fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A...

9
Priority Report A Specic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity by Inhibiting STAT3 Phosphorylation and Signaling Daejin Kim, In-Hyun Lee, Sunghyun Kim, Minsuk Choi, Hyungjun Kim, Sukyung Ahn, Phei Er Saw, Hyungsu Jeon, Yumi Lee, and Sangyong Jon Abstract STAT3 promotes the survival, proliferation, metastasis, immune escape, and drug resistance of cancer cells, making its targeting an appealing prospect. However, although multiple inhibitors of STAT3 and its regulatory or effector pathway elements have been developed, bioactive agents have been somewhat elusive. In this report, we report the identication of a specic STAT3-binding peptide (APT STAT3 ) through phage display of a novel "aptide" library. APT STAT3 bound STAT3 with high specicity and afnity (231 nmol/L). Addition of a cell-penetrating motif to the peptide to yield APT STAT3 -9R enabled uptake by murine B16F1 melanoma cells. Treatment of various types of cancer cells with APT STAT3 -9R blocked STAT3 phosphorylation and reduced expression of STAT targets, including cyclin D1, Bcl-xL, and survivin. As a result, APT STAT3 -9R suppressed the viability and proliferation of cancer cells. Furthermore, intratumoral injection of APT STAT3 -9R exerted potent antitumor activity in both xenograft and allograft tumor models. Our results offer a preclinical proof-of-concept for APT STAT3 as a tractable agent for translation to target the broad array of cancers harboring constitutively activated STAT3. Cancer Res; 74(8); 18. Ó2014 AACR. Introduction STAT3, a member of the STAT family, has received consid- erable attention for the important role it plays in signaling pathways linked to inammation-associated diseases and can- cers (1). Notably, unlike the transient activation of STAT3 characteristic of normal cells, STAT3 tends to be constitutively activated in most cancer cells. In these cells, STAT3 is associated with tumorigenesis and malignancy because its downstream signaling results in production of a number of cytokines that regulate proliferation, angiogenesis, survival, and metastasis (2). For these reasons, many research groups have focused on developing inhibitors that can block upstream or downstream elements in the STAT3 signaling pathway (3, 4). Upon activa- tion, STAT3 becomes phosphorylated and undergoes dimer- ization. The active, dimeric form enters the nucleus, in which it binds to target DNA-response elements, leading to the expres- sion of proteins related to cancer progression and malignancy. Thus, inhibitors reported to date, including small molecules (59), decoy oligodeoxynucleotides (ODN; ref. 10), siRNAs (11), and peptides (12, 13) and their peptidomimetics (14, 15), target one of the key steps required for STAT3-mediated gene expres- sion. Several small molecules that can inhibit phosphorylation and subsequent dimerization by binding to the SH2 domain of STAT3 have been shown to suppress cancer cell growth in vitro and reduce tumor burden in vivo, and indeed a few such inhibitors are now in various phases of clinical trials (3). Decoy ODNs that can intercept the active form of the STAT3 dimer also show potential as inhibitors in in vivo tumor models (16). Peptide-based STAT3 inhibitors have also been reported. These inhibitory phospho-peptides (P YLKTK, Y LPQTV), based on a truncated gp130 sequence, bind to the SH2 domain of STAT3 and block dimerization (17, 18). However, the relatively low potency and limited cell permeability of such phospho-peptides have hampered their further use in an in vivo setting. Peptide libraries have been screened for interaction with the DNA- binding domain (DBD) of STAT3, and the identied peptides have been shown to inhibit STAT3 DNA-binding activity (19). Very recently, a short peptide derived from the helices of the N- terminal domain of STAT3 has been shown to inhibit STAT3 transcriptional activity (20). However, although these peptide inhibitors have shown cytotoxicity in various cancer cells by blocking STAT3 signaling, to date there have been few reports demonstrating antitumor efcacy in vivo (3, 21). Aptides, developed by our laboratory, constitute a platform technology that enables screening and identication of high- afnity and -specicity peptides for various biologic targets (22). An aptide consists of a tryptophan zipper scaffold that forms a robust b-hairpin structure and a target-binding site containing Authors' Afliation: KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Sangyong Jon, KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Daejeon 305-701, Republic of Korea. Phone: 82-42-350-2634; Fax: 82-42-350-4450; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-13-2187 Ó2014 American Association for Cancer Research. Cancer Research www.aacrjournals.org OF1 Research. on June 18, 2020. © 2014 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Transcript of fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A...

Page 1: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

Priority Report

A Specific STAT3-Binding Peptide Exerts AntiproliferativeEffects and Antitumor Activity by Inhibiting STAT3Phosphorylation and Signaling

Daejin Kim, In-Hyun Lee, Sunghyun Kim, Minsuk Choi, Hyungjun Kim, Sukyung Ahn, Phei Er Saw,Hyungsu Jeon, Yumi Lee, and Sangyong Jon

AbstractSTAT3 promotes the survival, proliferation, metastasis, immune escape, and drug resistance of cancer cells,

making its targeting an appealing prospect. However, althoughmultiple inhibitors of STAT3 and its regulatory oreffector pathway elements have been developed, bioactive agents have been somewhat elusive. In this report, wereport the identification of a specific STAT3-binding peptide (APTSTAT3) through phage display of a novel "aptide"library. APTSTAT3 bound STAT3 with high specificity and affinity (�231 nmol/L). Addition of a cell-penetratingmotif to the peptide to yield APTSTAT3-9R enabled uptake bymurine B16F1melanoma cells. Treatment of varioustypes of cancer cells with APTSTAT3-9R blocked STAT3 phosphorylation and reduced expression of STAT targets,including cyclin D1, Bcl-xL, and survivin. As a result, APTSTAT3-9R suppressed the viability and proliferation ofcancer cells. Furthermore, intratumoral injection of APTSTAT3-9R exerted potent antitumor activity in bothxenograft and allograft tumor models. Our results offer a preclinical proof-of-concept for APTSTAT3 as a tractableagent for translation to target the broad array of cancers harboring constitutively activated STAT3. CancerRes; 74(8); 1–8. �2014 AACR.

IntroductionSTAT3, a member of the STAT family, has received consid-

erable attention for the important role it plays in signalingpathways linked to inflammation-associated diseases and can-cers (1). Notably, unlike the transient activation of STAT3characteristic of normal cells, STAT3 tends to be constitutivelyactivated inmost cancer cells. In these cells, STAT3 is associatedwith tumorigenesis and malignancy because its downstreamsignaling results in production of a number of cytokines thatregulate proliferation, angiogenesis, survival, andmetastasis (2).For these reasons, many research groups have focused ondeveloping inhibitors that can block upstream or downstreamelements in the STAT3 signaling pathway (3, 4). Upon activa-tion, STAT3 becomes phosphorylated and undergoes dimer-ization. The active, dimeric form enters the nucleus, in which itbinds to target DNA-response elements, leading to the expres-sion of proteins related to cancer progression and malignancy.Thus, inhibitors reported to date, including small molecules

(5–9), decoy oligodeoxynucleotides (ODN; ref. 10), siRNAs (11),and peptides (12, 13) and their peptidomimetics (14, 15), targetone of the key steps required for STAT3-mediated gene expres-sion. Several small molecules that can inhibit phosphorylationand subsequent dimerization by binding to the SH2 domain ofSTAT3 have been shown to suppress cancer cell growth in vitroand reduce tumor burden in vivo, and indeed a few suchinhibitors are now in various phases of clinical trials (3). DecoyODNs that can intercept the active form of the STAT3 dimeralso show potential as inhibitors in in vivo tumor models (16).Peptide-based STAT3 inhibitors have also been reported. Theseinhibitory phospho-peptides (P�YLKTK, Y�LPQTV), based on atruncated gp130 sequence, bind to the SH2 domain of STAT3and block dimerization (17, 18). However, the relatively lowpotency and limited cell permeability of such phospho-peptideshave hampered their further use in an in vivo setting. Peptidelibraries have been screened for interaction with the DNA-binding domain (DBD) of STAT3, and the identified peptideshave been shown to inhibit STAT3 DNA-binding activity (19).Very recently, a short peptide derived from the helices of the N-terminal domain of STAT3 has been shown to inhibit STAT3transcriptional activity (20). However, although these peptideinhibitors have shown cytotoxicity in various cancer cells byblocking STAT3 signaling, to date there have been few reportsdemonstrating antitumor efficacy in vivo (3, 21).

Aptides, developed by our laboratory, constitute a platformtechnology that enables screening and identification of high-affinity and -specificity peptides for various biologic targets (22).An aptide consists of a tryptophan zipper scaffold that forms arobust b-hairpin structure and a target-binding site containing

Authors' Affiliation: KAIST Institute for the BioCentury, Department ofBiological Sciences, Korea Advanced Institute of Science and Technology,Daejeon, Republic of Korea

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

CorrespondingAuthor:Sangyong Jon,KAIST Institute for theBioCentury,Department of Biological Sciences, Korea Advanced Institute of Scienceand Technology, 291 Daehak-ro, Daejeon 305-701, Republic of Korea.Phone: 82-42-350-2634; Fax: 82-42-350-4450; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-13-2187

�2014 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org OF1

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 2: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

12 randomizable amino acids that enables library construction(Fig. 1A). Phage display of an aptide library has enabled us toidentify a number of aptides specific to various protein targets(22). Most such aptides showed affinities in the tens of nano-molar range, even before affinity maturation, and as suchrepresent a general source of high-affinity binders. Here, wereport the screening and identification of a STAT3-specificaptide (APTSTAT3) and evaluate its potential as an inhibitor ofSTAT3 signaling in cancer cells in vitro and in tumor-bearingmice in vivo. To the best of our knowledge, APTSTAT3 is the firstreported peptide inhibitor to show therapeutic efficacy in in vivoanimal models by blocking STAT3 activation.

Materials and MethodsCell lines and culture

The A549 human lung carcinoma, B16F1 mouse melanoma,HepG2 human hepatocellular carcinoma, MDA-MB-231 breast

cancers, and U87MG human neuronal glioblastoma cells wereobtained from the American Type Culture Collection. A549cells were maintained in Ham F12K. B16F1 and MDA-MD-231cells weremaintained in RPMI-1640medium.HepG2 cells weremaintained in Dulbecco's Modified Eagle Medium. U87MGcells were maintained in Minimum Essential Medium. The cellculture media were supplemented with 10% heat-inactivatedFBS.

MaterialsAll peptides, including APTSTAT3-9R and APRscr-9R, were

custom synthesized by AnyGen. STAT3 protein was obtainedfrom SignalChem. Antibodies against STAT3, STAT1, STAT5,phosphorylated STAT3 (P-STAT3), P-STAT5, P-STAT1 AKT, P-AKT, glyceraldehyde-3-phosphate dehydrogenase (GAPDH),Bcl-xL, survivin, and cyclin D1 were obtained from Cell Sig-naling Technology.

Figure 1. Characterization of APTSTAT3, a specific STAT3-binding peptide. A, schematic depiction of the structure of a STAT3-specific aptide (APTSTAT3)and control scrambled peptide (APTscr) with amino acid sequences and a cell permeable form of the STAT3-specific aptide (APTSTAT3-9R). B, phageELISAs using a phage displaying APTSTAT3 sequence against various proteins (STAT3, streptavidin, BSA, VEGF, TNF-a, and anti-His6-tag-antibody; left).C, concentration–escalation ELISAs using APTSTAT3, APTSTAT3-9R, and APTscr-9R peptides against STAT3 protein. Antiaptide (trpzip) mAb wasused to detect. D, confocal microscopy images of A549 human lung carcinoma cells after treatment with none (control), FITC-APTSTAT3-9R, andFITC-APTscr-9R.

Kim et al.

Cancer Res; 74(8) April 15, 2014 Cancer ResearchOF2

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 3: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

Screening and identification of STAT3-specific bindersA STAT3-specific aptide was identified using a phage dis-

play-based screening method, as described previously (22).Briefly, STAT3 was immobilized on 96-well plates (Corning)overnight at 4�C. After incubating phage library pools onSTAT3-immobilized plates for 1 hour at 25�C, unbound phageswere removed by washing with PBS containing 0.5% Tween-20(PBS-T), and bound phages were eluted with 0.2 mol/L glycinebuffer (pH 2.2), followed by immediate neutralization withTris-HCl (pH 8.8). The eluted phages were amplified usinglog-phase cultures of Escherichia coli ER2537 (New EnglandBiolabs). The above mentioned screening steps were iteratedwith the amplified phages several times until specific phagesenriched. After biopanning, phage ELISAs were carried out.Bound phages were detected by incubating with horseradishperoxidase (HRP)–conjugated anti-M13 antibody (GE Health-care) followed by monitoring absorbance at 450 nm after incu-bation with the chromogenic substrate tetramethylbenzidine.

DNA-binding assayFollowing treatment of cells with APTSTAT3-9R, APRscr-9R, or

PBS for 6 hours, cell extracts were prepared and analyzed forSTAT3 DNA-binding activity in triplicate using a TransAMSTAT3 Transcription Factor Assay Kit, following the manu-facturer's protocol (Active Motif).

Quantitative reverse transcription-PCRTotal RNA was isolated using Ribo EX (GeneAll) and

subjected for reverse transcription using Reverse Transcrip-tase (Promega) according to the manufacturer's instructions.Quantitative real-time PCR were carried out as describedpreviously (23).

Western blot analysisCell lysates were prepared in PRO-PREP protein extraction

solution. Protein concentrations were determined by Bradfordassay. A sample of 30 mg of each protein was loaded in 15%SDS–PAGE gels. After gel electrophoresis, proteins were trans-ferred to polyvinylidene difluoride membrane (Millipore). Themembranes were blocked in 5% bovine serum albumen (BSA)in TBS containing 0.1% Tween 20 at room temperature for 2hours followed by overnight incubation at 4�C with primaryantibodies in TBST containing 5% BSA. The membranes werethen incubated with HRP-conjugated secondary antibody inTBST containing 5% BSA for 30 minutes at room temperature.Anti-mouse and -rabbit antibody were obtained from SantaCruz Biotechnology. Specific proteins were detected by Che-miDoc (Bio-Rad).

Cell viability assayCells were plated in a 96-well plate (5,000 cells/well) and

incubated for 12 hours. After incubation the cells were treatedwith nine-arginine (9R), APTSTAT3-9R, and APRscr-9R for 12hours. After 12 hours incubation, 20 mL MTT (5 mg/mL) wasadded to each well. After 4 hours, 100 mL dimethyl sulfoxidewas added to each well, and absorbance was measured at 570nm using a Spectra Macplus microplate reader (MolecularDevices).

Colony formation assayCells in growthmediumwere cultured at 500 cells per well in

12-well plates and treated with APTSTAT3-9R, APRscr-9R, or PBSon the following day. Cells were then allowed to grow for 2weeks until colonies were visible. Colonies were stained byincubating with 0.1% Coomassie Brilliant Blue R-250 for 20minutes.

Flow cytometry analysisTheAnnexinV–fluorescein isothiocyanate (FITC)Apoptosis

Detection Kit (BDBiosciences) was used to detect apoptosis byflow cytometry. Cells were exposed to APTSTAT3-9R, APRscr-9R,or PBS treatment at 30 mmol/L, and after 6 hours, they wereharvested (including detached cells), and processed accordingto the manufacturer's instructions.

Antitumor efficacy in the A549 xenograft tumormodel

A549 cells (1 � 107 cells) were harvested and resuspendedin 50 mL F12K media and injected subcutaneously into theright flank of 6-week-old female BALB/c nude mice. Approx-imately 14 days later, when tumor volumes had reachedapproximately 50 mm3, the mice were randomly divided intothree groups (n ¼ 5 mice/group): APTSTAT3-9R, APRscr-9R,and PBS. Mice were intratumorally injected four times withAPTSTAT3-9R and APRscr-9R (8 mg/kg in 50 mL PBS) or PBS.At the same time, tumor volume was determined by mea-suring with calipers and calculated according to the formula,A� B2 � 0:5, where A is the longest dimension of the tumorand B is the shortest dimension of the tumor. After 30 days,mice were sacrificed and tumors were excised for use inother assays. All mice were maintained at an animal facilityunder pathogen-free conditions. Experimental proceduresand handling of mice were carried out in accordance withexperimental animal guidelines approved by our Institution-al Animal Care and Use Committee.

Statistical analysisThe statistical significance of differences among groups was

calculated by one-way ANOVA. A P value of < 0.001 wasconsidered statistically significant.

Results and DiscussionA specific STAT3-binding peptide and its cell permeablederivative

Using phage display of an aptide library, we were able toscreen and identify a specific STAT3-binding peptide (Fig.1A). A total of four rounds of selection were performed toenrich for phage clones that showed specificity for STAT3 inphage ELISAs (Supplementary Fig. S1). Sequencing of thebound phages yielded only an aptide sequence. The affinityand specificity of the identified STAT3-binding aptide (APT-

STAT3) were measured. A surface plasmon resonance assayrevealed that APTSTAT3 bound to STAT3 with a dissociationconstant (Kd) of 231 nmol/L; a control peptide containingthe same trpzip scaffold but with a scrambled sequence inthe target-binding site, APTscr, did not bind STAT3 (Sup-plementary Fig. S2). Phage ELISAs showed that the phage

STAT3-Targeting, High-Affinity Peptide for Cancer Therapeutics

www.aacrjournals.org Cancer Res; 74(8) April 15, 2014 OF3

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 4: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

displaying APTSTAT3 sequence was highly specific to thetarget STAT3 compared with other proteins, whereas thephage displaying APTscr sequence showed no appreciablebinding to any of the proteins tested (Fig. 1B). To impart cellpermeability to APTSTAT3, we fused APTSTAT3 with a 9R, acell-penetrating peptide (Fig. 1A; ref. 24). ELISAs using anantiaptide monoclonal antibody (mAb) showed thatAPTSTAT3-9R was also able to specifically bind STAT3 witha comparable binding ability to APTSTAT3, as evidenced by aconcentration-dependent increase in the ELISA signal,whereas the 9R-modified scrambled aptide (APTscr-9R)showed little signal enhancement (Fig. 1C). Next, we exam-ined whether the attachment of 9R to the aptides wouldenable cell penetration and, thus, intracellular localization.As clearly shown in Fig. 1D, most B16F1 melanoma cellstreated with FITC-labeled APTSTAT3-9R or APTscr-9R showedintense fluorescence signals in the cytosol, demonstratingcellular uptake and confirming the cell-penetrating ability ofthe 9R-conjugated aptides.

APTSTAT3-9R inhibits STAT3 activation and downstreamsignaling by specifically blocking STAT3phosphorylation

To examine whether binding of APTSTAT3-9R to STAT3 isable to affect STAT3-mediated signaling pathway, we carriedout STAT3–DNA-binding assays. Human lung carcinoma cells(A549) were chosen because they are known to maintain highlevels of constitutively activated STAT3 (1). The cells weretreated with various concentrations of APTSTAT3-9R (7.5, 15,and 30 mmol/L) or APTscr-9R (30 mmol/L) for 6 hours and thenDNA-binding activity of the activated STAT3 dimer existing inthe cell extracts was measured using a STAT3 TranscriptionFactor Assay Kit (see Supplementary Information). APTSTAT3-9R significantly reduced STAT3–DNA-binding activity in adose-dependent manner, whereas APTscr-9R even at a concen-tration of 30 mmol/L did not affect STAT3–DNA-bindingactivity in A549 cells (Fig. 2A). This result suggests thatAPTSTAT3 is not a just binder but an effector involved ininhibition of homo- or heterodimerization of P-STAT3. To

Figure 2. The inhibitory effects of APTSTAT3-9R on STAT3 signaling. A, DNA-binding assay for STAT3 in cell extracts of A549 human lung carcinomafollowing treatment with APTSTAT3-9R (7.5, 15, and 30 mmol/L) or APTscr-9R (30 mmol/L) for 6 hours. B, quantification of mRNA level of STAT3 downstreamtarget genes, including Bcl-xL, cyclin D1, and survivin, in A549 cells following treatment with APTSTAT3-9R (7.5, 15, and 30 mmol/L) or APTscr-9R (30 mmol/L) for6 hours. C, dose-dependent Western blot assays for STAT3 downstream target proteins (Bcl-xL, cyclin D1, and survivin) and P-STAT3 (Y705) in A549 cellsfollowing treatment with APTSTAT3-9R (7.5, 15, and 30mmol/L) or APTscr-9R (30 mmol/L) for 6 hours. D,Western blot assays for STAT3, P-STAT3 (Y705), and othersignaling in A549 cells following treatment with APTSTAT3-9R and APTscr-9R (30 mmol/L) for 6 hours. �, P < 0.005 (one-way ANOVA); ��, P < 0.001.

Kim et al.

Cancer Res; 74(8) April 15, 2014 Cancer ResearchOF4

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 5: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

further examine the inhibitory effect of APTSTAT3-9R on STAT3downstream signaling, we measured mRNA expression of theSTAT3 target genes, cyclin D1, Bcl-xL, and survivin (10), byquantitative reverse transcription-PCR. Treatment withAPTSTAT3-9R decreased the mRNA levels of the indicatedtargets in a dose-dependent manner in A549 cells, whereasAPTscr-9R (30 mmol/L) treatment had little effect (Fig. 2B).Next, we estimated the expression of these genes at the proteinlevel by Western blotting after treatments of A549 cells withAPTSTAT3-9R (7.5, 15, and 30 mmol/L) or APTscr-9R (30 mmol/L)for 6 hours. As shown in Fig. 2C, the expression level of eachprotein was markedly decreased by treatment with APTSTAT3-9R in a dose-dependent manner compared with that inuntreated controls. As expected, cells treated with nonrelevantAPTscr-9R showed no appreciable difference in the levels ofthese proteins compared with controls. Furthermore, a similartrend of results was also obtained in various murine andhuman cancer cell lines, including B16F1, HepG2, MDA-MB-231, and U87MG that are known to express constitutivelyactivated STAT3, confirming the inhibitory effect ofAPTSTAT3-9R on STAT3 downstream signaling (SupplementaryFigs. S3–S5). Interestingly, we found that APTSTAT3-9R effec-tively inhibited phosphorylation of STAT3 (Fig. 2D) but did notaffect the level of AKT phosphorylation, indicating specificityof the aptide. This suggests that APTSTAT3-9R may be able tosuppress expression of STAT3-associated target genes byspecifically inhibiting STAT3 phosphorylation.On one hand, there are other proteins belonging to STAT

family, some of which have some degree of sequence homologyto STAT3. Thus, we further examined the specificity ofAPTSTAT3 over other STATs. Phage ELISAs using a phagedisplaying APTSTAT3 sequence against STAT3, STAT1, andSTAT5 revealed preferential binding of the aptide phage tothe target STAT3 over STAT1 or STAT5 (Supplementary Fig.S6A). Furthermore, as shown in the Western blot analysis datain A549 cells following treatment with APTSTAT3-9R or APTscr-9R (30 mmol/L; Supplementary Fig. S6B), APTSTAT3-9R signif-icantly inhibited only STAT3 phosphorylation but did notaffect phosphorylation of other STAT1 and STAT5, indicatinghigh specificity of the STAT3 aptide. On the other hand, it isknown that the SH2 domain of STAT3 plays a key role inmembrane recruitment, recognition of the phosphorylatedsite, and subsequent homodimerization of P-STAT3 (7, 21).To elucidate the binding site of APTSTAT3 in STAT3, we carriedout binding assays using phage ELISAs for STAT3 and itsdomains: DBD, SH2 domain, and transactivation domain (TA).A phage displaying APTSTAT3 sequence showed much higherbinding to SH2 domain than other domains (TA and DBD),which was close to the level of binding to parent STAT3;however, moderate binding to DBD was also observed (Sup-plementary Fig. S7A). This finding suggests that the SH2domain may be a putative binding site of the aptide. Further-more, to obtain a piece of clues about the mode-of-action ofAPTSTAT3, we carried out a JAK2 kinase activity assay as thekinase is a key enzyme responsible for STAT phosphorylation.As illustrated in Supplementary Fig. S7B and S7C, phosphor-ylation of the plate-immobilized STAT3 by JAK2 was signifi-cantly inhibited with increasing amounts of APTSTAT3-9R,

whereas the control scrambled peptide minimally affected theJAK2 activity. Although more studies such as the X-ray crys-tallographic study on the STAT3–APTSTAT3 complex arerequired to probe the exact binding site and the mode-of-action of APTSTAT3, we speculate that tight binding of APTSTAT3to the SH2 domainmay inhibit the interaction between STAT3and JAK2 during the membrane recruitment step, therebycausing inhibition of STAT3 phosphorylation.

APTSTAT3-9R suppresses cell viability andproliferation ofcancer cells

To examine whether the inhibitory effect of APTSTAT3-9R onSTAT3 signaling affects cancer cell viability, we performedMTT assays. The IC50 value of APTSTAT3-9Rwas estimated to be10 to 20 mmol/L in A549 cells; in contrast, those of APTScr-9Rand 9R alone were estimated to be >100 mmol/L (Fig. 3A). Asimilar level of IC50 values was attained in B16F1 and HepG2cells treated with APTSTAT3-9R (Supplementary Figs. S3D andS4C). In addition, we carried out colony formation assay toassess whether APTSTAT3-9R suppresses cancer cell prolifera-tion. Proliferation of A549 and B16F1 cancer cells was mon-itored for 2 weeks after cell seeding (500 cells/well) in thepresence of APTSTAT3-9R or APTscr-9R (30 mmol/L). Colonyformation was significantly suppressed in the APTSTAT3-9R–treated group but not in the APTscr-9R group (Fig. 3B andSupplementary Fig. S3E). Furthermore, we confirmed thatAPTSTAT3-9R treatment induced apoptosis of A549 cells byapproximately 3-fold increase compared with APTscr-9R inflow cytometry assessment (Fig. 3C). Collectively, these resultsindicate that APTSTAT3-9R effectively suppresses cell survivaland growth.

APTSTAT3-9R suppresses tumor growth in vivoEncouraged by the potency of APTSTAT3-9R in inhibiting cell

proliferation in vitro, we explored the antitumor efficacy of theaptide in vivo. A human lung carcinoma xenograft model wasprepared by subcutaneous implantation of A549 cancer cells.After tumors had reached a volume of approximately 50 mm3

(2 weeks), APTSTAT3-9R or APTscr-9R (8 mg/kg) was intratu-morally injected every other day for a total of four injections.Tumor burdenwas significantly reduced (�65% relative to thecontrol) in the APTSTAT3-9R–treated group (Fig. 4A) com-pared with that in the PBS control group; however, there waslittle difference in tumor size between APTscr-9R and PBSgroups. Images of excised tumors at day 30 further indicatesignificant difference between the aptide-treated group andcontrol groups (Fig. 4B). We found that the amount of P-STAT3 in the excised tumor tissues of the APTSTAT3-9R–treated group was substantially diminished, whereas APTscr

-9R did not show any appreciable inhibition of STAT3 phos-phorylation in the tumor tissues (Fig. 4C). Furthermore,TUNEL (terminal deoxynucleotidyl transferase–mediateddUTP nick end labeling) assays on excised tumor tissuesrevealed that, unlike PBS or the scrambled aptide–treatedgroups, the APTSTAT3-9R–treated group showed a significantlevel of apoptosis in tumor sections, presumably due toinhibition of STAT3 activation and downstream signaling(Fig. 4D). In addition to the A549 xenograft mouse model,

STAT3-Targeting, High-Affinity Peptide for Cancer Therapeutics

www.aacrjournals.org Cancer Res; 74(8) April 15, 2014 OF5

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 6: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

we also evaluated the anticancer efficacy of APTSTAT3-9R intwo additional tumor models, B16F1 melanoma allograft andU87MG glioblastoma xenograft models. Because B16F1 mel-anoma tumor is known to be aggressive and rapidly grows,APTSTAT3-9R was injected to the same foot pad in which themelanoma cells were implanted 2 days ago as suggested in theprotocol published elsewhere (25). The APTSTAT3-9R substan-tially reduced tumor burden by approximately 49% and, thus,extended survival time by more than 15 days compared withthe control (PBS-treated) group (Supplementary Fig. S8). Inthe case of the U87MG glioblastoma xenograft tumor modelthe first intratumoral injection of each group was initiatedwhen the tumor volumes had reached approximately 120mm3

and terminated after a total of four times injection. The tumorgrowth was significantly inhibited by treatment of the STAT3-specific aptide but little inhibition was seen from the scram-bled aptide–treated group (�45% vs. �7% inhibition; Supple-mentary Fig. S9). Collectively, these results indicate thatAPTSTAT3-9R could effectively suppress tumor growth in the

in vivo allograft and xenograft tumor models by inhibitingSTAT3 phosphorylation.

ConclusionIn summary, we screened and identified a specific STAT3-

binding peptide (APTSTAT3) using aptide platform technology.With the addition of a cell-penetrating motif, the resultingAPTSTAT3-9R aptide was able to suppress the viability andproliferation of cancer cells by blocking STAT3 phosphoryla-tion, thereby inhibiting STAT3 downstream signaling. Further-more, intratumorally injected APTSTAT3-9R showed potentantitumor activity in both xenograft and allograft tumormodels. To the best of our knowledge, APTSTAT3-9R is the firstexample of a peptide-based STAT3 inhibitor demonstratingpotent antitumor efficacy in vivo. Taken together, these find-ings suggest that APTSTAT3 holds potential for the treatment ofcancers maintaining high level of constitutively activatedSTAT3. Further works are currently undergoing to examine

Figure 3. The inhibitory effects of APTSTAT3-9R on cell viability and proliferation in A549 human lung carcinoma. A, cytotoxicity of APTSTAT3-9R, APTscr-9R, and9R toward A549 human lung carcinoma cells following treatment with each peptide for 12 hours, measured by the MTT assay. Cell viability wasexpressed as a percentage (Means � SD; n ¼ 5) relative to the control (100%). B, anchorage-independent proliferation of A549 cells following treatmentwith APTSTAT3-9R or APTscr-9R (30 mmol/L), as determined by colony-forming assays. Colonies were stained with Coomassie blue. C, apoptosis ofA549 cells treated with APTSTAT3-9R or APTscr-9R (30 mmol/L) for 6 hours was assessed by using flow cytometry. Annexin V–FITC and propidium iodide (PI)staining were denoted in x- and y-axis, respectively. The number represents the percentage of apoptotic cells (upper right quadrant).

Kim et al.

Cancer Res; 74(8) April 15, 2014 Cancer ResearchOF6

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 7: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

the feasibility of the aptide in treating STAT3-associatedinflammation diseases.

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

Authors' ContributionsConception and design: D. Kim, I.-H. Lee, S. JonDevelopment of methodology: D. Kim, I.-H. Lee, S. KimAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): D. Kim, M. Choi, H. Kim, S. Ahn, P.E. Saw, H. Jeon,Y. LeeAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):D. Kim, M. Choi, H. Kim, S. Ahn, H. Jeon, Y. Lee, S. Jon

Writing, review, and/or revision of the manuscript: D. Kim, P.E. Saw, S. JonStudy supervision: S. Jon

Grant SupportThis work was supported by the Radiation Technology R&D Program

(2013043391) and Global Research Laboratory (2013056349) through the Nation-al Research Foundation of Korea funded by the Ministry of Science, ICT, andFuture Planning.

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 indicate thisfact.

Received August 2, 2013; revised January 17, 2014; accepted February 3, 2014;published OnlineFirst February 27, 2014.

References1. YuH, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a

leading role for STAT3. Nat Rev Cancer 2009;9:798–809.2. YuH,KortylewskiM,Pardoll D.Crosstalk between cancer and immune

cells: role of STAT3 in the tumourmicroenvironment. Nat Rev Immunol2007;7:41–51.

3. Yue P, Turkson J. Targeting STAT3 in cancer: how successful are we?Expert Opin Investig Drugs 2009;18:45–56.

4. Benekli M, Baumann H, Wetzler M. Targeting signal transducer andactivator of transcription signaling pathway in leukemias. J Clin Oncol2009;27:4422–32.

Figure 4. Antitumor activity of APTSTAT3-9R in an A549 xenograft model. A, the tumor growth study in the A549 xenograft model. Nude mice bearing A549tumors (�50 mm3) were treated four times with APTSTAT3-9R (8 mg/kg), APTscr-9R (8 mg/kg), or PBS. Tumor volumes were measured every other day usingcalipers.Mean tumor size�SEM (n¼5). Arrows indicate the injection day. B, a photographof tumors excised frommice after 30 daysof treatment. C,Westernblot assays for STAT3 and P-STAT3 (Y705) in the excised tumor tissues. D, TUNEL assays of tumor sections. ��, P < 0.001 (one-way ANOVA).

STAT3-Targeting, High-Affinity Peptide for Cancer Therapeutics

www.aacrjournals.org Cancer Res; 74(8) April 15, 2014 OF7

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 8: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

5. Blaskovich MA, Sun JZ, Cantor A, Turkson J, Jove R, Sebti SM.Discovery of JSI-124 (Cucurbitacin I), a selective janus kinase/signaltransducer and activator of transcription 3 signaling pathway inhibitorwith potent antitumor activity against humanandmurine cancer cells inmice. Cancer Res 2003;63:1270–9.

6. SiddiqueeK, ZhangS,GuidaWC,BlaskovichMA,GreedyB, LawrenceHR, et al. Selective chemical probe inhibitor of Stat3, identified throughstructure-based virtual screening, inducesantitumor activity. ProcNatlAcad Sci U S A 2007;104:7391–6.

7. Lin L, Hutzen B, Zuo MX, Ball S, Deangelis S, Foust E, et al. NovelSTAT3 phosphorylation inhibitors exhibit potent growth-suppressiveactivity in pancreatic and breast cancer cells. Cancer Res 2010;70:2445–54.

8. Xu S, Grande F, Garofalo A, Neamati N. Discovery of a novel orallyactive small-molecule gp130 inhibitor for the treatment of ovariancancer. Mol Cancer Ther 2013;12:937–49.

9. Zhang X, Sun Y, Pireddu R, Yang H, Urlam MK, Lawrence HR, et al. Anovel inhibitor of STAT3 homodimerization selectively suppressesSTAT3 activity and malignant transformation. Cancer Res 2013;73:1922–33.

10. Zhang XL, Jian Z, Wang LH, Wei HM, Tian ZG. Therapeutic effects ofSTAT3 decoy oligodeoxynucleotide on human lung cancer in xeno-graft mice. Bmc Cancer 2007;7:149.

11. AlshamsanA,HamdyS, Samuel J, El-Kadi AOS, Lavasanifar A, UludagH. The inductionof tumor apoptosis inB16melanoma followingSTAT3siRNA delivery with a lipid-substituted polyethylenimine. Biomaterials2010;31:1420–8.

12. Borghouts C, Kunz C, Delis N, Groner B. Monomeric recombinantpeptide aptamers are required for efficient intracellular uptake andtarget inhibition. Mol Cancer Res 2008;6:267–81.

13. ZhaoW, JaganathanS, Turkson J. A cell-permeable Stat3SH2domainmimetic inhibits Stat3 activation and induces antitumor cell effects invitro. J Biol Chem 2010;285:35855–65.

14. Turkson J, Kim JS, Zhang SM, Yuan J, Huang M, Glenn M, et al. Novelpeptidomimetic inhibitors of signal transducer and activator of tran-scription 3 dimerization and biological activity. Mol Cancer Ther2004;3:261–9.

15. Shahani VM, Yue PB, Fletcher S, Sharmeen S, Sukhai MA, Luu DP,et al. Design, synthesis and in vitro characterization of novel hybrid

peptidomimetic inhibitors of STAT3 protein. Bioorg Med Chemi2011;19:1823–38.

16. Xi SC, Gooding WE, Grandis JR. In vivo antitumor efficacy of STAT3blockade using a transcription factor decoy approach: implications forcancer therapy. Oncogene 2005;24:970–9.

17. Turkson J, Ryan D, Kim JS, Zhang Y, Chen Z, Haura E, et al.Phosphotyrosyl peptides block Stat3-mediated DNA-binding activ-ity, gene regulation, and cell transformation. J Biol Chem 2001;276:45443–55.

18. ColemanDR, Ren ZY,Mandal PK, Cameron AG, Dyer GA, Muranjan S,et al. Investigation of the binding determinants of phosphopeptidestargeted to the Src homology 2 domain of the signal transducer andactivator of transcription 3. Development of a high-affinity peptideinhibitor. J Med Chem 2005;48:6661–70.

19. Nagel-Wolfrum K, Buerger C, Wittig I, Butz K, Hoppe-Seyler F, GronerB. The interaction of specific peptide aptamers with the DNA-bindingdomain and the dimerization domain of the transcription factor Stat3inhibits transactivation and induces apoptosis in tumor cells. MolCancer Res 2004;2:170–82.

20. Timofeeva OA, Gaponenko V, Lockett SJ, Tarasov SG, Jiang S,Michejda CJ, et al. Rationally designed inhibitors identify STAT3 N-domain as a promising anticancer drug target. Acs Chem Biol 2007;2:799–809.

21. Debnath B, Xu SL, Neamati N. Small molecule inhibitors of signaltransducer and activator of transcription 3 (Stat3) protein. JMedChem2012;55:6645–68.

22. Kim S, Kim D, Jung HH, Lee IH, Kim JI, Suh JY, et al. Bio-inspireddesign and potential biomedical applications of a novel class of high-affinity peptides. Angew Chem-Int Ed 2012;51:1890–4.

23. ZhangX, Zhang J,WangL,WeiH, Tian Z. Therapeutic effects of STAT3decoy oligodeoxynucleotide on human lung cancer in xenograft mice.BMC Cancer 2007;7:149.

24. Futaki S, Suzuki T, Ohashi W, Yagami T, Tanaka S, Ueda K, et al.Arginine-richpeptides—Anabundant source ofmembrane-permeablepeptides having potential as carriers for intracellular protein delivery.J Biol Chem 2001;276:5836–40.

25. Lee IH, Park YT, RohK,ChungH, Kwon IC, JeongSY. Stable paclitaxelformulations in oily contrast medium. J Control Release 2005;102:415–25.

Cancer Res; 74(8) April 15, 2014 Cancer ResearchOF8

Kim et al.

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187

Page 9: fic STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity … · A Specific STAT3-Binding Peptide Exerts Antiproliferative Effects and Antitumor Activity

Published OnlineFirst February 27, 2014.Cancer Res   Daejin Kim, In-Hyun Lee, Sunghyun Kim, et al.   Phosphorylation and SignalingEffects and Antitumor Activity by Inhibiting STAT3 A Specific STAT3-Binding Peptide Exerts Antiproliferative

  Updated version

  10.1158/0008-5472.CAN-13-2187doi:

Access the most recent version of this article at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/early/2014/03/26/0008-5472.CAN-13-2187To request permission to re-use all or part of this article, use this link

Research. on June 18, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2014; DOI: 10.1158/0008-5472.CAN-13-2187