Molecular features and survival outcomes of the intrinsic subtypes within HER2-positive breast cancer

Aleix Prat, Lisa A Carey, Barbara Adamo, Maria Vidal, Josep Tabernero, Javier Cortés, Joel S Parker, Charles M Perou, José Baselga, Aleix Prat, Lisa A Carey, Barbara Adamo, Maria Vidal, Josep Tabernero, Javier Cortés, Joel S Parker, Charles M Perou, José Baselga

Abstract

Background: The clinical impact of the biological heterogeneity within HER2-positive (HER2+) breast cancer is not fully understood. Here, we evaluated the molecular features and survival outcomes of the intrinsic subtypes within HER2+ breast cancer.

Methods: We interrogated The Cancer Genome Atlas (n = 495) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets (n = 1730) of primary breast cancers for molecular data derived from DNA, RNA and protein, and determined intrinsic subtype. Clinical HER2 status was defined according to American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines or DNA copy-number aberration by single nucleotide polymorphism arrays. Cox models tested the prognostic significance of each variable in patients not treated with trastuzumab (n = 1711).

Results: Compared with clinically HER2 (cHER2)-negative breast cancer, cHER2+ breast cancer had a higher frequency of the HER2-enriched (HER2E) subtype (47.0% vs 7.1%) and a lower frequency of Luminal A (10.7% vs 39.0%) and Basal-like (14.1% vs 23.4%) subtypes. The likelihood of cHER2-positivity in HER2E, Luminal B, Basal-like and Luminal A subtypes was 64.6%, 20.0%, 14.4% and 7.3%, respectively. Within each subtype, only 0.3% to 3.9% of genes were found differentially expressed between cHER2+ and cHER2-negative tumors. Within cHER2+ tumors, HER2 gene and protein expression was statistically significantly higher in the HER2E and Basal-like subtypes than either luminal subtype. Neither cHER2 status nor the new 10-subtype copy number-based classification system (IntClust) added independent prognostic value to intrinsic subtype.

Conclusions: When the intrinsic subtypes are taken into account, cHER2-positivity does not translate into large changes in the expression of downstream signaling pathways, nor does it affect patient survival in the absence of HER2 targeting.

© The Author 2014. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
Distribution of the intrinsic molecular subtypes of breast cancer in clinical HER2 status (cHER2)-negative and cHER2+ disease in the combined the cancer genome atlas and molecular taxonomy of breast cancer international consortium dataset. cHER2 = clinical HER2 status.
Figure 2.
Figure 2.
Gene expression patterns of the intrinsic molecular subtypes of breast cancer based on clinical HER2 status (cHER2) status in the molecular taxonomy of breast cancer international consortium (METABRIC) dataset. A) Relative transcript abundance of estrogen receptor gene (ESR1) and HER2 gene (ERBB2) across the intrinsic subtypes and based on HER2 status. The boxes represent the interquartile range (25th and 75th percentiles), and the horizontal line in the box represents the median value. The whiskers show the range of largest and smallest values. B) Hierarchical clustering of the cHER2+ and cHER2-negative intrinsic subtypes (total of 8 classes) with 13497 most variable genes in the METABRIC dataset. Sample and gene expression data from tumor samples of the same subtype, and of the same cHER2 status, have been combined into a single category. For each gene in a class, we calculated the standardized mean difference between the gene′s expression in that class, vs its overall mean expression in the dataset using an 8-class Significance Analyses of Microarrays on the METABRIC dataset. On the right panel, a heatmap of genes located in a selected gene cluster is shown. For both heatmaps, red color represents relative high gene expression, green represents relative low gene expression, and black represents median gene expression.
Figure 3.
Figure 3.
Kaplan-Meier 10-year breast cancer-specific survival analyses in the molecular taxonomy of breast cancer international consortium dataset (all patients). A) Based on cHER2 status. B) Based on intrinsic subtype and cHER2 status. Tables of the numbers of patients at risk in each group at various time points are below each graph. Statistically significant differences were determined by two-sided log-rank test.

References

    1. Baselga J. Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist. 2002;7(Suppl 4):2–8.
    1. Slamon D, Clark G, Wong S, et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235(4785): 177–182.
    1. Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational Study of the Efficacy and Safety of Humanized Anti-HER2 Monoclonal Antibody in Women Who Have HER2-Overexpressing Metastatic Breast Cancer That Has Progressed After Chemotherapy for Metastatic Disease. J Clin Oncol. 1999;17(9):2639–2648.
    1. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2. N Engl J Med. 2001; 344(11):783–792.
    1. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Breast Cancer. N Engl J Med. 2005;353(16):1659–1672.
    1. Baselga J, Cortés J, Kim S-B, et al. Pertuzumab plus Trastuzumab plus Docetaxel for Metastatic Breast Cancer. N Engl J Med. 2012;366(2):109–119.
    1. Burstein HJ, Harris LN, Marcom PK, et al. Trastuzumab and Vinorelbine as First-Line Therapy for HER2-Overexpressing Metastatic Breast Cancer: Multicenter Phase II Trial With Clinical Outcomes, Analysis of Serum Tumor Markers as Predictive Factors, and Cardiac Surveillance Algorithm. J Clin Oncol. 2003;21(15):2889–2895.
    1. Joensuu H, Kellokumpu-Lehtinen P-L, Bono P, et al. Adjuvant Docetaxel or Vinorelbine with or without Trastuzumab for Breast Cancer. N Engl J Med. 2006;354(8):809–820.
    1. Rimawi MF, Mayer IA, Forero A, et al. Multicenter Phase II Study of Neoadjuvant Lapatinib and Trastuzumab With Hormonal Therapy and Without Chemotherapy in Patients With Human Epidermal Growth Factor Receptor 2–Overexpressing Breast Cancer: TBCRC 006. J Clin Oncol. 2013;31(14):1726–1731.
    1. Gianni L, Pienkowski T, Im Y-H, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(1):25–32.
    1. Prat A, Baselga J. Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade and Hormonal Therapy for the Treatment of Primary HER2-Positive Breast Cancer: One More Step Toward Chemotherapy-Free Therapy. J Clin Oncol. 2013;31(14):1703–1706.
    1. TCGA. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.
    1. Parker JS, Mullins M, Cheang MCU, et al. Supervised Risk Predictor of Breast Cancer Based on Intrinsic Subtypes. J Clin Oncol. 2009;27(8):1160–1167.
    1. Prat A, Perou CM. Deconstructing the molecular portraits of breast cancer. Mol Oncol. 2011;5(1):5–23.
    1. Prat A, Baselga J. The role of hormonal therapy in the management of hormonal-receptor-positive breast cancer with co-expression of HER2. Nat Clin Prac Oncol. 2008;5(9):531–542.
    1. Montemurro F, Di Cosimo S, Arpino G. Human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor-positive breast cancer: new insights into molecular interactions and clinical implications. Ann Oncol. 2013;24(11):2715–2724.
    1. Staaf J, Jonsson G, Ringner M, et al. High-resolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer. Breast Cancer Res. 2010;12(3):R25.
    1. Carey LA, Dees EC, Sawyer L, et al. The Triple Negative Paradox: Primary Tumor Chemosensitivity of Breast Cancer Subtypes. Clin Cancer Res. 2007;13(8):2329–2334.
    1. Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295(21):2492–2502.
    1. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752.
    1. Prat A, Ellis M, Perou C. Practical implications of gene-expression-based assays for breast oncologists. Nat Rev Clin Oncol. 2011;9(1):48–57.
    1. Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A. 2003;100(14):8418–8423.
    1. Millikan R, Newman B, Tse C-K, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat. 2008;109(1):123–139.
    1. Prat A, Cheang MCU, Martín M, et al. Prognostic Significance of Progesterone Receptor–Positive Tumor Cells Within Immunohistochemically Defined Luminal A Breast Cancer. J Clin Oncol. 2013;31(2):203–209.
    1. Nielsen TO, Parker JS, Leung S, et al. A Comparison of PAM50 Intrinsic Subtyping with Immunohistochemistry and Clinical Prognostic Factors in Tamoxifen-Treated Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res. 2010;16(21):5222–5232.
    1. Cheang MCU, Voduc KD, Tu D, et al. Responsiveness of Intrinsic Subtypes to Adjuvant Anthracycline Substitution in the NCIC.CTG MA.5 Randomized Trial. Clin Cancer Res. 2012;18(8):2402–2412.
    1. Chia SK, Bramwell VH, Tu D, et al. A 50-Gene Intrinsic Subtype Classifier for Prognosis and Prediction of Benefit from Adjuvant Tamoxifen. Clin Cancer Res. 2012;18(16):4465–4472.
    1. Esserman L, Berry D, Cheang M, et al. Chemotherapy response and recurrence-free survival in neoadjuvant breast cancer depends on biomarker profiles: results from the I-SPY 1 TRIAL (CALGB 150007/150012; ACRIN 6657). Breast Cancer Res Treat. 2012;132(3):1049–1062.
    1. Ellis MJ, Suman VJ, Hoog J, et al. Randomized Phase II Neoadjuvant Comparison Between Letrozole, Anastrozole, and Exemestane for Postmenopausal Women With Estrogen Receptor–Rich Stage 2 to 3 Breast Cancer: Clinical and Biomarker Outcomes and Predictive Value of the Baseline PAM50-Based Intrinsic Subtype—ACOSOG Z1031. J Clin Oncol. 2011;29(17):2342–2349.
    1. Dowsett M, Sestak I, Lopez-Knowles E, et al. Comparison of PAM50 Risk of Recurrence Score With Oncotype DX and IHC4 for Predicting Risk of Distant Recurrence After Endocrine Therapy. J Clin Oncol. 2013;31(22):2783–2790.
    1. Curtis C, Shah SP, Chin S-F, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346–352.
    1. Goldhirsch A, Winer EP, Coates AS, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24(9):2206–2223.
    1. Wolff AC, Hammond MEH, Schwartz JN, et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer. J Clin Oncol. 2007;25(1):118–145.
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001;98(9):5116–5121.
    1. Baselga J, Swain S. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9(7):463–475.
    1. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–137.
    1. Yakes FM, Chinratanalab W, Ritter CA, et al. Herceptin-induced Inhibition of Phosphatidylinositol-3 Kinase and Akt Is Required for Antibody-mediated Effects on p27, Cyclin D1, and Antitumor Action. Cancer Res. 2002;62(14):4132–4141.
    1. Perez EA, Romond EH, Suman VJ, 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.
    1. Vaz-Luis I, Ottesen R, Hughes M, et al. Impact of hormone receptor status on patterns of recurrence and clinical outcomes among patients with human epidermal growth factor-2-positive breast cancer in the National Comprehensive Cancer Network: a prospective cohort study. Breast Cancer Res. 2012;14(5):R129.
    1. Gianni L, Eiermann W, Semiglazov V, et al. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet. 2010;375(9712):377–384.
    1. Guarneri V, Frassoldati A, Bottini A, et al. Preoperative Chemotherapy Plus Trastuzumab, Lapatinib, or Both in Human Epidermal Growth Factor Receptor 2–Positive Operable Breast Cancer: Results of the Randomized Phase II CHER-LOB Study. J Clin Oncol. 2012;30(16):1989–1995.
    1. Baselga J, Bradbury I, Eidtmann H, et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2012;379(9816):633–640.
    1. Untch M, Loibl S, Bischoff J, et al. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy (GeparQuinto, GBG 44): a randomised phase 3 trial. Lancet Oncol. 2012;13(2):135–144.
    1. Carey L, Berry D, Ollila D, et al. Clinical and translational results of CALGB 40601: A neoadjuvant phase III trial of weekly paclitaxel and trastuzumab with or without lapatinib for HER2-positive breast cancer. Proc Am Soc Clin Oncol: a500. 2013.
    1. Prat A, Bianchini G, Pickl M, et al. The research-based PAM50 subtype predictor identifies higher pathological responses and improved survival outcomes in HER2-positive breast cancer in the NOAH study. Clin Cancer Res. 2014;20(2):511–521.

Source: PubMed

3
Předplatit