RT Journal Article SR Electronic T1 DAGM: a novel modelling framework to assess the risk of HER2-negative breast cancer based on germline rare coding mutations JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2021.01.05.21249253 DO 10.1101/2021.01.05.21249253 A1 Mei Yang A1 Yanhui Fan A1 Zhi-Yong Wu A1 Jin Gu A1 Zhendong Feng A1 Qiangzu Zhang A1 Shunhua Han A1 Zhonghai Zhang A1 Xu Li A1 Yi-Ching Hsueh A1 Xiaoling Li A1 Jieqing Li A1 Meixia Hu A1 Weiping Li A1 Hongfei Gao A1 Ciqiu Yang A1 Chunming Zhang A1 Liulu Zhang A1 Teng Zhu A1 Minyi Cheng A1 Fei Ji A1 Juntao Xu A1 Hening Cui A1 Guangming Tan A1 Michael Q. Zhang A1 Changhong Liang A1 Zaiyi Liu A1 You-Qiang Song A1 Gang Niu A1 Kun Wang YR 2021 UL http://medrxiv.org/content/early/2021/01/09/2021.01.05.21249253.abstract AB Background Breast cancers can be divided into HER2-negative and HER2-positive subtypes according to the 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 accurately assess its potential risk and to understand the potential 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 the HER2 signalling pathway was up-regulated in the germline of HER2-negative patients, and those with high APSP risk scores had suppressed immunity. These findings were validated using RNA sequencing, phosphoproteome analysis, and CyTOF. Moreover, using germline mutations, DAGM could evaluate the risk of developing 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); and the Natural Science Foundation of Guangdong Province (grant no. 2017A030313882 to KW)Evidence before this study The majority of hereditary breast cancers are caused by BRCA1/2 mutations, and the presence of these mutations is strongly associated with an increased risk of breast cancer. Meanwhile, BRCA1/2 gene mutations are rarely found in sporadic breast cancers and only account for a modest percentage of all breast cancer patients. Polygenic risk score (PRS), a widely-used approach for stratifying individuals according to their risk of a certain kind of complex disease, has been used to predict subjects at high risk for breast cancer. However, relying on SNPs from genome-wide association studies (GWAS) without including gene expressions or pathway activities, PRS is not very suitable for cross-population prediction and describes disease risk in terms of genomic mutations without alluding to the underlying pathogenic mechanism(s). Therefore, there is still an urgent need for a population-independent comprehensive method to accurately assess the risk of breast cancer and to gain insights on potential mechanism(s).Added value of this study When subjecting germline rare coding mutations (gRCMs) to DAGM framework, which results in the corresponding APSP and APSP risk score. Both APSP and APSP risk score can identify HER2-negative from HER2-positive breast cancers. These findings suggest HER2-negative breast cancer does not develop accidentally, but rather is defined by a genomic evolutionary strategy. Furthermore, this study also revealed the up-regulation of HER2 signalling pathway in germlines of HER2-negative breast cancers and the immune suppression in subjects with high APSP risk score, shedding new light on the potential mechanisms of developing HER2-negative breast cancer. Moreover, our APSP risk score was able to relatively accurately evaluate the risk of developing HER2-negative breast cancer for each female, including not only BRCA1/2 carriers, but also non-carriers.Implications of all the available evidence The present study suggests that HER2 signalling pathway activity, as an aggressive factor, contribute to the development of different types of breast cancers, either via the combined effects of multiple germline mutations in HER2-negative germlines or via amplifying the gene itself in HER2-positive tumour cells. This provides a theoretical basis for the prevention, diagnosis, and treatment of breast cancers. At the same time, the study provides preliminary methods for assessing the relative risk of HER2-negative breast cancer for females with or without BRCA1/2 mutations. Finally, our findings provide a new perspective and theoretical basis for identifying high-risk female subjects, based on the high APSP risk score, for early screening and prevention of HER2-negative breast cancer.Competing Interest StatementThe authors have declared no competing interest.Funding StatementThis 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); and the Natural Science Foundation of Guangdong Province (grant no. 2017A030313882 to KW) Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Research Ethics Committee at Guangdong General Hospital, Guangdong Academy of Medicals Sciences.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).Yes I have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe raw exome sequencing data reported in this paper have been deposited in the Genome Sequence Archive (GSA) in National Genomics Data Centre, Beijing Institute of Genomics, Chinese Academy of Sciences, under accession number HRA000285, which is publicly accessible at https://bigd.big.ac.cn/gsa. All other data and materials are available upon request.APSPactivity profiles of signaling pathwayAUCarea under the receiver operating characteristic curveCCLECancer Cell Line EncyclopediaDAGMdamage assessment framework of genomic mutationsEREstrogen receptorGDFglobal driving forceGSEAGene Set Enrichment AnalysisGWASgenome-wide association studyHER2human epidermal growth factor 2PBMCperipheral blood mononuclear cellsPCCPearson correlation coefficientsPRProgesterone receptorPRSpolygenic risk scoreRCMrare coding mutationsSVMSupport Vector MachineTCGAThe Cancer Genome AtlasTNBCTriple-negative breast cancerWESwhole-exome sequencing.