Open Access

Impact on breast cancer susceptibility and clinicopathological traits of common genetic polymorphisms in TP53, MDM2 and ATM genes in Sardinian women

  • Authors:
    • Matteo Floris
    • Giovanna Pira
    • Paolo Castiglia
    • Maria Laura Idda
    • Maristella Steri
    • Maria Rosaria De Miglio
    • Andrea Piana
    • Andrea Cossu
    • Antonio Azara
    • Caterina Arru
    • Giovanna Deiana
    • Carlo Putzu
    • Valeria Sanna
    • Ciriaco Carru
    • Antonello Serra
    • Marco Bisail
    • Maria Rosaria Muroni
  • View Affiliations

  • Published online on: August 8, 2022     https://doi.org/10.3892/ol.2022.13451
  • Article Number: 331
  • Copyright: © Floris 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

Common variants of genes involved in DNA damage correction [tumor protein p53 (TP53), murine double 2 homolog oncoprotein (MDM2) and ataxia‑telengiectasia mutated (ATM)] may serve a role in cancer predisposition. The purpose of the present study was to investigate the association of five variants in these genes with breast cancer risk and clinicopathological traits in a cohort of 261 women from northern Sardinia. Polymorphic variants in TP53 (rs17878362, rs1042522 and rs1625895), MDM2 (rs2279744) and ATM (rs1799757) were determined by PCR and TaqMan single nucleotide polymorphism assay in patients with breast cancer (n=136) and healthy controls (n=125). Association with clinicopathological (e.g., age at diagnosis, lymph node involvement, clinical stage) and lifestyle factors (e.g., smoking status, alcohol intake, contraceptive use) was also evaluated. TP53 rs17878362 and rs1625895 polymorphisms were associated with decreased risk of BC diagnosis in patients older than 50 years (codominant and recessive models) and post‑menopause (recessive model). Furthermore, there was a significant association between lymph node status (positive vs. negative) and ATM rs1799757‑delT in dominant and additive models and between MDM2 rs2279744‑allele and use of oral contraceptives. This analysis suggested that TP53 rs17878362 and rs1625895 may affect age of onset of breast cancer and ATM rs1799757 and MDM2 rs2279744 may be associated with lymph node status and prolonged use of oral contraceptives, respectively.

Introduction

Breast cancer (BC) is a key public health issue worldwide. It is the most frequently diagnosed malignancy in women and has surpassed lung cancer as the leading cause of global cancer incidence, representing 11.7% of all cancer cases diagnosed in 2020 (1). Global BC mortality shows a continuing downward trend due to widespread mammography screening, leading to early diagnosis, and the advent of personalized medicine (2,3).

The etiology of BC is multifactorial and highly complex. Numerous epidemiological studies have indicated that this disease is caused by a combination of multiple genetic and environmental, making the analysis of causative factors complex (49). Gene expression profiling studies have established the heterogeneous molecular nature of BC, which is considered as a collection of distinct ‘intrinsic’ subtypes (including Luminal A, Luminal B, ERBB2+ and Basal-like) characterized by variable biological and clinical behavior and response to treatment (10,11).

Furthermore, linkage studies have identified germline mutations in high-penetrance (capable of causing the disease by itself) BC susceptibility genes, including BRCA1, BRCA2, tumor protein p53 (TP53), partner and localizer of BRCA2, ataxia-telengiectasia mutated (ATM) and checkpoint kinase (CHEK)2, which are responsible for 5–10% of BC in the general population (12,13).

However, 90–95% of BC cases are sporadic forms in which general risk factors and single nucleotide polymorphisms (SNPs) in key genes involved in BC serve a role (14). Genome-wide association studies have identified numerous low-penetrance alleles with variable frequency in different ethnic groups that are present in a high percentage of individuals and contribute to breast carcinogenesis (1517) SNPs are implicated in genetic predisposition or resistance to a particular disease. Numerous SNPs have been associated with protein expression or changes in gene function via amino acid or indirect epigenetic changes (1820).

Somatic mutations of the tumor suppressor gene, which encodes TP53, are the most frequent genetic alterations associated with several types of human cancer, such as colorectum, pancreas, ovarian, lung carcinomas (2123). In humans, this gene encodes a 53 kDa nuclear phosphoprotein comprising 393 amino acids that form five highly conserved regions and four functional domains (24) with tetrameric transcription factor function that are involved in regulation of cell cycle progression, maintenance of genomic integrity, autophagy, inhibition of angiogenesis, differentiation, senescence and apoptotic cell death (2527).

SNPs associated with BC have been identified in TP53, murine double 2 homolog oncoprotein (MDM2) and ATM. SNPs in TP53, MDM2 and ATM contribute to individual susceptibility to cancer risk via alterations in gene expression regulation and exhibit wide geographical and ethnic variation (2832). Furthermore, they have been the subject of functional case-control studies on BC to assess their role as genetic determinants on cancer risk, progression, treatment outcome and survival in patients with cancer (3335).

The most well-characterized intronic SNP in TP53 is PIN 16bp (rs17878362), a 16-base pair insertion at nucleotide 11,951 within intron 3 (36). The r.13494 G to A nucleotide transversion (rs1625895) is a rare polymorphism located in intron 6, which is one of the hotspots for TP53 mutation (37). TP53 R72P (rs1042522) is the most widely investigated SNP in cancer genetic epidemiology (3840). It is a non-synonymous change of arginine (R72 form CGC) to proline (P72 form CCC) in exon 4 codon 72. This SNP is located in a proline-rich region of the putative SH3 binding domain; the two isoforms, R and P, differ in their biochemical and biological properties. The R72 variant exhibits markedly superior potential to induce apoptosis due to increased mitochondrial localization (41). On the other hand, the P72 variant has greater binding affinity with the transcriptional machinery to activate transcription, inducing higher levels of arrest in G1 phase and facilitating repair of damaged DNA (4244) TP53 is a tumor suppressor gene that, under normal conditions, exerts its protective role via activation of a range of anti-proliferative responses triggered by DNA damage, hypoxia, oxidative stress and oncogene activation (21,45,46). Somatic mutations in TP53 gene occur in ~31% of all cancers and 23% of BC cases; it is the second most frequently mutated gene after PI3KCA protooncogene (47,48).

MDM2 oncogene consists of 12 exons encoding a protein composed of 491 amino acids containing a P53 binding domain in the N-terminal and a RING domain responsible for E3 ligase activity at the C-terminal (49). In addition, mutations and polymorphic variants that alter the key role of MDM2 in control of P53 function manifest as cancer-associated phenotypes (50). Although multiple SNPs have been described in MDM2, the most characterized is the 309 T>G (rs2279744) variant at nucleotide 309 downstream of exon 1 in the P2 promoter (28,29,38,5154).

ATM was mapped to chromosome 11q22-23 and contains 66 exons which encode a large protein (350 kDa) of 3,056 amino acids (55). ATM protein is a member of the PI3K-related protein kinase family. It has serine-threonine kinase activity that phosphorylates numerous substrates including proteins encoded by BC susceptibility genes such as TP53, BRCA1 and CHEK2 (5658). ATM protein serves multiple roles in human cell biology: Its function is essential for recognition and repair of double-strand breaks in DNA, oxidative stress, control of cell cycle checkpoints, transcriptional regulation and apoptosis control (30). The polymorphism IVS24-9 delT (rs1799757) alters the acceptor splice site of intron 24 and increases BC risk by inducing genetic instability and normal response to DNA damage (59,60).

Several studies have been published on the potential association between polymorphic variants, especially variants of TP53 (and, to a lesser extent, MDM2 and ATM) and BC risk in different populations, showing controversial results, with TP53 associated with BC risk and with no effect on BC risk (53,6165).

The present case-control study aimed to analyze the effect of common genetic variants in TP53, MDM2 and ATM and general risk factors for BC in a cohort from the north of Sardinia. Given its peculiar history of genetically isolated population, the Sardinian population represents European genetic variability while also including variants that are particularly frequent due to genetic drift or natural selection, and thus is an ideal population for genetic studies (66). The present study evaluated the role on BC susceptibility conferred by TP53 rs17878362, rs1042522 and rs1625895, MDM2 rs2279744 and ATM rs1799757 by allele frequency analysis and haplotype association and assessed their association with clinicopathological and lifestyle traits. The present study also verified the frequency of cumulative effects between three TP53 polymorphic variants and SNPs of MDM2 and ATM. A further aim of the study was to evaluate the role of causal risk factors that contribute to BC development in a cohort of patients and healthy women from northern Sardinia.

Materials and methods

Sampling plan

The case-control study, conducted between May 2017 and January 2020, involved a total of 261 unrelated women (aged 26–86): 136 patients with BC registered at the Medical Oncology Unit in Sassari University Hospital, Sassari, Italy, and a control group of 125 healthy women, who had never previously been affected by any tumor, registered at the Unit of Occupational Medicine of the University of Sassari, Sassari, Italy. Written informed consent was obtained from each participant and the protocol was reviewed and approved by the Azienda Sanitaria Locale Sassari Bioethics Committee (approval no. 2468/CE, 14/03/2017). The study was conducted in accordance with the code of ethics of the World Medical Association (Declaration of Helsinki).

To recruit only individual representative of the Sardinian population, the participants were selected on the basis of the place of birth of their grandparents (all born in Sardinia). All participants completed a questionnaire to collect information on etiological factors underlying the onset of BC. In answering the questions, the participants were asked to consider their entire, or almost entire, life span.

The following data of patients were taken from medical records: age of disease onset, tumor histology, stage according to TNM classification, hormonal receptor, HER2 and Ki67 expression, distant metastasis and molecular subtype.

Polymorphism genotyping. EDTA treated blood samples (5 ml) were obtained from all participants upon enrolment and stored at −20°C until further use. Genomic DNA was extracted from 200 µl peripheral blood by QIAmp DNA Blood Mini kit (Qiagen GmbH) according to the manufacturer's instructions. TP53 Ins16bp was determined by PCR analysis in a final volume of 20 µl reaction mixture containing 100 ng DNA, 1X PCR buffer, 1.5 mM MgCl2, 0.2 mM each dNTP, 1.25 units Taq Gold DNA polymerase (Applied Biosystems; Thermo Fisher Scientific, Inc.) and 0.6 µM sense primer 5′-CCATGGGACTGACTTTTCTGC-3′ and antisense primer 5′-GGGGACTGTAGATGGGTGAA-3′. PCR conditions were as follows: initial denaturation at 95°C for 5 min, followed by 5 cycles of 95°C for 30 sec, 60°C for 30 sec and 72°C for 30 sec, 30 cycles of 95°C for 30 sec, 57°C for 30 sec and 72°C for 30 sec and final extension at 72°C for 5 min, as previously described (67). The PCR products were electrophoresed on a 3% Metaphor agarose gel, stained with ethidium bromide and visualized by UV trans-illumination. Genotypes of TP53 Arg72Pro (cat. no. C_2403545_10), TP53 r.13494G>A (cat. no. C_8727782_20), MDM2 309T>G (cat. no. C_15968533_20) and ATM IVS24-9 (cat. no. C_33307825_10) were detected using TaqMan SNP assay kits (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to manufacturer's instructions. Briefly, 12.5 ng genomic DNA in 5 µl was added to a 25 µl reaction well with 20 µl reaction mix containing forward and reverse primers and two allele-specific fluorescent labelled probes, one wild-type (VIC) and one variant allele specific (FAM). PCR and allele detection were performed using the ABI Prism 7300 Sequence Detection System.

Statistical analysis

Minor allele frequency of polymorphisms in the Sardinian population was verified using public Sardinia Pheweb (pheweb.irgb.cnr.it/). SNP deviations from Hardy-Weinberg Equilibrium (HWE) in the control population were assessed independently for each SNP using 1-degree-of-freedom χ2 test, with SNPassoc v. 2.0-11 (68). Student's paired t-test and Fisher's exact test implemented in the R base package v. 4.2.1 (https://www.r-project.org/) were used to evaluate the differences in descriptive variables at diagnosis, such as age or body mass index (BMI), between the cases and controls. For each SNP, genotypic and allelic association were tested with regression analysis under multiple inheritance models (additive, dominant, codominant, over-dominant and recessive), using a custom R script and SNPassoc package (association function), to compare patients with controls. The R package fsmb v. 0.7.3 was used to calculate odds ratio (OR) and 95% confidence interval (CI) according to the Woolf method (69): when 0 count was observed, Gart-adjusted logit interval was calculated. Before association, potential confounders (such as age and BMI) were tested by including them as covariates in the association models; accordingly, age and BMI were added as covariates in the analysis. Haplotype analysis was restricted to polymorphisms located on the same chromosome and genetic region (for example, variants encompassing a 1 Mb region). Measures of linkage disequilibrium (LD) between each pair of SNPs (D' and r2 statistic) and haplotype reconstruction were obtained in the 1000 genomes European population (https://www.internationalgenome.org/) (70) using the LDpair web tool (ldlink.nci.nih.gov/?tab=ldpair). The most common haplotype was selected as the reference. OR and 95% CI were calculated to estimate the degree of association between haplotype and risk of BC. A threshold of P≤0.05 was used to assess statistical significance for each genetic association. Bonferroni's correction was applied to adjust for multiple testing with a significance threshold of P≤0.01 (0.05/number of inheritance models tested for each genetic variant) for differences in descriptive variables at diagnosis between cases and controls. To evaluate the interaction between lifestyle factors and SNPs with BC risk, a logistic regression model was constructed.

Results

Comparison of questionnaire information between BC patients and controls

The primary hormonal/reproductive, lifestyle/environmental and familial cancer risk factors of cases and controls from the questionnaire analysis are summarized in Table I. The mean age of cases and controls was 56.80 and 50.71 years, respectively (P=1.122×10−5). There were significant differences in BMI (OR: 3.150, 95% CI: 1.666-5.957, P=5×10−4), alcohol intake (OR: 1.955, 95% CI: 1.024-3.733, P=2.85×10−2) and competitive sport during adolescence (OR: 2.648, 95% CI: 1.577-4.444, P=3×10−4) between BC cases and controls. Regarding the role of genetic factors, first-degree relatives (mother, sisters, brothers and cousins and aunts only of first-degree) of cases more frequently experienced onset of BC at an age <45 years (OR: 3.573, 95% CI: 2.002-6.379, P=1.77×10−5). Furthermore, there was more frequent occurrence in number of familial cases of BC ≥3 and cancers of any organ ≥4 (OR: 4.139, 95% CI: 1.818-9.426, P=7×10−4 and OR: 1.848, 95% CI: 1.113-3.067, P=2.4×10−2 respectively), as well as more frequent occurrence of benign pre-tumor lesions (OR: 2.438, 95% CI: 1.207-4.924, P=1.7×10−2) between cases compared with controls. There were no statistically significant differences between cases and controls regarding all hormonal/reproductive (only age of menarche and nulliparity exhibited a small role; P=8×10−2 and P=9×10−2 respectively) and other lifestyle/environmental risk factors, although secondhand smoke exposure (P=7×10−2) appeared to have a small effect. Questionnaire analysis data showed a relevant impact on BC risk of both lifestyle and genetic factors in patients compared to controls.

Table I.

Hormonal/reproductive, lifestyle/environmental and familial cancer risk factors in patients and controls.

Table I.

Hormonal/reproductive, lifestyle/environmental and familial cancer risk factors in patients and controls.

Risk factorCases (n.136) n, %Controls (n.125) n, %OR (CI 95%)P (chisq)
Age at menarche <13 y70, 51.550, 40.01.591 (0.974-2.600)0.08306
Nulliparity38, 27.948, 38.40.622 (0.370-1.046)0.09611
Age at first bird ≥35 y12, 8.817, 13.60.615 (0.281-1.345)0.3033
No breastfeeding58, 42.658, 46.40.859 (0.527-1.401)0.6278
CO ≥10 years53, 39.049, 39.20.990 (0.602-1.629)1
aAge at menopause >51 y21, 15.419, 15.21.019 (0.519-2.000)1
BMI >2743, 31.616, 12.83.150 (1.666-5.957)0.0004963
Drinking31, 22.817, 13.61.876 (0.980-3.592)0.07919
bSmoke ≥10 p/y37, 27.234, 27.21.000 (0.580-1.726)1
Passive smoke78, 57.457, 45.61.604 (0.984-2.617)0.07603
No sport91, 66.974, 59.21.394 (0.841-2.309)0.2452
No sport in adolescence100, 73.564, 51.22.648 (1.577-4.444)0.000317
Chemicals exposure27, 19.917, 13.61.574 (0.811-3.053)0.237
cBenign breast lesions30, 22.113, 10.42.438 (1.207-4.924)0.01781
Bilateral BC in family23, 16.916, 12.81.387 (0.695-2.765)0.449
BC <45 y in family57, 41.921, 16.83.573 (2.002-6.379)1.77e-05
Ovary cancer in family19, 14.010, 8.01.868 (0.833-4.189)0.1815
N. BC ≥3 in family30, 22.18, 6.44.139 (1.818-9.426)0.0006557
N. all tum ≥4 in family62, 45.639, 31.21.848 (1.113-3.067)0.02401

a Only natural menopause was considered (55 BC patients and 60 healthy controls).

b Smoking status at the time of recruitment or in the past. Pack/years (p/y) is calculated as follows: (Years of smoking × number of cigarettes/day)/20.

c Parameters considered in first-degree relatives (parent, sibling, or child). OR, odds ratio; y, years; OC, oral contraceptives; BMI, body mass index; BC, breast cancer.

Clinicopathological characteristics of patients with BC

Clinical and pathological data of all patients are summarized in Table II. In brief, the analysis of BC cases in the north Sardinia cohort revealed that most cases were diagnosed in woman aged >40 years (83.82%) and who were pre-menopausal (56.68%). The most common histological type was invasive ductal carcinoma (85.29%), and more frequently the tumors showed early clinical stage (0-II, 66.18%); 72.79% of tumors expressed estrogen receptor (ER), 67.65% expressed progesterone receptor (PgR) and 30.88% were HER2+. Finally, the molecular subtypes of our BC cohort were: 58 Luminal A (42.65%), 21 Luminal B-like Her-2 neg (15.44%), 21 Luminal B-like Her-2 pos (15.44%), 21 Her-2 overexpressing (15.44%) and 15 Triple negative (11.03%).

Table II.

Clinicopathological features of breast cancer cases.

Table II.

Clinicopathological features of breast cancer cases.

VariableCases (%), n=136
Age at diagnosis, years (Mean=52.10)
  ≤4022.00 (16.18)
  >40114.00 (83.82)
Menopause status at diagnosis
  Pre-73.00 (56.68)
  Post-63 (46.32)
Histological subtype
  Ductal116.00 (85.29)
  Lobular10.00 (7.35)
  Other10.00 (7.35)
ER expression
  Positive99.00 (72.79)
  Negative37.00 (27.21)
PgR expression
  Positive92.00 (67.65)
  Negative44.00 (32.35)
HER2
  Positive42.00 (30.88)
  Negative89.00 (65.44)
  Missing5.00 (3.68)
Ki67
  ≤30%104.00 (76.47)
  >30%31.00 (22.79)
  Missing1.00 (0.73)
Distant metastasis
  M0112.00 (82.35)
  M122.00 (16.18)
  Missing2.00 (1.47)
Clinical stage
  Early (0, I, II)90.00 (66.18)
  Advanced (III, IV)39.00 (28.68)
  Missing7.00 (5.15)
Molecular subtype
  Luminal A58.00 (42.65)
  Luminal B-like HER2-negative21.00 (15.44)
  Luminal B-like HER2-positive21.00 (15.44)
  HER2-overexpressing21.00 (15.44)
  Triple negative15.00 (11.03)

[i] ER, estrogen receptor; PgR, progesterone receptor. Missing=number of patients for which this particular value is not available.

Genetic variants and BC risk association

TP53 (rs17878362) wild-type allele resulted in 153 bp fragment; 169 bp fragment was the variant allele (Fig. S1). None of the SNPs were discarded there was no deviation from HWE in the control population (TP53 rs17878362 HWEp=0.795, rs1042522 HWEp=0.826, and rs1625895 HWEp=1, ATM rs1799757, HWEp=0.306 and MDM2 rs2279744 HWEp=0.848). The distribution of allele frequencies of these SNPs in controls and BC cases in the Sardinian population is shown in Table III. In the distribution of alleles analyzed under multiple genetic inheritance models (codominant, dominant, recessive, over-dominant and additive), no statistically significant effect on the risk of BC was found in the two cohorts (association analysis adjusted for age and BMI). Haplotype analysis combining TP53 genotypes and those of MDM2 and ATM polymorphisms did not reveal significant differences associated with BC (data not shown). The SNPs of TP53, ATM and MDM2 considered in the present study have not shown effects on BC susceptibility.

Table III.

Genotype and allele frequencies of TP53, MDM2 and ATM variants in BC cases and controls (adjusted for age and BMI).

Table III.

Genotype and allele frequencies of TP53, MDM2 and ATM variants in BC cases and controls (adjusted for age and BMI).

A, TP53 (rs17878362). Ref. allele, NoIns; alt. allele, Ins16

ModelGenotypeCases (%),n=136.00Controls (%),n=125.00OR (95% CI)P-value
AlleleDel220.00 (80.88)196.00 (78.40)1.00
Ins52.00 (19.12)54.00 (21.60)1.16 (0.74-1.83)0.51
CodominantDel/del88.00 (64.70)76.00 (60.80)1.00
Del/ins44.00 (32.35)44.00 (35.20)0.85 (0.49-1.47)
Ins/ins4.00 (2.94)5.00 (4.00)0.55 (0.13-2.40)0.65
DominantDel/del88.00 (64.70)76.00 (60.80)1.00
Del/ins + ins/ins48.00 (35.29)49.00 (39.20)0.82 (0.48-1.39)0.45
RecessiveDel/del + del/ins132.00 (97.06)120.00 (96.00)1.00
Ins/ins4.00 (2.94)5.00 (4.00)0.59 (0.14-2.50)0.47
Over-dominantDel/del + ins/ins92.00 (67.65)81.00 (64.80)1.00
Del/ins44.00 (32.35)44.00 (35.20)0.87 (0.50-1.51)0.62
Log-additive0,1,2136.00 (52.10)125.00 (47.90)0.81 (0.51-1.29)0.38

B, TP53 (rs1042522). Ref. allele, C; alt. allele, G

Model GenotypeCases (%), n=136.00Controls (%), n=125.00OR (95% CI)P-value

AlleleG (Arg)202.00 (74.26)179.00 (71.60)1.00
C (Pro)70.00 (25.73)71.00 (28.40)1.14 (0.76-1.72)0.55
CodominantG/G75.00 (55.15)63.00 (50.40)1.00
G/C52.00 (38.23)53.00 (42.40)0.76 (0.44-1.30)
C/C9.00 (6.62)9.00 (7.20)0.60 (0.21-1.72)0.46
DominantG/G75.00 (55.15)63.00 (54.40)1.00
G/C + C/C61.00 (44.85)62.00 (49.60)0.74 (0.44-1.23)0.24
RecessiveG/G + G/C127.00 (93.38)116.00 (92.80)1.00
C/C9.00 (6.62)9.00 (7.20)0.68 (0.24-1.89)0.45
Over-dominantG/G + C/C84.00 (61.76)72.00 (57.60)1.00
G/C52.00 (38.23)53.00 (42.40)0.81 (0.48-1.36)0.42
Log-additive(0,1,2)136.00 (52.10)125.00 (47.90)0.77 (0.50-1.17)0.21

C, TP53 (rs1625895). Ref. allele, A; alt. allele, G

Model GenotypeCases (%), n=136.00Controls (%), n=125.00OR (95% CI)P-value

AlleleG221.00 (81.25)200.00 (80.00)1.00
A51.00 (18.75)50.00 (20.00)1.08 (0.68-1.71)0.74
CodominantG/G88.00 (64.71)80.00 (64.00)1.00
G/A45.00 (33.09)40.00 (32.00)0.92 (0.53-1.61)
A/A3.00 (2.20)5.00 (4.00)0.37 (0.07-1.82),0.45
Dominant,G/G88.00 (64.71)80.00 (64.00)1.00
G/A + A/A48.00 (35.29)45.00 (36.00)0.86 (0.50-1.47),0.57
Recessive,G/G + G/A133.00 (97.79)120.00 (96.00)1.00
A/A3.00 (2.20)5.00 (4.00)0.38 (0.08-1.85),0.22
Over-dominantG/G + A/A91.00 (66.91)85.00 (68.00)1.00
G/A45.00 (33.09)40.00 (32.00)0.96 (0.55-1.67),0.89
log-Additive(0,1,2)136.00 (52.10)125.00 (47.90)0.81 (0.50-1.30),0.38

D, MDM2 (rs2279744). Ref. allele, T; alt. allele, G

Model GenotypeCases (%), n=136.00Controls (%), n=125.00OR (95% CI)P-value

AlleleT176.00 (64.71)157.00 (62.80)1.00
G96.00 (35.29)93.00 (37.20)1.08 (0.75-1.58)0.71
CodominantT/T61.00 (44.85)50.00 (40.00)1.00
T/G54.00 (39.71)57.00 (45.60)0.81 (0.46-1.42)
G/G21.00 (15.44)18.00 (14.40)0.98 (0.45-2.11),0.75
Dominant,T/T61.00 (44.85)50.00 (40.00)1.00
TG + G/G75.00 (55.15)75.00 (60.00)0.85 (0.51-1.43),0.55
Recessive,TT + T/G115.00 (84.56)107.00 (85.60)1.00
G/G21.00 (15.44)18.00 (14.40)1.09 (0.53-2.22),0.82
Over-dominantT/T + G/G82.00 (60.29)68.00 (54.40)1.00
T/G54.00 (39.71)57.00 (45.60)0.82 (0.49-1.37),0.45
log-Additive(0,1,2)136.00 (52.10)125.00 (47.90)0.94 (0.66-1.36),0.76

E, ATM (rs1799757). Ref. allele, T; alt. allele, delT

Model GenotypeCases (%), n=136.00Controls (%), n=125.00OR (95% CI)P-value

AlleleT235.00 (86.40)213.00 (85.20)1.00
delT37.00 (13.60)37.00 (14.80)1.10 (0.65-1.86)0.71
CodominantT/T100.00 (73.53)89.00 (71.20)1.00
T/delT35.00 (25.73)35.00 (28.00)1.00 (0.56-1.79)
delT/delT1.00 (0.73)1.00 (0.80)0.37 (0.02-6.29),0.80
Dominant,T/T100.00 (75.53)89.00 (71.20)1.00
T/delT + delT/delT36.00 (26.47)36.00 (28.80)0.97 (0.54-1.73),0.92
Recessive,T/T + T/delT135.00 (99.26)124.00 (99.20)1.00
delT/delT1.00 (0.73)1.00 (0.80)0.37 (0.02-6.27),0.50
Over-dominantT/T + delT/delT101.00 (74.26)90.00 (72.00)1.00
T/delT35.00 (25.73)35.00 (28.00)1.01 (0.56-1.80),0.98
log-Additive(0,1,2)136.00 (52.10)125.00 (47.90)0.94 (0.54-1.63),0.82

[i] Del, deletion; Ins, insertion; BMI, body mass index.

Association between genotypes and clinical features and lifestyle

The present study evaluated the potential association between SNP genotypes in codominant, dominant, recessive, over-dominant and log-additive inheritance models and clinicopathological parameters such as familiar BC cases and other primary tumors, age at diagnosis, lymph node involvement, clinical stage, ER, Ki67 and HER2 status and menopause status at diagnosis. TP53 rs17878362 and rs1625895 polymorphisms were associated with a decreased risk of BC diagnosis at an age >50 years in codominant (ins/ins vs. del/del OR 0.01, 95% CI: 0.00-0.30, P=4.2×10−2; A/A vs. G/G OR 0.01, 95% CI: 0.00-0.28, P=4.8×10−2) and recessive models (ins/ins vs. del/ins OR: 0.01, 95% CI: 0.00-0.28, P=1.1×10−2; A/A vs. A/G OR 0.01, 95% CI: 0.00-0.28, P=1.4×10−2, Table IV). SNPs were associated with a decreased risk of BC diagnosis in post-menopausal cases in the recessive inheritance model (ins/ins vs. del/ins OR 0.03, 95% CI: 0.00-0.68, P=2.4×10−2; A/A vs. A/G OR 0.03, 95% CI: 0.00-0.69, P=4.0×10−2; association analysis adjusted for age and BMI; Table IV). Furthermore, there was an association between lymph node (positive vs. negative) status and allele delT in ATM rs1799757 dominant and additive models (T/delT-delT/delT vs. T/T OR 0.43, 95% CI: 0.19-0.98, P=4.0×10−2 and OR 0.43, 95% CI: 0.19-0.94, P=3.0×10−2, respectively, Table IV). The study also evaluated the potential association between distribution of SNP genotypes and lifestyle traits such as smoking status, alcohol intake and oral contraceptive (OC) use. There was an interaction between use of OC for <10 and ≥10 years and the rs2279744 polymorphism of MDM2 in BC cases (83/53) and controls (76/49). GG genotype with OC intake for >10 years was associated with an increase in BC risk (OR: 3.43, 95% CI: 0.92-12.78, P=4.8×10−2, association analysis adjusted for age and BMI; Table V). Of the 21 homozygotes of the G allele, 5 were ER-negative and 16 ER-positive with a frequency of the G allele equal to 0.31 and 0.38 respectively (data not shown). Furthermore, association analysis suggested an association between TP53 rs1042522 Pro allele/MDM2 rs2279744 T allele combination (P=5.6×10−2) and TP53 rs1042522 recessive Arg/Arg-Pro/Arg recessive model and premenopausal status (P=5.3×10−2; Table SI and SII association analysis adjusted for age and BMI). The present study showed that the investigated SNPs affect some clinical characteristics and are associated with prolonged OC intake in BC patients.

Table IV.

Association of TP53 polymorphisms and age and menopausal status and ATM polymorphism and lymph nodes status at diagnosis (adjusted by age and BMI).

Table IV.

Association of TP53 polymorphisms and age and menopausal status and ATM polymorphism and lymph nodes status at diagnosis (adjusted by age and BMI).

VariableSNPModels and allelenOR (95% CI)P-value
Age at diagnosis, years ≤50/>50TP53 rs17878362,Codominant Del/del45/431.00
(n=67/69) Del/ins19/251.05 (0.38-2.89)
Ins/ins3/10.01 (0.00-0.30) 4.2×10−2
Recessive64/681.00
Del/del-del/ins
Ins/ins3/10.01 (0.00-0.28) 1.1×10−2
TP53 rs1625895Codominant G/G5/431.00
A/G20/250.96 (0.35-2.64)
A/A2/10.01 (0.00-0.28) 4.8×10−2
Recessive G/G-A/G65/681.00
A/A2/10.01 (0.00-0.28) 1.4×10−2
Menopausal status at diagnosisTP53 rs17878362,Recessive70/621.00
Pre-/post- (n=73/63) Del/del-del/ins
Ins/ins3/10.03 (0.00-0.68) 3.4×10−2
Recessive G/G-A/G71/621.00
A/A2/10.03 (0.00-0.69) 4.0×10−2
Lymph nodes at diagnosisATM rs1799757Dominant T/T44/521.00
Negative/positive (n=67/64), T/delT-delT/delT23/120.43 (0.19-0.98) 4.0×10−2
Log additive (0,1,2) 0.43 (0.19-0.94) 3.0×10−2

[i] Del, deletion; Ins, insertion; BMI, body mass index.

Table V.

Association of polymorphism rs2279744 of MDM2 and duration of oral contraceptive use in cases and controls (adjusted by age and BMI).

Table V.

Association of polymorphism rs2279744 of MDM2 and duration of oral contraceptive use in cases and controls (adjusted by age and BMI).

Oral contraceptive use, yearsAlleleControl (n=125)Cases (n=136)OR (95% CI)P-value
<10T/T30371.00
T/G32361.04 (0.51-2.12)
G/G14100.60 (0.22-1.60)
≥10T/T20241.57 (0.69-3.57)
T/G25180.80 (0.36-1.80)
G/G4113.43 (0.92-12.78) 4.8×10−2

Discussion

BC etiology is extremely complex, it might be partially explained by individual genetic susceptibility, as well as by numerous extrinsic factors linked to lifestyle, which modify the normal biology of the mammary glandular epithelium during the woman's life. The present study investigated the association of 5 variants in TP53, MDM2 and ATM (rs17878362, rs1042522, rs1625895, rs2279744, rs1799757) with BC susceptibility, clinicopathological and lifestyle traits in a cohort of Sardinian women. TP53 rs17878362 and rs1625895 polymorphisms were associated with decreased risk of BC diagnosis both in patients aged >50 years and those who were post-menopausal. Moreover, there was a significant association between lymph node status and ATM rs1799757-delT and MDM2 rs2279744-allele and OC use.

When analyzing BC risk in the two groups using multiple genetic models, there was no statistically significant difference for polymorphisms TP53 rs17878362, rs1042522 and rs1625895. Published data report controversial results regarding the role of these SNPs in BC predisposition, showing increase or decrease of BC susceptibility risk, when other factors are not taken into consideration. This may be due to factors such as differences in geographical location and ethnicity, methodological approach and composition of the analyzed cohort. In this regard, large collaborative multi-center studies should be undertaken with the same methodology with particular attention paid to the ancestral genetic origins of the population under consideration (7186).

Regarding the rs17878362 Ins16bp and rs1625895 13494G>A TP53 polymorphisms, the presence of Ins-allele and AA-allele, respectively, was associated with a decreased probability of BC diagnosis in those aged >50 years (codominant model and recessive model) and with pre-menopausal status (recessive model). Certain evidence supports the hypothesis that non-coding genetic variations may be important in regulating P53 activity by initiating, for example, aberrant splicing of pre-messenger RNA and production of mRNA that is translated into a defective protein (8789). Both rs17878362 Ins16bp and rs1625895 13494G> A are intronic polymorphisms with a potential biological association with certain types of cancer, such as lung, colorectal and ovarian (67,78,83,90,91). As for the first, the variant A2 allele (16p duplication) causes an alteration in mRNA processing (90). Based on the present results, it might be hypothesized that the risk factors promoting BC combined with Ins and A alleles of the rs17878362 and rs1625895 polymorphisms respectively, affect the age of BC onset.

Here, the MDM2 309T> G polymorphism did not reveal any role in BC risk; however, there was a significant association between the GG variant and BC risk increase in those using OC for ≥10 years. In 2004, Bond et al (28) demonstrated an increase in the binding affinity of the consensus sequence of MDM2 promoter with the transcription factor Sp1 in conditions of homozygosity of the G allele of SNP309T>G, producing an 8-fold increase in MDM2 mRNA and a 4-fold increase in Mdm2 protein, resulting in the attenuation of P53 pathway both in vitro and in vivo. As this polymorphism is localized in a promoter region regulated by a hormonal signaling pathway and the G allele of SNP309T> G increases the affinity of co-transcriptional activator for nuclear hormone receptor (such as Sp1), there is accelerated hormone-dependent tumor formation (92). To the best of our knowledge, the present study is the first to report an association between the MDM2 rs2279744 GG genotype and a history of OC use for ≥10 years, these results are supported by data previously described.

ATM IVS24-9 polymorphism did not increase susceptibility to BC in the present case-control study. However, heterozygous T/-T and homozygous -T/-T genotypes in dominant and log-additive models were significantly associated with negative status of lymph node involvement at diagnosis. To the best of our knowledge, however, functional and case-control studies are scarce (59,9396). The IVS24-9 polymorphism is a splice acceptor site that increases BC risk by favoring genetic instability and normal response to DNA damage (97); additional studies are needed to define the putative role of lymph node negativity. ATM has numerous polymorphic sites and some may confer risk of BC and other tumors (30,95,96), although these conclusions are limited and conflicting and must therefore be confirmed by further functional and case-control studies.

Regarding analysis of lifestyle and demographic factors, there was only a small association between the age of menarche onset and nulliparity; however this was not significant. Research conducted in different populations and with a higher number of samples indicate increased relative risk associated with longer fertile period (early menarche and late menopause), nullity or low parity, first full-term pregnancy after 30–35 years, failure to breastfeed and OC use (4–7,98–100). However, other epidemiological investigations provide conflicting data, attributing little value to these factors and considering them important only for surveillance (8,101103). Here, there was a strong association between high BMI, frequent (daily/often) alcohol intake and absence of physical activity during adolescence and increased risk of BC. A sedentary lifestyle combined with being overweight/obese has adverse health effects and leads to an increased BC risk, particularly in postmenopausal women (104106). This is due to altered cellular sensitivity to insulin, inflammation and cytokine production, overexpression of leptinin adipose tissue, bioavailability of sex hormones and activation/variation of epigenetic mechanisms (107109). In obese women, the adipose tissue represents an important source of endogenous estrogens due to conversion of androgenic precursors (110). Secondly, the adipose microenvironment is similar to the tumor microenvironment in cellular composition, low-grade chronic inflammation and high ratio of reactive species oxygen to antioxidants (111).

Physical activity and BC have been linked in numerous studies (112,113). This is reported to be associated with decreased BC risk in post-menopausal women and mortality (114,115). Practicing intense physical activity in adolescence decreases BC risk by delaying the age of menarche onset and decreasing the amount of bioavailable circulating hormones (116,117). Acting on these modifiable risk factors, through regular physical activity and control of body weight, could contribute to risk reduction by modifying metabolic and hormonal status.

The assessment of genetic risk share indicates that BC cases occurring at a relatively early age in first-degree relatives, high number of familial BC (≥3) and primary tumors in other locations (≥4) and benign breast lesions were risk factors for BC. Similar results have been reported in other studies (118121).

In conclusion, genetic analysis of TP53 rs17878362, rs1042522, and rs1625895, MDM2 rs2279744 and ATM rs1799757 suggested that no polymorphic allele, alone or in combination within haplotypes, was associated with BC risk in Sardinian women. However, TP53 Ins16bp and 13494G>A SNPs showed a significant association with age of BC onset and menopausal status in BC patients. However, the most significant result was MDM2 309T>G polymorphism. To the best of our knowledge, the present study is the first to suggest an increased risk among GG-carrier patients who have taken OC for >10 years. The present results contribute to the characterization of the genetic BC susceptibility profile. However, caution is required when drawing conclusions and further studies are needed to elucidate the role of these polymorphisms in predisposition and as predictors of treatment outcome and prognostic markers in BC.

The descriptive analysis of questionnaires confirmed the key role of lifestyle/environmental and genetic/familial causal factors in increasing the relative risk of BC. At present, knowledge of breast carcinogenesis remains incomplete and although causative modifiable and immutable factors are known, it is not possible to identify subgroups of women who will develop BC, except those showing a high familial genetic risk.

Future large-scale studies should simultaneously consider intrinsic and extrinsic risk determinants, their interaction and their association with the individual genetic predisposition.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Dr Emma Dempsey (Centro Linguistico di Ateneo-University of Sassari, Sassari, Italy) for the review of the English language.

Funding

The present study was supported by the Fondazione Banco di Sardegna, ‘Salute Pubblica, Medicina Preventiva e Riabilitativa’, Anno 2019 and ‘Fondo di Ateneo per la ricerca’ Università degli studi di Sassari 2019.

Availability of data and materials

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

Authors' contributions

MRM conceived and coordinated the study and drafted the manuscript. PC, MF, MRM, GP, MI, AA and MS designed the methodology, collected, analyzed and visualized data. GP, AC, MRDM, GD, CA and CC performed experiments. MRM, AS, MB, AP, CP, VS and MB were involved in sample selection, collection and analysis of results. MRDM, MF, MI, MS, AA and AP edited the manuscript. MF and MRM confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by Azienda Sanitaria Locale (ASL) Sassari Bioethical Committee and written informed consent was obtained from each participant, according to Italian Legislation (approval no. 2468/CE 14/03/2017). All data are treated according to the EU General Data Protection Regulation 2016/679.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Yaffe MJ, Mittmann N, Alagoz O, Trentham-Dietz A, Tosteson AN and Stout NK: The effect of mammography screening regimen on incidence-based breast cancer mortality. J Med Screen. 25:197–204. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Alberg AJ, Lam AP and Helzlsouer KJ: Epidemiology, prevention, and early detection of breast cancer. Curr Opin Oncol. 11:435–441. 1999. View Article : Google Scholar

4 

Momenimovahed Z and Salehiniya H: Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer (Dove Med Press). 11:151–164. 2019.PubMed/NCBI

5 

Wu MH, Chou YC, Yu JC, Yu CP, Wu CC, Chu CM, Yang T, Lai CH, Hsieh CY, You SL, et al: Hormonal and body-size factors in relation to breast cancer risk: A prospective study of 11,889 women in a low-incidence area. Ann Epidemiol. 16:223–229. 2006. View Article : Google Scholar

6 

Dai Q, Liu B and Du Y: Meta-analysis of the risk factors of breast cancer concerning reproductive factors and oral contraceptive use. Front Med China. 3:452–458. 2009. View Article : Google Scholar

7 

Golubnitschaja O, Debald M, Yeghiazaryan K, Kuhn W, Pešta M, Costigliola V and Grech G: Breast cancer epidemic in the early twenty-first century: Evaluation of risk factors, cumulative questionnaires and recommendations for preventive measures. Tumor Biol. 37:12941–12957. 2016. View Article : Google Scholar

8 

Tamakoshi K, Yatsuya H, Wakai K, Suzuki S, Nishio K, Lin Y, Niwa Y, Kondo T, Yamamoto A, Tokudome S, et al: Impact of menstrual and reproductive factors on breast cancer risk in Japan: Results of the JACC study. Cancer Sci. 96:57–62. 2005. View Article : Google Scholar

9 

Daly AA, Rolph R, Cutress RI and Copson ER: A Review of modifiable risk factors in young women for the prevention of breast cancer. Breast cancer (Dove Med Press). 13:241–257. 2021.PubMed/NCBI

10 

Uva P, Cossu-Rocca P, Loi F, Pira G, Murgia L, Orrù S, Floris M, Muroni MR, Sanges F, Carru C, et al: miRNA-135b contributes to triple negative breast cancer molecular heterogeneity: Different expression profile in basal-like versus non-basal-like phenotypes. Int J Med Sci. 15:536–548. 2018. View Article : Google Scholar

11 

Perou CM, Sørile T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Dalivandan ST, Plummer J and Gayther SA: Risks and Function of breast cancer susceptibility alleles. Cancers (Basel). 13:39532021. View Article : Google Scholar : PubMed/NCBI

13 

Dumitrescu RG and Cotarla I: Understanding breast cancer risk-where do we stand in 2005? J Cell Mol Med. 9:208–221. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Deng N, Zhou H, Fan H and Yuan Y: Single nucleotide polymorphisms and cancer susceptibility. Oncotarget. 8:110635–110649. 2017. View Article : Google Scholar

15 

Palomba G, Loi A, Porcu E, Cossu A, Zara I, Budroni M, Dei M, Lai S, Mulas A, Olmeo N, et al: Genome-wide association study of susceptibility loci for breast cancer in Sardinian population. BMC Cancer. 15:2015. View Article : Google Scholar

16 

Narod SA: Genetic variants associated with breast-cancer risk. Lancet Oncol. 12:415–416. 2011. View Article : Google Scholar

17 

Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, Pukkala E, Skytthe A and Hemminki K: Environmental and heritable factors in the causation of cancer-analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 343:78–85. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Castiglia P, Sanna V, Azara A, De Miglio MR, Murgia L, Pira G, Sanges F, Fancellu A, Carru C, Bisail M and Muroni MR: Methylenetetrahydrofolate reductase (MTHFR) C677T and A1298C polymorphisms in breast cancer: A sardinian preliminary case-control study. Int J Med Sci. 16:1089–1095. 2019. View Article : Google Scholar

19 

Floris M, Sanna D, Castiglia P, Putzu C, Sanna V, Pazzola A, De Miglio MR, Sanges F, Pira G, Azara A, et al: MTHFR, XRCC1 and OGG1 genetic polymorphisms in breast cancer: A case-control study in a population from North Sardinia. BMC Cancer. 20:2342020. View Article : Google Scholar : PubMed/NCBI

20 

Pilato B, Martinucci M, Danza K, Pinto R, Petriella D, Lacalamita R, Bruno M, Lambo R, D'Amico C, Paradiso A and Tommasi S: Mutations and polymorphic BRCA variants transmission in breast cancer familial members. Breast Cancer Res Treat. 125:651–657. 2011. View Article : Google Scholar

21 

Levine AJ and Oren M: The first 30 years of p53: Growing ever more complex. Nat Rev Cancer. 9:749–758. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Leroy B, Girard L, Hollestelle A, Minna JD, Gazdar AF and Soussi T: Analysis of TP53 mutation status in human cancer cell lines: A reassessment. Hum Mutat. 35:756–765. 2014. View Article : Google Scholar

23 

Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al: COSMIC: Mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res. 39:(Database Issue). D945–D950. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Harris CC and Hollstein M: Clinical implications of the p53 tumor-suppressor gene. N Engl J Med. 329:1318–1327. 1993. View Article : Google Scholar : PubMed/NCBI

25 

Vogelstein B and Kinzler KW: Cancer genes and the pathways they control. Nat Med. 10:789–799. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Marchenko ND, Zaika A and Moll UM: Death signal-induced localization of p53 protein to mitochondria: A potential role in apoptotic signaling. J Biol Chem. 275:16202–16212. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Chipuk JE, Bouchier-Hayes L, Kuwana T, Newmeyer DD and Green DR: PUMA couples the nuclear and cytoplasmic proapoptotic function of p53. Science. 309:1732–1735. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, et al: A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell. 119:591–602. 2004. View Article : Google Scholar

29 

Miedl H, Lebhard J, Ehart L and Schreiber M: Association of the MDM2 SNP285 and SNP309 genetic variants with the risk, age at onset and prognosis of breast cancer in Central European women: A hospital-based case-control study. Int J Mol Sci. 20:5092019. View Article : Google Scholar

30 

Estiar MA and Mehdipour P: ATM in breast and brain tumors: A comprehensive review. Cancer Biol Med. 15:210–227. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Børresen-Dale AL: TP53 and breast cancer. Hum Mutat. 21:292–300. 2003. View Article : Google Scholar

32 

Grochola LF, Zeron-Medina J, Mériaux S and Bond GL: Single-nucleotide polymorphisms in the p53 signaling pathway. Cold Spring Harb Perspect Biol. 2:a0010322010. View Article : Google Scholar

33 

Whibley C, Pharoah PD and Hollstein M: p53 polymorphisms: Cancer implications. Nat Rev Cancer. 9:95–107. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Vannini I, Zoli W, Tesei A, Rosetti M, Sansone P, Storci G, Passardi A, Massa I, Ricci M, Gusolfino D, et al: Role of p53 codon 72 arginine allele in cell survival in vitro and in the clinical outcome of patients with advanced breast cancer. Tumor Biol. 29:145–151. 2008. View Article : Google Scholar

35 

Toyama T, Zhang Z, Nishio M, Hamaguchi M, Kondo N, Iwase H, Iwata H, Takahashi S, Yamashita H and Fujii Y: Association of TP53 codon 72 polymorphism and the outcome of adjuvant therapy in breast cancer patients. Breast Cancer Res. 9:R342007. View Article : Google Scholar : PubMed/NCBI

36 

Lazar V, Hazard F, Bertin F, Janin N, Bellet D and Bressac B: Simple sequence repeat polymorphism within the p53 gene. Oncogene. 8:1703–1705. 1993.PubMed/NCBI

37 

Peller S, Kopilova Y, Slutzki S, Halevy A, Kvitko K and Rotter V: A Novel Polymorphism in Intron 6 of the Human p53 Gene: A possible association with cancer predisposition and susceptibility. DNA Cell Biol. 14:983–990. 1995. View Article : Google Scholar

38 

Schmidt MK, Reincke S, Broeks A, Braaf LM, Hogervorst FB, Tollenaar RA, Johnson N, Fletcher O, Peto J, Tommiska J, et al: Do MDM2 SNP309 and TP53 R72P interact in breast cancer susceptibility? A large pooled series from the breast cancer association consortium. Cancer Res. 67:9584–9590. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Gonçalves ML, Borja SM, Cordeiro JA, Saddi VA, Ayres FM, Vilanova-Costa CA and Silva AM: Association of the TP53 codon 72 polymorphism and breast cancer risk: A meta-analysis. Springerplus. 3:7492014. View Article : Google Scholar

40 

Cheng H, Ma B, Jiang R, Wang W, Guo H, Shen N, Li D, Zhao Q, Wang R, Yi P, et al: Individual and combined effects of MDM2 SNP309 and TP53 Arg72Pro on breast cancer risk: An updated meta-analysis. Mol Biol Rep. 39:9265–9274. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Dumont P, Leu JIJ, Della Pietra AC III, George DL and Murphy M: The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet. 33:357–365. 2003. View Article : Google Scholar

42 

Thomas M, Kalita A, Labrecque S, Pim D, Banks L and Matlashewski G: Two polymorphic variants of wild-type p53 differ biochemically and biologically. Mol Cell Biol. 19:1092–1100. 1999. View Article : Google Scholar

43 

Pim D and Banks L: P53 polymorphic variants at codon 72 exert different effects on cell cycle progression. Int J Cancer. 108:196–199. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Siddique M and Sabapathy K: Trp53-dependent DNA-repair is affected by the codon 72 polymorphism. Oncogene. 25:3489–3500. 2006. View Article : Google Scholar : PubMed/NCBI

45 

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. View Article : Google Scholar

46 

Zhang Y and Lozano G: P53: Multiple facets of a rubik's cube. Annu Rev Cancer Biol. 1:185–201. 2017. View Article : Google Scholar

47 

Walerych D, Napoli M, Collavin L and Del Sal G: The rebel angel: Mutant p53 as the driving oncogene in breast cancer. Carcinogenesis. 33:2007–2017. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Cossu-Rocca P, Orrù S, Muroni MR, Sanges F, Sotgiu G, Ena S, Pira G, Murgia L, Manca A, Uras MG, et al: Analysis of PIK3CA mutations and activation pathways in triple negative breast cancer. PLoS One. 10:e01417632015. View Article : Google Scholar : PubMed/NCBI

49 

Huun J, Gansmo LB, Mannsåker B, Iversen GT, Sommerfelt-Pettersen J, Øvrebø JI, Lønning PE and Knappskog S: The functional roles of the MDM2 splice variants P2-MDM2-10 and MDM2-∆5 in breast cancer cells. Transl Oncol. 10:806–817. 2017. View Article : Google Scholar

50 

Bond G, Hu W and Levine A: MDM2 is a central node in the p53 pathway: 12 years and counting. Curr Cancer Drug Targets. 5:3–8. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Isakova J, Talaibekova E, Aldasheva N, Vinnikov D and Aldashev A: The association of polymorphic markers Arg399Gln of XRCC1 gene, Arg72Pro of TP53 gene and T309G of MDM2 gene with breast cancer in Kyrgyz females. BMC Cancer. 17:7582017. View Article : Google Scholar : PubMed/NCBI

52 

Gao J, Kang AJ, Lin S, Dai ZJ, Zhang SQ, Liu D, Zhao Y, Yang PT, Wang M and Wang XJ: Association between MDM2 rs 2279744 polymorphism and breast cancer susceptibility: A meta-analysis based on 9,788 cases and 11,195 controls. Ther Clin Risk Manag. 10:269–277. 2014.PubMed/NCBI

53 

Yilmaz M, Tas A, Donmez G, Kacan T and Silig Y: Significant association of the MDM2 T309G polymorphism with breast cancer risk in a Turkish Population. Asian Pac J Cancer Prev. 19:1059–1062. 2018.PubMed/NCBI

54 

Wilkening S, Bermejo JL and Hemminki K: MDM2 SNP309 and cancer risk: A combined analysis. Carcinogenesis. 28:2262–2267. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Gatti RA, Berkel I, Boder E, Braedt G, Charmley P, Concannon P, Ersoy F, Foroud T, Jaspers NG, Lange K, et al: Localization of an ataxia-telangiectasia gene to chromosome 11q22-23. Nature. 336:577–580. 1988. View Article : Google Scholar : PubMed/NCBI

56 

Kruse JP and Gu W: SnapShot: p53 posttranslational modifications. Cell. 133:930–30.e1. 2008. View Article : Google Scholar

57 

Chen L, Gilkes DM, Pan Y, Lane WS and Chen J: ATM and Chk2-dependent phosphorylation of MDMX contribute to p53 activation after DNA damage. EMBO J. 24:3411–3422. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Lee JH and Paull TT: Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene. 26:7741–7748. 2007. View Article : Google Scholar : PubMed/NCBI

59 

González-Hormazábal P, Bravo T, Blanco R, Valenzuela CY, Gómez F, Waugh E, Peralta O, Ortuzar W, Reyes JM and Jara L: Association of common ATM variants with familial breast cancer in a South American population. BMC Cancer. 8:1172008. View Article : Google Scholar

60 

Heikkinen K, Rapakko K, Karppinen SM, Erkko H, Nieminen P and Winqvist R: Association of common ATM polymorphism with bilateral breast cancer. Int J Cancer. 116:69–72. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Zhao L, Yin XX, Qin J, Wang W and He XF: Association between the TP53 polymorphisms and breast cancer risk: An updated meta-analysis. Front Genet. 13:8074662022. View Article : Google Scholar

62 

Diakite B, Kassogue Y, Dolo G, Wang J, Neuschler E, Kassogue O, Keita ML, Traore CB, Kamate B, Dembele E, et al: p.Arg72Pro polymorphism of P53 and breast cancer risk: A meta-analysis of case-control studies. BMC Med Genet. 21:2062020. View Article : Google Scholar

63 

Diakite B, Kassogue Y, Dolo G, Kassogue O, Keita ML, Joyce B, Neuschler E, Wang J, Musa J, Traore CB, et al: Association of PIN3 16-bp duplication polymorphism of TP53 with breast cancer risk in Mali and a meta-analysis. BMC Med Genet. 21:1422020. View Article : Google Scholar

64 

Jalilvand A, Yari K, Aznab M, Rahimi Z, Salahshouri Far I and Mohammadi P: A case-control study on the SNP309T → G and 40-bp Del1518 of the MDM2 gene and a systematic review for MDM2 polymorphisms in the patients with breast cancer. J Clin Lab Anal. 34:e235292020. View Article : Google Scholar

65 

Vodolazhsky DI, Mayakovskaya AV, Kubyshkin AV, Aliev KA and Fomochkina II: Clinical significance of gene polymorphisms for hereditary predisposition to breast and ovarian cancer (review of literature). Klin Lab Diagn. 66:760–767. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Chiang CWK, Marcus JH, Sidore C, Biddanda A, Al-Asadi H, Zoledziewska M, Pitzalis M, Busonero F, Maschio A, Pistis G, et al: Genomic history of the Sardinian population. Nat Genet. 50:1426–1434. 2018. View Article : Google Scholar

67 

Osorio A, Martínez-Delgado B, Pollán M, Cuadros M, Urioste M, Torrenteras C, Melchor L, Díez O, De La Hoya M, Velasco E, et al: A haplotype containing the p53 polymorphisms Ins16bp and Arg72Pro modifies cancer risk in BRCA2 mutation carriers. Hum Mutat. 27:242–248. 2006. View Article : Google Scholar

68 

González JR, Armengol L, Solé X, Guinó E, Mercader JM, Estivill X and Moreno V: SNPassoc: An R package to perform whole genome association studies. Bioinformatics. 23:644–645. 2007. View Article : Google Scholar

69 

Schlesselman JJ: Basic methods of analysis. Case-Control Studies: Design, Conduct, Analysis: Design, Conduct, Analysis. Oxford University Press; Oxford, UK: pp. 1761982

70 

1000 Genomes Project Consortium, . Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA, et al: A global reference for human genetic variation. Nat. 526:68–74. 2015. View Article : Google Scholar

71 

De Vecchi G, Verderio P, Pizzamiglio S, Manoukian S, Bernard L, Pensotti V, Volorio S, Ravagnani F, Radice P and Peterlongo P: The p53 Arg72Pro and Ins16bp polymorphisms and their haplotypes are not associated with breast cancer risk in BRCA-mutation negative familial cases. Cancer Detect Prev. 32:140–143. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Gohari-Lasaki S, Gharesouran J, Ghojazadeh M, Montazeri V and Mohaddes Ardebili SM: Lack of influence of TP53 Arg72Pro and 16bp duplication polymorphisms on risk of breast cancer in iran. Asian Pacific J Cancer Prev. 16:2971–2974. 2015. View Article : Google Scholar

73 

Eskandari-Nasab E, Hashemi M, Amininia S, Ebrahimi M, Rezaei M and Hashemi SM: Effect of TP53 16-bp and β-TrCP 9-bp INS/DEL polymorphisms in relation to risk of breast cancer. Gene. 568:181–185. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Costa S, Pinto D, Pereira D, Rodrigues H, Cameselle-Teijeiro J, Medeiros R and Schmitt F: Importance of TP53 codon 72 and intron 3 duplication 16bp polymorphisms in prediction of susceptibility on breast cancer. BMC Cancer. 8:322008. View Article : Google Scholar : PubMed/NCBI

75 

Ayoubi SE, Elkarroumi M, El Khachibi M, Hassani Idrissi H, Ayoubi H, Ennachit S, Arazzakou M and Nadifi S: The 72Pro variant of the tumor protein 53 is associated with an increased breast cancer risk in the Moroccan Population. Pathobiology. 85:247–253. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Hu Z, Li X, Qu X, He Y, Ring BZ, Song E and Su L: Intron 3 16 bp duplication polymorphism of TP53 contributes to cancer susceptibility: A meta-analysis. Carcinogenesis. 31:643–647. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Akkiprik M, Sonmez O, Gulluoglu BM, Caglar HB, Kaya H, Demirkalem P, Abacioglu U, Sengoz M, Sav A and Ozer A: Analysis of p53 gene polymorphisms and protein over-expression in patients with breast cancer. Pathol Oncol Res. 15:359–368. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Hrstka R, Coates PJ and Vojtesek B: Polymorphisms in p53 and the p53 pathway: Roles in cancer susceptibility and response to treatment. J Cell Mol Med. 13:440–453. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Hao W, Xu X, Shi H, Zhang C and Chen X: No association of TP53 codon 72 and intron 3 16-bp duplication polymorphisms with breast cancer risk in Chinese Han women: New evidence from a population-based case-control investigation. Eur J Med Res. 23:472018. View Article : Google Scholar : PubMed/NCBI

80 

Morten BC, Chiu S, Oldmeadow C, Lubinski J, Scott RJ and Avery-Kiejda KA: The intron 3 16 bp duplication polymorphism of p53 (rs17878362) is not associated with increased risk of developing triple-negative breast cancer. Breast Cancer Res Treat. 173:727–733. 2019. View Article : Google Scholar

81 

Campbell IG, Eccles DM, Dunn B, Davis M and Leake V: P53 polymorphism in ovarian and breast cancer. Lancet. 347:393–394. 1996. View Article : Google Scholar

82 

Mavridou D, Gornall R, Campbell IG and Eccles DM: TP53 intron 6 polymorphism and the risk of ovarian and breast cancer. Br J Cancer. 77:676–677. 1998. View Article : Google Scholar : PubMed/NCBI

83 

Dahabreh IJ, Schmid CH, Lau J, Varvarigou V, Murray S and Trikalinos TA: Genotype misclassification in genetic association studies of the rs1042522 TP53 (Arg72Pro) polymorphism: A systematic review of studies of breast, lung, colorectal, ovarian, and endometrial cancer. Am J Epidemiol. 177:1317–1325. 2013. View Article : Google Scholar

84 

Liu J, Tang X, Li M, Lu C, Shi J, Zhou L, Yuan Q and Yang M: Functional MDM4 rs4245739 genetic variant, alone and in combination with P53 Arg72Pro polymorphism, contributes to breast cancer susceptibility. Breast Cancer Res Treat. 140:151–157. 2013. View Article : Google Scholar

85 

Sharma S, Sambyal V, Guleria K, Manjari M, Sudan M, Uppal MS, Singh NR, Bansal D and Gupta A: TP53 polymorphisms in sporadic North Indian breast cancer patients. Asian Pacific J Cancer Prev. 15:6871–6879. 2014. View Article : Google Scholar

86 

Vymetalkova V, Soucek P, Kunicka T, Jiraskova K, Brynychova V, Pardini B, Novosadova V, Polivkova Z, Kubackova K, Kozevnikovova R, et al: Genotype and haplotype analyses of TP53 gene in breast cancer patients: Association with risk and clinical outcomes. PLoS One. 10:e01344632015. View Article : Google Scholar : PubMed/NCBI

87 

Perriaud L, Marcel V, Sagne C, Favaudon V, Guédin A, De Rache A, Guetta C, Hamon F, Teulade-Fichou MP, Hainaut P, et al: Impact of G-quadruplex structures and intronic polymorphisms rs17878362 and rs1642785 on basal and ionizing radiation-induced expression of alternative p53 transcripts. Carcinogenesis. 35:2706–2715. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Lai MY, Chang HC, Li HP, Ku CK, Chen PJ, Sheu JC, Huang GT, Lee PH and Chen DS: Splicing mutations of the p53 gene in human hepatocellular carcinoma. Cancer Res. 53:1653–1656. 1993.PubMed/NCBI

89 

Takahashi T, D'Amico D, Chiba I, Buchhagen DL and Minna JD: Identification of intronic point mutations as an alternative mechanism for p53 inactivation in lung cancer. J Clin Invest. 86:363–369. 1990. View Article : Google Scholar

90 

Gemignani F, Moreno V, Landi S, Moullan N, Chabrier A, Gutiérrez-Enríquez S, Hall J, Guino E, Peinado MA, Capella G and Canzian F: A TP53 polymorphism is associated with increased risk of colorectal cancer and with reduced levels of TP53 mRNA. Oncogene. 23:1954–1956. 2004. View Article : Google Scholar : PubMed/NCBI

91 

Biroš E, Kalina I, Kohút A, Štubňa J and Šalagovič J: Germ line polymorphisms of the tumor suppressor gene p53 and lung cancer. Lung Cancer. 31:157–162. 2001. View Article : Google Scholar

92 

Bond GL, Hirshfield KM, Kirchhoff T, Alexe G, Bond EE, Robins H, Bartel F, Taubert H, Wuerl P, Hait W, et al: MDM2 SNP309 accelerates tumor formation in a gender-specific and hormone-dependent manner. Cancer Res. 66:5104–5110. 2006. View Article : Google Scholar : PubMed/NCBI

93 

Jones JS, Gu X, Lynch PM, Rodriguez-Bigas M, Amos CI and Frazier ML: ATM Polymorphism and hereditary nonpolyposis colorectal cancer (HNPCC) age of onset (United States). Cancer Causes Control. 16:749–753. 2005. View Article : Google Scholar

94 

Thorstenson YR, Shen P, Tusher VG, Wayne TL, Davis RW, Chu G and Oefner PJ: Global analysis of ATM polymorphism reveals significant functional constraint. Am J Hum Genet. 69:396–412. 2001. View Article : Google Scholar

95 

Tommiska J, Jansen L, Kilpivaara O, Edvardsen H, Kristensen V, Tamminen A, Aittomäki K, Blomqvist C, Børresen-Dale AL and Nevanlinna H: ATM variants and cancer risk in breast cancer patients from Southern Finland. BMC Cancer. 6:2092006. View Article : Google Scholar : PubMed/NCBI

96 

Einarsdóttir K, Rosenberg LU, Humphreys K, Bonnard C, Palmgren J, Li Y, Li Y, Chia KS, Liu ET, Hall P, et al: Comprehensive analysis of the ATM, CHEK2 and ERBB2 genes in relation to breast tumour characteristics and survival: A population-based case-control and follow-up study. Breast Cancer Res. 8:R672006. View Article : Google Scholar

97 

Lavin MF, Birrell G, Chen P, Kozlov S, Scott S and Gueven N: ATM signaling and genomic stability in response to DNA damage. Mutat Res. 569:123–132. 2005. View Article : Google Scholar : PubMed/NCBI

98 

Veronesi U, Goldhirsch A, Boyle P, Orecchia R and Viale G: Breast Cancer. Discov Med. 5:271–277. 2005.PubMed/NCBI

99 

Collaborative Group on Hormonal Factors in Breast Cancer, . Breast cancer and breastfeeding: Collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 360:187–195. 2002. View Article : Google Scholar

100 

World Health Organization (WHO), . IARC Monographs on the Identification of Carcinogenic Hazards to Humans, List of classifications by cancer sites with sufficient or limited evidence in humans. IARC Monographs Volumes 1–132. https://monographs.iarc.who.int/wp-content/uploads/2019/07/Classifications_by_cancer_site.pdfFebruary 1–2022

101 

Horn J, Åsvold BO, Opdahl S, Tretli S and Vatten LJ: Reproductive factors and the risk of breast cancer in old age: A Norwegian cohort study. Breast Cancer Res Treat. 139:237–243. 2013. View Article : Google Scholar

102 

Lagerlund M, Sontrop JM and Zackrisson S: Do reproductive and hormonal risk factors for breast cancer associate with attendance at mammography screening? Cancer Causes Control. 24:1687–1694. 2013. View Article : Google Scholar

103 

Fioretti F, Tavani A, Bosetti C, La Vecchia C, Negri E, Barbone F, Talamini R and Franceschi S: Risk factors for breast cancer in nulliparous women. Br J Cancer. 79:1923–1928. 1999. View Article : Google Scholar : PubMed/NCBI

104 

Akram M, Iqbal M, Daniyal M and Khan AU: Awareness and current knowledge of breast cancer. Biol Res. 50:332017. View Article : Google Scholar : PubMed/NCBI

105 

Trentham-Dietz A, Newcomb PA, Egan KM, Titus-Ernstoff L, Baron JA, Storer BE, Stampfer M and Willett WC: Weight change and risk of postmenopausal breast cancer (United States). Cancer Causes Control. 11:533–542. 2000. View Article : Google Scholar

106 

Miller ER, Wilson C, Chapman J, Flight I, Nguyen AM, Fletcher C and Ramsey I: Connecting the dots between breast cancer, obesity and alcohol consumption in middle-aged women: Ecological and case control studies. BMC Public Health. 18:4602018. View Article : Google Scholar : PubMed/NCBI

107 

Alegre MM, Knowles MH, Robison RA and O'Neill KL: Mechanics behind breast cancer prevention-focus on obesity, exercise and dietary fat. Asian Pacific J Cancer Prev. 14:2207–2212. 2013. View Article : Google Scholar

108 

Zeng H, Irwin ML, Lu L, Risch H, Mayne S, Mu L, Deng Q, Scarampi L, Mitidieri M, Katsaros D and Yu H: Physical activity and breast cancer survival: An epigenetic link through reduced methylation of a tumor suppressor gene L3MBTL1. Breast Cancer Res Treat. 133:127–135. 2012. View Article : Google Scholar

109 

Jardé T, Perrier S, Vasson MP and Caldefie-Chézet F: Molecular mechanisms of leptin and adiponectin in breast cancer. Eur J Cancer. 47:33–43. 2011. View Article : Google Scholar

110 

Siiteri PK: Adipose tissue as a source of hormones. Am J Clin Nutr. 45 (1 Suppl):S277–S282. 1987. View Article : Google Scholar

111 

Zimta AA, Tigu AB, Muntean M, Cenariu D, Slaby O and Berindan-Neagoe I: Molecular links between central obesity and breast cancer. Int J Mol Sci. 20:53642019. View Article : Google Scholar

112 

Lynch BM, Neilson HK and Friedenreich CM: Physical activity and breast cancer prevention. Recent Results Cancer Res. 186:13–42. 2011. View Article : Google Scholar : PubMed/NCBI

113 

Wu Y, Zhang D and Kang S: Physical activity and risk of breast cancer: A meta-analysis of prospective studies. Breast Cancer Res Treat. 137:869–882. 2013. View Article : Google Scholar

114 

Lee J: A meta-analysis of the association between physical activity and breast cancer mortality. Cancer Nurs. 42:271–285. 2019. View Article : Google Scholar : PubMed/NCBI

115 

McTiernan A, Kooperberg C, White E, Wilcox S, Coates R, Adams-Campbell LL, Woods N and Ockene J; Women's Health Initiative Cohort Study, : Recreational physical activity and the risk of breast cancer in postmenopausal women: The women's health initiative cohort study. JAMA. 290:1331–1336. 2003. View Article : Google Scholar : PubMed/NCBI

116 

Lagerros YT, Hsieh SF and Hsieh CC: Physical activity in adolescence and young adulthood and breast cancer risk: A quantitative review. Eur J Cancer Prev. 13:5–12. 2004. View Article : Google Scholar : PubMed/NCBI

117 

Hankinson SE, Colditz GA and Willett WC: Towards an integrated model for breast cancer etiology: The lifelong interplay of genes, lifestyle, and hormones. Breast Cancer Res. 6:213–218. 2004. View Article : Google Scholar : PubMed/NCBI

118 

Hartmann LC, Sellers TA, Frost MH, Lingle WL, Degnim AC, Ghosh K, Vierkant RA, Maloney SD, Pankratz VS, Hillman DW, et al: Benign breast disease and the risk of breast cancer. N Engl J Med. 353:229–237. 2005. View Article : Google Scholar : PubMed/NCBI

119 

Key TJ, Verkasalo PK and Banks E: Epidemiology of breast cancer. Lancet Oncol. 2:133–140. 2001. View Article : Google Scholar

120 

Collaborative Group on Hormonal Factors in Breast Cancer, . Familial breast cancer: Collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet. 358:1389–1399. 2001. View Article : Google Scholar

121 

McPherson K, Steel CM and Dixon JM: ABC of breast diseases: Breast cancer-Epidemiology, risk factors, and genetics. BMJ. 321:624–628. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2022
Volume 24 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Floris M, Pira G, Castiglia P, Idda ML, Steri M, De Miglio MR, Piana A, Cossu A, Azara A, Arru C, Arru C, et al: Impact on breast cancer susceptibility and clinicopathological traits of common genetic polymorphisms in <em>TP53</em>, <em>MDM2</em> and <em>ATM</em> genes in Sardinian women. Oncol Lett 24: 331, 2022
APA
Floris, M., Pira, G., Castiglia, P., Idda, M.L., Steri, M., De Miglio, M.R. ... Muroni, M.R. (2022). Impact on breast cancer susceptibility and clinicopathological traits of common genetic polymorphisms in <em>TP53</em>, <em>MDM2</em> and <em>ATM</em> genes in Sardinian women. Oncology Letters, 24, 331. https://doi.org/10.3892/ol.2022.13451
MLA
Floris, M., Pira, G., Castiglia, P., Idda, M. L., Steri, M., De Miglio, M. R., Piana, A., Cossu, A., Azara, A., Arru, C., Deiana, G., Putzu, C., Sanna, V., Carru, C., Serra, A., Bisail, M., Muroni, M. R."Impact on breast cancer susceptibility and clinicopathological traits of common genetic polymorphisms in <em>TP53</em>, <em>MDM2</em> and <em>ATM</em> genes in Sardinian women". Oncology Letters 24.4 (2022): 331.
Chicago
Floris, M., Pira, G., Castiglia, P., Idda, M. L., Steri, M., De Miglio, M. R., Piana, A., Cossu, A., Azara, A., Arru, C., Deiana, G., Putzu, C., Sanna, V., Carru, C., Serra, A., Bisail, M., Muroni, M. R."Impact on breast cancer susceptibility and clinicopathological traits of common genetic polymorphisms in <em>TP53</em>, <em>MDM2</em> and <em>ATM</em> genes in Sardinian women". Oncology Letters 24, no. 4 (2022): 331. https://doi.org/10.3892/ol.2022.13451