Open Access

Detection of genetic mutations in patients with breast cancer from Saudi Arabia using Ion AmpliSeq™ Cancer Hotspot Panel v.2.0

  • Authors:
    • Safia A. Messaoudi
    • Nourah A. Al Sharhan
    • Bandar Alharthi
    • Saranya R. Babu
    • Abrar B. Alsaleh
    • Alanoud M. Alasiri
    • Mourad Assidi
    • Abdelbaset Buhmeida
    • Wassim Y. Almawi
  • View Affiliations

  • Published online on: February 14, 2022     https://doi.org/10.3892/br.2022.1509
  • Article Number: 26
  • Copyright: © Messaoudi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Next‑Generation Sequencing allows for quick and precise sequencing of multiple genes concurrently. Recently, this technology has been employed for the identification of novel gene mutations responsible for disease manifestation among breast cancer (BC) patients, the most common type of cancer amongst Arabian women, and the major cause of disease‑associated death in women worldwide. Genomic DNA was extracted from the peripheral blood of 32 Saudi Arabian BC patients with histologically confirmed invasive BC stages I‑III and IV, as well from 32 healthy Saudi Arabian women using a QIAamp® DNA Mini Kit. The isolated DNA was quantified using a Qubit™ dsDNA BR Assay Kit with a Qubit 2.0 Fluorometer. Ion semiconductor sequencing technology with an Ion S5 System and AmpliSeq™ Cancer Hotspot Panel v2 were utilized to analyze ~2,800 mutations described in the Catalogue of Somatic Mutations in Cancer from 50 oncogenes and tumor suppressor genes. Ion Reporter Software v.5.6 was used to evaluate the genomic alterations in all the samples after alignment to the hg19 human reference genome. The results showed that out of the 50 genes, 26 mutations, including 17 (65%) missense point mutations (single nucleotide variants), and 9 (35%) frameshift (insertion/deletion) mutations, were identified in 11 genes across the cohort in 61 samples (95%). Mutations were predominantly focused on two genes, PIK3CA and TP53, in the BC genomes of the sample set. PIK3CA mutation, c.1173A>G located in exon 9, was identified in 15 patients (46.9%). The TP53 mutations detected were a missense mutation (c.215C>G) in 26 patients (86.70%) and 1 frameshift mutation (c.215_216insG) in 1 patient (3.33%), located within exon 3 and 5, respectively. This study revealed specific mutation profiles for every BC patient, Thus, the results showed that Ion Torrent DNA Sequencing technology may be a possible diagnostic and prognostic method for developing personalized therapy based on the patient's individual BC genome.

Introduction

Breast cancer (BC) is the most common malignancy in females and the second leading cause of cancer-related death after lung cancer worldwide (1). BC has a significant impact on a women's health (2), and its incidence rates have been steadily increasing in recent years in Arab-speaking communities, with a significant number of cases being diagnosed in the first instance at advanced stages of the disease (3). BC incidence varies widely globally (4), and its incidence amongst Saudi Arabian women has progressively increased (3). BC is a complex and multifactorial disease, and genetic, hormonal and environmental factors contribute to its pathogenesis (5). The interplay between the genetic background and the environment in BC development has been proposed, but with mixed results on the importance of each (6-8). Female sex, age and ethnicity are the strongest risk factors associated with increased incidence (9). In addition, obesity (10), age at first delivery of a child (11), early menarche/late menopause (12), ionizing radiation exposure (13), breastfeeding (14), past/current estrogen treatment (15), breast tissue density (16), smoking and alcohol consumption (17) and steroid hormone receptors (18) are all known risk factors of BC.

BC aggregates in families, and an estimated 5-10% of BC cases are hereditary in origin caused by a myriad of susceptibility genes, transmitted from parent to child (19). These include rare variants in BRCA1, BRCA2, PALB2, ATM and CHEK2 genes, which reportedly confer a moderate-high lifetime risk of the disease. Other variants of >70 loci, which were identified through genome-wide association study, and large-scale replication studies (20), were also reported to confer heightened risk of disease, though to varying extents.

While hereditary BC is linked to a well-established set of susceptibility genes, the exact contribution of these genes to disease pathogenesis remains largely unknown (21). BRCA1 (chromosome 17) and BRCA2 (chromosome 13), described as regulators of DNA repair, transcription and cell cycle progression in response to DNA damage, were confirmed genetic loci associated with genetic susceptibility to BC (22,23). In this regard, it was shown that pathogenic BRCA1/BRCA2 mutations account for almost 30% of BC cases in high-risk families (24). Polymorphisms in other genes are also involved in BC, but to variable extents (25), suggesting that genetic variations may explain the heterogeneous nature of BC, and thus inter-individual differences with regard to tumor behavior (20).

AKT1, PIK3CA, PTEN and TP53 have been identified as recurrently mutated genes, and somatic mutations in these genes are found at a high frequency in BC patients. According to the Catalogue of Somatic Mutations in Cancer (COSMIC) database (26), high prevalence rates of PIK3CA (26.4%), TP53 (24.7%), PTEN (3.8%), and AKT1 (2.8%) were reported for BC.

In addition to the aforementioned mutated genes, two kinds of genomic instability have been often reported in BC and seldom in proliferative breast disease: Microsatellite instability (MSI) and loss of heterozygosity (27). Clinical testing for MSI involves immunohistochemistry and PCR testing for four proteins of the mismatch repair pathway: MSH2, MSH6, MLH1 and PMS2. MSI has been documented in BC, but at a lower frequency compared with other types of cancer. Previous studies reported that 0.9% of primary Triple Negative BC (28) and 1.53% of BC of all subtypes (29) have MSI.

In view of its heterogeneous etiology, the manifestations of BC vary widely among individual patients, with each patient having a unique profile, hence highlighting the potential value of precision medicine and individualized therapies for effective management (30-32). Next-Generation sequencing (NGS) was recently employed to improve identification of novel gene mutations responsible for disease manifestation amongst BC patients (33-36).

Ion Torrent™ technology allows the parallel sequencing of several genes, thus overcoming the problems inherent with conventional sequencing. In this study, Ion semiconductor sequencing technology with the Ion S5 System and AmpliSeq™ Cancer Hotspot Panel v2 was used to analyze ~2,800 COSMIC mutations from 50 oncogenes and tumor suppressor genes in a cohort of 32 BC cases from Saudi Arabia. Therefore, the present study aims to investigate the efficiency of AmpliSeq™ Cancer Hotspot Panel v2 on the detection of mutations in the genomic DNA extracted from the whole blood of the Saudi BC patients.

Patients and methods

Study subjects

Ethical approval for the present study was obtained from the Ethics Committee of King Fahad Medical City (KFMC; Riyadh, Saudi Arabia; IRB approval no. FWA00018774), and the study was performed in accordance with the guidelines described in the Helsinki Declaration (37). A total of 32 Saudi Arabian patients with BC (mean age 48.5±8.2 years; median age 45 years [age range, 31-85 years; interquartile range (IQR) 42.5-55.5)], and with histologically confirmed invasive BC, were recruited from Medical Oncology Department, KFMC. None of the patients had a history of other cancer types and were not subjected to chemo-, radio or hormone therapy. In addition, 32 healthy Saudi women with no familial history of any cancer types served as the controls (mean age 49.1±11.0 years; median age, 47.5 years (age range, 35-71 years; IQR, 42.5-55), were recruited into this retrospective case-controlled study from the blood bank (Table I). Demographic and clinical data of BC patients and control women were collected from the hospital records. All participants provided signed informed consent prior to inclusion in this study.

Table I

Demographics and clinical characteristics of the cohorts.

Table I

Demographics and clinical characteristics of the cohorts.

CharacteristicsHealthy controls, n=32Patients BC, n=32P-value
Mean age, yearsc49.09±11.0248.80±8.280.904d
BMI, kg/m2b27.60±5.6632.89±7.960.004b,d
Oral contraceptives use  0.5e
     Yes1920 
     No1312 
Breastfeeding  0.011a,e
     Yes199 
     No1323 
Tumor size  -
     <2 cm-4 
     ≥2 cm-28 
Tumor stage  -
     I-3 
     II-11 
     III-13 
     IV-5 
Histological classification  -
     IDC-29 
     ILC-2 
     DCIS-1 
Tumor location   
     Left-28 
     Right-4 
ER status  -
     ER+-23 
     ER--9 
PR status  -
     PR+-21 
     PR--11 
HER2 status  -
     HER2+-13 
     HER2--19 

[i] aP<0.05,

[ii] bP<0.01.

[iii] cMean ± standard deviation.

[iv] dUnpaired Student's t-test.

[v] eχ2 test. HER2, Human epidermal growth factor receptor; ER, Estrogen receptor; PR, Progesterone receptor; DCIS, Ductal carcinoma in situ; ILC, invasive lobular carcinoma; IDC, invasive ductal carcinoma.

DNA extraction and quantification

Peripheral blood (2 ml) was collected in EDTA tubes from each participant. Genomic DNA was extracted using a QIAamp® DNA Mini Kit according to manufacturer's instructions (Qiagen GmbH) then quantified using a Qubit™ dsDNA BR Assay Kit on a Qubit 2.0 Fluorometer (Thermo Fisher Scientific, Inc.), following the manufacturer's instructions.

Library preparation

Manual library preparations were performed using an Ion AmpliSeq™ Cancer Hotspot Panel v.2, Ion Xpress barcoded adapters, and an Ion AmpliSeq Library Kit 2.0 (Thermo Fisher Scientific, Inc.). The panel consisted of 207 amplicons, covering ~20,000 bases surveying hotspot regions, including up to 2,855 COSMIC mutations in 50 oncogenes and tumor suppressor genes, all with known cancer associations. The genes included in this panel were: ABL1, AKT1, ALK, APC, ATM, BRAF, CDH1, CDKN2A, CSF1R, CTNNB1, EGFR, ERBB2, ERBB4, EZH2, FBXW7, FGFR1, FGFR2, FGFR3, FLT3, GNA11, GNAS, GNAQ, HNF1A, HRAS, IDH1, IDH2, JAK2, JAK3, KDR, KIT, KRAS, MET, MLH1, MPL, NOTCH1, NPM1, NRAS, PDGFRA, PIK3CA, PTEN, PTPN11, RB1, RET, SMAD4, SMARCB1, SMO, SRC, STK11, TP53 and VHL.

Multiplex PCR was performed using 10 ng genomic DNA with a premixed primer pool and Ion AmpliSeq HiFi master mix (Ion AmpliSeq Library Kit 2.0). The amplicons were treated with 2 µl FuPa reagent to partially digest the primer sequences and phosphorylate the amplicons. Amplicons were ligated to adapters with the diluted barcodes of Ion Xpress Barcode Adapters kit (Thermo Fisher Scientific, Inc.). The adapter-ligated amplicons (library) were purified using the Agencourt AMPure XP reagent (Beckman Coulter, Inc.). Quantification of the final libraries was performed using an Ion Library TaqMan™ Quantitation Kit (Thermo Fisher Scientific, Inc.) on the Applied Biosystems® 7500 Real-Time PCR System, following the manufacturer's protocols.

Template preparation and chip loading

After library dilution to ~100 pM, the clonal amplification of barcoded DNA library (AmpliSeq libraries) onto ion spheres was performed on an Ion Chef™ Instrument using Ion 520™ & Ion 530™ Kit-Chef, according to the manufacturer's instructions (Thermo Fisher Scientific, Inc.). Template-positive spheres from barcoded libraries were multiplexed and loaded onto Ion 530™ Chips following the manufacturer's protocol, and sequencing was run on the Ion Gene Studio S5 system (Thermo Fisher Scientific, Inc.).

Statistical analysis

Samples were evaluated for genomic alterations, including single nucleotide variants (SNVs), and insertions and deletions, using Ion Reporter Software v.5.6 (Thermo Fisher Scientific, Inc.), after alignment to the hg19 human reference genome. Of note, higher quality requirements for variant analysis and selection, high-quality SNVs and insertion/deletion variants were strictly followed in this study and were defined as: i) FILTER=PASS, ii) QUAL ≥100, iii) depth coverage ≥20X, and iv) variant fraction ≥20%. The sequencing data analysis using such approach yielded high-quality variants that did not necessary require additional confirmatory testing (such as through Sanger sequencing validation) as recommended by Arteche-López group (38). Furthermore, the ‘bam’ files of each clinically actionable variant were carefully reviewed in order to provide additional confidence to the accuracy and reliability of the NGS calls.

Qualitative and quantitative data were analyzed using SPSS v.21 (IBM Corp.). Qualitative data are presented as the frequency and percentage of total, and these data were compared using a χ2 goodness-of-fit test, while continuous variables are presented as the mean ± SD, and were compared using an unpaired two-tailed Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Study subjects

The clinical and demographic characteristics of the study subjects are summarized in Table I. Patients were clinically characterized in terms of tumor size, location, stage, histological classification and presence/absence of tumor markers, including estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). In addition, age, body mass index (BMI), use of oral contraception and breastfeeding were compared between the two groups. No statistically significant differences were noted between patients and controls regarding mean age (P=0.90), and oral contraceptive use (P=0.50). However, a significant difference was noted between BC patients and healthy subjects for mean BMI, which was higher in patients compared with the control group (P=0.004), and breastfeeding (P=0.011), which was higher in the healthy control group.

Invasive ductal carcinoma of no specific type (n=29, 90.6%) was the predominant histological type of primary tumor, followed by invasive lobular carcinoma (n=2, 6.3%), and ductal carcinoma in situ (n=1, 3.1%). The majority of cases were stage III (n=13, 40.6%) and stage II (n=11, 34.4%), whereas stage I (n=3, 9.3%) and stage IV (n=5, 15.6%) were less common. In addition, 4 patients (12.5%) had ER+, PR+, HER2- tumors; 21 patients (65.6%) had ER+, and/or PR+, HER2+ tumors, 4 patients (12.5%) had ER-, PR-, HER2+ tumors, and 3 patients (9.4%) had triple-negative (ER-, PR-, HER2-) tumors.

Somatic mutations

There were only three unclassified samples, two from patients (sample #2 and sample #13) and 1 from a control individual (sample #4) that did not possess any identified mutations (Fig. 1). The observed mutations were detected with varied frequencies across BC patients (Table II) and healthy controls (Table III); 26 mutations, including 17 (65%) missense point mutations (SNV), and 9 (35%) frameshift (insertion/deletion) mutations in 11 genes (out of 50); TP53, PIK3CA, KDR, KIT, ATM, HRAS, ERBB2, FGFR3, GNAQ, APC and JAK3 (Fig. 2) across the cohort in the 61 samples (95%).

Table II

Mutational status of breast cancer patient samples analyzed using the Ion AmpliSeq™ Cancer Hotspot Panel v2.

Table II

Mutational status of breast cancer patient samples analyzed using the Ion AmpliSeq™ Cancer Hotspot Panel v2.

No.Tumor histologic typeStageHormone receptor statusGenesMutations detectedEffectAmino acid change
1IDCIII HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      KDRc.1416A>TMissensep.Gln472His
      GNAQc.625C>AMissensep.Gln209Lys
      TP53c.215C>GMissensep.Pro72Arg
3IDCIV HER2-ER-PR-PIK3CAc.1173A>GMissensep.Ile391Met
      HRASc.84_85insT,Frameshiftp.Val29fs,
      HRASc.80_81insCFrameshiftp.Val29fs
      TP53c.215C>GMissensep.Pro72Arg
      JAK3c.394C>AMissensep.Pro132Thr
4IDCIII HER2-ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      KDRc.1416A>TMissensep.Gln472His
      TP53c.215C>GMissensep.Pro72Arg
5IDCII HER2+ER+PR+KITc.1621A>CMissensep.Met541Leu
      TP53 c.21+I9:K95C>GMissensep.Pro72Arg
6IDCIII HER2+ER+PR+KDRc.1416A>TMissensep.Gln472His
      TP53c.215C>GMissensep.Pro72Arg
7IDCIII HER2+ER+PR-APCc.3949G>CMissensep.Glu1317Gln
      ATMc.2572T>CMissensep.Phe858Leu
      TP53c.215C>GMissensep.Pro72Arg
8IDCII HER2+ER+PR+KDRc.1416A>TMissensep.Gln472His
      TP53c.21GMissensep.Pro72Arg
9IDCII HER2-ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      TP53c.215C>GMissensep.Pro72Arg
10IDCII HER2+ER+PR+TP53c.215C>GMissensep.Pro72Arg
11IDCIV HER2-ER-PR-TP53c.215C>GMissensep.Pro72Arg
12DCISIII HER2+ER-PR-PIK3CAc.1173A>GMissensep.Ile391Met
      KDRc.1416A>TMissensep.Gln472His
      TP53c.215C>GMissensep.Pro72Arg
14IDCI HER2+ER-PR-TP53c.215C>GMissensep.Pro72Arg
15IDCIII HER2+ER+PR+ATMc.2525C>GMissensep.Thr842Ser
      TP53c.215C>GMissensep.Pro72Arg
16IDCIV HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
     PR+APCc.3920T>AMissensep.Ile1307Lys
      APC c.3920_3921delTAFrameshiftp.Ile1307fs
      APCc.3920delTFrameshiftp.Ile1307fs
      TP53c.215C>GMissensep.Pro72Arg
17IDCIV HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      KITc.1621A>CMissensep.Met541Leu
      TP53c.215_216insGFrameshiftp.Val73fs
      TP53c.215C>GMissensep.Pro72Arg
18IDCII HER2+ER+PR+KDRc.1416A>TMissensep.Gln472His
      TP53c.215C>GMissensep.Pro72Arg
19IDCIII HER2-ER-PR-PIK3CAc.1173A>GMissensep.Ile391Met
      TP53c.215C>GMissensep.Pro72Arg
20IDCII HER2+ER+PR+TP53c.215C>GMissensep.Pro72Arg
21ILCI HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      FGFR3c.2389G>CMissensep.Ala797Pro
      ATMc.7313C>TMissensep.Thr2438Ile
      TP53c.215C>GMissensep.Pro72Arg
22IDCIII HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      TP53c.215C>GMissensep.Pro72Arg
23IDCIII HER2+ER+PR+TP53c.215C>GMissensep.Pro72Arg
24IDCIII HER2+ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      HRASc.84_85insTFrameshiftp.Val29fs,
      HRASc.80_81insCFrameshiftp.Val29fs
      TP53c.215C>GMissensep.Pro72Arg
25IDCIII HER2-ER-PR-KITc.1621A>CMissensep.Met541Leu
      TP53c.215C>GMissensep.Pro72Arg
26ILCIII HER2+ER+PR+KITc.1621A>CMissensep.Met541Leu
      TP53c.215C>GMissensep.Pro72Arg
27IDCII HER2+ER+PR+PIK3CAc.233A>GMissensep.Glu78Gly
      PIK3CAc.1173A>GMissensep.Ile391Met
28IDCIV HER2+ER+PR+PIK3CAc.233A>GMissensep.Glu78Gly
      ATMc.1810C>TMissensep.Pro604Ser
      TP53c.215C>GMissensep.Pro72Arg
29IDCII HER2-ER+PR+PIK3CAc.1173A>GMissensep.Ile391Met
      KDRc.1416A>TMissensep.Gln472His
30ILCII HER2-ER+PR+KDRc.1416A>TMissensep.Gln472His
      ATMc.3905G>TMissensep.Gly1302Val
31IDCII HER2+ER-PR-TP53c.215C>GMissensep.Pro72Arg
32IDCII HER2+ER-PR-PIK3CAc.1173A>GMissensep.Ile391Met
      KDRc.1416A>TMissensep.Gln472His

[i] Del, deletion; ins, insertion; fs, frameshift; HER2, Human epidermal growth factor receptor; ER, Estrogen receptor; PR, Progesterone receptor; DCIS, Ductal carcinoma in situ; ILC, invasive lobular carcinoma; IDC, invasive ductal carcinoma.

Table III

Mutational status of healthy control samples analyzed using the Ion AmpliSeq™ Cancer Hotspot Panel v2.

Table III

Mutational status of healthy control samples analyzed using the Ion AmpliSeq™ Cancer Hotspot Panel v2.

No.GenesMutations detectedEffectAmino acid change
1KDRc.1416A>TMissensep.Gln472His
 HRASc.80_81insCFrameshiftp.Val29fs
 HRASc.80_81insCFrameshiftp.Val29fs
 ERBB2c.2380G>TMissensep.Val794Leu
2KDRc.1416A>TMissensep.Gln472His
 TP53c.215C>GMissensep.Pro72Arg
3TP53c.215C>GMissensep.Pro72Arg
   c.209_215delCTCCCCCinsTCCCCCGFrameshift p.Ala70_Pro72delinsValProArg
5TP53c.215C>GMissensep.Pro72Arg
6PIK3CAc.1173A>GMissensep.Ile391Met
 APCc.3920T>AMissensep.Ile1307Lys
 TP53c.215C>GMissensep.Pro72Arg
7TP53c.215C>GMissensep.Pro72Arg
8PIK3CAc.233A>GMissensep.Glu78Gly
 FGFR3c.2389G>CMissensep.Ala797Pro
 KDRc.1416A>TMissensep.Gln472His
9TP53c.215C>GMissensep.Pro72Arg
10TP53c.215C>GMissensep.Pro72Arg
11APCc.3920T>AMissensep.Ile1307Lys
 APCc.3920delTFrameshiftp.Ile1307fs
12ATMc.1811delCFrameshiftp.Pro604fs
 ATMc.1810C>TMissensep.Pro604Ser
 TP53c.215C>GMissensep.Pro72Arg
 PIK3CAc.1173A>GMissensep.Ile391Met
 KITc.1621A>CMissensep.Met541Leu
13KDRc.1416A>TMissensep.Gln472His
 APCc.3920T>AMissensep.Ile1307Lys
 APC c.3920_3921delTAFrameshiftp.Ile1307fs
14PIK3CAc.233A>GMissensep.Glu78Gly
 TP53c.215C>GMissensep.Pro72Arg
15PIK3CAc.1173A>GMissensep.Ile391Met
 GNAQc.625C>AMissensep.Gln209Lys
 TP53c.215C>GMissensep.Pro72Arg
 TP53 c.209_215delCTCCCCCinsTCCCCCGFrameshift p.Ala70_Pro72delinsValProArg
16KDRc.1416A>TMissensep.Gln472His
 TP53c.215C>GMissensep.Pro72Arg
17PIK3CAc.1173A>GMissensep.Ile391Met
18PIK3CAc.1173A>GMissensep.Ile391Met
 KDRc.1416A>TMissensep.Gln472His
 ATMc.2572T>CMissensep.Phe858Leu
 TP53c.215C>GMissensep.Pro72Arg
19KDRc.1416A>TMissensep.Gln472His
 JAK3c.394C>AMissensep.Pro132Thr
20TP53c.215C>GMissensep.Pro72Arg
21ATMc.2572T>CMissensep.Phe858Leu
 TP53c.215C>GMissensep.Pro72Arg
22PIK3CAc.1173A>GMissensep.Ile391Met
 KDRc.1416A>TMissensep.Gln472His
23KITc.1621A>CMissensep.Met541Leu
 KDRc.1416A>TMissensep.Gln472His
 TP53c.215C>GMissensep.Pro72Arg
24PIK3CAc.1173A>GMissensep.Ile391Met
 APCc.3920T>AMissensep.Ile1307Lys
 TP53c.215C>GMissensep.Pro72Arg
25PIK3CAc.1173A>GMissensep.Ile391Met
 TP53c.215C>GMissensep.Pro72Arg
26KITc.1621A>CMissensep.Met541Leu
 TP53c.215C>GMissensep.Pro72Arg
27TP53c.215C>GMissensep.Pro72Arg
28TP53c.215C>GMissensep.Pro72Arg
29PIK3CAc.1173A>GMissensep.Ile391Met
 APCc.3920delTFrameshiftp.Ile1307fs
 TP53c.215C>GMissensep.Pro72Arg
30TP53c.215C>GMissensep.Pro72Arg
31TP53c.215C>GMissensep.Pro72Arg
32TP53c.215C>GMissensep.Pro72Arg

[i] Del, deletion; ins, insertion; fs, frameshift.

Amongst BC patients, 27 patients (84.8%) were positive for >1 somatic mutation, compared with 17 control subjects (53.1%) (Fig. 1). The most frequently observed concurrent mutations were some combination of c.215C>G (TP53), c.1173A>G (PIK3CA), c.1416A>T (KDR) and c.1621A>C (KIT) in patients (Fig. 3A), and c.215C>G (TP53), c.1173A>G (PIK3CA) and c.1416A>T (KDR) in healthy controls (Fig. 3B).

The site of the most frequent mutations within TP53 and PIK3CA differed between samples. The most common TP53 mutation detected was a missense mutation (c.215C>G) in 26 patients (86.70%), with only 1 frameshift mutation (c.215_216insG) identified in 1 patient (3.33%). The most common PIK3CA mutation detected was c.1173A>G, located in exon 9, and was identified in 15 (50%) patients.

Table IV summarizes the distribution of the most frequent missense mutations in TP53 and PIK3CA (c.215C>G and c.1173A>G, respectively), between patients and healthy control samples. A significant difference (P=0.020) was observed in the frequency of c.215C>G (Pro72Arg), which was higher in patients (0.87) than in controls (0.61). Similarly, a significant difference was observed in the frequency of c.1173A>G (Pro72Arg) (P=0.041) that was higher in patients (0.53) compared with the healthy control participants (0.25).

Table IV

Frequency distributions of the most frequent mutations TP53 and PIK3CA between the breast cancer patients and the healthy controls.

Table IV

Frequency distributions of the most frequent mutations TP53 and PIK3CA between the breast cancer patients and the healthy controls.

GeneMutationAmino acid changeChromosomeExonNCBI 1000 Genomes Browser IDVariant frequency in the patientsVariant frequency in the healthy controls P-valueb
TP53c.215C>Gp.Pro72Arg173rs10425220.870.610.020a
PIK3CAc.1173A>Gp.Ile391Met39rs22304610.530.250.041a

[i] aP<0.05.

[ii] bχ2 test.

A unique mutational status was identified for every BC patient except for patients #8, #10, #18, and #20 (Table II).

Discussion

BC is the most common type of cancer amongst Arabian women, and the major cause of disease-associated mortality in women worldwide (39). In the Middle East, Arabian women face a significantly higher risk of mortality, as the cancer is often diagnosed at a later stage in disease progression (40-45).

Current efforts to manage BC include on improving prevention, diagnosis and an increased armamentarium of effective treatment choices for patients with BC (46). Due to the heterogeneity of BC and the interactions between genetic and environmental factors, each patient's tumor possibly exhibits an unique gene mutation profile (47). By profiling an individual's cancer genome, it becomes possible to differentiate the oncogenic mechanisms that regulate cancer and, therefore, the genetic biomarkers that may be specifically associated with the disease state (5).

In this study, massively parallel sequencing was performed to identify frequent mutations in 32 BC Saudi Arabian patients and 32 healthy controls using Ion Torrent sequencing technology.

In this study, 32 BC patients were clinically characterized in terms of tumor size, location, stage, histological classification, and the presence or absence of tumor markers such as ER, PR, and HER2. In addition, four common risk factors, including age, BMI, oral contraceptives use and breastfeeding, were also evaluated and compared between healthy controls and BC patients. The statistical analysis showed no significant difference in the mean age between BC patients and healthy controls as the two study groups were already age matched.

BMI was significantly higher in BC patients compared with the healthy control group. This finding is in agreement with several reports, highlighting BMI as one of the most important risk factors for the BC (48,49). Chronic low-levels of inflammation that are usually observed in obese people can lead to BC through the increased likelihood of DNA damage (50). Additionally, fat cells can produce an excess amount of estrogen and adipokines, which can stimulate cell growth, as observed in BC (49,51). Similarly, a significant difference was observed for breastfeeding, which was higher in control group compared to the BC patients group, supporting the previously published literature linking breastfeeding to reduced BC risk (52).

PIK3CA and TP53 are the most frequently mutated genes, and harbor most of the mutations in this cohort. The p53 tumor suppressor gene, located on chromosome 17p13, is 20 kb long, encompassing 11 exons encoding a 53 kDa phosphoprotein (53). In the present study the detected mutations in this gene were 1 missense mutations (c.215C>G) in 26 patients (86.70%) and 1 frameshift mutation (c.215_216insG) in 1 patient (3.33%). These results are in agreement with the current release of the International Agency for Research on Cancer TP53 database (http://www-p53.iarc.fr/), which also shows that all TP53 mutations were missense mutations in the coding region (54). The detected mutations were located within exons 3 and 5, encoding the Proline-rich domain, which plays a role in p53-mediated apoptosis and in the DNA-binding and oligomerization domain. These regions are required for interactions with FBX042, HIPK1 and AXIN1, the DNA major groove, and a domain containing a nuclear export signal (46,55,56). Dysfunction of p53 can cause defective DNA replication and malignant transformation, common in dysplasia's of BC (53). The p53 gene exhibits several genetic alterations in patients with BC (57). This highlights the need to administer effective treatments such as cell-cycle inhibitors in the form of target therapies and combinatorial target therapies against the wide range of TP53 mutations.

In the current study, the high TP53 mutation rate in the cohort could be explained by the high number of ER+ cases, given that 67% of the observed TP53 mutations occurred in the ER+ tumors. ER status is tightly associated with the molecular subtypes, and a significantly higher TP53 mutation rate was demonstrated in the basal-like subtype, mainly ER-, and HER2-enriched (both ER- and ER+) tumors compared with the primarily ER+ luminal type (56,58). A recent study by Bai et al (59) conducted in 2021 using NGS to detect TP53 mutations in the cell free DNA in Chinese metastatic BC (MBC) patients indicated that TP53 mutations could be used as a prognostic marker for worse outcomes in MBC and for the response of adjuvant endocrine therapy. TP53-mutated MBC patients had a significantly worse outcome than TP53 wild-type patients, especially those in the HR+/HER2- and triple-negative BC (TNBC) cohorts. TP53 mutations were also associated with endocrine resistance (59).

In the present study TP53 mutations were not associated with HER2- tumors, which is comparable to the previously published research. TP53 mutation status was an independent predictive factor of survival especially in HR+/HER2- and TNBC cohorts, but not in the HER2+ cohort (59,60).

In the present study, the somatic TP53 mutation c.215C>G p.(Pro72Arg) was the most frequently detected mutation in all BC patients, particularly those with Stage III BC. A previous study showed that TP53 mutation NM_000546.5:c.824G>A p.(Cys275Tyr) was the most common mutation detected in 82 patients with Stage I-III BC who underwent NGS using tissue and blood samples, and they showed that TP53 pathogenic somatic mutations were associated with an 8-fold risk of recurrence in the univariate Cox regression analysis (61). The same study showed that the coexistence of TP53 and PIK3CA mutations was a common finding in BC patients (61). PIK3CA mutation (c.1173A>G) and p.(Ile391Met) located in exon 9, was identified in 15 (50%) patients. This exon encodes the helical domain, and mutations, represented by single amino acid substitutions, in this domain are associated with increased lipid kinase activity, and thus induce oncogenic transformation (62,63).

The PI3K pathway has been identified as a major player in cancer development and progression (62-66). PI3K is a heterodimeric enzyme composed of a p110α catalytic subunit encoded by the PIK3CA gene and a p85 regulatory subunit encoded by the PIK3R1 gene (67). In the present study, 47% of the BC patients were carriers of PIK3CA mutations. This result corroborates the findings of the previous work, reporting that PIK3CA mutations occur in 20-40% of BC and ≈30% of tumors of the prostate, cervix and endometrium (68,69). Several studies have suggested that PIK3CA mutations are more frequent in ER+ and HER2+ BC cases (68,69). Accordingly, the low mutation rate can be explained by a bias in the subtype distribution of the cohort. In the present study, 2 out of 5 (40%) of the PIK3CA mutations occurred in ER+ primary tumors. The small size of the cohort may have influenced this distribution. Martínez-Sáez et al (70) showed that 28% of PIK3CA mutations identified in circulating tumor DNA (ctDNA) in 48 patients with advanced HR+/HER2- BC were not part of the therascreen® PIK3CA test (QIAGEN GmbH), a FDA approved kit used to select patients who possessed PIK3CA mutations in tumor tissue specimens and/or in circulating tumor DNA (ctDNA) isolated from plasma specimens (71). Therascreen PIK3CA detects 11 PIK3CA hotspot mutations, mostly found in exons 9 and 20(71).

It is important to remember that only a subset of genes was examined in the present study, and that for a deeper understanding of the mutational profiles of BC patients, considerably more extensive sequencing is required. The hotspot panel was not specifically developed for BC, and it includes areas and genes that are more commonly mutated in other cancer types (72).

Recently, Hempel et al (73) utilized a wide NGS panel to investigate 41 MBC samples and found that PIK3CA mutations appear in 34% of the patients. A recent study reported that 22% of the total population and 28% of patients with HR+ BC have a PIK3CA mutation (74). Further research using a large NGS panel targeting 1,021 genes in 193 MBC samples by Tang et al (75) detected 36 (18.7%) mutations in the kinase domain and 26 (13.5%) substitutions in the helical domain, with 10 (5.2%) additional alterations distributed in the remaining PIK3CA sequence.

PIK3CA mutations in the ctDNA of patients with BC have also been reported (76). Board et al (77) was able to detect PIK3CA mutations in the vast majority (80%) of ctDNA samples from PIK3CA-mutated MBC, but not in early BC (78).

In conclusion, the results of this investigation showed that Ion Torrent DNA Sequencing technology using AmpliSeq Cancer Hotspot Panel v2 was found to be a suitable method to perform molecular characterization of the genotype of BC patients and healthy controls using peripheral blood samples. The present study revealed specific mutational profiles for every BC patient; for this reason, it may be a possible to improve diagnosis and prognosis, and recommend personalized treatments for each BC patient based on the mutational profile. The primary benefits of NGS are that it allows for the identification of multiple mutations at the same time, eliminating the need for sequential individual tests. Therefore, this technique could be routinely implemented in cancer diagnosis, as it can precisely identify fusions, SNPs, copy number variants, and insertion/deletions. Additionally, confirmation of NGS variants must be carefully investigated and validated through Sanger sequencing to avoid false positive outcomes. The results of the present study add to the existing body of knowledge and practice in the diagnosis and treatment of BC patients.

Acknowledgements

Not applicable.

Funding

Funding: This work was supported by Naif Arab University for Security Sciences, Kingdom of Saudi Arabia.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

SAM conceived the study, curated the data, analyzed the data, performed the experiments and wrote and reviewed the manuscript. NAAS, BA and SRB contributed to the design of the study, performed the experiments and drafted the manuscript. ABA and AMA performed the experiments and contributed to data analysis and interpretation. MA, AB and WYA conceived the study, performed data interpretation, and wrote and critically reviewed the manuscript. SAM, MA and AB confirm the authenticity of all the raw data. All the authors have read and approved the final manuscript.

Ethics approval and consent to participate

Ethical approval for this study was obtained from the Ethics Committee of King Fahad Medical City (KFMC; Riyadh, Saudi Arabia; IRB approval no. FWA00018774), and the study was performed in accordance with the guidelines described in the Helsinki Declaration. All participants provided signed informed consent prior to inclusion in this study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Stewart BW and Wild CP (eds): World Cancer Report 2014. International Agency for Research on Cancer, WHO, 2014.

2 

Testa U, Castelli G and Pelosi E: Breast cancer: A molecularly heterogenous disease needing subtype-specific treatments. Med Sci (Basel). 8(18)2020.PubMed/NCBI View Article : Google Scholar

3 

Almutlaq BA, Almuazzi RF, Almuhayfir AA, Alfouzan AM, Alshammari BT, AlAnzi HS and Ahmed HG: Breast cancer in Saudi Arabia and its possible risk factors. J Cancer Policy. 12:83–89. 2017.

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018.PubMed/NCBI View Article : Google Scholar

5 

Zhang J, Späth SS, Marjani SL, Zhang W and Pan X: Characterization of cancer genomic heterogeneity by next-generation sequencing advances precision medicine in cancer treatment. Precis Clin Med. 1:29–48. 2018.PubMed/NCBI View Article : Google Scholar

6 

Fasching PA, Ekici AB, Adamietz BR, Wachter DL, Hein A, Bayer CM, Häberle L, Loehberg CR, Jud SM, Heusinger K, et al: Breast cancer risk-genes, environment and clinics. Geburtshilfe Frauenheilkd. 71:1056–1066. 2011.PubMed/NCBI View Article : Google Scholar

7 

Rudolph A, Chang-Claude J and Schmidt MK: Gene-environment interaction and risk of breast cancer. Br J Cancer. 114:125–133. 2016.PubMed/NCBI View Article : Google Scholar

8 

Hiatt RA, Haslam SZ and Osuch J: Breast Cancer and the Environment Research Centers. The breast cancer and the environment research centers: Transdisciplinary research on the role of the environment in breast cancer etiology. Environ Health Perspect. 117:1814–1822. 2009.PubMed/NCBI View Article : Google Scholar

9 

Winters S, Martin C, Murphy D and Shokar NK: Breast cancer epidemiology, prevention, and screening. Prog Mol Biol Transl Sci. 151:1–32. 2017.PubMed/NCBI View Article : Google Scholar

10 

Parkin DM and Boyd L: 8. Cancers attributable to overweight and obesity in the UK in 2010. Br J Cancer. 105 (Suppl 2):S34–S37. 2011.PubMed/NCBI View Article : Google Scholar

11 

Nelson HD, Zakher B, Cantor A, Fu R, Griffin J, O'Meara ES, Buist DS, Kerlikowske K, van Ravesteyn NT, Trentham-Dietz A, et al: Risk factors for breast cancer for women aged 40 to 49 years: A systematic review and meta-analysis. Ann Intern Med. 156:635–648. 2012.PubMed/NCBI View Article : Google Scholar

12 

Collaborative Group on Hormonal Factors in Breast Cancer. Menarche, menopause, and breast cancer risk: Individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol. 13:1141–1151. 2012.PubMed/NCBI View Article : Google Scholar

13 

Fenga C: Occupational exposure and risk of breast cancer. Biomed Rep. 4:282–292. 2016.PubMed/NCBI View Article : Google Scholar

14 

Elkum N, Al-Tweigeri T, Ajarim D, Al-Zahrani A, Amer SM and Aboussekhra A: Obesity is a significant risk factor for breast cancer in Arab women. BMC Cancer. 14(788)2014.PubMed/NCBI View Article : Google Scholar

15 

Gnant M, Mlineritsch B, Schippinger W, Luschin-Ebengreuth G, Pöstlberger S, Menzel C, Jakesz R, Seifert M, Hubalek M, Bjelic-Radisic V, et al: Endocrine therapy plus zoledronic acid in premenopausal breast cancer. N Engl J Med. 360:679–691. 2009.PubMed/NCBI View Article : Google Scholar

16 

McCormack VA and dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: A meta-analysis. Cancer Epidemiol Biomarkers Prev. 15:1159–1169. 2006.PubMed/NCBI View Article : Google Scholar

17 

Druesne-Pecollo N, Touvier M, Barrandon E, Chan DS, Norat T, Zelek L, Hercberg S and Latino-Martel P: Excess body weight and second primary cancer risk after breast cancer: A systematic review and meta-analysis of prospective studies. Breast Cancer Res Treat. 135:647–654. 2012.PubMed/NCBI View Article : Google Scholar

18 

Abderrahman B and Jordan VC: Rethinking extended adjuvant antiestrogen therapy to increase survivorship in breast cancer. JAMA Oncol. 4:15–16. 2018.PubMed/NCBI View Article : Google Scholar

19 

Apostolou P and Fostira F: Hereditary breast cancer: The era of new susceptibility genes. Biomed Res Int. 2013(747318)2013.PubMed/NCBI View Article : Google Scholar

20 

Michailidou K, Beesley J, Lindstrom S, Canisius S, Dennis J, Lush MJ, Maranian MJ, Bolla MK, Wang Q, Shah M, et al: Genome-wide association analysis of more than 120,000 individuals identifies 15 new susceptibility loci for breast cancer. Nat Genet. 47:373–380. 2015.PubMed/NCBI View Article : Google Scholar

21 

Luen S, Virassamy B, Savas P, Salgado R and Loi S: The genomic landscape of breast cancer and its interaction with host immunity. Breast. 29:241–250. 2016.PubMed/NCBI View Article : Google Scholar

22 

Mundhofir FE, Wulandari CE, Prajoko YW and Winarni TI: BRCA1 gene mutation screening for the hereditary breast and/or ovarian cancer syndrome in breast cancer cases: A first high resolution DNA melting analysis in Indonesia. Asian Pac J Cancer Prev. 17:1539–1546. 2016.PubMed/NCBI View Article : Google Scholar

23 

Yoshida K and Miki Y: Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage. Cancer Sci. 95:866–871. 2004.PubMed/NCBI View Article : Google Scholar

24 

Couch FJ, Nathanson KL and Offit K: Two decades after BRCA: Setting paradigms in personalized cancer care and prevention. Science. 343:1466–1470. 2014.PubMed/NCBI View Article : Google Scholar

25 

Byler S, Goldgar S, Heerboth S, Leary M, Housman G, Moulton K and Sarkar S: Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res. 34:1071–1077. 2014.PubMed/NCBI

26 

Forbes SA, Beare D, Boutselakis H, Bamford S, Bindal N, Tate J, Cole CG, Ward S, Dawson E, Ponting L, et al: COSMIC: Somatic cancer genetics at high-resolution. Nucleic Acids Res. 45:D777–D783. 2017.PubMed/NCBI View Article : Google Scholar

27 

Marcus L, Lemery SJ, Keegan P and Pazdur R: FDA approval summary: Pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 25:3753–3758. 2019.PubMed/NCBI View Article : Google Scholar

28 

Kurata K, Kubo M, Mori H, Kawaji H, Motoyama Y, Kuroki L, Yamada M, Kaneshiro K, Kai M and Nakamura M: Abstract P1-06-11: Microsatellite instability in triple negative breast cancers. Cancer Res. 79 (Suppl 4)(P1-06-11)2019.

29 

Bonneville R, Krook MA, Kautto EA, Miya J, Wing MR, Chen HZ, Reeser JW, Yu L and Roychowdhury S: Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol: Oct 3, 2017 (Epub ahead of print). doi: 10.1200/PO.17.00073.

30 

Le Du F, Eckhardt BL, Lim B, Litton JK, Moulder S, Meric-Bernstam F, Gonzalez-Angulo AM and Ueno NT: Is the future of personalized therapy in triple-negative breast cancer based on molecular subtype? Oncotarget. 6:12890–12908. 2015.PubMed/NCBI View Article : Google Scholar

31 

Liu YR, Jiang YZ, Xu XE, Yu KD, Jin X, Hu X, Zuo WJ, Hao S, Wu J, Liu GY, et al: Comprehensive transcriptome analysis identifies novel molecular subtypes and subtype-specific RNAs of triple-negative breast cancer. Breast Cancer Res. 18(33)2016.PubMed/NCBI View Article : Google Scholar

32 

Sachdev JC, Sandoval AC and Jahanzeb M: Update on precision medicine in breast cancer. Cancer Treat Res. 178:45–80. 2019.PubMed/NCBI View Article : Google Scholar

33 

Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S and Wakai T: Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci. 110:6–15. 2019.PubMed/NCBI View Article : Google Scholar

34 

Smith NG, Gyanchandani R, Shah OS, Gurda GT, Lucas PC, Hartmaier RJ, Brufsky AM, Puhalla S, Bahreini A, Kota K, et al: Targeted mutation detection in breast cancer using MammaSeq™. Breast Cancer Res. 21(22)2019.PubMed/NCBI View Article : Google Scholar

35 

Desmedt C, Voet T, Sotiriou C and Campbell PJ: Next-generation sequencing in breast cancer: First take home messages. Curr Opin Oncol. 24:597–604. 2012.PubMed/NCBI View Article : Google Scholar

36 

Ma R, Gong J and Jiang X: Novel applications of next-generation sequencing in breast cancer research. Genes Dis. 4:149–153. 2017.PubMed/NCBI View Article : Google Scholar

37 

Shrestha B and Dunn L: The declaration of Helsinki on medical research involving human subjects: A review of seventh revision. J Nepal Health Res Counc. 17:548–552. 2020.PubMed/NCBI View Article : Google Scholar

38 

Arteche-López A, Ávila-Fernández A, Romero R, Riveiro-Álvarez R, López-Martínez MA, Giménez-Pardo A, Vélez-Monsalve C, Gallego-Merlo J, García-Vara I, Almoguera B, et al: Sanger sequencing is no longer always necessary based on a single-center validation of 1109 NGS variants in 825 clinical exomes. Sci Rep. 11(5697)2021.PubMed/NCBI View Article : Google Scholar

39 

Donnelly TT, Khater AH, Al-Bader SB, Al Kuwari MG, Malik M, Al-Meer N, Singh R and Fung T: Factors that influence awareness of breast cancer screening among Arab women in Qatar: Results from a cross sectional survey. Asian Pac J Cancer Prev. 15:10157–10164. 2014.PubMed/NCBI View Article : Google Scholar

40 

Azaiza F and Cohen M: Health beliefs and rates of breast cancer screening among Arab women. J Womens Health (Larchmt). 15:520–530. 2006.PubMed/NCBI View Article : Google Scholar

41 

Baron-Epel O, Friedman N and Lernau O: Reducing disparities in mammography-use in a multicultural population in Israel. Int J Equity Health. 8(19)2009.PubMed/NCBI View Article : Google Scholar

42 

Bener A, Ayub H, Kakil R and Ibrahim W: Patterns of cancer incidence among the population of Qatar: A worldwide comparative study. Asian Pac J Cancer Prev. 9:19–24. 2008.PubMed/NCBI

43 

Najjar H and Easson A: Age at diagnosis of breast cancer in Arab nations. Int J Surg. 8:448–452. 2010.PubMed/NCBI View Article : Google Scholar

44 

Soskolne V, Marie S and Manor O: Beliefs, recommendations and intentions are important explanatory factors of mammography screening behavior among Muslim Arab women in Israel. Health Educ Res. 22:665–676. 2007.PubMed/NCBI View Article : Google Scholar

45 

Tarabeia J, Baron-Epel O, Barchana M, Liphshitz I, Ifrah A, Fishler Y and Green MS: A comparison of trends in incidence and mortality rates of breast cancer, incidence to mortality ratio and stage at diagnosis between Arab and Jewish women in Israel, 1979-2002. Eur J Cancer Prev. 16:36–42. 2007.PubMed/NCBI View Article : Google Scholar

46 

Bai X, Zhang E, Ye H, Nandakumar V, Wang Z, Chen L, Tang C, Li J, Li H, Zhang W, et al: PIK3CA and TP53 gene mutations in human breast cancer tumors frequently detected by ion torrent DNA sequencing. PLoS One. 9(e99306)2014.PubMed/NCBI View Article : Google Scholar

47 

Cho SH, Jeon J and Kim SI: Personalized medicine in breast cancer: A systematic review. J Breast Cancer. 15:265–272. 2012.PubMed/NCBI View Article : Google Scholar

48 

van den Brandt PA, Spiegelman D, Yaun SS, Adami HO, Beeson L, Folsom AR, Fraser G, Goldbohm RA, Graham S, Kushi L, et al: Pooled analysis of prospective cohort studies on height, weight, and breast cancer risk. Am J Epidemiol. 152:514–527. 2000.PubMed/NCBI View Article : Google Scholar

49 

Gallagher EJ and LeRoith D: Obesity and diabetes: The increased risk of cancer and cancer-related mortality. Physiol Rev. 95:727–748. 2015.PubMed/NCBI View Article : Google Scholar

50 

Gregor MF and Hotamisligil GS: Inflammatory mechanisms in obesity. Annu Rev Immunol. 29:415–445. 2011.PubMed/NCBI View Article : Google Scholar

51 

Seo BR, Bhardwaj P, Choi S, Gonzalez J, Andresen Eguiluz RC, Wang K, Mohanan S, Morris PG, Du B, Zhou XK, et al: Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci Transl Med. 7(301ra130)2015.PubMed/NCBI View Article : Google Scholar

52 

Anstey EH, Shoemaker ML, Barrera CM, O'Neil ME, Verma AB and Holman DM: Breastfeeding and breast cancer risk reduction: Implications for black mothers. Am J Prev Med. 53 (Suppl 1):S40–S46. 2017.PubMed/NCBI View Article : Google Scholar

53 

Levine AJ: p53, the cellular gatekeeper for growth and division. Cell. 88:323–331. 1997.PubMed/NCBI View Article : Google Scholar

54 

Langerød A, Zhao H, Borgan Ø, Nesland JM, Bukholm IR, Ikdahl T, Kåresen R, Børresen-Dale AL and Jeffrey SS: TP53 mutation status and gene expression profiles are powerful prognostic markers of breast cancer. Breast Cancer Res. 9(R30)2007.PubMed/NCBI View Article : Google Scholar

55 

Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P and Olivier M: Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: Lessons from recent developments in the IARC TP53 database. Hum Mutat. 28:622–629. 2007.PubMed/NCBI View Article : Google Scholar

56 

Silwal-Pandit L, Vollan HK, Chin SF, Rueda OM, McKinney S, Osako T, Quigley DA, Kristensen VN, Aparicio S, Børresen-Dale AL, et al: TP53 mutation spectrum in breast cancer is subtype specific and has distinct prognostic relevance. Clin Cancer Res. 20:3569–3580. 2014.PubMed/NCBI View Article : Google Scholar

57 

Ko LJ and Prives C: p53: Puzzle and paradigm. Genes Dev. 10:1054–1072. 1996.PubMed/NCBI View Article : Google Scholar

58 

Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012.PubMed/NCBI View Article : Google Scholar

59 

Bai H, Yu J, Jia S, Liu X, Liang X and Li H: Prognostic value of the TP53 mutation location in metastatic breast cancer as detected by next-generation sequencing. Cancer Manag Res. 13:3303–3316. 2021.PubMed/NCBI View Article : Google Scholar

60 

Ren J, Wang B and Li J: Integrating proteomic and phosphoproteomic data for pathway analysis in breast cancer. BMC Syst Biol. 12 (Suppl 8)(S130)2018.PubMed/NCBI View Article : Google Scholar

61 

Andrikopoulou A, Terpos E, Chatzinikolaou S, Apostolidou K, Ntanasis-Stathopoulos I, Gavriatopoulou M, Dimopoulos MA and Zagouri F: TP53 mutations determined by targeted NGS in breast cancer: A case-control study. Oncotarget. 12:2206–2214. 2021.PubMed/NCBI View Article : Google Scholar

62 

Bader AG, Kang S and Vogt PK: Cancer-specific mutations in PIK3CA are oncogenic in vivo. Proc Natl Acad Sci USA. 103:1475–1479. 2006.PubMed/NCBI View Article : Google Scholar

63 

Kang S, Bader AG and Vogt PK: Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci USA. 102:802–807. 2005.PubMed/NCBI View Article : Google Scholar

64 

Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C and González-Barón M: PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 30:193–204. 2004.PubMed/NCBI View Article : Google Scholar

65 

Noorolyai S, Shajari N, Baghbani E, Sadreddini S and Baradaran B: The relation between PI3K/AKT signalling pathway and cancer. Gene. 698:120–128. 2019.PubMed/NCBI View Article : Google Scholar

66 

Janku F, Yap TA and Meric-Bernstam F: Targeting the PI3K pathway in cancer: Are we making headway? Nat Rev Clin Oncol. 15:273–291. 2018.PubMed/NCBI View Article : Google Scholar

67 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009.PubMed/NCBI View Article : Google Scholar

68 

Cizkova M, Susini A, Vacher S, Cizeron-Clairac G, Andrieu C, Driouch K, Fourme E, Lidereau R and Bièche I: PIK3CA mutation impact on survival in breast cancer patients and in ERα, PR and ERBB2-based subgroups. Breast Cancer Res. 14(R28)2012.PubMed/NCBI View Article : Google Scholar

69 

Saal LH, Holm K, Maurer M, Memeo L, Su T, Wang X, Yu JS, Malmström PO, Mansukhani M, Enoksson J, et al: PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 65:2554–2559. 2005.PubMed/NCBI View Article : Google Scholar

70 

Martínez-Sáez O, Chic N, Pascual T, Adamo B, Vidal M, González-Farré B, Sanfeliu E, Schettini F, Conte B, Brasó-Maristany F, et al: Frequency and spectrum of PIK3CA somatic mutations in breast cancer. Breast Cancer Res. 22(45)2020.PubMed/NCBI View Article : Google Scholar

71 

QIAGEN Manchester, Ltd. therascreen® PIK3CA RGQ PCR Kit Instructions for Use (Handbook), 2019 (cited 2022 16 january); Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf19/P190001C.pdf.

72 

Bellevicine C, Sgariglia R, Nacchio M, De Luca C, Pisapia P, Pepe F and Troncone G: Molecular testing of thyroid fine-needle aspiration: Local issues and solutions. An interventional cytopathologist perspective. J Mol Pathol. 2:233–240. 2021.

73 

Hempel D, Ebner F, Garg A, Trepotec Z, Both A, Stein W, Gaumann A, Güttler L, Janni W, DeGregorio A, et al: Real world data analysis of next generation sequencing and protein expression in metastatic breast cancer patients. Sci Rep. 10(10459)2020.PubMed/NCBI View Article : Google Scholar

74 

Mosele F, Stefanovska B, Lusque A, Tran Dien A, Garberis I, Droin N, Le Tourneau C, Sablin MP, Lacroix L, Enrico D, et al: Outcome and molecular landscape of patients with PIK3CA-mutated metastatic breast cancer. Ann Oncol. 31:377–386. 2020.PubMed/NCBI View Article : Google Scholar

75 

Tang Y, Li J, Xie N, Yang X, Liu L, Wu H, Tian C, He Y, Wang X, He Q, et al: PIK3CA gene mutations in the helical domain correlate with high tumor mutation burden and poor prognosis in metastatic breast carcinomas with late-line therapies. Aging (Albany NY). 12:1577–1590. 2020.PubMed/NCBI View Article : Google Scholar

76 

Anderson EJ, Mollon LE, Dean JL, Warholak TL, Aizer A, Platt EA, Tang DH and Davis LE: A systematic review of the prevalence and diagnostic workup of PIK3CA mutations in HR+/HER2-metastatic breast cancer. Int J Breast Cancer. 2020(3759179)2020.PubMed/NCBI View Article : Google Scholar

77 

Board RE, Wardley AM, Dixon JM, Armstrong AC, Howell S, Renshaw L, Donald E, Greystoke A, Ranson M, Hughes A and Dive C: Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res Treat. 120:461–467. 2010.PubMed/NCBI View Article : Google Scholar

78 

Fusco N, Malapelle U, Fassan M, Marchiò C, Buglioni S, Zupo S, Criscitiello C, Vigneri P, Dei Tos AP, Maiorano E and Viale G: PIK3CA mutations as a molecular target for hormone receptor-positive, HER2-negative metastatic breast cancer. Front Oncol. 11(644737)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2022
Volume 16 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Messaoudi SA, Al Sharhan NA, Alharthi B, Babu SR, Alsaleh AB, Alasiri AM, Assidi M, Buhmeida A and Almawi WY: Detection of genetic mutations in patients with breast cancer from Saudi Arabia using Ion AmpliSeq™ Cancer Hotspot Panel v.2.0. Biomed Rep 16: 26, 2022
APA
Messaoudi, S.A., Al Sharhan, N.A., Alharthi, B., Babu, S.R., Alsaleh, A.B., Alasiri, A.M. ... Almawi, W.Y. (2022). Detection of genetic mutations in patients with breast cancer from Saudi Arabia using Ion AmpliSeq™ Cancer Hotspot Panel v.2.0. Biomedical Reports, 16, 26. https://doi.org/10.3892/br.2022.1509
MLA
Messaoudi, S. A., Al Sharhan, N. A., Alharthi, B., Babu, S. R., Alsaleh, A. B., Alasiri, A. M., Assidi, M., Buhmeida, A., Almawi, W. Y."Detection of genetic mutations in patients with breast cancer from Saudi Arabia using Ion AmpliSeq™ Cancer Hotspot Panel v.2.0". Biomedical Reports 16.4 (2022): 26.
Chicago
Messaoudi, S. A., Al Sharhan, N. A., Alharthi, B., Babu, S. R., Alsaleh, A. B., Alasiri, A. M., Assidi, M., Buhmeida, A., Almawi, W. Y."Detection of genetic mutations in patients with breast cancer from Saudi Arabia using Ion AmpliSeq™ Cancer Hotspot Panel v.2.0". Biomedical Reports 16, no. 4 (2022): 26. https://doi.org/10.3892/br.2022.1509