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:

Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer

Abstract

The extent of heterogeneity among driver gene mutations present in naturally occurring metastases—that is, treatment-naive metastatic disease—is largely unknown. To address this issue, we carried out 60× whole-genome sequencing of 26 metastases from four patients with pancreatic cancer. We found that identical mutations in known driver genes were present in every metastatic lesion for each patient studied. Passenger gene mutations, which do not have known or predicted functional consequences, accounted for all intratumoral heterogeneity. Even with respect to these passenger mutations, our analysis suggests that the genetic similarity among the founding cells of metastases was higher than that expected for any two cells randomly taken from a normal tissue. The uniformity of known driver gene mutations among metastases in the same patient has critical and encouraging implications for the success of future targeted therapies in advanced-stage disease.

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

Figure 1: Distributions of metastatic disease for four patients with pancreatic cancer.
Figure 2: Features of phylogenies and driver genes in Pam01, Pam02, Pam03, and Pam04.
Figure 3: Somatic evolution of normal tissues.
Figure 4: Inferred phylogeny and localization of primary tumor sections and metastases for patient Pam02.

Similar content being viewed by others

References

  1. Greaves, M. & Maley, C.C. Clonal evolution in cancer. Nature 481, 306–313 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Alizadeh, A.A. et al. Toward understanding and exploiting tumor heterogeneity. Nat. Med. 21, 846–853 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Yates, L.R. et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med. 21, 751–759 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. de Bruin, E.C. et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346, 251–256 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhang, J. et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 346, 256–259 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hong, M.K.H. et al. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 6, 6605 (2015).

    CAS  PubMed  Google Scholar 

  9. Kumar, A. et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 22, 369–378 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jones, S. et al. Comparative lesion sequencing provides insights into tumor evolution. Proc. Natl. Acad. Sci. USA 105, 4283–4288 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sanborn, J.Z. et al. Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination. Proc. Natl. Acad. Sci. USA 112, 10995–11000 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Tomasetti, C., Vogelstein, B. & Parmigiani, G. Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation. Proc. Natl. Acad. Sci. USA 110, 1999–2004 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Makohon-Moore, A. & Iacobuzio-Donahue, C.A. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat. Rev. Cancer 16, 553–565 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Barber, L.J. et al. Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor. J. Pathol. 229, 422–429 (2013).

    CAS  PubMed  Google Scholar 

  19. Diaz, L.A. Jr. et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486, 537–540 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Misale, S. et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486, 532–536 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Waddell, N. et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 518, 495–501 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Hoogstraat, M. et al. Genomic and transcriptomic plasticity in treatment-naive ovarian cancer. Genome Res. 24, 200–211 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Krasnitz, A., Sun, G., Andrews, P. & Wigler, M. Target inference from collections of genomic intervals. Proc. Natl. Acad. Sci. USA 110, E2271–E2278 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Siegel, R.L., Miller, K.D. & Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 66, 7–30 (2016).

    PubMed  Google Scholar 

  26. Embuscado, E.E. et al. Immortalizing the complexity of cancer metastasis: genetic features of lethal metastatic pancreatic cancer obtained from rapid autopsy. Cancer Biol. Ther. 4, 548–554 (2005).

    CAS  PubMed  Google Scholar 

  27. Bailey, P. et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016).

    CAS  PubMed  Google Scholar 

  28. Chang, M.T. et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat. Biotechnol. 34, 155–163 (2016).

    CAS  PubMed  Google Scholar 

  29. Douville, C. et al. CRAVAT: cancer-related analysis of variants toolkit. Bioinformatics 29, 647–648 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Iacobuzio-Donahue, C.A., Velculescu, V.E., Wolfgang, C.L. & Hruban, R.H. Genetic basis of pancreas cancer development and progression: insights from whole-exome and whole-genome sequencing. Clin. Cancer Res. 18, 4257–4265 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Borazanci, E. et al. Adenosquamous carcinoma of the pancreas: molecular characterization of 23 patients along with a literature review. World J. Gastrointest. Oncol. 7, 132–140 (2015).

    PubMed  PubMed Central  Google Scholar 

  32. Kinde, I., Wu, J., Papadopoulos, N., Kinzler, K.W. & Vogelstein, B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl. Acad. Sci. USA 108, 9530–9535 (2011).

    PubMed  PubMed Central  Google Scholar 

  33. Yachida, S. & Iacobuzio-Donahue, C.A. The pathology and genetics of metastatic pancreatic cancer. Arch. Pathol. Lab. Med. 133, 413–422 (2009).

    PubMed  Google Scholar 

  34. Olive, K.P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Campbell, P.J. et al. Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing. Proc. Natl. Acad. Sci. USA 105, 13081–13086 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Biankin, A.V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Schutte, M. et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 57, 3126–3130 (1997).

    CAS  PubMed  Google Scholar 

  39. Santarius, T., Shipley, J., Brewer, D., Stratton, M.R. & Cooper, C.S. A census of amplified and overexpressed human cancer genes. Nat. Rev. Cancer 10, 59–64 (2010).

    CAS  PubMed  Google Scholar 

  40. Maley, C.C. et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat. Genet. 38, 468–473 (2006).

    CAS  PubMed  Google Scholar 

  41. Fernández, L.C., Torres, M. & Real, F.X. Somatic mosaicism: on the road to cancer. Nat. Rev. Cancer 16, 43–55 (2016).

    PubMed  Google Scholar 

  42. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Reiter, J.G. et al. Reconstructing metastatic seeding patterns of human cancers. Nat. Commun. http://dx.doi.org/10.1038/ncomms14114 (2017).

  44. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Waclaw, B. et al. A spatial model predicts that dispersal and cell turnover limit intratumour heterogeneity. Nature 525, 261–264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Maddipati, R. & Stanger, B.Z. Pancreatic cancer metastases harbor evidence of polyclonality. Cancer Discov. 5, 1086–1097 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Gibson, W.J. et al. The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis. Nat. Genet. 48, 848–855 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Jones, P.A. & Baylin, S.B. The epigenomics of cancer. Cell 128, 683–692 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Williams, M.J., Werner, B., Barnes, C.P., Graham, T.A. & Sottoriva, A. Identification of neutral tumor evolution across cancer types. Nat. Genet. 48, 238–244 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Bozic, I., Gerold, J.M. & Nowak, M.A. Quantifying clonal and subclonal passenger mutations in cancer evolution. PLoS Comput. Biol. 12, e1004731 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Witkiewicz, A.K. et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 6, 6744 (2015).

    CAS  PubMed  Google Scholar 

  53. Jiao, Y. et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat. Genet. 45, 1470–1473 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J.R. Stat. Soc. 57, 289–300 (1995).

    Google Scholar 

  55. Salari, R. et al. Inference of tumor phylogenies with improved somatic mutation discovery. J. Comput. Biol. 20, 933–944 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. El-Kebir, M., Oesper, L., Acheson-Field, H. & Raphael, B.J. Reconstruction of clonal trees and tumor composition from multi-sample sequencing data. Bioinformatics 31, i62–i70 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Popic, V. et al. Fast and scalable inference of multi-sample cancer lineages. Genome Biol. 16, 91 (2015).

    PubMed  PubMed Central  Google Scholar 

  58. Boeva, V. et al. Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics 28, 423–425 (2012).

    CAS  PubMed  Google Scholar 

  59. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Lappalainen, I. et al. The European Genome-phenome Archive of human data consented for biomedical research. Nat. Genet. 47, 692–695 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Memorial Sloan Kettering Cancer Center Molecular Cytology core facility for immunohistochemistry staining. This work was supported by Office of Naval Research grant N00014-16-1-2914, the Bill and Melinda Gates Foundation (OPP1148627), and a gift from B. Wu and E. Larson (M.A.N.), National Institutes of Health grants CA179991 (C.A.I.-D. and I.B.), F31 CA180682 (A.P.M.-M.), CA43460 (B.V.), and P50 CA62924, the Monastra Foundation, the Virginia and D.K. Ludwig Fund for Cancer Research, the Lustgarten Foundation for Pancreatic Cancer Research, the Sol Goldman Center for Pancreatic Cancer Research, the Sol Goldman Sequencing Center, ERC Start grant 279307: Graph Games (J.G.R., D.K., and C.K.), Austrian Science Fund (FWF) grant P23499-N23 (J.G.R., D.K., and C.K.), and FWF NFN grant S11407-N23 RiSE/SHiNE (J.G.R., D.K., and C.K.).

Author information

Authors and Affiliations

Authors

Contributions

C.I.D. and A.M.M. performed the autopsies. C.I.D., A.P.M.-M., R.H.H., L.D.W., B.V., K.W.K., N.P., M.Z., F.W., and Y.J. designed experiments. A.M.M., J.R., I.B., F.W., J.H., and M.A. performed biostatistical analyses. A.M.M., M.Z., B.J., and Z.A.K. performed the experiments. J.G.R., I.B., J.H., D.K., and K.C. performed computational analysis. J.R., I.B., B.A., and M.A.N. performed modeling. All authors interpreted the data. C.A.I.-D., A.M.M., and B.V. wrote the manuscript, J.R., I.B., and M.A.N. provided input to the manuscript, and all authors read and approved the final manuscript.

Corresponding author

Correspondence to Christine A Iacobuzio-Donahue.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Hierarchical clustering for four cases.

Samples are indicated along the y axis, and variants are listed along the top of the diagram. Colors correspond to discrete tumor samples and follow the rainbow spectrum from the Treeomics phylogenies, scaling from ancestral to descendant, as indicated by the evolutionary relationships. Samples that are uncolored represent those analyzed by hierarchical clustering only. The variant status in each sample is shown in blue (for present), dark red (for absent), or light red (for unknown due to low coverage). Hierarchical clustering using UPGMA (unweighted pair group method with arithmetic mean) is indicated on the right y axes. Primary tumors are labeled as “PT” followed by a number, lymph node metastases are labeled as “NoM” followed by a number, liver metastases are labeled as “LiM” followed by a number, lung metastases are labeled as “LuM” followed by a number, and peritoneal metastases are labeled as “PeM” followed by a number. (a) Pam01. (b) Pam02. (c) Pam03. (d) Pam04.

Supplementary Figure 2 Immunohistochemical analysis of proteins encoded by four major driver genes.

Protein expression was evaluated in tumor tissues. Driver genes evaluated by immunohistochemistry are listed in columns, and tumor tissues from each case are presented in rows. In all images, red arrows point to positively staining non-neoplastic cells (internal controls) and black arrows point to positively staining nuclei in neoplastic cells. The patterns observed for the individual metastases sequenced in each patient were reproducibly observed in the primary carcinoma and other metastases of the same patient, as we have previously demonstrated59. Scale bar, 10 μm.

Supplementary Figure 3 Distributions of copy number variations in Pam02.

Circos plots showing statistically significant CNVs in Pam02 whole-genome samples. For each sample ring, the y axis spans –2 to 2, with 0 representing a normal diploid copy number in unaffected regions, deletions represented as –1 or –2, and amplifications represented as 1 or 2. CNVs of >2 were scored as 2. All values were log2 transformed for visualization. The outermost ring shows the chromosomes in clockwise order. Deletions are shown in blue, while amplifications are shown in red. Gene names are those described in Supplementary Table 7. From innermost to outermost, the samples are PT18, PT4, PT9, LiM6, LiM5, LiM2, LiM8, LiM3, LiM7, and LiM1.

Supplementary Figure 4 Distributions of copy number variations in Pam03.

Circos plots showing statistically significant CNVs in whole-genome samples. For each sample ring, the y axis goes from –2 to 2, with a central black line representing a normal diploid copy number in unaffected regions, deletions represented as –1 or –2, and amplifications represented as 1 or 2. CNVs of >2 were scored as 2. The values are log2 transformed for visualization. The outermost ring shows the chromosomes in clockwise order. Deletions are shown in blue, while amplifications are shown in red. Gene names are those described in Supplementary Table 7. The innermost ring is PT12, followed by PT10, PT11, LuM3, LiM2, LiM4, LiM5, LiM3, LiM1, LuM1, and LuM2.

Supplementary Figure 5 Distributions of copy number variations in Pam04.

Circos plots showing statistically significant CNVs in whole-genome samples. For each sample ring, the y axis goes from –2 to 2, with a central black line representing a normal diploid copy number in unaffected regions, deletions represented as –1 or –2, and amplifications represented as 1 or 2. CNVs of >2 were scored as 2. The values are log2 transformed for visualization. The outermost ring shows the chromosomes in clockwise order. Deletions are shown in blue, while amplifications are shown in red. Gene names are those described in Supplementary Table 7. The innermost ring is PT27, followed by PT2, PT26, PeM3, PeM2, PeM1, PeM6, PeM5, and PeM4.

Supplementary Figure 6 B-allele frequencies for four Pam01 metastases.

For tumors that were whole-genome sequenced, B-allele frequencies are plotted for >3,000 SNPs per chromosome. Each chromosome is aligned sequentially and colored according to the color spectrum. The y axis represents frequency; the normal range is represented by the middle blue bar. Major loss-of-heterozygosity events (black arrowheads) are observable in all metastases. The differences in the patterns of changes in B-allele frequency are likely caused by the varying neoplastic cell content in the different samples as well as other artifacts.

Supplementary Figure 7 B-allele frequencies for Pam02 primary tumor sections and metastases.

For tumors that were whole-genome sequenced, B-allele frequencies are plotted for >3,000 SNPs per chromosome. Each chromosome is aligned sequentially and colored according to the color spectrum. The y axis represents frequency; the normal range is represented by the middle blue bar. Major loss-of-heterozygosity events (black arrowheads) are observable in all primary tumor sections and metastases. The differences in the patterns of change in B-allele frequency are likely caused by the varying neoplastic cell content in the different samples as well as other artifacts.

Supplementary Figure 8 Structural variants identified in Pam01–Pam04 samples.

Each type of structural variant is assigned a distinct color. Samples are labeled along the x axis, while numbers of structural variants are shown along the y axis.

Supplementary Figure 9 Distributions of metastatic disease in the Pam13 and Pam16 patients with cancer.

Anatomical locations of the primary carcinomas and discrete metastases used for whole-exome sequencing.

Supplementary Figure 10 Inferred phylogeny of primary tumor sections and metastases for patient Pam03.

(a) Time is represented on the left axis, and divergence is represented on the x axis. Colors correspond to discrete tumor samples and follow the rainbow spectrum, scaling from ancestral to descendant, as indicated by the evolutionary relationships. The ATM mutation was originally inferred to be wild type in primary tumor section PT1; however, targeted sequencing had insufficient coverage in PT1 and hence Treeomics misplaced the mutation (shown correctly here). (b) See Supplementary Table 3 for sample identity. Primary tumors are labeled at “PT” followed by a number, and the remaining samples are metastases labeled by organ. Hypothetical subclones are indicated by “SC” followed by the subclone number. The numbers of acquired mutations are in blue with a plus sign. Percentages denote bootstrapping values. Phylogeny was inferred by Treeomics43. (c) The dimensions of the original primary tumor in centimeters. (d) Primary tumor slices are numbered according to the original sectioning and plane order. See Supplementary Table 3 for sample identity. Metastases are labeled by organ followed by a metastasis number.

Supplementary Figure 11 Inferred phylogeny of primary tumor sections and metastases for patient Pam04.

(a) Time is represented on the left axis, and divergence is represented on the x axis. Colors correspond to discrete tumor samples and follow the rainbow spectrum, scaling from ancestral to descendant, as indicated by the evolutionary relationships. The KRAS mutation was originally inferred to be wild type in primary tumor section PT1; however, targeted sequencing had insufficient coverage in PT1 and hence Treeomics misplaced the mutation (shown correctly here). (b) See Supplementary Table 3 for sample identity. Primary tumors are labeled at “PT” followed by a number, and the remaining samples are metastases labeled by organ. Hypothetical subclones are indicated by “SC” followed by the subclone number. The numbers of acquired mutations are in blue with a plus sign. Percentages denote bootstrapping values. Phylogeny was inferred by Treeomics43. (c) The dimensions of the original primary tumor in centimeters. (d) Primary tumor slices are numbered according to the original sectioning and plane order. See Supplementary Table 3 for sample identity. Metastases are labeled by organ followed by a metastasis number.

Supplementary Figure 12 Inferred phylogeny for Pam01.

Time is represented on the left axis, and divergence is represented on the x axis. See Supplementary Table 3 for sample identity. Colors correspond to discrete tumor samples and follow the rainbow spectrum, scaling from ancestral to descendant, as indicated by the evolutionary relationships. Hypothetical subclones are indicated by “SC” followed by the subclone number. The numbers of acquired mutations are in blue with a plus sign. Percentages denote bootstrapping values. Phylogeny was inferred by Treeomics43.

Supplementary Figure 13 Inferred phylogeny for Pam13.

Time is represented on the left axis, and divergence is represented on the x axis. See Supplementary Table 3 for sample identity. Colors correspond to discrete tumor samples and follow the rainbow spectrum, scaling from ancestral to descendant, as indicated by the evolutionary relationships. Hypothetical subclones are indicated by “SC” followed by the subclone number. The numbers of acquired mutations are in blue with a plus sign. Percentages denote bootstrapping values. Phylogeny was inferred by Treeomics43. The KRAS variant was visualized in every tumor sample during manual review.

Supplementary Figure 14 Inferred phylogeny for Pam16.

Time is represented on the left axis, and divergence is represented on the x axis. See Supplementary Table 3 for sample identity. Colors correspond to discrete tumor samples and follow the rainbow spectrum, scaling from ancestral to descendant, as indicated by the evolutionary relationships. Hypothetical subclones are indicated by “SC” followed by the subclone number. The numbers of acquired mutations are in blue with a plus sign. Percentages denote bootstrapping values. Phylogeny was inferred by Treeomics43.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14, Supplementary Tables 1 and 2, and Supplementary Note (PDF 2788 kb)

Supplementary Table 3

Samples analyzed. (XLSX 24 kb)

Supplementary Table 4

Average coverage per base. (XLSX 45 kb)

Supplementary Table 5

Summary of somatic copy number alterations identified in whole-genome sequencing samples. (XLSX 41 kb)

Supplementary Table 6

Major driver gene mutations identified in each patient. (XLSX 49 kb)

Supplementary Table 7

SCNAs identified in known PDAC driver genes. (XLSX 177 kb)

Supplementary Table 8

Candidate structural variants identified in Pam01. (XLSX 298 kb)

Supplementary Table 9

Candidate structural variants identified in Pam02. (XLSX 943 kb)

Supplementary Table 10

Candidate structural variants identified in Pam03. (XLSX 1079 kb)

Supplementary Table 11

Candidate structural variants identified in Pam04. (XLSX 639 kb)

Supplementary Table 12

Variants validated by targeted sequencing. (XLSX 102 kb)

Supplementary Table 13

Jaccard similarity coefficients of metastases based on stringently filtered whole-genome sequencing and whole-exome sequencing. (XLSX 46 kb)

Supplementary Table 14

Similarity coefficients of normal organs from Blokzjil et al. (XLSX 9 kb)

Supplementary Table 15

Genetic distances among metastases based on targeted sequencing. (XLSX 44 kb)

Supplementary Table 16

Jaccard similarity coefficients of metastases based on targeted sequencing (founder mutations excluded). (XLSX 44 kb)

Supplementary Table 17

Genetic distances among metastases based on whole-genome sequencing. (XLSX 47 kb)

Supplementary Table 18

Variants identified by whole-exome sequencing in the validation set. (XLSX 87 kb)

Supplementary Table 19

Primers. (XLSX 41 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Makohon-Moore, A., Zhang, M., Reiter, J. et al. Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer. Nat Genet 49, 358–366 (2017). https://doi.org/10.1038/ng.3764

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3764

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer