Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer

Abstract

Small-cell lung cancer (SCLC) is an aggressive lung tumor subtype with poor prognosis1,2,3. We sequenced 29 SCLC exomes, 2 genomes and 15 transcriptomes and found an extremely high mutation rate of 7.4 ± 1 protein-changing mutations per million base pairs. Therefore, we conducted integrated analyses of the various data sets to identify pathogenetically relevant mutated genes. In all cases, we found evidence for inactivation of TP53 and RB1 and identified recurrent mutations in the CREBBP, EP300 and MLL genes that encode histone modifiers. Furthermore, we observed mutations in PTEN, SLIT2 and EPHA7, as well as focal amplifications of the FGFR1 tyrosine kinase gene. Finally, we detected many of the alterations found in humans in SCLC tumors from Tp53 and Rb1 double knockout mice4. Our study implicates histone modification as a major feature of SCLC, reveals potentially therapeutically tractable genomic alterations and provides a generalizable framework for the identification of biologically relevant genes in the context of high mutational background.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Identification of SCNAs, chimeric transcripts and genomic rearrangements in human and mouse SCLC tumors.
Figure 2: Comparison of resected and autopsy samples and identification of candidate driver mutations.
Figure 3: Recurrent mutations affecting SLIT2, CREBBP and EP300.
Figure 4: Functional analysis of CREBBP and EP300.
Figure 5: Mutation spectra of TP53 and RB1 and genetic pathways altered in SCLC.

Similar content being viewed by others

References

  1. Gustafsson, B.I., Kidd, M., Chan, A., Malfertheiner, M.V. & Modlin, I.M. Bronchopulmonary neuroendocrine tumors. Cancer 113, 5–21 (2008).

    CAS  PubMed  Google Scholar 

  2. Travis, W.D. Lung tumours with neuroendocrine differentiation. Eur. J. Cancer 45 (suppl. 1), 251–266 (2009).

    PubMed  Google Scholar 

  3. van Meerbeeck, J.P., Fennell, D.A. & De Ruysscher, D.K. Small-cell lung cancer. Lancet 378, 1741–1755 (2011).

    PubMed  Google Scholar 

  4. Park, K.S. et al. A crucial requirement for Hedgehog signaling in small cell lung cancer. Nat. Med. 17, 1504–1508 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. World Health Organization. Pathology and Genetics of Tumours of the Lung, Pleura, Thymus and Heart (eds. Travis, W.D., Brambilla, E., Müller-Hermilink, H.K. & Harris, C.C.) 344 (IARC Press, Lyon, France, 2004).

  6. Tiseo, M. & Ardizzoni, A. Current status of second-line treatment and novel therapies for small cell lung cancer. J. Thorac. Oncol. 2, 764–772 (2007).

    PubMed  Google Scholar 

  7. Kwak, E.L. et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 363, 1693–1703 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. 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, 2129–2139 (2004).

    CAS  PubMed  Google Scholar 

  9. Paez, J.G. et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304, 1497–1500 (2004).

    CAS  PubMed  Google Scholar 

  10. Pao, W. & Chmielecki, J. Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer. Nat. Rev. Cancer 10, 760–774 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. 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, 13306–13311 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Soda, M. et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448, 561–566 (2007).

    CAS  PubMed  Google Scholar 

  13. Bergethon, K. et al. ROS1 rearrangements define a unique molecular class of lung cancers. J. Clin. Oncol. 30, 863–870 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kohno, T. et al. KIF5B-RET fusions in lung adenocarcinoma. Nat. Med. 18, 375–377 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Lipson, D. et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat. Med. 18, 382–384 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Takeuchi, K. et al. RET, ROS1 and ALK fusions in lung cancer. Nat. Med. 18, 378–381 (2012).

    CAS  PubMed  Google Scholar 

  17. Dutt, A. et al. Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer. PLoS One 6, e20351 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Hammerman, P.S. et al. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discov. 1, 78–89 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Weiss, J. et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci. Transl. Med. 2, 62ra93 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Bass, A.J. et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat. Genet. 41, 1238–1242 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Kim, Y.H. et al. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene 25, 130–138 (2006).

    CAS  PubMed  Google Scholar 

  22. Wistuba, I.I., Gazdar, A.F. & Minna, J.D. Molecular genetics of small cell lung carcinoma. Semin. Oncol. 28, 3–13 (2001).

    CAS  PubMed  Google Scholar 

  23. Gazzeri, S. et al. Activation of myc gene family in human lung carcinomas and during heterotransplantation into nude mice. Cancer Res. 51, 2566–2571 (1991).

    CAS  PubMed  Google Scholar 

  24. Zhao, X. et al. Homozygous deletions and chromosome amplifications in human lung carcinomas revealed by single nucleotide polymorphism array analysis. Cancer Res. 65, 5561–5570 (2005).

    CAS  PubMed  Google Scholar 

  25. Voortman, J. et al. Array comparative genomic hybridization–based characterization of genetic alterations in pulmonary neuroendocrine tumors. Proc. Natl. Acad. Sci. USA 107, 13040–13045 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hassan, M.I., Naiyer, A. & Ahmad, F. Fragile histidine triad protein: structure, function, and its association with tumorogenesis. J. Cancer Res. Clin. Oncol. 136, 333–350 (2010).

    CAS  PubMed  Google Scholar 

  27. Calbo, J. et al. A functional role for tumor cell heterogeneity in a mouse model of small cell lung cancer. Cancer Cell 19, 244–256 (2011).

    CAS  PubMed  Google Scholar 

  28. Dooley, A.L. et al. Nuclear factor I/B is an oncogene in small cell lung cancer. Genes Dev. 25, 1470–1475 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Meuwissen, R. et al. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell 4, 181–189 (2003).

    CAS  PubMed  Google Scholar 

  30. Schaffer, B.E. et al. Loss of p130 accelerates tumor development in a mouse model for human small-cell lung carcinoma. Cancer Res. 70, 3877–3883 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sutherland, K.D. et al. Cell of origin of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 19, 754–764 (2011).

    CAS  PubMed  Google Scholar 

  32. Iaquinta, P.J. & Lees, J.A. Life and death decisions by the E2F transcription factors. Curr. Opin. Cell Biol. 19, 649–657 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Berger, M.F. et al. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ding, L. et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Pleasance, E.D. et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 463, 191–196 (2010).

    CAS  PubMed  Google Scholar 

  38. Pleasance, E.D. et al. A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature 463, 184–190 (2010).

    CAS  PubMed  Google Scholar 

  39. Puente, X.S. et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475, 101–105 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  41. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Karro, J.E., Peifer, M., Hardison, R.C., Kollmann, M. & von Grunberg, H.H. Exponential decay of GC content detected by strand-symmetric substitution rates influences the evolution of isochore structure. Mol. Biol. Evol. 25, 362–374 (2008).

    CAS  PubMed  Google Scholar 

  43. Hecht, S.S. Progress and challenges in selected areas of tobacco carcinogenesis. Chem. Res. Toxicol. 21, 160–171 (2008).

    PubMed  Google Scholar 

  44. Rodin, S.N. & Rodin, A.S. Origins and selection of p53 mutations in lung carcinogenesis. Semin. Cancer Biol. 15, 103–112 (2005).

    CAS  PubMed  Google Scholar 

  45. Horowitz, J.M. et al. Frequent inactivation of the retinoblastoma anti-oncogene is restricted to a subset of human tumor cells. Proc. Natl. Acad. Sci. USA 87, 2775–2779 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Mori, N. et al. Variable mutations of the RB gene in small-cell lung carcinoma. Oncogene 5, 1713–1717 (1990).

    CAS  PubMed  Google Scholar 

  47. Bamford, S. et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 91, 355–358 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wong, K., Park, H.T., Wu, J.Y. & Rao, Y. Slit proteins: molecular guidance cues for cells ranging from neurons to leukocytes. Curr. Opin. Genet. Dev. 12, 583–591 (2002).

    CAS  PubMed  Google Scholar 

  49. Zhou, W.J. et al. Slit-Robo signaling induces malignant transformation through Hakai-mediated E-cadherin degradation during colorectal epithelial cell carcinogenesis. Cell Res. 21, 609–626 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Xian, J. et al. Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc. Natl. Acad. Sci. USA 98, 15062–15066 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Tseng, R.C. et al. SLIT2 attenuation during lung cancer progression deregulates β-catenin and E-cadherin and associates with poor prognosis. Cancer Res. 70, 543–551 (2010).

    CAS  PubMed  Google Scholar 

  52. Taguchi, A. et al. Lung cancer signatures in plasma based on proteome profiling of mouse tumor models. Cancer Cell 20, 289–299 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Oricchio, E. et al. The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell 147, 554–564 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Holmberg, J., Clarke, D.L. & Frisen, J. Regulation of repulsion versus adhesion by different splice forms of an Eph receptor. Nature 408, 203–206 (2000).

    CAS  PubMed  Google Scholar 

  55. Muraoka, M. et al. p300 gene alterations in colorectal and gastric carcinomas. Oncogene 12, 1565–1569 (1996).

    CAS  PubMed  Google Scholar 

  56. Liu, X. et al. The structural basis of protein acetylation by the p300/CBP transcriptional coactivator. Nature 451, 846–850 (2008).

    CAS  PubMed  Google Scholar 

  57. Pasqualucci, L. et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 471, 189–195 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Kishimoto, M. et al. Mutations and deletions of the CBP gene in human lung cancer. Clin. Cancer Res. 11, 512–519 (2005).

    CAS  PubMed  Google Scholar 

  59. Morin, R.D. et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Inthal, A. et al. CREBBP HAT domain mutations prevail in relapse cases of high hyperdiploid childhood acute lymphoblastic leukemia. Leukemia 26, 1797–1803 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Mullighan, C.G. et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 471, 235–239 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Tillinghast, G.W. et al. Analysis of genetic stability at the EP300 and CREBBP loci in a panel of cancer cell lines. Genes Chromosom. Cancer 37, 121–131 (2003).

    CAS  PubMed  Google Scholar 

  63. Ng, P.C. & Henikoff, S. SIFT: predicting amino acid changes that affect protein function. Nucleic Acids Res. 31, 3812–3814 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Kang-Decker, N. et al. Loss of CBP causes T cell lymphomagenesis in synergy with p27Kip1 insufficiency. Cancer Cell 5, 177–189 (2004).

    CAS  PubMed  Google Scholar 

  65. Kasper, L.H. et al. Conditional knockout mice reveal distinct functions for the global transcriptional coactivators CBP and p300 in T-cell development. Mol. Cell Biol. 26, 789–809 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Kasper, L.H. et al. CBP/p300 double null cells reveal effect of coactivator level and diversity on CREB transactivation. EMBO J. 29, 3660–3672 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Thirman, M.J. et al. Rearrangement of the MLL gene in acute lymphoblastic and acute myeloid leukemias with 11q23 chromosomal translocations. N. Engl. J. Med. 329, 909–914 (1993).

    CAS  PubMed  Google Scholar 

  68. Yang, X.J. The diverse superfamily of lysine acetyltransferases and their roles in leukemia and other diseases. Nucleic Acids Res. 32, 959–976 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Yokomizo, A. et al. PTEN/MMAC1 mutations identified in small cell, but not in non-small cell lung cancers. Oncogene 17, 475–479 (1998).

    CAS  PubMed  Google Scholar 

  70. Han, S.Y. et al. Functional evaluation of PTEN missense mutations using in vitro phosphoinositide phosphatase assay. Cancer Res. 60, 3147–3151 (2000).

    CAS  PubMed  Google Scholar 

  71. Yamamoto, H. et al. PIK3CA mutations and copy number gains in human lung cancers. Cancer Res. 68, 6913–6921 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Zakowski, M.F., Ladanyi, M. & Kris, M.G. EGFR mutations in small-cell lung cancers in patients who have never smoked. N. Engl. J. Med. 355, 213–215 (2006).

    CAS  PubMed  Google Scholar 

  73. Sequist, L.V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 3, 75ra26 (2011).

    PubMed  PubMed Central  Google Scholar 

  74. Gu, W., Shi, X.L. & Roeder, R.G. Synergistic activation of transcription by CBP and p53. Nature 387, 819–823 (1997).

    CAS  PubMed  Google Scholar 

  75. Brooks, C.L. & Gu, W. Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr. Opin. Cell Biol. 15, 164–171 (2003).

    CAS  PubMed  Google Scholar 

  76. Sakaguchi, K. et al. DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev. 12, 2831–2841 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kruse, J.P. & Gu, W. Modes of p53 regulation. Cell 137, 609–622 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Grossman, S.R. et al. Polyubiquitination of p53 by a ubiquitin ligase activity of p300. Science 300, 342–344 (2003).

    CAS  PubMed  Google Scholar 

  79. Lill, N.L., Grossman, S.R., Ginsberg, D., DeCaprio, J. & Livingston, D.M. Binding and modulation of p53 by p300/CBP coactivators. Nature 387, 823–827 (1997).

    CAS  PubMed  Google Scholar 

  80. Wong, K. et al. Signal transduction in neuronal migration: roles of GTPase activating proteins and the small GTPase Cdc42 in the Slit-Robo pathway. Cell 107, 209–221 (2001).

    CAS  PubMed  Google Scholar 

  81. Bordoli, L. et al. Functional analysis of the p300 acetyltransferase domain: the PHD finger of p300 but not of CBP is dispensable for enzymatic activity. Nucleic Acids Res. 29, 4462–4471 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Li, H., Ruan, J. & Durbin, R. Mapping short DNA sequencing reads and calling variants using mapping quality scores. Genome Res. 18, 1851–1858 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Ding, L. et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455, 1069–1075 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Scheble, V.J. et al. ERG rearrangement is specific to prostate cancer and does not occur in any other common tumor. Mod. Pathol. 23, 1061–1067 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Schildhaus, H.-U. et al. Definition of a fluorescence in situ hybridization score identifies high- and low-level FGFR1 amplification types in squamous cell lung cancer. Mod. Pathol. published online, doi:10.1038/modpathol.2012.102 (8 June 2012).

  87. Laframboise, T., Harrington, D. & Weir, B.A. PLASQ: a generalized linear model–based procedure to determine allelic dosage in cancer cells from SNP array data. Biostatistics 8, 323–336 (2007).

    PubMed  Google Scholar 

  88. Olshen, A.B., Venkatraman, E.S., Lucito, R. & Wigler, M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 5, 557–572 (2004).

    PubMed  Google Scholar 

  89. Sos, M.L. et al. Predicting drug susceptibility of non-small cell lung cancers based on genetic lesions. J. Clin. Invest. 119, 1727–1740 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Querings, S. et al. Benchmarking of mutation diagnostics in clinical lung cancer specimens. PLoS One 6, e19601 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are indebted to the individuals donating their tumor specimens as part of the Clinical Lung Cancer Genome Project initiative. Additional biospecimens for this study were obtained from the Victorian Cancer Biobank (Melbourne, Australia). The Institutional Review Board (IRB) of each participating institution approved collection and use of all specimens in this study. We also thank our colleagues at The Cancer Genome Atlas Research Network (TCGA) and A.L. Kung for invaluable discussion and many helpful comments. This work was supported by the German Ministry of Science and Education (BMBF) as part of the NGFNplus program (grant 01GS08100 to R.K.T. and 01GS08101 to J.W. and P.N.), by the Max Planck Society (M.I.F.A.NEUR8061 to R.K.T.), by the Deutsche Forschungsgemeinschaft (DFG) through SFB832 (TP6 to R.K.T. and TP5 and Z1 to L.C.H. and R.B.) and TH1386/3-1 (to R.K.T. and M.L.S.), by the European Union's Framework Programme CURELUNG (HEALTH-F2-2010-258677 to R.K.T., J.F., E.B., C. Brambilla, S.L., B.B. and J.W.), by Stand Up To Cancer–American Association for Cancer Research Innovative Research Grant (SU2C-AACR-IR60109 to R.K.T. and W.P.), by the Behrens-Weise Foundation (to R.K.T.) and by an anonymous foundation to R.K.T. M.L.S. is a fellow of the International Association for the Study of Lung Cancer (IASLC). P.K.B. and L.H.K. thank the St. Jude Cell and Tissue Imaging facility and acknowledge support from the US National Institutes of Health (NIH) Cancer Center (grant P30 CA021765) and the American Lebanese Syrian Associated Charities of St. Jude Children's Research Hospital. F.C. was supported by Associazione Italiana Ricerca sul Cancro (AIRC, grant IG 9425).

Author information

Authors and Affiliations

Authors

Contributions

M.P., L.F.-C., M.L.S., J.G., D.S., L.H.K., F.L.,T.Z., R.M., J.V., P.S., J. Sage, R. Schneider, R.B., S.P., L.C.H., P.K.B. and R.K.T. conceived and designed the experiments. L.F.-C., M.L.S., J.G., D.S., L.H.K., D.P., R.M., M.K., I.D., C.M., V.D.C., H.-U.S., J.A., I.B., C. Becker, B.d.W., D.B., F.G., I.W., S. Heynck, J.M.H. and P.M.S. performed experiments. M.P., L.F.-C., M.L.S., J.G., D.S., L.H.K., D.P., F.L., R. Sun, T.Z., R.M., V.D.C., B.d.W., J.V., X.L., W.P., M.M., J. Sage, R. Schneider, S.P., L.C.H., P.K.B., S. Haas and R.K.T. analyzed the data. M.P., R. Sun, S.A., S.L.C., K.C., S.B., G.G., K.-S.P., D.R., C.G., M.F., L.P., G.W., Z.W., P.R., I.P., Y.C., E. Stoelben, C. Ludwig, P.S., H.H., T.M., M.B., W.E.-R., L.A.M., V.M.F., H.G., W.T., H.S., E.T., E. Smit, D.A.M.H., P.J.F.S., F.C., C. Ligorio, S.D., J.F., S.S., O.T.B., M.L.-I., J. Sänger, J.H.C., A.S., H.M., W.W., B.S., J.-C.S., P.V., B.B., E.B., C. Brambilla, S.L., P.L., M.H., J. Sage, J. Shendure, R. Schneider, R.B., S.P., L.C.H., J.W., P.N., L.C.H., P.K.B. and S. Haas contributed reagents, materials or analysis tools. M.P., L.F.-C. and R.K.T. wrote the manuscript.

Corresponding author

Correspondence to Roman K Thomas.

Ethics declarations

Competing interests

R.K.T. reports the following potential sources of conflict of interest: consulting and lecture fees (Sanofi-Aventis, Merck, Bayer, Eli Lilly, Roche, Boehringer Ingelheim, Johnson & Johnson, AstraZeneca, Atlas-Biolabs and Daiichi-Sankyo) and research support (AstraZeneca, Merck and EOS). R.K.T. is a founder and shareholder of Blackfield, a company involved in cancer genome analysis services and cancer genomics–based drug discovery. M.P. and J.M.H. are shareholders of Blackfield. J.M.H. is a full-time employee of Blackfield.

Supplementary information

Supplementary Text and Figures

Supplementary Note and Supplementary Figures 1–12 (PDF 8342 kb)

Supplementary Tables

Supplementary Tables 1–13 (XLS 8649 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Peifer, M., Fernández-Cuesta, L., Sos, M. et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet 44, 1104–1110 (2012). https://doi.org/10.1038/ng.2396

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2396

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing