Top Banner
Gene signatures predictive of response to therapy in NSCLC Gene expression signatures predictive of bevacizumab/erlotinib therapeutic benefit in advanced non-squamous non-small cell lung cancer patients (SAKK 19/05 trial) Anca Franzini 1 , Florent Baty 1 , Ina I. Macovei 2 , Oliver Dürr 3 , Cornelia Droege 4 , Daniel Betticher 5 , Bogdan D. Griogriu 2 , Dirk Klingbiel 6 , Francesco Zappa 7 , and Martin H. Brutsche 1 1 Department of Pulmonary Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland 2 Department of Pulmonary Diseases, University of Medicine and Pharmacy, Iasi, Romania 3 Institute of Data Analysis and Process Design, Zürich University of Applied Sciences, Winterthur, Switzerland 4 Männedorf Hospital, Männedorf, Switzerland 5 Cantonal Hospital Fribourg, Fribourg, Switzerland 6 Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland 7 Department of Medical Oncology, Clinica Luganese, Lugano, Switzerland Corresponding author: Martin H. Brustche, Cantonal Hospital St. Gallen, 95 Rorschacher Strasse, 9007 St. Gallen, Switzerland. Phone: 004171-494-1004; Fax: 004171-494-6118; E-mail: [email protected] Grant Support Swiss Cancer League & Swiss Cancer Center (KLS-2880-02-2012): Martin H Brutsche, KSSG Medical Research Center (MFZF_2014_001): Anca Franzini, Romanian–Swiss Research Program (IZERZO_142235/1): Ina I. Macovei. Abstract Purpose: We aimed to identify gene expression signatures associated with angiogenesis and hypoxia pathways with predictive value for treatment response to bevacizumab/erlotinib (BE) of non- squamous advanced NSCLC patients. Experimental design: Whole genome gene expression profiling was performed on 42 biopsy samples (from SAKK 19/05 trial) using Affymetrix exon arrays, and associations with the following endpoints: time-to-progression (TTP) under therapy, tumor-shrinkage (TS), and overall survival (OS) were investigated. Next, we performed gene set enrichment analyses using genes associated with the angiogenic process and hypoxia response to evaluate their predictive value for patients’ outcome. Results: Our analysis revealed that both the angiogenic and hypoxia response signatures were enriched within the genes predictive of BE response, TS and OS. Higher gene expression levels (GELs) of the 10-gene angiogenesis-associated signature and lower levels of the 10-gene hypoxia response signature predicted improved TTP under BE, 7.1 months vs. 2.1 months for low vs. high-risk patients (P = 0.005), and median TTP 6.9 months vs. 2.9 months (P = 0.016), respectively. The hypoxia Research. on June 12, 2021. © 2015 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135
28

Gene expression signatures predictive of bevacizumab/erlotinib … · 2015. 4. 28. · Gene signatures predictive of response to therapy in NSCLC Gene expression signatures predictive

Jan 30, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • Gene signatures predictive of response to therapy in NSCLC

    Gene expression signatures predictive of bevacizumab/erlotinib therapeutic benefit in advanced non-squamous non-small cell lung cancer patients (SAKK 19/05 trial)

    Anca Franzini1, Florent Baty1, Ina I. Macovei2, Oliver Dürr3, Cornelia Droege4, Daniel Betticher5, Bogdan D. Griogriu2, Dirk Klingbiel6, Francesco Zappa7, and Martin H. Brutsche1

    1 Department of Pulmonary Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland

    2 Department of Pulmonary Diseases, University of Medicine and Pharmacy, Iasi, Romania

    3 Institute of Data Analysis and Process Design, Zürich University of Applied Sciences, Winterthur, Switzerland

    4 Männedorf Hospital, Männedorf, Switzerland

    5 Cantonal Hospital Fribourg, Fribourg, Switzerland

    6 Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland

    7 Department of Medical Oncology, Clinica Luganese, Lugano, Switzerland

    Corresponding author: Martin H. Brustche, Cantonal Hospital St. Gallen, 95 Rorschacher Strasse, 9007 St. Gallen, Switzerland. Phone: 004171-494-1004; Fax: 004171-494-6118; E-mail: [email protected]

    Grant Support

    Swiss Cancer League & Swiss Cancer Center (KLS-2880-02-2012): Martin H Brutsche,

    KSSG Medical Research Center (MFZF_2014_001): Anca Franzini,

    Romanian–Swiss Research Program (IZERZO_142235/1): Ina I. Macovei.

    Abstract

    Purpose: We aimed to identify gene expression signatures associated with angiogenesis and hypoxia pathways with predictive value for treatment response to bevacizumab/erlotinib (BE) of non-squamous advanced NSCLC patients.

    Experimental design: Whole genome gene expression profiling was performed on 42 biopsy samples (from SAKK 19/05 trial) using Affymetrix exon arrays, and associations with the following endpoints: time-to-progression (TTP) under therapy, tumor-shrinkage (TS), and overall survival (OS) were investigated. Next, we performed gene set enrichment analyses using genes associated with the angiogenic process and hypoxia response to evaluate their predictive value for patients’ outcome.

    Results: Our analysis revealed that both the angiogenic and hypoxia response signatures were enriched within the genes predictive of BE response, TS and OS. Higher gene expression levels (GELs) of the 10-gene angiogenesis-associated signature and lower levels of the 10-gene hypoxia response signature predicted improved TTP under BE, 7.1 months vs. 2.1 months for low vs. high-risk patients (P = 0.005), and median TTP 6.9 months vs. 2.9 months (P = 0.016), respectively. The hypoxia

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    2

    response signature associated with higher TS at 12 weeks and improved OS (17.8 months vs. 9.9 months for low vs. high risk patients, P = 0.001). Conclusions: We were able to identify gene expression signatures derived from the angiogenesis and hypoxia response pathways with predictive value for clinical outcome in advanced non-squamous NSCLC patients. This could lead to the identification of clinically relevant biomarkers, which will allow for selecting the subset of patients who benefit from the treatment and predict drug response.

    STATEMENT OF TRANSLATIONAL RELEVANCE

    Clinical outcome of non-small cell lung cancer (NSCLC) could be improved by molecular stratification of patients. Antiangiogenic therapy is approved for treatment of advanced cancers, however because only a subset of patients treated with angiogenesis inhibitors show objective clinical response, there is an increased need for predictive biomarkers. We identified 10-gene signatures associated with angiogenesis and hypoxia-response pathways, which have predictive value for response to combined anti-VEGF/anti-EGFR in non-squamous advanced NSCLC. Subclassification of the patients using these signatures indicated that patients most likely to respond (low-risk) showed higher levels of angiogenesis associated genes mainly involved in maintaining the vascular barrier integrity and lower levels of hypoxia-response genes. Moreover, they showed increased percentile tumor shrinkage and improved overall survival. The inverse trend was observed for the high-risk patients. These findings open the possibility for clinical use of these signatures as predictive biomarkers for identifying patients who would benefit from antiangiogenic therapies.

    Introduction

    Solid tumor cells in their primary goal to avoid senescence and achieve uncontrolled

    proliferation co-opt neighboring stromal cells to assist them with the expansion of the

    malignant tissue.1, 2 These tumor-associated stromal components have been demonstrated

    to have an important role in tumor growth, disease progression3 as well as in response and

    resistance to therapeutic agents.4, 5 Moreover, tumors are strongly dependent on

    angiogenesis, the formation of neovessels from the preexisting vasculature, to obtain the

    nutrients and oxygen essential for their rapid growth.6 Consequently, antiangiogenic

    therapeutic strategies were extensively exploited in the last decade in search for more

    efficient cancer treatments.7

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    3

    Though multiple therapeutic strategies have been developed against vascular endothelial

    growth factor (VEGF),8 bevacizumab,9 a monoclonal antibody targeting VEGFA, is the first

    approved antiangiogenic drug for clinical use. Bevacizumab is mostly used in combination

    with standard chemotherapy for treatment of metastatic cancers,10-12 and as single agent in

    recurrent glioblastoma.13 However, survival benefits are observed only in a subset of

    patients, as a significant number of patients show only modest response presumably

    because of intrinsic or rapidly acquired resistance to antiangiogenic therapy.14 One proposed

    mechanism of resistance to antiangiogenic agents is the onset of hypoxia within the tumor

    as a result of vessel regression during the course of antiangiogenic therapy.15, 16 Moreover,

    effective inhibition of neovascularization using antiangiogenic therapy was shown in some

    cases to change the phenotype of tumors by increasing their invasion and metastatic

    potential.17 Additionally, significant rates of adverse effects were reported for patients

    receiving anti-VEGF therapy, as well as a mortality rate of 1%, which was a direct

    consequence of bevacizumab administration.18, 19 Thus, understanding all cascades of

    vascular signaling involved in the response to antiangiogenic therapy and subsequent

    resistance is critical to achieve full potential of this therapeutic approach.

    Despite these obvious limitations of antiangiogenic therapy, because a subset of cancer

    patients treated with angiogenesis inhibitors show objective clinical response, there is an

    increased need to identify robust predictive biomarkers,20 which could allow for selecting

    the subgroup of patients who would benefit from the treatment.

    In a recent study, aimed at identifying novel biomarkers for response to combined anti-

    VEGFA/anti-EGFR (epidermal growth factor receptor) therapy in non-squamous NSCLC by

    exploring gene expression at exon-level, we identified EGFR exon 18 as a predictive marker

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    4

    for patients with metastatic non-squamous NSCLC who have received no previous therapy.21

    The gene expression profiles obtained from this set of microarray data are derived, however,

    from highly heterogeneous clinical biopsies consisting of both tumor and activated stromal

    cells. In a previous study, Baty et al. revealed that prediction of survival was independent of

    tumor cell content present in each NSCLC biopsy.22 This suggests a strong predictive

    contribution from the tumor microenvironment compartments in NSCLC. Additionally,

    recent studies have demonstrated that tumor microenvironment can provide independent

    and reliable predictors of clinical outcome.23 A key constituent of the tumor

    microenvironment is the blood vasculature, which undergoes angiogenesis to sustain the

    high proliferative rate of the tumor, and is the direct target of antiangiogenic therapies.

    Because there is a high degree of cross-talk between epidermal growth factor receptor

    EGF(R) and VEGF(R) pathways, they have been identified as potentially synergistic for dual

    targeting.24 EGFR pathway is involved in growth factor–induced angiogenesis,

    transcriptionally up-regulating VEGF expression.25 Additionally, multiple studies

    demonstrated that hypoxia can trigger the angiogenic switch in solid tumors.26 Therefore,

    we aimed to investigate whether angiogenesis and hypoxia-associated gene expression

    signatures could predict the combined anti-VEGF/anti-EGFR treatment response in advanced

    non-squamous therapy naїve NSCLC patients unselected for EGFR mutation status. Our

    analysis identified 10-gene angiogenesis-associated and hypoxia-response signatures

    predictive of therapeutic response to bevacizumab/erlotinib (BE) having time-to-progression

    (TTP) and tumor shrinkage (TS) as endpoints. We also identified for 10-gene signatures with

    prognostic value. These signatures hold great potential for clinical application allowing for

    identification of biomarkers, which can identify the patients most likely/ less likely to

    respond to targeted therapy.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    5

    Methods

    Study design. Our study is based on clinical bronchoscopic biopsies available from 42

    patients for which genome-wide gene expression was studied in a microarray platform.

    These patients (88% adenocarcinoma, 57% female, 31% never smoker) were enrolled in the

    Swiss Group for Clinical Cancer Research (SAKK) 19/05 phase II trial.27 Identification of

    predictive gene-expression signatures from RNA gene expression analysis was a predefined

    goal of this trial. The detailed clinical information of these patients was published

    previously.21 For these patients with stage IIIB or IV (93%) non-squamous NSCLC, BE was

    used as first-line therapy (independent of the EGFR mutation status) followed by standard

    platinum-based/gemcitabine chemotherapy (CT) after disease progression, (Figure 1). Time

    to disease progression and percentage tumor shrinkage at 12 weeks (assessed by CT scans)

    were defined according to RECIST criteria. To confirm that there was no sample selection

    bias for the 42 patients included in our study, we performed statistical analysis and found no

    significant differences between the study group and the patients with no available biopsies.

    The results are summarized in Supplementary Table 1.

    Gene expression analysis

    Total RNA from 42 bronchoscopic biopsy samples were extracted using miRNeasy Mini Kit

    (Qiagen) according to the manufacturer's recommendations. Affymetrix Human Exon 1.0 ST

    arrays (Affymetrix, Santa Clara, CA, USA) were used for mRNA hybridization. The gene level

    probesets were preprocessed, quality-checked and normalized using the Robust Multi-array

    Average (RMA) procedure.28 Data were expressed as log2 ratio of fluorescence intensities of

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    6

    the sample and the reference, for each element of the array. Additional experimental details

    for this section were previously reported by us.21

    Statistical analysis of gene expression.

    Survival and time-to-event analysis were performed by applying univariate Cox proportional

    hazards regression and principal component analysis (metagene approach) according to a

    previously described method.29 We built a binary score (low/high risk) using the median of

    the metagene scores. The classification accuracy of our algorithm was assessed by leave-

    one-out cross-validation (LOOCV). Time-to-event and survival results were displayed using

    Kaplan-Meier curves, and log-rank tests are reported. Hierarchical cluster analysis was

    carried out using the Euclidean distance together with the complete linkage agglomerative

    method. The median follow up time was estimated using the reverse Kaplan-Meier method.

    All statistical tests were performed using the R statistical software version 3.1.0

    (http://www.R-project.org). A P value of 0.05 was set as threshold for significance for all

    study outcomes.

    Gene set enrichment analysis (GSEA). We performed enrichment analysis using GSEA v2.1.0

    software (http://www.broad.mit.edu/gsea) and GSEA-preranked function.30 The gene lists

    were ranked by using as metric –log10(P) resulted from the Cox-regression analysis for all

    endpoints.

    Quantitative real-time PCR analysis. Forty samples of RNA isolated from non-squamous

    NSCLC biopsies and previously analyzed by microarrays were available for reverse

    transcription (RT). RT was performed using QuanTiect® Reverse Transcription Kit from

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    7

    Qiagen starting from 10 ng RNA for each qPCR reaction. The quantity and integrity of RNAs

    were assessed using an UVS-99 micro-volume spectrophotometer (ACTGene). For this study

    we investigated the expression levels of nine angiogenesis-associated predictive genes. For

    RT-qPCR experiments we used predesigned optimized primer sets (annealing temperature

    55 °C) for GPR116, EMCN, ITGA9, GNG11, KDR, PECAM1, S1PR1, JAM2 and RHOJ (QuantiTect

    Primer Assay, Qiagen). For sequences please refer to the Supplementary Table 2.

    All samples were processed in duplicate for the qPCR with the LightCycler®480 SYBR Green I

    Master in a 20 µL reaction volume containing 4 µL water, 1 µL PCR primer, 10 µL Master mix

    and 5 µL cDNA (10 ng). Quantitative real-time PCR experiments were performed on a

    LightCycler® 480 II instrument using the initial denaturation at 95 °C for 10 minutes followed

    by 45 cycles: 95 °C for 10 sec, 55 °C for 20 sec, 72 °C for 20 sec. Controls containing no

    reverse transcriptase were included for each sample. The mRNA expression levels were

    calculated relative to HPRT1 house-keeping gene and the relative quantification of the gene

    expression was performed using 2─ΔCp method.31 The correlation between the RT-qPCR gene

    expression levels (GELs) and microarray GELs was measured by means of Spearman's

    correlation coefficient.

    Results

    10-gene angiogenesis-associated and hypoxia-response signatures predict response to BE

    therapy. We hypothesized that the genes which could associate with the response to BE

    therapy are genes involved in angiogenic signaling pathways. To investigate this, we

    performed GSEA30 using a core gene signature (43 genes) specific for angiogenesis

    transcriptional program predefined by integrative meta-analysis of the expression profiles of

    over 1,000 primary human cancers.32

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    8

    GSEA revealed a significant enrichment of the angiogenesis-associated genes within the

    genes that associate with TTP under BE therapy endpoint (mean rank = 4246, P=0.004,

    Figure 2A). As VEGF expression is directly activated under hypoxic conditions by the

    transcription factor hypoxia inducible factor 1 alpha (HIF1α), we decided to additionally

    investigate hypoxia-response genes by GSEA. For this analysis we used a previously derived

    common hypoxia metagene (51 genes) across cancer types.33 Within the genes which

    associate with TTP under BE therapy, we identified by GSEA a significant enrichment of the

    hypoxia-response genes (mean rank = 4798, P = 0.001, Figure 2B). The top 12-ranked

    angiogenesis-associated and hypoxia-response genes, which significantly correlate with TTP

    under BE therapy are given in Figure 2C and 2D.

    We then applied unsupervised hierarchical clustering to the top 10-ranked

    angiogenesis-associated genes to investigate whether there are gene expression patterns

    correlating with the BE treatment response. The clustering output for the angiogenesis-

    associated signature is displayed in Figure 3A. We identified three distinct gene clusters:

    Cluster A1 (low risk, 9 patients, 21 %) was characterized by an increased expression of the

    angiogenesis-associated signature and associated with improved TTP under BE 7.1 months,

    95% confidence interval (95% CI): 4.0 ─ ∞; Cluster A2 (high risk, 12 patients, 29 %) was

    characterized by a decreased expression of the angiogenesis-associated gene signature and

    associated with reduced response to BE treatment with a mean TTP under BE of 2.1 months

    (95% CI: 1.4 ─ ∞). The patients within the third cluster A3 (medium risk, 21 patients, 50 %)

    showed intermediate angiogenesis-associated GELs and a median TTP under BE of 4.1

    months (95%CI: 3.1 ─ 7). The Kaplan-Meier TTP curves are shown in Figure 3C. GELs for the

    top 10-ranked angiogenesis-associated genes for patients included in each cluster are given

    in Supplementary Figure 1.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    9

    Hierarchical clustering showing the variability of gene expression of hypoxia-response top

    10-ranked genes is given in Figure 3B. For this signature, we identified two significant

    clusters: Cluster H1 (18 patients, 43 %) with lower hypoxia-response GELs and Cluster H2 (24

    patients, 57 %) with hypoxia-response higher GELs (Supplementary Figure 2).

    Dichotomization of the patients into low-risk (Cluster H1) and high-risk (Cluster H2),

    subgroups based on the gene expression levels of the hypoxia-response signature revealed a

    marked difference in TTP under BE between the two groups (Figure 3D). We obtained a

    median TTP for the high-risk patients of 2.9 months (95%CI: 1.8 ─ 4.1) and of 6.9 months

    (95% CI: 4.0 ─ 9.7) for the low-risk patients.

    Hypoxia-response gene signature predictive of tumor shrinkage after BE treatment in non-squamous NSCLC patients.

    An additional secondary endpoint of interest was tumor shrinkage (TS) measured at 12

    weeks after BE treatment, which indicates clinically relevant direct anti-tumor activity.

    Because of lack of measurements at 12 weeks, 14 patients had to be excluded from this

    analysis. We analyzed the genes correlating with TS in our patients using GSEA using both

    the angiogenesis-associated and hypoxia-response gene signatures (Figure 4A and B). Both

    gene signatures where significantly enriched in the gene set correlating with TS (mean rank =

    5104, P = 0.002 and mean rank = 7795, P = 0.038, respectively). The resulting top 12-ranked

    genes for both signatures are given in Figure 4C and D. Further, we classified the patients

    based on the metagene score calculated for the top 10-ranked genes for each gene

    signature. For the angiogenesis-associated signature, the resulting two patients groups (low-

    risk and high-risk) showed a non-significantly different median TS, 13.7 % interquartile range

    (IRQ): -0.8 ─ 26.2 vs. 0 %, IQR: -3.2 ─ 16.4, P = 0.755. In contrast, when dichotomization was

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    10

    performed using the 10-gene hypoxia-response signature, the low-risk patients had a

    significantly higher median TS than the high-risk patients (16.1 %, IRQ: 0 ─ 26.2 vs. -0.4 %,

    IRQ: -2.5 ─ 2.6, P = 0.013), indicating a higher potential for assessing treatment response

    using this signature. A heat map showing the gene expression levels of the top 10-ranked

    hypoxia response genes predictive of tumor shrinkage is given in Supplementary Figure 3.

    Angiogenesis-associated and hypoxia-response gene signatures have prognostic value for

    non-squamous NSCLC patients. Lastly, we analyzed the prognostic value of both

    angiogenesis and hypoxia gene signatures by investigating the genes correlating with the

    overall survival (OS). The median follow-up time was 24.9 months (95% CI, 23.9 - ∞). The

    results of the GSEA for OS are given in Figure 5A and 5B, and the derived top 12-ranked

    genes are given in Figure 5C and 5D, respectively.

    Both gene signatures where significantly enriched in the gene set correlating with OS (mean

    rank = 5064, P = 0.031 and mean rank = 4555, P = 0.001, respectively). Using the top

    10-ranked genes for both signatures to dichotomize the patients in low-risk (longer OS) and

    high-risk (shorter OS) revealed marked differences in OS. Figures 5E and 5F show the Kaplan-

    Meier OS curves for both gene signatures used. Using the angiogenesis-associated gene

    signature led to a median OS for the high-risk patients of 10.6 months [95%CI, 5.2 ─ 19.4]

    and for the low-risk patients of 14.1 months [95% CI, 10.5 ─ ∞], P = 0.035. The hypoxia-

    response gene signature showed a higher prognostic value (P = 0.001) resulting in a median

    OS for the high-risk patients of 9.9 months [95% CI 4.8 ─ 13.4] and 17.8 months [95% CI 16.6

    ─ ∞] for the low-risk patients. A heat map displaying the gene expression levels of the top

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    11

    10-ranked hypoxia response genes significantly associated with OS is given in Supplementary

    Figure 4.

    Validation of microarray gene expression levels (GELs) by RT-qPCR. RT-qPCR was used to

    assess the GELs that were established by microarray analysis. RT-qPCR was performed on 40

    out of 42 RNA samples and for nine genes, plus three control genes. The Spearman’s rank

    correlation coefficients (r) were between 0.51─0.71. The correlation was significant for each

    gene. We obtained P values for these associations of < 0.0008, demonstrating good

    agreement between the two complementary methods in quantifying the gene expression

    levels (Figure 6). This outcome indicates a statistically significant correlation between the

    microarray GELs and GELs assessed by RT-qPCR, which is a more convenient and less

    expensive technique for routine application in a clinical setting. These results are in

    agreement with the expected level of correlation considering the fact that there is no

    designed sequence overlap between the qPCR primers and the microarray probes.

    Discussion

    There are several gene expression signatures identified as prognostic biomarkers for lung

    cancer, mostly for lung adenocarcinoma.34 However, gene expression signatures with

    predictive value for treatment response to antiangiogenic therapy are still lacking.

    Therefore, the aim of this study was to evaluate the predictive potential of angiogenesis-

    associated and hypoxia-response gene signatures for the benefit of BE treatment. We

    performed GSEA for each endpoint, which led to the identification of specific angiogenesis

    and hypoxia–derived 10-gene signatures. These signatures were then evaluated for their

    predictive (TTP und TS endpoints) and prognostic value (OS) and tested in an independent

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    12

    data set. Our findings might shed light on the mechanism of antiangiogenic treatment

    response and resistance. Moreover, they may lead to the identification of the causal

    mechanism behind the high proportion of patients treated with angiogenesis inhibitors

    showing partial response, as demonstrated by increased progression free survival (PFS),

    however, with no improvement in their OS.

    We took a closer look at the angiogenesis-associated genes with predictive value for TTP

    under BE therapy. The expression of the first ranked gene, adhesion G-protein-coupled

    receptor 116 (GPR116), was shown to be significantly correlated with tumor progression,

    recurrence, and poor prognosis in human breast cancer. However, here we identify this gene

    to play a protective role and its expression to be associated with lower risk of disease

    recurrence in NSCLC. The biological role of GPR116 in angiogenesis and endothelial

    proliferation remains to be assessed, however this protein is highly expressed in normal

    human lung tissue and it was recently demonstrated to regulate lung surfactant

    homeostasis.35 The second ranked gene, endomucin (EMCN), is an endothelial sialomucin

    and an endothelial-specific marker, involved in cell-cell and cell-extracellular matrix

    interactions.36 Integrin α9 (ITGA9), which forms a heterodimeric receptor with activated β1

    integrin, has been demonstrated to bind directly to VEGFA, and to contribute to

    angiogenesis.37 α9 β1 integrin ligand, tenascin-C, enhances secretion of S1P (sphingosine 1-

    phosphate) in endothelial cells. In turn, S1P promotes endothelial cell barrier integrity,38

    acting as an anti-permeability agent39 and modulating vessel integrity through its cognate

    receptor S1PR1. S1PR1 inhibits VEGFR2 signaling and suppresses endothelial hypersprouting

    via stabilization of junctional VE-cadherin, which leads to enhanced cell-cell adhesion.40

    PECAM-1 and JAM2 are adhesive proteins that accumulate in adherens junctions and

    maintain the restrictiveness of the endothelial barrier. RhoJ, an endothelial-enriched Rho

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    13

    GTPase, regulates angiogenesis and vessel integrity.41 GNG11 is a member of the γ subunit

    family of heteromeric G-protein, which regulates cellular senescence in response to

    environmental stimuli.42 To obtain the ranking of the discriminating power for each gene, we

    performed optimized between-group classification (OBC)43 sensitivity analysis for this

    signature. OBC suggests that for angiogenesis-associated signature S1PR1, GPR116, and

    PECAM1 have the highest discriminating power for TTP under BE (Supplementary Figure 5a ).

    The hypoxia-response genes with predictive value for TTP under BE were mostly genes

    involved in the glycolytic and other metabolic pathways44: PFKP (phosphofructokinase,

    platelet), LDHA (lactate dehydrogenase A), GPI (glucose-6-phosphate isomerase), ALDOA

    (aldolase A), ACOT7 (acyl-CoA thioesterase 7), PGK1 (phosphoglycerate kinase), SLC25A32

    (solute carrier family 25, mitochondrial folate carrier, member 32) and SLC2A1 (solute carrier

    family 2, glucose transporter, member 1). The first-ranked hypoxia-response gene DDIT4

    (DNA-damage-inducible transcript 4 protein, also known as REDD1) has been shown to be

    implicated in inhibition of the mTORC1 (mammalian target of rapamycin kinase) signaling

    pathway, which is relevant in tumor suppression;45 MIF (macrophage migration inhibitory

    factor) has been revealed to act within the tumor microenvironment to stimulate

    angiogenesis and promote immune evasion;46 ADM (adrenomedullin) is involved in

    promoting tumor progression by sustaining proliferation and angiogenesis.47 For this

    signature, OBC indicated that DDIT4 and MIF have the highest discriminatory power

    (Supplementary Figure 5b). Importantly, the hypoxia-response signature shows both high

    predictive and prognostic value and could potentially guide future clinical decisions.

    Our gene expression analyses suggest that the low-risk patients (most likely to respond to

    antiangiogenic combined BE therapy and have better outcome) have a tumor

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    14

    microenvironment that sustains controlled angiogenesis, developing blood vessels with

    increased levels of integrity and reduced permeability. The controlled angiogenesis is

    associated with lower risk of hypoxia within the tumors (lower levels of hypoxia response

    genes). On the other hand, the high-risk patients (less likely to respond to BE treatment and

    have worse outcome) have a tumor microenvironment that sustains aberrant angiogenesis

    with reduced vascular stability and increased vascular permeability. This phenotype is

    further associated with increased tumor hypoxia and an earlier onset of disease progression.

    Moreover, we observed that the 10-gene hypoxia-response signature has a higher

    prognostic power than the angiogenesis-associated signature. This most likely indicates that

    CT has a relatively high contribution to OS, which is expected taking into account that

    hypoxic tumors are less responsive to CT than normoxic tumors.48

    Additionally, we performed LOOCV from the original dataset in order to test the robustness

    of the gene signatures associated with TTP under BE and OS endpoints. The perturbations

    resulting from LOOCV had an insignificant impact on our findings demonstrating the

    robustness of the discriminatory power of our gene signatures (Supplementary Table 3).

    Unfortunately, we could not investigate the performance of our gene expression signatures

    using an identical independent data set, as an additional gene expression data set from

    pretreatment biopsies of treatment-naïve NSCLC patients receiving the same therapeutic

    scheme is not available for validation. However, we tested our 10-gene signatures

    correlating with TTP under BE using a data set comprising GELs of biopsies from a pretreated

    NSCLC population (BATTLE-1 study,49 trial registration ID: NCT00409968, raw data GEO series

    accession number: GSE33072). We analyzed the association between our 10-gene

    expression signatures and PFS for two treatment arms: erlotinib (25 patients, all EGFR-WT)

    and sorafenib (31 patients, all EGFR-WT; the patients with squamous cell or adenosquamous

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    15

    carcinoma were excluded). This analysis revealed that our 10 gene angiogenesis-associated

    signature had no predictive value for either erlotinib or sorafenib response as second line

    therapy. Likewise, the 10-gene hypoxia-response signature had no predictive value for

    erlotinib response. However, for the second line sorafenib-treated patient group (majority

    erlotinib resistant), we found that the 10-gene hypoxia-response signature discriminated

    between low-risk (responders) and high-risk patients (non-responders): PFS 3.65 months

    (95%CI: 1.87 ─ 8.74) and 2.61 months (95%CI: 1.81 ─ 3.61), respec vely (P = 0.0186,

    Supplementary Figure 6). Sorafenib, a multikinase inhibitor, achieves antiangiogenic effects

    by blocking VEGFR and PDGFR. In addition, several studies suggest that sorafenib exerts a

    negative regulatory effect on angiogenesis by suppressing expression of VEGF via inhibition

    of HIF-1α accumulation and activation.50 Although caution in extrapolating data from one

    clinical trial to the other is required, our findings suggest that the hypoxia-response

    signature may be a predictive biomarker of anti-VEGF(R) treatment response (such as

    bevacizumab and sorafenib) and has no predictive power for erlotinib activity. The lack of

    predictive value of the 10-gene angiogenesis in the context of second line sorafenib

    treatment could indicate that the expression of the angiogenesis-associated genes

    significantly changes from treatment naïve tumors to CT resistant tumors, whereas hypoxia-

    response GELs are affected to a lesser extent. Importantly, only 18 patients in sorafenib-

    treated group had biopsies originating from the lung; the gene expression signatures of

    tumor-associated vascular endothelial cells originating from other organs could vary greatly.

    Nonetheless, our findings suggest that the 10-gene hypoxia-response has a high predictive

    value of treatment response even in the context of second line antiangiogenic therapy and a

    great potential for future clinical use.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    16

    The positive correlation between pretreatment angiogenesis-associated and hypoxia-

    response GELs from tumor biopsies and clinical outcomes following BE treatment derived

    from our analyses supports further evaluation of these candidate gene signatures as

    potential biomarkers for the selection of the patient subpopulation most likely to obtain

    benefit from antiangiogenic therapy. There are, however, several limitations that accompany

    our study. Our study comprises a relatively low number of patients, and control groups (no

    treatment, and bevacizumab-only and erlotinib-only treatment) are absent. Further

    validation with larger number of patients and adequate control arms is needed.

    Nevertheless, we found highly statistically significant differences in the hypoxia-response

    GELs of responders vs. non-responders to antiangiogenic therapy in both our data set and an

    additional independent data set. This is very promising and suggests that the identified

    signatures may be clinically useful for further stratifying non-squamous NSCLC patients and

    allow for personalized treatment to avoid unnecessary costs and patient exposure to

    toxicity.

    Conclusions

    We identified 10-gene angiogenesis and hypoxia signatures, which can predict the subgroup

    of patients with higher likelihood of responding to angiogenic therapy. These patients had

    higher GELs of the genes mainly involved in maintaining the vascular barrier integrity and

    lower levels of hypoxia-response genes. Moreover, these patients showed improved OS and

    75 % of them experienced a high tumor shrinkage level (between 16 - 76 % TS) at 12 weeks

    after the beginning of treatment. Although there is a very important implication to patient

    selection for antiangiogenic therapy, the results of this study are preliminary and need to be

    further validated.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    17

    Disclosure of Potential Conflicts of Interest

    No potential conflicts of interest were disclosed.

    Authors' Contributions

    Conception and design: A. Franzini, M. H. Brutsche

    Development of methodology: A. Franzini, M. H. Brutsche

    Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Franzini, I. I. Macovei, C. Droege, D. Betticher, F. Zappa

    Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Franzini, F. Baty, I. I. Macovei, O. Dürr, D. Klingbiel, M. H. Brutsche

    Writing, review, and/or revision of the manuscript: A. Franzini, F. Baty, B. D. Grigoriu, D. Klingbiel, M. H. Brutsche

    Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Franzini, B. D. Grigoriu, C. Droege, D. Betticher, F. Zappa, M. H. Brutsche

    Study supervision: A. Franzini, M. H. Brutsche

    Supplementary Information

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

    Grant Support

    This work was supported by the Swiss Cancer League & Swiss Cancer Center (KLS-2880-02-2012), KSSG Medical Research Center (MFZF_2014_001) and Romanian–Swiss Research Program (IZERZO_142235/1).

    Acknowledgements

    We thank all the investigators involved in the SAKK 19/05 trial for collecting the samples and SAKK for funding the Affymetrix arrays.

    References

    1. Horimoto Y, Polanska UM, Takahashi Y, Orimo A. Emerging roles of the tumor-associated stroma in promoting tumor metastasis. Cell Adh Migr 2012;6:193-202. 2. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74. 3. Brabek J, Mierke CT, Rosel D, Vesely P, Fabry B. The role of the tissue microenvironment in the regulation of cancer cell motility and invasion. Cell Commun Signal 2010;8:22. 4. McMillin DW, Negri JM, Mitsiades CS. The role of tumour-stromal interactions in modifying drug response: challenges and opportunities. Nat Rev Drug Discov 2013;12:217-28. 5. Meads MB, Gatenby RA, Dalton WS. Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat Rev Cancer 2009;9:665-74. 6. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353-64.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    18

    7. Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 2011;17:1359-70. 8. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer 2008;8:579-91. 9. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004;3:391-400. 10. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335-42. 11. Escudier B, Bellmunt J, Negrier S, Bajetta E, Melichar B, Bracarda S, et al. Phase III trial of bevacizumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol 2010;28:2144-50. 12. Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011;365:2484-96. 13. Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 2009;27:740-5. 14. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 2008;8:592-603. 15. Dang DT, Chun SY, Burkitt K, Abe M, Chen S, Havre P, et al. Hypoxia-inducible factor-1 target genes as indicators of tumor vessel response to vascular endothelial growth factor inhibition. Cancer Res 2008;68:1872-80. 16. Rapisarda A, Melillo G. Overcoming disappointing results with antiangiogenic therapy by targeting hypoxia. Nat Rev Clin Oncol 2012;9:378-90. 17. Ebos JM, Kerbel RS. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol 2011;8:210-21. 18. Miles DW, Chan A, Dirix LY, Cortes J, Pivot X, Tomczak P, et al. Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2010;28:3239-47. 19. Robert NJ, Dieras V, Glaspy J, Brufsky AM, Bondarenko I, Lipatov ON, et al. RIBBON-1: randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer. J Clin Oncol 2011;29:1252-60. 20. Gyanchandani R, Kim S. Predictive biomarkers to anti-VEGF therapy: progress toward an elusive goal. Clin Cancer Res 2013;19:755-7. 21. Baty F, Rothschild S, Fruh M, Betticher D, Droge C, Cathomas R, et al. EGFR exon-level biomarkers of the response to bevacizumab/erlotinib in non-small cell lung cancer. PLoS One 2013;8:e72966. 22. Baty F, Facompre M, Kaiser S, Schumacher M, Pless M, Bubendorf L, et al. Gene profiling of clinical routine biopsies and prediction of survival in non-small cell lung cancer. Am J Respir Crit Care Med 2010;181:181-8. 23. Edlund K, Lindskog C, Saito A, Berglund A, Ponten F, Goransson-Kultima H, et al. CD99 is a novel prognostic stromal marker in non-small cell lung cancer. Int J Cancer 2012;131:2264-73. 24. Larsen AK, Ouaret D, El Ouadrani K, Petitprez A. Targeting EGFR and VEGF(R) pathway cross-talk in tumor survival and angiogenesis. Pharmacol Ther 2011;131:80-90. 25. Maity A, Pore N, Lee J, Solomon D, O'Rourke DM. Epidermal growth factor receptor transcriptionally up-regulates vascular endothelial growth factor expression in human glioblastoma cells via a pathway involving phosphatidylinositol 3'-kinase and distinct from that induced by hypoxia. Cancer Res 2000;60:5879-86. 26. Fong GH. Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis 2008;11:121-40. 27. Zappa F, Droege C, Betticher D, von Moos R, Bubendorf L, Ochsenbein A, et al. Bevacizumab and erlotinib (BE) first-line therapy in advanced non-squamous non-small-cell lung cancer (NSCLC) (stage

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    19

    IIIB/IV) followed by platinum-based chemotherapy (CT) at disease progression: a multicenter phase II trial (SAKK 19/05). Lung Cancer 2012;78:239-44. 28. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003;4:249-64. 29. Bair E, Tibshirani R. Semi-supervised methods to predict patient survival from gene expression data. PLoS Biol 2004;2:E108. 30. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 2005;102:15545-50. 31. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001;25:402-8. 32. Masiero M, Simoes FC, Han HD, Snell C, Peterkin T, Bridges E, et al. A core human primary tumor angiogenesis signature identifies the endothelial orphan receptor ELTD1 as a key regulator of angiogenesis. Cancer Cell 2013;24:229-41. 33. Buffa FM, Harris AL, West CM, Miller CJ. Large meta-analysis of multiple cancers reveals a common, compact and highly prognostic hypoxia metagene. Br J Cancer 2010;102:428-35. 34. Zhu C-Q, Tsao M-S. Prognostic markers in lung cancer: is it ready for prime time? Transl Lung Cancer Res 2014;3:149-58. 35. Yang MY, Hilton MB, Seaman S, Haines DC, Nagashima K, Burks CM, et al. Essential regulation of lung surfactant homeostasis by the orphan G protein-coupled receptor GPR116. Cell Rep 2013;3:1457-64. 36. Ueno M, Igarashi K, Kimura N, Okita K, Takizawa M, Nobuhisa I, et al. Endomucin is expressed in embryonic dorsal aorta and is able to inhibit cell adhesion. Biochem Biophys Res Commun 2001;287:501-6. 37. Vlahakis NE, Young BA, Atakilit A, Hawkridge AE, Issaka RB, Boudreau N, et al. Integrin alpha9beta1 directly binds to vascular endothelial growth factor (VEGF)-A and contributes to VEGF-A-induced angiogenesis. J Biol Chem 2007;282:15187-96. 38. Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, et al. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest 2001;108:689-701. 39. Argraves KM, Gazzolo PJ, Groh EM, Wilkerson BA, Matsuura BS, Twal WO, et al. High density lipoprotein-associated sphingosine 1-phosphate promotes endothelial barrier function. J Biol Chem 2008;283:25074-81. 40. Gaengel K, Niaudet C, Hagikura K, Lavina B, Muhl L, Hofmann JJ, et al. The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between VE-cadherin and VEGFR2. Dev Cell 2012;23:587-99. 41. Kim C, Yang H, Fukushima Y, Saw PE, Lee J, Park JS, et al. Vascular RhoJ is an effective and selective target for tumor angiogenesis and vascular disruption. Cancer Cell 2014;25:102-17. 42. Hossain MN, Sakemura R, Fujii M, Ayusawa D. G-protein gamma subunit GNG11 strongly regulates cellular senescence. Biochem Biophys Res Commun 2006;351:645-50. 43. Baty F, Bihl MP, Perriere G, Culhane AC, Brutsche MH. Optimized between-group classification: a new jackknife-based gene selection procedure for genome-wide expression data. BMC Bioinformatics 2005;6:239. 44. Doherty JR, Cleveland JL. Targeting lactate metabolism for cancer therapeutics. J Clin Invest 2013;123:3685-92. 45. Ben Sahra I, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, et al. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res 2011;71:4366-72. 46. Girard E, Strathdee C, Trueblood E, Queva C. Macrophage migration inhibitory factor produced by the tumour stroma but not by tumour cells regulates angiogenesis in the B16-F10 melanoma model. Br J Cancer 2012;107:1498-505.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    20

    47. Nikitenko LL, Fox SB, Kehoe S, Rees MC, Bicknell R. Adrenomedullin and tumour angiogenesis. Br J Cancer 2006;94:1-7. 48. Harrison L, Blackwell K. Hypoxia and anemia: factors in decreased sensitivity to radiation therapy and chemotherapy? Oncologist 2004;9 Suppl 5:31-40. 49. Kim ES, Herbst RS, Wistuba, II, Lee JJ, Blumenschein GR, Jr., Tsao A, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov 2011;1:44-53. 50. Liu LP, Ho RL, Chen GG, Lai PB. Sorafenib inhibits hypoxia-inducible factor-1alpha synthesis: implications for antiangiogenic activity in hepatocellular carcinoma. Clin Cancer Res 2012;18:5662-71.

    Figure legends:

    Figure 1: Treatment course for patients with no prior therapy included in the SAKK 19/05 phase II trial, study endpoints and microenvironment compartments analyzed by GSEA; TTP ─ me to progression, TS ─ tumor shrinkage, OS ─ overall survival.

    Figure 2: Enrichment plot from GSEA shows statistically significant enrichment of the angiogenesis-associated genes (P=0.004) (A) and in the hypoxia-response genes (P=0.001) (B) in the gene set predictive of TTP under BE. Top 12-ranked angiogenesis-associated genes (C) and hypoxia-response genes (D) derived from GSEA.

    Figure 3: Hierarchical clustering of top 10-ranked angiogenesis-associated genes (A) and hypoxia-response genes (B) significantly associated with TTP under BE showing gene expression variation among patients. The heat maps represent relative intensity values of gene expression levels. Kaplan–Meier TTP under BE curves of the three patients groups (low-risk, medium-risk and high-risk) defined by the 10-gene angiogenesis-associated signature, P = 0.013 for Cluster A1 vs. Cluster A2, (C) and two patient groups (low-risk and high-risk) defined by the 10-gene hypoxia-response signature, P = 0.016 (D). The P values of these associations were determined by log-rank test.

    Figure 4: Angiogenesis and hypoxia-associated gene signatures associate with tumor shrinkage in NSCLC. Enrichment plot from GSEA shows statistically significant enrichment of the angiogenesis-associated genes (P=0.002) (A) and in the hypoxia-response genes (P=0.038) (B) in the gene signature correlating with TS. Top 12-ranked angiogenesis-associated genes (C) and hypoxia-response (D) derived from GSEA, which correlate with TS. Correlation between the two patients groups defined by the 10-gene angiogenesis-associated signature (E) and hypoxia-response signature (F) (high-risk and low-risk; n = 28) and tumor shrinkage at 12 weeks after BE treatment (box plots). The boxes represent the median ± interquartile range (IQR). Whiskers delimit the highest and lowest non-outlier data points (defined as greater/less than 1.5 × IQR).

    Figure 5: Angiogenesis and hypoxia-associated gene signatures predicting OS in NSCLC. Enrichment plot from GSEA shows statistically significant enrichment of the angiogenesis-associated genes (P = 0.031), (A) and hypoxia-response genes (P = 0.001) (B) in the gene signature predictive of OS. Top 12-ranked angiogenesis-associated genes (C) and top 12-

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Gene signatures predictive of response to therapy in NSCLC

    21

    ranked hypoxia-response genes (D) derived from GSEA, which significantly correlate with OS. Kaplan–Meier OS curves of the two patients groups (low-risk and high-risk) defined by the 10-gene angiogenesis-associated signature (P = 0.035) (E) and 10-gene hypoxia signature (P = 0.001) (F). The P values of the associations were determined by log-rank test.

    Figure 6: Comparison between microarray and RT-qPCR GELs. Spearman's correlations were performed between relative gene expression levels (GELs) determined by RT-qPCR (x-axis) and RNA microarray (y-axis) for nine angiogenesis-associated genes for 40 out of the 42 patients. The mRNA levels for each gene of interest were determined by RT qPCR and correlated with microarray expression scores determined after data processing, both calculated relative to HPRT1 gene. r, Spearman’s rank correlation coefficient.

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Bevacizumab/Erlotinib (BE) therapy

    n × 3 weeks until progression

    Endpoints: TS TTP under BE OS

    Standard chemotherapy (CT)

    6 × 6 weeks or until progression

    Biopsies from patients with stage

    IIIB or IV non-squamous

    NSCLC with no prior therapy

    Compartments: Angiogenesis Hypoxia

    12 weeks

    Figure 1

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • 0.1

    0.3

    0.5

    Angiogenesis

    Enri

    chm

    en

    t sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    45

    Rank in Ordered Dataset

    Ranked lis

    t m

    etr

    ic

    0.1

    0.3

    0.5

    Enri

    chm

    en

    t sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    45

    Rank in Ordered Dataset

    Ranked lis

    t m

    etr

    ic

    HypoxiaA BFigure 2

    C DGene symbol Rank in gene list Rank metric score Enrichment score (ES)

    GPR116 403 1.924 0.027

    EMCN 611 1.700 0.060

    ITGA9 648 1.667 0.103

    LDB2 707 1.629 0.143

    MEF2C 805 1.561 0.179

    GNG11 829 1.546 0.219

    CALCRL 865 1.522 0.257

    KDR 987 1.455 0.289

    PECAM1 1019 1.435 0.326

    S1PR1 1117 1.391 0.357

    JAM2 1288 1.313 0.382

    RHOJ 1364 1.289 0.412

    Gene symbol Rank in gene list Rank metric score Enrichment score (ES)

    DDIT4 18 3.539 0.082

    MIF 33 3.276 0.157

    PFKP 53 2.991 0.226

    ADM 69 2.835 0.291

    LDHA 132 2.513 0.346

    GPI 137 2.494 0.404

    ALDOA 238 2.211 0.450

    ACOT7 373 1.975 0.488

    SLC25A32 976 1.460 0.487

    PGK1 1110 1.394 0.512

    TUBA1B 1238 1.331 0.535

    SLC2A1 1420 1.268 0.554Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • 002

    078

    102

    055

    103

    056

    094

    060

    070

    091

    090

    058

    077

    051

    080

    065

    038

    099

    057

    063

    088

    064

    082

    083

    097

    081

    096

    098

    074

    095

    049

    093

    069

    068

    067

    076

    023

    101

    061

    084

    075

    087

    CALCRL

    EMCN

    GNG11

    GPR116

    PECAM1

    ITGA9

    MEF2C

    S1PR1

    LDB2

    KDR

    −2 0 2

    Row Z−Score

    Color Key

    Cluster A1 Cluster A2 Cluster A3

    Angiogenesis

    103

    102

    094

    093

    002

    078

    058

    083

    082

    077

    023

    060

    074

    056

    099

    055

    084

    098

    080

    091

    038

    096

    057

    063

    097

    061

    051

    049

    075

    095

    081

    067

    101

    070

    076

    087

    068

    065

    090

    088

    064

    069

    SLC25A32

    PFKP

    ACOT7

    DDIT4

    ADM

    PGK1

    LDHA

    ALDOA

    MIF

    GPI

    −2 0 2 4

    Row Z−Score

    Color Key

    Cluster H1 Cluster H2

    Hypoxia

    0 2 4 6 8 10 12 14

    0

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    Time (months)

    Pro

    po

    rtio

    no

    fp

    ati

    en

    tsw

    ith

    ou

    tp

    rog

    resio

    n Cluster H1

    Cluster H2

    Cluster H2 18 17 13 10 7 2 1 0

    Cluster H1 24 15 9 3 2 2 1 0

    Patients at risk:

    A B

    C D

    Figure 3

    0 2 4 6 8 10 12 14

    0

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    Time (months)

    Pro

    po

    rtio

    no

    fp

    ati

    en

    tsw

    ith

    ou

    tp

    rog

    resio

    n Cluster A1

    Cluster A2

    Cluster A3

    Cluster A1 9 9 7 5 4 2 1 0

    Cluster A2 12 6 3 2 1 0 0 0

    Cluster A3 21 17 12 6 4 2 1 0

    Patients at risk:

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • 0.1

    0.3

    0.5

    En

    rich

    me

    nt sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    4

    Rank in Ordered Dataset

    Ra

    nke

    d lis

    t m

    etr

    ic

    Angiogenesis

    0.1

    0.2

    0.3

    0.4

    0.5

    En

    rich

    me

    nt sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    4

    Rank in Ordered Dataset

    Ra

    nke

    d lis

    t m

    etr

    ic

    Hypoxia

    −20

    0

    20

    40

    60

    80

    Patients at risk (metagene score)

    TS

    (%)

    FALSE. TRUE.

    18 10 −20

    0

    20

    40

    60

    80

    Patients at risk (metagene score)

    TS

    (%)

    FALSE. TRUE.

    18 10

    A B

    C

    E

    D

    F

    Figure 4

    Gene symbol Rank in gene list Rank metric score Enrichment score (ES)

    LDB2 622 1.818 0.010

    VWF 664 1.785 0.053

    MYCT1 669 1.784 0.098

    TEK 700 1.763 0.141

    EMCN 734 1.745 0.183

    CDH5 749 1.735 0.227

    TIE1 788 1.716 0.268

    JAM2 1072 1.559 0.291

    ZNF423 1139 1.533 0.326

    ROBO4 1197 1.506 0.361

    ADCY4 1398 1.425 0.386

    PTPRB 1690 1.328 0.402

    Gene symbol Rank in gene list Rank metric score Enrichment score (ES)

    MRPL15 61 2.960 0.090

    PSMA7 82 2.836 0.179

    ANKRD37 170 2.465 0.252

    MIF 228 2.285 0.321

    LDHA 473 1.951 0.368

    GPI 605 1.829 0.418

    TUBA1B 1212 1.501 0.430

    YKT6 1522 1.382 0.456

    PFKP 2118 1.206 0.459

    ACOT7 3154 0.991 0.429

    ALDOA 3247 0.975 0.455

    PGK1 3600 0.922 0.463

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • 0.1

    0.3

    0.5

    En

    rich

    me

    nt sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    45

    Rank in Ordered Dataset

    Ra

    nke

    d lis

    t m

    etr

    ic

    Angiogenesis

    0.0

    0.2

    0.4

    0.6

    En

    rich

    me

    nt sco

    re (

    ES

    )

    0 5000 10000 15000

    01

    23

    45

    Rank in Ordered Dataset

    Ra

    nke

    d lis

    t m

    etr

    ic

    Hypoxia

    0 6 12 18 24 300

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    Time (months)

    Cu

    mu

    lati

    ve

    su

    rviv

    al(%

    )

    Low risk

    High risk

    Low risk 21 18 13 8 4 3 2

    High risk 21 17 12 6 4 2 1

    Patients at risk:

    0 6 12 18 24 300

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    Time (months)

    Cu

    mu

    lati

    ve

    su

    rviv

    al(%

    )

    Low risk

    High risk

    Low risk 21 19 16 12 5 2 1

    High risk 21 14 10 3 2 2 2

    Patients at risk:

    A B

    C D

    E F

    Figure 5

    Gene symbol Rank in gene list Rank metric score Enrichment score (ES)

    GPR116 504 1.798 0.030

    MYCT1 811 1.563 0.064

    ITGA9 1228 1.346 0.084

    RHOJ 1369 1.297 0.118

    KDR 2137 1.059 0.108

    CDH5 2260 1.031 0.135

    GNG11 2354 1.009 0.163

    CALCRL 2379 1.004 0.195

    LDB2 2485 0.987 0.222

    ESAM 2601 0.962 0.247

    JAM2 3091 0.879 0.247

    ROBO4 3220 0.859 0.268

    Gene symbol Rank in gene list Rank metric score Enrichment score (ES)

    MIF 100 2.699 0.061

    ALDOA 265 2.177 0.105

    MRPL15 290 2.124 0.156

    LDHA 333 2.059 0.204

    ADM 387 1.957 0.249

    GPI 388 1.955 0.297

    DDIT4 442 1.880 0.341

    ACOT7 466 1.844 0.385

    SHCBP1 757 1.598 0.407

    PFKP 931 1.489 0.434

    PSMA7 977 1.460 0.467

    NDRG1 1307 1.320 0.480

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • 0 5 10 150.0

    0.5

    1.0

    1.5

    2.0

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.6437

    P < 0.0001

    GNG11

    0.0 0.2 0.4 0.6 0.80.0

    0.5

    1.0

    1.5

    2.0

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.7032

    P < 0.0001

    EMCN

    0.0 0.5 1.0 1.50.8

    1.0

    1.2

    1.4

    1.6

    1.8

    2.0

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.6283

    P < 0.0001

    ITGA9

    0.0 0.1 0.2 0.30.0

    0.5

    1.0

    1.5

    2.0

    2.5

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.7131

    P < 0.0001

    KDR

    0 2 4 6 8 100.0

    0.5

    1.0

    1.5

    2.0

    2.5

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.6534

    P < 0.0001

    PECAM1

    0 1 2 3 40.0

    0.5

    1.0

    1.5

    2.0

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.5102

    P = 0.0008

    S1PR1

    0.0 0.5 1.0 1.5 2.00.0

    0.5

    1.0

    1.5

    2.0

    2.5

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.7030

    P < 0.0001

    JAM2

    0.00 0.02 0.04 0.06 0.08 0.100.0

    0.5

    1.0

    1.5

    2.0

    2.5

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.6588

    P < 0.0001

    RHOJ

    0.0 0.5 1.0 1.5 2.00.0

    0.5

    1.0

    1.5

    2.0

    2.5

    GELs, RT-qPCR/HPRT1

    GE

    L,

    mic

    roa

    rra

    y/H

    PR

    T1

    r = 0.6912

    P < 0.0001

    GPR116Figure 6

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst April 28, 2015.Clin Cancer Res Anca Franzini, Florent Baty, Ina I Macovei, et al. lung cancer patients (SAKK 19/05 trial)therapeutic benefit in advanced non-squamous non-small cell Gene expression signatures predictive of bevacizumab/erlotinib

    Updated version

    10.1158/1078-0432.CCR-14-3135doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2015/04/29/1078-0432.CCR-14-3135.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

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

    .http://clincancerres.aacrjournals.org/content/early/2015/04/28/1078-0432.CCR-14-3135To request permission to re-use all or part of this article, use this link

    Research. on June 12, 2021. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 28, 2015; DOI: 10.1158/1078-0432.CCR-14-3135

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-14-3135http://clincancerres.aacrjournals.org/content/suppl/2015/04/29/1078-0432.CCR-14-3135.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2015/04/28/1078-0432.CCR-14-3135http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure4Figure 5Figure 6