Skip to main content
Erschienen in: Breast Cancer Research 1/2021

Open Access 01.12.2021 | Research article

Association of germline genetic variants with breast cancer-specific survival in patient subgroups defined by clinic-pathological variables related to tumor biology and type of systemic treatment

verfasst von: Anna Morra, Maria Escala-Garcia, Jonathan Beesley, Renske Keeman, Sander Canisius, Thomas U. Ahearn, Irene L. Andrulis, Hoda Anton-Culver, Volker Arndt, Paul L. Auer, Annelie Augustinsson, Laura E. Beane Freeman, Heiko Becher, Matthias W. Beckmann, Sabine Behrens, Stig E. Bojesen, Manjeet K. Bolla, Hermann Brenner, Thomas Brüning, Saundra S. Buys, Bette Caan, Daniele Campa, Federico Canzian, Jose E. Castelao, Jenny Chang-Claude, Stephen J. Chanock, Ting-Yuan David Cheng, Christine L. Clarke, Sarah V. Colonna, Fergus J. Couch, Angela Cox, Simon S. Cross, Kamila Czene, Mary B. Daly, Joe Dennis, Thilo Dörk, Laure Dossus, Alison M. Dunning, Miriam Dwek, Diana M. Eccles, Arif B. Ekici, A. Heather Eliassen, Mikael Eriksson, D. Gareth Evans, Peter A. Fasching, Henrik Flyger, Lin Fritschi, Manuela Gago-Dominguez, José A. García-Sáenz, Graham G. Giles, Mervi Grip, Pascal Guénel, Melanie Gündert, Eric Hahnen, Christopher A. Haiman, Niclas Håkansson, Per Hall, Ute Hamann, Steven N. Hart, Jaana M. Hartikainen, Arndt Hartmann, Wei He, Maartje J. Hooning, Reiner Hoppe, John L. Hopper, Anthony Howell, David J. Hunter, Agnes Jager, Anna Jakubowska, Wolfgang Janni, Esther M. John, Audrey Y. Jung, Rudolf Kaaks, Machteld Keupers, Cari M. Kitahara, Stella Koutros, Peter Kraft, Vessela N. Kristensen, Allison W. Kurian, James V. Lacey, Diether Lambrechts, Loic Le Marchand, Annika Lindblom, Martha Linet, Robert N. Luben, Jan Lubiński, Michael Lush, Arto Mannermaa, Mehdi Manoochehri, Sara Margolin, John W. M. Martens, Maria Elena Martinez, Dimitrios Mavroudis, Kyriaki Michailidou, Roger L. Milne, Anna Marie Mulligan, Taru A. Muranen, Heli Nevanlinna, William G. Newman, Sune F. Nielsen, Børge G. Nordestgaard, Andrew F. Olshan, Håkan Olsson, Nick Orr, Tjoung-Won Park-Simon, Alpa V. Patel, Bernard Peissel, Paolo Peterlongo, Dijana Plaseska-Karanfilska, Karolina Prajzendanc, Ross Prentice, Nadege Presneau, Brigitte Rack, Gad Rennert, Hedy S. Rennert, Valerie Rhenius, Atocha Romero, Rebecca Roylance, Matthias Ruebner, Emmanouil Saloustros, Elinor J. Sawyer, Rita K. Schmutzler, Andreas Schneeweiss, Christopher Scott, Mitul Shah, Snezhana Smichkoska, Melissa C. Southey, Jennifer Stone, Harald Surowy, Anthony J. Swerdlow, Rulla M. Tamimi, William J. Tapper, Lauren R. Teras, Mary Beth Terry, Rob A. E. M. Tollenaar, Ian Tomlinson, Melissa A. Troester, Thérèse Truong, Celine M. Vachon, Qin Wang, Amber N. Hurson, Robert Winqvist, Alicja Wolk, Argyrios Ziogas, Hiltrud Brauch, Montserrat García-Closas, Paul D. P. Pharoah, Douglas F. Easton, Georgia Chenevix-Trench, Marjanka K. Schmidt, NBCS Collaborators, ABCTB Investigators, kConFab Investigators

Erschienen in: Breast Cancer Research | Ausgabe 1/2021

Abstract

Background

Given the high heterogeneity among breast tumors, associations between common germline genetic variants and survival that may exist within specific subgroups could go undetected in an unstratified set of breast cancer patients.

Methods

We performed genome-wide association analyses within 15 subgroups of breast cancer patients based on prognostic factors, including hormone receptors, tumor grade, age, and type of systemic treatment. Analyses were based on 91,686 female patients of European ancestry from the Breast Cancer Association Consortium, including 7531 breast cancer-specific deaths over a median follow-up of 8.1 years. Cox regression was used to assess associations of common germline variants with 15-year and 5-year breast cancer-specific survival. We assessed the probability of these associations being true positives via the Bayesian false discovery probability (BFDP < 0.15).

Results

Evidence of associations with breast cancer-specific survival was observed in three patient subgroups, with variant rs5934618 in patients with grade 3 tumors (15-year-hazard ratio (HR) [95% confidence interval (CI)] 1.32 [1.20, 1.45], P = 1.4E−08, BFDP = 0.01, per G allele); variant rs4679741 in patients with ER-positive tumors treated with endocrine therapy (15-year-HR [95% CI] 1.18 [1.11, 1.26], P = 1.6E−07, BFDP = 0.09, per G allele); variants rs1106333 (15-year-HR [95% CI] 1.68 [1.39,2.03], P = 5.6E−08, BFDP = 0.12, per A allele) and rs78754389 (5-year-HR [95% CI] 1.79 [1.46,2.20], P = 1.7E−08, BFDP = 0.07, per A allele), in patients with ER-negative tumors treated with chemotherapy.

Conclusions

We found evidence of four loci associated with breast cancer-specific survival within three patient subgroups. There was limited evidence for the existence of associations in other patient subgroups. However, the power for many subgroups is limited due to the low number of events. Even so, our results suggest that the impact of common germline genetic variants on breast cancer-specific survival might be limited.
Begleitmaterial
Additional file 2: Supplementary Methods, Supplementary Table S5, Supplementary Table S6, Supplementary Table S7, Supplementary Figure S1, Supplementary Figure S2, Supplementary Figure S3, Supplementary Figure S4, Supplementary Figure S5, Supplementary Figure S6, Supplementary Figure S7, Supplementary Figure S8, Supplementary Figure S9, Supplementary Figure S10, Supplementary Figure S11, Supplementary Figure S12. The Supplementary methods section includes details about: multiple imputation of missing data; power calculations. Supplementary Table S5. Shows an overview of number of breast cancer patients, breast cancer deaths and follow-up information by subgroup and endpoint. Supplementary Table S6. Shows a list of imputed variables with corresponding percentage of missing values, imputation method and processing. Supplementary Table S7. Shows an overview of the GWAS significant associations (P < 5 E-08) and noteworthy (BFDP< 0.15) associations from the unadjusted 15-year and 5-year breast cancer-specific survival analyses by subgroup. Supplementary Figure S1. Shows the Q-Q plots of the meta-analysis results of all variants for 15-year breast cancer-specific survival. Supplementary Figure S2. Shows the Q-Q plots of the meta-analysis results of all variants for 5-year breast cancer-specific survival. Supplementary Figure S3. Shows the Q-Q plots of the meta-analysis results for 15-year breast cancer-specific survival and the corresponding genome inflation factors by minor allele frequency. Supplementary Figure S4. Shows the Q-Q plots of the meta-analysis results for 5-year breast cancer-specific survival and the corresponding genome inflation factors by minor allele frequency. Supplementary Figure S5. Shows the regional plots of genome-wide significant (P < 5E-08) independent associated variants from the 15-year and 5-year genome-wide breast cancer-specific survival analyses. Supplementary Figure S6. Shows the regional plots of noteworthy (BFDP< 0.15), non-genome-wide significant (P > 5E-08) variants from the 15-year and 5-year genome-wide breast cancer-specific survival analyses. Supplementary Figure S7 shows the unadjusted association of variant rs5934618 with 15-year breast cancer-specific survival by tumor grade and in all breast cancer patients. Supplementary Figure S8. Shows the functional annotation and position of variants rs5934618, rs4830644, rs3810742, rs4830642, and rs72611496 relative to TBL1X. Supplementary Figure S9. Shows Kaplan-Meier distant metastasis-free survival plots (based on KMPlotter data) for high versus low expression level of gene TBL1X by tumor grade. Supplementary Figure S10. Shows the association of genetic variants with TBL1X expression, based on GTEx v8 data on samples of normal breast tissue from 396 individuals (male and female). Supplementary Figure S11. Shows a Kaplan-Meier distant metastasis-free survival plot for high versus low expression level of gene GRIP2, restricted to patients with an ER- tumor who received chemotherapy (based on KMPlotter data). Supplementary Figure S12. Shows a Kaplan-Meier distant metastasis-free survival plot for high versus low expression level of gene ARAP2, restricted to patients with an ER- tumor who received chemotherapy (based on KMPlotter data).
Additional file 3: Supplementary Table S8, Supplementary Table S9, Supplementary Table S10, Supplementary Table S11, and Supplementary Table S12. Supplementary Table S8. Shows the BFDPs under two more restrictive prior probabilities of true association (10-5 and 10-6) for the results presented in Table 1. Supplementary Table S9. Shows the BFDPs under two more restrictive prior probabilities of true association (10-5 and 10-6) for the results presented in Table 2. Supplementary Table S10. Shows power calculation by subgroup, at the two-sided 5E-08 level for varying genotype hazard ratio (GHR) and minor allele frequency (MAF), based on number of cases and event rate from the 15-year breast cancer-specific analyses. Supplementary Table S11. Shows power calculation by subgroup, at the two-sided 5E-08 level for varying genotype hazard ratio (GHR) and minor allele frequency (MAF), based on number of cases and event rate from the 5-year breast cancer-specific analyses. Supplementary Table S12. Shows the subgroup-specific associations detected by previous studies and corresponding estimates from the current study.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13058-021-01450-7.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BCAC
Breast Cancer Association Consortium
BFDP
Bayesian false discovery probability
CI
Confidence interval
CMF
Cyclophosphamide Methotrexate Fluorouracil
EBCTCG
Early Breast Cancer Trialists’ Collaborative Group
ER
Estrogen receptor
eQTL
Expression quantitative trait locus
eSNP
Expression single-nucleotide polymorphism
FUMA
Functional mapping and annotation
GWAS
Genome-wide association study
HER2
Human epidermal growth factor receptor 2
HR
Hazard ratio or Hormone receptor
HRC
Haplotype Reference Consortium
MAF
Minor allele frequency
MICE
Multiple imputation by chained equations
PR
Progesterone receptor
SNV
Single-nucleotide variant

Introduction

Inherited common genetic variation is likely to influence survival in breast cancer patients [1]. Results from pre-clinical experiments have shown different metastatic behaviors in mice with different genetic backgrounds [27]. In addition, familial studies of breast cancer patients have shown that women with a first-degree relative with a poor prognosis breast cancer have a worse prognosis compared to women with a first-degree relative with a good prognosis cancer [8]. Moreover, genome-wide and candidate gene association studies have discovered common genetic variants associated with specific subtypes of breast cancer based on the expression of the estrogen receptor (ER) [911], progesterone receptor (PR), and the amplification of the human epidermal growth factor receptor 2 (HER2) [12, 13], which are known breast cancer prognostic factors [14, 15]. Finally, a number of studies have suggested that specific common germline genetic variants affect breast cancer prognosis both overall and within subgroups of patients [1624].
Despite the supporting evidence, it remains challenging to identify common germline variants associated with breast cancer-specific survival. This may partially be explained by the good prognosis of breast cancer patients, which leads to underpowered analyses. Even large studies based on worldwide consortia cannot reach the number of breast cancer deaths necessary to detect small to moderate associations at a genome-wide significant level [19, 20, 25]. However, breast cancer is a heterogeneous disease, and it is possible that stronger associations between common germline variants and breast cancer-specific survival are present in certain patient subgroups, but cannot be detected in breast cancer overall. Previous studies provide modest evidence supporting this hypothesis [16, 19, 20].
The aim of our study was to evaluate the evidence for associations of inherited common genetic variants with breast cancer-specific survival within more homogeneous subgroups of breast cancer patients, defined by prognostic factors representative of tumor biology and/or by the type of systemic treatment. To this end, we performed genome-wide association analyses within clinically relevant, defined subgroups of patients based on hormone receptors, tumor grade, age at diagnosis, and type of systemic treatment [26, 27]. We also explored the subgroup-specific associations identified by previous studies [16, 19, 23, 28, 29], to confirm or refute those results.

Materials and methods

Study sample

We selected female breast cancer patients of European ancestry from studies participating in the Breast Cancer Association Consortium (BCAC). We included patients with available information about vital status and number of years from diagnosis to last follow-up who were diagnosed with a primary invasive breast cancer of any stage and were at least 18 years old at diagnosis. The final study sample consisted of 91,686 breast cancer patients from 70 BCAC studies. A description of the included studies is given in Additional file 1: Supplementary Table S1.
Information about histopathology, survival, and treatment was collected by individual studies and pooled and harmonized at the Netherlands Cancer Institute before incorporation into the BCAC database at the University of Cambridge (version 12, July 2019). All studies were approved by the relevant ethics committees and informed consent was obtained from all patients.

Patient subgroups

The subgroups of interest were defined based on age at diagnosis, estrogen receptor (ER) status, progesterone receptor (PR) status, human epidermal growth factor receptor 2 (HER2) status, tumor grade, and the use and type of systemic treatment, as available in the BCAC database. For age at diagnosis and tumor grade, we focused on subgroups characterized by worse prognosis. We thus defined 15 subgroups: (a) patients younger than age 40 years at diagnosis; (b) patients with grade 3 tumors; (c) patients with ER-positive (ER+) tumors, who received endocrine therapy (any kind); (d) patients diagnosed with ER-negative (ER−) tumors, who received chemotherapy (any kind); (e) patients with tumors that were hormone receptor (HR) positive (ER+ or PR+) and HER2-negative (HER2−); (f) patients with HR-positive (HR+), HER2− tumors, who received chemotherapy (any kind); (g) patients with HR+, HER2− tumors, who did not receive chemotherapy; (h) patients with HR+, HER2-positive (HER2+) tumors; (i) patients with HR-negative (HR−), HER2+ tumors, (j) patients with HR−, HER2− tumors; (k) patients who received Tamoxifen; (l) patients who received an aromatase inhibitor; (m) patients who received a Cyclophosphamide Methotrexate Fluorouracil (CMF)-like chemotherapy regimen; (n) patients who received taxanes; (o) patients who received anthracyclines.
The rationale and references to the literature supporting the choice of each subgroup for inclusion in a genome-wide association study on survival are given in Additional file 1: Supplementary Table S2. We did not include the subgroup of HER2+ tumors treated with Trastuzumab because of a relatively small number of patients and low event rate, leading to analyses that are more underpowered than those presented.
Patients with metastatic breast tumors at diagnosis (1.1 % of all included patients) were excluded from the subgroup analyses whose definition was based on the use and type of systemic therapy as generally they are treated with palliative intent [15, 30, 31].
In addition to the subgroup analyses, we also performed a genome-wide analysis of 15-year breast cancer-specific survival in all breast cancer patients. We performed this analysis to evaluate whether associations between common germline variants and breast cancer-specific survival in subgroups could be detected in the full dataset of patients. Genome-wide analyses for survival (unstratified by subtype) were previously performed [20] based on 12 GWAS datasets, but these included fewer patients from iCOGS and OncoArray (n = 84,757), with shorter follow-up, than were available in the current dataset. We focused our analyses on the iCOGS and OncoArray datasets, because the remaining 10 GWAS datasets used in the previous study did not include information about tumor characteristics, beyond ER status, or treatment, which were crucial for the subgroup analyses.
Due to the presence of missing values in the variables used to define the subgroups, not all patients could be classified by each subgroup. The number of patients included in each subgroup, together with the number of breast cancer-specific deaths, patient/tumor characteristics, treatment, and follow-up information, are shown in Additional file 1: Supplementary Tables S3-S4, and Additional file 2: Supplementary Table S5.

Imputation of missing values in clinical and pathological variables

For secondary adjusted analyses, we imputed missing values in the clinical and pathological variables using the Multiple imputation by Chained Equations (MICE) R package (v. 3.2.0), as described in Additional file 2: Supplementary Methods. A list of imputed variables and corresponding percentages of missing values and imputation methods is provided in Additional file 2: Supplementary Table S6.

Genotyping and imputation of genetic variants, ancestry analysis, and quality controls

Methods related to genotyping and genotype imputation have been described previously [17, 18, 20]. In brief, patients were genotyped with two different arrays: iCOGS and OncoArray [17, 32]. Only samples that were inferred to have European ancestry, based on genotype data, were included in the analyses. Non-genotyped variants were initially imputed based on the 1000 Genomes Project Phase 3 (October 2014) release as reference panel. More recently, non-genotyped single-nucleotide variants (SNVs) were re-imputed using a reference panel from the Haplotype Reference Consortium (HRC) [33] in order to improve imputation quality, especially for rarer variants. Analyses were performed on genotyped variants or imputed variants with a minor allele frequency (MAF) > 0.01. Imputed variants were included in the analyses if they had imputation r2 > 0.7. Approximately 10 million variants were analyzed.

Statistical analyses

The outcome in the analyses was breast cancer-specific survival (time to death due to breast cancer). Hazard ratios (HR) and 95% confidence intervals (CI) were estimated using delayed entry Cox regression models, where the time at risk was considered as starting from the time of study entry if the study entry was after diagnosis (22.9% within 1 year after diagnosis, and 27.3% more than 1 year after diagnosis) and from diagnosis if the time of study entry was missing (24.5%), at diagnosis (16.9%) or before diagnosis (8.4%). The time-to-event was right censored at the time of last follow-up, or at 15 years after diagnosis, whichever came first. Patients who died of unknown cause or causes other than breast cancer were censored at the time of death if death occurred before 15 years from diagnosis or at 15 years otherwise.
With reference to the results of Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) [34], we additionally performed analyses within the subgroups whose definition was based on the use and type of systemic therapy, where we restricted the maximum follow-up time to 5 years after diagnosis (Additional file 2: Supplementary Table S5). The goal of those analyses was to investigate the potential short-term effects of germline variants on patients who received specific types of systemic treatment, since the effect might not be constant over time and treatment plans tend to focus on the first 5 years after diagnosis [15, 31].
Cox regression analysis was performed within each subgroup of interest, separately, and was stratified by country. All the analyses were performed separately by genotyping platform (iCOGS vs OncoArray), and the results were combined via a fixed-effects meta-analysis. The standard errors of the HR estimates were re-computed based on the likelihood ratio test statistic, as done previously [20] (Figs. 1 and 2). For variants that satisfied the inclusion criteria (MAF>0.01 and r2 > 0.7) on only one genotyping platform, we included the result for that specific platform (Tables 1 and 2). However, for variants with an association P < 5E−08, we also computed HR and 95% confidence interval in the other genotyping platform to verify that the direction of the association was the same (Additional file 2: Supplementary Table S7).
Table 1
GWAS significant (P<5×10-8) and noteworthy (BFDP<0.15) results by subgroup, and corresponding results from adjusted analyses
Subgroup
Variant
Chr
Position
Allelesa
AAF
Unadjusted analyses
Adjusted analyses
HR [95% CI]
P value
BFDP
HR [95% CI]
P value
BFDP
Grade 3 tumors
rs5934618b
X
9437463
A/G
0.08
1.39 [1.24,1.56]
1.7E-08
0.02
1.36 [1.21,1.53]e
3.0E-07
0.17
rs4830644b
X
9434808
A/G
0.08
1.39 [1.24,1.56]
2.0E-08
0.02
1.35 [1.20,1.52]e
4.8E-07
0.23
rs3810742b, c
X
9432603
T/C
0.08
1.38 [1.24,1.55]
2.0E-08
0.02
1.35 [1.20,1.52]e
4.2E-07
0.20
rs4830642b
X
9431786
T/C
0.08
1.38 [1.24,1.55]
2.9E-08
0.02
1.35 [1.20,1.52]e
5.8E-07
0.26
rs72611496b
X
9434264
G/A
0.08
1.38 [1.24,1.55]
4.3E-08
0.03
1.34 [1.19,1.51]e
1.2E-06
0.40
rs66871326
2
209048052
AAGGAG/A
0.76
0.85 [0.80,0.90]
2.1E-07
0.11
0.86 [0.81,0.92]e
1.8E-06
0.49
ER+ or PR+, and HER2-
rs8030394
15
71637241
C/T
0.99
2.47 [1.81,3.37]
1.1E-08
0.42
2.38 [1.74,3.27]f
7.6E-08
0.72
rs112641969
15
71715016
A/G
0.02
0.46 [0.35,0.61]
4.6E-08
0.46
0.48 [0.36,0.64]f
3.7E-07
0.78
rs16955466
15
71637757
C/T
0.01
0.40 [0.29,0.55]
1.5E-08
0.49
0.42 [0.31,0.58]f
1.8E-07
0.82
rs7165279
15
71636591
T/C
0.99
2.41 [1.77,3.28]
2.7E-08
0.54
2.33 [1.70,3.19]f
1.4E-07
0.78
rs111962948
15
71656213
G/T
0.01
0.41 [0.29,0.56]
3.0E-08
0.61
0.43 [0.31,0.60]f
5.6E-07
0.91
rs112813972
15
71577932
T/C
0.02
0.40 [0.28,0.55]
4.0E-08
0.70
0.42 [0.30,0.59]f
3.7E-07
0.90
ER+ or PR+, and HER2- treated with CT
rs62192052
2
230372348
C/T
0.02
0.15 [0.08, 0.28]
2.6E-09
0.99
0.15 [0.08, 0.29]g
5.5E-09
0.99
rs74423556c
2
230325234
C/G
0.02
0.16 [0.08,0.30]
2.1E-08
0.99
0.16 [0.08,0.31]g
3.8E-08
1.00
rs145983608
2
230296944
A/G
0.02
0.15 [0.08,0.30]
3.8E-08
1.00
0.15 [0.08,0.31]g
1.1E-07
1.00
ER+ or PR+, and HER2- not treated with CT
rs56248395b
11
20084391
C/T
0.13
2.33 [1.72,3.15]
4.8E-08
0.59
2.23 [1.66,2.99]g
1.2E-07
0.69
ER+ treated with ET
rs4679741
3
155003603
T/G
0.49
1.18 [1.11,1.26]
1.6E-07
0.09
1.20 [1.13,1.28] h
1.1E-08
0.01
ER- treated with CT
rs78754389d
4
35962454
G/A
0.07
1.79 [1.46,2.20]
1.7E-08
0.07
1.67 [1.39,2.00] i
4.1E-08
0.09
rs1106333
3
14562127
C/A
0.06
1.68 [1.39,2.03]
5.6E-08
0.12
1.70 [1.41,2.05] i
4.4E-08
0.11
rs117685664d
8
26989084
C/T
0.03
0.26 [0.16,0.42]
4.6E-08
0.97
0.50 [0.35,0.70]i
6.4E-05
0.99
Tamoxifen
rs72775397d
5
94266932
C/T
0.28
1.36 [1.21,1.53]
1.8E-07
0.11
1.11 [1.03,1.19]j
6.6E-03
1.00
Anthracylines
rs34072391
7
30243729
C/CA
0.52
1.27 [1.17,1.39]
6.2E-08
0.04
1.26 [1.15,1.37]k
3.4E-07
0.16
Abbreviations: Chr chromosome, ER estrogen receptor, PR progesterone receptor, HER2 human epidermal growth factor receptor 2, ET Endocrine therapy, CT chemotherapy, AAF alternative allele frequency, HR hazard ratio, CI confidence interval, BFDP Bayesian False Discovery Probability
Note: BFDP is computed assuming the prior probability of true association equal to 10-4for all variants, which implies a number of expected true associations in the order of 102. Results with BFDP<0.15 in the adjusted analyses are bolded. aReference/Alternative alleles, bAnalyses only include OncoArray data since the variants had imputation r2 <0.7 on iCOGS. More detailed analyses are reported in Table 2 and Supplementary Table 7, cVariant genotyped on OncoArray, dFrom the 5-years breast cancer specific survival analysis, eAdjusted for age at diagnosis, lymph node status, tumor size, distant metastases status, ER status, HER2 status, (neo)adjuvant CT, fAdjusted for age at diagnosis, lymph node status, tumor size, tumor grade, distant metastases status, and (neo)adjuvant CT, gAdjusted for age at diagnosis, lymph node status, tumor size, and tumor grade, hAdjusted for age at diagnosis, lymph node status, tumor size, tumor grade, HER2 status, (neo)adjuvant CT, iAdjusted for age at diagnosis, lymph node status, tumor size, tumor grade, and HER2 status, jAdjusted for age at diagnosis, lymph node status, tumor size, tumor grade, HER2 status ,and (neo)adjuvant CT, kAdjusted for age at diagnosis, lymph node status, tumor size, tumor grade, ER status, and HER2 status
Table 2
Meta-analysis results for variants analysed on OncoArray only in the unadjusted analyses in Table 1
Subgroup
Variant
Chr
Position
Allelesa
AAF
Unadjusted meta-analysis
Adjusted meta-analysis
HR [95% CI]
P value
BFDP
HR [95% CI]
P value
BFDP
Grade 3 tumors
rs5934618
X
9437463
A/G
0.08
1.32 [1.20,1.45]
1.4E-08
0.01
1.31 [1.18, 1.44]b
7.9E-08
0.05
rs4830644
9434808
A/G
0.08
1.32 [1.20,1.45]
2.1E-08
0.01
1.30 [1.18, 1.43]b
1.7E-07
0.10
rs3810742
9432603
T/C
0.08
1.31 [1.19,1.44]
2.7E-08
0.02
1.29 [1.17, 1.42]b
1.8E-07
0.10
rs4830642
9431786
T/C
0.08
1.31 [1.19,1.44]
2.8E-08
0.02
1.29 [1.17, 1.42]b
2.2E-07
0.12
rs72611496
9434264
G/A
0.08
1.32 [1.20,1.45]
2.3E-08
0.02
1.30 [1.18, 1.43]b
2.5E-07
0.13
ER+ or PR+, and HER2- not treated with CT
rs56248395
11
20084391
C/T
0.13
1.53 [1.25,1.89]
5.2E-05
0.97
1.52 [1.24,1.87]c
6.4E-05
0.98
Abbreviations: Chr chromosome, AAF alternative allele frequency, HR hazard ratio, CI confidence interval, BFDP Bayesian False Discovery Probability
Note: BFDP is computed assuming the prior probability of true association equal to 10-4 for all variants, which implies a number of expected true associations in the order of 102. Results with BFDP<0.15 in the adjusted analyses are bolded. aReference/Alternative alleles, bAdjusted for age at diagnosis, lymph node status, tumor size, distant metastases status, ER status, HER2 status, (neo)adjuvant CT, cAdjusted for age at diagnosis, lymph node status, tumor size, and tumor grade
Inflation of the likelihood ratio test statistics was estimated, within each subgroup, by dividing the median of the observed test statistics values by the median of a \( {\chi}_1^2 \) distribution (Additional file 2: Supplementary Figures S1 and S2). To assess the noteworthiness of the observed associations, we made use of the Bayesian false discovery probability (BFDP) measure [35]. To compute BFDPs, we set the prior probability of true association to 10−4 [36, 37], as done previously [20], and chose the prior distribution of the log hazard ratio of interest (effect size of a variant) to be a Normal distribution with mean 0 and standard error equal to 0.2 [36]. We describe associations with BFDP < 0.15 as “noteworthy” [20]. For each noteworthy result at a prior of 10−4, we also provided BFDPs under two, more restrictive, prior probabilities of true association (10−5 and 10−6; Additional file 3: Supplementary Tables S8-S9) [36, 37]. In addition, we estimated the power to detect genetic variant associated with 15-year and 5-year breast cancer-specific survival by subgroup (Additional file 3: Supplementary Table S10 and S11) as described in Additional file 2: Supplementary Methods.
For each genome-wide significant (P < 5E−08) [38] and/or noteworthy (BFDP < 0.15) association observed in the primary unadjusted subgroup analyses, we performed secondary analyses adjusted for age at diagnosis, tumor characteristics, and type of systemic treatment not used in the definition of the specific subgroup in which the association was detected (Tables 1 and 2). Secondary adjusted analyses were performed to account for residual heterogeneity; we used imputed covariates in order to keep the same sample size.
For each genome-wide significant or noteworthy association in the primary unadjusted analyses, we looked at the functional annotation of the surrounding genomic area, using the Functional Mapping and Annotation of Genome-Wide Association Studies (FUMA GWAS) tool [39] (Additional file 2: Supplementary Figures S5 and S6). We also tested whether the expression of the nearest genes correlated with distant metastasis-free survival in breast cancer patients using KMplotter [40, 41].
PancanQTL [42] was used to identify cis-expression quantitative trait locus (eQTLs), trans-eQTLs, and survival eQTLs in breast cancer to see whether the genome-wide significant or noteworthy genetic variants from the primary analyses could be linked with the expression levels of genes affecting survival. In addition, for all the genome-wide significant and/or noteworthy associations detected in the primary analyses, we searched the GWAS catalog [43] to see whether there was already evidence of those being associated with breast cancer or other traits.

Results

After a median follow-up of 8.1 years, there were a total of 7531 breast cancer deaths among 91686 breast cancer patients (Additional file 1: Supplementary Tables S3-S4 and Additional file 2: Supplementary Table S5).
In the 15-year breast cancer-specific survival analyses, power for detecting genome-wide significant associations was < 0.45 for effect sizes (HRs) < 1.20 in all subgroups investigated and for all minor allele frequencies. The power was highest in the subgroups of grade 3 tumors, ER+ tumors treated with endocrine therapy, HR+ and HER2− tumors, and patients who received tamoxifen (Additional file 3: Supplementary Table S10). In the 5-year breast cancer-specific survival analyses, power was highest in the subgroups of patients with an ER− tumor who received chemotherapy, patients who received tamoxifen, and patients who received anthracyclines (Additional file 3: Supplementary Table S11).
Genome-wide significant and/or noteworthy associations with 15-year or 5-year breast cancer-specific survival were observed in the unadjusted analyses based on all patients (Additional file 2: Supplementary Table S7) and analyses of eight out of the 15 subgroups investigated (Tables 1 and 2; Figs. 1 and 2). The genomic inflation factor of the unadjusted genome-wide analyses varied from 0.981 to 1.028 (Additional file 2: Supplementary Figures S1- S4); it is therefore unlikely that the association results were affected by cryptic population substructure.
Two genome-wide significant associations were observed in the unstratified analysis based on patients genotyped using OncoArray (Additional file 2: Supplementary Table S7), namely variants rs57714252 (P = 4.7E−08) and rs4129285 (P = 4.9E−08), both situated in an intergenic region of chromosome 4 (Additional file 2: Supplementary Figure S5). These results were only based on the OncoArray data, since on iCOGS the variants did not satisfy the inclusion criteria for genotypes (iCOGS imputation r2 = 0.62). The corresponding estimates in the iCOGS data were in the opposite direction compared to the OncoArray estimates, and the results from the meta-analysis were not genome-wide significant and showed a large BFDP (Additional file 2: Supplementary Table S7).
Genome-wide significant associations were observed in the analysis restricted to patients diagnosed with a grade 3 tumor, with five correlated variants (Tables 1 and 2) located on chromosome X (Additional file 2: Supplementary Figure S5) in intron 1 of TBL1X. For the most significant variant, rs5934618, the alternative G allele was associated with increased risk of breast cancer death in unadjusted analyses (meta-analysis hazard ratio (HR) [95% confidence interval (CI)] 1.32 [1.20, 1.45], P = 1.4E−08, BFDP = 0.01; Table 2). The meta-analysis result remained substantially unchanged after adjusting for age at diagnosis, additional tumor characteristics, and treatment with (neo)adjuvant chemotherapy (HR [95% CI] 1.31 [1.18, 1.44], P = 7.9E−08, BFDP = 0.05; Table 2). The variant was not associated with the outcome in lower-grade tumors or in all patients combined (heterogeneity by grade P = 1.5E−03; Additional file 2: Supplementary Figure S7). All the five variants overlap chromatin features H3K4me3 and H3K27ac (associated with active transcription start sites) in multiple mammary cell types from normal breast tissue (Additional file 2: Supplementary Figure S8). Furthermore, there was evidence of TBL1X expression being associated with distant metastasis-free survival (HR [95%CI] for high vs low expression 1.71 [1.20,2.44], P = 2.7E−03) specifically in grade 3 patients, but not in patients with lower-grade disease (Additional file 2: Supplementary Figure S9). However, there was no evidence of association with TBL1X expression in normal breast tissue with any of the variants identified in our genome-wide analyses (Additional file 2: Supplementary Figure S10).
In the same subgroup of grade 3 tumors, we observed a noteworthy, non-genome-wide significant association with variant rs66871326, located on chromosome 2 in an intron of C2orf80. For variant rs66871326, the alternative A allele was associated with decreased risk of breast cancer death (HR [95% CI] 0.85 [0.80,0.90], P = 2.1E−07, BFDP = 0.11). The corresponding BFDP increased to 0.49 after adjusting for age at diagnosis, additional tumor characteristics, and treatment with (neo)adjuvant chemotherapy (Table 1).
We identified six variants on chromosome 15 with genome-wide significant associations within the subgroup of patients diagnosed with an ER+ or PR+, HER2− tumor (Table 1). We identified two independent variants, namely rs8030394 and rs112813972, both situated in an intronic region of THSD4 (Additional file 2: Supplementary Figure S5). For the most significant variant, rs8030394, the T allele was associated with increased risk of breast cancer death (HR [95% CI]: 2.47 [1.81, 3.37], P = 1.1E−08, BFDP = 0.42). For the second variant, rs112813972, the C allele was associated with decreased risk of death (HR [95% CI] 0.40 [0.28,0.55], P = 4.0E−08, BFDP = 0.70). These associations were not genome-wide significant after adjusting for age at diagnosis, additional tumor characteristics, and treatment with (neo)adjuvant chemotherapy, and the corresponding BFDPs increased to 0.72 and 0.90, respectively (Table 1).
We observed genome-wide significant associations from the 15-year breast cancer-specific analyses in the subgroups of patients with an ER+ or PR+ and HER2− tumor who did and did not receive chemotherapy. Three correlated variants on chromosome 2 were identified within the subgroup of patients who received chemotherapy. The most significant variant, rs62192052, is located in an intronic region of DNER (Additional file 2: Supplementary Figure S5) and was associated with decreased risk of death (HR [95% CI] 0.15 [0.08, 0.28], P = 2.6E−09, per T allele; Table 1). Although the result remained genome-wide significant after adjusting for age at diagnosis and additional tumor characteristics, the BFDP from both the unadjusted and adjusted analysis was ≥ 0.99 (Table 1; Supplementary Table S8), indicating that this association is almost certainly a false positive. Variant rs56248395, located on chromosome 11 in an intron of NAV2 (Additional file 2: Supplementary Figure S5), was associated with breast cancer death in the subgroup of patients with an ER+ or PR+ and HER2− tumor who did not receive chemotherapy (HR [95% CI] 2.33 [1.72,3.15], P = 4.8E−08, per T allele; Table 1). This result had BFDP ≥ 0.59 and was only based on the OncoArray data, since on iCOGS the variants did not satisfy the inclusion criteria for genotypes (iCOGS imputation r2 = 0.66; Additional file 2: Supplementary Table S7). The corresponding estimates in the iCOGS data (HR [95% CI] 1.07 [0.80,1.41], P = 6.6E−01; Additional file 2: Supplementary Table S7) and from the meta-analysis (HR [95% CI] 1.53 [1.25,1.89], P = 5.2E−05; Table 2) were not genome-wide significant and not noteworthy (meta-analysis BFDP≥0.97; Table 2; Supplementary Table S9).
We observed three additional single SNP noteworthy associations from the 15-year breast cancer-specific survival analyses. The intergenic variant rs4679741 on chromosome 3 was associated with breast cancer death in the subgroup of patients with an ER+ tumor treated with endocrine therapy (HR [95% CI] 1.18 [1.11, 1.26], P = 1.6E−07, BFDP = 0.09, per G allele). This result became genome-wide significant after adjusting for age at diagnosis, tumor characteristics, and treatment with chemotherapy (HR [95% CI] 1.20 [1.13, 1.28], P = 1.1E−08, BFDP = 0.01). The BFDP of this association remained < 0.15 when considering 10−5 as prior probability of true association (Additional file 3: Supplementary Table S8). PanCanQTL did not show any cis-eQTLs, trans-eQTLs nor survival eQTLs for this variant. Variant rs1106333 on chromosome 3, whose nearest gene is GRIP2, was associated with risk of dying of breast cancer in the subgroup of patients with an ER− tumor who received chemotherapy (HR [95% CI] 1.68 [1.39,2.03], P = 5.6E−08, BFDP = 0.12 per A allele). This result also became genome-wide significant after adjusting for additional prognostic factors (HR [95% CI] 1.70 [1.41,2.05], P = 4.4E−08, BFDP = 0.11). PanCanGTL revealed the presence of a cis-eQTL linking variant rs1106333 with GRIP2 expression in prostate adenocarcinoma but not in breast cancer. There was no evidence of association of GRIP2 expression levels with distant metastasis-free survival within ER− breast cancer patients treated with chemotherapy based on KMPlotter data (Additional file 2: Supplementary Figure S11). The last association of interest was observed in the subgroup of patients who received anthracyclines with intergenic variant rs34072391 on chromosome 7, but was not noteworthy after adjustment for additional prognostic factors (Table 1).
In the 5-year survival analyses focused on the treatment subgroups, we observed two genome-wide significant associations within the subgroup of patients diagnosed with an ER− tumor who received chemotherapy. The most significant variant was rs78754389, located on chromosome 4 in an intronic region of gene ARAP2 (Additional file 2: Supplementary Figure S5; HR [95% CI] 1.79 [1.46,2.20], P = 1.7E−08, BFDP = 0.07 per A allele). This result remained both genome-wide significant and noteworthy after adjusting for age at diagnosis and additional tumor characteristics (HR [95% CI] 1.67 [1.39,2.00], P = 4.1E−08, BFDP = 0.09; Table 1). However, PanCanQTL did not show any cis-eQTLs, trans-eQTLs nor survival eQTLs for rs78754389 and there was no evidence of association of ARAP2 expression levels with distant metastasis-free survival within ER− breast cancer patients treated with chemotherapy based on KMPlotter data (Additional file 2: Supplementary Figure S12). The second genome-wide significant variant was rs117685664, located on chromosome 8 (HR [95% CI] 0.26 [0.16,0.42], P = 4.6E−08, per T allele). This association was not genome-wide significant after accounting for the age at diagnosis and additional tumor characteristics (Table 1), and the corresponding BFDPs from unadjusted and adjusted analysis were 1.00 and 0.99, respectively, indicating a false positive finding.
We also observed one additional noteworthy but not genome-wide significant association in the 5-year breast cancer-specific survival analyses in the subgroup of patients who received Tamoxifen with variant rs72775397, situated in the 3′ untranslated region of MCTP1 (HR [95% CI] 1.36 [1.21,1.53], P = 1.8E−07, BFDP = 0.11, per C allele). The association was attenuated after adjustment for additional prognostic factors (HR [95% CI] 1.11 [1.04,1.20], P = 3.8E−03, BFDP = 1.00).
We did not identify any genome-wide significant or noteworthy association in any of the remaining seven subgroups investigated (Figs. 1 and 2). In addition, none of the above reported associations have a BFDP < 0.15 when considering 10−6 as prior probability of true association (Additional file 3: Supplementary Tables S8). Moreover, none of the subgroup-specific genome-wide significant associations detected by previous studies using a smaller version (both in terms of number of cases and of length of follow-up) of the iCOGS and/or OncoArray BCAC datasets were replicated at P < 0.001 (Additional file 3: Supplementary Table S12).

Discussion

We investigated the association of over 10 million common germline genetic variants with breast cancer-specific survival within 15 patient subgroups based on prognostic factors representative of tumor biology or related to the type of systemic treatment. Our hypothesis was that focusing on more homogeneous subgroups of breast cancer patients might reveal otherwise undetected associations. Besides type of systemic treatment, the definition of the subgroups was based on current clinically used biological characteristics for tumor subtyping and treatment decisions: patient’s age, tumor histological grade, ER, PR, and HER2 status ( [31]; Supplementary table S2). We did not have gene expression or copy number aberration data available to classify tumors on the basis of specific biological processes [44, 45]; however, relevant survival differences have been reported among the four subtypes based on ER, PR, and HER2 status [27, 46, 47].
A concern about performing GWAS on several subgroups of patients is the increased proportion of false discoveries, also known as type I errors. For this reason and to overcome additional limitations of the association p values [3537], we made use of the BFDP approach and only considered as robust candidates those associations with BFDP < 0.15 at a prior probability of 10−4.
We found evidence of four loci potentially associated with breast cancer survival: one in the subgroup of patients diagnosed with a grade 3 tumor, one in the subgroup of patients with an ER+ tumor and treated with endocrine therapy, and two in the subgroup of patients with an ER− tumor and treated with chemotherapy.
The most significant variant identified in the subgroup of grade 3 tumors, rs5934618, is situated in intron 1 of TBL1X, a gene which encodes the Transducin (beta)-like 1X-linked protein. Both TBL1X and the closely related gene TBLR1 have been implicated in the activation of the Wnt/beta-catenin signaling pathway, which has been reported to be overactivated in the progression and proliferation of several tumors, including breast tumors, where it has been linked with reduced overall survival [4850]. Rs5934618 and the other four correlated variants identified in our genome-wide analysis overlap with chromatin features H3K4me3 and H3K27ac in normal breast; these histone marks are generally characteristics of gene promoters and/or enhancers and might indicate that one or more of these variants act through modulating expression of TBL1X. There was no direct evidence that any of these variants are expression single-nucleotide polymorphisms (eSNPs) for TBL1X, but this might reflect the tissues examined or that the variants only regulate the gene in a specific context.
The remaining three variants potentially associated with breast cancer survival were as follows: rs4679741, identified in the subgroup of patients diagnosed with an ER+ tumor and treated with endocrine therapy; rs1106333 and rs78754389, identified in the subgroup of patients diagnosed with an ER− tumor and treated with chemotherapy. For variant rs4679741, it is unclear which the potential target genes might be, while there was no evidence linking the other two variants to the expression of the closest genes in breast cancer nor evidence of association between those genes and survival within the specific subgroups of breast cancer patients. Nevertheless, there are several mechanisms through which the four identified variants could affect survival. For example, they could act through regulation of an unannotated long noncoding RNA [51, 52] or microRNA [53, 54]. Further functional studies including epigenetic mechanisms are needed in order to gain more insights about the detected associations and to ascertain the potential underlying biological mechanisms.
We did not find strong evidence of germline variants associated with breast cancer-specific survival in any of the other subgroups of patients investigated. In addition, we did not replicate any of the subgroup-specific associations identified by previous studies. One of these associations, with variant rs4458204, was previously detected in the subgroup of patients with an ER− tumor who received chemotherapy [16]. The estimated HR (95% CI) was 1.81 (1.49–2.19) with association P = 1.9E−09. In our analysis of the same subgroup, we obtained a much lower HR estimate and the association was no longer statistically significant (HR (95% CI) 1.14 (0.99, 1.32), P = 6.0E−02), suggesting that the previous result was a false positive. Even though there is some overlap in terms of patients between the previous study and our current study, the latter is based on a substantially larger number of breast cancer patients and it includes more complete follow-up data.
A major strength of our study is the sample size, which was the largest to date and provided reasonable power to detect associations with breast cancer-specific survival within specific subgroups of patients. On the other hand, our study is subject to several limitations that are intrinsic to large consortium studies: these include variation in study design, time periods of diagnosis, and duration of follow-up, all of which can contribute to within subgroup heterogeneity. Some broad treatment-related subgroups, namely ER+ treated with any endocrine therapy and ER− treated with any chemotherapy, may include different treatments due to the wide period of diagnosis included in our study. On the other hand, the majority of patients were diagnosed between 2000 and 2009 (69.9% and 64.5% for ER+ treated with any endocrine therapy and ER− treated with any chemotherapy, respectively). If any impact on the results, the variation in treatment over time might have hampered the detection of associations between variants and survival in these subgroups. Several studies did not report the cause of death for all patients. Out of 14,606 deaths observed within the first 15 years after diagnosis, 7531 (51.6%) were due to breast cancer. Of the remaining 7075 deaths, 4905 (33.6%) were due to causes other than breast cancer, and for 2170 deaths (14.8%) it was unknown whether they were due to breast cancer or to other causes. This will have led to a loss of power, given that most of the deaths of unknown cause are likely to have been due to breast cancer. A related weakness of the study is its dependence on accuracy of cause of death certification and on coding practices of underlying cause of death in different countries. However, despite potential inaccuracies in cause of death, we considered it more valid to focus on deaths reported as due to breast cancer than by considering all deaths together, which would include those due to other causes. An additional limitation of the study is that in most subgroups we had very limited power to detect highly significant associations, particularly for small to moderate effect sizes (HRs 1.05–1.30), even for variants of relatively high minor allele frequency (MAF = 0.20). Therefore, we may have missed variants with low to moderate associations with survival.

Conclusions

In conclusion, we found evidence of four loci associated with breast cancer-specific survival within specific patient subgroups. The variants identified appear to be independent of known additional prognostic factors, as shown in the results of the adjusted analyses based on imputed clinic-pathological variables, and could, after proper validation, improve prognostic estimates and potentially help in better stratifying patients in treatment subgroups. However, the power for many subgroups is limited due to the low number of events. Even so, given the lack of evidence of strong associations in many of the patient subgroups investigated, and the fact that previously reported variants were not confirmed, our results suggest that the impact of common germline genetic variant on breast cancer-specific survival might be limited.

Acknowledgements

We thank all the individuals who took part in these studies and all the researchers, clinicians, technicians, and administrative staff who have enabled this work to be carried out. ABCFS thank Maggie Angelakos, Judi Maskiell, Gillian Dite. ABCTB Investigators: Christine Clarke, Deborah Marsh, Rodney Scott, Robert Baxter, Desmond Yip, Jane Carpenter, Alison Davis, Nirmala Pathmanathan, Peter Simpson, J. Dinny Graham, Mythily Sachchithananthan. Samples are made available to researchers on a non-exclusive basis. BCEES thanks Allyson Thomson, Christobel Saunders, Terry Slevin, BreastScreen Western Australia, Elizabeth Wylie, Rachel Lloyd. The BCINIS study would not have been possible without the contributions of Dr. K. Landsman, Dr. N. Gronich, Dr. A. Flugelman, Dr. W. Saliba, Dr. F. Lejbkowicz, Dr. E. Liani, Dr. I. Cohen, Dr. S. Kalet, Dr. V. Friedman, Dr. O. Barnet of the NICCC in Haifa, and all the contributing family medicine, surgery, pathology and oncology teams in all medical institutes in Northern Israel. BIGGS thanks Niall McInerney, Gabrielle Colleran, Andrew Rowan, Angela Jones. The BREOGAN study would not have been possible without the contributions of the following: Manuela Gago-Dominguez, Jose Esteban Castelao, Angel Carracedo, Victor Muñoz Garzón, Alejandro Novo Domínguez, Maria Elena Martinez, Sara Miranda Ponte, Carmen Redondo Marey, Maite Peña Fernández, Manuel Enguix Castelo, Maria Torres, Manuel Calaza (BREOGAN), José Antúnez, Máximo Fraga and the staff of the Department of Pathology and Biobank of the University Hospital Complex of Santiago-CHUS, Instituto de Investigación Sanitaria de Santiago, IDIS, Xerencia de Xestion Integrada de Santiago-SERGAS; Joaquín González-Carreró and the staff of the Department of Pathology and Biobank of University Hospital Complex of Vigo, Instituto de Investigacion Biomedica Galicia Sur, SERGAS, Vigo, Spain. The BSUCH study acknowledges the Principal Investigator, Barbara Burwinkel, and, thanks Peter Bugert, Medical Faculty Mannheim. CCGP thanks Styliani Apostolaki, Anna Margiolaki, Georgios Nintos, Maria Perraki, Georgia Saloustrou, Georgia Sevastaki, Konstantinos Pompodakis. CGPS thanks staff and participants of the Copenhagen General Population Study. For the excellent technical assistance: Dorthe Uldall Andersen, Maria Birna Arnadottir, Anne Bank, Dorthe Kjeldgård Hansen. The Danish Cancer Biobank is acknowledged for providing infrastructure for the collection of blood samples for the cases. Investigators from the CPS-II cohort thank the participants and Study Management Group for their invaluable contributions to this research. They also acknowledge the contribution to this study from central cancer registries supported through the Centers for Disease Control and Prevention National Program of Cancer Registries, as well as cancer registries supported by the National Cancer Institute Surveillance Epidemiology and End Results program. The authors would like to thank the California Teachers Study Steering Committee that is responsible for the formation and maintenance of the Study within which this research was conducted. A full list of California Teachers Study team members is available at https://​www.​calteachersstudy​.​org/​team. DIETCOMPLYF thanks the patients, nurses and clinical staff involved in the study. The DietCompLyf study was funded by the charity Against Breast Cancer (Registered Charity Number 1121258) and the NCRN. We thank the participants and the investigators of EPIC (European Prospective Investigation into Cancer and Nutrition). ESTHER thanks Hartwig Ziegler, Sonja Wolf, Volker Hermann, Christa Stegmaier, Katja Butterbach. FHRISK thanks NIHR for funding. GC-HBOC thanks Stefanie Engert, Heide Hellebrand, Sandra Kröber and LIFE - Leipzig Research Centre for Civilization Diseases (Markus Loeffler, Joachim Thiery, Matthias Nüchter, Ronny Baber). The GENICA Network: Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Germany [HB, RH, Wing-Yee Lo], German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany [HB], gefördert durch die Deutsche Forschungsgemeinschaft (DFG) im Rahmen der Exzellenzstrategie des Bundes und der Länder - EXC 2180 - 390900677 [HB], Department of Internal Medicine, Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn, Germany [Yon-Dschun Ko, Christian Baisch], Institute of Pathology, University of Bonn, Germany [Hans-Peter Fischer], Molecular Genetics of Breast Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany [Ute Hamann], Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), Bochum, Germany [TB, Beate Pesch, Sylvia Rabstein, Anne Lotz]; and Institute of Occupational Medicine and Maritime Medicine, University Medical Center Hamburg-Eppendorf, Germany [Volker Harth]. HABCS would like to thank Peter Schürmann, Natalia Bogdanova, Nikki Adrian Krentel, Regina Meier, Frank Papendorf, Michael Bremer, Johann H. Karstens, Hans Christiansen and Peter Hillemanns for their contributions to this study. HEBCS thanks Johanna Kiiski, Carl Blomqvist, Sofia Khan, Kristiina Aittomäki, Kirsimari Aaltonen, Karl von Smitten, Irja Erkkilä. ICICLE thanks Kelly Kohut, Michele Caneppele, Maria Troy. KARMA and SASBAC thank the Swedish Medical Research Counsel. KBCP thanks Eija Myöhänen, Helena Kemiläinen. kConFab/AOCS wish to thank Heather Thorne, Eveline Niedermayr, all the kConFab research nurses and staff, the heads and staff of the Family Cancer Clinics, and the Clinical Follow Up Study (which has received funding from the NHMRC, the National Breast Cancer Foundation, Cancer Australia, and the National Institute of Health (USA)) for their contributions to this resource, and the many families who contribute to kConFab. LMBC thanks Gilian Peuteman, Thomas Van Brussel, EvyVanderheyden and Kathleen Corthouts. MABCS thanks Milena Jakimovska (RCGEB “Georgi D. Efremov”), Emilija Lazarova, Marina Iljovska (University Clinic of Radiotherapy and Oncology), Katerina Kubelka-Sabit, Dzengis Jashar and Mitko Karadjozov (Adzibadem-Sistina Hospital) for their contributions and commitment to this study. MARIE thanks Petra Seibold, Dieter Flesch-Janys, Judith Heinz, Nadia Obi, Alina Vrieling, Sabine Behrens, Ursula Eilber, Muhabbet Celik, Til Olchers and Stefan Nickels. MBCSG (Milan Breast Cancer Study Group): Paolo Radice, Siranoush Manoukian, Jacopo Azzollini, Erica Rosina, Daniela Zaffaroni, Bernardo Bonanni, Irene Feroce, Mariarosaria Calvello, Aliana Guerrieri Gonzaga, Monica Marabelli, Davide Bondavalli and the personnel of the Cogentech Cancer Genetic Test Laboratory. The MCCS was made possible by the contribution of many people, including the original investigators, the teams that recruited the participants and continue working on follow-up, and the many thousands of Melbourne residents who continue to participate in the study. We thank the coordinators, the research staff and especially the MMHS participants for their continued collaboration on research studies in breast cancer. The following are NBCS Collaborators: Anne-Lise Børresen-Dale (Prof. Em.), Kristine K. Sahlberg (PhD), Lars Ottestad (MD), Rolf Kåresen (Prof. Em.) Dr. Ellen Schlichting (MD), Marit Muri Holmen (MD), Toril Sauer (MD), Vilde Haakensen (MD), Olav Engebråten (MD), Bjørn Naume (MD), Alexander Fosså (MD), Cecile E. Kiserud (MD), Kristin V. Reinertsen (MD), Åslaug Helland (MD), Margit Riis (MD), Jürgen Geisler (MD), OSBREAC and Grethe I. Grenaker Alnæs (MSc). For NHS and NHS2 the study protocol was approved by the institutional review boards of the Brigham and Women’s Hospital and Harvard T.H. Chan School of Public Health, and those of participating registries as required. We would like to thank the participants and staff of the NHS and NHS2 for their valuable contributions as well as the following state cancer registries for their help: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, IN, IA, KY, LA, ME, MD, MA, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, SC, TN, TX, VA, WA, WY. The authors assume full responsibility for analyses and interpretation of these data. OBCS thanks Katri Pylkäs, Arja Jukkola, Saila Kauppila, Meeri Otsukka, Leena Keskitalo and Kari Mononen for their contributions to this study. The OFBCR thanks Teresa Selander, Nayana Weerasooriya and Steve Gallinger. ORIGO thanks E. Krol-Warmerdam, and J. Blom for patient accrual, administering questionnaires, and managing clinical information. The LUMC survival data were retrieved from the Leiden hospital-based cancer registry system (ONCDOC) with the help of Dr. J. Molenaar. PBCS thanks Louise Brinton, Mark Sherman, Neonila Szeszenia-Dabrowska, Beata Peplonska, Witold Zatonski, Pei Chao, Michael Stagner. The ethical approval for the POSH study is MREC /00/6/69, UKCRN ID: 1137. We thank staff in the Experimental Cancer Medicine Centre (ECMC) supported Faculty of Medicine Tissue Bank and the Faculty of Medicine DNA Banking resource. PREFACE thanks Sonja Oeser and Silke Landrith. PROCAS thanks NIHR for funding. The RBCS thanks Jannet Blom, Saskia Pelders, Wendy J.C. Prager – van der Smissen, and the Erasmus MC Family Cancer Clinic. SBCS thanks Sue Higham, Helen Cramp, Dan Connley, Ian Brock, Sabapathy Balasubramanian and Malcolm W.R. Reed. We thank the SEARCH and EPIC teams. SKKDKFZS thanks all study participants, clinicians, family doctors, researchers and technicians for their contributions and commitment to this study. We thank the SUCCESS Study teams in Munich, Duessldorf, Erlangen and Ulm. SZBCS thanks Ewa Putresza. UCIBCS thanks Irene Masunaka. UKBGS thanks Breast Cancer Now and the Institute of Cancer Research for support and funding of the Generations Study, and the study participants, study staff, and the doctors, nurses and other health care providers and health information sources who have contributed to the study. We acknowledge NHS funding to the Royal Marsden/ICR NIHR Biomedical Research Centre. The authors thank the WHI investigators and staff for their dedication and the study participants for making the program possible.

Declarations

The study was performed in accordance with the Declaration of Helsinki. All individual studies included in the analyses were approved by the appropriate institutional ethical review boards. All study participants provided informed consent.
Not applicable.

Competing interests

Matthias W. Beckmann and Peter A. Fasching conduct research funded by Amgen, Novartis, and Pfizer (not related to this study). Peter A. Fasching received Honoraria from Roche, Novartis, and Pfizer (not related to this study). Allison W. Kurian’s institution received a research funding from Myriad genetics for an unrelated project (not related to this study). The other authors declare no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Additional file 2: Supplementary Methods, Supplementary Table S5, Supplementary Table S6, Supplementary Table S7, Supplementary Figure S1, Supplementary Figure S2, Supplementary Figure S3, Supplementary Figure S4, Supplementary Figure S5, Supplementary Figure S6, Supplementary Figure S7, Supplementary Figure S8, Supplementary Figure S9, Supplementary Figure S10, Supplementary Figure S11, Supplementary Figure S12. The Supplementary methods section includes details about: multiple imputation of missing data; power calculations. Supplementary Table S5. Shows an overview of number of breast cancer patients, breast cancer deaths and follow-up information by subgroup and endpoint. Supplementary Table S6. Shows a list of imputed variables with corresponding percentage of missing values, imputation method and processing. Supplementary Table S7. Shows an overview of the GWAS significant associations (P < 5 E-08) and noteworthy (BFDP< 0.15) associations from the unadjusted 15-year and 5-year breast cancer-specific survival analyses by subgroup. Supplementary Figure S1. Shows the Q-Q plots of the meta-analysis results of all variants for 15-year breast cancer-specific survival. Supplementary Figure S2. Shows the Q-Q plots of the meta-analysis results of all variants for 5-year breast cancer-specific survival. Supplementary Figure S3. Shows the Q-Q plots of the meta-analysis results for 15-year breast cancer-specific survival and the corresponding genome inflation factors by minor allele frequency. Supplementary Figure S4. Shows the Q-Q plots of the meta-analysis results for 5-year breast cancer-specific survival and the corresponding genome inflation factors by minor allele frequency. Supplementary Figure S5. Shows the regional plots of genome-wide significant (P < 5E-08) independent associated variants from the 15-year and 5-year genome-wide breast cancer-specific survival analyses. Supplementary Figure S6. Shows the regional plots of noteworthy (BFDP< 0.15), non-genome-wide significant (P > 5E-08) variants from the 15-year and 5-year genome-wide breast cancer-specific survival analyses. Supplementary Figure S7 shows the unadjusted association of variant rs5934618 with 15-year breast cancer-specific survival by tumor grade and in all breast cancer patients. Supplementary Figure S8. Shows the functional annotation and position of variants rs5934618, rs4830644, rs3810742, rs4830642, and rs72611496 relative to TBL1X. Supplementary Figure S9. Shows Kaplan-Meier distant metastasis-free survival plots (based on KMPlotter data) for high versus low expression level of gene TBL1X by tumor grade. Supplementary Figure S10. Shows the association of genetic variants with TBL1X expression, based on GTEx v8 data on samples of normal breast tissue from 396 individuals (male and female). Supplementary Figure S11. Shows a Kaplan-Meier distant metastasis-free survival plot for high versus low expression level of gene GRIP2, restricted to patients with an ER- tumor who received chemotherapy (based on KMPlotter data). Supplementary Figure S12. Shows a Kaplan-Meier distant metastasis-free survival plot for high versus low expression level of gene ARAP2, restricted to patients with an ER- tumor who received chemotherapy (based on KMPlotter data).
Additional file 3: Supplementary Table S8, Supplementary Table S9, Supplementary Table S10, Supplementary Table S11, and Supplementary Table S12. Supplementary Table S8. Shows the BFDPs under two more restrictive prior probabilities of true association (10-5 and 10-6) for the results presented in Table 1. Supplementary Table S9. Shows the BFDPs under two more restrictive prior probabilities of true association (10-5 and 10-6) for the results presented in Table 2. Supplementary Table S10. Shows power calculation by subgroup, at the two-sided 5E-08 level for varying genotype hazard ratio (GHR) and minor allele frequency (MAF), based on number of cases and event rate from the 15-year breast cancer-specific analyses. Supplementary Table S11. Shows power calculation by subgroup, at the two-sided 5E-08 level for varying genotype hazard ratio (GHR) and minor allele frequency (MAF), based on number of cases and event rate from the 5-year breast cancer-specific analyses. Supplementary Table S12. Shows the subgroup-specific associations detected by previous studies and corresponding estimates from the current study.
Literatur
3.
Zurück zum Zitat Crawford NPS, Alsarraj J, Lukes L, Walker RC, Officewala JS, Yang HH, Lee MP, Ozato K, Hunter KW <em>Bromodomain 4</em> activation predicts breast cancer survival. Proceedings of the National Academy of Sciences. 2008;105(17):6380-6385, doi: https://doi.org/10.1073/pnas.0710331105. Crawford NPS, Alsarraj J, Lukes L, Walker RC, Officewala JS, Yang HH, Lee MP, Ozato K, Hunter KW <em>Bromodomain 4</em> activation predicts breast cancer survival. Proceedings of the National Academy of Sciences. 2008;105(17):6380-6385, doi: https://​doi.​org/​10.​1073/​pnas.​0710331105.
7.
Zurück zum Zitat Winter JM, Gildea DE, Andreas JP, Gatti DM, Williams KA, Lee M, et al. Mapping complex traits in a diversity outbred f1 mouse population identifies germline modifiers of metastasis in human prostate cancer. Cell Syst. 2017;4(1):31–45.e6.CrossRef Winter JM, Gildea DE, Andreas JP, Gatti DM, Williams KA, Lee M, et al. Mapping complex traits in a diversity outbred f1 mouse population identifies germline modifiers of metastasis in human prostate cancer. Cell Syst. 2017;4(1):31–45.e6.CrossRef
16.
Zurück zum Zitat Li J, Lindström LS, Foo JN, Rafiq S, Schmidt MK, Pharoah PDP, et al. 2q36.3 is associated with prognosis for oestrogen receptor-negative breast cancer patients treated with chemotherapy. Nat Commun. 2014;5(1):4051.CrossRef Li J, Lindström LS, Foo JN, Rafiq S, Schmidt MK, Pharoah PDP, et al. 2q36.3 is associated with prognosis for oestrogen receptor-negative breast cancer patients treated with chemotherapy. Nat Commun. 2014;5(1):4051.CrossRef
23.
Zurück zum Zitat Kiyotani K, Mushiroda T, Tsunoda T, Morizono T, Hosono N, Kubo M, et al. A genome-wide association study identifies locus at 10q22 associated with clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients in Japanese. Hum Mol Genet. 2011;21(7):1665–72. https://doi.org/10.1093/hmg/ddr597.CrossRefPubMed Kiyotani K, Mushiroda T, Tsunoda T, Morizono T, Hosono N, Kubo M, et al. A genome-wide association study identifies locus at 10q22 associated with clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients in Japanese. Hum Mol Genet. 2011;21(7):1665–72. https://​doi.​org/​10.​1093/​hmg/​ddr597.CrossRefPubMed
28.
Zurück zum Zitat Fagerholm R, Schmidt MK, Khan S, Rafiq S, Tapper W, Aittomäki K, et al. The SNP rs6500843 in 16p13.3 is associated with survival specifically among chemotherapy-treated breast cancer patients. Oncotarget. 2015;6(10):7390–407. Fagerholm R, Schmidt MK, Khan S, Rafiq S, Tapper W, Aittomäki K, et al. The SNP rs6500843 in 16p13.3 is associated with survival specifically among chemotherapy-treated breast cancer patients. Oncotarget. 2015;6(10):7390–407.
30.
Zurück zum Zitat Cardoso F, Senkus E, Costa A, Papadopoulos E, Aapro M, André F, et al. 4th ESO–ESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 4)††These guidelines were developed by the European School of Oncology (ESO) and the European Society for Medical Oncology (ESMO). Ann Oncol. 2018;29(8):1634–57. https://doi.org/10.1093/annonc/mdy192.CrossRefPubMed Cardoso F, Senkus E, Costa A, Papadopoulos E, Aapro M, André F, et al. 4th ESO–ESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 4)††These guidelines were developed by the European School of Oncology (ESO) and the European Society for Medical Oncology (ESMO). Ann Oncol. 2018;29(8):1634–57. https://​doi.​org/​10.​1093/​annonc/​mdy192.CrossRefPubMed
33.
Zurück zum Zitat the Haplotype Reference Consortium, McCarthy S, Das S, Kretzschmar W, Delaneau O, Wood AR, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nature Genet. 2016;48:1279.CrossRef the Haplotype Reference Consortium, McCarthy S, Das S, Kretzschmar W, Delaneau O, Wood AR, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nature Genet. 2016;48:1279.CrossRef
34.
Zurück zum Zitat EBCTCG. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–717.CrossRef EBCTCG. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–717.CrossRef
37.
Zurück zum Zitat Burton PR, Clayton DG, Cardon LR, Craddock N, Deloukas P, Duncanson A, et al. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Burton PR, Clayton DG, Cardon LR, Craddock N, Deloukas P, Duncanson A, et al. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls.
42.
Zurück zum Zitat Gong J, Mei S, Liu C, Xiang Y, Ye Y, Zhang Z, et al. PancanQTL: systematic identification of cis-eQTLs and trans-eQTLs in 33 cancer types. Nucleic Acids Res. 2017;46(D1):D971–D6.CrossRef Gong J, Mei S, Liu C, Xiang Y, Ye Y, Zhang Z, et al. PancanQTL: systematic identification of cis-eQTLs and trans-eQTLs in 33 cancer types. Nucleic Acids Res. 2017;46(D1):D971–D6.CrossRef
43.
Zurück zum Zitat Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2018;47(D1):D1005–D12.CrossRef Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2018;47(D1):D1005–D12.CrossRef
47.
Zurück zum Zitat Howlader N, Cronin KA, Kurian AW, Andridge R. Differences in breast cancer survival by molecular subtypes in the United States. Cancer Epidemiology Biomarkers &amp. Prevention. 2018;27(6):619–26. Howlader N, Cronin KA, Kurian AW, Andridge R. Differences in breast cancer survival by molecular subtypes in the United States. Cancer Epidemiology Biomarkers &amp. Prevention. 2018;27(6):619–26.
50.
Zurück zum Zitat Inagawa S, Itabashi M, Adachi S, Kawamoto T, Hori M, Shimazaki J, et al. Expression and prognostic roles of beta-catenin in hepatocellular carcinoma: correlation with tumor progression and postoperative survival. Clin Cancer Res. 2002;8(2):450–6.PubMed Inagawa S, Itabashi M, Adachi S, Kawamoto T, Hori M, Shimazaki J, et al. Expression and prognostic roles of beta-catenin in hepatocellular carcinoma: correlation with tumor progression and postoperative survival. Clin Cancer Res. 2002;8(2):450–6.PubMed
Metadaten
Titel
Association of germline genetic variants with breast cancer-specific survival in patient subgroups defined by clinic-pathological variables related to tumor biology and type of systemic treatment
verfasst von
Anna Morra
Maria Escala-Garcia
Jonathan Beesley
Renske Keeman
Sander Canisius
Thomas U. Ahearn
Irene L. Andrulis
Hoda Anton-Culver
Volker Arndt
Paul L. Auer
Annelie Augustinsson
Laura E. Beane Freeman
Heiko Becher
Matthias W. Beckmann
Sabine Behrens
Stig E. Bojesen
Manjeet K. Bolla
Hermann Brenner
Thomas Brüning
Saundra S. Buys
Bette Caan
Daniele Campa
Federico Canzian
Jose E. Castelao
Jenny Chang-Claude
Stephen J. Chanock
Ting-Yuan David Cheng
Christine L. Clarke
Sarah V. Colonna
Fergus J. Couch
Angela Cox
Simon S. Cross
Kamila Czene
Mary B. Daly
Joe Dennis
Thilo Dörk
Laure Dossus
Alison M. Dunning
Miriam Dwek
Diana M. Eccles
Arif B. Ekici
A. Heather Eliassen
Mikael Eriksson
D. Gareth Evans
Peter A. Fasching
Henrik Flyger
Lin Fritschi
Manuela Gago-Dominguez
José A. García-Sáenz
Graham G. Giles
Mervi Grip
Pascal Guénel
Melanie Gündert
Eric Hahnen
Christopher A. Haiman
Niclas Håkansson
Per Hall
Ute Hamann
Steven N. Hart
Jaana M. Hartikainen
Arndt Hartmann
Wei He
Maartje J. Hooning
Reiner Hoppe
John L. Hopper
Anthony Howell
David J. Hunter
Agnes Jager
Anna Jakubowska
Wolfgang Janni
Esther M. John
Audrey Y. Jung
Rudolf Kaaks
Machteld Keupers
Cari M. Kitahara
Stella Koutros
Peter Kraft
Vessela N. Kristensen
Allison W. Kurian
James V. Lacey
Diether Lambrechts
Loic Le Marchand
Annika Lindblom
Martha Linet
Robert N. Luben
Jan Lubiński
Michael Lush
Arto Mannermaa
Mehdi Manoochehri
Sara Margolin
John W. M. Martens
Maria Elena Martinez
Dimitrios Mavroudis
Kyriaki Michailidou
Roger L. Milne
Anna Marie Mulligan
Taru A. Muranen
Heli Nevanlinna
William G. Newman
Sune F. Nielsen
Børge G. Nordestgaard
Andrew F. Olshan
Håkan Olsson
Nick Orr
Tjoung-Won Park-Simon
Alpa V. Patel
Bernard Peissel
Paolo Peterlongo
Dijana Plaseska-Karanfilska
Karolina Prajzendanc
Ross Prentice
Nadege Presneau
Brigitte Rack
Gad Rennert
Hedy S. Rennert
Valerie Rhenius
Atocha Romero
Rebecca Roylance
Matthias Ruebner
Emmanouil Saloustros
Elinor J. Sawyer
Rita K. Schmutzler
Andreas Schneeweiss
Christopher Scott
Mitul Shah
Snezhana Smichkoska
Melissa C. Southey
Jennifer Stone
Harald Surowy
Anthony J. Swerdlow
Rulla M. Tamimi
William J. Tapper
Lauren R. Teras
Mary Beth Terry
Rob A. E. M. Tollenaar
Ian Tomlinson
Melissa A. Troester
Thérèse Truong
Celine M. Vachon
Qin Wang
Amber N. Hurson
Robert Winqvist
Alicja Wolk
Argyrios Ziogas
Hiltrud Brauch
Montserrat García-Closas
Paul D. P. Pharoah
Douglas F. Easton
Georgia Chenevix-Trench
Marjanka K. Schmidt
NBCS Collaborators
ABCTB Investigators
kConFab Investigators
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2021
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-021-01450-7

Weitere Artikel der Ausgabe 1/2021

Breast Cancer Research 1/2021 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.