13
Therapeutics, Targets, and Chemical Biology MET-Independent Lung Cancer Cells Evading EGFR Kinase Inhibitors Are Therapeutically Susceptible to BH3 Mimetic Agents Weiwen Fan 1 , Zhe Tang 2 , Lihong Yin 1 , Bei Morrison 1 , Said Hafez-Khayyata 3 , Pingfu Fu 4,8 , Honglian Huang 6 , Rakesh Bagai 2 , Shan Jiang 2 , Adam Kresak 8 , Scott Howell 9 , Amit Vasanji 7 , Chris A. Flask 8,10 , Balazs Halmos 11 , Henry Koon 2,8 , and Patrick C. Ma 1,5,8 Abstract Targeted therapies for cancer are inherently limited by the inevitable recurrence of resistant disease after initial responses. To define early molecular changes within residual tumor cells that persist after treatment, we analyzed drug-sensitive lung adenocarcinoma cell lines exposed to reversible or irreversible epidermal growth factor receptor (EGFR) inhibitors, alone or in combination with MET-kinase inhibitors, to characterize the adaptive response that engenders drug resistance. Tumor cells displaying early resistance exhibited dependence on MET-independent activation of BCL-2/BCL-XL survival signaling. Further, such cells displayed a quiescence- like state associated with greatly retarded cell proliferation and cytoskeletal functions that were readily reversed after withdrawal of targeted inhibitors. Findings were validated in a xenograft model, showing BCL-2 induction and p-STAT3[Y705] activation within the residual tumor cells surviving the initial antitumor response to targeted therapies. Disrupting the mitochondrial BCL-2/BCL-XL antiapoptotic machinery in early survivor cells using BCL-2 Homology Domain 3 (BH3) mimetic agents such as ABT-737, or by dual RNAi-mediated knockdown of BCL-2/BCL-XL, was sufficient to eradicate the early-resistant lung-tumor-cells evading targeted inhibitors. Similarly, in a xenograft model the preemptive cotreatment of lung tumor cells with an EGFR inhibitor and a BH3 mimetic eradicated early TKI-resistant evaders and ultimately achieved a more durable response with prolonged remission. Our findings prompt prospective clinical investigations using BH3-mimetics combined with targeted receptor kinase inhibitors to optimize and improve clinical outcomes in lung-cancer treatment. Cancer Res; 71(13); 4494505. Ó2011 AACR. Introduction Lung cancer is the second most common cancer and continues to have the highest cancer-mortality rates. Receptor tyrosine kinase (RTK) is a main class of druggable molecular targets, such as epidermal growth factor receptor (EGFR; 1, 2), MET (3, 4), which can be therapeutically inhibited in human cancer therapy. EGFR tyrosine kinase inhibitors (TKIs), gefi- tinib and erlotinib, are approved targeted agents against nonsmall cell lung cancer (NSCLC), with enhanced efficacy toward tumors that express somatic sensitizing kinase domain mutations (e.g., L858R, exon 19 deletions; 57). One of the most formidable challenges of targeted therapy is the invariable tumor secondary resistance after initial response. MET genomic amplification has been implicated in about 20% of acquired EGFR TKI resistance (8, 9) whereas the EGFR gatekeeper T790M kinase mutation (1012) accounts for approximately half of the resistant cases. Further targeting strategies to overcome EGFR TKI resistance include the use of irreversible TKIs (10, 13, 14), pan-EGFR/ERBB kinase inhibi- tors (15), and MET inhibitors (8, 16). The MET receptor has been shown to be an important molecule in a variety of malignancies (3, 17) and has recently been validated as an attractive therapeutic target in cancer therapy, including lung cancer (4, 1823). Reversible small molecule inhibitors to target against MET have been developed for novel anticancer therapeutic intervention (20, 21, 2426). Studies from our group and others have recently showed the cross-talk signal- ing network between EGFR and MET, and also the role of MET Authors' Affiliations: 1 Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic; 2 Division of Hematology/Oncology, Department of Medicine, 3 Department of Pathology, and 4 Department of Biostatistics, 5 Cleveland Clinic Lerner College of Medicine, Case Western Reserve University; 6 Department of Immunology, 7 Animal Imaging Core, Lerner Research Institute, Cleveland Clinic; 8 Case Comprehensive Cancer Center; 9 Center of Visual Research, 10 Case Center of Imaging Research, University Hospitals Case Medical Center, Cleveland, Ohio; 11 Division of Hematology/Oncology, Department of Medicine, Columbia University, New York, New York Note: W. Fan and Z. Tang have contributed equally to the work. Current address for Z. Tang: First Affiliated Hospital of Zhengzhou Uni- versity, Zhengzhou, Henan, China 450001. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Patrick C. Ma, Taussig Cancer Institute, Cleve- land Clinic, 9500 Euclid Avenue, R40, Cleveland, OH 44195. Phone: 216- 445-5545; Fax: 216-636-2498; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-10-2668 Ó2011 American Association for Cancer Research. Cancer Research Cancer Res; 71(13) July 1, 2011 4494 Research. on June 27, 2021. © 2011 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

MET-Independent Lung Cancer Cells Evading EGFR Kinase … · Therapeutics, Targets, and Chemical Biology MET-Independent Lung Cancer Cells Evading EGFR Kinase Inhibitors Are Therapeutically

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

  • Therapeutics, Targets, and Chemical Biology

    MET-Independent Lung Cancer Cells Evading EGFR KinaseInhibitors Are Therapeutically Susceptible to BH3 MimeticAgents

    Weiwen Fan1, Zhe Tang2, Lihong Yin1, Bei Morrison1, Said Hafez-Khayyata3, Pingfu Fu4,8,Honglian Huang6, Rakesh Bagai2, Shan Jiang2, Adam Kresak8, Scott Howell9, Amit Vasanji7,Chris A. Flask8,10, Balazs Halmos11, Henry Koon2,8, and Patrick C. Ma1,5,8

    AbstractTargeted therapies for cancer are inherently limited by the inevitable recurrence of resistant disease after

    initial responses. To define early molecular changes within residual tumor cells that persist after treatment, weanalyzed drug-sensitive lung adenocarcinoma cell lines exposed to reversible or irreversible epidermal growthfactor receptor (EGFR) inhibitors, alone or in combination with MET-kinase inhibitors, to characterize theadaptive response that engenders drug resistance. Tumor cells displaying early resistance exhibited dependenceon MET-independent activation of BCL-2/BCL-XL survival signaling. Further, such cells displayed a quiescence-like state associated with greatly retarded cell proliferation and cytoskeletal functions that were readily reversedafter withdrawal of targeted inhibitors. Findings were validated in a xenograft model, showing BCL-2 inductionand p-STAT3[Y705] activation within the residual tumor cells surviving the initial antitumor response totargeted therapies. Disrupting the mitochondrial BCL-2/BCL-XL antiapoptotic machinery in early survivor cellsusing BCL-2 Homology Domain 3 (BH3) mimetic agents such as ABT-737, or by dual RNAi-mediated knockdownof BCL-2/BCL-XL, was sufficient to eradicate the early-resistant lung-tumor-cells evading targeted inhibitors.Similarly, in a xenograft model the preemptive cotreatment of lung tumor cells with an EGFR inhibitor and aBH3 mimetic eradicated early TKI-resistant evaders and ultimately achieved a more durable response withprolonged remission. Our findings prompt prospective clinical investigations using BH3-mimetics combinedwith targeted receptor kinase inhibitors to optimize and improve clinical outcomes in lung-cancer treatment.Cancer Res; 71(13); 4494–505. �2011 AACR.

    Introduction

    Lung cancer is the second most common cancer andcontinues to have the highest cancer-mortality rates. Receptortyrosine kinase (RTK) is a main class of druggable molecular

    targets, such as epidermal growth factor receptor (EGFR; 1, 2),MET (3, 4), which can be therapeutically inhibited in humancancer therapy. EGFR tyrosine kinase inhibitors (TKIs), gefi-tinib and erlotinib, are approved targeted agents againstnonsmall cell lung cancer (NSCLC), with enhanced efficacytoward tumors that express somatic sensitizing kinasedomain mutations (e.g., L858R, exon 19 deletions; 5–7). Oneof the most formidable challenges of targeted therapy is theinvariable tumor secondary resistance after initial response.MET genomic amplification has been implicated in about 20%of acquired EGFR TKI resistance (8, 9) whereas the EGFRgatekeeper T790M kinase mutation (10–12) accounts forapproximately half of the resistant cases. Further targetingstrategies to overcome EGFR TKI resistance include the use ofirreversible TKIs (10, 13, 14), pan-EGFR/ERBB kinase inhibi-tors (15), and MET inhibitors (8, 16). The MET receptor hasbeen shown to be an important molecule in a variety ofmalignancies (3, 17) and has recently been validated as anattractive therapeutic target in cancer therapy, including lungcancer (4, 18–23). Reversible small molecule inhibitors totarget against MET have been developed for novel anticancertherapeutic intervention (20, 21, 24–26). Studies from ourgroup and others have recently showed the cross-talk signal-ing network between EGFR andMET, and also the role of MET

    Authors' Affiliations: 1Department of Translational Hematology andOncology Research, Taussig Cancer Institute, Cleveland Clinic; 2Divisionof Hematology/Oncology, Department of Medicine, 3Department ofPathology, and 4Department of Biostatistics, 5Cleveland Clinic LernerCollege of Medicine, Case Western Reserve University; 6Department ofImmunology, 7Animal Imaging Core, Lerner Research Institute, ClevelandClinic; 8Case Comprehensive Cancer Center; 9Center of Visual Research,10Case Center of Imaging Research, University Hospitals Case MedicalCenter, Cleveland, Ohio; 11Division of Hematology/Oncology, Departmentof Medicine, Columbia University, New York, New York

    Note: W. Fan and Z. Tang have contributed equally to the work.

    Current address for Z. Tang: First Affiliated Hospital of Zhengzhou Uni-versity, Zhengzhou, Henan, China 450001.

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

    Corresponding Author: Patrick C. Ma, Taussig Cancer Institute, Cleve-land Clinic, 9500 Euclid Avenue, R40, Cleveland, OH 44195. Phone: 216-445-5545; Fax: 216-636-2498; E-mail: [email protected]

    doi: 10.1158/0008-5472.CAN-10-2668

    �2011 American Association for Cancer Research.

    CancerResearch

    Cancer Res; 71(13) July 1, 20114494

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • inhibition in combination with EGFR inhibitor in lung cancerin overcoming MET amplified resistance (8) or T790M–EGFRmediated resistance (16) to EGFR–TKI.Further knowledge into additional mechanisms of tumor-

    cell resistance to targeted inhibitors should prove to be of greatsignificance in the quest for novel effective treatment strategiesto impact the long-term prognosis of lung cancer. Majority ofthe reported studies investigating mechanisms of tumor resis-tance centered on late time window after chronic exposure toTKIs at escalating dosing concentrations when secondaryresistant clones ultimately arose and propagated from theparental drug-sensitive cell populations. Nonetheless, a deepunderstanding of the entire spectrum of tumor cells mechan-istic strategies to escape or evade targeted therapeutics inresistance, especially during the early inhibitory phase, remainsto be better defined at present (27, 28). Here, we investigatedthe "early" molecular events in lung tumor cells under targetedEGFR alone or combined with MET-kinase inhibitors treat-ment. Our results identified that a resurgence of prosurvival-antiapoptotic signalingwas evident in the surviving tumorwithearly evasion against the targeted kinase inhibitors, thatinvolved a TKI-induced dependence of activated STAT3, andits transcriptional target BCL-2/BCL-XL, with therapeutictranslational values. Our results show that proapoptoticBCL-2 Homology Domain 3 (BH3)-mimetic, such as ABT-737, can be effective in eradicating these "early" TKI-resistantlung tumor evader cells, thereby potentially enhancing thelong-term efficacy of targeted EGFR lung cancer therapy.

    Materials and Methods

    Cell culture and immunoblottingLung cancer cell lines were obtained directly from American

    Type Culture Collection (ATCC) and grown under standardcell culture conditions. Cell lines characterization and authenti-cation were carried out by the ATCCMolecular AuthenticationCenter, using cytochrome c oxidase subunit I (COI) for inter-species identification and short tandem repeat (STR) anlaysis(DNA fingerprinting) for intraspecies identification. Sodiumdodecylsulfatepolyacrylamidegelelectrophoresis (SDS—PAGE)and Western blotting were carried out as previously described(16, 29). The primary antibodies used are as follows: phospho-MET[Y1234/1235]: Cell Signaling Technology (CST), MET(C-12, Santa Cruz Biotechnology), phospho-EGFR[Y1068]; CST,EGFR (Santa Cruz), phosphotyrosine (p-Tyr), phospho-AKT[S473], AKT, phospho-MAPK(ERK1/2)[T202/Y204]—all fromCST,MAPK(ERK1/2):Biosource,phospho-STAT3[Y705];CST, STAT3, BCL-2, BCL-XL (all fromZymed), cleaved-Caspase-3[Asp175], cleaved-PARP[Asp214], phospho-STAT5[Y694]—allfrom CST, survivin (Zymed) and actin (Santa Cruz).

    Chemicals and inhibitorsEGFR inhibitor (reversible) erlotinib was prepared as pre-

    viously described (29, 30). MET inhibitors SU11274,PHA665752 and EGFR inhibitor (irreversible) CL-387,785 wereobtained from EMD–Calbiochem. BCL-2 family inhibitorsABT-737, obatoclax mesylate (GX15–070), and HA14–1 wereobtained from Selleck.

    Cellular cytotoxicity, viability, and survival assaysCellular cytotoxicity and viability assays were carried out

    using CellTiter 96 AQueous. One Solution Cell Proliferation(MTS) assay (Promega), according to the manufacturer'sinstruction at 72 hours after treatment with indicated inhi-bitors in 10% fetal bovine serum (FBS) media. For the studiesof cells under 9 days of pretreatment under targeted (EGFR/MET) inhibitors, the indicated inhibitors in the culture mediawere replenished at least every 2–3 days (which was verified tobe indistinguishable from daily TKI replacement) prior to cellharvesting at the end of the inhibitory culture for subsequentcellular assays.

    For cell survival assay using crystal violet staining method,H1975 cells or HCC827 cells were treated as described inSupplementary Materials and Methods with indicated TKIsfor 6 days, followed by indicated BH3-mimetic inhibition withor without concurrent TKI for 3 extra days.

    Time-lapsed video microscopy: Image analysis ofcytoskeletal functions

    HCC827 cells were plated on cell-culture dishes in a tem-perature-controlled chamber at 37�C in an atmosphere of 5%CO2 for time-lapsed video microscopy (TLVM) analysis ofcytoskeletal functions and determination of cellular mitoticactivities as previously described (16) and also in Supplemen-tary Materials.

    In vivo xenograft model and bioluminescence imagingof human lung cancer

    Lung cancer xenograft. Firefly-luciferase(luc)-expressingHCC827 and H1975 lung cancer cells, and their correspondingmurine xenograft models were established as previouslydescribed (see also Supplementary Materials and Methods)according to institution-approved protocols and guidelines(16). Immunohistochemical (IHC) analysis of the tumor xeno-graft was carried out in the Tissue Procurement and HistologyCore Facility, Case Comprehensive Cancer Center, using anti-human BCL-2 (Abcam), antihuman p-STAT3[Y705] (rabbitmonoclonal antibody, D3A7, CST) primary antibodies. Fordetails see also Supplementary Materials and Methods.

    Tumor microarrayHuman lung cancer tumor microarray was purchased from

    Zymed-Invitrogen (MaxArray Human Lung Cancer TissueMicroarray Slides, Cat. No. 75–4083). IHC staining usingantihuman BCL-2 antibody was carried out as describedabove, and graded using 4-tier scoring system (0, 1þ, 2þ,and 3þ) by a dedicated thoracic pathologist (S. H.-K.). For thelung cancer tumor microarray (TMA) analysis, the TMAused in the analysis consisted of the followings: SquamousCell (n ¼ 25), Adenocarcinoma (n ¼ 21), Large Cell (n ¼ 3),SCLC (n ¼ 5), Carcinoid (n ¼ 2), Mesothelioma (n ¼ 2).

    BCL-2/BCL-XL DNA transfection and RNA interferencestudies

    Human BCL-2 plasmid vector was a generous gift from Dr.Clark Distelhorst (Case Western Reserve University). Trans-fection of the BCL-2 expression vector into HCC827 cells was

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4495

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • carried out using Fugene 6 according to the manufacturer'sinstructions (Roche). RNA interference (RNAi) knockdownstudies were conducted using the Thermo Scientific/Dharma-con RNAi Technologies, including siGENOME siRNA-NT (non-targeting; Cat.#D-001210–02), siRNAs against human BCL-2(Cat.#L-003307–00), and BCL-XL (Cat.#L-003458–00). ForHCC827 cells (Fig. 6A–B), cells were plated at full confluenceon 48-well plates, then cultured for 9 days in serum-containingmedia (a) without inhibitor, or with treatment of (b) Erlotinibalone for 9 days, or (c–f) Erlotinib together with the followingsin combination: (c) ABT-737 (2 mmol/L) concurrently at Day 0,(d) siRNA-nontargeting (siNT), (e) siRNA-BCL-2, and (f) dualsiRNA-BCL-2/BCL-XL RNAi knockdown. Cells were then fixedin methanol and stained with 0.1% crystal violet as above atthe end of day 9 to visualize the early TKI-resistant tumorsurvivor cells emerged under various conditions. Experimentswere carried out in triplicate.

    Statistical analysisIn the BCL-2 transfection study and erlotinib cellular cyto-

    toxicity assay in the HCC827 cells (Fig. 5D), the results undereach transfection condition were first summarized by the areaunder the curve (AUC). The differences of AUC betweentransfection conditions were then examined by Z-test. Statis-tical data analysis of the in vivo study using HCC827-lucxenograft murine model (Fig. 6E) was carried out using themixed model to examine the difference of read-out[bioluminescence imaging (BLI) region of interest (ROI)]among the four study groups (I–IV), by the in vivo xenograftgrowth rate–changing rate over time [i.e., the change of read-out (BLI ROI) divided by time (day)]. To ensure the normalityassumption for the mixed model used is satisfied, the read-outs were transformed by natural log function, i.e., loge(read-out), prior to fitting the data using Mixed Model. Tumorrecurrence was defined as 20% increase of tumor BLI-fluxfrom the nadir and the difference of recurrence rates betweenGroup II (ABT-737 alone), Group III (Erlotinib alone), andGroup IV (ABT-737 plus Erlotinib) was examined by Fisher'sexact test. All tests were two-sided and P-values � 0.05 wereconsidered statistically significant.

    Results

    Tumor resistance emerged "early" from EGFR-reversible-TKI-sensitive lung adenocarcinoma evadingerlotinib:MET-independent BCL-2/BCL-XL signaling

    The lung adenocarcinoma cell lines HCC827 and PC-9 areboth highly sensitive to reversible-EGFR inhibitors (erlotinib/gefitinib), owing to the oncogenic sensitizing EGFR exon 19deletion (E746_A750 del). Here, we focused to study the "early"molecular alterations in tumor cells under TKI treatment, inan attempt to uncover potential therapeutic "Achilles’ heel"for the tumor cells that may survive the TKI within the earlytime-window.

    We first adopted the HCC827 cell line in the in vitro "early"TKI-resistance studies, with the cells cultured under ongoingerlotinib (1 mmol/L) inhibitory treatment up to 9 days.We chose the concentration of erlotinib to be used at

    approximately IC70–75 in the 72-hrs cell viability assay. Byday 9 of inhibition, there were cell subpopulations ("early"survivors, HCC827_ERL-D9.R) that evaded and survived theTKI treatment. These "early" survivor cells exhibited a dra-matic shift of TKI-sensitivity phenotype toward higherresistance (�100-fold), compared with the TKI-naïve par-ental cells (Fig. 1A). After an initial inhibited state, there wasalso reactivated BCL–2/BCL–XL, within the background of atyrosine-phosphoproteomic reactivated cellular state of aunique profile different from the parental cells (Fig. 1B–C).Importantly, the tumor cells that survived up to days 6–9 ofthe EGFR–TKI treatment evidently signaled independentlyof EGFR and MET (Fig. 1C). Following an initial inhibitoryperiod, we observed a rather early p-STAT3[Y705] reactiva-tion despite ongoing erlotinib treatment. These restoredprosurvival-antiapoptotic markers correlated well withtime-dependent downregulated cleaved-caspase 3 andcleaved-PARP, indicative of a suppressed caspase-dependentintrinsic apoptosis mechanism among these TKI-evadercells. These results were further verified in PC-9 cell line,with the erlotinib-surviving PC-9 cells (PC-9_ERL-D9.R) atDay-9 inhibition exhibiting MET-independent upregulatedp-STAT3[Y705]/BCL-2/BCL-XL signaling, and "early" TKI-resistance (Supplementary Fig. S1).

    EGFR-irreversible-TKI-sensitive lung adenocarcinomaH1975 cells also showed "early" tumor resistant evasionfrom CL-387,785

    We also tested the H1975 cell line against the irreversible-EGFR-TKI CL-387,785 as an alternate model. Similarly, the"early" TKI-surviving H1975 cells (H1975_CL-D9.R) that wereharvested after 9 days of CL-387,785 (1mmol/L) exposure werefound remarkably more resistant (Fig. 2A), accompanied witha prosurvival-antiapoptotic signaling upregulation as early asbeyond day 3 of inhibition (Fig. 2B). Consistent with thefindings in both HCC827 and PC-9 cells, the H1975_CL-D9.R cells also signaled independent of MET. Intriguingly, theH1975_CL-D9.R cells were found to exhibit reactivated p-EGFR[Y1068] beyond day 3 and onward despite ongoingCL-387,785, in the presence of complete p-MET inhibition.Thus, the downstream prosurvival-antiapoptotic signaling inthe survivor cells may still be at least partially driven by EGFRsignaling. We verified that the reactivated EGFR phosphoryla-tion, and its downstream signals emerged in the H1975_CL-D9.R cells, could be inhibited by a higher concentration (5mmol/L) of the same inhibitor (Fig. 2C). Nonetheless, p-STAT3and BCL-2 were not effectively inhibited by the retreatment,highlighting their potential importance in promoting thecellular resistant survival, independent of both EGFR andMET signaling.

    Early EGFR–TKI resistance exhibited "adaptive"phenotypes that were highly "reversible"

    Using TLVM analysis, we found that the HCC827_ERL-D9.R exhibited a cellular "quiescence-like" state, with dramati-cally inhibited proliferative and cytoskeletal functions whilein evasion against erlotinib. Promptly after erlotinib-with-drawal, we found that the early resistant cells could readily

    Fan et al.

    Cancer Res; 71(13) July 1, 2011 Cancer Research4496

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • be reverted to a highly activated state of cellular motility(Fig. 3A) and mitotic proliferation (Fig. 3B; see also Supple-mentary Movies). We further tested to see if the Day-9 "early"resistant cells could maintain their resistant phenotype aftera brief period of TKI withdrawal. Interestingly, after only7 days of withdrawal of the corresponding TKIs, bothHCC827_ERL-D9.R and H1975_CL-D9.R cells quicklyreverted back to a highly TKI-sensitive phenotype, indis-tinguishable from parental-cell populations, respectively(Fig. 3C and E). Importantly, upon a washout period ofTKI-withdrawal, these early resistant escape survivor tumorcells reexhibited TKI-induced p-EGFR inhibition as in theTKI-naïve parental cells (Fig. 3D).

    In vivo activated STAT3/BCL-2 prosurvival-antiapoptotic signal axis in "early" TKI-resistant lungtumor survivor cells

    We extended our studies using in vivo xenograft model toexamine tumor cells that survived initial treatment withtargeted RTK-inhibitors. HCC827 xenograft was inhibited witherlotinib for 3 days, during which remarkable tumor responsewas evident as expected. Consistent with our in vitro data, theSTAT3 downstream transcriptional target BCL-2 expressionwas found induced in the TKI-evading survivor cells (Fig. 4A).Interestingly, these early TKI-resistant cells were localizedalong the peripheral rind of the tumor xenograft (Fig. 4A–B). P-STAT3[Y705] was predominantly membranous and less

    A

    B

    C

    W.B.

    W.B.

    EGFR (Total)P-EGFR

    EGFR-TKl:

    Erlotinib

    P-MET

    P-STAT3

    BCL-2

    BCL-XL

    P-AKT

    P-MAPK

    c-Caspase3

    c-PARP

    P-STAT5

    Survivin

    Cyclin D1

    Actin

    HCC827

    HCC827

    Hours

    WB: anti-p-Tyr

    kDa

    250

    150

    100

    75

    50

    37

    25

    Erlotinib (1 µmol/L)

    6 D

    ays

    9 D

    ays

    72482484210

    Hours

    6 D

    ays

    9 D

    ays

    72482484210

    EGFR-TKl:

    Erlotinib (1 µmol/L)

    Hours

    6 D

    ays

    9 D

    ays

    72482484210

    HCC827HCC827_ERL-D9.R

    HCC827

    Erlotinib Conc. (µmol/L)

    Per

    cent

    cel

    l via

    bilit

    y (M

    TS

    Ass

    ay)

    (%)

    100

    90

    80

    70

    60

    50

    40

    30

    20

    10

    00 0.001 0.01 0.1 1 10

    110

    120

    MET (Total)

    STAT3 (Total)

    AKT (Total)

    MAPK (Total)

    Figure 1. Targeted inhibition of EGFR-TKI-sensitive HCC827 lung adenocarcinoma cells resulted in emergence of "early" tumor TKI-resistant evadercells with MET-independent prosurvival-antiapoptotic signaling. A, significant shift of cell viability toward insensitivity in HCC827 cells that survived 9 days oferlotinib (1mmol/L) treatment. HCC827_ERL-D9.R cells displayed �100-fold increase in IC50 corresponding with a dramatically higher resistant phenotypeagainst the EGFR-TKI. B, activated tyrosine-phosphoproteome of early resistant HCC827 cells (HCC827_ERL-D9.R) evading erlotinib up to 9 days.C, activation of p-STAT3[Y705]/BCL-2/BCL-XL signaling in HCC827_ERL-D9.R cells.

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4497

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • so cytoplasmic in the untreated HCC827 cells. Conversely, theactivated p-STAT3 signal was predominantly nuclear in theresidual HCC827 tumor early survivor cells circumscribingalong the xenograft periphery (Fig. 4B).

    Past studies suggested that non-T790M-EGFR mediatedacquired gefitinib/erlotinib resistance in sensitive-lung can-cer cells may include genomic MET amplification (8, 9) orhepatocyte growth factor (HGF) overexpression (31). Cur-rently, there are various clinical trials investigating thestrategy of combining EGFR- and MET-inhibitors to enhancetherapeutic efficacy and overcome acquired EGFR-TKI resis-tance in NSCLC. We have previously characterized the METinhibitors SU11274 (20) and PHA665752 (21) in lung cancernovel therapy, which were utilized in the present study.Similar to the EGFR-TKI monotherapy model above, therewas an upregulated BCL-2/BCL-XL prosurvival-antiapopto-tic signaling in H1975 TKI-evader cells after 9 days ofdual CL-387,785/PHA665752 inhibition, alongwith restoredp-STAT3 activation (Fig. 4C), and associating with a moreTKI-resistant phenotype in the evader cells (P ¼ 0.0118;Supplementary Fig. S2).

    We recently reported the efficacy of combined SU11274-erlotinib (MET-EGFR/ERBB TKI) in vivo H1975 xenograftmodel in overcoming T790M-EGFR drug-resistance, withresultant near-complete BLI-radiographic complete response(16). Here, we further evaluated the microscopic residualTKI-evading H1975-luc tumor cells and found they residedprimarily along the tumor periphery juxtaposing the murinehost-microenvironment. These TKI-evading survivor tumorcells also exhibited upregulated BCL-2 expression (Fig. 4D).

    BCL-2 signal pathway expression and its potentialtherapeutic utility in NSCLC inhibition by BH3-mimetic

    BCL-2 was found to be expressed at varying levels in NSCLCcell lines and lung tumor tissues, albeit at a significantlylower level range than in SCLC (Fig. 5A–B). Interestingly,unlike BCL-2, the expression levels of BCL-XL appeared tobe more comparable among NSCLC and SCLC cell lines(Fig. 5A). TMA analysis showed that BCL-2 expression inNSCLC was primarily nuclear, whereas that in SCLC wasstrongly positive both nuclear and cytoplasmic. Stronger

    P-EGFR

    P-EGFR

    P-MET

    P-STAT3

    P-STAT3

    BCL-2

    BCL-2

    BCL-XL

    BCL-XL

    P-AKT

    P-AKT

    P-MAPK

    P-MAPK

    c-PARP

    Survivin

    Actin

    Actin

    H1975

    H1975

    + +

    (+)–

    + +

    (+)–

    H1975

    100

    75

    50

    25

    0

    Per

    cent

    cel

    l via

    bilit

    y (M

    TS

    Ass

    ay)

    (%)

    CL-387,785 (µmol/L)

    CL-387,785 (D1–9)

    CL-387,785 (D9)

    0 0.001 0.01 0.1 51 10

    EGFR (Total)

    EGFR-TKl:

    CL-387,785 (1 µmol/L)

    EGFR-TKl:

    CL-387,785

    MET (Total)

    STAT3 (Total)

    AKT (Total)

    MAPK (Total)

    W.B.

    W.B.

    W.B.

    A

    C

    B

    W.B.

    0 1 4 24 3 90 1 4 24 3 9

    DaysHrHr Days

    EGFR (Total)

    EGFR/ERBB-TKl:

    1 µmol/L (D1–9)

    5 µmol/L (at D9, x8h)

    STAT3 (Total)

    AKT (Total)

    MAPK (Total)

    H1975

    H1975_CL-9D.R

    Figure 2. Targeted inhibition of irreversible EGFR-TKI-sensitive H1975lung adenocarcinoma cells also results in "early’ emergence of tumorresistant escape survivor cells with activated prosurvival-antiapoptoticsignaling. A, significant resistant shift of cell viability in H1975 cellsthat survived 9 days of CL-387,785 treatment. B, activation ofp-STAT3[Y705]/BCL-2/BCL-XL signaling in H1975 cells that survived9 days of CL-387,785 treatment. C, reactivated p-EGFR anddownstream signaling in day 9-resistant-survivors H1975 cells againstCL-387,785 (1 mmol/L) could be inhibited by higher dose ofCL-387,785 (5 mmol/L) in retreatment, but not p-STAT3 orBCL-2.

    Fan et al.

    Cancer Res; 71(13) July 1, 2011 Cancer Research4498

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • BCL-2 nuclear expression was observed in squamous cellcomparing with adenocarcinoma subtype. Our results aboveprovide a rationale to target BCL-2 family signaling throughproapoptotic BH3-mimetic, such as ABT-737 (32–34), in orderto optimize targeted therapies. ABT-737 has been well-char-acterized recently and shown to antagonize BCL-2/BCL-XL,thereby inducing a proapoptotic effect through the mitochon-drial intrinsic apoptosis pathway. NSCLC cell lines wererelatively insensitive to ABT-737 (IC50 > 5 mmol/L), whereasthe SCLC H345 cell line tested was as expected highly sensitive(IC50 < 0.2 mmol/L; Fig. 5C). HCC827 cells with forced over-expression of transfected BCL-2 was sufficient to induce a

    significantly higher erlotinib-resistance, with �100-foldincrease in IC50 (Fig. 5D).

    ABT-737 inhibition in concert with targeted kinaseinhibitors, both in vitro and in vivo, eradicated "early"TKI-resistant tumor evaders and further inhibitedsubsequent tumor recurrence

    We hypothesized that preemptive inhibition and eradica-tion of "early" resistant tumor cells in RTK targeted therapymay impact on the long-term outcome of targeted therapy. Weadopted RNAi knockdown of BCL-2/BCL-XL using siRNAmethods here to test in parallel with ABT-737 (Fig. 6A–B).

    A D

    B C E

    “Quiescence”

    H1975

    – + +

    – – +

    n/a

    CL-387,785(1 µmol/L x 9 Days)

    CL-387,785 Re-Treatment(1 µmol/L, 8 h)

    Withdrawal CL-387,785 x 7 Days

    “Proliferative” with “Activated cytoskeletal functions”

    HCC827-TLVM

    HCC827

    Parental cell

    TKI x 9D -> Washout x 7D

    TKI x 9D -> Washout x 7DParental cell

    P = N.S. P = N.S.

    H1975

    HCC827

    50

    40

    30

    Mito

    tic in

    dex

    (%)

    20

    10

    0 Per

    cent

    cel

    l via

    bilit

    y (M

    TS

    Ass

    ay)

    Per

    cent

    cel

    l via

    bilit

    y (M

    TS

    Ass

    ay)

    0

    0 0.001 0.01 0.1

    Erlotinib (µmol/L)

    1 100

    100

    75

    50

    25

    0

    CL-387,785 (µmol/L)10310.30.1

    25

    50

    75

    100

    Pare

    ntal

    Erlot

    inib

    x9D

    Off-T

    KI x3

    D

    Off-T

    KI x1

    1D

    OFFErlotinib

    (i)

    (ii)

    (iii)

    W.B.

    P-EGFR

    P-MET

    P-MAPK

    Cleaved-PARP

    Actin

    Erlotinib(1 µmol/L)x9 Days

    Erlotinibwithdrawalx11 Days

    Figure 3. "Adaptive" emergence of "early" TKI-resistant tumor survivor cells in HCC827 under erlotinib, and H1975 under CL-387,785 inhibitorypressure. HCC827 cells (A–C): A, TLVM: HCC827_ERL-D9.R cells exhibited a "quiescence-like" state with highly inhibited cell proliferation as wellas cellular cytoskeletal functions that were both readily reversible upon TKI-washout for 11 days. B, cell mitotic index (M.I.) of HCC827 cells under varioustreatment conditions. Error bar, S.E.M. (n¼ 4). *, P < 0.0001; **, P¼ 0.002; †, P¼ 0.03; z, P¼ 0.008. C, HCC827_ERL-D9.R cells were resensitized to erlotinibinhibition after 7 days of TKI-washout culture conditions, indistinguishable from the parental HCC827 cells sensitivity (P > 0.05, N.S.). H1975 cells (D–E):D, H1975_CL-D9.R cells were resensitized to TKI in cellular signaling inhibition promptly upon withdrawal of CL-387,785. E, H1975_CL-D9.R cells wereresensitized to CL-387,785 inhibition upon 7 days of TKI-washout, indistinguishable from the parental H1975 cells sensitivity (P > 0.05, N.S.).

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4499

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • Dramatic reduction in the early TKI-resistant tumor survivorcells was achievable by dual BCL-2/BCL-XL RNAi knockdownin conjunction with erlotinib, but not by mere knockdown ofBCL-2 alone. ABT-737, when used concurrently with erlotinibto inhibit HCC827 cells, also dramatically reduced emergenceof early TKI-resistant tumor survivor cells against erlotinib(Fig. 6B).

    We further carried out in vitro ABT-737 inhibition studieson the NSCLC H1975 TKI-evading tumor cells that wereprimed to upregulate BCL-2/BCL-XL prosurvival signalingby (i) EGFR/ERBB inhibitor (CL-387,785; Fig. 6C), and (ii) dualEGFR–MET inhibitors (erlotinib plus SU11274; Fig. 6D). ABT-737, at a concentration relatively insensitive against H1975parental cells, completely eradicated the early CL-387,785-resistant H1975 evader cells (Fig. 6C, top), either alone or incombination with CL-387,785. Importantly, we showed thatthe "early" TKI-resistant tumor cells were primed to be moresusceptible to ABT-737 inhibition, exhibiting a muchenhanced proapoptotic marker cleaved-PARP induction bythe BH3-mimetic (Fig. 6C, bottom). Moreover, the dual-TKI-resistant H1975_ERL/SU-D9.R tumor cells could also be tar-

    geted by ABT-737 to further induce apoptosis (Fig. 6D). OtherBH3-mimetic BCL-2 family inhibitors tested in our study,obatoclax (35) and HA14–1 (36), also showed efficacy(Fig. 6D). Similar to H1975 cells, early TKI-evading resistantHCC827 cells also displayed therapeutic susceptibility to BH3-mimetic in vitro (Fig. 6E).

    Finally, we tested if the addition of the BH3-mimetic ABT-737 in vivo would prolong the duration of response inerlotinib-treated drug-sensitive HCC827-luc xenograft(Fig. 6E). The in vivo ABT-737 treatment dosage in our studywas chosen based on reported literature on the therapeuticrange in the tested sensitive cancer models (37). The tumorgrowth rate in recurrence of the ABT-737þErlotinib-treatedgroup (IV) was significantly lower than that of the Erlotinib-alone group–III [P ¼ 0.0009; Fig. 6E, Table 1; also Supple-mentary Fig. S3]. ABT-737-alone (Group II) did not accountfor the tumor regression and abrogation of tumor recurrenceas seen in the ABT-737þErlotinib group (Group IV; P ¼0.0004). Finally, the HCC827 tumor recurrence rates atday 18 and day 32 for Group III (Erlotinib-alone) animalswere 50% (P ¼ 0.014) and 62.5% (P ¼ 0.004), respectively,

    A

    HC

    C82

    7H

    CC

    827

    B

    C

    D

    BCL-2Control

    H1975

    BCL-2

    BCL-2

    Erlotinib

    HCC827 Xenograft: BCL-2

    HCC827 Xenograft: P-STAT3[Y705]

    MET/EGFR-TKIs:

    PHA665752 (1 µmol/L)CL-387,785 (1 µmol/L)

    P-EGFR

    EGFR

    P-MET

    MET

    P-STAT3

    STAT3

    BCL-2

    BCL-XL

    Actin

    W.B.

    ControlControl

    SU11274 + Erlotinib

    Erlotinib

    BCL-2P-STAT3

    H19

    75 BCL-2

    BCL-2

    10x

    10x

    Hr

    0 1 4 24 72 9 D

    ays

    5x

    5x

    5x

    20x

    10x

    10x

    P-STAT3

    P-STAT3

    10x

    10x20x 5x

    Figure 4. In vivo xenograft lung tumor survivor cells evading against targeted inhibitors, with activated p-STAT3/BCL-2 antiapoptotic signaling. In vivoHCC827 xenograft model treated with erlotinib (n ¼ 6) revealed "early" TKI-resistant tumor survivor cells (A) with induced BCL-2 expression especiallyin the nuclear subcellular localization (red arrows), and (B) with nuclear translocation (red arrows) of p-STAT3[Y705] from the untreated cytoplasmic localization(yellow arrows). C, combined MET–EGFR TKIs treatment of H1975 cells in vitro also led to an induction of MET-independent activated p-STAT3[Y705],and BCL-2/BCL-XL signaling in TKI-evading survivor cells against 9 days of combined-TKIs (PHA665752 [1 mmol/L] and CL-387,785 [1 mmol/L]). D, inducedBCL-2 expression in H1975-xenograft residual dual SU11274/erlotinib-TKI-evader survivor cells.

    Fan et al.

    Cancer Res; 71(13) July 1, 2011 Cancer Research4500

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • both significantly higher than Group IV (ABT-737þErlotinib;0%; Fig. 6E; Table 1).

    Discussion

    In recent years, molecularly-targeted cancer therapy hasrenewed our hope for cancer cure. Nonetheless, the challengesof clinical tumor resistance, both intrinsic and acquired,remain formidable and substantially limit long-term efficacy.Classic secondary mutational resistance [e.g., T790M-EGFRagainst gefitinib/erlotinib in lung cancer (11)], and receptorkinase class-switching [e.g., from EGFR-addiction to MET/HGF (8, 9, 16), IGF1-R (27, 38), or AXL (39) signaling] have beenidentified in earlier studies that emphasized on "acquired"drug-resistance at "late-stages" of chronic drug treatment. Ourstudy here focuses on the molecular changes of drug-sensitivetumor cell population within the "early" time-window oftargeted TKI treatment. We identified and further character-ized the "early" adaptive functional TKI-resistant lung adeno-carcinoma cells that survived and evaded EGFR/MET-TKI,as early as within 9 days of therapy. During our manu-script preparation, Settleman and colleagues reported theidentification of "drug-tolerant state" in cancer cell subpopu-lations that was maintained through engagement of IGF-1Rsignaling and an epigenetic alteration of chromatin state that

    requires the histone demethylase RBP2/KDM5A/Jarid1A (27).Our report here lends further support to the emerging evi-dence of the existence of tumor cell subpopulations withadaptive resistant-escape under therapeutic inhibitory stress.These early adaptive resistant survivors likely serve as thefounder population as minimal residual disease in solidcancers under therapeutic pressure, which ultimately leadsto frankly recurrent resistant disease on therapy in the future.

    Despite the new insights into nonmutational early resis-tance (28), detailed underlying regulatory mechanism(s) thatdirectly mediate the emergence of such early resurgent resis-tant cells against the inhibitors remain to be fully defined. Ourstudy here provided the first evidence that the "early" emer-gence of resistant tumor survivors evading EGFR/ERBB-METTKIs is independent of MET receptor signaling activation,contrasting previous reports ofMET genomic amplification asacquired resistance mechanism in HCC827 cells that escape"chronic" dose-escalating gefitinib inhibition at "late stages"after many months of treatment (8, 9). We present findingshere that the BCL-2-family signaling in the mitochondrial(intrinsic) programmed-cell death pathway may indeed repre-sent the central mechanism as tumor cells’ newly-dependentaddiction, in promoting "early tumor evasion" to survivetargeted therapeutics. Here, we also provide additional in vivotherapeutic study evidence to validate the efficacy of targeting

    ANSCLC

    Exposure:

    (–)

    “Light”

    “Heavy”

    BCL-2

    BCL-2

    TMA: IHC

    TMA analysis

    W.B.

    Small cell Adenocarcinoma Squamous cell

    Human lung cancer

    Lung cancer TMA: BCL-2

    BCL-2 Expression

    HCC827-BCL-2

    ABT-737 (µmol/L)

    00.

    078

    0.15

    60.

    313

    0.62

    51.

    25 2.5 5

    HCC827

    (†, ‡)

    (*)

    BCL-2

    ParentalMock

    SCLC

    NSCLC

    0 1+ 2+ 3+

    3+

    2+

    1+

    0

    BCL-XL

    HC

    C82

    7

    PC

    -9

    A54

    9

    H59

    5

    H14

    37

    H19

    75

    H19

    93

    H69

    H28

    H20

    52

    MS

    TO-2

    11H

    H34

    5

    H69

    H28

    H20

    52

    MS

    TO-2

    11H

    H34

    5

    H18

    38

    H14

    37

    H19

    75

    H19

    93

    H18

    38

    H44

    1

    HC

    C82

    7

    PC

    -9

    A54

    9

    H44

    1

    SCLC MesotheliomaB C

    D1009080

    7060

    5040

    3020

    100

    Per

    cent

    TM

    A tu

    mor

    sam

    ple

    (%)

    Per

    cent

    cel

    l via

    bilit

    y (M

    TS

    )

    Squa

    mou

    s cell

    Larg

    e ce

    ll

    Aden

    ocar

    cinom

    a

    100

    50

    00 0.001 0.01

    Erlotinib (µmol/L)0.1 1 10

    Moc

    kPa

    rent

    al

    BCL-

    2 Tra

    nsfec

    ted

    BCL-2

    Actin

    0

    25

    50

    75

    100

    H441

    H1975

    H345

    HCC827

    Cel

    l via

    bilit

    y (%

    Con

    trol

    )

    Figure 5. BCL-2/BCL-XL signaling expression and inhibition in lung cancer. A, BCL-2/BCL-XL expression in lung cancer. Top, BCL-2 and BCL-XL expressionin thoracic malignancy cell lines. Bottom, BCL-2 TMA-IHC staining in SCLC, adenocarcinoma, and squamous cell carcinoma of the lung. B, pattern of lungcancer TMA BCL-2 expression. C, cell viability assay by ABT-737 in HCC827, H441 and H1975 (NSCLC), and H345 (SCLC) cells. D, forced BCL-2overexpression in HCC827 cells desensitized the cells to erlotinib. BCL-2-transfected versus mock-transfected cells: †, P < 0.0001, or versus parental cells:z, P < 0.0001. Mock-transfected versus parental cells: *, P ¼ 0.99.

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4501

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • HCC827A

    B

    E

    C DR

    NA

    i

    Moc

    ksi

    -NT

    si-B

    CL-

    2si

    -BC

    L-2/

    BCL-

    XL

    Moc

    ksi

    -NT

    si-B

    CL-

    2si

    -BC

    L-2/

    BCL-

    XL

    BCL-2

    BCL-XL

    Actin

    BCL-2

    6 Days

    Treatment

    H1975 cells

    H1975

    HCC827-luc in vivo bioluminescence imaging

    W.B.

    – – + +

    – – + +– + – +

    W.B.

    DAY:Groups

    I

    I II

    II

    III

    III A

    B

    A

    B

    IV

    IV

    I

    II

    III

    A

    B

    A

    B

    Time

    IV

    0 4 18 32

    – + +

    – + ––

    +

    ––– +

    +

    ++

    +

    +

    – – – +– +–

    H1975

    H1975 cells

    U

    U

    U ER

    L/S

    U

    Oba

    tocl

    ax

    HA

    14-1

    HA

    14-1

    + E

    RL/

    SU

    Oba

    tocl

    ax +

    ER

    L/S

    U

    AB

    T-73

    7 +

    ER

    L/S

    U

    AB

    T-73

    7

    UCL-387,785 x 6D

    CL-387,785 (1 µmol/L)x 9 Days

    SU11274 + Erlotinibx 9 Days

    SU11274 + Erlotinib

    Cleaved-PARPCleaved-PARP

    ActinActin

    CL-387,785 (1 µmol/L)

    ABT-737 (2 µmol/L) ABT-737 (2 µmol/L)Oblatoclax (2 µmol/L)

    Treatment at Days 9 (x8h): Treatment at Days 9 (x8h):

    Erlotinib + SU11274 x 6D

    CL-3

    87,7

    85

    ABT-7

    37 +

    CL-

    387,

    785

    ABT-7

    37

    Erlot

    inib

    Oblat

    oclax

    HA14

    -1

    HA14

    -1 +

    Erlo

    tinib

    Oblat

    oclax

    + E

    rlotin

    ib

    ABT-7

    37 +

    Erlo

    tinib

    ABT-7

    37

    Day 7–9:

    Day 1–6: Day 1–6:

    Treatment

    Day 7–9:

    Post-Transfection RNAi

    (a) (b) (c)

    (d) (e) (f)

    2 Days

    U ERL ERL+ABT-737

    ERL+si-BCL-2/BCL-XL

    HCC827 Cells

    Treatment

    (–)

    (–) Erlotinib (1 µmol/L)

    Day 7−9:

    Day 1−6:

    ERL+si-BCL-2ERL+si-NT

    In vivo treatment groups:

    BCL-XL

    Actin

    ImageMin=-2.8587e+06Max=8.2892e+06

    p/sec/cm2/sr

    Color barMin=-2000

    Max=2.9905e+06

    0

    0.5

    1.0

    1.5

    x106

    2.0

    2.5

    bkg subflat-fieldedcosmic

    Erlotinib

    ABT-737

    HCC827-luc xenograft

    4000000

    3000000

    BLI

    Flu

    x (R

    OI)

    1600000

    1200000

    800000

    400000

    00 1 2 3 4

    Weeks

    EGFR-TKI ± BH3-Mimetics

    5 6 7 8

    * P = 0.0009† P < 0.0001†† P = 0.0004

    * P = 0.0009† P < 0.0001

    Figure 6. BH3-mimetic therapeutic inhibition of the BCL-2/BCL-XL programmed cell death pathway "Achilles’ heel" to eradicate "early" TKI-resistantlung tumor survivor cells. A, siRNA-mediated knockdown of BCL-2 and BCL-XL in HCC827 cells. WCLs at day 2 and day 6 post-siRNA transfection were thenextracted for Western blotting to verify efficient gene knockdown of the target protein(s) expression. B, BCL-2/BCL-XL RNAi knockdown or BH3-mimeticABT-737 (2 mmol/L) in conjunction with erlotinib (1 mmol/L) remarkably suppressed the emergence of "early" EGFR–TKI resistant tumor-evader cells inHCC827. Representative photomicrographs from the triplicate experiments are shown here. Mag: 50�. C, proapoptotic BH3-mimetic ABT-737 eradicated theH1975 early tumor prosurvival resistance against CL-387,785. H1975 cells that were pretreated with 6 days of CL-387,785 (1 mmol/L) were replatedat full confluence, followed by further treatments as indicated for 3 additional days in triplicate, either with CL- 387,785 (1 mmol/L), ABT-787 (2 mmol/L),or ABT-737þCL-387,785, followed by crystal violet cell staining (top). U, untreated control. Bottom, induction of proapoptotic marker cleaved-PARPby BH3-mimetic in the CL-387,785-resistant early tumor survivor H1975 cells. D, ABT-737, Obatoclax, and HA14-1 eradicated the H1975 early tumorprosurvival resistance against dual-TKIs inhibition by erlotinib/SU11274 (ERL/SU). The experiment was carried out with H1975 cells similar to (C)above, except that cells were pretreated with dual EGFR–MET inhibitors here, i.e., erlotinib (1 mmol/L)/SU11274 (1 mmol/L). BH3-mimetic used in treatmentdays 7–9 were all 2 mmol/L in concentration. Top, crystal violet cell survival staining assay. Bottom, BH3-mimetic treatment of the dual ERL/SU-resistanttumor cells induced a proapoptotic response. E, in vivo EGFR inhibition with erlotinib in conjunction with BH3-mimetic ABT-737 led to significantlymore durable tumor response and prolonged remission in HCC827-luc lung adenocarcinoma xenograft. Left, schematic outline of treatment conditions of thein vivo HCC827-luc xenografts. Middle top, in vitro HCC827_ERL-D9.R early resistant TKI-evader cells emerged after 6 days of erlotinib (1 mmol/L) treatment,and were eradicated by cotargeting BH3-mimetic inhibition using ABT-737 (2 mmol/L), Obatoclax (2 mmol/L), or HA14-1 (2 mmol/L)þ/-ongoing erlotinibfrom days 7 to 9. Middle bottom, in vivo HCC827- luc tumor xenograft growth, under treatment conditions as in Groups I–IV, was monitored by BLI asdescribed in the section Materials and Methods. Error bar, � SEM. Erlotinib-alone (III) versus ABT-737þErlotinib (IV): *, P ¼ 0.0009. Erlotinib-alone (III) versusDiluent Control (I), P < 0.0001 (†); ABT-737þErlotinib (IV) versus Diluent Control (I), P < 0.0001 (†). Group II (ABT-737-alone) versus Group IV(ABT-737þErlotinib), P ¼ 0.0004 (††). Right, BH3-mimetic ABT-737 in vivo treatment in conjunction with EGFR-TKI (Group IV) significantly abolishedlung tumor recurrence.

    Fan et al.

    Cancer Res; 71(13) July 1, 2011 Cancer Research4502

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • the BCL-2 family antiapoptotic machinery in the residualtumor survivors under TKI(s) therapeutic inhibition(Fig. 6E). Collectively, our results further raise the promiseof the feasibility in "drugging" the drug-resistant residualtumor cells (40), particularly within an early therapeuticwindow-of-opportunity. Hence, targeting the mitochondrialantiapoptotic machinery as the secondary "Achilles’ heel"newly-emerged in the early-resistant tumor cells appears tobe an attractive therapeutic strategy. On the other hand,concurrent EGFR-TKI and ABT-737 treatment also signifi-cantly suppressed the emergence of early TKI-resistantHCC827 cells, evident as early as within 6 to 9 days (Fig. 6)with the efficacy lasting up to 4 to 6 weeks (data not shown).We believe that the novel therapeutic strategy in targeting theadaptive drug-evader tumor survivor cells emergent withinthe "early treatment time-window" is attractive, as theseevader cells are most likely more "homogeneous" molecularlythan those found eventually as overtly resistant disease after"chronic" TKI inhibition for months. "Late" TKI-resistanttumor cells likely already had undergone divergent resistantmolecular evolution in progression, hence more heteroge-neous, during the long time lapsed under chronic TKI stress.Our data suggest that the early tumor survival against TKI is

    an "adaptive"mechanism, rather than a selection of preexistingresistant cell clones. It remains unclear at present as to whatdefinitively regulates and determines the cell fates early undertargeted inhibition, and which cells among the parental drug-sensitive cell populationwould emerge as resistant survivors inthe beginning of the tumor evolution under therapeutic stres-sors. Nonetheless, the contribution of intrinsic molecularheterogeneity and nongenetic variation within individual cellsamong the parental cell population may still play at least apartial role in the ultimate cell-fate determination. Interest-ingly, BCL-2 has recently been implicated as inhibitor of DNArepairmechanism (41–44), whichmay potentially enhance andfacilitate themolecular evolution of tumor progression beyondthe "early" nonmutational resistance.Persistent STAT3 activation has been detected in a variety

    of hematopoietic malignancies and solid tumors (45–47). Weobserved phosphorylated-STAT3(p-STAT3) in the residualtumor survivor cells both in vitro and in vivo under targetedkinase inhibitors. Our results suggest that "early" reactivationof STAT3 at tyrosine-705 (important in STAT3 dimerizationand subsequent nuclear translocation) may be an important

    central transcriptional programming event prior to the ulti-mate resurgence of resistant tumor survivors. Recent attemptsto develop therapeutic inhibitors to target STAT3 have provento be rather difficult. Nonetheless, a number of BH3-mimeticthat target the key STAT3 downstream transcriptional targets,such as BCL-2/BCL-XL, have shown promise in preclinical andclinical studies, including ABT-737, and the newer pan-BCL-2family inhibitors ABT-263, and obatoclax (37, 48, 49). Thelatter pan-BCL-2 family inhibitors may potentially be moreadvantageous over ABT-737 in their effective inhibition ofMCL-1, shown to induce ABT-737 resistance (50).

    To our knowledge, our study represents the first in vivoevidence that therapeutic targeting early resurgent resistanttumor survivor cells evading cancer targeted inhibitors isfeasible through inhibiting the mitochondrial antiapoptoticBCL-2/BCL-XL signaling in NSCLC, impacting on the thera-peutic outcome. Our results here provide support to furtherdevelop BH3-mimetic beyond basally BCL-2 overexpressingtumors such as SCLC and lymphomas, and extend to NSCLCas a therapeutic strategy to unleash the full potential and tooptimize long-term clinical outcome of oncogenic kinaseinhibitors. We propose that the combinational approach usingBH3-mimetic and RTK inhibitors should be investigatedfurther in the context of NSCLC human clinical trial studies.

    Disclosure of Potential Conflicts of Interest

    No potential conflicts of interest were disclosed.

    Acknowledgments

    We thank Drs. Zhenghe Wang, Stan Gerson, and George Stark for helpfuldiscussion and suggestions. We also thank Mr. Joseph Molter for technicalassistance.

    Grant Support

    This work is supported by National Institutes of Health/National CancerInstitute–K08 Award (5K08-CA102545–05, 3K08-CA102545–05S1; P.C. Ma), Cle-veland Clinic Taussig Cancer Institute, Sol Siegal Lung Cancer Research GrantProgram, Case Comprehensive Cancer Center (Gene Expression and Genotyp-ing Core, Confocal Microscopy Core, Xenograft and Athymic Animal Core,Tissue Procurement and Histology Core, and Animal Imaging Core Facilities;P30-CA43703–12), and Northern-East Ohio Small Animal Imaging ResourcesProgram (R24-CA110943).

    Received July 21, 2010; revised May 3, 2011; accepted May 3, 2011;published OnlineFirst May 9, 2011.

    Table 1. Inhibition of HCC827-luc tumor in vivo xenograft recurrence rates by BH3-mimetic ABT-737treatment in conjunction with EGFR-inhibitor (Fig. 6E)

    Treatment Groups Xenograft Recurrence Rates(%)

    Day 18 Day 32

    III A 50% (2/4) 50%a (4/8) 75% (3/4) 62.5%b (5/8)B 50% (2/4) 50% (2/4)

    IV A 0% (0/6) 0% (0/12) 0% (0/6) 0% (0/12)B 0% (0/6) 0% (0/6)

    aP ¼ 0.014.bP ¼ 0.004.

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4503

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • References1. TsaoMS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, et al.

    Erlotinib in lung cancer–molecular and clinical predictors of outcome.N Engl J Med 2005;353:133–44.

    2. Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N EnglJ Med 2008;358:1160–74.

    3. Ma PC, Maulik G, Christensen J, Salgia R. c-Met:structure, functionsand potential for therapeutic inhibition. Cancer Metastasis Rev2003;22:309–25.

    4. Eder JP, Vande Woude GF, Boerner SA, LoRusso PM. Novel ther-apeutic inhibitors of the c-Met signaling pathway in cancer. ClinCancer Res 2009;15:2207–14.

    5. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V,Thongprasert S, et al. Erlotinib in previously treated non-small-celllung cancer. N Engl J Med 2005;353:123–32.

    6. Lynch TJ, Adjei AA, Bunn PA Jr, Eisen TG, Engelman J, Goss GD, et al.Summary statement:novel agents in the treatment of lung cancer:advances in epidermal growth factor receptor-targeted agents. ClinCancer Res 2006;12:4365s-71s.

    7. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factorreceptor mutations in lung cancer. Nat Rev Cancer 2007;7:169–81.

    8. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO,et al. MET amplification leads to gefitinib resistance in lung cancer byactivating ERBB3 signaling. Science 2007;316:1039–43.

    9. Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, et al. METamplification leads to gefitinib resistance in lung cancer by activatingERBB3 signaling. Science 2007;316:1039–43.

    10. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al.Acquired resistance of lung adenocarcinomas to gefitinib or erlotinibis associated with a second mutation in the EGFR kinase domain.PLoS Med 2005;2:e73.

    11. Kobayashi S, Boggon TJ, Dayaram T, J€anne PA, Kocher O, MeyersonM, et al. EGFR mutation and resistance of non-small-cell lung cancerto gefitinib. N Engl J Med 2005;352:786–92.

    12. Regales L, Balak MN, Gong Y, Politi K, Sawai A, Le C, et al. Devel-opment of new mouse lung tumor models expressing EGFR T790Mmutants associated with clinical resistance to kinase inhibitors. PLoSOne 2007;2:e810.

    13. Kobayashi S, Ji H, Yuza Y,MeyersonM,Wong KK, Tenen DG, et al. Analternative inhibitor overcomes resistance caused by amutation of theepidermal growth factor receptor. Cancer Res 2005;65:7096–101.

    14. Wong KK, Fracasso PM, Bukowski RM, Lynch TJ, Munster PN,Shapiro GI, et al. A phase I study with neratinib (HKI-272), an irrever-sible pan ErbB receptor tyrosine kinase inhibitor, in patients with solidtumors. Clin Cancer Res 2009;15:2552–8.

    15. Engelman JA, Zejnullahu K, Gale CM, Lifshits E, Gonzales AJ, Shi-mamura T, et al. PF00299804, an irreversible pan-ERBB inhibitor, iseffective in lung cancer models with EGFR and ERBB2 mutations thatare resistant to gefitinib. Cancer Res 2007;67:11924–32.

    16. Tang Z, Du R, Jiang S, Wu C, Barkauskas DS, Richey J, et al. DualMET-EGFR combinatorial inhibition against T790M-EGFR-mediatederlotinib-resistant lung cancer. Br J Cancer 2008;99:911–22.

    17. Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, et al.Germline and somatic mutations in the tyrosine kinase domain of theMET proto-oncogene in papillary renal carcinomas. Nat Genet1997;16:68–73.

    18. Christensen JG, Burrows J, Salgia R. c-Met as a target for humancancer and characterization of inhibitors for therapeutic intervention.Cancer Lett 2005;225:1–26.

    19. Ma PC, Kijima T, Maulik G, Fox EA, Sattler M, Griffin JD, et al. c-METmutational analysis in small cell lung cancer:novel juxtamembranedomain mutations regulating cytoskeletal functions. Cancer Res2003;63:6272–81.

    20. Ma PC, Jagadeeswaran R, Jagadeesh S, Tretiakova MS, Nallasura V,Fox EA, et al. Functional expression and mutations of c-Met and itstherapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Res 2005;65:1479–88.

    21. Ma PC, Schaefer E, Christensen JG, Salgia R. A selective smallmolecule c-MET Inhibitor, PHA665752, cooperates with rapamycin.Clin Cancer Res 2005;11:2312–9.

    22. Ma PC, Tretiakova MS, MacKinnon AC, Ramnath N, Johnson C,Dietrich S, et al. Expression and mutational analysis of MET in humansolid cancers. Genes Chromosomes Cancer 2008;47:1025–37.

    23. Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, et al. Globalsurvey of phosphotyrosine signaling identifies oncogenic kinases inlung cancer. Cell 2007;131:1190–203.

    24. Ma PC, Tretiakova MS, Nallasura V, Jagadeeswaran R, Husain AN,Salgia R. Downstream signalling and specific inhibition of c-MET/HGFpathway in small cell lung cancer:implications for tumour invasion. BrJ Cancer 2007;97:368–77.

    25. Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, et al.An orally available small-molecule inhibitor of c-Met, PF-2341066,exhibits cytoreductive antitumor efficacy through antiproliferative andantiangiogenic mechanisms. Cancer Res 2007;67:4408–17.

    26. Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR,et al. Cytoreductive antitumor activity of PF-2341066, a novel inhibitorof anaplastic lymphoma kinase and c-Met, in experimental models ofanaplastic large-cell lymphoma. Mol Cancer Ther 2007;6:3314–22.

    27. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S,et al. A chromatin-mediated reversible drug-tolerant state in cancercell subpopulations. Cell 2010;141:69–80.

    28. Workman P, Travers J. Cancer:drug-tolerant insurgents. Nature2010;464:844–5.

    29. Choong NW, Dietrich S, Seiwert TY, Tretiakova MS, Nallasura V,Davies GC, et al. Gefitinib response of erlotinib-refractory lung cancerinvolving meninges–role of EGFR mutation. Nat Clin Pract Oncol2006;3:50–7.

    30. Tang Z, Jiang S, Du R, Petri ET, El-Telbany A, Chan PS, et al.Disruption of the EGFR E884-R958 ion pair conserved in the humankinome differentially alters signaling and inhibitor sensitivity. Onco-gene 2009;28:518–33.

    31. Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura T, et al.Hepatocyte growth factor induces gefitinib resistance of lung ade-nocarcinoma with epidermal growth factor receptor-activating muta-tions. Cancer Res 2008;68:9479–87.

    32. Chonghaile TN, Letai A. Mimicking the BH3 domain to kill cancer cells.Oncogene 2008;27Suppl 1:S149–57.

    33. Mason KD, Khaw SL, Rayeroux KC, Chew E, Lee EF, Fairlie WD, et al.The BH3 mimetic compound, ABT-737, synergizes with a range ofcytotoxic chemotherapy agents in chronic lymphocytic leukemia.Leukemia 2009;23:2034–41.

    34. Kuroda J, Puthalakath H, Cragg MS, Kelly PN, Bouillet P,Huang DC, et al. Bim and Bad mediate imatinib-induced killingof Bcr/Abl þleukemic cells, and resistance due to their loss isovercome by a BH3 mimetic. Proc Natl Acad Sci U S A2006;103:14907–12.

    35. Trudel S, Li ZH, Rauw J, Tiedemann RE, Wen XY, Stewart AK.Preclinical studies of the pan-Bcl inhibitor obatoclax (GX015–070)in multiple myeloma. Blood 2007;109:5430–8.

    36. Manero F, Gautier F, Gallenne T, Cauquil N, Gr�ee D, Cartron PF, et al.The small organic compound HA14–1 prevents Bcl-2 interaction withBax to sensitize malignant glioma cells to induction of cell death.Cancer Res 2006;66:2757–64.

    37. Zeitlin BD, Zeitlin IJ, Nor JE. Expanding circle of inhibition:small-molecule inhibitors of Bcl-2 as anticancer cell and antiangiogenicagents. J Clin Oncol 2008;26:4180–8.

    38. Guix M, Faber AC, Wang SE, Olivares MG, Song Y, Qu S, et al.Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cellsis mediated by loss of IGF-binding proteins. J Clin Invest 2008;118:2609–19.

    39. Mahadevan D, Cooke L, Riley C, Swart R, Simons B, Della Croce K,et al. A novel tyrosine kinase switch is a mechanism of imatinibresistance in gastrointestinal stromal tumors. Oncogene 2007;26:3909–19.

    40. Dannenberg JH, Berns A. Drugging drug resistance. Cell 2010;141:18–20.

    41. Jin Z, May WS, Gao F, Flagg T, Deng X. Bcl2 suppresses DNA repairby enhancing c-Myc transcriptional activity. J Biol Chem 2006;281:14446–56.

    Fan et al.

    Cancer Res; 71(13) July 1, 2011 Cancer Research4504

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • 42. Hou Y, Gao F, Wang Q, Zhao J, Flagg T, Zhang Y, et al. Bcl2 impedesDNA mismatch repair by directly regulating the hMSH2-hMSH6 het-erodimeric complex. J Biol Chem 2007;282:9279–87.

    43. Wang Q, Gao F, May WS, Zhang Y, Flagg T, Deng X. Bcl2 negativelyregulates DNA double-strand-break repair through a nonhomologousend-joining pathway. Mol Cell 2008;29:488–98.

    44. Zhao J, Gao F, Zhang Y, Wei K, Liu Y, Deng X. Bcl2 inhibits abasic siterepair by down-regulating APE1 endonuclease activity. J Biol Chem2008;283:9925–32.

    45. Bowman T, Garcia R, Turkson J, Jove R. STATs in oncogenesis.Oncogene 2000;19:2474–88.

    46. Darnell JE. Validating Stat3 in cancer therapy. Nat Med 2005;11:595–6.

    47. Yu H, Pardoll D, Jove R. STATs in cancer inflammation andimmunity:a leading role for STAT3. Nat Rev Cancer 2009;9:798–809.

    48. Kutuk O, Letai A. Alteration of the mitochondrial apoptotic pathway iskey to acquired paclitaxel resistance and can be reversed by ABT-737. Cancer Res 2008;68:7985–94.

    49. Gong Y, Somwar R, Politi K, Balak M, Chmielecki J, Jiang X, et al.Induction of BIM is essential for apoptosis triggered by EGFR kinaseinhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoSMed 2007;4:e294.

    50. Yecies D, Carlson NE, Deng J, Letai A. Acquired resistance to ABT-737 in lymphoma cells that up-regulate MCL-1 and BFL-1. Blood2010;115:3304–13.

    BH3-Mimetic Susceptible Early TKI-Resistant Tumor Cells

    www.aacrjournals.org Cancer Res; 71(13) July 1, 2011 4505

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/

  • 2011;71:4494-4505. Published OnlineFirst May 9, 2011.Cancer Res Weiwen Fan, Zhe Tang, Lihong Yin, et al. AgentsInhibitors Are Therapeutically Susceptible to BH3 Mimetic MET-Independent Lung Cancer Cells Evading EGFR Kinase

    Updated version

    10.1158/0008-5472.CAN-10-2668doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://cancerres.aacrjournals.org/content/suppl/2011/05/09/0008-5472.CAN-10-2668.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://cancerres.aacrjournals.org/content/71/13/4494.full#ref-list-1

    This article cites 49 articles, 20 of which you can access for free at:

    Citing articles

    http://cancerres.aacrjournals.org/content/71/13/4494.full#related-urls

    This article has been cited by 4 HighWire-hosted articles. Access the articles at:

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

    SubscriptionsReprints 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/71/13/4494To request permission to re-use all or part of this article, use this link

    Research. on June 27, 2021. © 2011 American Association for Cancercancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 9, 2011; DOI: 10.1158/0008-5472.CAN-10-2668

    http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-10-2668http://cancerres.aacrjournals.org/content/suppl/2011/05/09/0008-5472.CAN-10-2668.DC1http://cancerres.aacrjournals.org/content/71/13/4494.full#ref-list-1http://cancerres.aacrjournals.org/content/71/13/4494.full#related-urlshttp://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/71/13/4494http://cancerres.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice