Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2818 2023-05-09 13:58:24 |
2 Format correct Meta information modification 2818 2023-05-10 05:47:17 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
El Hejjioui, B.; Bouguenouch, L.; Melhouf, M.A.; El Mouhi, H.; Bennis, S. Gene Analysis of Breast Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/44041 (accessed on 18 August 2024).
El Hejjioui B, Bouguenouch L, Melhouf MA, El Mouhi H, Bennis S. Gene Analysis of Breast Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/44041. Accessed August 18, 2024.
El Hejjioui, Brahim, Laila Bouguenouch, Moulay Abdelilah Melhouf, Hind El Mouhi, Sanae Bennis. "Gene Analysis of Breast Cancer" Encyclopedia, https://encyclopedia.pub/entry/44041 (accessed August 18, 2024).
El Hejjioui, B., Bouguenouch, L., Melhouf, M.A., El Mouhi, H., & Bennis, S. (2023, May 09). Gene Analysis of Breast Cancer. In Encyclopedia. https://encyclopedia.pub/entry/44041
El Hejjioui, Brahim, et al. "Gene Analysis of Breast Cancer." Encyclopedia. Web. 09 May, 2023.
Gene Analysis of Breast Cancer
Edit

Breast cancer (BC) is a disease characterized by various clinical behaviors and biological characteristics. Breast cancer (BC) is heterogenous, showing variable morphologic and biological features; thus, it has different clinical behaviors and responses to treatment. Based on molecular and histological evidence, BC could be categorized into three groups: (1) BC expressing hormone receptor (estrogen receptor (ER+) or progesterone receptor (PR+)) commonly noted as luminal tumors and are responsive to endocrine therapy, (2) BC expressing human epidermal receptor 2 (HER2+) which is characterized by the overexpression of HER2 oncogene and is treated with trastuzumab, (3) Triple-negative breast cancer (TNBC) (ER−, PR−, HER2−) subtype, which is associated with high mortality rates and is not responsive to some drug treatment approaches.

breast cancer gene mutations

1. Introduction

Several genes present a potential target for studying breast cancer, The genes studied have been listed in Table 1, and the signaling pathways of these genes were mentioned in Figure 1.

Figure 1. Illustration of several signaling pathways implicated in the oncogenesis of breast cancer. (1) ERBB2 associated protein HER2 signaling pathway, (2) p53 protein implicated in the apoptosis, (3) BRCA1/2 couple implicated in DNA repair mechanisms such as Homologous recombination, (4) ESR1 engaged as a transcription factor.
Table 1. Summary of molecular and clinical utility of ctDNA in breast cancer.
Gene Patient Cohort Molecule Testing Technique Main Findings Clinical Significance References
ESR1 541 postmenopausal women with a diagnosis of MBC. Analyzed ESR1 mutations (Y537S and D538G) on cell-free DNA (cfDNA) using droplet digital polymerase chain reaction (ddPCR). D538G (21.1%)
Y537S (13.3%)
30 had both mutations.
These mutations were associated with shorter overall survival:
- wild-type, 32.1 months
- D538G, 25.99 months
- Y537S, 19.98 months
- Both mutations, 15.15 months.
[1]
  86 estrogen receptor-positive BC patients.185 plasma samples (151 plasma samples from 69 MBC patients, and 34 plasma samples from 17 primary BC (PBC) patients). Multiplex droplet digital PCR assays in a snapshot and serially. cfDNA ESR1 and PIK3CA mutations were found in 28.9% and 24.6% of MBC patients, respectively. All patients with ESR1 mutations had resistance to prior AI (aromatase inhibitor) therapy.
85% of patients with ESR1 mutations had resistance to prior SERM (Selective estrogen receptor modulators) therapy.
[2]
BRCA1/2 828 patients with advanced breast, ovarian, prostate, or pancreatic cancer.
(the study was conducted in accordance with the Declaration of Helsinki).
Plasma-based NGS assay. Of 828 patients, 60 (7.2%) had at least one BRCA1/2 loss-of-function mutation, 42 patients with germline mutations and 18 (14 patients had breast cancer) with somatic mutations only. NGS analysis of cfDNA identified high rates of therapeutically relevant mutations, including deleterious BRCA1/2 somatic mutations missed by germline testing. [3]
  24 patients with proven BRCA1/2 germline mutations (19 ovarian cancer patients and 5 patients with MBC who received prior treatment with platinum-based chemotherapy and/or PARP inhibitors). Targeted massively parallel sequencing of tumor DNA from ovarian cancer patients, cfDNA from ovarian and breast cancer patients, and their germline DNA. Identification of BRCA1 or BRCA2 reversion mutations in the cfDNA of 4 ovarian cancer patients (21%) and 2 breast cancer patients (40%). cfDNA sequencing can help identify putative BRCA1/2 reversion mutations which may facilitate patient selection for PARP inhibition therapy. [4]
PIK3CA Thirty patients with advanced BC (ABC); PIK3CA mutation analysis was performed using ddPCR. The presence of a PI3K mutation in liquid biopsy correlates with worse PFS in patients with ABC receiving CDK4/6i. Integration of PI3K status assessment with other molecular information could improve the management of patients with aggressive breast cancer and better suggest the best therapeutic strategy. [5]
TP53 46 patients with nonmetastatic triple-negative breast cancer; Characterization of TP53 gene mutations in tumor tissue through massively parallel sequencing (MPS). Monitoring of previously characterized mutations based on ctDNA analysis by ddPCR at four time points: pre-NCT, post-cycle, pre-surgery, and post-surgery. Results show a marked decrease in ctDNA levels and positivity rate during chemotherapy cycles. The high prevalence of TP53 mutations in TNBC is a potential biomarker for ctDNA monitoring during NCT, and therefore is a tool for TNBC management. [6]
  113 lung and 18 breast cancer patients NGS analysis of ctDNA: Panel for hot spot regions in 11 genes for lung cancer and 10 genes for breast cancer. Variations in the TP53 gene were detected at a high frequency in both tumor types, followed by the PIK3CA gene in breast cancer. Based on NGS and ddPCR techniques, liquid biopsy could be a very effective method for managing terminal cancer cases and monitoring treatment responses. [7]
  68 patients with metastatic breast cancer (MBC). cfDNA and gDNA (Genomic DNA) analysis by next-generation sequencing (NGS) TP53 mutations occurred in 10 (45.45%) TNBC patients, 9 (36.00%) HER2+ patients, and 7 (22.22%) HR+ patients.
TP53 represents the gene with the highest number of somatic mutations.
Mutations in TP53 cDNA and PIK3CA genes likely limit survival and promote disease progression. [8]
ERBB2 636 women with HER2 nonamplified MBC. ctDNA analysis by NGS. Results of this study indicate the efficacy of neratinib for HER2-mutated nonamplified breast cancer. This study supports the potential use of ctDNA to identify patients with HER2-mutated breast cancer to establish a new standard of care. [9]
  Multicohort, phase 2a, platform trial of ctDNA testing in 18 UK hospitals.1051 patients were registered in the study. ddPCR and NGS are used to detect ctDNA mutations.
Patients were recruited into four parallel treatment cohorts corresponding to the mutations identified in the ctDNA (ESR1; HER2; AKT1 and PTEN).
The findings of this study demonstrate the clinically relevant activity of targeted therapies against rare HER2 and AKT1 mutations. The results of this research show that ctDNA analysis, with the technologies used in this study, is accurate enough to be routinely adopted into clinical practice. [10]

2. BRCA1/2 Genes

Breast cancer with BRCA mutations is characterized by its aggressiveness, BRCA1 mutated breast cancer is frequently high grade and triple negative, BRCA2 related breast cancer is on average of a higher histological grade than sporadic cases [11]. Somatic mutations in BRCA1/2 occur in approximately 3% of all sporadic breast cancers [12].
The BRCA1 gene encodes a nuclear phosphoprotein that acts as a tumor suppressor gene by maintaining genomic stability [13]. The encoded protein combines with other tumor suppressors, DNA damage sensors, and signal transducers to form a large multi-unit protein complex known as the BRCA1 genome-associated surveillance complex [14].
The BRCA2 gene is involved in the maintenance of genomic stability and more specifically the homologous recombination (HR) pathway that repairs double-stranded DNA breaks. BRCA2 is located on chromosome 13q12.13, it is a large gene comprising 27 exons coding for 3418 amino acids [15].
In The Cancer Genome Atlas breast cancer study, which performed exome sequencing of tumor and normal samples from a selected cohort of breast cancer patients, a 1/3 somatic to 2/3 germline ratio was found [16] and a similar ratio of somatic to germline mutations has been found in other studies [17].
In a large-scale study in the United States, Vidula et al. analyzed cDNA by NGS and demonstrated that this approach can identify three classes of clinically relevant BRCA1/2 mutations: germline mutations, somatic loss of function, and reversions. This research supports the notion that systematic analysis of cfDNA in patients with advanced cancer can help identify potential candidates for appropriate genotype-based therapy [3].
In line with this, another study conducted in 2017 to analyze cfDNA by massively parallel sequencing, this study targeted all exons of 141 genes and all exons and introns of BRCA1 and BRCA2, as well as functional studies to assess the impact of putative BRCA1/2 reversion mutations on BRCA1/2 function, the findings of this research support the utility of cfDNA sequencing to identify putative BRCA1/2 reversion mutations and facilitate patient selection for PARP inhibition therapy [4].

3. ESR1 Gene

Over 75% of primary breast cancers are ER-positive, for which hormone therapy is the gold standard in the treatment armamentarium [18][19]. The estrogen receptor (ER) is a transcription factor involved in cell proliferation and activation [20]. Estrogen receptor alpha is encoded by the ESR1 gene. The acquisition of alterations in this gene has been identified as an important resistance mechanism to endocrine therapy [21]. ESR1 mutations rarely occur in primary tumors (~1%) but are relatively common (10–50%) in metastatic cancers presenting resistance to endocrine therapy, this is explained by the therapeutic pressure that stimulates the emergence of activating mutations of ESR1 in the metastatic tissue. These gene mutations are associated with shorter progression-free survival [20]. Therefore, it is important to test for ESR1 somatic mutations in ctDNA in order to detect the onset of this molecular resistance even before clinical progression. To predict treatment outcomes, because this may dictate patient management, including monitoring and modifications of the treatment plan, eventually this approach can be used to guide sequential treatment options in patients [22][23]. The potential value of screening for ESR1 mutations in metastatic cancer has increased.
In a Japanese study in 2017, involving 86 ER+ breast cancer patients, including 69 patients diagnosed with metastatic breast cancer (MBC) and 17 patients with primary breast cancer (PBC), the results showed the absence of ESR1 mutations in the PBC group, whereas in the MBC group ESR1 cfDNA mutations were detected in 28.9%. The clinical impact of these mutations was significantly important: all ESR1 mutation-positive patients had resistance to previous treatments with aromatase inhibitors (AIs) compared to 71.4% of ESR1 WT patients; 85% of ESR1 mutation-positive patients had resistance to previous treatment with selective estrogen receptor modulators (SERMs) compared to 51% of ESR1 WT patients. Otherwise, cfDNA monitoring in a subgroup of 52 patients showed that loss of ESR1 mutations was associated with a longer response time [2].
In January 2020, a Chinese comparative study was conducted comparing 297 tumor samples of primary breast cancer patients and 43 blood samples of metastatic breast cancer (MBC) patients. Next-generation sequencing (NGS) was used by targeting the whole exon of the ESR1 gene, and the result’s findings are consistent with previous studies: ESR1 mutations were more frequently detected in circulating tumor DNA of MBC patients than in PBC patients, and the ESR1 mutation frequency in patients using aromatase inhibitors (AIs) was significantly higher than those who were not using AIs [24].
A French large study targeting twelve genes by whole exome sequencing has identified ESR1 as a pilot gene specific to MBC, they confirmed that the ESR1 gene mutation is the most frequent “metastasis-specific” mutation observed in MBC, by identifying that 100% of ESR1 mutation-positive cases are ER + and present resistance to endocrine therapy [25]. In 2020, a confirmative study by Shibayama and Al reported that ESR1 mutations were found to be a prognostic predictor of acquired resistance to endocrine therapy [26].
Collectively, these studies demonstrate how the genetic profile of MBC is different from that of primary breast tumors. Hence, metastatic cancer profiling must become a primary step in the definition of optimal treatments for patients; thus, monitoring and surveillance studies will be critical to confirm the effectiveness after treatment. A final point regarding these findings was how easily and feasibly this biomarker could be obtained, plasma samples can be easily collected for ctDNA analysis, allowing for dynamic testing that will increase our understanding of the disease progression and the design of strategies to improve outcomes.

4. PIK3CA Gene

The PIK3CA gene encodes the A isoform of the catalytic subunit (p110a) of the phosphatidylinositol 3-kinases class IA. Phosphatidylinositol 3-kinases (PI3K) are involved in the conversion of phosphatidylinositol 4,5-biphosphate (PIP2) to phosphatidylinositol 3,4,5-triphosphate (PIP3). This PI3K class has a critical role in the control of various cellular processes such as cell growth and proliferation, metabolism, and migration via the PI3K/AKT/mTOR pathway [27].
In approximately 40% of HR + breast cancer cases, the most common molecular alterations are the activating mutations of the PI3K subunit in the PIK3CA gene, these mutations induce hyperactivation of the p110α catalytic subunit, leading to constitutive phosphorylation of the AKT and its forward effectors [28][29].
Several preclinical studies indicate that the PIK3/AKT/mTOR pathway alterations may correlate with resistance to CDK4/6 inhibitors (CDK4/6i). Cyclin-dependent kinase 4 and 6 (CDK4/6i) inhibitors have shown clinical efficacy in ER-positive MBC, although their cytostatic effects are limited by primary and acquired resistance, putative mechanisms of resistance to CDK4/6i have been identified [5][30].
An Italian study completed in January 2021 showed that patients with a PIK3CA mutation in the blood at the start of CDK4/6i treatment had a significantly shorter Progression Free Survival (PFS) compared to patients without a mutation. Therefore, PI3K status should be considered as a potential predictive biomarker of CDK4/6i resistance. The integration of PI3K status assessment with other molecular information in a surveillance system can improve the accuracy of predicting Overall Survival (OS) and PFS of patients with metastatic breast cancer and may suggest the best treatment strategy. On the other hand, it should be noted that PI3K status as such cannot be the only responsible for CDK4/6i resistance, and thus, special attention should be given to all mutations that also promote the activation of the PI3K/AKT/mTOR signaling pathway [5].
In the same sense, targeted therapy drugs in the final phase of clinical trials, specifically targeting PI3K, are designed to be administered to patients whose tumor has a PIK3CA gene alteration, which makes their detection particularly important in tumor genetics.

5. TP53 Gene

Tumor suppressor TP53 is considered the genome gatekeeper. Several studies indicate that TP53 mutations increase the risk of cancer and, once cancer occurs, these mutations promote invasion, metastasis, and chemoresistance [31]. In invasive breast cancer, TP53 (p. 53) is the most frequently mutated gene, which is mutated in approximately 80% of triple-negative breast cancer (TNBC) [32].
The presence or absence of TP53 mutations is routinely investigated in clinical practice on tumor tissues, using immunohistochemistry or sequencing techniques. However, these methods have various limitations, such as invasiveness and inability to identify tumor heterogeneity and progression, which underline the need for a more robust and sophisticated technology [33]. Currently, TP53 gene mutations can be detected by cfDNA analysis given the ability of this biomarker to detect tumor heterogeneity, which is a limitation of tumor biopsies [34][35].
In 2017, a French study was realized including 47 patients with non-metastatic triple-negative breast cancer (TNBC) treated with neoadjuvant chemotherapy (NCT), and the study targeted the TP53 gene at several time points: before NCT; after 1 cycle; before surgery, and after surgery. The results indicated a decrease in ctDNA levels during NCT; however, a small decrease in ctDNA levels during NCT was significantly associated with shorter survival. This research confirmed that the high prevalence of TP53 mutations in TNBC is a potential biomarker for ctDNA monitoring during NCT, and that ctDNA may become a clinically useful prognostic tool for managing TNBC patients treated with NCT [6].
In July 2019, Savli and colleagues showed that variations in the TP53 gene are strongly observed in breast cancer samples, this study demonstrated that TP53 pathogenic variants detection and monitoring by ctDNA analysis is recommended as a useful biomarker for predictive studies, for tumor growth monitoring and personalized treatment strategy planning [7].
Screening for TP53 mutations in ctDNA can provide monitoring for early detection of genetic events underlying drug resistance and can also inform therapy approaches [8], FEI MA et al., (ClinicalTrials.gov NCT01937689) have exploited ctDNA profiling before and after treatment with an oral anti-HER1/HER2 tyrosine kinase inhibitor for 18 HER2-positive metastatic breast cancer patients, and they identified that mutations in TP53 and PI3K/Akt/mTOR pathway genes were strongly implicated in resistance to HER1/HER2 blocking [36].
The overall clinical trials confirm that p53 is both a potential prognostic and predictive biomarker and a therapeutic target for breast cancer patients, particularly for TN subtype patients.

6. ERBB2 Gene

The human epidermal growth factor receptor 2 (HER2) is overexpressed in 20–30% of breast cancer patients [37], this amplification is “acquired” in approximately 2–5% of metastatic breast cancers that originally had primary cancers that were not HER2 amplified [38]. Blocking HER2 activity through trastuzumab provides a better outcome for HER2-positive patients. However, resistance in women with metastatic breast cancer (MBC) is usually observed [39].
As targeted anti-HER2 therapies are developed, the assessment of HER2 status will be more important to stratify patients to the most appropriate treatment regimens [38], and this requires repetitive tumor sampling to identify whether the cancer’s genetic profile has changed after previous treatment [40]. The analysis of circulating DNA has the potential to screen for HER2 (ERBB2) amplification acquisition in a non-invasive manner.
In 2017, a phase II study (Clinicaltrials.gov, NCT01670877) was conducted to evaluate the clinical benefit rate (CBR) of NERATINIB as monotherapy through determining HER2 mutations based on circulating DNA analysis, whereby the results indicated the efficacy of NERATINIB in HER2 mutated non-amplified breast cancer and justify that cfDNA sequencing may offer a non-invasive strategy to identify patients with HER2 mutated cancers [9]. In a similar direction, and at a larger scale, a multicenter, open-label, multicohort, phase 2a clinical trial (ClinicalTrials.gov, NCT03182634) in 18 UK hospitals was conducted, the results confirm the clinically relevant activity of targeted therapies against rare activating mutations in breast cancer, in HER2 mutant breast cancer (ERBB2) identified by cfDNA tests, NERATINIB had comparable activity to that observed when guided by tissue tests [10].
In October 2020, Kleftogiannis et al. developed a new error-correcting cfDNA sequencing approach using bioinformatics strategies to identify tumor-associated genomic alterations. This assay was also performed effectively on the detection of copy number variations (CNVs) in the ERBB2 oncogene. This result creates opportunities for better tumor characterization, in which sequential plasma samples can be collected to represent CNVs and variants more precisely over time [41].

References

  1. Chandarlapaty, S.; Chen, D.; He, W.; Sung, P.; Samoila, A.; You, D.; Bhatt, T.; Patel, P.; Voi, M.; Gnant, M.; et al. Prevalence of ESR1 Mutations in Cell-Free DNA and Outcomes in Metastatic Breast Cancer: A Secondary Analysis of the BOLERO-2 Clinical Trial. JAMA Oncol. 2016, 2, 1310–1315.
  2. Takeshita, T.; Yamamoto, Y.; Yamamoto-Ibusuki, M.; Tomiguchi, M.; Sueta, A.; Murakami, K.; Omoto, Y.; Iwase, H. Analysis of ESR1 and PIK3CA mutations in plasma cell-free DNA from ER-positive breast cancer patients. Oncotarget 2017, 8, 52142–52155.
  3. Vidula, N.; Rich, T.A.; Sartor, O.; Yen, J.; Hardin, A.; Nance, T.; Lilly, M.B.; Nezami, M.A.; Patel, S.P.; Carneiro, B.A.; et al. Routine Plasma-Based Genotyping to Comprehensively Detect Germline, Somatic, and Reversion BRCA Mutations among Patients with Advanced Solid Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2546–2555.
  4. Weigelt, B.; Comino-Méndez, I.; de Bruijn, I.; Tian, L.; Meisel, J.L.; García-Murillas, I.; Fribbens, C.; Cutts, R.; Martelotto, L.G.; Ng, C.K.; et al. Diverse BRCA1 and BRCA2 Reversion Mutations in Circulating Cell-Free DNA of Therapy-Resistant Breast or Ovarian Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 6708–6720.
  5. Del Re, M.; Crucitta, S.; Lorenzini, G.; De Angelis, C.; Diodati, L.; Cavallero, D.; Bargagna, I.; Cinacchi, P.; Fratini, B.; Salvadori, B.; et al. PI3K mutations detected in liquid biopsy are associated to reduced sensitivity to CDK4/6 inhibitors in metastatic breast cancer patients. Pharmacol. Res. 2021, 163, 105241.
  6. Riva, F.; Bidard, F.-C.; Houy, A.; Saliou, A.; Madic, J.; Rampanou, A.; Hego, C.; Milder, M.; Cottu, P.; Sablin, M.-P.; et al. Patient-Specific Circulating Tumor DNA Detection during Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Clin. Chem. 2017, 63, 691–699.
  7. Savli, H.; Sertdemir, N.; Aydin, D.; Dursun, B.; Kurtas, O.; Reka, S.; Sunnetci-Akkoyunlu, D.; Eren-Keskin, S.; Uygun, K.; Ozden, E.; et al. TP53, EGFR and PIK3CA gene variations observed as prominent biomarkers in breast and lung cancer by plasma cell-free DNA genomic testing. J. Biotechnol. 2019, 300, 87–93.
  8. Hu, Z.-Y.; Xie, N.; Tian, C.; Yang, X.; Liu, L.; Li, J.; Xiao, H.; Wu, H.; Lu, J.; Gao, J.; et al. Identifying Circulating Tumor DNA Mutation Profiles in Metastatic Breast Cancer Patients with Multiline Resistance. EBioMedicine 2018, 32, 111–118.
  9. Ma, C.X.; Bose, R.; Gao, F.; Freedman, R.A.; Telli, M.L.; Kimmick, G.; Winer, E.; Naughton, M.; Goetz, M.P.; Russell, C.; et al. Neratinib Efficacy and Circulating Tumor DNA Detection of HER2 Mutations in HER2 Nonamplified Metastatic Breast Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 5687–5695.
  10. Turner, N.C.; Kingston, B.; Kilburn, L.S.; Kernaghan, S.; Wardley, A.M.; Macpherson, I.R.; Baird, R.D.; Roylance, R.; Stephens, P.; Oikonomidou, O.; et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): A multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020, 21, 1296–1308.
  11. Baretta, Z.; Mocellin, S.; Goldin, E.; Olopade, O.I.; Huo, D. Effect of BRCA germline mutations on breast cancer prognosis. Medicine (Baltimore) 2016, 95, e4975.
  12. Pascual, T.; Gonzalez-Farre, B.; Teixidó, C.; Oleaga, L.; Oses, G.; Ganau, S.; Chic, N.; Riu, G.; Adamo, B.; Galván, P.; et al. Significant Clinical Activity of Olaparib in a Somatic BRCA1-Mutated Triple-Negative Breast Cancer with Brain Metastasis. JCO Precis. Oncol. 2019, 3, 1–6.
  13. Claus, E.B.; Risch, N.; Thompson, W.D. Genetic analysis of breast cancer in the cancer and steroid hormone study. Am. J. Hum. Genet. 1991, 48, 232–242.
  14. Wang, Y.; Cortez, D.; Yazdi, P.; Neff, N.; Elledge, S.J.; Qin, J. BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes Dev. 2000, 14, 927–939.
  15. Algebaly, A.S.; Suliman, R.S.; Al-Qahtani, W.S. Comprehensive study for BRCA1 and BRCA2 entire coding regions in breast cancer. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2021, 23, 74–81.
  16. Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70.
  17. Winter, C.; Nilsson, M.P.; Olsson, E.; George, A.M.; Chen, Y.; Kvist, A.; Törngren, T.; Vallon-Christersson, J.; Hegardt, C.; Häkkinen, J.; et al. Targeted sequencing of BRCA1 and BRCA2 across a large unselected breast cancer cohort suggests that one-third of mutations are somatic. Ann. Oncol. 2016, 27, 1532–1538.
  18. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990.
  19. Clatot, F.; Perdrix, A.; Sefrioui, D.; Sarafan-Vasseur, N.; Di Fiore, F. Intérêt clinique de la détection des mutations circulantes d’ESR1 chez les patientes traitées pour un cancer du sein métastatique exprimant les récepteurs hormonaux. Bull. Cancer (Paris) 2018, 105, 46–54.
  20. Zundelevich, A.; Dadiani, M.; Kahana-Edwin, S.; Itay, A.; Sella, T.; Gadot, M.; Cesarkas, K.; Farage-Barhom, S.; Saar, E.G.; Eyal, E.; et al. ESR1 mutations are frequent in newly diagnosed metastatic and loco-regional recurrence of endocrine-treated breast cancer and carry worse prognosis. Breast Cancer Res. 2020, 22, 16.
  21. Leiser, M. ESR1 Mutations Predict Survival, Endocrine Therapy Failure in Early Breast Cancer, 28 Avril 2020. Consulté le: 5 décembre 2022. . Available online: https://www.healio.com/news/hematology-oncology/20200428/esr1-mutations-predict-survival-endocrine-therapy-failure-in-early-breast-cancer (accessed on 24 September 2022).
  22. Song, P.; Wu, L.R.; Yan, Y.H.; Zhang, J.X.; Chu, T.; Kwong, L.N.; Patel, A.A.; Zhang, D.Y. Limitations and opportunities of technologies for the analysis of cell-free DNA in cancer diagnostics. Nat. Biomed. Eng. 2022, 6, 232–245.
  23. Smyth, L.M.; Reichel, J.B.; Tang, J.; Patel, J.A.A.; Meng, F.; Selcuklu, D.S.; Houck-Loomis, B.; You, D.; Samoila, A.; Schiavon, G.; et al. Utility of Serial cfDNA NGS for Prospective Genomic Analysis of Patients on a Phase I Basket Study. JCO Precis. Oncol. 2021, 5, 6–16.
  24. Zhu, W.; Ren, C.; Wang, Y.; Wen, L.; Zhang, G.; Liao, N. Prevalence of ESR1 Mutation in Chinese ER-Positive Breast Cancer. OncoTargets Ther. 2020, 13, 615–621.
  25. Lefebvre, C.; Bachelot, T.; Filleron, T.; Pedrero, M.; Campone, M.; Soria, J.-C.; Massard, C.; Lévy, C.; Arnedos, M.; Lacroix-Triki, M.; et al. Mutational Profile of Metastatic Breast Cancers: A Retrospective Analysis. PLoS Med. 2016, 13, e1002201.
  26. Shibayama, T.; Low, S.-K.; Ono, M.; Kobayashi, T.; Kobayashi, K.; Fukada, I.; Ito, Y.; Ueno, T.; Ohno, S.; Nakamura, Y.; et al. Clinical significance of gene mutation in ctDNA analysis for hormone receptor-positive metastatic breast cancer. Breast Cancer Res. Treat. 2020, 180, 331–341.
  27. Mosele, F.; Stefanovska, B.; Lusque, A.; Tran Dien, A.; Garberis, I.; Droin, N.; Le Tourneau, C.; Sablin, M.-P.; Lacroix, L.; Enrico, D.; et al. Outcome and molecular landscape of patients with PIK3CA-mutated metastatic breast cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2020, 31, 377–386.
  28. Saal, L.H.; Holm, K.; Maurer, M.; Memeo, L.; Su, T.; Wang, X.; Yu, J.S.; Malmström, P.-O.; Mansukhani, M.; Enoksson, J.; et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005, 65, 2554–2559.
  29. Zhao, J.J.; Cheng, H.; Jia, S.; Wang, L.; Gjoerup, O.V.; Mikami, A.; Roberts, T.M. The p110alpha isoform of PI3K is essential for proper growth factor signaling and oncogenic transformation. Proc. Natl. Acad. Sci. USA 2006, 103, 16296–16300.
  30. Herrera-Abreu, M.T.; Palafox, M.; Asghar, U.; Rivas, M.A.; Cutts, R.J.; Garcia-Murillas, I.; Pearson, A.; Guzman, M.; Rodriguez, O.; Grueso, J.; et al. Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor-Positive Breast Cancer. Cancer Res. 2016, 76, 2301–2313.
  31. Li, H.; Zhang, J.; Tong, J.H.M.; Chan, A.W.H.; Yu, J.; Kang, W.; To, K.F. Targeting the Oncogenic p53 Mutants in Colorectal Cancer and Other Solid Tumors. Int. J. Mol. Sci. 2019, 20, 5999.
  32. Duffy, M.J.; Synnott, N.C.; Crown, J. Mutant p53 in breast cancer: Potential as a therapeutic target and biomarker. Breast Cancer Res. Treat. 2018, 170, 213–219.
  33. Garrido-Castro, A.C.; Lin, N.U.; Polyak, K. Insights into Molecular Classifications of Triple-Negative Breast Cancer: Improving Patient Selection for Treatment. Cancer Discov. 2019, 9, 176–198.
  34. Rodriguez, B.J.; Córdoba, G.D.; Aranda, A.G.; Álvarez, M.; Vicioso, L.; Pérez, C.L.; Hernando, C.; Bermejo, B.; Parreño, A.J.; Lluch, A.; et al. Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis. J. Clin. Med. 2019, 8, 1183.
  35. Madic, J.; Kiialainen, A.; Bidard, F.-C.; Birzele, F.; Ramey, G.; Leroy, Q.; Rio Frio, T.; Vaucher, I.; Raynal, V.; Bernard, V.; et al. Circulating tumor DNA and circulating tumor cells in metastatic triple negative breast cancer patients. Int. J. Cancer 2015, 136, 2158–2165.
  36. Ma, F.; Zhu, W.; Guan, Y.; Yang, L.; Xia, X.; Chen, S.; Li, Q.; Guan, X.; Yi, Z.; Qian, H.; et al. ctDNA dynamics: A novel indicator to track resistance in metastatic breast cancer treated with anti-HER2 therapy. Oncotarget 2016, 7, 66020–66031.
  37. Cooke, T.; Reeves, J.; Lanigan, A.; Stanton, P. HER2 as a prognostic and predictive marker for breast cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2001, 12 (Suppl. 1), S23–S28.
  38. Houssami, N.; Macaskill, P.; Balleine, R.L.; Bilous, M.; Pegram, M.D. HER2 discordance between primary breast cancer and its paired metastasis: Tumor biology or test artefact? Insights through meta-analysis. Breast Cancer Res. Treat. 2011, 129, 659–674.
  39. Abraham, J.; Montero, A.J.; Jankowitz, R.C.; Salkeni, M.A.; Beumer, J.H.; Kiesel, B.F.; Piette, F.; Adamson, L.M.; Nagy, R.J.; Lanman, R.B.; et al. Safety and Efficacy of T-DM1 Plus Neratinib in Patients With Metastatic HER2-Positive Breast Cancer: NSABP Foundation Trial FB-10. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 2601–2609.
  40. Gevensleben, H.; Garcia-Murillas, I.; Graeser, M.K.; Schiavon, G.; Osin, P.; Parton, M.; Smith, I.E.; Ashworth, A.; Turner, N.C. Noninvasive detection of HER2 amplification with plasma DNA digital PCR. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 3276–3284.
  41. Kleftogiannis, D.; Ho, D.; Liew, J.X.; Poon, P.S.Y.; Gan, A.; Ng, R.C.-H.; Tan, B.K.-T.; Tay, K.H.; Lim, S.H.; Tan, G.S.; et al. Detection of genomic alterations in breast cancer with circulating tumour DNA sequencing. Sci. Rep. 2020, 10, 16774.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 313
Revisions: 2 times (View History)
Update Date: 10 May 2023
1000/1000
Video Production Service