1932

Abstract

The genetic determinants of many diseases, including monogenic diseases and cancers, have been identified; nevertheless, targeted therapy remains elusive for most. High-throughput screening (HTS) of small molecules, including high-content analysis (HCA), has been an important technology for the discovery of molecular tools and new therapeutics. HTS can be based on modulation of a known disease target (called reverse chemical genetics) or modulation of a disease-associated mechanism or phenotype (forward chemical genetics). Prominent target-based successes include modulators of transthyretin, used to treat transthyretin amyloidoses, and the BCR-ABL kinase inhibitor Gleevec, used to treat chronic myelogenous leukemia. Phenotypic screening successes include modulators of cystic fibrosis transmembrane conductance regulator, splicing correctors for spinal muscular atrophy, and histone deacetylase inhibitors for cancer. Synthetic lethal screening, in which chemotherapeutics are screened for efficacy against specific genetic backgrounds, is a promising approach that merges phenotype and target. In this article, we introduce HTS technology and highlight its contributions to the discovery of drugs and probes for monogenic diseases and cancer.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-genom-083117-021452
2018-08-31
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/genom/19/1/annurev-genom-083117-021452.html?itemId=/content/journals/10.1146/annurev-genom-083117-021452&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Aflaki E, Borger DK, Moaven N, Stubblefield BK, Rogers SA et al. 2016. A new glucocerebrosidase chaperone reduces α-synuclein and glycolipid levels in iPSC-derived dopaminergic neurons from patients with Gaucher disease and Parkinsonism. J. Neurosci. 36:7441–52
    [Google Scholar]
  2. 2.  Aflaki E, Stubblefield BK, Maniwang E, Lopez G, Moaven N et al. 2014. Macrophage models of Gaucher disease for evaluating disease pathogenesis and candidate drugs. Sci. Transl. Med. 6:240ra73
    [Google Scholar]
  3. 3.  Aiken CT, Tobin AJ, Schweitzer ES 2004. A cell-based screen for drugs to treat Huntington's disease. Neurobiol. Dis. 16:546–55
    [Google Scholar]
  4. 4.  Almaca J, Faria D, Sousa M, Uliyakina I, Conrad C et al. 2013. High-content siRNA screen reveals global ENaC regulators and potential cystic fibrosis therapy targets. Cell 154:1390–400
    [Google Scholar]
  5. 5.  Amberger JS, Bocchini CA, Schiettecatte F, Scott AF, Hamosh A 2015. OMIM.org: Online Mendelian Inheritance in Man (OMIM®), an online catalog of human genes and genetic disorders. Nucleic Acids Res 43:D789–98
    [Google Scholar]
  6. 6.  Arkin MR, Ang KK, Chen S, Davies J, Merron C et al. 2014. UCSF Small Molecule Discovery Center: innovation, collaboration and chemical biology in the Bay Area. Comb. Chem. High Throughput Screen. 17:333–42
    [Google Scholar]
  7. 7.  Baell JB, Holloway GA 2010. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 53:2719–40
    [Google Scholar]
  8. 8.  Baell JB, Walters MA 2014. Chemical con artists foil drug discovery. Nature 513:481–83
    [Google Scholar]
  9. 9.  Banasik M, Komura H, Shimoyama M, Ueda K 1992. Specific inhibitors of poly(ADP-ribose) synthetase and mono(ADP-ribosyl)transferase. J. Biol. Chem. 267:1569–75
    [Google Scholar]
  10. 10.  Bard J, Wall MD, Lazari O, Arjomand J, Munoz-Sanjuan I 2014. Advances in Huntington disease drug discovery: novel approaches to model disease phenotypes. J. Biomol. Screen. 19:191–204
    [Google Scholar]
  11. 11.  Berthelier V, Wetzel R 2006. Screening for modulators of aggregation with a microplate elongation assay. Methods Enzymol 413:313–25
    [Google Scholar]
  12. 12.  Besnard J, Jones PS, Hopkins AL, Pannifer AD 2015. The Joint European Compound Library: boosting precompetitive research. Drug Discov. Today 20:181–86
    [Google Scholar]
  13. 13.  Billin AN, Bantscheff M, Drewes G, Ghidelli-Disse S, Holt JA et al. 2016. Discovery of novel small molecules that activate satellite cell proliferation and enhance repair of damaged muscle. ACS Chem. Biol. 11:518–29
    [Google Scholar]
  14. 14.  Bittker JA, Ross NT, eds. 2017. High Throughput Screening Methods: Evolution and Refinement Cambridge, UK: R. Soc. Chem.
  15. 15.  Blat Y, Blat S 2015. Drug discovery of therapies for Duchenne muscular dystrophy. J. Biomol. Screen. 20:1189–203
    [Google Scholar]
  16. 16.  Bollag G, Tsai J, Zhang J, Zhang C, Ibrahim P et al. 2012. Vemurafenib: the first drug approved for BRAF-mutant cancer. Nat. Rev. Drug Discov. 11:873–86
    [Google Scholar]
  17. 17.  Bordet T, Buisson B, Michaud M, Drouot C, Galea P et al. 2007. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J. Pharmacol. Exp. Ther. 322:709–20
    [Google Scholar]
  18. 18.  Botelho HM, Uliyakina I, Awatade NT, Proenca MC, Tischer C et al. 2015. Protein traffic disorders: an effective high-throughput fluorescence microscopy pipeline for drug discovery. Sci. Rep. 5:9038
    [Google Scholar]
  19. 19.  Bowman TV, Zon LI 2010. Swimming into the future of drug discovery: in vivo chemical screens in zebrafish. ACS Chem. Biol. 5:159–61
    [Google Scholar]
  20. 20.  Boyle MP, Bell SC, Konstan MW, McColley SA, Rowe SM et al. 2014. A CFTR corrector (lumacaftor) and a CFTR potentiator (ivacaftor) for treatment of patients with cystic fibrosis who have a phe508del CFTR mutation: a phase 2 randomised controlled trial. Lancet Respir. Med. 2:527–38
    [Google Scholar]
  21. 21.  Bray MA, Vokes MS, Carpenter AE 2015. Using CellProfiler for automatic identification and measurement of biological objects in images. Curr. Protoc. Mol. Biol. 109:14.17.1–13
    [Google Scholar]
  22. 22.  Bruni G, Lakhani P, Kokel D 2014. Discovering novel neuroactive drugs through high-throughput behavior-based chemical screening in the zebrafish. Front. Pharmacol. 5:153
    [Google Scholar]
  23. 23.  Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D et al. 2005. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434:913–17
    [Google Scholar]
  24. 24.  Buchdunger E, Cioffi CL, Law N, Stover D, Ohno-Jones S et al. 2000. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J. Pharmacol. Exp. Ther. 295:139–45
    [Google Scholar]
  25. 25.  Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M et al. 1996. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res 56:100–4
    [Google Scholar]
  26. 26.  Bulman CA, Bidlow CM, Lustigman S, Cho-Ngwa F, Williams D et al. 2015. Repurposing auranofin as a lead candidate for treatment of lymphatic filariasis and onchocerciasis. PLOS Negl. Trop. Dis. 9:e0003534
    [Google Scholar]
  27. 27.  Butko MT, Moree B, Mortensen RB, Salafsky J 2016. Detection of ligand-induced conformational changes in oligonucleotides by second-harmonic generation at a supported lipid bilayer interface. Anal. Chem. 88:10482–89
    [Google Scholar]
  28. 28.  Caicedo JC, Cooper S, Heigwer F, Warchal S, Qiu P et al. 2017. Data-analysis strategies for image-based cell profiling. Nat. Methods 14:849–63
    [Google Scholar]
  29. 29.  Calder AN, Androphy EJ, Hodgetts KJ 2016. Small molecules in development for the treatment of spinal muscular atrophy. J. Med. Chem. 59:10067–83
    [Google Scholar]
  30. 30.  Capuzzi SJ, Muratov EN, Tropsha A 2017. Phantom PAINS: problems with the utility of alerts for Pan-Assay INterference CompoundS. J. Chem. Inf. Model. 57:417–27
    [Google Scholar]
  31. 31.  Carroll M, Ohno-Jones S, Tamura S, Buchdunger E, Zimmermann J et al. 1997. CGP 57148, a tyrosine kinase inhibitor, inhibits the growth of cells expressing BCR-ABL, TEL-ABL, and TEL-PDGFR fusion proteins. Blood 90:4947–52
    [Google Scholar]
  32. 32.  Chakravorty S, Hegde M 2017. Gene and variant annotation for Mendelian disorders in the era of advanced sequencing technologies. Annu. Rev. Genom. Hum. Genet. 18:229–56
    [Google Scholar]
  33. 33.  Cherry JJ, Evans MC, Ni J, Cuny GD, Glicksman MA, Androphy EJ 2012. Identification of novel compounds that increase SMN protein levels using an improved SMN2 reporter cell assay. J. Biomol. Screen. 17:481–95
    [Google Scholar]
  34. 34.  Cherry JJ, Osman EY, Evans MC, Choi S, Xing X et al. 2013. Enhancement of SMN protein levels in a mouse model of spinal muscular atrophy using novel drug-like compounds. EMBO Mol. Med. 5:1103–18
    [Google Scholar]
  35. 35.  Cho S, Dreyfuss G 2010. A degron created by SMN2 exon 7 skipping is a principal contributor to spinal muscular atrophy severity. Genes Dev 24:438–42
    [Google Scholar]
  36. 36.  Chong CR, Chen X, Shi L, Liu JO, Sullivan DJ Jr. 2006. A clinical drug library screen identifies astemizole as an antimalarial agent. Nat. Chem. Biol. 2:415–16
    [Google Scholar]
  37. 37.  Chopra V, Fox JH, Lieberman G, Dorsey K, Matson W et al. 2007. A small-molecule therapeutic lead for Huntington's disease: preclinical pharmacology and efficacy of C2-8 in the R6/2 transgenic mouse. PNAS 104:16685–89
    [Google Scholar]
  38. 38.  Clancy JP, Rowe SM, Accurso FJ, Aitken ML, Amin RS et al. 2012. Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for the F508del-CFTR mutation. Thorax 67:12–18
    [Google Scholar]
  39. 39.  Coelho T, Merlini G, Bulawa CE, Fleming JA, Judge DP et al. 2016. Mechanism of action and clinical application of tafamidis in hereditary transthyretin amyloidosis. Neurol. Ther. 5:1–25
    [Google Scholar]
  40. 40.  Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ 2014. Drugging the undruggable RAS: mission possible?. Nat. Rev. Drug Discov. 13:828–51
    [Google Scholar]
  41. 41.  De Matteis MA, Vicinanza M, Venditti R, Wilson C 2013. Cellular assays for drug discovery in genetic disorders of intracellular trafficking. Annu. Rev. Genom. Hum. Genet. 14:159–90
    [Google Scholar]
  42. 42.  Deininger M, Buchdunger E, Druker BJ 2005. The development of imatinib as a therapeutic agent for chronic myeloid leukemia. Blood 105:2640–53
    [Google Scholar]
  43. 43.  Delvecchio C, Tiefenbach J, Krause HM 2011. The zebrafish: a powerful platform for in vivo, HTS drug discovery. Assay Drug Dev. Technol. 9:354–61
    [Google Scholar]
  44. 44.  Desai UA, Pallos J, Ma AA, Stockwell BR, Thompson LM et al. 2006. Biologically active molecules that reduce polyglutamine aggregation and toxicity. Hum. Mol. Genet. 15:2114–24
    [Google Scholar]
  45. 45.  Dillon KJ, Smith GC, Martin NM 2003. A FlashPlate assay for the identification of PARP-1 inhibitors. J. Biomol. Screen. 8:347–52
    [Google Scholar]
  46. 46.  Dolma S, Lessnick SL, Hahn WC, Stockwell BR 2003. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell 3:285–96
    [Google Scholar]
  47. 47.  Drilon A, Somwar R, Wagner JP, Vellore NA, Eide CA et al. 2016. A novel crizotinib-resistant solvent-front mutation responsive to cabozantinib therapy in a patient with ROS1-rearranged lung cancer. Clin. Cancer Res. 22:2351–58
    [Google Scholar]
  48. 48.  Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM et al. 1996. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat. Med. 2:561–66
    [Google Scholar]
  49. 49.  Du L, Damoiseaux R, Nahas S, Gao K, Hu H et al. 2009. Nonaminoglycoside compounds induce readthrough of nonsense mutations. J. Exp. Med. 206:2285–97
    [Google Scholar]
  50. 50.  Du L, Jung ME, Damoiseaux R, Completo G, Fike F et al. 2013. A new series of small molecular weight compounds induce read through of all three types of nonsense mutations in the ATM gene. Mol. Ther. 21:1653–60
    [Google Scholar]
  51. 51.  Erlanson DA, Fesik SW, Hubbard RE, Jahnke W, Jhoti H 2016. Twenty years on: the impact of fragments on drug discovery. Nat. Rev. Drug Discov. 15:605–19
    [Google Scholar]
  52. 52.  Erlanson DA, Jahnke W, eds. 2016. Fragment-Based Drug Discovery: Lessons and Outlook Weinheim, Ger.: Wiley-VCH
  53. 53.  Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA et al. 2005. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434:917–21
    [Google Scholar]
  54. 54.  Ferrins L, Pollastri MP 2018. The importance of collaboration between industry, academics, and nonprofits in tropical disease drug discovery. ACS Infect Dis 4:445–48
    [Google Scholar]
  55. 55.  Freeman AK, Ritt DA, Morrison DK 2013. Effects of Raf dimerization and its inhibition on normal and disease-associated Raf signaling. Mol. Cell 49:751–58
    [Google Scholar]
  56. 56.  Friend C, Scher W, Holland JG, Sato T 1971. Hemoglobin synthesis in murine virus-induced leukemic cells in vitro: stimulation of erythroid differentiation by dimethyl sulfoxide. PNAS 68:378–82
    [Google Scholar]
  57. 57.  Fuentealba RA, Marasa J, Diamond MI, Piwnica-Worms D, Weihl CC 2012. An aggregation sensing reporter identifies leflunomide and teriflunomide as polyglutamine aggregate inhibitors. Hum. Mol. Genet. 21:664–80
    [Google Scholar]
  58. 58.  Gambacorti-Passerini C 2008. Part I: milestones in personalised medicine—imatinib. Lancet Oncol 9:600
    [Google Scholar]
  59. 59.  Garbaccio RM, Parmee ER 2016. The impact of chemical probes in drug discovery: a pharmaceutical industry perspective. Cell Chem. Biol. 23:10–17
    [Google Scholar]
  60. 60.  Gintjee TJ, Magh AS, Bertoni C 2014. High throughput screening in Duchenne muscular dystrophy: from drug discovery to functional genomics. Biology 3:752–80
    [Google Scholar]
  61. 61.  Goldin E, Zheng W, Motabar O, Southall N, Choi JH et al. 2012. High throughput screening for small molecule therapy for Gaucher disease using patient tissue as the source of mutant glucocerebrosidase. PLOS ONE 7:e29861
    [Google Scholar]
  62. 62.  Griffin A, Hamling KR, Knupp K, Hong S, Lee LP, Baraban SC 2017. Clemizole and modulators of serotonin signalling suppress seizures in Dravet syndrome. Brain 140:669–83
    [Google Scholar]
  63. 63.  Harvey AL, Edrada-Ebel R, Quinn RJ 2015. The re-emergence of natural products for drug discovery in the genomics era. Nat. Rev. Drug Discov. 14:111–29
    [Google Scholar]
  64. 64.  Heiser V, Engemann S, Brocker W, Dunkel I, Boeddrich A et al. 2002. Identification of benzothiazoles as potential polyglutamine aggregation inhibitors of Huntington's disease by using an automated filter retardation assay. PNAS 99:Suppl. 416400–6
    [Google Scholar]
  65. 65.  Hu Y, Wu B, Zillmer A, Lu P, Benrashid E et al. 2010. Guanine analogues enhance antisense oligonucleotide-induced exon skipping in dystrophin gene in vitro and in vivo. Mol. Ther. 18:812–18
    [Google Scholar]
  66. 66.  Irwin JJ, Duan D, Torosyan H, Doak AK, Ziebart KT et al. 2015. An aggregation advisor for ligand discovery. J. Med. Chem. 58:7076–87
    [Google Scholar]
  67. 67.  Janzen WP 2014. Screening technologies for small molecule discovery: the state of the art. Chem. Biol. 21:1162–70
    [Google Scholar]
  68. 68.  Johannes JW, Almeida L, Daly K, Ferguson AD, Grosskurth SE et al. 2015. Discovery of AZ0108, an orally bioavailable phthalazinone PARP inhibitor that blocks centrosome clustering. Bioorg. Med. Chem. Lett. 25:5743–47
    [Google Scholar]
  69. 69.  Kaelin WG Jr 2005. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5:689–98
    [Google Scholar]
  70. 70.  Kaniskan HU, Konze KD, Jin J 2015. Selective inhibitors of protein methyltransferases. J. Med. Chem. 58:1596–629
    [Google Scholar]
  71. 71.  Karoulia Z, Gavathiotis E, Poulikakos PI 2017. New perspectives for targeting RAF kinase in human cancer. Nat. Rev. Cancer 17:676–91
    [Google Scholar]
  72. 72.  Katayama R, Kobayashi Y, Friboulet L, Lockerman EL, Koike S et al. 2015. Cabozantinib overcomes crizotinib resistance in ROS1 fusion-positive cancer. Clin. Cancer Res. 21:166–74
    [Google Scholar]
  73. 73.  Kendall GC, Mokhonova EI, Moran M, Sejbuk NE, Wang DW et al. 2012. Dantrolene enhances antisense-mediated exon skipping in human and mouse models of Duchenne muscular dystrophy. Sci. Transl. Med. 4:164ra60
    [Google Scholar]
  74. 74.  Ketley A, Chen CZ, Li X, Arya S, Robinson TE et al. 2014. High-content screening identifies small molecules that remove nuclear foci, affect MBNL distribution and CELF1 protein levels via a PKC-independent pathway in myotonic dystrophy cell lines. Hum. Mol. Genet. 23:1551–62
    [Google Scholar]
  75. 75.  Knowles MR, Drumm M 2012. The influence of genetics on cystic fibrosis phenotypes. Cold Spring Harb. Perspect. Med. 2:a009548
    [Google Scholar]
  76. 76.  Langenau DM 2016. Cancer and Zebrafish: Mechanisms, Techniques, and Models Cham, Switz.: Springer
  77. 77.  Lefebvre S, Burglen L, Reboullet S, Clermont O, Burlet P et al. 1995. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 80:155–65
    [Google Scholar]
  78. 78.  Liu J, Zhou Y, Qi X, Chen J, Chen W et al. 2017. CRISPR/Cas9 in zebrafish: an efficient combination for human genetic diseases modeling. Hum. Genet. 136:1–12
    [Google Scholar]
  79. 79.  Lord CJ, Ashworth A 2017. PARP inhibitors: synthetic lethality in the clinic. Science 355:1152–58
    [Google Scholar]
  80. 80.  Lorson CL, Hahnen E, Androphy EJ, Wirth B 1999. A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. PNAS 96:6307–11
    [Google Scholar]
  81. 81.  Lunn MR, Stockwell BR 2005. Chemical genetics and orphan genetic diseases. Chem. Biol. 12:1063–73
    [Google Scholar]
  82. 82.  Ma CX, Cai S, Li S, Ryan CE, Guo Z et al. 2012. Targeting Chk1 in p53-deficient triple-negative breast cancer is therapeutically beneficial in human-in-mouse tumor models. J. Clin. Investig. 122:1541–52
    [Google Scholar]
  83. 83.  Macarron R, Banks MN, Bojanic D, Burns DJ, Cirovic DA et al. 2011. Impact of high-throughput screening in biomedical research. Nat. Rev. Drug Discov. 10:188–95
    [Google Scholar]
  84. 84.  Marks PA, Breslow R 2007. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat. Biotechnol. 25:84–90
    [Google Scholar]
  85. 85.  Martinez Molina D, Nordlund P 2016. The cellular thermal shift assay: a novel biophysical assay for in situ drug target engagement and mechanistic biomarker studies. Annu. Rev. Pharmacol. Toxicol. 56:141–61
    [Google Scholar]
  86. 86.  Mathews Griner LA, Guha R, Shinn P, Young RM, Keller JM et al. 2014. High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells. PNAS 111:2349–54
    [Google Scholar]
  87. 87.  McMahon B, Aflaki E, Sidransky E 2016. Chaperoning glucocerebrosidase: a therapeutic strategy for both Gaucher disease and Parkinsonism. Neural Regen. Res. 11:1760–61
    [Google Scholar]
  88. 88.  Menear KA, Adcock C, Boulter R, Cockcroft XL, Copsey L et al. 2008. 4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1. J. Med. Chem. 51:6581–91
    [Google Scholar]
  89. 89.  Mistry PK, Lopez G, Schiffmann R, Barton NW, Weinreb NJ, Sidransky E 2017. Gaucher disease: progress and ongoing challenges. Mol. Genet. Metab. 120:8–21
    [Google Scholar]
  90. 90.  Moffat JG, Rudolph J, Bailey D 2014. Phenotypic screening in cancer drug discovery—past, present and future. Nat. Rev. Drug Discov. 13:588–602
    [Google Scholar]
  91. 91.  Moorwood C, Khurana TS 2013. Duchenne muscular dystrophy drug discovery—the application of utrophin promoter activation screening. Expert Opin. Drug Discov. 8:569–81
    [Google Scholar]
  92. 92.  Mullard A 2017. 2016 FDA drug approvals. Nat. Rev. Drug Discov. 16:73–76
    [Google Scholar]
  93. 93.  Nakajima H, Kim YB, Terano H, Yoshida M, Horinouchi S 1998. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Exp. Cell Res. 241:126–33
    [Google Scholar]
  94. 94.  Naryshkin NA, Weetall M, Dakka A, Narasimhan J, Zhao X et al. 2014. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy. Science 345:688–93
    [Google Scholar]
  95. 95.  Nierobisz LS, Cheatham B, Buehrer BM, Sexton JZ 2013. High-content screening of human primary muscle satellite cells for new therapies for muscular atrophy/dystrophy. Curr. Chem. Genom. Transl. Med. 7:21–29
    [Google Scholar]
  96. 96.  O'Leary DA, Sharif O, Anderson P, Tu B, Welch G et al. 2009. Identification of small molecule and genetic modulators of AON-induced dystrophin exon skipping by high-throughput screening. PLOS ONE 4:e8348
    [Google Scholar]
  97. 97.  O'Neil NJ, Bailey ML, Hieter P 2017. Synthetic lethality and cancer. Nat. Rev. Genet. 18:613–23
    [Google Scholar]
  98. 98.  Ong T, Ramsey BW 2016. New therapeutic approaches to modulate and correct cystic fibrosis transmembrane conductance regulator. Pediatr. Clin. N. Am. 63:751–64
    [Google Scholar]
  99. 99.  Ottesen EW 2017. ISS-N1 makes the first FDA-approved drug for spinal muscular atrophy. Transl. Neurosci. 8:1–6
    [Google Scholar]
  100. 100.  Palacino J, Swalley SE, Song C, Cheung AK, Shu L et al. 2015. SMN2 splice modulators enhance U1-pre-mRNA association and rescue SMA mice. Nat. Chem. Biol. 11:511–17
    [Google Scholar]
  101. 101.  Pattenden SG, Simon JM, Wali A, Jayakody CN, Troutman J et al. 2016. High-throughput small molecule screen identifies inhibitors of aberrant chromatin accessibility. PNAS 113:3018–23
    [Google Scholar]
  102. 102.  Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH et al. 2010. How to improve R&D productivity: the pharmaceutical industry's grand challenge. Nat. Rev. Drug Discov. 9:203–14
    [Google Scholar]
  103. 103.  Plantie E, Migocka-Patrzalek M, Daczewska M, Jagla K 2015. Model organisms in the fight against muscular dystrophy: lessons from Drosophila and zebrafish. Molecules 20:6237–53
    [Google Scholar]
  104. 104.  Plass C, Pfister SM, Lindroth AM, Bogatyrova O, Claus R, Lichter P 2013. Mutations in regulators of the epigenome and their connections to global chromatin patterns in cancer. Nat. Rev. Genet. 14:765–80
    [Google Scholar]
  105. 105.  Pollak M 2014. Overcoming drug development bottlenecks with repurposing: repurposing biguanides to target energy metabolism for cancer treatment. Nat. Med. 20:591–93
    [Google Scholar]
  106. 106.  Pollitt SK, Pallos J, Shao J, Desai UA, Ma AA et al. 2003. A rapid cellular FRET assay of polyglutamine aggregation identifies a novel inhibitor. Neuron 40:685–94
    [Google Scholar]
  107. 107.  Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C et al. 2011. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature 480:387–90
    [Google Scholar]
  108. 108.  Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N 2010. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature 464:427–30
    [Google Scholar]
  109. 109.  Pruss RM, Giraudon-Paoli M, Morozova S, Berna P, Abitbol JL, Bordet T 2010. Drug discovery and development for spinal muscular atrophy: lessons from screening approaches and future challenges for clinical development. Future Med. Chem. 2:1429–40
    [Google Scholar]
  110. 110.  Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC et al. 2011. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N. Engl. J. Med. 365:1663–72
    [Google Scholar]
  111. 111.  Ramsey BW, Welsh MJ 2017. Progress along the pathway of discovery leading to treatment and cure of cystic fibrosis. Am. J. Respir. Crit. Care Med. 195:1092–99
    [Google Scholar]
  112. 112.  Reaper PM, Griffiths MR, Long JM, Charrier JD, MacCormick S et al. 2011. Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nat. Chem. Biol. 7:428–30
    [Google Scholar]
  113. 113.  Richon VM, Emiliani S, Verdin E, Webb Y, Breslow R et al. 1998. A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases. PNAS 95:3003–7
    [Google Scholar]
  114. 114.  Richon VM, Webb Y, Merger R, Sheppard T, Jursic B et al. 1996. Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. PNAS 93:5705–8
    [Google Scholar]
  115. 115.  Schindelin J, Rueden CT, Hiner MC, Eliceiri KW 2015. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol. Reprod. Dev. 82:518–29
    [Google Scholar]
  116. 116.  Schirle M, Jenkins JL 2016. Identifying compound efficacy targets in phenotypic drug discovery. Drug Discov. Today 21:82–89
    [Google Scholar]
  117. 117.  Schreiber SL, Kotz JD, Li M, Aube J, Austin CP et al. 2015. Advancing biological understanding and therapeutics discovery with small-molecule probes. Cell 161:1252–65
    [Google Scholar]
  118. 118.  Senger MR, Fraga CA, Dantas RF, Silva FP Jr. 2016. Filtering promiscuous compounds in early drug discovery: Is it a good idea?. Drug Discov. Today 21:868–72
    [Google Scholar]
  119. 119.  Shannon KM, Fraint A 2015. Therapeutic advances in Huntington's disease. Mov. Disord. 30:1539–46
    [Google Scholar]
  120. 120.  Sharma J, Pareek V, Liu H, Cheng H 2016. Emerging treatment for ALK-positive lung cancer. Expert Opin. Emerg. Drugs 21:147–55
    [Google Scholar]
  121. 121.  Shimizu-Motohashi Y, Miyatake S, Komaki H, Takeda S, Aoki Y 2016. Recent advances in innovative therapeutic approaches for Duchenne muscular dystrophy: from discovery to clinical trials. Am. J. Transl. Res. 8:2471–89
    [Google Scholar]
  122. 122.  Shiryaev SA, Mesci P, Pinto A, Fernandes I, Sheets N et al. 2017. Repurposing of the anti-malaria drug chloroquine for Zika virus treatment and prophylaxis. Sci. Rep. 7:15771
    [Google Scholar]
  123. 123.  Shoichet BK 2006. Screening in a spirit haunted world. Drug Discov. Today 11:607–15
    [Google Scholar]
  124. 124.  Sittampalam GS, Coussens NP, Brimacombe K, Grossman A, Arkin M, eds. 2017. Assay Guidance Manual Bethesda, MD: Eli Lilly and Natl. Cent. Adv. Transl. Sci. https://www.ncbi.nlm.nih.gov/books/NBK53196
  125. 125.  Sleigh JN, Buckingham SD, Esmaeili B, Viswanathan M, Cuppen E et al. 2011. A novel Caenorhabditis elegans allele, smn-1(cb131), mimicking a mild form of spinal muscular atrophy, provides a convenient drug screening platform highlighting new and pre-approved compounds. Hum. Mol. Genet. 20:245–60
    [Google Scholar]
  126. 126.  Specht EA, Braselmann E, Palmer AE 2017. A critical and comparative review of fluorescent tools for live-cell imaging. Annu. Rev. Physiol. 79:93–117
    [Google Scholar]
  127. 127.  Swinney DC 2013. The contribution of mechanistic understanding to phenotypic screening for first-in-class medicines. J. Biomol. Screen. 18:1186–92
    [Google Scholar]
  128. 128.  Thorne N, Shen M, Lea WA, Simeonov A, Lovell S et al. 2012. Firefly luciferase in chemical biology: a compendium of inhibitors, mechanistic evaluation of chemotypes, and suggested use as a reporter. Chem. Biol. 19:1060–72
    [Google Scholar]
  129. 129.  Tinsley JM, Fairclough RJ, Storer R, Wilkes FJ, Potter AC et al. 2011. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PLOS ONE 6:e19189
    [Google Scholar]
  130. 130.  Tiscornia G, Vivas EL, Matalonga L, Berniakovich I, Barragan Monasterio M et al. 2013. Neuronopathic Gaucher's disease: induced pluripotent stem cells for disease modelling and testing chaperone activity of small compounds. Hum. Mol. Genet. 22:633–45
    [Google Scholar]
  131. 131.  Titus SA, Southall N, Marugan J, Austin CP, Zheng W 2012. High-throughput multiplexed quantitation of protein aggregation and cytotoxicity in a Huntington's disease model. Curr. Chem. Genom. 6:79–86
    [Google Scholar]
  132. 132.  Torre BG, Albericio F 2017. The pharmaceutical industry in 2016. An analysis of FDA drug approvals from a perspective of the molecule type. Molecules 22:368
    [Google Scholar]
  133. 133.  Tsai J, Lee JT, Wang W, Zhang J, Cho H et al. 2008. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. PNAS 105:3041–46
    [Google Scholar]
  134. 134.  Ueda H, Manda T, Matsumoto S, Mukumoto S, Nishigaki F et al. 1994. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum no. 968. III. Antitumor activities on experimental tumors in mice. J. Antibiot. 47:315–23
    [Google Scholar]
  135. 135.  Ueda H, Nakajima H, Hori Y, Fujita T, Nishimura M et al. 1994. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum no. 968. I. Taxonomy, fermentation, isolation, physico-chemical and biological properties, and antitumor activity. J. Antibiot. 47:301–10
    [Google Scholar]
  136. 136.  Ueda H, Nakajima H, Hori Y, Goto T, Okuhara M 1994. Action of FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum no. 968, on Ha-ras transformed NIH3T3 cells. Biosci. Biotechnol. Biochem. 58:1579–83
    [Google Scholar]
  137. 137.  Valenzano KJ, Khanna R, Powe AC, Boyd R, Lee G et al. 2011. Identification and characterization of pharmacological chaperones to correct enzyme deficiencies in lysosomal storage disorders. Assay Drug Dev. Technol. 9:213–35
    [Google Scholar]
  138. 138.  Van Goor F, Hadida S, Grootenhuis PD, Burton B, Cao D et al. 2009. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. PNAS 106:18825–30
    [Google Scholar]
  139. 139.  Van Goor F, Hadida S, Grootenhuis PD, Burton B, Stack JH et al. 2011. Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. PNAS 108:18843–48
    [Google Scholar]
  140. 140.  Van Goor F, Straley KS, Cao D, Gonzalez J, Hadida S et al. 2006. Rescue of ΔF508-CFTR trafficking and gating in human cystic fibrosis airway primary cultures by small molecules. Am. J. Physiol. Lung Cell Mol. Physiol. 290:L1117–30
    [Google Scholar]
  141. 141.  Van Rossum A, Holsopple M 2016. Enzyme replacement or substrate reduction? A review of Gaucher disease treatment options. Hosp. Pharm. 51:553–63
    [Google Scholar]
  142. 142.  Varma H, Voisine C, DeMarco CT, Cattaneo E, Lo DC et al. 2007. Selective inhibitors of death in mutant huntingtin cells. Nat. Chem. Biol. 3:99–100
    [Google Scholar]
  143. 143.  Wang J, Gines S, MacDonald ME, Gusella JF 2005. Reversal of a full-length mutant huntingtin neuronal cell phenotype by chemical inhibitors of polyglutamine-mediated aggregation. BMC Neurosci 6:1
    [Google Scholar]
  144. 144.  Wang W, Duan W, Igarashi S, Morita H, Nakamura M, Ross CA 2005. Compounds blocking mutant huntingtin toxicity identified using a Huntington's disease neuronal cell model. Neurobiol. Dis. 20:500–8
    [Google Scholar]
  145. 145.  Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ et al. 2007. PTC124 targets genetic disorders caused by nonsense mutations. Nature 447:87–91
    [Google Scholar]
  146. 146.  Woll MG, Qi H, Turpoff A, Zhang N, Zhang X et al. 2016. Discovery and optimization of small molecule splicing modifiers of survival motor neuron 2 as a treatment for spinal muscular atrophy. J. Med. Chem. 59:6070–85
    [Google Scholar]
  147. 147.  Xiao J, Marugan JJ, Zheng W, Titus S, Southall N et al. 2011. Discovery, synthesis, and biological evaluation of novel SMN protein modulators. J. Med. Chem. 54:6215–33
    [Google Scholar]
  148. 148.  Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS et al. 2007. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447:864–68
    [Google Scholar]
  149. 149.  Zhang C, Bollag G 2010. Scaffold-based design of kinase inhibitors for cancer therapy. Curr. Opin. Genet. Dev. 20:79–86
    [Google Scholar]
  150. 150.  Zhang JH, Chung TD, Oldenburg KR 1999. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J. Biomol. Screen. 4:67–73
    [Google Scholar]
  151. 151.  Zhang N, Bailus BJ, Ring KL, Ellerby LM 2016. iPSC-based drug screening for Huntington's disease. Brain Res 1638:42–56
    [Google Scholar]
  152. 152.  Zhang X, Smith DL, Meriin AB, Engemann S, Russel DE et al. 2005. A potent small molecule inhibits polyglutamine aggregation in Huntington's disease neurons and suppresses neurodegeneration in vivo. PNAS 102:892–97
    [Google Scholar]
  153. 153.  Zimmermann J, Caravatti G, Mett H, Meyer T, Muller M et al. 1996. Phenylamino-pyrimidine (PAP) derivatives: a new class of potent and selective inhibitors of protein kinase C (PKC). Arch. Pharm. 329:371–76
    [Google Scholar]
/content/journals/10.1146/annurev-genom-083117-021452
Loading
/content/journals/10.1146/annurev-genom-083117-021452
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error