An assessment of prognostic immunity markers in breast cancer

Benlong Yang, Jeff Chou, Yaozhong Tao, Dengbin Wu, Xinhong Wu, Xueqing Li, Yan Li, Yiwei Chu, Feng Tang, Yanxia Shi, Linlin Ma, Tong Zhou, William Kaufmann, Lisa A Carey, Jiong Wu, Zhiyuan Hu, Benlong Yang, Jeff Chou, Yaozhong Tao, Dengbin Wu, Xinhong Wu, Xueqing Li, Yan Li, Yiwei Chu, Feng Tang, Yanxia Shi, Linlin Ma, Tong Zhou, William Kaufmann, Lisa A Carey, Jiong Wu, Zhiyuan Hu

Abstract

Tumor-infiltrating lymphocytes (TIL) and immunity gene signatures have been reported to be significantly prognostic in breast cancer but have not yet been applied for calculation of risk of recurrence in clinical assays. A compact set of 17 immunity genes was derived herein from an Affymetrix-derived gene expression dataset including 1951 patients (AFFY1951). The 17 immunity genes demonstrated significant prognostic stratification of estrogen receptor (ER)-negative breast cancer patients with high proliferation gene expression. Further analysis of blood and breast cancer single-cell RNA-seq datasets revealed that the 17 immunity genes were derived from TIL that were inactive in the blood and became active in tumor tissue. Expression of the 17 immunity genes was significantly (p < 2.2E-16, n = 91) correlated with TILs percentage on H&E in triple negative breast cancer. To demonstrate the impact of tumor immunity genes on prognosis, we built a Cox model to incorporate breast cancer subtypes, proliferation score and immunity score (72 gene panel) with significant prediction of outcomes (p < 0.0001, n = 1951). The 72 gene panel and its risk evaluation model were validated in two other published gene expression datasets including Illumina beads array data METABRIC (p < 0.0001, n = 1997) and whole transcriptomic mRNA-seq data TCGA (p = 0.00019, n = 996) and in our own targeted RNA-seq data TARGETSEQ (p < 0.0001, n = 303). Further examination of the 72 gene panel in single cell RNA-seq of tumors demonstrated tumor heterogeneity with more than two subtypes observed in each tumor. In conclusion, immunity gene expression was an important parameter for prognosis and should be incorporated into current multi-gene assays to improve assessment of risk of distant metastasis in breast cancer.

Conflict of interest statement

The Authors declare no competing interests.

Figures

Fig. 1
Fig. 1
RNA-seq gene expression of 17 immunity genes and 19 proliferation genes in published PBMC single-cell dataset (Macosko et al. Cell 2015) and breast cancer solid tumor single cells (Chung et al. Nature Communications 2017). (A) Expression of immunity and proliferation genes in different PBMC cell types including B cells, CD4 T cells, CD8 T cells, NK cells, Monocytes and dendritic cells. (B) Expression of immunity and proliferation genes of single cells including breast tumors’ immune single cells labeled as T_cell + Mac + M & B_cell (Mac = macrophages, M = monocytes) and carcinoma single cells groups 1 to 3 (mixed carcinoma single cells from different tumors), BC02 and BC05 (carcinoma single cells from each individual tumor)
Fig. 2
Fig. 2
Survival plots of Immunity Score in different patient groups identified by proliferation and ER status in the AFFY1951 training dataset, two test datasets METABRIC and TARGETSEQ. Immunity Score demonstrated strongest outcome prediction in patients who were ER-negative and proliferation high in AFFY1951 (A) (p < 0.0001, n = 467), METABRIC (C) (p = 0.00018, n = 398) and TARGETSEQ (E) (p = 0.044, n = 65), but was insignificant in ER-positive and proliferation high patients in AFFY1951 (B) (p = 0.079, n = 585), METABRIC (D) (p = 0.22, n = 603) and TARGETSEQ (F) (p = 0.46, n = 97). High proliferation groups had proliferation scores no less than 50 and low proliferation groups had proliferation scores less than 50. Survival analysis of ER-negative or ER-positive and low proliferation patients were demonstrated in Supplementary Figure 1
Fig. 3
Fig. 3
Comparison of survival analysis of iRDM and PAM50 in AFFY1951 breast cancer training dataset. Subtypes and risk groups are color-coded: Basal-like (Red), HER2E (Hot Pink), Immuno (Yellow), Luminal A (Dark Blue), Luminal B (Sky Blue), Normal-like (Green); low (Green), med (Dark Blue), and high (Red) risks. Kaplan-Meier plots were used to show Distant Metastasis-Free Survival (DMFS) by subtypes for iRDM (A) (p < 0.0001, n = 1655) and PAM50 (B) (p < 0.0001, n = 1463) and risk groups for iRDM (C) (p < 0.0001, n = 1951) and PAM50 (D) (p < 0.0001, n = 1951)
Fig. 4
Fig. 4
Validation of iRDM subtype and risk survival analysis in three independent test datasets. Survival plots of iRDM subtypes in METABRIC (A) (p < 0.0001, n = 1997), TCGA (C) (p = 0.022, n = 996), TARGETSEQ (E) (p = 0.00021, n = 303) and survival plots of risk groups (high, low, med) in METABRIC (B) (p < 0.0001, n = 1997), TCGA (D) (p = 0.00019, n = 996), TARGETSEQ (E) (p < 0.0001, n = 303) were shown. Subtypes and risk groups are color-coded: Basal-like (Red), HER2E (Hot Pink), Immuno (Yellow), Luminal A (Dark Blue), Luminal B (Sky Blue), Normal-like (Green); low (Green), med (Dark Blue), and high (Red) risks
Fig. 5
Fig. 5
Heatmap of iRDM subtypes analyzed in 549 single cells from 11 primary breast tumors and two lymph node metastases. All tumors showed two or more iRDM subtypes. Sub, subtype; Cell, single cell number; TIL, percentage of tumor-infiltrating lymphocytes. Color-coded individual cell subtypes: Basal-like (Red), HER2E (Hot Pink), Immuno (Yellow), Luminal A (Dark Blue), Luminal B (Sky Blue), Normal-like (Green), Mixed (Black)

References

    1. Hu Z, et al. A compact VEGF signature associated with distant metastases and poor outcomes. BMC Med. 2009;7:9. doi: 10.1186/1741-7015-7-9.
    1. Lesokhin AM, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016;34:2698–2704. doi: 10.1200/JCO.2015.65.9789.
    1. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    1. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Powles T, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:558–562. doi: 10.1038/nature13904.
    1. Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2015;371:2189–2199. doi: 10.1056/NEJMoa1406498.
    1. Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J. Immunother. Cancer. 2016;4:59. doi: 10.1186/s40425-016-0165-6.
    1. Salgado R, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann. Oncol. 2015;26:259–271. doi: 10.1093/annonc/mdu450.
    1. Denkert C, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018;19:40–50. doi: 10.1016/S1470-2045(17)30904-X.
    1. Miller LD, et al. Immunogenic Subtypes of Breast Cancer Delineated by Gene Classifiers of Immune Responsiveness. Cancer Immunol. Res. 2016;4:600–610. doi: 10.1158/2326-6066.CIR-15-0149.
    1. Ramaswamy S, Ross KN, Lander ES, Golub TR. A molecular signature of metastasis in primary solid tumors. Nat. Genet. 2003;33:49–54. doi: 10.1038/ng1060.
    1. van de Vijver MJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 2002;347:1999–2009. doi: 10.1056/NEJMoa021967.
    1. van ‘t Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–536. doi: 10.1038/415530a.
    1. Wang Y, et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005;365:671–679. doi: 10.1016/S0140-6736(05)70933-8.
    1. Paik S, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N. Engl. J. Med. 2004;351:2817–2826. doi: 10.1056/NEJMoa041588.
    1. Perou CM, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093.
    1. Weigelt B, et al. Molecular portraits and 70-gene prognosis signature are preserved throughout the metastatic process of breast cancer. Cancer Res. 2005;65:9155–9158. doi: 10.1158/0008-5472.CAN-05-2553.
    1. Sparano JA, Paik S. Development of the 21-gene assay and its application in clinical practice and clinical trials. J. Clin. Oncol. 2008;26:721–728. doi: 10.1200/JCO.2007.15.1068.
    1. Slodkowska EA, Ross JS. MammaPrint 70-gene signature: another milestone in personalized medical care for breast cancer patients. Expert Rev. Mol. Diagn. 2009;9:417–422. doi: 10.1586/erm.09.32.
    1. Filipits M, et al. The PAM50 risk-of-recurrence score predicts risk for late distant recurrence after endocrine therapy in postmenopausal women with endocrine-responsive early breast cancer. Clin. Cancer Res. 2014;20:1298–1305. doi: 10.1158/1078-0432.CCR-13-1845.
    1. Prat A, et al. Research-based PAM50 subtype predictor identifies higher responses and improved survival outcomes in HER2-positive breast cancer in the NOAH study. Clin. Cancer Res. 2014;20:511–521. doi: 10.1158/1078-0432.CCR-13-0239.
    1. Dieci, M.V. et al. Integrated evaluation of PAM50 subtypes and immune modulation of pCR in HER2-positive breast cancer patients treated with chemotherapy and HER2-targeted agents in the CherLOB trial. Ann Oncol (2016).
    1. Parker JS, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 2009;27:1160–1167. doi: 10.1200/JCO.2008.18.1370.
    1. Dowsett M, 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. 2009;31:2783–2790. doi: 10.1200/JCO.2012.46.1558.
    1. Alvarado MD, et al. A Prospective Comparison of the 21-Gene Recurrence Score and the PAM50-Based Prosigna in Estrogen Receptor-Positive Early-Stage Breast Cancer. Adv. Ther. 2015;32:1237–1247. doi: 10.1007/s12325-015-0269-2.
    1. Macosko EZ, et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015;161:1202–1214. doi: 10.1016/j.cell.2015.05.002.
    1. Chung W, et al. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nature Communications. 2017;8:15081. doi: 10.1038/ncomms15081.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352. doi: 10.1038/nature10983.
    1. Hu Z, et al. The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genom. 2006;7:96. doi: 10.1186/1471-2164-7-96.
    1. Sorlie T, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl. Acad. Sci. USA. 2003;100:8418–8423. doi: 10.1073/pnas.0932692100.
    1. Prat A, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12:R68. doi: 10.1186/bcr2635.
    1. Alistar A, et al. Dual roles for immune metagenes in breast cancer prognosis and therapy prediction. Genome Med. 2014;6:80. doi: 10.1186/s13073-014-0080-8.
    1. Wolchok JD, Chan TA. Cancer: Antitumour immunity gets a boost. Nature. 2014;515:496–498. doi: 10.1038/515496a.
    1. Iglesia MD, et al. Prognostic B-cell signatures using mRNA-seq in patients with subtype-specific breast and ovarian cancer. Clin. Cancer Res. 2014;20:3818–3829. doi: 10.1158/1078-0432.CCR-13-3368.
    1. Taylor NA, et al. Treg depletion potentiates checkpoint inhibition in claudin-low breast cancer. J. Clin. Invest. 2017;127:3472–3483. doi: 10.1172/JCI90499.
    1. Iglesia Michael D., Parker Joel S., Hoadley Katherine A., Serody Jonathan S., Perou Charles M., Vincent Benjamin G. Genomic Analysis of Immune Cell Infiltrates Across 11 Tumor Types. Journal of the National Cancer Institute. 2016;108(11):djw144. doi: 10.1093/jnci/djw144.
    1. Mao Y, et al. The Prognostic Value of Tumor-Infiltrating Lymphocytes in Breast Cancer: A Systematic Review and Meta-Analysis. PLoS One. 2016;11:e0152500. doi: 10.1371/journal.pone.0152500.
    1. Yue Q, et al. The prognostic value of Foxp3 + tumor-infiltrating lymphocytes in patients with glioblastoma. J. Neurooncol. 2013;116:251–259. doi: 10.1007/s11060-013-1314-0.
    1. Loi S, et al. Predicting prognosis using molecular profiling in estrogen receptor-positive breast cancer treated with tamoxifen. BMC Genom. 2008;9:239. doi: 10.1186/1471-2164-9-239.
    1. Loi S, et al. PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc. Natl. Acad. Sci. USA. 2010;107:10208–10213. doi: 10.1073/pnas.0907011107.
    1. Irizarry RA, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264. doi: 10.1093/biostatistics/4.2.249.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–127. doi: 10.1093/biostatistics/kxj037.
    1. Chou JW, Zhou T, Kaufmann WK, Paules RS, Bushel PR. Extracting gene expression patterns and identifying co-expressed genes from microarray data reveals biologically responsive processes. BMC Bioinforma. 2007;8:427. doi: 10.1186/1471-2105-8-427.
    1. Zhou T, et al. Profiles of global gene expression in ionizing-radiation-damaged human diploid fibroblasts reveal synchronization behind the G1 checkpoint in a G0-like state of quiescence. Environ. Health Perspect. 2006;114:553–559. doi: 10.1289/ehp.8026.
    1. Nagalla S, et al. Interactions between immunity, proliferation and molecular subtype in breast cancer prognosis. Genome Biol. 2013;14:R34. doi: 10.1186/gb-2013-14-4-r34.
    1. TCGA. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin. Cancer Res. 2004;10:7252–7259. doi: 10.1158/1078-0432.CCR-04-0713.
    1. Tibshirani R, Hastie T, Narasimhan B, Chu G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc. Natl. Acad. Sci. USA. 2002;99:6567–6572. doi: 10.1073/pnas.082099299.
    1. Fan C, et al. Concordance among gene-expression-based predictors for breast cancer. N. Engl. J. Med. 2006;355:560–569. doi: 10.1056/NEJMoa052933.
    1. Newcombe RG. Two-sided confidence intervals for the single proportion: comparison of seven methods. Stat. Med. 1998;17:857–872. doi: 10.1002/(SICI)1097-0258(19980430)17:8<857::AID-SIM777>;2-E.
    1. Benito M, et al. Adjustment of systematic microarray data biases. Bioinformatics. 2004;20:105–114. doi: 10.1093/bioinformatics/btg385.

Source: PubMed

3
Sottoscrivi