scispace - formally typeset
Search or ask a question

Showing papers by "Yon-Dschun Ko published in 2021"


Journal ArticleDOI
TL;DR: The results of this study define the genes that are most clinically useful for inclusion on panels for the prediction of breast cancer risk, as well as provide estimates of the risks associated with protein-truncating variants, to guide genetic counseling.
Abstract: BACKGROUND Genetic testing for breast cancer susceptibility is widely used, but for many genes, evidence of an association with breast cancer is weak, underlying risk estimates are imprecise, and reliable subtype-specific risk estimates are lacking. METHODS We used a panel of 34 putative susceptibility genes to perform sequencing on samples from 60,466 women with breast cancer and 53,461 controls. In separate analyses for protein-truncating variants and rare missense variants in these genes, we estimated odds ratios for breast cancer overall and tumor subtypes. We evaluated missense-variant associations according to domain and classification of pathogenicity. RESULTS Protein-truncating variants in 5 genes (ATM, BRCA1, BRCA2, CHEK2, and PALB2) were associated with a risk of breast cancer overall with a P value of less than 0.0001. Protein-truncating variants in 4 other genes (BARD1, RAD51C, RAD51D, and TP53) were associated with a risk of breast cancer overall with a P value of less than 0.05 and a Bayesian false-discovery probability of less than 0.05. For protein-truncating variants in 19 of the remaining 25 genes, the upper limit of the 95% confidence interval of the odds ratio for breast cancer overall was less than 2.0. For protein-truncating variants in ATM and CHEK2, odds ratios were higher for estrogen receptor (ER)-positive disease than for ER-negative disease; for protein-truncating variants in BARD1, BRCA1, BRCA2, PALB2, RAD51C, and RAD51D, odds ratios were higher for ER-negative disease than for ER-positive disease. Rare missense variants (in aggregate) in ATM, CHEK2, and TP53 were associated with a risk of breast cancer overall with a P value of less than 0.001. For BRCA1, BRCA2, and TP53, missense variants (in aggregate) that would be classified as pathogenic according to standard criteria were associated with a risk of breast cancer overall, with the risk being similar to that of protein-truncating variants. CONCLUSIONS The results of this study define the genes that are most clinically useful for inclusion on panels for the prediction of breast cancer risk, as well as provide estimates of the risks associated with protein-truncating variants, to guide genetic counseling.

407 citations


Journal ArticleDOI
TL;DR: Variation in projected absolute lifetime risk of breast cancer associated with classical risk factors was greater for women with higher genetic risk (PRS313 and family history) and, on average, 17.5% higher in the highest vs lowest deciles of genetic risk.
Abstract: We evaluated the joint associations between a new 313-variant PRS (PRS313) and questionnaire-based breast cancer risk factors for women of European ancestry, using 72 284 cases and 80 354 controls from the Breast Cancer Association Consortium. Interactions were evaluated using standard logistic regression and a newly developed case-only method for breast cancer risk overall and by estrogen receptor status. After accounting for multiple testing, we did not find evidence that per-standard deviation PRS313 odds ratio differed across strata defined by individual risk factors. Goodness-of-fit tests did not reject the assumption of a multiplicative model between PRS313 and each risk factor. Variation in projected absolute lifetime risk of breast cancer associated with classical risk factors was greater for women with higher genetic risk (PRS313 and family history) and, on average, 17.5% higher in the highest vs lowest deciles of genetic risk. These findings have implications for risk prevention for women at increased risk of breast cancer.

45 citations



Journal ArticleDOI
03 Jul 2021-Cancers
TL;DR: In this paper, the authors analyzed the predictive value of frailty and sarcopenia as assessment tools for physiological integrity in patients with non-small cell lung cancer (NSCLC) who had undergone surgery for brain metastasis.
Abstract: Neurosurgical resection represents an important therapeutic pillar in patients with brain metastasis (BM). Such extended treatment modalities require preoperative assessment of patients' physical status to estimate individual treatment success. The aim of the present study was to analyze the predictive value of frailty and sarcopenia as assessment tools for physiological integrity in patients with non-small cell lung cancer (NSCLC) who had undergone surgery for BM. Between 2013 and 2018, 141 patients were surgically treated for BM from NSCLC at the authors' institution. The preoperative physical condition was assessed by the temporal muscle thickness (TMT) as a surrogate parameter for sarcopenia and the modified frailty index (mFI). For the ≥65 aged group, median overall survival (mOS) significantly differed between patients classified as 'frail' (mFI ≥ 0.27) and 'least and moderately frail' (mFI < 0.27) (15 months versus 11 months (p = 0.02)). Sarcopenia revealed significant differences in mOS for the <65 aged group (10 versus 18 months for patients with and without sarcopenia (p = 0.036)). The present study confirms a predictive value of preoperative frailty and sarcopenia with respect to OS in patients with NSCLC and surgically treated BM. A combined assessment of mFI and TMT allows the prediction of OS across all age groups.

7 citations


Journal ArticleDOI
TL;DR: In this article, postoperative prolonged mechanical ventilation (PMV) was found to be a significant prognostic factor for OS after surgical treatment in patients with brain metastasis, independent of other predictive factors such as age, location, and preoperative physical status.
Abstract: Objective: Surgical resection represents a common treatment modality in patients with brain metastasis (BM). Postoperative prolonged mechanical ventilation (PMV) might have an enormous impact on the overall survival (OS) of these patients suffering from advanced cancer disease. We therefore have analyzed our institutional database with regard to a potential impact of PMV on OS of patients who had undergone surgery for brain metastases. Methods: 360 patients with surgically treated brain metastases were included. The definition of PMV consisted of postoperative mechanical ventilation lasting for more than 48 hours. Analysis of survival incorporating established prognostic factors such as age, location of BM, and preoperative physical status was performed. Results: 14 of 360 patients with BM (4%) suffered from postoperative PMV after surgical treatment of BM. Patients with PMV presented in a significantly more impaired neurological condition preoperatively than patients without (p<0.0001). Multivariate analysis determined PMV to be a significant prognostic factor for OS after surgical treatment in patients with BM, independent of other predictive factors (p<0.0001). Conclusions: The present study demonstrates postoperative PMV as significantly related to poor OS in patients with surgically treated BM. Postoperative PMV is a so far underestimated prognostic predictor, but might be utilized for optimized patient management early in the postoperative phase. For this purpose, the results of the present study should encourage the initiation of further scientific efforts.

7 citations


Journal ArticleDOI
TL;DR: In this paper, the pre-operative frailty was associated with poor survival in elderly patients with brain metastases requiring surgery, and the authors used the modified Frailty Index (mFI) as predictors for reduced OS in older patients undergoing surgical treatment for BM.
Abstract: Object In the light of an aging population and ongoing advances in cancer control, the optimal management in geriatric patients with brain metastases (BM) poses an increasing challenge, especially due to the scarce data available. We therefore analyzed our institutional data with regard to factors influencing overall survival (OS) in geriatric patients with BM. Methods Between 2013 and 2018, patients aged ≥ 65 years with surgically treated BM were included in this retrospective analysis. In search of preoperatively identifiable risk factors for poor OS, in addition to the underlying cancer, the preoperative frailty of patients was analyzed using the modified Frailty Index (mFI). Results A total of 180 geriatric patients with surgically treated BM were identified. Geriatric patients categorized as least-frail achieved a median OS of 18 months, whereas frailest patients achieved an OS of only 3 months (p 5 mg/l" (p=0.01) and "frailest patients (mFI ≥ 0.27)" (p=0.002) as predictors for reduced OS in older patients undergoing surgical treatment for BM. Conclusions In this retrospective series, pre-operative frailty was associated with poor survival in elderly patients with BM requiring surgery. Our analyses warrant thorough counselling and support of affected elderly patients and their families.

6 citations


Journal ArticleDOI
TL;DR: In this article, an alternative diagnostic approach for automatic and label-free detection of mutations in lung adenocarcinoma tissue using quantum cascade laser-based infrared imaging is presented.
Abstract: Therapeutic decisions in lung cancer critically depend on the determination of histologic types and oncogene mutations. Therefore, tumor samples are subjected to standard histologic and immunohistochemical analyses and examined for relevant mutations using comprehensive molecular diagnostics. In this study, an alternative diagnostic approach for automatic and label-free detection of mutations in lung adenocarcinoma tissue using quantum cascade laser–based infrared imaging is presented. For this purpose, a five-step supervised classification algorithm was developed, which was not only able to detect tissue types and tumor lesions, but also the tumor type and mutation status of adenocarcinomas. Tumor detection was verified on a data set of 214 patient samples with a specificity of 97% and a sensitivity of 95%. Furthermore, histology typing was verified on samples from 203 of the 214 patients with a specificity of 97% and a sensitivity of 94% for adenocarcinoma. The most frequently occurring mutations in adenocarcinoma (KRAS, EGFR, and TP53) were differentiated by this technique. Detection of mutations was verified in 60 patient samples from the data set with a sensitivity and specificity of 95% for each mutation. This demonstrates that quantum cascade laser infrared imaging can be used to analyze morphologic differences as well as molecular changes. Therefore, this single, one-step measurement provides comprehensive diagnostics of lung cancer histology types and most frequent mutations.

5 citations


Journal ArticleDOI
TL;DR: A prespecified interim analysis following induction therapy with the quadruplet regimen isatuximab, carfilzomib, lenalidomide and dexamethasone in newly diagnosed HR MM patients, finding Isa-KRd induction induces rapid and deep responses and the overall safety profile is consistent with previous reports.
Abstract: Background: High-risk (HR) multiple myeloma (MM) has an impaired prognostic outcome. Addition of anti-CD38 monoclonal antibodies to standard-of-care regimens improved response rates and depth of response. Here, we report a prespecified interim analysis (IA) following induction therapy with the quadruplet regimen isatuximab, carfilzomib, lenalidomide and dexamethasone (Isa-KRd) in newly diagnosed (ND) HR MM patients (pts) in the phase II multicentric GMMG-CONCEPT trial (NCT03104842). Methods: 153 pts with HR NDMM were enrolled into the trial, the IA reports on the first 50 pts evaluable for IA. Pts receive Isa-KRd in induction, consolidation and Isa-KR maintenance. Transplant eligible pts undergo high-dose therapy. This IA reports on overall response rates (ORR) during induction. Findings: 50 pts were included in the IA population for ORR. HR MM was defined by del17p in 52%, t(4;14) in 38%, t(14;16) in 12% and > 3 copies 1q21 in 42% and ISS stage 2/3 disease. 46/50 pts completed induction treatment. ORR was 100%, with 5 pts (10·0%) showing partial response (PR), 22 (44·0%; including 4 in arm B) very good partial response (VGPR) and 23 (46·0%) complete response (CR). Hematologic grade 3/4 treatment-emergent adverse events (≥ 10%) were neutropenia (34·0%), leukopenia (26·0%) and thrombocytopenia (14·0%). Main non-hematologic toxicities grade 3/4 were hypertension (12·0%) and infection (8·0%). Interpretation: We report for the first time on a trial investigating Isa-KRd quadruplet treatment in solely HR NDMM. Isa-KRd induction induces rapid and deep responses. The overall safety profile is consistent with previous reports. Trial Registration: The trial is registered at clinicaltrials.gov (NCT03104842). Funding Statement: Study drug and financial support by Amgen, Celgene-BMS and Sanofi. Declaration of Interests: Dr. Leypoldt reports grants and non-financial support from Celgene-BMS, grants and non-financial support from Sanofi, grants and non-financial support from Amgen, during the conduct of the study; non-financial support from GSK, non-financial support from Abbvie, outside the submitted work; Dr. Asemissen has nothing to disclose. Dr. Besemer has nothing to disclose. Dr. Hanel reports personal fees from Celgene, personal fees from Novartis, personal fees from Takeda, personal fees from Amgen, during the conduct of the study; Dr. Blau has nothing to disclose. Dr. Gorner has nothing to disclose. Dr. Ko has nothing to disclose. Dr. Reinhardt reports personal fees from Abbvie, grants from Gilead, personal fees from Merck, other from CDL Therapeutics GmbH, outside the submitted work; Dr. Staib reports grants, personal fees, non-financial support and other from Abbvie, grants, personal fees, non-financial support and other from Amgen, grants, personal fees, non-financial support and other from Celgene, grants, personal fees, non-financial support and other from Janssen-Cilag, grants, personal fees, non-financial support and other from Novartis, grants, personal fees, non-financial support and other from Gilead, grants, personal fees, non-financial support and other from Pfizer, grants, personal fees, non-financial support and other from Roche, outside the submitted work; Dr. Mann has nothing to disclose. Dr. Lutz has nothing to disclose. Dr. Munder reports personal fees and non-financial support from Janssen, personal fees and non-financial support from Amgen, grants from Incyte, personal fees and non-financial support from BMS, personal fees from Abbvie, personal fees from Sanofi, personal fees from GSK, personal fees from Takeda, outside the submitted work; Dr. Graeven reports personal fees from Amgen, personal fees and non-financial support from Boehringer Ingelheim, personal fees from Daichi Sankyo, personal fees from Servier, personal fees from Celgene, personal fees from Astra Zeneca, personal fees from Johnson Johnson, non-financial support from Merck, personal fees from MSD, personal fees from BMS, during the conduct of the study; Dr. Peceny reports grants and personal fees from Sanofi Genzyme, grants from Novartis, grants from DRK Blutspendedienst NSTOB, grants from Boehringer Ingelheim Pharma GmbH & Co KG, grants from Celgene, outside the submitted work; Dr. Salwender reports personal fees from Bristol-Myers Squibb/Celgene, personal fees from Janssen Cilag, personal fees from Glaxo Smith Kline, personal fees from Oncopeptides, personal fees from Takeda, personal fees from Sanofi, personal fees from AbbVie, personal fees from Amgen, outside the submitted work; Dr. Jauch has nothing to disclose. Dr. Zago has nothing to disclose. Axel Benner has nothing to disclose. Dr. Tichy has nothing to disclose. Dr. Bokemeyer reports personal fees from Sanofi Aventis, personal fees from Merck KgA, personal fees from Bristol-Myers Squibb, personal fees from Merck Sharp & Dohme, personal fees from Lilly Imclone, personal fees from Bayer Healthcare, personal fees from GSO Contract research, personal fees from AOK-Rheinland-Hamburg, personal fees from Novartis, outside the submitted work; Dr. Goldschmidt reports grants, personal fees, non-financial support and other from Amgen, grants, personal fees, non-financial support and other from BMS, grants, personal fees, non-financial support and other from Celgene, grants, personal fees, and other from Chugai, grants, personal fees, non-financial support and other from Janssen, grants, personal fees, non-financial support and other from Sanofi, other from Incyte, other from Molecular Partners, other from Merck Sharp and Dohme (MSD), other from Mundipharma, grants, personal fees, non-financial support and other from Takeda, personal fees and other from Novartis, personal fees from Adaptive Biotechnology, personal fees from GlaxoSmithKline (GSK), outside the submitted work. Dr. Weisel reports grants from AMGEN, grants from Celgene, grants from Sanofi, during the conduct of the study; grants, personal fees and non-financial support from Amgen, personal fees and non-financial support from BMS, grants, personal fees and non-financial support from Celgene, personal fees from Adaptive Biotech, grants, personal fees and non-financial support from Janssen, personal fees and non-financial support from GSK, personal fees from Karyopharm, grants, personal fees and non-financial support from Sanofi, personal fees and non-financial support from Takeda, personal fees from Oncopeptides, personal fees from Roche, outside the submitted work. Ethics Approval Statement: All patients provided written informed consent. The trial was approved by the competent authorities and the Tuebingen University Ethics Committee.

1 citations


Posted ContentDOI
TL;DR: This immunomethylomic study shows that a shift in the ratios/proportions of leukocyte subtypes is associated with TNBC, with decreased NK cell showing the strongest association.
Abstract: BACKGROUND A shift in the proportions of blood immune cells is a hallmark of cancer development. Here, we investigated whether methylation-derived immune cell type ratios and methylation-derived neutrophil-to-lymphocyte ratios (mdNLRs) are associated with triple-negative breast cancer (TNBC). METHODS Leukocyte subtype-specific unmethylated/methylated CpG sites were selected, and methylation levels at these sites were used as proxies for immune cell type proportions and mdNLR estimation in 231 TNBC cases and 231 age-matched controls. Data were validated using the Houseman deconvolution method. Additionally, the natural killer (NK) cell ratio was measured in a prospective sample set of 146 TNBC cases and 146 age-matched controls. RESULTS The mdNLRs were higher in TNBC cases compared with controls and associated with TNBC (odds ratio (OR) range (2.66-4.29), all Padj. < 1e-04). A higher neutrophil ratio and lower ratios of NK cells, CD4 + T cells, CD8 + T cells, monocytes, and B cells were associated with TNBC. The strongest association was observed with decreased NK cell ratio (OR range (1.28-1.42), all Padj. < 1e-04). The NK cell ratio was also significantly lower in pre-diagnostic samples of TNBC cases compared with controls (P = 0.019). CONCLUSION This immunomethylomic study shows that a shift in the ratios/proportions of leukocyte subtypes is associated with TNBC, with decreased NK cell showing the strongest association. These findings improve our knowledge of the role of the immune system in TNBC and point to the possibility of using NK cell level as a non-invasive molecular marker for TNBC risk assessment, early detection, and prevention.

1 citations


Journal ArticleDOI
TL;DR: In this paper, the authors identified several risk factors for a reduced survival in patients receiving a surgical treatment of brain metastases derived from non-small cell lung cancer (NSCLC).
Abstract: Background: Brain metastases (BM) indicate advanced states of cancer disease and cranial surgery represents a common treatment modality. In the present study, we aimed to identify the risk factors for a reduced survival in patients receiving a surgical treatment of BM derived from non-small cell lung cancer (NSCLC). Methods: A total of 154 patients with NSCLC that had been surgically treated for BM at the authors’ institution between 2013 and 2018 were included for a further analysis. A multivariate analysis was performed to identify the predictors of a poor overall survival (OS). Results: The median overall survival (mOS) was 11 months (95% CI 8.2–13.8). An age > 65 years, the infratentorial location of BM, elevated preoperative C-reactive protein levels, a perioperative red blood cell transfusion, postoperative prolonged mechanical ventilation (>48 h) and the occurrence of postoperative adverse events were identified as independent factors of a poor OS. Conclusions: The present study identified several predictors for a worsened OS in patients that underwent surgery for BM of NSCLC. These findings might guide a better risk/benefit assessment in the course of metastatic NSCLC therapy and might help to more sufficiently cope with the challenges of cancer therapy in these advanced stages of disease.

Posted ContentDOI
Leila Dorling1, Sara Carvalho1, Jamie Allen1, Michael T. Parsons2, Cristina Fortuno2, Anna González-Neira, Stephan M. Heijl, Muriel A. Adank3, Thomas U. Ahearn4, Irene L. Andrulis5, Irene L. Andrulis6, Päivi Auvinen7, Heiko Becher8, Matthias W. Beckmann9, Sabine Behrens10, Marina Bermisheva11, Natalia Bogdanova12, Stig E. Bojesen13, Stig E. Bojesen14, Manjeet K. Bolla1, Michael Bremer12, Ignacio Briceño15, Nicola J. Camp16, Archie Campbell17, Jose E. Castelao, Jenny Chang-Claude8, Jenny Chang-Claude10, Stephen J. Chanock4, Georgia Chenevix-Trench2, J. Margriet Collée18, Kamila Czene19, Joe Dennis1, Thilo Dörk12, Mikael Eriksson19, D. Gareth Evans, Peter A. Fasching9, Peter A. Fasching20, Jonine D. Figueroa17, Jonine D. Figueroa4, Henrik Flyger13, Marike Gabrielson19, Manuela Gago-Dominguez21, Montserrat Garcia-Closas4, Graham G. Giles22, Graham G. Giles23, Graham G. Giles24, Gord Glendon5, Pascal Guénel25, Melanie Gündert10, Melanie Gündert26, Andreas Hadjisavvas27, Eric Hahnen28, Per Hall19, Ute Hamann10, Elaine F. Harkness29, Elaine F. Harkness30, Mikael Hartman31, Mikael Hartman32, Frans B. L. Hogervorst3, Antoinette Hollestelle33, Reiner Hoppe34, Reiner Hoppe35, Anthony Howell29, Anthony Howell30, kConFab Investigators23, kConFab Investigators36, Sgbcc Investigators, Anna Jakubowska37, Anna Jakubowska38, Audrey Y. Jung10, Elza Khusnutdinova39, Elza Khusnutdinova11, Sung-Won Kim40, Yon-Dschun Ko, Vessela N. Kristensen41, Vessela N. Kristensen42, Inge M. M. Lakeman43, Jingmei Li44, Jingmei Li32, Annika Lindblom19, Annika Lindblom45, Maria A. Loizidou27, Artitaya Lophatananon29, Jan Lubinski38, Craig Luccarini1, Michael J. Madsen16, Arto Mannermaa7, Mehdi Manoochehri10, Sara Margolin19, Dimitrios Mavroudis, Roger L. Milne22, Roger L. Milne24, Roger L. Milne23, Nur Aishah Taib46, Nur Aishah Taib47, Kenneth Muir29, Heli Nevanlinna48, William G. Newman30, Jan C. Oosterwijk49, Sue K. Park50, Sue K. Park51, Paolo Peterlongo, Paolo Radice, Emmanouil Saloustros, Elinor J. Sawyer52, Rita K. Schmutzler28, Mitul Shah1, Xueling Sim32, Melissa C. Southey24, Melissa C. Southey22, Melissa C. Southey23, Harald Surowy10, Harald Surowy26, Maija Suvanto48, Ian Tomlinson53, Ian Tomlinson54, Diana Torres55, Diana Torres10, Thérèse Truong25, Christi J. van Asperen43, Regina Waltes12, Qin Wang1, Xiaohong R. Yang4, Paul D.P. Pharoah1, Marjanka K. Schmidt3, Javier Benitez, Bas Vroling56, Alison M. Dunning1, Soo Hwang Teo47, Soo Hwang Teo46, Anders Kvist57, Miguel de la Hoya58, Peter Devilee43, Amanda B. Spurdle2, Maaike P.G. Vreeswijk43, Douglas F. Easton1 
University of Cambridge1, QIMR Berghofer Medical Research Institute2, Netherlands Cancer Institute3, National Institutes of Health4, Lunenfeld-Tanenbaum Research Institute5, University of Toronto6, University of Eastern Finland7, University of Hamburg8, University of Erlangen-Nuremberg9, German Cancer Research Center10, Russian Academy of Sciences11, Hannover Medical School12, Copenhagen University Hospital13, University of Copenhagen14, Universidad de La Sabana15, Huntsman Cancer Institute16, University of Edinburgh17, Erasmus University Medical Center18, Karolinska Institutet19, University of California, Los Angeles20, University of California, San Diego21, Cancer Council Victoria22, University of Melbourne23, Monash University24, Université Paris-Saclay25, Heidelberg University26, The Cyprus Institute of Neurology and Genetics27, University of Cologne28, University of Manchester29, Manchester Academic Health Science Centre30, University Health System31, National University of Singapore32, Erasmus University Rotterdam33, University of Tübingen34, Bosch35, Peter MacCallum Cancer Centre36, Laboratory of Molecular Biology37, Pomeranian Medical University38, Bashkir State University39, University of Saint Mary40, University of Oslo41, Oslo University Hospital42, Leiden University Medical Center43, Genome Institute of Singapore44, Karolinska University Hospital45, The Breast Cancer Research Foundation46, University of Malaya47, University of Helsinki48, University Medical Center Groningen49, Seoul National University50, New Generation University College51, King's College London52, Wellcome Trust Centre for Human Genetics53, University of Birmingham54, Pontifical Xavierian University55, Radboud University Nijmegen56, Lund University57, Hospital Clínico San Carlos58
15 Sep 2021-medRxiv
TL;DR: In this paper, the authors evaluated breast cancer risks according to five in-silico prediction-of-deleteriousness algorithms, functional protein domain, and frequency, using logistic regression models and also mixture models in which a subset of variants was assumed to be risk-associated.
Abstract: BACKGROUND Protein truncating variants in ATM, BRCA1, BRCA2, CHEK2 and PALB2 are associated with increased breast cancer risk, but risks associated with missense variants in these genes are uncertain. METHODS Combining 59,639 breast cancer cases and 53,165 controls, we sampled training (80%) and validation (20%) sets to analyze rare missense variants in ATM (1,146 training variants), BRCA1 (644), BRCA2 (1,425), CHEK2 (325) and PALB2 (472). We evaluated breast cancer risks according to five in-silico prediction-of-deleteriousness algorithms, functional protein domain, and frequency, using logistic regression models and also mixture models in which a subset of variants was assumed to be risk-associated. RESULTS The most predictive in-silico algorithms were Helix (BRCA1, BRCA2 and CHEK2) and CADD (ATM). Increased risks appeared restricted to functional protein domains for ATM (FAT and PIK domains) and BRCA1 (RING and BRCT domains). For ATM, BRCA1 and BRCA2, data were compatible with small subsets (approximately 7%, 2% and 0.6%, respectively) of rare missense variants giving similar risk to those of protein truncating variants in the same gene. For CHEK2, data were more consistent with a large fraction (approximately 60%) of rare missense variants giving a lower risk (OR 1.75, 95% CI (1.47-2.08)) than CHEK2 protein truncating variants. There was little evidence for an association with risk for missense variants in PALB2. The best fitting models were well calibrated in the validation set. CONCLUSIONS These results will inform risk prediction models and the selection of candidate variants for functional assays, and could contribute to the clinical reporting of gene panel testing for breast cancer susceptibility.

Posted ContentDOI
Joe Dennis1, Tyrer Jp1, Logan C. Walker2, Kyriaki Michailidou3, Kyriaki Michailidou1, Leila Dorling1, Manjeet K. Bolla1, Qinghua Wang1, Thomas U. Ahearn4, Irene L. Andrulis5, Irene L. Andrulis6, Hoda Anton-Culver7, Antonenkova Nn, Arndt8, Kristan J. Aronson9, Beane Freeman Le4, Matthias W. Beckmann10, Sabine Behrens8, Javier Benítez, Marina Bermisheva11, Natalia Bogdanova12, Stig E. Bojesen13, Stig E. Bojesen14, H Brenner8, Jose Esteban Castelao, Jenny Chang-Claude8, Jenny Chang-Claude15, Georgia Chenevix-Trench16, Christine L. Clarke17, Collee Jm18, Fergus J. Couch19, Cox A20, Simon S. Cross20, Kamila Czene21, Peter Devilee22, Thilo Dörk12, Laure Dossus23, A. H. Eliassen24, A. H. Eliassen25, Mikael Eriksson21, D G Evans26, Peter A. Fasching10, Peter A. Fasching27, Jonine D. Figueroa28, Jonine D. Figueroa4, Olivia Fletcher29, Henrik Flyger14, Lin Fritschi30, Marike Gabrielson21, Manuela Gago-Dominguez31, M Garcia-Closas4, Graham G. Giles32, Graham G. Giles33, Graham G. Giles34, Anna González-Neira, Pascal Guénel35, Eric Hahnen36, Christopher A. Haiman37, Per Hall21, Antoinette Hollestelle38, Reiner Hoppe39, Reiner Hoppe40, John L. Hopper32, Anthony Howell41, Abctb Investigators17, Investigators k42, Investigators k32, Agnes Jager38, Anna Jakubowska43, Anna Jakubowska44, Esther M. John45, Nichola Johnson29, Michael Jones29, Audrey Y. Jung8, Rudolph Kaaks8, Renske Keeman46, Elza Khusnutdinova47, Elza Khusnutdinova11, Cari M. Kitahara, Yon-Dschun Ko, Kosma48, Stella Koutros4, P. Kraft25, Vessela N. Kristensen49, Vessela N. Kristensen50, Kubelka-Sabit K, Allison W. Kurian45, James V. Lacey51, D Lambrechts52, Nicole L. Larson19, Martha S. Linet, Alicja Lukomska43, Arto Mannermaa48, Siranoush Manoukian, Sara Margolin21, Dimitrios Mavroudis, Roger L. Milne34, Roger L. Milne32, Roger L. Milne33, Taru A. Muranen53, Rachel A. Murphy54, Heli Nevanlinna53, Janet E. Olson19, Håkan Olsson55, Tjoung-Won Park-Simon12, Charles M. Perou56, Paolo Peterlongo, Dijana Plaseska-Karanfilska, Katri Pylkäs57, Gadi Rennert58, Emmanouil Saloustros, Dale P. Sandler4, Elinor J. Sawyer59, Marjanka K. Schmidt46, Rita K. Schmutzler36, Shibli R58, Ann Smeets52, Penny Soucy60, Melissa C. Southey32, Melissa C. Southey33, Melissa C. Southey34, Anthony J. Swerdlow29, Rulla M. Tamimi25, Rulla M. Tamimi61, Jack A. Taylor4, Lauren R. Teras62, Mary Beth Terry63, Ian Tomlinson64, Ian Tomlinson65, Melissa A. Troester56, Thérèse Truong35, Celine M. Vachon19, Camilla Wendt21, Robert Winqvist57, Alicja Wolk21, Alicja Wolk66, Xiaohong R. Yang4, Wei Zheng67, Argyrios Ziogas7, Jacques Simard60, Alison M. Dunning1, Pharoah Pd1, Doug Easton1 
University of Cambridge1, University of Otago2, The Cyprus Institute of Neurology and Genetics3, National Institutes of Health4, University of Toronto5, Lunenfeld-Tanenbaum Research Institute6, University of California, Irvine7, German Cancer Research Center8, Queen's University9, University of Erlangen-Nuremberg10, Russian Academy of Sciences11, Hannover Medical School12, University of Copenhagen13, Copenhagen University Hospital14, University of Hamburg15, QIMR Berghofer Medical Research Institute16, University of Sydney17, Erasmus University Medical Center18, Mayo Clinic19, University of Sheffield20, Karolinska Institutet21, Leiden University Medical Center22, International Agency for Research on Cancer23, Brigham and Women's Hospital24, Harvard University25, Manchester Academic Health Science Centre26, University of California, Los Angeles27, University of Edinburgh28, Institute of Cancer Research29, Curtin University30, University of California, San Diego31, University of Melbourne32, Monash University33, Cancer Council Victoria34, Université Paris-Saclay35, University of Cologne36, University of Southern California37, Erasmus University Rotterdam38, Bosch39, University of Tübingen40, University of Manchester41, Peter MacCallum Cancer Centre42, Pomeranian Medical University43, Laboratory of Molecular Biology44, Stanford University45, Netherlands Cancer Institute46, Saint Petersburg State University47, University of Eastern Finland48, Oslo University Hospital49, University of Oslo50, City of Hope National Medical Center51, Katholieke Universiteit Leuven52, University of Helsinki53, University of British Columbia54, Lund University55, University of North Carolina at Chapel Hill56, University of Oulu57, Technion – Israel Institute of Technology58, King's College London59, Laval University60, Cornell University61, American Cancer Society62, Columbia University63, Wellcome Trust Centre for Human Genetics64, University of Birmingham65, Uppsala University66, Vanderbilt University67
22 Jun 2021-bioRxiv
TL;DR: This is the first genome-wide analysis of rare CNVs in a large breast cancer case-control dataset and detected associations with exonic deletions in established breast cancer susceptibility genes and suggestive associations with non-codingCNVs in known and novel loci with large effects sizes.
Abstract: BackgroundCopy number variants (CNVs) are pervasive in the human genome but potential disease associations with rare CNVs have not been comprehensively assessed in large datasets. We analysed rare CNVs in genes and non-coding regions for 86,788 breast cancer cases and 76,122 controls of European ancestry with genome-wide array data. ResultsGene burden tests detected the strongest association for deletions in BRCA1 (P= 3.7E-18). Nine other genes were associated with a p-value < 0.01 including known susceptibility genes CHEK2 (P= 0.0008), ATM (P= 0.002) and BRCA2 (P= 0.008). Outside the known genes we detected associations with p-values < 0.001 for either overall or subtype-specific breast cancer at nine deletion regions and four duplication regions. Three of the deletion regions were in established common susceptibility loci. ConclusionsThis is the first genome-wide analysis of rare CNVs in a large breast cancer case-control dataset. We detected associations with exonic deletions in established breast cancer susceptibility genes. We also detected suggestive associations with non-coding CNVs in known and novel loci with large effects sizes. Larger sample sizes will be required to reach robust levels of statistical significance.

Posted ContentDOI
Thomas U. Ahearn1, Haoyu Zhang2, Haoyu Zhang1, Kyriaki Michailidou3  +186 moreInstitutions (85)
01 Feb 2021-bioRxiv
TL;DR: In this article, the authors used two-stage polytomous logistic regression models to evaluate variants in relation to multiple tumor features (ER, progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and grade) adjusting for each other.
Abstract: Genome-wide association studies (GWAS) have identified multiple common breast cancer susceptibility variants. Many of these variants have differential associations by estrogen receptor (ER), but how these variants relate with other tumor features and intrinsic molecular subtypes is unclear. Among 106,571 invasive breast cancer cases and 95,762 controls of European ancestry with data on 173 breast cancer variants identified in previous GWAS, we used novel two-stage polytomous logistic regression models to evaluate variants in relation to multiple tumor features (ER, progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and grade) adjusting for each other, and to intrinsic-like subtypes. Eighty-five of 173 variants were associated with at least one tumor feature (false discovery rate