Simultaneous evolutionary expansion and constraint of genomic heterogeneity in multifocal lung cancer

Pengfei Ma, Yujie Fu, Mei-Chun Cai, Ying Yan, Ying Jing, Shengzhe Zhang, Minjiang Chen, Jie Wu, Ying Shen, Liang Zhu, Hong-Zhuan Chen, Wei-Qiang Gao, Mengzhao Wang, Zhenyu Gu, Trever G Bivona, Xiaojing Zhao, Guanglei Zhuang, Pengfei Ma, Yujie Fu, Mei-Chun Cai, Ying Yan, Ying Jing, Shengzhe Zhang, Minjiang Chen, Jie Wu, Ying Shen, Liang Zhu, Hong-Zhuan Chen, Wei-Qiang Gao, Mengzhao Wang, Zhenyu Gu, Trever G Bivona, Xiaojing Zhao, Guanglei Zhuang

Abstract

Recent genomic analyses have revealed substantial tumor heterogeneity across various cancers. However, it remains unclear whether and how genomic heterogeneity is constrained during tumor evolution. Here, we sequence a unique cohort of multiple synchronous lung cancers (MSLCs) to determine the relative diversity and uniformity of genetic drivers upon identical germline and environmental background. We find that each multicentric primary tumor harbors distinct oncogenic alterations, including novel mutations that are experimentally demonstrated to be functional and therapeutically targetable. However, functional studies show a strikingly constrained tumorigenic pathway underlying heterogeneous genetic variants. These results suggest that although the mutation-specific routes that cells take during oncogenesis are stochastic, genetic trajectories may be constrained by selection for functional convergence on key signaling pathways. Our findings highlight the robust evolutionary pressures that simultaneously shape the expansion and constraint of genomic diversity, a principle that holds important implications for understanding tumor evolution and optimizing therapeutic strategies.Across cancer types tumor heterogeneity has been observed, but how this relates to tumor evolution is unclear. Here, the authors sequence multiple synchronous lung cancers, highlighting the evolutionary pressures that simultaneously shape the expansion and constraint of genomic heterogeneity.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
Clonal architecture and mutational landscape of MSLC. a Computed tomography (CT) diagnosis and clonal architecture of multifocal tumors in four MSLC patients. Heat maps showed the presence (blue) or absence (gray) of all non-silent somatic mutations in every tumor region. Phylogenetic trees indicated the clonal structure of sequenced tumor regions in each patient. Scale bar, 1 cm. b Mutational landscape of all 16 sequenced tumor regions. Putative driver genes with somatic mutations were classified according to the functional categories. c Frequencies for each of the six substitutions at all 16 possible trinucleotide contexts were displayed in a heat map. All specimens were separated into two major clusters based on the mutational signatures
Fig. 2
Fig. 2
Interfocal and intrafocal genetic heterogeneity of MSLC. a A schematic overview of tumor heterogeneity analysis at interpatient, interfocal and intrafocal levels. b Mutation spectra of four MSLC patients were summarized by pie charts and shared mutations between different subjects were showed in Venn diagram. c Overexpression of indicated wild-type or mutant genes in BEAS-2B or LXF-289 cells. Gene expression and ERK phosphorylation were measured by immunoblotting. d Indicated wild-type or mutant genes were lentivirally introduced into BEAS-2B or LXF-289 cells. Cells were cultured for two weeks and stained with crystal violet. Scale bar, 5 mm. e Tumor growth of BEAS-2B xenografts that ectopically expressed indicated mutant genes. Each line represented mean tumor volume of the respective group, and error bars indicated standard deviation (10 mice per group). Scale bar, 10 mm. f A heat map presented the CCF of putative driver mutations in each sequenced region of the MSLC tumors. g Mutation spectra of early mutations (trunk) and late mutations (branch), and mutational signatures of different tumor regions in M-seq samples of RJLC1-T1 and RJLC4-T1
Fig. 3
Fig. 3
Constrained tumorigenic pathways among multicentric lesions. a A summary of the most prominent driver mutation identified in each primary tumor of MSLC. b EGFR or KRAS was knocked out using CRISPR-Cas9 system in lung cancer cell lines and replaced with HER2, c-MET, ARAF, BRAF, or MEK mutants. c EGFR was knocked out using CRISPR-Cas9 system in PC9 cells and replaced with HER2YMVA. AKT and ERK phosphorylation were measured by western blot analysis. d Cells were treated with a serial dilution of indicated inhibitors for a week and stained with crystal violet. Scale bar, 5 mm. The corresponding cell lysates were analyzed by immunoblotting. e KRAS was knocked out using CRISPR-Cas9 system in NCI-H1944 cells and replaced with BRAFD594G, c-METΔE14, ARAFS214C, or MEK1E102-I103 del. ERK phosphorylation was measured by western blot analysis. Indicated cells were treated with a serial dilution of trametinib for a week and stained with crystal violet. Scale bar, 5 mm. The corresponding cell lysates were analyzed by immunoblotting
Fig. 4
Fig. 4
Independent validation of oncogenic pathway convergence. a CT diagnosis of synchronous lung and breast tumors in RJLC5 and RJLC6. Scale bar, 1 cm. b Circos plots highlighting different structural variations of lung and breast tumors in RJLC5 and RJLC6. RJLC5-Lung harbored c-METΔE14 and RJLC5-Breast harbored HER2Y777L; RJLC6-Lung harbored EGFR-KDD and RJLC6-Breast harbored HER4N855K. c CT diagnosis of MSLC and circos plots representation of somatic mutations in RJLC7. EGFRL858R identified in RJLC7-T1 was confirmed using ARMS PCR. Scale bar, 1 cm. d Time line of RJLC7 MSLC diagnosis and treatment. Baseline and 21-month CT scan for RJLC7 following treatment with gefitinib. Scale bar, 1 cm. e A schematic model showing that simultaneous evolutionary expansion and constraint of genomic heterogeneity collaboratively shape tumorigenic process and clonal architecture

References

    1. Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–28. doi: 10.1126/science.959840.
    1. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–313. doi: 10.1038/nature10762.
    1. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability--an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 2010;11:220–228. doi: 10.1038/nrm2858.
    1. Cahill DP, Kinzler KW, Vogelstein B, Lengauer C. Genetic instability and darwinian selection in tumours. Trends Cell Biol. 1999;9:M57–M60. doi: 10.1016/S0962-8924(99)01661-X.
    1. Gerlinger M, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205.
    1. Sottoriva A, et al. A big bang model of human colorectal tumor growth. Nat. Genet. 2015;47:209–216. doi: 10.1038/ng.3214.
    1. Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014;346:256–259. doi: 10.1126/science.1256930.
    1. de Bruin EC, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014;346:251–256. doi: 10.1126/science.1253462.
    1. Yates LR, et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med. 2015;21:751–759. doi: 10.1038/nm.3886.
    1. Hao JJ, et al. Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat. Genet. 2016;48:1500–1507. doi: 10.1038/ng.3683.
    1. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015;27:15–26. doi: 10.1016/j.ccell.2014.12.001.
    1. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501:338–345. doi: 10.1038/nature12625.
    1. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat. Rev. Cancer. 2012;12:323–334. doi: 10.1038/nrc3261.
    1. Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Annu. Rev. Pathol. 2013;8:277–302. doi: 10.1146/annurev-pathol-020712-163923.
    1. Greaves M. Evolutionary determinants of cancer. Cancer Discov. 2015;5:806–820. doi: 10.1158/-15-0439.
    1. Gerlinger M, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 2014;46:225–233. doi: 10.1038/ng.2891.
    1. Kumar A, et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 2016;22:369–378. doi: 10.1038/nm.4053.
    1. Suzuki H, et al. Mutational landscape and clonal architecture in grade II and III gliomas. Nat. Genet. 2015;47:458–468. doi: 10.1038/ng.3273.
    1. Juric D, et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature. 2015;518:240–244. doi: 10.1038/nature13948.
    1. Fisher R, et al. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15:433. doi: 10.1186/s13059-014-0433-z.
    1. Martini N, Melamed MR. Multiple primary lung cancers. J. Thorac. Cardiovasc. Surg. 1975;70:606–612.
    1. Gazdar AF, Minna JD. Multifocal lung cancers--clonality vs field cancerization and does it matter? J. Natl Cancer Inst. 2009;101:541–543. doi: 10.1093/jnci/djp059.
    1. Rostad H, Strand TE, Naalsund A, Norstein J. Resected synchronous primary malignant lung tumors: a population-based study. Ann. Thorac. Surg. 2008;85:204–209. doi: 10.1016/j.athoracsur.2007.07.091.
    1. Flieder DB, et al. Pathologic findings of lung tumors diagnosed on baseline CT screening. Am. J. Surg. Pathol. 2006;30:606–613. doi: 10.1097/01.pas.0000202040.51967.d0.
    1. Trousse D, et al. Synchronous multiple primary lung cancer: an increasing clinical occurrence requiring multidisciplinary management. J. Thorac. Cardiovasc. Surg. 2007;133:1193–1200. doi: 10.1016/j.jtcvs.2007.01.012.
    1. Murphy SJ, et al. Identification of independent primary tumors and intrapulmonary metastases using DNA rearrangements in non-small-cell lung cancer. J. Clin. Oncol. 2014;32:4050–4058. doi: 10.1200/JCO.2014.56.7644.
    1. Wang X, et al. Evidence for common clonal origin of multifocal lung cancers. J. Natl Cancer Inst. 2009;101:560–570. doi: 10.1093/jnci/djp054.
    1. Liu Y, et al. Genomic heterogeneity of multiple synchronous lung cancer. Nat. Commun. 2016;7:13200. doi: 10.1038/ncomms13200.
    1. Swanton C, Govindan R. Clinical implications of genomic discoveries in lung cancer. N. Engl. J. Med. 2016;374:1864–1873. doi: 10.1056/NEJMra1504688.
    1. Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–550. doi: 10.1038/nature13385.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Rosenthal R, McGranahan N, Herrero J, Taylor BS, Swanton C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 2016;17:31. doi: 10.1186/s13059-016-0893-4.
    1. Helleday T, Eshtad S, Nik-Zainal S. Mechanisms underlying mutational signatures in human cancers. Nat. Rev. Genet. 2014;15:585–598. doi: 10.1038/nrg3729.
    1. Paik PK, et al. Response to MET inhibitors in patients with stage IV lung adenocarcinomas harboring MET mutations causing exon 14 skipping. Cancer Discov. 2015;5:842–849. doi: 10.1158/-14-1467.
    1. Frampton GM, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015;5:850–859. doi: 10.1158/-15-0285.
    1. Imielinski M, et al. Oncogenic and sorafenib-sensitive ARAF mutations in lung adenocarcinoma. J. Clin. Invest. 2014;124:1582–1586. doi: 10.1172/JCI72763.
    1. Heidorn SJ, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 2010;140:209–221. doi: 10.1016/j.cell.2009.12.040.
    1. Roth A, et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods. 2014;11:396–398. doi: 10.1038/nmeth.2883.
    1. Miller CA, et al. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput. Biol. 2014;10:e1003665. doi: 10.1371/journal.pcbi.1003665.
    1. Gerlinger M, et al. Cancer: evolution within a lifetime. Annu. Rev. Genet. 2014;48:215–236. doi: 10.1146/annurev-genet-120213-092314.
    1. Greaves M. Darwinian medicine: a case for cancer. Nat. Rev. Cancer. 2007;7:213–221. doi: 10.1038/nrc2071.
    1. Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature. 2009;458:719–724. doi: 10.1038/nature07943.
    1. Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C. Translational implications of tumor heterogeneity. Clin. Cancer Res. 2015;21:1258–1266. doi: 10.1158/1078-0432.CCR-14-1429.
    1. Lipinski KA, et al. Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer. 2016;2:49–63. doi: 10.1016/j.trecan.2015.11.003.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. McKenna A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110.
    1. Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576. doi: 10.1101/gr.129684.111.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603.
    1. Boeva V, et al. Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics. 2012;28:423–425. doi: 10.1093/bioinformatics/btr670.
    1. Chen K, et al. BreakDancer: an algorithm for high-resolution mapping of genomic structural variation. Nat. Methods. 2009;6:677–681. doi: 10.1038/nmeth.1363.
    1. Krzywinski M, et al. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–1645. doi: 10.1101/gr.092759.109.
    1. Carter SL, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 2012;30:413–421. doi: 10.1038/nbt.2203.
    1. Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 2015;33:1152–1158. doi: 10.1038/nbt.3344.
    1. Nielsen M, Andreatta M. NetMHCpan-3.0; improved prediction of binding to MHC class I molecules integrating information from multiple receptor and peptide length datasets. Genome Med. 2016;8:33. doi: 10.1186/s13073-016-0288-x.
    1. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods. 2014;11:783–784. doi: 10.1038/nmeth.3047.

Source: PubMed

3
S'abonner