Comprehensive genomic characterization of squamous cell lung cancers

Cancer Genome Atlas Research Network

Abstract

Lung squamous cell carcinoma is a common type of lung cancer, causing approximately 400,000 deaths per year worldwide. Genomic alterations in squamous cell lung cancers have not been comprehensively characterized, and no molecularly targeted agents have been specifically developed for its treatment. As part of The Cancer Genome Atlas, here we profile 178 lung squamous cell carcinomas to provide a comprehensive landscape of genomic and epigenomic alterations. We show that the tumour type is characterized by complex genomic alterations, with a mean of 360 exonic mutations, 165 genomic rearrangements, and 323 segments of copy number alteration per tumour. We find statistically recurrent mutations in 11 genes, including mutation of TP53 in nearly all specimens. Previously unreported loss-of-function mutations are seen in the HLA-A class I major histocompatibility gene. Significantly altered pathways included NFE2L2 and KEAP1 in 34%, squamous differentiation genes in 44%, phosphatidylinositol-3-OH kinase pathway genes in 47%, and CDKN2A and RB1 in 72% of tumours. We identified a potential therapeutic target in most tumours, offering new avenues of investigation for the treatment of squamous cell lung cancers.

Figures

Figure 1. Significantly mutated genes in lung…
Figure 1. Significantly mutated genes in lung SqCC
Significantly mutated genes (q-value

Figure 2. Somatically altered pathways in squamous…

Figure 2. Somatically altered pathways in squamous cell lung cancer

Left, Alterations in oxidative response…
Figure 2. Somatically altered pathways in squamous cell lung cancer
Left, Alterations in oxidative response pathway genes by somatic mutation as defined by somatic mutation, copy number alteration or up- or down-regulation. Frequencies of alteration are expressed as a percentage of all cases, with background in red for activated genes and blue for inactivated genes. Right, Alterations in genes that regulate squamous differentiation, as defined in the left panel.

Figure 3. Gene expression subtypes integrated with…

Figure 3. Gene expression subtypes integrated with genomic alterations

Tumors are displayed as columns, grouped…

Figure 3. Gene expression subtypes integrated with genomic alterations
Tumors are displayed as columns, grouped by gene expression subtype. Subtypes were compared by Kruskal-Wallis tests for continuous features and by Fisher’s exact tests for categorical features. Displayed features displayed showed significant association with gene expression subtype (P<0.05), except for CDKN2A alterations. deltaN expression percentage represents transcript isoform usage between the TP63 isoforms, deltaN and tap63, as determined by RNA-sequencing. Chromosomal instability (CIN) is defined by the mean of the absolute values of chromosome arm copy numbers from the GISTIC, output. Absolute values are used so that amplification and deletion alterations are counted equally. Hypermethylation scores and iCluster assignments are described in Supplementary Figure S6.1 and S7.A1, respectively. CIN, methylation, gene expression, and deltaN values were standardized for display using z-score transformation.

Figure 4. Multi-faceted characterization of mechanisms of…

Figure 4. Multi-faceted characterization of mechanisms of CDKN2A loss

a, Schematic view of the exon…
Figure 4. Multi-faceted characterization of mechanisms of CDKN2A loss
a, Schematic view of the exon structure of CDKN2A demonstrating the types of alterations identified in the study. The locations of point mutation are denoted by black and green circles. b,CDKN2A expression (y-axis) versus CDKN2A copy number (x-axis). Samples are represented by circles and colored-coded by specific type of CDKN2A alteration. c, Diagram of the KIAA1797-CDKN2A fusion identified by whole genome sequencing. d,CDKN2A alterations and expression levels (binary) in each sample.

Figure 5. Alterations in targetable oncogenic pathways…

Figure 5. Alterations in targetable oncogenic pathways in lung SqCCs

Pathway diagram showing the percentage…

Figure 5. Alterations in targetable oncogenic pathways in lung SqCCs
Pathway diagram showing the percentage of samples with alterations in the PI3K/RTK/RAS pathways. Alterations are defined by somatic mutations, homozygous deletions, high-level, focal amplifications, and, in some cases, by significant up- or down-regulation of gene expression (AKT3, FGFR1, PTEN).
Figure 2. Somatically altered pathways in squamous…
Figure 2. Somatically altered pathways in squamous cell lung cancer
Left, Alterations in oxidative response pathway genes by somatic mutation as defined by somatic mutation, copy number alteration or up- or down-regulation. Frequencies of alteration are expressed as a percentage of all cases, with background in red for activated genes and blue for inactivated genes. Right, Alterations in genes that regulate squamous differentiation, as defined in the left panel.
Figure 3. Gene expression subtypes integrated with…
Figure 3. Gene expression subtypes integrated with genomic alterations
Tumors are displayed as columns, grouped by gene expression subtype. Subtypes were compared by Kruskal-Wallis tests for continuous features and by Fisher’s exact tests for categorical features. Displayed features displayed showed significant association with gene expression subtype (P<0.05), except for CDKN2A alterations. deltaN expression percentage represents transcript isoform usage between the TP63 isoforms, deltaN and tap63, as determined by RNA-sequencing. Chromosomal instability (CIN) is defined by the mean of the absolute values of chromosome arm copy numbers from the GISTIC, output. Absolute values are used so that amplification and deletion alterations are counted equally. Hypermethylation scores and iCluster assignments are described in Supplementary Figure S6.1 and S7.A1, respectively. CIN, methylation, gene expression, and deltaN values were standardized for display using z-score transformation.
Figure 4. Multi-faceted characterization of mechanisms of…
Figure 4. Multi-faceted characterization of mechanisms of CDKN2A loss
a, Schematic view of the exon structure of CDKN2A demonstrating the types of alterations identified in the study. The locations of point mutation are denoted by black and green circles. b,CDKN2A expression (y-axis) versus CDKN2A copy number (x-axis). Samples are represented by circles and colored-coded by specific type of CDKN2A alteration. c, Diagram of the KIAA1797-CDKN2A fusion identified by whole genome sequencing. d,CDKN2A alterations and expression levels (binary) in each sample.
Figure 5. Alterations in targetable oncogenic pathways…
Figure 5. Alterations in targetable oncogenic pathways in lung SqCCs
Pathway diagram showing the percentage of samples with alterations in the PI3K/RTK/RAS pathways. Alterations are defined by somatic mutations, homozygous deletions, high-level, focal amplifications, and, in some cases, by significant up- or down-regulation of gene expression (AKT3, FGFR1, PTEN).

References

    1. [Accessed February, 2012];WHO Statistics. < >.
    1. Soda M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–566. doi: 10.1038/nature05945. nature05945 [pii]
    1. Paez JG, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–1500. doi: 10.1126/science.10993141099314. [pii]
    1. Lynch TJ, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139. doi: 10.1056/NEJMoa040938. NEJMoa040938 [pii]
    1. 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 U S A. 2004;101:13306–13311. doi: 10.1073/pnas.04052201010405220101. [pii]
    1. Felip E, Gridelli C, Baas P, Rosell R, Stahel R. Metastatic non-small-cell lung cancer: consensus on pathology and molecular tests, first-line, second-line, and third-line therapy: 1st ESMO Consensus Conference in Lung Cancer; Lugano 2010. Ann Oncol. 2011;22:1507–1519. doi: 10.1093/annonc/mdr150. mdr150 [pii]
    1. Ju YS, et al. A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-genome and transcriptome sequencing. Genome Res. 2012 doi: 10.1101/gr.133645.111. gr.133645.111 [pii]
    1. Rekhtman N, et al. Clarifying the spectrum of driver oncogene mutations in biomarker-verified squamous carcinoma of lung: lack of EGFR/KRAS and presence of PIK3CA/AKT1 mutations. Clin Cancer Res. 2012 doi: 10.1158/1078-0432.CCR-11-2109. 1078-0432.CCR-11-2109 [pii]
    1. Bass AJ, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 2009;41:1238–1242. doi: 10.1038/ng.465. ng.465 [pii]
    1. Ramos AH, et al. Amplification of chromosomal segment 4q12 in non-small cell lung cancer. Cancer Biol Ther. 2009;8:2042–2050. 9764 [pii]
    1. Shibata T, et al. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A. 2008;105:13568–13573. doi: 10.1073/pnas.0806268105. 0806268105 [pii]
    1. Kan Z, et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature. 2010;466:869–873. doi: 10.1038/nature09208. nature09208 [pii]
    1. Hammerman PS, Sos ML, Ramos A, Xu C. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discovery. 2011;1
    1. Weiss J, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2:62ra93. doi: 10.1126/scitranslmed.3001451. 2/62/62ra93 [pii]
    1. Dutt A, et al. Inhibitor-Sensitive FGFR1 Amplification in Human Non-Small Cell Lung Cancer. PLoS One. 2011;6:e20351. doi: 10.1371/journal.pone.0020351. PONE-D-11-00027 [pii]
    1. Kenfield SA, Wei EK, Stampfer MJ, Rosner BA, Colditz GA. Comparison of aspects of smoking among the four histological types of lung cancer. Tob Control. 2008;17:198–204. doi: 10.1136/tc.2007.022582. tc.2007.022582 [pii]
    1. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615. doi: 10.1038/nature10166. nature10166 [pii]
    1. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385. nature07385 [pii]
    1. Tonon G, et al. High-resolution genomic profiles of human lung cancer. Proc Natl Acad Sci U S A. 2005;102:9625–9630. doi: 10.1073/pnas.0504126102. 0504126102 [pii]
    1. Beroukhim R, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. doi: 10.1038/nature08822. nature08822 [pii]
    1. Mermel CH, et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12:R41. doi: 10.1186/gb-2011-12-4-r41. gb-2011-12-4-r41 [pii]
    1. Stransky N, et al. The mutational landscape of head and neck squamous cell carcinoma. Science. 2011;333:1157–1160. doi: 10.1126/science.1208130. science.1208130 [pii]
    1. Chapman MA, et al. Initial genome sequencing and analysis of multiple myeloma. Nature. 2011;471:467–472. doi: 10.1038/nature09837. nature09837 [pii]
    1. Agrawal N, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333:1154–1157. doi: 10.1126/science.1206923. science.1206923 [pii]
    1. Bass AJ, et al. Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat Genet. 2011;43:964–968. doi: 10.1038/ng.936. ng.936 [pii]
    1. Berger MF, et al. The genomic complexity of primary human prostate cancer. Nature. 2011;470:214–220. doi: 10.1038/nature09744. nature09744 [pii]
    1. Stephens PJ, et al. Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature. 2009;462:1005–1010. doi: 10.1038/nature08645. nature08645 [pii]
    1. Singh A, et al. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med. 2006;3:e420. doi: 10.1371/journal.pmed.0030420. 06-PLME-RA-0322R2 [pii]
    1. Singh A, Bodas M, Wakabayashi N, Bunz F, Biswal S. Gain of Nrf2 function in non-small-cell lung cancer cells confers radioresistance. Antioxid Redox Signal. 2010;13:1627–1637. doi: 10.1089/ars.2010.3219.
    1. Vaske CJ, et al. Inference of patient-specific pathway activities from multidimensional cancer genomics data using PARADIGM. Bioinformatics. 2010;26:i237–245. doi: 10.1093/bioinformatics/btq182. btq182 [pii]
    1. Aster JC, Blacklow SC, Pear WS. Notch signalling in T-cell lymphoblastic leukaemia/lymphoma and other haematological malignancies. J Pathol. 2011;223:262–273. doi: 10.1002/path.2789.
    1. Wang NJ, et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci U S A. 2011;108:17761–17766. doi: 10.1073/pnas.1114669108. 1114669108 [pii]
    1. Osada H, Tatematsu Y, Yatabe Y, Horio Y, Takahashi T. ASH1 gene is a specific therapeutic target for lung cancers with neuroendocrine features. Cancer Res. 2005;65:10680–10685. doi: 10.1158/0008-5472.CAN-05-1404. 65/23/10680 [pii]
    1. Wilkerson MD, et al. Lung squamous cell carcinoma mRNA expression subtypes are reproducible, clinically important, and correspond to normal cell types. Clin Cancer Res. 2010;16:4864–4875. doi: 10.1158/1078-0432.CCR-10-0199. 1078-0432.CCR-10-0199 [pii]
    1. Bishop JA, et al. p40 (DeltaNp63) is superior to p63 for the diagnosis of pulmonary squamous cell carcinoma. Mod Pathol. 2011 doi: 10.1038/modpathol.2011.173. modpathol2011173 [pii]
    1. Massion PP, et al. Significance of p63 amplification and overexpression in lung cancer development and prognosis. Cancer Res. 2003;63:7113–7121.
    1. Shen R, Olshen AB, Ladanyi M. Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics. 2009;25:2906–2912. doi: 10.1093/bioinformatics/btp543. btp543 [pii]
    1. Wikman H, Kettunen E. Regulation of the G1/S phase of the cell cycle and alterations in the RB pathway in human lung cancer. Expert Rev Anticancer Ther. 2006;6:515–530. doi: 10.1586/14737140.6.4.515.
    1. Kancha RK, Peschel C, Duyster J. The epidermal growth factor receptor-L861Q mutation increases kinase activity without leading to enhanced sensitivity toward epidermal growth factor receptor kinase inhibitors. J Thorac Oncol. 2011;6:387–392. doi: 10.1097/JTO.0b013e3182021f3e. 01243894-201102000-00020 [pii]
    1. Reva B, Antipin Y, Sander C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 2011;39:e118. doi: 10.1093/nar/gkr407. gkr407 [pii]
    1. Govindan R. Summary of the proceedings from the 10th annual meeting of molecularly targeted therapy in non-small cell lung cancer. J Thorac Oncol. 2010;5:S433. doi: 10.1097/JTO.0b013e3181fd6fe1. 01243894-201012002-00001 [pii]
    1. Ding L, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075. doi: 10.1038/nature07423. nature07423 [pii]
    1. Ciriello G, Cerami E, Sander C, Schultz N. Mutual exclusivity analysis identifies oncogenic network modules. Genome Res. 2012;22:398–406. doi: 10.1101/gr.125567.111. gr.125567.111 [pii]
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. S0092-8674(11)00127-9 [pii]
    1. Brahmer JR, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–3175. doi: 10.1200/JCO.2009.26.7609. JCO.2009.26.7609 [pii]
    1. Lynch TJ, Bondarenko IN, Luft A, Serwatowski P, Barlesi F, Chacko RT, Sebastian M, Siegel J, Cuillerot J, Reck M. Phase II trial of ipilimumab (IPI) and paclitaxel/carboplatin (P/C) in first-line stage IIIb/IV non-small cell lung cancer (NSCLC) J Clin Oncol. 2010;28:7531.

Source: PubMed

3
Abonnieren