+ All Categories
Home > Documents > DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each...

DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each...

Date post: 19-Jul-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
16
Small Molecule Therapeutics Discovery of New Small Molecules Targeting the Vitronectin- Binding Site of the Urokinase Receptor That Block Cancer Cell Invasion Vincenza Elena Anna Rea 1 , Antonio Lavecchia 2 , Carmen Di Giovanni 2 , Francesca Wanda Rossi 1 , Anna Gorrasi 3 , Ada Pesapane 1 , Amato de Paulis 1 , Pia Ragno 3 , and Nunzia Montuori 1 Abstract Besides focusing urokinase (uPA) proteolytic activity on the cell membrane, the uPA receptor (uPAR) is able to bind vitronectin, via a direct binding site. Furthermore, uPAR interacts with other cell surface receptors, such as integrins, receptor tyrosine kinases, and chemotaxis receptors, triggering cell-signaling pathways that promote tumor progression. The ability of uPAR to coordinate binding and degradation of extracellular matrix (ECM) and cell signaling makes it an attractive therapeutic target in cancer. We used structure-based virtual screening (SB-VS) to search for small molecules targeting the uPAR-binding site for vitronectin. Forty-one compounds were identified and tested on uPAR-negative HEK-293 epithelial cells transfected with uPAR (uPAR-293 cells), using the parental cell line transfected with the empty vector (V-293 cells) as a control. Compounds 6 and 37 selectively inhibited uPAR-293 cell adhesion to vitronectin and the resulting changes in cell morphology and signal transduction, without exerting any effect on V-293 cells. Compounds 6 and 37 inhibited uPAR-293 cell binding to vitronectin with IC 50 values of 3.6 and 1.2 mmol/L, respectively. Compounds 6 and 37 targeted S88 and R91, key residues for uPAR binding to vitronectin but also for uPAR interaction with the fMLF family of chemotaxis receptors (fMLF-Rs). As a consequence, compounds 6 and 37 impaired uPAR-293 cell migration toward fetal calf serum (FCS), uPA, and fMLF, likely by inhibiting the interaction between uPAR and FPR1, the high affinity fMLF-R. Both compounds blocked in vitro ECM invasion of several cancer cell types, thus representing new promising leads for pharmaceuticals in cancer. Mol Cancer Ther; 12(8); 1402–16. Ó2013 AACR. Introduction The urokinase (uPA)-mediated plasminogen activation system, which generates the potent serine-protease plas- min, is involved in various pathologic processes, includ- ing angiogenesis, inflammation, wound healing, and metastasis (1, 2). The key molecule of this system, the uPA receptor (uPAR), is anchored to the plasma membrane by a glyco- sylphosphatidylinositol moiety and is formed by three homologous domains (DI, DII, and DIII, from the N-ter- minus). The uPA-binding site is located in the DI domain, but the full-length molecule is required for an efficient binding (3, 4). uPAR enhances pericellular proteolysis by serving as a docking site to uPA, thus triggering a cascade of proteolytic events that leads to the active degradation of extracellular matrix (ECM) components (5). Despite the lack of a transmembrane domain, uPAR can activate intracellular signaling through lateral interac- tions with other cell surface receptors, such as integrins, receptor tyrosine kinases, and G-protein–coupled chemo- taxis receptors (6). uPAR ability to regulate integrin activity plays a key role in cell adhesion, migration, proliferation, and surviv- al (7, 8). Recently, integrin-binding sites have been iden- tified in uPAR domain DII (residues 130–142; ref. 9) and in uPAR domain DIII (residues 240–248; ref. 10). Moreover, uPAR cross-talk with EGF receptor (EGFR) is extensive and may regulate the shift from tumor cell dormancy to proliferation (11, 12). uPAR interaction with receptors for fMet-Leu-Phe (fMLF-Rs; ref. 13) is required for both uPA- and fMLF- dependent cell migration and occurs through a chemo- tactic domain located in the DI–DII linker region, the SRSRY sequence (amino acids, 88–92; ref. 14). Because a soluble cleaved form of uPAR, exposing at the N-terminus of the SRSRY sequence is a ligand for fMLF-Rs (15), it has been proposed that uPA binding to uPAR determines a conformational modification of the receptor with the Authors' Afliations: Departments of 1 Translational Medical Sciences and 2 Pharmacy, "Drug Discovery" Laboratory, "Federico II" University, Naples; and 3 Department of Chemistry and Biology, University of Salerno, Salerno, Italy V.E.A. Rea and A. Lavecchia equally contributed to this work. Corresponding Author: Nunzia Montuori, Department of Translational Medical Sciences, "Federico II" University, via S. Pansini, 5, 80131 Naples, Italy. Phone: 39-0817463309; Fax: 39-0817463308; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-12-1249 Ó2013 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 12(8) August 2013 1402 on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249
Transcript
Page 1: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

Small Molecule Therapeutics

Discovery of New Small Molecules Targeting the Vitronectin-Binding Site of the Urokinase Receptor That Block CancerCell Invasion

Vincenza Elena Anna Rea1, Antonio Lavecchia2, Carmen Di Giovanni2, Francesca Wanda Rossi1,Anna Gorrasi3, Ada Pesapane1, Amato de Paulis1, Pia Ragno3, and Nunzia Montuori1

AbstractBesides focusing urokinase (uPA) proteolytic activity on the cell membrane, the uPA receptor (uPAR) is able

to bindvitronectin, via a direct binding site. Furthermore, uPAR interactswith other cell surface receptors, such

as integrins, receptor tyrosine kinases, and chemotaxis receptors, triggering cell-signaling pathways that

promote tumor progression. The ability of uPAR to coordinate binding anddegradation of extracellularmatrix

(ECM) and cell signaling makes it an attractive therapeutic target in cancer. We used structure-based virtual

screening (SB-VS) to search for small molecules targeting the uPAR-binding site for vitronectin. Forty-one

compounds were identified and tested on uPAR-negative HEK-293 epithelial cells transfected with uPAR

(uPAR-293 cells), using the parental cell line transfected with the empty vector (V-293 cells) as a control.

Compounds 6 and 37 selectively inhibited uPAR-293 cell adhesion to vitronectin and the resulting changes in

cell morphology and signal transduction, without exerting any effect on V-293 cells. Compounds 6 and 37

inhibited uPAR-293 cell binding to vitronectin with IC50 values of 3.6 and 1.2 mmol/L, respectively.

Compounds 6 and 37 targeted S88 and R91, key residues for uPAR binding to vitronectin but also for uPAR

interaction with the fMLF family of chemotaxis receptors (fMLF-Rs). As a consequence, compounds 6 and 37

impaired uPAR-293 cell migration toward fetal calf serum (FCS), uPA, and fMLF, likely by inhibiting the

interaction between uPAR andFPR1, the high affinity fMLF-R. Both compounds blocked in vitroECM invasion

of several cancer cell types, thus representing new promising leads for pharmaceuticals in cancer.Mol Cancer

Ther; 12(8); 1402–16. �2013 AACR.

IntroductionThe urokinase (uPA)-mediated plasminogen activation

system, which generates the potent serine-protease plas-min, is involved in various pathologic processes, includ-ing angiogenesis, inflammation, wound healing, andmetastasis (1, 2).

The key molecule of this system, the uPA receptor(uPAR), is anchored to the plasma membrane by a glyco-sylphosphatidylinositol moiety and is formed by threehomologous domains (DI, DII, and DIII, from the N-ter-minus). The uPA-binding site is located in the DI domain,but the full-length molecule is required for an efficientbinding (3, 4). uPAR enhances pericellular proteolysis by

serving as a docking site to uPA, thus triggering a cascadeof proteolytic events that leads to the active degradation ofextracellular matrix (ECM) components (5).

Despite the lack of a transmembrane domain, uPAR canactivate intracellular signaling through lateral interac-tions with other cell surface receptors, such as integrins,receptor tyrosine kinases, and G-protein–coupled chemo-taxis receptors (6).

uPAR ability to regulate integrin activity plays a keyrole in cell adhesion, migration, proliferation, and surviv-al (7, 8). Recently, integrin-binding sites have been iden-tified in uPARdomainDII (residues 130–142; ref. 9) and inuPAR domain DIII (residues 240–248; ref. 10). Moreover,uPAR cross-talk with EGF receptor (EGFR) is extensiveand may regulate the shift from tumor cell dormancy toproliferation (11, 12).

uPAR interaction with receptors for fMet-Leu-Phe(fMLF-Rs; ref. 13) is required for both uPA- and fMLF-dependent cell migration and occurs through a chemo-tactic domain located in the DI–DII linker region, theSRSRY sequence (amino acids, 88–92; ref. 14). Because asoluble cleaved formof uPAR, exposing at theN-terminusof the SRSRY sequence is a ligand for fMLF-Rs (15), it hasbeen proposed that uPA binding to uPAR determines aconformational modification of the receptor with the

Authors'Affiliations: Departmentsof 1TranslationalMedical Sciencesand2Pharmacy, "Drug Discovery" Laboratory, "Federico II" University, Naples;and 3Department of Chemistry and Biology, University of Salerno, Salerno,Italy

V.E.A. Rea and A. Lavecchia equally contributed to this work.

Corresponding Author: Nunzia Montuori, Department of TranslationalMedical Sciences, "Federico II" University, via S. Pansini, 5, 80131 Naples,Italy. Phone: 39-0817463309; Fax: 39-0817463308; E-mail:[email protected]

doi: 10.1158/1535-7163.MCT-12-1249

�2013 American Association for Cancer Research.

MolecularCancer

Therapeutics

Mol Cancer Ther; 12(8) August 20131402

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 2: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

exposure of the chemotactic SRSRY domain that, in turn,binds and activates members of the fMLF-R family, thusinducing chemotaxis (16).Furthermore, uPAR itself is an adhesion receptor; in-

deed, it binds vitronectin, an abundant component of pro-visional ECM (17, 18). uPAR interactions with integrinsand vitronectin are positively regulated by uPA (19, 20),and both uPA and vitronectin can induce uPAR-mediatedcytoskeletal reorganization and cell migration (21, 22).Toward the goal of discovering uPAR-inhibitory com-

pounds, we focused on uPAR binding to vitronectin.Indeed, it has been recently reported that uPAR promotesmetastasis of human malignancies by engaging vitronec-tin through the activation of a cell signaling to Rac-1 (23).In epithelial cell lines, uPAR induces phenotypic changesconsistent with hypoxia-induced epithelial–mesenchy-mal transition (EMT), through a direct binding to vitro-nectin (24). Abnormal uPAR levels, like occur in cancer,may encourage EMT through vitronectin binding, thusfacilitating tumor invasion and metastasis (25).The X-ray structure of the ternary complex between

uPAR, the aminoterminal fragment of uPA (ATF) and thesomatomedin B domain of vitronectin (SMB) has beenrecently determined (26). There is now evidence thatuPAR–vitronectin interaction is entirely mediated by acomposite epitope exposed on the DI/DII interface ofuPAR (residues R30, W32, S56, R58, I63, S65, S88, R91,R116, and Q114; ref. 27).Therefore, we used structure-based virtual screening

(SB-VS; ref. 28) of the National Cancer Institute (NCI,National Health Institutes, Bethesda, MD) Diversity SetII to identify small molecules targeting this site and dis-rupting uPAR binding to vitronectin.Here, we describe the successful outcome of this search

and the initial biologic evaluation of the two most prom-ising compounds from this effort.

Materials and MethodsComputational chemistryMolecular modeling and graphics manipulations were

conducted using Maestro software (Maestro, version 9.2;Schr€odinger, LLC) and PyMOL packages (29) running ona E4 Computer Engineering E1080 workstation providedof a Intel Core i7-930 Quad-Core processor.

Protein preparationTheX-ray coordinates of humanuPAR–ATF–SBM tern-

ary complex (PDB code: 3BT1; ref. 27)were extracted fromthe Protein Data Bank (30). The structure was then pre-pared using the Protein Preparation Wizard of theSchr€odinger graphical interface Maestro. Hydrogenatoms were added to the protein consistent with theneutral physiologic pH (7.0). Thus, theR andKside chainswere cationic,whereas theEandDside chain carboxylateswere anionic. The protonation and flip states of the imid-azole rings of the hydrogen (H) residues were adjustedtogether with the side chain amides of N and Q residuesand the OH and SH orientations to optimize such inter-

actions. X-ray water molecules and ATF and SMB ligandswere removed during protein preparation, the last step ofwhich was energy minimization of the entire structure.The minimization was terminated when the root meansquare deviation (rmsd) of the heavy atoms in the energy-minimized structure relative to the starting (X-ray) coor-dinates exceeded 0.3 A. This ensures that the integrity ofthe X-ray structure is preserved in further modelingstudies while eliminating potential stereochemical shortcontacts that may exist in the protein structure. Further-more, this process also ensures that the hydrogen atomsare placed in optimized geometries.

Ligand preparation and filteringThree-dimensional (3D) structures ofNCIDiversity Set

II (1,364 compounds) were extracted by the website (31)and prepared using LigPrep software v2.5 (LigPrep,version 2.5; Schr€odinger, LLC) with Epik to expandprotonation and tautomeric states at 7.0 � 2.0 pH units.In post-LigPrep steps, unspecified stereoisomers wereretained up to 4 low-energy stereoisomers and sample5/6-membered rings retainedup to one conformationperligand as default parameters suggested inMaestro virtualscreening workflow. High-energy ionization/tautomerstates were removed from the generated conformations.So, about 2,000 structures including stereoisomers, tauto-mers, and ionization states were ready to be submitted tothe subsequent docking runs.

SB-VS protocolThe Grid-based Ligand Docking from Energetics

(GLIDE) virtual screening application in Schr€odingerMolecularModeling Suitewas used to screen compoundsusing two levels of docking precision. Amodified versionof the Chemscore function is used by GLIDE to assign ascore to each ligand in all poses. In the first step, GLIDEwas run in standard precision mode. The 20% of the top-scoring ligands (272 compounds)were kept and redockedusing the GLIDE extra precision mode, which similarlyretained the 20% of the best-ranked compounds (54 com-pounds). The extra precision–docking procedure ofGLIDE,which incorporates amore accurate, finer-graineddocking algorithm, was designed to eliminate false-posi-tives that survive the standard precision stage. Aftervisual inspection of the top-ranked compounds, 41 hitswere chosen for biologic evaluation. Flexible dockingwasallowed in all stages and default parameters from theVirtual Screening Workflow were used in all dockingstudies, in addition to the aforementionedmodificationsto the percentage of compounds entering each stage. Allfinal scores and poses came from GLIDE extra precision.The grid for docking studies was chosen sufficientlylarge to enclose all residues involved in the vitronectin–uPAR interactions within a cubic box of dimensions46A � 46A � 46A. The enclosing box was centered onthe vitronectin-binding site setting the bounding boxwith the sizes of 14 A � 14 A � 14 A. A van der Waalsradius scaling factor of 0.80 for atoms with a partial

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1403

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 3: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

atomic charge (absolute value) less than 0.15 was usedto soften the potential for nonpolar parts of the receptor.Compounds 6 and 37 identified by SB-VS were flexiblydocked using the same protein grid prepared for virtualscreening protocol. Ten poses were collected for eachligand and ranked according to predicted GLIDE extraprecision score.

Chemical inhibitorsAll compounds were obtained from the NCI/DTP

Open Chemical Repository (32). The compounds identi-fied by Virtual Screening were dissolved in dimethylsulfoxide (DMSO) and stored at�20�C, at a concentrationof 0.01 mol/L.

Cell cultures and transfectionsThe uPAR-negative (17) human embryonic kidney

cell line HEK-293 [American Type Culture Collection(ATCC)-certified from LGC Standards) was grown inDulbecco’s modified Eagle medium (DMEM; GIBCO)supplemented with 10% FBS. The sarcoma-derivedHT1080, the PC3 prostate cancer, the HCT colon cancer,and the MDAMB231 breast cancer cell lines (ATCC-certified from LGC Standards) were grown in DMEM(GIBCO) supplemented with 10% FBS. Authenticationof cell lines was conducted by ATCC using a short-tandem repeat assay. Upon receipt, all cell lines werestored in liquid nitrogen and passaged for less than 6months before use in this study.

uPAR cDNA was cloned in a pcDNA3 vector withresistance to Geneticin (Invitrogen), and the resultingplasmid was named uPAR-pcDNA3. HEK-293 cells werestably transfected with uPAR-pcDNA3 or with the emptyvector pcDNA3, as described previously (14).

Western blot analysisCells were lysed in PBS (0.08 mol/L NaCl, 0.002 mol/L

KCl, 0.0115 mol/L Na2HPO4, and 0.002 mol/L KH2PO4,pH 7.2) containing 1% Triton X-100, in the presence of aprotease inhibitor cocktail containing AEBSF, Aprotinin,Bestatin, E-64, Leupeptin, and Pepstatin A (Sigma-Aldrich) and a phosphatase inhibitor cocktail containingmicrocystin LR, cantharidin, and bromotetramisole(Sigma-Aldrich). Protein concentration of lysates wasdetermined using a colorimetric assay (Bio-Rad). Equalamounts of protein were subjected to SDS-PAGE andtransferred to polyvinylidene difluoride (PVDF) filters(Millipore). Membranes were incubated for 20 hours at4�C with the 399 polyclonal anti-uPAR antibody (Amer-ican Diagnostica), mouse monoclonal anti-p-Erk or rabbitpolyclonal anti-Erk2 antibodies (Santa Cruz Biotechnol-ogy), or with nonimmune immunoglobulins (Ig; JacksonImmunoResearch), as a negative control. Filters werefurther incubated for 30 minutes at room temperaturewith horseradish peroxidase (HRP)–conjugated anti-rab-bit or anti-mouse antibodies (Bio-Rad). The reaction wasrevealedbyanenhanced chemiluminescence (ECL)detec-tion kit (Amersham Biosciences).

Cell adhesion assayThe adhesion assays were conducted on 96-well flat-

bottomed plates for cell (Nunc). Wells were coated with 1mg of vitronectin (Becton Dickinson Biosciences), fibronec-tin (Roche), or with 100 mL of heat-denatured 1% bovineserum albumin (BSA) in PBS, as a negative control, andincubatedovernight at 4�C. The plateswere then blocked 1hour at room temperature with 1% heat-denatured BSA inPBS. Cells were detached with trypsin, resuspended inDMEM containing 10% FBS and incubated for 1 hour at37�C, 5% CO2, to allow receptor recovery. Cells were thenwashedwith serum-freeDMEM, counted, distributed intothewells at adensity of 105 cells perwell, and incubated for1 hour at 37�C in the presence of inhibitors, or DMSO, as avehicle control. Attached cells were fixed with 3% para-formaldehyde in PBS for 10 minutes and then incubatedwith 2% methanol for 10 minutes. Cells were finallystained for 10 minutes with 0.5% crystal violet in 20%methanol. Stain was eluted by 0.1 mol/L sodium citratein 50% ethanol, pH 4.2, and the absorbance at 540 nmwasmeasured by a spectrophotometer.

Cytotoxicity assayV-293 and uPAR-293 cells were plated, at 5 � 104 cells

per well, in 96-well plates and grown for 24 hours. Cellswere serum-starved overnight using DMEM containing0.1% BSA, and then incubated for 1 and 24 hours at 37�C,5% CO2 in the same medium containing the selectedsmall molecules or DMSO, as a vehicle control. Of note,20mL/well of CellTiter 96AQueousOne SolutionReagent(Promega) was added and, after incubation at 37�C for4 hours, the absorbance was determined by an ELISAreader (Bio-Rad) at a wavelength of 490 nm.

Cell migration and invasion assaysCell migration assays were conducted in Boyden

chambers using 8-mm pore size polyvinylpyrrolidone(PVPF)-free chemotaxis filters (Wathman Int.), coatedwith 50 mg/mL collagen or 5 mg/mL vitronectin V-293and uPAR-293 cells (2 � 105) were plated in the upperchamber in DMEM 0.1% BSA containing the selectedsmall molecules or DMSO, as a vehicle control. DMEM0.1% BSA alone or containing the ATF (1 � 10�8 mol/L),fMLF (1 � 10�7 mol/L) or 10% FBS was added in thelower chamber and the cellswere allowed tomigrate for 4hours at 37�C, 5% CO2.

For the invasion assay, filters were coated with 50mg/mL Matrigel (BD Biosciences) and incubated for30 minutes at 37�C for gelling. A total of 2 � 105 cells,plated in the upper chamber in DMEM 0.1% BSA con-taining the selected small molecules or DMSO, wereallowed to migrate toward DMEMmedium supplemen-ted with 10% FCS, or toward DMEM medium supple-mented with 0.1% BSA, as a control, for 18 hours at37�C, 5% CO2.

At the end of both experiments, cells on the lowersurface of the filter were fixed in ethanol, stained withhematoxylin, and counted at �200 magnification (10

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1404

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 4: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

random fields/filter). Cell migration and invasion wereexpressed as a percentage increase over the control.

CoimmunoprecipitationuPAR-293 cells (5 � 106/sample) were plated in 100-

mmdishes for 24 hours; then, the cells were incubated for16hours inDMEM0.1%BSAcontaining the selected smallmolecules or DMSO, as a vehicle control, in serum-freemedium. Cells were lysed in radioimmunoprecipitationassay (RIPA) buffer (150 mmol/LNaCl, 50 mmol/L Tris–HCl, pH7.5, 1%deoxycholate acid, 0.1%SDS, 1%TritonX-100, protease, andphosphatases inhibitors) and incubatedwith nonimmune serum and 10% protein A–conjugatedsepharose (GE Healthcare) for 2 hours at 4�C. After cen-trifugation, the supernatants were incubated with 2mg/mL of the R4 monoclonal anti-uPAR antibody, kindlyprovided by Dr. G. Hoyer-Hansen (The Finsen Laborato-ry, Copenhagen, Denmark), or with nonimmune Igs, for 2hours at 4�C and then with 10% protein A–sepharose for30minutes at room temperature. The immunoprecipitateswerewashed inRIPAbuffer, subjected to 10%SDS-PAGE,and analyzed byWestern blot analysis using a polyclonalantibody directed against the high-affinity fMLF receptor,FPR1 (Santa Cruz Biotechnology), or nonimmune Igs, as anegative control.

Rac-1 pull-down assayuPAR-293 cells were starved for 24 hours and then

plated on vitronectin-coated wells (5 mg/mL) for theindicated time, in the presence of compounds 6 and 37,or DMSO as a vehicle control. After a quick wash withice-cold PBS, cells were lysed with glutathione S-trans-ferase (GST)-FISH buffer [50 mmol/L Tris–HCl pH 7.4,2 mmol/L MgCl2, 1%NP-40, 10% glycerol, 100 mmol/LNaCl, 1 mg/mL leupeptin, 1 mg/mL pepstatin, 1 mg/mLaprotinin, 1 mmol/L phenylmethylsulfonylfluoride(PMSF), and 2 mmol/L dithiothreitol (DTT)]. After 10minutes at 4�C under agitation, cells were scraped andlysates were cleared by centrifugation in a precooledrotor. Five hundred microgram of total protein extractwas mixed with 10 mg of GST-PAK-CRIB domain, whichspecifically recognizes the GTP-bound forms of Rac-1,coupled to glutathione-sepharose beads (Upstate Bio-technology) and incubated 30 minutes at 4�C underagitation. Beads were then rinsed three times rapidlywith 1 mL of ice-cold GST-FISH buffer. The amounts oftotal Rac and Rac-GTP were estimated by immunoblotagainst Rac-1 (Upstate Biotechnology).

uPAR biotinylationRecombinant soluble uPAR (suPAR; American Diag-

nostica) was biotinylated and purified with the Amer-sham ECL protein biotinylation module according to themanufacturer’s instructions (GE Healthcare).

Binding of soluble uPAR to immobilized vitronectinHigh-binding plates with 96 flat-bottomedwells (Corn-

ing) were coated with 0.5 mg/well vitronectin diluted in

PBS, or BSA as a negative control, and incubated at 4�Covernight. After a wash in PBS, residual binding siteswere blocked with 200 mL of 1% BSA in PBS, for 1 hourat room temperature. Of note, 25 nmol/L biotinylateds-uPAR (diluted in PBS, 1 mg/mL BSA), alone or in thepresence of compounds 6 and 37, was placed into coatedwells. The plates were kept at 4�C for 1 hour, washedwithPBS containing 0.1% Tween 20 and then peroxidase-labeled avidin (Amersham), diluted 1:1,500 in PBS, and10 mg/mL BSA was added. After additional washings,the peroxidase substrate o-phenylenediamine (Sigma)was added and allowed to react for 3 minutes. The reac-tions were terminated with 1 mol/L H2SO4 and theproduct was quantified by measuring absorbance at492 nm using an automated plate reader (Bio-Rad), asdescribed previously (17). Results were expressed as apercentage decrease over the control, that is, the bindingin the absence of compounds.

ResultsIdentification by SB-VS of small molecules directedto the uPAR-binding site for vitronectin

uPAR overexpression functions as a biomarker forcancer progression and metastasis in many forms ofhuman malignancy (33). It has been recently proposedthat uPAR can promote metastasis not only by a uPA-dependent mechanism but also through a direct bindingto vitronectin followed by activation of a specific signaltransduction (23–25).

To identify new potential small molecules capable ofinterfering in the vitronectin–uPAR interaction, we con-ducted a SB-VS experiment using the X-ray crystal struc-ture of the human uPAR–ATF–SBM ternary complex (27)determined at 2.8 A resolution (3BT1 in the Protein DataBank).

Before starting the search, the binding site for vitronec-tin was identified on the uPAR crystal structure; it isexposed on the DI/DII interface of the receptor andcomprises residues R30, W32, S56, R58, I63, S65, S88,R91, R116, and Q114 (27). Among these, W32, R58, I63,R91, and Y92 were identified as key residues for uPAR/vitronectin binding by alanine-scanning mutagenesis(18, 25–27). In details, residues F13, Y28, and D22 ofvitronectin form an open pocket to bind R91 of uPAR.Y27 and Y28 of vitronectin insert into a large cavity ofuPAR, showing shape complementarity of this interface.The phenyl ring of Y28 contacts uPAR residues I63 andW32, mainly through hydrophobic interactions. Interest-ingly, the vitronectin-binding site contains some residuesincluded in the uPAR chemotactic sequence (SRSRY;amino acids, 88–92), required for cell migration towardfMLF and uPA (14–16).

Docking simulations were conducted through theGLIDE software (available from Schr€odinger LLC; refs.34, 35), selecting the NCI Diversity Set II as virtuallibrary of compounds. Then, GLIDE standard precisionmode docked each chemical structure into the uPAR:

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1405

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 5: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

vitronectin–binding site retaining the 20% of the top-scoring ligands. The resulting 272 compounds were thenredocked and scored with Glide extra precision to esti-mate binding affinity and rank the ligands. The result-ing 54 top-ranked compounds were analyzed by visualinspection because it has broadly shown that dockingscoring functions are often more successful at predict-ing a binding pose than the actual binding affinity (36).

Therefore, compounds were prioritized by taking intoaccount their binding mode as well as the overall matchamong binding modes of all the stereoisomers, tauto-mers, and ionization states of each compound and,second, their docking score. As for the visual inspection,compounds were checked for a good protein–ligandcomplementarity. Moreover, ligands able to make inter-actions with residues known to be important by muta-genesis studies or because they interact with knownsubstrates such as vitronectin or fMLF were prioritized.To promote the selection of structurally diverse com-pounds, potential hits were grouped into chemical clas-ses by visual inspection, and one molecule was selectedfor each class.

Finally, compounds were checked for ready sampleavailability from the compound provider, and 41 struc-turally diverse compounds (Fig. 1) were requested andtested in a cell-based assay.

Four compounds selected by SB-VS inhibited uPAR-transfected epithelial cell binding to vitronectin

Human embryonic kidney (HEK-293) cells, unlikemostcancer cells, do not express uPAR or uPA endogenously(14, 17, 18, 23). Therefore, as amodel tomimic the effects ofuPARoverexpression in cancer cells and to investigate theability of SB-VS–selected compounds to inhibit uPAR-mediated cell binding to vitronectin, HEK-293 cells werestably transfected with a human uPAR cDNA (uPAR-293cells) or with an empty vector (V-293 cells).

uPAR expression was then evaluated by Western blotanalysis, using an anti-uPAR polyclonal antibody, inuPAR-293 andV-293 cell lysates. uPAR-293 cells expressedhigh levels of full-length uPAR, whereas V-293 cells wereuPAR-negative (Fig. 2A).

In uPAR-293 cells, uPAR and integrins form stablecomplexes that both inhibit the native integrin-mediated

Figure 1. Chemical structures of compounds selected from NCI Diversity Set II by SB-VS.

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1406

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 6: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

cell adhesion to fibronectin and promote adhesion tovitronectin, via the direct vitronectin-binding site onuPAR (14, 17). Therefore, we evaluated uPAR-293 celladhesion to fibronectin and vitronectin, using the uPAR-negative V-293 cell line as a control. As expected, uPAR-293 cells showed a strongly increased cell adhesion tovitronectin, as comparedwithV-293 cells; on the contrary,the native cell adhesion to fibronectin was inhibited byuPAR overexpression in uPAR-293 cells, as comparedwith V-293 cells (Fig. 2B).To identify potential molecules able to inhibit uPAR

binding to vitronectin, uPAR-293 cells were subjected toadhesion experiments on vitronectin-coated wells in thepresence of 41 SB-VS–selected compounds (50 mmol/L)or DMSO, as a negative control. Although compounds9–12 and 14, increased cell adhesion to vitronectin, only4 compounds significantly inhibited uPAR-293 cell adhe-sion to vitronectin; namely, compounds 6 [4,40-dimethyl-[1,10-biphenyl]-2,20,5,50-tetraol], 13 [1-(phenanthren-9-yl)guanidine], 24 [8-((4-(tert-butyl) phenoxy)methyl)-1,3-dimethyl-1H-purine-2,6(3H,9H)-dione], and 37 [piperidin-2-yl(2-(trifluoromethyl)-6-(4-(trifluoromethyl) phenyl)pyr-idin-4-yl)methanol] (Fig. 2C).

Two compounds specifically inhibited uPAR-mediated cell binding to vitronectin independentlyof cell cytotoxicity

To ensure that inhibition of uPAR-293 cell adhesion tovitronectin was not due to cell killing, the toxicity of theabove selectedcompoundswasmeasuredbya cell viabilityassay. Serum-starved uPAR-293 cells were incubated for 1and 24 hourswith compounds 6, 13, 24, and 37, at the sameconcentration used in the adhesion assay (50 mmol/L), orwith DMSO, as a control. Cell viability was thenmeasuredby a MTS-PES assay. Compounds 13 and 24 showed asignificant level of cell toxicity (Fig. 3A), also confirmed onuPAR-negative V-293 cells (not shown), and were with-drawn from the study for their unspecific toxic effects.

To show the specificity of uPAR as a target, the abilityof compounds 6 and 37 to inhibit in vitro binding ofrecombinant suPAR to immobilized vitronectin was eval-uated. Compounds 6 and 37 inhibited suPAR bindingto vitronectin, albeit with a lower efficiency (Fig. 3B). In-deed, suPAR differs from membrane Glycosylphosphati-dylinositol (GPI)-linked uPAR in the conformation andflexibility of the molecule, being the linker region betweendomains D1 and D2 less accessible (37). The possibility of

A B

C

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1

VN FN

OD

OD

0

0.5

1.0

2.0

1.5

**

**

INH - 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

V-293 uPAR-293

- uPAR

- Actin

*

*

Figure 2. Increased cell adhesion to vitronectin in uPAR-transfected HEK-293 cells can be inhibited by SB-VS–selected compounds. A, HEK-293cellswere transfectedwithuPARcDNA (uPAR-293) orwith empty vector (V-293) and their lysateswere analyzedby10%SDS-PAGEandWesternblot analysiswith a polyclonal anti-uPAR antibody. uPAR-293 cells expressed high amounts of uPAR. B, V-293 (&) and uPAR-293 (&) cells were plated onto vitronectin(VN)- and fibronectin (FN)-coated wells for 1 hour, attached cells were fixed and stained with crystal violet; the stain was eluted and the absorbance [opticaldensity (OD)] at 540 nm was measured by a spectrophotometer. Values represent the mean � SD of 3 experiments carried out in triplicate (�, P < 0.05).uPAR-293 cells showed increased cell adhesion to vitronectin and decreased cell adhesion to fibronectin, as compared with V-293 cells. C, uPAR-293 cellswere plated on vitronectin-coated wells for 1 hour in the presence of 41 compounds identified by SB-VS (50 mmol/L) or DMSO (�), as a vehicle control.Attached cells were fixed and stained with crystal violet; the stain was eluted and the absorbance (OD) at 540 nm was measured by a spectrophotometer.Values represent themean�SDof 3 experiments carried out in triplicate (�,P < 0.05). Four compounds (6, 14, 24, and 37) significantly inhibited uPAR-293 celladhesion to vitronectin.

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1407

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 7: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

conformational effects, imposed by the C-terminal hydro-phobic group, is a widespread phenomenon among GPI-anchored proteins (37); therefore, we decided to studycompounds efficiency in uPAR-293 cells, using theuPAR-negative V-293 cell line as a control. Compounds6 and 37 inhibited uPAR-293 cell adhesion to vitronectinwithout affecting V-293 cell adhesion to the same substrate(Fig. 3C). Indeed, in the absence of uPAR, HEK-293 cellbinding tovitronectin ismediatedby specific integrins (38).

To further show the specificity of compounds 6 and 37for uPAR–vitronectin interaction, their ability to inhibituPAR-293 cell adhesionwas evaluatedbothonvitronectinand fibronectin. Compounds 6 and 37 specifically inhib-

ited uPAR-293 cell adhesion to vitronectin without affect-ing adhesion to fibronectin of the same cells (Fig. 3D).

Thus, compounds 6 and 37 are specific inhibitors ofuPAR binding to vitronectin.

Compounds 6 and 37 inhibit uPAR-dependent cellbinding to vitronectin in a dose-dependent mannerwith a submicromolar affinity

uPAR-923 cell adhesion to vitronectin was also evalu-ated in the presence of decreasing concentration of com-pounds 6 and 37, to evaluate the dose-dependency of theirinhibitory activity. IC50 values of 6 and 37 were 3.6and 1.2 mmol/L, respectively, as calculated by nonlinear

0

0.2

0.4

0.6

0.8

1

1.2

DMSO 6 37

OD

*

*

VN

C

A

D

B

0

1

2

3

1h 24h

*O

D

50 µmol/L µmol/L100

0

20

40

60

80

100

120

% o

f in

hib

itio

n

*

FN VN

OD

**

0

0.2

0.4

0.6

0.8

1

1.2

uPAR-293

**

*

**

Figure 3. Two top-scoring molecules from the in silico screening show specificity for uPAR–vitronectin interaction and are not toxic to the cells.A, serum-starved uPAR-293 cells were incubated for 1 and 24 hours with 50 mmol/L compound 6 ( ), 13 ( ), 24 ( ), and 37 ( ) or DMSO (&), as a vehiclecontrol; cell survivalwas then evaluatedby addingMTS-PESand, after incubation at 37�C for 4hours, the absorbance [optical density (OD)]wasdeterminedat490 nmby a spectrophotometer. Values are themean�SDof 3 experiments carried out in triplicate (�,P <0.05). Compounds 6 and 37did not affect uPAR-293cell viability, whereas compounds 13 and 24 were cytotoxic. B, biotinylated suPAR was placed for 1 hour at 4�C on vitronectin (VN)-coated wells in thepresence of 50 and 100 mmol/L compounds 6 ( ) and 37 ( ) or DMSO (&), as a vehicle control. Bound suPARwas revealed byHRP-avidin andOPD staining;the absorbance at 490 nm was measured by a spectrophotometer. Values of suPAR binding in BSA-coated wells were subtracted to obtain specificbinding. Values represent themean�SDof 3 experiments carried out in triplicate (�,P < 0.05). Hundred percent values represent suPARbinding to vitronectinin the absence of compounds. C, V-293 (&) and uPAR-293 (&) cells were plated on vitronectin-coated wells in the presence of 50 mmol/L compounds6 or 37, or DMSO, as negative control. Attached cells were fixed and stained with crystal violet; the stain was eluted and the absorbance (OD) at 540 nmwasmeasured by a spectrophotometer. Values represent the mean � SD of 3 experiments carried out in triplicate (�, P < 0.05). Compounds 6 and 37inhibited uPAR-293 cell adhesion to vitronectin without exerting any effect on uPAR-negative V-293 cells. D, uPAR-293 cells were plated on fibronectin (FN)-and vitronectin-coated wells in the presence of 50 mmol/L compounds 6 ( ) or 37 ( ) or DMSO (&), as a negative control. The attached cells were fixedand stained with crystal violet; the stain was eluted and the absorbance at 540 nm was measured by a spectrophotometer. Values represent the mean� SDof 3 experiments carried out in triplicate (�, P < 0.05). Compounds 6 and 37 inhibited uPAR-293 cell adhesion to vitronectin without affecting their adhesionto fibronectin.

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1408

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 8: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

regression curves using the sigmoidal dose–responseanalysis of the GraphPad Prism software (Fig. 4A andB). Ki of compounds 6 and 37 were 0.58 and 0.19 mmol/L,respectively, as calculated by the Cheng and Prusoffequation from EC50, using GraphPad Prism (39).Thus, two novel small molecules specifically inhibiting

uPAR-mediated cell binding to vitronectin were identi-fied by SB-VS (Table 1).

Compounds 6 and 37 abolished the changes in cellmorphology and the signal transduction induced byuPAR-mediated cell adhesion to vitronectinuPAR triggers cell attachment to the matrix through a

direct interactionwith the SMBdomain of vitronectin (27).

This initial adhesion is followed by engagement of integ-rins, changes in cell morphology, migration, and signaltransduction (18, 23, 25).

When uPAR-293 cells were seeded on vitronectin, theyunderwent marked changes in morphology, includingcell flattening and extensive lamellipodia formation. Inthe presence of compounds 6 and 37, at 2� IC50, uPAR-293cells retained a rounded cell body, failed to form lamelli-podia and only a round adhesion patch could be observedunder the cell body (Fig. 5A).

The limited cell contact with matrix in the presence ofcompounds 6 and 37 determined a strongly reduced celladhesion to vitronectin. Indeed, uPAR-293 cell adhesionto increasing concentration of vitronectin, in the absence

A B

Log [6 (mol/L)]

% in

hib

itio

n o

f c

ell b

ind

ing

to

VN

% in

hib

itio

n o

f c

ell b

ind

ing

to

VN

8-7-6-5-4-

0

50

100

Log [37 (mol/L)]

8-7-6-5-4-

0

50

100

Figure 4. Compounds 6 and 37inhibit uPAR-mediated celladhesion to vitronectin (VN) in adose-dependent manner. uPAR-293 cells were plated for 1 hour onvitronectin-coated wells in thepresence of decreasingconcentrations of compounds 6 (A)or 37 (B); attached cells werefixed andstainedwith crystal violet,the stain was eluted, and theabsorbance at 540 nmwas measured by aspectrophotometer. Valuesrepresent the mean � SD ofexperiments carried out intriplicate.

Table 1. The two low-micromolar uPAR-vitronectin inhibitors indentified by SB-VS

Cpd Structure NCI ID IsomerGLIDE GScore,kcal/mol

uPAR IC50,mmol/L

uPAR Ki,mmol/L Log P

6 2805 �6.54 3.6 0.58 2.93

37 305798 1.23 0.19 4.28

R,S �6.83R,R �6.81S,S �6.74S,R �6.70

NOTE: The second and third columns show compound structures, ID used throughout the text and NSC number. The fourth columnshows the 4 diastereoisomers of compound 37. The fifth column shows the estimated free energy of binding (kcal/mol). The sixth andseventh columns show the IC50 andKi values asmeasured in adhesion assays of uPAR-923 cell to vitronectin. The final column showsthe predicted Log P value of each compound.

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1409

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 9: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

of inhibitors, showed a KD of 26 nmol/L, similar to thatreported for in vitro uPAR:vitronectin binding (30nmol/L; ref. 17); the addition of compounds 6 and 37,at their 2� IC50, resulted in a significant loss of uPAR-293 cell binding to vitronectin (Fig. 5B).

After a direct binding to vitronectin, uPAR can activateextracellular signal–regulated kinases (ERK), most prob-ably through its lateral interaction with integrins (18);therefore, we investigated whether compounds 6 and37 were able to block ERK activation in response tovitronectin. Serum-starved uPAR-293 cells were platedon vitronectin-coated wells for 20minutes in the presence

of the 2 molecules, at 2� IC50, or DMSO, as a negativecontrol; then, cell lysates were analyzed by Western blotanalysis with an anti-phospho-ERK antibody. Both com-pounds strongly reduced vitronectin-mediated ERK acti-vation, as compared with DMSO-treated cells, in which astrong vitronectin-induced ERK activation was observed(Fig. 5C).

It has been reported that uPAR, through its bindingto vitronectin, activates the small GTPase Rac-1, thusstimulating cell migration (23); indeed uPAR-293cells plated on vitronectin show extensive lamellipoidaformation (18, 23, 25; Fig. 5A, arrows). Therefore, we

A

B C

D

-5.5 -5.0 -4.5 -4.0 -3.5

0.0

0.5

1.0

1.5

Log [VN (µmol/L)]

OD

INH: - - 6 37

uPAR-293

VN: 0 min 20 min

-pERKs

-ERK2

DMSO 6 37

uPAR-293

uPAR-293

VN: 0 min 20 min

-GTP/Rac-1

-Rac-1

INH: - - 6 37

Figure 5. Compounds 6 and 37 abolish the changes in cell morphology and the signal transduction induced by uPAR-mediated cell adhesion tovitronectin (VN). A, uPAR-293 cells were plated on vitronectin-coated wells for 1 hour in the presence of compounds 6 or 37, at their 2� IC50, or DMSO, as avehicle control; attached cells were fixed with 3% paraformaldehyde for 10 minutes and photographed by a contrast phase microscope. In the presence ofcompounds 6 and 37, uPAR-293 cells did not spread on vitronectin and failed to form lamellipodia. B, uPAR-293 cells were plated for 1 hour on wellscoated with increasing concentrations of vitronectin in the presence of compounds 6 (&) or 37 (&), at their 2� IC50, or DMSO (*), as a vehicle control;attached cells were fixed and stained with crystal violet, the stain was eluted and the absorbance at 540 nm was measured by a spectrophotometer. Valuesrepresent themean�SDof experiments carried out in triplicate. TheKDof uPAR-293 cell binding to vitronectin in the presence of vehicle alone is similar to thatreported for in vitro uPAR–vitronectin interaction (26 nmol/L) and is increased 2-fold by compound 6 and 4-fold by compound 37. C, serum-starveduPAR-293 cells were plated on vitronectin-coated wells in the presence of compounds 6 or 37, inhibitors (INH) of cell adhesion to VN, at their 2� IC50, orDMSO, as negative control. The cells were harvested at the indicated times and lysed for Western blot analysis with anti-phospho-ERKs and anti-ERK 2 (as aloading control) antibodies. uPAR-293 cell adhesion to vitronectin activatedERKs; this effectwasabolishedbyboth compounds.D, serum-starveduPAR-293cells were plated on vitronectin-coated wells in the presence of compounds 6 or 37 (INH), at their 2� IC50, or DMSO, as negative control. The cells wereharvested at the indicated times andwere subjected to Rac-1-GTP pull-down assay, as described in theMaterials andMethods. The amounts of total (Rac-1)and active (Rac-1-GTP) protein were estimated by immunoblotting with an anti-Rac-1 antibody. uPAR-293 cell adhesion to vitronectin activated Rac-1; thiseffect was abolished by both compounds 6 and 37.

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1410

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 10: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

investigated, by pull-down assays, whether com-pounds 6 and 37 were able to block Rac-1 activationin response to vitronectin. Serum-starved uPAR-293cells were plated on vitronectin-coated wells for 20minutes in the presence of both ligands, at 2� IC50, orDMSO, as a negative control. Active Rac-1 (Rac-1/GTP)was precipitated from cell lysates using the p21-bind-ing domain (PBD) of its target, PAK1, bound to agarosebeads; Rac-1/GTP was eluted from beads and estimat-ed by Western blot analysis with a monoclonal anti-Rac-1 antibody. Both compounds strongly reducedvitronectin-mediated Rac-1 activation, as comparedwith DMSO-treated cells, in which Rac-1 was stronglyactivated (Fig. 5D).Therefore, compounds 6 and 37 specifically inhibited

uPAR-mediated cell binding to vitronectin and its down-stream signals.

Structural basis for uPAR inhibition by compounds 6and 37

To understand the structural basis of the binding of thenewly discovered compounds 6 and 37 to uPAR, wescrutinized their binding poses by means of GLIDE pro-gram with the procedure described in the Materials andMethods. Figure 6 depicts the predicted bindingmodes ofthe 2 compounds in the vitronectin-binding site of uPAR.

All the top-ranked poses found by GLIDE for com-pound 6 showa twisted conformation of the ligand,whichis stabilized by an intramolecular H-bond between thetwo phenolic OH groups. Themolecule extends deep intothe vitronectin-binding site of uPAR, making several H-bonds and hydrophobic interactions with key residues ofthe site. In particular, the OH group at position 2 ispredicted to form 2 H-bonds with both S88 OH groupand theguanidinylNeofR116. TheOHgroupatposition 5

A

B C

D E

Figure 6. Binding modes ofcompounds 6 (A), (R,S)-37 (B), (R,R)-37 (C), (S,S)-37 (D), and (S,R)-37(E) into the vitronectin uPAR-binding site as calculated byGLIDE. For clarity, only interactingresidues are displayed and labeled.Compound 6 (cyan), compound 37(green) and interacting keyresidues (yellow) are representedas stick models, whereas theprotein is shown as a sky-blueribbon model. Chemotacticepitope residues S88, R91, andY92 are displayed in magenta. H-bonds are shown as dashed redlines.

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1411

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 11: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

also engages a H-bond with R91 side chain. Finally, theligand 50 OH group establishes a bidentate H-bond withthe guanidinium group of R30. In addition, the uPAR/6complex is further stabilized byhydrophobic contacts thatinvolve the 40-methyl group of the ligand and residues I63and W32 of the receptor.

Interestingly, the docking results showed that,although with a different arrangement, the two phenylrings of compound 6 nicely mimic the vitronectin resi-dues Y27 and Y28, which in the crystal structure of theuPAR–ATF–SMB complex insert into a large cavity onuPAR’s surface making H-bonding and hydrophobicinteractions (27).

It is also noteworthy that compound 6 interacts withR91 and S88, two key residues of the uPAR chemotacticepitope, which could interfere with the fMLF-R recogni-tion and affect uPAR-dependent cell migration.

Because compound 37 has two chiral centers and thechemical sample from the NCI is a racemic mixture withundetermined diastereomeric ratio, we docked the 4 dia-stereomers (R,S)-37, (R,R)-37, (S,S)-37, and (S,R)-37 gen-erated by LigPrep software (see Materials and Methods)into the uPAR–vitronectin–binding site and scored themby GLIDE extra precision. Whether one or all four of thediastereomers of compound 37 contributed to the inhibi-tion is not entirely clear, as the top-ranked poses for eachisomer within the vitronectin-binding site were not dif-ferent and gave comparable G-scores (Table 1).

Asdepicted inFig. 6B–E, oneof the 4-CF3fluorine atomsof the diastereomers interacts tightly with the cationicterminus of both R30 andR116 side chains. The remaining2 fluorine atoms of (R,S)-37 and (R,R)-37 also experience aslightly elongated H-bond with both S56 and S65 OHgroup. The 4-CF3 phenyl ring of the 4 isomers formshydrophobic contacts with W32 and I63 and is stabilizedby edge-on-face aromatic stacking interactions withW32.The 2-CF3 group of (R,S)-37, (R,R)-37 and (S,S)-37 isoptimally oriented to make H-bonds with the cationicterminus of R91side chain. On the contrary, the 2-CF3group of (S,R)-37 engages aH-bondwith R58 residue. TheF���H–NH2 and F���H–O distances are below 4 A, respec-tively, and the corresponding F���H–NH2 and F���H–Oangles are comprised between 107.0� and 151.0�. All thesevalues decrease in the range considered acceptable forF���H–X (X ¼ O, N, or S) H-bonds (36, 37). It has beenargued that fluorine rarely engages in H-bonds in small-molecule X-ray crystal structures (40, 41). However, inprotein pockets where ligands are immobilized by avariety of forces, they seem to be more common. F���Xdistances beyond 3.0 A can be regarded as dipole–dipoleinteractions that most likely provide small stabilizingcontributions (�1 kcal/mol) for the observed bindingposes.

The alcoholic OH group of (R,S)-37 and (R,R)-37 formsH-bonds with the R58 NH2 group and the S112 CObackbone, whereas in (S,S)-37 and (S,R)-37 it forms H-bonds with the Q114 CO group and the R116 NH2 group.With the exception of (R,R)-37, the protonated piperidine

N1 of (R,S)-37, (S,S)-37, and (S,R)-37 donates a H-bond toQ114 CO side chain.

The predicted G-scores could suggest that the (R,S)-37isomer binds slightly more strongly than the other ones.But further studies are needed to determinewhich isomeris the more active form for binding to uPAR.

From the docking results it seems evident that com-pound 37 entirely fills the vitronectin recognition pocketand engages numerousH-bonding and hydrophobic con-tacts with the receptor, explaining the slightly betteraffinity in comparison with compound 6 (Table 1). How-ever, compound 37, differently from compound 6, con-tacts only partially the chemotactic SRSRY sequence inter-acting only with the R91 side chain.

Compounds 6 and 37 inhibit the structural andfunctional interaction between uPAR and fMLF-Rs,thus blocking tumor cell invasion

Membrane-bound uPAR functionally interacts withfMLF-Rs through its SRSRY sequence (residues 88–92)and this interaction is required for both fMLF- and uPA-dependent cell migration (14–16, 23, 42, 43). Therefore, wetested whether compounds 6 and 37 were able to affectuPAR-dependent cell migration, as they also target keyresidues of the uPAR chemotactic domain, namely S88and R91.

To this aim, in vitro cell migration experiments werecarried out on uPAR-293 cells. These cells acquire theability to migrate toward uPA and fMLF by expressinguPAR; on the contrary, uPAR-negative V-293 cells do notmigrate toward uPA and fMLF, albeit they express fMLF-Rs (14).

Both compounds strongly inhibited uPAR-293 cellmigration toward FCS, whose main chemotactic compo-nent is vitronectin, aswell as towarduPAand fMLF,whenplated on collagen-coated membranes; as a control, V-293cell migration toward FCS was not affected (Fig. 7A). Asexpected, given the ability of compounds 6 and 37 toinhibit uPAR-mediated cell adhesion to vitronectin,migration of uPAR-293 cells on membranes coated withvitronectin confirmed the results obtained with collagen-coated membranes (Fig. 7A).

Therefore, compounds 6 and 37, targeting R91 of uPAR,not only block cell adhesion to vitronectin, but also impaircell migration toward serum, uPA, and fMLF by specif-ically inhibiting the functional interaction between uPARand fMLF-Rs.

To show that compounds 6 and 37 acted by inhibitingthe structural interaction between uPAR and fMLF-Rs,coimmunoprecipitation experiments were carried out inuPAR-293 cells, constitutively expressing high levels ofFPR1, the high affinity receptor for fMLF.

uPAR-293 cells were incubated for 20 hours in DMEMcontaining 0.1% BSA in the presence of compounds 6and 37, at 2� IC50, or DMSO, as a negative control. Cellslysateswere incubatedwith theR4monoclonal anti-uPARantibody or with nonimmune Igs, as a control. Immuno-complexes were purified with protein-A sepharose and

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1412

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 12: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

subjected to Western blot analysis with a polyclonalantibody recognizing FPR1.InDMSO-treateduPAR-293 cells, immunoprecipitation

with anti-uPARantibodies andWestern blot analysiswithanti-FPR1 antibodies revealed a band corresponding toFPR1 that was absent in the same lysate immunoprecipi-tated with nonimmune Igs, thus indicating the existenceof a structural interaction between uPAR and FPR1. Thisinteraction was completely blocked by compound 6 andslightly reduced by compound 37. As a control, no bands

were evidenced in uPAR-293 lysates imunoprecipitatedwith nonimmune Igs and subjected to Western blot anal-ysis with anti-FPR1 antibodies.

The inhibition of uPAR–FPR1 interaction in cells treatedwith compounds 6 and 37 was specific, as evidenced bythe presence of the same amount of FPR1 in all cell lysatestested by Western blot analysis with the anti-FPR1 anti-body, as a loading control (Fig. 7B).

A growing body of evidence suggests that the uPA–uPAR system promotes tumor metastasis by several

uPAR-293

IP: Igs Anti-uPAR

WB: Nonimmune Ig

Igs Anti-uPAR

Anti-FPR

INH: – – 6 37 – – 6 37

-FPR

-FPR

Stimulus FBS FBS ATF fMLF FBS ATF fMLF

* *

% o

f cell m

igra

tio

n (

ove

r co

ntr

ol)

**

**

0

50

100

150

200

250A

B D

C

CG VN

**

* * * *

HT1080

INH: – – 6 37

–FBS +FBS

– 6 37 – 6 37 – 6 370

50

100

150

200

250

* * ** * *

INH – 6 37

+FBS

0

150

300

450

600

750

% o

f c

ell

in

va

sio

n (

ove

r co

ntr

ol)

**

Figure 7. Compounds 6 and 37 inhibit the structural and functional interaction between uPAR and fMLF-Rs, thus blocking tumor cell invasion. A, V-293 (&)and uPAR-293 (&) cells were incubatedwith DMSO, as a vehicle control, or with compounds 6 ( ) and 37 ( ), at 2� IC50, plated in Boyden chambers coatedwith collagen (CG) or vitronectin (VN) and allowed to migrate toward FBS, the ATF or fMLF. Hundred percent values represent cell migration in theabsenceof chemoattractants. The valuesare themean�SDof 3experiments carriedout in triplicate (�,P<0.05). TheFBS-, ATF-, and fMLF-dependent uPAR-293 cell migration was strongly inhibited by compounds 6 and 37 both on collagen and vitronectin. B, serum-starved uPAR-293 cells were incubated for 16hours with compounds 6 or 37 (INH), at 2� IC50, or DMSO, as negative control. Cell lysates were immunoprecipitated with the R4 anti-uPAR monoclonalantibody or with nonimmune serum. The immunoprecipitated samples were electrophoresed on 10%SDS-PAGE and analyzed byWestern blot analysis withan anti-FPR1 antibody (top right) or with nonimmune serum (top left). Nonimmunoprecipitated cell lysates were subjected to Western blot analysis with theanti-FPR1 antibody (bottom right) or with nonimmune serum (bottom left), as a loading control. uPAR-coimmunoprecipitates with FPR1; treatment withcompounds 6 or 37 significantly reduced uPAR association to FPR1. C, highly invasive HT1080 sarcoma cells were treated with DMSO (�) or withcompounds 6 or 37 (INH), at their 2� IC50, and allowed to invade Matrigel, using as chemoattractants cell culture medium without serum (�FCS), to evaluatespontaneous invasion, or with 10% serum (þFBS). Invading cells on the lower side of the membrane were fixed, stained, and photographed; arepresentative image is shown. D, HT1080 sarcoma cells ( ), HCT human colon cancer cells ( ), PC3 human prostate cancer cells ( ), and MDAMB231human breast cancer cells ( ) were treated with DMSO (�) or with compounds 6 or 37, at their 2� IC50, and allowed to invade Matrigel, using aschemoattractants cell culturemediumwithout serum (�FCS) orwith 10%serum (þFBS). Invading cells on the lower side of themembranewere fixed, stained,counted (magnification,�200), and the results were expressed as a percentage of the invasion in the absence of chemoattractant (�FBS). The values are themean � SD of 3 experiments carried out in triplicate (�, P < 0.05). FBS-induced cancer cell invasiveness was significantly reduced by compounds 6 and 37.

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1413

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 13: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

different mechanisms, and not solely through the break-down of the ECM (30, 41). Among uPAR partners, thefMLF-R family of G-protein–coupled chemotaxis recep-tors is crucial in mediating cancer cell invasion andmetastasis (42, 43). Therefore, we sought to investigatewhether compounds 6 and 37 were able to inhibitthe invasion through reconstituted basal membranes ofthe highly invasive human fibrosarcomaHT1080 cell line,constitutively expressing high uPAR levels (42). Asshown in Fig. 7C, both compounds strongly reducedMatrigel invasion, as compared with DMSO-treated cells.

The same result was obtained on HCT human coloncancer cells, PC3 human prostate cancer cells, and onMDAMB231 breast cancer cells (Fig. 7D).

Therefore, compounds 6 and 37, two newly identifiedsmall-molecule inhibitors of uPAR, are able to blockcancer cell invasion by targeting the direct uPAR bindingto vitronectin and its structural and functional interactionwith fMLF-Rs.

DiscussionuPAR expression and function have been implicated in

nearly every step of tumor formation and progression(30, 44). Therefore, the identification of reagents withfavorable pharmacokinetic characteristics capable ofinterfering with uPAR-mediated signaling is an area ofgreat interest.

Many of the events of uPAR signaling are dependenton its binding to extracellular ligands, such as uPA andvitronectin. The main goal of this study was to identifysmall molecules that, by inhibiting uPAR binding tovitronectin, would block tumor cell migration and ECMinvasion.

Our work was based on previous studies demonstrat-ing that, although uPAR plays a pivotal role in the acti-vation of protease cascades at the cell surface, it promotesmetastasis of human malignancies mostly by engagingvitronectin through the activation of a cell signaling toRac-1 (23, 24, 45) and by encouraging EMT (18, 24, 25).

We posed that inhibiting uPAR binding to vitronectincould be strongly effective in cancer therapy. Indeed,uPA–uPAR complex formation enhances pericellularproteolysis through the activation of plasminogen and,in turn, matrix metalloproteinases (MMP). However,recent experimental evidence indicates that some mem-bers of the MMP family behave as tumor-suppressorenzymes and may therefore be regarded as antitargetsin cancer therapy (46). Moreover, small-molecule inhi-bitors of uPA binding to uPAR have been recentlydescribed (47, 48).

Another important advantage of our approach is thespecificity of the target; indeed, uPAR binding to vitro-nectin takes place when uPAR is overexpressed (Fig. 2), acondition mostly of malignant tumors. Moreover, vitro-nectin, a circulating adhesive protein, becomes abundant-ly associated with ECM sites upon tissue remodeling,injury/repair, or under disease conditions (49).

Combined biologic and biochemical approachesidentified the composite site of interaction betweenuPAR and vitronectin (26, 27). This information facili-tated the in silico screening of compounds targeting theuPAR-binding site for vitronectin and the computation-al evaluation of the binding to the target protein.Through this approach, which has already been usedto successfully identify inhibitors of Cdc25B dual spec-ificity phosphatases (50), we identified 2 small mole-cules able to inhibit the uPAR-dependent cell binding tovitronectin and, when applied to cancer cells, to blockECM invasion.

Compounds 6 and 37 disrupted cell adhesion to vitro-nectin of uPAR-transfected HEK-293 cells, and stoppedthe activation of signals to ERK andRac-1 in the same cells(Figs. 3 and 4). Cells that do not express uPAR, such asnontransfected HEK-293 cells, were insensitive to theeffects of compounds 6 and 37.

Docking studies showed that both compounds makeseveral H-bonds and hydrophobic interactions withmany residues of the vitronectin-binding site of uPAR(Fig. 6). Compounds 6 deeply extends into the vitro-nectin-binding site of uPAR, nicely mimicking vitro-nectin itself; compounds 37 entirely fills the vitronectinrecognition pocket of the receptor, explaining theslightly better affinity in comparison with compound6 (Table 1).

Amongothers, compounds 6 and 37 also target residuescomprised within the uPAR chemotactic domain (aminoacids, 88–92), able to mediate uPAR interaction with thefMLF family of chemotaxis receptors (fMLF-Rs). Indeed,both compounds 6 and 37 not only blocked cell adhesionto vitronectin, but also impaired the uPAR and fMLF-Rs–dependent cell migration (Fig. 7A).

Compound 6, which interacts with R91 and S88,completely abolished also the structural interactionbetween uPARand the high-affinity fMLF receptor, FPR1.Compound 37 which, differently from compound 6, par-tially contacts the chemotactic SRSRY sequence interact-ing only with the R91 side chain, slightly inhibited thestructural interaction between uPAR and FPR (Fig. 7B),even though it exerted the same functional effect ofcompound 6 on cell migration. It is noteworthy thatR91, the crucial residue in uPAR binding to vitronectin,as shown by Ala-scanning studies (18), is also, as shownby our results, the key residue in uPAR interaction withfMLF-Rs.

It was previously thought that, due to the largesurfaces area involved in protein–protein interface(51), small molecules would not be optimal candidatesfor disruption of these interactions. More recently, itwas concluded that "hotspots" might be responsible forthe high-affinity protein–protein interaction (52). To thebest of our knowledge, the disruption by a small mol-ecule of uPAR binding to vitronectin and its conse-quence on intracellular signaling and biologic outcomeshas not been already described, adding novelty to ourapproach.

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1414

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 14: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

Moreover, compounds 6 and 37 also inhibited uPARinteraction with fMLF-Rs that plays a fundamental role intumor cell migration, invasion, and metastasis(18, 22, 23, 42, 43). Indeed, both compounds did not causeacute cancer cell death (Fig. 3A), but blocked ECM inva-sion by cells from several cancer types (Fig. 7C and D);thus, theymaybe effective in preventing the occurrence ofmetastasis.In summary, this study used an integrated drug

discovery pipeline consisting of molecular modelingapproaches followed by experimental validation. Weconducted a screening of a full diversity compoundlibrary based on predicted binding to the uPAR:vitro-nectin–binding site and identified two novel putativecompounds for the treatment of cancer diseases. Inaddition, the functional selectivity and specificity of theselected compounds will allow for further insights inuPAR function and signaling.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: V.E.A. Rea, A. Lavecchia, P. Ragno, N. MontuoriDevelopment of methodology: V.E.A. Rea, F.W. Rossi, N. MontuoriAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.):V.E.A. Rea, A. Gorrasi, A. Pesapane, A. de PaulisAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):V.E.A.Rea,A. Lavecchia,C.DiGiovanni,A. PesapaneWriting, review, and/or revision of the manuscript: A. Lavecchia,P. Ragno, N. MontuoriAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): V.E.A. ReaStudy supervision: V.E.A. Rea, N. Montuori

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received January 2, 2013; revised May 20, 2013; accepted May 20, 2013;published OnlineFirst May 22, 2013.

References1. Blasi F, Carmeliet P. uPAR: a versatile signalling orchestrator. Nat Rev

Mol Cell Biol 2002;3:932–43.2. Ragno P. The urokinase receptor: a ligand or a receptor? Story of a

sociable molecule. Cell Mol Life Sci 2006;63:1028–37.3. Llinas P, Le Du MH, Ga

�rdsvoll H, Danø K, Ploug M, Gilquin B, et al.

Crystal structure of the humanurokinase plasminogen activator recep-tor bound to an antagonist peptide. EMBO J 2005;24:1655–63.

4. Barinka C, Parry G, Callahan J, Shaw DE, Kuo A, Bdeir K, et al.Structural basis of interaction between urokinase-type plasminogenactivator and its receptor. J Mol Biol 2006;363:482–95.

5. Montuori N, Cosimato V, Rinaldi L, Rea VE, Alfano D, Ragno P. uPARregulatespericellular proteolysis throughamechanism involving integ-rins and fMLF-receptors. Thromb Haemost 2013;109:309–18.

6. Montuori N, Ragno P. Multiple activities of a multifaceted receptor:roles of cleaved and soluble uPAR. Front Biosci 2009;14:2494–503.

7. Van der Pluijm G, Sijmons B, Vloedgraven H, van der Bent C, DrijfhoutJW, Verheijen J, et al. Urokinase-receptor/integrin complexes arefunctionally involved in adhesion and progression of human breastcancer in vivo. Am J Pathol 2001;159:971–82.

8. Tang CH, Wei Y. The urokinase receptor and integrins in cancerprogression. Cell Mol Life Sci 2008;65:1916–32.

9. Degryse B, Resnati M, Czekay RP, Loskutoff DJ, Blasi F. Domain 2 ofthe urokinase receptor contains an integrin-interacting epitope withintrinsic signaling activity: generation of a new integrin inhibitor. J BiolChem 2005;280:24792–803.

10. Chaurasia P, Aguirre-Ghiso JA, Liang OD, Gardsvoll H, Ploug M,Ossowski L. A region in urokinase plasminogen receptor domain IIIcontrolling a functional association with alpha5beta1 integrin andtumor growth. J Biol Chem 2006;281:14852–63.

11. Liu D, Aguirre Ghiso J, Estrada Y, Ossowski L. EGFR is a transducer ofthe urokinase receptor initiated signal that is required for in vivo growthof a human carcinoma. Cancer Cell 2002;1:445–57.

12. Jo M, Thomas KS, Marozkina N, Amin TJ, Silva CM, Parsons SJ, et al.Dynamic assembly of the urokinase-type plasminogen activator sig-naling receptor complex determines the mitogenic activity of uroki-nase-type plasminogen activator. J Biol Chem 2005;280:17449–57.

13. Le Y, Murphy PM, Wang JM. Formyl-peptide receptors revisited.Trends Immunol 2002;23:541–8.

14. Montuori N,CarrieroMV,SalzanoS,RossiG,RagnoP. Thecleavageofthe urokinase receptor regulates its multiple functions. J Biol Chem2002;277:46932–9.

15. Resnati M, Pallavicini I, Wang JM, Oppenheim J, Serhan CN, RomanoM, et al. The fibrinolytic receptor for urokinase activates the G protein-

coupled chemotactic receptor FPRL1/LXA4R. Proc Natl Acad Sci U SA 2002;99:1359–64.

16. de Paulis A, Montuori N, Prevete N, Fiorentino I, Rossi FW,Visconte V, et al. Urokinase induces basophil chemotaxis througha urokinase receptor epitope that is an endogenous ligand forformyl peptide receptor-like 1 and -like 2. J Immunol 2004;173:5739–48.

17. Wei Y, Waltz DA, Rao N, Drummond RJ, Rosenberg S, Chapman HA.Identification of the urokinase receptor as an adhesion receptor forvitronectin. J Biol Chem 1994;269:32380–8.

18. Madsen CD, Ferraris GM, Andolfo A, Cunningham O, Sidenius N.uPAR-induced cell adhesion and migration: vitronectin provides thekey. J Cell Biol 2007;177:927–39.

19. Sidenius N, Andolfo A, Fesce R, Blasi F. Urokinase regulates vitro-nectin binding by controlling urokinase receptor oligomerization. J BiolChem 2002;277:27982–90.

20. Carriero MV, Del Vecchio S, Capozzoli M, Franco P, Fontana L,Zannetti A, et al. Urokinase receptor interacts with alpha(v)beta5vitronectin receptor, promoting urokinase-dependent cell migrationin breast cancer. Cancer Res 1999;59:5307–14.

21. Degryse B, Orlando S, Resnati M, Rabbani SA, Blasi F. Urokinase/urokinase receptor and vitronectin/alpha(v)beta(3) integrin induce che-motaxis and cytoskeleton reorganization through different signalingpathways. Oncogene 2001;20:2032–43.

22. Gargiulo L, Longanesi-Cattani I, Bifulco K, Franco P, Raiola R, Cam-piglia P, et al. Cross-talk between fMLP and vitronectin receptorstriggered by urokinase receptor-derived SRSRY peptide. J Biol Chem2005;280:25225–32.

23. Jo M, Takimoto S, Montel V, Gonias SL. The urokinase receptorpromotes cancer metastasis independently of urokinase-type plas-minogen activator in mice. Am J Pathol 2009;175:190–200.

24. Lester RD, Jo M, Montel V, Takimoto S, Gonias SL. uPAR inducesepithelialmesenchymal transition in hypoxic breast cancer cells. J CellBiol 2007;178:425–36.

25. Madsen CD, Sidenius N. The interaction between urokinase receptorand vitronectin in cell adhesion and signalling. Eur J Cell Biol 2008;87:617–29.

26. Gardsvoll H, Ploug M. Mapping of the vitronectin-binding site on theurokinase receptor: involvement of a coherent receptor interfaceconsisting of residues from both domain I and the flanking interdomainlinker region. J Biol Chem 2007;282:13561–72.

27. Huai Q, Zhou A, Lin L, Mazar AP, Parry GC, Callahan J, et al. Crystalstructures of two human vitronectin, urokinase and urokinase receptorcomplexes. Nat Struct Mol Biol 2008;15:422–3.

Small Molecules Inhibiting uPAR Binding to Vitronectin

www.aacrjournals.org Mol Cancer Ther; 12(8) August 2013 1415

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 15: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

28. Lyne PD. Structure-based virtual screening: an overview. Drug DiscovToday 2002;7:1047–55.

29. DeLano WL. The PyMOL molecular graphics system on worldwideweb. 2005. Available from: http://www.pymol.org.

30. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H,et al. The protein data bank. Nucleic Acids Res 2000;28:235–42.

31. Available from: http://dtp.nci.nih.gov/branches/dscb/diversity_explanation.html.

32. Available from: http://dtp.cancer.gov.33. Mazar AP. Urokinase plasminogen activator receptor choreographs

multiple ligand interactions: implications for tumor progression andtherapy. Clin Cancer Res 2008;14:5649–55.

34. Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT,et al. Glide: a new approach for rapid, accurate docking and scoring. 1.Method and assessment of docking accuracy. J Med Chem 2004;47:1739–49.

35. Halgren TA, Murphy RB, Friesner RA, Beard HS, Frye LL, Pollard WT,et al. Glide: a new approach for rapid, accurate docking and scoring. 2.Enrichment factors in database screening. J Med Chem 2004;47:1750–9.

36. Jacobsson M, G€aredal M, Schultz J, Karl�en A. Identification of Plas-modium falciparum spermidine synthase active site binders throughstructure-based virtual screening. J Med Chem 2008;51:2777–86.

37. Høyer-Hansen G, Pessara U, Holm A, Pass J, Weidle U, Danø K, et al.Urokinase-catalysed cleavage of the urokinase receptor requires anintact glycolipid anchor. Biochem J 2001;358:673–9.

38. Bodary SC, McLean JW. The integrin beta 1 subunit associates withthe vitronectin receptor alpha v subunit to form a novel vitronectinreceptor in a human embryonic kidney cell line. J Biol Chem 1990;265:5938–41.

39. Cheng Y, Prusoff WH. Relationship between the inhibition constant(K1) and the concentration of inhibitor which causes 50 per centinhibition (I50) of an enzymatic reaction. Biochem Pharmacol 1973;22:3099–108.

40. Dunitz JD, Taylor R. Organic fluorine hardly ever accepts hydrogenbonds. Chem Eur J 1997;3:89.

41. Dunitz JD. Organic fluorine: odd man out. Chem Biol Chem 2004;5:614–21.

42. Carriero MV, Longanesi-Cattani I, Bifulco K, Maglio O, Lista L, BarbieriA, et al. Structure-based design of an urokinase-type plasminogenactivator receptor-derived peptide inhibiting cell migration and lungmetastasis. Mol Cancer Ther 2009;8:2708–17

43. Bifulco K, Longanesi-Cattani I, Franco P, Pavone V, Mugione P, DiCarluccio G, et al. Single amino acid substitutions in the chemotacticsequence of urokinase receptor modulate cell migration and invasion.PLoS ONE 2012;7:e44806.

44. Laufs S, Schumacher J, Allgayer H. Urokinase-receptor (u-PAR): anessential player in multiple games of cancer: a review on its role intumor progression, invasion, metastasis, proliferation/dormancy, clin-ical outcome and minimal residual disease. Cell Cycle 2006;5:1760–71.

45. Kjoller L, Hall A. Rac mediates cytoskeletal rearrangements andincreased cell motility induced by urokinase-type plasminogen acti-vator receptor binding to vitronectin. J Cell Biol 2001;152:1145–57.

46. Decock J, Thirkettle S, Wagstaff L, Edwards DR. Matrix metallopro-teinases: protective roles in cancer. J Cell Mol Med 2011;15:1254–65.

47. Wang F, Li J, Sinn AL, Knabe WE, Khanna M, Jo I, et al. Virtualscreening targeting the urokinase receptor, biochemical and cell-based studies, synthesis, pharmacokinetic characterization, andeffect on breast tumor metastasis. J Med Chem 2011;54:7193–205.

48. Wang F, Eric Knabe W, Li L, Jo I, Mani T, Roehm H, et al. Design,synthesis, biochemical studies, cellular characterization, and struc-ture-based computational studies of small molecules targeting theurokinase receptor. Bioorg Med Chem 2012;20:4760–73.

49. Preissner KT, Reuning U. Vitronectin in vascular context: facets of amultitalented matricellular protein. Semin Thromb Hemost 2011;37:408–24.

50. Lavecchia A, Di Giovanni C, Pesapane A, Montuori N, Ragno P,Martucci NM, et al. Discovery of new inhibitors of Cdc25B dualspecificity phosphatases by structure-based virtual screening. J MedChem 2012;55:4142–58.

51. Lo Conte L, Chothia C, Janin J. The atomic structure of protein-proteinrecognition sites. J Mol Biol 1999;285:2177–98.

52. Arkin MR, Wells JA. Small-molecule inhibitors of protein–protein inter-actions: progressing towards the dream. Nat Rev Drug Discov 2004;3:301–17.

Rea et al.

Mol Cancer Ther; 12(8) August 2013 Molecular Cancer Therapeutics1416

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249

Page 16: DiscoveryofNewSmallMoleculesTargetingtheVitronectin- Binding … · 2013-08-07 · score to each ligand in all poses. In the first step, GLIDE was run in standard precision mode.

2013;12:1402-1416. Published OnlineFirst May 22, 2013.Mol Cancer Ther   Vincenza Elena Anna Rea, Antonio Lavecchia, Carmen Di Giovanni, et al.   Cancer Cell InvasionVitronectin-Binding Site of the Urokinase Receptor That Block Discovery of New Small Molecules Targeting the

  Updated version

  10.1158/1535-7163.MCT-12-1249doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/12/8/1402.full#ref-list-1

This article cites 49 articles, 17 of which you can access for free at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mct.aacrjournals.org/content/12/8/1402To request permission to re-use all or part of this article, use this link

on September 11, 2020. © 2013 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst May 22, 2013; DOI: 10.1158/1535-7163.MCT-12-1249


Recommended