regulation of hif1a under hypoxia by ape1/ref-1...

12
Cancer Biology and Signal Transduction Regulation of HIF1a under Hypoxia by APE1/Ref-1 Impacts CA9 Expression: Dual Targeting in Patient-Derived 3D Pancreatic Cancer Models Derek P. Logsdon 1 , Michelle Grimard 2 , Meihua Luo 2 , SaShahda 3,4 , Yanlin Jiang 2 , Yan Tong 5 , Zhangsheng Yu 5 , Nicholas Zyromski 4,6 , Ernestina Schipani 7 , Fabrizio Carta 8 , Claudiu T. Supuran 8 , Murray Korc 4,9,10 , Mircea Ivan 3,4,11 , Mark R. Kelley 1,2,4,10 , and Melissa L. Fishel 1,2,4 Abstract Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related mortality in the United States. Aggressive treatment regimens have not changed the disease course, and the median survival has just recently reached a year. Several mechan- isms are proposed to play a role in PDAC therapeutic resistance, including hypoxia, which creates a more aggressive phenotype with increased metastatic potential and impaired therapeutic efcacy. AP Endonuclease-1/Redox Effector Factor 1 (APE1/Ref-1) is a multifunctional protein possessing a DNA repair function in base excision repair and the ability to reduce oxidized transcription factors, enabling them to bind to their DNA target sequences. APE1/Ref-1 regulates several transcription factors involved in survival mechanisms, tumor growth, and hypoxia signaling. Here, we explore the mechanisms underlying PDAC cell responses to hypoxia and modulation of APE1/Ref-1 redox signaling activity, which regulates the transcriptional activation of hypoxia-inducible factor 1 alpha (HIF1a). Carbonic anhydrase IX (CA9) is regulated by HIF1a and functions as a part of the cellular response to hypoxia to regulate intracellular pH, thereby promoting cell survival. We hypothesized that modulating APE1/Ref-1 function will block activation of downstream transcription factors, STAT3 and HIF1a, interfering with the hypoxia-induced gene expression. We dem- onstrate APE1/Ref-1 inhibition in patient-derived and established PDAC cells results in decreased HIF1amediated induction of CA9. Furthermore, an ex vivo three-dimensional tumor coculture model demonstrates dramatic enhancement of APE1/Ref-1induced cell killing upon dual targeting of APE1/Ref-1 and CA9. Both APE1/Ref-1 and CA9 are under clinical development; there- fore, these studies have the potential to direct novel PDAC ther- apeutic treatment. Mol Cancer Ther; 15(11); 272232. Ó2016 AACR. Introduction Pancreatic ductal adenocarcinoma (PDAC) continues to be the fourth leading cause of cancer-related mortality in the United States in both genders with a ve-year survival rate of 5%7% (1). Approximately 80% of patients present with advanced disease due to local invasion or metastatic disease (1, 2). Treatment options for this patient population are limited to palliative chemotherapy, and even the most aggressive treatment strategies fail to extend life beyond one year for most patients (1, 3). The disappointing result in pancreatic cancer may be explained by the complexity of this disease. Several mechanisms of resistance have been proposed to play a role in resistance to therapy. The presence of cancer-associated broblasts (CAF) as well as other cell types within a desmoplastic stroma, hypoperfusion of the tumor, hypoxia, multidrug resistance, and other mechanisms have been reported (46). Lack of clinical efcacy is due, at least in part, to the brosis that accompanies the disease. In the studies presented here, tumorstroma effects were monitored as the importance of the stroma in PDAC is well established (7, 8). Targeting one pathway is very unlikely to alter the natural course of this disease. However, affecting critical survival mechanisms of this cancer is crucial to produce any positive results (9, 10). Hypoxic conditions in pancreatic tumors are associated with poor prognosis. Oxygen deprivation leads to stabilization of hypoxia-inducible factor 1 alpha (HIF1a), a transcription factor that upregulates a variety of factors that contribute to increased drug resistance, proliferation, and migration/inva- sion in tumor cells (1113). HIF-1 transcriptional activity depends on stabilization of its a subunit, which is targeted 1 Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. 2 Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana. 3 Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, India- napolis, Indiana. 4 Pancreatic Cancer Signature Center, Indianapolis, Indiana. 5 Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana. 6 Department of Surgery, Indiana Uni- versity School of Medicine, Indianapolis, Indiana. 7 Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan. 8 Neurofarba Department, Section of Medicinal Chemistry, University of Florence, Florence, Italy. 9 Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana. 10 Department of Biochemistry and Molecular Biology, Indiana Univer- sity School of Medicine, Indianapolis, Indiana. 11 Department of Micro- biology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Melissa L. Fishel, Department of Pediatrics, Herman B Wells Center for Pediatric Research, 1044 W Walnut St, R4-312, Indianapolis, IN 46202. Phone: 317-274-8810; E-mail: m[email protected] doi: 10.1158/1535-7163.MCT-16-0253 Ó2016 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 15(11) November 2016 2722 on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Upload: buithuy

Post on 19-Jun-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

Cancer Biology and Signal Transduction

Regulation of HIF1a under Hypoxia by APE1/Ref-1Impacts CA9 Expression: Dual Targeting inPatient-Derived 3D Pancreatic Cancer ModelsDerek P. Logsdon1, Michelle Grimard2, Meihua Luo2, Safi Shahda3,4, Yanlin Jiang2,Yan Tong5, Zhangsheng Yu5, Nicholas Zyromski4,6, Ernestina Schipani7, Fabrizio Carta8,Claudiu T. Supuran8, Murray Korc4,9,10, Mircea Ivan3,4,11, Mark R. Kelley1,2,4,10, andMelissa L. Fishel1,2,4

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leadingcause of cancer-related mortality in the United States. Aggressivetreatment regimens have not changed the disease course, and themedian survival has just recently reached a year. Several mechan-isms are proposed to play a role in PDAC therapeutic resistance,including hypoxia, which creates a more aggressive phenotypewith increased metastatic potential and impaired therapeuticefficacy. AP Endonuclease-1/Redox Effector Factor 1 (APE1/Ref-1)is a multifunctional protein possessing a DNA repair function inbase excision repair and the ability to reduceoxidized transcriptionfactors, enabling them to bind to their DNA target sequences.APE1/Ref-1 regulates several transcription factors involved insurvival mechanisms, tumor growth, and hypoxia signaling. Here,we explore the mechanisms underlying PDAC cell responses tohypoxia and modulation of APE1/Ref-1 redox signaling activity,

which regulates the transcriptional activationof hypoxia-induciblefactor 1 alpha (HIF1a). Carbonic anhydrase IX (CA9) is regulatedbyHIF1a and functions as apart of the cellular response tohypoxiato regulate intracellular pH, thereby promoting cell survival. Wehypothesized that modulating APE1/Ref-1 function will blockactivation of downstream transcription factors, STAT3 andHIF1a,interfering with the hypoxia-induced gene expression. We dem-onstrate APE1/Ref-1 inhibition in patient-derived and establishedPDAC cells results in decreased HIF1a–mediated induction ofCA9. Furthermore, an ex vivo three-dimensional tumor coculturemodel demonstrates dramatic enhancement of APE1/Ref-1–induced cell killing upon dual targeting of APE1/Ref-1 and CA9.Both APE1/Ref-1 and CA9 are under clinical development; there-fore, these studies have the potential to direct novel PDAC ther-apeutic treatment. Mol Cancer Ther; 15(11); 2722–32. �2016 AACR.

IntroductionPancreatic ductal adenocarcinoma (PDAC) continues to be the

fourth leading cause of cancer-related mortality in the United

States in both genders with a five-year survival rate of 5%–7% (1).Approximately 80% of patients present with advanced diseasedue to local invasion or metastatic disease (1, 2). Treatmentoptions for this patient population are limited to palliativechemotherapy, and even the most aggressive treatment strategiesfail to extend life beyond one year for most patients (1, 3). Thedisappointing result in pancreatic cancer may be explained by thecomplexity of this disease. Several mechanisms of resistance havebeen proposed to play a role in resistance to therapy. The presenceof cancer-associated fibroblasts (CAF) as well as other cell typeswithin a desmoplastic stroma, hypoperfusion of the tumor,hypoxia, multidrug resistance, and other mechanisms have beenreported (4–6). Lack of clinical efficacy is due, at least in part, tothe fibrosis that accompanies the disease. In the studies presentedhere, tumor–stroma effects were monitored as the importance ofthe stroma in PDAC is well established (7, 8). Targeting onepathway is very unlikely to alter the natural course of this disease.However, affecting critical survival mechanisms of this cancer iscrucial to produce any positive results (9, 10).

Hypoxic conditions in pancreatic tumors are associated withpoor prognosis. Oxygen deprivation leads to stabilization ofhypoxia-inducible factor 1 alpha (HIF1a), a transcriptionfactor that upregulates a variety of factors that contribute toincreased drug resistance, proliferation, and migration/inva-sion in tumor cells (11–13). HIF-1 transcriptional activitydepends on stabilization of its a subunit, which is targeted

1Department of Pharmacology and Toxicology, Indiana UniversitySchool of Medicine, Indianapolis, Indiana. 2Department of Pediatrics,Wells Center for Pediatric Research, Indiana University School ofMedicine, Indianapolis, Indiana. 3Division of Hematology/Oncology,Department of Medicine, Indiana University School ofMedicine, India-napolis, Indiana. 4Pancreatic Cancer Signature Center, Indianapolis,Indiana. 5Department of Biostatistics, Indiana University School ofMedicine, Indianapolis, Indiana. 6Department of Surgery, Indiana Uni-versity School of Medicine, Indianapolis, Indiana. 7Department ofOrthopaedic Surgery, University of Michigan, Ann Arbor, Michigan.8Neurofarba Department, Section of Medicinal Chemistry, Universityof Florence, Florence, Italy. 9Divisionof Endocrinology, Department ofMedicine, IndianaUniversity School of Medicine, Indianapolis, Indiana.10Department of Biochemistry and Molecular Biology, Indiana Univer-sity School of Medicine, Indianapolis, Indiana. 11Department of Micro-biology and Immunology, Indiana University School of Medicine,Indianapolis, Indiana.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

Corresponding Author: Melissa L. Fishel, Department of Pediatrics, Herman BWells Center for Pediatric Research, 1044 WWalnut St, R4-312, Indianapolis, IN46202. Phone: 317-274-8810; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-16-0253

�2016 American Association for Cancer Research.

MolecularCancerTherapeutics

Mol Cancer Ther; 15(11) November 20162722

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 2: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

for degradation under normoxic conditions by proline hydrox-ylation and subsequent von Hippel-Lindau protein (VHL)-mediated ubiquitination. Stable HIF1a dimerizes with theconstitutively expressed b subunit to activate genes with hyp-oxia-response elements (HRE) in their promoters (14, 15). NoHIF-1–specific inhibitors currently exist, so targeting its vitaltranscriptional targets and the enzymes that regulate HIF-1activity are promising ways to modulate hypoxia signaling incancer cells (15, 16).

Apurinic/apyrimidinic endonuclease/redox factor-1 (APE1/Ref-1) is a multifunctional protein that is involved in responsesto oxidative stress, acting on both oxidative and alkylative DNAdamage (via its endonuclease activity in base excision repair;refs. 17–19) and augmenting activity of various transcriptionfactors (via its redox signaling; refs. 20–23) as well as contributingto clearance of RNAwith damaged bases (24). APE1/Ref-1 expres-sion and redox activity are increased in PDAC tissue, and itsupregulation increases tumor cell migration, proliferation, andsurvival (17, 23). We previously demonstrated that APE1/Ref-1redox activity regulates several transcription factors involved inpancreatic cancer signaling, including HIF-1a, as well as STAT3and NFkB (20, 21).

Carbonic anhydrase IX (CA9) is a transmembrane proteinthat regulates pH in tumor cells under low oxygen conditionsand contributes to cell proliferation and transformation(25, 26). CA9 was reported to be an endogenous sensor toHIF-1 activity (27) and CA9 inhibitors were more potent in 3Dculture due to the regions of hypoxia (28). One report dem-onstrated a strong correlation in vivo between pSTAT3 and CA9expression and postulated that the regulation of CA9 by STAT3could be important in invasion through IL6 signaling (29).Both of these transcription factors are regulated by APE1/Ref-1,so the impact of APE1/Ref-1 redox activity on HIF1 activity aswell as CA9 expression was evaluated in PDAC followingexposure to hypoxia.

Here we describe the mechanisms by which APE1/Ref-1regulates hypoxia signaling through HIF1a-mediated transcrip-tion. Inhibiting APE1/Ref-1 redox signaling with the selectiveinhibitor APX3330 (also called E3330) resulted in decreasedSTAT3 activity (20) and HIF1a activity (21) leading todecreased expression of CA9, a major HIF-1 target within cells(27). We also demonstrate that hypoxia stimulates interactionsbetween APE1/Ref-1 and its redox targets, HIF1a and STAT3,but not NFkB in both PDAC tumor and stromal cells support-ing our approach.

We also show for the first time evidence using patient-derivedtumor cells in the presence of appropriate stromal components,CAFs, three-dimensional spheroid size, and proliferation aredramatically reduced upon combination treatment with APE1/Ref-1 inhibitor, APX3330 and CA9 inhibitor, SLC-0111 (Clin-ical Trial NCT02215850; refs. 30, 31). Upon blockade ofmultiple hypoxia signaling pathways with APX3330 andSLC-0111, we observe a dramatic effect on 3D tumor spheroidgrowth even in the presence of the protective environment ofthe CAFs. APE1/Ref-1 redox inhibitor APX3330 (20, 22, 32, 33)is slated for phase I clinical trials in late 2016, and the CA9inhibitor is currently in clinical trials. By decreasing APE1/Ref-1redox activity, we can impair the tumor cells' response tohypoxia (21) and potentially improve the response to therapy.Therefore, the combination of these agents may have proximateclinical applications.

Materials and MethodsCell culture

Cells were maintained in culture as described previously(20–22). Patient-derived tumor cells, Pa03C, Pa02C, andPanc10.05 and CAF19 cells, were a kind gift from Dr. AnirbanMaitra (The Johns Hopkins University, Baltimore, MD; ref. 34).Upon receipt of the cells in 2011 as well as in June of 2015, weused STR analysis (CellCheck with IDEXX BioResearch) to con-firm that we indeed received the aforementioned cells fromDr. Anirban Maitra and that they were mycoplasma-free. MIA-PaCa2 cells were purchased from and authenticated by ATCC.Cancer-associated fibroblasts, UH1303-02, were isolated usingthe outgrowth method from patient tumor tissue as describedpreviously (35). All patient-derived lines were passaged only10 times before new stocks frozen prior to authentication wereresuscitated. Hypoxia exposure was achieved using a RuskinnInvivo2 200 hypoxia workstation. CMV-EGFP-WT APE1/Ref-1lentiviral construct was used to overexpress APE1/Ref-1 asdescribed previously (36). To detect the cells for imaging, aCMV-EGFP lentiviral construct was used as described previous-ly (36). In addition, 150 pfu/cell of the pCL7TdTOMwo lenti-viral vector was incubated with Pa03C and Panc10.05 cells for48 hours to make cells stably express TdTomato.

Western blot analysisWestern blots were performed as described previously (20–23)

with antibodies for APE1/Ref-1 (Novus Biologicals), HIF1a (Gen-eTex), STAT1, STAT3, (Cell Signaling Technology), NFkB(Abcam), CA9 (Santa Cruz Biotechnology), Vinculin (Sigma),and Actin (NeoMarkers).

CoimmunoprecipitationSamples were coimmunoprecipitated using the Pierce Co-IP

kit (Thermo Scientific) with modifications as described previ-ously (22).

TransfectionPDAC andCAF cells were transfectedwithAPE1/Ref -1 siRNA as

described previously (20, 22, 23). siRNAs used were: #1 orscrambled control (previously reported) and two LifeTech vali-dated siRNAs (#2: s1445 and #4: s1447; ref. 22). APE1/Ref-1siRNA #1 was used as the standard siRNA unless otherwisespecified.

Transient luciferase reporter assaysMIA-PaCa-2 cells were cotransfected with constructs contain-

ing luciferase driven by HIF1a and a Renilla luciferase reportervector as described previously (22) using X-tremeGENE 9 DNAtransfection reagent (Roche) along with siRNA as describedabove. Firefly and Renilla luciferase activities were assayed usingthe Dual Luciferase Reporter Assay System (Promega Corp.) asbefore (20, 22).

qRT-PCR reactionsqRT-PCR was used to measure the mRNA expression levels of

CA9 as described previously (21). Cells were treated with APE1/Ref-1 siRNA or increasing amounts of APX3330 in the presence orabsence of hypoxia (1% and 0.2% O2) for 24 hours, then totalRNA was extracted from cells using the Qiagen RNeasy Mini Kit(Qiagen; ref. 37). First-strand cDNA synthesis and quantitative

Dual-Targeting APE1/Ref-1 and CA9 in Hypoxic PDAC Cells

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2723

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 3: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

PCR were performed as described previously (21). The relativequantitative mRNA level was determined using the comparativeCt method using 18S rRNA, RPLP0, and B2M as reference genes(37, 38). The primers for CA9, 18S, RPLP0, and B2M are com-mercially available (Applied Biosystems).

InhibitorsCompounds were prepared and used as described previously:

APX3330 (32, 33) and SLC-0111 (30, 31). The concentrations ofAPX3330 used are within clinically tolerated levels establishedpreviously by Eisai pharmaceutical company through a priordevelopment program that evaluated the toxicology and phaseI/II safety and clinical profile in noncancer patients. In addition,the SLC-0111 analogue FC13-555A was synthesized as describedin SupplementaryMethods. The structure of each inhibitor can beseen in Supplementary Fig. S1.

Cell proliferationPDAC cell proliferation in monolayer was measured using the

Alamar Blue assay as described previously (22). Cells treated withAPX3330 and SLC-0111 were exposed to hypoxia for 6 daysfollowed by addition of AlamarBlue reagent (Invitrogen) andsubsequent fluorescence analysis. Fold change refers to the fluo-rescence reading for cells treated with indicated inhibitors com-pared with cells growing in normal media.

pH assayIntracellular pH was evaluated using the pHrodo Red AM

Intracellular pH Indicator (LifeTech). PDAC cells treated withAPX3330 and SLC-0111 were exposed to hypoxia for 48 hoursfollowed by analysis with pHrodo Red AM dye. Intracellular pHCalibration Buffers (LifeTech)were used to create a standard curveof fluorescence intensity for determination of pH values. Resultswere normalized to MTS analysis to account for changes inproliferation as before (20–22). Fluorescent imageswere acquiredusing a confocal/two-photon Olympus Fluoview FV-1000 MPEsystem (Olympus American) at the Indiana Center for BiologicalMicroscopy facility.

Statistical analysisqPCR data points for scrambled, siAPE1/Ref-1, and hypoxia

treatments were analyzed using the 2�DDCt method and analysisof covariance (ANCOVA) models as described previously(22, 39). Data points in tests with multiple treatment groupswere analyzed using post hoc multiple comparisons tests (Tukey,Dunnett, or Sidak as appropriate). For evaluation of data curvesusing multiple drugs, an extra-sum-of-squares F test was used tocompare the goodness-of-fit of a nonlinear regression curveshared between groups with that of separate curves for eachgroup. Differences between the treatment groups and controlgroupwere considered significant if P < 0.05 following Bonferronicorrections was appropriate. Statistical analyses were performedusing SAS (Version 9.3, Copyright 2010 SAS Institute Inc.) andPrism (Version 6.0f, Copyright 2014 GraphPad Software Inc.).

HIF-1�/� MEF generationHIF-1–floxed mouse embryonic fibroblast (MEF) cells were

generated as reported previously (40) and transduced with Ad-CMV-Cre (Cre adenovirus) or Ad-GFP (control) vector (VectorBioLabs) for 24 hours using 5 ng/mL polybrene to produce

HIF-1–deficient cells (40). PCR was used to verify the deletionof HIF (Supplementary Fig. S2).

3D CoculturesUltra-low attachment 96-well plates (Corning Inc., Life

Sciences) were used to generate three-dimensional (3D) tumorspheroids in the presence and absence of CAFs (75 mL/well) asdescribed previously (41, 42). Cells were stably transduced withEGFP (green) or TdTomato (red) as indicated to preserve thegenetic characteristics of the low passage patient cells (34, 42).Cells were resuspended in colorless DMEM growth media con-taining 3% reduced growth factor Matrigel (RGF, BD Biosciences)and 5% FBS. After plating, cells were treated on days 4 and 8 withmedia containing 5% serum, 3% RGF Matrigel, and inhibitors asindicated. On day 12, spheroids were analyzed using ThermoArrayScan high-content imaging system (43). Images of 3Dstructures were captured by ArrayScan using a 2.5� objective forTdTomato and EGFP; then 2D projections were processed toquantify differences in total intensity and total area of both CAFsand tumor.

ResultsAPE1/Ref-1 interactions with HIF1a and STAT3 are stimulatedby hypoxia

Wepreviously published data demonstrating decreased STAT3,HIF1a, and NFkB activity following knockdown of APE1/Ref-1and/or inhibition of APE1/Ref-1 redox signalingwith the selectiveinhibitor APX3330 (also called E3330; refs. 20, 21). Similarly, wefound that inhibition of APE1/Ref-1 led to a decrease in a majorHIF-1 target within cells, CA9 (21, 28). To further dissect the roleof APE1/Ref-1 in hypoxia signaling and more clearly determinewhether hypoxia stimulates interactions between APE1/Ref-1 andits redox targets, endogenous APE1/Ref-1 was immunoprecipi-tated from lysates of two low-passage PDAC cell lines (Panc10.05and Pa03C) under normoxic and hypoxic (0.2% O2) conditions.These cell lines that are representative of ductal adenocarcinoma,were previously sequenced (34), and have the common G12Dmutation in KRAS as well as missense mutations in p53.Panc10.05 was derived from a primary PDAC tumor, is wild-type(wt) for p16(INK4A), but has a SMAD4deletion; however, Pa03Cwas isolated from ametastatic lesion in the liver and is wt for bothp16(INK4A) and SMAD4. IPs were probed for HIF1a, STAT3, andNFkB.HIF1a and STAT3, but not NFkB, were detected in the pull-down fractions under hypoxic conditions, but these interactionswere not detected under normoxic conditions (Fig. 1A and B).Controls of TNFa (NFkB) and IL6 (STAT3) were performed toshow that interactions between APE1/Ref-1 and the transcriptionfactors it regulates do indeed occur under normoxic conditionswith appropriate stimulation (Supplementary Fig. S3). Interac-tions of APE1/Ref-1 with HIF1a and STAT3 were obvious underhypoxic conditions.

With overexpression of APE1/Ref-1, the interactionwithHIF1aand STAT3 remained intact while NFkB was still not detected inIPs from cells overexpressing APE1/Ref-1 following exposure tohypoxia indicating that the amount of APE1/Ref-1 was notlimiting in the above panels (Fig. 1C and D). To demonstratethat APE1/Ref-1's interactions with the transcription factors arespecific to signaling in hypoxia, we probed for another STATfamily member, STAT1. IPs from Panc10.05 cells were probedfor STAT1, which was not detected regardless of the levels of

Logsdon et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2724

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 4: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

APE1/Ref-1 or oxygen conditions (Fig. 1C). Because of the com-plexity of the disease and the signaling between various cell typesin the pancreatic tumor microenvironment, we investigatedAPE1/Ref-1 interactions with HIF1a, STAT3, and NFkB in CAFs.These CAFs are nontumorigenic and although they have acti-vated signaling pathways due to their association with thetumor, they are nontransformed. The results were identical tothe result with PDAC cells: APE1/Ref-1 interacts with HIF1aand STAT3 under hypoxia, but not NFkB (SupplementaryFig. S4). In light of CA9 inhibitors beginning clinical trials andour previous data demonstrating transcriptional regulation ofCA9 following APE1/Ref-1 blockade (21), we focused thesestudies on HIF-1a signaling and the regulation of the down-stream molecule CA9 through APE1/Ref-1.

APE1/Ref-1 protein expression contributes to hypoxia-inducedHIF1a-mediated transcription

To show that the interactions between APE1/Ref-1 and HIF1aare functionally important, we evaluated the contribution ofAPE1/Ref-1 to HIF-1 transcriptional activity by cotransfectingMIA PaCa-2 cells with HIF1a-driven firefly luciferase or pLuc-MCS (vector control) alongside APE1/Ref-1 siRNA or scrambledcontrol and exposing cells to hypoxia for 24 hours. APE1/Ref-1knockdown resulted in a significant reduction (�47%) in hyp-oxia-induced HIF1a activity (Fig. 2A and B).

Effects of APE1/Ref-1 on HIF transcriptional activity werefurther evaluated by examining hypoxia-mediated transcription

of HIF-1 target, CA9. We compared CA9 mRNA levels in twoPDAC cell lines and one pancreatic CAF cell line following APE1/Ref-1 knockdown and exposure to hypoxia. Bar graph representsthe fold change of mRNA expression level of CA9. Hypoxia-induced CA9mRNA levels were attenuated by APE1/Ref-1 knock-down in all cell lines at both levels of hypoxia (Fig. 2C–E).Variability in the amount of induction in different cell lines maybe partially attributable to the extremely low baseline CA9expression under normoxic conditions. APE1/Ref-1 knockdownsimilarly attenuated CA9mRNA levels under hypoxic conditionsin two additional primary PDAC cell lines (Supplementary Fig.S5). These results were validated in MIA-PaCa-2 cells exposed tohypoxia using two additional APE1/Ref-1–targeting siRNAs, andsimilar results were obtained (Fig. 2F and G). To verify thereduction in CA9 also occurred at the protein level, hypoxia-induced CA9 protein levels were evaluated via Western blotanalysis following APE1/Ref-1 knockdown in PDAC cells andpancreatic CAF cells. APE1/Ref-1 knockdown resulted in anapproximately 70% reduction in hypoxia-induced CA9 proteinlevels (Fig. 2H and I).

Hypoxia-induced CA9 transcription is HIF-1–dependentTo confirm that the effects of APE1/Ref-1 and hypoxia on CA9

transcription were mediated by HIF-1 activity, we evaluatedhypoxia-induced CA9 mRNA levels in HIF-1–deficient (�/�)MEFs following APE1/Ref-1 knockdown. As expected, in HIF-1–proficientMEFs,CA9 is induced30-fold comparedwithnormal

Figure 1.

APE1/Ref-1 interactions with HIF1a and STAT3 are stimulated by hypoxia in PDAC cells. Cell extracts were prepared from Panc10.05 (A) and Pa03C (B)cells and also from cells that overexpress APE1/Ref-1 (C and D) following exposure to hypoxia (0.2%) for 24 hours. Extracts were immunoprecipitatedwith anti-APE1/Ref-1 antibody or IgG. The immunoprecipitated complexes were then probed for HIF1a, STAT3, NFkB, STAT1, or APE1/Ref-1. n ¼ 3–4 perexperiment. Typical results shown.

Dual-Targeting APE1/Ref-1 and CA9 in Hypoxic PDAC Cells

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2725

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 5: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

oxygen controls. In HIF-1�/� MEFs, CA9 mRNA levels were notinduced by exposure to hypoxia (Fig. 3A), or affected by APE1/Ref-1 knockdown (Fig. 3B), indicating that CA9 transcription isHIF-1–dependent, regardless of APE1/Ref-1 expression or oxygenlevels. HIF-1 depletion in these cells was confirmed by PCR(Supplementary Fig. S2).

Inhibition of APE1/Ref-1 redox signaling affects CA9transcription

As a multifunctional protein, APE1/Ref-1 is also involved inbase excision repair (BER) of DNA lesions, RNA quality control,and reduction–oxidation (redox) regulation. Knockdown ofAPE1/Ref-1 affects all of these functions. We therefore examinedwhether the redox function is responsible for the APE1/Ref-1–mediated regulation of hypoxia signaling pathways using anAPE1/Ref-1–specific redox inhibitor that does not affect otherAPE1/Ref-1 functions (44, 45) and is slated for clinical trial in thesummer of 2016. We previously showed that APX3330 decreases

CA9 mRNA levels in Panc-1 and MIA-PaCa2 cells exposed tohypoxia (21). Here we expand these results to primary cells andCAF cells, as well as 3D cocultures. After treatment with APX3330and exposure to hypoxia, CA9mRNA levels in Pa03C cells and inpancreatic CAF cells were attenuated in a dose-dependentmanner(Fig. 3C and D). In addition, CA9 protein expression wasmeasured in a 3D coculture model following inhibition ofAPE1/Ref-1 with APX3330. While CA9 was not detected undernormoxic conditions in the patient-derived Pa03C cells inmonolayer, when grown as spheroids, these cells now expressCA9. Tumor spheroids grown in the presence of CAFs morestrongly upregulated CA9 expression approximately 3-fold,likely due to larger spheroids containing larger regions ofhypoxia, as well as the more complex signaling present withthe stromal elements. Inhibition of APE1/Ref-1 redox signalingwith APX3330 led to decreased CA9 expression in 3D tumorcultures in a dose-dependent manner, both in the presence andabsence of CAF cells (Fig. 3E). These data support the use of the

Figure 2.

APE1/Ref-1 protein expression contributes to hypoxia-induced HIF1a-mediated transcription. MIA-PaCa2 cells were assayed for HIF1 activity using a luciferaseand Renilla reporter assay following APE1/Ref-1 knockdown. A, knockdown of APE1/Ref-1 was confirmed via Western blot analysis. B, HIF1a-driven luciferaseexpression was evaluated following hypoxia (24 hours, 0.2% oxygen vs. normoxia controls; n ¼ 3). C–E, following APE1/Ref-1 knockdown and 24 hours inhypoxia, CA9mRNA levels were evaluated via qPCR in the cell lines described (n¼ 3). F, APE1/Ref-1 knockdown in MIA-PaCa2 cells with three different siRNAs wasconfirmed via Western blot analysis. G, CA9 mRNA levels were evaluated via qPCR in SC and knocked-down samples from three siRNAs following hypoxicconditions (24 hours, 0.2% oxygen vs. normoxia controls; representative experiment of n ¼ 3). H and I, CA9 protein levels were evaluated via Western blotanalysis in Panc10.05 (10.05) and CAF19 cells after transfection with SC or APE1/Ref-1 siRNA and hypoxia (24 hours, 1% oxygen, representative blots of n ¼ 3).� , P < 0.001 (Tukeymultiple comparisons test); �� , P < 0.01; #, P < 0.001 (ANCOVA); NS, not significant. For CA9Western blots (H and I), P <0.05 for SC versus siAPEunder hypoxia (Tukey multiple comparisons test).

Logsdon et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2726

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 6: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

3D coculture system for preclinical studies validating noveltargets like CA9 and APE1/Ref-1 in PDAC.

Importantly, we evaluated CA9 and APE1/Ref-1 protein expres-sion following exposure to hypoxia (0.2%oxygen) in three PDACcell lines and found that, while CA9 levels increased over time,APE1/Ref-1 levels did not change significantly (SupplementaryFig. S6), indicating that hypoxia-induced CA9 expression is notsecondary to APE1/Ref-1 upregulation.

Dual targeting of CA9 and APE1/Ref-1 acidifies PDAC cells andinhibits cell viability under hypoxia

CA9 regulates intracellular pH under hypoxic conditions, andAPE1/Ref-1 redox activity contributes to hypoxia-induced CA9expression. We analyzed intracellular pH in hypoxia-exposedPDAC cells after treatment with CA9 inhibitor, SLC-0111, andthe APE1/Ref-1 redox inhibitor, APX3330, using the pHrodo RedAMfluorescent pH indicator as a functional endpoint for carbonicanhydrase activity under hypoxic conditions. The analysis ofintracellular pH was performed at an early timepoint (48-hourexposure to inhibitors and hypoxia) to avoid drastic changes incell survival. Still, some viability changes were observed (Supple-

mentary Fig. S7), so these changes were taken into account in theanalysis and normalization of the intracellular pH data. Dualtreatment with SLC-0111 and APX3330 results in a greaterdecrease in intracellular pH than treatment with either inhibitoralone (Fig. 4A and B).

Inhibition of APE1/Ref-1 redox activity results in a dose-depen-dent decrease in PDAC cell viability following treatment of cellswith APX3330 and hypoxia. Remarkably, the effect of APE1/Ref-1inhibition on cell viability is greatly enhanced by treating with theCA9 inhibitor, SLC-0111, in addition to APX3330 treatmentunder hypoxia (Fig. 4C). In support of these results, new CA9inhibitors are being developed and the combination of APX3330with SLC-0111 analogue, FC13-555A, is also significantly effec-tive at killing PDAC cells in monolayer (Fig. 4D), supporting thehypothesis that blockade of hypoxia signaling proteins will bedeleterious to PDAC cells.

Dual targeting of CA9 and APE1/Ref-1 inhibits PDAC tumorgrowth in a 3D coculture model

To more accurately mimic the tumor microenvironment, weutilized a 3D coculture model of PDAC that included the low

Figure 3.

APE1/Ref-1 redox signaling affectsCA9 transcription in a HIF-1-dependent manner. A, HIF-1–proficient (þ/þ) and HIF-1-deficient(�/�) mouse embryonic fibroblasts(MEF) were exposed to 0.2% oxygenfor 24 hours, and CA9 mRNA levelswere evaluated by qPCR,representative experiment of n ¼ 3.B, HIF-1�/� MEFs were transfectedwith SC or APE1/Ref-1–directed siRNAand incubated at 0.2% oxygen for 24hours and CA9 mRNA levels wereevaluated via qPCR, representativeexperiment of n ¼ 3. C and D, CA9mRNA levels were evaluated via qPCRfollowing APX3330 treatment andhypoxia (24 hours, 1% oxygen;representative graph of n ¼ 3).E, Pa03C cells were collected frommonolayer (2D) cultures and 3D tumorspheroid cultures grown in thepresence or absence ofCAFs followingtreatment with APX3330, and CA9protein levels were evaluated viaWestern blot analysis (representativeblot of n ¼ 2). �� , P < 0.001 (Tukeymultiple comparisons test).

Dual-Targeting APE1/Ref-1 and CA9 in Hypoxic PDAC Cells

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2727

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 7: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

passage patient-derived tumor cells as well as cancer-associatedfibroblasts. As demonstrated above, the levels of CA9were greaterin these tumor spheroids when grown with CAF cells, and CA9expression was attenuated by treatment with APX3330 (Fig. 3E).Inhibition of CA9 with SLC-0111 was more potent in the 3Dmodel with dramatic effects on tumor cell killing observed atlower doses than inmonolayer, asmeasured by reductions in areaof patient-derived cells (Fig. 5A and B). Cell killing was moredramatic in the tumor cells than in the CAFs, especially whenCAF19 cells were in coculture with Pa03C cells. Although theseCAFs have aberrantly activated signaling pathways, they arenontumorigenic and suggest that tumor cells are more greatlyaffected by the dual targeting approach as compared with normalcells. Similar trends were seen when measuring fluorescenceintensity (data not shown). Importantly, inhibition of CA9 caneffectively kill tumor cells even when in the protective environ-ment of the CAFs.

To determine whether the blockade of STAT3 and HIF-medi-ated transcription alongside the inhibition of CA9 activity wouldpotentiate PDAC cell death, we combined APX3330 and SLC-0111 in the 3D coculture model. We can assess the effects of dualtargeting on both the tumor alone and the tumor and CAFs incoculture due to the different fluorescent labels in each cell type.

As seen in hypoxia-exposed 2D cultures, addition of CA9 inhi-bition to APE1/Ref-1 redox inhibition resulted in dramatic poten-tiation of the cell killing in the tumor spheroids. Spheroidscomposed of patient-derived PDAC cells (Pa03C or Panc10.05,labeled red) and CAF cells (labeled green) were treated withAPX3330 and SLC-0111 (Fig. 5E and F), and the graphicalrepresentation, that is, area of red and green fluorescence wereevaluated separately as markers for each cell type and is shownin Fig. 5C andD.Dramatic enhancement of theAPX3330-inducedblockade of spheroid growth was observed with the addition ofCA9 inhibition. The observed decrease in tumor cell area withAPX3330 treatment was significantly different in the presence ofSLC-0111, validating the effects seen in hypoxia-exposed 2Dcultures. Similar trends were seen when measuring red and greenfluorescence intensity (data not shown).

DiscussionElevated APE1/Ref-1 expression is associated with numerous

cancers, including pancreatic, ovarian, gastric, breast, lung, glio-blastoma, liver, and colon (18, 19, 46), and analysis of publiclyavailable data from The Cancer Genome Atlas (TCGA, cancer-genome.nih.gov) reveals a significant decrease in survival of

Figure 4.

Dual targeting of CA9 and APE1/Ref-1 acidifies PDAC cells and inhibits cell viability under hypoxia. A, Panc10.05 cells were treated with APX3330 and SLC-0111and exposed to hypoxia (0.2% O2) for 48 hours prior to analysis of intracellular pH (Avg � SE, n ¼ 3). Representative images from pH experiments areshown in B. C and D, viability assay of Pa02C cells treated with the indicated concentrations of APX3330 and CA9 inhibitors, SLC-0111, or FC13-555A andexposed to hypoxia (0.2%) for 6 days (avg � SE, n ¼ 6). Fold change refers to the comparison of each data point to the fluorescence of untreated tumor cells.� , P < 0.05; �� , P < 0.01 (Dunnett multiple comparisons test); #, P < 0.05; ##, P < 0.01 (Sidak multiple comparisons test). In addition, differences in nonlinearregression curves between treatment groups were confirmed using extra-sum-of-squares F tests followed by Bonferroni corrections in each experiment(P < 0.05 for all dual treatment curves vs. single-agent curves).

Logsdon et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2728

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 8: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

PDAC patients with elevated APE1/Ref-1 expression (Supplemen-tary Fig. S8A, more information in Supplementary Methods;refs. 47, 48). In tumor cells, reduction–oxidation (redox) of thiolsof cysteines in various tumor-promoting transcription factorssuch as STAT3, NFkB, and HIF-1 by APE1/Ref-1 is a crucial stepin the activation of these factors. These TFs are all importanttargets in cancer therapy and particularly PDAC, but have beenshown to be particularly hard to drug (18, 20, 21, 46, 49, 50).While previous studies by us and others (23, 51, 52) havedemonstrated the effect of APE1/Ref-1 knockdown on tumor cell

growth and survival, additional studies have demonstrated theeffectiveness of a small molecule, APX3330, in targeting andblocking the redox signaling activity of APE1/Ref-1. APX3330has been shown in multiple in vitro and in vivo models ofpancreatic cancer to be effective in reducing tumor volume andmetastases as both a single agent and in combination withgemcitabine (20, 21, 53). The mechanism of action has beenextensively investigated and the drug has a direct and selectiveinteraction with APE1/Ref-1 as demonstrated by chemical foot-printing, mass spectrometry, and other biochemical analyses

Figure 5.

Dual targeting of CA9 and APE1/Ref-1 inhibits PDAC tumor growth in a 3D coculture model. Pa03C (A, C, and E) and Panc10.05 (B, D, and F) tumor cells weregrown in 3D cultures in the presence and absence of CAFs. Spheroids were treated with SLC-0111 alone (A and B) and in combination with APX3330(C andD), and the area of tumor (red) andCAF (green)were quantified following 12 days in culture, n¼ 3–4. Representative images fromdual treatment experimentsare shown in E and F. Differences in nonlinear regression curves between treatment groups were confirmed using extra-sum-of-squares F tests followed byBonferroni corrections in dual-treatment experiments (P < 0.01 for each curve versus the curve for APX3330 alone in tumor cells alone; P < 0.01 for curveswith 50 mmol/L SLC-0111 versus the curve for APX3330 alone in tumor þ CAF cocultures).

Dual-Targeting APE1/Ref-1 and CA9 in Hypoxic PDAC Cells

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2729

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 9: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

(33, 44, 54). Importantly, while APX3330 blocks APE1/Ref-1'sredox function, it has no effect on APE1/Ref-1 endonuclease DNArepair activity (45). Although multiple pathways may be modu-lated, unacceptable toxicity following APE1/Ref-1 inhibition hasnot been observed in animal or human studies (21, 55). APX3330is slated for a phase Ia/Ib clinical trial of a dose-escalation study ofAPX3330 in patients with advanced solid tumors and a dose-expansion cohort of patientswith advanced PDAC inmid-2016 inPDAC patients. Therefore, targeting APE1/Ref-1 with APX3330has a great deal of potential in cancer therapy.

We previously demonstrated that APE1/Ref-1 contributes toSTAT3 activation and the consequent tumor-promoting effects ofSTAT3 in PDAC cells (20). The cooperative activities of STAT3 andHIF-1 have been demonstrated in a variety of cancers (56, 57);however, the finding that APE1/Ref-1 binding to STAT3 is stim-ulated by exposure to hypoxia in PDAC cells, presented here forthe first time, further indicates the importance of both APE1/Ref-1and STAT3 as potential therapeutic targets in PDAC. These find-ings will be further pursued with preclinical STAT3 inhibitors thatare being developed for eventual clinical trials (42). Furthermore,our results demonstrating that APX3330 treatment decreaseshypoxia-induced HIF-1 transcriptional activity and CA9 mRNAlevels (21) is exciting as CA9 inhibitors are either entering or are inclinical trials. This latter finding is of great interest, not onlybecause it is closer to patient applicability, but builds upon ourstrategy of blocking various signaling pathways atmultiple pointsalong pathways influenced by APE1/Ref-1.

CA9 is one of only two tumor-associated carbonic anhydraseisoenzymes known, and it has similarly been established as apotential therapeutic cancer target (25, 26, 30). CA9 is notdetected in most normal tissues, but its expression in renal andother cancers often indicates locally advanced, hypoxic tumorsand poor treatment response (58, 59). Variations in CA9 expres-sion by region in tumor samples make whole-tissue analysisdifficult (60, 61), but analysis of publicly available data fromOncomine (oncomine.org) using microdissected samples reveals

upregulation of CA9 in PDAC tissue samples, as compared withnormal pancreas and pancreatic cancer precursor samples (Sup-plementary Fig. S8B–S8D; refs. 62, 63). Furthermore, the CA9inhibitor SLC-0111 is in clinical trials to evaluate its safety andefficacy in patients with advanced solid tumors, including pan-creatic cancer (clinicaltrials.gov ID: NCT02215850). Therefore,our strategy of inhibiting theHIF-CA9 axis at two points; blockingHIF-1 production of CA9 with APX3330 as well as blocking theactivity of any CA9 that is produced using SLC-0111 (Fig. 6), is anovel approach to the targeting of hypoxic PDAC cells. However,sensitivity to the combination of APX3330 and SLC-0111will notbe limited to hypoxic cells. APX3330 is targeting other signalingpathways that are activated in tumor cells that are fully oxygen-ated, and SLC-0111 can also inhibit CA12, another tumor-asso-ciated carbonic anhydrase (31). Our findings establish this strat-egy resulting in additive cell acidification and inhibition ofhypoxic PDAC cell proliferation. This combination approach issimilar to the results we previously published using APX3330 andSTAT3 inhibition as a dual hit strategy in PDAC cells (20) andAPX3330 (HIF-1 inhibition) andavastin (bevacizumab) for VEGFsignaling inhibition as an antiangiogenesis combination strategy(45, 64, 65).

As discussed, APX3330 will soon be entering clinical trials forgeneral solid tumors as well as pancreatic cancer. As SLC-0111 isalready in clinical trials with other CA9 inhibitors queued up toenter the clinic, our data demonstrating a novel two-prongedapproach targeting highly hypoxic pancreatic cancer could beclinically applicable in the foreseeable future. In addition, givendata demonstrating elevated expression of APE1/Ref-1, STAT3,and CA9 in a variety of other solid tumors (59, 66), thesecombination approaches should have applicability beyond pan-creatic cancer (30, 32, 59).

In conclusion, the data presented here provide continuedevidence of the close relationship between APE1/Ref-1, STAT3,and HIF-1 signaling and CA9 production in PDAC as well as thefirst evidence that the combination of two small-molecule

Figure 6.

Schematic demonstrating the effectsof APE1/Ref-1-CA9 dual targeting oncellular acidification and downstreamsignaling. Inhibition of APE1/Ref-1redox signaling with APX3330 resultsin decreased DNA binding oftranscription factors including HIF-1(as well as STAT3 and NFkB). HIF-1transactivation is stimulated underlow oxygen conditions, resulting intranscription of tumor-promotingfactors, including CA9. CA9 catalyzesthe conversion of carbon dioxide andwater to bicarbonate and hydrogenions, resulting in stabilization ofintracellular pH, which promotes cellsurvival under hypoxic conditions.Inhibition of CA9 activity with SLC-0111promotes cell killing via acidification,and this inhibition is enhanced byAPX3330 due to a decrease in CA9expression as well as inhibition ofother key signaling pathways viaSTAT3, NFkB, and AP-1.

Logsdon et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2730

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 10: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

inhibitors, each showing minimal toxicity, may be an importantnext step in the treatment of PDAC, a disease for which effectivetreatment remains elusive.

Disclosure of Potential Conflicts of InterestM.R. Kelley reports receiving a commercial research grant, from has owner-

ship interest (including patents) in, and is a consultant/advisory boardmemberfor ApeX Therapeutics. No potential conflicts of interest were disclosed by theother authors.

Authors' ContributionsConception and design: D.P. Logsdon, C.T. Supuran, M. Ivan, M.R. Kelley,M.L. FishelDevelopment of methodology: D.P. Logsdon, M. Luo, Y. Jiang, E. Schipani,M. Korc, M. Ivan, M.R. KelleyAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): D.P. Logsdon, M. Grimard, M.R. Kelley, M.L. FishelAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): D.P. Logsdon, M. Grimard, Y. Tong, Z. Yu, M. Ivan,M.R. Kelley, M.L. FishelWriting, review, and/or revision of the manuscript: D.P. Logsdon, S. Shahda,Z. Yu, N. Zyromski, C.T. Supuran, M. Korc, M. Ivan, M.R. Kelley, M.L. FishelAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): D.P. Logsdon, M.R. Kelley, M.L. FishelStudy supervision: Z. Yu, M.R. Kelley, M.L. Fishel

Other (provided consultation on clinical relevance to current standardpractice): S. ShahdaOther (synthesis of compounds): F. Carta

AcknowledgmentsThe authors thank Dr. Bryan Schneider, Department of Hematology/Oncol-

ogy for use of laboratory equipment and Dr. Malgorzata Kamocka for help withcell imaging. We also thank Christopher Below from Faculty 6, Natural Sciencesat Brandenburg University of Technology for help with figure designing.

Grant SupportThis work was financially supported by the NCI CA122298 (to M.L. Fishel),

CA167291 (to M.L. Fishel, M. Korc, M. Ivan, and M.R. Kelley), NCI CA075059(to M. Korc), and the NIH R21NS091667 (to M.R. Kelley). Additional financialsupport was provided by RalphW. andGraceM. Showalter Research Trust Fund(to M.L. Fishel), the Earl and Betty Herr Professor in Pediatric OncologyResearch, Hyundai Hope on Wheels, Jeff Gordon Children's Foundation, andthe Riley Children's Foundation (to M.R. Kelley).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received April 26, 2016; revised August 2, 2016; accepted August 3, 2016;published OnlineFirst August 17, 2016.

References1. Sclafani F, Iyer R, Cunningham D, Starling N. Management of metastatic

pancreatic cancer: current treatment options and potential new therapeutictargets. Crit Rev Oncol Hematol 2015;95:318–36.

2. McIntyre CA, Winter JM. Diagnostic evaluation and staging of pancreaticductal adenocarcinoma. Semin Oncol 2015;42:19–27.

3. Thota R, Pauff JM, Berlin JD. Treatment of metastatic pancreatic adeno-carcinoma: a review. Oncology 2014;28:70–4.

4. Tamburrino A, Piro G, Carbone C, Tortora G, Melisi D. Mechanisms ofresistance to chemotherapeutic and anti-angiogenic drugs as novel targetsfor pancreatic cancer therapy. Front Pharmacol 2013;4:56.

5. McCarroll JA, Naim S, Sharbeen G, Russia N, Lee J, Kavallaris M, et al. Roleof pancreatic stellate cells in chemoresistance in pancreatic cancer. FrontPhysiol 2014;5:141.

6. Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA.The pancreas cancer microenvironment. Clin Cancer Res 2012;18:4266–76.

7. Lo A, Wang LC, Scholler J, Monslow J, Avery D, Newick K, et al. Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromalcells. Cancer Res 2015;75:2800–10.

8. RhimAD,Oberstein PE, ThomasDH,Mirek ET, PalermoCF, Sastra SA, et al.Stromal elements act to restrain, rather than support, pancreatic ductaladenocarcinoma. Cancer Cell 2014;25:735–47.

9. Xu Z, Pothula SP, Wilson JS, Apte MV. Pancreatic cancer and its stroma: aconspiracy theory. World J Gastroenterol 2014;20:11216–29.

10. Bijlsma MF, van Laarhoven HW. The conflicting roles of tumor stroma inpancreatic cancer and their contribution to the failure of clinical trials: asystematic review and critical appraisal. Cancer Metastasis Rev 2015;34:97–114.

11. Arora S, Bhardwaj A, Singh S, Srivastava SK, McClellan S, Nirodi CS,et al. An undesired effect of chemotherapy: gemcitabine promotespancreatic cancer cell invasiveness through reactive oxygen species-dependent, nuclear factor kappaB- and hypoxia-inducible factor1alpha-mediated up-regulation of CXCR4. J Biol Chem 2013;288:21197–207.

12. Blum R, Kloog Y. Metabolism addiction in pancreatic cancer. Cell DeathDis 2014;5:e1065.

13. Zhao T, Ren H, Li J, Chen J, Zhang H, Xin W, et al. LASP1 is a HIF1alphatarget gene critical for metastasis of pancreatic cancer. Cancer Res2015;75:111–9.

14. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al.Ubiquitination of hypoxia-inducible factor requires direct binding to the

beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2000;2:423–7.

15. Masoud GN, Li W. HIF-1 pathway: role, regulation and intervention forcancer therapy. Acta Pharm Sin B 2015;5:378–89.

16. Kim SY, Yang EG. Recent advances in developing inhibitors for hypoxia-inducible factor prolyl hydroxylases and their therapeutic implications.Molecule 2015;20:20551–68.

17. SharbeenG,McCarroll J, Goldstein D, Phillips PA. Exploiting base excisionrepair to improve therapeutic approaches for pancreatic cancer. Front Nutr2015;2:10.

18. FishelML, KelleyMR. TheDNAbase excision repair proteinApe1/Ref-1 as atherapeutic and chemopreventive target. Mol Aspects Med 2007;28:375–95.

19. Kelley MR, Logsdon D, Fishel ML. Targeting DNA repair pathways forcancer treatment: what's new? Future Oncol 2014;10:1215–37.

20. Cardoso AA, Jiang Y, Luo M, Reed AM, He Y, Kelley MR, et al. APE1/Ref-1redox function regulates STAT3 transcriptional activity and APE1/Ref-1-STAT3 dual-targeting synergize to effectively inhibit pancreatic cancer cellsurvival. PLoS One 2012;10:e47462

21. Fishel ML, Jiang Y, Rajeshkumar NV, Scandura G, Sinn AL, He Y, et al.Impact of APE1/Ref-1 redox inhibition on pancreatic tumor growth. MolCancer Ther 2011;10:1698–708.

22. Fishel ML, Wu X, Devlin CM, Logsdon DP, Jiang Y, Luo M, et al.Apurinic/apyrimidinic endonuclease/redox factor-1 (APE1/Ref-1)redox function negatively regulates NRF2. J Biol Chem 2015;290:3057–68.

23. Jiang Y, Zhou S, Sandusky GE, KelleyMR, FishelML. Reduced expression ofDNA repair and redox signaling protein APE1/Ref-1 impairs humanpancreatic cancer cell survival, proliferation, and cell cycle progression.Cancer Invest 2010;28:885–95.

24. Poletto M, Malfatti MC, Dorjsuren D, Scognamiglio PL, Marasco D,Vascotto C, et al. Inhibitors of the apurinic/apyrimidinic endonuclease1 (APE1)/nucleophosmin (NPM1) interaction that display anti-tumorproperties. Mol Carcinog 2016;55:688–704.

25. Sedlakova O, Svastova E, Takacova M, Kopacek J, Pastorek J, Pastor-ekova S. Carbonic anhydrase IX, a hypoxia-induced catalytic compo-nent of the pH regulating machinery in tumors. Front Physiol2014;4:400.

26. McIntyre A, Patiar S, Wigfield S, Li JL, Ledaki I, Turley H, et al. Carbonicanhydrase IX promotes tumor growth and necrosis in vivo and inhibitionenhances anti-VEGF therapy. Clin Cancer Res 2012;18:3100–11.

Dual-Targeting APE1/Ref-1 and CA9 in Hypoxic PDAC Cells

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2731

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 11: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

27. Kaluz S, Kaluzova M, Liao SY, Lerman M, Stanbridge EJ. Transcriptionalcontrol of the tumor- and hypoxia-marker carbonic anhydrase 9: a onetranscription factor (HIF-1) show? BiochimBiophysActa 2009;1795:162–72.

28. Swietach P, Wigfield S, Cobden P, Supuran CT, Harris AL, Vaughan-JonesRD. Tumor-associated carbonic anhydrase 9 spatially coordinates intra-cellular pH in three-dimensional multicellular growths. J Biol Chem2008;283:20473–83.

29. Schoppmann SF, Jesch B, Friedrich J, Jomrich G, Maroske F, Birner P.Phosphorylation of signal transducer and activator of transcription 3(STAT3) correlates with Her-2 status, carbonic anhydrase 9 expressionand prognosis in esophageal cancer. Clin Exp Metastasis 2012;29:615–24.

30. Supuran CT, Winum JY. Designing carbonic anhydrase inhibitors for thetreatment of breast cancer. Expert Opin Drug Discov 2015;10:591–7.

31. Supuran CT. Howmany carbonic anhydrase inhibitionmechanisms exist?J Enzyme Inhib Med Chem 2016;31:345–60.

32. Fishel ML, Colvin ES, Luo M, Kelley MR, Robertson KA. Inhibition of theredox function of APE1/Ref-1 in myeloid leukemia cell lines results in ahypersensitive response to retinoic acid-induced differentiation and apo-ptosis. Exp Hematol 2010;38:1178–88.

33. SuDG,Delaplane S, LuoM, Rempel DL, Vu B, KelleyMR, et al. Interactionsof APE1 with a redox inhibitor: evidence for an alternate conformation ofthe enzyme. Biochemistry 2011;50:82–92.

34. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Coresignaling pathways in human pancreatic cancers revealed by global geno-mic analyses. Science 2008;321:1801–6.

35. Walter K, Omura N, Hong SM, Griffith M, Goggins M. Pancreatic cancerassociated fibroblasts display normal allelotypes. Cancer Biol Ther2008;7:882–8.

36. Kim HS, Guo C, Thompson EL, Jiang Y, Kelley MR, Vasko MR, et al. APE1,the DNA base excision repair protein, regulates the removal of platinumadducts in sensory neuronal cultures byNER.Mutat Res 2015;779:96–104.

37. Jiang Y, Guo C, Fishel ML,Wang ZY, VaskoMR, Kelley MR. Role of APE1 indifferentiated neuroblastoma SH-SY5Y cells in response to oxidative stress:use of APE1 small molecule inhibitors to delineate APE1 functions. DNARepair (Amst) 2009;8:1273–82.

38. Livak KJ, Schmittgen TD. Analysis of relative gene expression data usingreal-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods(San Diego, Calif) 2001;25:402–8.

39. Yuan JS, Reed A, Chen F, Stewart CN Jr. Statistical analysis of real-time PCRdata. BMC Bioinformatics 2006;7:85.

40. Rankin EB, Wu C, Khatri R, Wilson TL, Andersen R, Araldi E, et al. The HIFsignaling pathway in osteoblasts directly modulates erythropoiesisthrough the production of EPO. Cell 2012;149:63–74.

41. Sempere LF, Gunn JR, Korc M. A novel 3-dimensional culture systemuncovers growth stimulatory actions by TGFbeta in pancreatic cancer cells.Cancer Biol Ther 2011;12:198–207.

42. Arpin CC, Mac S, Jiang Y, Cheng H, Grimard M, Page BD, et al. Applyingsmall molecule signal transducer and activator of trancription-3 (STAT3)protein inhibitors as pancreatic cancer therapeutics. Mol Cancer Ther2016;15:794–805.

43. LindblomP, Berg AL, ZhangH,Westerberg R, Tugwood J, LundgrenH, et al.Tesaglitazar, a dual PPAR-alpha/gamma agonist, hamster carcinogenicity,investigative animal and clinical studies. Toxicol Pathol 2012;40:18–32.

44. Zhang J LM, Marasco D, Logsdon D, LaFavers KA, Chen Q, Reed A, et al.Inhibition of apurinic/apyrimidinic endonuclease I's redox activity revis-ited. Biochemistry 2013;52:2955–66.

45. Luo M, Delaplane S, Jiang A, Reed A, He Y, Fishel M, et al. Role of themultifunctional DNA repair and redox signaling protein Ape1/Ref-1 incancer and endothelial cells: small-molecule inhibition of the redoxfunction of Ape1. Antioxid Redox Signal 2008;10:1853–67.

46. Kelley MR, Georgiadis MM, Fishel ML. APE1/Ref-1 role in redox signaling:translational applications of targeting the redox function of the DNArepair/redox protein APE1/Ref-1. Curr Mol Pharmacol 2012;5:36–53.

47. Cerami E,Gao J,DogrusozU,Gross BE, Sumer SO, Aksoy BA, et al. The cBiocancer genomics portal: an open platform for exploring multidimensionalcancer genomics data. Cancer Discov 2012;2:401–4.

48. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al.Integrative analysis of complex cancer genomics and clinical profiles usingthe cBioPortal. Sci Signal 2013;6:pl1.

49. Dibra D, Mishra L, Li S. Molecular mechanisms of oncogene-inducedinflammation and inflammation-sustained oncogene activation in gastro-intestinal tumors: an underappreciated symbiotic relationship. BiochimBiophys Acta 2014;1846:152–60.

50. Prabhu L, Mundade R, Korc M, Loehrer PJ, Lu T. Critical role of NF-kappaBin pancreatic cancer. Oncotarget 2014;5:10969–75.

51. Wang D, Luo M, Kelley MR. Human apurinic endonuclease 1 (APE1)expression and prognostic significance in osteosarcoma: enhanced sensi-tivity of osteosarcoma to DNA damaging agents using silencing RNA APE1expression inhibition. Mol Cancer Ther 2004;3:679–86.

52. Cun Y,DaiN, XiongC, LiM, Sui J, QianC, et al. Silencing of APE1 enhancessensitivity of human hepatocellular carcinoma cells to radiotherapy in vitroand in a xenograft model. PLoS One 2013;8:e55313.

53. Zou GM, Maitra A. Small-molecule inhibitor of the AP endonuclease1/REF-1 E3330 inhibits pancreatic cancer cell growth and migration.Mol Cancer Ther 2008;7:2012–21.

54. Luo M, Zhang J, He H, Su D, Chen Q, Gross ML, et al. Characterization ofthe redox activity and disulfide bond formation in apurinic/apyrimidinicendonuclease. Biochemistry 2012;51:695–705.

55. Vasko MR, Guo C, Thompson EL, Kelley MR. The repair function of themultifunctional DNA repair/redox protein APE1 is neuroprotective afterionizing radiation. DNA Repair 2011;10:942–52.

56. Pawlus MR, Wang L, Hu CJ. STAT3 and HIF1alpha cooperatively activateHIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene 2014;33:1670–9.

57. Gariboldi MB, Ravizza R, Monti E. The IGFR1 inhibitor NVP-AEW541disrupts a pro-survival and pro-angiogenic IGF-STAT3-HIF1 pathway inhuman glioblastoma cells. Biochem Pharmacol 2010;80:455–62.

58. McDonald PC, Winum JY, Supuran CT, Dedhar S. Recent developments intargeting carbonic anhydrase IX for cancer therapeutics. Oncotarget2012;3:84–97.

59. Oosterwijk E. Carbonic anhydrase expression in kidney and renal cancer:implications for diagnosis and treatment. Subcell Biochem 2014;75:181–98.

60. Juhasz M, Chen J, Lendeckel U, Kellner U, Kasper HU, Tulassay Z, et al.Expression of carbonic anhydrase IX in human pancreatic cancer. AlimentPharmacol Ther 2003;18:837–46.

61. Kivela AJ, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, et al.Expression of transmembrane carbonic anhydrase isoenzymes IX and XIIin normal human pancreas and pancreatic tumours. Histochem Cell Biol2000;114:197–204.

62. Logsdon CD, Simeone DM, Binkley C, Arumugam T, Greenson JK, Gior-dano TJ, et al. Molecular profiling of pancreatic adenocarcinoma andchronic pancreatitis identifies multiple genes differentially regulated inpancreatic cancer. Cancer Res 2003;63:2649–57.

63. PeiH, Li L, Fridley BL, Jenkins GD, Kalari KR, LingleW, et al. FKBP51 affectscancer cell response to chemotherapy by negatively regulating Akt. CancerCell 2009;16:259–66.

64. Li Y, Liu X, Zhou T, KelleyMR, Edwards P, GaoH, et al. Inhibition of APE1/Ref-1 redox activity rescues human retinal pigment epithelial cells fromoxidative stress and reduces choroidal neovascularization. Redox Biol2014;2:485–94.

65. Jiang A, Gao H, Kelley MR, Qiao X. Inhibition of APE1/Ref-1 redox activitywith APX3330 blocks retinal angiogenesis in vitro and in vivo. Vision Res2011;51:93–100.

66. Furtek SL, Backos DS, Matheson CJ, Reigan P. Strategies and approaches oftargeting STAT3 for cancer treatment. ACS Chem Biol 2016;11:308–18.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2732

Logsdon et al.

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253

Page 12: Regulation of HIF1a under Hypoxia by APE1/Ref-1 …mct.aacrjournals.org/content/molcanther/15/11/2722.full.pdfMolecular Cancer Therapeutics 2722 Mol Cancer Ther; 15(11) November 2016

2016;15:2722-2732. Published OnlineFirst August 17, 2016.Mol Cancer Ther   Derek P. Logsdon, Michelle Grimard, Meihua Luo, et al.   Cancer ModelsExpression: Dual Targeting in Patient-Derived 3D Pancreatic

under Hypoxia by APE1/Ref-1 Impacts CA9αRegulation of HIF1

  Updated version

  10.1158/1535-7163.MCT-16-0253doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2016/08/17/1535-7163.MCT-16-0253.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/15/11/2722.full#ref-list-1

This article cites 66 articles, 15 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/15/11/2722.full#related-urls

This article has been cited by 1 HighWire-hosted articles. Access the articles 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/15/11/2722To request permission to re-use all or part of this article, use this link

on July 10, 2018. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 17, 2016; DOI: 10.1158/1535-7163.MCT-16-0253