Citation

Silva KSF (2019) Genetic Counseling, Polymorphisms and Breast Cancer. J Fam Med Dis Prev 5:098. doi.org/10.23937/2469-5793/1510098

REVIEW ARTICLE | OPEN ACCESS DOI: 10.23937/2469-5793/1510098

Genetic Counseling, Polymorphisms and Breast Cancer

Kleber Santiago Freitas e Silva*

Biological Sciences Institute, Federal University of Goiás, Brazil

Abstract

Breast cancer, a highly penetrant hereditary disorder, is the most common cancer in women worldwide. Approximately 10% of breast cancer cases are hereditary and 15% of patients with invasive breast cancer have a first-degree relative with the same disorder. Genetic counseling has become an important tool of the health care system providing information and support to families at risk of a genetic disorder. Oncology research teams have designed breast cancer screening guidelines for high-risk patients. Along the past decade, several genes have been identified as genetically related to breast cancer inheritance. BRCA1 and BRCA2 are considered the most important genes related to inheritance predisposition of breast cancer, along with PTEN and TP53 [1]. The BRCA1 and BRCA2 structures are different but their functions are interconnected and related to DNA repair. The susceptibility of breast cancer for patients with the BRCA1 mutation is up to 87% for older women. Another gene, TP53, codes for a protein that acts as the guardian of the genome, binds to DNA in order to perform transcriptional regulatory functions, regulation of the cell cycle and apoptosis among other functions. According to genetic counseling and epidemiologic studies, the risk of developing cancer for patients with TP53 polymorphisms is 90%. Regarding the gene PTEN, germline mutations increases the risk of breast cancer and about 80% of patients with breast cancer carry germline mutations in the gene. PTEN is an oncogene and codes for a protein with phosphatase activity, related to the regulation of cell cycle, controlling cells growth and able to promote cell cycle arrest. Currently, genetic counseling endeavor to identify patients at risk of genetic anomalies, study family history and inheritance patterns, calculate risks of recurrence, and provide information regarding testing and treatment procedures. Therefore, breast cancer patients and their families are presented with possibilities of screening for BRCA1, BRCA2, TP53, and PTEN mutations, and preventive care such as chemoprevention and prophylactic surgery.

Keywords

Genetic Counseling, Polymorphisms, Breast Cancer, BRCA1, BRCA2, TP53, PTEN

Introduction

Over the last decade, there were more than 500,000 cases of deaths from breast cancer in the world [2]. Breast cancer is the most common cancer in women worldwide. It has been investigated for more than 2000 years throughout history and its relation to a genetic component was described in 1700 in a young female with breast cancer whose maternal uncle and grandmother had died of the same type of cancer [3].

Breast cancer is a highly penetrant hereditary disorder. Germinal mutations occur in cancer susceptibility genes and is inherited either from the paternal or maternal origin. Approximately 10% of breast cancer cases are hereditary [4] and 15% of patients with invasive breast cancer have a first-degree relative with the same disorder [5]. Genetic counseling has become an important tool of the health care system providing information and support to families at risk of a genetic disorder. Oncology research teams have designed breast cancer screening guidelines for high-risk patients [6-9].

Genetic counseling can lead to the detection of early-stage breast cancer tumors. In Europe, for example, breast cancer screening reduced patient mortality by 48% in patients during the first decade of the twenty-first century [10]. In Italy, breast cancer screening let to a significant survival rate for patients after 5 years from the date of diagnosis [11]. A positive family history for breast cancer is one of the strongest predictors of a patient's risk of developing the disease [12]. Therefore, genetic counseling is routinely provided to patients with increased risk of carrying genetic mutations [13].

Along the past decade, several genes have been identified as genetically related to breast cancer inheritance. The most studied ones are ATM (Ataxia Telangiectasia-Mutated) [14], BRCA1 (Breast cancer gene 1), BRCA2 (Breast Cancer gene 2) [15], BRIP1 (BRCA1 interacting protein C-terminal helicase 1) [16], CHEK2 (Checkpoint kinase 2) [17], PALB2 (Partner and localizer of BRCA2) [18], PTEN (Phosphatase and tensin homolog) [19], TP53 (Tumor Protein p53) [1] and RAD51C (RAD51 paralog C) [20]. In the present review, we explore the involvement of high-penetrance genes (BRAC1, BRCA2, TP53 and PTEN) with breast cancer, familial history of the disease and genetic counseling.

Breast Cancer Gene

The molecular and genetic basis of inherited breast cancer risk started to be unraveled after the discovery of BRCA1 [21,22] and BRCA2 [23] in the 90's. Currently, BRCA1 and BRCA2 [24] are considered the most important genes related to inheritance predisposition of breast cancer, along with PTEN [19] and TP53 [1]. BRAC1 and BRAC2 are oncogenes, also known as caretaker genes, responsible for the error-free repair and maintenance of genomic stability through a homologous recombination process [15].

The BRCA1 gene codes for a protein (BRAC1) that interact with other proteins such as tumor suppressors, DNA damage sensitive proteins and cell signaling protein (Figure 1). The multiprotein complex formed is so-called the BRCA1-associated genome surveillance complex (BASC) [25]. Moreover, BRCA1 interacts with RNA polymerase and histones in order to drive transcriptional and transcriptional regulation [26], cell cycle progression and ubiquitination [27]. The BRCA1 protein contains two main domains. A zinc finger domain related to its ubiquitination function [28] and a BRCA1 C Terminus domain (BRCT) related to its DNA repair function [29].

Figure 1: Schematic view of BRCA1 (chromosome 17) and BRCA2 (chromosome 13) genes and their functions to maintain genomic stability through transcriptional regulation, protein ubiquitination, cell cycle checkpoints and DNA repair. View Figure 1

The BRCA1 and BRCA2 structures are different but their functions are interconnected and related to DNA repair. BRCA2 is key to homologous recombination as its protein takes part in a nucleoprotein complex in order to identify regions of homology in the DNA, recruit partners such as RAD51C and PALB2 [30,31], repair strand breaks and maintain genome stability. BRCA2 has an important role in meiosis, a process of cell division that produces sperm and eggs. During meiosis, chromosomes exchange genetic material and are prone to undergo DNA strand break and BRCA2 is essential for fixing such damage in the DNA. Studies performed in mice have shown that individuals with mutations in BRCA2 have serious growth problems, are sterile and fail to produce spermatozoa [32].

Breast cancer incidence and death rates increase with age. The cumulative incidence of breast cancer is 0.4% and although the disease is not very common in young patients, it is the most frequent type of cancer in women from 15 to 40-years-old [33]. The susceptibility of breast cancer for patients with the BRCA1 mutation is up to 87% for women that are over 70-years-old, with little variation among ethnic groups [34]. Genetic counseling and cancer susceptibility evaluation are of great significance for the diagnosis of breast cancer patients. BRCA1 and BRCA2 mutation screening is performed for at-risk families through genetic counseling. Patients and their family need to comprehend the significance of the counseling in order to make decisions regarding prevention strategies. This way, the outcome of genetic counseling is able to guide clinical decisions and disease management [35].

Tumor Protein p53

The TP53 gene has a sequence that is highly conserved across species [36]. The protein p53 acts as the guardian of the genome, binds to DNA in order to perform transcriptional regulatory functions [37], regulation of the cell cycle, apoptosis [38], inhibition of angiogenesis [39] and maintaining genome integrity [40] (Figure 2). TP53 becomes induced as a response not only to DNA damage, but also oxidative stress, osmotic shock, ribonucleotide levels reduction, and anomaly regarding expression of tumor suppressor genes [41]. TP53 is a tumor suppressor, preventing tumorigenesis [42]. Mutations on the p53 DNA binding motif is an indication of cancer susceptibility and should be investigated through genetic counseling [43]. TP53 is the most frequently mutated gene in cancer. It is related to familial breast cancer, especially when it involves germinal mutations [44].

Figure 2: Schematic view of the TP53 gene functions. The protein p53 when expressed at normal levels recruits partner and regulates several processes that regulate oncogenesis, such as angiogenesis, cell cycle arrest or progression, apoptosis, and oxidative stress. TP53 is activated both by endogenous or exogenous signals. View Figure 2

Polymorphic genes, especially oncogenes, increase the susceptibility to cancer. There is a common polymorphism regarding the TP53 gene, characterized by the substitution of the amino acid arginine for a proline at codon 72. Innumerous studies have tried to find a link between TP53 polymorphism and diseases [45-47], including cancer [48-51]. Alternative methods of cancer treatment have been applied, such as the exogenous p53 transduction [52,53] to increase the levels of the protein and restoration of endogenous wild-type p53 activity premature aging [54]. The former alternative shows some downsides such as premature aging and the latter show promising results since it promotes tumor cells remission without harming surrounding cells and preventing metastases. After extensive investigation, methods such those could be indicated by genetic counselors in order to prevent diseases.

Genetic counseling families at risk of breast cancer due to mutation on TP53 is never a simple task due to the wide likelihood of the clinical anomalies related to it. Moreover, the idiopathic causes may lie on association with other tumor suppressor genes [55]. According to genetic counseling and epidemiologic studies, the risk of developing cancer for patients with TP53 polymorphisms is 90%, and breast cancer is included in that estimative [56]. Women show up to 85% risk of developing breast cancer before they reach the 45 years of age [55,56]. In addition, 20% of TP53 mutation related tumors occur before the age of 20 years [55]. The complexity of TP53 related cancers may bring psychosocial issues for the affected families and genetic counseling may help them with decisions regarding prevention strategies.

Phosphatase and Tensin Homolog

PTEN is an oncogene and codes for a protein with phosphatase activity, related to the regulation of cell cycle, controlling cells growth and able to promote cell cycle arrest [57] (Figure 3). PTEN genetic alterations may lead to tumor onset due to loss of its enzyme product and inability to control cell mitosis and at the same time, those cells show a reduced apoptosis rate. Phosphatidylinositol triphosphate (PIP3) is the substrate of the protein coded by PTEN. PIP3 is a signal molecule during tumorigenesis that takes part in cascade pathways related to cell growth and invasion [58]. PTEN regulates growth down-regulating PIP3 intracellular levels, thus, avoiding growth and invasion of cells that lost control over mitosis [59,60].

Figure 3: Schematic view of factors that may change PTEN expression and alter its functions, consequently leading to a possible tumor cell onset. View Figure 3

Genetic anomalies related to PTEN is found in glioblastoma [61], endometrial [62], prostate [63,64], lung [65,66] and breast cancer [67-69]. Several studies have shown that PTEN is related to the onset and development of breast cancer. The underlying mechanisms involved in breast cancer tumorigenesis due to PTEN mutations include a large variety of genomic processes. The most common ones are germline [70] and somatic mutations [71], epigenetic regulation [72], PTEN protein-protein interactions [73], PTEN protein degradation and post-translational modifications [74,75] that causes repressed expression of the PTEN protein and loss of heterozygosity [76].

Assessment of family history helps to determine the risk of breast cancer. It has been shown that germline mutations in PTEN increases the risk of breast cancer. About 80% of patients with breast cancer carry germline mutations in PTEN [77]. In addition, the rate of PTEN deletion is 40% and the polymorphisms within PTEN sequence is less than 5% [78]. Breast cancer patients with loss of at least one allele of the PTEN gene show very poor prognosis [76,79]. Women with mutation in the PTEN gene have up to 76% risk for benign breast anomalies [80]. Interestingly, males with genetic disorders in PTEN have been diagnosed with breast cancer [77,81].

Genetic counseling is recommended for families with historical cases of PTEN anomalies and breast cancer. PTEN anomalies is clinically assessed by criteria developed by the International Cowden Consortium in 1995 and the National Comprehensive Cancer Network Genetics/High Risk Cancer Surveillance Panel [82]. Genetic counselors normally suggest that women with historical family of PTEN breast cancer should breast self-exam every month and clinical breast examinations should be performed semiannually. In addition, clinical mammography should be performed annually [82,83].

Concluding Remarks

Here, we explored the involvement of high-penetrance genes (BRAC1, BRCA2, TP53 and PTEN) with breast cancer, familial history of the disease and genetic counseling. BRAC1 and BRAC2 are genome caretaker genes, responsible for error-free repair and maintenance of genomic stability. BRCA1 and BRCA2 mutation screening should be performed for at-risk families through genetic counseling and clinical assessment, since mutations affecting them account to a large number of breast cancer. Mutations on the p53 DNA binding motif increase cancer susceptibility and should also be investigated through genetic counseling. Germline mutations in PTEN increases the risk of breast cancer and women with family history of the disease show 50% risk of developing breast cancer and 80% of them carry germline mutations in PTEN.

The number of patients with family history of breast cancer engaging genetic counseling is increasing. This should lead an improvement of breast cancer surveillance and a more effective preventive care. Currently, genetic counseling endeavor to identify patients at risk of genetic anomalies, study family history and inheritance patterns, calculate risks of recurrence, and provide information regarding testing and treatment procedures. Therefore, breast cancer patients and their families are presented with possibilities of screening for BRCA1, BRCA2, TP53, and

PTEN

mutations, and preventive care such as chemoprevention and prophylactic surgery.

Declaration

The author declares no financial support was sought in this project and there is no conflicts of interest.

References

  1. Meric-Bernstam F, Zheng X, Shariati M, Damodaran S, Wathoo C, et al. (2018) Survival Outcomes by TP53 Mutation Status in Metastatic Breast Cancer. JCO Precis Oncol 2018.
  2. Ghoncheh M, Pournamdar Z, Salehiniya H (2016) Incidence and Mortality and Epidemiology of Breast Cancer in the World. Asian Pac J Cancer Prev 17: 43-46.
  3. Eisinger F, Sobol H, Serin D, Whorton JC (1998) Hereditary breast cancer, circa 1750. Lancet 351: 1366.
  4. Tung N, Lin NU, Kidd J, Allen BA, Singh N, et al. (2016) Frequency of Germline Mutations in 25 Cancer Susceptibility Genes in a Sequential Series of Patients With Breast Cancer. J Clin Oncol 34: 1460-1468.
  5. Couch FJ, Nathanson KL, Offit K (2014) Two decades after BRCA: setting paradigms in personalized cancer care and prevention. Science 343: 1466-1470.
  6. Foulkes WD, Smith IE, Reis-Filho JS (2010) Triple-negative breast cancer. N Engl J Med 363: 1938-1948.
  7. Evans DG, Graham J, O'Connell S, Arnold S, Fitzsimmons D (2013) Familial breast cancer: summary of updated NICE guidance. BMJ 346: 3829.
  8. Balmaña J, Díez O, Rubio IT, Cardoso F, ESMO Guidelines Working Group (2011) BRCA in breast cancer: ESMO Clinical Practice Guidelines. Ann Oncol 22: 31-34.
  9. La Verde N, Corsi F, Moretti A, Peissel B, Dalu D, et al. (2016) A targeted approach to genetic counseling in breast cancer patients: the experience of an Italian local project. Tumori 102: 45-50.
  10. Broeders M, Moss S, Nyström L, Njor S, Jonsson H, et al. (2012) The impact of mammographic screening on breast cancer mortality in Europe: a review of observational studies. J Med Screen 19: 14-25.
  11. Cedolini C, Bertozzi S, Londero AP, Bernardi S, Seriau L, et al. (2014) Type of breast cancer diagnosis, screening, and survival. Clin Breast Cancer 14: 235-240.
  12. Collaborative Group on Hormonal Factors in Breast Cancer (2001) 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.
  13. Robson ME, Bradbury AR, Arun B, Domchek SM, Ford JM (2015) American Society of Clinical Oncology Policy Statement Update: Genetic and Genomic Testing for Cancer Susceptibility. J Clin Oncol 33: 3660-3667.
  14. Wen L, Liu L, Wen L, Yu T, Wei F (2018) Artesunate promotes G2/M cell cycle arrest in MCF7 breast cancer cells through ATM activation. Breast Cancer 25: 681-686.
  15. Gudmundsdottir K, Ashworth A (2006) The roles of BRCA1 and BRCA2 and associated proteins in the maintenance of genomic stability. Oncogene 25: 5864-5874.
  16. Gupta I, Ouhtit A, Al-Ajmi A, Rizvi SGA, Al-Riyami H (2018) BRIP1 overexpression is correlated with clinical features and survival outcome of luminal breast cancer subtypes. Endocr Connect 7: 65-77.
  17. Liang M, Zhang Y, Sun C, Rizeq FK, Min M, et al. (2018) Association Between CHEK2*1100delC and Breast Cancer: A Systematic Review and Meta-Analysis. Mol Diagn Ther.
  18. Guacci A, Cordella A, Rocco T, Giurato G, Nassa G, et al. (2018) Identification of a novel truncating mutation in PALB2 gene by a multigene sequencing panel for mutational screening of breast cancer risk-associated and related genes. J Clin Lab Anal.
  19. Chai C, Wu H, Wang B, Eisenstat DD, Leng RP (2018) MicroRNA-498 promotes proliferation and migration by targeting the tumor suppressor PTEN in breast cancer cells. Carcinogenesis 39: 1185-1196.
  20. Liao SG, Liu L, Wang YJ (2018) Effect of RAD51C expression on the chemosensitivity of Eμ-Myc p19Arf-/- cells and its clinical significance in breast cancer. Oncol Lett 15: 6107-6114.
  21. Hall JM, Lee MK, Newman B, Morrow JE, Anderson LA, et al. (1990) Linkage of early-onset familial breast cancer to chromosome 17q21. Science 250: 1684-1689.
  22. Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K, et al. (1994) A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266: 66-71.
  23. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, et al. (1995) Identification of the breast cancer susceptibility gene BRCA2. Nature 378: 789-792.
  24. Lalloo F, Evans DG (2012) Familial breast cancer. Clin Genet 82: 105-114.
  25. Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, et al. (2000) BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes Dev 14: 927-939.
  26. Moiola C, De Luca P, Cotignola J, Gardner K, Vazquez E, et al. (2012) Dynamic coregulatory complex containing BRCA1, E2F1 and CtIP controls ATM transcription. Cell Physiol Biochem 30: 596-608.
  27. Starita LM, Parvin JD (2003) The multiple nuclear functions of BRCA1: transcription, ubiquitination and DNA repair. Curr Opin Cell Biol 15: 345-350.
  28. Brzovic PS, Rajagopal P, Hoyt DW, King MC, Klevit RE (2001) Structure of a BRCA1-BARD1 heterodimeric RING-RING complex. Nat Struct Biol 8: 833-837.
  29. Williams RS, Green R, Glover JN (2001) Crystal structure of the BRCT repeat region from the breast cancer-associated protein BRCA1. Nat Struct Biol 8: 838-842.
  30. Xia B, Sheng Q, Nakanishi K, Ohashi A, Wu J, et al. (2006) Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2. Mol Cell 22: 719-729.
  31. O'Donovan PJ, Livingston DM (2010) BRCA1 and BRCA2: breast/ovarian cancer susceptibility gene products and participants in DNA double-strand break repair, Carcinogenesis 31: 961-967.
  32. Connor F, Bertwistle D, Mee PJ, Ross GM, Swift S, et al. (1997) Tumorigenesis and a DNA repair defect in mice with a truncating Brca2 mutation. Nat Genet 17: 423-430.
  33. Fidler MM, Gupta S, Soerjomataram I, Ferlay J, Steliarova-Foucher E, et al. (2017) Cancer incidence and mortality among young adults aged 20-39 years worldwide in 2012: a population-based study. Lancet Oncol 18: 1579-1589.
  34. Barnes DR, Antoniou AC (2012) Unravelling modifiers of breast and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: update on genetic modifiers. J Intern Med 271: 331-343.
  35. Davis TC, Williams MV, Marin E, Parker RM, Glass J (2002) Health literacy and cancer communication. CA Cancer J Clin 52: 134-149.
  36. Soussi T, Caron de Fromentel C, May P (1990) Structural aspects of the p53 protein in relation to gene evolution. Oncogene 5: 945-952.
  37. Walker KK, Levine AJ (1996) Identification of a novel p53 functional domain that is necessary for efficient growth suppression. Proc Natl Acad Sci U S A 93: 15335-15340.
  38. Wawryk-Gawda E, Chylińska-Wrzos P, Lis-Sochocka M, Chłapek K, Bulak K, et al. (2014) P53 protein in proliferation, repair and apoptosis of cells. Protoplasma 251: 525-533.
  39. Song R, Tian K, Wang W, Wang L (2015) P53 suppresses cell proliferation, metastasis, and angiogenesis of osteosarcoma through inhibition of the PI3K/AKT/mTOR pathway. Int J Surg 20: 80-87.
  40. Lane D, Levine A (2010) p53 Research: the past thirty years and the next thirty years. Cold Spring Harb Perspect Biol 2: a000893.
  41. Han ES, Muller FL, Pérez VI, Qi W, Liang H, et al. (2008) The in vivo gene expression signature of oxidative stress. Physiol Genomics 34: 112-126.
  42. Surget S, Khoury MP, Bourdon JC (2013) Uncovering the role of p53 splice variants in human malignancy: a clinical perspective. Onco Targets Ther 7: 57-68.
  43. Cho Y, Gorina S, Jeffrey PD, Pavletich NP (1994) Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 265: 346-355.
  44. Sved J, Bird A (1990) The expected equilibrium of the CpG dinucleotide in vertebrate genomes under a mutation model. Proc Natl Acad Sci U S A 87: 4692-4696.
  45. Silva KS, Moura KK (2016) Genetic polymorphisms in patients with endometriosis: an analytical study in Goiânia (Central West of Brazil). Genet Mol Res 15.
  46. Lagares MH, Silva KSF, Barbosa AM, Rodrigues DA, Costa IR, et al. (2017) Analysis of p53 gene polymorphism (codon 72) in symptomatic patients with atherosclerosis. Genet Mol Res 16.
  47. de Morais MP, Curado RF, E Silva KS, Moura KK, Arruda JT (2016) Male idiopathic infertility and the TP53 polymorphism in codon 72. Genet Mol Res 15.
  48. Alawadi S, Ghabreau L, Alsaleh M, Abdulaziz Z, Rafeek M, et al. (2011) P53 gene polymorphisms and breast cancer risk in Arab women. Med Oncol 28: 709-715.
  49. Piao JM, Kim HN, Song HR, Kweon SS, Choi JS, et al. (2011) p53 codon 72 polymorphism and the risk of lung cancer in a Korean population. Lung Cancer 73: 264-267.
  50. Jiang DK, Yao L, Ren WH, Wang WZ, Peng B, et al. (2011) TP53 Arg72Pro polymorphism and endometrial cancer risk: a meta-analysis. Med Oncol 28: 1129-1135.
  51. Huang CY, Su CT, Chu JS, Huang SP, Pu YS, et al. (2011) The polymorphisms of P53 codon 72 and MDM2 SNP309 and renal cell carcinoma risk in a low arsenic exposure area. Toxicol Appl Pharmacol 257: 349-355.
  52. Tyner SD, Venkatachalam S, Choi J, Jones S, Ghebranious N, et al. (2002) p53 mutant mice that display early ageing-associated phenotypes, Nature 415: 45-53.
  53. Moretti F, Nanni S, Farsetti A, Narducci M, Crescenzi M, et al. (2000) Effects of exogenous p53 transduction in thyroid tumor cells with different p53 status. J Clin Endocrinol Metab 85: 302-308.
  54. Ikeda J, Tada M, Ishii N, Saya H, Tsuchiya K, et al. (2001) Restoration Of Endogenous Wild-Type P53 Activity In A Glioblastoma Cell Line With Intrinsic Temperature-Sensitive P53 Induces Growth Arrest But Not Apoptosis. Int J Cancer 94: 35-43.
  55. Hwang SJ, Lozano G, Amos CI, Strong LC (2003) Germline p53 mutations in a cohort with childhood sarcoma: sex differences in cancer risk. Am J Hum Genet 72: 975-983.
  56. Chompret A, Brugières L, Ronsin M, Gardes M, Dessarps-Freichey F, et al. (2000) Feunteun, P53 germline mutations in childhood cancers and cancer risk for carrier individuals. Br J Cancer 82: 1932-1937.
  57. Chu EC, Tarnawski AS (2004) PTEN regulatory functions in tumor suppression and cell biology. Med Sci Monit 10: RA235-241.
  58. Shaw RJ, Cantley LC (2006) Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441: 424-430.
  59. Engelman JA, Luo J, Cantley LC (2006) The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet 7: 606-619.
  60. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB (2005) Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4: 988-1004.
  61. Duan S, Yuan G, Liu X, Ren R, Li J, et al. (2015) PTEN deficiency reprogrammes human neural stem cells towards a glioblastoma stem cell-like phenotype. Nat Commun 6: 10068.
  62. Yang HP, Meeker A, Guido R, Gunter MJ, Huang GS, et al. (2015) PTEN expression in benign human endometrial tissue and cancer in relation to endometrial cancer risk factors. Cancer Causes Control 26: 1729-1736.
  63. Mithal P, Allott E, Gerber L, Reid J, Welbourn W, et al. (2014) PTEN loss in biopsy tissue predicts poor clinical outcomes in prostate cancer. Int J Urol 21: 1209-1214.
  64. Wang L, Xiong H, Wu F, Zhang Y, Wang J, et al. (2014) Hexokinase 2-mediated Warburg effect is required for PTEN- and p53-deficiency-driven prostate cancer growth. Cell Rep 8: 1461-1474.
  65. Cui M, Augert A, Rongione M, Conkrite K, Parazzoli S, et al. (2014) PTEN is a potent suppressor of small cell lung cancer. Mol Cancer Res 12: 654-659.
  66. Sacco JJ, Yau TY, Darling S, Patel V, Liu H, et al. (2014) The deubiquitylase Ataxin-3 restricts PTEN transcription in lung cancer cells. Oncogene 33: 4265-4272.
  67. Chiang KC, Chen HY, Hsu SY, Pang JH, Wang SY, et al. (2015) PTEN insufficiency modulates ER+ breast cancer cell cycle progression and increases cell growth in vitro and in vivo. Drug Des Devel Ther 9: 4631-4638.
  68. Guo Y, Chang H, Li J, Xu XY, Shen L, et al. (2015) Thymosin alpha 1 suppresses proliferation and induces apoptosis in breast cancer cells through PTEN-mediated inhibition of PI3K/Akt/mTOR signaling pathway. Apoptosis 20: 1109-1121.
  69. Kechagioglou P, Papi RM, Provatopoulou X, Kalogera E, Papadimitriou E, et al. (2014) Gounaris, Tumor suppressor PTEN in breast cancer: heterozygosity, mutations and protein expression. Anticancer Res 34: 1387-1400.
  70. Smith IN, Thacker S, Jaini R, Eng C (2018) Dynamics and structural stability effects of germline PTEN mutations associated with cancer versus autism phenotypes. J Biomol Struct Dyn 14: 1-17.
  71. Bhaumik S, Ahmad F, Das BR (2016) Somatic mutation analysis of KRAS, BRAF, HER2 and PTEN in EGFR mutation-negative non-small cell lung carcinoma: determination of frequency, distribution pattern and identification of novel deletion in HER2 gene from Indian patients. Med Oncol 33: 117.
  72. Chen Z, Che Q, Jiang FZ, Wang HH, Wang FY, et al. (2015) Piwil1 causes epigenetic alteration of PTEN gene via upregulation of DNA methyltransferase in type I endometrial cancer. Biochem Biophys Res Commun 463: 876-880.
  73. Nakanishi A, Kitagishi Y, Ogura Y, Matsuda S (2014) The tumor suppressor PTEN interacts with p53 in hereditary cancer (Review). Int J Oncol 44: 1813-1819.
  74. Ho J, Bassi C, Stambolic V (2015) Characterization of nuclear PTEN and its post translational modifications. Methods 77-78: 104-111.
  75. Li N, Zhang Y, Han X, Liang K, Wang J, et al. (2015) Poly-ADP ribosylation of PTEN by tankyrases promotes PTEN degradation and tumor growth. Genes Dev 29: 157-170.
  76. Singh B, Ittmann MM, Krolewski JJ (1998) Sporadic breast cancers exhibit loss of heterozygosity on chromosome segment 10q23 close to the Cowden disease locus. Genes Chromosomes Cancer 21: 166-171.
  77. Fackenthal JD, Marsh DJ, Richardson AL, Cummings SA, Eng C, et al. (2001) Male breast cancer in Cowden syndrome patients with germline PTEN mutations. J Med Genet 38: 159-164.
  78. Tan MH, Mester JL, Ngeow J, Rybicki LA, Orloff MS, et al. (2012) Lifetime cancer risks in individuals with germline PTEN mutations. Clin Cancer Res 18: 400-407.
  79. Garcia JM, Silva JM, Dominguez G, Gonzalez R, Navarro A, et al. (1999) Allelic loss of the PTEN region (10q23) in breast carcinomas of poor pathophenotype. Breast Cancer Res Treat 57: 237-243.
  80. Eng C (2003) PTEN: one gene, many syndromes. Hum Mutat 22: 183-198.
  81. Brownstein MH, Wolf M, Bikowski JB (1978) Cowden's disease: a cutaneous marker of breast cancer. Cancer 41: 2393-2398.
  82. Daly MB, Axilbund JE, Bryant E, Buys S, Eng C, et al. (2006) National Comprehensive Cancer Network, Genetic/familial high-risk assessment: breast and ovarian. J Natl Compr Canc Netw 4: 156-176.
  83. Pilarski R, Eng C (2004) Will the real Cowden syndrome please stand up (again)? Expanding mutational and clinical spectra of the PTEN hamartoma tumour syndrome. J Med Genet 41: 323-326.

Citation

Silva KSF (2019) Genetic Counseling, Polymorphisms and Breast Cancer. J Fam Med Dis Prev 5:098. doi.org/10.23937/2469-5793/1510098