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:

Genetics and Genomics

Stepwise evolutionary genomics of early-stage lung adenocarcinoma manifesting as pure, heterogeneous and part-solid ground-glass nodules

Abstract

Background

This study was designed to unravel the genomic landscape and evolution of early-stage subsolid lung adenocarcinomas (SSN-LUADs) manifesting as pure ground-glass nodules (pGGNs), heterogeneous ground-glass nodules (HGGNs) and part-solid nodules (PSNs).

Methods

Samples subjected to either broad-panel next-generation sequencing (NGS) or whole-exome sequencing (WES) were included. Clinicopathologic and genomic features were compared among pGGN, HGGN and PSN, while tumour evolutionary trajectories and mutational signatures were evaluated in the entire cohort.

Results

In total, 247 SSN-LUAD samples subjected to broad-panel NGS and 125 to WES were identified. Compared with PSNs, HGGNs had significantly lower tumour mutation count (P < 0.001), genomic alteration count (P < 0.001), and intra-tumour heterogeneity (P = 0.005). Statistically significant upward trends were observed in alterations involving driver mutations and oncogenic pathways from pGGNs to HGGNs to PSNs. EGFR mutation was proved to be a key early event in the progression of SSN-LUADs, with subsequently two evolutionary trajectories involving either RBM10 or TP53 mutation in the cancer-evolution models.

Conclusions

This study provided evidence for unravelling the previously unknown genomic underpinnings associated with SSN-LUAD evolution from pGGN to HGGN to PSN, proving that HGGN was an intermediate SSN form between pGGN and PSN genetically.

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: Association between radiological subtype and genomic features in the broad-panel NGS cohort.
Fig. 2: Analyses of driver genes detected by dNdScv algorithm in the broad-panel NGS cohort.
Fig. 3: Oncogenic pathway and therapeutic actionability analyses in the broad-panel NGS cohort.
Fig. 4: Cancer-evolution models of early-stage SSN-LUADs.
Fig. 5: Mutational signature analyses in the WES cohort.

Similar content being viewed by others

Data availability

All data are available from the authors upon reasonable request.

References

  1. Zhang Y, Jheon S, Li H, Zhang H, Xie Y, Qian B, et al. Results of low-dose computed tomography as a regular health examination among Chinese hospital employees. J Thorac Cardiovasc Surg. 2020;160:824–31.

  2. Kim YW, Kwon BS, Lim SY, Lee YJ, Park JS, Cho YJ, et al. Lung cancer probability and clinical outcomes of baseline and new subsolid nodules detected on low-dose CT screening. Thorax. 2021;76:980–8.

  3. Sawada S, Yamashita N, Sugimoto R, Ueno T, Yamashita M. Long-term outcomes of patients with ground-glass opacities detected using CT scanning. Chest. 2017;151:308–15.

  4. Travis WD, Asamura H, Bankier AA, Beasley MB, Detterbeck F, Flieder DB, et al. The IASLC lung cancer staging project: proposals for coding T categories for Subsolid Nodules and assessment of tumor size in part-solid tumors in the forthcoming eighth edition of the TNM classification of lung cancer. J Thorac Oncol. 2016;11:1204–23.

  5. MacMahon H, Naidich DP, Goo JM, Lee KS, Leung A, Mayo JR, et al. Guidelines for management of incidental pulmonary nodules detected on CT images: from the Fleischner Society 2017. Radiology. 2017;284:228–43.

  6. Asamura H, Hishida T, Suzuki K, Koike T, Nakamura K, Kusumoto M, et al. Radiographically determined noninvasive adenocarcinoma of the lung: survival outcomes of Japan Clinical Oncology Group 0201. J Thorac Cardiovasc Surg. 2013;146:24–30.

  7. Bankier AA, MacMahon H, Goo JM, Rubin GD, Schaefer-Prokop CM, Naidich DP. Recommendations for measuring pulmonary nodules at CT: A statement from the Fleischner Society. Radiology. 2017;285:584–600.

  8. Kakinuma R, Noguchi M, Ashizawa K, Kuriyama K, Maeshima AM, Koizumi N, et al. Natural history of pulmonary subsolid nodules: a prospective multicenter study. J Thorac Oncol. 2016;11:1012–28.

  9. Yin J, Xi J, Liang J, Zhan C, Jiang W, Lin Z, et al. Solid components in the mediastinal window of computed tomography define a distinct subtype of subsolid nodules in clinical stage I lung cancers. Clin Lung Cancer. 2021;22:324–31.

  10. Li Y, Li X, Li H, Zhao Y, Liu Z, Sun K, et al. Genomic characterisation of pulmonary subsolid nodules: mutational landscape and radiological features. Eur Respir J. 2020;55. https://doi.org/10.1183/13993003.01409-2019

  11. Wang Y, Yu M, Yang JX, Cao DY, Zhang Y, Zhou HM, et al. Genomic comparison of endometrioid endometrial carcinoma and its precancerous lesions in Chinese patients by high-depth next generation sequencing. Front Oncol. 2019;9:123.

  12. Cheng Y, Zhang Y, Yuan Y, Wang J, Liu K, Yu B, et al. The comprehensive analyses of genomic variations and assessment of TMB and PD-L1 expression in Chinese lung adenosquamous carcinoma. Front Genet. 2020;11:1794.

  13. Xiao W, Zhang G, Chen B, Chen X, Wen L, Lai J, et al. Characterization of frequently mutated cancer genes and tumor mutation burden in Chinese breast cancer. Front Oncol. 2021;11:1107.

  14. Mroz EA, Rocco JW. A novel measure of intratumor genetic heterogeneity, is high in poor-outcome classes of head and neck squamous cell carcinoma. Oral Oncol. 2013;49:211–5.

  15. Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013; https://doi.org/10.1038/nature12213

  16. Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173:371–85.

  17. Network CGAR. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–50.

  18. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell. 2018;173:321–37.

  19. Zhou J, Sanchez-Vega F, Caso R, Tan KS, Brandt WS, Jones GD, et al. Analysis of tumor genomic pathway alterations using broad-panel next-generation sequencing in surgically resected lung adenocarcinoma. Clin Cancer Res. 2019;14:2763–7.

  20. Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017; https://doi.org/10.1200/PO.17.00011

  21. Ramazzotti D, Caravagna G, Olde Loohuis L, Graudenzi A, Korsunsky I, Mauri G, et al. CAPRI: efficient inference of cancer progression models from cross-sectional data. Bioinformatics. 2015;31:3016–26.

  22. De Sano L, Caravagna G, Ramazzotti D, Graudenzi A, Mauri G, Mishra B, et al. TRONCO: an R package for the inference of cancer progression models from heterogeneous genomic data. Bioinformatics. 2016;32:1911–3.

  23. Caravagna G, Graudenzi A, Ramazzotti D, Sanz-Pamplona R, De Sano L, Mauri G, et al. Algorithmic methods to infer the evolutionary trajectories in cancer progression. Proc Natl Acad Sci USA. 2016;113:E4025–34.

  24. Brunet JP, Tamayo P, Golub TR, Mesirov JP. Metagenes and molecular pattern discovery using matrix factorization. Proc Natl Acad Sci USA. 2004;101:4164–9.

  25. Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix factorization. BMC Bioinformatics. 2010;11:1–9.

  26. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;534:47–54.

  27. Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ, Stratton MR. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 2013;35:951–9.

  28. Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26:1572–3.

  29. Kucab JE, Zou X, Morganella S, Joel M, Nanda AS, Nagy E, et al. A compendium of mutational signatures of environmental agents. Cell. 2019;177:821–36.

  30. Yan D, Wu S, Zhou S, Tong G, Li F, Wang Y, et al. Characteristics, sources and health risk assessment of airborne particulate PAHs in Chinese cities: a review. Environ Pollut. 2019;248:804–14.

  31. Chen YJ, Roumeliotis TI, Chang YH, Chen CT, Han CL, Lin MH, et al. Proteogenomics of non-smoking lung cancer in East Asia delineates molecular signatures of pathogenesis and progression. Cell. 2020;182:226–44.

  32. Myers R, Brauer M, Dummer T, Atkar-Khattra S, Yee J, Melosky B, et al. High-ambient air pollution exposure among never smokers versus ever smokers with lung cancer. J Thorac Oncol. 2021;16:1850–8.

Download references

Funding

This work was financially supported by the National Natural Science Foundation of China (grant 82002410, HL), the Major Research Plan of National Natural Science of China (grant 92059203, JW) and Peking University People’s Hospital Scientific Research Development Funds (grant RDY2020-02, HL).

Author information

Authors and Affiliations

Authors

Contributions

HL: conceptualisation, project administration, formal analysis and writing—original draft; ZS: methodology, software, formal analysis and writing—original draft; RX: formal analysis and writing—original draft; QQ: resources, investigation and data curation; XL: project administration and supervision; HH: methodology, software, formal analysis and visualisation; XW: investigation; JZ: investigation; ZW: investigation; PY: investigation; FY: project administration and supervision; JW: project administration and supervision.

Corresponding authors

Correspondence to Xiao Li or Jun Wang.

Ethics declarations

Competing interests

HH and KL were employed by the company Berry Oncology, Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Ethics approval and consent to participate

This study was approved by the Institutional Review Board at Peking University People’s Hospital (2020PHB363-01). The study was performed in accordance with the Declaration of Helsinki.

Consent to publish

No individual person’s data is presented in this manuscript.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, H., Sun, Z., Xiao, R. et al. Stepwise evolutionary genomics of early-stage lung adenocarcinoma manifesting as pure, heterogeneous and part-solid ground-glass nodules. Br J Cancer 127, 747–756 (2022). https://doi.org/10.1038/s41416-022-01821-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-01821-7

Search

Quick links