pd-l1 studies across tumor types, its differential ... · 19.05.2017  · or combination therapy...

11
Cancer Therapy: Preclinical PD-L1 Studies Across Tumor Types, Its Differential Expression and Predictive Value in Patients Treated with Immune Checkpoint Inhibitors Harriet M. Kluger 1 , Christopher R. Zito 2 , Gabriela Turcu 1,3 , Marina K. Baine 4 , Hongyi Zhang 1 , Adebowale Adeniran 4 , Mario Sznol 1 , David L. Rimm 4 , Yuval Kluger 4 , Lieping Chen 5 , Justine V. Cohen 1 , and Lucia B. Jilaveanu 1 Abstract Purpose: With recent approval of inhibitors of PD-1 in mel- anoma, nonsmall cell lung cancer (NSCLC) and renal cell carcinoma, extensive efforts are under way to develop biomarkers predictive of response. PD-L1 expression has been most widely studied, and is more predictive in NSCLC than renal cell carci- noma or melanoma. We therefore studied differences in expres- sion patterns across tumor types. Experimental Design: We used tissue microarrays with tumors from NSCLC, renal cell carcinoma, or melanoma and a panel of cell lines to study differences between tumor types. Predictive studies were conducted on samples from 65 melanoma patients treated with PD-1 inhibitors alone or with CTLA-4 inhibitors, character- ized for outcome. PD-L1 expression was studied by quantitative immunouorescence using two well-validated antibodies. Results: PD-L1 expression was higher in NSCLC specimens than renal cell carcinoma, and lowest in melanoma (P ¼ 0.001), and this nding was conrmed in a panel of cell lines. In melanoma tumors, PD-L1 was expressed either on tumor cells or immune-inltrating cells. The association between PD-L1 expression in immune-inltrating cells and progres- sion-free or overall-survival in melanoma patients treated with ipilimumab and nivolumab was stronger than PD-L1 expres- sion in tumor cells, and remained signicant on multivariable analysis. Conclusions: PD-L1 expression in melanoma tumor cells is lower than NSCLC or renal cell carcinoma cells. The higher response rate in melanoma patients treated with PD-1 inhibi- tors is likely related to PD-L1 in tumor-associated inammatory cells. Further studies are warranted to validate the predictive role of inammatory cell PD-L1 expression in melanoma and determine its biological signicance. Clin Cancer Res; 110. Ó2017 AACR. Introduction Immune checkpoint inhibitors have become the mainstay of treatment for melanoma and other tumor types. The rst immune checkpoint inhibitor to gain approval, ipilimumab, inhibits CTLA-4 on cytotoxic T cells, resulting in durable responses in 11% to 19% of patients with advanced melanoma and prolonging overall survival (OS; refs. 13). Treatment with ipilimumab, however, causes grade 3 to 4 immune-related adverse events in approximately 30% of patients at the FDA-approved dose of 3 mg/kg, diminishing the riskbenet ratio of this drug. Inhibitors of PD-1 or its ligand, PD-L1, have similarly been studied in advanced melanoma and other tumor types, and have now been approved for a number of diseases including melano- ma, renal cell carcinoma, bladder cancer, nonsmall cell lung cancer (NSCLC), head and neck cancer, and Hodgkin's lympho- ma (412). Response rates to PD-1 and PD-L1 inhibitors in melanoma were higher than those of ipilimumab, and the toxicity prole more favorable, with response rates in the range of 30% to 40% and approximately 15% of patients having grade 3 to 4 immune-related adverse events (1, 46). The combination of ipilimumab and nivolumab has been studied in a number of diseases, and is now approved for advanced melanoma. The response rate with the combination was superior to that of either drug alone (57.6% in the rst line setting), and the rate of grade 3 to 4 adverse events was 55%, more than double that of monotherapy (1, 13, 14). Biomarkers pre- dictive of response or resistance are therefore needed to improve patient selection, and given that this is a relatively new regimen with limited patient follow-up, predictive biomarkers have barely been studied. To date, despite a number of attempts to identify biomarkers predictive of response to ipilimumab monotherapy, no biomark- er has consistently been shown to be associated with response or clinical benet (15, 16). Given the broader use of inhibitors of 1 Department of Medicine, Yale University School of Medicine, New Haven, Connecticut. 2 Department of Biology, School of Health and Natural Sciences, University of Saint Joseph, West Hartford, Connecticut. 3 Department of Der- matology, Carol Davila University of Medicine and Pharmacy, Bucharest, Roma- nia. 4 Department of Pathology, Yale University School of Medicine, New Haven, Connecticut. 5 Department of Immunobiology, Yale University School of Med- icine, New Haven, Connecticut. H.M. Kluger and C.R. Zito contributed equally to this article. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Harriet M. Kluger, Yale University School of Medicine, 333 Cedar Street, PO BOX 208028, New Haven, CT 06520. Phone: 203-737- 2572, Fax: 203-785-3788; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-16-3146 Ó2017 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org OF1 Research. on March 29, 2021. © 2017 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

Upload: others

Post on 19-Oct-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

  • Cancer Therapy: Preclinical

    PD-L1 Studies Across Tumor Types, Its DifferentialExpression and Predictive Value in PatientsTreated with Immune Checkpoint InhibitorsHarriet M. Kluger1, Christopher R. Zito2,Gabriela Turcu1,3, Marina K. Baine4, Hongyi Zhang1,Adebowale Adeniran4, Mario Sznol1, David L. Rimm4, Yuval Kluger4, Lieping Chen5,Justine V. Cohen1, and Lucia B. Jilaveanu1

    Abstract

    Purpose: With recent approval of inhibitors of PD-1 in mel-anoma, non–small cell lung cancer (NSCLC) and renal cellcarcinoma, extensive efforts are under way to develop biomarkerspredictive of response. PD-L1 expression has been most widelystudied, and is more predictive in NSCLC than renal cell carci-noma or melanoma. We therefore studied differences in expres-sion patterns across tumor types.

    Experimental Design:Weused tissuemicroarrays with tumorsfromNSCLC,renalcell carcinoma,ormelanomaandapanelof celllines to study differences between tumor types. Predictive studieswere conducted on samples from 65 melanoma patients treatedwith PD-1 inhibitors alone or with CTLA-4 inhibitors, character-ized for outcome. PD-L1 expression was studied by quantitativeimmunofluorescence using two well-validated antibodies.

    Results: PD-L1 expression was higher in NSCLC specimensthan renal cell carcinoma, and lowest inmelanoma (P¼ 0.001),

    and this finding was confirmed in a panel of cell lines. Inmelanoma tumors, PD-L1 was expressed either on tumorcells or immune-infiltrating cells. The association betweenPD-L1 expression in immune-infiltrating cells and progres-sion-free or overall-survival in melanoma patients treated withipilimumab and nivolumab was stronger than PD-L1 expres-sion in tumor cells, and remained significant on multivariableanalysis.

    Conclusions: PD-L1 expression in melanoma tumor cellsis lower than NSCLC or renal cell carcinoma cells. The higherresponse rate in melanoma patients treated with PD-1 inhibi-tors is likely related to PD-L1 in tumor-associated inflammatorycells. Further studies are warranted to validate the predictiverole of inflammatory cell PD-L1 expression in melanoma anddetermine its biological significance. Clin Cancer Res; 1–10. �2017AACR.

    IntroductionImmune checkpoint inhibitors have become the mainstay of

    treatment formelanoma and other tumor types. The first immunecheckpoint inhibitor to gain approval, ipilimumab, inhibitsCTLA-4 on cytotoxic T cells, resulting in durable responses in11% to19%ofpatientswith advancedmelanomaandprolongingoverall survival (OS; refs. 1–3). Treatment with ipilimumab,however, causes grade 3 to 4 immune-related adverse events in

    approximately 30% of patients at the FDA-approved dose of 3mg/kg, diminishing the risk–benefit ratio of this drug.

    Inhibitors of PD-1 or its ligand, PD-L1, have similarly beenstudied in advanced melanoma and other tumor types, and havenow been approved for a number of diseases including melano-ma, renal cell carcinoma, bladder cancer, non–small cell lungcancer (NSCLC), head and neck cancer, and Hodgkin's lympho-ma (4–12). Response rates to PD-1 and PD-L1 inhibitors inmelanomawere higher than those of ipilimumab, and the toxicityprofile more favorable, with response rates in the range of 30% to40% and approximately 15% of patients having grade 3 to 4immune-related adverse events (1, 4–6).

    The combination of ipilimumab and nivolumab has beenstudied in a number of diseases, and is now approved foradvanced melanoma. The response rate with the combinationwas superior to that of either drug alone (57.6% in the first linesetting), and the rate of grade 3 to 4 adverse events was 55%,morethan double that of monotherapy (1, 13, 14). Biomarkers pre-dictive of response or resistance are therefore needed to improvepatient selection, and given that this is a relatively new regimenwith limited patient follow-up, predictive biomarkers have barelybeen studied.

    To date, despite a number of attempts to identify biomarkerspredictive of response to ipilimumabmonotherapy, no biomark-er has consistently been shown to be associated with responseor clinical benefit (15, 16). Given the broader use of inhibitors of

    1Department of Medicine, Yale University School of Medicine, New Haven,Connecticut. 2Department of Biology, School of Health and Natural Sciences,University of Saint Joseph, West Hartford, Connecticut. 3Department of Der-matology, Carol Davila University of Medicine and Pharmacy, Bucharest, Roma-nia. 4Department of Pathology, Yale University School of Medicine, New Haven,Connecticut. 5Department of Immunobiology, Yale University School of Med-icine, New Haven, Connecticut.

    H.M. Kluger and C.R. Zito contributed equally to this article.

    Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

    Corresponding Author: Harriet M. Kluger, Yale University School of Medicine,333 Cedar Street, PO BOX 208028, New Haven, CT 06520. Phone: 203-737-2572, Fax: 203-785-3788; E-mail: [email protected]

    doi: 10.1158/1078-0432.CCR-16-3146

    �2017 American Association for Cancer Research.

    ClinicalCancerResearch

    www.aacrjournals.org OF1

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://crossmark.crossref.org/dialog/?doi=10.1158/1078-0432.CCR-16-3146&domain=pdf&date_stamp=2017-5-19http://clincancerres.aacrjournals.org/

  • PD-1 or PD-L1 in multiple tumor types, intense efforts are underway to identify predictors of response. Expression of PD-L1 ontumor cells has been themost widely studied predictive biomark-er, and has been shown to correlate with response to therapy inmultiple tumor types, although the correlation is insufficient inmost tumor types, includingmelanoma and renal cell carcinoma,for clinical use. Other predictive biomarkers that have beenstudied in melanoma tumors include tumor mutation burden,T-cell receptor repertoire, T-cell infiltrate, gene expression profiles,and presence of MHC molecules. Inflammatory gene expressionsignatures within the tumor, particularly those associated withIFNg secretion, are associated with response to PD-1 inhibitors(17). Tumors with a greater mutation load might be moresensitive, particularly BRCA2 mutations (18). Presence of CD8þ

    T cells at the periphery of the melanoma tumor bed is associatedwith a greater likelihood of response to PD-1 inhibitors, as ispresence of tumor specificMHCclass IImolecules (19, 20). PD-L1expression, however, is the one biomarker that has consistentlybeen shown to be associated with response in multiple trials andclinical settings, albeit insufficiently correlated to be broadly usedalone as a companion diagnostic.

    Most predictive biomarker studies involving PD-L1 expressionhave used standard IHC, as reviewed (21–23). These studies haveused a variety of antibodies and cutpoints for positivity (24). Forexample, in the randomized trial of nivolumab versus chemo-therapy in the second line setting, 43.6% of patients with >5%tumor cell staining for PD-L1 had a response, compared to 20.3%of those with

  • ipilimumab and nivolumab and 52% received anti-PD-1 mono-therapy. RECIST 1.1 criteria were used for clinical assessment andclassification of response.

    To assess antibody specificity and for comparison of stainingacross diseases we used control arrays containing placental andtonsil tissue (known to be positive for PD-L1 expression), pelletsfrom MEL-624 cell lines, overexpressing or not overexpressingPD-L1, and cases of metastatic melanoma, NSCLC and clear cellrenal cell carcinoma, as previously described (27).

    Immunofluorescence and Automated Quantitative AnalysisStaining was performed for automated analysis, as previously

    described (27, 29, 33). Briefly, TMA slideswere deparaffinized andhydrated followed by antigen retrieval. For the E1L3N antibody(catalogno. 13684;Cell Signaling Technology), slideswere boiledfor 20 minutes in a pressure cooker containing 6.5mmol/Lsodium citrate (pH 6.0). Endogenous peroxidase activity wasblocked using hydrogen peroxide solution. Unspecific stainingwas blocked in 0.3% bovine serum albumin solution beforeovernight incubation with anti-PDL1 antibody diluted in TBSsupplementedwith 0.05%Tween20 (1:250, at 4�C). Todetect thecell membrane/cytoplasmic compartment and to create a tumormask, we simultaneously used a cocktail of mouse anti-S100 andanti-HMB45 antibodies, diluted at 1:100 in the overnight solu-tion (BioGenex) for melanoma samples. The tumor mask forNSCLC and renal cell carcinoma was generated usingmouse anti-cytokeratin (catalog no. M3515; DAKO) or antibody cocktail ofanti-cytokeratin, CAIX (gift from Jan Zavada), CD10 (catalog no.M7308;DAKO), and streptavidinHRP (catalog no. S2438; Sigma-Aldrich), respectively. Slides were then incubated with goat anti-mouse IgG conjugated to Alexa Flour 546 diluted at 1:200(Molecular Probes, Inc.) in anti-rabbit amplification reagent(Envision; catalog no. K4003; DAKO).

    For clone 5H1 (generated by Dr. Lieping Chen), antigenretrieval was performed in Tris-EDTA buffer (pH 9.0; DAKO)supplemented with 0.3% Tween 20 (34). Following peroxidaseblocking, slides were incubated at room temperature for 15minutes in ACE blocking buffer. To block endogenous biotin,slides were first incubated in Avidin solution followed by biotinblocking reagent (Vector Laboratories) for 15 minutes at 37�C.Slides were incubated overnight with clone 5H1 (1:400 at 4�C)and rabbit anti-S100 (1:100, catalog no. Z0311; DAKO) in ACEblock. A second TMA control slide, incubated overnight in ACEblock only was used to verify lack of false-positive staining. Abiotynilated anti-mouse IgG (Vector laboratories) was used as asecondary reagent (1:500 for 45 minutes at room temperature),followed by incubationwith the ABCKit (Vector Laboratories) for15 minutes. Amplification reagent and streptavidin-HRP wereused for signal amplification as recommended (DAKO). Forvisualization of S100 slides were incubated with Alexa Fluor546-conjugated anti-rabbit IgG.

    For T-cell detection, the following primary antibodies wereutilized: rabbit anti-CD3 (1: 3200, catalog no. A0452; DAKO),mouse anti-CD4 (1:400, catalog no.M7310; DAKO),mouse anti-CD8 (1:200, catalog no. M7103; DAKO), andmouse anti-FOXP3(1:200, catalog no. ab20034; Abcam). Staining was carried outusing our standardprotocol. To amplify the signal goat anti-rabbitand/or anti-mouse Envision were used followed by fluorophoretyramides. Slides were incubatedwith benzoic hydrazide solution(100 mmol/L in PBS) containing 50 mmol/L hydrogen peroxidebefore second amplification to quench residual HRP.

    Target stainingwas visualizedwith cyanine-5-tyramide (PD-L1,CD4 and CD8, or cyanine-3-tyramide (CD4 and FOXP3) (Perkin-Elmer). A nuclear mask was created by incubating the slides with4,6-diamidine-2-phenylindole (DAPI; 1:500; Invitrogen). Cover-slips were mounted with ProLong Gold antifade medium (Invi-trogen/Life Technologies).

    Quantitative Determination of Target ExpressionImage capturing and quantitative measurements were con-

    ducted using methods and algorithms previously described(33). Tumor was distinguished from the surrounding stromalelements by S-100 signal for melanoma and cytokeratin forNSCLC or renal cell carcinoma. The tumor mask was created byconverting the anti-S100 image via automated processing andthresholding. DAPI signal defining the nuclear compartment, wasutilized to create a total tissue mask (tumor and stroma). Thestromal compartment was obtained by subtracting the tumormask from the total tissue mask. Quantification of PD-L1 signalfor each antibody in the tumor or stromal compartment (totalsignal intensity/area of the compartment) was performed, andoutput obtained on a scale of 0 to 255. To assess the degree oftumor-infiltrating lymphocytes (TIL), we used the percentage ofeither CD3-, CD4- or CD8-, FOXP3þ T-cell area within the histo-spot. Tumor spots were excluded if they contained insufficienttissue (

  • 5H1, generated by Dr. Lieping Chen and validated in previouspublications (25, 27, 29), which binds the extracellular domain ofPD-L1, and a rabbit monoclonal antibody (clone E1L3N fromCell Signaling), which recognizes the intracellular domain or totalendogenous level of the protein. To verify antibody specificity andsensitivity, we first used quantitative immunofluorescent (QIF)analysis of control TMAs containing placenta, tonsil tissue(known to be positive for PD-L1 expression), and pellets fromMEL-624 cell lines, overexpressing or not overexpressing PD-L1,as previously described (27). Control arrays also included12 casesof metastatic melanoma, 12 NSCLC, and 21 renal cell carcinomaspecimens, which allowed comparison of staining patterns andintensity across the three diseases. Staining was confined either tothe cell surface (membranous staining) and in some cases to thecytoplasm and was specific for both antibodies as judged by ourpositive/negative cell line controls (Figure 1). As expected, PD-L1was expressed at high levels inMEL624PD-L1þ cells but not in theparental MEL624 cells. In melanoma specimens, PD-L1 wasexpressed either on tumor cells or in the tumormicroenvironmenton immune-infiltrating cells. In contrast, in NSCLC and renal cellcarcinoma specimens from our control arrays, PD-L1 expressionwas predominantly on tumor cells. An example of a PD-L1þ casefor each tumor type is shown in Figure 1, corresponding to thestrongest expressing histospots for each of the three diseases. Forcomparative studies across tumor types, we quantified the fluo-rescent signal (either maximum or mean intensity values) withinthe entire histospot. Expression levels in the corresponding speci-mens by the two antibodies (5H1 and E1L3N) was highly cor-related (R¼ 0.89). The expression of PD-L1 inNSCLC tumorswassignificantly higher by ANOVA when compared to renal cellcarcinoma and melanoma specimens (mean AQUA score of21.5 vs. 16 vs. 4, respectively for clone 5H1, P ¼ 0.001; 21.5 vs.12.2 vs. 9.6, respectively for clone E1L3N, P ¼ 0.0008; Supple-mentary Figure S1A and B).

    To further verify differential PD-L1 expression in tumor cellsfrom the three histologies as opposed to stromal and immuneinfiltrating cells, we probed lysates from a panel of tumor-derived melanoma cultures, and NSCLC or renal cell carcinomacell lines by Western blot analysis (Figure 2). Both antibodieswere highly specific for their target, demonstrating a band thatcorresponded to a molecular weight of 48 kDa. Equal loadingand protein content among samples was confirmed by b-actinsignal. PD-L1 levels varied significantly by tumor cell type.Expression levels were high or moderately high in six of tenNSCLC and two of six renal cell carcinoma lysates tested. Bycomparison, only two of the nine melanoma cell lines dem-onstrated PD-L1 expression and levels in these samples weremuch lower than in NSCLC and renal cell carcinoma lysates.For semiquantitative assessment of protein levels, signal wasnormalized to b-actin and fold increase above backgroundsignal was determined for each sample using ImageJ software.Expression levels in the corresponding cell lines by the twoantibodies (5H1 and E1L3N) was highly correlated (R ¼ 0.94).NSCLC cell lines had significantly higher PD-L1 levels thanrenal cell carcinoma or melanoma lysates by ANOVA (meanlevels: 14,162 vs. 5,905 vs. 3,032, respectively for clone 5H1,P ¼ 0.047; 21,612 vs. 11,158 vs. 1,828, respectively for cloneE1L3N, P ¼ 0.014; Supplementary Figure S1C and S1D).

    Association between PD-L1 expression in melanoma tumorversus tumor microenvironment cells and response toPD-1-based therapy

    To assess the association between PD-L1 expression on thecell surface of either tumor cells (melanoma cells) or infiltratingimmune cells (stromal cells) and the likelihood of response toPD-1 blockade (alone or in combination with ipilimumab), weconstructed a TMA containing three cores from different areasof pretreatment tumors from 65 melanoma patients. The

    Figure 1.

    Examples of strong PD-L1 fluorescent staining. Staining is shown in paraffin-embedded pellets of MEL624 transfected to overexpress PD-L1 or parental cells,NSCLC, renal cell carcinoma, and melanoma specimens. Either an anti-S100 antibody or anti-cytokeratin (CK) antibody and fluorophore tyramidewere used to distinguish tumor cells (green) from the surrounding stroma and leukocytes. 5H1 antibody (top) or E1L3N antibody (bottom) andCyanine-5-tyramide (red) was used to visualize PD-L1. Overlaid images show PD-L1 expression within the entire histospot comprising both the tumor(orange/yellow) and stroma (red), at 10� magnification.

    Kluger et al.

    Clin Cancer Res; 2017 Clinical Cancer ResearchOF4

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/

  • diameter of the histocores is similar to that of biopsies, ren-dering TMAs a useful surrogate for clinical evaluation of met-astatic samples, which is often based on biopsy rather thanmetastatectomy. The TMA was stained with either 5H1 orE1L3N antibodies and immunofluorescence scores were gen-erated using methods previously described (29). Each variablewas measured separately in either the tumor or stromal com-partment for the two antibodies.

    For patients who had two or three assessable histospots, acomposite score was formed by calculating the mean value. Themean value was compared to themaximum score in these cases todetermine whether there were significant differences, and wefound a very strong correlation between mean and maximumvalues across measurements, with correlation coefficients rangingfrom0.94 to0.98; Supplementary Figure S2). Therefore, the rest ofthe analyses are based onmean values only. For patients who hadonly one histospot, a single value was used.

    To assess the variability of PD-L1 staining between clones 5H1andE1L3N, themeanQIF scores (mean intensity eitherwithin thetumor compartment or stroma) for each case from the corre-sponding TMAswere compared and found to be highly correlated(R ¼ 0.86 and R ¼ 0.78, respectively; Supplementary Figure S3).

    To assess the association between PD-L1 expression andresponse to therapy, we compared PD-L1 expression in twopatient groups: responders (complete response and partialresponse) and nonresponders (stable disease and progressivedisease). Our analysis in the group of patients receiving mono-therapy had a number of limitations; several were treated withpembrolizumab in an expanded access trial, some had receivedprior nivolumab, and most had received multiple other lines oftherapy, and the response rate was therefore lower than expected.We note that 11 of 31 cases with PD-L1 scores available foranalysis were treated with ipilimumab within 3months of receiv-ing anti-PD-1 monotherapy and were excluded from the analysisdue to the potential interaction between the drugs. In the smallergroup of 20 remaining patients treated with PD-1 inhibitormonotherapy, high PD-L1 expression in the stromal compart-ment was weakly associated with response to therapy, but this didnot reach statistical significance (P ¼ 0.07 by Chi-square analysisof scores dichotomized by the median value, data not shown).Given the limitations of this cohort, both in terms of the lower

    than expected response rate and the small cohort size, the remain-der of the analysis focuses on patients treated with the combina-tion of ipilimumab and nivolumab.

    In patients treated with combination therapy (ipilimumab andnivolumab), the time between tissue acquisition and initiation ofimmune checkpoint inhibitors ranged from 1 to 34 months(median-10, mean–11). Twenty-two patients received no inter-current systemic therapies, six received one line of prior therapyand three received two prior regimens. Prior therapies includedIL2 (five patients), IFN (one patient), vemurafenib (threepatients), dacarbazine, carboplatin plus paclitaxel, and IL21 plusnivolumab (one each). Unpaired t tests showed that expression ofPD-L1 was significantly higher in patients who achieved anobjective response (complete or partial) than in nonrespondingpatients (Fig. 3). PD-L1 expression in the stromal compartment,however, better discriminated between responders and nonre-sponders than expression in tumor cells, with either the 5H1 orE1L3N antibodies.

    We next assessed the association between continuous PD-L1scores and PFS or OS by Cox univariate analysis. High PD-L1expression was associated with improved PFS and OS and thisresult was consistent between the two antibodies, particularlywhen PD-L1 was analyzed in the stromal compartment (Table 1).Kaplan–Meier survival curves for PD-L1 scores dichotomized bythe median and PFS or OS are shown in Figures 4 and 5. Witheither antibody, PD-L1 expression in nontumor cells was morestrongly associated with outcome than expression in the S100positive cells. In addition, we note that the percent area of PD-L1-expressing cells in this cohort ranged from 0% to 27.88% (medi-an-1.93%,mean-4.72%). Tumors with PD-L1 intensity above themedian had a corresponding mean PD-L1þ percent area of 8.47compared to 0.44 in low expressers.

    On multivariable analysis, which included age, gender, LDHand American Joint Committee on Cancer (AJCC)M stage, PD-L1expression was an independent predictor of prolonged PFS or OS(Supplementary Tables S2 and S3, respectively). All other vari-ables included in the model were not associated with PFS or OS.To assess the association between PD-L1 expression and otherclinical parameters, we used the Student t-test. There was nosignificant association between PD-L1 and age, gender, LDH orAJCC M stage (data not shown).

    NSCLC

    H82

    7H

    CC

    2935

    H20

    30H

    358

    H52

    0H

    596

    PC9

    H46

    0SW

    900

    HC

    C22

    79M

    EL62

    4(+)

    MEL

    624(

    -)76

    9-P

    786-

    OC

    AK

    I-2C

    AK

    I-1A

    498

    AC

    HN

    YUH

    OIN

    YUM

    AC

    YUM

    UL

    YUM

    UT

    YUSA

    CYU

    SIT

    YUR

    IFYU

    GA

    SP

    E1L3

    N A

    ntib

    ody

    5H1

    Ant

    ibod

    y

    RCC Melanoma

    b-Actin

    b-Actin

    PD-L1

    PD-L1

    Figure 2.

    PD-L1 levels in a panel of tumor-derivedmetastaticmelanoma, NSCLC,or renal cell carcinoma cell lines.MEL-624 cell lines, overexpressing ornot overexpressing PD-L1 were usedas controls (boxed). Protein wasextracted from NSCLC, renal cellcarcinoma, and melanoma cell linesand subjected to SDS-PAGE andWestern blot analysis to detect PD-L1with either the 5H1 or E1L3N antibody.The level of PD-L1 normalized to theb-actin level in each sample wasmeasured by ImageJ software.

    PD-L1 in the Tumor Microenvironment

    www.aacrjournals.org Clin Cancer Res; 2017 OF5

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/

  • Quantification and characterization of TILs and associationwith PD-L1 expression

    To characterize T cell infiltrates in the pretreatment specimensand assess correlations with response to therapy, the TMAs werestained with antibodies to CD3, CD4, CD8 and Foxp3. Stainingwas membranous for CD3, CD8 and CD4 while FOXP3 showedexclusively nuclear staining. TIL subpopulations were character-ized by the percent area of each, CD3, CD4, CD8 and Foxp3expressing cells within the total area of the histospot, as done inprevious studies (28). Our group and others have previouslyshown that PDL1 expression correlates with the overall T cellinfiltrate (29). In this cohort we further confirmed the associationbetween high PD-L1 expression in tumor cells or stroma and highTIL densities as measured by CD3 positivity (SupplementaryFigure S4). TIL populations were split in two groups with highand low TIL content determined by the median % area value. Noassociation was found between the density of TIL subtypes or TILratios and response to therapy, PFS or OS.

    DiscussionPD-L1 expression is being used as a predictive biomarker for

    immune checkpoint inhibitors or incorporated into multipara-metric predictive biomarker models in numerous diseases. Herewe show that expression of PD-L1 varies across tumor types. Theexpression was globally markedly higher in NSCLC tumors thanrenal cell carcinoma tumors, and expression was lower in

    melanomas. Moreover, the pattern of expression appears to differbetween the three tumor types, with more expression on non-tumor cells within the tumor microenvironment in melanomasamples than in NSCLC or renal cell carcinoma. Most clinicallyused assays assess the overall percent of positively staining cellswithin the entire tumor to develop predictive biomarker models,and we therefore compared expression of PD-L1 in the tumormicroenvironment and in tumor cells in pretreatment samplesfrompatients treatedwith immune checkpoint inhibitors. Expres-sion in the tumor microenvironment appears to better predictresponse to therapy.

    The confusion related to PD-L1 as a biomarker is multifac-eted, and due to technical issues of location and size of biopsyspecimen, antibody variability and interpretation of results.Clinical trials using PD-L1 or PD-1 inhibitors have used avariety cut-points to determine "positive" versus "negative"expression, ranging from as low as 1% in the phase III trialof nivolumab versus everolimus in renal cell carcinoma, inwhich 75% of patients had

  • domain of PD-L1 (5H1) and the other than binds to theintracellular domain, E1L3N. Both antibodies were shown tobe highly specific for the target, and staining patterns betweenthe two did correlate in general. When comparing the threetumor types for differences in global PD-L1 expression usinguniform conditions with either antibody, we found that PD-L1expression was significantly higher in NSCLC than renal cellcarcinoma or melanoma, and somewhat higher in renal cellcarcinoma than melanoma. These differences might account for

    the higher cut-points used in some NSCLC trials compared tomelanoma, as our data indicate that a cut-point of 50% inmelanoma, would render all patients "negative" for PD-L1expression.

    Expressionof PD-L1was stronger in apanel ofNSCLCcell lines,intermediate in renal cell carcinoma cell lines, whereasmelanomacell lines were least likely to express PD-L1, and PD-L1þ cell lineshad lower expression intensity than NSCLC or renal cell carcino-ma, further indicating that activity of PD-1 or PD-L1 inhibitors in

    Figure 4.

    Kaplan–Meier curves showing theassociation between PD-L1expression and PFS. The medianPD-L1 intensity score,measured eitherin tumor cells or stroma by the 5H1 orE1L3N antibodies, was utilized todichotomize our cohort into low/highcategories. Patients with high PD-L1levels in the stromal compartment hada longer median PFS compared to lowexpressers.

    Figure 5.

    Kaplan–Meier curves showing theassociation between PD-L1 expressionand OS. PD-L1 scores weredichotomized by the median score inthe tumor or stromal compartment.High PD-L1 level in the stromalcompartment was significantlyassociated with prolonged OS.

    PD-L1 in the Tumor Microenvironment

    www.aacrjournals.org Clin Cancer Res; 2017 OF7

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/

  • melanoma might be due to the interaction between PD-L1 innontumor cells and PD-1 in TILs.

    Our second purpose was to study the clinical significance ofPD-L1 expression in the tumor versus the tumor microenvi-ronment in melanoma samples. We used TMAs to determinePD-L1 expression in up to three locations in the tumor, as thediameter of TMA histocores is similar to that of biopsies,rendering them a useful surrogate for clinical evaluation ofmetastatic samples, which is often based on biopsy rather thanmetastatectomy. Our studies were therefore not designed todetermine location of PD-L1þ cells within the tumor bed, andcannot be compared to findings of Taube and colleagues whohave shown that an abundance of PD-L1þ cells at the tumoredge, rather than deep with the tumor, is associated withresponse to therapy (35, 37).

    We examined PD-L1 expression in either melanoma cells ornontumor cells separately for each antibody. Our studiesincluded only 34 patients who received anti-PD-1 monother-apy, of whom only 20 had not received ipilimumab within 3months prior to initiation of PD-1 inhibitors. In this smallgroup of 20, PD-L1 expression was weakly associated withresponse to therapy, with a P value that only trended towardsignificance. Given the small sample size and the relatively lowresponse rate to therapy (

  • Grant SupportThis work was supported in part by NIH grantsR0-1 CA158167, NIH

    R01CA216846, and K24CA172123 (to H. Kluger, PI), Yale SPORE in SkinCancer, P50 CA121974 (to R. Halaban, PI), NIH R01CA204002, CTSA GrantNumber KL2 TR000140 (to L. Jilaveanu, PI) from the National Center forResearch Resources (NCRR) and the National Center for Advancing Transla-tional Science (NCATS), components of theNational Institutes ofHealth (NIH),and NIH roadmap for Medical Research, a grant from the Lung Cancer research

    Foundation-LUNGevity and Melanoma Research Alliance, Award #308721 (toL. Jilaveanu, PI), and in part by a Research Scholar Grant (130157-RSG-16-216-01-TBG) from the American Cancer Society (to L. Jilaveanu, PI). The content ofthis publication is solely the responsibility of the authors and does notnecessarily represent the official views of the National Institutes of Health.

    Received December 14, 2016; revised January 5, 2017; accepted February 17,2017; published OnlineFirst February 21, 2017.

    References1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al.

    Combined Nivolumab and Ipilimumab or Monotherapy in UntreatedMelanoma. N Engl J Med 2015;373:23–34.

    2. Hodi FS,O'Day SJ,McDermott DF,Weber RW, Sosman JA,Haanen JB, et al.Improved survivalwith ipilimumab in patients withmetastaticmelanoma.N Engl J Med 2010;363:711–23.

    3. Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C, et al.Ipilimumab plus dacarbazine for previously untreated metastatic mela-noma. N Engl J Med 2011;364:2517–26.

    4. Hamid O, Robert C, Daud A, Hodi FS, HwuWJ, Kefford R, et al. Safety andtumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl JMed 2013;369:134–44.

    5. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al.Nivolumab in previously untreated melanoma without BRAF mutation.N Engl J Med 2015;372:320–30.

    6. Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al.Nivolumab versus chemotherapy in patients with advanced melanomawho progressed after anti-CTLA-4 treatment (CheckMate 037): a rando-mised, controlled, open-label, phase 3 trial. Lancet Oncol 2015;16:375–84.

    7. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S,et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. NEngl J Med 2015;373:1803–13.

    8. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV,Necchi A, et al. Atezolizumab in patients with locally advanced andmetastatic urothelial carcinoma who have progressed following treatmentwith platinum-based chemotherapy: a single-arm, multicentre, phase 2trial. Lancet 2016;387:1909–20.

    9. Yared JA, Hardy N, Singh Z, Hajj S, Badros AZ, Kocoglu M, et al. Majorclinical response to nivolumab in relapsed/refractory Hodgkin lymphomaafter allogeneic stem cell transplantation. Bone Marrow Transplant 2016;51:850–2.

    10. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al.Nivolumab versus docetaxel in advanced nonsquamous non-small-celllung cancer. N Engl J Med 2015;373:1627–39.

    11. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al.Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl JMed 2015;372:2018–28.

    12. Ferris RL, BlumenscheinGJr., Fayette J, Guigay J, Colevas AD, Licitra L, et al.Nivolumab for recurrent squamous-cell carcinoma of the head andneck. NEngl J Med 2016;375:1856–67.

    13. PostowMA, Chesney J, Pavlick AC, Robert C, Grossmann K,McDermott D,et al. Nivolumab and ipilimumab versus ipilimumab in untreated mela-noma. N Engl J Med 2015;372:2006–17.

    14. Wolchok JD, Kluger H, CallahanMK, PostowMA, Rizvi NA, Lesokhin AM,et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med2013;369:122–33.

    15. Yuan J, Adamow M, Ginsberg BA, Rasalan TS, Ritter E, Gallardo HF, et al.Integrated NY-ESO-1 antibody and CD8þ T-cell responses correlate withclinical benefit in advanced melanoma patients treated with ipilimumab.Proc Natl Acad Sci U S A 2011;108:16723–8.

    16. Martens A, Wistuba-Hamprecht K, Geukes Foppen M, Yuan J, Postow MA,Wong P, et al. Baseline peripheral blood biomarkers associated withclinical outcomeof advancedmelanomapatients treatedwith ipilimumab.Clin Cancer Res 2016;22:2908–18.

    17. Ribas A, Robert C, Hodi FS, Wolchok JD, Joshua AM, Hwu W-J, et al.Association of response to programmed death receptor 1 (PD-1) blockadewith pembrolizumab (MK-3475) with an interferon-inflammatory

    immune gene signature. ASCO Annual Meeting; J Clin Oncol 2015;33:suppl; abstr 3001.

    18. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al.Genomic and transcriptomic features of response to anti-PD-1 therapy inmetastatic melanoma. Cell 2016;165:35–44.

    19. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al.PD-1 blockade induces responses by inhibiting adaptive immune resis-tance. Nature 2014;515:568–71.

    20. Johnson DB, Estrada MV, Salgado R, Sanchez V, Doxie DB, Opalenik SR,et al. Melanoma-specific MHC-II expression represents a tumour-autono-mous phenotype and predicts response to anti-PD-1/PD-L1 therapy. NatCommun 2016;7:10582.

    21. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers andits association with clinical outcomes. Onco Targets Ther 2016;9:5023–39.

    22. Festino L, Botti G, Lorigan P, Masucci GV, Hipp JD, Horak CE, et al. CancerTreatment with Anti-PD-1/PD-L1 Agents: Is PD-L1 expression a biomarkerfor patient selection? Drugs 2016;76:925–45.

    23. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven bio-markers to guide immune checkpoint blockade in cancer therapy. Nat RevCancer 2016;16:275–87.

    24. McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V,Rehman J, et al. Quantitative assessment of the heterogeneity of PD-L1expression in non-small-cell lung cancer. JAMA Oncol 2016;2:46–54.

    25. TopalianSL,Hodi FS, Brahmer JR,Gettinger SN, SmithDC,McDermottDF,et al. Safety, activity, and immune correlates of anti-PD-1 antibody incancer. N Engl J Med 2012;366:2443–54.

    26. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al.Predictive correlates of response to the anti-PD-L1 antibody MPDL3280Ain cancer patients. Nature 2014;515:563–7.

    27. Jilaveanu LB, Shuch B, Zito CR, Parisi F, Barr M, Kluger Y, et al. PD-L1expression in clear cell renal cell carcinoma: an analysis of nephrectomyand sites of metastases. J Cancer 2014;5:166–72.

    28. Baine MK, Turcu G, Zito CR, Adeniran AJ, Camp RL, Chen L, et al.Characterization of tumor infiltrating lymphocytes in paired primaryand metastatic renal cell carcinoma specimens. Oncotarget 2015;6:24990–5002.

    29. Kluger HM, Zito CR, Barr ML, Baine MK, Chiang VL, Sznol M, et al.Characterization of PD-L1 expression and associated T-cell infiltrates inmetastatic melanoma samples from variable anatomic sites. Clin CancerRes 2015;21:3052–60.

    30. Jilaveanu LB, Zito CR, Aziz SA, Conrad PJ, Schmitz JC, Sznol M, et al. C-Rafis associated with disease progression and cell proliferation in a subset ofmelanomas. Clin Cancer Res 2009;15:5704–13.

    31. McCarthy MM, DiVito KA, Sznol M, Kovacs D, Halaban R, Berger AJ, et al.Expression of tumor necrosis factor–related apoptosis-inducing ligandreceptors 1 and 2 in melanoma. Clin Cancer Res 2006;12:3856–63.

    32. McCarthy MM, Pick E, Kluger Y, Gould-Rothberg B, Lazova R, Camp RL,et al. HSP90 as a marker of progression in melanoma. Ann Oncol2008;19:590–4.

    33. Camp RL, Chung GG, Rimm DL. Automated subcellular localization andquantification of protein expression in tissue microarrays. Nat Med2002;8:1323–7.

    34. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al.Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mecha-nism of immune evasion. Nat Med 2002;8:793–800.

    35. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association ofPD-1, PD-1 ligands, and other features of the tumor immune

    www.aacrjournals.org Clin Cancer Res; 2017 OF9

    PD-L1 in the Tumor Microenvironment

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/

  • microenvironment with response to anti-PD-1 therapy. Clin Cancer Res2014;20:5064–74.

    36. Mahoney KM, Sun H, Liao X, Hua P, Callea M, Greenfield EA, et al. PD-L1antibodies to its cytoplasmic domain most clearly delineate cell mem-branes in immunohistochemical staining of tumor cells. Cancer ImmunolRes 2015;3:1308–15.

    37. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al.Colocalization of inflammatory response with B7-h1 expression in humanmelanocytic lesions supports an adaptive resistance mechanism ofimmune escape. Sci Transl Med 2012;4:127ra37.

    38. Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, et al.Programmed death-ligand 1 expression and response to the

    anti-programmed death 1 antibody pembrolizumab in melanoma.J Clin Oncol 2016;34:4102–9.

    39. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7family, co-stimulates T-cell proliferation and interleukin-10 secretion. NatMed 1999;5:1365–9.

    40. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. Loss oftumor suppressor PTEN function increases B7-H1 expression and immu-noresistance in glioma. Nat Med 2007;13:84–8.

    41. Gowrishankar K, Gunatilake D, Gallagher SJ, Tiffen J, Rizos H, Hersey P.Inducible but not constitutive expression of PD-L1 in human melanomacells is dependent on activation of NF-kappaB. PLoS One 2015;10:e0123410.

    Clin Cancer Res; 2017 Clinical Cancer ResearchOF10

    Kluger et al.

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst February 21, 2017.Clin Cancer Res Harriet M. Kluger, Christopher R. Zito, Gabriela Turcu, et al. Immune Checkpoint InhibitorsExpression and Predictive Value in Patients Treated with PD-L1 Studies Across Tumor Types, Its Differential

    Updated version

    10.1158/1078-0432.CCR-16-3146doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2017/07/04/1078-0432.CCR-16-3146.DC2

    http://clincancerres.aacrjournals.org/content/suppl/2017/02/21/1078-0432.CCR-16-3146.DC1Access the most recent supplemental material at:

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

    Subscriptions

    Reprints and

    [email protected] at

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

    Permissions

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

    .http://clincancerres.aacrjournals.org/content/early/2017/05/19/1078-0432.CCR-16-3146To request permission to re-use all or part of this article, use this link

    Research. on March 29, 2021. © 2017 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 21, 2017; DOI: 10.1158/1078-0432.CCR-16-3146

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-16-3146http://clincancerres.aacrjournals.org/content/suppl/2017/02/21/1078-0432.CCR-16-3146.DC1http://clincancerres.aacrjournals.org/content/suppl/2017/07/04/1078-0432.CCR-16-3146.DC2http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2017/05/19/1078-0432.CCR-16-3146http://clincancerres.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