The molecular diversity of Luminal A breast tumors

Giovanni Ciriello, Rileen Sinha, Katherine A Hoadley, Anders S Jacobsen, Boris Reva, Charles M Perou, Chris Sander, Nikolaus Schultz, Giovanni Ciriello, Rileen Sinha, Katherine A Hoadley, Anders S Jacobsen, Boris Reva, Charles M Perou, Chris Sander, Nikolaus Schultz

Abstract

Breast cancer is a collection of diseases with distinct molecular traits, prognosis, and therapeutic options. Luminal A breast cancer is the most heterogeneous, both molecularly and clinically. Using genomic data from over 1,000 Luminal A tumors from multiple studies, we analyzed the copy number and mutational landscape of this tumor subtype. This integrated analysis revealed four major subtypes defined by distinct copy-number and mutation profiles. We identified an atypical Luminal A subtype characterized by high genomic instability, TP53 mutations, and increased Aurora kinase signaling; these genomic alterations lead to a worse clinical prognosis. Aberrations of chromosomes 1, 8, and 16, together with PIK3CA, GATA3, AKT1, and MAP3K1 mutations drive the other subtypes. Finally, an unbiased pathway analysis revealed multiple rare, but mutually exclusive, alterations linked to loss of activity of co-repressor complexes N-Cor and SMRT. These rare alterations were the most prevalent in Luminal A tumors and may predict resistance to endocrine therapy. Our work provides for a further molecular stratification of Luminal A breast tumors, with potential direct clinical implications.

Figures

Fig. 1
Fig. 1
a Schematic stratification of breast cancer subtypes based on receptor status, ER and Her2, and PAM50 mRNA-derived signatures. b The table shows statistically significant intersections between the PAM50 subtypes (arranged horizontally) and copy number-driven clusters (arranged vertically) from the METABRIC and TCGA datasets. c Average number of mutations per sample (white) and number of recurrently mutated genes (black) are shown for the four major PAM50 subtypes. Luminal tumors have fewer mutations per samples, but they tend to affect similar genes. d Boxplot statistics of disease survival is shown for deceased patients from the METABRIC dataset across the four major PAM50 subtypes. While Luminal A tumors have the longest average survival, they also have the largest diversity
Fig. 2
Fig. 2
Copy number clustering of Luminal A breast tumors. a Hierarchical clustering of copy number data from 209 Luminal A tumors from the TCGA dataset reveals four distinct patterns of alterations, plus a mixed subgroup. Chromosomes are arranged from left to right, and tumors are arranged vertically and grouped according to cluster membership. Red indicates copy number gain, blue copy number losses, with color intensity proportional to absolute copy number values. b Cluster centroids were used to classify the METABRIC dataset (721 samples). Clusters in the METABRIC dataset show similar proportions to the TCGA counterparts, and the quality of the clusters is confirmed by statistically significant IGP. c Clusters determined from the METABRIC dataset are compared with the breast cancer subtypes proposed in [12]. Lines connect clusters with non-empty overlap with a thickness proportional to the extent of overlap
Fig. 3
Fig. 3
Landscape of Luminal A somatic mutations. a An unbiased enrichment analysis shows that PIK3CA mutations are significantly enriched in the 1q/16q subgroup, MAP3K1 mutations in the Chr8-associated, and TP53 mutations in the CNH subtgroup. All recurrent Luminal A mutations are displayed (one mutated gene per row). Mutations are color coded based on type (dark blue frame-shift, splice-site, and nonsense/light blue missense) and recurrent hotspots (red). All TCGA tumors, grouped by copy-number subtype, are shown in columns, together with mutated cases from the Sanger [13], WashU [11], and Broad [16] datasets. bMAP3K1 mutations are strongly associated with a subset of the Chr8-associated cluster characterized by 8p−/8q+/16p+/16q−. The heatmap shows copy number profiles for all the Chr.8-associated samples (arranged vertically). The panel on the right shows that high level amplification in 8p11 and mutations at MAP3K1 are largely mutually exclusive and characterize distinct subgroups of tumors in this Luminal A subtype. c Most patients affected by MAP3K1 mutations have more than one mutation, suggesting bi-allelic inactivation. The plot shows all samples with at least one MAP3K1 mutation (X axis), and the actual number of MAP3K1 mutations for each sample (Y axis)
Fig. 4
Fig. 4
CNH tumors. a Survival analysis across two independent datasets shows significantly worse outcome for the CNH Luminal A tumors. b Unbiased enrichment analysis of genomic alterations found CNH tumors to be enriched for TP53 mutations, focal amplification of MYC, 5q loss, 20q gain, and depleted for PIK3CA mutations. c Differential expression analysis shows that significantly up-regulated genes in CNH tumors are enriched for regulators of mitosis and Aurora kinase pathway components. The heatmap shows all genes that are differentially expressed in CNH tumors when compared to other Luminal A samples (red indicates high expression, green low expression). Aurora kinase is a mitotic serine/threonine kinase that phosphorylates multiple proteins including PLK1 and Cdc25; it is required for CDK1 activation and regulates mitotic events
Fig. 5
Fig. 5
Pathway analysis. Altered pathways across Luminal A tumors identified by the MEMo algorithm. a MEMo identified multiple modules recapitulating Akt, MAPK, and Ras signaling. Gene activation is shown in shades of red, inactivation in shades of blue. b MEMo found network modules highlighting multiple alterations of nuclear co-repressors. Genes are arranged vertically, and altered tumors from left to right. c Nuclear co-repressors and co-activators regulate ER transcription and Tamoxifen anti-proliferative effects. d Alterations identified by MEMo compromise co-repressor activities and may predict response to therapy

References

    1. Paik S, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;351(27):2817–2826. doi: 10.1056/NEJMoa041588.
    1. Paik S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006;24(23):3726–3734. doi: 10.1200/JCO.2005.04.7985.
    1. Romond EH, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353(16):1673–1684. doi: 10.1056/NEJMoa052122.
    1. Slamon DJ, Romond EH, Perez EA (2006) Advances in adjuvant therapy for breast cancer. Clin Adv Hematol Oncol 4(3 Suppl 7): suppl 1, 4–9; discussion suppl 10; quiz 2 p following suppl 10
    1. Perez EA, et al. Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2-positive breast cancer: joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol. 2011;29(25):3366–3373. doi: 10.1200/JCO.2011.35.0868.
    1. Sorlie T, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98(19):10869–10874. doi: 10.1073/pnas.191367098.
    1. Perou CM, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. doi: 10.1038/35021093.
    1. Parker JS, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27(8):1160–1167. doi: 10.1200/JCO.2008.18.1370.
    1. Fan C, et al. Concordance among gene-expression-based predictors for breast cancer. N Engl J Med. 2006;355(6):560–569. doi: 10.1056/NEJMoa052933.
    1. TCGA Comprehensive molecular portraits of human breast tumors. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Ellis MJ, et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature. 2012;486(7403):353–360.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346–352.
    1. Stephens PJ, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486(7403):400–404.
    1. Nik-Zainal S, et al. The life history of 21 breast cancers. Cell. 2012;149(5):994–1007. doi: 10.1016/j.cell.2012.04.023.
    1. Nik-Zainal S, et al. Mutational processes molding the genomes of 21 breast cancers. Cell. 2012;149(5):979–993. doi: 10.1016/j.cell.2012.04.024.
    1. Banerji S, et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012;486(7403):405–409. doi: 10.1038/nature11154.
    1. Haque R, et al. Impact of breast cancer subtypes and treatment on survival: an analysis spanning two decades. Cancer Epidemiol Biomarkers Prev. 2012;21(10):1848–1855. doi: 10.1158/1055-9965.EPI-12-0474.
    1. Chin K, et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006;10(6):529–541. doi: 10.1016/j.ccr.2006.10.009.
    1. Russnes HG, et al. Genomic architecture characterizes tumor progression paths and fate in breast cancer patients. Sci Transl Med. 2010;2(38):38ra47. doi: 10.1126/scitranslmed.3000611.
    1. Hicks J, et al. Novel patterns of genome rearrangement and their association with survival in breast cancer. Genome Res. 2006;16(12):1465–1479. doi: 10.1101/gr.5460106.
    1. Cerami E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404. doi: 10.1158/-12-0095.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57(1):289–300.
    1. Olshen AB, et al. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5(4):557–572. doi: 10.1093/biostatistics/kxh008.
    1. Zhang J, CNTools: Convert segment data into a region by sample matrix to allow for other high level computational analyses. R package (Version 1.6.0.)
    1. Gentleman RC, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5(10):R80. doi: 10.1186/gb-2004-5-10-r80.
    1. R-Development-Core-Team (2010) R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna
    1. Kapp AV, Tibshirani R. Are clusters found in one dataset present in another dataset? Biostatistics. 2007;8(1):9–31. doi: 10.1093/biostatistics/kxj029.
    1. Dees ND, et al. MuSiC: identifying mutational significance in cancer genomes. Genome Res. 2012;22:1589–1598. doi: 10.1101/gr.134635.111.
    1. Beroukhim R, et al. Assessing the significance of chromosomal aberrations in cancer: methodology and application to glioma. Proc Natl Acad Sci USA. 2007;104(50):20007–20012. doi: 10.1073/pnas.0710052104.
    1. Dennis G, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4(5):P3. doi: 10.1186/gb-2003-4-5-p3.
    1. Ciriello G, et al. Mutual exclusivity analysis identifies oncogenic network modules. Genome Res. 2012;22(2):398–406. doi: 10.1101/gr.125567.111.
    1. Flagiello D, et al. Highly recurrent der(1;16)(q10;p10) and other 16q arm alterations in lobular breast cancer. Genes Chromosomes Cancer. 1998;23(4):300–306. doi: 10.1002/(SICI)1098-2264(199812)23:4<300::AID-GCC4>;2-N.
    1. Tsuda H, et al. der(16)t(1;16)/der(1;16) in breast cancer detected by fluorescence in situ hybridization is an indicator of better patient prognosis. Genes Chromosomes Cancer. 1999;24(1):72–77. doi: 10.1002/(SICI)1098-2264(199901)24:1<72::AID-GCC10>;2-M.
    1. van de Vijver MJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347(25):1999–2009. doi: 10.1056/NEJMoa021967.
    1. Whitfield ML, et al. Common markers of proliferation. Nat Rev Cancer. 2006;6(2):99–106. doi: 10.1038/nrc1802.
    1. Zhang A, et al. Identification of a novel family of ankyrin repeats containing cofactors for p160 nuclear receptor coactivators. J Biol Chem. 2004;279(32):33799–33805. doi: 10.1074/jbc.M403997200.
    1. Zhang A, Li CW, Chen JD. Characterization of transcriptional regulatory domains of ankyrin repeat cofactor-1. Biochem Biophys Res Commun. 2007;358(4):1034–1040. doi: 10.1016/j.bbrc.2007.05.017.
    1. Keeton EK, Brown M. Cell cycle progression stimulated by tamoxifen-bound estrogen receptor-alpha and promoter-specific effects in breast cancer cells deficient in N-CoR and SMRT. Mol Endocrinol. 2005;19(6):1543–1554. doi: 10.1210/me.2004-0395.
    1. Cutrupi S, et al. Targeting of the adaptor protein Tab 2 as a novel approach to revert tamoxifen resistance in breast cancer cells. Oncogene. 2012;31:4353–4361. doi: 10.1038/onc.2011.627.
    1. Girault I, et al. Expression analysis of estrogen receptor alpha coregulators in breast carcinoma: evidence that NCOR1 expression is predictive of the response to tamoxifen. Clin Cancer Res. 2003;9(4):1259–1266.
    1. Green AR, et al. The prognostic significance of steroid receptor co-regulators in breast cancer: co-repressor NCOR2/SMRT is an independent indicator of poor outcome. Breast Cancer Res Treat. 2008;110(3):427–437. doi: 10.1007/s10549-007-9737-y.
    1. Ellis MJ, Perou CM. The genomic landscape of breast cancer as a therapeutic roadmap. Cancer Discov. 2013;3(1):27–34. doi: 10.1158/-12-0462.
    1. Zardavas D, Baselga J, Piccart M. Emerging targeted agents in metastatic breast cancer. Nat Rev Clin Oncol. 2013;10(4):191–210. doi: 10.1038/nrclinonc.2013.29.
    1. Gautschi O, et al. Aurora kinases as anticancer drug targets. Clin Cancer Res. 2008;14(6):1639–1648. doi: 10.1158/1078-0432.CCR-07-2179.
    1. Nishida N, et al. High copy amplification of the Aurora-A gene is associated with chromosomal instability phenotype in human colorectal cancers. Cancer Biol Ther. 2007;6(4):525–533. doi: 10.4161/cbt.6.4.3817.
    1. Dar AA, et al. Aurora kinase inhibitors—rising stars in cancer therapeutics? Mol Cancer Ther. 2010;9(2):268–278. doi: 10.1158/1535-7163.MCT-09-0765.
    1. Juric D, Baselga J. Tumor genetic testing for patient selection in phase I clinical trials: the case of PI3K inhibitors. J Clin Oncol. 2012;30(8):765–766. doi: 10.1200/JCO.2011.39.6390.
    1. Small GW, et al. Mitogen-activated protein kinase phosphatase-1 is a mediator of breast cancer chemoresistance. Cancer Res. 2007;67(9):4459–4466. doi: 10.1158/0008-5472.CAN-06-2644.
    1. Bose R, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3(2):224–237. doi: 10.1158/-12-0349.
    1. Ross JS, et al. Relapsed classic E-cadherin (CDH1) mutated invasive lobular breast cancer demonstrates a high frequency of HER2 (ERBB2) gene mutations. Clin Cancer Res. 2013;19:2668–2676. doi: 10.1158/1078-0432.CCR-13-0295.
    1. Prat A, et al. Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist. 2013;18(2):123–133. doi: 10.1634/theoncologist.2012-0397.
    1. O’Toole SA, et al. Therapeutic targets in triple negative breast cancer. J Clin Pathol. 2013;66:530–542. doi: 10.1136/jclinpath-2012-201361.
    1. Pacheco JM, et al. Racial differences in outcomes of triple-negative breast cancer. Breast Cancer Res Treat. 2013;138(1):281–289. doi: 10.1007/s10549-012-2397-6.

Source: PubMed

3
Subscribe