Next Article in Journal
“Third Window” and “Single Window” Effects Impede Surgical Success: Analysis of Retrofenestral Otosclerosis Involving the Internal Auditory Canal or Round Window
Previous Article in Journal
Effects of Prevalent and Incident Chronic Kidney Disease on Cardiovascular Events in Patients with Atrial Fibrillation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis

by
Begona Jimenez Rodriguez
1,2,
Gema Diaz Córdoba
3,
Alicia Garrido Aranda
2,
Martina Álvarez
2,
Luis Vicioso
4,
Casilda Llácer Pérez
1,2,
Cristina Hernando
5,
Begoña Bermejo
5,6,
Ana Julve Parreño
5,
Ana Lluch
6,7,
Matthew B. Ryder
8,
Frederick S. Jones
8,
Johannes Fredebohm
8,
Frank Holtrup
8,
María Isabel Queipo-Ortuño
1,2,* and
Emilio Alba
1,2,6
1
Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, 29010 Málaga, Spain
2
Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
3
Radiology Department, Hospital Clínico Universitario Virgen de la Victoria de Málaga, 29010 Málaga, Spain
4
Histopathology Department, Hospital Clínico Universitario Virgen de la Victoria de Málaga, 29010 Málaga, Spain
5
Hospital Clínico de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
6
Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
7
Hospital Clínico de Valencia, Universidad de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
8
Sysmex Inostics, GmbH, 20251 Hamburg, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2019, 8(8), 1183; https://doi.org/10.3390/jcm8081183
Submission received: 18 July 2019 / Revised: 2 August 2019 / Accepted: 6 August 2019 / Published: 7 August 2019
(This article belongs to the Section Oncology)

Abstract

:
Circulating tumor DNA (ctDNA) has emerged as a non-invasive “liquid biopsy” for early breast cancer diagnosis. We evaluated the suitability of ctDNA analysis in the diagnosis of early breast cancer after mammography findings, comparing PIK3CA and TP53 mutations between tumor biopsies and pre-biopsy circulating DNA. Matched plasma and frozen fresh tissue biopsies from patients with Breast Imaging-Reporting and Data System (BIRADS) 4c/5 mammography findings and subsequent diagnosis of primary breast cancer were analyzed using NGS TruSeq Custom Amplicon Low Input Panel (Illumina) and plasma SafeSEQ (Sysmex Inostics). The same plasma and tumor mutations were observed in eight of 29 patients (27.6%) with four in TP53 and five in PIK3CA mutations. Sequencing analysis also revealed four additional ctDNA mutations (three in TP53 and one in PIK3CA) previously not identified in three patients tissue biopsy. One of these patients had mutations in both genes. Age, tumor grade and size, immunohistochemical (IHC) subtype, BIRADS category, and lymph node positivity were significantly associated with the detectability of these blood tumor-derived mutations. In conclusion, ctDNA analysis could be used in early breast cancer diagnosis, providing critical clinical information to improve patient diagnosis.

1. Introduction

Breast cancer (BC) is the most common cancer in females worldwide, the second most common cause of death from cancer among males and females (only preceded by lung cancer), and a leading cause of premature mortality from cancer as measured by average and total years of life lost [1]. GLOBOCAN estimated that there would be about 2.1 million newly diagnosed cases of female breast cancer in 2018, accounting for almost one in four cancer cases among women worldwide [2]. As earlier detection and treatment of this cancer can improve survival, great effort should be placed on improving screening methods [3].
The current standard of care for BC screening is mammography [4]. Radiologists use the BIRADS (Breast Imaging-Reporting and Data System) criteria to describe the probability of a mammographic finding to be a malignant tumor [5], although, currently, it is still necessary to perform a biopsy of the lesion to confirm the diagnosis. Tissue biopsies are invasive, costly, time-consuming, and in some cases not amenable for repetition. Moreover, in many cases, these biopsies do not reflect the intratumor heterogeneity, which may have important consequences for personalized-medicine approaches that commonly rely on single tumor-biopsy samples to portray tumor mutational landscapes [6].
To overcome the limitations of tissue biopsies, recent studies have shown that molecular driver alterations in tumor tissues can be detected by liquid biopsy for circulating tumor DNA (ctDNA) [7], which comprise small fragments of double-stranded DNA (160–180 base pairs) released in to the bloodstream from cells through the process of necrosis and apoptosis in tumor tissues [8]. Increased levels of cell-free DNA (cfDNA) in the blood are frequently observed in cancer patients [9]. In addition, ctDNA levels have been correlated with tumor burden [10,11]. Previous studies have shown that ctDNA analysis is a good tool to assess the tumor genomic landscape in metastatic breast cancer patients [10,12]. Moreover, Madic et al. reported that TP53 mutations were detected in the plasma of metastatic triple negative BC patients [13]. Interestingly, it was demonstrated that mutation tracking in ctDNA of breast cancer patients can identify early breast cancer patients at high risk of relapse [14]. Consistent with the previous data, Riva et al. found that a slow decrease of ctDNA level during neoadjuvant chemotherapy was strongly associated with shorter survival [15].
With the advance of next-generation sequencing (NGS) technologies, mutation analysis has become feasible and effective for clinical application in breast cancer [16]. According to the Cancer Genome Atlas (TCGA), the two most frequently mutated genes in primary BC are tumor protein p53 (TP53) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), which are generally conserved mutations between primary tumors and metastatic disease [17].
Digital-PCR (dPCR) has become widely used to detect plasma DNA mutations due to its high accuracy, sensitivity, and specificity. However, dPCR is limited to the detection of a relatively small number of hotspot target alleles for which primers and/or probes are specifically designed but not useful for identifying unreported novel mutations. In a previous study in patients with early-stage breast cancer, the ctDNA detection rate was low, presumably due to the limited number of gene and loci analyzed [18]. In contrast, targeted NGS like SafeSEQ has recently emerged as a new tool to increase the sensitivity of massively parallel sequencing system instruments for the identification of rare variants in plasma DNA. This sequencing strategy uses single-molecule barcoding before PCR amplification to reduce sequencing error and increase accuracy [19,20,21]. To our knowledge, SafeSEQ has so far been applied to plasma samples of metastatic colorectal cancer [22] and to gastrointestinal stromal tumors (GIST) patients [23].
Nevertheless, it is still uncertain whether the driver molecular alterations in ctDNA can be detected by plasma cfDNA NGS based assays in primary breast cancer patients at screening and how these are related to the tumor tissue mutation found in the same patient.
The aim of the present study was to investigate whether ctDNA analysis can be used in early breast cancer diagnosis, by sequencing plasma DNA taken before tumor biopsy in patients with mammographic findings. Then, we have compared PIK3CA and TP53 mutations between tumor biopsies and pre-biopsy circulating DNA as well as we have explored clinicopathological variables that may affect the detectability of these tumor-derived mutations in blood.

2. Experimental Section

2.1. Study Design and Patient Population

The pilot study included 29 patients with BIRADS 4c/5 mammography findings and subsequent diagnosis of primary breast cancer recruited at Virgen de la Victoria Hospital Málaga and Clinic University Hospital in Valencia. Blood samples were collected immediately (less than an hour) before tissue biopsy and prior to receiving any type of treatment. The matched tumor tissues were collected through core needle biopsies of breast tumors, which were subsequently fresh-frozen. Immunohistochemical (IHC) analysis was performed to quantify expression of human epidermal growth factor receptor 2 (HER2), hormone receptors (HR) and Ki67. Estrogen receptor (ER) and progesterone receptor (PR) were considered positive if tumors had more than 1% nuclear-stained cells. HER2 staining was scored on a scale of 0 to 3+, according to the HercepTest guidelines [24]. HER2 status was considered positive when graded as 3+, while 0 to 1+ were negative and 2+ was an inconclusive result and silver in situ hybridization (SISH) was performed in those cases to confirm positivity. Hormone receptor positive tumor plus Ki 67 index of <14% was considered a luminal A tumor while >14% was considered luminal B. IHC breast cancer subtypes were defined using a combination of these IHC markers as follows: Luminal A (ER-positive and/or PR-positive, HER2-negative and Ki-67 < 14%), luminal B (ER-positive and/or PR-positive, HER2-negative and Ki-67 ≥ 14%), HER2 positive (ER negative, PR negative, HER2 positive), and triple negative (ER, PR, and HER2 negative).
The study was approved by the research ethics committees at Virgen de la Victoria Hospital and Clínico de Valencia Hospital, all patients provided written informed consent. Research was conducted according to Good Clinical Practice and the Declaration of Helsinki.

2.2. DNA Extraction

Tumor DNA was isolated from fresh frozen tissue samples using the DNeasy Blood and Tissue Kit (Qiagen, Valencia, CA, USA) following the manufacturer’s instructions. Plasma blood samples of 10 mL were collected in STRECK tubes immediately before the tissue biopsy. Within 2 h after collection, plasma was separated from whole blood samples through centrifugation for 10 min at 3000 rpm at room temperature and stored at −80 °C until further use. The plasma samples were defrosted and were centrifuged again for 10 min at 13,000 rpm at room temperature prior to DNA extraction to remove debris. Isolation of DNA from 2 mL of plasma was performed using QIAamp Circulating Nucleic Acid Kit (Qiagen) according to the manufacturer’s instructions and eluted in 140 μL of AVE buffer (Qiagen). The total amount of amplifiable human genomic DNA isolated from plasma samples was quantified using a modified version of human long interspersed element 1 (LINE-1) real-time PCR assay and reported as genome equivalents (GE) [25,26].

2.3. Targeted Sequencing

The NGS study on fresh frozen tissue samples was performed using a customized design of TruSeq Custom Amplicon Low Input Panel (Illumina), which includes the full region of the PIK3CA and TP53 genes (the most commonly mutated genes in early-stage BC according to TCGA). Libraries were constructed using Tru-Seq reagents from Illumina according to the standard protocol provided by Illumina. All DNA samples were diluted to the same initial concentration (25 ng/μL). In order to artificially increase the genetic diversity, 10% DNA from phage PhiX was added to the library of genomic DNA before loading on the flow-cell. Exon regions were captured in a solution using the Agilent SureSelect v.5 kit according to the manufacturer’s instructions (Agilent, Santa Clara, CA, USA). Paired-end sequencing was performed using a HiSeq 2500 Genome Analyzer (Illumina, San Diego, CA, USA). The sequences were aligned to the human genome reference sequence (hg19) using the Eland algorithm of CASAVA 1.8 software (Illumina, San Diego, CA, USA). The chastity filter of the BaseCall software of Illumina was used to select sequence reads for subsequent analysis. The ELANDv2 algorithm of CASAVA 1.8 software (Illumina, San Diego, CA, USA) was then applied to identify point mutations and small insertions and deletions. Potential somatic mutations were filtered and manually curated.

2.4. Plasma DNA Sequencing (SafeSEQ)

Plasma sequencing was performed using Plasma SafeSEQ (Sysmex Inostics, Hamburg, Germany). SafeSEQ analysis was performed on up to 10,000 GE (~33 ng DNA) per sample. Briefly, human genomic DNA isolated from plasma samples was amplified (15 cycles) in 10 replicate PCR wells using primers containing unique identifier sequences (UIDs), which consisted of 14 random bases with an equal probability of A, C, T, and G, to allow for the distinction of each template molecule. The amplified reactions were purified using AMPure XP beads (Beckman Coulter) and eluted in 250 mL of Buffer EB (Qiagen). A fraction of purified PCR product was then amplified in the second round of PCR with universal primers. The PCR products were purified with AMPure XP and sequenced on an Illumina MiSeq instrument. High-quality sequence reads were selected based on quality scores, which were generated by the Illumina sequencing instrument to indicate the probability that an error was made in base calling. The template-specific portion of the reads was matched to reference sequences. Reads from a common template molecule were then grouped based on the UIDs that were incorporated as molecular barcodes. Variants calls from the SafeSEQ assay were considered real if ≥90% of the PCR fragments with the same UID contain an identical mutation. Wells with fewer than 200 UIDs as a result of poor amplification were excluded from analysis. Once the analysis was performed, the following criteria were followed to call a mutation: (1) Variant allele frequency (VAF) of at least 0.05% and (2). The mutation needed to be detected in at least two out of 10 replicate wells.

2.5. Statistical Analysis

The statistical analysis was performed with SPSS (19.0, SPSS Inc., Chicago, IL, USA). For association analyses between clinicopathological variables and PIK3CA and TP53 derived-mutations in blood, Spearmen correlation and Fisher’s exact test (for categorical variables) were used. The threshold for statistical significance was set at p < 0.05.

3. Results

3.1. Patient Characteristics

The clinicopathological features of the tumors of the enrolled breast cancer patients are shown in Table 1. The average age of the patients was 64 years (range 44–92 years). The 29 primary breast tumors examined included 24 (82.7%) invasive ductal carcinoma (CDI), two (6.9%) invasive lobular carcinoma (CLI), two (6.9%) papillary carcinomas and one tubular carcinoma (3.4%). Fourteen tumors (48.3%) showed diameters of less than 2 cm, while 15 (51.7%) showed diameters of more than 2 cm. Axillary lymph node metastasis was detected by core needle biopsy in nine patients (31.0%). Ten (34.5%) patients without lymph node metastasis had tumors with diameters of more than 2 cm. Immunohistochemistry was used to determine IHC subtypes of tumors as follows, 14 (48.3%) luminal A tumors, 11 (37.9%) luminal B tumors, three (10.3%) triple-negative tumors and one (3.4%) HER2-positive tumor. With respect to hormone receptor status, 25 (86.2%) of 29 BC patients were estrogen receptor (ER) positive and 17 (58.6%) of 29 patients were progesterone receptor (PR) negative.
Concerning the tumor grade, the majority of the patients presented grade 2 tumors (16 patients, 55%) while seven patients (24.5%) had grade 3 tumors and five (17%) patients had grade 1 tumors. The tumor grade was unknown in one patient (3.5%).

3.2. Analysis of PIK3CA and TP53 Mutations in Fresh Frozen Tissue Samples of Primary Breast

Mutations of the PIK3CA and TP53 genes were analyzed in genomic DNA from fresh frozen tissue samples of 29 primary breast cancer using the Illumina platform and the TruSeq Custom Amplicon Low Input Panel. Samples were successfully sequenced, and data quality was high, probably because DNA was isolated from fresh tissue material and had not been degraded. A total of 34 somatic mutations were detected in the 29 primary breast cancer patients. Of the 29 patients, 19 (65.5%) had PIK3CA mutations, six (20.7%) had TP53 mutations, and four (13.8%) had both PIK3CA and TP53 mutations. One (3.4%) patient had two PIK3CA mutations. In summary, all tumor samples were found to harbor at least one mutation.
In total, PIK3CA somatic mutations were detected in 23 of 29 tumors (79.3%), including eight tumors with c.3140A > G (27.6%), five with c.1633G > A (17.2%), three with c.3140A > T (10.3%), three with c.1624G > A (10.34%), two with c.1637A > G (6.9%), one with c.3145G > C (3.4%) and one with both c.3129G > A (3.44%) and c.3130A > C (3.4%) mutations (Table 2). With respect to the TP53 gene, mutations were detected in 10 of 29 tumors (34.5%), including two tumors with c.524G > A (6.9%) and eight tumors with c.637C > T (3.4%), c.398T > A (3.4%), c.743G > T (3.4%), c.1028_1029delAG (3.4%), c.842A > C (3.4%), c.832C > T (3.4%), c.734G > T (3.4%), and c.587G > C (3.4%), respectively (Table 2). The VAF of the mutated PIK3CA allele in these tumors was 22.4% (range 7.4–82.4%) and 18.0% (range 29.3–75.2%) for TP53.
The PIK3CA mutations were detected in 13 (92.8%) of 14 luminal A tumors, nine (81.8%) of 11 luminal B tumors (one harboring two different PIK3CA mutations), and one (33.3%) of three triple-negative tumors. The TP53 mutations were found in three (21.4%) of 14 luminal A tumors, four (36.4%) of 11 luminal B tumors, two (66.7%) of three triple-negative and one (100%) of one Her2-positive tumor. Interestingly, the TP53 mutations coexisted with PIK3CA mutations in two of three luminal A tumors and in two of four luminal B tumors.

3.3. PIK3CA and TP53 Mutations Detected in ctDNA

Plasma samples from 29 primary early stage breast cancer patients were analyzed by SafeSEQ. The analysis revealed the presence of 13 mutations in the ctDNA of 10 patients out of 29 (34%). There were six different PIK3CA mutations (c.3140A > T, c.3145G > C, c.1651C > A, c.3140A > G, c.1633G > A, c.1624G > A) and seven different TP53 mutations (c.398T > A, c.641A > G, c.743G > T, c.587G > C, c.637C > T, c.659A > G, c.748C > T). The median VAF for TP53 was 1.03% (range 0.09–20.56%) and 0.17% for PIK3CA (range 3.6–0.01%).
One patient with a grade 3 luminal B tumor had mutations on both genes, TP53 (c.743G > T) and PIK3CA (c.3140A > T). One patient with a grade 2 luminal A tumor had two different PIK3CA mutations (c.1651C > A and c.1633G > A) and one patient with a grade 2 luminal A tumor had two different TP53 mutations (c.641A > G and c.748C > T) (Table 3). The rest of TP53 mutations were mostly detected in patients with luminal B tumors (three of six (50%)), followed by one (16.7%) patient with triple-negative tumor and two (33.3%) patients with luminal A tumors. With respect to PIK3CA mutations, were detected mostly in patients with luminal A tumors (three of five (60%)), while two (40%) patients with PIK3CA mutations had luminal B tumors.

3.4. Comparison of PIK3CA and TP53 Mutations Detection Results from Tumor Tissue and ctDNA

The same plasma and tumor somatic mutations were observed in eight (27.6%) of 29 patients. Five PIK3CA and four TP53 mutations were detected in eight matched pairs of primary tumors and plasma samples 3 TP53 (c.398T > A, c.587G > C, c.637C > T), 4 PIK3CA (c.3140A > G, c.1633G > A, C.1624G > A, C.3145G > C), and one patient with mutations in both genes (TP53 c.743G > T, PIK3CA c.3140A > T) (Table 4).
Moreover, base transition (purine-pyrimidine and pyrimidine-purine) was more frequent than transversions (pyrimidine-pyrimidine and purine-purine). All these concordant somatic mutations were pathogenic, eight of them were missense mutations and one was a nonsense mutation. PIK3CA concordant mutations were detected in three (21.4%) of 14 luminal A tumors, and two (18.2%) of 11 luminal B tumors, while TP53 concordant mutations were found in three (27.3%) of 11 luminal B tumors, and one (33.3%) of three triple-negative tumors. Moreover, all patients that showed these concordant mutations had grade 2–3 invasive ductal carcinoma.
Interestingly, there were three TP53 mutations (c.641A > G, c.659A > G, c.748C > T) and one PIK3CA mutation (c.1651C > A) detected in plasma ctDNA of three different patients that were not detected in the matched tissue sample, all of them found in grade 2 luminal A tumors.

3.5. Clinicopathological Variables Associated with the Detectability of Tumor-Derived PIK3CA and TP53 Mutations in Blood

We analyzed the association between the detectability of tumor PIK3CA and TP53 mutations in ctDNA and patient clinicopathological variables (Table 5). The analysis indicated that the presence of ctDNA mutations was significantly associated with a lower age of the patients (p = 0.040), higher tumor size (p = 0.033) and tumor grade (p = 0.041), the presence of lymph node positivity (p < 0.001), a BIRADS category 5 (p = 0.004) and the IHC cancer subtype (p = 0.025).

4. Discussion

In this pilot study, we have demonstrated the possible implementation of plasma tumor DNA detection as noninvasive means for early breast cancer screening.
Data are currently limited about whether ctDNA analyses would be applicable to the screening process in early breast cancer detection, in part because the low tumor burden of early stage disease makes the detection of ctDNA difficult [18,20] and very low levels of plasma ctDNA are not normally detectable [25,27]. In our study and with the aim of improving this detection problem, the novel approach of SafeSEQ was employed. This technique is able to detect rare mutations with an allele frequency <0.001% [19].
In our population of 29 breast cancer screening patients, the NGS analysis of the tumor biopsies detected PIK3CA mutations in 79.3% (23/29) and TP53 in 34.5% (10/29) of primary breast cancer patients. While, one third (10/29, 34%) of the patients were found to have mutations in plasma samples. In total, 13 mutations were found, six (46.1%) different PIK3CA mutations and seven (53.8%) different TP53 mutations. The VAF in ctDNA was low (0.05%–3.60%) with the exception of one case with 20.56% in a patient later found to have lung metastasis in the CT scan performed as part of the staging process.
Of the 13 plasma mutations, nine were concordant with the mutations found in matched tissue samples (five PIK3CA and four TP53 mutations) in eight of 29 patients. Of these nine mutations, four PIK3CA mutations within the PI3Ka (p.E545K and p.E542K) and PI3Kc (p.H1047R and p.H1047L) have been previously reported as important hotspots in breast cancer [28,29,30,31]. These findings may highlight the potential role of ctDNA to capture the molecular abnormalities of the tumor at the very first stage of the disease, even before having a diagnosis.
On the other hand, in this study four additional mutation, three TP53 (c.641A > G, c.659A > G, c.748C > T) and one PIK3CA (c.1651C > A) not found in the tissue were found in plasma of three different patients, which did not have any undetected metastases or additional undiagnosed tumors. All these TP53 mutations have been reported in the catalog of somatic mutations in cancer (COSMIC) as driver mutations. This finding may be related to the capability of ctDNA to capture the tumor heterogeneity, which is indeed a limitation of tumor biopsies [32].
To date, a few applications of the liquid biopsies have been integrated into daily clinical practice, such as for molecular profiling of the tumor and monitoring resistance [33]. In the case of breast cancer, some studies have demonstrated its potential role in tracking therapeutic response and detection of relapse [14,34,35]. In a study with patients with more advanced locoregional disease, Riva et al. detected TP53 plasma mutations in 27 of 36 triple-negative patients (75%) before neoadjuvant chemotherapy treatment using dPCR [13]. Likewise, in another study PIK3CA (p.H1047R, p.E545K, and p.E542K) mutations in ctDNA were found in 22% of 110 stage I-III BC patients [36].
Two recent studies have also investigated the potential of liquid biopsy for the early detection of different type of cancer. Cohen et al. applied a screening method combining mutation detection in ctDNA with circulating protein markers in 1005 patients with nonmetastatic, clinically detectable tumors across eight common tumor types (ovary, liver, stomach, pancreas, esophagus, colorectal, lung, and breast). The median sensitivity of this test among the eight cancer types evaluated was 70% and ranged from 98% in ovarian cancers to 33% in breast cancers. At this sensitivity, the specificity was >99%, only seven of the 812 individuals without known cancers scored positive [37]. Chan et al. used detection of Epstein–Barr Virus (EBV) DNA in plasma to screen for nasopharyngeal carcinoma in 20.174 Chinese patients. Overall, the sensitivity and specificity of this approach were 97.1% and 98.6%, respectively [38].
In addition, several clinicopathological variables have been found associated with PIK3CA and TP53 plasma ctDNA mutation detectability in early breast cancer patients. We found a trend for a higher plasma ctDNA mutation burden in patients with clinical characteristics associated with more aggressive disease factors such as higher tumor grade (grade II-III), and tumor size, BIRADS category 5, the presence of positive lymph nodes and the IHC subtype present, being more frequently mutated in luminal A tumors followed by luminal B tumors. Similar results have been described in a previous study in primary breast cancer patients where clinicopathological features, such as N stage and hormone receptor status, were associated with the detectability of tumor-derived mutations in blood [39]. Then, this finding may help to explain the absence of ctDNA mutations for some patients in the early breast cancer screening.
Regarding clonal hematopoiesis, detectable clonal expansions most frequently involved somatic mutations in DNMT3A, ASXL1, and TET2 genes [40]. Furthermore, recent studies demonstrated that the genes more commonly associated with clonal hematopoiesis of indeterminate potential (CHIP) were DNMT3A, ASXL1, TET2, JAK2, SF3B1, CBL, GNAS, and IDH2 [41]. Interestingly, when healthy individuals were assessed for CHIP, no alterations in driver genes related to solid cancers were detected [42]. For these reasons, clonal hematopoiesis was not analyzed in our population, since TP53 and PIK3CA are commonly mutated genes in breast cancer.

5. Conclusions

In conclusion, to our knowledge, this is the first pilot study done in patients at the breast cancer screening process before undergoing any invasive diagnostic procedure or treatment, which shows that ctDNA analysis could be used in early breast cancer diagnosis. This study has demonstrated that ctDNA may reflect the PIK3CA and TP53 tumor-derived mutations present in very early breast cancer patients. Nevertheless, several clinicopathological variables can affect the detectability of these ctDNA mutations. Although our study population is small and larger-scale studies will be necessary to validate our results, we can conclude that early ctDNA testing can provide critical clinical information that may improve patient diagnosis.

Author Contributions

Conception and design: B.J.R., M.I.Q. and E.A. Study supervision: M.I.Q.O. and E.A. Development of methodology: B.J.R., F.S.J., M.I.Q.O. and E.A. Acquisition of data (acquired and managed patients, provided facilities, etc.): B.J.R., G.D.C., L.V., C.L.P., C.H., B.B., A.J.P., A.L., M.B.R., J.F. and F.H. Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B.J.R., M.A., F.S.J., M.I.Q.O. and E.A. Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): B.J.R., G.D.C., L.V., C.L.P., C.H., B.B., A.J.P., A.L., M.B.R., J.F. and F.H. Writing, review, and/or revision of the manuscript: The corresponding author (M.I.Q.O.) and all of the authors have read and approved the final submitted manuscript and they participated in writing and reviewing the manuscript.

Funding

Mariía Isabel Queipo-Ortunńo is a recipient of a “Miguel Servet Type II” program (CPI13/00003) from ISCIII, co-funded by the Fondo Europeo de Desarrollo Regional-FEDER, Madrid, Spain and also belongs to the regional “Nicolas Monardes” research program of the Consejeriía de Salud (C-0030-2018), Junta de Andaluciía, Spain. Begoña Jiménez Rodríguez is a recipient of “Fundación SEOM-Font Vella grant for Breast Cancer Research Projects”, thanks to this grant a part of this study was funded.

Acknowledgments

The authors are thankful to all women who participated in the study. We thank Ian Johnstone for his help with the English language version of the text.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BCBreast cancer;
BIRADSBreast Imaging-Reporting and Data System;
ctDNACirculating tumor DNA;
NGSNext generation sequencing;
TCGACancer Genome;
TP53tumor protein p53;
PIK3CAphosphatidylinositol-4;5-bisphosphate 3-kinase catalytic subunit alpha;
dPCRDigital-PCR;
GISTGastrointestinal stromal tumors;
IHCImmunohistochemical;
HER2human epidermal growth factor receptor 2;
HRHormone receptors;
EREstrogen receptor;
PRProgesterone receptor;
SISHSilver in situ hybridization;
VAFVariant allele frequency;
CDIInvasive ductal carcinoma;
CLIInvasive lobular carcinoma;
EBVEpstein–Barr Virus;
OROdds ratio;
CIConfidence interval;
COSMICCatalog of somatic mutations in cancer;
CHIPClonal hematopoiesis of indeterminate potential

References

  1. Smith, R.A.; Manassaram-Baptiste, D.; Brooks, D.; Doroshenk, M.; Fedewa, S.; Saslow, D.; Brawley, O.W.; Wender, R. Cancer screening in the United States; 2018: A review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J. Clin. 2018, 68, 297–316. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. Loomans-Kropp, H.A.; Umar, A. Cancer prevention and screening: The next step in the era of precision medicine. NPJ Precis. Oncol. 2019, 3, 3. [Google Scholar] [CrossRef] [PubMed]
  4. Seely, J.M.; Alhassan, T. Screening for breast cancer in 2018-what should we be doing today? Curr. Oncol. 2018, 25 (Suppl. 1), S115–S124. [Google Scholar] [CrossRef] [PubMed]
  5. American College of Radiology. ACR BI-RADS 5th Edition Changes. Available online: http://www.acr.org/∼/media/acr/documents/pdf/qualitysafety/resources/birads/birads_v5_changes.pdf (accessed on 1 January 2017).
  6. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; Tarpey, P. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef] [PubMed]
  7. Schwaederle, M.; Husain, H.; Fanta, P.T.; Piccioni, D.E.; Kesari, S.; Schwab, R.B.; Patel, S.P.; Harismendy, O.; Ikeda, M.; Parker, B.A.; et al. Use of Liquid Biopsies in Clinical Oncology: Pilot Experience in 168 Patients. Clin. Cancer. Res. 2016, 22, 5497–5505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Diaz, L.A., Jr.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef] [PubMed]
  9. Valpione, S.; Gremel, G.; Mundra, P.; Middlehurst, P.; Galvani, E.; Girotti, M.R.; Lee, R.J.; Garner, G.; Dhomen, N.; Lorigan, P.C.; et al. Plasma total cell-free DNA (cfDNA) is a surrogate biomarker for tumour burden and a prognostic biomarker for survival in metastatic melanoma patients. Eur. J. Cancer 2018, 88, 1–9. [Google Scholar] [CrossRef]
  10. Dawson, S.J.; Rosenfeld, N.; Caldas, C. Circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 369, 93–94. [Google Scholar] [CrossRef]
  11. McEvoy, A.C.; Warburton, L.; Al-Ogaili, Z.; Celliers, L.; Calapre, L.; Pereira, M.R.; Khattak, M.A.; Meniawy, T.M.; Millward, M.; Ziman, M.; et al. Correlation between circulating tumour DNA and metabolic tumour burden in metastatic melanoma patients. BMC Cancer 2018, 18, 726. [Google Scholar] [CrossRef]
  12. Alimirzaie, S.; Bagherzadeh, M.; Akbari, M.R. Liquid biopsy in breast cancer: A comprehensive review. Clin. Genet. 2019, 95, 643–660. [Google Scholar] [CrossRef]
  13. Madic, J.; Kiialainen, A.; Bidard, F.C.; Birzele, F.; Ramey, G.; Leroy, Q.; Frio, T.R.; Vaucher, I.; Raynal, V.; Bernard, V.; et al. Circulating tumor DNA and circulating tumor cells in metastatic triple negative breast cancer patients. Int. J. Cancer. 2015, 136, 2158–2165. [Google Scholar] [CrossRef]
  14. Garcia-Murillas, I.; Schiavon, G.; Weigelt, B.; Ng, C.; Hrebien, S.; Cutts, R.J.; Cheang, M.; Osin, P.; Nerurkar, A.; Kozarewa, I.; et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci. Transl. Med. 2015, 7, 302ra133. [Google Scholar] [CrossRef]
  15. Riva, F.; Bidard, F.C.; Houy, A.; Saliou, A.; Madic, J.; Rampanou, A.; Hego, C.; Milder, M.; Cottu, P.; Sablin, M.P.; et al. Patient-Specific Circulating Tumor DNA Detection during Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Clin. Chem. 2017, 63, 691–699. [Google Scholar] [CrossRef] [Green Version]
  16. Lv, P.P.; Meng, Y.; Lv, M.; Feng, C.; Liu, Y.; Li, J.Y.; Yu, D.Q.; Shen, Y.; Hu, X.L.; Gao, Q.; et al. Altered thyroid hormone profile in offspring after exposure to high estradiol environment during the first trimester of pregnancy: A cross-sectional study. BMC Med. 2014, 12, 240. [Google Scholar] [CrossRef]
  17. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [Green Version]
  18. Beaver, J.A.; Jelovac, D.; Balukrishna, S.; Cochran, R.L.; Croessmann, S.; Zabransky, D.J.; Wong, H.Y.; Toro, P.V.; Cidado, J.; Blair, B.G.; et al. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin. Cancer Res. 2014, 20, 2643–2650. [Google Scholar] [CrossRef]
  19. Kinde, I.; Wu, J.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl. Acad. Sci. USA 2011, 108, 9530–9535. [Google Scholar] [CrossRef] [Green Version]
  20. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra24. [Google Scholar] [CrossRef]
  21. Buono, G.; Gerratana, L.; Bulfoni, M.; Provinciali, N.; Basile, D.; Giuliano, M.; Corvaja, C.; Arpino, G.; Del Mastro, L.; De Placido, S.; et al. Circulating tumor DNA analysis in breast cancer: Is it ready for prime-time? Cancer. Treat. Rev. 2019, 73, 73–83. [Google Scholar] [CrossRef] [Green Version]
  22. Tie, J.; Kinde, I.; Wang, Y.; Wong, H.L.; Roebert, J.; Christie, M.; Tacey, M.; Wong, R.; Singh, M.; Karapetis, C.S.; et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015, 26, 1715–1722. [Google Scholar] [CrossRef]
  23. Fredebohm, J.; Mehnert, D.H.; Löber, A.K.; Holtrup, F.; Van Rahden, V.; Angenendt, P.; Diehl, F. Detection and Quantification of KIT Mutations in ctDNA by Plasma Safe-SeqS. Adv. Exp. Med. Biol. 2016, 924, 187–189. [Google Scholar]
  24. Wolff, A.C.; Hammond, M.E.; Schwartz, J.N.; Hagerty, K.L.; Allred, D.C.; Cote, R.J. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J. Clin. Oncol. 2007, 25, 118–145. [Google Scholar] [CrossRef]
  25. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef]
  26. Rago, C.; Huso, D.L.; Diehl, F.; Karim, B.; Liu, G.; Papadopoulos, N.; Samuels, Y.; Velculescu, V.E.; Vogelstein, B.; Kinzler, K.W.; et al. Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Res. 2007, 67, 9364–9370. [Google Scholar] [CrossRef]
  27. Reinert, T.; Schøler, L.V.; Thomsen, R.; Tobiasen, H.; Vang, S.; Nordentoft, I.; Lamy, P.; Kannerup, A.S.; Mortensen, F.V.; Stribolt, K.; et al. Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut 2016, 65, 625–634. [Google Scholar] [CrossRef]
  28. Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S.M.; Riggins, G.J.; et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004, 304, 554. [Google Scholar] [CrossRef]
  29. Silwal-Pandit, L.; Langerød, A.; Børresen-Dale, A.L.; Silwal-Pandit, L.; Langerod, A.; Borresen-Dale, A.L. TP53 Mutations in Breast and Ovarian Cancer. Cold Spring Harb. Perspect. Med. 2017, 7, a026252. [Google Scholar] [CrossRef]
  30. Chang, M.T.; Asthana, S.; Gao, S.P.; Lee, B.H.; Chapman, J.S.; Kandoth, C.; Gao, 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]
  31. Cheng, J.; Fu, S.; Wei, C.; Tania, M.; Khan, M.; Imani, S.; Zhou, B.; Chen, H.; Xiao, X.; Wu, J.; et al. Evaluation of PIK3CA mutations as a biomarker in Chinese breast carcinomas from Western China. Cancer Biomark. 2017, 19, 85–92. [Google Scholar] [CrossRef]
  32. Cai, Z.X.; Chen, G.; Zeng, Y.Y.; Dong, X.Q.; Lin, M.J.; Huang, X.H.; Zhang, D.; Liu, X.L.; Liu, J.F. Circulating tumor DNA profiling reveals clonal evolution and real-time disease progression in advanced hepatocellular carcinoma. Int. J. Cancer 2017, 141, 977–985. [Google Scholar] [CrossRef]
  33. Malapelle, U.; Sirera, R.; Jantus-Lewintre, E.; Reclusa, P.; Calabuig-Fariñas, S.; Blasco, A.; Pisapia, P.; Rolfo, C.; Camps, C. Profile of the Roche cobas EGFR mutation test v2 for non-small cell lung cancer. Expert Rev. Mol. Diagn. 2017, 17, 209–215. [Google Scholar] [CrossRef]
  34. Sparano, J.; O’neill, A.; Alpaugh, K.; Wolff, A.C.; Northfelt, D.W.; Dang, C.T.; Sledge, G.W.; Miller, K.D. Association of Circulating Tumor Cells with Late Recurrence of Estrogen Receptor-Positive Breast Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol. 2018, 4, 1700–1706. [Google Scholar] [CrossRef]
  35. Dawson, S.J.; Tsui, D.W.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef]
  36. Oshiro, C.; Kagara, N.; Naoi, Y.; Shimoda, M.; Shimomura, A.; Maruyama, N.; Shimazu, K.; Kim, S.J.; Noguchi, S. PIK3CA mutations in serum DNA are predictive of recurrence in primary breast cancer patients. Breast Cancer Res. Treat. 2015, 150, 299–307. [Google Scholar] [CrossRef]
  37. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [Green Version]
  38. Chan, K.A.; Woo, J.K.; King, A.; Zee, B.C.; Lam, W.J.; Chan, S.L.; Chu, S.W.; Mak, C.; Tse, I.O.; Leung, S.Y.; et al. Analysis of Plasma Epstein-Barr Virus DNA to Screen for Nasopharyngeal Cancer. N. Engl. J. Med. 2017, 377, 513–522. [Google Scholar] [CrossRef]
  39. Zhou, Y.; Xu, Y.; Gong, Y.; Zhang, Y.; Lu, Y.; Wang, C.; Yao, R.; Li, P.; Guan, Y.; Wang, J.; et al. Clinical factors associated with circulating tumor DNA (ctDNA) in primary breast cancer. Mol. Oncol. 2019, 13, 1033–1046. [Google Scholar] [CrossRef]
  40. Genovese, G.; Kähler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [Green Version]
  41. Hrebien, S.; Citi, V.; Garcia-Murillas, I.; Cutts, R.; Fenwick, K.; Kozarewa, I.; McEwen, R.; Ratnayake, J.; Maudsley, R.; Carr, T.H.; et al. Early ctDNA dynamics as a surrogate for progression-free survival in advanced breast cancer in the BEECH trial. Ann. Oncol. 2019, 30, 945–952. [Google Scholar] [CrossRef]
  42. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan241. [Google Scholar] [CrossRef]
Table 1. Clinicopathological characteristics of primary breast cancer patients.
Table 1. Clinicopathological characteristics of primary breast cancer patients.
Diagnostic Age (years)
Median (range)
64 years (44–92)
Mammographic Tumor Size n (%)
<2 cm15 (51.7)
2 to 5 cm13 (44.8)
>5 cm1 (3.4)
Tumor type
CDI24 (82.7)
CLI2 (6.9)
Papilar carcinoma2 (6.9)
Tubular carcinoma1 (3.4)
Tumor grade n (%)
I5 (17.2)
II16 (55.2)
III7 (24.13)
Unknown1 (3.4)
Axilar lymph node
Positive9 (31.3)
Negative20 (68.9)
Progesterone receptor n (%)
Positive12 (41.4)
Negative17 (58.6)
Estrogen receptor n (%)
Positive25 (86.20)
Negative4 (13.79)
HER2 status n (%)
Positive1 (3.4)
Negative28 (96.6)
IHC subtype n (%)
Luminal A tumor14 (48.3)
Luminal B tumor11 (37.9)
HER2-positive tumor1 (3.4)
Triple negative tumor3 (10.3)
BIRADS category
4c14 (48.3)
515 (51.7)
IDC: Invasive ductal carcinoma; ILC: Invasive lobular carcinoma; HER2: Human epidermal growth factor receptor 2; IHC: Immunohistochemistry; BIRADS: Breast Imaging-Reporting and Data System.
Table 2. Pathogenic mutations of PIK3CA and TP53 genes in tumors samples of primary breast cancer patients.
Table 2. Pathogenic mutations of PIK3CA and TP53 genes in tumors samples of primary breast cancer patients.
Patient IDGeneHGVSProteinChrhg19Tumor VAF (%)COSMIC
2MSPIK3CAc.3140A > Tp.H1047L317895208531.2COSM776
2MSTP53c.524G > Ap.R175H17757840658.3COSM10648
7MSPIK3CAc.3140A > Gp.H1047R317895208531COSM775
9MSTP53c.637C > Tp.R213Ter17757821246.9COSM10654
10MSPIK3CAc.3140A > Gp.H1047R317895208517COSM775
13MSTP53c.398T > Ap.M133K17757853275.2COSM11781
14MSPIK3CAc.3140A > Gp.H1047R317895208525.3COSM775
16MSPIK3CAc.3140A > Tp.H1047L317895208528.2COSM776
16MSTP53c.743G > Tp.R248L17757753843.1COSM6549
17MSPIK3CAc.3140A > Gp.H1047R317895208530.1COSM775
19MSPIK3CAc.1624G > Ap.E542K317893608247.7COSM760
22MSTP53c.1028_1029delAGp.E343AfsTer317757399773.5COSM5752326
23MSPIK3CAc.1633G > Ap.E545K317893609130.6COSM763
23MSTP53c.524G > Ap.R175H17757840668.9COSM10648
30MSPIK3CAc.1633G > Ap.E545K317893609156.2COSM763
31MSPIK3CAc.1633G > Ap.E545K317893609110.6COSM763
32MSPIK3CAc.3140A > Gp.H1047R317895208512.7COSM775
35MSPIK3CAc.1633G > Ap.E545K317893609128.9COSM763
36MSPIK3CAc.3140A > Tp.H1047L317895208544.9COSM776
40MSPIK3CAc.1624G > Ap.E542K317893608225.5COSM760
41MSTP53c.832C > Tp.P278S17757710629.1COSM10939
43MSTP53c.734G > Tp.G245V17757754745.3COSM11196
44MSTP53c.587G > Cp.R196P17757826235.8COSM43814
47MSPIK3CAc.3140A > Gp.H1047R317895208510.6COSM775
50MSPIK3CAc.3129G > Ap.M1043I317895207433.4COSM29313
50MSPIK3CAc.3130A > Cp.N1044H317895207533.4-----
52MSPIK3CAc.1637A > Gp.Q546R317893609511.6COSM12459
56MSPIK3CAc.1624G > Ap.E542K317893608216.9COSM760
65MSPIK3CAc.3140A > Gp.H1047R317895208523.6COSM775
67MSPIK3CAc.3145G > Cp.G1049R317895209082.4COSM12597
67MSTP53c.842A > Cp.D281A17757709644.6COSM11665
68MSPIK3CAc.3140A > Gp.H1047R317895208515.9COSM775
79MSPIK3CAc.1637A > Gp.Q546R31789360957.4COSM12459
80MSPIK3CAc.1633G > Ap.E545K317893609129.5COSM763
HGVS: Human Genome Variation Society; Chr: Chromosome; COSMIC: Catalog of somatic mutations in cancer; Tumor VAF: Tumor variant allele frequency.
Table 3. TP53 and PIK3CA mutations in ctDNA from primary breast cancer patients.
Table 3. TP53 and PIK3CA mutations in ctDNA from primary breast cancer patients.
Patient IDGeneHGVSSafeSEQ VAF (%)Proteinhg19COSMIC
7MSPIK3CAc.3140A > G0.14%p.H1047R178952085COSM775
9MSTP53c.637C > T0.91%p.R213Ter7578212COSM10654
13MSTP53c.398T > A20.56%p.M133K7578532COSM11781
16MSPIK3CAc.3140A > T3.60%p.H1047L178952085COSM776
16MSTP53c.743G > T2.11%p.R248L7577538COSM6549
30MSPIK3CAc.1651C > A0.17%p.L551I178936109----
30MSPIK3CAc.1633G > A0.05%p.E545K178936091COSM763
10MSTP53c.641A > G3.88%p.H214R7578208COSM43687c
10MSTP53c.748C > T0.09%p.P250S7577533COSM43695c
31MSTP53c.659A > G0.16%p.Y220C7578190COSM10758c
40MSPIK3CAc.1624G > A0.20%p.E542K178936082COSM760
44MSTP53c.587G > C1.03%p.R196P7578262COSM43814
67MSPIK3CAc.3145G > C0.39%p.G1049R178952090COSM12597
HGVS: Human Genome Variation Society; COSMIC: Catalog of somatic mutations in cancer; SafeSEQ VAF: Plasma variant allele frequency.
Table 4. Concordant PIK3CA and TP53 somatic mutations between tumors and ctDNA in primary breast cancer patients.
Table 4. Concordant PIK3CA and TP53 somatic mutations between tumors and ctDNA in primary breast cancer patients.
Patient IDDiagnostic MammographyMarkersTypeGeneHGVSSafeSEQ VAF (%)ProteinTumor VAF (%)COSMICChr
13MSIDC, grade 3ER+/HER2+Luminal BTP53c.398T > A20.56p.M133K75.2COSM1178117
16MSIDC, grade 3ER+/HER2+Luminal BPIK3CAc.3140A > T3.60p.H1047L28.2COSM7763
16MSIDC, grade 3ER+/HER2+Luminal BTP53c.743G > T2.11p.R248L43.1COSM654917
44MSIDC, grade 3ER+/HER2+Luminal BTP53c.587G > C1.03p.R196P35.8COSM4381417
9MSIDC, grade 3ER-/PR-/HER2-Triple negativeTP53c.637C > T0.91p.R213Ter46.9COSM1065417
67MSIDC, grade 2ER+/HER2−Luminal APIK3CAc.3145G > C0.39p.G1049R82.4COSM125973
7MSIDC, grade 2ER+/HER2−Luminal APIK3CAc.3140A > G0.14p.H1047R31COSM7753
30MSIDC, grade 2ER+/HER2−Luminal APIK3CAc.1633G > A0.05p.E545K56.2COSM7633
40MSIDC, grade 1ER+/HER2+Luminal BPIK3CAc.1624G > A0.20p.E542K25.5COSM7603
IDC: Invasive ductal carcinoma; HGVS: Human Genome Variation Society; SafeSEQ VAF: Plasma variant allele frequency; Tumor VAF: Tumor variant allele frequency; COSMIC: Catalog of somatic mutations in cancer; Chr: Chromosome.
Table 5. Clinicopathological variables influencing ctDNA mutation. detection in early breast cancer patients.
Table 5. Clinicopathological variables influencing ctDNA mutation. detection in early breast cancer patients.
CharacteristicsTotalctDNA-Positive b* p Value
nn (%)
Diagnostic age (years) 0.040
<3697 (77.8)
36–50104 (40)
>66102 (20)
Tumor size 0.033
<2 cm152 (13.3)
2 to 5 cm1310 (76.9)
>5 cm11 (10)
Tumor grade 0.041
I51 (20)
II166 (37.5)
III76 (85.7)
Unknown10 (0)
Axilar lymph node <0.001
Positive99 (100)
Negative204 (20)
Progesterone receptor 0.615
Positive125 (41.7)
Negative178 (47)
Estrogen receptor 0.571
Positive2512 (48)
Negative41 (25)
HER2 status 0.655
Positive11 (10)
Negative2812 (42.8)
IHC subtype 0.033
Luminal A tumor147 (50)
Luminal B tumor115 (45.4)
HER2-positive tumor10 (0)
Triple-negative31 (33.3)
BIRADS category 0.004
4141 (7.1)
5c1512 (80)
b ctDNA was defined as positive if tumor-derived mutations for TP53 and PIK3CA could be detected in blood. * p-value < 0.05 represents statistical significance.

Share and Cite

MDPI and ACS Style

Jimenez Rodriguez, B.; Diaz Córdoba, G.; Garrido Aranda, A.; Álvarez, M.; Vicioso, L.; Llácer Pérez, C.; Hernando, C.; Bermejo, B.; Julve Parreño, A.; Lluch, A.; et al. Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis. J. Clin. Med. 2019, 8, 1183. https://doi.org/10.3390/jcm8081183

AMA Style

Jimenez Rodriguez B, Diaz Córdoba G, Garrido Aranda A, Álvarez M, Vicioso L, Llácer Pérez C, Hernando C, Bermejo B, Julve Parreño A, Lluch A, et al. Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis. Journal of Clinical Medicine. 2019; 8(8):1183. https://doi.org/10.3390/jcm8081183

Chicago/Turabian Style

Jimenez Rodriguez, Begona, Gema Diaz Córdoba, Alicia Garrido Aranda, Martina Álvarez, Luis Vicioso, Casilda Llácer Pérez, Cristina Hernando, Begoña Bermejo, Ana Julve Parreño, Ana Lluch, and et al. 2019. "Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis" Journal of Clinical Medicine 8, no. 8: 1183. https://doi.org/10.3390/jcm8081183

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