucleotide polymorphism ,Drug resistance ,Polymorphism, Single Nucleotide ,lcsh:Infectious and parasitic diseases ,Antimalarials ,03 medical and health sciences ,Fitness ,medicine ,Autophagy ,Parasite hosting ,SNP ,lcsh:RC109-216 ,Amino Acid Sequence ,Artemisinin ,Gene ,Genetics ,biology ,Research ,atg18 ,biology.organism_classification ,Artemisinins ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Artemisinin resistance ,Cardiovascular and Metabolic Diseases ,Parasitology ,Sequence Alignment ,medicine.drug - Abstract
Background Artemisinin-resistant Plasmodium falciparum has been reported throughout the Greater Mekong subregion and threatens to disrupt current malaria control efforts worldwide. Polymorphisms in kelch13 have been associated with clinical and in vitro resistance phenotypes; however, several studies suggest that the genetic determinants of resistance may involve multiple genes. Current proposed mechanisms of resistance conferred by polymorphisms in kelch13 hint at a connection to an autophagy-like pathway in P. falciparum. Results A SNP in autophagy-related gene 18 (atg18) was associated with long parasite clearance half-life in patients following artemisinin-based combination therapy. This gene encodes PfAtg18, which is shown to be similar to the mammalian/yeast homologue WIPI/Atg18 in terms of structure, binding abilities, and ability to form puncta in response to stress. To investigate the contribution of this polymorphism, the atg18 gene was edited using CRISPR/Cas9 to introduce a T38I mutation into a k13-edited Dd2 parasite. The presence of this SNP confers a fitness advantage by enabling parasites to grow faster in nutrient-limited settings. The mutant and parent parasites were screened against drug libraries of 6349 unique compounds. While the SNP did not modulate the parasite’s susceptibility to any of the anti-malarial compounds using a 72-h drug pulse, it did alter the parasite’s susceptibility to 227 other compounds. Conclusions These results suggest that the atg18 T38I polymorphism may provide additional resistance against artemisinin derivatives, but not partner drugs, even in the absence of kelch13 mutations, and may also be important in parasite survival during nutrient deprivation. Electronic supplementary material The online version of this article (10.1186/s12936-018-2532-x) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
43. New WS9326A Derivatives and One New Annimycin Derivative with Antimalarial Activity are Produced by Streptomyces asterosporus DSM 41452 and Its Mutant
- Author
-
Thomas Paululat, Songya Zhang, Claudia Jessen-Trefzer, Jing Zhu, Richard T. Eastman, David L. Zechel, and Andreas Bechthold
- Subjects
Streptomyces asterosporus ,Stereochemistry ,Mutant ,Molecular Conformation ,Mutagenesis (molecular biology technique) ,Biology ,010402 general chemistry ,Peptides, Cyclic ,01 natural sciences ,Biochemistry ,Article ,Antimalarials ,Lactones ,Polyketide ,Gene cluster ,Insertion ,Molecular Biology ,010405 organic chemistry ,Organic Chemistry ,Cytochrome P450 ,Monooxygenase ,Amides ,Streptomyces ,0104 chemical sciences ,Mutation ,Fatty Acids, Unsaturated ,biology.protein ,Molecular Medicine - Abstract
In this study, we report that Streptomyces asterosporus DSM 41452 is a producer of new molecules related to the nonribosomal cyclodepsipeptide WS9326A and the polyketide annimycin. S. asterosporus DSM 41452 is shown to produce six cyclodepsipeptides and peptides, WS9326A to G. Notably, the compounds WS9326F and WS9326G have not been described before. The genome of S. asterosporus DSM 41452 was sequenced, and a putative WS9326A gene cluster was identified. Gene-deletion experiments confirmed that this cluster was responsible for the biosynthesis of WS9326A to G. Additionally, a gene-deletion experiment demonstrated that sas16 encoding a cytochrome P450 monooxygenase was involved in the synthesis of the novel (E)-2,3-dehydrotyrosine residue found in WS9326A and its derivatives. An insertion mutation within the putative annimycin gene cluster led to the production of a new annimycin derivative, annimycin B, which exhibited modest inhibitory activity against Plasmodium falciparum.
- Published
- 2017
- Full Text
- View/download PDF
44. Application of niclosamide and analogs as small molecule inhibitors of Zika virus and SARS-CoV-2 infection
- Author
-
Zina Itkin, Paul Shinn, Wei Zheng, Miao Xu, Catherine Z. Chen, Carleen Klumpp-Thomas, Sam Michael, Guo Li Ming, Pranav Shah, Hui Guo, Wenwei Huang, Lu Chen, Min Shen, Emily M. Lee, Xin Hu, Xiao Lu, Richard T. Eastman, Ruili Huang, Donald C. Lo, Hengli Tang, Xin Xu, Anton Simeonov, Khalida Shamim, and Hongjun Song
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Viral Nonstructural Proteins ,Pharmacology ,ZIKV, Zika virus ,PAMPA, parallel artificial membrane permeability assay ,01 natural sciences ,Biochemistry ,Zika virus ,NS-1 assay ,chemistry.chemical_compound ,Drug Stability ,Chlorocebus aethiops ,Drug Discovery ,MOI, multiplicity of infection ,Niclosamide ,media_common ,Molecular Structure ,biology ,Serine Endopeptidases ,Small molecule ,Molecular Docking Simulation ,ADME, absorption ,Salicylanilide ,Drug repositioning ,Flavivirus ,distribution, metabolism and excretion ,Microsomes, Liver ,Molecular Medicine ,Protein Binding ,medicine.drug ,Drug ,media_common.quotation_subject ,small molecule ,Microbial Sensitivity Tests ,Antiviral Agents ,SAR, structure-activity relationship ,Article ,Structure-Activity Relationship ,Viral Proteins ,medicine ,Animals ,Humans ,Structure–activity relationship ,Vero Cells ,Molecular Biology ,ComputingMethodologies_COMPUTERGRAPHICS ,Binding Sites ,SARS-CoV-2 ,010405 organic chemistry ,Organic Chemistry ,biology.organism_classification ,Rats ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry ,RLM, rat liver microsomal stability ,salicylanilide - Abstract
Graphical abstract, Zika virus has emerged as a potential threat to human health globally. A previous drug repurposing screen identified the approved anthelminthic drug niclosamide as a small molecule inhibitor of Zika virus infection. However, as antihelminthic drugs are generally designed to have low absorption when dosed orally, the very limited bioavailability of niclosamide will likely hinder its potential direct repurposing as an antiviral medication. Here, we conducted SAR studies focusing on the anilide and salicylic acid regions of niclosamide to improve physicochemical properties such as microsomal metabolic stability, permeability and solubility. We found that the 5-bromo substitution in the salicylic acid region retains potency while providing better drug-like properties. Other modifications in the anilide region with 2’-OMe and 2’-H substitutions were also advantageous. We found that the 4’-NO2 substituent can be replaced with a 4’-CN or 4’-CF3 substituents. Together, these modifications provide a basis for optimizing the structure of niclosamide to improve systemic exposure for application of niclosamide analogs as drug lead candidates for treating Zika and other viral infections. Indeed, key analogs were also able to rescue cells from the cytopathic effect of SARS-CoV-2 infection, indicating relevance for therapeutic strategies targeting the COVID-19 pandemic.
- Published
- 2021
- Full Text
- View/download PDF
45. Artemisinin resistance phenotypes and K13 inheritance in a
- Author
-
Juliana M, Sá, Sarah R, Kaslow, Michael A, Krause, Viviana A, Melendez-Muniz, Rebecca E, Salzman, Whitney A, Kite, Min, Zhang, Roberto R, Moraes Barros, Jianbing, Mu, Paul K, Han, J Patrick, Mershon, Christine E, Figan, Ramoncito L, Caleon, Rifat S, Rahman, Tyler J, Gibson, Chanaki, Amaratunga, Erika P, Nishiguchi, Kimberly F, Breglio, Theresa M, Engels, Soundarapandian, Velmurugan, Stacy, Ricklefs, Judith, Straimer, Nina F, Gnädig, Bingbing, Deng, Anna, Liu, Ababacar, Diouf, Kazutoyo, Miura, Gregory S, Tullo, Richard T, Eastman, Sumana, Chakravarty, Eric R, James, Kenneth, Udenze, Suzanne, Li, Daniel E, Sturdevant, Robert W, Gwadz, Stephen F, Porcella, Carole A, Long, David A, Fidock, Marvin L, Thomas, Michael P, Fay, B Kim Lee, Sim, Stephen L, Hoffman, John H, Adams, Rick M, Fairhurst, Xin-Zhuan, Su, and Thomas E, Wellems
- Subjects
Antimalarials ,Gene Expression Regulation ,Mutation ,Plasmodium falciparum ,Drug Resistance ,Protozoan Proteins ,Animals ,Aotidae ,Malaria, Falciparum ,Biological Sciences ,Artemisinins ,Crosses, Genetic - Abstract
Concerns about malaria parasite resistance to treatment with artemisinin drugs (ARTs) have grown with findings of prolonged parasite clearance t(1/2)s (>5 h) and their association with mutations in Plasmodium falciparum Kelch-propeller protein K13. Here, we describe a P. falciparum laboratory cross of K13 C580Y mutant with C580 wild-type parasites to investigate ART response phenotypes in vitro and in vivo. After genotyping >400 isolated progeny, we evaluated 20 recombinants in vitro: IC(50) measurements of dihydroartemisinin were at similar low nanomolar levels for C580Y- and C580-type progeny (mean ratio, 1.00; 95% CI, 0.62–1.61), whereas, in a ring-stage survival assay, the C580Y-type progeny had 19.6-fold (95% CI, 9.76–39.2) higher average counts. In splenectomized Aotus monkeys treated with three daily doses of i.v. artesunate, t(1/2) calculations by three different methods yielded mean differences of 0.01 h (95% CI, −3.66 to 3.67), 0.80 h (95% CI, −0.92 to 2.53), and 2.07 h (95% CI, 0.77–3.36) between C580Y and C580 infections. Incidences of recrudescence were 57% in C580Y (4 of 7) versus 70% in C580 (7 of 10) infections (−13% difference; 95% CI, −58% to 35%). Allelic substitution of C580 in a C580Y-containing progeny clone (76H10) yielded a transformant (76H10(C580Rev)) that, in an infected monkey, recrudesced regularly 13 times over 500 d. Frequent recrudescences of ART-treated P. falciparum infections occur with or without K13 mutations and emphasize the need for improved partner drugs to effectively eliminate the parasites that persist through the ART component of combination therapy.
- Published
- 2018
46. Screening the Pathogen Box for Molecules Active against Plasmodium Sexual Stages Using a New Nanoluciferase-Based Transgenic Line of P. berghei Identifies Transmission-Blocking Compounds
- Author
-
Daniel Y. Bargieri, Marisé S. Ramos, Richard T. Eastman, Juliana Calit, Irina Dobrescu, Xiomara A. Gaitán, and Miriam H. Borges
- Subjects
0301 basic medicine ,Plasmodium berghei ,Transgene ,Biology ,Plasmodium ,03 medical and health sciences ,Antimalarials ,Mice ,parasitic diseases ,medicine ,Gametocyte ,Animals ,Pharmacology (medical) ,Antimalarial Agent ,Luciferases ,Pathogen ,Pharmacology ,Mice, Inbred BALB C ,Transmission (medicine) ,medicine.disease ,biology.organism_classification ,Virology ,Malaria ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Culicidae ,Susceptibility ,Parasitic disease ,Nanoparticles - Abstract
Malaria remains an important parasitic disease with a large morbidity and mortality burden. Plasmodium transmission-blocking (TB) compounds are essential for achieving malaria elimination efforts. Recent efforts to develop high-throughput screening (HTS) methods to identify compounds that inhibit or kill gametocytes, the Plasmodium sexual stage infectious to mosquitoes, have yielded insight into new TB compounds. However, the activities of these compounds against gametes, formed in the first minutes of mosquito infection, are typically not assessed, unless screened in a standard membrane feeding assay, a labor-intensive assay. We demonstrate here the generation of a Plasmodium model for drug screens against gametes and fertilization. The new P. berghei line, named Ookluc, was genetically and pharmacologically validated and scalable for HTS. Screening the Pathogen Box from the Medicines for Malaria Venture using the new model identified promising TB compounds. The use of Ookluc in different libraries of compounds may aid in the identification of transmission-blocking drugs not assessed in screens against asexual stages or gametocytes.
- Published
- 2018
47. Sulfamethoxazole Levels in HIV-Exposed Uninfected Ugandan Children
- Author
-
Scott R. Penzak, Erin E. Gabriel, Jillian Neal, David Serwadda, Richard T. Eastman, Aggrey Anok, Kevin Newell, Steven J. Reynolds, Jingo Kasule, Charlotte V. Hobbs, and Patrick E. Duffy
- Subjects
Male ,Sulfamethoxazole ,Drug Resistance ,HIV Infections ,urologic and male genital diseases ,Pre-exposure prophylaxis ,chemistry.chemical_compound ,0302 clinical medicine ,Uganda ,030212 general & internal medicine ,Malaria, Falciparum ,biology ,Incidence (epidemiology) ,Incidence ,Articles ,female genital diseases and pregnancy complications ,Infectious Diseases ,Anti-Retroviral Agents ,Antifolate ,Female ,medicine.drug ,medicine.medical_specialty ,030231 tropical medicine ,Plasmodium falciparum ,Context (language use) ,03 medical and health sciences ,Antimalarials ,HIV Seroprevalence ,Virology ,Internal medicine ,Trimethoprim, Sulfamethoxazole Drug Combination ,medicine ,Humans ,Insecticide-Treated Bednets ,business.industry ,HIV ,Infant ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Trimethoprim ,chemistry ,Mutation ,Folic Acid Antagonists ,Parasitology ,Pre-Exposure Prophylaxis ,business ,Malaria - Abstract
Trimethoprim–sulfamethoxazole (TMP–SMX) prophylaxis in HIV-uninfected, exposed (HUE) children variably reduces clinical malaria burden despite antifolate resistance, but data regarding achieved serum levels and adherence are lacking. Serum samples from 70 HUE children aged 3–12 months from Rakai, Uganda, enrolled in an observational study were assayed for random SMX levels using a colorimetric assay. Adherence with TMP–SMX prophylaxis data (yes/no) was also collected. Of 148 visits with concurrent SMX levels available, 56% had self-reported adherence with TMP–SMX therapy. Among these 82 visits, mean (standard deviation) level was 19.78 (19.22) µg/mL, but 33% had SMX levels below half maximal inhibitory concentrations (IC50) for Plasmodium falciparum with some, but not all, of the reported antifolate resistance mutations reported in Uganda. With TMP–SMX prophylaxis, suboptimal adherence is concerning. Sulfamethoxazole levels below IC50s required to overcome malaria parasites with multiple antifolate resistance mutations may be significant. Further study of TMP–SMX in this context is needed.
- Published
- 2018
48. Actinoramide A Identified as a Potent Antimalarial from Titration-Based Screening of Marine Natural Product Extracts
- Author
-
Avi Raveh, Teatulohi Matainaho, Giselle Tamayo-Castillo, George E. Chlipala, Ryan MacArthur, Shugeng Cao, Patricia Dranchak, Richard T. Eastman, Ken Chih-Chien Cheng, David H. Sherman, Pamela J. Schultz, Jon Clardy, James Inglese, Jing Yuan, Rajarshi Guha, Matthew T. Okoneski, and Xin-zhuan Su
- Subjects
Costa Rica ,Prioritization ,Geologic Sediments ,Plasmodium falciparum ,Pharmaceutical Science ,Marine Biology ,Streptomyces ,Article ,Analytical Chemistry ,Antimalarials ,Papua New Guinea ,chemistry.chemical_compound ,Drug Discovery ,Botany ,Nuclear Magnetic Resonance, Biomolecular ,Phylogeny ,Organism ,Pharmacology ,Natural products ,Biological Products ,Natural product ,Molecular Structure ,biology ,Tetrapeptide ,Organic Chemistry ,New guinea ,biology.organism_classification ,Streptomyces species ,Complementary and alternative medicine ,chemistry ,Biochemistry ,Potent Antimalarial ,Molecular Medicine ,Titration ,Marine Extracts ,Oligopeptides - Abstract
Methods to identify the bioactive diversity within natural product extracts (NPEs) continue to evolve. NPEs constitute complex mixtures of chemical substances varying in structure, composition, and abundance. NPEs can therefore be challenging to evaluate efficiently with high-throughput screening approaches designed to test pure substances. Here we facilitate the rapid identification and prioritization of antimalarial NPEs using a pharmacologically driven, quantitative high-throughput-screening (qHTS) paradigm. In qHTS each NPE is tested across a concentration range from which sigmoidal response, efficacy, and apparent EC50s can be used to rank order NPEs for subsequent organism reculture, extraction, and fractionation. Using an NPE library derived from diverse marine microorganisms we observed potent antimalarial activity from two Streptomyces sp. extracts identified from thousands tested using qHTS. Seven compounds were isolated from two phylogenetically related Streptomyces species: Streptomyces ballenaensis collected from Costa Rica and Streptomyces bangulaensis collected from Papua New Guinea. Among them we identified actinoramides A and B, belonging to the unusually elaborated nonproteinogenic amino-acid-containing tetrapeptide series of natural products. In addition, we characterized a series of new compounds, including an artifact, 25-epi-actinoramide A, and actinoramides D, E, and F, which are closely related biosynthetic congeners of the previously reported metabolites. National Institutes of Health/[U01 TW007404]/NIH/Estados Unidos Instituto Nacional de Biodiversidad/[R-CM-INBio-82-2009-OT]/INBio/Costa Rica UCR::Vicerrectoría de Investigación::Unidades de Investigación::Ciencias Básicas::Centro de Investigaciones en Productos Naturales (CIPRONA)
- Published
- 2015
- Full Text
- View/download PDF
49. Mechanism of splenic cell death and host mortality in a Plasmodium yoelii malaria model
- Author
-
Nicolas Riteau, Kevin W. Bock, Norinne Lacerda-Queiroz, Ian N. Moore, Dragana Jankovic, Carole A. Long, Alan Sher, Richard T. Eastman, Marlene Orandle, and Xin-zhuan Su
- Subjects
0301 basic medicine ,Chemokine ,Biopsy ,T cell ,030231 tropical medicine ,lcsh:Medicine ,Apoptosis ,Parasitemia ,Disease ,Article ,Host-Parasite Interactions ,Mice ,03 medical and health sciences ,0302 clinical medicine ,parasitic diseases ,medicine ,Animals ,Humans ,Lymphocytes ,Mortality ,lcsh:Science ,Mice, Knockout ,Life Cycle Stages ,Multidisciplinary ,Cell Death ,biology ,lcsh:R ,Plasmodium yoelii ,medicine.disease ,biology.organism_classification ,Malaria ,DNA-Binding Proteins ,CXCL1 ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Cytokines ,lcsh:Q ,Female ,Inflammation Mediators ,Biomarkers ,Spleen - Abstract
Malaria is a fatal disease that displays a spectrum of symptoms and severity, which are determined by complex host-parasite interactions. It has been difficult to study the effects of parasite strains on disease severity in human infections, but the mechanisms leading to specific disease phenotypes can be investigated using strains of rodent malaria parasites that cause different disease symptoms in inbred mice. Using a unique mouse malaria model, here we investigated the mechanisms of splenic cell death and their relationship to control of parasitemia and host mortality. C57BL/6 mice infected with Plasmodium yoelii nigeriensis N67C display high levels of pro-inflammatory cytokines and chemokines (IL-6, IFN-γ, TNF-α, CXCL1, and CCL2) and extensive splenic damage with dramatic reduction of splenic cell populations. These disease phenotypes were rescued in RAG2−/−, IFN-γ−/−, or T cell depleted mice, suggesting IFN-γ and T cell mediated disease mechanisms. Additionally, apoptosis was one of the major pathways involved in splenic cell death, which coincides with the peaks of pro-inflammatory cytokines. Our results demonstrate the critical roles of T cells and IFN-γ in mediating splenic cell apoptosis, parasitemia control, and host lethality and thus may provide important insights for preventing/reducing morbidity associated with severe malaria in humans.
- Published
- 2017
- Full Text
- View/download PDF
50. Plasmodium genetic loci linked to host cytokine and chemokine responses
- Author
-
Sittiporn Pattaradilokrat, Mariam Quinones, Zhao K, Osamu Kaneko, Yanwei Qi, Jiang H, Li N, Zhu J, Carole A. Long, Xin-zhuan Su, Sethu C. Nair, Jian Wu, Jian Li, Richard T. Eastman, Huaman Mc, and Martine Zilversmit
- Subjects
Chemokine ,Plasmodium ,030231 tropical medicine ,Immunology ,Genes, Protozoan ,Quantitative Trait Loci ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,cytokine ,Animals ,China ,Genetics (clinical) ,Chemokine CCL2 ,Crosses, Genetic ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Polymorphism, Genetic ,pathogenesis ,chemokine ,Epistasis, Genetic ,Plasmodium yoelii ,biology.organism_classification ,medicine.disease ,3. Good health ,Malaria ,virulence ,Disease Models, Animal ,inflammation ,Genetic Loci ,Host-Pathogen Interactions ,biology.protein ,Cytokines ,Christian ministry ,Female ,genetic mapping ,Chemokines ,Genome, Protozoan - Abstract
Both host and parasite factors contribute to disease severity of malaria infection; however, the molecular mechanisms responsible for the disease and the host-parasite interactions involved remain largely unresolved. To investigate the effects of parasite factors on host immune responses and pathogenesis, we measured levels of plasma cytokines/chemokines (CCs) and growth rates in mice infected with two Plasmodium yoelii strains having different virulence phenotypes and in progeny from a genetic cross of the two parasites. Quantitative trait loci (QTL) analysis linked levels of many CCs, particularly IL-1β, IP-10, IFN-γ, MCP-1 and MIG, and early parasite growth rate to loci on multiple parasite chromosomes, including chromosomes 7, 9, 10, 12 and 13. Comparison of the genome sequences spanning the mapped loci revealed various candidate genes. The loci on chromosomes 7 and 13 had significant (P0.005) additive effects on IL-1β, IL-5 and IP-10 responses, and the chromosome 9 and 12 loci had significant (P=0.017) interaction. Infection of knockout mice showed critical roles of MCP-1 and IL-10 in parasitemia control and host mortality. These results provide important information for a better understanding of malaria pathogenesis and can be used to examine the role of these factors in human malaria infection.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.