39
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Quantitative, Wide-Spectrum Kinase Profiling in Live Cells for Assessing the Effect of Cellular ATP on Target Engagement

      research-article
      1 , 2 , 1 , 1 , 1 , 1 , 1 , 1 , 2 , 1 , 1 , 1 , 1 , 1 , 3 , 4 , 5 , 4 , 6 , 7 , 8 , 9 , 3 , 6 , 7 , 3 , 4 , 5 , 5 , 2 , 1 , 1 , 1 , 10 ,
      Cell Chemical Biology
      Cell Press
      target engagement, BRET, NanoBRET, NanoLuc, kinase, profiling, selectivity, crizotinib, dasatinib, ATP

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          For kinase inhibitors, intracellular target selectivity is fundamental to pharmacological mechanism. Although a number of acellular techniques have been developed to measure kinase binding or enzymatic inhibition, such approaches can fail to accurately predict engagement in cells. Here we report the application of an energy transfer technique that enabled the first broad-spectrum, equilibrium-based approach to quantitatively profile target occupancy and compound affinity in live cells. Using this method, we performed a selectivity profiling for clinically relevant kinase inhibitors against 178 full-length kinases, and a mechanistic interrogation of the potency offsets observed between cellular and biochemical analysis. For the multikinase inhibitor crizotinib, our approach accurately predicted cellular potency and revealed improved target selectivity compared with biochemical measurements. Due to cellular ATP, a number of putative crizotinib targets are unexpectedly disengaged in live cells at a clinically relevant drug dose.

          Graphical Abstract

          Highlights

          • The approach enables quantitative profiling of 178 full-length kinases

          • Compared with biochemical approaches, this is a better predictor of cellular potency

          • An unexpected intracellular selectivity is observed for certain kinase inhibitors

          • A mechanistic analysis of ATP interference on target engagement is performed

          Abstract

          Vasta et al. describe a broad-spectrum approach (BRET) to quantitatively measure target engagement for kinases inside live cells. Compared with biochemical measurements, the analysis revealed an improved intracellular selectivity profile for clinically relevant kinase inhibitors. This serves as a mechanistic tool to determine the effect of cellular ATP on engagement potency.

          Related collections

          Most cited references32

          • Record: found
          • Abstract: found
          • Article: not found

          Tracking cancer drugs in living cells by thermal profiling of the proteome.

          The thermal stability of proteins can be used to assess ligand binding in living cells. We have generalized this concept by determining the thermal profiles of more than 7000 proteins in human cells by means of mass spectrometry. Monitoring the effects of small-molecule ligands on the profiles delineated more than 50 targets for the kinase inhibitor staurosporine. We identified the heme biosynthesis enzyme ferrochelatase as a target of kinase inhibitors and suggest that its inhibition causes the phototoxicity observed with vemurafenib and alectinib. Thermal shifts were also observed for downstream effectors of drug treatment. In live cells, dasatinib induced shifts in BCR-ABL pathway proteins, including CRK/CRKL. Thermal proteome profiling provides an unbiased measure of drug-target engagement and facilitates identification of markers for drug efficacy and toxicity. Copyright © 2014, American Association for the Advancement of Science.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Structure based drug design of crizotinib (PF-02341066), a potent and selective dual inhibitor of mesenchymal-epithelial transition factor (c-MET) kinase and anaplastic lymphoma kinase (ALK).

            Because of the critical roles of aberrant signaling in cancer, both c-MET and ALK receptor tyrosine kinases are attractive oncology targets for therapeutic intervention. The cocrystal structure of 3 (PHA-665752), bound to c-MET kinase domain, revealed a novel ATP site environment, which served as the target to guide parallel, multiattribute drug design. A novel 2-amino-5-aryl-3-benzyloxypyridine series was created to more effectively make the key interactions achieved with 3. In the novel series, the 2-aminopyridine core allowed a 3-benzyloxy group to reach into the same pocket as the 2,6-dichlorophenyl group of 3 via a more direct vector and thus with a better ligand efficiency (LE). Further optimization of the lead series generated the clinical candidate crizotinib (PF-02341066), which demonstrated potent in vitro and in vivo c-MET kinase and ALK inhibition, effective tumor growth inhibition, and good pharmaceutical properties.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Ligand binding assays at equilibrium: validation and interpretation.

              The focus of this review paper is factors affecting data interpretation in ligand binding assays under equilibrium conditions. Protocols for determining K(d) (the equilibrium dissociation constant) and K(dA) (the equilibrium inhibitor constant) for receptor ligands are discussed. The basic theory describing the interaction of a radiotracer and an unlabelled competitor ligand with a receptor is developed. Inappropriate experimental design may result in ligand depletion and non-attainment of equilibrium, distorting the calculation of K(d) and K(dA) . Strategies, both theoretical and practical, will be given to avoid and correct such errors, thus leading to the determination of reliable values for these constants. In determining K(dA) from competition binding studies, two additional concepts are discussed. First, the necessity to measure an adequate specific binding signal from the bound radiotracer ligand limits the range of affinity constants that can be measured: a particular set of assay conditions may lead to an upper limit on the apparent affinity of unlabelled ligands. Second, an extension of the basic assay methodology can indicate whether the interaction between the tracer and a test ligand is mediated by a competitive or an allosteric mechanism. Finally, the review ends with a discussion of two factors that are often overlooked: buffer composition and the temperature at which the assay is conducted, and the impact these can have on affinity measurements and the understanding of drug interactions. © 2010 The Authors. Journal compilation © 2010 The British Pharmacological Society.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Chem Biol
                Cell Chem Biol
                Cell Chemical Biology
                Cell Press
                2451-9456
                2451-9448
                15 February 2018
                15 February 2018
                : 25
                : 2
                : 206-214.e11
                Affiliations
                [1 ]Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
                [2 ]Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, CA 93401, USA
                [3 ]Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
                [4 ]Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
                [5 ]Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
                [6 ]Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK
                [7 ]Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
                [8 ]State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
                [9 ]School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
                Author notes
                []Corresponding author matt.robers@ 123456promega.com
                [10]

                Lead Contact

                Article
                S2451-9456(17)30391-4
                10.1016/j.chembiol.2017.10.010
                5814754
                29174542
                bb8a7265-c69f-4dbb-ab92-90335d0a8897
                © 2017 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 21 June 2017
                : 24 August 2017
                : 24 October 2017
                Categories
                Article

                target engagement,bret,nanobret,nanoluc,kinase,profiling,selectivity,crizotinib,dasatinib,atp

                Comments

                Comment on this article