Influence of mutation type on prognostic and predictive values of TP53 status in primary breast cancer patients

  • Authors:
    • Petr Dobes
    • Jan Podhorec
    • Oldrich  Coufal
    • Andrea  Jureckova
    • Katarina  Petrakova
    • Borivoj  Vojtesek
    • Roman Hrstka
  • View Affiliations

  • Published online on: July 21, 2014     https://doi.org/10.3892/or.2014.3346
  • Pages: 1695-1702
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

High rates of mutation in the TP53 tumor suppressor gene have been found in many human cancers, including breast tumors, making p53 one of the most studied proteins in oncology. However, the prognostic and predictive value of alterations in this gene remains ambiguous. To analyze the clinical value of somatic TP53 mutations, we collected clinical and molecular data on 210 women with primary breast cancer. We found significant associations of p53 mutations with tumor grade, metastasis, molecular subtype, Her2 status and inverse correlations with estrogen and progesterone receptor status. Cox proportional hazard analysis confirmed a strong prognostic value of p53 mutation for overall survival rate and highlighted significant interactions with lymph node involvement and tumor size. In relation to treatment options, TP53 mutations were associated with poor response to anthracyclines and radiotherapy. Categorization of TP53 mutations according to their type and location revealed that patients with nonsense mutation have the poorest prognosis in comparison with wild-type cases and other types of mutations in this gene. Classification of TP53 mutations with respect to the degree of disturbance of protein structure showed association of disruptive mutations with poorer patients' outcome in contrast to wild-type and non-disruptive mutations. In conclusion, the present study confirms p53 as a potential predictive and prognostic factor in oncology practice and highlights the growing evidence that distinct types of mutations have different clinical impacts.

Introduction

Breast cancer is the most frequent invasive tumor diagnosed in women, showing growing incidence primarily in advanced countries. In the Czech Republic the average incidence of breast cancer in the population of women during the period 2005–2010 was 65.9 cases per 100,000, with an average mortality of 16.4. In contrast, in the years 1985–1990 the average incidence was 42.9, with average mortality 19.7 (data retrieved from The Czech National Cancer Registry) (1). As indicated, although the incidence has increased by more than one-third in the respective time periods, mainly due to improved screening programs and diagnostics, conversely, mortality has decreased due to earlier detection and more effective treatment. A similar trend is also observed in other European countries; for example, Cancer Research UK reports for the same time periods show an increase in the incidence rate by ~36%, however mortality rate decreased by almost one third (2). Although the success in breast cancer screening, treatment and familial cancer diagnosis highlight the effort of the international cancer research community, the cause of a large majority of breast cancers is unknown and the prospects for curing invasive or chemoresistant breast cancers remains relatively low.

Breast cancer has several important markers with worldwide clinical utility: the estrogen receptor, progesterone receptor and ErbB2 status. These three gene products play an important role in driving breast cancer development and anti-estrogen or anti-ErbB2 therapies are proving to be useful in treating many breast tumors. Nevertheless, successful breast cancer treatment is complicated by the fact that significant chemoresistance and metastasis can obstruct the current treatments. Thus, although therapeutic success is highly related to expression of hormonal receptors, it remains difficult to predict those individuals who will not respond to conventional therapy and/or will show metastatic progression. Significant efforts have been made to link the common oncogenic or tumor suppressor mutations in cancers to predict patient prognosis and/or tumor responses. One of the most studied genes in oncology for the last 35 years is TP53. The product of this gene, p53 protein has been named ‘guardian of the genome’. This protein mediates the response to various forms of genotoxic stress, and is involved in cell cycle regulation (3), DNA repair (4), senescence and apoptosis (3), and substantially prevents cells from malignant transformation. Its role as a tumor suppressor is reflected in its high rate of mutations in human cancers and there have been many studies evaluating the prognostic role of p53 for the outcome of different types of cancer (5). It was shown that genetic alterations leading to p53 inactivation cause abnormal pathological behavior (6) and even that mutations in p53 by dominant negative manner suppress the normal functions of the wild-type (wt) protein (7).

Considerable data indicate that mutant p53 proteins not only lose their tumor suppressive functions, but also gain new oncogenic properties that are independent of wt p53. This ‘gain of function’ received its first support when transfection of mutant p53 into TP53 null cells was shown to enhance their ability to form tumors in mice. Then, a number of studies demonstrated a wide range of oncogenic properties for p53 mutants and provided key mechanistic insights to explain these phenomena (8).

p53 is composed of three domains: i) the trans-activation domain [amino acids (AAs) 1–95], ii) the DNA binding domain (DBD) (AA 102–292), and iii) the oligomerization domain (AA 300–393) (9). The mutation rate of p53 in breast cancer is ~30% (10) and the most common p53 mutations for breast cancer, similar to other types of cancer, are missense mutations in the DBD (11). A closer look at missense mutations shows they can be separated on DNA-binding motif (DBM) mutations involving: i) AAs from L2 and L3 loops (AAs 164–194 and 237–250) and ii) mutations in LSH (loop-sheet- helix) motif (AAs 119–135 and 272–287), and non-DBM mutations including other AAs (12). Missense mutations in DBM were shown to be associated with poorer prognosis (1215) in comparison to missense mutations in non-DBM regions. Another method to sub-divide TP53 mutations that have clinical value is the classification into ‘nondisruptive’ and ‘disruptive’ mutations, which include either truncating mutations or produce dissimilar AAs in the L2 or L3 loops (13).

In the present study, we investigated the role of TP53 mutations for hospital-based study in breast cancer and we correlated them with standard clinicopathological characteristics. Furthermore, we compared specific groups of mutations in TP53 with clinicopathological variables and patient outcome. We found that the prognostic significance of TP53 mutation status in breast cancer is dependent on the specific mutation type, and that nonsense mutations in contrast to mutations in the DNA-binding domain are the most important for estimating the prognosis of breast cancer patients.

Materials and methods

Clinical samples and processing

Our hospital-based study included 210 primary breast tumors with determined p53 status from patients treated at the Masaryk Memorial Cancer Institute (MMCI). The median age of patients was 59 years (mean 59 with standard deviation 12). Samples were collected within 20 min of surgical removal and immediately evaluated by a pathologist according to standardized hospital protocol. Tissue pieces of ~3×3×8 mm were cut from redundant tumor tissue after standard surgicopathological processing and stored in RNA later for 3–5 days at 4°C and then frozen at −80°C. Subsequently, total cellular RNA was extracted using RNeasy Isolation kit (Qiagen) according to the manufacturer’s instructions. RNA concentration and purity were controlled by UV spectrophotometry (A260:A280>2.0; A260:A230>1.8). RNA integrity was checked using Agilent 2100 Bioanalyzer and only non-degraded RNA characterized by RNA integrity number (RIN) ≥7 with no DNA contamination signs was processed. TP53 mRNA was then amplified using the SuperScript™ III One-Step RT-PCR System with Platinum® Taq High Fidelity (Invitrogen), as previously described (14). PCR products were purified by MinElute™ PCR Purification kit (Qiagen) and sequenced using the ABI PRISM BigDye® Terminator v3.1 Cycle Sequencing Kit on an ABI 3130 genetic analyzer (Applied Biosystems).

The main clinicopathological variables including tumor type, grade and nuclear grade, were determined according to Elston and Ellis (15); estrogen receptor (ER), progesterone receptor (PR) and Her2/neu status were extracted from pathological records obtained from the MMCI database. The tumors were also categorized according to IHC subtype with respect to ER, PR, Her2 and Ki67-expression. Her2 positivity was primarily determined by immunohistochemistry and in equivocal cases Her2 status was analyzed for gene amplification by FISH.

Patients and evaluation and treatment regimens

The present study was approved by the local ethics committee in the MMCI and informed consent was obtained from each patient. Clinical data including patient follow-up were evaluated by an oncologist from the hospital’s patient records. The initial staging consisted of chest X-rays, radionuclide bone scans, abdominal ultrasound or computed tomography, bilateral mammography and determination of carcinoembryonic antigen (CEA) and CA15–3 (Mucin 1). All patients lacked evidence of distant metastases at the time of surgery. Primary treatment followed approximately up to one month post-surgery (partial/total mastectomy) and was administered with respect to clinicopathological parameters. With respect to other clinical indications, hormonal therapy was applied either in monotherapy or in combination with other treatment modalities. Adjuvant chemotherapy was administered with respect to expected response to systematic treatment according to standard protocols at MMCI and mainly consisted of a combination of taxanes, anthracyclines and cyclophosphamide. In total, 156 patients received radiation following completion of chemotherapy.

Classification of TP53 mutations

Missense mutations represent single base-pair substitutions that result in the translation of a different AA in that position in the context of the full-length protein. Nonsense mutations are created by single base substitutions (stop codon) or small insertions and deletions leading to a frameshift and creating a truncated protein. TP53 mutations can also be categorized according to other criteria such as the location of the mutation and the predicted AA alterations. Following a study by Poeta et al (13), we also classified mutations as disruptive and nondisruptive, where disruptive mutations are DNA sequence alterations that i) introduce a stop codon or ii) occur within the L2, or L3 binding domains and replace an AA from one polarity/charge category with an AA from another category. Conversely, nondisruptive mutation is any mutation occurring outside the L2 or L3 binding domain (except stop mutations) or mutations within the L2 or L3 binding domains that result in replacement of an AA with another from the same polarity/charge category.

Statistical analysis

Statistical analysis was performed using IBM SPSS Statistics 20.0. χ2 test was used to evaluate associations of molecular and clinicopathological variables with TP53 status. Both disease-free survival (DFS) and overall survival (OS) curves were generated by the Kaplan-Meier method. DFS was defined as the time from the date of surgery to the date of death or relapse of disease. OS was defined as the time from diagnosis to death or last record. Patients who had not died or who were lost to follow-up were censored when they were last known to be alive. The log-rank test was used for comparison of statistical significance for differences among the survival curves for both DFS and OS. A value of p<0.05 was evaluated as statistically significant. Multivariate Cox regression models with backward selection were used to test the prognostic effect of TP53 status after adjusting for other important prognostic variables.

Results

Relationship between p53 status and clinicopathological variables

Mutational status of p53 was determined by direct sequencing in a group of 210 breast cancer patients. In parallel all clinicopathological variables available from patient records were statistically evaluated in relation to p53 status. The frequency of p53 mutations in our cohort of breast cancer patients was 25.7% which is consistent with other reports (10). Cases with mutations that did not change the resulting AA (silent mutations) were counted as wt p53. Table I shows the associations of clinicopathological parameters with p53 status. We found that mutant p53 was significantly associated with tumor grade (p<0.001), metastasis (p=0.016), molecular subtype (p<0.001), Her2 status (p=0.004) and inversely correlated with ER- and PR-positivity (p<0.001 for both). In parallel, the proportions of TP53 missense, nonsense and silent mutations were determined and calculated with respect to clinicopathological variables (Table I). No significant changes were observed in statistical associations of clinicopathological parameters and type of p53 mutations.

Table I

TP53 mutations in relation to clinicopathological parameters.

Table I

TP53 mutations in relation to clinicopathological parameters.

ParametersNWild-type p53Mutant p53P-valueMissense mutationNonsense mutationSilent mutationWild-typeP-value
Tumor histology0.0790.229
 Ductal NOS155110453788102
 Other12931209
 Lobular2926312125
 Medullary5232102
 Tubular3300003
 Total2041505441139141
Tumor grade<0.001<0.001
 G16966321264
 G24237523136
 G3985246379745
 Total20915554411310145
Tumor size0.4680.616
 pT11269729245691
 pT2674720146443
 pT31510532010
 Total20815454411310144
Nodal status0.9940.861
 pN0856322184558
 pN11208931238584
 Total20515253411210142
Metastasis0.0160.017
 No174135393278127
 Yes36211596219
 Total21015654411310146
Duplicity0.5440.349
 01781354333109126
 11711633011
 213944018
 Total20815553401310145
ER status<0.001<0.001
 ER451926215316
 ER+164136282087129
 Total20915554411310145
PR status<0.001<0.001
 PR562927225425
 PR+151124271986118
 Total20715354411310143
Her2 status0.0040.004
 Her21611263524117119
 Her2+442519172322
 Total20515154411310141
IHC based subtype<0.001<0.001
 ER Her2+14598114
 ER+ Her2 Ki67a low6359422356
 ER+ Her2 Ki67 not low523715105235
 ER+ Her2+30201091218
 Triple-negative271215114210
 Total18613353401310123

a Ki67 threshold was 15%.

Mutations in p53 and prognosis

To study the impact of p53 mutational status on patient outcome, DFS and OS were determined (Fig. 1). We found that the presence of mutations resulting in change in p53 protein significantly decreased both DFS and OS (p=0.015 and p<0.001, respectively). Grade, tumor size, lymph node involvement, metastasis and Her2 status also showed significant impact on both DFS and OS (data not shown).

Multivariate analysis including the most important patient characteristics (histological type, grade, pT, pN, ER, PR, Her2, p53 status), identified only p53 mutational status, pT (p=0.013) and pN as statistically significant factors for DFS (Table II), indicating that the presence of p53 mutation has independent prognostic value. Only p53 mutational status and pT showed statistical significance in relation to OS (Table II).

Table II

Multivariate Cox proportional hazards models of disease-free survival and overall survival.

Table II

Multivariate Cox proportional hazards models of disease-free survival and overall survival.

DFSHR (95% CI)P-valueOSHR (95% CI)P-value
p53 protein statusp53 protein status
Wild-type p531Wild-type p531
Mutant p532.453 (1.219–4.935)0.012Mutant p535.378 (2.138–13.524)0.000
Tumor size (pT)Tumor size (pT)
 pT11pT11
 pT21.890 (0.887–4.029)0.099pT22.947 (1.067–8.135)0.037
 pT34.755 (1.655–13.658)0.004pT311.238 (3.094–40.813)0.000
Nodal status
 Negative1
 Positive2.890 (1.165–7.166)0.022

[i] CI, confidence interval; HR, hazard ratio; DFS, disease-free survival; OS, overall survival.

Clinical utility of p53 status

Retrospective analysis of the impact of p53 mutations on patient response to first round adjuvant therapy revealed significantly better DFS of patients with wt p53 (p=0.046; Fig. 2A). On the other hand, p53 mutations had no effect on DFS in patients who did not receive adjuvant treatment (p=0.235; Fig. 2B). Notably, the presence of p53 mutations showed significant association with poorer OS for both treated and untreated patients (Fig. 2C and D).

The predictive values of p53 status were also retrospectively analyzed in relation to individual specific treatment options represented by anthracyclines, taxanes, tamoxifen, trastuzumab and radiotherapy. Better outcome was found in wt p53 patients treated with anthracyclines (p=0.060) and patients who underwent radiotherapy (p=0.002) compared to patients with p53 mutation (Fig. 3).

The effect of different types of TP53 mutations

Since our data clearly demonstrated the impact of TP53 mutations on prognosis of breast cancer patients and their treatment response, we focused only on cases with mutation in TP53 gene to categorize mutations according to type and location. In this group of 63 tumors, 63.5% contained missense mutations, 20.6% nonsense mutations and 15.9% silent mutations. Kaplan-Meier analysis clearly showed that patients with nonsense mutation have the poorest prognosis in comparison with other types of mutants, as well as wt cases (Fig. 4A and B). No significant differences in either DFS and OS were found when comparing missense mutations to DBM (L2/L3 and LSH) with missense mutations outside of the DBM (Fig. 4C and D). Patients with missense TP53 mutations in DBM showed significantly poorer OS compared to breast cancer patients bearing wt TP53 (p=0.016), while patients with missense mutations outside of p53 DBM showed only marginal significance (p=0.077) (Fig. 4D).

Mutations were also classified into two groups, disruptive and nondisruptive, according to the degree of disturbance of protein structure predicted from the crystal structure of the p53-DNA complexes as described previously in patients with squamous-cell carcinoma of the head and neck (13). The comparison of disruptive and nondisruptive p53 mutations showed no statistically significant impact on patient DFS as well as OS (Fig. 5). On the other hand, in comparison with wt p53, only disruptive mutations in case of DFS (p=0.015) and both disruptive and nondisruptive p53 mutations for OS (p=0.009 and p<0.001) showed significantly poorer outcome (Fig. 5).

Discussion

Breast cancer represents the most common female malignancy, responsible for over 400,000 deaths yearly worldwide. This is a disease with a complex, heterogeneous genetic and biochemical background and no single genomic or metabolic condition may be considered as decisive for its formation and progression. However, a few key factors can be pointed out including TP53 tumor suppressor gene. Mutant p53 is known to affect multiple oncogenic processes during breast tumorigenesis including tumor formation and development, growth and metastasis. Due to loss of tumor suppressor function and pro-oncogenic properties of mutant p53, many studies focused on analysis of TP53 mutations as a prognostic and/or predictive biomarker (8). Mutations in the TP53 gene were found to be more frequent in high-grade, large-size, node-positive breast tumors that typically lack functional ER and PR (12). In accordance to these findings, recent analysis of ~2,000 breast tumors found the lowest frequency of TP53 mutations in luminal subtype A ~5%, followed by subtypes luminal B ~13%, Her2 ~22% and basal-like ~34% (16). The present study is compatible with those data (Table I).

To date, only limited information is available on the value of mutant p53 in breast cancer. Since p53 mutations occur mostly in ER- and PR-negative tumors, where chemotherapy usually combined with radiotherapy is applied, it is reasonable to study the impact of p53 status on these treatment regimens. In agreement with our data, several reports demonstrated that breast carcinomas expressing mutant p53 show lower sensitivity to treatment by anthracyclines (10,17,18). On the other hand, treatment by taxanes with respect to p53 status does not exhibit convincing data. Despite promising preliminary data (19,20), a recent large phase-3 clinical trial has shown similar sensitivity to taxanes in breast tumors bearing both wt and mutant p53 (21).

Increasingly more studies are focused on the localization and/or type of TP53 mutations and their impact on patient prognosis. Due to the function of p53 as a transcriptional factor, many studies focus on missense mutations located in DBM (chiefly in the L2/L3 loops) showing association with a poorer outcome compared to mutations outside these motifs (17,22,23). Similarly in a large cohort of unselected breast tumors, Olivier et al found that missense mutations in DBM are associated with poorer survival than missense mutations outside DBM (12). Notably, they also found that truncating (nonsense) mutations compared to missense mutations in DBM have fairly similar prognostic value. However, retrospective study on the prognosis and predictive values of TP53 somatic mutations in the BIG 02–98 randomized phase III trial, led by the same group, concluded that i) missense mutations are not associated with poor survival, ii) discrimination of missense mutations according to location inside DBM and outside DBM does not significantly affect patient outcome, and iii) only truncated (nonsense) mutations may serve as an independent predictor of poor DFS and OS (24). Our findings are consistent with their results showing significantly poorer effect of nonsense p53 mutations on patient outcome in comparison with both all TP53 missense mutations and missense mutations located in the DBM only. Our results are also consistent with a study by Lai et al showing significant association of nonsense mutations with breast cancer-specific mortality (HR with 95% CI: 9.43, 1.29–69.12) (25). Our data is also indirectly supported by Geisler et al showing that mutations in DBM are associated with poorer prognosis; nevertheless, approximately half of the mutations classified in this manner are represented by truncating mutations (17,22). In contrast, categorization of p53 mutations according to the location and the predicted amino acid alterations showed no significant difference in impact on patient survival between disruptive and nondisruptive p53 mutations.

In conclusion, our results in general confirm a negative prognostic value of p53 mutations. However, the limitations of assessing the predictive value of a molecular marker in treatment regimens that combine drugs with different modes of action regarding this marker should be considered. With this in mind, our study demonstrates that clinical outcome for breast cancer patients is associated with different TP53 mutation types and indicates potential utilization of TP53 mutational status to predict response to treatment by anthracyclines and radiotherapy. However, further functional studies are required to clarify the impact of particular p53 mutations on patient outcome and response to specific therapy.

Acknowledgements

This study was supported by the European Regional Development Fund and the State Budget of the Czech Republic RECAMO CZ.1.05/2.1.00/03.0101, MH CZ-DRO (MMCI, 00209805), BBMRI_CZ (LM2010004) and IGA NT13794- 4/2012. The authors thank Dr P.J. Coates for his critical reading of the manuscript.

References

1 

Dušek L, Mužík J, Gelnarová E, Fínek J, Vyzula R and Abrahámová J: Cancer incidence and mortality in the Czech Republic. Klin Onkol. 23:311–324. 2010.

2 

Cancer Research UK. www.cancerresearchuk.orghttps://www.cancerresearchuk.org. Accessed March, 2013

3 

Kastan MB, Canman CE and Leonard CJ: P53, cell cycle control and apoptosis: implications for cancer. Cancer Metastasis Rev. 14:3–15. 1995. View Article : Google Scholar

4 

Kastan MB, Onyekwere O, Sidransky D, Vogelstein B and Craig RW: Participation of p53 protein in the cellular response to DNA damage. Cancer Res. 51:6304–6311. 1991.PubMed/NCBI

5 

Soussi T and Béroud C: Assessing TP53 status in human tumours to evaluate clinical outcome. Nat Rev Cancer. 1:233–239. 2001.

6 

Greenblatt M, Bennett W, Hollstein M and Harris C: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 54:4855–4878. 1994.

7 

Caron de Fromentel C and Soussi T: TP53 tumor suppressor gene: a model for investigating human mutagenesis. Genes Chromosomes Cancer. 4:1–15. 1992.PubMed/NCBI

8 

Brosh R and Rotter V: When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer. 9:701–713. 2009.PubMed/NCBI

9 

Cho Y, Gorina S, Jeffrey PD and Pavletich NP: Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science. 265:346–355. 1994. View Article : Google Scholar : PubMed/NCBI

10 

Bertheau P, Espié M, Turpin E, et al: TP53 status and response to chemotherapy in breast cancer. Pathobiology. 75:132–139. 2008. View Article : Google Scholar

11 

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

12 

Olivier M, Langerød A, Carrieri P, et al: The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer. Clin Cancer Res. 12:1157–1167. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Poeta ML, Manola J, Goldwasser MA, et al: TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med. 357:2552–2561. 2007. View Article : Google Scholar

14 

Hrstka R, Beranek M, Klocova K, Nenutil R and Vojtesek B: Intronic polymorphisms in TP53 indicate lymph node metastasis in breast cancer. Oncol Rep. 22:1205–1211. 2009.PubMed/NCBI

15 

Elston C and Ellis I: Assessment of histological grade. The Breast. 13. Churchill Livingstone; Edinburgh, New York: pp. 356–384. 1998

16 

Curtis C, Shah SP, Chin SF, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 486:346–352. 2012.PubMed/NCBI

17 

Geisler S, Lønning PE, Aas T, et al: Influence of TP53 gene alterations and c-erbB-2 expression on the response to treatment with doxorubicin in locally advanced breast cancer. Cancer Res. 61:2505–2512. 2001.

18 

Chrisanthar R, Knappskog S, Løkkevik E, et al: CHEK2 mutations affecting kinase activity together with mutations in TP53 indicate a functional pathway associated with resistance to epirubicin in primary breast cancer. PLoS One. 3:e30622008. View Article : Google Scholar : PubMed/NCBI

19 

Wahl AF, Donaldson KL, Faircnild C, et al: Loss of normal p53 function confers sensitization to Taxol by increasing G2/M arrest and apoptosis. Nat Med. 2:72–79. 1996. View Article : Google Scholar : PubMed/NCBI

20 

Chrisanthar R, Knappskog S, Løkkevik E, et al: Predictive and prognostic impact of TP53 mutations and MDM2 promoter genotype in primary breast cancer patients treated with epirubicin or paclitaxel. PLoS One. 6:e192492011.PubMed/NCBI

21 

Bonnefoi H, Piccart M, Bogaerts J, et al: TP53 status for prediction of sensitivity to taxane versus non-taxane neoadjuvant chemotherapy in breast cancer (EORTC 10994/BIG 1–00): a randomised phase 3 trial. Lancet Oncol. 12:527–539. 2011. View Article : Google Scholar

22 

Geisler S, Børresen-Dale AL, Johnsen H, et al: TP53 gene mutations predict the response to neoadjuvant treatment with 5-fluorouracil and mitomycin in locally advanced breast cancer. Clin Cancer Res. 9:5582–5588. 2003.

23 

Powell B, Soong R, Iacopetta B, Seshadri R and Smith DR: Prognostic significance of mutations to different structural and functional regions of the p53 gene in breast cancer. Clin Cancer Res. 6:443–451. 2000.PubMed/NCBI

24 

Fernández-Cuesta L, Oakman C, Falagan-Lotsch P, et al: Prognostic and predictive value of TP53 mutations in nodepositive breast cancer patients treated with anthracycline-or anthracycline/taxane-based adjuvant therapy: results from the BIG 02–98 phase III trial. Breast Cancer Res. 14:R702012.

25 

Lai H, Ma F, Trapido E, Meng L and Lai S: Spectrum of p53 tumor suppressor gene mutations and breast cancer survival. Breast Cancer Res Treat. 83:57–66. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October 2014
Volume 32 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dobes P, Podhorec J, Coufal O, Jureckova A, Petrakova K, Vojtesek B and Hrstka R: Influence of mutation type on prognostic and predictive values of TP53 status in primary breast cancer patients. Oncol Rep 32: 1695-1702, 2014
APA
Dobes, P., Podhorec, J., Coufal, O., Jureckova, A., Petrakova, K., Vojtesek, B., & Hrstka, R. (2014). Influence of mutation type on prognostic and predictive values of TP53 status in primary breast cancer patients. Oncology Reports, 32, 1695-1702. https://doi.org/10.3892/or.2014.3346
MLA
Dobes, P., Podhorec, J., Coufal, O., Jureckova, A., Petrakova, K., Vojtesek, B., Hrstka, R."Influence of mutation type on prognostic and predictive values of TP53 status in primary breast cancer patients". Oncology Reports 32.4 (2014): 1695-1702.
Chicago
Dobes, P., Podhorec, J., Coufal, O., Jureckova, A., Petrakova, K., Vojtesek, B., Hrstka, R."Influence of mutation type on prognostic and predictive values of TP53 status in primary breast cancer patients". Oncology Reports 32, no. 4 (2014): 1695-1702. https://doi.org/10.3892/or.2014.3346