Next Article in Journal
AGTR1 Is Overexpressed in Neuroendocrine Neoplasms, Regulates Secretion and May Potentially Serve as a Target for Molecular Imaging and Therapy
Next Article in Special Issue
The 40bp Indel Polymorphism rs150550023 in the MDM2 Promoter is Associated with Intriguing Shifts in Gene Expression in the p53-MDM2 Regulatory Hub
Previous Article in Journal
Natural Killer Cells in Immunotherapy: Are We Nearly There?
Previous Article in Special Issue
Gene Panel Tumor Testing in Ovarian Cancer Patients Significantly Increases the Yield of Clinically Actionable Germline Variants beyond BRCA1/BRCA2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

NGS Panel Testing of Triple-Negative Breast Cancer Patients in Cyprus: A Study of BRCA-Negative Cases

1
Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Agios Dhometios, Nicosia, Cyprus
2
Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Agios Dhometios, Nicosia, Cyprus
3
Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Agios Dhometios, Nicosia, Cyprus
4
Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Agios Dhometios, Nicosia, Cyprus
5
Department of Medical Oncology, Bank of Cyprus Oncology Center, 32 Acropoleos Avenue, 2012 Strovolos, Nicosia, Cyprus
6
Clinical Genetics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Agios Dhometios, Nicosia, Cyprus
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(11), 3140; https://doi.org/10.3390/cancers12113140
Submission received: 18 September 2020 / Revised: 5 October 2020 / Accepted: 23 October 2020 / Published: 27 October 2020
(This article belongs to the Special Issue Molecular Genetics of Breast and Ovary Cancer)

Abstract

:

Simple Summary

In Cyprus, approximately 9% of triple-negative (negative in common breast cancer receptors—estrogen, progesterone, and human epidermal growth factor receptor 2 (HER2) receptors) breast cancer (TNBC) patients carry inherited mutations in the BRCA1/2 breast cancer (BC) susceptibility genes. These mutations increase the risk of BC. However, the contribution of other BC susceptibility genes has not yet been determined. To this end, we aimed to investigate the prevalence of mutations in BRCA1/2-negative TNBC patients in Cyprus. Ninety-five cancer susceptibility genes were sequenced for 163 TNBC patients. The frequency of non-BRCA mutations and especially PALB2 in TNBC patients in Cyprus appears to be higher compared to other populations, and half of the mutation-positive patients were diagnosed over the age of 60 years. Based on these results, we believe that PALB2 and TP53 along with BRCA1/2 genetic testing could be beneficial for a large proportion of TNBC patients in Cyprus, irrespective of their age of diagnosis.

Abstract

In Cyprus, approximately 9% of triple-negative (estrogen receptor-negative, progesterone receptor-negative, and human epidermal growth factor receptor 2-negative) breast cancer (TNBC) patients are positive for germline pathogenic variants (PVs) in BRCA1/2. However, the contribution of other genes has not yet been determined. To this end, we aimed to investigate the prevalence of germline PVs in BRCA1/2-negative TNBC patients in Cyprus, unselected for family history of cancer or age of diagnosis. A comprehensive 94-cancer-gene panel was implemented for 163 germline DNA samples, extracted from the peripheral blood of TNBC patients. Identified variants of uncertain clinical significance were evaluated, using extensive in silico investigation. Eight PVs (4.9%) were identified in two high-penetrance TNBC susceptibility genes. Of these, seven occurred in PALB2 (87.5%) and one occurred in TP53 (12.5%). Interestingly, 50% of the patients carrying PVs were diagnosed over the age of 60 years. The frequency of non-BRCA PVs (4.9%) and especially PALB2 PVs (4.3%) in TNBC patients in Cyprus appears to be higher compared to other populations. Based on these results, we believe that PALB2 and TP53 along with BRCA1/2 genetic testing could be beneficial for a large proportion of TNBC patients in Cyprus, irrespective of their age of diagnosis.

Graphical Abstract

1. Introduction

Breast cancer (BC) is the most common cancer and the leading cause of cancer-related mortality in women worldwide [1]. Triple-negative breast cancer (TNBC) is a distinct, highly aggressive form of BC, characterized by lack of expression of the estrogen and progesterone receptors (ER/PR) as well as of human epidermal growth factor receptor 2 (HER2) in breast tumors [2]. It accounts for approximately 10–20% of all BC cases, and it occurs more frequently in young or premenopausal women and women of African American, Hispanic, and Ashkenazi Jewish ancestry [2,3,4]. Although TNBC lacks a specific therapeutic approach due to the absence of receptors for conventional targeted therapies, systemic cytotoxic chemotherapy can achieve good pathologic complete response (pCR) rates [5]. Survival rates are lower compared to other forms of breast cancer, with the majority of patients presenting with early recurrence and a high frequency of metastasis, especially to the central nervous system (20%) and viscera (29%) [3,6,7]. Accordingly, the five-year survival rate of patients with TNBC is estimated at 70% compared with >80% for all other breast cancer subtypes [8].
A high percentage of TNBC patients carry a germline pathogenic variant (PV) in one of the highly penetrant BC susceptibility genes, BRCA1 and BRCA2 [9]. Additionally, studies have also examined the contribution of other BC susceptibility genes including ATM, BARD1, CHEK2, PALB2, PTEN, RAD51D, and TP53 to TNBC susceptibility [9,10,11,12,13,14]. Other cancer predisposition genes including BRIP1, CDH1, CDKN2A, MLH1, MRE11A, MSH2, MSH6, NBN, NF1, PMS2, RAD50, RAD51C, FANCM, XRCC2, and MUTYH—biallelic mutations—have also been investigated in TNBC [9,10,11,12,13,14,15]. Recently, it has been reported that 14.4% of TNBC patients carried PVs in established BC genes as well as other cancer susceptibility genes [9,14]. Of these, 6% harbored PVs in non-BRCA TNBC high-risk genes (odds ratio (OR) > 5.0) including BARD1, PALB2, and RAD51D and TNBC moderate-risk genes (OR > 2.0) including BRIP1, RAD51C, and TP53 [9].
In a recent study from Cyprus, it has been reported that approximately 9% of TNBC patients were positive for germline PVs in the BRCA1/2 genes [16]. However, the contribution of other BC susceptibility genes towards TNBC predisposition is not yet determined; thus, there is a great need to investigate the prevalence and distribution of germline PVs in other susceptibility genes.
In recent years, the expanding panel of BC susceptibility genes has necessitated the use of Next-Generation Sequencing (NGS). Consequently, NGS panel testing has been integrated into routine clinical practice. In this manuscript, we present the first NGS gene-panel investigation of TNBC patients in Cyprus who were negative for BRCA1 and BRCA2 germline PVs.

2. Results

2.1. Pathogenic Variants

Pathogenic variants were identified in two high-penetrance BC and TNBC susceptibility genes, in 8 out of 163 patients tested (4.9%) (Table 1). Of these, seven were detected in the PALB2 gene (87.5%) and one was detected in the TP53 gene (12.5%) (Table 1). Two PALB2-positive patients did not have a family history of breast, ovarian, pancreatic, or colorectal cancer, whereas two other patients had a strong family history (at least three first- or second-degree relatives diagnosed with breast, ovarian, or pancreatic cancer) (Table 1). It is noted that two PALB2 PVs (c.487_488del and c.1685-2A>G) were present in 3 and 2 unrelated TNBC patients, respectively (Table 1). Besides family history, it is noteworthy that half of the PV carriers, (4 out of 8) were diagnosed with TNBC over the age of 60 years (Table 1) (p-value = 0.027, two-tailed t-test) (Figure S1).

2.2. Classification of Variants of Uncertain Clinical Significance (VUS)

In this study, 65 unique VUSs were identified and, of these, 21 were classified as deleterious by at least 75% of the in silico prediction tools (Table S1 and Figure S2). It is noted that five of the VUSs predicted as deleterious, are novel and have not been reported before in ClinVar, dbSNP, and LOVD databases or in any publications (Table S1). The American College of Medical Genetics and Genomics (ACMG) guidelines (Table S2) were followed for the classification of the 21 VUSs predicted as deleterious, according to which five out of the 21 variants were classified as likely benign, and 16 variants as VUS (Table S3).
Comprehensive in silico classification of the structural, functional, stability, and flexibility impact of the 21 VUSs predicted as deleterious was carried out (Tables S4 and S5). Despite its classification as a VUS, the missense TP53 p.(Gly154Asp) variant is of particular interest (Figure 1) (ClinGen TP53 Expert Panel Specifications to the ACMG/AMP variant interpretation guidelines) [17], since there is extensive evidence that supports its pathogenicity (Table S3). At the protein level, the Gly154 residue (Figure 1a) resides in the p53 DNA-binding domain (PF00870, amino acids 102-292), in which a large proportion of missense variants are classified as pathogenic (29%, 198 out of 674). Additionally, the Gly154 residue constitutes a mutational hotspot, since 25 somatic occurrences have been reported in the cancerhotspots.org database [18,19]. This variant is predicted to destabilize the protein structure and to disrupt the proper function of the TP53 protein, as it probably negatively affects the secondary structure of the protein and subsequently DNA binding (Table S4). As predicted, the phi (φ)/psi (ψ) angles are in the favored region for the wild-type Gly154 residue but in the outlier region for the mutant Asp154 residue. Additionally, the substitution causes a predicted decrease in molecule flexibility (−0.028 kcal.mol−1K−1) (Figure 1b). The wild-type amino acid may participate in intermolecular interactions (hydrogen bonds) with surrounding amino acids (Figure 1c), which would be probably hampered and shifted upon mutagenesis (Figure 1d). However, functional studies are needed for the accurate classification of this variant as well as all the other VUSs predicted as deleterious.

3. Discussion

This study is the first gene-panel NGS investigation of patients with TNBC in Cyprus who are negative for PVs in the BRCA1 and BRCA2 genes. The results present for the first time the prevalence of germline PVs in BC and other cancer predisposition genes in patients diagnosed with TNBC in Cyprus.
Among the 163 BRCA1/2-negative TNBC patients tested, 4.9% were positive for PVs in the ΤNBC susceptibility genes PALB2 and TP53. This frequency is higher compared to a large multiethnic study of 10,901 TNBC patients, which demonstrated that only 3.1% of the BRCA1/2-negative study participants carried PVs in high-risk (BARD1, PALB2, and RAD51D) and moderate-risk (TP53, BRIP1, and RAD51C) TNBC susceptibility genes [9]. Similar observations are made when comparing the frequency of PVs reported here to that reported in the study of González-Rivera et al. in which 3.3% of the BRCA1/2-negative TNBC patients carried PVs in the high-penetrance TNBC susceptibility genes BARD1 and RAD51D [20]. A large-scale study evaluated the performance of gene-panel testing in 35,409 women with a single diagnosis of BC. It has been reported that 3.3% of BRCA1/2-negative TNBC patients carried a PV in high- (BARD1, PALB2, and RAD51D) or moderate-risk (TP53, BRIP1, and RAD51C) TNBC susceptibility genes [10]. The frequency of PVs in TNBC susceptibility genes in Cyprus is about three times higher than the reported frequency in Chinese BRCA-negative TNBC patients, where only 1.36% were positive for PVs in the high-penetrance TNBC susceptibility genes PALB2, RAD51D, and BARD1 [21]. The frequency of PVs reported here is also different compared to the frequency of PVs (1%) in TNBC susceptibility genes (BARD1, PALB2, RAD51D, BRIP1, RAD51C, and TP53) reported in a recently published study of the Georgia and California Cancer Registries [22]. In contrast, our results are similar to the frequency of PVs in TNBC susceptibility genes (4.8%) reported in Greek patients with TNBC [23]. However, the patients included in the Greek study were highly selected, having a strong family history (at least three breast, ovarian, or pancreatic cancer patients in the same family) and/or diagnosed earlier than the age of 35 years. This could in part explain the relatively high frequency of PVs reported in Greek samples. Nevertheless, this was not the case for our study cohort, where TNBC patients were unselected for age of onset and family history.
It is estimated that VUSs account for around 40% of the total number of variants identified in extended gene-panel sequencing studies, and their classification remains a major challenge [24]. In this study, nineteen patients (11.7%) carried VUS predicted as deleterious in established BC and other cancer susceptibility genes, while they did not carry any PV in a gene with confirmed BC association. In order to decipher further which VUS may be actionable, a detailed in silico investigation was carried out. The TP53 VUS p.(Gly154Asp) identified in a patient with a family history of breast cancer is of particular interest. The variant resides in the p53 DNA binding domain, where 90% of all known TP53 pathogenic variants are located [25] and is predicted to alter the DNA binding capacity of the p53 protein. Based on in silico prediction tools, it is likely that this missense variant is pathogenic, but functional studies and/or co-segregation or case-control studies are needed for the accurate classification of this and other VUS in TNBC patients.
The average age at initial TNBC diagnosis of our patient cohort is 50.55 ± 10.35, with a range from 27 to 75 years. This is similar to the average age of onset of the 10,901 TNBC patients recruited by the TNBC Consortium (TNBCC) and the Ambry Genetics clinical testing laboratory (49.9 ± 11.3 years) [9]. Among the 10,901 patients, 17.7% were diagnosed with TNBC over the age of 60 years, which is very similar to our population (17.8%) [9]. An interesting observation is that, among 29 study participants diagnosed over the age of 60 years, 13.8% were found positive for PVs in established BC. In contrast, in the large-scale multiethnic study of TNBC patients, 10.2% of patients diagnosed over the age of 60 years tested positive for germline PVs in established BC and other cancer susceptibility genes [9]. Considering that only half of the TNBC PV carriers fulfilled the NCCN guidelines, our results support the notion that all TNBC patients in Cyprus should be referred for genetic testing, irrespective of their age at diagnosis.
According to Cybulski et al., it is estimated that 34% of BC patients with a germline PALB2 PV have a TNBC subtype [26]. PALB2 PVs were the most prevalent in our cohort of BRCA1/2-negative TNBC samples (4.3%). Two large multiethnic studies of the TNBCC in collaboration with Ambry Genetics clinical testing laboratory and Myriad Genetics laboratory reported lower frequencies of PALB2 PVs amongst TNBC patients (1.3% and 1.4%, respectively) [9,10]. A possible founder effect for two of the PALB2 PVs identified (c.487_488del and c.1685-2A>G) in our study could partly explain the higher prevalence of PALB2 PVs in our TNBC population. However, additional studies are required to draw more definite conclusions. The absence of a CHEK2 PV in our TNBC cohort agrees with the reported association of CHEK2 PVs with ER-positive BC and the absence of association with TNBC [9].
It is well known that BRCA1/2 carriers with TNBC can benefit from targeted therapeutic strategies. Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) can be used as a targeted treatment for germline BRCA1/2-positive BC patients with HER2-negative metastatic breast tumors [27]. Tumors harboring inherited or somatic pathogenic variants in other non-BRCA genes implicated in DNA repair pathways may also respond with a BRCA-like behavior to this particular class of drugs. An ongoing phase II clinical trial is investigating the antitumor efficacy of PARPi in patients with advanced HER2-negative or TNBC and a somatic or germline PV in a non-BRCA1/2 Homologous Recombination pathway gene [28]. It is anticipated that, in the near future, patients harboring PVs in genes other than BRCA1/2 may also benefit from these and other targeted therapies.

4. Materials and Methods

4.1. Study Population

This study involved patients with unilateral TNBC, referred to the Clinical Genetics Department of the Cyprus Institute of Neurology and Genetics for genetic counseling from 2004 to 2019. According to NCCN guidelines (version 1.2020), genetic testing should be offered to patients diagnosed with TNBC at age ≤60 years. The testing criteria were slightly modified from the international guidelines to reflect the situation in Cyprus. TNBC patients were referred for genetic counseling and subsequently underwent genetic testing irrespective of their age of disease onset. This study includes 163 TNBC patients who tested negative for germline PVs (including large genomic rearrangements) in the BRCA1 and BRCA2 genes and were unselected for family history of breast and/or ovarian cancer. Each study participant signed an informed consent form and agreed to undergo genetic testing.
The characteristics of the TNBC patients included in the study are summarized in Table 2.

4.2. Multigene Panel-Based Mutation Analysis

DNA samples from TNBC patients that tested negative for germline PVs in the BRCA1 and BRCA2 genes were analyzed further by targeted NGS, using a panel of 94 cancer susceptibility genes (Illumina TruSight Cancer Sequencing panel: #FC-121-0202). The panel contains oligos targeting and enriches more than 1700 exons, including coding regions and noncoding exon-flanking regions spanning the 94 cancer susceptibility genes (Table S6). The TruSight Rapid Capture kit was used for the preparation of the NGS library according to the manufacturer’s protocol (Illumina, #FC-140-1106). Sequencing was performed on the NextSeq 500 or the MiSeq Sequencing Platforms (Illumina, San Diego, CA, USA) with 75-bp or 150-bp paired-end reads. Screening for large rearrangements was not performed.

4.3. BARD1 Mutation Screening

Since the established high-risk TNBC susceptibility gene BARD1 is not included in the TruSight Cancer Sequencing panel, BARD1 mutation screening was performed for all samples included in this study by Sanger sequencing. All the coding exons and proximal splice site regions of the BARD1 gene (LRG_297) were sequenced as described previously [29].

4.4. Bioinformatics Analysis

Sequence reads (2 × 150 bp or 2 × 75 bp) were aligned to the reference GRCh37 (hg19) human genome assembly using the Burrows-Wheeler Aligner—Maximal Exact Match (BWA-MEM) algorithm. Duplicates were detected by Picard (http://broadinstitute.github.io/picard/), and bases were recalibrated using the GATK suite and according to the best practice guidelines (https://www.broadinstitute.org/gatk). Variants were called with the GATK HaplotypeCaller and GenotypeGVCF, filtered following the GATK guidelines and functionally annotated using ANNOVAR (https://doc-openbio.readthedocs.io/projects/annovar/en/latest/).

4.5. Variant Selection

Multiple filtering criteria were used to identify PVs and variants of uncertain clinical significance (VUS). Variants with read depth of less than 30 were excluded from further analysis. Rare variants with minor allele frequency (MAF)—taken from the Exome Aggregation Consortium (ExAC) (https://gnomad.broadinstitute.org/)—less than or equal to 1% were selected for further investigation. Variation databases (ClinVar, https://www.ncbi.nlm.nih.gov/clinvar/; LOVD, http://www.lovd.nl/3.0/home; and dbSNP, https://www.ncbi.nlm.nih.gov/projects/SNP/) were explored to detect already published variants and their structural/functional effects. Only variants having a damaging effect on the protein sequence or structure—frameshift indels, splicing variants (±3 positions), nonsense, and missense variants reported as pathogenic or likely pathogenic in the literature—were considered as pathogenic. The regSNP-splicing tool (http://regsnps-splicing.ccbb.iupui.edu/) was used to prioritize synonymous variants based on their proximity to splice donor or acceptor sites. Alternative splicing events were tested by MaxEntScan (http://hollywood.mit.edu/burgelab/maxent/), BDGP: Splice Site Prediction by Neural Network (http://www.fruitfly.org/seq_tools/splice.html), Human Splicing Finder (HSF3.0) (http://www.umd.be/HSF3/), and NetGene2 (http://www.cbs.dtu.dk/services/NetGene2/). All PVs were confirmed by Sanger Sequencing as described elsewhere [29].

4.6. Classification of Variants of Uncertain Clinical Significance (VUS)

Further analysis of VUS was performed only for variants reported in established BC susceptibility genes (BRCA1, BRCA2, PALB2, BARD1, RAD51D, ATM, CHEK2, PTEN, and TP53) and other cancer susceptibility genes (CDH1, BRIP1, CDKN2A, MLH1, MSH2, MSH6, NBN, NF1, PMS2, RAD51C, XRCC2, STK11, FANCM, and MUTYH-biallelic). Protein features such as amino acid conservation among species, wild-type/mutant amino acid biophysical and/or biochemical properties, amino acid localization within the protein, and potential impact on mRNA were investigated using several in silico prediction tools [30,31,32,33,34,35,36,37,38,39]. Variants considered as deleterious were those classified as such by at least 75% of the prediction tools. The American College of Medical Genetics and Genomics (ACMG) standards and guidelines for the interpretation of sequence variants were followed for variant classification [40]. In addition, a comprehensive in silico classification scheme for the interpretation of the stability [41,42,43,44,45,46,47,48,49,50,51,52,53,54], structural [55,56,57,58], functional [55,56,57,58], and flexibility [51] impact of VUS predicted as deleterious was carried out, using experimentally solved or in silico predicted protein structures [59,60]. The detailed workflow followed for the in silico classification of VUS is described in the Supplementary Methods. All VUSs predicted as deleterious were confirmed by Sanger Sequencing as described elsewhere [29].

4.7. Statistical Analysis

The R (v3.3.2) (https://www.r-project.org/) statistical computing language was used for statistical analysis. The Shapiro–Wilk’s test was used to test whether the underlying distribution was normal. Carriers and non-carriers of PVs were compared for numerical variables with independent samples two-tailed t-test (for normally distributed values). Two-tailed p-values lower than 0.05 were considered to be statistically significant.

5. Conclusions

In conclusion, our study identified eight PVs in ΤΝΒC susceptibility genes in eight unrelated BRCA1/2-negative TNBC patients in Cyprus (4.9%), unselected for family history and age at disease diagnosis. Although our sample size is small, the reported frequency is higher compared to other populations and may have important clinical implications. Additionally, the frequency of PALB2 PVs (4.3%) appears to be higher compared to other populations (1.3–1.4%), while half of the PV-positive patients were diagnosed over the age of 60 years. We suggest that PALB2 and TP53 along with BRCA1/2 genetic testing could be beneficial for a substantial proportion of TNBC patients in Cyprus, irrespective of their age at diagnosis.

Supplementary Materials

The following are available online at https://www.mdpi.com/2072-6694/12/11/3140/s1, Supplementary Methods: Classification of variants with uncertain clinical significance, Figure S1: Plots demonstrating the distribution of age at TNBC diagnosis of BRCA1/2-negative women in Cyprus, Table S1: Summary of the predicted deleterious VUSs in established BC and other cancer susceptibility genes, Figure S2: Box plot and histogram demonstrating the distribution of pathogenic variants and in silico predicted deleterious VUSs in established BC and other cancer susceptibility genes among all study samples, Table S2: Criteria as specified by the American College of Medical Genetics (ACMG) standards and guidelines for the interpretation of sequence variants, Table S3: Classification of the in silico predicted deleterious VUS based on the ACMG standards and guidelines for interpretation of sequence variants, Table S4: In silico assessment of the structural, functional, stability, and flexibility impact of the predicted deleterious VUSs in BC susceptibility genes, Table S5: In silico assessment of the structural, functional, stability, and flexibility impact of the predicted deleterious VUSs in other cancer susceptibility genes, Table S6: TruSight Cancer (Illumina) target genes in alphabetical order.

Author Contributions

Conceptualization, M.Z., M.A.L., K.M., K.K., and A.H.; data curation, M.Z., M.A.L., K.M., Y.M., F.K., E.K., G.A.T., E.S., G.M.S., K.K., and A.H.; formal analysis, M.Z. and K.M.; funding acquisition, G.M.S. and A.H.; investigation, M.Z., M.A.L., K.M., K.K., and A.H.; methodology, M.Z., M.A.L., K.M., P.P., C.M., and A.H.; project administration, K.K. and A.H.; resources, G.M.S., K.K., and A.H.; software, M.Z. and K.M.; supervision, K.K. and A.H.; validation, M.Z., M.A.L., K.M., G.M.S., K.K., and A.H.; visualization, M.Z., M.A.L., K.M., G.M.S., K.K., and A.H.; writing—original draft, M.Z.; writing—review and editing, M.Z., M.A.L., K.M., G.A.T., G.M.S., K.K., and A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Cyprus Institute of Neurology and Genetics, and M.Z. and G.S. were supported by the European Commission Research Executive Agency (REA) Grant BIORISE (grant number 669026) under the Spreading Excellence, Widening Participation, Science with and for Society Framework.

Acknowledgments

We would like to thank all the patients and their families for participating in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Foulkes, W.D.; Smith, I.E.; Reis-Filho, J.S. Triple-Negative Breast Cancer. N. Engl. J. Med. 2010, 363, 1938–1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Afghahi, A.; Telli, M.L.; Kurian, A.W. Genetics of triple-negative breast cancer: Implications for patient care. Curr. Probl. Cancer 2016, 40, 130–140. [Google Scholar] [CrossRef] [PubMed]
  4. Hubalek, M.; Czech, T.; Müller, H. Biological Subtypes of Triple-Negative Breast Cancer. Breast Care (Basel) 2017, 12, 8–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Poggio, F.; Bruzzone, M.; Ceppi, M.; Pondé, N.F.; La Valle, G.; Del Mastro, L.; De Azambuja, E.; Lambertini, M. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: A systematic review and meta-analysis. Ann. Oncol. 2018, 29, 1497–1508. [Google Scholar] [CrossRef] [PubMed]
  6. Polivka, J.; Kralickova, M.; Polivka, J.; Kaiser, C.; Kuhn, W.; Golubnitschaja, O. Mystery of the brain metastatic disease in breast cancer patients: Improved patient stratification, disease prediction and targeted prevention on the horizon? EPMA J. 2017, 8, 119–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yao, Y.; Chu, Y.; Xu, B.; Hu, Q.; Song, Q. Risk factors for distant metastasis of patients with primary triple-negative breast cancer. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed]
  8. Blows, F.M.; Driver, K.E.; Schmidt, M.K.; Broeks, A.; van Leeuwen, F.E.; Wesseling, J.; Cheang, M.C.; Gelmon, K.; Nielsen, T.O.; Blomqvist, C.; et al. Subtyping of Breast Cancer by Immunohistochemistry to Investigate a Relationship between Subtype and Short and Long Term Survival: A Collaborative Analysis of Data for 10,159 Cases from 12 Studies. PLoS Med. 2010, 7, e1000279. [Google Scholar] [CrossRef]
  9. Shimelis, H.; LaDuca, H.; Hu, C.; Hart, S.N.; Na, J.; Thomas, A.; Akinhanmi, M.; Moore, R.M.; Brauch, H.; Cox, A.; et al. Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J. Natl. Cancer Inst. 2018, 110, 855. [Google Scholar] [CrossRef]
  10. Buys, S.S.; Sandbach, J.F.; Gammon, A.; Patel, G.; Kidd, J.; Brown, K.L.; Sharma, L.; Saam, J.; Lancaster, J.; Daly, M.B. A study of over 35,000 women with breast cancer tested with a 25-gene panel of hereditary cancer genes. Cancer 2017, 123, 1721–1730. [Google Scholar] [CrossRef] [Green Version]
  11. Ollier, M.; Radosevic-Robin, N.; Kwiatkowski, F.; Ponelle, F.; Viala, S.; Privat, M.; Uhrhammer, N.; Bernard-Gallon, D.; Penault-Llorca, F.F.; Bignon, Y.-J.J.; et al. DNA repair genes implicated in triple negative familial non-BRCA1/2 breast cancer predisposition. Am. J. Cancer Res. 2015, 5, 2113–2126. [Google Scholar] [PubMed]
  12. Tung, N.; Lin, N.U.; Kidd, J.; Allen, B.A.; Singh, N.; Wenstrup, R.J.; Hartman, A.R.; Winer, E.P.; Garber, J.E. Frequency of germline mutations in 25 cancer susceptibility genes in a sequential series of patients with breast cancer. J. Clin. Oncol. 2016, 34, 1460–1468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Sun, J.; Meng, H.; Yao, L.; Lv, M.; Bai, J.; Zhang, J.; Wang, L.; Ouyang, T.; Li, J.; Wang, T.; et al. Germline Mutations in Cancer Susceptibility Genes in a Large Series of Unselected Breast Cancer Patients. Clin. Cancer Res. 2017, 23, 6113–6119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Couch, F.J.; Hart, S.N.; Sharma, P.; Toland, A.E.; Wang, X.; Miron, P.; Olson, J.E.; Godwin, A.K.; Pankratz, V.S.; Olswold, C.; et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J. Clin. Oncol. 2015, 33, 304–311. [Google Scholar] [CrossRef]
  15. Kiiski, J.I.; Pelttari, L.M.; Khan, S.; Freysteinsdottir, E.S.; Reynisdottir, I.; Hart, S.N.; Shimelis, H.; Vilske, S.; Kallioniemi, A.; Schleutker, J.; et al. Exome sequencing identifies FANCM as a susceptibility gene for triple-negative breast cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 15172–15177. [Google Scholar] [CrossRef] [Green Version]
  16. Loizidou, M.A.; Hadjisavvas, A.; Pirpa, P.; Spanou, E.; Delikurt, T.; Tanteles, G.A.; Daniel, M.; Kountourakis, P.; Malas, S.; Ioannidis, G.; et al. BRCA1 and BRCA2 mutation testing in Cyprus; a population based study. Clin. Genet. 2017, 91, 611–615. [Google Scholar] [CrossRef]
  17. ClinGen TP53 Expert Panel Specifications to the ACMG/AMP Variant Interpretation Guidelines Version 1. Available online: https://www.clinicalgenome.org/site/assets/files/3876/clingen_tp53_acmg_specifications_v1.pdf (accessed on 6 August 2019).
  18. Chang, M.T.; Bhattarai, T.S.; Schram, A.M.; Bielski, C.M.; Donoghue, M.T.A.; Jonsson, P.; Chakravarty, D.; Phillips, S.; Kandoth, C.; Penson, A.; et al. Accelerating discovery of functional mutant alleles in cancer. Cancer Discov. 2018, 8, 174–183. [Google Scholar] [CrossRef] [Green Version]
  19. Chang, M.T.; Asthana, S.; Gao, S.P.; Lee, B.H.; Chapman, J.S.; Kandoth, C.; Gao, J.J.; Socci, N.D.; Solit, D.B.; Olshen, A.B.; et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat. Biotechnol. 2016, 34, 155–163. [Google Scholar] [CrossRef]
  20. González-Rivera, M.; Lobo, M.; López-Tarruella, S.; Jerez, Y.; del Monte-Millán, M.; Massarrah, T.; Ramos-Medina, R.; Ocaña, I.; Picornell, A.; Garzón, S.S.; et al. Frequency of germline DNA genetic findings in an unselected prospective cohort of triple-negative breast cancer patients participating in a platinum-based neoadjuvant chemotherapy trial. Breast Cancer Res. Treat. 2016, 156, 507–515. [Google Scholar] [CrossRef]
  21. Yan, Y.; Liang, H.; Light, M.; Li, T.; Deng, Y.; Li, M.; Li, S.; Qin, X. XPD Asp312Asn and Lys751Gln polymorphisms and breast cancer susceptibility: A meta-analysis. Tumor Biol. 2014, 35, 1907–1915. [Google Scholar] [CrossRef] [Green Version]
  22. Kurian, A.W.; Ward, K.C.; Howlader, N.; Deapen, D.; Hamilton, A.S.; Mariotto, A.; Miller, D.; Penberthy, L.S.; Katz, S.J. Genetic Testing and Results in a Population-Based Cohort of Breast Cancer Patients and Ovarian Cancer Patients. J. Clin. Oncol. 2019, 37, 1305–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Fostira, F.; Kostantopoulou, I.; Apostolou, P.; Papamentzelopoulou, M.S.; Papadimitriou, C.; Faliakou, E.; Christodoulou, C.; Boukovinas, I.; Razis, E.; Tryfonopoulos, D.; et al. One in three highly selected Greek patients with breast cancer carries a loss-of-function variant in a cancer susceptibility gene. J. Med. Genet. 2020, 57, 53–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Federici, G.; Soddu, S. Variants of uncertain significance in the era of high-throughput genome sequencing: A lesson from breast and ovary cancers. J. Exp. Clin. Cancer Res. 2020, 39, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Végran, F.; Rebucci, M.; Chevrier, S.; Cadouot, M.; Boidot, R.; Lizard-Nacol, S. Only Missense Mutations Affecting the DNA Binding Domain of P53 Influence Outcomes in Patients with Breast Carcinoma. PLoS ONE 2013, 8, e55103. [Google Scholar] [CrossRef] [PubMed]
  26. Cybulski, C.; Kluźniak, W.; Huzarski, T.; Wokołorczyk, D.; Kashyap, A.; Jakubowska, A.; Szwiec, M.; Byrski, T.; Dębniak, T.; Górski, B.; et al. Clinical outcomes in women with breast cancer and a PALB2 mutation: A prospective cohort analysis. Lancet Oncol. 2015, 16, 638–644. [Google Scholar] [CrossRef]
  27. Caulfield, S.E.; Davis, C.C.; Byers, K.F. Olaparib: A Novel Therapy for Metastatic Breast Cancer in Patients With a BRCA1/2 Mutation. J. Adv. Pract. Oncol. 2019, 10, 167–174. [Google Scholar]
  28. Afghahi, A.; Chang, P.-J.; Ford, J.; Telli, M. The Talazoparib Beyond BRCA (TBB) trial: A phase II clinical trial of talazoparib in BRCA1 and BRCA2 wild-type patients with (i) advanced triple-negative breast cancer (TNBC) and homologous recombination deficiency (HRD) as assessed by myriad genetics HRD. Cancer Res. 2016, 76. [Google Scholar] [CrossRef]
  29. Loizidou, M.; Marcou, Y.; Anastasiadou, V.; Newbold, R.; Hadjisavvas, A.; Kyriacou, K. Contribution of BRCA1 and BRCA2 germline mutations to the incidence of early-onset breast cancer in Cyprus. Clin. Genet. 2007, 71, 165–170. [Google Scholar] [CrossRef]
  30. Vaser, R.; Adusumalli, S.; Ngak Leng, S.; Sikic, M.; Ng, P.C. SIFT missense predictions for genomes. Nat. Protoc. 2015, 11. [Google Scholar] [CrossRef]
  31. Adzhubei, I.A.; Schmidt, S.; Peshkin, L.; Ramensky, V.E.; Gerasimova, A.; Bork, P.; Kondrashov, A.S.; Sunyaev, S.R. A Method and Server for Predicting Damaging Missense Mutations. Nat. Methods 2010, 7, 248–249. [Google Scholar] [CrossRef] [Green Version]
  32. Chun, S.; Fay, J.C. Identification of deleterious mutations within three human genomes. Genome Res. 2009, 19, 1553–1561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Reva, B.; Antipin, Y.; Sander, C. Predicting the functional impact of protein mutations: Application to cancer genomics. Nucleic Acids Res. 2011, 39, e118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Choi, Y.; Sims, G.E.; Murphy, S.; Miller, J.R.; Chan, A.P. Predicting the Functional Effect of Amino Acid Substitutions and Indels. PLoS ONE 2012, 7, e46688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Schwarz, J.M.; Rödelsperger, C.; Schuelke, M.; Seelow, D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat. Methods 2010, 7, 575–576. [Google Scholar] [CrossRef]
  36. Kircher, M.; Witten, D.M.; Jain, P.; O’Roak, B.J.; Cooper, G.M.; Shendure, J. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 2014, 46, 310–315. [Google Scholar] [CrossRef] [Green Version]
  37. Tavtigian, S.V.; Deffenbaugh, A.M.; Yin, L.; Judkins, T.; Scholl, T.; Samollow, P.B.; de Silva, D.; Zharkikh, A.; Thomas, A. Comprehensive statistical study of 452 BRCA1 missense substitutions with classification of eight recurrent substitutions as neutral. J. Med. Genet. 2006, 43, 295–305. [Google Scholar] [CrossRef] [Green Version]
  38. Salgado, D.; Desvignes, J.-P.; Rai, G.; Blanchard, A.; Miltgen, M.; Pinard, A.; Lévy, N.; Collod-Béroud, G.; Béroud, C. UMD-Predictor: A High-Throughput Sequencing Compliant System for Pathogenicity Prediction of any Human cDNA Substitution. Hum. Mutat. 2016, 37, 439–446. [Google Scholar] [CrossRef] [Green Version]
  39. Feng, B.-J. PERCH: A Unified Framework for Disease Gene Prioritization. Hum. Mutat. 2017, 38, 243–251. [Google Scholar] [CrossRef] [Green Version]
  40. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; 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, 405–423. [Google Scholar] [CrossRef]
  41. Schymkowitz, J.; Borg, J.; Stricher, F.; Nys, R.; Rousseau, F.; Serrano, L. The FoldX web server: An online force field. Nucleic Acids Res. 2005, 33, W382–W388. [Google Scholar] [CrossRef] [Green Version]
  42. Capriotti, E.; Fariselli, P.; Casadio, R. I-Mutant2.0: Predicting stability changes upon mutation from the protein sequence or structure. Nucleic Acids Res. 2005, 33, W306–W310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Lindberg, M.J.; Bystrom, R.; Boknas, N.; Andersen, P.M.; Oliveberg, M. Systematically perturbed folding patterns of amyotrophic lateral sclerosis (ALS)-associated SOD1 mutants. Proc. Natl. Acad. Sci. USA 2005, 102, 9754–9759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Randles, L.G.; Lappalainen, I.; Fowler, S.B.; Moore, B.; Hamill, S.J.; Clarke, J. Using Model Proteins to Quantify the Effects of Pathogenic Mutations in Ig-like Proteins. J. Biol. Chem. 2006, 281, 24216–24226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sheu, S.Y.; Yang, D.Y.; Selzle, H.L.; Schlag, E.W. Energetics of hydrogen bonds in peptides. Proc. Natl. Acad. Sci. USA 2003, 100, 12683–12687. [Google Scholar] [CrossRef] [Green Version]
  46. Seifi, M.; Walter, M.A. Accurate prediction of functional, structural, and stability changes in PITX2 mutations using in silico bioinformatics algorithms. PLoS ONE 2018, 13, e0195971. [Google Scholar] [CrossRef] [Green Version]
  47. Cheng, J.; Randall, A.; Baldi, P. Prediction of Protein Stability Changes for Single-Site Mutations Using Support Vector Machines. Proteins 2006, 62, 1125–1132. [Google Scholar] [CrossRef]
  48. Parthiban, V.; Gromiha, M.M.; Schomburg, D. CUPSAT: Prediction of protein stability upon point mutations. Nucleic Acids Res. 2006, 34, 239–242. [Google Scholar] [CrossRef] [Green Version]
  49. Dehouck, Y.; Kwasigroch, J.M.; Gilis, D.; Rooman, M. PoPMuSiC 2.1: A web server for the estimation of protein stability changes upon mutation and sequence optimality. BMC Bioinform. 2011, 12, 151. [Google Scholar] [CrossRef]
  50. Rodrigues, C.H.; Pires, D.E.; Ascher, D.B.; RenéRen, I.; Rachou, R.; Oswaldo Cruz, F. DynaMut: Predicting the impact of mutations on protein conformation, flexibility and stability. Nucleic Acids Res. 2018, 46. [Google Scholar] [CrossRef]
  51. Frappier, V.; Chartier, M.; Najmanovich, R.J. ENCoM server: Exploring protein conformational space and the effect of mutations on protein function and stability. Nucleic Acids Res. 2015, 43, 395–400. [Google Scholar] [CrossRef] [Green Version]
  52. Pires, D.E.V.; Ascher, D.B.; Blundell, T.L. DUET: A server for predicting effects of mutations on protein stability using an integrated computational approach. Nucleic Acids Res. 2014, 42, W314–W319. [Google Scholar] [CrossRef] [PubMed]
  53. Pandurangan, A.P.; Ochoa-Montaño, B.; Ascher, D.B.; Blundell, T.L. SDM: A server for predicting effects of mutations on protein stability. Nucleic Acids Res. 2017, 45, W229–W235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Pires, D.E.V.; Ascher, D.B.; Blundell, T.L. mCSM: Predicting the effects of mutations in proteins using graph-based signatures. Bioinformatics 2014, 30, 335–342. [Google Scholar] [CrossRef] [Green Version]
  55. Al-Numair, N.S.; Martin, A.C.R. The SAAP pipeline and database: Tools to analyze the impact and predict the pathogenicity of mutations. BMC Genom. 2013, 14, S4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ittisoponpisan, S.; Islam, S.A.; Khanna, T.; Alhuzimi, E.; David, A.; Sternberg, M.J.E. Can Predicted Protein 3D Structures Provide Reliable Insights into whether Missense Variants Are Disease Associated? J. Mol. Biol. 2019, 431, 2197–2212. [Google Scholar] [CrossRef]
  57. Yates, C.M.; Filippis, I.; Kelley, L.A.; Sternberg, M.J.E. SuSPect: Enhanced prediction of single amino acid variant (SAV) phenotype using network features. J. Mol. Biol. 2014, 426, 2692–2701. [Google Scholar] [CrossRef]
  58. Bromberg, Y.; Rost, B. SNAP: Predict effect of non-synonymous polymorphisms on function. Nucleic Acids Res. 2007, 35, 3823–3835. [Google Scholar] [CrossRef] [Green Version]
  59. Yang, J.; Zhang, Y. Protein Structure and Function Prediction Using I-TASSER. Curr. Protoc. Bioinform. 2015, 52, 5–8. [Google Scholar] [CrossRef]
  60. Guzenko, D.; Lafita, A.; Monastyrskyy, B.; Kryshtafovych, A.; Duarte, J.M. Assessment of protein assembly prediction in CASP13. Proteins 2019. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Structural and flexibility analysis of p.(Gly154Asp), a TP53-predicted deleterious Variants of Uncertain Clinical Significance (VUS): (a) schematic representation of the location of the mutated Gly154 amino acid on the wild-type TP53 protein (Protein Data Bank (PDB) ID: 1GZH); (b) visual representation of the differences in vibrational entropy energy upon mutagenesis; (c) atomic interactions of the wild-type (Gly154) amino acid; and (d) atomic interactions of the mutant (Asp154) amino acid. Analysis was performed using the DynaMut webserver (http://biosig.unimelb.edu.au/dynamut/).
Figure 1. Structural and flexibility analysis of p.(Gly154Asp), a TP53-predicted deleterious Variants of Uncertain Clinical Significance (VUS): (a) schematic representation of the location of the mutated Gly154 amino acid on the wild-type TP53 protein (Protein Data Bank (PDB) ID: 1GZH); (b) visual representation of the differences in vibrational entropy energy upon mutagenesis; (c) atomic interactions of the wild-type (Gly154) amino acid; and (d) atomic interactions of the mutant (Asp154) amino acid. Analysis was performed using the DynaMut webserver (http://biosig.unimelb.edu.au/dynamut/).
Cancers 12 03140 g001
Table 1. Summary of the detected pathogenic variants and patient characteristics.
Table 1. Summary of the detected pathogenic variants and patient characteristics.
GeneExoncDNA ChangeAmino Acid ChangeFH Age Frequency
Per PV (%)
§ Frequency
Per Gene (%)
PALB24c.487_488delp.(Val163Leufs*4)2 × BC513/8 (37.5%)7/8 (87.5%)
PALB24c.487_488delp.(Val163Leufs*4)4 × BC65
PALB24c.487_488delp.(Val163Leufs*4)-67
PALB25c.1685-2A>Gp.?7 × BC542/8 (25%)
PALB25c.1685-2A>Gp.?-48
PALB212c.3350+4A>Gp.?BC501/8 (12.5%)
PALB23c.172_175delp.(Gln60Argfs*7)BC671/8 (12.5%)
TP536c.584T>Cp.(Ile195Thr)OC681/8 (12.5%)1/8 (12.5%)
FH: Family history of cancer, with first- or second-degree relatives diagnosed with breast, ovarian, pancreatic, or colorectal cancer. Age at diagnosis with Triple-Negative Breast Cancer (years). Patients carrying the pathogenic variant (PV)/patients tested positive for PVs in established breast cancer (BC) susceptibility genes. § Patients carrying PVs per gene/patients tested positive for PVs in established BC susceptibility genes. Mutation nomenclature according to LRG_308t1 (PALB2) and LRG_321t1 (TP53). BC, breast cancer; OC, ovarian cancer; PV, pathogenic variant.
Table 2. Study population characteristics.
Table 2. Study population characteristics.
CharacteristicsNo. of Patients (%) PV Carriers (%)
Total female patients163 (100%)8 (4.9%)
Age at diagnosis, y
Mean ± SD (range)50.6 ± 10.4 (27–75)58.8 ± 8.8 (48–68)
<2000
20–293 (1.8%)0
30–3922 (13.5%)0
40–4952 (31.9%)1 (12.5%)
50–5948 (29.4%)3 (37.5%)
≥6038 (23.3%)4 (50%)
* Family history of cancer
Breast or ovarian cancer83 (50.9%)6 (75%)
Pancreatic or colorectal cancer (No BC or OC)17 (10.4%)0
No breast, ovarian, pancreatic or colorectal63 (38.7%)2 (25%)
* First- or second-degree relatives PVs, Pathogenic variants (frameshift; splice site variants, ±3 positions; nonsense; missense variants reported as pathogenic or likely pathogenic) reported in established breast cancer susceptibility genes (PALB2 and TP53). BC, breast cancer; OC, ovarian cancer.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zanti, M.; Loizidou, M.A.; Michailidou, K.; Pirpa, P.; Machattou, C.; Marcou, Y.; Kyriakou, F.; Kakouri, E.; Tanteles, G.A.; Spanou, E.; et al. NGS Panel Testing of Triple-Negative Breast Cancer Patients in Cyprus: A Study of BRCA-Negative Cases. Cancers 2020, 12, 3140. https://doi.org/10.3390/cancers12113140

AMA Style

Zanti M, Loizidou MA, Michailidou K, Pirpa P, Machattou C, Marcou Y, Kyriakou F, Kakouri E, Tanteles GA, Spanou E, et al. NGS Panel Testing of Triple-Negative Breast Cancer Patients in Cyprus: A Study of BRCA-Negative Cases. Cancers. 2020; 12(11):3140. https://doi.org/10.3390/cancers12113140

Chicago/Turabian Style

Zanti, Maria, Maria A. Loizidou, Kyriaki Michailidou, Panagiota Pirpa, Christina Machattou, Yiola Marcou, Flora Kyriakou, Eleni Kakouri, George A. Tanteles, Elena Spanou, and et al. 2020. "NGS Panel Testing of Triple-Negative Breast Cancer Patients in Cyprus: A Study of BRCA-Negative Cases" Cancers 12, no. 11: 3140. https://doi.org/10.3390/cancers12113140

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop