The target landscape of clinical kinase drugs

An atlas for drug interactions Kinase inhibitors are an important class of drugs that block certain enzymes involved in diseases such as cancer and inflammatory disorders. There are hundreds of kinases within the human body, so knowing the kinase “target” of each drug is essential for developing successful treatment strategies. Sometimes clinical trials can fail because drugs bind more than one target. Yet sometimes off-target effects can be beneficial, and drugs can be repurposed for treatment of additional diseases. Klaeger et al. performed a comprehensive analysis of 243 kinase inhibitors that are either approved for use or in clinical trials. They provide an open-access resource of target summaries that could help researchers develop better drugs, understand how existing drugs work, and design more effective clinical trials. Science, this issue p. eaan4368 The druggable kinome is unraveled. INTRODUCTION Molecularly targeted drugs such as imatinib and crizotinib have revolutionized the treatment of certain blood and lung cancers because of their remarkable clinical success. Over the past 20 years, protein kinases have become a major class of drug targets because these signaling biomolecules are often deregulated in disease, particularly in cancer. Today, 37 small kinase inhibitors (KIs) are approved medicines worldwide and more than 250 drug candidates are undergoing clinical evaluation. RATIONALE Although it is commonly accepted that most KIs target more than one protein, the extent to which this information is available to the public varies greatly between drugs. It would seem important to thoroughly characterize the target spectrum of any drug because additional off-targets may offer opportunities, not only for repurposing but also to explain undesired side effects. To this end, we used a chemical proteomic approach (kinobeads) and quantitative mass spectrometry to characterize the target space of 243 clinical KIs that are approved drugs or have been tested in humans. RESULTS The number of targets for a given drug differed substantially. Whereas some compounds showed exquisite selectivity, others targeted more than 100 kinases simultaneously, making it difficult to attribute their biological effects to any particular mode of action. Also of note is that recently developed irreversible KIs can address more kinases than their intended targets epidermal growth factor receptor (EGFR) and Bruton’s tyrosine kinase (BTK). Collectively, the evaluated KIs targeted 220 kinases with submicromolar affinity, offering a view of the druggable kinome and enabling the development of a universal new selectivity metric termed CATDS (concentration- and target-dependent selectivity). All drug profiles can be interactively explored in ProteomicsDB and a purpose-built shinyApp. Many uses of this unique data and analysis resource by the scientific community can be envisaged, of which we can only highlight a few. The profiles identified many new targets for established drugs, thus improving our understanding of how these drugs might exert their phenotypic effects. For example, we evaluated novel salt-inducible kinase 2 (SIK2) inhibitors for their ability to modulate tumor necrosis factor–α (TNFα) and interleukin-10 (IL-10) production, which may allow repurposing these drugs for inflammatory conditions. Integrating target space information with phosphoproteomic analysis of several EGFR inhibitors enabled the identification of drug response markers inside and outside the canonical EGFR signaling pathway. Off-target identification may also inform drug discovery projects using high-value clinical molecules as lead compounds. We illustrate such a case by a novel structure-affinity relationship analysis of MELK inhibitors based on target profiles and cocrystal structures. To assess the repurposing potential of approved or clinically advanced compounds, we used cell-based assays and mouse xenografts to show that golvatinib and cabozantinib may be used for the treatment of acute myeloid leukemia (AML) based on their FLT3 inhibitory activity. CONCLUSION This work provides a rich data resource describing the target landscape of 243 clinically tested KIs. It is the most comprehensive study to date and illustrates how the information may be used in basic research, drug discovery, or clinical decision-making. Schematic representation of identifying the druggable kinome. A chemical proteomic approach revealed quantitative interaction profiles of 243 clinically evaluated small-molecule KIs covering half of the human kinome. Results can be interactively explored in ProteomicsDB and inform basic biology, drug discovery, and clinical decision-making. Kinase inhibitors are important cancer therapeutics. Polypharmacology is commonly observed, requiring thorough target deconvolution to understand drug mechanism of action. Using chemical proteomics, we analyzed the target spectrum of 243 clinically evaluated kinase drugs. The data revealed previously unknown targets for established drugs, offered a perspective on the “druggable” kinome, highlighted (non)kinase off-targets, and suggested potential therapeutic applications. Integration of phosphoproteomic data refined drug-affected pathways, identified response markers, and strengthened rationale for combination treatments. We exemplify translational value by discovering SIK2 (salt-inducible kinase 2) inhibitors that modulate cytokine production in primary cells, by identifying drugs against the lung cancer survival marker MELK (maternal embryonic leucine zipper kinase), and by repurposing cabozantinib to treat FLT3-ITD–positive acute myeloid leukemia. This resource, available via the ProteomicsDB database, should facilitate basic, clinical, and drug discovery research and aid clinical decision-making.

[1]  J. Pouysségur,et al.  Effect of mutations affecting Na+:H+ antiport activity on tumorigenic potential of hamster lung fibroblasts , 1988, Journal of cellular biochemistry.

[2]  J M Thornton,et al.  LIGPLOT: a program to generate schematic diagrams of protein-ligand interactions. , 1995, Protein engineering.

[3]  G. Murshudov,et al.  Refinement of macromolecular structures by the maximum-likelihood method. , 1997, Acta crystallographica. Section D, Biological crystallography.

[4]  D. Sgouras,et al.  Transcriptional Repressor ERF Is a Ras/Mitogen-Activated Protein Kinase Target That Regulates Cellular Proliferation , 1999, Molecular and Cellular Biology.

[5]  P. Cohen,et al.  Specificity and mechanism of action of some commonly used protein kinase inhibitors. , 2000, The Biochemical journal.

[6]  B. Druker,et al.  Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. , 2000, The Journal of pharmacology and experimental therapeutics.

[7]  D. Gilliland,et al.  FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model. , 2002, Blood.

[8]  F. Diederich,et al.  Interactions with aromatic rings in chemical and biological recognition. , 2003, Angewandte Chemie.

[9]  J. Warmus,et al.  Structures of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition , 2004, Nature Structural &Molecular Biology.

[10]  K Henrick,et al.  Electronic Reprint Biological Crystallography Secondary-structure Matching (ssm), a New Tool for Fast Protein Structure Alignment in Three Dimensions Biological Crystallography Secondary-structure Matching (ssm), a New Tool for Fast Protein Structure Alignment in Three Dimensions , 2022 .

[11]  P. Cohen,et al.  ERF Nuclear Shuttling, a Continuous Monitor of Erk Activity That Links It to Cell Cycle Progression , 2004, Molecular and Cellular Biology.

[12]  L. Meijer,et al.  Crystal Structure of Pyridoxal Kinase in Complex with Roscovitine and Derivatives* , 2005, Journal of Biological Chemistry.

[13]  L. Wodicka,et al.  A small molecule–kinase interaction map for clinical kinase inhibitors , 2005, Nature Biotechnology.

[14]  N. Perkins,et al.  Regulation of NF‐κB and p53 through activation of ATR and Chk1 by the ARF tumour suppressor , 2005, The EMBO journal.

[15]  R. Cardone,et al.  The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis , 2005, Nature Reviews Cancer.

[16]  N. Perkins,et al.  Cisplatin mimics ARF tumor suppressor regulation of RelA (p65) nuclear factor-kappaB transactivation. , 2006, Cancer research.

[17]  P. Evans,et al.  Scaling and assessment of data quality. , 2006, Acta crystallographica. Section D, Biological crystallography.

[18]  L. Otterbein,et al.  N-(5-chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5- (tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine, a novel, highly selective, orally available, dual-specific c-Src/Abl kinase inhibitor. , 2006, Journal of medicinal chemistry.

[19]  R. Jove,et al.  Dasatinib inhibits migration and invasion in diverse human sarcoma cell lines and induces apoptosis in bone sarcoma cells dependent on SRC kinase for survival. , 2007, Cancer research.

[20]  P. Cohen,et al.  The selectivity of protein kinase inhibitors: a further update. , 2007, The Biochemical journal.

[21]  P. Graczyk Gini coefficient: a new way to express selectivity of kinase inhibitors against a family of kinases. , 2007, Journal of medicinal chemistry.

[22]  M. Mann,et al.  Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips , 2007, Nature Protocols.

[23]  Bernhard Kuster,et al.  Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors , 2007, Nature Biotechnology.

[24]  Randy J. Read,et al.  Phaser crystallographic software , 2007, Journal of applied crystallography.

[25]  P. Hwu,et al.  Visualizing fewer than 10 mouse T cells with an enhanced firefly luciferase in immunocompetent mouse models of cancer , 2008, Proceedings of the National Academy of Sciences.

[26]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

[27]  Mindy I. Davis,et al.  A quantitative analysis of kinase inhibitor selectivity , 2008, Nature Biotechnology.

[28]  M. Mann,et al.  Mislocalized activation of oncogenic RTKs switches downstream signaling outcomes. , 2009, Molecular cell.

[29]  P. Cossart,et al.  Human BAHD1 promotes heterochromatic gene silencing , 2009, Proceedings of the National Academy of Sciences.

[30]  G. Superti-Furga,et al.  The structure of the leukemia drug imatinib bound to human quinone reductase 2 (NQO2) , 2009, BMC Structural Biology.

[31]  J. Cox,et al.  Proteomics strategy for quantitative protein interaction profiling in cell extracts , 2009, Nature Methods.

[32]  C. Eyers Universal sample preparation method for proteome analysis , 2009 .

[33]  A. Cheng,et al.  Analysis of kinase inhibitor selectivity using a thermodynamics-based partition index. , 2010, Journal of medicinal chemistry.

[34]  Joost C. M. Uitdehaag,et al.  A theoretical entropy score as a single value to express inhibitor selectivity , 2011, BMC Bioinformatics.

[35]  Brendan MacLean,et al.  Skyline: an open source document editor for creating and analyzing targeted proteomics experiments , 2010, Bioinform..

[36]  P. Emsley,et al.  Features and development of Coot , 2010, Acta crystallographica. Section D, Biological crystallography.

[37]  Vincent B. Chen,et al.  Correspondence e-mail: , 2000 .

[38]  J. Mauer,et al.  Phosphoinositide-Dependent Kinase 1 Provides Negative Feedback Inhibition to Toll-Like Receptor-Mediated NF-κB Activation in Macrophages , 2010, Molecular and Cellular Biology.

[39]  Wolfgang Kabsch,et al.  Integration, scaling, space-group assignment and post-refinement , 2010, Acta crystallographica. Section D, Biological crystallography.

[40]  E. Estey,et al.  Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[41]  Theonie Anastassiadis,et al.  Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity , 2011, Nature biotechnology.

[42]  N. Pannu,et al.  REFMAC5 for the refinement of macromolecular crystal structures , 2011, Acta crystallographica. Section D, Biological crystallography.

[43]  Yongchang Shi,et al.  Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth , 2011, Molecular Cancer Therapeutics.

[44]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[45]  Randy J. Read,et al.  Overview of the CCP4 suite and current developments , 2011, Acta crystallographica. Section D, Biological crystallography.

[46]  N. Gray,et al.  In situ kinase profiling reveals functionally relevant properties of native kinases. , 2011, Chemistry & biology.

[47]  Mindy I. Davis,et al.  Comprehensive analysis of kinase inhibitor selectivity , 2011, Nature Biotechnology.

[48]  Owen Johnson,et al.  iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM , 2011, Acta crystallographica. Section D, Biological crystallography.

[49]  M. Mann,et al.  Andromeda: a peptide search engine integrated into the MaxQuant environment. , 2011, Journal of proteome research.

[50]  P. Hajduk,et al.  Navigating the kinome. , 2011, Nature chemical biology.

[51]  P. Andrew Karplus,et al.  Linking Crystallographic Model and Data Quality , 2012, Science.

[52]  Patrick G. A. Pedrioli,et al.  Phosphorylation of CRTC3 by the salt-inducible kinases controls the interconversion of classically activated and regulatory macrophages , 2012, Proceedings of the National Academy of Sciences.

[53]  Bernhard Kuster,et al.  Systematic Identification of the HSP90 Regulated Proteome , 2012, Molecular & Cellular Proteomics.

[54]  Joost C. M. Uitdehaag,et al.  A guide to picking the most selective kinase inhibitor tool compounds for pharmacological validation of drug targets , 2012, British journal of pharmacology.

[55]  I. Jeremias,et al.  In Vivo Imaging Enables High Resolution Preclinical Trials on Patients’ Leukemia Cells Growing in Mice , 2012, PloS one.

[56]  Randy J. Read,et al.  Phenix - a comprehensive python-based system for macromolecular structure solution , 2012 .

[57]  P. Evans Resolving Some Old Problems in Protein Crystallography , 2012, Science.

[58]  Derek J. Bailey,et al.  Parallel Reaction Monitoring for High Resolution and High Mass Accuracy Quantitative, Targeted Proteomics* , 2012, Molecular & Cellular Proteomics.

[59]  B. Domon,et al.  Targeted Proteomic Quantification on Quadrupole-Orbitrap Mass Spectrometer* , 2012, Molecular & Cellular Proteomics.

[60]  E. Barillot,et al.  Prognostic impact of vitamin B6 metabolism in lung cancer. , 2012, Cell reports.

[61]  W. Hiddemann,et al.  Casitas B-lineage lymphoma mutants activate AKT to induce transformation in cooperation with class III receptor tyrosine kinases. , 2013, Experimental hematology.

[62]  Patrick G. A. Pedrioli,et al.  PGE2 Induces Macrophage IL-10 Production and a Regulatory-like Phenotype via a Protein Kinase A–SIK–CRTC3 Pathway , 2013, The Journal of Immunology.

[63]  Bernhard Kuster,et al.  DMSO enhances electrospray response, boosting sensitivity of proteomic experiments , 2013, Nature Methods.

[64]  Ron Prywes,et al.  Serum regulation of Id1 expression by a BMP pathway and BMP responsive element. , 2013, Biochimica et biophysica acta.

[65]  G. Canevari,et al.  Structural insight into maternal embryonic leucine zipper kinase (MELK) conformation and inhibition toward structure-based drug design. , 2013, Biochemistry.

[66]  B. Cravatt,et al.  Determining target engagement in living systems. , 2013, Nature chemical biology.

[67]  Mathias Wilhelm,et al.  Global proteome analysis of the NCI-60 cell line panel. , 2013, Cell reports.

[68]  K. Diederichs,et al.  Better models by discarding data? , 2013, Acta crystallographica. Section D, Biological crystallography.

[69]  Michal Vieth,et al.  What general conclusions can we draw from kinase profiling data sets? , 2013, Biochimica et biophysica acta.

[70]  Bernhard Kuster,et al.  Comparing immobilized kinase inhibitors and covalent ATP probes for proteomic profiling of kinase expression and drug selectivity. , 2013, Journal of proteome research.

[71]  Zhifu Sun,et al.  Network-based approach identified cell cycle genes as predictor of overall survival in lung adenocarcinoma patients. , 2013, Lung cancer.

[72]  W. Hiddemann,et al.  Exome sequencing identifies recurring FLT3 N676K mutations in core-binding factor leukemia. , 2012, Blood.

[73]  G. Kayser,et al.  Simultaneous Multi-Antibody Staining in Non-Small Cell Lung Cancer Strengthens Diagnostic Accuracy Especially in Small Tissue Samples , 2013, PloS one.

[74]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration , 2012, Briefings Bioinform..

[75]  Martin Augustin,et al.  A broad activity screen in support of a chemogenomic map for kinase signalling research and drug discovery. , 2013, The Biochemical journal.

[76]  Julian Blagg,et al.  A public-private partnership to unlock the untargeted kinome. , 2013, Nature chemical biology.

[77]  L. Garraway,et al.  Oncogenic and drug sensitive NTRK1 rearrangements in lung cancer , 2013, Nature Medicine.

[78]  K. Clark Protein kinase networks that limit TLR signalling. , 2014, Biochemical Society transactions.

[79]  Lars Juhl Jensen,et al.  A Comparison of Protein Kinases Inhibitor Screening Methods Using Both Enzymatic Activity and Binding Affinity Determination , 2014, PloS one.

[80]  Daniel J. O'Connell,et al.  Small-molecule screening identifies inhibition of salt-inducible kinases as a therapeutic strategy to enhance immunoregulatory functions of dendritic cells , 2014, Proceedings of the National Academy of Sciences.

[81]  Andrea Lombardi Borgia,et al.  The TPM3‐NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition , 2014, Molecular oncology.

[82]  Heiner Koch,et al.  Comprehensive and Reproducible Phosphopeptide Enrichment Using Iron Immobilized Metal Ion Affinity Chromatography (Fe-IMAC) Columns , 2014, Molecular & Cellular Proteomics.

[83]  L. Peshkin,et al.  Exploiting polypharmacology for drug target deconvolution , 2014, Proceedings of the National Academy of Sciences.

[84]  G. Drewes,et al.  Tracking cancer drugs in living cells by thermal profiling of the proteome , 2014, Science.

[85]  S. Dry,et al.  Roles of bone morphogenetic protein signaling in osteosarcoma , 2014, International Orthopaedics.

[86]  George Papadatos,et al.  The ChEMBL bioactivity database: an update , 2013, Nucleic Acids Res..

[87]  S. Knapp,et al.  Exploration of Type II Binding Mode: A Privileged Approach for Kinase Inhibitor Focused Drug Discovery? , 2014, ACS chemical biology.

[88]  B. Kuster,et al.  Mass-spectrometry-based draft of the human proteome , 2014, Nature.

[89]  Marco Y. Hein,et al.  Accurate Proteome-wide Label-free Quantification by Delayed Normalization and Maximal Peptide Ratio Extraction, Termed MaxLFQ * , 2014, Molecular & Cellular Proteomics.

[90]  W. Hiddemann,et al.  Activating FLT3 Mutants Show Distinct Gain-of-Function Phenotypes In Vitro and a Characteristic Signaling Pathway Profile Associated with Prognosis in Acute Myeloid Leukemia , 2014, PloS one.

[91]  H. Leonhardt,et al.  Tyrosin Kinase Inhibition Restores the Membrane Localization of FLT3-ITD , 2015 .

[92]  M. Clausen,et al.  FDA-approved small-molecule kinase inhibitors. , 2015, Trends in pharmacological sciences.

[93]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[94]  Vasilis Ntziachristos,et al.  Early recognition of lung cancer by integrin targeted imaging in K‐ras mouse model , 2015, International journal of cancer.

[95]  Nan Wang,et al.  A Phosphoproteomic Comparison of B-RAFV600E and MKK1/2 Inhibitors in Melanoma Cells* , 2015, Molecular & Cellular Proteomics.

[96]  L. Cesaro,et al.  Protein kinase CK2 potentiates translation efficiency by phosphorylating eIF3j at Ser127. , 2015, Biochimica et biophysica acta.

[97]  A. Giaccia,et al.  Analysis of p53 transactivation domain mutants reveals Acad11 as a metabolic target important for p53 pro-survival function. , 2015, Cell reports.

[98]  G. Superti-Furga,et al.  A cellular screen identifies ponatinib and pazopanib as inhibitors of necroptosis , 2015, Cell Death and Disease.

[99]  B. Kuster,et al.  Optimized chemical proteomics assay for kinase inhibitor profiling. , 2015, Journal of proteome research.

[100]  W. Hiddemann,et al.  Individualized Treatment Strategy with Small-Molecular Inhibitors inAcute Myeloid Leukemia with Concurrent FLT3-ITD and FLT3-TKDMutation , 2015 .

[101]  A. Prescott,et al.  The clinically approved drugs dasatinib and bosutinib induce anti-inflammatory macrophages by inhibiting the salt-inducible kinases , 2014, The Biochemical journal.

[102]  Doriano Fabbro,et al.  Ten things you should know about protein kinases: IUPHAR Review 14 , 2015, British journal of pharmacology.

[103]  Zifei Zhou,et al.  The importance of Src signaling in sarcoma. , 2015, Oncology letters.

[104]  Y. Mishina,et al.  Toward the Validation of Maternal Embryonic Leucine Zipper Kinase: Discovery, Optimization of Highly Potent and Selective Inhibitors, and Preliminary Biology Insight. , 2016, Journal of Medicinal Chemistry.

[105]  Jing Liang,et al.  FOXK2 Elicits Massive Transcription Repression and Suppresses the Hypoxic Response and Breast Cancer Carcinogenesis. , 2016, Cancer cell.

[106]  Heiner Koch,et al.  Phosphoproteome Profiling Reveals Molecular Mechanisms of Growth-Factor-Mediated Kinase Inhibitor Resistance in EGFR-Overexpressing Cancer Cells. , 2016, Journal of proteome research.

[107]  W. Taylor,et al.  OTSSP167 Abrogates Mitotic Checkpoint through Inhibiting Multiple Mitotic Kinases , 2016, PloS one.

[108]  S. Armstrong,et al.  Impact of FLT3-ITD location on sensitivity to TKI-therapy in vitro and in vivo , 2016, Leukemia.

[109]  Chris de Graaf,et al.  KLIFS: a structural kinase-ligand interaction database , 2015, Nucleic Acids Res..

[110]  Yusuke Nakamura,et al.  Preclinical evaluation of biomarkers associated with antitumor activity of MELK inhibitor , 2016, Oncotarget.

[111]  John P. Overington,et al.  Comprehensive characterization of the Published Kinase Inhibitor Set , 2016, Nature Biotechnology.

[112]  Marco Y. Hein,et al.  The Perseus computational platform for comprehensive analysis of (prote)omics data , 2016, Nature Methods.

[113]  B. Kuster,et al.  Chemical Proteomics and Structural Biology Define EPHA2 Inhibition by Clinical Kinase Drugs. , 2016, ACS chemical biology.

[114]  Chien-Yuan Chen,et al.  Cabozantinib is selectively cytotoxic in acute myeloid leukemia cells with FLT3-internal tandem duplication (FLT3-ITD). , 2016, Cancer letters.

[115]  C. Gilliéron,et al.  SIK inhibition in human myeloid cells modulates TLR and IL‐1R signaling and induces an anti‐inflammatory phenotype , 2016, Journal of leukocyte biology.

[116]  Robert Preissner,et al.  Chemical Proteomics Reveals Ferrochelatase as a Common Off-target of Kinase Inhibitors. , 2016, ACS chemical biology.

[117]  Michael L. Gatza,et al.  Proteogenomics connects somatic mutations to signaling in breast cancer , 2016, Nature.

[118]  Nathanael S Gray,et al.  MELK is not necessary for the proliferation of basal-like breast cancer cells , 2017, eLife.

[119]  A. Lin,et al.  CRISPR/Cas9 mutagenesis invalidates a putative cancer dependency targeted in on-going clinical trials , 2017, eLife.

[120]  A. Drilon,et al.  Somatic chromosomal engineering identifies BCAN-NTRK1 as a potent glioma driver and therapeutic target , 2017, Nature Communications.

[121]  Florent Baty,et al.  Dose-Response Analysis Using R , 2015, PLoS ONE.