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.

  • Article
  • Published:

Genomic and evolutionary classification of lung cancer in never smokers

Abstract

Lung cancer in never smokers (LCINS) is a common cause of cancer mortality but its genomic landscape is poorly characterized. Here high-coverage whole-genome sequencing of 232 LCINS showed 3 subtypes defined by copy number aberrations. The dominant subtype (piano), which is rare in lung cancer in smokers, features somatic UBA1 mutations, germline AR variants and stem cell-like properties, including low mutational burden, high intratumor heterogeneity, long telomeres, frequent KRAS mutations and slow growth, as suggested by the occurrence of cancer drivers’ progenitor cells many years before tumor diagnosis. The other subtypes are characterized by specific amplifications and EGFR mutations (mezzo-forte) and whole-genome doubling (forte). No strong tobacco smoking signatures were detected, even in cases with exposure to secondhand tobacco smoke. Genes within the receptor tyrosine kinase–Ras pathway had distinct impacts on survival; five genomic alterations independently doubled mortality. These findings create avenues for personalized treatment in LCINS.

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

Access options

Buy this article

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

Fig. 1: TMB across LCINS from the Sherlock-Lung study and 33 cancer types from the TCGA study.
Fig. 2: Genomic characteristics of LCINS.
Fig. 3: Genomic classification of LCINS based on SCNAs.
Fig. 4: Landscape of mutational processes in Sherlock-Lung.
Fig. 5: Comparison of mutational spectra between passive and nonpassive smokers in Sherlock-Lung.
Fig. 6: Diagrams of estimated ordering of significant SCNAs (including chromosome gains/losses and mutations) relative to WGD in three lung cancer subtypes based on the SCNA clusters forte, mezzo-forte and piano.
Fig. 7: Reconstruction of the evolutionary history of LCINS.
Fig. 8: Association between genomic aberrations and clinical outcomes in never smoker patients with lung cancer.

Similar content being viewed by others

Data availability

The 232 normal and tumor-paired raw data (BAM files) of the WGS datasets have been deposited in the dbGaP under accession no. phs001697.v1.p1. Researchers will need to obtain authorization from the dbGaP to download these data. The RNA-seq raw data (FASTQ files) have been submitted to the Gene Expression Omnibus under accession no. GSE171415. The germline variant dataset from the EAGLE whole-exome sequencing study can be access at the dbGaP with accession no. phs002496.v1.p1. In addition, histological images of these tumors can be found at https://episphere.github.io/svs. Public datasets were used in this study including gnomAD v.2.1.1/ExAC v.0.3.1 (https://gnomad.broadinstitute.org/), 1000 Genomes (phase 3 v.5, https://www.internationalgenome.org/) and dbSNP (v.138, https://www.ncbi.nlm.nih.gov/snp/).

Code availability

The code for the WGS subclonal copy number caller can be found at https://github.com/Wedge-lab/battenberg (v.2.2.8). The code for somatic mutation filtering can be found at https://github.com/xtmgah/Sherlock-Lung. The code for the Dirichlet process-based methods for subclonal reconstruction of tumors can be found at https://github.com/Wedge-lab/dpclust (v.2.2.8). The code for the mutational signature analysis can be found at https://pypi.org/project/sigproextractor/ (SigProfilerExtractor v.0.0.5.77). The code for inferring the order of genomic events can be found at https://github.com/hturner/PlackettLuce (v.0.2-2). The code for the chronological timing analysis can be found at https://gerstung-lab.github.io/PCAWG-11/. The code for P-MACD can be found at https://github.com/NIEHS/P-MACD.

References

  1. The Cancer Atlas: Lung Cancer (American Cancer Society, 2021); https://canceratlas.cancer.org/the-burden/lung-cancer/

  2. Cho, J. et al. Proportion and clinical features of never-smokers with non-small cell lung cancer. Chin. J. Cancer 36, 20 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Campbell, J. D. et al. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat. Genet. 48, 607–616 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Collisson, E. A. et al. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

    Article  CAS  Google Scholar 

  5. Chen, J. et al. Genomic landscape of lung adenocarcinoma in East Asians. Nat. Genet. 52, 177–186 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Govindan, R. et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 150, 1121–1134 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lee, J. J.-K. et al. Tracing oncogene rearrangements in the mutational history of lung adenocarcinoma. Cell 177, 1842–1857.e21 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Shi, J. et al. Somatic genomics and clinical features of lung adenocarcinoma: a retrospective study. PLoS Med. 13, e1002162 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Wang, C. et al. Whole-genome sequencing reveals genomic signatures associated with the inflammatory microenvironments in Chinese NSCLC patients. Nat. Commun. 9, 2054 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Fernandez-Cuesta, L. et al. Frequent mutations in chromatin-remodelling genes in pulmonary carcinoids. Nat. Commun. 5, 3518 (2014).

    Article  PubMed  CAS  Google Scholar 

  12. Campbell, P. J. et al. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020).

    Article  CAS  Google Scholar 

  13. Wu, K. et al. Frequent alterations in cytoskeleton remodelling genes in primary and metastatic lung adenocarcinomas. Nat. Commun. 6, 10131 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Carrot-Zhang, J. et al. Whole-genome characterization of lung adenocarcinomas lacking the RTK/RAS/RAF pathway. Cell Rep. 34, 108707 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Landi, M. T. et al. Tracing lung cancer risk factors through mutational signatures in never smokers: the Sherlock-Lung study. Am. J. Epidemiol. 190, 962–976 (2021).

    Article  PubMed  Google Scholar 

  16. Skoulidis, F. et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 5, 860–877 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Moll, U. M. & Petrenko, O. The MDM2-p53 interaction. Mol. Cancer Res. 1, 1001–1008 (2003).

    CAS  PubMed  Google Scholar 

  18. Wala, J. A. et al. Selective and mechanistic sources of recurrent rearrangements across the cancer genome. Preprint at bioRxiv https://doi.org/10.1101/187609 (2017).

  19. Reznik, E. et al. Mitochondrial DNA copy number variation across human cancers. eLife 5, e10769 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  20. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271.e11 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Moudry, P. et al. Ubiquitin-activating enzyme UBA1 is required for cellular response to DNA damage. Cell Cycle 11, 1573–1582 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Martínez-Jiménez, F. A compendium of mutational cancer driver genes. Nat. Rev. Cancer 20, 555–572 (2020).

    Article  PubMed  CAS  Google Scholar 

  24. Huang, K.-L. et al. Pathogenic germline variants in 10,389 adult cancers. Cell 173, 355–370.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Staaf, J. et al. Whole-genome sequencing of triple-negative breast cancers in a population-based clinical study. Nat. Med. 25, 1526–1533 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bergstrom, E. N. et al. SigProfilerMatrixGenerator: a tool for visualizing and exploring patterns of small mutational events. BMC Genomics 20, 685 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Petljak, M. et al. Characterizing mutational signatures in human cancer cell lines reveals episodic APOBEC mutagenesis. Cell 176, 1282–1294.e20 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jager, M. et al. Deficiency of nucleotide excision repair is associated with mutational signature observed in cancer. Genome Res. 29, 1067–1077 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Singh, V. K., Rastogi, A., Hu, X., Wang, Y. & De, S. Mutational signature SBS8 predominantly arises due to late replication errors in cancer. Commun. Biol. 3, 421 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Roberts, S. A. et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat. Genet. 45, 970–976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kucab, J. E. et al. A compendium of mutational signatures of environmental agents. Cell 177, 821–836.e16 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tokiwa, H. & Sera, N. Contribution of nitrated polycyclic aromatic hydrocarbons in diesel particles to human lung cancer induction. Polycycl. Aromat. Compd. 21, 231–245 (2000).

    Article  CAS  Google Scholar 

  34. Saini, N. et al. Mutation signatures specific to DNA alkylating agents in yeast and cancers. Nucleic Acids Res. 48, 3692–3707 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chan, K. et al. An APOBEC3A hypermutation signature is distinguishable from the signature of background mutagenesis by APOBEC3B in human cancers. Nat. Genet. 47, 1067–1072 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Barthel, F. P. et al. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat. Genet. 49, 349–357 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Feuerbach, L. et al. TelomereHunter—in silico estimation of telomere content and composition from cancer genomes. BMC Bioinformatics 20, 272 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Davies, H. et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 23, 517–525 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhao, E. Y. et al. Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clin. Cancer Res. 23, 7521–7530 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. Letouzé, E. et al. Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat. Commun. 8, 1315 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Shinde, J. et al. Palimpsest: an R package for studying mutational and structural variant signatures along clonal evolution in cancer. Bioinformatics 34, 3380–3381 (2018).

    CAS  PubMed  Google Scholar 

  42. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Halvorsen, A. R. et al. TP53 mutation spectrum in smokers and never smoking lung cancer patients. Front. Genet. 7, 85 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Gu, J. et al. TP53 mutation is associated with a poor clinical outcome for non-small cell lung cancer: evidence from a meta-analysis. Mol. Clin. Oncol. 5, 705–713 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. López, S. et al. Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution. Nat. Genet. 52, 283–293 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Bielski, C. M. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 50, 1189–1195 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, International Agency for Research on Cancer. A Review of Human Carcinogens: Personal Habits and Indoor Combustions (International Agency for Research on Cancer, 2012).

  49. United States Public Health Service. Office of the Surgeon General. The Health Consequences of Involuntary Exposure to Tobacco Smoke: A Report of the Surgeon General (US Department of Health and Human Services, Public Health Service, Office of the Surgeon General, 2006).

  50. Lopez-Bigas, N. & Gonzalez-Perez, A. Are carcinogens direct mutagens? Nat. Genet. 52, 1137–1138 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Cho, I. J. et al. Mechanisms, hallmarks, and implications of stem cell quiescence. Stem Cell Reports 12, 1190–1200 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fukada, S.-I., Ma, Y. & Uezumi, A. Adult stem cell and mesenchymal progenitor theories of aging. Front. Cell Dev. Biol. 2, 10 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Li, L. & Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science 327, 542–545 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kim, C. F. B. et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121, 823–835 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Van Meter, M. E. M. et al. K-RasG12D expression induces hyperproliferation and aberrant signaling in primary hematopoietic stem/progenitor cells. Blood 109, 3945–3952 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kubara, K. et al. Status of KRAS in iPSCs impacts upon self-renewal and differentiation propensity. Stem Cell Reports 11, 380–394 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bax, M. et al. The ubiquitin proteasome system is a key regulator of pluripotent stem cell survival and motor neuron differentiation. Cells 8, 581 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  58. Leon, T. Y. Y. et al. Transcriptional regulation of RET by Nkx2-1, Phox2b, Sox10, and Pax3. J. Pediatr. Surg. 44, 1904–1912 (2009).

    Article  PubMed  Google Scholar 

  59. Grey, W. et al. Activation of the receptor tyrosine kinase, RET, improves long-term hematopoietic stem cell outgrowth and potency. Blood 136, 2535–2547 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Fonseca-Pereira, D. et al. The neurotrophic factor receptor RET drives haematopoietic stem cell survival and function. Nature 514, 98–101 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Zhao, B. et al. ARID1A promotes genomic stability through protecting telomere cohesion. Nat. Commun. 10, 4067 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Sun, X. et al. Suppression of the SWI/SNF component Arid1a promotes mammalian regeneration. Cell Stem Cell 18, 456–466 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. van der Vaart, A. & van den Heuvel, S. Switching on regeneration. Stem Cell Investig. 3, 41 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Wu, S., Zhang, R. & Bitler, B. G. Arid1a controls tissue regeneration. Stem Cell Investig. 3, 35 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Nagl, N. G. Jr, Wang, X., Patsialou, A., Van Scoy, M. & Moran, E. Distinct mammalian SWI/SNF chromatin remodeling complexes with opposing roles in cell-cycle control. EMBO J. 26, 752–763 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chiba, S. Notch signaling in stem cell systems. Stem Cells 24, 2437–2447 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Yoshida, K. et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature 578, 266–272 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Maeda, Y., Davé, V. & Whitsett, J. A. Transcriptional control of lung morphogenesis. Physiol. Rev. 87, 219–244 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Alanis, D. M., Chang, D. R., Akiyama, H., Krasnow, M. A. & Chen, J. Two nested developmental waves demarcate a compartment boundary in the mouse lung. Nat. Commun. 5, 3923 (2014).

    Article  PubMed  Google Scholar 

  70. Singh, I. et al. Hmga2 is required for canonical WNT signaling during lung development. BMC Biol. 12, 21 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Laughney, A. M. et al. Regenerative lineages and immune-mediated pruning in lung cancer metastasis. Nat. Med. 26, 259–269 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Duffy, M. J. et al. p53 as a target for the treatment of cancer. Cancer Treat. Rev. 40, 1153–1160 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Shaikh, M. F. et al. Emerging role of MDM2 as target for anti-cancer therapy: a review. Ann. Clin. Lab. Sci. 46, 627–634 (2016).

    CAS  PubMed  Google Scholar 

  74. Chuang, J. C. et al. ERBB2-mutated metastatic non-small cell lung cancer: response and resistance to targeted therapies. J. Thorac. Oncol. 12, 833–842 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Harvey, R. D., Adams, V. R., Beardslee, T. & Medina, P. Afatinib for the treatment of EGFR mutation-positive NSCLC: a review of clinical findings. J. Oncol. Pharm. Pract. 26, 1461–1474 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Park, K. et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol. 17, 577–589 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Shen, X. et al. A systematic analysis of the resistance and sensitivity of HER2YVMA receptor tyrosine kinase mutant to tyrosine kinase inhibitors in HER2-positive lung cancer. J. Recept. Signal Transduct. Res. 36, 89–97 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Miyazaki, M. et al. The p53 activator overcomes resistance to ALK inhibitors by regulating p53-target selectivity in ALK-driven neuroblastomas. Cell Death Discov. 4, 56 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Dey, P. et al. Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer. Nature 542, 119–123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Muller, F. L., Aquilanti, E. A. & DePinho, R. A. Collateral lethality: a new therapeutic strategy in oncology. Trends Cancer 1, 161–173 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Hsiehchen, D. et al. DNA repair gene mutations as predictors of immune checkpoint inhibitor response beyond tumor mutation burden. Cell Rep. Med. 1, 100034 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hellmann, M. D. et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 378, 2093–2104 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ready, N. et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J. Clin. Oncol. 37, 992–1000 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Canon, J. et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 (2019).

    Article  CAS  PubMed  Google Scholar 

  86. Yang, L. et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 5, 8 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Medema, J. P. & Vermeulen, L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature 474, 318–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  88. Jørsboe, E., Hanghøj, K. & Albrechtsen, A. fastNGSadmix: admixture proportions and principal component analysis of a single NGS sample. Bioinformatics 33, 3148–3150 (2017).

    Article  PubMed  CAS  Google Scholar 

  89. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kim, S. et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat. Methods 15, 591–594 (2018).

    Article  CAS  PubMed  Google Scholar 

  91. Freed, D., Pan, R. & Aldana, R. TNscope: accurate detection of somatic mutations with haplotype-based variant candidate detection and machine learning filtering. Preprint at bioRxiv https://doi.org/10.1101/250647 (2018).

  92. Zhu, B. et al. The genomic and epigenomic evolutionary history of papillary renal cell carcinomas. Nat. Commun. 11, 3096 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Karczewski, K. J. et al. The mutational constraints spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ramos, A. H. et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 36, E2423–E2429 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Hasan, M. S., Wu, X., Watson, L. T. & Zhang, L. UPS-indel: a universal positioning system for indels. Sci. Rep. 7, 14106 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Dentro, S. C., Wedge, D. C. & Van Loo, P. Principles of reconstructing the subclonal architecture of cancers. Cold Spring Harb. Perspect. Med. 7, a026625 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Scott, A. D. et al. CharGer: clinical Characterization of Germline variants. Bioinformatics 35, 865–867 (2019).

    Article  CAS  PubMed  Google Scholar 

  100. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–D868 (2016).

    Article  CAS  PubMed  Google Scholar 

  101. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041.e21 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Muiños, F., Martínez-Jiménez, F., Pich, O., Gonzalez-Perez, A. & Lopez-Bigas, N. In silico saturation mutagenesis of cancer genes. Nature 596, 428–432 (2021).

    Article  PubMed  CAS  Google Scholar 

  103. Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 12, R41 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Dewhurst, S. M. et al. Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 4, 175–185 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Yang, L. et al. Diverse mechanisms of somatic structural variations in human cancer genomes. Cell 153, 919–929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature 578, 112–121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Ding, Z. et al. Estimating telomere length from whole genome sequence data. Nucleic Acids Res. 42, e75 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 33, 1152–1158 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Bolli, N. et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat. Commun. 5, 2997 (2014).

    Article  PubMed  CAS  Google Scholar 

  111. Luce, R. D. Individual Choice Behavior: a Theoretical Analysis (Wiley, 1959).

  112. Plackett, R. L. The analysis of permutations. Appl. Stat. 24, 193 (1975).

    Article  Google Scholar 

Download references

Acknowledgements

This work has been supported by the Intramural Research Program of the Division of Cancer Epidemiology and Genetics, National Cancer Institute, and the Intramural Research Program of the National Institute of Environmental Health Sciences (project nos. Z01 ES050159 to S.H.W. and Z1AES103266 to D.A.G.), National Institutes of Health (NIH). This project was funded in whole or in part with federal funds from the National Cancer Institute, NIH, under contract nos. 75N91019D00024 and HHSN261201800001I. The content of this publication does not necessarily reflect the views or policies of the U.S. Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the U.S. Government. The research was supported by the Wellcome Trust Core Award, grant no. 203141/Z/16/Z with funding from the National Institute for Health Research Oxford Biomedical Research Centre. L.B.A. is an Abeloff V scholar and he is personally supported by an Alfred P. Sloan Research Fellowship and a Packard Fellowship for Science and Engineering. Research at the L.B.A. laboratory was supported by a National Institute of Environmental Health Sciences grant no. R01ES032547. The views expressed are those of the authors and not necessarily those of the National Health Service, National Institute for Health Research or Department of Health. The collection of samples from the Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Université Laval was supported by the IUCPQ Foundation. The GR Program 2010-2316264 supported L.A.M. for sample collection by the Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Casa Sollievo della Sofferenza. A.L.M. is supported by a Damon Runyon Cancer Research Foundation postdoctoral fellowship (no. DRG:2368-19) and a Postdoctoral Enrichment Program Award from the Burroughs Wellcome Fund (no. 1019903). C.F.K. is supported in part by grant no. R35HL150876-01, the Thoracic Foundation, Ellison Foundation, American Lung Association (no. LCD-619492) and the Harvard Stem Cell Institute. N.L-B. acknowledges funding from the European Research Council (consolidator grant no. 682398). P.H. is supported in part by the Association pour la Recherche contre le Cancer (CANC’AIR GENExposomics project). This work has been supported in part by the Tissue Core at the H. Lee Moffitt Cancer Center & Research Institute, a comprehensive cancer center designated by the National Cancer Institute and funded in part by a Moffitt Cancer Center Support Grant (no. P30-CA076292). B.E.G.R. is supported by NIH grant nos. 1P50 CA196530-01 and NIH 1K08 CA151645-01. We thank the Sherlock-Lung study scientific advisory board (M. Meyerson, J. Samet, M. Spitz, R. Summers, M. Thun and W. Travis) for their support. We also thank Y. Rubanova from Toronto University for her help with the TrackSig analysis. We thank the staff at the IUCPQ Université Laval Biobank, Nice Biobank Centre de Ressources Biologiques, Yale University and Moffitt Cancer Center & Research Institute for their valuable assistance in collecting samples and corresponding clinical data. This work utilized the computational resources of the NIH high-performance computational capabilities Biowulf cluster (http://hpc.nih.gov).

Author information

Authors and Affiliations

Authors

Contributions

M.T.L. and T.Z. conceptualized the study. T.Z., D.C.W., J. Shi., B.Z., N.A-P., N.L-B., B.Z., S.H.W., Y.P., H.C., T.R., D.R.S., D.A.G., L.B.A. and M.T.L. devised the methodology. T.Z., N.A-P., W.Z., P.H.H., R.L., K.H.-H., A.G.-P., F.M.-J., A.C., I.P., J. Sang, J. Shi, J.K., N.S., L.J.K., S.M.A.I., B.O., A.K., A.L.M. and C.F.K. carried out the formal analysis. A.H., N.C., J.C., D.H. and K.M.B. carried out the laboratory work. M.O., S.M.L., M.D., P.L., P.M.S.B. and J.S.A. carried out the pathology work. P.J., Y.B., P.H., D.C., A.C.P., L.A.M., B.E.G.R., M.L.P., M.C., M.B.S., N.E.C., M.L. and S.J.C. managed the resources. M.K., L.M. and J.R. curated the data. T.Z. and M.T.L. wrote the original draft. D.C.W., S.J.C., Y.B., Q.L., N.R., M.G-C., D.A.G., L.B.A., N.L-B., B.Z., J. Sang, J. Shi, T.Z., P.H.H. and M.T.L. reviewed and edited the draft. All authors carried out the data visualization. M.T.L. supervised the study.

Corresponding author

Correspondence to Maria Teresa Landi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Genomic alterations of RTK-RAS pathway in Sherlock-Lung.

a, Oncoplot showing mutual exclusivity of genes within the RTK-RAS pathway, which were used to define the RTK-RAS status. The bottom bar shows tumor histological types. b, Comparison of genomic features between RTK-RAS negative and positive tumors. Left four panels: tumor mutational burden, percentage of genome with SCNAs, SV burden and T/N TL ratio. P-values are calculated using the two-sided Mann-Whitney U test; Middle three panels: enrichments for Kataegis events, WGD events, and BRCA2 LOH. P-values and OR are calculated using Fisher’s exact test (two-sided); Right panel: Contributions of each SBS signature.

Extended Data Fig. 2 Genomic alterations of TP53 pathway in Sherlock-Lung.

a, Oncoplot showing the mutual exclusivity between TP53 mutations and MDM2 amplification, which was used to define the TP53 proficient and deficient groups. The bottom bar shows tumor histological types. b, Comparison of genomic features between TP53-proficient and TP53-deficient tumors. Left three panels: tumor mutation burden, percentage of genome with SCNA and SV burden. P-values are calculated using the two-sided Mann-Whitney U test. Middle four panels: enrichments for BRCA1 LOH, Kataegis events, WGD events, and HLA LOH. P-values and OR are calculated using Fisher’s exact test (two-sided). Right panel: Contributions of each SBS signature.

Extended Data Fig. 3 Recurrence of SV breakpoints in Sherlock-Lung.

The frequencies of chromosomal breakpoints are calculated using 5 Mb as a window across the whole genome.

Extended Data Fig. 4 Summary of genomic features in LCINS based on different SCNA clusters.

Panels from top to bottom describe: 1) most frequently mutated or potential driver genes; 2) oncogenic fusions; 3) somatic mutations in surfactant associated genes; 4) significant focal SCNAs; 5) significant arm-level SCNAs; 6) genes with rare germline mutations; 7) and 8) other genomic features. The numbers on the right panel show the overall frequency (1-7) or median values (8). NRPCC: the number of reads per clonal copy.

Extended Data Fig. 5 Genes with signals of positive selection in Sherlock-Lung.

a, The scatter plot showing significantly mutated genes according to IntOGen q-value <0.05 (y-axis) and mutational frequency in the cohort (x-axis). Genes are colored according to their inferred mode of action in tumorigenesis. b, Recurrent non-synonymous driver mutations (in ≥2 patients).

Extended Data Fig. 6 Dominant endogenous processes in Sherlock-Lung.

a, Density plot of cosine similarity between original mutational profile and reconstructed mutational profile using reference signatures from (top to bottom): 65 COSMIC SBS signatures, 22 COSMIC SBS signatures for endogenous processes, 53 MutaGene SBS signatures of environmental exposures, and a combined set of signatures including the 22 endogenous and 53 environmental exposure signatures. b, Comparison of the cosine similarity between the original mutational profiles and reconstructed mutational profiles using endogenous and exogenous signatures (similar to a). Each dot represents one sample. The size and color represent the total number of mutations and tumor histological type, respectively.

Extended Data Fig. 7 Association between T/N TL ratio and somatic alterations in Sherlock-Lung.

a, Distribution of mean telomere lengths (TL) in Sherlock-Lung (dark blue, overall and by histological type), TCGA LUAD (green, overall and by smoking status) and TCGA other cancer types (Grey). Total sample numbers for each type are shown at the top. Error bars, 95% CIs from linear mixed model. b, Scatterplot showing association between T/N TL ratio and somatic alterations. Association P-values (two-sided t-test; FDR adjusted using Benjamini-Hochberg method) are shown on the y-axis. Genomic alterations with FDR <=0.1 or T/N TL ratio >1.1 or <0.9 are labeled and further highlighted in red when significant (FDR=0.05; horizontal dashed line). c, The proportion of each SCNA cluster among the group of tumors with somatic alterations significantly associated with shorten T/N TL including Chr22q Loss, Chr9p/q Loss or HLA LOH.

Extended Data Fig. 8 Homologous recombination deficiency (HRD) in Sherlock-Lung.

a, HRDetect scores of Sherlock-Lung samples. HRD-high: >0.7, HRD-low: < 0.005. b, Comparison of the number of total indels, microhomology deletions, SVs, and SNVs between samples with HRDetect score below 0.7 (group N) and above 0.7 (group Y). P-values are calculated using the two-sided Mann-Whitney U test. For box plots, center lines show the medians; box limits indicate the 25th and 75th percentiles; whiskers extend 1.5 times the interquartile range from the 25th and 75th percentiles.

Extended Data Fig. 9 Genomic alterations in HRD associated genes in Sherlock-Lung.

a, Oncoplot of genomic alterations in HRD associated genes, including germline mutations, somatic mutations and LOH. Samples with biallelic alterations are represented by bars with two different colors. The bottom bar shows tumor histological types. b, Boxplots of HRDetect scores (top) and SBS mutation loads (bottom) in tumors with and without LOH of six HR associated genes. FDR are calculated using the two-sided Mann-Whitney U test with multiple testing correction based on the Benjamini & Hochberg method. For box plots, center lines show the medians; box limits indicate the 25th and 75th percentiles; whiskers extend 1.5 times the interquartile range from the 25th and 75th percentiles.

Supplementary information

Supplementary Information

Supplementary Notes, Methods and Figs. 1–38.

Reporting Summary

Peer Review Information

Supplementary Tables

Supplementary Tables 1–8

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, T., Joubert, P., Ansari-Pour, N. et al. Genomic and evolutionary classification of lung cancer in never smokers. Nat Genet 53, 1348–1359 (2021). https://doi.org/10.1038/s41588-021-00920-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00920-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research