Skip to main content
Erschienen in: BMC Cancer 1/2018

Open Access 01.12.2018 | Research article

Variants of cancer susceptibility genes in Korean BRCA1/2 mutation-negative patients with high risk for hereditary breast cancer

verfasst von: Ji Soo Park, Seung-Tae Lee, Eun Ji Nam, Jung Woo Han, Jung-Yun Lee, Jieun Kim, Tae Il Kim, Hyung Seok Park

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

We evaluated the incidence and spectrum of pathogenic and likely pathogenic variants of cancer susceptibility genes in BRCA1/2 mutation-negative Korean patients with a high risk for hereditary breast cancer using a comprehensive multigene panel that included 35 cancer susceptibility genes.

Methods

Samples from 120 patients who were negative for BRCA1/2 mutations, but had been diagnosed with breast cancer that was likely hereditary, were prospectively evaluated for the prevalence of high-penetrance and moderate-penetrance germline mutations.

Results

Nine patients (7.5%) had at least one pathogenic or likely pathogenic variant. Ten variants were identified in these patients: TP53 in two patients, PALB2 in three patients, BARD1 in two patients, BRIP1 in two patients, and MRE11A in one patient. We also identified 30 types of 139 variants of unknown significance (VUS). High-penetrance germline mutations, including TP53 and PALB2, tended to occur with high frequency in young (< 35 years) breast cancer patients (4/19, 21.1%) than in those diagnosed with breast cancer at ≥35 years of age (1/101, 1.0%; p = 0.003).

Conclusions

These combined results demonstrate that multigene panels offer an alternative strategy for identifying veiled pathogenic and likely pathogenic mutations in breast cancer susceptibility genes.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12885-017-3940-y) contains supplementary material, which is available to authorized users.
Abkürzungen
ACMG
American College of Medical Genetics and Genomics
AJCC
American Joint Committee on Cancer
BAM
Binary alignment map
BARD1
BRCA1-associated RING domain 1
BRIP1
BRCA1-interacting protein C-terminal helicase 1
CHEK2
Cell cycle checkpoint kinase 2
CI
Confidence interval
CNV
Copy number variants
ExAC
Exome Aggregation Consortium
F-CSGE
Fluorescent-conformation sensitive gel electrophoresis
HBOC
Hereditary breast and ovarian cancers
MLPA
Multiplex ligation-dependent probe amplification
NGS
Next generation sequencing
OR
Odds ratio
PM
Pathogenic criterion weighted as moderate
PP
Pathogenic criterion weighted as supporting
PVS
Pathogenic criterion weighted as very strong
SAM
Sequence alignment map
VUS
Variants of unknown significance

Background

The identification of BRCA1 and BRCA2 germline mutations as predictors of cancer susceptibility significantly improved the diagnosis and prevention of hereditary breast and ovarian cancers (HBOC). Recent advances in genetic testing have enabled the discovery of novel genes that increase the risk of cancer in patients with familial predisposition. Multiple research laboratories have evaluated these cancer-associated mutations in patients who are negative for BRCA1/2 mutations, but still have a high risk of HBOC. These efforts have identified mutations in moderate-risk genes, such as ATM, BRIP1, CHEK2, BARD1, MRE11A, NBN, RAD50, RAD51, and XRCC2, as well as those in high-penetrance genes, including TP53, PTEN, STK11, CDH1, and PALB2, have been reported across diverse ethnic populations [1].
Next generation sequencing (NGS) can provide detailed genetic information via multi-gene panel assays [2]. However, the application of NGS multigene panel test in a clinical setting represents a considerable challenge. It is necessary to not only validate this novel technique, but also to select candidate susceptibility genes. Furthermore, mutations indicative of cancer susceptibility vary across ethnicities; therefore, it is important to understand the clinical and genetic characteristics of multiple susceptibility genes identified by NGS multigene panels in each ethnic population.
In this study, we used comprehensive multigene panels that included 35 known or suspected cancer susceptibility genes to examine BRCA1/2 mutation-negative Korean patients who had clinical features indicative of hereditary breast cancer. We also investigated the feasibility of multigene panel testing for Korean patients, and evaluated potential clinicopathological risk factors related to germline mutations other than BRCA1/2.

Methods

Study population

The study population included 182 Korean BRCA1/2 mutation-negative breast cancer patients with a familial predisposition who were referred to the Cancer Prevention Center, Yonsei Cancer Center, Seoul, Korea between March 1, 2015 and November 11, 2016. Sixty-two patients opted to not participate. Finally, a total of 120 patients were enrolled in the study. Suspected clinical features of hereditary breast cancer were defined as follows: (1) at least one case of breast or ovarian cancer in first- or second-degree relatives; (2) a first diagnosis of breast cancer before age 40; (3) bilateral breast cancer; and (4) co-diagnosis of breast and ovarian cancers in the same patient.

Panel-based mutation analysis

Germline DNA was extracted from the participants’ peripheral blood samples. We used a customized targeted capture sequencing panel (OncoRisk®, Celemics, Seoul, Korea) which included all coding sequences and intron-exon boundaries of the coding exon from 35 cancer predisposition genes (BRCA1, BRCA2, PALB2, BARD1, BRIP1, RAD51C, RAD51D, RAD50, NBN, MRE11A, ATM, CHEK2, TP53, PTEN, APC, BLM, BMPR1A, CDH1, CDK4, CDKN2A, EPCAM, MEN1, MLH1, MSH2, MSH6, MUTYH, PMS2, POLE, PRSS1, RET, SLX4, SMAD4, STK11, VLH, and WT1). Products with each capture reaction were sequenced by 100 base pair paired-end reads on a MiSeq platform (Illumina, San Diego, CA). High-quality sequencing data with an average depth of 500−1000 folds were obtained.
We identified all single base pair substitutions, insertion-deletions, and copy number variants (CNVs) in each gene. Split-read-based detection of large insertions and deletions was conducted using the Pindel and Manta algorithms. CNVs detected by ExomeDepth software [3] were further crosschecked with our custom pipelines, which retrieved base-level depth of coverage for each binary alignment map (BAM) file using SAMtools software (http://​samtools.​sourceforge.​net) and normalized the depths in the same batch (Additional file 1: Figure S1). All likely deleterious mutations were validated by Sanger sequencing, and all possible large rearrangements were confirmed by the multiplex ligation-dependent probe amplification (MLPA) method (Additional file 1: Figure S2).
Genetic variants were classified using a five-tier system following guidelines from the American College of Medical Genetics and Genomics (ACMG) as follows: pathogenic, likely pathogenic, variants of unknown significance (VUS), likely benign, or benign/polymorphism [4]. We used the Sorting Intolerant From Tolerant (SIFT, http://​sift.​bii.​a-star.​edu.​sg/​) and Polymorphism Phenotyping-2 (PolyPhen-2, http://​genetics.​bwh.​harvard.​edu/​pph2) to generate in silico predictions of several of the identified nonsynonymous variants. Using large rearrangements of exons, pathogenic and likely pathogenic variants were considered as mutations, for consistency with previous studies [5].

Results

Baseline characteristics of the patients are presented in Additional file 2: Table S1. A total of 7.5% (9/120) of patients were found to carry at least one pathogenic or likely pathogenic variant. A total of ten gene variants (Fig. 1a) were identified in nine patients: TP53 in two patients, PALB2 in three patients, BARD1 in two patients, BRIP1 in two patients, and MRE11A in one patient. We detected a large deletion from exon 2−9 in the TP53 gene, and the other pathogenic variants identified were as follows: PALB2 (c.3267_3268delGT, p.Phe1090SerfsTer6, rs587781890; c.2257C > T, p.Arg753Ter, rs180177110; and c.695delC, p.Gly232ValfsTer6); BARD1 (c.1345C > T, p.Gln449Ter); BRIP1 (c.1066C > T, p.Arg356Ter, rs730881633; and exon 5–6 deletion); and MRE11A (c.1773_1774delAA, p.Gly593LysfsTer4). Likely pathogenic variants were found in TP53 (c.733G > A, p.Gly245Ser, rs28934575). Pathogenic variants in PALB2 and MRE11A were identified in a 34-year-old patient who was co-diagnosed with breast and gastric cancer (Table 1). Three of the pathogenic variants identified in this study were not reported previously.
Table 1
Characteristics of patients with pathogenic or likely pathogenic variants
Case number
Site/histology of breast cancer
Breast cancer subtype
Breast cancer stage (AJCC 7th ed)
Concomitant cancers
Affected gene
Nucleotide change
Amino acid change
dbSNP
Variant effect
Family cancer history (family member, age)
MAF by ExAC (n = 60,704)
MAF by ExAC Asian (n = 12,583)
MAF by KRGDB(n = 622)
Confirmation method
Pathogenicity
Reference
1
L/IDC
ER+/PR+/HER2-
IIA
TP53
exon2–9 deletion
N/A
Large deletion
Breast ca (mother, 32)
N/A
N/A
N/A
MLPA
Pathogenic
 
2
B/IDC
ER+/PR+/HER2-
IIA
PALB2
c.3267_3268delGT
p.Phe1090SerfsTer6
rs587781890
Frameshift
Breast ca (aunt, 47),
Colon ca (GF, 60),
Stomach ca (GM, 60)
**
**
**
Sanger sequencing
Likely pathogenic
 
3
R/IDC
ER+/PR+/HER2-
IIB
AoV
PALB2
c.2257C > T
p.Arg753Ter
rs180177110
Nonsense
Breast ca (sister, 53)
3.29 × 10−5
**
**
Sanger sequencing
Pathogenic
 
4*
L/poorly differentiated
TNBC
IA
Stomach
PALB2
c.695delG
p.Gly232ValfsTer6
Frameshift
Stomach ca (GF, 90),
Liver ca (uncle, 60)
**
**
**
Sanger sequencing
Likely pathogenic
 
4*
L/poorly differentiated
TNBC
IA
Stomach
MRE11A
c.1773_1774delAA
p.Gly593LysfsTer4
Frameshift
Stomach ca (GF, 90),
Liver ca (uncle, 60)
**
**
**
Sanger sequencing
Likely pathogenic
 
5†
L/mucinous
TNBC
IA
BARD1
c.1345C > T
p.Gln449Ter
Nonsense
Breast ca (sister1, 67; sister2, 47)
**
**
**
Sanger sequencing
Likely pathogenic
 
6†
L/IDC
ER+/PR-/HER2-
IIA
BARD1
c.1345C > T
p.Gln449Ter
Nonsense
Breast ca (sister1, 67; sister2, 58)
**
**
**
Sanger sequencing
Likely pathogenic
 
7
L/IDC
ER-/PR-/HER2+
IA
BRIP1
exon5–6 deletion
N/A
Largedeletion
Ovarian ca (mother, 35)
N/A
N/A
N/A
MLPA
Pathogenic
 
8
R/IDC
ER-/PR-/HER2+
IA
Cervix uteri
BRIP1
c.1066C > T
p.Arg356Ter
rs730881633
Nonsense
Breast ca (sister, 40)
**
**
**
Sanger sequencing
Likely pathogenic
 
9
B/IDC
ER-/PR-/HER2+
IIA
TP53
c.733G > A
p.Gly245Ser
rs28934575
Missense
Stomach ca (father, 56); Pancreatic ca (father, 73)
8.24 × 10−6
**
**
Sanger sequencing
Likely pathogenic (Table S2)
[23]
Abbreviation: AJCC, American Joint Committee on Cancer; AoV, ampulla of Vater; B: bilateral; ca: cancer; dbSNP, single nucleotide polymorphism database; DCIS, ductal carcinoma in situ; ER, estrogen receptor; ExAC, Exome Aggregation Consortium; HER2, human epidermal growth factor receptor 2; IDC, invasive ductal carcinoma; KRGDB, Korean Reference Genome database; L, left; N/A, not assessable; MAF, minor allele frequency; MLPA, multiplex ligation-dependent probe amplification; Polyphen, Polymorphism Phenotyping-2; PR, progesterone receptor; R, right; SIFT, Sorting Intolerant From Tolerant; TNBC, triple negative breast cancer
*Case 4 had pathogenic variants in PALB2 and MRE11A. Case 5 and Case 6 are siblings. **There was no case with the relevant variant in the databases with respect to the general population
A total of 87 patients (72.5%) had at least one VUS (median, 1; range, 0–3). A total of 139 VUS were identified in 30 cancer susceptibility genes, including SLX4 (n = 11), BLM (n = 10), POLE (n = 10), ATM (n = 9), CDH1 (n = 9), CHEK2 (n = 9), BRCA2 (n = 8), RAD50 (n = 7), BRIP1 (n = 6), EPCAM (n = 5), PALB2 (n = 5), PRSS1 (n = 5), TP53 (n = 5), APC (n = 4), MLH1 (n = 4), RET (n = 4), MRE11A (n = 3), MSH2 (n = 3), MSH6 (n = 3), MUTYH (n = 3), RAD51D (n = 3), STK11 (n = 3), BMPR1A (n = 2), BRCA1 (n = 2), CDKN2A (n = 1), MEN1 (n = 1), NBN (n = 1), PMS2 (n = 1), VHL (n = 1), and WT1 (n = 1) (Fig. 1b).
First diagnosis of breast cancer at a relatively young age (<35 years) was correlated with pathogenic or likely-pathogenic variants in high-penetrance cancer susceptibility genes. Pathogenic variants in high-penetrance genes were detected in 21.1% (4/19) of these patients, which was significantly higher than that for patients who were first diagnosed with breast cancer at age ≥ 35 years (1/101, 1.0%, p = 0.003) (Table 2).
Table 2
Association between the clinicopathological features of suspected hereditary breast cancer and the pathogenic or likely pathogenic variants of non-BRCA cancer predisposition genes (n = 120 patients)
Clinicopathological features
 
High-penetrance mutations
Moderate-penetrance mutations
None or VUS
 
  
Number ofpatients
%
Number ofpatients
%
Number ofpatients
%
p-value
Breast cancer site
 
Bilateral
2
18.2
0
0
9
81.8
0.106*
 
Unilateral
3
2.8
4
3.7
102
93.5
 
Breast cancer subtype (n = 117, excluding patients with unknown breast cancer subtypes)
 
TNBC
0
0
1
4.5
21
95.5
>0.99*
 
hormone + and/or HER2+
4
4.2
3
3.2
88
92.6
 
Concomitant diagnosis with ovarian cancer
 
Yes
0
0
0
0
3
100
>0.99*
 
No
5
4.3
4
3.4
108
92.3
 
Age at first diagnosis of breast cancer
 
< 35 years
4
21.1
0
0
15
78.9
0.003*
 
≥ 35 years
1
1.0
4
4.0
96
95.0
 
Family history of young (< 50 years old at diagnosis) breast and/or ovarian cancer patients within 2nd degree family
 
Yes
2
6.3
3
9.4
27
84.3
0.053*
 
No
3
3.4
1
1.1
84
95.5
 
Abbreviations: HER2, human epidermal growth factor receptor 2; TNBC, triple negative breast cancer; VUS, variant of unknown significance. *Analyzed using Fisher’s exact test

Discussion

Previous studies using multigene panel tests identified cancer susceptibility genes in 2.1−16.8% of BRCA1/2 mutation-negative patients [511]. Our tests of high-penetrance genes identified a large exon deletion in TP53, and pathogenic and likely pathogenic variants in TP53 and PALB2 (Table 1). We also identified a frameshift mutation of MRE11A c.1773_1774delAA (p.Gly593LysfsTer4) in a patient with a PALB2 mutation. The MRE11 protein functions in non-homologous end-joining and homologous recombination, which occur during the repair of double-stranded DNA breaks [12]. Therefore, the risk for patients with concurrent dysfunction in PALB2 and MRE11A is unclear and should be assessed in future studies. Because the two frameshift variants in PALB2 (c.3267_3268delGT, p.Phe1090SerfsTer6, rs587781890; and c.695delG, p.Gly232ValfsTer6) were not found in the control group, the variants met the criteria to be likely pathogenic according to the ACMG guideline (PVS1 and PM2) (Table 1) [4]. One nonsense variant in PALB2 (c.2257C > T p.Arg753Ter, rs180177110) had a higher prevalence in affected patients compared to the control group [odds ratio (OR), 127.0; 95% confidence interval (CI), 14.1–1140.1; p < 0.0001]. Therefore, this variant conformed to the criteria to be classified as pathogenic according to ACMG guidelines (PVS1 and PS4) (Table 1) [4]. In addition, a missense variant in TP53, c.733G > A (p.Gly245Ser, rs28934575) was classified as a pathogenic or likely pathogenic variant in the ClinVar database (http://​www.​ncbi.​nlm.​nih.​gov/​clinvar/​), and met the criteria for a likely pathogenic variant according to the ACMG guidelines (PM2, PM5, PP2, PP3, and PP5) (Additional file 2: Table S2) [4].
Pathogenic or likely pathogenic variants also were detected in BRCA1-associated RING domain 1 (BARD1) and BRCA1-interacting protein C-terminal helicase 1 (BRIP1). BARD1 and BRIP1 encode proteins that interact with the BRCA1 protein during the repair of DNA double- stranded break, and pathogenic variants of these genes have been investigated [13]. However, there is a controversy as to whether these rare variants are clinically associated with a risk of breast cancer [11, 14]. In a previous study that screened for BRIP1 mutations among 235 Korean patients with BRCA1/2 mutation-negative high-risk breast cancers using fluorescent-conformation sensitive gel electrophoresis (F-CSGE), there was no case of a protein-truncating BRIP1 mutation, which suggests that the prevalence of BRIP1 mutations is likely to be low in the Korean population [15].
Cell cycle checkpoint kinase 2 (CHEK2) is a well-established moderate-penetrance breast cancer gene. Several studies have shown that essentially no case of CHEK2 (c.1100delC) was observed in Asian populations, in contrast to the observed prevalence in European populations [1619]. Liu and colleagues reported that the CHEK2 c.1111C > T (p.His371Tyr, rs531398630) variant was observed in 4.24% (5/118) of Chinese familial breast cancer cases without BRCA1/2 mutations, and was associated with dysfunctional phosphorylation of T68 in the SQ/TQ rich domain, which is an activation point following DNA damage [18]. We also identified CHEK2 c.1111C > T variants in 2.5% (3/120) of Korean breast cancer patients without BRCA1/2 mutations (Additional file 2: Table S2). Population-based investigations are required to establish the prevalence of this variant, especially in Asian patients. We identified the CHEK2 c.908 + 2delT variant in one patient, and it was classified as likely pathogenic according to the ACMG guideline (Additional file 2: Table S2). However, we did not classify this variant as a positive result because the experimental study was not sufficient.
In the current study, clinically important likely pathogenic or pathogenic variants of high-penetrance genes were identified in only five (4.2%) patients (TP53 in two patients, and PALB2 in three patients). These variants were identified in 4 of 19 patients (21.1%) with early-onset breast cancer (< 35 years old at onset) (Table 2). A previous study identified cancer susceptibility mutations in 11% of BRCA1/2-negative patients with early-onset breast cancer (diagnosed at <40 years of age) [20]. Considering the frequency of pathogenic variants of high-penetrance genes in patients with early-onset cancer, clinicians should be encouraged to consider performing multigene panel tests for these patients if their conventional BRCA1/2 tests are negative.
This study has several limitations. The primary limitation is the small number of patients (n = 120), which provides only limited data for cancer susceptibility genes in Korean patients with breast cancer. A large-scale cohort study will be required to establish the accurate prevalence and spectrum of pathogenic variants in these patients. The majority of patients (87 of the 120, 72.5%) had VUS. A functional and population-based study will be necessary to clarify the clinical meaning of these VUS. Despite these limitations, to the best of our knowledge, this is the first prospective study to apply customized multigene panels to BRCA1/2 mutation-negative Korean patients with a high risk for HBOC. A recent study conducted by Couch et al. assessed the commercial multigene panel test results of 65,057 patients with breast cancer; however, the frequency, phenotypic association, and cancer risks related to each variant were analyzed among Caucasian women only [11]. Regarding diversity of prevalence of the genetic variants, more prospective studies will be required among diverse ethnic populations.

Conclusions

Wider application of multigene panel tests that include high-penetrance cancer susceptibility genes, so-called “beyond BRCA1/2 genes”, will likely provide clinically relevant information for some patients with high risk for hereditary cancer [1, 13, 21]. However, these panels can produce abundant and conflicting results in clinical practice. To efficiently utilize these data, clinical databases should be established with respect to ethnic backgrounds, and genetic results should be carefully applied for high-risk patients.

Acknowledgements

We are very grateful for the participation in this study of patients and staffs from Breast Cancer Center and Cancer Prevention Center at Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea. The results of this study were presented as a poster at the 15th St.Gallen International Breast Cancer Conference 2017 held on March 15th-18th, 2017, Vienna, Austria [22]. This manuscript includes the abstract presented at the 15th St.Gallen International Breast Cancer Conference 2017.

Funding

This research was supported by the Korea Breast Cancer Foundation (KBCF-2015E002) and the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2016R1D1A1B03934564).

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.
The prospective study was approved by institutional review board at Severance Hospital, Seoul, Korea (IRB approval number 4–2015-0819). We obtained informed consent in writing from all patients who participated in this study.
Not applicable.

Competing interests

The authors declare no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Young EL, Feng BJ, Stark AW, Damiola F, Durand G, Forey N, Francy TC, Gammon A, Kohlmann WK, Kaphingst KA, et al. Multigene testing of moderate-risk genes: be mindful of the missense. J Med Genet. 2016;53(6):366–76.CrossRefPubMedPubMedCentral Young EL, Feng BJ, Stark AW, Damiola F, Durand G, Forey N, Francy TC, Gammon A, Kohlmann WK, Kaphingst KA, et al. Multigene testing of moderate-risk genes: be mindful of the missense. J Med Genet. 2016;53(6):366–76.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Park HS, Park JS, Nam EJ, Lee S-T, Han JW, Kim TI. Clinical implications of genetic testing for hereditary breast and ovarian cancer syndrome in the era of genomic medicine: Clinician’s perspectives. J Breast Dis. 2016;4(1):1–9.CrossRef Park HS, Park JS, Nam EJ, Lee S-T, Han JW, Kim TI. Clinical implications of genetic testing for hereditary breast and ovarian cancer syndrome in the era of genomic medicine: Clinician’s perspectives. J Breast Dis. 2016;4(1):1–9.CrossRef
3.
Zurück zum Zitat Plagnol V, Curtis J, Epstein M, Mok KY, Stebbings E, Grigoriadou S, Wood NW, Hambleton S, Burns SO, Thrasher AJ, et al. A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics. 2012;28(21):2747–54.CrossRefPubMedPubMedCentral Plagnol V, Curtis J, Epstein M, Mok KY, Stebbings E, Grigoriadou S, Wood NW, Hambleton S, Burns SO, Thrasher AJ, et al. A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics. 2012;28(21):2747–54.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRefPubMedPubMedCentral Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, Vaccari EM, Bissonnette J, Booker JK, Cremona ML, et al. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Genet Med. 2016;18(8):823–32.CrossRefPubMed Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, Vaccari EM, Bissonnette J, Booker JK, Cremona ML, et al. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Genet Med. 2016;18(8):823–32.CrossRefPubMed
6.
Zurück zum Zitat Hirotsu Y, Nakagomi H, Sakamoto I, Amemiya K, Oyama T, Mochizuki H, Omata M. Multigene panel analysis identified germline mutations of DNA repair genes in breast and ovarian cancer. Mol Genet Genomic Med. 2015;3(5):459–66.CrossRefPubMedPubMedCentral Hirotsu Y, Nakagomi H, Sakamoto I, Amemiya K, Oyama T, Mochizuki H, Omata M. Multigene panel analysis identified germline mutations of DNA repair genes in breast and ovarian cancer. Mol Genet Genomic Med. 2015;3(5):459–66.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, McGuire V, Ladabaum U, Kobayashi Y, Lincoln SE, et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol. 2014;32(19):2001–9.CrossRefPubMedPubMedCentral Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, McGuire V, Ladabaum U, Kobayashi Y, Lincoln SE, et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol. 2014;32(19):2001–9.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Tung N, Battelli C, Allen B, Kaldate R, Bhatnagar S, Bowles K, Timms K, Garber JE, Herold C, Ellisen L, et al. Frequency of mutations in individuals with breast cancer referred for BRCA1 and BRCA2 testing using next-generation sequencing with a 25-gene panel. Cancer. 2015;121(1):25–33.CrossRefPubMed Tung N, Battelli C, Allen B, Kaldate R, Bhatnagar S, Bowles K, Timms K, Garber JE, Herold C, Ellisen L, et al. Frequency of mutations in individuals with breast cancer referred for BRCA1 and BRCA2 testing using next-generation sequencing with a 25-gene panel. Cancer. 2015;121(1):25–33.CrossRefPubMed
9.
Zurück zum Zitat Wong ES, Shekar S, Met-Domestici M, Chan C, Sze M, Yap YS, Rozen SG, Tan M-H, Ang P, Ngeow J. Inherited breast cancer predisposition in Asians: multigene panel testing outcomes from Singapore. NPJ Genomic Medicine. 2016;1:15003.CrossRefPubMedPubMedCentral Wong ES, Shekar S, Met-Domestici M, Chan C, Sze M, Yap YS, Rozen SG, Tan M-H, Ang P, Ngeow J. Inherited breast cancer predisposition in Asians: multigene panel testing outcomes from Singapore. NPJ Genomic Medicine. 2016;1:15003.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Crawford B, Adams SB, Sittler T, van den Akker J, Chan S, Leitner O, Ryan L, Gil E, van 't Veer L: Multi-gene panel testing for hereditary cancer predisposition in unsolved high-risk breast and ovarian cancer patients. Breast Cancer Res Treat 2017, 163(2):383–390. Crawford B, Adams SB, Sittler T, van den Akker J, Chan S, Leitner O, Ryan L, Gil E, van 't Veer L: Multi-gene panel testing for hereditary cancer predisposition in unsolved high-risk breast and ovarian cancer patients. Breast Cancer Res Treat 2017, 163(2):383–390.
11.
Zurück zum Zitat Couch FJ, Shimelis H, Hu C, Hart SN, Polley EC, Na J, Hallberg E, Moore R, Thomas A, Lilyquist J, et al. Associations between cancer predisposition testing panel genes and breast cancer. JAMA Oncol. 2017; Couch FJ, Shimelis H, Hu C, Hart SN, Polley EC, Na J, Hallberg E, Moore R, Thomas A, Lilyquist J, et al. Associations between cancer predisposition testing panel genes and breast cancer. JAMA Oncol. 2017;
12.
Zurück zum Zitat Xie A, Kwok A, Scully R. Role of mammalian Mre11 in classical and alternative nonhomologous end joining. Nat Struct Mol Biol. 2009;16(8):814–8.CrossRefPubMedPubMedCentral Xie A, Kwok A, Scully R. Role of mammalian Mre11 in classical and alternative nonhomologous end joining. Nat Struct Mol Biol. 2009;16(8):814–8.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Economopoulou P, Dimitriadis G, Psyrri A. Beyond BRCA: new hereditary breast cancer susceptibility genes. Cancer Treat Rev. 2015;41(1):1–8.CrossRefPubMed Economopoulou P, Dimitriadis G, Psyrri A. Beyond BRCA: new hereditary breast cancer susceptibility genes. Cancer Treat Rev. 2015;41(1):1–8.CrossRefPubMed
14.
Zurück zum Zitat Easton DF, Lesueur F, Decker B, Michailidou K, Li J, Allen J, Luccarini C, Pooley KA, Shah M, Bolla MK, et al. No evidence that protein truncating variants in BRIP1 are associated with breast cancer risk: implications for gene panel testing. J Med Genet. 2016;53(5):298–309.CrossRefPubMedPubMedCentral Easton DF, Lesueur F, Decker B, Michailidou K, Li J, Allen J, Luccarini C, Pooley KA, Shah M, Bolla MK, et al. No evidence that protein truncating variants in BRIP1 are associated with breast cancer risk: implications for gene panel testing. J Med Genet. 2016;53(5):298–309.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Kim H, Cho DY, Choi DH, Jung GH, Shin I, Park W, Huh SJ, Nam SJ, Lee JE, Gil WH, et al. Analysis of BRIP1 variants among Korean patients with BRCA1/2 mutation-negative high-risk breast cancer. Cancer Res Treat. 2016;48(3):955–61.CrossRefPubMedPubMedCentral Kim H, Cho DY, Choi DH, Jung GH, Shin I, Park W, Huh SJ, Nam SJ, Lee JE, Gil WH, et al. Analysis of BRIP1 variants among Korean patients with BRCA1/2 mutation-negative high-risk breast cancer. Cancer Res Treat. 2016;48(3):955–61.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Chen W, Yurong S, Liansheng N. Breast cancer low-penetrance allele 1100delC in the CHEK2 gene: not present in the Chinese familial breast cancer population. Adv Ther. 2008;25(5):496–501.CrossRefPubMed Chen W, Yurong S, Liansheng N. Breast cancer low-penetrance allele 1100delC in the CHEK2 gene: not present in the Chinese familial breast cancer population. Adv Ther. 2008;25(5):496–501.CrossRefPubMed
17.
Zurück zum Zitat Choi DH, Cho DY, Lee MH, Park HS, Ahn SH, Son BH, Haffty BG. The CHEK2 1100delC mutation is not present in Korean patients with breast cancer cases tested for BRCA1 and BRCA2 mutation. Breast Cancer Res Treat. 2008;112(3):569–73.CrossRefPubMed Choi DH, Cho DY, Lee MH, Park HS, Ahn SH, Son BH, Haffty BG. The CHEK2 1100delC mutation is not present in Korean patients with breast cancer cases tested for BRCA1 and BRCA2 mutation. Breast Cancer Res Treat. 2008;112(3):569–73.CrossRefPubMed
18.
Zurück zum Zitat Liu Y, Liao J, Xu Y, Chen W, Liu D, Ouyang T, Li J, Wang T, Fan Z, Fan T, et al. A recurrent CHEK2 p.H371Y mutation is associated with breast cancer risk in Chinese women. Hum Mutat. 2011;32(9):1000–3.CrossRefPubMed Liu Y, Liao J, Xu Y, Chen W, Liu D, Ouyang T, Li J, Wang T, Fan Z, Fan T, et al. A recurrent CHEK2 p.H371Y mutation is associated with breast cancer risk in Chinese women. Hum Mutat. 2011;32(9):1000–3.CrossRefPubMed
19.
Zurück zum Zitat Zhang S, Phelan CM, Zhang P, Rousseau F, Ghadirian P, Robidoux A, Foulkes W, Hamel N, McCready D, Trudeau M, et al. Frequency of the CHEK2 1100delC mutation among women with breast cancer: an international study. Cancer Res. 2008;68(7):2154–7.CrossRefPubMed Zhang S, Phelan CM, Zhang P, Rousseau F, Ghadirian P, Robidoux A, Foulkes W, Hamel N, McCready D, Trudeau M, et al. Frequency of the CHEK2 1100delC mutation among women with breast cancer: an international study. Cancer Res. 2008;68(7):2154–7.CrossRefPubMed
20.
Zurück zum Zitat Maxwell KN, Wubbenhorst B, D'Andrea K, Garman B, Long JM, Powers J, Rathbun K, Stopfer JE, Zhu J, Bradbury AR, et al. Prevalence of mutations in a panel of breast cancer susceptibility genes in BRCA1/2-negative patients with early-onset breast cancer. Genet Med. 2015;17(8):630–8.CrossRefPubMed Maxwell KN, Wubbenhorst B, D'Andrea K, Garman B, Long JM, Powers J, Rathbun K, Stopfer JE, Zhu J, Bradbury AR, et al. Prevalence of mutations in a panel of breast cancer susceptibility genes in BRCA1/2-negative patients with early-onset breast cancer. Genet Med. 2015;17(8):630–8.CrossRefPubMed
21.
Zurück zum Zitat Easton DF, Pharoah PD, Antoniou AC, Tischkowitz M, Tavtigian SV, Nathanson KL, Devilee P, Meindl A, Couch FJ, Southey M, et al. Gene-panel sequencing and the prediction of breast-cancer risk. N Engl J Med. 2015;372(23):2243–57.CrossRefPubMedPubMedCentral Easton DF, Pharoah PD, Antoniou AC, Tischkowitz M, Tavtigian SV, Nathanson KL, Devilee P, Meindl A, Couch FJ, Southey M, et al. Gene-panel sequencing and the prediction of breast-cancer risk. N Engl J Med. 2015;372(23):2243–57.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Park H, Park J, Nam E, Han J, Kim J, Kim T, Cho Y. The prevalence of mutations of breast cancer susceptibility genes in Korean BRCA1/2 mutation-negative patients with a high risk for hereditary breast cancer. Breast. 2017;32:S130.CrossRef Park H, Park J, Nam E, Han J, Kim J, Kim T, Cho Y. The prevalence of mutations of breast cancer susceptibility genes in Korean BRCA1/2 mutation-negative patients with a high risk for hereditary breast cancer. Breast. 2017;32:S130.CrossRef
23.
Zurück zum Zitat Monti P, Ciribilli Y, Jordan J, Menichini P, Umbach DM, Resnick MA, Luzzatto L, Inga A, Fronza G. Transcriptional functionality of germ line p53 mutants influences cancer phenotype. Clin Cancer Res. 2007;13(13):3789–95.CrossRefPubMedPubMedCentral Monti P, Ciribilli Y, Jordan J, Menichini P, Umbach DM, Resnick MA, Luzzatto L, Inga A, Fronza G. Transcriptional functionality of germ line p53 mutants influences cancer phenotype. Clin Cancer Res. 2007;13(13):3789–95.CrossRefPubMedPubMedCentral
Metadaten
Titel
Variants of cancer susceptibility genes in Korean BRCA1/2 mutation-negative patients with high risk for hereditary breast cancer
verfasst von
Ji Soo Park
Seung-Tae Lee
Eun Ji Nam
Jung Woo Han
Jung-Yun Lee
Jieun Kim
Tae Il Kim
Hyung Seok Park
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3940-y

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Update Onkologie

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