DAGM: A novel modelling framework to assess the risk of HER2-negative breast cancer based on germline rare coding mutations

Mei Yang, Yanhui Fan, Zhi-Yong Wu, Jin Gu, Zhendong Feng, Qiangzu Zhang, Shunhua Han, Zhonghai Zhang, Xu Li, Yi-Ching Hsueh, Yanxiang Ni, Xiaoling Li, Jieqing Li, Meixia Hu, Weiping Li, Hongfei Gao, Ciqiu Yang, Chunming Zhang, Liulu Zhang, Teng Zhu, Minyi Cheng, Fei Ji, Juntao Xu, Hening Cui, Guangming Tan, Michael Q Zhang, Changhong Liang, Zaiyi Liu, You-Qiang Song, Gang Niu, Kun Wang, Mei Yang, Yanhui Fan, Zhi-Yong Wu, Jin Gu, Zhendong Feng, Qiangzu Zhang, Shunhua Han, Zhonghai Zhang, Xu Li, Yi-Ching Hsueh, Yanxiang Ni, Xiaoling Li, Jieqing Li, Meixia Hu, Weiping Li, Hongfei Gao, Ciqiu Yang, Chunming Zhang, Liulu Zhang, Teng Zhu, Minyi Cheng, Fei Ji, Juntao Xu, Hening Cui, Guangming Tan, Michael Q Zhang, Changhong Liang, Zaiyi Liu, You-Qiang Song, Gang Niu, Kun Wang

Abstract

Background: Breast cancers can be divided into HER2-negative and HER2-positive subtypes according to different status of HER2 gene. Despite extensive studies connecting germline mutations with possible risk of HER2-negative breast cancer, the main category of breast cancer, it remains challenging to obtain accurate risk assessment and to understand the potential underlying mechanisms.

Methods: We developed a novel framework named Damage Assessment of Genomic Mutations (DAGM), which projects rare coding mutations and gene expressions into Activity Profiles of Signalling Pathways (APSPs).

Findings: We characterized and validated DAGM framework at multiple levels. Based on an input of germline rare coding mutations, we obtained the corresponding APSP spectrum to calculate the APSP risk score, which was capable of distinguish HER2-negative from HER2-positive cases. These findings were validated using breast cancer data from TCGA (AUC = 0.7). DAGM revealed that HER2 signalling pathway was up-regulated in germline of HER2-negative patients, and those with high APSP risk scores had exhibited immune suppression. These findings were validated using RNA sequencing, phosphoproteome analysis, and CyTOF. Moreover, using germline mutations, DAGM could evaluate the risk for HER2-negative breast cancer, not only in women carrying BRCA1/2 mutations, but also in those without known disease-associated mutations.

Interpretation: The DAGM can facilitate the screening of subjects at high risk of HER2-negative breast cancer for primary prevention. This study also provides new insights into the potential mechanisms of developing HER2-negative breast cancer. The DAGM has the potential to be applied in the prevention, diagnosis, and treatment of HER2-negative breast cancer.

Funding: This work was supported by the National Key Research and Development Program of China (grant no. 2018YFC0910406 and 2018AAA0103302 to CZ); the National Natural Science Foundation of China (grant no. 81202076 and 82072939 to MY, 81871513 to KW); the Guangzhou Science and Technology Program key projects (grant no. 2014J2200007 to MY, 202002030236 to KW); the National Key R&D Program of China (grant no. 2017YFC1309100 to CL); Shenzhen Science and Technology Planning Project (grant no. JCYJ20170817095211560 574 to YN); and the Natural Science Foundation of Guangdong Province (grant no. 2017A030313882 to KW and S2013010012048 to MY); Hefei National Laboratory for Physical Sciences at the Microscale (grant no. KF2020009 to GN); and RGC General Research Fund (grant no. 17114519 to YQS).

Keywords: Activity profiles of signalling pathways (APSP); Damage assessment of genomic mutations (DAGM); Germline rare coding mutations; HER2 signalling pathway; HER2-negative breast cancer; Immune suppression; Risk assessment.

Conflict of interest statement

Declaration of Competing Interest The authors declare that they have no competing interests.

Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Workflow of the DAGM framework. The DAGM framework mainly composed of three steps. In the first step (grey colour), driving force (DF) and global driving force (GDF) are established for future analysis based on the rare coding mutations and gene expression from the COSMIC Cell Lines project. When obtaining the germline rare coding mutations of a subject, we calculated the combined effect of all these mutations (Step 2). In the last step, the activity profile of signalling pathways (APSPs) is evaluated for this subject.
Fig. 2
Fig. 2
Germline APSP spectrum distinguishes HER2-negative breast cancer patients from HER2-positive patients. (a) Heatmap of the germline APSPs of 60 pathways for 721 subjects. Each row represents one pathway and each column represents one subject. (b) Hierarchical clustering dendrogram. HER2-negative subtype (Luminal B (HER2-negative), Luminal A, and TNBC) and HER2-positive subtype (ERBB2-positive and Luminal B (HER2-positive)) were well separated by hierarchical clustering with Euclidean distance as the distance measure. (c) Receiver operating characteristic (ROC) curve for distinguishing HER2-negative breast cancer patients from HER2-positive patients by APSPs. Area under the curve (AUC) was 0.77. (d) Receiver operating characteristic (ROC) curve for distinguishing HER2-negative breast cancer patients from controls by APSPs. Area under the curve (AUC) was 0.76.
Fig. 3
Fig. 3
Upregulated HER2 signalling pathway in germlines of Her2-negative breast cancer patients. (a) Difference of activity profiles of signalling pathways (APSPs) between breast cancer patients and controls. To test the significance of the difference, P-values were calculated by permutation (1,000,000 times). The FDR corrected P-values were also calculated to correct the multiple comparisons. The asterisk denotes FDR corrected P-value 

Fig. 4

Identification of HER2-negative breast cancer…

Fig. 4

Identification of HER2-negative breast cancer using germline APSP risk score. (a) The boxplot…

Fig. 4
Identification of HER2-negative breast cancer using germline APSP risk score. (a) The boxplot of APSPs for eight pathway panels. The panel of immune system is at the top of three significantly down-regulated panels (labelled by a star) that were then used to calculate APSP risk scores. (b) The distribution of APSP risk scores of HER2-negative breast cancer patients (upper panel), controls (middle panel) and HER2-positive breast cancer patients (bottom panel). The APSP risk scores in HER2-negative breast cancer patients were significantly higher than those in the controls (T-test, P-value = 3.96e-27) and HER2-positive breast cancer patients (T-test, P-value = 4.88e-22). (c) The APSP risk scores were significantly higher in HER2-negative breast cancer patients with or without BRCA1/2 mutations than the control subjects. (d) Receiver operating characteristic (ROC) curve for APSP risk score in distinguishing HER2-negative breast cancer patients from HER2-positive patients. Area under the curve (AUC) was 0.79. (e) Receiver operating characteristic (ROC) curve for APSP risk score in distinguishing HER2-negative breast cancer patients from controls. Area under the curve (AUC) was 0.74.

Fig. 5

Enhanced immune suppression and risk…

Fig. 5

Enhanced immune suppression and risk stratification of HER2-negative breast cancer. Mass cytometry analysis…

Fig. 5
Enhanced immune suppression and risk stratification of HER2-negative breast cancer. Mass cytometry analysis revealed percentage of exhausted CD8+ T cells in female subjects of low and high APSP risk scores. (b) Mass cytometry analysis revealed percentage of exhausted CD8+ T cells in TNBC patients and control subjects. (c) Stratification of HER2-negative patients (red dots) as well as control subjects (green dots) into six groups, G1 to G6, according to the different standard deviations of APSP risk scores from their mean value. G1 and G6 included subject with APSP risk score two standard deviations lower and higher than the mean value, respectively. G2 and G5 included subject with APSP risk score between one and two standard deviations lower and higher than the mean value, respectively. G3 and G4 included subject with APSP risk score within one standard deviation lower and higher than the mean value, respectively. (d) Odds ratios for HER2 negative breast cancer display exponential distribution when the subjects were stratified into six groups (G1 to G6) as in (c). The grey circles represent the sample sizes of each group.
Fig. 4
Fig. 4
Identification of HER2-negative breast cancer using germline APSP risk score. (a) The boxplot of APSPs for eight pathway panels. The panel of immune system is at the top of three significantly down-regulated panels (labelled by a star) that were then used to calculate APSP risk scores. (b) The distribution of APSP risk scores of HER2-negative breast cancer patients (upper panel), controls (middle panel) and HER2-positive breast cancer patients (bottom panel). The APSP risk scores in HER2-negative breast cancer patients were significantly higher than those in the controls (T-test, P-value = 3.96e-27) and HER2-positive breast cancer patients (T-test, P-value = 4.88e-22). (c) The APSP risk scores were significantly higher in HER2-negative breast cancer patients with or without BRCA1/2 mutations than the control subjects. (d) Receiver operating characteristic (ROC) curve for APSP risk score in distinguishing HER2-negative breast cancer patients from HER2-positive patients. Area under the curve (AUC) was 0.79. (e) Receiver operating characteristic (ROC) curve for APSP risk score in distinguishing HER2-negative breast cancer patients from controls. Area under the curve (AUC) was 0.74.
Fig. 5
Fig. 5
Enhanced immune suppression and risk stratification of HER2-negative breast cancer. Mass cytometry analysis revealed percentage of exhausted CD8+ T cells in female subjects of low and high APSP risk scores. (b) Mass cytometry analysis revealed percentage of exhausted CD8+ T cells in TNBC patients and control subjects. (c) Stratification of HER2-negative patients (red dots) as well as control subjects (green dots) into six groups, G1 to G6, according to the different standard deviations of APSP risk scores from their mean value. G1 and G6 included subject with APSP risk score two standard deviations lower and higher than the mean value, respectively. G2 and G5 included subject with APSP risk score between one and two standard deviations lower and higher than the mean value, respectively. G3 and G4 included subject with APSP risk score within one standard deviation lower and higher than the mean value, respectively. (d) Odds ratios for HER2 negative breast cancer display exponential distribution when the subjects were stratified into six groups (G1 to G6) as in (c). The grey circles represent the sample sizes of each group.

References

    1. Jiang X, Tang H, Chen T. Epidemiology of gynecologic cancers in China. J Gynecol Oncol. 2018;29:e7.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2018;68:7–30. 2018.
    1. Dai X, Li T, Bai Z, Yang Y, Liu X, Zhan J. Breast cancer intrinsic subtype classification, clinical use and future trends. Am J Hum Genet. 2015;5:2929–2943.
    1. DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA. Convergence of incidence rates between black and white women. CA Cancer J Clin. 2015;66:31–42. 2016.
    1. Godoy-Ortiz A, Sanchez-Muñoz A, Chica Parrado MR, Álvarez M, Ribelles N, Rueda Dominguez A. Deciphering HER2 breast cancer disease: biological and clinical implications. Front Oncol. 2019;9
    1. Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389:1134–1150.
    1. Voduc KD, Cheang MCU, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28:1684–1691.
    1. Sun J, Meng H, Yao L, Lv M, Bai J, Zhang J. Germline mutations in cancer susceptibility genes in a large series of unselected breast cancer patients. Clin Cancer Res. 2017;23:6113–6119.
    1. Wu S, Zhang Q, Zhang F, Meng F, Liu S, Zhou R. HER2 recruits AKT1 to disrupt STING signalling and suppress antiviral defence and antitumour immunity. Nat Cell Biol. 2019;21:1027–1040.
    1. Sivick KE, Desbien AL, Glickman LH, Reiner GL, Corrales L, Surh NH. Magnitude of therapeutic STING activation determines CD8 T cell-mediated anti-tumor immunity. Cell Rep. 2018;25:3074–3085. e5.
    1. Lei J, Rudolph A, Moysich KB, Rafiq S, Behrens S, Goode EL. Assessment of variation in immunosuppressive pathway genes reveals TGFBR2 to be associated with prognosis of estrogen receptor-negative breast cancer after chemotherapy. Breast Cancer Res. 2015;17:18.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550.
    1. Sugrue LP, Desikan RS. What are polygenic scores and why are they important? JAMA. 2019;321:1820–1821.
    1. Mavaddat N, Michailidou K, Dennis J, Lush M, Fachal L, Lee A. Polygenic risk scores for prediction of breast cancer and breast cancer subtypes. Am J Hum Genet. 2019;104:21–34.
    1. Li H, Feng B, Miron A, Chen X, Beesley J, Bimeh E. Breast cancer risk prediction using a polygenic risk score in the familial setting: a prospective study from the Breast Cancer Family Registry and kConFab. Genet Med. 2017;19:30–35.
    1. Läll K, Lepamets M, Palover M, Esko T, Metspalu A, Tõnisson N. Polygenic prediction of breast cancer: comparison of genetic predictors and implications for risk stratification. BMC Cancer. 2019;19:557.
    1. Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet. 2018;50:1219–1224.
    1. Wang B, Bao S, Zhang Z, Zhou X, Wang J, Fan Y. A rare variant in MLKL confers susceptibility to ApoE ɛ4-negative Alzheimer's disease in Hong Kong Chinese population. Neurobiol Aging. 2018;68:160. e1-60.e7.
    1. Li H. 2013. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. . arXiv:1303.3997v2:
    1. McKenna AH, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A. The genome analysis toolkit: a mapreduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38 e164-e64.
    1. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37:907–915.
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–930.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140.
    1. The UniProt Consortium UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res. 2018;47 D506-D15.
    1. Ghandi M, Huang FW, Jané-Valbuena J, Kryukov GV, Lo CC, McDonald ER. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature. 2019;569:503–508.
    1. Kuhn M. Building predictive models in R using the caret package. J Stat Softw. 2008;28:1–26.
    1. Noble WS. What is a support vector machine? Nature Biotechnol. 2006;24:1565–1567.
    1. LaValley MP. Logistic regression. Circulation. 2008;117:2395–2399.
    1. Murtagh F, Legendre P. Ward’s hierarchical agglomerative clustering method: which algorithms implement Ward’s criterion? J Classif. 2014;31:274–295.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc B. 1995;57:289–300.
    1. Chen JJ, Roberson PK, Schell MJ. The false discovery rate: a key concept in large-scale genetic studies. Cancer Control. 2010;17:58–62.
    1. Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I, Forbes SA. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat Rev Cancer. 2018;18:696–705.
    1. Liu S-H, Shen P-C, Chen C-Y, Hsu A-N, Cho Y-C, Lai Y-L. DriverDBv3: a multi-omics database for cancer driver gene research. Nucleic Acids Res. 2019
    1. Levy-Lahad E, Friedman E. Cancer risks among BRCA1 and BRCA2 mutation carriers. Br J Cancer. 2007;96:11–15.
    1. Wong-Brown MW, Meldrum CJ, Carpenter JE, Clarke CL, Narod SA, Jakubowska A. Prevalence of BRCA1 and BRCA2 germline mutations in patients with triple-negative breast cancer. Breast Cancer Res Treat. 2015;150:71–80.
    1. Antoniou A, Pharoah PDP, Narod S, Risch HA, Eyfjord JE, Hopper JL. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet. 2003;72:1117–1130.
    1. Shieh Y, Fejerman L, Lott PC, Marker K, Sawyer SD, Hu D. A polygenic risk score for breast cancer in US Latinas and Latin American women. J Natl Cancer Inst. 2019;112:590–598.
    1. Bauer S, Robinson PN, Gagneur J. Model-based gene set analysis for Bioconductor. Bioinformatics. 2011;27:1882–1883.
    1. Tai W, Mahato R, Cheng K. The role of HER2 in cancer therapy and targeted drug delivery. J Control Release. 2010;146:264–275.
    1. Andrechek ER. HER2/Neu tumorigenesis and metastasis is regulated by E2F activator transcription factors. Oncogene. 2015;34:217–225.
    1. Zaczek A, Brandt B, Bielawski KP. The diverse signaling network of EGFR, HER2, HER3 and HER4 tyrosine kinase receptors and the consequences for therapeutic approaches. Histol Histopathol. 2005;20:1005–1015.
    1. Giral H, Landmesser U, Kratzer A. Into the wild: GWAS exploration of non-coding RNAs. Front Cardiovasc Med. 2018;5

Source: PubMed

3
Subskrybuj