667
Views
2
CrossRef citations to date
0
Altmetric
Review

Protein–Protein Interactions as Drug Targets

&
Pages 2195-2219 | Published online: 29 Oct 2015

References

  • Milroy L-G , GrossmannTN, HennigS, BrunsveldL, OttmannC. Modulators of protein–protein interactions. Chem. Rev.114, 4695–4748 (2014).
  • Arkin MR , TangY, WellsJA. Small-molecule inhibitors of protein–protein interactions: progressing toward the reality. Chem. Biol.21 (9), 1102–1114 (2014).
  • Kuenemann MA , SperandioO, LabbéCM, LagorceD, MitevaMA, VilloutreixBO. In silico design of low molecular weight protein–protein interaction inhibitors: overall concept and recent advances. Prog. Biophys. Mol. Biol. doi:10.1016/j.pbiomolbio.2015.02.006 (2015) ( Epub ahead of print).
  • Laraia L , McKenzieG, SpringDR, VenkitaramanAR, HugginsDJ. Overcoming chemical, biological, and computational challenges in the development of inhibitors targeting protein–protein interactions. Chem. Biol.22 (6), 689–703 (2015).
  • Venkatesan K , RualJ, VazquezAet al. An empirical framework for binary interactome mapping. Nat. Methods6 (1), 83–90 (2008).
  • Stumpf M , ThorneT, de SilvaEet al. Estimating the size of the human interactome. Proc. Natl Acad. Sci. USA105, 6959–6964 (2008).
  • Zhang QC , PetreyD, DengLet al. Structure-based prediction of protein–protein interactions on a genome-wide scale. Nature490 (7421), 556–560 (2012).
  • Tse C , ShoemakerAR, AdickesJet al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res.68 (9), 3421–3428 (2008).
  • Cleary JM , LimaCMSR, HurwitzHIet al. A Phase I clinical trial of navitoclax, a targeted high-affinity Bcl-2 family inhibitor, in combination with gemcitabine in patients with solid tumors. Invest. New Drugs32, 937–945 (2014).
  • Vassilev LT , VuBT, GravesBet al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science303 (5659), 844–848 (2004).
  • Biswas S , KillickE. The clinical development of p53-reactivating drugs in sarcomas-charting future therapeutic approaches and understanding the clinical molecular toxicology of nutlins. Expert Opin. Investig. Drugs23 (5), 692–645 (2014).
  • Filippakopoulos P , KnappS. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat. Rev. Drug Discov.13 (5), 337–356 (2014).
  • Higueruelo AP , JubbH, BlundellTL. Protein-protein interactions as druggable targets: recent technological advances. Curr. Opin. Pharmacol.13 (5), 791–796 (2013).
  • Uvebrant K , da Graça ThrigeD, RosénAet al. Discovery of selective small-molecule CD80 inhibitors. J. Biomol. Screen.12 (4), 464–472 (2007).
  • Sun H , LuJ, LiuLet al. Nonpeptidic and potent small-molecule inhibitors of cIAP-1/2 and XIAP proteins. J. Med. Chem.53 (17), 6361–6367 (2010).
  • Kwon HS , KimD-R, YangEGet al. Inhibition of VEGF transcription through blockade of the hypoxia inducible factor-1α-p300 interaction by a small molecule. Bioorg. Med. Chem. Lett.22 (16), 5249–5252 (2012).
  • Molzan M , KasperS, RöglinLet al. Stabilization of physical RAF/14–3–3 interaction by cotylenin A as treatment strategy for RAS mutant cancers. ACS Chem. Biol.8, 1869–1875 (2013).
  • Ottmann C , WeyandM, SassaTet al. A structural rationale for selective stabilization of anti-tumor interactions of 14–3–3 proteins by cotylenin A. J. Mol. Biol.386 (4), 913–919 (2009).
  • Thiel P , KaiserM, OttmannC. Small-molecule stabilization of protein–protein interactions: an underestimated concept in drug discovery?Angew. Chem. Int. Ed. Engl.51 (9), 2012–2018 (2012).
  • Jin L , WangW, FangG. Targeting protein–protein interaction by small molecules. Annu. Rev. Pharmacol. Toxicol.54, 435–456 (2014).
  • Bier D , ThielP, BrielsJ, OttmannC. Stabilization of protein–protein interactions in chemical biology and drug discovery. Prog. Biophys. Mol. Biol. doi:10.1016/j.pbiomolbio.2015.05.002 (2015) ( Epub ahead of print).
  • Allen JG , BourbeauMP, WohlhieterGEet al. Discovery and optimization of chromenotriazolopyrimidines as potent inhibitors of the mouse double minute 2-tumor protein 53 protein–protein interaction. J. Med. Chem.52 (22), 7044–7053 (2009).
  • Koblish HK , ZhaoS, FranksCFet al. Benzodiazepinedione inhibitors of the Hdm2:p53 complex suppress human tumor cell proliferation in vitro and sensitize tumors to doxorubicin in vivo. Mol. Cancer Ther.5 (1), 160–169 (2006).
  • Shangary S , QinD, McEachernDet al. Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition. Proc. Natl Acad. Sci. USA105 (10), 3933–3938 (2008).
  • Czarna A , BeckB, SrivastavaSet al. Robust generation of lead compounds for protein–protein interactions by computational and MCR chemistry: p53/Hdm2 antagonists. Angew. Chem. Int. Ed. Engl.49 (31), 5352–5356 (2010).
  • Davey DD , AdlerM, ArnaizDet al. Design, synthesis, and activity of 2-imidazol-1-ylpyrimidine derived inducible nitric oxide synthase dimerization inhibitors. J. Med. Chem.50 (6), 1146–1157 (2007).
  • Chu G-H , Le BourdonnecB, GuMet al. Design and synthesis of imidazopyrimidine derivatives as potent iNOS dimerization inhibitors. Open Med Chem J.3, 8–13 (2009).
  • Ohtsuka M , KonnoF, HondaHet al. PPA250 [3-(2,4-Difluorophenyl)-6-{2-[4-(1H-imidazol-1-ylmethyl) phenoxy] ethoxy}-2-phenylpyridine], a novel orally effective inhibitor of the dimerization of inducible nitric-oxide synthase, exhibits an anti-inflammatory effect in animal models of chronic. J. Pharmacol. Exp. Ther.303 (1), 52–57 (2002).
  • Erbe DV , WangS, XingY, TobinJF. Small molecule ligands define a binding site on the immune regulatory protein B7.1. J. Biol. Chem.277 (9), 7363–7368 (2002).
  • Yoakim C , OgilvieWW, GoudreauNet al. Discovery of the first series of inhibitors of human papillomavirus type 11: inhibition of the assembly of the E1–E2–Origin DNA complex. Bioorg. Med. Chem. Lett.13 (15), 2539–2541 (2003).
  • Deng Y , ShippsGW, ZhaoLet al. Modulating the interaction between CDK2 and cyclin A with a quinoline-based inhibitor. Bioorg. Med. Chem. Lett.24 (1), 199–203 (2014).
  • Benicchi T , IozziS, SvahnAet al. A homogeneous HTRF assay for the identification of inhibitors of the TWEAK-Fn14 protein interaction. J. Biomol. Screen.17 (7), 933–945 (2012).
  • Kenny CH , DingW, KelleherKet al. Development of a fluorescence polarization assay to screen for inhibitors of the FtsZ/ZipA interaction. Anal. Biochem.323 (2), 224–233 (2003).
  • Von Kleist L , StahlschmidtW, BulutHet al. Role of the clathrin terminal domain in regulating coated pit dynamics revealed by small molecule inhibition. Cell146 (3), 471–484 (2011).
  • Moerke NJ , AktasH, ChenHet al. Small-molecule inhibition of the interaction between the translation initiation factors eIF4E and eIF4G. Cell128 (2), 257–267 (2007).
  • Papadopoulos E , JenniS, KabhaEet al. Structure of the eukaryotic translation initiation factor eIF4E in complex with 4EGI-1 reveals an allosteric mechanism for dissociating eIF4G. Proc. Natl Acad. Sci. USA111 (31), E3187–E3195 (2014).
  • Mehraein-Ghomi F , KegelSJ, ChurchDRet al. Targeting androgen receptor and JunD interaction for prevention of prostate cancer progression. Prostate74 (7), 792–803 (2014).
  • Miyata Y , ChangL, BainorAet al. High-throughput screen for Escherichia coli heat shock protein 70 (Hsp70/DnaK): ATPase assay in low volume by exploiting energy transfer. J. Biomol. Screen.15 (10), 1211–1219 (2010).
  • Wisén S , BertelsenE, ThompsonAet al. Binding of a small molecule at a protein–protein interface regulates the chaperone activity of hsp70–hsp40. ACS Chem. Biol.5 (6), 611–622 (2010).
  • Zimmermann G , PapkeB, IsmailSet al. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature497 (7451), 638–642 (2013).
  • Gautier B , MitevaM a, GoncalvesVet al. Targeting the proangiogenic VEGF-VEGFR protein–protein interface with drug-like compounds by in silico and in vitro screening. Chem. Biol.18 (12), 1631–1639 (2011).
  • Chen Z , RuanQ, HanSet al. Discovery of structure-based small molecular inhibitor of αB-crystallin against basal-like/triple-negative breast cancer development in vitro and in vivo. Breast Cancer Res. Treat.145 (1), 45–59 (2014).
  • Starzec A , MitevaMA, LadamP, VilloutreixBO, PerretGY. Discovery of novel inhibitors of vascular endothelial growth factor-A-Neuropilin-1 interaction by structure-based virtual screening. Bioorg. Med. Chem.22 (15), 4042–4048 (2014).
  • Liu W-Q , MegaleV, BorrielloLet al. Synthesis and structure–activity relationship of non-peptidic antagonists of neuropilin-1 receptor. Bioorg. Med. Chem. Lett.24 (17), 4254–4259 (2014).
  • Wang G , WangX, YuHet al. Small-molecule activation of the TRAIL receptor DR5 in human cancer cells. Nat. Chem. Biol.9 (2), 84–89 (2013).
  • Song Y , Margolles-ClarkE, BayerA, BuchwaldP. Small-molecule modulators of the OX40-OX40 ligand co-stimulatory protein–protein interaction. Br. J. Pharmacol.117 (21), 4955–4969 (2014).
  • Rose R , ErdmannS, BovensSet al. Identification and structure of small-molecule stabilizers of 14–3–3 protein–protein interactions. Angew. Chem. Int. Ed. Engl.49 (24), 4129–4132 (2010).
  • Graves B , ThompsonT, XiaMet al. Activation of the p53 pathway by small-molecule-induced MDM2 and MDMX dimerization. Proc. Natl Acad. Sci. USA109 (29), 11788–11793 (2012).
  • Malashkevich VN , DulyaninovaNG, RamagopalUAet al. Phenothiazines inhibit S100A4 function by inducing protein oligomerization. Proc. Natl Acad. Sci. USA107 (19), 8605–8610 (2010).
  • Gerritz SW , CianciC, KimSet al. Inhibition of influenza virus replication via small molecules that induce the formation of higher-order nucleoprotein oligomers. Proc. Natl Acad. Sci. USA108 (37), 15366–15371 (2011).
  • Classen S , OllandS, BergerJ. Structure of the topoisomerase II ATPase region and its mechanism of inhibition by the chemotherapeutic agent ICRF-187. Proc. Natl Acad. Sci. USA100 (19), 543–549 (2003).
  • Bulawa CE , ConnellyS, DevitMet al. Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc. Natl Acad. Sci. USA109 (24), 9629–9634 (2012).
  • Zhang M , PascalJM, ZhangJ-F. Unstructured to structured transition of an intrinsically disordered protein peptide in coupling Ca2+-sensing and SK channel activation. Proc. Natl Acad. Sci. USA110 (12), 4828–4833 (2013).
  • Jose RA , VoetA, BroosKet al. An integrated fragment based screening approach for the discovery of small molecule modulators of the VWF-GPIbα interaction. Chem. Commun. (Camb.)48 (92), 11349–11351 (2012).
  • Villoutreix BO , LabbéC, LagorceD, LacondeG, SperandioO. A leap into the chemical space of protein–protein interaction inhibitors. Curr. Pharm. Des.18 (30), 4648–4667 (2012).
  • Metz A , CigliaE, GohlkeH. Modulating protein–protein interactions: from structural determinants of binding to druggability prediction to application. Curr. Pharm. Des.18 (30), 4630–4647 (2012).
  • Aeluri M , ChamakuriS, DasariBet al. Small molecule modulators of protein–protein interactions: selected case studies. Chem. Rev.114 (9), 4640–4694 (2014).
  • Griffith OW , StuehrDJ. Nitric oxide synthases: properties and catalytic mechanism. Annu. Rev. Physiol.57, 707–736 (1995).
  • Stuehr DJ . Mammalian nitric oxide synthases. Biochim. Biophys. Acta1411 (2–3), 217–230 (1999).
  • O'dell T , HuangP, DawsonTet al. Endothelial NOS and the blockade of LTP by NOS inhibitors in mice lacking neuronal NOS. Science265, 542–546 (1994).
  • Moncada S , HiggsE. Molecular mechanisms and therapeutic strategies related to nitric oxide. FASEB J.9, 1319–1330 (1995).
  • Kröncke K , FehselK, Kolb-BachofenV. Inducible nitric oxide synthase and its product nitric oxide, a small molecule with complex biological activities. Biol. Chem. Hoppe Seyler376 (6), 327–343 (1995).
  • Ghosh DK , StuehrDJ. Macrophage NO synthase: characterization of isolated oxygenase and reductase domains reveals a head-to-head subunit interaction. Biochemistry34 (3), 801–807 (1995).
  • Alderton WK , CooperCE, KnowlesRG. Nitric oxide synthases: structure, function and inhibition. Biochem. J.357 (Pt 3), 593–615 (2001).
  • Siddhanta U , PrestaA, FanB, WolanD, RousseauDL, StuehrDJ. Domain swapping in inducible nitric-oxide synthase: elektron transfer occurs between flavin and heme groups located on adjecent subjunits in the dimer domain. J. Biol. Chem.273, 18950–18958 (1998).
  • Vallance P , LeiperJ. Blocking NO synthesis: how, where and why?Nat. Rev. Drug Discov.1 (12), 939–950 (2002).
  • Bryk R , WolffD. Mechanism of inducible nitric oxide synthase inactivation by aminoguanidine and L-N6-(1-Iminoethyl)lysine. Biochemistry37 (97), 4844–4852 (1998).
  • Edwards RM , StackEJ, TriznaW. Interaction of l-arginine analogs with l-arginine uptake in rat renal brush border membrane vesicles. J. Pharmacol. Exp. Ther.285 (3), 1019–1022 (1998).
  • Sennequier N , WolanD, StuehrD. Antifungal imidazoles block assembly of inducible NO synthase into an active dimer. J. Biol. Chem.274 (2), 930–938 (1999).
  • McMillan K , AdlerM, AuldDSet al. Allosteric inhibitors of inducible nitric oxide synthase dimerization discovered via combinatorial chemistry. Proc. Natl Acad. Sci. USA97 (4), 1506–1511 (2000).
  • Sohn M-J , HurG-M, ByunH-S, KimW-G. Cyclo(dehydrohistidyl-l-tryptophyl) inhibits nitric oxide production by preventing the dimerization of inducible nitric oxide synthase. Biochem. Pharmacol.75 (4), 923–930 (2008).
  • Zhang W , RamamoorthyY, KilicarslanT, NolteH, TyndaleR, SellersE. Inhibition of cytochromes P450 by antifungal imidazole derivatives. Drug Metab. Dispos.30 (3), 314–318 (2002).
  • Bonnefous C , PayneJE, RoppeJet al. Discovery of inducible nitric oxide synthase (iNOS) inhibitor development candidate KD7332, part 1: Identification of a novel, potent, and selective series of quinolinone iNOS dimerization inhibitors that are orally active in rodent pain models. J. Med. Chem.52 (9), 3047–3062 (2009).
  • Symons KT , MassariME, NguyenPMet al. KLYP956 Is a non-imidazole-based orally active inhibitor of nitric-oxide synthase dimerization. Mol. Pharmacol.76 (1), 153–162 (2009).
  • Nagpal L , HaqueMM, SahaAet al. Mechanism of inducible nitric-oxide synthase dimerization inhibition by novel pyrimidine imidazoles. J. Biol. Chem.288 (27), 19685–19697 (2013).
  • Harris ES , McIntyreTM, PrescottSM, ZimmermanGA. The leukocyte integrins. J. Biol. Chem.275 (31), 23409–23412 (2000).
  • Reilly P , WoskaJJ, JeanfavreD, McNallyE, RothleinR, BormannB. The native structure of intercellular adhesion molecule-1 (ICAM-1) is a dimer. Correlation with binding to LFA-1. J. Immunol.155, 529–532 (1995).
  • Butcher EC . Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity. Cell67, 1033–1036 (1991).
  • Grakoui A . The immunological synapse: a molecular machine controlling T cell activation. Science285 (5425), 221–227 (1999).
  • Shimaoka M , SpringerTA. Therapeutic antagonists and the conformational regulation of the β2 integrins. Curr. Top. Med. Chem.4, 1485–1495 (2004).
  • Yonekawa K , HarlanJM. Targeting leukocyte integrins in human diseases. Exp. Cell. Res.77, 129–140 (2005).
  • Miele M , BennettC, MillerB, WelchD. Enhanced metastativ ability of TNF-α-treated malighangt melanoma cells is reduced by intercellular adhesion molecule-1 (ICAM-1, CD54) antisense oligonucleotides. Exp. Cell. Res.214, 231–241 (1994).
  • Cather JC , CatherJC, MenterA. Modulating T cell responses for the treatment of psoriasis: a focus on efalizumab. Expert Opin. Biol. Ther.3 (2), 361–370 (2003).
  • Gauvreau GM , BeckerAB, BouletL-Pet al. The effects of an anti-CD11a mAb, efalizumab, on allergen-induced airway responses and airway inflammation in subjects with atopic asthma. J. Allergy Clin. Immunol.112 (2), 331–338 (2003).
  • Zimmerman T , BlancoFJ. Inhibitors targeting the LFA-1/ICAM-1 cell-adhesion interaction: design and mechanism of action. Curr. Pharm. Des.14 (22), 2128–2139 (2008).
  • Kallen J , WelzenbachK, RamagePet al. Structural basis for LFA-1 inhibition upon lovastatin binding to the CD11a I-domain. J. Mol. Biol.292 (1), 1–9 (1999).
  • Weitz-Schmidt G , WelzenbachK, DawsonJ, KallenJ. Improved lymphocyte function-associated antigen-1 (LFA-1) inhibition by statin derivatives: molecular basis determined by x-ray analysis and monitoring of LFA-1 conformational changes in vitro and ex vivo. J. Biol. Chem.279 (45), 46764–47771 (2004).
  • Kelly TA , JeanfavreDD, McNeilDWet al. Cutting edge: a small molecule antagonist of LFA-1-mediated cell adhesion. J. Immunol.163 (10), 5173–5177 (1999).
  • Last-Barney K , DavidsonW, CardozoMet al. Binding site elucidation of hydantoin-based antagonists of LFA-1 using multidisciplinary technologies: evidence for the allosteric inhibition of a protein–protein interaction. J Am Chem Soc.123 (24), 5643–5650 (2001).
  • Wu J-P , EmeighJ, GaoD aet al. Second-generation lymphocyte function-associated antigen-1 inhibitors: 1H-imidazo[1,2-alpha]imidazol-2-one derivatives. J. Med. Chem.47 (22), 5356–5366 (2004).
  • Panzenbeck MJ , JeanfavreDD, KellyT aet al. An orally active, primate selective antagonist of LFA-1 inhibits delayed-type hypersensitivity in a humanized-mouse model. Eur. J. Pharmacol.534 (1–3), 233–240 (2006).
  • Potin D , LaunayM, NicolaiEet al. De novo design, synthesis, and in vitro activity of LFA-1 antagonists based on a bicyclic[5.5]hydantoin scaffold. Bioorg. Med. Chem. Lett.15 (4), 1161–1164 (2005).
  • Dodd DS , SheriffS, ChangCJet al. Design of LFA-1 antagonists based on a 2,3-dihydro-1H-pyrrolizin-5(7aH)-one scaffold. Bioorg. Med. Chem. Lett.17 (7), 1908–1911 (2007).
  • Potin D , LaunayM, MonatlikFet al. Discovery and development of 5-[(5S,9R)-9-(4-cyanophenyl)-3-(3,5-dichlorophenyl)-1-methyl-2,4-dioxo-1,3,7-triazaspiro[4.4]non- 7-yl-methyl]-3-thiophenecarboxylic Acid (BMS-587101)-A small molecule antagonist of leukocyte function associated antigen-1. J. Med. Chem.49, 6946–6949 (2006).
  • Watterson SH , XiaoZ, DoddDSet al. Small molecule antagonist of leukocyte function associated antigen-1 (LFA-1): structure–activity relationships leading to the identification of 6-((5S,9R)-9-(4-cyanophenyl)-3-(3,5-dichlorophenyl)-1-methyl-2,4-dioxo-1,3,7-triazaspiro[4.4]nonan-7-yl)nicotin. J. Med. Chem.53 (9), 3814–3830 (2010).
  • Wattanasin S , AlbertR, EhrhardtCet al. 1,4-Diazepane-2-ones as novel inhibitors of LFA-1. Bioorg. Med. Chem. Lett.13 (3), 499–502 (2003).
  • Wattanasin S , KallenJ, MyersSet al. 1,4-Diazepane-2,5-diones as novel inhibitors of LFA-1. Bioorg. Med. Chem. Lett.15 (4), 1217–1220 (2005).
  • Liu G , LinkJT, PeiZet al. Discovery of novel p-Arylthio cinnamides as antagonists of leukocyte identification of an additional binding pocket based on an anilino diaryl sulfide lead. J. Med. Chem.43, 4025–4040 (2000).
  • Winn M , ReillyEB, LiuGet al. Discovery of Nnovel p-Arylthio cinnamides as antagonists of leukocyte structure–activity relationship of substituents on the benzene ring of the cinnamide. J. Med. Chem.44 (Scheme 4), 4393–4403 (2001).
  • Guckian KM , LinEY-S, SilvianL, FriedmanJE, ChinD, ScottDM. Design and synthesis of a series of meta aniline-based LFA-1 ICAM inhibitors. Bioorg. Med. Chem. Lett.18 (19), 5249–5251 (2008).
  • Lin EY-S , GuckianKM, SilvianLet al. Structure–activity relationship of ortho- and meta-phenol based LFA-1 ICAM inhibitors. Bioorg. Med. Chem. Lett.18 (19), 5245–5248 (2008).
  • Shoda M , HaradaT, YanoKet al. Virtual screening leads to the discovery of an effective antagonist of lymphocyte function-associated antigen-1. ChemMedChem2 (4), 515–521 (2007).
  • Gadek TR , BurdickDJ, McDowellRSet al. Generation of an LFA-1 antagonist by the transfer of the ICAM-1 immunoregulatory epitope to a small molecule. Science295 (5557), 1086–9 (2002).
  • San Sebastián E , ZimmermanT, ZubiaAet al. Design, synthesis, and functional evaluation of leukocyte function associated antigen-1 antagonists in early and late stages of cancer development. J. Med. Chem.56, 735–747 (2013).
  • Kollmann CS , BaiX, TsaiC-Het al. Application of encoded library technology (ELT) to a protein–protein interaction target: discovery of a potent class of integrin lymphocyte function-associated antigen 1 (LFA-1) antagonists. Bioorg. Med. Chem.22 (7), 2353–2365 (2014).
  • Hintersteiner M , KallenJ, SchmiedMet al. Identification and X-ray co-crystal structure of a small-molecule activator of LFA-1-ICAM-1 binding. Angew. Chem. Int. Ed. Engl.53 (17), 4322–4326 (2014).
  • Zarbock A , KempfT, WollertKC, VestweberD. Leukocyte integrin activation and deactivation: novel mechanisms of balancing inflammation. J. Mol. Med. (Berl.).90 (4), 353–359 (2012).
  • Gardino A , YaffeM. 14–3–3 proteins as signaling integration points for cell cycle control and apoptosis. Semin. Cell. Dev. Biol.22 (7), 688–695 (2011).
  • Robinson DN . 14–3–3, an integrator of cell mechanics and cytokinesis. Small GTPases1 (3), 165–169 (2010).
  • Messaritou G , GrammenoudiS, SkoulakisEMC. Dimerization is essential for 14–3–3zeta stability and function in vivo. J. Biol. Chem.285 (3), 1692–1700 (2010).
  • Morton DG , ShakesDC, NugentSet al. The caenorhabditis elegans par-5 gene encodes a 14–3–3 protein required for cellular asymmetry in the early embryo. Dev. Biol.241 (1), 47–58 (2002).
  • Aitken A . 14–3–3 proteins: a historic overview. Semin. Cancer Biol.16 (3), 162–172 (2006).
  • De Boer AH , van KleeffPJM, GaoJ. Plant 14–3–3 proteins as spiders in a web of phosphorylation. Protoplasma250 (2), 425–440 (2013).
  • Bridges D , MoorheadGBG. 14–3–3 proteins: a number of functions for a numbered protein. Sci STKE.2005 (296), re10 (2005).
  • Yaffe M , RittingerK, VoliniaSet al. The structural basis for 14–3–3: phosphopeptide binding specificity. Cell91, 961–971 (1997).
  • Coblitz B , WuM, ShikanoS, LiM. C-terminal binding: an expanded repertoire and function of 14–3–3 proteins. FEBS Lett.580 (6), 1531–1535 (2006).
  • Johnson C , CrowtherS, StaffordMJ, CampbellDG, TothR, MacKintoshC. Bioinformatic and experimental survey of 14–3–3-binding sites. Biochem. J.427 (1), 69–78 (2010).
  • Vassilev A , KanekoK, ShuH, ZhaoY, DePamphilisM. TEAD/TEF transcription factors utilize the activation domain of YAP65, a Src/Yes-associated protein localized in the cytoplasm. Genes Dev.15 (10), 1229–1241 (2001).
  • Schumacher B , SkwarczynskaM, RoseR, OttmannC. Structure of a 14–3–3σ-YAP phosphopeptide complex at 1.15 A resolution. Acta Crystallogr. Sect. F. Struct. Biol. Cryst. Commun.66 (Pt 9), 978–984 (2010).
  • Obsilova V , NedbalkovaE, SilhanJet al. The 14–3–3 protein affects the conformation of the regulatory domain of human tyrosine hydroxylase. Biochemistry47 (6), 1768–1777 (2008).
  • Dumaz N , MaraisR. Protein kinase A blocks Raf-1 activity by stimulating 14–3–3 binding and blocking Raf-1 interaction with RAS. J. Biol. Chem.278 (32), 29819–29823 (2003).
  • Molzan M , OttmannC. Synergistic binding of the phosphorylated S233- and S259-binding sites of C-RAF to one 14–3–3ζ dimer. J. Mol. Biol.423 (4), 486–495 (2012).
  • Liang X , Da PaulaAC, BozókyZet al. Phosphorylation-dependent 14–3–3 protein interactions regulate CFTR biogenesis. Mol. Biol. Cell23 (6), 996–1009 (2012).
  • Johnson C , TintiM, WoodNTet al. Visualization and biochemical analyses of the emerging mammalian 14–3–3-phosphoproteome. Mol Cell Proteomics10 (10), M110.005751 (2011).
  • Freeman A , MorrisonD. 14–3–3 proteins: diverse functions in cell proliferation and cancer progression. Semin. Cell. Dev. Biol.22 (7), 681–687 (2011).
  • Freed E , SymonsM, MacdonaldSG, McCormickF, RuggieriR. Binding of 14–3–3 proteins to the protein kinase Raf and effects on its activation. Science265 (5179), 1713–1716 (1994).
  • Conklin D , GalaktionovK, BeachD. 14–3–3 proteins associate with Cdc25 phosphatases. Proc. Natl Acad. Sci. USA92 (17), 7892–7896 (1995).
  • Rajagopalan S , JaulentAM, WellsM, VeprintsevDB, FershtAR. 14–3–3 activation of DNA binding of p53 by enhancing its association into tetramers. Nucleic Acids Res.36 (18), 5983–5991 (2008).
  • Schumacher B , MondryJ, ThielP, WeyandM, OttmannC. Structure of the p53 C-terminus bound to 14–3–3: implications for stabilization of the p53 tetramer. FEBS Lett.584 (8), 1443–1448 (2010).
  • Basu S , TottyNF, IrwinMS, SudolM, DownwardJ. Akt phosphorylates the Yes-associated protein, YAP, to induce interaction with 14–3–3 and attenuation of p73-mediated apoptosis. Mol. Cell.11 (1), 11–23 (2003).
  • Sluchanko NN , Seit-NebiAS, GusevNB. Phosphorylation of more than one site is required for tight interaction of human tau protein with 14–3–3zeta. FEBS Lett.583 (17), 2739–2742 (2009).
  • Plotegher N , KumarD, TessariIet al. The chaperone-like protein 14–3–3η interacts with human α-synuclein aggregation intermediates rerouting the amyloidogenic pathway and reducing α-synuclein cellular toxicity. Hum. Mol. Genet.23 (21), 5615–5629 (2014).
  • Dzamko N , DeakM, HentatiFet al. Inhibition of LRRK2 kinase activity leads to dephosphorylation of Ser(910)/Ser(935), disruption of 14–3–3 binding and altered cytoplasmic localization. Biochem. J.430 (3), 405–413 (2010).
  • Sato S , ChibaT, SakataEet al. 14–3–3eta is a novel regulator of parkin ubiquitin ligase. EMBO J.25 (1), 211–221 (2006).
  • Ramm G , LaranceM, GuilhausM, JamesDE. A role for 14–3–3 in insulin-stimulated GLUT4 translocation through its interaction with the RabGAP AS160. J. Biol. Chem.281 (39), 29174–29180 (2006).
  • Pierrat B , ItoM, HinzWet al. Uncoupling proteins 2 and 3 interact with members of the 14.3.3 family. Eur. J. Biochem.267 (9), 2680–2687 (2000).
  • Fu H , CoburnJ, CollierRJ. The eukaryotic host factor that activates exoenzyme S of Pseudomonas aeruginosa is a member of the 14–3–3 protein family. Proc. Natl Acad. Sci. USA90 (6), 2320–2324 (1993).
  • Ottmann C , YasminL, WeyandMet al. Phosphorylation-independent interaction between 14–3–3 and exoenzyme S: from structure to pathogenesis. EMBO J.26 (3), 902–913 (2007).
  • Glas A , BierD, HahneG, RademacherC, OttmannC, GrossmannTN. Constrained peptides with target-adapted cross-links as inhibitors of a pathogenic protein–protein interaction. Angew. Chem. Int. Ed. Engl.53 (9), 2489–2493 (2014).
  • Wilker E , YaffeMB. 14–3–3 proteins – a focus on cancer and human disease. J. Mol. Cell. Cardiol.37 (3), 633–642 (2004).
  • Aitken A , BaxterH, DuboisTet al. Specificity of 14–3–3 isoform dimer interactions and phosphorylation. Biochem. Soc. Trans.30, 351–360 (2002).
  • Mackintosh C . Dynamic interactions between 14–3–3 proteins and phosphoproteins regulate diverse cellular processes. Biochem. J.381 (Pt 2), 329–342 (2004).
  • Zhao J , MeyerkordCL, DuY, KhuriFR, FuH. 14–3–3 proteins as potential therapeutic targets. Semin. Cell. Dev. Biol.22 (7), 705–712 (2011).
  • Zhao J , DuY, HortonJRet al. Discovery and structural characterization of a small molecule 14–3–3 protein–protein interaction inhibitor. Proc. Natl Acad. Sci. USA108 (39), 16212–16216 (2011).
  • Mori M , VignaroliG, BottaM. Small molecules modulation of 14–3–3 protein–protein interactions. Drug Discov. Today Technol.10 (4), e541–e547 (2013).
  • Camoni L , Di LucenteC, ViscontiS, AducciP. The phytotoxin fusicoccin promotes platelet aggregation via 14–3–3-glycoprotein Ib-IX-V interaction. Biochem. J.436 (2), 429–436 (2011).
  • Ottmann C . Small-molecule modulators of 14–3–3 protein–protein interactions. Bioorg. Med. Chem.21, 4058–4062 (2013).
  • Milroy L-G , BrunsveldL, OttmannC. Stabilization and inhibition of protein–protein interactions: the 14–3–3 case study. ACS Chem. Biol.8 (1), 27–35 (2013).
  • Wu H , GeJ, YaoSQ. Microarray-assisted high-throughput identification of a cell-permeable small-molecule binder of 14–3–3 proteins. Angew. Chem. Int. Ed. Engl.49 (37), 6528–6532 (2010).
  • Arrendale A , KimK, ChoiJY, LiW, GeahlenRL, BorchRF. Synthesis of a phosphoserine mimetic prodrug with potent 14–3–3 protein inhibitory activity. Chem. Biol.19 (6), 764–771 (2012).
  • Röglin L , ThielP, KohlbacherO, OttmannC. Covalent attachment of pyridoxal-phosphate derivatives to 14–3–3 proteins. Proc Natl Acad Sci USA.109 (18), E1051–E1053; author reply E1054 (2012).
  • Thiel P , RöglinL, MeissnerN, HennigS, KohlbacherO, OttmannC. Virtual screening and experimental validation reveal novel small-molecule inhibitors of 14–3–3 protein–protein interactions. Chem. Commun. (Camb.)49 (76), 8468–8470 (2013).
  • Corradi V , ManciniM, ManettiF, PettaS, SantucciMA, BottaM. Identification of the first non-peptidic small molecule inhibitor of the c-Abl/14–3–3 protein–protein interactions able to drive sensitive and Imatinib-resistant leukemia cells to apoptosis. Bioorg. Med. Chem. Lett.20 (20), 6133–6137 (2010).
  • Yan Y-M , DaiH-Q, DuYet al. Identification of blapsins A and B as potent small-molecule 14–3–3 inhibitors from the insect Blaps japanensis. Bioorg. Med. Chem. Lett.22 (12), 4179–4181 (2012).
  • Woodcock JM , CoolenC, GoodwinKLet al. Destabilisation of dimeric 14–3–3 proteins as a novel approach to anti-cancer therapeutics. Oncotarget.6 (16), 14522–14536 (2015).
  • Fullone MR , ViscontiS, MarraM, FoglianoV, AducciP. Fusicoccin effect on the in vitro interaction between plant 14–3–3 proteins and plasma membrane H+-ATPase. J. Biol. Chem.273 (13), 7698–7702 (1998).
  • Ottmann C , MarcoS, JaspertNet al. Structure of a 14–3–3 coordinated hexamer of the plant plasma membrane H+-ATPase by combining X-ray crystallography and electron cryomicroscopy. Mol. Cell.25 (3), 427–440 (2007).
  • Richter A , RoseR, HedbergC, WaldmannH, OttmannC. An optimised small-molecule stabiliser of the 14–3–3-PMA2 protein–protein interaction. Chemistry18, 6520–6527 (2012).
  • De Vries-van Leeuwen IJ , da Costa PereiraD, FlachKDet al. Interaction of 14–3–3 proteins with the estrogen receptor alpha F domain provides a drug target interface. Proc. Natl Acad. Sci. USA110, 8894–8899 (2013).
  • Anders C , HiguchiY, KoschinskyKet al. A semisynthetic fusicoccane stabilizes a protein–protein interaction and enhances the expression of K+ channels at the cell surface. Chem. Biol.20 (4), 583–593 (2013).
  • Kasukabe T , Okabe-KadoJ, KatoN, SassaT, HonmaY. Effects of combined treatment with rapamycin and cotylenin A, a novel differentiation-inducing agent, on human breast carcinoma MCF-7 cells and xenografts. Breast Cancer Res.7 (6), R1097–R1110 (2005).
  • Jubb H , BlundellTL, AscherDB. Flexibility and small pockets at protein–protein interfaces: new insights into druggability. Prog. Biophys. Mol. Biol. doi:10.1016/j.pbiomolbio.2015.01.009 (2015) ( Epub ahead of print).
  • Dreze M , CharloteauxB, MilsteinSet al. “Edgetic” perturbation of a C. elegans BCL2 ortholog. Nat. Methods6 (11), 843–849 (2009).
  • Zhong Q , SimonisN, LiQ-Ret al. Edgetic perturbation models of human inherited disorders. Mol. Syst. Biol.5, 321 (2009).
  • Sahni N , YiS, ZhongQet al. Edgotype: a fundamental link between genotype and phenotype. Curr. Opin. Genet. Dev.23 (6), 649–657 (2013).
  • Csermely P , KorcsmárosT, KissHJM, LondonG, NussinovR. Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol. Ther.138 (3), 333–408 (2013).
  • Sahni N , YiS, TaipaleMet al. Widespread macromolecular interaction perturbations in human genetic disorders. Cell161 (3), 647–660 (2015).
  • Watson VG , DrakeKM, PengY, NapperAD. Development of a high-throughput screening-compatible assay for the discovery of inhibitors of the AF4-AF9 interaction using AlphaScreen technology. Assay Drug Dev. Technol.11 (4), 253–268 (2013).
  • Oltersdorf T , ElmoreSW, ShoemakerARet al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature435 (7042), 677–681 (2005).
  • Vo DD , GautierF, JuinP, GréeR. Structure–activity relationship of selected polyphenol derivatives as inhibitors of Bax/Bcl-xL interaction. Eur. J. Med. Chem.51, 286–293 (2012).
  • Corbel C , WangQ, BousserouelHet al. First BRET-based screening assay performed in budding yeast leads to the discovery of CDK5/p25 interaction inhibitors. Biotechnol. J.6 (7), 860–870 (2011).
  • Margolles-Clark E , Jacques-SilvaMC, GanesanLet al. Suramin inhibits the CD40–CD154 costimulatory interaction: A possible mechanism for immunosuppressive effects. Biochem. Pharmacol.77 (7), 1236–1245 (2009).
  • Margolles-Clark E , KenyonNS, RicordiC, BuchwaldP. Effective and specific inhibition of the CD40-CD154 costimulatory interaction by a naphthalenesulphonic acid derivative. Chem. Biol. Drug Des.76 (4), 305–313 (2010).
  • Silvian L , FriedmanJ, StrauchKet al. Small molecule inhibition of the TNF family cytokine CD40 ligand through a subunit fracture mechanism. ACS Chem. Biol.6 (6), 636–647 (2011).
  • Breuer S , EspinolaS, MorelliX, TorbettBE, AroldST, EngelsIH. A biochemical/biophysical assay Dyad for HTS-compatible triaging of inhibitors of the HIV-1 Nef/Hck SH3 interaction. Curr. Chem. Genomics Transl. Med.7, 16–20 (2013).
  • Thanos CD , DeLanoWL, WellsJA. Hot-spot mimicry of a cytokine receptor by a small molecule. Proc. Natl Acad. Sci. USA103 (42), 15422–15427 (2006).
  • Hu L , MageshS, ChenLet al. Discovery of a small-molecule inhibitor and cellular probe of Keap1–Nrf2 protein–protein interaction. Bioorg. Med. Chem. Lett.23 (10), 3039–3043 (2013).
  • Christ F , VoetA, MarchandAet al. Rational design of small-molecule inhibitors of the LEDGF/p75-integrase interaction and HIV replication. Nat. Chem. Biol.6 (6), 442–448 (2010).
  • Sun Q , BurkeJ, PhanJet al. Discovery of small molecules that bind to K-RAS and inhibit SOS-mediated activation. Angew. Chem. Int. Ed. Engl.51 (25), 6140–6143 (2012).
  • Cianci C , YuK, CombrinkK. Orally active fusion inhibitor of respiratory syncytial virus. Antimicrob. Agents Chemother.48, 413–422 (2004).
  • Andries K , MoeremansM, GeversTet al. Substituted benzimidazoles with nanomolar activity against respiratory syncytial virus. Antivir. Res.60 (3), 209–219 (2003).
  • Ungermannova D , LeeJ, ZhangG, DallmannHG, McHenryCS, LiuX. High-throughput screening AlphaScreen assay for identification of small-molecule inhibitors of ubiquitin E3 ligase SCF(Skp2-Cks1). J. Biomol. Screen.18 (8), 910–920 (2013).
  • Buller F , ZhangY, ScheuermannJ, SchäferJ, BühlmannP, NeriD. Discovery of TNF inhibitors from a DNA-encoded chemical library based on diels-alder cycloaddition. Chem. Biol.16 (10), 1075–1086 (2009).
  • Xie X , SunL, PessettoZYet al. Development of a fluorescence polarization based high-throughput assay to identify Casitas B-lineage lymphoma RING domain regulators. PLoS ONE8 (10), e78042 (2013).
  • Hamdan FF , AudetM, GarneauP, PelletierJ, BouvierM. High-throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1-based beta-arrestin2 recruitment assay. J. Biomol. Screen.10 (5), 463–475 (2005).
  • Kao RY , YangD, LauL-Set al. Identification of influenza A nucleoprotein as an antiviral target. Nat. Biotechnol.28 (6), 600–605 (2010).
  • Germain P , GaudonC, PogenbergVet al. Differential action on coregulator interaction defines inverse retinoid agonists and neutral antagonists. Chem. Biol.16 (5), 479–489 (2009).
  • Burns MC , SunQ, DanielsRNet al. Approach for targeting RAS with small molecules that activate SOS-mediated nucleotide exchange. Proc. Natl Acad. Sci. USA111 (9), 3401–3406 (2014).
  • Garrett S , HodgsonL, RybinAet al. A biosensor of S100A4 metastasis factor activation: inhibitor screening and cellular activation dynamics. Biochemistry47 (3), 986–996 (2008).
  • Zhang M , PascalJM, SchumannM, ArmenRS, ZhangJ-F. Identification of the functional binding pocket for compounds targeting small-conductance Ca2-activated potassium channels. Nat. Commun.3, 1021 (2012).

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.