scispace - formally typeset
Search or ask a question

Showing papers by "Xin Zhou published in 2018"


Journal ArticleDOI
15 Mar 2018-Nature
TL;DR: The data suggest that 7–8% of the children in this cohort carry an unambiguous predisposing germline variant and that nearly 50% of paediatric neoplasms harbour a potentially druggable event, which is highly relevant for the design of future clinical trials.
Abstract: Pan-cancer analyses that examine commonalities and differences among various cancer types have emerged as a powerful way to obtain novel insights into cancer biology. Here we present a comprehensive analysis of genetic alterations in a pan-cancer cohort including 961 tumours from children, adolescents, and young adults, comprising 24 distinct molecular types of cancer. Using a standardized workflow, we identified marked differences in terms of mutation frequency and significantly mutated genes in comparison to previously analysed adult cancers. Genetic alterations in 149 putative cancer driver genes separate the tumours into two classes: small mutation and structural/copy-number variant (correlating with germline variants). Structural variants, hyperdiploidy, and chromothripsis are linked to TP53 mutation status and mutational signatures. Our data suggest that 7-8% of the children in this cohort carry an unambiguous predisposing germline variant and that nearly 50% of paediatric neoplasms harbour a potentially druggable event, which is highly relevant for the design of future clinical trials.

958 citations


Journal ArticleDOI
28 Feb 2018-Nature
TL;DR: A pan-cancer study of somatic alterations, including single nucleotide variants, small insertions or deletions, structural variations, copy number alterations, gene fusions and internal tandem duplications in 1,699 paediatric leukaemias and solid tumours across six histotypes, provides a comprehensive genomic architecture for paediatric cancers.
Abstract: Analysis of molecular aberrations across multiple cancer types, known as pan-cancer analysis, identifies commonalities and differences in key biological processes that are dysregulated in cancer cells from diverse lineages. Pan-cancer analyses have been performed for adult but not paediatric cancers, which commonly occur in developing mesodermic rather than adult epithelial tissues. Here we present a pan-cancer study of somatic alterations, including single nucleotide variants, small insertions or deletions, structural variations, copy number alterations, gene fusions and internal tandem duplications in 1,699 paediatric leukaemias and solid tumours across six histotypes, with whole-genome, whole-exome and transcriptome sequencing data processed under a uniform analytical framework. We report 142 driver genes in paediatric cancers, of which only 45% match those found in adult pan-cancer studies; copy number alterations and structural variants constituted the majority (62%) of events. Eleven genome-wide mutational signatures were identified, including one attributed to ultraviolet-light exposure in eight aneuploid leukaemias. Transcription of the mutant allele was detectable for 34% of protein-coding mutations, and 20% exhibited allele-specific expression. These data provide a comprehensive genomic architecture for paediatric cancers and emphasize the need for paediatric cancer-specific development of precision therapies.

573 citations


Journal ArticleDOI
Sebastian M. Waszak, Paul A. Northcott1, Paul A. Northcott2, Ivo Buchhalter2, Giles W. Robinson1, Christian Sutter3, Susanne N. Groebner2, Kerstin Grund3, Laurence Brugières4, David T.W. Jones2, Kristian W. Pajtler5, Kristian W. Pajtler2, A. Sorana Morrissy6, Marcel Kool2, Dominik Sturm2, Dominik Sturm5, Lukas Chavez2, Aurélie Ernst2, Sebastian Brabetz2, Sebastian Brabetz5, Michael Hain2, Thomas Zichner, Maia Segura-Wang, Joachim Weischenfeldt7, Tobias Rausch, Balca R. Mardin, Xin Zhou1, Cristina Baciu8, Christian Lawerenz2, Jennifer A. Chan6, Pascale Varlet, Léa Guerrini-Rousseau4, Daniel W. Fults9, Wiesława Grajkowska, Peter Hauser10, Nada Jabado11, Young Shin Ra12, Karel Zitterbart13, Suyash Shringarpure14, Francisco M. De La Vega14, Carlos Bustamante14, Ho Keung Ng15, Arie Perry16, Tobey J. MacDonald17, Pablo Hernáiz Driever18, Anne Bendel19, Daniel C. Bowers20, Geoffrey McCowage21, Murali Chintagumpala22, Richard J. Cohn22, Tim Hassall22, Gudrun Fleischhack23, Tone Eggen, Finn Wesenberg24, Finn Wesenberg25, Maria Feychting26, Birgitta Lannering27, Joachim Schüz28, Christoffer Johansen7, Tina Veje Andersen, Martin Röösli29, Claudia E. Kuehni30, Michael A. Grotzer31, Kristina Kjaerheim, Camelia M. Monoranu32, Tenley C. Archer22, Tenley C. Archer33, Elizabeth S. Duke22, Scott L. Pomeroy33, Scott L. Pomeroy22, Redmond Shelagh30, Stephan Frank34, David Sumerauer35, Wolfram Scheurlen, Marina Ryzhova, Till Milde5, Till Milde2, Christian P. Kratz36, David Samuel22, Jinghui Zhang1, David A. Solomon16, Marco A. Marra37, Roland Eils2, Claus R. Bartram3, Katja von Hoff18, Katja von Hoff38, Stefan Rutkowski38, Vijay Ramaswamy39, Richard J. Gilbertson40, Andrey Korshunov5, Andrey Korshunov2, Michael D. Taylor, Peter Lichter2, David Malkin39, Amar Gajjar1, Jan O. Korbel, Stefan M. Pfister2, Stefan M. Pfister5 
TL;DR: The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup, and survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes.
Abstract: Summary Background Medulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines. Methods In this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MB WNT ), SHH (MB SHH ), group 3 (MB Group3 ), and group 4 (MB Group4 ). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma. Findings We included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU , and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MB SHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MB SHH subgroup). Patients with germline APC mutations developed MB WNT and accounted for most (five [71%] of seven) cases of MB WNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MB SHH . Germline TP53 mutations presented only in childhood patients in the MB SHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MB SHH , MB Group3 , and MB Group4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40–69) and 5-year overall survival was 65% (95% CI 52–81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes. Interpretation Genetic counselling and testing should be used as a standard-of-care procedure in patients with MB WNT and MB SHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics. Funding German Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Children's Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDT's Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.

229 citations


Journal ArticleDOI
TL;DR: The findings support referral of all survivors for genetic counseling for potential clinical genetic testing, which should be prioritized for nonirradiated survivors with any SN and for those with breast cancer or sarcoma in the field of prior irradiation.
Abstract: Purpose Childhood cancer survivors are at increased risk of subsequent neoplasms (SNs), but the germline genetic contribution is largely unknown. We assessed the contribution of pathogenic/likely pathogenic (P/LP) mutations in cancer predisposition genes to their SN risk. Patients and Methods Whole-genome sequencing (30-fold) was performed on samples from childhood cancer survivors who were ≥ 5 years since initial cancer diagnosis and participants in the St Jude Lifetime Cohort Study, a retrospective hospital-based study with prospective clinical follow-up. Germline mutations in 60 genes known to be associated with autosomal dominant cancer predisposition syndromes with moderate to high penetrance were classified by their pathogenicity according to the American College of Medical Genetics and Genomics guidelines. Relative rates (RRs) and 95% CIs of SN occurrence by mutation status were estimated using multivariable piecewise exponential regression stratified by radiation exposure. Results Participants were 3,006 survivors (53% male; median age, 35.8 years [range, 7.1 to 69.8 years]; 56% received radiotherapy), 1,120 SNs were diagnosed among 439 survivors (14.6%), and 175 P/LP mutations were identified in 5.8% (95% CI, 5.0% to 6.7%) of survivors. Mutations were associated with significantly increased rates of breast cancer (RR, 13.9; 95% CI, 6.0 to 32.2) and sarcoma (RR, 10.6; 95% CI, 4.3 to 26.3) among irradiated survivors and with increased rates of developing any SN (RR, 4.7; 95% CI, 2.4 to 9.3), breast cancer (RR, 7.7; 95% CI, 2.4 to 24.4), nonmelanoma skin cancer (RR, 11.0; 95% CI, 2.9 to 41.4), and two or more histologically distinct SNs (RR, 18.6; 95% CI, 3.5 to 99.3) among nonirradiated survivors. Conclusion The findings support referral of all survivors for genetic counseling for potential clinical genetic testing, which should be prioritized for nonirradiated survivors with any SN and for those with breast cancer or sarcoma in the field of prior irradiation.

95 citations



Journal ArticleDOI
TL;DR: Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins.

53 citations


Posted ContentDOI
29 Jul 2018-bioRxiv
TL;DR: It is shown that ATRX mutations and MYCN amplification are mutually exclusive across all ages and stages in neuroblastoma, and this synthetic lethality may eventually be exploited to improve outcomes for patients with high-risk Neuroblastoma.
Abstract: Aggressive cancers often have activating mutations in growth controlling oncogenes and inactivating mutations in tumor-suppressor genes. In neuroblastoma, amplification of the MYCN oncogene and inactivation of the ATRX tumor-suppressor gene correlate with high-risk disease and poor prognosis. Here we show that ATRX mutations and MYCN amplification are mutually exclusive across all ages and stages in neuroblastoma. Using human cell lines and mouse models, we found that elevated MYCN expression and ATRX mutations are incompatible. Elevated MYCN levels promote metabolic reprogramming, mitochondrial dysfunction, reactive-oxygen species generation, and DNA replicative stress. The combination of replicative stress caused by defects in the ATRX histone chaperone complex and that induced by MYCN mediated metabolic reprogramming leads to synthetic lethality. Therefore, ATRX and MYCN represent an unusual example, where inactivation of a tumor-suppressor gene and activation of an oncogene are incompatible. This synthetic lethality may eventually be exploited to improve outcomes for patients with high risk neuroblastoma.

32 citations


Journal ArticleDOI
Susanne Gröbner1, Barbara C. Worst, Joachim Weischenfeldt2, Joachim Weischenfeldt3, Ivo Buchhalter1, Kortine Kleinheinz1, Vasilisa A. Rudneva4, Vasilisa A. Rudneva3, Pascal Johann, Gnana Prakash Balasubramanian1, Maia Segura-Wang3, Sebastian Brabetz1, Sebastian Bender1, Barbara Hutter1, Dominik Sturm, Elke Pfaff, Daniel Hübschmann5, Daniel Hübschmann6, Daniel Hübschmann1, Gideon Zipprich1, Michael Heinold6, Michael Heinold1, Jürgen Eils1, Christian Lawerenz1, Serap Erkek, Sander Lambo1, Sebastian M. Waszak3, Claudia Blattmann1, Arndt Borkhardt7, Arndt Borkhardt1, Michaela Kuhlen7, Michaela Kuhlen1, Angelika Eggert8, Angelika Eggert1, Simone Fulda1, Manfred Gessler, Jenny Wegert, Roland Kappler9, Roland Kappler1, Daniel Baumhoer10, Stefan Burdach1, Stefan Burdach11, Renate Kirschner-Schwabe8, Renate Kirschner-Schwabe1, Udo Kontny1, Andreas E. Kulozik1, Andreas E. Kulozik5, Dietmar R. Lohmann1, Simone Hettmer12, Cornelia Eckert1, Cornelia Eckert8, Stefan S. Bielack, Michaela Nathrath1, Michaela Nathrath11, Charlotte M. Niemeyer1, Charlotte M. Niemeyer12, Gunther Richter1, Gunther Richter11, Johannes H. Schulte1, Johannes H. Schulte8, Reiner Siebert13, Frank Westermann1, Jan J. Molenaar, Gilles Vassal14, Hendrik Witt, Birgit Burkhardt, Christian P. Kratz15, Olaf Witt, Cornelis M. van Tilburg1, Christof M. Kramm16, Gudrun Fleischhack1, Gudrun Fleischhack17, Uta Dirksen17, Stefan Rutkowski18, Michael C. Frühwald19, Katja von Hoff18, Stephan Wolf1, Thomas Klingebiel1, Thomas Klingebiel19, Ewa Koscielniak, Pablo Landgraf, Jan Koster, Adam C. Resnick20, Jinghui Zhang4, Yanling Liu4, Xin Zhou4, Angela J. Waanders20, Danny A. Zwijnenburg, Pichai Raman20, Benedikt Brors1, Ursula D. Weber1, Paul A. Northcott4, Paul A. Northcott1, Kristian W. Pajtler, Marcel Kool1, Rosario M. Piro, Jan O. Korbel3, Matthias Schlesner1, Roland Eils6, Roland Eils1, David T.W. Jones1, Peter Lichter1, Lukas Chavez1, Marc Zapatka21, Marc Zapatka1, Stefan M. Pfister 
01 Jul 2018-Nature
TL;DR: In this Article, author Benedikt Brors was erroneously associated with affiliation number ‘8’ (Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, USA); the author’s two other affiliations were correct.
Abstract: In this Article, author Benedikt Brors was erroneously associated with affiliation number '8' (Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, USA); the author's two other affiliations (affiliations '3' and '7', both at the German Cancer Research Center (DKFZ)) were correct. This has been corrected online.

25 citations


Posted ContentDOI
06 Jun 2018-bioRxiv
TL;DR: PeCanPIE is a web- and cloud-based platform for annotation, identification, and classification of variations in known or putative disease genes, applied to classify variant pathogenicity in cancer predisposition genes in two large-scale investigations involving >4,000 pediatric cancer patients.
Abstract: Variant interpretation in the era of next-generation sequencing (NGS) is challenging. While many resources and guidelines are available to assist with this task, few integrated end-to-end tools exist. Here we present "PeCanPIE" — the Pediatric Cancer Variant Pathogenicity Information Exchange, a web- and cloud-based platform for annotation, identification, and classification of variations in known or putative disease genes. Starting from a set of variants in Variant Call Format (VCF), variants are annotated, ranked by putative pathogenicity, and presented for formal classification using a decision-support interface based on published guidelines from the American College of Medical Genetics and Genomics (ACMG). The system can accept files containing millions of variants and handle single-nucleotide variants (SNVs), simple insertions/deletions (indels), multiple-nucleotide variants (MNVs), and complex substitutions. PeCanPIE has been applied to classify variant pathogenicity in cancer predisposition genes in two large-scale investigations involving >4,000 pediatric cancer patients, and serves as a repository for the expert-reviewed results. While PeCanPIE9s web-based interface was designed to be accessible to non-bioinformaticians, its back end pipelines may also be run independently on the cloud, facilitating direct integration and broader adoption. PeCanPIE is publicly available and free for research use.

11 citations


Journal ArticleDOI
29 Nov 2018-Blood
TL;DR: The St. Jude Cloud is expanded to sickle cell disease data through the Sickle Genome Project (SGP) Data Portal to allow instantaneous raw data access (following data access committee approval), as well as visualization of genotype calls at individual level in a novel genome.

3 citations


Proceedings ArticleDOI
TL;DR: The mechanism underlying aberrant MYC overexpression, in a subset of neuroblastomas (NBL) lacking MYCN amplification, proves the power of this tool in facilitating the discovery of oncogenic SV drivers, and provides an integrative analysis tool to evaluate the pathogenic basis underlying the often complex SVs in noncoding genome.
Abstract: Somatic structural variations (SV) play an important role in tumorigenesis as they may cause oncogenic gene fusions or transcriptional activation of oncogenes by introducing aberrant promoter-enhancer interactions. Evaluating the oncogenic implications of SVs is challenging, especially for SVs in noncoding regions, which requires integrating data from 1) whole-genome sequencing (WGS), 2) RNA-seq from the tumor sample and series patient samples with same tumor type to compare with, 3) epigenetic profiling (ChIP-seq), and 4) genome-wide chromosome conformation capture studies such as Hi-C. To facilitate the discovery of oncogenic SVs, we developed GenomePaint, an interactive browser to integrate and visualize somatic SVs and copy number alterations (CNAs) analyzed by WGS, and gene expression data analyzed by RNA-seq in >2,000 pediatric cancers, as well as ChIP-seq and Hi-C data from pediatric tumors and cell lines. The genomic profiling of patient samples was generated by the St Jude/Washington University Pediatric Cancer Genome Project and the NCI-TARGET project, and includes all major subtypes of pediatric leukemia, solid tumors and brain tumors. A global view of the entire patient cohort using GenomePaint revealed SV hotspots (e.g. within first intron of TP53 in osterosarcoma) and recurrent CNAs (e.g. TAL1 deletion in T-ALL) in each cancer type. Each SV was integrated with results on gene expression, ChIP-seq and Hi-C, on the rearranged chromosome. Our discovery of the mechanism underlying aberrant MYC overexpression, in a subset of neuroblastomas (NBL) lacking MYCN amplification, proves the power of this tool in facilitating the discovery of oncogenic SV drivers. The sample with the highest MYC expression in this cohort harbors an SV between chr 8 and 4, with breakpoint on chr 8 located 50 Kb downstream of MYC. Similar SVs were detected in multiple NBL cell lines by WGS, which also express high levels of MYC. By analyzing Hi-C data from these cell lines, a new topologically associating domain (TAD) extending beyond the SV breakpoint was observed along the chimeric chromosome. This new TAD shows an aberrant interaction of a super-enhancer with a broad H3K27ac peak associated with HAND2/FBXO8 on chr 4 and the MYC promoter, demonstrating the rewired regulatory architecture introduced by SV as the driver for MYC dysregulation. In conclusion, our new GenomePaint interactive browser facilitates the analysis of SVs, CNAs and gene expression in concert studies by WGS and Hi-C in pediatric tumors. Coding mutations are rarely identified in many types of childhood tumors, even in the presence of pronounced chromosomal SVs, indicating new approaches are needed to unveil oncogenic mechanisms. GenomePaint provides an integrative analysis tool to evaluate the pathogenic basis underlying the often complex SVs in noncoding genome and function as oncogenic drivers in a large fraction of pediatric malignancies. Citation Format: Yu Liu, Xin Zhou, Jian Wang, Ying Shao, John Easton, Mark W. Zimmerman, Brian J. Abraham, A. Thomas Look, Jinghui Zhang. Exploring somatic DNA structural alteration and aberrant genomic interactions in cancer through GenomePaint [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1287.

Journal ArticleDOI
29 Nov 2018-Blood
TL;DR: Novel subtypes of B-ALL are identified with distinct GEP characterized by PAX5 alterations, which are identical to those observed in "double/triple hit" lymphoma, and are of pre-B immunophenotype.

Proceedings ArticleDOI
TL;DR: This is the first assessment of the joint effects of rare and common genetic variations implicated in the etiology of BC in the general population, among long-term survivors of childhood cancer.
Abstract: The allelic spectrum of the genetic architecture of breast cancer (BC) susceptibility includes at least 172 common variants with small effect sizes (per-allele odds ratio range: 1.03-1.31), plus rare variants with high (BRCA1, BRCA2, CHD1, PTEN, STK11, TP53) or moderate penetrance (ATM, CHEK2, NBN, NF1, PALB2). While these common variants confer modest risk individually, their combined effect in the form of a polygenic risk score (PRS) may be substantial. The SJLIFE whole-genome sequencing (WGS) data provide a unique opportunity to evaluate common and rare sets of genetic variants jointly, along with treatment exposures, for their contributions to subsequent BC risk in adult survivors of childhood cancer. This analysis utilized WGS data from 1131 females of European ancestry [median age at last follow-up: 34.9 years (range: 6.2-68.6)] of whom 47 were diagnosed with a subsequent BC (median age at BC 40.3 years, range: 25.5-53.0). The PRS (mean, 10.1; range, 8.3-12.2) was calculated using a weighted sum of the number of risk alleles and their log per-allele odds ratio from Michailidou et al. (Nature, Nov. 2017). A total of 34 (3.0%) survivors were carriers of pathogenic or likely pathogenic (P/LP) variants in the 11 BC predisposition genes (listed above). The standardized incidence ratio (SIR) for BC was 6.7 (95% CI, 5.0-8.9) for survivors relative to the SEER population. The SIR varied from 3.7 (95% CI, 1.4-8.1) for survivors with PRS in the 1st quintile to 3.6 (95% CI, 1.2-8.3), 7.3 (95% CI, 3.8-12.7), 7.6 (95% CI, 3.6-14.0), and 11.4 (95% CI, 6.8-18.1) in the 2nd, 3rd, 4th, and 5th quintiles, respectively. In the multivariable model adjusting for age at diagnosis, chest irradiation, alkylating agents, anthracyclines, attained age, and significant genotype eigenvectors, the relative rates (RR) of BC were 16.5 (95% CI, 6.4 - 42.6), 11.5 (95% CI, 4.4-29.9), and 47.8 (95% CI, 8.2-278.3) for carriers vs. non-carriers of the P/LP variants among all survivors, and survivors with and without chest irradiation, respectively. The RR per one standard deviation of PRS were 1.5 (95% CI, 1.1-1.9), 1.6 (95% CI, 1.2-2.0) and 1.3 (95% CI, 0.7-2.2), respectively, for the same three groups. Importantly, PRS was significantly associated with the rate of subsequent BC under the age of 45 (RR, 1.7; 95% CI, 1.3-2.2) but not over 45 (RR, 0.9; 95% CI, 0.6-1.5). To our knowledge, this is the first assessment of the joint effects of rare and common genetic variations implicated in the etiology of BC in the general population, among long-term survivors of childhood cancer. Clinically, we anticipate that an individual genetic profile utilizing common susceptibility loci in combination with rare P/LP variants will inform an improved strategy for personalized BC risk stratification and management for childhood cancer survivors. Further replication studies are warranted to confirm and refine our findings. Citation Format: Zhaoming Wang, Carmen L. Wilson, Qi Liu, John Easton, Heather L. Mulder, Michael Rusch, Michael Edmonson, Shawn Levy, Aman Patel, Ying Shao, Ti-Cheng Chang, Stephen V. Rice, Yadav Sapkota, Russell J. Brooke, Wonjong Moon, Evadnie Rampersaud, Xiaotu Ma, Cynthia Pepper, Xin Zhou, Xiang Chen, Wenan Chen, Angela Jones, Braden Boone, Matthew J. Ehrhardt, Rebecca M. Howell, Nicholas Phillips, Courtney Lewis, Chimene A. Kesserwan, Gang Wu, Kim E. Nichols, James R. Downing, Melissa M. Hudson, Jinghui Zhang, Yutaka Yasui, Leslie L. Robison. Monogenic and polygenic associations with subsequent breast cancer risk in survivors of childhood cancer: The St. Jude Lifetime Cohort Study (SJLIFE) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3007.