ϟ

Arja Jukkola‐Vuorinen

Here are all the papers by Arja Jukkola‐Vuorinen that you can download and read on OA.mg.
Arja Jukkola‐Vuorinen’s last known institution is . Download Arja Jukkola‐Vuorinen PDFs here.

Claim this Profile →
DOI: 10.1158/1078-0432.ccr-05-1029
2006
Cited 522 times
The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer.
Abstract To investigate the clinical value of somatic TP53 mutations in breast cancer, we assembled clinical and molecular data on 1,794 women with primary breast cancer with long-term follow-up and whose tumor has been screened for mutation in exons 5 to 8 of TP53 by gene sequencing. TP53 mutations were more frequent in tumors of ductal and medullar types, aggressive phenotype (high grade, large size, node positive cases, and low hormone receptor content) and in women <60 years old. TP53 mutations within exons 5 to 8 conferred an elevated risk of breast cancer-specific death of 2.27 (relative risk >10 years; P < 0.0001) compared with patients with no such mutation. The prognostic value of TP53 mutation was independent of tumor size, node status, and hormone receptor content, confirming and reconciling previous findings in smaller series. Moreover, an interaction between TP53 mutation and progesterone receptor (PR) status was revealed, TP53 mutation combined with the absence of progesterone receptor being associated with the worst prognosis. Whereas previous studies have emphasized the fact that missense mutations in the DNA-binding motifs have a worse prognosis than missense mutations outside these motifs, we show that non-missense mutations have prognostic value similar to missense mutations in DNA-binding motifs. Nonetheless, specific missense mutants (codon 179 and R248W) seem to be associated with an even worse prognosis. These results, obtained on the largest series analyzed thus far, show that TP53 mutations identified by gene sequencing have an independent prognostic value in breast cancer and could have potential uses in clinical practice.
DOI: 10.1038/nature05609
2007
Cited 413 times
A recurrent mutation in PALB2 in Finnish cancer families
DOI: 10.1038/ng.985
2011
Cited 286 times
A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor–negative breast cancer
Estrogen receptor (ER)-negative breast cancer shows a higher incidence in women of African ancestry compared to women of European ancestry. In search of common risk alleles for ER-negative breast cancer, we combined genome-wide association study (GWAS) data from women of African ancestry (1,004 ER-negative cases and 2,745 controls) and European ancestry (1,718 ER-negative cases and 3,670 controls), with replication testing conducted in an additional 2,292 ER-negative cases and 16,901 controls of European ancestry. We identified a common risk variant for ER-negative breast cancer at the TERT-CLPTM1L locus on chromosome 5p15 (rs10069690: per-allele odds ratio (OR) = 1.18 per allele, P = 1.0 × 10(-10)). The variant was also significantly associated with triple-negative (ER-negative, progesterone receptor (PR)-negative and human epidermal growth factor-2 (HER2)-negative) breast cancer (OR = 1.25, P = 1.1 × 10(-9)), particularly in younger women (<50 years of age) (OR = 1.48, P = 1.9 × 10(-9)). Our results identify a genetic locus associated with estrogen receptor negative breast cancer subtypes in multiple populations.
DOI: 10.1007/s10549-013-2442-0
2013
Cited 115 times
Vimentin, zeb1 and Sip1 are up-regulated in triple-negative and basal-like breast cancers: association with an aggressive tumour phenotype
DOI: 10.1038/sj.bjc.6605565
2010
Cited 113 times
8-Hydroxydeoxyguanosine: a new potential independent prognostic factor in breast cancer
8-Hydroxydeoxyguanosine (8-oxodG) is the commonly used marker of oxidative stress-derived DNA damage. 8-OxodG formation is regulated by local antioxidant capacity and DNA repair enzyme activity. Earlier studies have reported contradictory data on the function of 8-oxodG as a prognostic factor in different cancer types. We assessed pre-operative serum 8-oxodG levels with an enzyme-linked immunosorbent assay in a well-defined series of 173 breast cancer patients. 8-OxodG expression in the nuclei of cancer cells from 150 of these patients was examined by immunohistochemistry. The serum 8-oxodG levels and immunohistochemical 8-oxodG expression were in concordance with each other (P<0.05). Negative 8-oxodG immunostaining was an independent prognostic factor for poor breast cancer-specific survival according to the multivariate analysis (P<0.01). This observation was even more remarkable when ductal carcinomas only (n=140) were considered (P<0.001). A low serum 8-oxodG level was associated statistically significantly with lymphatic vessel invasion and a positive lymph node status. Low serum 8-oxodG levels and a low immunohistochemical 8-oxodG expression were associated with an aggressive breast cancer phenotype. In addition, negative 8-oxodG immunostaining was a powerful prognostic factor for breast cancer-specific death in breast carcinoma patients.
DOI: 10.1186/bcr3376
2013
Cited 113 times
The prognostic significance and value of cyclin D1, CDK4 and p16 in human breast cancer
Loss of the retinoblastoma protein tumor suppressor gene (RB) coding for a nuclear phosphoprotein that regulates the cell cycle is found in many human cancers and probably leads to disruption of the p16-cyclin D1-CDK4/6-RB pathway. Cyclin D1 is known to activate CDK4, which then phosphorylates the RB protein, leading to cell cycle progression. p16 inhibits CDK4, keeping RB hypophosphorylated and preventing cell cycle progression. The significance of these three markers, cyclin D1, CDK4 and p16, for breast cancer and carcinogenesis is nevertheless still controversial.The material consisted of 102 formalin-fixed human breast cancer samples, in which cyclin D1, CDK4 and p16 expression was evaluated immunohistochemically. The amounts of cyclin D1 mRNA present were analyzed by quantitative real time PCR.High cyclin D1 expression statistically significantly correlated with lower tumor grade, estrogen and progesterone receptor positivity and lower proliferation activity in breast tumors and increased breast cancer-specific survival and overall survival. Tumors with high cyclin D1 protein had 1.8 times higher expression of cyclin D1 mRNA. CDK4 expression did not correlate with cyclin D1 expression or the survival data. p16 expression was associated with Human Epidermal Growth Factor Receptor 2 (HER2) negativity and increased breast cancer-specific survival and disease-free survival. No statistical correlations between cyclin D1, CDK4 and p16 were found.Cyclin D1 was associated with a good breast cancer prognosis but functioned independently of CDK4. High cyclin D1 expression may be partially due to increased CCND1 transcription. p16 correlated with a better prognosis and may function without CDK4. In conclusion, it appears that cyclin D1, CDK4 and p16 function independently in human breast cancer.
DOI: 10.1158/0008-5472.can-11-1266
2011
Cited 112 times
Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer
Abstract Triple-negative breast cancers are an aggressive subtype of breast cancer with poor survival, but there remains little known about the etiologic factors that promote its initiation and development. Commonly inherited breast cancer risk factors identified through genome-wide association studies display heterogeneity of effect among breast cancer subtypes as defined by the status of estrogen and progesterone receptors. In the Triple Negative Breast Cancer Consortium (TNBCC), 22 common breast cancer susceptibility variants were investigated in 2,980 Caucasian women with triple-negative breast cancer and 4,978 healthy controls. We identified six single-nucleotide polymorphisms, including rs2046210 (ESR1), rs12662670 (ESR1), rs3803662 (TOX3), rs999737 (RAD51L1), rs8170 (19p13.1), and rs8100241 (19p13.1), significantly associated with the risk of triple-negative breast cancer. Together, our results provide convincing evidence of genetic susceptibility for triple-negative breast cancer. Cancer Res; 71(19); 6240–9. ©2011 AACR.
DOI: 10.1200/jco.2011.35.4639
2012
Cited 103 times
Adjuvant Capecitabine, Docetaxel, Cyclophosphamide, and Epirubicin for Early Breast Cancer: Final Analysis of the Randomized FinXX Trial
Purpose Capecitabine is an active agent in the treatment of breast cancer. It is not known whether integration of capecitabine into an adjuvant regimen that contains a taxane, an anthracycline, and cyclophosphamide improves outcome in early breast cancer. Patients and Methods Women with axillary node–positive or high-risk node-negative breast cancer were randomly assigned to receive either three cycles of docetaxel and capecitabine (TX) followed by three cycles of cyclophosphamide, epirubicin, and capecitabine (CEX; n = 753) or three cycles of docetaxel (T) followed by three cycles of cyclophosphamide, epirubicin, and fluorouracil (CEF; n = 747). The primary end point was recurrence-free survival (RFS). Results During a median follow-up time of 59 months, 214 RFS events occurred (local or distant recurrences or deaths; TX/CEX, n = 96; T/CEF, n = 118). RFS was not significantly different between the groups (hazard ratio [HR], 0.79; 95% CI, 0.60 to 1.04; P = .087; 5-year RFS, 86.6% for TX/CEX v 84.1% for T/CEF). Fifty-six patients assigned to TX/CEX died during the follow-up compared with 75 of patients assigned to T/CEF (HR, 0.73; 95% CI, 0.52 to 1.04; P = .080). In exploratory analyses, TX/CEX improved breast cancer–specific survival (HR, 0.64; 95% CI, 0.44 to 0.95; P = .027) and RFS in women with triple-negative disease and in women who had more than three metastatic axillary lymph nodes at the time of diagnosis. We detected little severe late toxicity. Conclusion Integration of capecitabine into a regimen that contains docetaxel, epirubicin, and cyclophosphamide did not improve RFS significantly compared with a similar regimen without capecitabine.
DOI: 10.1001/jamaoncol.2016.6120
2017
Cited 76 times
Adjuvant Capecitabine in Combination With Docetaxel, Epirubicin, and Cyclophosphamide for Early Breast Cancer
Capecitabine is not considered a standard agent in the adjuvant treatment of early breast cancer. The results of this study suggest that addition of adjuvant capecitabine to a regimen that contains docetaxel, epirubicin, and cyclophosphamide improves survival outcomes of patients with triple-negative breast cancer (TNBC).To investigate the effect of capecitabine on long-term survival outcomes of patients with early breast cancer, particularly in subgroups defined by cancer estrogen receptor (ER) and progesterone receptor (PR) content, and HER2 content (human epidermal growth factor receptor 2).This is an exploratory analysis of the multicenter FinXX randomized clinical trial that accrued 1500 women in Finland and Sweden between January 27, 2004, and May 29, 2007. About half received 3 cycles of docetaxel followed by 3 cycles of cyclophosphamide, epirubicin, and fluorouracil (T+CEF), while the other half received 3 cycles of docetaxel plus capecitabine followed by 3 cycles of cyclophosphamide, epirubicin, and capecitabine (TX+CEX). Data analysis took place between January 27, 2004, and December 31, 2015.Recurrence-free survival (RFS).Following random allocation, 747 women received T+CEF, and 753 women received TX+CEX. Five patients were excluded from the intention-to-treat population (3 had overt distant metastases at the time of randomization; 2 withdrew consent). The median age of the remaining 1495 patients was 53 years at the time of study entry; 157 (11%) had axillary node-negative disease; 1142 (76%) had ER-positive cancer; and 282 (19%) had HER2-positive cancer. The median follow-up time after random allocation was 10.3 years. There was no significant difference in RFS or overall survival between the groups (hazard ratio [HR], 0.88; 95% CI, 0.71-1.08; P = .23; and HR, 0.84, 95% CI, 0.66-1.07; P = .15; respectively). Breast cancer-specific survival tended to favor the capecitabine group (HR, 0.79; 95% CI, 0.60-1.04; P = .10). When RFS and survival of the patients were compared within the subgroups defined by cancer steroid hormone receptor status (ER and/or PR positive vs ER and PR negative) and HER2 status (positive vs negative), TX+CEX was more effective than T+CEF in the subset of patients with TNBC (HR, 0.53; 95% CI, 0.31-0.92; P = .02; and HR, 0.55, 95% CI, 0.31-0.96; P = .03; respectively).Capecitabine administration with docetaxel, epirubicin, and cyclophosphamide did not prolong RFS or survival compared with a regimen that contained only standard agents. Patients with TNBC had favorable survival outcomes when treated with the capecitabine-containing regimen in an exploratory subgroup analysis.clinicaltrials.gov Identifier: NCT00114816.
DOI: 10.1158/1078-0432.ccr-08-0210
2008
Cited 95 times
Penetrance Analysis of the <i>PALB2</i> c.1592delT Founder Mutation
Abstract Purpose: PALB2 is a recently identified breast cancer susceptibility gene. We have previously identified in the Finnish population a PALB2 c.1592delT founder truncation mutation that is associated with an increased risk of breast cancer. In the present study, we wanted to assess in more detail the increased risk (hazard ratio, HR) and the age-specific cumulative risk (penetrance) of c.1592delT with regard to susceptibility to breast and other forms of cancer. Experimental Design: Modified segregation analyses fitted under maximum likelihood theory were used to estimate age-specific cumulative risks and HRs using the families of mutation carriers identified from a consecutive series of breast cancer cases unselected for age at onset or family history. Results: We found a substantially increased risk of breast cancer [HR, 6.1; 95% confidence interval (95% CI), 2.2-17.2; P = 0.01] equivalent to a 40% (95% CI, 17-77) breast cancer risk by age 70 years, comparable to that for carriers of mutations in BRCA2. We found marginal evidence (P = 0.06) that the HR for breast cancer decreased with age by 4.2% per year (95% CI, 0.2-8.1), from 7.5-fold at age 30 years to 2.0-fold at age 60 years. Conclusions: Our results suggest that it may be appropriate to offer PALB2 c.1592delT mutation testing to Finnish women with breast cancer, especially those with an early age at onset or a family history of breast or related cancers, and to offer carriers the option of participation in extended disease surveillance programs.
DOI: 10.1093/hmg/dds159
2012
Cited 80 times
The role of genetic breast cancer susceptibility variants as prognostic factors
Recent genome-wide association studies identified 11 single nucleotide polymorphisms (SNPs) associated with breast cancer (BC) risk. We investigated these and 62 other SNPs for their prognostic relevance. Confirmed BC risk SNPs rs17468277 (CASP8), rs1982073 (TGFB1), rs2981582 (FGFR2), rs13281615 (8q24), rs3817198 (LSP1), rs889312 (MAP3K1), rs3803662 (TOX3), rs13387042 (2q35), rs4973768 (SLC4A7), rs6504950 (COX11) and rs10941679 (5p12) were genotyped for 25 853 BC patients with the available follow-up; 62 other SNPs, which have been suggested as BC risk SNPs by a GWAS or as candidate SNPs from individual studies, were genotyped for replication purposes in subsets of these patients. Cox proportional hazard models were used to test the association of these SNPs with overall survival (OS) and BC-specific survival (BCS). For the confirmed loci, we performed an accessory analysis of publicly available gene expression data and the prognosis in a different patient group. One of the 11 SNPs, rs3803662 (TOX3) and none of the 62 candidate/GWAS SNPs were associated with OS and/or BCS at P<0.01. The genotypic-specific survival for rs3803662 suggested a recessive mode of action [hazard ratio (HR) of rare homozygous carriers=1.21; 95% CI: 1.09-1.35, P=0.0002 and HR=1.29; 95% CI: 1.12-1.47, P=0.0003 for OS and BCS, respectively]. This association was seen similarly in all analyzed tumor subgroups defined by nodal status, tumor size, grade and estrogen receptor. Breast tumor expression of these genes was not associated with prognosis. With the exception of rs3803662 (TOX3), there was no evidence that any of the SNPs associated with BC susceptibility were associated with the BC survival. Survival may be influenced by a distinct set of germline variants from those influencing susceptibility.
DOI: 10.1093/hmg/ddr368
2011
Cited 74 times
Associations of common variants at 1p11.2 and 14q24.1 (RAD51L1) with breast cancer risk and heterogeneity by tumor subtype: findings from the Breast Cancer Association Consortium†
A genome-wide association study (GWAS) identified single-nucleotide polymorphisms (SNPs) at 1p11.2 and 14q24.1 (RAD51L1) as breast cancer susceptibility loci. The initial GWAS suggested stronger effects for both loci for estrogen receptor (ER)-positive tumors. Using data from the Breast Cancer Association Consortium (BCAC), we sought to determine whether risks differ by ER, progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), grade, node status, tumor size, and ductal or lobular morphology. We genotyped rs11249433 at 1p.11.2, and two highly correlated SNPs rs999737 and rs10483813 (r(2)= 0.98) at 14q24.1 (RAD51L1), for up to 46 036 invasive breast cancer cases and 46 930 controls from 39 studies. Analyses by tumor characteristics focused on subjects reporting to be white women of European ancestry and were based on 25 458 cases, of which 87% had ER data. The SNP at 1p11.2 showed significantly stronger associations with ER-positive tumors [per-allele odds ratio (OR) for ER-positive tumors was 1.13, 95% CI = 1.10-1.16 and, for ER-negative tumors, OR was 1.03, 95% CI = 0.98-1.07, case-only P-heterogeneity = 7.6 × 10(-5)]. The association with ER-positive tumors was stronger for tumors of lower grade (case-only P= 6.7 × 10(-3)) and lobular histology (case-only P= 0.01). SNPs at 14q24.1 were associated with risk for most tumor subtypes evaluated, including triple-negative breast cancers, which has not been described previously. Our results underscore the need for large pooling efforts with tumor pathology data to help refine risk estimates for SNP associations with susceptibility to different subtypes of breast cancer.
DOI: 10.1158/1055-9965.epi-06-0841
2007
Cited 82 times
Serum Macrophage Inhibitory Cytokine-1 Concentrations Correlate with the Presence of Prostate Cancer Bone Metastases
Abstract Macrophage-inhibitory cytokine-1 (MIC-1) is a divergent member of the transforming growth factor β superfamily. It is up-regulated by nonsteroidal anti-inflammatory drugs and is highly expressed in human prostate cancer leading to high serum MIC-1 concentrations with advanced disease. A role for MIC-1 has been implicated in the process of early bone formation, suggesting that it may also mediate sclerosis at the site of prostate cancer bone metastases. Consequently, the aim of this study was to retrospectively determine the relationship of serum MIC-1 concentration and other markers related to current and future prostate cancer bone metastasis in a cohort of 159 patients with prostate cancer. Serum markers included cross-linked carboxy-terminal telopeptide of type I collagen, prostate-specific antigen, and amino-terminal propeptide of type I procollagen (PINP). The mean values of all the biomarkers studied were significantly higher in patients with baseline bone metastases (BM+, n = 35), when compared with those without bone metastases (BM−, n = 124). In a multivariate logistic model, both MIC-1 and PINP independently predicted the presence of baseline bone metastasis. Based on receiver operator curve analysis, the best predictor for the presence of baseline bone metastasis was MIC-1, which was significantly better than carboxy-terminal telopeptide of type I collagen, prostate-specific antigen, and PINP. Patients who experienced bone relapse had significantly higher levels of baseline MIC-1 compared with patients who did not (1476.7 versus 988.4; P = 0.03). Current use of acetylsalicylic acid did not influence serum MIC-1 levels in this cohort. Although requiring validation prospectively, these results suggest that serum MIC-1 determination may be a valuable tool for the diagnosis of current and future bone metastases in patients with prostate cancer. (Cancer Epidemiol Biomarkers Prev 2007;16(3):532–7)
DOI: 10.1158/1541-7786.mcr-07-2005
2008
Cited 71 times
Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
Abstract Toll-like receptor 9 (TLR9) belongs to the innate immune system and recognizes microbial and vertebrate DNA. We showed previously that treatment with the TLR9-agonistic ODN M362 (a CpG sequence containing oligonucleotide) induces matrix metalloproteinase-13–mediated invasion in TLR9-expressing human cancer cell lines. Here, we further characterized the role of the TLR9 pathway in this process. We show that CpG oligonucleotides induce invasion in macrophages from wild-type C57/B6 and MyD88 knockout mice and in human MDA-MB-231 breast cancer cells lacking MyD88 expression. This effect was significantly inhibited in macrophages from TLR9 knockout mice and in human MDA-MB-231 breast cancer cells stably expressing TLR9 small interfering RNA or dominant-negative tumor necrosis factor receptor-associated factor 6 (TRAF6). Sequence modifications to the CpG oligonucleotides that targeted the stem loop and other secondary structures were shown to influence the invasion-inducing effect in MDA-MB-231 cells. In contrast, methylation of the cytosine residues of the parent CpG oligonucleotide did not affect the TLR9-mediated invasion compared with the unmethylated parent CpG oligonucleotide. Finally, expression of TLR9 was studied in clinical breast cancer samples and normal breast epithelium with immunohistochemistry. TLR9 staining localized in epithelial cells in both cancer and normal samples. The mean TLR9 staining intensity was significantly increased in the breast cancer cells compared with normal breast epithelial cells. In conclusion, our results suggest that TLR9 expression is increased in breast cancer and CpG oligonucleotide–induced cellular invasion is mediated via TLR9 and TRAF6, independent of MyD88. Further, our findings suggest that the structure and/or stability of DNA may influence the induction of TLR9-mediated invasion in breast cancer. (Mol Cancer Res 2008;6(10):1534–43)
DOI: 10.1111/j.1365-2559.2011.03835.x
2011
Cited 66 times
Divergent behaviour of oxidative stress markers 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxy-2-nonenal (HNE) in breast carcinogenesis
To clarify the role of oxidative stress during breast carcinogenesis by studying the expression of 8-hydroxydeoxyguanosine (8-OHdG) (a marker of oxidative DNA damage) and 4-hydroxy-2-nonenal (HNE) (a marker of lipid peroxidation) during the different phases of breast carcinogenesis.The study material consisted of a total of 219 patients: 31 with usual ductal hyperplasia (UDH), 25 with atypical ductal hyperplasia (ADH), 30 with ductal carcinoma in situ (DCIS) and 133 with invasive carcinoma. The expression of 8-OHdG and HNE were evaluated immunohistochemically. Both 8-OHdG (77.4%) and HNE (45.8%) expression was already seen in UDH lesions. Interestingly, the trend of these two immunostainings during breast carcinogenesis was diverse. 8-OHdG expression diminished significantly in invasive breast carcinomas compared to non-invasive lesions (P < 0.005 when set against non-invasive cohorts). Also within the same lesions, 8-OHdG expression was the most intensive in benign cells. Conversely, HNE immunostaining was strongest in invasive breast carcinomas (UDH versus invasive cohort, P = 0.015).4-hydroxy-2-nonenal as a marker of lipid peroxidation increases during breast carcinogenesis, reflecting the role of oxidative stress in the pathogenesis of breast cancer. However, 8-OHdG shows diminished levels in carcinomas, possibly resulting from the induction of DNA repair in these invasive lesions.
DOI: 10.1093/hmg/ddu311
2014
Cited 52 times
Common non-synonymous SNPs associated with breast cancer susceptibility: findings from the Breast Cancer Association Consortium
Candidate variant association studies have been largely unsuccessful in identifying common breast cancer susceptibility variants, although most studies have been underpowered to detect associations of a realistic magnitude. We assessed 41 common non-synonymous single-nucleotide polymorphisms (nsSNPs) for which evidence of association with breast cancer risk had been previously reported. Case-control data were combined from 38 studies of white European women (46 450 cases and 42 600 controls) and analyzed using unconditional logistic regression. Strong evidence of association was observed for three nsSNPs: ATXN7-K264R at 3p21 [rs1053338, per allele OR = 1.07, 95% confidence interval (CI) = 1.04-1.10, P = 2.9 × 10(-6)], AKAP9-M463I at 7q21 (rs6964587, OR = 1.05, 95% CI = 1.03-1.07, P = 1.7 × 10(-6)) and NEK10-L513S at 3p24 (rs10510592, OR = 1.10, 95% CI = 1.07-1.12, P = 5.1 × 10(-17)). The first two associations reached genome-wide statistical significance in a combined analysis of available data, including independent data from nine genome-wide association studies (GWASs): for ATXN7-K264R, OR = 1.07 (95% CI = 1.05-1.10, P = 1.0 × 10(-8)); for AKAP9-M463I, OR = 1.05 (95% CI = 1.04-1.07, P = 2.0 × 10(-10)). Further analysis of other common variants in these two regions suggested that intronic SNPs nearby are more strongly associated with disease risk. We have thus identified a novel susceptibility locus at 3p21, and confirmed previous suggestive evidence that rs6964587 at 7q21 is associated with risk. The third locus, rs10510592, is located in an established breast cancer susceptibility region; the association was substantially attenuated after adjustment for the known GWAS hit. Thus, each of the associated nsSNPs is likely to be a marker for another, non-coding, variant causally related to breast cancer risk. Further fine-mapping and functional studies are required to identify the underlying risk-modifying variants and the genes through which they act.
DOI: 10.1007/s10549-012-2181-7
2012
Cited 51 times
Low TLR9 expression defines an aggressive subtype of triple-negative breast cancer
DOI: 10.1093/jnci/djv081
2015
Cited 49 times
Identification of Novel Genetic Markers of Breast Cancer Survival
Survival after a diagnosis of breast cancer varies considerably between patients, and some of this variation may be because of germline genetic variation. We aimed to identify genetic markers associated with breast cancer-specific survival.We conducted a large meta-analysis of studies in populations of European ancestry, including 37954 patients with 2900 deaths from breast cancer. Each study had been genotyped for between 200000 and 900000 single nucleotide polymorphisms (SNPs) across the genome; genotypes for nine million common variants were imputed using a common reference panel from the 1000 Genomes Project. We also carried out subtype-specific analyses based on 6881 estrogen receptor (ER)-negative patients (920 events) and 23059 ER-positive patients (1333 events). All statistical tests were two-sided.We identified one new locus (rs2059614 at 11q24.2) associated with survival in ER-negative breast cancer cases (hazard ratio [HR] = 1.95, 95% confidence interval [CI] = 1.55 to 2.47, P = 1.91 x 10(-8)). Genotyping a subset of 2113 case patients, of which 300 were ER negative, provided supporting evidence for the quality of the imputation. The association in this set of case patients was stronger for the observed genotypes than for the imputed genotypes. A second locus (rs148760487 at 2q24.2) was associated at genome-wide statistical significance in initial analyses; the association was similar in ER-positive and ER-negative case patients. Here the results of genotyping suggested that the finding was less robust.This is currently the largest study investigating genetic variation associated with breast cancer survival. Our results have potential clinical implications, as they confirm that germline genotype can provide prognostic information in addition to standard tumor prognostic factors.
DOI: 10.1111/cen.13519
2017
Cited 49 times
Long‐term cardiovascular morbidity and mortality in patients treated for differentiated thyroid cancer
Thyroid hormone suppression therapy has been widely used in the treatment of thyroid cancer, but concerns have been raised about the cardiovascular risks of this treatment. The objective of this study was to evaluate long-term cardiovascular morbidity and mortality in patients treated for differentiated thyroid cancer (DTC) and to assess the effect of TSH suppression and radioiodine (RAI) treatment on the cardiovascular outcome.Retrospective cohort study.Patients (n = 901) treated for DTC between 1981 and 2002 at 2 Finnish University hospitals were compared with a randomly chosen reference group (n = 4485) matched for age, gender and the place of residence. Kaplan-Meier and Cox regression analyses were used to estimate the risk of morbidity or death due to different cardiovascular diseases (CVD) after the diagnosis of DTC.Morbidity due to any CVD (hazard ratio [HR] 1.16, 95% confidence interval [CI] 1.05-1.28) and due to all arrhythmias (HR 1.25, CI 1.06-1.48) and atrial fibrillation (AF) (HR 1.29, CI 1.06-1.57) was more frequent in the DTC patients than in the controls. The increased cardiovascular morbidity was confined to patients with a mean TSH level below 0.1 mU/L (HR 1.27, CI 1.03-1.58) and to those treated with RAI (HR 1.18, CI 1.05-1.31). Cardiovascular mortality, however, was lower among the patients than the controls (HR 0.73, CI 0.58-0.92), due to a lower mortality from coronary artery disease.Differentiated thyroid cancer patients have an increased CVD morbidity, which is mostly accountable to AF and to TSH suppression below 0.1 mU/L.
DOI: 10.1093/ije/dyx131
2017
Cited 47 times
Body mass index and breast cancer survival: a Mendelian randomization analysis
There is increasing evidence that elevated body mass index (BMI) is associated with reduced survival for women with breast cancer. However, the underlying reasons remain unclear. We conducted a Mendelian randomization analysis to investigate a possible causal role of BMI in survival from breast cancer.We used individual-level data from six large breast cancer case-cohorts including a total of 36 210 individuals (2475 events) of European ancestry. We created a BMI genetic risk score (GRS) based on genotypes at 94 known BMI-associated genetic variants. Association between the BMI genetic score and breast cancer survival was analysed by Cox regression for each study separately. Study-specific hazard ratios were pooled using fixed-effect meta-analysis.BMI genetic score was found to be associated with reduced breast cancer-specific survival for estrogen receptor (ER)-positive cases [hazard ratio (HR) = 1.11, per one-unit increment of GRS, 95% confidence interval (CI) 1.01-1.22, P = 0.03). We observed no association for ER-negative cases (HR = 1.00, per one-unit increment of GRS, 95% CI 0.89-1.13, P = 0.95).Our findings suggest a causal effect of increased BMI on reduced breast cancer survival for ER-positive breast cancer. There is no evidence of a causal effect of higher BMI on survival for ER-negative breast cancer cases.
DOI: 10.1200/jco.21.02054
2022
Cited 16 times
Adjuvant Capecitabine for Early Breast Cancer: 15-Year Overall Survival Results From a Randomized Trial
PURPOSE Few data are available regarding the influence of adjuvant capecitabine on long-term survival of patients with early breast cancer. METHODS The Finland Capecitabine Trial (FinXX) is a randomized, open-label, multicenter trial that evaluates integration of capecitabine to an adjuvant chemotherapy regimen containing a taxane and an anthracycline for the treatment of early breast cancer. Between January 27, 2004, and May 29, 2007, 1,500 patients with axillary node-positive or high-risk node-negative early breast cancer were accrued. The patients were randomly allocated to either TX-CEX, consisting of three cycles of docetaxel (T) plus capecitabine (X) followed by three cycles of cyclophosphamide, epirubicin, and capecitabine (CEX, 753 patients), or to T-CEF, consisting of three cycles of docetaxel followed by three cycles of cyclophosphamide, epirubicin, and fluorouracil (CEF, 747 patients). We performed a protocol-scheduled analysis of overall survival on the basis of approximately 15-year follow-up of the patients. RESULTS The data collection was locked on December 31, 2020. By this date, the median follow-up time of the patients alive was 15.3 years (interquartile range, 14.5-16.1 years) in the TX-CEX group and 15.4 years (interquartile range, 14.8-16.0 years) in the T-CEF group. Patients assigned to TX-CEX survived longer than those assigned to T-CEF (hazard ratio 0.81; 95% CI, 0.66 to 0.99; P = .037). The 15-year survival rate was 77.6% in the TX-CEX group and 73.3% in the T-CEF group. In exploratory subgroup analyses, patients with estrogen receptor–negative cancer and those with triple-negative cancer treated with TX-CEX tended to live longer than those treated with T-CEF. CONCLUSION Addition of capecitabine to a chemotherapy regimen that contained docetaxel, epirubicin, and cyclophosphamide prolonged the survival of patients with early breast cancer.
DOI: 10.1016/s1470-2045(09)70307-9
2009
Cited 60 times
Adjuvant capecitabine in combination with docetaxel and cyclophosphamide plus epirubicin for breast cancer: an open-label, randomised controlled trial
Standard adjuvant chemotherapy regimens for patients with moderate-to-high-risk early breast cancer typically contain a taxane, an anthracycline, and cyclophosphamide. We aimed to investigate whether integration of capecitabine into such a regimen enhances outcome.In this open-label trial, we randomly assigned (centrally by computer; stratified by node status, HER2 status, and centre) 1500 women with axillary node-positive or high-risk node-negative breast cancer to either three cycles of capecitabine and docetaxel followed by three cycles of cyclophosphamide, epirubicin, and capecitabine (capecitabine group, n=753), or to three cycles of docetaxel followed by three cycles of cyclophosphamide, epirubicin, and fluorouracil (control group, n=747). The primary endpoint was recurrence-free survival. A planned interim analysis was done after 3 years' median follow-up. Efficacy analyses were by modified intention to treat. The study is registered with ClinicalTrials.gov, number NCT00114816.Two patients in each group were excluded from efficacy analyses because of withdrawal of consent or distant metastases. After a median follow-up of 35 months (IQR 25.5-43.6), recurrence-free survival at 3 years was better with the capecitabine regimen than with control (93%vs 89%; hazard ratio 0.66, 95% CI 0.47-0.94; p=0.020). The capecitabine regimen was associated with more cases of grade 3 or 4 diarrhoea (46/740 [6%] vs 25/741 [3%]) and hand-foot syndrome (83/741 [11%] vs 2/741 [<1%]) and the control regimen with more occurrences of grade 3 or 4 neutropenia (368/375 [98%] vs 325/378 [86%]) and febrile neutropenia (65/741 [9%] vs 33/742 [4%]). More patients discontinued planned treatment in the capecitabine group than in the control group (178/744 [24%] vs 23/741 [3%]). Four patients in the capecitabine group and two in the control group died from potentially treatment-related causes.The capecitabine-containing chemotherapy regimen reduced breast cancer recurrence compared with a control schedule of standard agents. Capecitabine administration was frequently discontinued because of adverse effects.Roche, Sanofi-Aventis, AstraZeneca, Cancer Society of Finland.
DOI: 10.1002/pros.21115
2010
Cited 54 times
Expression of toll-like receptor-9 is increased in poorly differentiated prostate tumors
Abstract BACKGROUND Toll‐like receptor‐9 (TLR9) is a cellular receptor for bacterial and vertebrate DNA. In addition to cells of the immune system, it is also expressed in various human cancer cell lines, including prostate cancer. We demonstrated previously that synthetic TLR9 ligands induce matrix metalloproteinase‐13‐mediated invasion in TLR9‐expressing prostate cancer cells in vitro. Other studies have suggested possible sex steroid regulation of the function of the various TLRs. The role of TLR9 in the pathophysiology of prostate or any cancer is, however, unknown. METHODS Expression of TLR9, androgen receptor (AR), or the estrogen receptors α (ERα) and β (ERβ) were studied with immunohistochemistry in prostate cancer (n = 62) and benign prostatic hyperplasia (n = 45) specimens. TLR9 staining scores were compared with tumor stage, Gleason score, prostate‐specific antigen (PSA) concentrations before tissue sampling and with the staining scores of AR, ERα, and ERβ. RESULTS TLR9 expression was statistically significantly increased in prostate cancer epithelium and stroma, as compared with the same cellular compartments in benign hyperplasia. Significantly increased ( P = 0.04) TLR9 expression was detected in cancers with high Gleason score (&gt;7, n = 23), as compared with lower Gleason scores (≤7, n = 39). No statistically significant associations were detected between TLR9 expression scores and PSA concentrations or tumor staging. Prostate adenocarcinoma cells were all positive for TLR9, AR, and ERβ but negative for ERα expression. In cancer stroma cells, increased TLR9 expression was associated with increased ERα expression. CONCLUSIONS Expression of TLR9 is increased in prostate cancer specimens, especially in the most poorly differentiated forms. Prostate 70: 817–824, 2010. © 2010 Wiley‐Liss, Inc.
DOI: 10.1158/1078-0432.ccr-14-1887
2015
Cited 40 times
<i>KEAP1</i> Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
Defective oxidative stress response may increase cancer susceptibility. In tumors, these rescue mechanisms may cause chemo- and radioresistance impacting patient outcome. We previously showed that genetic variation in the nuclear factor erythroid 2-related factor 2 (NFE2L2) is associated with breast cancer risk and prognosis. Here we further studied this pathway by investigating Kelch-like ECH-associated protein 1 (KEAP1).Five tagging SNPs in the KEAP1 gene were genotyped in 996 breast cancer cases and 880 controls from two Finnish case-control sets. KEAP1 protein expression was studied in 373 invasive breast cancer tumors.rs34197572 genotype TT was associated with increased risk of breast cancer in the KBCP samples [P = 1.8×10(-4); OR, 7.314; confidence interval (CI), 2.185-24.478]. rs11085735 allele A was associated with lower KEAP1 protein expression (P = 0.040; OR,= 3.545) and high nuclear NRF2 expression (P = 0.009; OR, 2.445) and worse survival in all invasive cases (P = 0.023; HR, 1.634). When including treatment data, rs11085735 was associated with recurrence-free survival (RFS; P = 0.020; HR, 1.545) and breast cancer-specific survival (P = 0.016; HR, 1.683) and rs34197572 with overall survival (P = 0.045; HR, 1.304). rs11085735 associated with RFS also among tamoxifen-treated cases (P = 0.003; HR, 3.517). Among radiotherapy-treated cases, overall survival was associated with rs34197572 (P = 0.018; HR, 1.486) and rs8113472 (P = 0.025; HR, 1.455). RFS was associated with rs9676881 (P = 0.024; HR, 1.452) and rs1048290 (P = 0.020; HR, 1.468) among all invasive cases and among estrogen receptor (ER)-positive tamoxifen-treated cases (P = 0.018; HR, 2.407 and P = 0.015; HR, 2.476, respectively).The present findings suggest that the investigated SNPs have effects related to oxidative stress induced by cancer treatment, supporting involvement of the NRF2/KEAP1 pathway in breast cancer susceptibility and patient outcome.
DOI: 10.1080/2162402x.2014.1002726
2015
Cited 39 times
Tumor infiltrating CD8<sup>+</sup>T lymphocyte count is independent of tumor TLR9 status in treatment naïve triple negative breast cancer and renal cell carcinoma
Toll-like receptor 9 (TLR9) is a cellular DNA-receptor of the innate immune system that is widely expressed in cancers. We demonstrated that low tumor TLR9 expression predicts poor disease-specific survival in triple negative breast cancer (TNBC) and renal cell carcinoma (RCC). We hypothesized that this is because TLR9 expression affects tumor immunophenotype. To begin to test this, we compared the number of tumor infiltrating CD8+ T lymphocytes with TLR9 expression in treatment naïve breast cancer (n = 197) and RCC (n = 94) cohorts with known TLR9 expression status. CD8+ T lymphocyte counts were assayed with image analysis after immunohistochemistry (IHC). Tumor TLR9 expression was not correlated with CD8+ T cell counts in breast cancer or RCC. CD8+ T cell counts were significantly associated with tumor proliferation index in TNBC, but not in non-TNBC. CD8+ T cell counts were also significantly associated with tumor grade in non-TNBC, but not in TNBC. In RCC, CD8+ T cell counts were significantly associated with tumor stage. CD8+ T cell counts were significantly associated with prognosis in TNBC and RCC, but the presence of CD8+ T cells in these tumors had opposite effects on disease-specific survival: High CD8+ counts were associated with better prognosis in TNBC and worse prognosis in RCC. Among TNBC patients, those with low tumor TLR9 and low CD8+ T cell counts had the poorest prognosis (log-rank p = 0.0002 vs. high tumor TLR9 and high CD8+ T cell count). In conclusion, pre-treatment tumor TLR9 status is not associated with tumor infiltrating CD8+ T lymphocytes in TNBC or RCC. The combination of TLR9 and CD8+ TIL count might be a novel composite prognostic marker in TNBC.
DOI: 10.1016/j.ajhg.2015.05.002
2015
Cited 36 times
Polymorphisms in a Putative Enhancer at the 10q21.2 Breast Cancer Risk Locus Regulate NRBF2 Expression
Genome-wide association studies have identified SNPs near ZNF365 at 10q21.2 that are associated with both breast cancer risk and mammographic density. To identify the most likely causal SNPs, we fine mapped the association signal by genotyping 428 SNPs across the region in 89,050 European and 12,893 Asian case and control subjects from the Breast Cancer Association Consortium. We identified four independent sets of correlated, highly trait-associated variants (iCHAVs), three of which were located within ZNF365. The most strongly risk-associated SNP, rs10995201 in iCHAV1, showed clear evidence of association with both estrogen receptor (ER)-positive (OR = 0.85 [0.82–0.88]) and ER-negative (OR = 0.87 [0.82–0.91]) disease, and was also the SNP most strongly associated with percent mammographic density. iCHAV2 (lead SNP, chr10: 64,258,684:D) and iCHAV3 (lead SNP, rs7922449) were also associated with ER-positive (OR = 0.93 [0.91–0.95] and OR = 1.06 [1.03–1.09]) and ER-negative (OR = 0.95 [0.91–0.98] and OR = 1.08 [1.04–1.13]) disease. There was weaker evidence for iCHAV4, located 5′ of ADO, associated only with ER-positive breast cancer (OR = 0.93 [0.90–0.96]). We found 12, 17, 18, and 2 candidate causal SNPs for breast cancer in iCHAVs 1–4, respectively. Chromosome conformation capture analysis showed that iCHAV2 interacts with the ZNF365 and NRBF2 (more than 600 kb away) promoters in normal and cancerous breast epithelial cells. Luciferase assays did not identify SNPs that affect transactivation of ZNF365, but identified a protective haplotype in iCHAV2, associated with silencing of the NRBF2 promoter, implicating this gene in the etiology of breast cancer. Genome-wide association studies have identified SNPs near ZNF365 at 10q21.2 that are associated with both breast cancer risk and mammographic density. To identify the most likely causal SNPs, we fine mapped the association signal by genotyping 428 SNPs across the region in 89,050 European and 12,893 Asian case and control subjects from the Breast Cancer Association Consortium. We identified four independent sets of correlated, highly trait-associated variants (iCHAVs), three of which were located within ZNF365. The most strongly risk-associated SNP, rs10995201 in iCHAV1, showed clear evidence of association with both estrogen receptor (ER)-positive (OR = 0.85 [0.82–0.88]) and ER-negative (OR = 0.87 [0.82–0.91]) disease, and was also the SNP most strongly associated with percent mammographic density. iCHAV2 (lead SNP, chr10: 64,258,684:D) and iCHAV3 (lead SNP, rs7922449) were also associated with ER-positive (OR = 0.93 [0.91–0.95] and OR = 1.06 [1.03–1.09]) and ER-negative (OR = 0.95 [0.91–0.98] and OR = 1.08 [1.04–1.13]) disease. There was weaker evidence for iCHAV4, located 5′ of ADO, associated only with ER-positive breast cancer (OR = 0.93 [0.90–0.96]). We found 12, 17, 18, and 2 candidate causal SNPs for breast cancer in iCHAVs 1–4, respectively. Chromosome conformation capture analysis showed that iCHAV2 interacts with the ZNF365 and NRBF2 (more than 600 kb away) promoters in normal and cancerous breast epithelial cells. Luciferase assays did not identify SNPs that affect transactivation of ZNF365, but identified a protective haplotype in iCHAV2, associated with silencing of the NRBF2 promoter, implicating this gene in the etiology of breast cancer.
DOI: 10.1159/000151602
2008
Cited 44 times
Toll-Like Receptor-9 Expression Is Inversely Correlated with Estrogen Receptor Status in Breast Cancer
Toll-like receptor 9 (TLR9) recognizes microbial and vertebrate DNA. We previously demonstrated TLR9 expression in human breast cancer cell lines and showed that TLR9 ligands stimulate their in vitro invasion. The aim of this study was to characterize TLR9 expression in clinical breast cancer specimens. Immunohistochemical staining intensity was compared with known baseline prognostic factors and distant metastasis-free survival. TLR9 expression was detected in 98% of the tumors studied (n = 141). The mean TLR9 staining intensity was higher in ER– than in the highly ER+ breast cancers (p = 0.039). High-grade tumors had significantly increased TLR9 expression (p = 0.027) compared with lower-grade tumors. The highest TLR9 expression was detected in the mucinous and the lowest in the tubular breast cancers (p = 0.034). Distant metastasis-free survival was higher in the lower TLR9-expressing half of the cohort than in the higher TLR9-expressing group (p = 0.118). TLR9 expression did not correlate with menopausal, PgR or Her2 status, patient age, tumor proliferative or invasive characteristics.
DOI: 10.3892/ol.2013.1204
2013
Cited 35 times
Expression of Toll-like receptor-9 is associated with poor progression-free survival in prostate cancer
Toll-like receptor-9 (TLR9) is a member of the innate immune system and recognizes bacterial and vertebrate DNA in cells. In addition to being expressed in cells of the immune system, it is widely expressed in various types of human cancer, including prostate cancer. We have previously demonstrated that synthetic TLR9 ligands induce invasion in TLR9-expressing prostate cancer cells in vitro. However, the role of TLR9 in the pathophysiology of prostate cancer is unclear. The expression of TLR9 in radical prostatectomy samples (n=186) was studied using immunohistochemistry. TLR9 staining scores were compared with tumor stage, Gleason score and prostate-specific antigen (PSA) concentration prior to treatment and progression-free survival. Results revealed that 124 (66.7%) of the tumors were strongly positive, 59 (31.7%) were weakly positive and 3 (1.6%) were negative, for cytoplasmic TLR9 immunostaining in cancer cells. There was no significant association between cytoplasmic TLR9 expression and distributions of pT-class, prostatectomy sample margin status, Gleason score and preoperative PSA value. Prostate cancer-specific progression-free survival was significantly longer for patients whose tumors were graded as negative for cytoplasmic TLR9 expression, as compared with patients whose tumors were strongly immunopositive for cytoplasmic TLR9 (P=0.009). In the Cox regression analysis, high TLR9 expression was an independent marker of poor prognosis in prostate cancer. Expression of TLR9 is associated with poor progression-free survival in prostate cancer patients who were treated by radical prostatectomy with curative intent.
DOI: 10.1002/humu.22089
2012
Cited 35 times
11q13 is a susceptibility locus for hormone receptor positive breast cancer
Human MutationVolume 33, Issue 7 p. 1123-1132 Research Article 11q13 is a susceptibility locus for hormone receptor positive breast cancer† Diether Lambrechts, Diether Lambrechts Vesalius Research Center (VRC), VIB, KU Leuven, Leuven, Belgium Contributed equally to this work. Writing Group: Diether Lambrechts, Therese Truong, Christina Justenhoven, Douglas F. Easton, Pascal Guénel, and Hiltrud BrauchSearch for more papers by this authorTherese Truong, Therese Truong Environmental Epidemiology of Cancer, CESP Centre for Research in Epidemiology and Population Health, U1018, Inserm, F-94807, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, France Contributed equally to this work. Writing Group: Diether Lambrechts, Therese Truong, Christina Justenhoven, Douglas F. Easton, Pascal Guénel, and Hiltrud BrauchSearch for more papers by this authorChristina Justenhoven, Christina Justenhoven Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart; University Tübingen, Tübingen, Germany Contributed equally to this work. Writing Group: Diether Lambrechts, Therese Truong, Christina Justenhoven, Douglas F. Easton, Pascal Guénel, and Hiltrud BrauchSearch for more papers by this authorManjeet K. Humphreys, Manjeet K. Humphreys Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United KingdomSearch for more papers by this authorJean Wang, Jean Wang Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United KingdomSearch for more papers by this authorJohn L. Hopper, John L. Hopper Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, Melbourne, AustraliaSearch for more papers by this authorGillian S. Dite, Gillian S. Dite Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, Melbourne, AustraliaSearch for more papers by this authorCarmel Apicella, Carmel Apicella Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, Melbourne, AustraliaSearch for more papers by this authorMelissa C. Southey, Melissa C. Southey Department of Pathology, The University of Melbourne, Melbourne, AustraliaSearch for more papers by this authorMarjanka K. Schmidt, Marjanka K. Schmidt Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The NetherlandsSearch for more papers by this authorAnnegien Broeks, Annegien Broeks Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The NetherlandsSearch for more papers by this authorSten Cornelissen, Sten Cornelissen Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The NetherlandsSearch for more papers by this authorRichard van Hien, Richard van Hien Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The NetherlandsSearch for more papers by this authorElinor Sawyer, Elinor Sawyer Division of Cancer Studies, NIHR Comprehensive Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust in partnership with King's College London, London, United KingdomSearch for more papers by this authorIan Tomlinson, Ian Tomlinson Welcome Trust Centre for Human Genetics and Oxford Biomedical Research Centre, University of Oxford, United KingdomSearch for more papers by this authorMichael Kerin, Michael Kerin Clinical Science Institute. University Hospital Galway, Galway, IrelandSearch for more papers by this authorNicola Miller, Nicola Miller Clinical Science Institute. University Hospital Galway, Galway, IrelandSearch for more papers by this authorRoger L. Milne, Roger L. Milne Genetic and Molecular Epidemiology Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, SpainSearch for more papers by this authorM. Pilar Zamora, M. Pilar Zamora Servicio de Oncología Médica, Hospital Universitario La Paz, Madrid, SpainSearch for more papers by this authorJosé Ignacio Arias Pérez, José Ignacio Arias Pérez Servicio de Cirugía General y Especialidades, Hospital Monte Naranco, Oviedo, SpainSearch for more papers by this authorJavier Benítez, Javier Benítez Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, SpainSearch for more papers by this authorUte Hamann, Ute Hamann Molecular Genetics of Breast Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, GermanySearch for more papers by this authorYon-Dschun Ko, Yon-Dschun Ko Department of Internal Medicine, Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn, GermanySearch for more papers by this authorThomas Brüning, Thomas Brüning Institute for Prevention and Occupational Medicine of the German Social Accident Insurance (IPA), Bochum, GermanySearch for more papers by this authorThe GENICA Network, The GENICA Network Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart; University Tübingen; Molecular Genetics of Breast Cancer; Deutsches Krebsforschungszentrum (DKFZ), Heidelberg; Department of Internal Medicine, Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn; Institute of Pathology, Medical Faculty of the University of Bonn, Bonn; Institute for Prevention and Occupational Medicine of the German Social Accident Insurance (IPA), Bochum; Institute and Outpatient Clinic of Occupational Medicine, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, GermanySearch for more papers by this authorJenny Chang-Claude, Jenny Chang-Claude Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, GermanySearch for more papers by this authorUrsel Eilber, Ursel Eilber Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, GermanySearch for more papers by this authorRebecca Hein, Rebecca Hein Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, GermanySearch for more papers by this authorStefan Nickels, Stefan Nickels Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, GermanySearch for more papers by this authorDieter Flesch-Janys, Dieter Flesch-Janys Institute for Medical Biometrics and Epidemiology, University Clinic Hamburg-Eppendorf, Hamburg, GermanySearch for more papers by this authorShan Wang-Gohrke, Shan Wang-Gohrke Department of Obstetrics and Gynecology, University of Ulm, Ulm, GermanySearch for more papers by this authorEsther M. John, Esther M. John Cancer Prevention Institute of California, Fremont, California and Stanford University School of Medicine and Stanford Cancer Institute, Stanford, CaliforniaSearch for more papers by this authorAlexander Miron, Alexander Miron Dana-Farber Cancer Institute, Boston, MassachusettsSearch for more papers by this authorRobert Winqvist, Robert Winqvist Laboratory of Cancer Genetics, Department of Clinical Genetics and Biocenter Oulu, University of Oulu, Oulu, FinlandSearch for more papers by this authorKatri Pylkäs, Katri Pylkäs Laboratory of Cancer Genetics, Department of Clinical Genetics and Biocenter Oulu, University of Oulu, Oulu, FinlandSearch for more papers by this authorArja Jukkola-Vuorinen, Arja Jukkola-Vuorinen Department of Oncology, Oulu University Hospital, University of Oulu, Oulu, FinlandSearch for more papers by this authorMervi Grip, Mervi Grip Department of Surgery, Oulu University Hospital, University of Oulu, Oulu, FinlandSearch for more papers by this authorGeorgia Chenevix-Trench, Georgia Chenevix-Trench Queensland Institute of Medical Research, Brisbane, AustraliaSearch for more papers by this authorJonathan Beesley, Jonathan Beesley Queensland Institute of Medical Research, Brisbane, AustraliaSearch for more papers by this authorXiaoqing Chen, Xiaoqing Chen Queensland Institute of Medical Research, Brisbane, AustraliaSearch for more papers by this authorkConFab Investigators, kConFab Investigators Peter MacCallum Cancer Center, Melbourne, AustraliaSearch for more papers by this authorAustralian Ovarian Cancer Study Group, Australian Ovarian Cancer Study Group Peter MacCallum Cancer Center, Melbourne, AustraliaSearch for more papers by this authorFlorence Menegaux, Florence Menegaux Environmental Epidemiology of Cancer, CESP Centre for Research in Epidemiology and Population Health, U1018, Inserm, F-94807, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, FranceSearch for more papers by this authorEmilie Cordina-Duverger, Emilie Cordina-Duverger Environmental Epidemiology of Cancer, CESP Centre for Research in Epidemiology and Population Health, U1018, Inserm, F-94807, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, FranceSearch for more papers by this authorChen-Yang Shen, Chen-Yang Shen Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Taiwan Biobank, Taipei, TaiwanSearch for more papers by this authorJyh-Cherng Yu, Jyh-Cherng Yu Department of Surgery, Tri-Service General Hospital, Taipei, TaiwanSearch for more papers by this authorPei-Ei Wu, Pei-Ei Wu Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Taiwan Biobank, Taipei, TaiwanSearch for more papers by this authorMing-Feng Hou, Ming-Feng Hou Cancer Center and Department of Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, TaiwanSearch for more papers by this authorIrene L. Andrulis, Irene L. Andrulis Ontario Cancer Genetics Network, Cancer Care Ontario; Fred A. Litwin Center for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, CanadaSearch for more papers by this authorTeresa Selander, Teresa Selander Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, CanadaSearch for more papers by this authorGord Glendon, Gord Glendon Ontario Cancer Genetics Network, Cancer Care Ontario, Toronto, Ontario, CanadaSearch for more papers by this authorAnna Marie Mulligan, Anna Marie Mulligan Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, CanadaSearch for more papers by this authorHoda Anton-Culver, Hoda Anton-Culver Department of Epidemiology, University of California Irvine, Irvine, CaliforniaSearch for more papers by this authorArgyrios Ziogas, Argyrios Ziogas Department of Epidemiology, University of California Irvine, Irvine, CaliforniaSearch for more papers by this authorKenneth R. Muir, Kenneth R. Muir Health Sciences Research Institute, Warwick Medical School, Warwick University, Coventry, United KingdomSearch for more papers by this authorArtitaya Lophatananon, Artitaya Lophatananon Health Sciences Research Institute, Warwick Medical School, Warwick University, Coventry, United KingdomSearch for more papers by this authorSuthee Rattanamongkongul, Suthee Rattanamongkongul Department of Preventive Medicine, Srinakhrainwirot University, Ongkharak, Nakhon Nayok, ThailandSearch for more papers by this authorPuttisak Puttawibul, Puttisak Puttawibul Department of Surgery, Medical School, Prince Songkla University, Songkla, ThailandSearch for more papers by this authorMichael Jones, Michael Jones Section of Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United KingdomSearch for more papers by this authorNicholas Orr, Nicholas Orr Breakthrough Breast Cancer Research Centre, Chester Beatty Laboratories, The Institute of Cancer Research, London, United KingdomSearch for more papers by this authorAlan Ashworth, Alan Ashworth Breakthrough Breast Cancer Research Centre, Chester Beatty Laboratories, The Institute of Cancer Research, London, United KingdomSearch for more papers by this authorAnthony Swerdlow, Anthony Swerdlow Section of Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United KingdomSearch for more papers by this authorGianluca Severi, Gianluca Severi Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, AustraliaSearch for more papers by this authorLaura Baglietto, Laura Baglietto Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, AustraliaSearch for more papers by this authorGraham Giles, Graham Giles Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, Melbourne, Australia Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, AustraliaSearch for more papers by this authorMelissa Southey, Melissa Southey Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, AustraliaSearch for more papers by this authorFederik Marmé, Federik Marmé National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, GermanySearch for more papers by this authorAndreas Schneeweiss, Andreas Schneeweiss National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, GermanySearch for more papers by this authorChristof Sohn, Christof Sohn Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, GermanySearch for more papers by this authorBarbara Burwinkel, Barbara Burwinkel Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany Molecular Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, GermanySearch for more papers by this authorBetul T. Yesilyurt, Betul T. Yesilyurt Vesalius Research Center (VRC), VIB, KU Leuven, Leuven, BelgiumSearch for more papers by this authorPatrick Neven, Patrick Neven Multidisciplinary Breast Center, University Hospital Gasthuisberg, Leuven, BelgiumSearch for more papers by this authorRobert Paridaens, Robert Paridaens Multidisciplinary Breast Center, University Hospital Gasthuisberg, Leuven, BelgiumSearch for more papers by this authorHans Wildiers, Hans Wildiers Multidisciplinary Breast Center, University Hospital Gasthuisberg, Leuven, BelgiumSearch for more papers by this authorHermann Brenner, Hermann Brenner Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, GermanySearch for more papers by this authorHeiko Müller, Heiko Müller Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, GermanySearch for more papers by this authorVolker Arndt, Volker Arndt Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, GermanySearch for more papers by this authorChrista Stegmaier, Christa Stegmaier Saarland Cancer Registry, Saarbrücken, GermanySearch for more papers by this authorAlfons Meindl, Alfons Meindl Division of Gynaecology and Obstetrics, Technical University of Munich, Munich, GermanySearch for more papers by this authorSarah Schott, Sarah Schott Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, GermanySearch for more papers by this authorClaus R. Bartram, Claus R. Bartram Institute of Human Genetics, University of Heidelberg, Heidelberg, GermanySearch for more papers by this authorRita K. Schmutzler, Rita K. Schmutzler Division of Molecular Gyneco-Oncology, Department of Gynaecology and Obstetrics, Center of Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Cologne, GermanySearch for more papers by this authorAngela Cox, Angela Cox Institute for Cancer Studies, Department of Oncology, University of Sheffield, United KingdomSearch for more papers by this authorIan W. Brock, Ian W. Brock Institute for Cancer Studies, Department of Oncology, University of Sheffield, United KingdomSearch for more papers by this authorGraeme Elliott, Graeme Elliott Institute for Cancer Studies, Department of Oncology, University of Sheffield, United KingdomSearch for more papers by this authorSimon S. Cross, Simon S. Cross Academic Unit of Pathology, Department of Neuroscience, University of Sheffield, United KingdomSearch for more papers by this authorPeter A. Fasching, Peter A. Fasching University Breast Center, Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany; David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, CaliforniaSearch for more papers by this authorRuediger Schulz-Wendtland, Ruediger Schulz-Wendtland Institute of Diagnostic Radiology, University Hospital Erlangen, Erlangen, GermanySearch for more papers by this authorArif B. Ekici, Arif B. Ekici Institute of Human Genetics, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, GermanySearch for more papers by this authorMatthias W. Beckmann, Matthias W. Beckmann University Breast Center, Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, GermanySearch for more papers by this authorOlivia Fletcher, Olivia Fletcher Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United KingdomSearch for more papers by this authorNichola Johnson, Nichola Johnson Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United KingdomSearch for more papers by this authorIsabel dos Santos Silva, Isabel dos Santos Silva London School of Hygiene and Tropical Medicine, London, United KingdomSearch for more papers by this authorJulian Peto, Julian Peto London School of Hygiene and Tropical Medicine, London, United KingdomSearch for more papers by this authorHeli Nevanlinna, Heli Nevanlinna Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Biomedicum Helsinki, Helsinki, FinlandSearch for more papers by this authorTaru A. Muranen, Taru A. Muranen Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Biomedicum Helsinki, Helsinki, FinlandSearch for more papers by this authorKristiina Aittomäki, Kristiina Aittomäki Department of Clinical Genetics, Helsinki University Central Hospital, Helsinki, FinlandSearch for more papers by this authorCarl Blomqvist, Carl Blomqvist Department of Oncology, Helsinki University Central Hospital, Helsinki, FinlandSearch for more papers by this authorThilo Dörk, Thilo Dörk Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorPeter Schürmann, Peter Schürmann Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorMichael Bremer, Michael Bremer Department of Radiation Oncology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorPeter Hillemanns, Peter Hillemanns Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorNatalia V. Bogdanova, Natalia V. Bogdanova Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany Department of Radiation Oncology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorNatalia N. Antonenkova, Natalia N. Antonenkova N.N. Alexandrov Research Institute of Oncology and Medical Radiology, Minsk, BelarusSearch for more papers by this authorYuri I. Rogov, Yuri I. Rogov N.N. Alexandrov Research Institute of Oncology and Medical Radiology, Minsk, BelarusSearch for more papers by this authorJohann H. Karstens, Johann H. Karstens Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, GermanySearch for more papers by this authorElza Khusnutdinova, Elza Khusnutdinova Institute of Biochemistry and Genetics, Ufa Scientific Center of Russian Academy of Sciences, Ufa, RussiaSearch for more papers by this authorMarina Bermisheva, Marina Bermisheva Institute of Biochemistry and Genetics, Ufa Scientific Center of Russian Academy of Sciences, Ufa, RussiaSearch for more papers by this authorDarya Prokofieva, Darya Prokofieva Institute of Biochemistry and Genetics, Ufa Scientific Center of Russian Academy of Sciences, Ufa, RussiaSearch for more papers by this authorShamil Gancev, Shamil Gancev Bashkirian Medical University, Ufa, RussiaSearch for more papers by this authorAnna Jakubowska, Anna Jakubowska Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, PolandSearch for more papers by this authorJan Lubinski, Jan Lubinski Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, PolandSearch for more papers by this authorKatarzyna Jaworska, Katarzyna Jaworska Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, PolandSearch for more papers by this authorKatarzyna Durda, Katarzyna Durda Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, PolandSearch for more papers by this authorBørge G. Nordestgaard, Børge G. Nordestgaard Copenhagen General Population Study and Department of Clinical Biochemistry, Herlev University Hospital, University of Copenhagen, Copenhagen, DenmarkSearch for more papers by this authorStig E. Bojesen, Stig E. Bojesen Copenhagen General Population Study and Department of Clinical Biochemistry, Herlev University Hospital, University of Copenhagen, Copenhagen, DenmarkSearch for more papers by this authorCharlotte Lanng, Charlotte Lanng Department of Breast Surgery, Herlev University Hospital, University of Copenhagen, Copenhagen, DenmarkSearch for more papers by this authorArto Mannermaa, Arto Mannermaa School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland; Biocenter Kuopio and Department of Clinical Pathology, Kuopio University Hospital, Kuopio, FinlandSearch for more papers by this authorVesa Kataja, Vesa Kataja School of Medicine, Institute of Clinical Medicine, Oncology, University of Eastern Finland; Biocenter Kuopio and Department of Oncology, Kuopio University Hospital, Kuopio, FinlandSearch for more papers by this authorVeli-Matti Kosma, Veli-Matti Kosma School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland; Biocenter Kuopio and Department of Clinical Pathology, Kuopio University Hospital, Kuopio, FinlandSearch for more papers by this authorJaana M. Hartikainen, Jaana M. Hartikainen School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland; Biocenter Kuopio and Department of Clinical Pathology, Kuopio University Hospital, Kuopio, FinlandSearch for more papers by this authorPaolo Radice, Paolo Radice Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predicted Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT); IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, ItalySearch for more papers by this authorPaolo Peterlongo, Paolo Peterlongo Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predicted Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT); IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, ItalySearch for more papers by this authorSiranoush Manoukian, Siranoush Manoukian Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, ItalySearch for more papers by this authorLoris Bernard, Loris Bernard Department of Experimental Oncology, Istituto Europeo di Oncologia (IEO); Consortium for Genomics Technology (Cogentech) Milan, ItalySearch for more papers by this authorFergus J. Couch, Fergus J. Couch Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MinnesotaSearch for more papers by this authorJanet E. Olson, Janet E. Olson Department of Health Sciences Research, Mayo Clinic, Rochester, MinnesotaSearch for more papers by this authorXianshu Wang, Xianshu Wang Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MinnesotaSearch for more papers by this authorZachary Fredericksen, Zachary Fredericksen Department of Health Sciences Research, Mayo Clinic, Rochester, MinnesotaSearch for more papers by this authorGrethe Grenaker Alnæs, Grethe Grenaker Alnæs Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, NorwaySearch for more papers by this authorVessela Kristensen, Vessela Kristensen Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway Faculty of Medicine (Faculty Division Ahus), University of Oslo, NorwaySearch for more papers by this authorAnne-Lise Børresen-Dale, Anne-Lise Børresen-Dale Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway Faculty of Medicine (Faculty Division Ahus), University of Oslo, NorwaySearch for more papers by this authorPeter Devilee, Peter Devilee Department of Human Genetics, and Department of Pathology, Leiden University Medical Centre, Leiden, The NetherlandsSearch for more papers by this authorRobert A.E.M. Tollenaar, Robert A.E.M. Tollenaar Department of Surgery, Leiden University Medical Center, Leiden, The NetherlandsSearch for more papers by this authorCaroline M. Seynaeve, Caroline M. Seynaeve Department of Medical Oncology, Rotterdam Family Cancer Clinic, Erasmus MC-Daniel den Hoed Cancer Center, Rotterdam, The NetherlandsSearch for more papers by this authorMaartje J. Hooning, Maartje J. Hooning Department of Medical Oncology, Rotterdam Family Cancer Clinic, Erasmus MC-Daniel den Hoed Cancer Center, Rotterdam, The NetherlandsSearch for more papers by this authorMontserrat García-Closas, Montserrat García-Closas Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland; Division of Genetics and Epidemiology, Institute of Cancer Research and Breakthrough Breast Cancer Research Centre, London, United KingdomSearch for more papers by this authorStephen J. Chanock, Stephen J. Chanock Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MarylandSearch for more papers by this authorJolanta Lissowska, Jolanta Lissowska Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, PolandSearch for more papers by this authorMark E. Sherman, Mark E. Sherman Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MarylandSearch for more papers by this authorPer Hall, Per Hall Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, SwedenSearch for more papers by this authorJianjun Liu, Jianjun Liu Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, SwedenSearch for more papers by this authorKamila Czene, Kamila Czene Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, SwedenSearch for more papers by this authorDaehee Kang, Daehee Kang Seoul National University College of Medicine, Seoul, Republic of KoreaSearch for more papers by this authorKeun-Young Yoo, Keun-Young Yoo Seoul National University College of Medicine, Seoul, Republic of KoreaSearch for more papers by this authorDong-Young Noh, Dong-Young Noh Seoul National University College of Medicine, Seoul, Republic of KoreaSearch for more papers by this authorAnnika Lindblom, Annika Lindblom Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SwedenSearch for more papers by this authorSara Margolin, Sara Margolin Department of Oncology Pathology, Karolinska Institutet, Stockholm, SwedenSearch for more papers by this authorAlison M. Dunning, Alison M. Dunning Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United KingdomSearch for more papers by this authorPaul D.P. Pharoah, Paul D.P. Pharoah Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United KingdomSearch for more papers by this authorDouglas F. Easton, Douglas F. Easton Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United KingdomSearch for more papers by this authorPascal Guénel, Pascal Guénel Environmental Epidemiology of Cancer, CESP Centre for Research in Epidemiology and Population Health, U1018, Inserm, F-94807, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, FranceSearch for more papers by this authorHiltrud Brauch, Corresponding Author Hiltrud Brauch [email protected] Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart; University Tübingen, Tübingen, GermanyDr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, Germany.Search for more papers by this author Diether Lambrechts, Diether Lambrechts Vesalius Research Center (VRC), VIB, KU Leuven, Leuven, Belgium Contributed equally to this work. Writing Group: Diether Lambrechts, Therese Truong, Christina Justenhoven, Douglas F. Easton, Pascal Guénel, and Hiltrud BrauchSearch for more papers by this authorTherese Truong, Therese Truong Environmental Epidemiology of Cancer, CESP Centre for Research in Epidemiology and Population Health, U1018, Inserm, F-94807, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, France Contributed equally to this work. Writing Group: Diether Lambrechts, Therese Truong, Christina Ju
DOI: 10.1093/hmg/ddt581
2013
Cited 33 times
A large-scale assessment of two-way SNP interactions in breast cancer susceptibility using 46 450 cases and 42 461 controls from the breast cancer association consortium
Part of the substantial unexplained familial aggregation of breast cancer may be due to interactions between common variants, but few studies have had adequate statistical power to detect interactions of realistic magnitude. We aimed to assess all two-way interactions in breast cancer susceptibility between 70,917 single nucleotide polymorphisms (SNPs) selected primarily based on prior evidence of a marginal effect. Thirty-eight international studies contributed data for 46,450 breast cancer cases and 42,461 controls of European origin as part of a multi-consortium project (COGS). First, SNPs were preselected based on evidence (P < 0.01) of a per-allele main effect, and all two-way combinations of those were evaluated by a per-allele (1 d.f.) test for interaction using logistic regression. Second, all 2.5 billion possible two-SNP combinations were evaluated using Boolean operation-based screening and testing, and SNP pairs with the strongest evidence of interaction (P < 10(-4)) were selected for more careful assessment by logistic regression. Under the first approach, 3277 SNPs were preselected, but an evaluation of all possible two-SNP combinations (1 d.f.) identified no interactions at P < 10(-8). Results from the second analytic approach were consistent with those from the first (P > 10(-10)). In summary, we observed little evidence of two-way SNP interactions in breast cancer susceptibility, despite the large number of SNPs with potential marginal effects considered and the very large sample size. This finding may have important implications for risk prediction, simplifying the modelling required. Further comprehensive, large-scale genome-wide interaction studies may identify novel interacting loci if the inherent logistic and computational challenges can be overcome.
DOI: 10.1186/s12885-017-3631-8
2017
Cited 30 times
CHEK2 c.1100delC mutation is associated with an increased risk for male breast cancer in Finnish patient population
Several susceptibility genes have been established for female breast cancer, of which mutations in BRCA1 and especially in BRCA2 are also known risk factors for male breast cancer (MBC). The role of other breast cancer genes in MBC is less well understood.In this study, we have genotyped 68 MBC patients for the known breast or ovarian cancer associated mutations in the Finnish population in CHEK2, PALB2, RAD51C, RAD51D, and FANCM genes.CHEK2 c.1100delC mutation was found in 4 patients (5.9%), which is significantly more frequent than in the control population (OR: 4.47, 95% CI 1.51-13.18, p = 0.019). Four CHEK2 I157T variants were also detected, but the frequency did not significantly differ from population controls (p = 0.781). No RAD51C, RAD51D, PALB2, or FANCM mutations were found.These data suggest that the CHEK2 c.1100delC mutation is associated with an increased risk for MBC in the Finnish population.
DOI: 10.1186/s12885-018-4141-z
2018
Cited 28 times
High-level cytoplasmic claudin 3 expression is an independent predictor of poor survival in triple-negative breast cancer
The subtype of claudin-low breast cancer can be reliably determined only by gene-expression profiling. Attempts have been made to develop immunohistochemical surrogates, which nearly always focus on membranous claudin expression.We assessed the immunohistochemical expression of both membranous and cytoplasmic claudins 3, 4 and 7 in a series of 197 non-metastatic breast cancers, enriched with triple-negative breast cancers (TNBCs; 60%). The expression of epithelial-to-mesenchymal transition-regulating transcription factors Sip1, Zeb1 and vimentin had previously been determined in the same material.In multivariate analysis, strong cytoplasmic claudin 3 expression was associated with poor relapse-free survival (RFS), disease-free survival, distant disease-free survival, breast cancer-specific survival and overall survival among TNBC patients (for RFS, RR 5.202, 95% CI 1.210-22.369, p = 0.027, vs. T-class, RR 0.663, 95% CI 0.168-2.623, p = 0.558, and N-class, RR 3.940, 95% CI 0.933-16.631, p = 0.062). Cytoplasmic claudin 3 expression was also associated with strong nuclear Sip1 expression (p = 0.000053), TNBC phenotype (p = 0.012) and within them, non-basal-like phenotype (p = 0.026). Cytoplasmic claudin 7 was associated with dismal RFS (RR 6.328, 95% CI 1.401-28.593, p = 0.016, vs. T-class, RR 0.692, 95% CI 0.242-1.982, p = 0.493, and N-class, RR 2.981, 95% CI 1.1016-8.749, p = 0.047). Low cytoplasmic expression of claudins 3, 4 and 7 together also predicted poor RFS (RR 6.070, 95% CI 1.347-27.363, p = 0.019, vs. T-class, RR 0.677, 95% CI 0.237-1.934, p = 0.467, and N-class, RR 3.167, 95% CI 1.079-9.290, p = 0.036).Immunohistochemical expression levels of cytoplasmic claudins 3 and 7 appear to be novel prognostic factors in TNBC.
DOI: 10.1007/s10549-011-1750-5
2011
Cited 34 times
Expressions of individual PHDs associate with good prognostic factors and increased proliferation in breast cancer patients
DOI: 10.1258/acb.2011.011040
2011
Cited 32 times
Comparison of total and intact aminoterminal propeptide of type I procollagen assays in patients with breast cancer with or without bone metastases
Background Aminoterminal propeptide of type I procollagen (PINP) reflects bone formation. Two different antigens exist in human serum: intact PINP and a monomeric form. The intact PINP assay measures trimeric form and the total assay measures both forms. The structure and origin of the monomeric form is still unclear. Methods Automated intact and total PINP assays were compared in breast cancer patients with ( n = 60) or without bone metastases ( n = 226). In addition, cross-linked carboxyterminal telopeptide of type I collagen (ICTP) was measured from the same patients and compared with the concentration of PINP monomer (difference between intact and total PINP). Monomeric PINP was purified from human ascitic fluid and characterized by sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). Results The correlations were good ( r &gt; 0.948) between intact PINP and total P1NP in all patient groups. The correlation between the monomeric form and ICTP was lower in patients without bone metastases ( r = 0.507) than in patients with bone metastases ( r = 0.894). This indicates that the monomeric form reflects the degradation of type I collagen because bone metastases are osteolytic in nature. After several steps in the purification of the monomer form there was a single peak. Only the single band was visible in the SDS-PAGE gel. The alpha1-chain of intact PINP consists of 161 amino acids with a molecular weight of 14224.02. The purified monomer peptide in MALDI-TOF MS was smaller, 10576.41, and most likely cleaved after the arginine residue (amino acid number 120) with a trypsin-like protease. Conclusions Intact and total PINP assays give similar results in many conditions, but there are differences, for example in breast carcinoma, which should be recognized.
DOI: 10.1371/journal.pgen.1002734
2012
Cited 31 times
Rare Copy Number Variants Observed in Hereditary Breast Cancer Cases Disrupt Genes in Estrogen Signaling and TP53 Tumor Suppression Network
Breast cancer is the most common cancer in women in developed countries, and the contribution of genetic susceptibility to breast cancer development has been well-recognized. However, a great proportion of these hereditary predisposing factors still remain unidentified. To examine the contribution of rare copy number variants (CNVs) in breast cancer predisposition, high-resolution genome-wide scans were performed on genomic DNA of 103 BRCA1, BRCA2, and PALB2 mutation negative familial breast cancer cases and 128 geographically matched healthy female controls; for replication an independent cohort of 75 similarly mutation negative young breast cancer patients was used. All observed rare variants were confirmed by independent methods. The studied breast cancer cases showed a consistent increase in the frequency of rare CNVs when compared to controls. Furthermore, the biological networks of the disrupted genes differed between the two groups. In familial cases the observed mutations disrupted genes, which were significantly overrepresented in cellular functions related to maintenance of genomic integrity, including DNA double-strand break repair (P = 0.0211). Biological network analysis in the two independent breast cancer cohorts showed that the disrupted genes were closely related to estrogen signaling and TP53 centered tumor suppressor network. These results suggest that rare CNVs represent an alternative source of genetic variation influencing hereditary risk for breast cancer.
DOI: 10.1007/s10549-011-1590-3
2011
Cited 29 times
Estrogen receptor-α and sex steroid hormones regulate Toll-like receptor-9 expression and invasive function in human breast cancer cells
DOI: 10.1186/s13058-015-0570-7
2015
Cited 27 times
Common germline polymorphisms associated with breast cancer-specific survival
Previous studies have identified common germline variants nominally associated with breast cancer survival. These associations have not been widely replicated in further studies. The purpose of this study was to evaluate the association of previously reported SNPs with breast cancer-specific survival using data from a pooled analysis of eight breast cancer survival genome-wide association studies (GWAS) from the Breast Cancer Association Consortium. A literature review was conducted of all previously published associations between common germline variants and three survival outcomes: breast cancer-specific survival, overall survival and disease-free survival. All associations that reached the nominal significance level of P value <0.05 were included. Single nucleotide polymorphisms that had been previously reported as nominally associated with at least one survival outcome were evaluated in the pooled analysis of over 37,000 breast cancer cases for association with breast cancer-specific survival. Previous associations were evaluated using a one-sided test based on the reported direction of effect. Fifty-six variants from 45 previous publications were evaluated in the meta-analysis. Fifty-four of these were evaluated in the full set of 37,954 breast cancer cases with 2,900 events and the two additional variants were evaluated in a reduced sample size of 30,000 samples in order to ensure independence from the previously published studies. Five variants reached nominal significance (P <0.05) in the pooled GWAS data compared to 2.8 expected under the null hypothesis. Seven additional variants were associated (P <0.05) with ER-positive disease. Although no variants reached genome-wide significance (P <5 x 10−8), these results suggest that there is some evidence of association between candidate common germline variants and breast cancer prognosis. Larger studies from multinational collaborations are necessary to increase the power to detect associations, between common variants and prognosis, at more stringent significance levels.
DOI: 10.1186/s12885-017-3056-4
2017
Cited 26 times
Primary neuroendocrine breast carcinomas are associated with poor local control despite favourable biological profile: a retrospective clinical study
Breast carcinomas with neuroendocrine features (NEBC) are a very rare entity of mammary neoplasms, WHO classification of which has recently been revised. There are very limited data available about the clinical behaviour and treatment options of NEBC.We collected retrospectively patients with NEBC from Oulu and Helsinki University Hospitals in 2007-2015. There were 43 NEBC cases during the period.The incidence of NEBC from all breast cancers varied from 0.1% in Helsinki to 1.3% in Oulu. The mean tumor size was 2.2 cm and 23 patients (55.8%) had no lymph node metastases when diagnosed. In total, 4 patients (9.3%) had distant metastases at the time of diagnosis. High estrogen receptor (ER) expression was observed in 41 (97.7%) patients. When non-metastatic NEBC were compared to a prospective set of ductal carcinomas (n = 506), NEBC were more often HER2 negative (p = 0.046), ER positive (p = 0.0062) and the NEBC patients were older (p < 0.0005) than patients with ductal carcinomas. Plasma chromogranin A correlated only to higher age at diagnosis (p = 0.0028). Relapse-free survival (p = 0.0013), disease-free survival (p = 0.024) and overall survival (p = 0.0028) favoured ductal carcinomas compared to NEBC, while no difference was observed in distant disease-free survival or in breast cancer-specific survival.There is remarkable variation in the incidence of NEBC in Finland, which is likely to be explained by differences in the use of neuroendocrine marker immunostainings. Poor local control and worse overall survival may be linked to the more aggressive biology of the disease, despite its association with apparently indolent prognostic factors.
DOI: 10.1158/1541-7786.mcr-17-0452
2018
Cited 23 times
USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
Recent studies suggest that the ubiquitin-specific protease USP28 plays an important role in cellular repair and tissue remodeling, which implies that it has a direct role in carcinogenesis. The carcinogenic potential of USP28 was investigated in a comprehensive manner using patients, animal models, and cell culture. The findings demonstrate that overexpression of USP28 correlates with a better survival in patients with invasive ductal breast carcinoma. Mouse xenograft experiments with USP28-deficient breast cancer cells also support this view. Furthermore, lack of USP28 promotes a more malignant state of breast cancer cells, indicated by an epithelial-to-mesenchymal (EMT) transition, elevated proliferation, migration, and angiogenesis as well as a decreased adhesion. In addition to breast cancer, lack of USP28 in mice promoted an earlier onset and a more severe tumor formation in a chemical-induced liver cancer model. Mechanistically, the angio- and carcinogenic processes driven by the lack of USP28 appeared to be independent of HIF-1α, p53, and 53BP1.Implications: The findings of this study are not limited to one particular type of cancer but are rather applicable for carcinogenesis in a more general manner. The obtained data support the view that USP28 is involved in tumor suppression and has the potential to be a prognostic marker. Mol Cancer Res; 16(6); 1000-12. ©2018 AACR.
DOI: 10.1038/bjc.2011.518
2011
Cited 27 times
Absence of the DNA repair enzyme human 8-oxoguanine glycosylase is associated with an aggressive breast cancer phenotype
8-Oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) is the most abundant marker of DNA damage and it reflects oxidative stress. Human 8-oxoguanine glycosylase (hOGG1) is a DNA-repair enzyme that participates in 8-oxodG removal.hOGG1 protein expression was immunohistochemically studied in 96 patients with local or locally advanced breast cancer and in 20 lesions of non-malignant breast disease. 8-OxodG levels had been previously determined in all patients.hOGG1 was overexpressed in invasive vs non-invasive lesions (P=0.006). 8-OxodG and hOGG1 had a significant inverse association (P=0.046). Lack of hOGG1 expression was associated with the most poor prognostic factors of breast cancer. In addition, all triple-negative breast carcinomas (TNBCs) were hOGG1 negative (P=0.027 vs non-TNBCs). Patients with a lack of both hOGG1- and 8-oxodG immunostaining showed extremely poor breast cancer-specific survival compared with those with either 8-oxodG- or hOGG1-positive tumours (P<0.000005).The current results imply that absence of hOGG1 expression is associated with features of aggressive breast cancer. Tumours lacking both 8-oxodG and hOGG1 seem to indicate especially poor prognosis.
DOI: 10.3109/0284186x.2013.820840
2013
Cited 24 times
Outcome of patients with HER2-positive breast cancer treated with or without adjuvant trastuzumab in the Finland Capecitabine Trial (FinXX)
Little information is available about survival outcomes of patients with HER2-positive early breast cancer treated with adjuvant capecitabine-containing chemotherapy with or without trastuzumab.One thousand and five hundred patients with early breast cancer were entered to the Finland Capecitabine trial (FinXX) between January 2004 and May 2007, and were randomly assigned to receive either three cycles of adjuvant TX (docetaxel, capecitabine) followed by three cycles of CEX (cyclophosphamide, epirubicin, capecitabine; TX-CEX) or three cycles of docetaxel followed by three cycles of CEF (cyclophosphamide, epirubicin, fluorouracil; T-CEF). The primary endpoint was recurrence-free survival (RFS). The study protocol was amended in May 2005 while study accrual was ongoing to allow adjuvant trastuzumab for patients with HER2-positive cancer. Of the 284 patients with HER2-positive cancer accrued to FinXX, 176 (62.0%) received trastuzumab after amending the study protocol, 131 for 12 months and 45 for nine weeks. The median follow-up time was 6.7 years.Patients with HER2-positive cancer who received trastuzumab had better RFS than those who did not (five-year RFS 89.2% vs. 75.9%; HR 0.41, 95% CI 0.23-0.72; p = 0.001). Patients treated with trastuzumab for 12 months or nine weeks had similar RFS. There was no significant interaction between trastuzumab administration and the type of chemotherapy. Four (2.3%) patients treated with trastuzumab had heart failure or left ventricular dysfunction, three of these received capecitabine.Adjuvant trastuzumab improves RFS of patients treated with TX-CEX or T-CEF. Few patients had cardiac failure.
DOI: 10.1371/journal.pgen.1005816
2016
Cited 20 times
Targeted Next-Generation Sequencing Identifies a Recurrent Mutation in MCPH1 Associating with Hereditary Breast Cancer Susceptibility
Breast cancer is strongly influenced by hereditary risk factors, a majority of which still remain unknown. Here, we performed a targeted next-generation sequencing of 796 genes implicated in DNA repair in 189 Finnish breast cancer cases with indication of hereditary disease susceptibility and focused the analysis on protein truncating mutations. A recurrent heterozygous mutation (c.904_916del, p.Arg304ValfsTer3) was identified in early DNA damage response gene, MCPH1, significantly associating with breast cancer susceptibility both in familial (5/145, 3.4%, P = 0.003, OR 8.3) and unselected cases (16/1150, 1.4%, P = 0.016, OR 3.3). A total of 21 mutation positive families were identified, of which one-third exhibited also brain tumors and/or sarcomas (P = 0.0007). Mutation carriers exhibited significant increase in genomic instability assessed by cytogenetic analysis for spontaneous chromosomal rearrangements in peripheral blood lymphocytes (P = 0.0007), suggesting an effect for MCPH1 haploinsufficiency on cancer susceptibility. Furthermore, 40% of the mutation carrier tumors exhibited loss of the wild-type allele. These findings collectively provide strong evidence for MCHP1 being a novel breast cancer susceptibility gene, which warrants further investigations in other populations.
DOI: 10.1038/s41598-017-00766-9
2017
Cited 20 times
Case-control analysis of truncating mutations in DNA damage response genes connects TEX15 and FANCD2 with hereditary breast cancer susceptibility
Several known breast cancer susceptibility genes encode proteins involved in DNA damage response (DDR) and are characterized by rare loss-of-function mutations. However, these explain less than half of the familial cases. To identify novel susceptibility factors, 39 rare truncating mutations, identified in 189 Northern Finnish hereditary breast cancer patients in parallel sequencing of 796 DDR genes, were studied for disease association. Mutation screening was performed for Northern Finnish breast cancer cases (n = 578-1565) and controls (n = 337-1228). Mutations showing potential cancer association were analyzed in additional Finnish cohorts. c.7253dupT in TEX15, encoding a DDR factor important in meiosis, associated with hereditary breast cancer (p = 0.018) and likely represents a Northern Finnish founder mutation. A deleterious c.2715 + 1G > A mutation in the Fanconi anemia gene, FANCD2, was over two times more common in the combined Finnish hereditary cohort compared to controls. A deletion (c.640_644del5) in RNF168, causative for recessive RIDDLE syndrome, had high prevalence in majority of the analyzed cohorts, but did not associate with breast cancer. In conclusion, truncating variants in TEX15 and FANCD2 are potential breast cancer risk factors, warranting further investigations in other populations. Furthermore, high frequency of RNF168 c.640_644del5 indicates the need for its testing in Finnish patients with RIDDLE syndrome symptoms.
DOI: 10.1186/1471-2407-14-902
2014
Cited 18 times
Recurrent CYP2C19 deletion allele is associated with triple-negative breast cancer
Using a genome-wide approach, we have previously observed an increase in the frequency of rare copy number variants (CNVs) in familial and early-onset breast cancer cases when compared to controls. Moreover, the biological networks of the CNV disrupted genes differed between the two groups. Here, six of the previously observed CNVs were selected for further investigation. Four of these were singletons and disturbed the following genes: DCLRE1C, CASP3, DAB2IP and ITGA9, encoding proteins that are part of the TP53 and β-estradiol centered network. The two others were recurrent alleles and disrupted CDH19 and CYP2C19 genes. Of these, CDH19 encodes a cadherin functioning as a cell-cell adhesion receptor and CYP2C19 a CYP450 enzyme with a major function in estrogen catabolism.The exact breakpoints of the six previously observed CNV deletion alleles were defined by using qPCR, nested PCR and sequencing. The prevalence of these CNVs was investigated in 842 Northern Finnish breast cancer cases, unselected for family history of cancer and age at disease onset, as well as in 497 healthy female controls by using multiplex PCR. Also the association of the relatively common CDH19 and CYP2C19 deletion alleles with different clinical parameters was studied.No significant differences in the carrier frequencies between cases and controls were found for any of the studied CNVs. However, the deletion in CYP2C19 showed a significant association with triple-negative breast cancer (p=0.021).Our results indicate that inherited changes in CYP2C19 gene participating in estrogen catabolism have an influence on the molecular subtype of breast cancer.
DOI: 10.1038/ncomms5051
2014
Cited 18 times
2q36.3 is associated with prognosis for oestrogen receptor-negative breast cancer patients treated with chemotherapy
Large population-based registry studies have shown that breast cancer prognosis is inherited. Here we analyse single-nucleotide polymorphisms (SNPs) of genes implicated in human immunology and inflammation as candidates for prognostic markers of breast cancer survival involving 1,804 oestrogen receptor (ER)-negative patients treated with chemotherapy (279 events) from 14 European studies in a prior large-scale genotyping experiment, which is part of the Collaborative Oncological Gene-environment Study (COGS) initiative. We carry out replication using Asian COGS samples (n=522, 53 events) and the Prospective Study of Outcomes in Sporadic versus Hereditary breast cancer (POSH) study (n=315, 108 events). Rs4458204_A near CCL20 (2q36.3) is found to be associated with breast cancer-specific death at a genome-wide significant level (n=2,641, 440 events, combined allelic hazard ratio (HR)=1.81 (1.49–2.19); P for trend=1.90 × 10−9). Such survival-associated variants can represent ideal targets for tailored therapeutics, and may also enhance our current prognostic prediction capabilities. Studies have shown that breast cancer prognosis is hereditary. Here the authors show that a genetic variant in CCL20, a chemokine ligand involved in immune response, is significantly associated with breast cancer survival and may therefore represent an important therapeutic or prognostic target.
DOI: 10.1186/s13058-024-01812-x
2024
Serum protein profiling reveals an inflammation signature as a predictor of early breast cancer survival
Breast cancers exhibit considerable heterogeneity in their biology, immunology, and prognosis. Currently, no validated, serum protein-based tools are available to evaluate the prognosis of patients with early breast cancer.The study population consisted of 521 early-stage breast cancer patients with a median follow-up of 8.9 years. Additionally, 61 patients with breast fibroadenoma or atypical ductal hyperplasia were included as controls. We used a proximity extension assay to measure the preoperative serum levels of 92 proteins associated with inflammatory and immune response processes. The invasive cancers were randomly split into discovery (n = 413) and validation (n = 108) cohorts for the statistical analyses.Using LASSO regression, we identified a nine-protein signature (CCL8, CCL23, CCL28, CSCL10, S100A12, IL10, IL10RB, STAMPB2, and TNFβ) that predicted various survival endpoints more accurately than traditional prognostic factors. In the time-dependent analyses, the prognostic power of the model remained rather stable over time. We also developed and validated a 17-protein model with the potential to differentiate benign breast lesions from malignant lesions (Wilcoxon p < 2.2*10- 16; AUC 0.94).Inflammation and immunity-related serum proteins have the potential to rise above the classical prognostic factors of early-stage breast cancer. They may also help to distinguish benign from malignant breast lesions.
DOI: 10.1111/ejh.14219
2024
Front‐line and second‐line treatment for mantle cell lymphoma in clinical practice: A multicenter retrospective analysis
Abstract Background There are few reports of clinical practice treatment patterns and efficacy in mantle cell lymphoma (MCL). Materials and Methods We retrospectively studied a large, multicenter, cohort of patients with MCL diagnosed between 2000 and 2020 in eight institutions. Results 536 patients were registered (73% male, median of 70 years). Front‐line treatment was based on high‐dose cytarabine, bendamustine, and anthracyclines in 42%, 12%, and 15%, respectively. The median PFS for all patients was 45 months; 68, 34, and 30 months for those who received high‐dose cytarabine‐based, bendamustine‐based and anthracycline‐based therapy. 204 patients received second‐line. Bendamustine‐based treatment was the most common second‐line regimen (36% of patients). The median second‐line PFS (sPFS) for the entire cohort was 14 months; 19, 24, and 31 for bendamustine‐, platinum‐, and high‐dose cytarabine‐based regimens, with broad confidence intervals for these latter estimates. Patients treated with cytarabine‐based therapies in the front‐line and those with front‐line PFS longer than 24 months had a substantially superior sPFS. Conclusion Front‐line treatment in this cohort of MCL was as expected and with a median PFS of over 3.5 years. Second‐line treatment strategies were heterogeneous and the median second‐line PFS was little over 1 year.
DOI: 10.1016/j.breast.2010.05.001
2010
Cited 19 times
Long-term observational follow-up study of breast cancer diagnosed in women ≤40 years old
The prognosis of young breast cancer patients has been considered to be much poorer than in older patients. Two hundred and sixty-eight premenopausal women with a median follow-up time of 74.0 months were included in the study. 33.5% had oestrogen receptor-negative and 34.6% progesterone receptor-negative tumours. 15.2% of the tumours were HER2-positive. Five-year breast cancer-specific survival (BCSS) was 81.1% and the corresponding 10-year figure was 72.3%. 91.8% of all relapses occurred within seven years of surgery. Among the ≤35-year-old women, only 2 of 38 (5.3%) relapsed beyond seven years of follow-up. Lymph node ratio was the most significant independent prognostic factor of poor disease-free survival and BCSS. This study revealed a high relapse rate in the youngest women as early as during the first few years after diagnosis, although their prognosis as a whole was surprisingly good.
DOI: 10.1002/ijc.31259
2018
Cited 15 times
Rare missense mutations in <i>RECQL</i> and <i>POLG</i> associate with inherited predisposition to breast cancer
Several known breast cancer susceptibility genes with moderate‐to‐high risk alleles encode proteins involved in DNA damage response (DDR). As these explain less than half of the hereditary breast cancer cases, additional predisposing alleles are likely to be discovered. Many of the previous studies utilizing massive parallel sequencing have focused on the protein‐truncating variants, and the role of rare missense mutations has remained poorly addressed. To identify novel susceptibility factors, we have systematically analyzed the data from our parallel sequencing of 796 DDR genes in 189 Northern Finnish hereditary breast cancer patients for rare missense variants, predicted as deleterious. Thirty‐five variants were studied here for the disease association using Finnish breast cancer case ( n = 492–2,035) and control ( n = 277–1,539) cohorts. As a result, two missense variants in genes involved in DNA replication, RECQL p.I156M and POLG p.L392V, the former involving genomic and the latter mitochondrial DNA replication, showed significant association with risk of breast cancer. Rare RECQL p.I156M allele was observed in breast cancer cases only (6/1,946, 0.3%, p = 0.043), whereas POLG p.L392V was two times more frequent in breast cancer cases (53/2,238, 2.4%) compared to controls (18/1,539, 1.2%, OR = 2.1, 95% CI 1.2–3.5, p = 0.010). Based on the current genetic data, both RECQL p.I156M and POLG p.L392V represent novel breast cancer predisposing alleles.
DOI: 10.1111/bjh.18597
2022
Cited 6 times
Survival of patients with mantle cell lymphoma in the rituximab era: Retrospective binational analysis between 2000 and 2020
Summary Mantle cell lymphoma (MCL) is a rare peripheral B‐cell lymphoma characterised by eventual relapse and progression towards a more aggressive disease biology. With the introduction of rituximab‐ and cytarabine‐based immunochemotherapy regimens, the prognosis of the disease has changed dramatically over the last two decades. To assess the real‐world survival of patients with MCL, we used a population‐based cohort of 564 patients with MCL who were diagnosed and treated between 2000 and 2020. Patient data were collected from seven Finnish treatment centres and one Spanish treatment centre. For the entire patient population, we report a 2‐year overall survival (OS) rate of 77%, a 5‐year OS of 58%, and a 10‐year OS of 32%. The estimated median OS was 80 months after diagnosis. MCL is associated with increased mortality across the entire patient population. Additionally, we assessed the survival of patients after MCL relapse with the aim of establishing a cut‐off point of prognostic significance. Based on our statistical analysis of survival after the first relapse, disease progression within 24 months of the initial diagnosis should be considered as a strong indicator of poor prognosis.
DOI: 10.1007/s10549-016-3683-5
2016
Cited 12 times
Telomeric G-quadruplex-forming DNA fragments induce TLR9-mediated and LL-37-regulated invasion in breast cancer cells in vitro
DOI: 10.1371/journal.pone.0160316
2016
Cited 12 times
Fine-Mapping of the 1p11.2 Breast Cancer Susceptibility Locus
The Cancer Genetic Markers of Susceptibility genome-wide association study (GWAS) originally identified a single nucleotide polymorphism (SNP) rs11249433 at 1p11.2 associated with breast cancer risk. To fine-map this locus, we genotyped 92 SNPs in a 900kb region (120,505,799–121,481,132) flanking rs11249433 in 45,276 breast cancer cases and 48,998 controls of European, Asian and African ancestry from 50 studies in the Breast Cancer Association Consortium. Genotyping was done using iCOGS, a custom-built array. Due to the complicated nature of the region on chr1p11.2: 120,300,000–120,505,798, that lies near the centromere and contains seven duplicated genomic segments, we restricted analyses to 429 SNPs excluding the duplicated regions (42 genotyped and 387 imputed). Per-allelic associations with breast cancer risk were estimated using logistic regression models adjusting for study and ancestry-specific principal components. The strongest association observed was with the original identified index SNP rs11249433 (minor allele frequency (MAF) 0.402; per-allele odds ratio (OR) = 1.10, 95% confidence interval (CI) 1.08–1.13, P = 1.49 x 10-21). The association for rs11249433 was limited to ER-positive breast cancers (test for heterogeneity P≤8.41 x 10-5). Additional analyses by other tumor characteristics showed stronger associations with moderately/well differentiated tumors and tumors of lobular histology. Although no significant eQTL associations were observed, in silico analyses showed that rs11249433 was located in a region that is likely a weak enhancer/promoter. Fine-mapping analysis of the 1p11.2 breast cancer susceptibility locus confirms this region to be limited to risk to cancers that are ER-positive.
DOI: 10.1093/hmg/ddu300
2014
Cited 12 times
Genetic variation in mitotic regulatory pathway genes is associated with breast tumor grade
Mitotic index is an important component of histologic grade and has an etiologic role in breast tumorigenesis. Several small candidate gene studies have reported associations between variation in mitotic genes and breast cancer risk. We measured associations between 2156 single nucleotide polymorphisms (SNPs) from 194 mitotic genes and breast cancer risk, overall and by histologic grade, in the Breast Cancer Association Consortium (BCAC) iCOGS study (n = 39 067 cases; n = 42 106 controls). SNPs in TACC2 [rs17550038: odds ratio (OR) = 1.24, 95% confidence interval (CI) 1.16–1.33, P = 4.2 × 10−10) and EIF3H (rs799890: OR = 1.07, 95% CI 1.04–1.11, P = 8.7 × 10−6) were significantly associated with risk of low-grade breast cancer. The TACC2 signal was retained (rs17550038: OR = 1.15, 95% CI 1.07–1.23, P = 7.9 × 10−5) after adjustment for breast cancer risk SNPs in the nearby FGFR2 gene, suggesting that TACC2 is a novel, independent genome-wide significant genetic risk locus for low-grade breast cancer. While no SNPs were individually associated with high-grade disease, a pathway-level gene set analysis showed that variation across the 194 mitotic genes was associated with high-grade breast cancer risk (P = 2.1 × 10−3). These observations will provide insight into the contribution of mitotic defects to histological grade and the etiology of breast cancer.
DOI: 10.18632/oncotarget.13570
2016
Cited 11 times
Toll-like receptor 9 expression is associated with breast cancer sensitivity to the growth inhibitory effects of bisphosphonates <i>in vitro</i> and <i>in vivo</i>
Bisphosphonates are standard treatments for bone metastases.When given in the adjuvant setting, they reduce breast cancer mortality and recurrence in bone but only among post-menopausal patients.Optimal drug use would require biomarkerbased patient selection.Such biomarkers are not yet in clinical use.Based on the similarities in inflammatory responses to bisphosphonates and Toll-like receptor (TLR) agonists, we hypothesized that TLR9 expression may affect bisphosphonate responses in cells.We compared bisphosphonate effects in breast cancer cell lines with low or high TLR9 expression.We discovered that cells with decreased TLR9 expression are significantly more sensitive to the growth-inhibitory effects of bisphosphonates in vitro and in vivo.Furthermore, cancer growth-promoting effects seen with some bisphosphonates in some control shRNA cells were not detected in TLR9 shRNA cells.These differences were not associated with inhibition of Rap1A prenylation or p38 phosphorylation, which are known markers for bisphosphonate activity.However, TLR9 shRNA cells exhibited increased sensitivity to ApppI, a metabolite that accumulates in cells after bisphosphonate treatment.We conclude that decreased TLR9-expression sensitizes breast cancer cells to the growth inhibitory effects of bisphosphonates.Our results suggest that TLR9 should be studied as a potential biomarker for adjuvant bisphosphonate sensitivity among breast cancer patients.
DOI: 10.1016/j.wneu.2023.07.008
2023
Impact of Timing of Surgery and Adjuvant Treatment on Survival of Adult IDH–wild-type Glioblastoma: A Single-center Study of 392 Patients
The purpose of our study was to analyze the impact of time interval from referral to surgery and from surgery to adjuvant treatment on survival of adult IDH-wildtype glioblastomas. Data on 392 IDH-wildtype glioblastomas diagnosed at the Tampere University Hospital in 2004–2016 were obtained from the electronic patient record system. Piecewise Cox regression was used to calculate hazard ratios for different time intervals between referral and surgery, as well as between surgery and adjuvant treatments. The median survival time from primary surgery was 9.5 months (interquartile range: 3.8–16.0). Survival among patients with an interval exceeding four weeks from referral to surgery was no worse compared to <2 weeks (hazard ratio: 0.78; 95% confidence interval: 0.54–1.14). We found indications of poorer outcome when the interval from surgery to radiotherapy exceeded 30 days (hazard ratio: 1.42; 95% confidence interval: 0.91–2.21 for 31–44 days and 1.59; 0.94–2.67 for over 45 days). Interval from referral to surgery in the range of 4–10 weeks was not associated with decreased survivals in IDH-wildtype glioblastomas. In contrast, delay exceeding 30 days from surgery to adjuvant treatment may decrease long-term survival.
DOI: 10.3389/fgene.2013.00104
2013
Cited 8 times
The UGT1A6_19_GG genotype is a breast cancer risk factor
Validation of an association between the UGT1A6_19_T>G (rs6759892) polymorphism and overall breast cancer risk. A pilot study included two population-based case-control studies from Germany (MARIE-GENICA). An independent validation study comprised four independent breast cancer case-control studies from Finland (KBCP, OBCS), Germany (BBCC), and Sweden (SASBAC). The pooled analysis included 7418 cases and 8720 controls from all six studies. Participants were of European descent. Genotyping was done by MALDI-TOF MS and statistical analysis was performed by logistic regression adjusted for age and study. The increased overall breast cancer risk for women with the UGT1A6_19_GG genotype which was observed in the pilot study was confirmed in the set of four independent study collections (OR 1.13, 95% CI 1.05-1.22; p = 0.001). The pooled study showed a similar effect (OR 1.09, 95% CI 1.04-1.14; p = 0.001). The risk effect on the basis of allele frequencies was highly significant, the pooled analysis showed an OR of 1.11 (95% CI 1.06-1.16; p = 5.8 × 10(-6)). We confirmed the association of UGT1A6_19_GG with increased overall breast cancer risk and conclude that our result from a well powered multi-stage study adds a novel candidate to the panel of validated breast cancer susceptibility loci.
DOI: 10.1159/000493348
2018
Cited 8 times
Neuroendocrine Breast Carcinomas Share Prognostic Factors with Gastroenteropancreatic Neuroendocrine Tumors: A Putative Prognostic Role of Menin, p27, and SSTR-2A
&lt;b&gt;&lt;i&gt;Objectives:&lt;/i&gt;&lt;/b&gt; Due to the rarity of breast carcinomas with neuroendocrine features (NEBC), the knowledge on their biology is very limited but the identification of their biology and prognostic factors is essential to evaluate both pathogenesis and possible targeted treatment options. We assessed the expression of the well-characterized prognostic factors of gastroenteropancreatic neuroendocrine tumors (GEP-NET) in NEBC. &lt;b&gt;&lt;i&gt;Methods:&lt;/i&gt;&lt;/b&gt; We assessed the immunohistochemical expression of neuron-specific enolase (NSE), thymidylate synthase (TS), p27, CD56, menin, and somatostatin receptor type 2A (SSTR-2A) in a series of 36 NEBC and 45 invasive ductal carcinomas (IDC). &lt;b&gt;&lt;i&gt;Results:&lt;/i&gt;&lt;/b&gt; Nuclear and cytoplasmic TS, nuclear and cytoplasmic NSE, and nuclear p27 had significant overexpression in NEBC compared with IDC (for all, &lt;i&gt;p&lt;/i&gt; &amp;#x3c; 0.01). In NEBC, cytoplasmic SSTR-2A expression was associated with excellent distant disease-free survival (&lt;i&gt;p&lt;/i&gt; = 0.013), cytoplasmic menin expression with poorer relapse-free survival (&lt;i&gt;p&lt;/i&gt; = 0.022), and nuclear p27 with longer breast cancer-specific survival (&lt;i&gt;p&lt;/i&gt; = 0.022). &lt;b&gt;&lt;i&gt;Conclusions:&lt;/i&gt;&lt;/b&gt; There is a striking similarity in GEP-NET and NEBC regarding prognostic factors. GEP-NET and NEBC also appear to show similar expression patterns of the studied markers, while there are notable differences compared to IDC. Due to the wide expression of SSTR-2A, the treatment option with somatostatin analogs in NEBC should be evaluated.
DOI: 10.1038/bjc.2011.448
2011
Cited 7 times
Evaluation of variation in the phosphoinositide-3-kinase catalytic subunit alpha oncogene and breast cancer risk
Somatic mutations in phosphoinositide-3-kinase catalytic subunit alpha (PIK3CA) are frequent in breast tumours and have been associated with oestrogen receptor (ER) expression, human epidermal growth factor receptor-2 overexpression, lymph node metastasis and poor survival. The goal of this study was to evaluate the association between inherited variation in this oncogene and risk of breast cancer.A single-nucleotide polymorphism from the PIK3CA locus that was associated with breast cancer in a study of Caucasian breast cancer cases and controls from the Mayo Clinic (MCBCS) was genotyped in 5436 cases and 5280 controls from the Cancer Genetic Markers of Susceptibility (CGEMS) study and in 30 949 cases and 29 788 controls from the Breast Cancer Association Consortium (BCAC).Rs1607237 was significantly associated with a decreased risk of breast cancer in MCBCS, CGEMS and all studies of white Europeans combined (odds ratio (OR)=0.97, 95% confidence interval (CI) 0.95-0.99, P=4.6 × 10(-3)), but did not reach significance in the BCAC replication study alone (OR=0.98, 95% CI 0.96-1.01, P=0.139).Common germline variation in PIK3CA does not have a strong influence on the risk of breast cancer.
DOI: 10.1158/0008-5472.sabcs10-s4-1
2010
Cited 7 times
Abstract S4-1: FinXX Final 5-Year Analysis: Results of the Randomised, Open-Label, Phase III Trial in Medium-to-High Risk Early Breast Cancer
Abstract Background: The randomised, phase III FinXX trial (NCT00114816) investigated whether the integration of capecitabine (X) into a sequential docetaxel (T)→cyclophosphamide + epirubicin + 5-FU (CEF) adjuvant regimen might improve clinical outcome for patients with medium-to-high risk early breast cancer. The planned interim analysis after a median follow-up of 3 years found a significant recurrence-free survival (RFS) benefit with the X-containing regimen versus control (3-year RFS rate: 93% XTP→ukkalaCEX vs 89% T→CEF; hazard ratio 0.66 [95% CI: 0.47-0.94]; p=0.020) [Lancet Oncol 2009;10:1145-51]. Here we present final 5-year RFS data, the primary endpoint of the study. Methods: Eligible patients were aged 18-65 years, had a WHO performance status of 0 or 1 and histologically confirmed invasive breast cancer at medium-to-high risk of recurrence, defined as regional node-positive disease or node-negative disease with primary tumour diameter greater than 20mm and negative progesterone receptor assay. Previous neoadjuvant chemotherapy was not permitted. Patients were randomised in a 1:1 ratio to receive 3 cycles of T→3 cycles of CEF (T→CEF; T 80mg/m2 d1, q3w→C 600mg/m2 d1, E 75mg/m2 d1, F 600mg/m2 d1, q3w) or 3 cycles of XT→3 cycles of CEX (XT→CEX; X 900mg/m2 bid d1-15 + T 60mg/m2 d1, q3w→C 600mg/m2 d1, E 75mg/m2 d1, X 900mg/m2 bid d1-15, q3w). The primary endpoint was RFS, defined as the time from randomisation to the first time the patient is recorded as having disease recurrence or the date of death if the patient dies due to causes other than disease recurrence; secondary endpoints were overall survival and safety. Results: Between January 2004 and May 2007, 1,500 women from Finland and Sweden were randomised to T→CEF (n=747) or XT→CEX (n=753). Final analysis of the primary endpoint will be presented after a median follow-up of 5 years and approximately 200 events. Data are expected in October 2010. Conclusions: The FinXX trial was the first to report the efficacy and safety of X in combination with anthracycline/taxane-containing therapy in the adjuvant treatment of early breast cancer. Final efficacy data are expected in October 2010. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr S4-1.
DOI: 10.1158/0008-5472.sabcs-82
2009
Cited 7 times
Significant improvement in recurrence-free survival (RFS) when capecitabine (X) is integrated into docetaxel (T) → 5-FU + epirubicin + cyclophosphamide (CEF) adjuvant therapy for high-risk early breast cancer (BC): interim analysis of the FinXX-trial.
Abstract Abstract #82 Background: Adding X to T extends survival in patients (pts) with metastatic BC. The primary objective of the FinXX-trial is to compare RFS with XT→CEX vs. T→CEF as adjuvant therapy in pts with early BC. Secondary objectives are comparison of overall survival, distant RFS and safety. Methods: Between Jan 2004 and May 2007, 1500 pts were randomized to receive T x 3 → CEF x 3 (T 80mg/m² d1 → C 600mg/m² d1, E 75mg/m² d1, F 600mg/m² d1, q3 wks) or XT x 3 → CEX x 3 (T 60mg/m² d1 + X 900mg/m² bid d1–15 → C 600mg/m² d1, E 75mg/m² d1, X 900mg/m² bid d115, q3 wks). Results: 1496 pts were included in the ITT analysis (two pts withdrew consent, two had metastatic disease). The study population represented a high-risk group (90% node positive, 19% HER2 positive, 88% poorly or moderately differentiated tumors). The baseline characteristics were well balanced in the two treatment arms. After median follow-up of 3 years, there have been 134 events (deaths, distant or local relapses; Table). The significantly improved RFS rate was achieved despite a higher number of study treatment discontinuations in the X-containing arm (178 vs. 23 pts in the control arm; when XT was discontinued, the remaining XT cycles were replaced with an equal number of T cycles, and CEX cycles with CEF cycles). 75% of pts in the X-containing arm received all six planned cycles of therapy compared with 96% in the control arm. The adverse effect profiles of XT→CEX and T→CEF were in part different (Gr. 3/4 neutropenic fever/infection 10.0% vs. 20.2%, hand-foot syndrome 11.1% vs. 0.3%, diarrhea 6.2% vs. 3.4%, stomatitis 4.2% vs. 1.6%, myalgia 1.9% vs. 8.0%, respectively).&amp;#x2028; &amp;#x2028; Conclusions: The primary endpoint of this trial, to demonstrate a significant improvement in RFS, was achieved after median follow-up of 3 years. The hazard ratio of 0.66 (95% CI 0.47–0.94; p=0.020) for RFS translates into a 34% reduction in the rate of recurrences or deaths in pts receiving XT→CEX. This significant benefit was seen using an X dose of 900 mg/m² bid, and with a lower T dose in the XT arm than with T monotherapy. Overall survival data are not yet mature but a trend towards improvement (hazard ratio 0.66 [95% CI 0.401.07]; p=0.089) can be seen in the XT→CEX arm. The results suggest that the risk of breast cancer recurrence can be reduced substantially with integration of capecitabine into a taxane-/anthracycline-based regimen. Citation Information: Cancer Res 2009;69(2 Suppl):Abstract nr 82.
DOI: 10.1158/0008-5472.sabcs10-pd01-02
2010
Cited 6 times
Abstract PD01-02: Integration of Capecitabine into Anthracycline-and Taxane-Based Adjuvant Therapy for Triple-Negative Early Breast Cancer: Final Subgroup Analysis of the FinXX Study
Abstract Background: Standard adjuvant chemotherapy regimens for patients with moderate-to-high risk early breast cancer typically contain a taxane, an anthracycline and cyclophosphamide. The randomised, phase III FinXX study aimed to investigate whether the integration of capecitabine into such a regimen enhances outcome. Here we present final 5-year exploratory analyses from the subgroup of patients with triple-negative early breast cancer (TNBC). Methods: Patients aged 18-65 years, with histologically confirmed invasive node positive breast cancer or node negative progesterone receptor negative breast cancer &amp;gt; 20 mm, and with a WHO performance status of 0 or 1, with no previous neoadjuvant chemotherapy were randomised. Patients received 3 x XT (capecitabine 900mg/m2 bid d1-15 + docetaxel 60mg/m2 d1)≥3 x CEX (cyclophosphamide 600mg/m2 d1, + epirubicin 75mg/m2 d1 + X 900mg/m2 bid d1-15, q3w) or 3 x T (80mg/m2 d1)≥3 x CEF (C 600mg/m2 d1, E 75mg/m2 d1, 5-FU 600mg/m2 d1, q3w). The primary endpoint was RFS, defined as the time from randomisation to the first time the patient is recorded as having disease recurrence or the date of death if the patient dies due to causes other than disease recurrence. Secondary endpoints included overall survival and safety. Results: Between January 2004 and May 2007, 1,500 women from Finland and Sweden were randomised (XT→CEX n=753, T→CEF n=747). Of these, 202 patients (13.5%) had TNBC (XT→CEX n=93, T→CEF n=109). After a median follow-up of 59 months, RFS was significantly improved in the TNBC XT→CEX arm versus control (HR 0.48, 95% CI 0.26-0.88; p=0.018). The following endpoints were also significant: 5-year RFS in patients with TNBC versus those without TNBC (HR 0.50, 95% CI 0.36-0.69; P&amp;lt;0.001); 5-year distant disease-free survival in TNBC patients receiving XT→CEX versus control (HR 0.51, p=0.035); overall survival in TNBC patients receiving XT→CEX versus control (HR 0.42, p=0.019). Conclusions: The FinXX trial is the first to report the efficacy of capecitabine in combination with anthracycline-/taxane-containing therapy in the adjuvant treatment of early breast cancer. The exploratory subgroup analysis shows that XT→CEX is more effective than T→CEF in triple-negative BC, a population with a high unmet need; XT→CEX versus control resulted in a significant improvement in RFS and OS after 5 years. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr PD01-02.
DOI: 10.1200/jco.2016.34.15_suppl.1001
2016
Cited 4 times
Adjuvant capesitabine in combination with docetaxel (T), epirubicin (E), and cyclophosphamide (C) in the treatment of early breast cancer (BC): 10-year survival results from the randomized FinXX trial.
1001 Background: The role of capecitabine (X) in the adjuvant treatment of BC is not established, and few long-term survival data are available. In an analysis of the Finland Capecitabine Trial (FinXX), based on a median FU of 59 months (JCO 2012;30:11-8), patients (pts) treated with X-containing adjuvant chemotherapy did not have significantly longer recurrence-free survival (RFS) as compared with the control group (HR 0.79, 95% CI 0.60-1.04, P= .087), but in an exploratory analysis a significant benefit emerged in the subset with triple negative BC (TNBC; ER-, PgR-, and HER2-; HR 0.48, 95% CI 0.26-0.88). Methods: Eligible pts had either axillary node-positive or high-risk node-negative BC (tumor >20 mm and PgR-). Pts were randomly assigned centrally either to 3 3-wkly cycles of docetaxel (T, 80 mg/m2) followed by 3 3-wkly cycles of C600 E75 F600 (T-CEF) or to 3 3-wkly cycles of TX (T60 , X1800 for dd. 1-15) followed by 3 3-wkly cycles of C600 E75 X1800 (TX-CEX). A total of 1,500 patients were entered to FinXX from 20 sites in Finland or Sweden between Jan 27, 2004, and May 29, 2007 (T-CEF, 747; TX-CEX, 753). Five pts withdrew consent. The data collection cut-off date for this preplanned analysis was Dec 31, 2015. Results: During a median FU of 10.3 yrs 142 RFS events occurred in the TX-CEX group and 161 in the T-CEF group (HR 0.85, 95% CI 0.68-1.07; P= .168), and 120 and 141 pts died, respectively (HR 0.83, 95% CI 0.65-1.06, P= .132; 92 vs. 113 pts died from BC). Pts with TNBC (n=202) had more favorable RFS (HR 0.43, 0.24-0.79: P= .007) and survival (HR 0.55, 0.31-0.96, P= .037) when treated with TX-CEX compared to T-CEF; no significant benefit was found in other ER/PgR/HER2-defined subgroups. The numbers of contralateral BCs (T-CEF, 24; TX-CEX, 28) and other second cancers (T-CEF, 41; TX-CEX, 35) were similar. AML was diagnosed in 4 pts. Conclusions: Integration of X to the taxane-anthracycline backbone did not prolong RFS or survival. The survival benefit in favor of TX-CEX had persisted in the TNBC subgroup, but this finding should be viewed with caution due to the small subgroup size and the exploratory nature of the analysis. Clinical trial information: NCT00114816.
DOI: 10.1200/jco.2010.28.15_suppl.531
2010
Cited 4 times
Integration of capecitabine (X) into adjuvant therapy comprising docetaxel (T) followed by 5-FU, epirubicin, and cyclophosphamide (CEF): Efficacy in patients with triple-negative breast cancer (BC).
531 Background: Interim data from this phase III randomized trial (FinXX) evaluating the integration of capecitabine into anthracycline-/taxane-based adjuvant therapy for early BC were recently published (Lancet Oncol 2009); the present subgroup analysis explores the efficacy results in patients with triple-negative BC. Methods: 1,500 patients with axillary node-positive or high-risk node-negative (tumor size > 20 mm, progesterone receptor-negative) BC were randomized to the X-containing regimen: 3x XT (X 900 mg/m2 bid d1–14 and T 60 mg/m2 iv d1, q21d) followed by 3x CEX (C 600 mg/m2 iv d1, E 75 mg/m2 iv d1 and X 900 mg/m2 bid d1–14, q21d; n = 753), or the control regimen: 3x T (80 mg/m2 iv d1, q21d) followed by 3x CEF (C 600 mg/m2, E 75 mg/m2 and F 600 mg/m2 all d1, q21d; n = 747) in January 2004 to May 2007. The primary endpoint was recurrence-free survival (RFS) and the median follow-up time 3 yrs. Results: RFS was significantly increased with the X-containing therapy compared with control (92.5% vs. 88.9%; Cox proportional hazards model hazard ratio [HR] 0.66, 95% CI 0.47–0.94; p = 0.020). Overall, patients without triple-negative disease (n = 1,294) had longer RFS than those with triple-negative cancer (n = 202; HR 0.43, 95% CI 0.29–0.63; p < 0.001). Within the triple-negative BC subgroup, patients in the X arm (n = 93) achieved a longer RFS than those in the control arm (n = 109; HR: 0.43, 95% CI 0.21–0.90; p = 0.024; 3-yr RFS 87.7% vs. 76.6%, respectively), representing a 57% reduction in the risk of breast cancer recurrence or death. RFS of patients with triple-negative BC treated with the X-containing regimen did not differ significantly from RFS of the FinXX study participants whose cancer was not triple negative (HR: 0.74, 95% CI 0.38-1.41; p = 0.357). Conclusions: In this explorative analysis the integration of X into adjuvant combination chemotherapy reduced the risk of breast cancer recurrence substantially compared with a control regimen of standard agents in patients with triple-negative early BC. The result needs to be confirmed in studies that address adjuvant X-containing chemotherapy for triple-negative BC. Author Disclosure Employment or Leadership Position Consultant or Advisory Role Stock Ownership Honoraria Research Funding Expert Testimony Other Remuneration Roche, sanofi-aventis Roche, sanofi-aventis Roche, sanofi-aventis
DOI: 10.1200/jco.2007.25.18_suppl.11035
2007
Cited 3 times
The FinXX trial: Safety results in 600 patients (pts) randomized to either docetaxel (T) followed by cyclophosphamide (C) + epirubicin (E) + 5-FU (F) (CEF) or T + capecitabine (X) followed by CEX as adjuvant therapy for early breast cancer (BC)
11035 Background: Adding X to T extends survival in pts with metastatic BC. The primary objective of the FinXX trial is to compare recurrence-free survival with XT→CEX vs T→CEF as adjuvant therapy in pts with early stage BC. Secondary objectives were comparison of safety and overall survival. Methods: Between Jan 2004 and May 2005, 600 of a planned 1,500 pts were randomized to receive T x 3 → CEF x 3 (T 80 mg/m 2 d1 → C 600 mg/m 2 d1, E 75 mg/m 2 d1, F 600 mg/m 2 d1) or TX x 3 → CEX x 3 (T 60 mg/m 2 d1 + X 900 mg/m 2 bid d1–15 → C 600 mg/m 2 d1, E 75 mg/m 2 d1, X 900 mg/m 2 bid d1–15) q3 wks. Results: Grade 3/4 toxicities (CTCAE, v3) in &gt;5% of pts are shown below. 62 pts required G-CSF. Nos. of cycles with G-CSF were: T 34; XT 15; CEF 17; CEX 15. Adverse events (AEs) led to interruption of T in 7 pts (2%) and XT in 50 pts (16%). Median dose intensities at cycle 3 were T: 95%; XT: 84/97%; CEF: 98/97/98%; CEX: 98/98/84%. T dose reductions of =20% in cycle 3 were needed in 18% and 8% of pts receiving T and XT, respectively. 4 pts died during treatment: pulmonary embolism (n=1) with T and suicide, sudden death and colitis/sepsis (each n=1) with XT. Conclusions: XT→CEX is generally well tolerated. Treatment interruptions were more common with XT→CEX, but were effective in reducing development of grade 3/4 AEs. Neutropenic infections were less frequent with XT vs T and infrequent with XT, CEF and CEX. Rates of grade 3/4 diarrhea and HFS were acceptable with XT and minimal with CEX. Accrual is nearly complete; efficacy data are expected in 2009. [Table: see text] [Table: see text]
DOI: 10.1158/0008-5472.22388264.v1
2023
Supplementary Materials and Methods, Tables 1-5 from Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer
&lt;p&gt;PDDF file - 167K&lt;/p&gt;
DOI: 10.1158/1078-0432.22461747
2023
Supplementary table and figure legends from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary table and figure legends&lt;/p&gt;
DOI: 10.1158/1078-0432.22461744
2023
Supplementary tables S1-S10 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary tables S1-S10. S1. Significant associations of the KEAP1 protein expression and receptor statuses; S2. Association of the KEAP1 protein expression with NRF2 protein expression; S3. Analyzed polymorphisms in the KEAP1 gene; S4. Significant associations of the KEAP1 SNP genotypes with KEAP1 protein expression; S5. Associations of the KEAP1 SNP s11085735 genotypes with NRF2 protein expression; S6. Associations of the KEAP1 SNPs with breast cancer survival in multivariate analysis among invasive KBCP and OBCS breast cancer cases separately; S7. Variables significantly associated with breast cancer survival in multivariate analysis among invasive KBCP and OBCS breast cancer cases; S8. Significant associations with breast cancer survival in univariate analysis (Kaplan-Meier) according to KEAP1 SNP rs11085735 genotypes among KBCP and OBCS ER positive cases, KBCP ER positive cases and KBCP cases with low/negative ({less than or equal to}1.33) KEAP1 protein expression levelS9. Variables significantly associated with breast cancer survival in multivariate analysis among ER positive KBCP breast cancer cases; S10. Variables significantly associated with breast cancer survival in multivariate analysis among (KBCP) cases with lower KEAP1 protein expression&lt;/p&gt;
DOI: 10.1158/1078-0432.22461750
2023
Supplementary figures S2-S6 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary figures S2-S6. Supplementary Figure S2: A diagram depicting the location of the studied SNPs and miR-200a binding site in &lt;i&gt;KEAP1&lt;/i&gt; gene. Supplementary Figure S3: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among the KBCP and OBCS breast cancer cases. Supplementary Figure S4: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among the ER positive cases. Supplementary Figure S5: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among KBCP cases with lower (&lt;1.33) KEAP1 protein expression. Supplementary Figure S6: Association of KEAP1 protein expression with breast cancer survival in univariate Kaplan-Meier analysis.&lt;/p&gt;
DOI: 10.1158/1078-0432.22461753.v1
2023
Supplementary Figure S1 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary Figure S1. Immunohistochemical staining of KEAP1 in breast tumor tissue.&lt;/p&gt;
DOI: 10.1158/1078-0432.22461747.v1
2023
Supplementary table and figure legends from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary table and figure legends&lt;/p&gt;
DOI: 10.1158/1078-0432.22461750.v1
2023
Supplementary figures S2-S6 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary figures S2-S6. Supplementary Figure S2: A diagram depicting the location of the studied SNPs and miR-200a binding site in &lt;i&gt;KEAP1&lt;/i&gt; gene. Supplementary Figure S3: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among the KBCP and OBCS breast cancer cases. Supplementary Figure S4: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among the ER positive cases. Supplementary Figure S5: Association of &lt;i&gt;KEAP1&lt;/i&gt; rs11085735 with breast cancer survival among KBCP cases with lower (&lt;1.33) KEAP1 protein expression. Supplementary Figure S6: Association of KEAP1 protein expression with breast cancer survival in univariate Kaplan-Meier analysis.&lt;/p&gt;
DOI: 10.1158/1078-0432.22461744.v1
2023
Supplementary tables S1-S10 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary tables S1-S10. S1. Significant associations of the KEAP1 protein expression and receptor statuses; S2. Association of the KEAP1 protein expression with NRF2 protein expression; S3. Analyzed polymorphisms in the KEAP1 gene; S4. Significant associations of the KEAP1 SNP genotypes with KEAP1 protein expression; S5. Associations of the KEAP1 SNP s11085735 genotypes with NRF2 protein expression; S6. Associations of the KEAP1 SNPs with breast cancer survival in multivariate analysis among invasive KBCP and OBCS breast cancer cases separately; S7. Variables significantly associated with breast cancer survival in multivariate analysis among invasive KBCP and OBCS breast cancer cases; S8. Significant associations with breast cancer survival in univariate analysis (Kaplan-Meier) according to KEAP1 SNP rs11085735 genotypes among KBCP and OBCS ER positive cases, KBCP ER positive cases and KBCP cases with low/negative ({less than or equal to}1.33) KEAP1 protein expression levelS9. Variables significantly associated with breast cancer survival in multivariate analysis among ER positive KBCP breast cancer cases; S10. Variables significantly associated with breast cancer survival in multivariate analysis among (KBCP) cases with lower KEAP1 protein expression&lt;/p&gt;
DOI: 10.1158/1078-0432.c.6524889
2023
Data from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;div&gt;Abstract&lt;p&gt;&lt;b&gt;Purpose:&lt;/b&gt; Defective oxidative stress response may increase cancer susceptibility. In tumors, these rescue mechanisms may cause chemo- and radioresistance impacting patient outcome. We previously showed that genetic variation in the nuclear factor erythroid 2–related factor 2 (&lt;i&gt;NFE2L2&lt;/i&gt;) is associated with breast cancer risk and prognosis. Here we further studied this pathway by investigating Kelch-like ECH-associated protein 1 (KEAP1).&lt;/p&gt;&lt;p&gt;&lt;b&gt;Experimental Design:&lt;/b&gt; Five tagging SNPs in the &lt;i&gt;KEAP1&lt;/i&gt; gene were genotyped in 996 breast cancer cases and 880 controls from two Finnish case–control sets. KEAP1 protein expression was studied in 373 invasive breast cancer tumors.&lt;/p&gt;&lt;p&gt;&lt;b&gt;Results:&lt;/b&gt; rs34197572 genotype &lt;i&gt;TT&lt;/i&gt; was associated with increased risk of breast cancer in the KBCP samples [&lt;i&gt;P&lt;/i&gt; = 1.8×10&lt;sup&gt;−4&lt;/sup&gt;; OR, 7.314; confidence interval (CI), 2.185–24.478]. rs11085735 allele &lt;i&gt;A&lt;/i&gt; was associated with lower KEAP1 protein expression (&lt;i&gt;P&lt;/i&gt; = 0.040; OR,= 3.545) and high nuclear NRF2 expression (&lt;i&gt;P&lt;/i&gt; = 0.009; OR, 2.445) and worse survival in all invasive cases (&lt;i&gt;P&lt;/i&gt; = 0.023; HR, 1.634). When including treatment data, rs11085735 was associated with recurrence-free survival (RFS; &lt;i&gt;P&lt;/i&gt; = 0.020; HR, 1.545) and breast cancer–specific survival (&lt;i&gt;P&lt;/i&gt; = 0.016; HR, 1.683) and rs34197572 with overall survival (&lt;i&gt;P&lt;/i&gt; = 0.045; HR, 1.304). rs11085735 associated with RFS also among tamoxifen-treated cases (&lt;i&gt;P&lt;/i&gt; = 0.003; HR, 3.517). Among radiotherapy-treated cases, overall survival was associated with rs34197572 (&lt;i&gt;P&lt;/i&gt; = 0.018; HR, 1.486) and rs8113472 (&lt;i&gt;P&lt;/i&gt; = 0.025; HR, 1.455). RFS was associated with rs9676881 (&lt;i&gt;P&lt;/i&gt; = 0.024; HR, 1.452) and rs1048290 (&lt;i&gt;P&lt;/i&gt; = 0.020; HR, 1.468) among all invasive cases and among estrogen receptor (ER)-positive tamoxifen-treated cases (&lt;i&gt;P&lt;/i&gt; = 0.018; HR, 2.407 and &lt;i&gt;P&lt;/i&gt; = 0.015; HR, 2.476, respectively).&lt;/p&gt;&lt;p&gt;&lt;b&gt;Conclusions:&lt;/b&gt; The present findings suggest that the investigated SNPs have effects related to oxidative stress induced by cancer treatment, supporting involvement of the NRF2/KEAP1 pathway in breast cancer susceptibility and patient outcome. &lt;i&gt;Clin Cancer Res; 21(7); 1591–601. ©2015 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1078-0432.c.6524889.v1
2023
Data from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;div&gt;Abstract&lt;p&gt;&lt;b&gt;Purpose:&lt;/b&gt; Defective oxidative stress response may increase cancer susceptibility. In tumors, these rescue mechanisms may cause chemo- and radioresistance impacting patient outcome. We previously showed that genetic variation in the nuclear factor erythroid 2–related factor 2 (&lt;i&gt;NFE2L2&lt;/i&gt;) is associated with breast cancer risk and prognosis. Here we further studied this pathway by investigating Kelch-like ECH-associated protein 1 (KEAP1).&lt;/p&gt;&lt;p&gt;&lt;b&gt;Experimental Design:&lt;/b&gt; Five tagging SNPs in the &lt;i&gt;KEAP1&lt;/i&gt; gene were genotyped in 996 breast cancer cases and 880 controls from two Finnish case–control sets. KEAP1 protein expression was studied in 373 invasive breast cancer tumors.&lt;/p&gt;&lt;p&gt;&lt;b&gt;Results:&lt;/b&gt; rs34197572 genotype &lt;i&gt;TT&lt;/i&gt; was associated with increased risk of breast cancer in the KBCP samples [&lt;i&gt;P&lt;/i&gt; = 1.8×10&lt;sup&gt;−4&lt;/sup&gt;; OR, 7.314; confidence interval (CI), 2.185–24.478]. rs11085735 allele &lt;i&gt;A&lt;/i&gt; was associated with lower KEAP1 protein expression (&lt;i&gt;P&lt;/i&gt; = 0.040; OR,= 3.545) and high nuclear NRF2 expression (&lt;i&gt;P&lt;/i&gt; = 0.009; OR, 2.445) and worse survival in all invasive cases (&lt;i&gt;P&lt;/i&gt; = 0.023; HR, 1.634). When including treatment data, rs11085735 was associated with recurrence-free survival (RFS; &lt;i&gt;P&lt;/i&gt; = 0.020; HR, 1.545) and breast cancer–specific survival (&lt;i&gt;P&lt;/i&gt; = 0.016; HR, 1.683) and rs34197572 with overall survival (&lt;i&gt;P&lt;/i&gt; = 0.045; HR, 1.304). rs11085735 associated with RFS also among tamoxifen-treated cases (&lt;i&gt;P&lt;/i&gt; = 0.003; HR, 3.517). Among radiotherapy-treated cases, overall survival was associated with rs34197572 (&lt;i&gt;P&lt;/i&gt; = 0.018; HR, 1.486) and rs8113472 (&lt;i&gt;P&lt;/i&gt; = 0.025; HR, 1.455). RFS was associated with rs9676881 (&lt;i&gt;P&lt;/i&gt; = 0.024; HR, 1.452) and rs1048290 (&lt;i&gt;P&lt;/i&gt; = 0.020; HR, 1.468) among all invasive cases and among estrogen receptor (ER)-positive tamoxifen-treated cases (&lt;i&gt;P&lt;/i&gt; = 0.018; HR, 2.407 and &lt;i&gt;P&lt;/i&gt; = 0.015; HR, 2.476, respectively).&lt;/p&gt;&lt;p&gt;&lt;b&gt;Conclusions:&lt;/b&gt; The present findings suggest that the investigated SNPs have effects related to oxidative stress induced by cancer treatment, supporting involvement of the NRF2/KEAP1 pathway in breast cancer susceptibility and patient outcome. &lt;i&gt;Clin Cancer Res; 21(7); 1591–601. ©2015 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1078-0432.22461753
2023
Supplementary Figure S1 from &lt;i&gt;KEAP1&lt;/i&gt; Genetic Polymorphisms Associate with Breast Cancer Risk and Survival Outcomes
&lt;p&gt;Supplementary Figure S1. Immunohistochemical staining of KEAP1 in breast tumor tissue.&lt;/p&gt;
DOI: 10.1158/0008-5472.22388264
2023
Supplementary Materials and Methods, Tables 1-5 from Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer
&lt;p&gt;PDDF file - 167K&lt;/p&gt;
DOI: 10.1158/1541-7786.c.6541396
2023
Data from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
&lt;div&gt;Abstract&lt;p&gt;Toll-like receptor 9 (TLR9) belongs to the innate immune system and recognizes microbial and vertebrate DNA. We showed previously that treatment with the TLR9-agonistic ODN M362 (a CpG sequence containing oligonucleotide) induces matrix metalloproteinase-13–mediated invasion in TLR9-expressing human cancer cell lines. Here, we further characterized the role of the TLR9 pathway in this process. We show that CpG oligonucleotides induce invasion in macrophages from wild-type C57/B6 and MyD88 knockout mice and in human MDA-MB-231 breast cancer cells lacking MyD88 expression. This effect was significantly inhibited in macrophages from TLR9 knockout mice and in human MDA-MB-231 breast cancer cells stably expressing TLR9 small interfering RNA or dominant-negative tumor necrosis factor receptor-associated factor 6 (TRAF6). Sequence modifications to the CpG oligonucleotides that targeted the stem loop and other secondary structures were shown to influence the invasion-inducing effect in MDA-MB-231 cells. In contrast, methylation of the cytosine residues of the parent CpG oligonucleotide did not affect the TLR9-mediated invasion compared with the unmethylated parent CpG oligonucleotide. Finally, expression of TLR9 was studied in clinical breast cancer samples and normal breast epithelium with immunohistochemistry. TLR9 staining localized in epithelial cells in both cancer and normal samples. The mean TLR9 staining intensity was significantly increased in the breast cancer cells compared with normal breast epithelial cells. In conclusion, our results suggest that TLR9 expression is increased in breast cancer and CpG oligonucleotide–induced cellular invasion is mediated via TLR9 and TRAF6, independent of MyD88. Further, our findings suggest that the structure and/or stability of DNA may influence the induction of TLR9-mediated invasion in breast cancer. (Mol Cancer Res 2008;6(10):1534–43)&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1541-7786.22516525.v1
2023
Supplementary Figure S1 from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
Supplementary Figure S1 from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
DOI: 10.1158/1541-7786.c.6541396.v1
2023
Data from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
&lt;div&gt;Abstract&lt;p&gt;Toll-like receptor 9 (TLR9) belongs to the innate immune system and recognizes microbial and vertebrate DNA. We showed previously that treatment with the TLR9-agonistic ODN M362 (a CpG sequence containing oligonucleotide) induces matrix metalloproteinase-13–mediated invasion in TLR9-expressing human cancer cell lines. Here, we further characterized the role of the TLR9 pathway in this process. We show that CpG oligonucleotides induce invasion in macrophages from wild-type C57/B6 and MyD88 knockout mice and in human MDA-MB-231 breast cancer cells lacking MyD88 expression. This effect was significantly inhibited in macrophages from TLR9 knockout mice and in human MDA-MB-231 breast cancer cells stably expressing TLR9 small interfering RNA or dominant-negative tumor necrosis factor receptor-associated factor 6 (TRAF6). Sequence modifications to the CpG oligonucleotides that targeted the stem loop and other secondary structures were shown to influence the invasion-inducing effect in MDA-MB-231 cells. In contrast, methylation of the cytosine residues of the parent CpG oligonucleotide did not affect the TLR9-mediated invasion compared with the unmethylated parent CpG oligonucleotide. Finally, expression of TLR9 was studied in clinical breast cancer samples and normal breast epithelium with immunohistochemistry. TLR9 staining localized in epithelial cells in both cancer and normal samples. The mean TLR9 staining intensity was significantly increased in the breast cancer cells compared with normal breast epithelial cells. In conclusion, our results suggest that TLR9 expression is increased in breast cancer and CpG oligonucleotide–induced cellular invasion is mediated via TLR9 and TRAF6, independent of MyD88. Further, our findings suggest that the structure and/or stability of DNA may influence the induction of TLR9-mediated invasion in breast cancer. (Mol Cancer Res 2008;6(10):1534–43)&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1541-7786.22516525
2023
Supplementary Figure S1 from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
Supplementary Figure S1 from Toll-Like Receptor 9 Mediates CpG Oligonucleotide–Induced Cellular Invasion
DOI: 10.1158/1541-7786.c.6540270.v1
2023
Data from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;div&gt;Abstract&lt;p&gt;Recent studies suggest that the ubiquitin-specific protease USP28 plays an important role in cellular repair and tissue remodeling, which implies that it has a direct role in carcinogenesis. The carcinogenic potential of USP28 was investigated in a comprehensive manner using patients, animal models, and cell culture. The findings demonstrate that overexpression of USP28 correlates with a better survival in patients with invasive ductal breast carcinoma. Mouse xenograft experiments with USP28-deficient breast cancer cells also support this view. Furthermore, lack of USP28 promotes a more malignant state of breast cancer cells, indicated by an epithelial-to-mesenchymal (EMT) transition, elevated proliferation, migration, and angiogenesis as well as a decreased adhesion. In addition to breast cancer, lack of USP28 in mice promoted an earlier onset and a more severe tumor formation in a chemical-induced liver cancer model. Mechanistically, the angio- and carcinogenic processes driven by the lack of USP28 appeared to be independent of HIF-1α, p53, and 53BP1.&lt;/p&gt;&lt;p&gt;&lt;b&gt;Implications:&lt;/b&gt; The findings of this study are not limited to one particular type of cancer but are rather applicable for carcinogenesis in a more general manner. The obtained data support the view that USP28 is involved in tumor suppression and has the potential to be a prognostic marker. &lt;i&gt;Mol Cancer Res; 16(6); 1000–12. ©2018 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1541-7786.22512534
2023
Supplemental table 2 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Overview of macroscopic changes in liver morphology.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512537
2023
Supplemental table 1 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Primers used for qRT-PCR&lt;/p&gt;
DOI: 10.1158/1541-7786.22512543
2023
Figure S3 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Knockdown of USP28 affects HIF-1a, c-Myc, c-Jun and Notch1 protein levels and enhances EMT markers in BT-549 cells.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512540
2023
Figure S4 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Knockdown of USP28 downregulates HIF-1a, c-Myc, c-Jun and Notch1 protein levels but enhances cell proliferation in MCF7 cells.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512549
2023
Figure S1 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;USP28 expression in hepatocellular carcinoma and generation of an Usp28 knockout mouse.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512546
2023
Figure S2 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Lack of USP28 does not affect p53 and 53BP1 protein levels in human cells or mouse tissue samples.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512546.v1
2023
Figure S2 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Lack of USP28 does not affect p53 and 53BP1 protein levels in human cells or mouse tissue samples.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512534.v1
2023
Supplemental table 2 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Overview of macroscopic changes in liver morphology.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512540.v1
2023
Figure S4 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Knockdown of USP28 downregulates HIF-1a, c-Myc, c-Jun and Notch1 protein levels but enhances cell proliferation in MCF7 cells.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512543.v1
2023
Figure S3 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Knockdown of USP28 affects HIF-1a, c-Myc, c-Jun and Notch1 protein levels and enhances EMT markers in BT-549 cells.&lt;/p&gt;
DOI: 10.1158/1541-7786.22512537.v1
2023
Supplemental table 1 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;Primers used for qRT-PCR&lt;/p&gt;
DOI: 10.1158/1541-7786.22512549.v1
2023
Figure S1 from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;p&gt;USP28 expression in hepatocellular carcinoma and generation of an Usp28 knockout mouse.&lt;/p&gt;
DOI: 10.1080/0284186x.2023.2213444
2023
Survival with lung cancer in Finland has not improved during 2007–2019–a single center retrospective population-based real-world study
According to the CONCORD-3 study, the 5-year survival rate of lung cancer patients in Finland has not improved during the twenty-first century. In the present study, we evaluated the survival trends of lung cancer patients diagnosed and treated in one of the five university hospitals in Finland to determine possible explanatory factors behind the lack of improved survival.This retrospective population-based study included all lung cancer patients diagnosed in Tampere University Hospital in 2007-2019 (N = 3041). The study population was divided into two subcohorts: the patients diagnosed in 2007-2012 and those diagnosed in 2013-2019. The two subcohorts were then compared to analyze the temporal changes in survival and the distribution of prognostic factors.A comparison of the patients diagnosed in 2007-2012 and 2013-2019 showed that the patients' overall survival had remained unchanged. The median overall survival was 8.7 months in the earlier subcohort and 9.2 months in the later subcohort. The respective 5-year survival rates were 16.6% and 17.8%, and these differences were not statistically significant. The proportion of stage IV patients (approximately 59% in both subcohorts) and their risk of death were similar for the two subcohorts. According to the regression analysis, male gender, advanced stage, and poor Eastern Cooperative Oncology Group performance status were independent risk factors for death, while a never-smoking status and mutation-positive disease were associated with a decreased risk of death, but only in the later cohort.Echoing the results of CONCORD-3, this study confirmed that the real-world survival of unselected lung cancer populations in Finland has not improved over the last 15 years, mainly because of the unchanged proportions of patients with late-stage lung cancer. This calls for earlier recognition of lung cancer, achieved by screening and increasing awareness of the disease.
DOI: 10.1097/hs9.0000000000000954
2023
Real-world Data: MCL2 Protocol Demonstrates Excellent Treatment Results Among Patients With Mantle Cell Lymphoma Not Fulfilling the Original Trial Inclusion Criteria
Mantle cell lymphoma (MCL) is a rare and heterogeneous subgroup of B-cell lymphomas. The clinical course of MCL varies from an indolent form to an extremely aggressive phenotype. MCL is still an incurable disease. However, intensified chemoimmunotherapy regimens and targeted therapies have increased the overall survival (OS) of patients <65 years old from 5.6 years to 8.9 years.1 Today, the standard treatment for fit patients <65–70 years is high-dose cytarabine-containing immunochemotherapy regimens followed by autologous stem cell transplantation (ASCT).2 The MCL2 protocol developed by the Nordic Lymphoma Group is one of these approaches.3,4 The MCL2 protocol has shown a median OS of 12.7 years at the 15-year follow-up and a median progression-free survival (PFS) of 8.5 years.4 Similar results have been obtained in other high-dose regimen trials.5–7 This study aimed to evaluate the efficacy of the MCL2 protocol in a population-based retrospective analysis and to compare treatment results in patients fulfilling the trial eligibility criteria with those who would have been trial ineligible. We collected data on patients diagnosed with MCL from 7 hospitals in Finland and 1 hospital in Spain from 2000 to 2020. This study was approved by the Finnish Social and Health Data Permit Authority. In total, 520 patients were included in this study. Of these, 198 patients (38%) received treatment according to the MCL2 protocol (the MCL2 group). The MCL2 protocol started with the induction treatment of rituximab–maxi-CHOP, alternating with rituximab–high-dose-cytarabine. ASCT, preceded by BEAM therapy (carmustine–etoposide–cytarabine–melphalan), was used as a consolidation therapy after 6 immunochemotherapy cycles.3 Rituximab maintenance therapy has been used as an option after ASCT in recent years. The remaining 322 patients (the other group) received rituximab combined with CHOP/CHOP-like regimens, bendamustine, or high-dose cytarabine-based regimens, MCL-FI (a Finnish protocol for elderly patients),8 single-agent rituximab, or irradiation. To evaluate the impact of the patient selection criteria on survival, we divided the patients treated with the MCL2 protocol into trial-eligible and trial-ineligible subgroups, following the MCL2 trial inclusion criteria, which were an Eastern Cooperative Oncology Group (ECOG) performance test status of 0–2, stage II–IV disease, and age <66 years.3 To obtain comparable groups, we excluded the patients with missing trial inclusion parameters in all categories. The patient-specific characteristics of the other group, the MCL2 group, and the subgroups of trial-eligible and trial-ineligible patients, along with the actual MCL2 trial,3 are summarized in Table 1. The median age was higher in the trial-ineligible group (67 years) than in the trial-eligible group (59 years), and Mantle Cell Lymphoma International Prognostic Index (MIPI) high-risk disease was more common in the trial-ineligible group (37.7% versus 20.9%). Table 1 - Patient-specific Characteristics of the Other Group, all MCL2-treated, Trial-eligible (Stage II–IV, Age <66 Years, ECOG 0–2), and Trial-ineligible Patients Other MCL2 Trial Eligible Trial Ineligible ClinicalMCL2 Trial Patient number 322 198 129 69 160 Male 229 (69.0%) 153 (77.3%) 104 (80.6%) 49 (71.0%) 113 (70.6%) Median age, y 75 (43–91) 62 (27–79) 59 (32–65) 67 (27–79) 56 (32–65) <66 years 39 (12.1%) 145 (73.2%) 129 (100%) 16 (23.2%) 160 (100%) Stage I 27 (8.4%) <5 (2.5%) 0 <5 (7.1%) 0 II 26 (8.1%) 17 (8.5%) 9 (7.0%) 8 (11.4%) 24 (15%) III 63 (19.6%) 23 (11.6%) 15 (11.6%) 8 (11.4%) IV 206 (63.9%) 154 (77.4%) 105 (81.4%) 49 (68.1%) 136 (85%) ECOGa 0 80 (26.5%) 92 (47.4%) 65 (50.4%) 27 (41.5%) 148 (93%) 1 137 (45.4%) 77 (39.7%) 54 (41.8%) 23 (35.4%) 2 50 (16.6%) 15a (7.7%) 10 (7.8%) <5 (7.7%) 12 (7%) 3 33 (10.9%) 5 (2.6%) 0 5 (7.7%) 0 4 <5 (1.6%) <5 (2.6%) 0 <5 (7.7%) 0 MIPI risk groups (15.9% missing) (14.6% missing) (17.0% missing) (10.1% missing) (1.9% missing) Low 12 (3.7%) 45 (22.7%) 38 (29.5%) 7 (10.1%) 79 (50%) Intermediate 78 (24.2%) 71 (35.9%) 42 (32.6%) 29 (42.1%) 41 (26%) High 181 (56.2%) 53 (26.8%) 27 (20.9%) 26 (37.7%) 37 (24%) aIn one of the centers, ECOG 0–2 patients were assigned to the same group (performance status at least ECOG 2).ECOG = Eastern Cooperative Oncology Group; MCL = mantle cell lymphoma; MIPI = Mantle Cell Lymphoma International Prognostic Index.The MCL2 trial patient characteristics, as referenced in the last column, have been adapted from Geisler et al.3 The MCL subtype was not reported in most pathology reports; therefore, the subgroup unclassified was used in addition to indolent, conventional, blastoid, and pleomorphic histologies. The MCL subtypes were as follows: the trial-eligible group, 77 unclassified, 21 conventional, 2 pleomorphic, 23 blastoid, and 6 missing; the trial-ineligible group, 29 unclassified, 1 indolent, 8 conventional, 2 pleomorphic, 11 blastoid, and 18 missing; and the other group, 162 unclassified, 7 indolent, 46 conventional, 12 pleomorphic, 32 blastoid, and 63 missing. The OS (the time from diagnosis to death by any cause or last follow-up date), age- and gender-standardized net survival (NS), and PFS (the time from diagnosis to progression or death by any cause) were calculated according to the Kaplan–Meier method. Analyses were performed using IBM Statistical Package for the Social Sciences (SPSS) version 27.0 (IBM Corp, Armonk, NY) and R software version 4.1.1 (R Foundation for Statistical Computing, Vienna, Austria). A P-value of <0.05 was considered statistically significant. The MIPI risk classification was calculated according to Hoster et al.9 The estimated median OS for the entire MCL2 group was 12.8 years (95% confidence interval [CI], 7.9-17.6). The median OS of the other group was 4.3 years (95% CI, 3.3-5.4). The OS was significantly better in the MCL2 group than in the other group (P < 0.001). Of the 198 patients treated with the MCL2 protocol, 129 fulfilled all MCL2 trial inclusion criteria (Table 1). Age was the major discriminator that made many of the MCL2-treated patients ineligible. The 5-year OS rate of the trial-eligible group was 79% (95% CI, 71-87) compared with 59% (95% CI, 46-74) of the trial-ineligible group. For the other group, the 5-year OS rate was 47% (95% CI, 41-53) (Figure 1A; also, 2- and 10-year OS rates are included for additional information). The median OS of the trial-eligible patients was 14.3 years (95% CI, 7.20-21.3), and the median OS of the trial-ineligible patients was 9.8 years (95% CI, 6.30-13.3). The OS was significantly improved in the trial-eligible and trial-ineligible groups compared with the other group (P < 0.001). In addition, the median OS in the trial-eligible group was improved compared with the trial-ineligible group (P < 0.001).Figure 1.: Comparison of 2-, 5-, and 10-year OS, NS, and PFS of the MCL2-treated trial-eligible and trial-ineligible groups and the other group, who did not receive MCL2 treatment. (A) OS at 2, 5, and 10 years. The trial-eligible group: 93% (95% CI, 89-98), 79% (95% CI, 71-87), and 57% (95% CI, 45-72). The trial-ineligible group: 84% (95% CI, 75-94), 59% (95% CI, 46-74), and 49% (95% CI, 34-70). The other group: 70% (95% CI, 65-75), 47% (95% CI, 41-53), and 17% (95% CI, 11-25). The OS was significantly improved in the trial-eligible and trial-ineligible groups compared with the other group (P < 0.001 in both comparisons). The median OS in the trial-eligible group was also improved compared with the trial-ineligible group (P < 0.001). (B) NS at 2, 5, and 10 years. The trial-eligible group: 95% (95% CI, 90-99), 82% (95% CI, 74-91), and 64% (95% CI, 51-80). The trial-ineligible group: 86% (95% CI, 77-96), 64% (95% CI, 51-81), and 61% (95% CI, 43-87). The other group: 76% (95% CI, 71-82), 60% (95% CI, 53-69), and 29% (95% CI, 20-44). NS was significantly improved in the trial-eligible and trial-ineligible groups compared with the other group (P < 0.001 in both comparisons). There was no statistically significant difference between the trial-eligible and trial-ineligible groups (P = 0.368). (C) PFS at 2, 5, and 10 years. The trial-eligible group: 81% (95% CI, 74-89), 62% (95% CI, 53-73), and 40% (95% CI, 29-56). The trial-ineligible group: 72% (95% CI, 62-85), 46% (95% CI, 34-62), and 23% (95% CI, 11-45). The other group: 57% (95% CI, 51-63), 30% (95% CI, 25-37), and 2% (95% CI, 0-13). PFS was significantly improved both in the trial-eligible group (P < 0.001) and trial-ineligible (P = 0.003) group compared with the other group. The median PFS in the trial-eligible group was also improved compared with the trial-ineligible group (P = 0.030). CI = confidence interval; MCL = mantle cell lymphoma; NS = net survival; OS = overall survival; PFS = progression-free survival.To determine whether the observed expansion of trial inclusion criteria in clinical practice leads to increased mortality and differences in frontline disease control, NS and PFS rates were calculated (Figure 1B, C). The 5-year NS rate of the trial-eligible group was 82% (95% CI, 74-91) compared with 64% (95% CI, 51-81) of the trial-ineligible group. For the other group, the 5-year NS rate was 60% (95% CI, 53-69) (Figure 1B; 2- and 10-year NS rates are also included for additional information). The NS was significantly improved in the trial-eligible and trial-ineligible groups compared with the other group (P < 0.001). There was no significant difference between the trial-eligible and trial-ineligible groups (P = 0.368). The 5-year PFS rate was 46% (95% CI, 34-62) in the trial-ineligible group and 62% (95% CI, 53-73) in the trial-eligible group. The PFS rate in the other group was 30% (95% CI, 25-37) at 5 years (Figure 1C; 2- and 10-year PFS rates are also included for additional information). The estimated median PFS was 6.4 years (95% CI, 3.2-9.6) in the trial-eligible group, 4.4 years (95% CI, 2.6-6.8) in the trial-ineligible group, and 2.7 years (95% CI, 2.2-3.1) in the other group. The PFS rate was significantly improved both in the trial-eligible (P < 0.001) and trial-ineligible (P < 0.003) groups compared with the other group. There was also a significant PFS improvement in the trial-eligible group compared with the trial-ineligible group (P = 0.030). There was shorter survival in the trial-ineligible group after the first relapse, with a median OS of 4.2 years, compared with 1.8 years among trial-eligible patients (data not shown). After the first relapse, 73% of the trial-ineligible patients and 88% of the trial-eligible patients received salvage chemotherapy. Rituximab maintenance every 2–3 months for 2 years is offered as a treatment option after ASCT, and in other treatment modalities for patients with MCL. In our study population, 33 patients (26%) in the trial-eligible group and 27 (39%) in the trial-ineligible group received rituximab maintenance. In the other group, 81 (25%) patients received rituximab maintenance. Only 10 patients discontinued rituximab treatment. We found that patients selected to receive the MCL2 protocol were older (median age of 62 years, compared with the trial setting, where the median age was 56 years; Table 1), but younger than the other group, whose median age was 75 years. In the MCL2 trial3 and in our study, the patients selected to receive MCL2 treatment were fit and had stage II–IV disease. Many patients had a high-risk MIPI class in the other group (56.2%) compared with the MCL2 group (26.8%) and the MCL2 trial (24%; Table 1). A previous retrospective study comparing the efficacy of R-bendamustine, R-CHOP, and MCL2 as frontline therapies showed similar age, ECOG, and MIPI class distribution in MCL2-treated patients.10 We observed an increase in MIPI high-risk patients in the trial-ineligible group (37.7%), and the inclusion of ECOG 3–4 patients, showing that aggressive disease presentation is an important factor in selecting treatment strategies in clinical practice. A previous phase 3 trial showed that an intensified treatment that includes high-dose cytarabine postpones the time-to-treatment failure in patients <65 years of age compared with anthracycline-based induction.7 Also, the Nordic MCL2 phase 2 study demonstrated a promising median OS of 12.7 years and a PFS of 8.5 years. Since the MCL2 and MCL3 trials3,11 were conducted, the upper age limit for this therapy has gradually increased to 75 years at some centers, and the therapy has been offered to patients with ECOG > 2 and more comorbidities, as also shown in this study. To date, no data on whether the results of the MCL2 and MCL3 trials can be extrapolated to real-world populations exist. We demonstrated an even higher OS of 14.3 years in a real-world setting among trial-eligible patients. Among trial-ineligible patients, the median OS was also remarkable (9.8 years). Albertsson-Linblad and colleagues10 reported a median OS of 4.9 years in patients receiving any systemic therapy in the frontline and also showed that both MCL2 and RB are superior over R-CHOP. We observed that the PFS improvement was greater in trial-eligible patients than in trial-ineligible patients (6.4 versus 4.4 years). The trial-ineligible patients were older and probably frailer with more comorbidities; therefore, the trial-eligible patients may also have better frontline disease control. Notably, we found no significant difference in the NS, suggesting that the MCL2 approach outside clinical trial inclusion does not substantively increase mortality. However, there are several limitations in our study. This was a retrospective study and the MCL subtype was not reported in most pathology reports. Also the patients with missing trial inclusion criteria data were excluded, although the subgroups were balanced for the analyses. To conclude, this study confirmed that the MCL2 protocol is an effective treatment approach for real-world patients with MCL. Importantly, the data show the importance of the expansion of trial inclusion criteria in real-world practice, as patients over 65 years of age may also benefit from the MCL2 strategy. AUTHOR CONTRIBUTIONS MH and TK did research design, data collection, analysis, and writing. OK and ARö did research design, funding, analysis, and writing. MS did data collection, analysis, and writing. TS did data analysis and figure preparation. MK, RP, EK, EA, ARa, KS, AJ, EJ, SK, JMS, and HK did data collection. All authors have read and approved the article. DISCLOSURES The authors have no conflicts of interest to disclose. SOURCES OF FUNDING This work was funded by North Ostrobothnia Health Care District.
DOI: 10.1158/0008-5472.c.6502184
2023
Data from Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer
&lt;div&gt;Abstract&lt;p&gt;Triple-negative breast cancers are an aggressive subtype of breast cancer with poor survival, but there remains little known about the etiologic factors that promote its initiation and development. Commonly inherited breast cancer risk factors identified through genome-wide association studies display heterogeneity of effect among breast cancer subtypes as defined by the status of estrogen and progesterone receptors. In the Triple Negative Breast Cancer Consortium (TNBCC), 22 common breast cancer susceptibility variants were investigated in 2,980 Caucasian women with triple-negative breast cancer and 4,978 healthy controls. We identified six single-nucleotide polymorphisms, including rs2046210 (&lt;i&gt;ESR1&lt;/i&gt;), rs12662670 (&lt;i&gt;ESR1&lt;/i&gt;), rs3803662 (&lt;i&gt;TOX3&lt;/i&gt;), rs999737 (&lt;i&gt;RAD51L1&lt;/i&gt;), rs8170 (19p13.1), and rs8100241 (19p13.1), significantly associated with the risk of triple-negative breast cancer. Together, our results provide convincing evidence of genetic susceptibility for triple-negative breast cancer. &lt;i&gt;Cancer Res; 71(19); 6240–9. ©2011 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1541-7786.c.6540270
2023
Data from USP28 Deficiency Promotes Breast and Liver Carcinogenesis as well as Tumor Angiogenesis in a HIF-independent Manner
&lt;div&gt;Abstract&lt;p&gt;Recent studies suggest that the ubiquitin-specific protease USP28 plays an important role in cellular repair and tissue remodeling, which implies that it has a direct role in carcinogenesis. The carcinogenic potential of USP28 was investigated in a comprehensive manner using patients, animal models, and cell culture. The findings demonstrate that overexpression of USP28 correlates with a better survival in patients with invasive ductal breast carcinoma. Mouse xenograft experiments with USP28-deficient breast cancer cells also support this view. Furthermore, lack of USP28 promotes a more malignant state of breast cancer cells, indicated by an epithelial-to-mesenchymal (EMT) transition, elevated proliferation, migration, and angiogenesis as well as a decreased adhesion. In addition to breast cancer, lack of USP28 in mice promoted an earlier onset and a more severe tumor formation in a chemical-induced liver cancer model. Mechanistically, the angio- and carcinogenic processes driven by the lack of USP28 appeared to be independent of HIF-1α, p53, and 53BP1.&lt;/p&gt;&lt;p&gt;&lt;b&gt;Implications:&lt;/b&gt; The findings of this study are not limited to one particular type of cancer but are rather applicable for carcinogenesis in a more general manner. The obtained data support the view that USP28 is involved in tumor suppression and has the potential to be a prognostic marker. &lt;i&gt;Mol Cancer Res; 16(6); 1000–12. ©2018 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/0008-5472.c.6502184.v1
2023
Data from Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer
&lt;div&gt;Abstract&lt;p&gt;Triple-negative breast cancers are an aggressive subtype of breast cancer with poor survival, but there remains little known about the etiologic factors that promote its initiation and development. Commonly inherited breast cancer risk factors identified through genome-wide association studies display heterogeneity of effect among breast cancer subtypes as defined by the status of estrogen and progesterone receptors. In the Triple Negative Breast Cancer Consortium (TNBCC), 22 common breast cancer susceptibility variants were investigated in 2,980 Caucasian women with triple-negative breast cancer and 4,978 healthy controls. We identified six single-nucleotide polymorphisms, including rs2046210 (&lt;i&gt;ESR1&lt;/i&gt;), rs12662670 (&lt;i&gt;ESR1&lt;/i&gt;), rs3803662 (&lt;i&gt;TOX3&lt;/i&gt;), rs999737 (&lt;i&gt;RAD51L1&lt;/i&gt;), rs8170 (19p13.1), and rs8100241 (19p13.1), significantly associated with the risk of triple-negative breast cancer. Together, our results provide convincing evidence of genetic susceptibility for triple-negative breast cancer. &lt;i&gt;Cancer Res; 71(19); 6240–9. ©2011 AACR&lt;/i&gt;.&lt;/p&gt;&lt;/div&gt;
DOI: 10.6084/m9.figshare.23272331
2023
Survival with lung cancer in Finland has not improved during 2007–2019–a single center retrospective population-based real-world study
According to the CONCORD-3 study, the 5-year survival rate of lung cancer patients in Finland has not improved during the twenty-first century. In the present study, we evaluated the survival trends of lung cancer patients diagnosed and treated in one of the five university hospitals in Finland to determine possible explanatory factors behind the lack of improved survival. This retrospective population-based study included all lung cancer patients diagnosed in Tampere University Hospital in 2007–2019 (<i>N</i> = 3041). The study population was divided into two subcohorts: the patients diagnosed in 2007–2012 and those diagnosed in 2013–2019. The two subcohorts were then compared to analyze the temporal changes in survival and the distribution of prognostic factors. A comparison of the patients diagnosed in 2007–2012 and 2013–2019 showed that the patients’ overall survival had remained unchanged. The median overall survival was 8.7 months in the earlier subcohort and 9.2 months in the later subcohort. The respective 5-year survival rates were 16.6% and 17.8%, and these differences were not statistically significant. The proportion of stage IV patients (approximately 59% in both subcohorts) and their risk of death were similar for the two subcohorts. According to the regression analysis, male gender, advanced stage, and poor Eastern Cooperative Oncology Group performance status were independent risk factors for death, while a never-smoking status and mutation-positive disease were associated with a decreased risk of death, but only in the later cohort. Echoing the results of CONCORD-3, this study confirmed that the real-world survival of unselected lung cancer populations in Finland has not improved over the last 15 years, mainly because of the unchanged proportions of patients with late-stage lung cancer. This calls for earlier recognition of lung cancer, achieved by screening and increasing awareness of the disease.
DOI: 10.1158/1538-7445.sabcs14-p5-04-13
2015
Abstract P5-04-13: Toll like receptor-9 and CD73 may act on the same pathway to induce immunosuppression in triple negative breast cancer cells
Abstract Toll like-receptor-9 (TLR9) is an innate immune system DNA-receptor which is also widely expressed in breast cancer cell lines and in clinical breast cancer specimens. Although TLR9 ligands (such as bacterial DNA and synthetic, CpG-sequence containing oligonucleotides) induce TLR9-mediated invasion in breast cancer cells in vitro, the contribution of this protein to breast cancer pathophysiology remains unclear. We showed previously that tumor TLR9 expression is a highly significant prognostic factor in breast cancer, but only in tumors that are triple negative (TNBC). Specifically, low tumor TLR9 expression in TNBC is associated with a significantly shortened disease-specific survival. Our published, preclinical results further suggest that tumor TLR9 expression may be an important determinant of tumor immunophenotype and that patients with low-TLR9 TNBC may not gain the immunogenic benefit from chemotherapy. A possible mechanistic explanation for the change in the immunophenotype of low TLR9-TNBC tumors involves CD73, which is a 5’ectonucleotidase that converts extracellular adenosine monophosphate (AMP) into the highly immunosuppressive adenosine. CD73 is also expressed in breast cancer cells and in clinical specimens. High CD73 expression has been associated with poor prognosis, but similar with TLR9 expression, the prognostic significance was specific only for TNBC. There are no previous reports on TLR9 regulation of CD73 in any cancer. Interestingly, however, TLR9 deficiency was shown to promote CD73 expression of T-cells in a mouse model of diabetes. To begin to test the hypothesis that TLR9 affects CD73 expression in TNBC, we studied CD73 mRNA and protein expression in control and TLR9 siRNA TNBC cells in vitro. We discovered that TLR9 siRNA TNBC cells express significantly higher CD73 mRNA concentrations than control siRNA cells specifically in hypoxia. Similar findings were observed with immunofluorescence. Our results suggest that the lack or TLR9 in TNBC may allow high CD73 expression and through this mechanism result in an immunosuppressive phenotype of the tumors. The immunosuppression then would explain the poor prognosis associated with low TLR9-TNBC tumors. We are currently studying this with clinical TNBC specimens. Citation Format: Katri S Selander, Johanna M Tuomela, Mikko Mella, Joonas Kauppila, Jouko Sandholm, Gennady Yegutkin, Peeter Karihtala, Arja Jukkola-Vuorinen, Kirsi-Maria Haapasaari, Katri S Vuopala, Kevin W Harris. Toll like receptor-9 and CD73 may act on the same pathway to induce immunosuppression in triple negative breast cancer cells [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P5-04-13.