Advance in Reversible Covalent Kinase Inhibitors

Reversible covalent kinase inhibitors (RCKIs) are a class of novel kinase inhibitors attracting increasing attention because they simultaneously show the selectivity of covalent kinase inhibitors, yet avoid permanent protein-modification-induced adverse effects. Over the last decade, RCKIs have been reported to target different kinases, including atypical kinases. Currently, three RCKIs are undergoing clinical trials to treat specific diseases, for example, Pemphigus, an autoimmune disorder. In this perspective, first, RCKIs are systematically summarized, including characteristics of electrophilic groups, chemical scaffolds, nucleophilic residues, and binding modes. Second, we provide insights into privileged electrophiles, the distribution of nucleophiles and hence effective design strategies for RCKIs. Finally, we provide a brief perspective on future design strategies for RCKIs, including those that target proteins other than kinases.

[1]  J. M. Bradshaw,et al.  Discovery of Reversible Covalent Bruton's Tyrosine Kinase Inhibitors PRN473 and PRN1008 (Rilzabrutinib). , 2022, Journal of medicinal chemistry.

[2]  R. Roskoski Properties of FDA-approved small molecule protein kinase inhibitors: a 2022 update. , 2021, Pharmacological research.

[3]  D. Murrell,et al.  Bruton Tyrosine Kinase Inhibition and Its Role as an Emerging Treatment in Pemphigus , 2021, Frontiers in Medicine.

[4]  D. Fabbro,et al.  Trends in kinase drug discovery: targets, indications and inhibitor design , 2021, Nature Reviews Drug Discovery.

[5]  Jiahua Rao,et al.  Quantitative evaluation of explainable graph neural networks for molecular property prediction , 2021, Patterns.

[6]  P. Jänne,et al.  Kinase drug discovery 20 years after imatinib: progress and future directions , 2021, Nature Reviews Drug Discovery.

[7]  Colin J. Daniel,et al.  Sulfopin is a covalent inhibitor of Pin1 that blocks Myc-driven tumors in vivo , 2021, Nature Chemical Biology.

[8]  R. Roskoski Properties of FDA-approved small molecule phosphatidylinositol 3-kinase inhibitors prescribed for the treatment of malignancies. , 2021, Pharmacological research.

[9]  Rong Li,et al.  Strategic Design of Catalytic Lysine-Targeting Reversible Covalent BCR-ABL Inhibitors. , 2021, Angewandte Chemie.

[10]  Chris de Graaf,et al.  KLIFS: an overhaul after the first 5 years of supporting kinase research , 2020, Nucleic Acids Res..

[11]  Derek J. Essegian,et al.  The Clinical Kinase Index: A Method to Prioritize Understudied Kinases as Drug Targets for the Treatment of Cancer , 2020, Cell reports. Medicine.

[12]  W. Jahnke,et al.  Discovery of Roblitinib (FGF401) as a Reversible-Covalent Inhibitor of the Kinase Activity of Fibroblast Growth Factor Receptor 4. , 2020, Journal of medicinal chemistry.

[13]  G. Keserű,et al.  An electrophilic warhead library for mapping the reactivity and accessibility of tractable cysteines in protein kinases. , 2020, European journal of medicinal chemistry.

[14]  D. Murrell,et al.  Open trial of Bruton's tyrosine kinase inhibitor (PRN1008) in the treatment of canine pemphigus foliaceus. , 2020, Veterinary dermatology.

[15]  S. Constantinescu,et al.  Advances in covalent kinase inhibitors. , 2020, Chemical Society reviews.

[16]  S. Meroueh,et al.  Small-molecule covalent bond formation at tyrosine creates a binding site and inhibits activation of Ral GTPases , 2020, Proceedings of the National Academy of Sciences.

[17]  S. Albeck,et al.  Efficient Targeted Degradation via Reversible and Irreversible Covalent PROTACs , 2020, Journal of the American Chemical Society.

[18]  Michael L. Wang,et al.  Enhancing intracellular accumulation and target engagement of PROTACs with reversible covalent chemistry , 2019, Nature Communications.

[19]  N. Gray,et al.  Recent Advances in Selective and Irreversible Covalent Ligand Development and Validation. , 2019, Cell chemical biology.

[20]  J. Irwin,et al.  Discovery of Lysine-Targeted eIF4E Inhibitors through Covalent Docking. , 2019, Journal of the American Chemical Society.

[21]  Kathryn A. Porter,et al.  Kinase Atlas: Druggability Analysis of Potential Allosteric Sites in Kinases. , 2019, Journal of medicinal chemistry.

[22]  W. Liu,et al.  Covalent Inhibition in Drug Discovery , 2019, ChemMedChem.

[23]  I. E. Smith,et al.  PROTAC-Mediated Degradation of Bruton's Tyrosine Kinase Is Inhibited by Covalent Binding. , 2019, ACS chemical biology.

[24]  S. Laufer,et al.  Emerging and Re-Emerging Warheads for Targeted Covalent Inhibitors: Applications in Medicinal Chemistry and Chemical Biology. , 2018, Journal of medicinal chemistry.

[25]  M. Ono,et al.  Selective and reversible modification of kinase cysteines with chlorofluoroacetamides , 2019, Nature Chemical Biology.

[26]  Daniel P. Walker,et al.  Aminopyrazole Carboxamide Bruton's Tyrosine Kinase Inhibitors. Irreversible to Reversible Covalent Reactive Group Tuning. , 2018, ACS medicinal chemistry letters.

[27]  Yan Li,et al.  Comparative Assessment of Scoring Functions: The CASF-2016 Update , 2018, J. Chem. Inf. Model..

[28]  P. Bourne,et al.  Overview of Current Type I/II Kinase Inhibitors , 2018, Next Generation Kinase Inhibitors.

[29]  J. Bauman,et al.  Identification of Cyanamide-Based Janus Kinase 3 (JAK3) Covalent Inhibitors. , 2018, Journal of medicinal chemistry.

[30]  M. Katoh Fibroblast growth factor receptors as treatment targets in clinical oncology , 2018, Nature Reviews Clinical Oncology.

[31]  G. F. Ruda,et al.  Rapid Covalent-Probe Discovery by Electrophile-Fragment Screening , 2018, bioRxiv.

[32]  N. Westwood,et al.  The Meisenheimer Complex as a Paradigm in Drug Discovery: Reversible Covalent Inhibition through C67 of the ATP Binding Site of PLK1. , 2018, Cell chemical biology.

[33]  W. Fast,et al.  The Taxonomy of Covalent Inhibitors. , 2018, Biochemistry.

[34]  S. Knapp,et al.  The Cysteinome of Protein Kinases as a Target in Drug Development. , 2018, Angewandte Chemie.

[35]  N. Gray,et al.  Kinase inhibitors: the road ahead , 2018, Nature Reviews Drug Discovery.

[36]  Philip E Bourne,et al.  Progress with covalent small-molecule kinase inhibitors. , 2018, Drug discovery today.

[37]  H. Rupasinghe,et al.  Kinase-targeted cancer therapies: progress, challenges and future directions , 2018, Molecular Cancer.

[38]  K. Kang,et al.  Essential Role of Polo-like Kinase 1 (Plk1) Oncogene in Tumor Growth and Metastasis of Tamoxifen-Resistant Breast Cancer , 2018, Molecular Cancer Therapeutics.

[39]  P. Furet,et al.  2-Formylpyridyl Ureas as Highly Selective Reversible-Covalent Inhibitors of Fibroblast Growth Factor Receptor 4. , 2018, ACS medicinal chemistry letters.

[40]  J. M. Bradshaw,et al.  PRN473, an inhibitor of Bruton's tyrosine kinase, inhibits neutrophil recruitment via inhibition of macrophage antigen‐1 signalling , 2018, British journal of pharmacology.

[41]  J. Funk,et al.  A phase I trial of PRN1008, a novel reversible covalent inhibitor of Bruton's tyrosine kinase, in healthy volunteers , 2017, British journal of clinical pharmacology.

[42]  P. Furet,et al.  Approaches to selective fibroblast growth factor receptor 4 inhibition through targeting the ATP-pocket middle-hinge region. , 2017, MedChemComm.

[43]  L. Olsen,et al.  Aldehyde Oxidase: Reaction Mechanism and Prediction of Site of Metabolism , 2017, ACS omega.

[44]  K. Shokat,et al.  Drugging the 'undruggable' cancer targets , 2017, Nature Reviews Cancer.

[45]  D. Rauh,et al.  Characterization of Covalent-Reversible EGFR Inhibitors , 2017, ACS omega.

[46]  P. Bourne,et al.  Determining Cysteines Available for Covalent Inhibition Across the Human Kinome. , 2017, Journal of medicinal chemistry.

[47]  N. Turner,et al.  Advances and challenges in targeting FGFR signalling in cancer , 2017, Nature Reviews Cancer.

[48]  Tudor I. Oprea,et al.  A comprehensive map of molecular drug targets , 2016, Nature Reviews Drug Discovery.

[49]  S. Knapp,et al.  Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding Pocket , 2016, Cell chemical biology.

[50]  T. Baillie,et al.  Targeted Covalent Inhibitors for Drug Design. , 2016, Angewandte Chemie.

[51]  R. Zahedi,et al.  Insight into the Inhibition of Drug-Resistant Mutants of the Receptor Tyrosine Kinase EGFR. , 2016, Angewandte Chemie.

[52]  N. Hannett,et al.  Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. , 2016, Nature chemical biology.

[53]  R. Obach,et al.  Intrinsic reactivity profile of electrophilic moieties to guide covalent drug design: N-α-acetyl-L-lysine as an amine nucleophile , 2016 .

[54]  A. Olson,et al.  Proteome-wide covalent ligand discovery in native biological systems , 2016, Nature.

[55]  H. Gharwan,et al.  Kinase inhibitors and monoclonal antibodies in oncology: clinical implications , 2016, Nature Reviews Clinical Oncology.

[56]  Zheng Zhao,et al.  Delineation of Polypharmacology across the Human Structural Kinome Using a Functional Site Interaction Fingerprint Approach. , 2016, Journal of medicinal chemistry.

[57]  S. Knapp,et al.  The ins and outs of selective kinase inhibitor development. , 2015, Nature chemical biology.

[58]  Chengbo Zhan,et al.  Access to a new class of synthetic building blocks via trifluoromethoxylation of pyridines and pyrimidines , 2015, Chemical science.

[59]  Megan Cully Rational drug design: Tuning kinase inhibitor residence time , 2015, Nature reviews. Drug discovery.

[60]  D. Rauh,et al.  Structure-based design and synthesis of covalent-reversible inhibitors to overcome drug resistance in EGFR. , 2015, Bioorganic & medicinal chemistry.

[61]  J. M. Bradshaw,et al.  Prolonged and tunable residence time using reversible covalent kinase inhibitors , 2015, Nature chemical biology.

[62]  George Papadatos,et al.  ChEMBL web services: streamlining access to drug discovery data and utilities , 2015, Nucleic Acids Res..

[63]  Pengyu Y. Ren,et al.  Reversible Covalent Inhibition of eEF‐2K by Carbonitriles , 2014, Chembiochem : a European journal of chemical biology.

[64]  Nir London,et al.  Covalent Docking of Large Libraries for the Discovery of Chemical Probes , 2014, Nature chemical biology.

[65]  J. Taunton,et al.  Design of Reversible, Cysteine-Targeted Michael Acceptors Guided by Kinetic and Computational Analysis , 2014, Journal of the American Chemical Society.

[66]  Tove Tuntland,et al.  Implementation of pharmacokinetic and pharmacodynamic strategies in early research phases of drug discovery and development at Novartis Institute of Biomedical Research , 2014, Front. Pharmacol..

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

[68]  P. Kuzmič,et al.  Covalent EGFR inhibitor analysis reveals importance of reversible interactions to potency and mechanisms of drug resistance , 2013, Proceedings of the National Academy of Sciences.

[69]  S. Frye,et al.  Pseudo-cyclization through intramolecular hydrogen bond enables discovery of pyridine substituted pyrimidines as new Mer kinase inhibitors. , 2013, Journal of medicinal chemistry.

[70]  K. Kinzler,et al.  Cancer Genome Landscapes , 2013, Science.

[71]  Jack Taunton,et al.  Electrophilic fragment-based design of reversible covalent kinase inhibitors. , 2013, Journal of the American Chemical Society.

[72]  Nathanael S Gray,et al.  Developing irreversible inhibitors of the protein kinase cysteinome. , 2013, Chemistry & biology.

[73]  Pedro M. P. Gois,et al.  Iminoboronates: a new strategy for reversible protein modification. , 2012, Journal of the American Chemical Society.

[74]  Jack Taunton,et al.  Reversible targeting of noncatalytic cysteines with chemically tuned electrophiles , 2012, Nature chemical biology.

[75]  Adrian Whitty,et al.  The resurgence of covalent drugs , 2011, Nature Reviews Drug Discovery.

[76]  F. Fleming,et al.  Nitrile-containing pharmaceuticals: efficacious roles of the nitrile pharmacophore. , 2010, Journal of medicinal chemistry.

[77]  B. Brandhuber,et al.  Crystal Structure of Human AKT1 with an Allosteric Inhibitor Reveals a New Mode of Kinase Inhibition , 2010, PloS one.

[78]  David Baker,et al.  Quantitative reactivity profiling predicts functional cysteines in proteomes , 2010, Nature.

[79]  Douglas H. Thamm,et al.  The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy , 2010, Proceedings of the National Academy of Sciences.

[80]  P. Majumder,et al.  MK-2206, an Allosteric Akt Inhibitor, Enhances Antitumor Efficacy by Standard Chemotherapeutic Agents or Molecular Targeted Drugs In vitro and In vivo , 2010, Molecular Cancer Therapeutics.

[81]  Scott B Ficarro,et al.  A structure-guided approach to creating covalent FGFR inhibitors. , 2010, Chemistry & biology.

[82]  John T. Powers,et al.  Targeting Bcr–Abl by combining allosteric with ATP-binding-site inhibitors , 2010, Nature.

[83]  John R. Engen,et al.  Novel mutant-selective EGFR kinase inhibitors against EGFR T790M , 2009, Nature.

[84]  Adele Di Matteo,et al.  Structural basis for the binding of the anticancer compound 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol to human glutathione s-transferases. , 2009, Cancer research.

[85]  Andrew G. Leach,et al.  Beyond picomolar affinities: quantitative aspects of noncovalent and covalent binding of drugs to proteins. , 2009, Journal of medicinal chemistry.

[86]  R. Copeland,et al.  Residence time of receptor-ligand complexes and its effect on biological function. , 2008, Biochemistry.

[87]  F. Sicheri Faculty Opinions recommendation of Inhibition of the EGF receptor by binding of MIG6 to an activating kinase domain interface. , 2007 .

[88]  T. Zhou,et al.  Crystal Structure of the T315I Mutant of Abl Kinase , 2007, Chemical biology & drug design.

[89]  R. Copeland,et al.  Drug–target residence time and its implications for lead optimization , 2007, Nature Reviews Drug Discovery.

[90]  Jean-François Truchon,et al.  A generally applicable method for assessing the electrophilicity and reactivity of diverse nitrile-containing compounds. , 2007, Bioorganic & medicinal chemistry letters.

[91]  Jeffrey Jie-Lou Liao,et al.  Molecular recognition of protein kinase binding pockets for design of potent and selective kinase inhibitors. , 2007, Journal of medicinal chemistry.

[92]  Michael S. Cohen,et al.  Structural Bioinformatics-Based Design of Selective, Irreversible Kinase Inhibitors , 2005, Science.

[93]  A. Siraki,et al.  Aldehyde Sources, Metabolism, Molecular Toxicity Mechanisms, and Possible Effects on Human Health , 2005, Critical reviews in toxicology.

[94]  O. Silvennoinen,et al.  The Janus kinases (Jaks) , 2004, Genome Biology.

[95]  T. Hunter,et al.  The Protein Kinase Complement of the Human Genome , 2002, Science.

[96]  J. Powers,et al.  Irreversible inhibitors of serine, cysteine, and threonine proteases. , 2002, Chemical reviews.

[97]  P. Cohen,et al.  The origins of protein phosphorylation , 2002, Nature Cell Biology.

[98]  J. Adams,et al.  Kinetic and catalytic mechanisms of protein kinases. , 2001, Chemical reviews.

[99]  D. Fairlie,et al.  Protease inhibitors: current status and future prospects. , 2000, Journal of medicinal chemistry.

[100]  J. Gierse,et al.  Kinetic basis for selective inhibition of cyclo-oxygenases. , 1999, The Biochemical journal.

[101]  Francois Terrier,et al.  Rate and equilibrium studies in Jackson-Meisenheimer complexes , 1982 .

[102]  H. L. Holmes,et al.  Equilibrium reactions of n-butanethiol with some conjugated heteroenoid compounds , 1968 .

[103]  N. Ormaechea-Pérez,et al.  Adverse skin effects of imatinib, a tyrosine kinase inhibitor. , 2014, Actas dermo-sifiliograficas.

[104]  N. Schork,et al.  Kinase mutations in human disease: interpreting genotype–phenotype relationships , 2010, Nature Reviews Genetics.

[105]  N. Gray,et al.  Targeting cancer with small molecule kinase inhibitors , 2009, Nature Reviews Cancer.