This invention relates generally to EMT signatures and predictive markers for successful drug therapy, and more particularly, gene expression signatures and markers useful for characterizing the status of epithelial cancers and for predicting drug responses in patients having non-small cell lung cancer.
This application claims the benefit of and priority in U.S. Patent Application Ser. Nos. 61/470,625 filed on Apr. 1, 2011 and 61/472,098 filed Apr. 5, 2011. The applications are herein incorporated by reference.
None.
None.
Epithelial-mesenchymal transition (“EMT”) has been associated with metastatic spread and EGFR inhibitor resistance. However, currently, there is no standard method for assessing EMT. Hence, there is an unmet need for therapeutic strategies targeting mesenchymal cells and overcoming EMT-associated drug resistance. Furthermore, to date, EGFR mutation is the only validated marker for identifying and predicting a benefit in patients with wild type EGFR mutation in non-small cell lung cancer.
Signatures and biomarkers are needed to select patients that will experience greater benefit from a specific treatment regimen for non-small cell lung cancer and other cancers, potentially sparing patients who are less likely to benefit from receiving toxic therapy.
Epithelial-mesenchymal transition (“EMT”) gene expression signatures are provided herein. These signatures are useful for characterizing the status of epithelial cancers and for predicting certain drug responses in patients having non-small cell lung cancer (“NSCLC”). The gene signatures as well as certain individual biomarkers disclosed herein can be used to identify which NSCLC patients may benefit from certain drug treatments. The signatures may also be useful for predicting response to EGFR inhibitors in NSCLC as well as other tumor types. In addition, EGFR mutations could be used in conjunction with these EMT signatures and other biomarkers (sometimes referred to herein as “markers”) to identify patients at greater risk for relapse or metastatic spread after definitive (e.g. surgery, radiation) therapy.
As taught herein, we confirmed that certain signatures are associated with shorter progression and overall survival. These signatures together with other markers could be useful for improving the selection of patients likely to respond to a given treatment, particularly for NSCLC patients treated with EGFR inhibitors. The signatures also may be used for selecting patients to receive cisplatin-based chemotherapy.
The EMT signatures presented herein were developed using non-small cell lung cancer cell lines. These signatures been have validated using independent gene expression platforms, for NSCLC lines and head and neck cell lines. Clinical validation was performed using several clinical datasets including the BATTLE study, which confirmed the signature is as a marker of erlotinib resistance, and a set of head and neck patients who received PORT (“post-operative radiotherapy”).
The EMT gene expression signatures disclosed herein can also accurately classify cell lines as epithelial or mesenchymal-like across microarray platforms and several cancer types. Furthermore, as taught herein Axl and LCN2 have been identified as a novel EMT markers in NSCLC and Head and Neck Cancer (“HNC”). Hence, the EMT signature is a reliable predictor of erlotinib resistance and is more accurate than single mRNA or protein markers such as E-cadherin.
Epithelial-mesenchymal transition (“EMT”) is a biological program observed in several epithelial cancers including non-small lung cancer cells (“NSCLC”). EMT is associated with loss of cell adhesion molecules such as E-cadherin and increased invasion, migration, and proliferation in epithelial cancers. Huber M. A., et al., Molecular Requirements for Epithelial-Mesenchymal Transition During Tumor Progression, Curr Opin Cell Biol. 17:548-58 (2005); Thiery J. P., Epithelial-Mesenchymal Transitions in Tumour Progression. Nature Rev. 2:442-54 (2002); Thiery J. P., et al., Epithelial-Mesenchymal Transitions in Development and Disease, Cell 139:871-90 (2009); Hugo H., et al., Epithelial-Mesenchymal and Mesenchymal-Epithelial Transitions in Carcinoma Progression, J Cell Physiol. 213:374-83 (2007).
Previous profiling and mutational analyses have demonstrated the molecular heterogeneity of non-small cell lung cancer. For EGFR mutant and EML4-ALK fusion subgroups, mutation status predicts response to therapy with EGFR inhibitors or ALK inhibitors, respectively. Unfortunately only a minority of patients express these markers, with EGFR mutations detected in ˜10-15% of lung adenocarcinomas and EML4-ALK fusions in ˜4%. Koivunen, J. P., et al., EML4-ALK Fusion Gene and Efficacy of an ALK Kinase Inhibitor in Lung Cancer, Clin Cancer Res. 14:4275-83 (2008); Pao, W., et al., EGF Receptor Gene Mutations are Common in Lung Cancers From “Never Smokers” and are Associated With Sensitivity Of Tumors To Gefitinib And Erlotinib, Proc Natl Acad Sci USA 101:13306-11 (2004); Lynch, T. J., et al., Activating Mutations In The Epidermal Growth Factor Receptor Underlying Responsiveness Of Non-Small-Cell Lung Cancer To Gefitinib, N Engl J Med. 350:2129-39 (2004); Paez, J. G., et al., EGFR Mutations in Lung Cancer: Correlation With Clinical Response To Gefitinib Therapy, Science 304:1497-500 (2004); Tokumo, M., et al., The Relationship Between Epidermal Growth Factor Receptor Mutations And Clinicopathologic Features In Non-Small Cell Lung Cancers, Clin Cancer Res. 11:1167-73 (2005); Cappuzzo, F., et al., Epidermal Growth Factor Receptor Gene and Protein and Gefitinib Sensitivity In Non-Small-Cell Lung Cancer, J Natl Cancer Inst. 97:643-55 (2005); Soda, M., et al., Identification of the Transforming EML4-ALK Fusion Gene in Non-Small-Cell Lung Cancer, Nature 448:561-6 (2007).
For the majority of patients with wild-type EGFR, only a certain subgroup appears to benefit from EGFR inhibitor treatment. However, prior to the present discoveries, there were no validated markers for identifying these patients. Bell D. W., et al., Epidermal Growth Factor Receptor Mutations and Gene Amplification in Non-Small-Cell Lung Cancer: Molecular Analysis of the IDEAL/INTACT Gefitinib Trials, J Clin Oncol. 23:8081-92 (2005); Zhu C. Q., et al., Role of KRAS and EGFR as Biomarkers of Response to Erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21, J Clin Oncol. 26:4268-75 (2008); Mok T. S., et al., Gefitinib or Carboplatin-Paclitaxel in Pulmonary Adenocarcinoma, N Engl J Med. 361:947-57 (2009).
Thus, presented herein are gene expression signatures and other validated predictive markers to accurately predict response to EGFR-targeted therapy in patients with wild-type EGFR mutation status, as well as for other targeted therapies, and that can help identify potential strategies for improving the efficacy of these agents.
As used herein, gene expression signatures are sometimes referred to herein as “signatures,” “gene signatures,” “EMT gene signatures,” “signature genes” “EMT signature genes” or “EMT signatures,” or, in the singular as a “signature,” “gene signature,” “EMT gene signature,” “signature gene” “EMT signature gene” or “EMT signature.”
Mesenchymal markers have been associated with limited responses to EGFR inhibitors, whereas an epithelial phenotype is associated with response even in patients without EGFR receptor mutations. Yauch R. L., et al., Epithelial Versus Mesenchymal Phenotype Determines In Vitro Sensitivity and Predicts Clinical Activity of Erlotinib in Lung Cancer Patients, Clin Cancer Res. 11:8686-98 (2005); Thomson S., et al., Epithelial to Mesenchymal Transition is a Determinant of Sensitivity of Non-Small-Cell Lung Carcinoma Cell Lines and Xenografts to Epidermal Growth Factor Receptor Inhibition, Cancer Res. 65:9455-62 (2005); Frederick B. A., et al., Epithelial to Mesenchymal Transition Predicts Gefitinib Resistance in Cell Lines of Head and Neck Squamous Cell Carcinoma and Non-Small Cell Lung Carcinoma, Mol Cancer Ther. 6:1683-91 (2007); Nikolova D. A., et al., Cetuximab Attenuates Metastasis and U-PAR Expression in Non-Small Cell Lung Cancer: U-PAR and E-Cadherin are Novel Biomarkers of Cetuximab Sensitivity, Cancer Res. 69:2461-70 (2009).
For example, high E-cadherin and low vimentin/fibronectin (i.e., an epithelial phenotype) has been associated with erlotinib sensitivity in cell lines and xenografts with wild-type EGFR. Thomson S., et al., Epithelial to Mesenchymal Transition is a Determinant of Sensitivity of Non-Small-Cell Lung Carcinoma Cell Lines and Xenografts to Epidermal Growth Factor Receptor Inhibition, Cancer Res. 65:9455-62 (2005). Clinically, E-cadherin protein expression has been associated with longer time to progression and a trend toward longer overall survival following combination erlotinib/chemotherapy. Yauch R. L., et al., Epithelial Versus Mesenchymal Phenotype Determines In Vitro Sensitivity and Predicts Clinical Activity of Erlotinib in Lung Cancer Patients, Clin Cancer Res. 11:8686-98 (2005). The ability to identify tumors that have not undergone EMT may help identify patients most likely to benefit from EGFR inhibition, particularly in patients with wild type EGFR. In addition, targeting EMT or EMT-associated resistance pathways may reverse or prevent acquisition of EGFR inhibitor resistance, as illustrated by one study in which restoration of an epithelial phenotype in mesenchymal NSCLC cell lines restored sensitivity to the EGFR inhibitor gefitinib. Witta S. E., et al., Restoring E-Cadherin Expression Increases Sensitivity to Epidermal Growth Factor Receptor Inhibitors in Lung Cancer Cell Lines, Cancer Res. 66:944-50 (2006). Although a number of markers have been associated with EMT and EMT signatures have been described in other cancer types, there is no validated signature in NSCLC that can identify tumors that have undergone EMT.
In non-small cell lung cancer (“NSCLC”), EMT is associated with worse prognosis and resistance to EGFR inhibitors. Despite the clinical implications, no gold standard exists for classifying a cancer as epithelial or mesenchymal. Our goal was to develop robust, platform-independent EMT gene expression signatures and test the correlation of these signatures with drug response.
In one aspect, we conducted analysis of an integrated gene expression, proteomic, and drug response using cell lines and tumors from non-small cell lung cancer patients. A 76-gene EMT signature was developed and validated using gene expression profiles from four microarray platforms of NSCLC cell lines and patients treated in the BATTLE (“Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination”) study, and potential therapeutic targets associated with EMT were identified.
We found mesenchymal cells demonstrated significantly greater resistance to EGFR and PI3K/Akt pathway inhibitors, independent of EGFR mutation status, but not to sorafenib. Mesenchymal cells expressed increased levels of the receptor tyrosine kinase Axl and showed a trend towards greater sensitivity to the Axl inhibitor SGI-7079. The combination of SGI-7079 with erlotinib reversed erlotinib resistance in mesenchymal lines expressing Axl.
In NSCLC patients with non-mutated EGFR, the EMT signature predicted 8-week disease control in patients receiving erlotinib, but not other therapies. See,
Specifically, as set out in Example 1 below, to better characterize EMT and its association with drug response in NSCLC, we performed an integrated analysis of gene expression profiling from several microarray platforms as well as high-throughput functional proteomic profiling. See generally,
Cell Lines.
NSCLC cell lines were established by John D. Minna and Adi Gazdar (20, 21) or obtained through ATCC and grown in RPMI-1640 plus 10% FBS. Identities were confirmed by DNA fingerprinting.
Selection of Single Best EMT Marker Probes.
Because the NSCLC cell line panel was profiled on both Affymetrix and Illumina microarray platforms, we were able to select the single best Affymetrix probe sets for CDH1, VIM, CDH2, and FN1 on the basis of their correlations with other Affymetrix probes and Illumina WG v2 probes for the same gene transcript (
For N-cadherin (CDH2), Aff 203440_at and Aff 203411_s_at were highly correlated (r=0.802). Aff 203440_at was selected for the analysis because of its better correlation with the Illumina CDH2 probe (r=0.904 versus 0.730). Fibronectin (FN1) probe set 210495_x_at was selected from among four good Affymetrix probe sets because it had the highest correlation with the Illumina FN1 probes. Although the Affymetrix arrays include only one probe set for vimentin (VIM) (201426_s_at), measurements from that set correlated well (r=0.958) with that from the Illumina WGv2 VIM probe set (III 50671). The Affymetrix probe was therefore considered to be an accurate measure of VIM transcript expression.
Once the best probes were selected, EMT signature genes were selected based on their correlation with the four EMT genes (absolute r-value ≧0.65 for CDH1 and VIM, ≧0.52 for CDH2 and FN1) and their bimodal distribution across the training set, as described in results. By limiting the EMT signature to genes expressed among the cell lines at either relatively high or low levels, but not in between, we expected to increase the likelihood that the signature could separate patient tumors into distinct epithelial and mesenchymal groups. Hierarchical clustering and Principal Component Analysis (PCA) algorithms were used on mRNA expression data to evaluate the EMT signature.
Expression Profiling of Cell Lines.
Affymetrix microarray results were previously published and archived at the Gene Expression Omnibus repository (http://www.ncbi.nlm.nih.gov/geo/, GEO accession GSE4824). Zhou B. B., et al., Targeting ADAM-Mediated Ligand Cleavage to Inhibit HERS and EGFR Pathways in Non-Small Cell Lung Cancer, Cancer Cell 10:39-50 (2006); Edgar R., et al., Gene Expression Omnibus: NCBI Gene Expression and Hybridization Array Data Repository, Nucleic Acids Res. 30:207-10 (2002); Barrett T., et al., NCBI GEO: Archive for Functional Genomics Data Sets—10 Years On, Nucleic Acids Res. 39:D1005-10. Illumina v2 (GSE32989) and v3 (GSE32036) results have been deposited in the GEO repository. Microarray data was used to derive a platform-independent, 76-gene expression signature was derived as described in Supplemental Methods.
Gene Expression Profiling of BATTLE Tumors.
BATTLE (Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination) was a randomized, biomarker-based clinical trial for patients with recurrent or metastatic NSCLC in the second-line setting (Trial registration ID: NCT00409968). Kim E. S. H. R., The BATTLE Trial: Personalizing Therapy for Lung Cancer, Cancer Discovery 1:43-51 (2011). mRNA from tumors obtained via core-needle biopsy at enrollment were profiled on Human Gene 1.0 ST array, Affymetrix. Array results were deposited in the GEO repository (GSE33072).
Drug Sensitivity of Cell Lines.
For each drug, the concentration required to inhibit 50% growth (IC50) was measured by MTS assay ≧3 times in NSCLC cell lines. Average values were used for analysis as described. Gandhi J., et al., Alterations in Genes of the EGFR Signaling Pathway and Their Relationship to EGFR Tyrosine Kinase Inhibitor Sensitivity in Lung Cancer Cell Lines, PLoS One 4:e4576 (2009). Axl inhibitor SGI-7079 was generated as described in Supplemental Methods. The effect of erlotinib, SGI-7079, or the combination of erlotinib and SGI-7079 on proliferation was assayed using CellTiter-Glo Luminescent Cell Viability kit (Promega), as described. Chou T. C., et al., Quantitative Analysis of Dose-Effect Relationships: The Combined Effects of Multiple Drugs or Enzyme Inhibitors. Adv Enzyme Regul. 22:27-55 (1984); Johnson F. M., et al., Abrogation of Signal Transducer and Activator of Transcription 3 Reactivation After Src Kinase Inhibition Results in Synergistic Antitumor Effects, Clin Cancer Res. 13:4233-44 (2007).
Protein Profiling by Reverse-Phase Protein Array (RPPA) and Western Blot.
RPPA studies were performed as described. Byers L. A., et al., Reciprocal Regulation of C-Src And STATS in Non-Small Cell Lung Cancer, Clin Cancer Res. 15:6852-61 (2009). Protein lysate was collected from sub-confluent cultures after 24 hours in complete medium. RPPA slides were printed from lysates. Immunostaining was performed and analyzed, as described in Supplemental Methods. Primary antibodies included pEGFR (Y1173), pSTAT3 (Y705), pSTAT5 (Y694), pSTAT6 (Y641), pSrc (Y416), and E-cadherin (Cell Signaling); pHer2 (Y1248) (Upstate Biotechnology); Axl (Abcam), and Rab25 (Covance).
Generation and Characterization of AXL Inhibitor SGI-7079.
Purified recombinant AXL kinase was used to screen a library of structures with appropriate drug-like scaffolds to identify potential inhibitors. Hits from the screen were confirmed and r analyzed by selection criteria including Lipinski rules. One pyrrolopyrimidine-based compound was selected for structure-activity relationship efforts. Optimization of this scaffold and subsequent evaluation led to the generation of compound SG1-7079 as the lead candidate inhibitor (
RPPA Data Processing and Statistical Analysis.
MicroVigene software (VigeneTech, Carlisle, Mass.) and an R package developed in house were used to assess spot intensity. Protein levels were quantified by the SuperCurve method (http://bioinformatics.mdanderson.org/OOMPA) as previously described. Hu J., et al., Non-Parametric Quantification of Protein Lysate Arrays, Bioinformatics 23:1986-94 (2007); Nanjundan M., et al., Proteomic Profiling Identifies Pathways Dysregulated in Non-Small Cell Lung Cancer and an Inverse Association of AMPK and Adhesion Pathways With Recurrence, J Thorac Oncol. 5:1894-904 (2010). Data were log-transformed (base 2) and median-control normalized across all proteins within a sample. Differences in protein expression between epithelial and mesenchymal cell lines were compared by t-test. Pearson correlation between E-cadherin protein expression levels and first principal component of the EMT signature derived from mRNA expression data was then assessed. All statistical analyses were performed using R packages (version 2.10.0)
A 76-Gene EMT Signature Classifies NSCLC Cell Lines into Distinct Epithelial and Mesenchymal Groups.
Using a training set of 54 NSCLC cell lines profiled on Affymetrix U133A, U133B, and Plus2.0 arrays, we selected genes for the EMT gene expression signature based on two criteria aimed at increasing the robustness and potential applicability of the signature across different platforms. First, we identified genes whose mRNA expression levels were either positively or negatively correlated with the single best probe for at least one of four putative EMT markers—E-cadherin (CDH1), vimentin (VIM), N-cadherin (CDH2), and/or fibronectin 1 (FN1). For this analysis, the best probe to represent each of the four genes was selected based on its strong correlation with other probes for the same gene within a microarray platform and/or across platforms (see Methods). From that set, we selected only those genes whose mRNA expression followed a bimodal distribution pattern across cell lines (bimodal index >1.5). Wang J., et al., The Bimodality Index: A Criterion for Discovering and Ranking Bimodal Signatures From Cancer Gene Expression Profiling Data, Cancer Inform. 7:199-216 (2009).
indicates data missing or illegible when filed
Table 1 provided immediately below lists the ninety-six probes representing 76 unique bimodally distributed genes that correlated with E-cadherin (CDH1), vimentin (VIM), N-cadherin (CDH2), and/or fibronectin 1 (FN1) were identified in the NSCLC training set. Individual probes are ranked in the table by their correlation with E-cadherin. These probes and the associated information are also provided in
Specifically, as shown in
Cell lines in the mesenchymal group expressed higher levels of genes activated by EMT transcription factors ZEB1/2 and/or SNAIL1/2, including matrix metalloprotease-2 (MMP-2), vimentin, and ZEB1 itself (a target of SNAIL). Miyoshi A., et al., Snail And SIP1 Increase Cancer Invasion by Upregulating MMP Family in Hepatocellular Carcinoma Cells, Br J Cancer 90:1265-73 (2004); Yokoyama K., et al., Increased Invasion and Matrix Metalloproteinase-2 Expression by Snail-Induced Mesenchymal Transition in Squamous Cell Carcinomas, Int J Oncol. 22:891-8 (2003); Cano A., et al., The Transcription Factor Snail Controls Epithelial-Mesenchymal Transitions by Repressing E-Cadherin Expression, Nat Cell Biol. 2:76-83 (2002); Eger A., et al., Deltaefl is a Transcriptional Repressor of E-Cadherin and Regulates Epithelial Plasticity in Breast Cancer Cells, Oncogene 24:2375-85 (2005); Bindels S., et al., Regulation of Vimentin by SIP1 in Human Epithelial Breast Tumor Cells, Oncogene 25:4975-85 (2006); Guaita S., et al., Snail Induction of Epithelial to Mesenchymal Transition in Tumor Cells is Accompanied by MUC1 Repression and ZEB1 Expression, J Biol Chem. 277:39209-16 (2002). AXL, a receptor tyrosine kinase associated with EMT in breast and pancreatic cancer was also highly expressed in mesenchymal NSCLC cells. Gjerdrum C., et al., Axl is an Essential Epithelial-To-Mesenchymal Transition-Induced Regulator of Breast Cancer Metastasis and Patient Survival, Proc Natl Acad Sci USA 107:1124-9 (2010); Vuoriluoto K., et al., Vimentin Regulates EMT Induction by Slug and Oncogenic H-Ras and Migration by Governing Axl Expression in Breast Cancer, Oncogene 30:1436-48 (2011); Koorstra J. B., et al,. The Axl Receptor Tyrosine Kinase Confers an Adverse Prognostic Influence in Pancreatic Cancer and Represents a New Therapeutic Target, Cancer Biol Ther. 8:618-26 (2009).
In contrast, epithelial lines had higher expression of genes repressed by ZEB1 and SNAIL, such as CDH1, RAB25, MUC1, and claudins 4 (CLDN4) and 7 (CLDN7). Cano A., et al., The Transcription Factor Snail Controls Epithelial-Mesenchymal Transitions by Repressing E-Cadherin Expression, Nat Cell Biol. 2:76-83 (2002); Eger A., et al., Deltaefl is a Transcriptional Repressor of E-Cadherin and Regulates Epithelial Plasticity in Breast Cancer Cells, Oncogene 24:2375-85 (2005); Guaita S., et al., Snail Induction of Epithelial to Mesenchymal Transition in Tumor Cells is Accompanied by MUC1 Repression and ZEB1 Expression, J Biol Chem. 277:39209-16 (2002); Battle E., et al., The Transcription Factor Snail is a Repressor of E-Cadherin Gene Expression in Epithelial Tumour Cells, Nat Cell Biol. 2:84-9 (2000); De Craene B., et al., The Transcription Factor Snail Induces Tumor Cell Invasion Through Modulation of the Epithelial Cell Differentiation Program, Cancer Res. 65:6237-44 (2005); Ikenouchi J., et al., Regulation of Tight Junctions During the Epithelium-Mesenchyme Transition: Direct Repression of the Gene Expression of Claudins/Occludin by Snail, J Cell Sci. 116:1959-67 (2003).
The EGFR family member ERBB3 and SPINT2, a regulator of HGF, were also expressed at higher levels in epithelial lines. RAB25, a trafficking protein involved with EGFR recycling, was also strongly correlated with CDH1 expression (r=0.8) and had a high bimodal index (BI=2.88, top 3% of signature genes). Although Rab25 suppression has been described as a marker of EMT in breast cancer, this is the first time to our knowledge that it has been associated with an epithelial (versus mesenchymal) phenotype in NSCLC. Vuoriluoto K., et al., Vimentin Regulates EMT Induction by Slug and Oncogenic H-Ras and Migration by Governing Axl Expression in Breast Cancer, Oncogene 30:1436-48 (2011). As expected, all EGFR-mutant cell lines were classified by the EMT signature as epithelial, including H1975 and H820, which carry the resistance mutation T790M (
Because a major goal of this study was to develop a platform-independent signature, we tested performance of the EMT signature on the Illumina WGv2 microarray platform. As with the Affymetrix platform, distinct differences were observed in the expression of Illumina probes corresponding to the 76 EMT signature genes, as reflected by hierarchical clustering and first principal component analysis (
Next, we performed an integrated proteomic analysis to identify major differences in protein expression between epithelial and mesenchymal cells. Not surprisingly, out of more than 200 proteins and phosphoproteins assayed, E-cadherin differed the most between the groups (p<0.0001 by t-test) with mean E-cadherin levels 7.42-fold higher in epithelial lines, compared to mesenchymal. (
Previously, E-cadherin expression has been associated with greater benefit from erlotinib in NSCLC patients. Yauch R. L., et al., Epithelial Versus Mesenchymal Phenotype Determines In Vitro Sensitivity and Predicts Clinical Activity of Erlotinib in Lung Cancer Patients, Clin Cancer Res. 11:8686-98 (2005); Thomson S., et al., Epithelial to Mesenchymal Transition is a Determinant of Sensitivity of Non-Small-Cell Lung Carcinoma Cell Lines and Xenografts to Epidermal Growth Factor Receptor Inhibition, Cancer Res. 65:9455-62 (2005); Frederick B. A., et al., Epithelial to Mesenchymal Transition Predicts Gefitinib Resistance in Cell Lines of Head and Neck Squamous Cell Carcinoma and Non-Small Cell Lung Carcinoma, Mol Cancer Ther. 6:1683-91 (2007); Nikolova D. A., et al., Cetuxirnab Attenuates Metastasis and U-PAR Expression in Non-Small Cell Lung Cancer: U-PAR and E-Cadherin are Novel Biomarkers of Cetuximab Sensitivity, Cancer Res. 69:2461-70 (2009). Therefore, we tested the association between our EMT signature and cell line sensitivity to erlotinib. Mesenchymal cells were highly resistant to erlotinib, with IC50s 3.7-fold higher in mesenchymal versus epithelial cell lines (p=0.002 by t-test). (
Although cell lines with EGFR activating mutations were among the most sensitive to erlotinib, in the subset with wild-type EGFR and wild-type KRAS, the correlation between EMT signature and erlotinib response was maintained, with significantly greater resistance in mesenchymal lines (p=0.023, 2-fold higher mean IC50 values). Importantly, the EMT signature was a better predictor of erlotinib response than were mRNA probe sets for individual genes such as CDH1 or VIM (
As with EGFR inhibitors, mesenchymal NSCLC cell lines were also more resistant to PI3K/Akt pathway targeting drugs, such as the selective pan PI3K inhibitor GDC0941 (p=0.068, 1.9-fold higher IC50) and 8-amino-adenosine, an adenosine analog that inhibits Akt/mTOR signaling (p=0.003, 1.7-fold higher IC50) (
Because the receptor tyrosine kinase Axl was expressed at higher mRNA and protein levels in mesenchymal cell lines (
We then compared the sensitivity of mesenchymal cells to SGI-7079 versus erlotinib (
In two cell lines with highest Axl protein expression (Calu-1 and H2882), the combination was synergistic at higher concentrations of SGI-7079, possibly reflecting a need for higher dosing in cells with higher target expression levels.
EMT Signature in Patients with Relapsed or Metastatic NSCLC.
Finally, we tested the EMT signature in 139 previously-treated NSCLC patients with advanced NSCLC enrolled in the BATTLE-1 trial (Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination). Kim E. S. H. R., The BATTLE Trial: Personalizing Therapy for Lung Cancer, Cancer Discovery 1:43-51 (2011). Consistent with the cell line data—and despite all patients having advanced, metastatic disease—a majority of patients (approximately 2/3) had epithelial signatures (
EMT is a pervasive process among epithelial cancers that has been linked to morphologic changes, increased invasiveness, and metastatic potential. While a number of EMT markers have been identified, no robust gene signature capable of use across multiple platforms has been established. Furthermore, the mesenchymal phenotype has been linked with resistance to EGFR inhibitors, but it is unknown how EMT affects response to other drugs and effective therapeutic strategies for targeting mesenchymal cells are needed.
To address these needs, we developed and validated a robust, platform-independent gene expression signature capable of classifying NSCLC as epithelial or mesenchymal. The signature was selected using probes with high cross-platform correlations to increase the likelihood that the signature could be applied to different types of mRNA arrays or emerging technologies. The success of this approach was demonstrated in independent testing sets, with essentially identical classification of cell lines profiled on Affymetrix, Illumina v2 and v3 arrays. An integrated analysis of mRNA and proteomic expression confirmed strong correlation of the EMT signature with E-cadherin protein levels. Additionally, higher expression of activated EGFR signaling proteins was observed in epithelial cell lines. Moreover, as predicted, EGFR mutant cells all demonstrated an epithelial signature.
To investigate whether other drugs may preferentially target epithelial or mesenchymal cells we assessed the activity of several targeted drugs used commonly in NSCLC or in current clinical development. Consistent with prior studies, epithelial cells demonstrated greater sensitivity to the EGFR inhibitors erlotinib and gefitinib in vitro, independent of EGFR mutation status, while mesenchymal cells were highly resistant (
Next, we investigated Axl as a potential therapeutic target for the mesenchymal phenotype. We observed higher levels the receptor tyrosine kinase Axl in the mesenchymal phenotype at both the mRNA and protein level (
Finally, we tested the EMT signature in refractory NSCLC patients treated with erlotinib or sorafenib in the BATTLE study. Among erlotinib-treated patients (wild-type EGFR and KRAS), those with 8-week disease control, the primary study endpoint, had a more epithelial phenotype than those who did not have DC control (p=0.05, by t-test) (
This study established a robust, cross-platform EMT signature capable of classifying NSCLC cell lines and patient tumors as epithelial or mesenchymal. Consistent with prior studies, the mesenchymal phenotype is associated with resistance to EGFR inhibitors both in vitro and in patients with wild-type EGFR treated with erlotinib, a subgroup for which there is no established predictive marker. Similarly, we also showed that PI3K/AKT inhibitors are more active in epithelial cells. Finally, we identify Axl as a novel EMT marker in NSCLC and demonstrate that Axl inhibitors are active against cells with a mesenchymal phenotype and can reverse EGFR inhibitor resistance associated in mesenchymal cells. Together these findings suggest that assessment of EMT status may guide drug selection in NSCLC patients and dual Axl/EGFR inhibition may be an effective targeted strategy for overcoming EGFR inhibitor resistance associated with the mesenchymal phenotype. These findings merit further investigation in future clinical trials.
The EMT signature was derived in 54 DNA fingerprinted NSCLC cell lines profiled on Affymetrix U133A, B, and Plus2.0 arrays and tested on the Illumina WGv2 and WGv3 platforms and in an independent set of head and neck cancer lines (HNC). E-cadherin and other protein levels were quantified by reverse phase protein array and correlated with the first principal component of the EMT signature. IC50s were determined for NSCLC cell lines by MTS assay. Response to erlotinib was evaluated in patients treated in the BATTLE clinical trial using eight-week disease free status and progression free survival.
In the original EMT signature, genes were selected based on two criteria. First, they must be correlated with one of four EMT genes (CDH1, VIM, FN1 and CDH2). Second, they must be biomodally distributed. A third requirement was added to improve the signature. The third criteria is that the genes included in the signature come from “good quality” probes-defined as those probes having a correlation between Affymetrix and Illumina platform of r greater than 0.90. This refines the signature to the smallest number of genes with the greatest contribution to the EMT signature.
The classification of each cell line as epithelial or mesenchymal remained the same between the original and the refined signature, suggesting that the refined signature includes the “core EMT genes” contributing most significantly to the EMT signature.
Expression of 35 genes (the EMT signature) correlated with mRNA expression of known EMT markers E-cadherin, vimentin, N-cadherin, or fibronectin 1 and expression was bimodally distributed across the NSCLC panel.
mRNA levels for Axl, a tyrosine kinase receptor associated with EMT in breast cancer, had the most negative correlation with E-cadherin (r=−0.45) of any signature gene after ZEB 1 and vimentin and was positively correlated with vimentin (r=0.60) and N-cadherin (r-0.54) expression. Higher Axl total protein was confirmed in NSCLC and HNC mesenchymal-like cell lines. Classification as mesenchymal by the EMT signature was more strongly correlated with NSCLC erlotinib resistance (p=0.028) than E-cadherin mRNA or protein level. In contrast, an epithelial classification by the EMT signature was associated with improved 8-week disease control and PFS.
A five-gene signature for predicting benefit in patients with non-small cell lung cancer treated with erlotinib is provided herein. (
We conducted an analysis of tissue samples at MDACC from a trial of non-small cell lung cancer patients treated in the BATTLE trial. The analysis was conducted using the Affymetrix gene expression array platform. The five-gene signature was validated in a panel of NSCLC cell lines and predicts clinical response to erlotninib. (
We also investigated markers for identifying patients that would be most likely to benefit from erlotinib in patients with non-small cell lung cancer (NSCLC) treated in the BATTLE program. The Affymetrix platform was used to analyze gene expression from NSCLC patients treated in the BATTLE program. There were a total of 101 patients treated in the following arms: erlotinib (n=27), erlotinib+bexarotene (n=8), vandetinib (n=19) and sorafenib (n=47). A five gene signature that predicts clinical benefit (e.g. disease control) in patients that were EGFR and KRAS widtype was developed and validated in NSCLC cell lines. The genes including in the signature include the following probesets (gene name included if known): 219789_at (NPR3), 219790_s_at, 219054_at (C5orf23), 212531_at (LCN2), 205760_s_at (OGG1), and 205301_s_at. Of these genes, LCN2 has a very strong potential for predicting response to erlotinib on its own.
Despite a low response rate, erlotinib (E) improves survival in a subset of NSCLC patients with EGFR but there are no established markers for identifying patients likely to have clinical benefit.
We used pretreatment gene expression profiles (Affymetrix HG LOST) from 101 chemo-refractory patients in our Biomarkers-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) treated with E, E+bexarotene (EB), sorafenib (S), or vandetanib (V). 24 cases of with EGFR & KRAS tumors treated with E or EB were compared to train the signature (two-sided t-test), using the primary end-point of the trial[8-week disease control (8 with DC)]. Principal component (PC) analysis and a logistic regression model were used to develop the signature. Gene expression profiles from 108 NSCLC cell lines (Illumina), with available E IC50 (N=94) and DNA methylation profiling (N=66, Illumina), were used for in vitro studies.
113 genes were differentially expressed between patients with or without 8wDC (false discovery rate 30%; P=0.004). Leave-one-out cross validation with various gene list lengths produced the 5-gene signature, including lipocalin 2 (LCN2), with a specificity, sensitivity and accuracy of 80% to predict 8 with DC.
In patients treated with E or EB, using the median signature score, the 8 with DC rate in the signature-positive group was 83% compared with 0% in the signature-negative group; the signature did not predict 8wDC in patients treated with S or V (Mantel-Haenszel chi-squared test P=0.023). The improvement in 8 with DC in the signature-positive group translated to an increased progression-free survival (PFS) (hazard ratio=0.12, 95% confidence interval: 0.03-0.46, P=0.001; log-rank P=0.0004; median PFS: 12.5 weeks vs. 7.2 weeks). We tested the signature in an independent set of 47 with EGFR & KRAS cell lines. It predicted E sensitivity with an area under the curve of 78% (P=0.002). The first PC of the signature and the IC50 for E were correlated (r=−0.47, P=0.0009). In 108 NSCLC cell lines, LCN2 gene expression was bimodal and correlated with the IC50 for E (r=−0.46, P=0.001). Degree of methylation and expression level of LCN2 were inversely in with EGFR & KRAS NSCLC cells (r=−0.79, P<0.0001, N=33). Cell lines with completely unmethylated LCN2 were more sensitive to E compared to those with LCN2 full methylation (N=36) (P=0.006); the difference remained significant in with EGFR & KRAS cell lines (P=0.014). As noted above,
We identified a 5-gene signature predictive of PFS benefit in NSCLC patients with EGFR & KRAS treated with E, but not S or V. The signature was also predictive of E sensitivity in vitro. LCN2 was the strongest individual marker of sensitivity and may be epigenetically regulated.
We have discovered that LCN2 is a predictive marker of benefit in patients with non-small cell lung cancer treated with EGFR inhibitors. This discovery could help select patients that will experience greater benefit from a specific treatment regimen for NSCLC and other cancers, and potentially spare patients who are less likely to benefit from receiving toxic therapy.
LCN2 as a biomarker could be used for the purpose of better selecting patients likely to respond to a given treatment, particularly for NSCLC patients treated with erlotinib or other EGFR inhibitor. Subsets of non-small-cell lung cancer (NSCLC) are currently defined in part by mutations in key oncogenic drivers such as EGFR and KRAS. EGFR inhibitors such as erlotinib prolong progression-free survival (PFS) and/or overall survival in previously treated NSCLC patients. Among these patients, the subset bearing EGFR mutations (˜10-15%) have a high likelihood of major objective tumor responses, while those bearing KRAS mutations (˜15-20%) are likely to be resistant to EGFR TKIs.
Patients bearing wild-type (wt) EGFR and KRAS do, however, appear to benefit overall from EGFR TKIs. For this group, which constitutes roughly two thirds of patients, there are currently no established markers to predict a clinical benefit from EGFR TKIs. Our hypothesis was that using a gene expression signature will allow the identification of a subgroup of patients with with EGFR&KRAS tumors that benefit from EGFR TKIs.
Therefore, we investigated markers for identifying patients that would be most likely to benefit from erlotinib in patients with non-small cell lung cancer (NSCLC) treated in the BATTLE program. The Affymetrix platform was used to analyze gene expression from NSCLC patients treated in the BATTLE program. There were a total of 101 patients treated in the following arms: erlotinib (n=27), erlotinib+bexarotene (n=8), vandetinib (n=19) and sorafenib (n=47).
As a result, and noted above, a five gene signature that predicts clinical benefit (e.g. disease control) in patients that were EGFR and KRAS wildtype was developed and validated in NSCLC cell lines. The genes included in the signature have the following probe sets (gene name included if known): 219789_at (NPR3), 219790_s_at, 219054_at (C5orf23), 212531_at (LCN2), 205760_s_at (OGG1), and 205301_s_at.
Furthermore, our data identified that one of the genes in this 5-gene signature, LCN2, is a potential biomarker for predicting response to EGFR inhibitors. LCN2 gene, protein and secreted form as detected in plasma was a biomarker of response. LCN2 is also a marker for EGFR inhibitors and other inhibitors of the EGFR family such as HER2 (trastuzumab) and an important marker for epithelial phenotype and PI3K activation and dependence. As noted above,
This invention was made with government support under awarded under P50 CA070907 by the National Institutes of Health/National Cancer Institute, and under W81XWH-07-1-0306 and W81XWH-06-1-0303 by the Department of Defense. The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind | 371c Date |
---|---|---|---|---|
PCT/US12/31873 | 4/2/2012 | WO | 00 | 10/1/2013 |
Number | Date | Country | |
---|---|---|---|
61470625 | Apr 2011 | US | |
61472098 | Apr 2011 | US |