Skip to main content

Advertisement

Log in

Genomic landscape of ductal carcinoma in situ and association with progression

  • Preclinical study
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Purpose

The detection rate of breast ductal carcinoma in situ (DCIS) has increased significantly, raising the concern that DCIS is overdiagnosed and overtreated. Therefore, there is an unmet clinical need to better predict the risk of progression among DCIS patients. Our hypothesis is that by combining molecular signatures with clinicopathologic features, we can elucidate the biology of breast cancer progression, and risk-stratify patients with DCIS.

Methods

Targeted exon sequencing with a custom panel of 223 genes/regions was performed for 125 DCIS cases. Among them, 60 were from cases having concurrent or subsequent invasive breast cancer (IBC) (DCIS + IBC group), and 65 from cases with no IBC development over a median follow-up of 13 years (DCIS-only group). Copy number alterations in chromosome 1q32, 8q24, and 11q13 were analyzed using fluorescence in situ hybridization (FISH). Multivariable logistic regression models were fit to the outcome of DCIS progression to IBC as functions of demographic and clinical features.

Results

We observed recurrent variants of known IBC-related mutations, and the most commonly mutated genes in DCIS were PIK3CA (34.4%) and TP53 (18.4%). There was an inverse association between PIK3CA kinase domain mutations and progression (Odds Ratio [OR] 10.2, p < 0.05). Copy number variations in 1q32 and 8q24 were associated with progression (OR 9.3 and 46, respectively; both p < 0.05).

Conclusions

PIK3CA kinase domain mutations and the absence of copy number gains in DCIS are protective against progression to IBC. These results may guide efforts to distinguish low-risk from high-risk DCIS.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Meijnen P, Oldenburg HS, Peterse JL et al (2008) Clinical outcome after selective treatment of patients diagnosed with ductal carcinoma in situ of the breast. Ann Surg Oncol 15:235–243

    PubMed  Google Scholar 

  2. Cutuli B, Cohen-Solal-le Nir C, de Lafontan B et al (2002) Breast-conserving therapy for ductal carcinoma in situ of the breast: the French Cancer Centers’ experience. Int J Radiat Oncol Biol Phys 53:868–879

    PubMed  Google Scholar 

  3. Wapnir IL, Dignam JJ, Fisher B et al (2011) Long-term outcomes of invasive ipsilateral breast tumor recurrences after lumpectomy in NSABP B-17 and B-24 randomized clinical trials for DCIS. J Natl Cancer Inst 103:478–488

    PubMed  PubMed Central  Google Scholar 

  4. Cuzick J, Sestak I, Pinder SE et al (2011) Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: long-term results from the UK/ANZ DCIS trial. Lancet Oncol 12:21–29

    CAS  PubMed  Google Scholar 

  5. Ernster VL, Barclay J, Kerlikowske K et al (1996) Incidence of and treatment for ductal carcinoma in situ of the breast. JAMA 275:913–918

    CAS  PubMed  Google Scholar 

  6. Fisher ER, Dignam J, Tan-Chiu E et al (1999) Pathologic findings from the National Surgical Adjuvant Breast Project (NSABP) eight-year update of Protocol B-17: intraductal carcinoma. Cancer 86:429–438

    CAS  PubMed  Google Scholar 

  7. Newburger DE, Kashef-Haghighi D, Weng Z et al (2013) Genome evolution during progression to breast cancer. Genome Res 23:1097–1108

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Weng Z, Spies N, Zhu SX et al (2015) Cell-lineage heterogeneity and driver mutation recurrence in pre-invasive breast neoplasia. Genome Med 7:28

    PubMed  PubMed Central  Google Scholar 

  9. Johnson CE, Gorringe KL, Thompson ER et al (2012) Identification of copy number alterations associated with the progression of DCIS to invasive ductal carcinoma. Breast Cancer Res Treat 133:889–898

    CAS  PubMed  Google Scholar 

  10. Rane SU, Mirza H, Grigoriadis A et al (2015) Selection and evolution in the genomic landscape of copy number alterations in ductal carcinoma in situ (DCIS) and its progression to invasive carcinoma of ductal/no special type: a meta-analysis. Breast Cancer Res Treat 153:101–121

    PubMed  Google Scholar 

  11. Kroigard AB, Larsen MJ, Laenkholm AV et al (2015) Clonal expansion and linear genome evolution through breast cancer progression from pre-invasive stages to asynchronous metastasis. Oncotarget 6:5634–5649

    PubMed  PubMed Central  Google Scholar 

  12. Afghahi A, Forgo E, Mitani AA et al (2015) Chromosomal copy number alterations for associations of ductal carcinoma in situ with invasive breast cancer. Breast Cancer Res 17:108

    PubMed  PubMed Central  Google Scholar 

  13. Troxell ML, Brunner AL, Neff T et al (2012) Phosphatidylinositol-3-kinase pathway mutations are common in breast columnar cell lesions. Mod Pathol 25:930–937

    CAS  PubMed  Google Scholar 

  14. Xu R, Perle MA, Inghirami G et al (2002) Amplification of Her-2/neu gene in Her-2/neu-overexpressing and -nonexpressing breast carcinomas and their synchronous benign, premalignant, and metastatic lesions detected by FISH in archival material. Mod Pathol 15:116–124

    PubMed  Google Scholar 

  15. Weber SC, Seto T, Olson C et al (2012) Oncoshare: lessons learned from building an integrated multi-institutional database for comparative effectiveness research. AMIA Annu Symp Proc 2012:970–978

    PubMed  PubMed Central  Google Scholar 

  16. Kurian AW, Mitani A, Desai M et al (2014) Breast cancer treatment across health care systems: linking electronic medical records and state registry data to enable outcomes research. Cancer 120:103–111

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  18. McKenna A, Hanna M, Banks E et al (2010) The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20:1297–1303

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Cibulskis K, Lawrence MS, Carter SL et al (2013) Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31:213–219

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Lai Z, Markovets A, Ahdesmaki M et al (2016) VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res 44:e108

    PubMed  PubMed Central  Google Scholar 

  21. Garrison E, Marth G (2012) Haplotype-based variant detection from short-read sequencing. arXiv:12073907

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

    PubMed  PubMed Central  Google Scholar 

  23. Gao Y, Niu Y, Wang X et al (2009) Genetic changes at specific stages of breast cancer progression detected by comparative genomic hybridization. J Mol Med (Berl) 87:145–152

    CAS  Google Scholar 

  24. R development Core Team (2015) R: a language and environment for statistical computing. The R Foundation for Statistical Computing, Vienna

    Google Scholar 

  25. Porta-Pardo E, Godzik A (2014) e-Driver: a novel method to identify protein regions driving cancer. Bioinformatics 30:3109–3114

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Porta-Pardo E, Garcia-Alonso L, Hrabe T et al (2015) A pan-cancer catalogue of cancer driver protein interaction interfaces. PLoS Comput Biol 11:e1004518

    PubMed  PubMed Central  Google Scholar 

  27. Yang F, Petsalaki E, Rolland T et al (2015) Protein domain-level landscape of cancer-type-specific somatic mutations. PLoS Comput Biol 11:e1004147

    PubMed  PubMed Central  Google Scholar 

  28. Curtis C, Shah SP, Chin SF et al (2012) The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486:346–352

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cancer Genome Atlas N (2012) Comprehensive molecular portraits of human breast tumours. Nature 490:61–70

    Google Scholar 

  30. Horlings HM, Weigelt B, Anderson EM et al (2013) Genomic profiling of histological special types of breast cancer. Breast Cancer Res Treat 142:257–269

    CAS  PubMed  Google Scholar 

  31. Ciriello G, Gatza ML, Beck AH et al (2015) Comprehensive molecular portraits of invasive lobular breast cancer. Cell 163:506–519

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Desmedt C, Zoppoli G, Gundem G et al (2016) Genomic characterization of primary invasive lobular breast cancer. J Clin Oncol 34:1872–1881

    CAS  PubMed  Google Scholar 

  33. Yates LR, Gerstung M, Knappskog S et al (2015) Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med 21:751–759

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Abba MC, Gong T, Lu Y et al (2015) A molecular portrait of high-grade ductal carcinoma in situ. Cancer Res 75:3980–3990

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kim SY, Jung SH, Kim MS et al (2015) Genomic differences between pure ductal carcinoma in situ and synchronous ductal carcinoma in situ with invasive breast cancer. Oncotarget 6:7597–7607

    PubMed  PubMed Central  Google Scholar 

  36. Pang JB, Savas P, Fellowes AP et al (2017) Breast ductal carcinoma in situ carry mutational driver events representative of invasive breast cancer. Mod Pathol 30:952–963

    CAS  PubMed  Google Scholar 

  37. Hernandez L, Wilkerson PM, Lambros MB et al (2012) Genomic and mutational profiling of ductal carcinomas in situ and matched adjacent invasive breast cancers reveals intra-tumour genetic heterogeneity and clonal selection. J Pathol 227:42–52

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Liao S, Desouki MM, Gaile DP et al (2012) Differential copy number aberrations in novel candidate genes associated with progression from in situ to invasive ductal carcinoma of the breast. Genes Chromosomes Cancer 51:1067–1078

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hwang ES, Lal A, Chen YY et al (2011) Genomic alterations and phenotype of large compared to small high-grade ductal carcinoma in situ. Hum Pathol 42:1467–1475

    CAS  PubMed  Google Scholar 

  40. Shah V, Nowinski S, Levi D et al (2017) PIK3CA mutations are common in lobular carcinoma in situ, but are not a biomarker of progression. Breast Cancer Res 19:7

    PubMed  PubMed Central  Google Scholar 

  41. Sakr RA, Weigelt B, Chandarlapaty S et al (2014) PI3 K pathway activation in high-grade ductal carcinoma in situ–implications for progression to invasive breast carcinoma. Clin Cancer Res 20:2326–2337

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhao JJ, Liu Z, Wang L et al (2005) The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol 3-kinases in human mammary epithelial cells. Proc Natl Acad Sci USA 102:18443–18448

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Zardavas D, Phillips WA, Loi S (2014) PIK3CA mutations in breast cancer: reconciling findings from preclinical and clinical data. Breast Cancer Res 16:201

    PubMed  PubMed Central  Google Scholar 

  44. Fry EA, Taneja P, Inoue K (2017) Oncogenic and tumor-suppressive mouse models for breast cancer engaging HER2/neu. Int J Cancer 140:495–503

    CAS  PubMed  Google Scholar 

  45. Ang DC, Warrick AL, Shilling A et al (2014) Frequent phosphatidylinositol-3-kinase mutations in proliferative breast lesions. Mod Pathol 27:740–750

    CAS  PubMed  Google Scholar 

  46. Allred DC, Clark GM, Molina R et al (1992) Overexpression of HER-2/neu and its relationship with other prognostic factors change during the progression of in situ to invasive breast cancer. Hum Pathol 23:974–979

    CAS  PubMed  Google Scholar 

  47. Latta EK, Tjan S, Parkes RK et al (2002) The role of HER2/neu overexpression/amplification in the progression of ductal carcinoma in situ to invasive carcinoma of the breast. Mod Pathol 15:1318–1325

    CAS  PubMed  Google Scholar 

  48. Jang M, Kim E, Choi Y et al (2012) FGFR1 is amplified during the progression of in situ to invasive breast carcinoma. Breast Cancer Res 14:R115

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Park K, Han S, Kim HJ et al (2006) HER2 status in pure ductal carcinoma in situ and in the intraductal and invasive components of invasive ductal carcinoma determined by fluorescence in situ hybridization and immunohistochemistry. Histopathology 48:702–707

    CAS  PubMed  Google Scholar 

  50. Borgquist S, Zhou W, Jirstrom K et al (2015) The prognostic role of HER2 expression in ductal breast carcinoma in situ (DCIS); a population-based cohort study. BMC Cancer 15:468

    PubMed  PubMed Central  Google Scholar 

  51. Hoque A, Sneige N, Sahin AA et al (2002) Her-2/neu gene amplification in ductal carcinoma in situ of the breast. Cancer Epidemiol Biomark Prev 11:587–590

    CAS  Google Scholar 

  52. Miron A, Varadi M, Carrasco D et al (2010) PIK3CA mutations in in situ and invasive breast carcinomas. Cancer Res 70:5674–5678

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Dunlap J, Le C, Shukla A et al (2010) Phosphatidylinositol-3-kinase and AKT1 mutations occur early in breast carcinoma. Breast Cancer Res Treat 120:409–418

    CAS  PubMed  Google Scholar 

  54. Gorringe KL, Hunter SM, Pang JM et al (2015) Copy number analysis of ductal carcinoma in situ with and without recurrence. Mod Pathol 28:1174–1184

    CAS  PubMed  Google Scholar 

  55. Bhat-Nakshatri P, Goswami CP, Badve S et al (2016) Molecular insights of pathways resulting from two common PIK3CA mutations in breast cancer. Cancer Res 76:3989–4001

    CAS  PubMed  Google Scholar 

  56. Dogruluk T, Tsang YH, Espitia M et al (2015) Identification of variant-specific functions of PIK3CA by rapid phenotyping of rare mutations. Cancer Res 75:5341–5354

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Short-read sequencing assays were performed by the OHSU Massively Parallel Sequencing Shared Resource. We thank Norman Cyr for his artistic contribution to Fig. 2.

Funding

This work is supported by NIH R01CA193694, the Susan and Richard Levy Gift Fund; the Suzanne Pride Bryan Fund for Breast Cancer Research; the Breast Cancer Research Foundation; the Jan Weimer Junior Faculty Chair in Breast Oncology; the BRCA Foundation; and the National Cancer Institute’s Surveillance, Epidemiology and End Results Program under contract HHSN261201000140C awarded to the Cancer Prevention Institute of California. The project was supported by an NIH CTSA award number UL1 RR025744. The collection of cancer incidence data used in this study was supported by the California Department of Health Services as part of the statewide cancer reporting program mandated by California Health and Safety Code Section 103885; the National Cancer Institute’s Surveillance, Epidemiology, and End Results Program under contract HHSN261201000140C awarded to the Cancer Prevention Institute of California, contract HHSN261201000035C awarded to the University of Southern California, and contract HHSN261201000034C awarded to the Public Health Institute; and the Centers for Disease Control and Prevention’s National Program of Cancer Registries, under agreement #1U58 DP000807-01 awarded to the Public Health Institute. The ideas and opinions expressed herein are those of the authors, and endorsements by the University or State of California, the California Department of Health Services, the National Cancer Institute, or the Centers for Disease Control and Prevention or their contractors and subcontractors are neither intentional nor should be inferred.

Author information

Authors and Affiliations

Authors

Contributions

RBW conceived and designed the study. CYL conceived and carried out experiments, as well as analyzed the data. S Vennam and HS analyzed the sequencing data (S Venname: single-nucleotide alteration; HS: copy number variations). EL and S Varma carried out the FISH experiments. NJW carried out the targeted exon sequencing. NP, SH, MD, and TS performed biostatistics analysis. MLT conceived the study. AK developed and led the Oncoshare data resource. All authors were involved in writing the paper and gave final approval to the submitted and published versions.

Corresponding author

Correspondence to Robert B. West.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interests.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. As stated in the Materials and Methods section, the study was performed with Health Insurance Portability and Accountability Act (HIPAA)-compliant Stanford University Institutional Review Board (IRB) approval (Protocol number 32496). Because archival tissue was used, a waiver of consent was obtained.

Additional information

Publisher's Note

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

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, CY., Vennam, S., Purington, N. et al. Genomic landscape of ductal carcinoma in situ and association with progression. Breast Cancer Res Treat 178, 307–316 (2019). https://doi.org/10.1007/s10549-019-05401-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-019-05401-x

Keywords

Navigation