DeepMalaria: Artificial Intelligence Driven Discovery of Potent Antiplasmodials

Antimalarial drugs are becoming less effective due to the emergence of drug resistance. Resistance has been reported for all available malaria drugs, including artemisinin, thus creating a perpetual need for alternative drug candidates. The traditional drug discovery approach of high throughput screening (HTS) of large compound libraries for identification of new drug leads is time-consuming and resource intensive. While virtual in silico screening is a solution to this problem, however, the generalization of the models is not ideal. Artificial intelligence (AI), utilizing either structure-based or ligand-based approaches, has demonstrated highly accurate performances in the field of chemical property prediction. Leveraging the existing data, AI would be a suitable alternative to blind-search HTS or fingerprint-based virtual screening. The AI model would learn patterns within the data and help to search for hit compounds efficiently. In this work, we introduce DeepMalaria, a deep-learning based process capable of predicting the anti-Plasmodium falciparum inhibitory properties of compounds using their SMILES. A graph-based model is trained on 13,446 publicly available antiplasmodial hit compounds from GlaxoSmithKline (GSK) dataset that are currently being used to find novel drug candidates for malaria. We validated this model by predicting hit compounds from a macrocyclic compound library and already approved drugs that are used for repurposing. We have chosen macrocyclic compounds as these ligand-binding structures are underexplored in malaria drug discovery. The in silico pipeline for this process also consists of additional validation of an in-house independent dataset consisting mostly of natural product compounds. Transfer learning from a large dataset was leveraged to improve the performance of the deep learning model. To validate the DeepMalaria generated hits, we used a commonly used SYBR Green I fluorescence assay based phenotypic screening. DeepMalaria was able to detect all the compounds with nanomolar activity and 87.5% of the compounds with greater than 50% inhibition. Further experiments to reveal the compounds’ mechanism of action have shown that not only does one of the hit compounds, DC-9237, inhibits all asexual stages of Plasmodium falciparum, but is a fast-acting compound which makes it a strong candidate for further optimization.

[1]  D. Chakrabarti,et al.  4-Nitro styrylquinoline is an antimalarial inhibiting multiple stages of Plasmodium falciparum asexual life cycle , 2017, International journal for parasitology. Drugs and drug resistance.

[2]  S. N. Bukhari,et al.  The Evolving Role of Natural Products from the Tropical Rainforests as a Replenishable Source of New Drug Leads , 2015 .

[3]  D. Newman,et al.  Natural Products as Sources of New Drugs from 1981 to 2014. , 2016, Journal of natural products.

[4]  Weiyuan Cui WHO urges the phasing out of artemisinin based monotherapy for malaria to reduce resistance , 2011, BMJ : British Medical Journal.

[5]  P. Ermert Design, Properties and Recent Application of Macrocycles in Medicinal Chemistry. , 2017, Chimia.

[6]  S. Ralph,et al.  Delayed death in the malaria parasite Plasmodium falciparum is caused by disruption of prenylation-dependent intracellular trafficking , 2019, PLoS biology.

[7]  M. Blanco Building upon Nature's Framework: Overview of Key Strategies Toward Increasing Drug-Like Properties of Natural Product Cyclopeptides and Macrocycles. , 2019, Methods in molecular biology.

[8]  E. Ashley,et al.  Drugs in Development for Malaria , 2018, Drugs.

[9]  Alán Aspuru-Guzik,et al.  Convolutional Networks on Graphs for Learning Molecular Fingerprints , 2015, NIPS.

[10]  J. Vederas,et al.  Drug Discovery and Natural Products: End of an Era or an Endless Frontier? , 2009, Science.

[11]  S. Hochreiter,et al.  DeepTox: Toxicity prediction using deep learning , 2017 .

[12]  D. Saunders,et al.  Dihydroartemisinin-piperaquine failure in Cambodia. , 2014, The New England journal of medicine.

[13]  R. Fairhurst,et al.  Artemisinin-Resistant Plasmodium falciparum Malaria. , 2016, Microbiology spectrum.

[14]  Hugo Ceulemans,et al.  Large-scale comparison of machine learning methods for drug target prediction on ChEMBL† †Electronic supplementary information (ESI) available: Overview, Data Collection and Clustering, Methods, Results, Appendix. See DOI: 10.1039/c8sc00148k , 2018, Chemical science.

[15]  D. Chakrabarti,et al.  Spirocyclic chromanes exhibit antiplasmodial activities and inhibit all intraerythrocytic life cycle stages , 2016, International journal for parasitology. Drugs and drug resistance.

[16]  David L. Selwood Macrocycles, the edge of drug‐likeness chemical space or Goldilocks zone? , 2017, Chemical biology & drug design.

[17]  Danny W. Wilson,et al.  Defining the Timing of Action of Antimalarial Drugs against Plasmodium falciparum , 2013, Antimicrobial Agents and Chemotherapy.

[18]  James R. Brown,et al.  Thousands of chemical starting points for antimalarial lead identification , 2010, Nature.

[19]  Sumudu P Leelananda,et al.  Computational methods in drug discovery , 2016, Beilstein journal of organic chemistry.

[20]  Nic Fleming,et al.  How artificial intelligence is changing drug discovery , 2018, Nature.

[21]  Andrey Kazennov,et al.  The cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncology , 2016, Oncotarget.

[22]  M. Cooper,et al.  Natural product and natural product derived drugs in clinical trials. , 2014, Natural product reports.

[23]  D. Wirth,et al.  High-Throughput Plasmodium falciparum Growth Assay for Malaria Drug Discovery , 2006, Antimicrobial Agents and Chemotherapy.

[24]  D C Swinney,et al.  Phenotypic vs. Target‐Based Drug Discovery for First‐in‐Class Medicines , 2013, Clinical pharmacology and therapeutics.

[25]  A. Rojek,et al.  Resource-Poor Countries with Fragile Health Care and Disease Surveillance and Response Systems , 2016 .

[26]  F. Koehn,et al.  The evolving role of natural products in drug discovery , 2005, Nature Reviews Drug Discovery.

[27]  A. Zhavoronkova,et al.  Artificial intelligence for aging and longevity research , 2018 .

[28]  Joseph Gomes,et al.  MoleculeNet: a benchmark for molecular machine learning† †Electronic supplementary information (ESI) available. See DOI: 10.1039/c7sc02664a , 2017, Chemical science.

[29]  Lei Jia,et al.  Chemi-net: a graph convolutional network for accurate drug property prediction , 2018, ArXiv.

[30]  L. Gerena,et al.  Assessment and Continued Validation of the Malaria SYBR Green I-Based Fluorescence Assay for Use in Malaria Drug Screening , 2007, Antimicrobial Agents and Chemotherapy.

[31]  Jyoti Chhibber-Goel,et al.  New leads for drug repurposing against malaria. , 2019, Drug discovery today.

[32]  P. Sanseau,et al.  Drug repurposing: progress, challenges and recommendations , 2018, Nature Reviews Drug Discovery.

[33]  D. Kwiatkowski,et al.  Spread of artemisinin resistance in Plasmodium falciparum malaria. , 2014, The New England journal of medicine.

[34]  Manish K. Gupta,et al.  Impact of Artificial Neural Networks in QSAR and Computational Modeling , 2016 .

[35]  Izhar Wallach,et al.  AtomNet: A Deep Convolutional Neural Network for Bioactivity Prediction in Structure-based Drug Discovery , 2015, ArXiv.

[36]  Andrew Y. Ng,et al.  Cardiologist-Level Arrhythmia Detection with Convolutional Neural Networks , 2017, ArXiv.

[37]  Michael Wainberg,et al.  Deep learning in biomedicine , 2018, Nature Biotechnology.

[38]  Stephen P. Hale,et al.  The exploration of macrocycles for drug discovery — an underexploited structural class , 2008, Nature Reviews Drug Discovery.

[39]  Christopher P. Austin,et al.  Cell Viability Assays -- Assay Guidance Manual , 2004 .

[40]  Jeremy N. Burrows,et al.  The Open Access Malaria Box: A Drug Discovery Catalyst for Neglected Diseases , 2013, PloS one.

[41]  Anne E Carpenter,et al.  Leveraging machine vision in cell-based diagnostics to do more with less , 2019, Nature Materials.

[42]  Ernest Fraenkel,et al.  Revealing disease-associated pathways by network integration of untargeted metabolomics , 2016, Nature Methods.

[43]  S. Ralph,et al.  Delayed Death by Plastid Inhibition in Apicomplexan Parasites. , 2019, Trends in parasitology.

[44]  Vijay S. Pande,et al.  Massively Multitask Networks for Drug Discovery , 2015, ArXiv.

[45]  P. Chiba,et al.  The SYBR Green I malaria drug sensitivity assay: performance in low parasitemia samples. , 2010, The American journal of tropical medicine and hygiene.

[46]  Ian Collins,et al.  Macrocycles in new drug discovery. , 2012, Future medicinal chemistry.

[47]  O. Mercereau‐Puijalon,et al.  Screening and evaluation of inhibitors of Plasmodium falciparum merozoite egress and invasion using cytometry. , 2012, Methods in molecular biology.

[48]  Alex Zhavoronkov,et al.  Artificial intelligence for aging and longevity research: Recent advances and perspectives , 2019, Ageing Research Reviews.

[49]  A. Whitty,et al.  Progress towards the broad use of non-peptide synthetic macrocycles in drug discovery. , 2017, Organic & biomolecular chemistry.

[50]  Edward W. Lowe,et al.  Computational Methods in Drug Discovery , 2014, Pharmacological Reviews.

[51]  D. Pei,et al.  Macrocycles as protein-protein interaction inhibitors. , 2017, The Biochemical journal.

[52]  Miguel Prudêncio,et al.  Liver-stage malaria parasites vulnerable to diverse chemical scaffolds , 2012, Proceedings of the National Academy of Sciences.

[53]  Thomas Blaschke,et al.  The rise of deep learning in drug discovery. , 2018, Drug discovery today.

[54]  Vijay S. Pande,et al.  Molecular graph convolutions: moving beyond fingerprints , 2016, Journal of Computer-Aided Molecular Design.

[55]  Haipeng Shen,et al.  Artificial intelligence in healthcare: past, present and future , 2017, Stroke and Vascular Neurology.

[56]  Dinggang Shen,et al.  A cybernetic eye for rare disease. , 2017, Nature biomedical engineering.

[57]  E. Winzeler,et al.  The genomic architecture of antimalarial drug resistance , 2019, Briefings in functional genomics.

[58]  Jun O. Liu,et al.  A clinical drug library screen identifies astemizole as an antimalarial agent , 2006, Nature chemical biology.

[59]  John Fox,et al.  Artificial intelligence-enabled healthcare delivery , 2018, Journal of the Royal Society of Medicine.

[60]  A. Hamilton,et al.  Macrocyclic anion receptors based on directed hydrogen bonding interactions , 2003 .

[61]  Gisbert Schneider,et al.  Automating drug discovery , 2017, Nature Reviews Drug Discovery.

[62]  Brian K. Shoichet,et al.  Virtual screening of chemical libraries , 2004, Nature.

[63]  David Rogers,et al.  Extended-Connectivity Fingerprints , 2010, J. Chem. Inf. Model..