521
Views
8
CrossRef citations to date
0
Altmetric
Reviews

Non-stoichiometric inhibition in integrated lead finding – a literature review

Pages 149-162 | Received 09 Oct 2015, Accepted 03 Dec 2015, Published online: 31 Dec 2015

References

  • Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
  • Macarron R, Banks MN, Bojanic D, et al. Impact of high-throughput screening in biomedical research. Nat Rev Drug Discov. 2011;10(3):188–195.
  • Wigglesworth MJ, Murray DC, Blackett CJ, et al. Increasing the delivery of next generation therapeutics from high throughput screening libraries. Curr Opin Chem Biol. 2015;26:104–110.
  • Hasson SA, Inglese J. Innovation in academic chemical screening: filling the gaps in chemical biology. Curr Opin Chem Biol. 2013;17(3):329–338.
  • Baell JB. Screening-based translation of public research encounters painful problems. ACS Med Chem Lett. 2015;6(3):229–234.
  • Whitty A. Growing PAINS in academic drug discovery. Future Med Chem. 2011;3(7):797–801.
  • Coan KE, Ottl J, Klumpp M. Non-stoichiometric inhibition in biochemical high-throughput screening. Expert Opin Drug Discov. 2011;6(4):405–417.
  • Dahlin JL, Walters MA. How to triage PAINS-full research. Assay Drug Dev Technol. 2015. DOI:10.1089/adt.2015.674.
  • Sliwoski G, Kothiwale S, Meiler J, et al. Computational methods in drug discovery. Pharmacol Rev. 2014;66(1):334–395.
  • Dahlin JL, Walters MA. The essential roles of chemistry in high-throughput screening triage. Future Med Chem. 2014;6(11):1265–1290.
  • Baell J, Walters MA. Chemistry: chemical con artists foil drug discovery. Nature. 2014;513(7519):481–483.

• Easy-to-read, sometimes humorous and provocative, introduction to the topic.

  • Crisman TJ, Parker CN, Jenkins JL, et al. Understanding false positives in reporter gene assays: in silico chemogenomics approaches to prioritize cell-based HTS data. J Chem Inf Model. 2007;47(4):1319–1327.
  • Nissink JWM, Blackburn S. Quantification of frequent-hitter behavior based on historical high-throughput screening data. Future Med Chem. 2014;6(10):1113–1126.
  • Anighoro A, Bajorath J, Rastelli G. Polypharmacology: challenges and opportunities in drug discovery. J Med Chem. 2014;57(19):7874–7887.
  • Ciceri P, Muller S, O’Mahony A, et al. Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat Chem Biol. 2014;10(4):305–312.
  • Garuti L, Roberti M, Bottegoni G. Multi-kinase inhibitors. Curr Med Chem. 2015;22(6):695–712.
  • Zhang JH, Chung TD, Oldenburg KR. Confirmation of primary active substances from high throughput screening of chemical and biological populations: a statistical approach and practical considerations. J Comb Chem. 2000;2(3):258–265.
  • Wu Z, Liu D, Sui Y. Quantitative assessment of hit detection and confirmation in single and duplicate high-throughput screenings. J Biomol Screen. 2008;13(2):159–167.
  • Huth JR, Mendoza R, Olejniczak ET, et al. ALARM NMR: a rapid and robust experimental method to detect reactive false positives in biochemical screens. J Am Chem Soc. 2005;127(1):217–224.
  • Sink R, Gobec S, Pecar S, et al. False positives in the early stages of drug discovery. Curr Med Chem. 2010;17(34):4231–4255.
  • Hermann JC, Chen Y, Wartchow C, et al. Metal impurities cause false positives in high-throughput screening campaigns. ACS Med Chem Lett. 2013;4(2):197–200.
  • Adam GC, Meng J, Rizzo JM, et al. Use of high-throughput mass spectrometry to reduce false positives in protease uHTS screens. J Biomol Screen. 2015;20(2):212–222.
  • Wigle TJ, Swinger KK, Campbell JE, et al. A high-throughput mass spectrometry assay coupled with redox activity testing reduces artifacts and false positives in lysine demethylase screening. J Biomol Screen. 2015;20(6):810–820.
  • Mansson C, Arosio P, Hussein R, et al. Interaction of the molecular chaperone DNAJB6 with growing amyloid-beta 42 (Abeta42) aggregates leads to sub-stoichiometric inhibition of amyloid formation. J Biol Chem. 2014;289(45):31066–31076.
  • Lulf S, Matz J, Rouyez MC, et al. Structural basis for the inhibition of HIV-1 Nef by a high-affinity binding single-domain antibody. Retrovirology. 2014;11:24.
  • Habig M, Blechschmidt A, Dressler S, et al. Efficient elimination of nonstoichiometric enzyme inhibitors from HTS hit lists. J Biomol Screen. 2009;14(6):679–689.

• Illustrates how the apparent potency of non-stoichiometric inhibitors shifts upon varying enzyme concentration

  • Henrich CJ, Beutler JA. Matching the power of high throughput screening to the chemical diversity of natural products. Nat Prod Rep. 2013;30(10):1284–1298.
  • Schorpp K, Rothenaigner I, Salmina E, et al. Identification of small-molecule frequent hitters from AlphaScreen high-throughput screens. J Biomol Screen. 2013;19(5):715–726.
  • Klink TA, Staeben M, Twesten K, et al. Development and validation of a generic fluorescent methyltransferase activity assay based on the transcreener AMP/GMP assay. J Biomol Screen. 2012;17(1):59–70.
  • Dahlin JL, Nissink JW, Strasser JM, et al. PAINS in the assay: chemical mechanisms of assay interference and promiscuous enzymatic inhibition observed during a sulfhydryl-scavenging HTS. J Med Chem. 2015;58(5):2091–2113.

•• Contains numerous examples of various counterscreens to eliminate different types of non-stoichiometric inhibitors.

  • Simard JR, Plant M, Emkey R, et al. Development and implementation of a high-throughput AlphaLISA assay for identifying inhibitors of EZH2 methyltransferase. Assay Drug Dev Technol. 2013;11(3):152–162.
  • Brimacombe KR, Walsh MJ, Liu L, et al. Identification of ML251, a potent inhibitor of T. brucei and T. cruzi phosphofructokinase. ACS Med Chem Lett. 2014;5(1):12–17.
  • Nguyen GH, Dexheimer TS, Rosenthal AS, et al. A small molecule inhibitor of the BLM helicase modulates chromosome stability in human cells. Chem Biol. 2013;20(1):55–62.
  • Han Y, Lyman K, Clutter M, et al. Identification of small-molecule inhibitors of hyperpolarization-activated cyclic nucleotide-gated channels. J Biomol Screen. 2015. DOI:10.1177/1087057115589590.
  • Meng J, Lai MT, Munshi V, et al. Screening of HIV-1 protease using a combination of an ultra-high-throughput fluorescent-based assay and RapidFire mass spectrometry. J Biomol Screen. 2015;20(5):606–615.
  • Kashem MA, Wa C, Wolak JP, et al. A high-throughput scintillation proximity assay for sphingosine-1-phosphate lyase. Assay Drug Dev Technol. 2014;12(5):293–302.
  • Gurard-Levin ZA, Scholle MD, Eisenberg AH, et al. High-throughput screening of small molecule libraries using SAMDI mass spectrometry. ACS Comb Sci. 2011;13(4):347–350.
  • Rathore R, Corr JJ, Lebre DT, et al. Extending matrix-assisted laser desorption/ionization triple quadrupole mass spectrometry enzyme screening assays to targets with small molecule substrates. Rapid Commun Mass Spectrom. 2009;23(20):3293–3300.
  • Thorne N, Shen M, Lea WA, et al. Firefly luciferase in chemical biology: a compendium of inhibitors, mechanistic evaluation of chemotypes, and suggested use as a reporter. Chem Biol. 2012;19(8):1060–1072.
  • Shum D, Bhinder B, Radu C, et al. An image-based biosensor assay strategy to screen for modulators of the microRNA 21 biogenesis pathway. Comb Chem High Throughput Screen. 2012;15(7):529–541.
  • Ho PI, Yue K, Pandey P, et al. Reporter enzyme inhibitor study to aid assembly of orthogonal reporter gene assays. ACS Chem Biol. 2013;8(5):1009–1017.
  • Cheng KC, Inglese J. A coincidence reporter-gene system for high-throughput screening. Nat Methods. 2012;9(10):937.
  • Coan KE, Maltby DA, Burlingame AL, et al. Promiscuous aggregate-based inhibitors promote enzyme unfolding. J Med Chem. 2009;52(7):2067–2075.
  • Sassano MF, Doak AK, Roth BL, et al. Colloidal aggregation causes inhibition of G protein-coupled receptors. J Med Chem. 2013;56(6):2406–2414.
  • Coan KE, Shoichet BK. Stability and equilibria of promiscuous aggregates in high protein milieus. Mol Biosyst. 2007;3(3):208–213.
  • Wang J, Matayoshi E. Solubility at the molecular level: development of a critical aggregation concentration (CAC) assay for estimating compound monomer solubility. Pharm Res. 2012;29(7):1745–1754.
  • Redhead M, Satchell R, Morkunaite V, et al. A combinatorial biophysical approach; FTSA and SPR for identifying small molecule ligands and PAINs. Anal Biochem. 2015;479:63–73.

•• Analyzes the typical results measured in biophysical binding assays for stoichiometric and non-stoichiometric inhibitors. A valuable reference.

  • Zorn JA, Wille H, Wolan DW, et al. Self-assembling small molecules form nanofibrils that bind procaspase-3 to promote activation. J Am Chem Soc. 2011;133(49):19630–19633.
  • Julien O, Kampmann M, Bassik MC, et al. Unraveling the mechanism of cell death induced by chemical fibrils. Nat Chem Biol. 2014;10(11):969–976.
  • Owen SC, Doak AK, Wassam P, et al. Colloidal aggregation affects the efficacy of anticancer drugs in cell culture. ACS Chem Biol. 2012;7(8):1429–1435.
  • LaPlante SR, Carson R, Gillard J, et al. Compound aggregation in drug discovery: implementing a practical NMR assay for medicinal chemists. J Med Chem. 2013;56(12):5142–5150.
  • Dahlin JL, Nissink JW, Francis S, et al. Post-HTS case report and structural alert: promiscuous 4-aroyl-1,5-disubstituted-3-hydroxy-2H-pyrrol-2-one actives verified by ALARM NMR. Bioorg Med Chem Lett. 2015;25(21):4740–4752.
  • Kashyap S, Sandler J, Peters U, et al. Using ‘biased-privileged’ scaffolds to identify lysine methyltransferase inhibitors. Bioorg Med Chem. 2014;22(7):2253–2260.

• Demonstrates how the potency of a stoichiometric inhibitor is unaffected by enzyme concentration.

  • Owen SC, Doak AK, Ganesh AN, et al. Colloidal drug formulations can explain ‘bell-shaped’ concentration-response curves. ACS Chem Biol. 2014;9(3):777–784.
  • Rohrig UF, Majjigapu SR, Chambon M, et al. Detailed analysis and follow-up studies of a high-throughput screening for indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. Eur J Med Chem. 2014;84:284–301.
  • Dahlin JL, Sinville R, Solberg J, et al. A cell-free fluorometric high-throughput screen for inhibitors of Rtt109-catalyzed histone acetylation. PLoS One. 2013;8(11):e78877.
  • Jadhav A, Ferreira RS, Klumpp C, et al. Quantitative analyses of aggregation, autofluorescence, and reactivity artifacts in a screen for inhibitors of a thiol protease. J Med Chem. 2010;53(1):37–51.
  • Irwin JJ, Duan D, Torosyan H, et al. An aggregation advisor for ligand discovery. J Med Chem. 2015;58(17):7076–7087.
  • Bauer RA. Covalent inhibitors in drug discovery: from accidental discoveries to avoided liabilities and designed therapies. Drug Discov Today. 2015. DOI:10.1016/j.drudis.2015.05.005.
  • Singh J, Petter RC, Baillie TA, et al. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10(4):307–317.
  • Morisseau C, Sahdeo S, Cortopassi G, et al. Development of an HTS assay for EPHX2 phosphatase activity and screening of nontargeted libraries. Anal Biochem. 2013;434(1):105–111.
  • McCallum MM, Nandhikonda P, Temmer JJ, et al. High-throughput identification of promiscuous inhibitors from screening libraries with the use of a thiol-containing fluorescent probe. J Biomol Screen. 2013;18(6):705–713.
  • Bruno S, Pinto A, Paredi G, et al. Discovery of covalent inhibitors of glyceraldehyde-3-phosphate dehydrogenase, a target for the treatment of malaria. J Med Chem. 2014;57(17):7465–7471.
  • Mirkovic B, Sosic I, Gobec S, et al. Redox-based inactivation of cysteine cathepsins by compounds containing the 4-aminophenol moiety. PLoS One. 2011;6(11):e27197.
  • Johnston PA. Redox cycling compounds generate H2O2 in HTS buffers containing strong reducing reagents: real hits or promiscuous artifacts? Curr Opin Chem Biol. 2011;15(1):174–182.
  • Martin KR, Narang P, Medina-Franco JL, et al. Integrating virtual and biochemical screening for protein tyrosine phosphatase inhibitor discovery. Methods. 2014;65(2):219–228.
  • Johnston PA, Soares KM, Shinde SN, et al. Development of a 384-well colorimetric assay to quantify hydrogen peroxide generated by the redox cycling of compounds in the presence of reducing agents. Assay Drug Dev Technol. 2008;6(4):505–518.
  • Lor LA, Schneck J, McNulty DE, et al. A simple assay for detection of small-molecule redox activity. J Biomol Screen. 2007;12(6):881–890.
  • Stewart MD, Igumenova TI. Reactive cysteine in the structural Zn(2+) site of the C1B domain from PKCalpha. Biochemistry. 2012;51(37):7263–7277.
  • Bacchi A, Carcelli M, Compari C, et al. Investigating the role of metal chelation in HIV-1 integrase strand transfer inhibitors. J Med Chem. 2011;54(24):8407–8420.
  • Camp D, Davis RA, Evans-Illidge EA, et al. Guiding principles for natural product drug discovery. Future Med Chem. 2012;4(9):1067–1084.

• Outlines some pecularities of natural products that are critical when screening such samples.

  • Priyadarsini KI. Chemical and structural features influencing the biological activity of curcumin. Curr Pharm Des. 2013;19(11):2093–2100.
  • Tomasic T, Peterlin Masic L. Rhodanine as a scaffold in drug discovery: a critical review of its biological activities and mechanisms of target modulation. Expert Opin Drug Discov. 2012;7(7):549–560.
  • Beauchemin C, Moerke NJ, Faloon P, et al. Assay development and high-throughput screening for inhibitors of Kaposi’s sarcoma-associated herpesvirus N-terminal latency-associated nuclear antigen binding to nucleosomes. J Biomol Screen. 2014;19(6):947–958.
  • Roche O, Schneider P, Zuegge J, et al. Development of a virtual screening method for identification of ‘frequent hitters’ in compound libraries. J Med Chem. 2002;45(1):137–142.
  • Baell JB, Holloway GA. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J Med Chem. 2010;53(7):2719–2740.
  • Baell JB. Observations on screening-based research and some concerning trends in the literature. Future Med Chem. 2010;2(10):1529–1546.
  • Lajiness MS, Maggiora GM, Shanmugasundaram V. Assessment of the consistency of medicinal chemists in reviewing sets of compounds. J Med Chem. 2004;47(20):4891–4896.
  • Bruns RF, Watson IA. Rules for identifying potentially reactive or promiscuous compounds. J Med Chem. 2012;55(22):9763–9772.
  • Hu Y, Bajorath J. Compound promiscuity: what can we learn from current data? Drug Discov Today. 2013;18(13–14):644–650.
  • Che J, King FJ, Zhou B, et al. Chemical and biological properties of frequent screening hits. J Chem Inf Model. 2012;52(4):913–926.
  • Moberg A, Zander Balderud L, Hansson E, et al. Assessing HTS performance using BioAssay Ontology: screening and analysis of a bacterial phospho-N-acetylmuramoyl-pentapeptide translocase campaign. Assay Drug Dev Technol. 2014;12(9–10):506–513.
  • Wassermann AM, Lounkine E, Davies JW, et al. The opportunities of mining historical and collective data in drug discovery. Drug Discov Today. 2015;20(4):422–434.

•• Suggests valuable novel approaches for state-of-the-art in silico hit list analysis.

  • Renner S, Popov M, Schuffenhauer A, et al. Recent trends and observations in the design of high-quality screening collections. Future Med Chem. 2011;3(6):751–766.
  • Kadakkuzha BM, Spicer TP, Chase P, et al. High-throughput screening for small molecule modulators of motor protein Kinesin. Assay Drug Dev Technol. 2014;12(8):470–480.
  • Wu X, Lan L, Wilson DM, et al. Identification and validation of novel small molecule disruptors of HuR-mRNA interaction. ACS Chem Biol. 2015;10(6):1476–1484.
  • Jacobs AC, Calkins MJ, Jadhav A, et al. Inhibition of DNA glycosylases via small molecule purine analogs. PLoS One. 2013;8(12):e81667.
  • Wen Y, Xu L, Chen FL, et al. Discovery of a novel inhibitor of NAD(P)(+)-dependent malic enzyme (ME2) by high-throughput screening. Acta Pharmacol Sin. 2014;35(5):674–684.
  • Swingle MR, Honkanen RE. Development and validation of a robust and sensitive assay for the discovery of selective inhibitors for serine/threonine protein phosphatases PP1alpha (PPP1C) and PP5 (PPP5C). Assay Drug Dev Technol. 2014;12(8):481–496.
  • Benicchi T, Iozzi S, Svahn A, et al. A homogeneous HTRF assay for the identification of inhibitors of the TWEAK-Fn14 protein interaction. J Biomol Screen. 2012;17(7):933–945.
  • Raoof A, Depledge P, Hamilton NM, et al. Toxoflavins and deazaflavins as the first reported selective small molecule inhibitors of tyrosyl-DNA phosphodiesterase II. J Med Chem. 2013;56(16):6352–6370.
  • Drake KM, Watson VG, Kisielewski A, et al. A sensitive luminescent assay for the histone methyltransferase NSD1 and other SAM-dependent enzymes. Assay Drug Dev Technol. 2014;12(5):258–271.
  • Zhang JH, Kang ZB, Ardayfio O, et al. Application of titration-based screening for the rapid pilot testing of high-throughput assays. J Biomol Screen. 2013;19(5):651–660.
  • Zander Balderud L, Murray D, Larsson N, et al. Using the BioAssay Ontology for analyzing high-throughput screening data. J Biomol Screen. 2015;20(3):402–415.
  • Schmidt C. GSK/Sirtris compounds dogged by assay artifacts. Nat Biotechnol. 2010;28(3):185–186.

• A classic example of how assay artifacts can derail drug discovery.

  • Soares KM, Blackmon N, Shun TY, et al. Profiling the NIH Small Molecule Repository for compounds that generate H2O2 by redox cycling in reducing environments. Assay Drug Dev Technol. 2010;8(2):152–174.
  • Feng BY, Simeonov A, Jadhav A, et al. A high-throughput screen for aggregation-based inhibition in a large compound library. J Med Chem. 2007;50(10):2385–2390.
  • Seidler J, McGovern SL, Doman TN, et al. Identification and prediction of promiscuous aggregating inhibitors among known drugs. J Med Chem. 2003;46(21):4477–4486.
  • Mendgen T, Steuer C, Klein CD. Privileged scaffolds or promiscuous binders: a comparative study on rhodanines and related heterocycles in medicinal chemistry. J Med Chem. 2012;55(2):743–753.

• Challenges some common assumptions about a presumably ‘promiscuous’ scaffold with experimental evidence.

  • Thorne N, Auld DS, Inglese J. Apparent activity in high-throughput screening: origins of compound-dependent assay interference. Curr Opin Chem Biol. 2010;14(3):315–324.
  • Chen K, Michelsen K, Kurzeja RJ, et al. Discovery of small-molecule glucokinase regulatory protein modulators that restore glucokinase activity. J Biomol Screen. 2014;19(7):1014–1023.
  • Bergsdorf C, Ottl J. Affinity-based screening techniques: their impact and benefit to increase the number of high quality leads. Expert Opin Drug Discov. 2010;5(11):1095–1107.
  • O’Connell TN, Ramsay J, Rieth SF, et al. Solution-based indirect affinity selection mass spectrometry: a general tool for high-throughput screening of pharmaceutical compound libraries. Anal Chem. 2014;86(15):7413–7420.
  • Whitehurst CE, Yao Z, Murphy D, et al. Application of affinity selection-mass spectrometry assays to purification and affinity-based screening of the chemokine receptor CXCR4. Comb Chem High Throughput Screen. 2012;15(6):473–485.
  • Deng Y, Shipps GW Jr, Cooper A, et al. Discovery of novel, dual mechanism ERK inhibitors by affinity selection screening of an inactive kinase. J Med Chem. 2014;57(21):8817–8826.
  • Holdgate G, Geschwindner S, Breeze A, et al. Biophysical methods in drug discovery from small molecule to pharmaceutical. Methods Mol Biol. 2013;1008:327–355.
  • Clemente JC, Nulton E, Nelen M, et al. Screening and characterization of human monoglyceride lipase active site inhibitors using orthogonal binding and functional assays. J Biomol Screen. 2012;17(5):629–640.
  • Genick CC, Barlier D, Monna D, et al. Applications of biophysics in high-throughput screening hit validation. J Biomol Screen. 2014;19(5):707–714.
  • Simeonov A. Recent developments in the use of differential scanning fluorometry in protein and small molecule discovery and characterization. Expert Opin Drug Discov. 2013;8(9):1071–1082.
  • Vivoli M, Novak HR, Littlechild JA, et al. Determination of protein-ligand interactions using differential scanning fluorimetry. J Vis Exp. 2014. DOI:10.3791/51809(91):51809.
  • DeSantis K, Reed A, Rahhal R, et al. Use of differential scanning fluorimetry as a high-throughput assay to identify nuclear receptor ligands. Nucl Recept Signal. 2012;10:e002.
  • Sorrell FJ, Greenwood GK, Birchall K, et al. Development of a differential scanning fluorimetry based high throughput screening assay for the discovery of affinity binders against an anthrax protein. J Pharm Biomed Anal. 2010;52(5):802–808.
  • Martinez-Munoz L, Barroso R, Paredes AG, et al. Methods to immobilize GPCR on the surface of SPR sensors. Methods Mol Biol. 2015;1272:173–188.
  • Frenzel D, Gluck JM, Brener O, et al. Immobilization of homogeneous monomeric, oligomeric and fibrillar Abeta species for reliable SPR measurements. PLoS One. 2014;9(3):e89490.
  • Lee H, Zhu T, Patel K, et al. High-throughput screening (HTS) and hit validation to identify small molecule inhibitors with activity against NS3/4A proteases from multiple hepatitis C virus genotypes. PLoS One. 2013;8(10):e75144.
  • Lee H, Mittal A, Patel K, et al. Identification of novel drug scaffolds for inhibition of SARS-CoV 3-Chymotrypsin-like protease using virtual and high-throughput screenings. Bioorg Med Chem. 2014;22(1):167–177.
  • Mikuni S, Kodama K, Sasaki A, et al. Screening for FtsZ dimerization inhibitors using fluorescence cross-correlation spectroscopy and surface resonance plasmon analysis. PLoS One. 2015;10(7):e0130933.
  • McKinstry-Wu AR, Bu W, Rai G, et al. Discovery of a novel general anesthetic chemotype using high-throughput screening. Anesthesiology. 2015;122(2):325–333.
  • Seidel SA, Wienken CJ, Geissler S, et al. Label-free microscale thermophoresis discriminates sites and affinity of protein-ligand binding. Angew Chem Int Ed Engl. 2012;51(42):10656–10659.
  • Sillerud LO, Larson RS. Advances in nuclear magnetic resonance for drug discovery. Methods Mol Biol. 2012;910:195–266.
  • Zheng H, Handing KB, Zimmerman MD, et al. X-ray crystallography over the past decade for novel drug discovery: where are we heading next? Expert Opin Drug Discov. 2015;10(9):975–989.
  • Chan-Penebre E, Kuplast KG, Majer CR, et al. A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Nat Chem Biol. 2015;11(6):432–437.

•• A prime example of how various approaches and techniques can be integrated into a meaningful flowchart for efficient lead finding.

• A good introduction to the concept of ‘target engagement’ and its role at various stages of the drug discovery pipeline.

  • Paweletz CP, Andersen JN, Pollock R, et al. Identification of direct target engagement biomarkers for kinase-targeted therapeutics. PLoS One. 2011;6(10):e26459.
  • Simon GM, Niphakis MJ, Cravatt BF. Determining target engagement in living systems. Nat Chem Biol. 2013;9(4):200–205.
  • Visser SA, Aurell M, Jones RD, et al. Model-based drug discovery: implementation and impact. Drug Discov Today. 2013;18(15–16):764–775.
  • Pedro-Rosa L, Buckner FS, Ranade RM, et al. Identification of potent inhibitors of the Trypanosoma brucei methionyl-tRNA synthetase via high-throughput orthogonal screening. J Biomol Screen. 2015;20(1):122–130.
  • Dranchak P, MacArthur R, Guha R, et al. Profile of the GSK published protein kinase inhibitor set across ATP-dependent and-independent luciferases: implications for reporter-gene assays. PLoS One. 2013;8(3):e57888.
  • Grohar PJ, Woldemichael GM, Griffin LB, et al. Identification of an inhibitor of the EWS-FLI1 oncogenic transcription factor by high-throughput screening. J Natl Cancer Inst. 2011;103(12):962–978.
  • McDermott SP, Eppert K, Notta F, et al. A small molecule screening strategy with validation on human leukemia stem cells uncovers the therapeutic efficacy of kinetin riboside. Blood. 2012;119(5):1200–1207.
  • Martinez Molina D, Jafari R, Ignatushchenko M, et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science. 2013;341(6141):84–87.
  • Jafari R, Almqvist H, Axelsson H, et al. The cellular thermal shift assay for evaluating drug target interactions in cells. Nat Protoc. 2014;9(9):2100–2122.
  • Jensen AJ, Martinez Molina D, Lundback T. CETSA: a target engagement assay with potential to transform drug discovery. Future Med Chem. 2015;7(8):975–978.

• Contains some interesting thoughts on how to apply target engagement assays.

  • Savitski MM, Reinhard FB, Franken H, et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science. 2014;346(6205):1255784.
  • Auld DS, Davis CA, Jimenez M, et al. Examining ligand-based stabilization of proteins in cells with MEK1 kinase inhibitors. Assay Drug Dev Technol. 2015;13(5):266–276.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.