Targeted Covalent Inhibitors for Drug Design.

In contrast to the traditional mechanism of drug action that relies on the reversible, noncovalent interaction of a ligand with its biological target, a targeted covalent inhibitor (TCI) is designed such that the initial, reversible association is followed by the formation of a covalent bond between an electrophile on the ligand and a nucleophilic center in the protein. Although this approach offers a variety of potential benefits (high potency and extended duration of action), concerns over the possible toxicological consequences of protein haptenization have hindered the development of the TCI concept. Recently, approaches to mitigate the risk of serious adverse reactions to this new class of agent have emerged, thus stimulating interest in the field and leading to authorization of the first cadre of TCIs to be marketed. The covalent inhibitor approach is rapidly gaining acceptance as a valuable tool in drug discovery, and is poised to make a major impact on the design of enzyme inhibitors and receptor modulators.

[1]  Declan Butler,et al.  Scientists in the dark after French clinical trial proves fatal , 2016, Nature.

[2]  Aubry K. Miller,et al.  Spiroepoxytriazoles Are Fumagillin-like Irreversible Inhibitors of MetAP2 with Potent Cellular Activity. , 2016, ACS chemical biology.

[3]  W. Humphreys,et al.  Label-Free Bottom-Up Proteomic Workflow for Simultaneously Assessing the Target Specificity of Covalent Drug Candidates and Their Off-Target Reactivity to Selected Proteins. , 2016, Chemical research in toxicology.

[4]  S. Vasudevan,et al.  Identification of covalent active site inhibitors of dengue virus protease , 2015, Drug design, development and therapy.

[5]  G. Kocic,et al.  An overview of recent dipeptidyl peptidase-IV inhibitors: linking their structure and physico-chemical properties with sar, pharmacokinetics and toxicity. , 2015, Current topics in medicinal chemistry.

[6]  K. Geoghegan,et al.  A tag-free collisionally induced fragmentation approach to detect drug-adducted proteins by mass spectrometry. , 2015, Rapid communications in mass spectrometry : RCM.

[7]  J. McCarter,et al.  Systematic Study of the Glutathione (GSH) Reactivity of N-Arylacrylamides: 1. Effects of Aryl Substitution. , 2015, Journal of medicinal chemistry.

[8]  A. Vojtek,et al.  Targeted inhibition of the Shroom3–Rho kinase protein–protein interaction circumvents Nogo66 to promote axon outgrowth , 2015, BMC Neuroscience.

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

[10]  M. Safo,et al.  Identification of a novel class of covalent modifiers of hemoglobin as potential antisickling agents. , 2015, Organic & biomolecular chemistry.

[11]  P. Jänne,et al.  EGFR Mutations and Resistance to Irreversible Pyrimidine-Based EGFR Inhibitors , 2015, Clinical Cancer Research.

[12]  S. Sieber,et al.  A mass spectrometry platform for a streamlined investigation of proteasome integrity, posttranslational modifications, and inhibitor binding. , 2015, Chemistry & biology.

[13]  Y. Shibata,et al.  The Role of Extrahepatic Metabolism in the Pharmacokinetics of the Targeted Covalent Inhibitors Afatinib, Ibrutinib, and Neratinib , 2015, Drug Metabolism and Disposition.

[14]  L. Leclercq,et al.  Absorption, Metabolism, and Excretion of Oral 14C Radiolabeled Ibrutinib: An Open-Label, Phase I, Single-Dose Study in Healthy Men , 2015, Drug Metabolism and Disposition.

[15]  Ruth Nussinov,et al.  The design of covalent allosteric drugs. , 2015, Annual review of pharmacology and toxicology.

[16]  T. Baillie The contributions of Sidney D. Nelson to drug metabolism research , 2015, Drug metabolism reviews.

[17]  Elisabetta Marini,et al.  Electrophilic warhead-based design of compounds preventing NLRP3 inflammasome-dependent pyroptosis. , 2014, Journal of medicinal chemistry.

[18]  Wei Huang,et al.  A chemical tuned strategy to develop novel irreversible EGFR-TK inhibitors with improved safety and pharmacokinetic profiles. , 2014, Journal of medicinal chemistry.

[19]  G. S. Walker,et al.  Chemical and computational methods for the characterization of covalent reactive groups for the prospective design of irreversible inhibitors. , 2014, Journal of medicinal chemistry.

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

[21]  C. Klein,et al.  Promiscuity and selectivity in covalent enzyme inhibition: a systematic study of electrophilic fragments. , 2014, Journal of medicinal chemistry.

[22]  E. Novellino,et al.  Discovery of covalent inhibitors of glyceraldehyde-3-phosphate dehydrogenase, a target for the treatment of malaria. , 2014, Journal of medicinal chemistry.

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

[24]  Benjamin F. Cravatt,et al.  A roadmap to evaluate the proteome-wide selectivity of covalent kinase inhibitors , 2014, Nature chemical biology.

[25]  M. Bogyo,et al.  Activity-based profiling of proteases. , 2014, Annual review of biochemistry.

[26]  L. Xing,et al.  Bruton's TK inhibitors: structural insights and evolution of clinical candidates. , 2014, Future medicinal chemistry.

[27]  A. Kulkarni,et al.  A proposed screening paradigm for discovery of covalent inhibitor drugs. , 2014, Drug metabolism letters.

[28]  Stefan G. Kathman,et al.  A Fragment-Based Method to Discover Irreversible Covalent Inhibitors of Cysteine Proteases , 2014, Journal of medicinal chemistry.

[29]  A. Cavalli,et al.  Predicting the reactivity of nitrile-carrying compounds with cysteine: a combined computational and experimental study. , 2014, ACS medicinal chemistry letters.

[30]  S. Ševčíková,et al.  Proteasome inhibitors – molecular basis and current perspectives in multiple myeloma , 2014, Journal of cellular and molecular medicine.

[31]  J. Rudolph,et al.  Selektive Inhibierung von mutiertem Ras‐Protein durch kovalent bindende Liganden , 2014 .

[32]  D. Stokoe,et al.  Selective inhibition of mutant Ras protein through covalent binding. , 2014, Angewandte Chemie.

[33]  D. Boger,et al.  Design, Synthesis, and Characterization of α-Ketoheterocycles That Additionally Target the Cytosolic Port Cys269 of Fatty Acid Amide Hydrolase , 2014, Journal of medicinal chemistry.

[34]  J. Taunton,et al.  Targeting protein kinases with selective and semipromiscuous covalent inhibitors. , 2014, Methods in enzymology.

[35]  Robert Mah,et al.  Drug discovery considerations in the development of covalent inhibitors. , 2014, Bioorganic & medicinal chemistry letters.

[36]  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.

[37]  J. Gesell,et al.  Discovery of an irreversible HCV NS5B polymerase inhibitor. , 2013, Bioorganic & medicinal chemistry letters.

[38]  R. Obach,et al.  Benchmarking in vitro covalent binding burden as a tool to assess potential toxicity caused by nonspecific covalent binding of covalent drugs. , 2013, Chemical research in toxicology.

[39]  Matthew Grist,et al.  Structure- and reactivity-based development of covalent inhibitors of the activating and gatekeeper mutant forms of the epidermal growth factor receptor (EGFR). , 2013, Journal of medicinal chemistry.

[40]  Katharine Sanderson,et al.  Irreversible kinase inhibitors gain traction , 2013, Nature Reviews Drug Discovery.

[41]  J. Callahan,et al.  Keap calm, and carry on covalently. , 2013, Journal of medicinal chemistry.

[42]  Weida Tong,et al.  High lipophilicity and high daily dose of oral medications are associated with significant risk for drug‐induced liver injury , 2013, Hepatology.

[43]  A. Look,et al.  Discovery of a selective irreversible BMX inhibitor for prostate cancer. , 2013, ACS chemical biology.

[44]  D. Boger,et al.  Rational design of fatty acid amide hydrolase inhibitors that act by covalently bonding to two active site residues. , 2013, Journal of the American Chemical Society.

[45]  D. Naisbitt,et al.  Idiosyncratic Adverse Drug Reactions: Current Concepts , 2013, Pharmacological Reviews.

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

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

[48]  Richard J. Marhöfer,et al.  Docking-based virtual screening of covalently binding ligands: an orthogonal lead discovery approach. , 2013, Journal of medicinal chemistry.

[49]  Juswinder Singh,et al.  Discovery of a potent and isoform-selective targeted covalent inhibitor of the lipid kinase PI3Kα. , 2013, Journal of medicinal chemistry.

[50]  T. Monks,et al.  Reactive Intermediates , 2013, Toxicologic pathology.

[51]  A. Kalgutkar Reactive Drug Metabolites , 2012 .

[52]  J. Taunton,et al.  Selective targeting of distinct active site nucleophiles by irreversible SRC-family kinase inhibitors. , 2012, Journal of the American Chemical Society.

[53]  David R. Anderson,et al.  Covalent inhibitors of interleukin-2 inducible T cell kinase (itk) with nanomolar potency in a whole-blood assay. , 2012, Journal of medicinal chemistry.

[54]  M. Johansson Reversible Michael additions: covalent inhibitors and prodrugs. , 2012, Mini reviews in medicinal chemistry.

[55]  M. Soellner,et al.  Irreversible inhibitors of c-Src kinase that target a nonconserved cysteine. , 2012, ACS chemical biology.

[56]  A. Kalgutkar,et al.  Drug discovery for a new generation of covalent drugs , 2012, Expert opinion on drug discovery.

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

[58]  Allard Kaptein,et al.  Irreversible protein kinase inhibitors: balancing the benefits and risks. , 2012, Journal of medicinal chemistry.

[59]  H. Overkleeft,et al.  Activity-based protein profiling: an enabling technology in chemical biology research. , 2012, Current opinion in chemical biology.

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

[61]  Raymond E Moellering,et al.  How chemoproteomics can enable drug discovery and development. , 2012, Chemistry & biology.

[62]  B. Kuster,et al.  Mass spectrometry-based proteomics in preclinical drug discovery. , 2012, Chemistry & biology.

[63]  Taebo Sim,et al.  Discovery of potent and selective covalent inhibitors of JNK. , 2012, Chemistry & biology.

[64]  A. Gilbert,et al.  Targeted Covalent Enzyme Inhibitors , 2012 .

[65]  B. Cravatt,et al.  The pharmacological landscape and therapeutic potential of serine hydrolases , 2012, Nature Reviews Drug Discovery.

[66]  T. Ebner,et al.  Afatinib pharmacokinetics and metabolism after oral administration to healthy male volunteers , 2012, Cancer Chemotherapy and Pharmacology.

[67]  A. Ardizzoni,et al.  Epidermal growth factor receptor irreversible inhibitors: chemical exploration of the cysteine-trap portion. , 2011, Mini reviews in medicinal chemistry.

[68]  A. Stepan,et al.  Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: a perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. , 2011, Chemical research in toxicology.

[69]  G. Bottegoni,et al.  Irreversible protein kinase inhibitors. , 2011, Current medicinal chemistry.

[70]  J. Taunton,et al.  Irreversible Nek2 kinase inhibitors with cellular activity. , 2011, Journal of medicinal chemistry.

[71]  F. J. Luque,et al.  Switching reversibility to irreversibility in glycogen synthase kinase 3 inhibitors: clues for specific design of new compounds. , 2011, Journal of medicinal chemistry.

[72]  B. Hug,et al.  Pharmacokinetics of oral neratinib during co-administration of ketoconazole in healthy subjects. , 2011, British journal of clinical pharmacology.

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

[74]  D. Moras,et al.  Cysteine mapping in conformationally distinct kinase nucleotide binding sites: application to the design of selective covalent inhibitors. , 2011, Journal of medicinal chemistry.

[75]  Juswinder Singh,et al.  Selective irreversible inhibition of a protease by targeting a noncatalytic cysteine. , 2011, Nature chemical biology.

[76]  J. Scatina,et al.  Reversible covalent binding of neratinib to human serum albumin in vitro. , 2010, Drug metabolism letters.

[77]  Juswinder Singh,et al.  Targeted covalent drugs of the kinase family. , 2010, Current opinion in chemical biology.

[78]  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.

[79]  R. Talaat,et al.  Characterization of HKI-272 Covalent Binding to Human Serum Albumin , 2010, Drug Metabolism and Disposition.

[80]  B. Cravatt,et al.  Strategies for discovering and derisking covalent, irreversible enzyme inhibitors. , 2010, Future medicinal chemistry.

[81]  R. Petter,et al.  Acylating drugs: redesigning natural covalent inhibitors. , 2010, Current opinion in chemical biology.

[82]  John J. M. Wiener,et al.  Recent advances in the design of cathepsin S inhibitors. , 2010, Current topics in medicinal chemistry.

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

[84]  M. Wenk,et al.  Activity-based proteome profiling of potential cellular targets of Orlistat--an FDA-approved drug with anti-tumor activities. , 2010, Journal of the American Chemical Society.

[85]  G. Schüürmann,et al.  Kinetic glutathione chemoassay to quantify thiol reactivity of organic electrophiles--application to alpha,beta-unsaturated ketones, acrylates, and propiolates. , 2009, Chemical research in toxicology.

[86]  P. Macfaul,et al.  A simple in vitro assay for assessing the reactivity of nitrile containing compounds. , 2009, Bioorganic & medicinal chemistry letters.

[87]  M. Potashman,et al.  Covalent modifiers: an orthogonal approach to drug design. , 2009, Journal of medicinal chemistry.

[88]  D. Mansuy,et al.  Metabolic oxidative cleavage of thioesters: evidence for the formation of sulfenic acid intermediates in the bioactivation of the antithrombotic prodrugs ticlopidine and clopidogrel. , 2009, Chemical research in toxicology.

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

[90]  D. Liebler,et al.  Reversibility of covalent electrophile-protein adducts and chemical toxicity. , 2008, Chemical research in toxicology.

[91]  J. Hughes,et al.  Physiochemical drug properties associated with in vivo toxicological outcomes. , 2008, Bioorganic & medicinal chemistry letters.

[92]  Chandan Saha,et al.  Relationship between daily dose of oral medications and idiosyncratic drug‐induced liver injury: Search for signals , 2008, Hepatology.

[93]  U. McDermott,et al.  The T790M “gatekeeper” mutation in EGFR mediates resistance to low concentrations of an irreversible EGFR inhibitor , 2008, Molecular Cancer Therapeutics.

[94]  M. Meyerson,et al.  The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP , 2008, Proceedings of the National Academy of Sciences.

[95]  J. Moses,et al.  The growing applications of click chemistry. , 2007, Chemical Society reviews.

[96]  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.

[97]  S. Demo,et al.  Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. , 2005, Blood.

[98]  John Cl Erve,et al.  Chemical toxicology: reactive intermediates and their role in pharmacology and toxicology , 2006, Expert opinion on drug metabolism & toxicology.

[99]  T. Baillie,et al.  Future of toxicology-metabolic activation and drug design: challenges and opportunities in chemical toxicology. , 2006, Chemical research in toxicology.

[100]  B. Cravatt,et al.  Protein-reactive natural products. , 2005, Angewandte Chemie.

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

[102]  Wei Duan,et al.  Drug Bioactivation Covalent Binding to Target Proteins and Toxicity Relevance , 2005, Drug metabolism reviews.

[103]  G. Sachs,et al.  Chemistry of covalent inhibition of the gastric (H+, K+)-ATPase by proton pump inhibitors. , 2004, Journal of the American Chemical Society.

[104]  T. Baillie,et al.  Drug-protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development. , 2004, Chemical research in toxicology.

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

[106]  M. Pirmohamed,et al.  Are chemically reactive metabolites responsible for adverse reactions to drugs? , 2002, Current drug metabolism.

[107]  W. Denny Irreversible inhibitors of the erbB family of protein tyrosine kinases. , 2002, Pharmacology & therapeutics.

[108]  F. Guengerich,et al.  Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity. , 2001, Chemical research in toxicology.

[109]  O. H. Chan,et al.  Tyrosine kinase inhibitors. 17. Irreversible inhibitors of the epidermal growth factor receptor: 4-(phenylamino)quinazoline- and 4-(phenylamino)pyrido[3,2-d]pyrimidine-6-acrylamides bearing additional solubilizing functions. , 2000, Journal of medicinal chemistry.

[110]  J. Way,et al.  Covalent modification as a strategy to block protein-protein interactions with small-molecule drugs. , 2000, Current opinion in chemical biology.

[111]  W. Denny,et al.  Specific, irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class of tyrosine kinase inhibitor. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[112]  A. Burlingame,et al.  Identification of the Hepatic Protein Targets of Reactive Metabolites of Acetaminophen in Vivo in Mice Using Two-dimensional Gel Electrophoresis and Mass Spectrometry* , 1998, The Journal of Biological Chemistry.

[113]  R. Guerciolini Mode of action of orlistat. , 1997, International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity.

[114]  A Whitty,et al.  Structure-based design of a potent, selective, and irreversible inhibitor of the catalytic domain of the erbB receptor subfamily of protein tyrosine kinases. , 1997, Journal of medicinal chemistry.

[115]  T. Baillie,et al.  Glutathione: a vehicle for the transport of chemically reactive metabolites in vivo , 1991 .

[116]  J. Groten,et al.  Reversible interaction of a reactive intermediate derived from furazolidone with glutathione and protein. , 1988, Toxicology and applied pharmacology.

[117]  T. Baillie,et al.  Identification of the major covalent adduct formed in vitro and in vivo between acetaminophen and mouse liver proteins. , 1985, Molecular pharmacology.

[118]  S. Nelson,et al.  Synthesis, decomposition kinetics, and preliminary toxicological studies of pure N-acetyl-p-benzoquinone imine, a proposed toxic metabolite of acetaminophen. , 1982, Journal of medicinal chemistry.