Next Article in Journal
Low Temperature Hall Effect Investigation of Conducting Polymer-Carbon Nanotubes Composite Network
Next Article in Special Issue
DNA Damage and Repair in Epithelium after Allogeneic Hematopoietic Stem Cell Transplantation
Previous Article in Journal
Optimization of Lipase Production by Burkholderia sp. Using Response Surface Methodology
Previous Article in Special Issue
Alzheimer’s Disease-Associated Neurotoxic Peptide Amyloid-Β Impairs Base Excision Repair in Human Neuroblastoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A DNA Repair BRCA1 Estrogen Receptor and Targeted Therapy in Breast Cancer

Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
Int. J. Mol. Sci. 2012, 13(11), 14898-14916; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms131114898
Submission received: 24 September 2012 / Revised: 1 November 2012 / Accepted: 12 November 2012 / Published: 14 November 2012
(This article belongs to the Special Issue DNA Damage and Repair in Degenerative Diseases)

Abstract

:
BRCA1 is a key mediator of DNA repair pathways and participates in the maintenance of the genomic integrity of cells. The control of DNA damage repair mechanisms by BRCA1 is of great interest since molecular defects in this pathway may reflect a predictive value in terms of a cell’s sensitivity to DNA damaging agents or anticancer drugs. BRCA1 has been found to exhibit a hormone-dependent pattern of expression in breast cells. Wild-type BRCA1 is required for the inhibition of the growth of breast tumor cells in response to the pure steroidal ERα antagonist fulvestrant. Also a loss of BRCA1-mediated transcriptional activation of ERα expression results in increased resistance to ERα antagonists. Platinum-based drugs, poly(ADP-ribose) polymerase (PARP) inhibitors, and their combination are currently included in chemotherapy regimens for breast cancer. Preclinical and clinical studies in a BRCA1-defective setting have recently indicated a rationale for the use of these compounds against hereditary breast cancers. Initial findings indicate that neoadjuvant use of cisplatin results in high rates of complete pathological response in patients with breast cancer who have BRCA1 mutations. Cisplatin produces a better response in triple-negative breast cancer (TNBC) than in non-TNBC diseases in both the neoadjuvant and adjuvant settings. This implies that TNBC cells may harbor a dysfunctional BRCA1 repair pathway.

1. Breast Cancer Suppressor Gene 1 (BRCA1) and its Encoded Protein

BRCA1 is a tumor suppressor gene consisting of 5592 base pairs spanning 24 exons; 22 exons of which encode a 220 kDa protein of 1863 amino acids together with two non-coding exons. The encoded BRCA1 is functionally characterized into three major domains including a N-terminal RING domain, a nuclear localization signal domain (NLS) and a BRCA1 C-terminal domain (BRCT domain) [1,2]. The BRCA1 protein has multi-functions in at least four major areas of cellular processes, including DNA repair, transcriptional activation, cell cycle regulation, chromatin remodeling and protein ubiquitination (Figure 1).
The N-terminal RING domain contains the conservative sequences of cysteine and histidine residues necessary for specific coordination with two Zn2+ ions. This region of BRCA1 interacts with a BARD1 (BRCA1 associated RING domain 1) to form a heterodimeric complex. The BRCA1-BARD1 complex requires both parts for their mutual stability. They are co-localized in nuclear dots during the S phase but not the G phase of the cell cycle as well as in nuclear foci [3]. The progression to the S phase by aggregation of nuclear BRCA1 and BARD1 implied the importance of both proteins for a DNA repair function [4]. The BRCA1-BARD1 complex also exhibits enzymatic activity of an E3 ubiquitin ligase that specifically transfers ubiquitin to protein substrates which are essential for cellular viability [3,5]. The central region of BRCA1 also called the nuclear localization signal domain, covers exon 11 (approximately 3500 bp) and constitutes approximately 60 percent of the coding region of the gene. Deletion of exon 11 results in removal of the nuclear localization signal of BRCA1. Biophysical characterization revealed that this domain is intrinsically disordered or is negatively unfolded under physiological conditions. This might potentially allow the BRCA1 central region to act as a long flexible scaffold, to mediate interactions with DNA, and perhaps a number of other proteins involved in DNA damage response and repair [6]. The reported binding partner proteins to the central region are c-Myc, RB, p53, FANCA, RAD50, RAD51, and BRCA2 [7]. The C-terminal domain of BRCA1 (residues 1646–1863) contains two BRCT (BRCA1 C-terminal) domains in tandem (motif 1: amino acids 1653–1736); motif 2: amino acids 1760–1855). These domains serve as multipurpose protein-protein interaction modules that bind to other BRCT repeats or other protein domains with apparently unrelated structures [8].
Since all these processes are involved in the maintenance of genomic stability, BRCA1 has been implicated as a key regulator of cellular response to DNA damage [9,10]. One important function of BRCA1 is the repair of DNA damage. The link between BRCA1 and DNA repair has demonstrated that BRCA1 colocalizes with a homologous recombinase, RAD51 [11]. BRCA1 is implicated in playing a crucial role in DNA repair through several mechanisms, including homologous recombination (HR) repair, the less error-prone mechanism of repairing DNA double-strand breaks (DSBs) [12]. Induction of DSB, the most destructive and cytotoxic DNA lesion, by irradiation or anticancer agents is a major strategy employed for breast cancer treatment. To repair the lesions, cells perform a DNA-damage response that includes chromatin remodeling, activation of cell-cycle checkpoints, DNA repair and, allowing time for the DNA repair to occur. If the responses fail, cells undergo apoptosis as a last resort to sustain genomic stability. DSB preferentially causes breast cancer cells to undergo apoptosis, especially when relevant repair pathways, such as those mediated by BRCA1, are perturbed [13]. Several lines of evidence have indicated that inactivation of the genes required for a DNA double-strand break (DSB) repair pathway including ATM, MDC, BRCA1, BRCA2, and RAD51, causes cells to become hypersensitive to DSB-inducing agents.
The molecular mechanism of the HR repair pathway after DSB has recently been dramatically revealed (Figure 2). The Mre11-Rad50-Nbs1 (MRN) complex that acts as a DSB sensor, first recognizes DSB and recruits ATM, a PI3 kinase, that is frequently mutated in ataxia telangiectasia patients, to the site of DNA damage. ATM phophorylates the histone variant H2AX (γ-H2AX) [14,15] which can now directly recruit MDC1. ATM further phosphorylates MDC1 [1618], then recruits an E3 ubiquitin ligase, RNF8, that catalyzes lysine 63, K63-linked polyubiquitin chains at the sites of DNA damage [1922]. The K63-linked ubiquitin polymer next recruits the BRCA1-Abraxas-RAP80 complex through the RAP80 component, a protein that contains two UIM (ubiquitin interacting motif) domains [2325]. BRCA1 can form RING heterodimer E3 ligase activity with BARD1, and this is required for the recruitment of BRCA2 and RAD51 to the damaged sites to initiate HR repair through a sister chromatid exchange [2628]. Many cancer-predisposing mutations in the BRCA1 RING domain, that inhibited E3 ligase activity and its ability to accumulate at a damaged site, were defective in homologous recombination, which is critical for tumor suppression [27,29,30].
The significance of the DNA repair function of BRCA1 through HR was observed from experiments that showed that BRCA1-deficient mouse embryonic stem cells displayed defective homologous repair of chromosomal recombination with increased frequency of non-homologous recombination. This impairment could be corrected by the reconstitution of a wild-type BRCA1 [31]. Antisense or siRNA-based inhibition of endogenous BRCA1 expression promoted increased sensitivity to cisplatin that was associated with decreased DNA repair and increased apoptosis [3234]. This indicated that the reduced BRCA1 expression observed in sporadic cancers may also be exploited for DNA damage-based chemotherapy [35,36]. Expression of BRCA1 was found to be about two-fold lower in sporadic triple-negative breast cancers compared to estrogen receptor (ER)-positive cancers [37]. In addition, upregulation of microRNA (miR-182) that targets BRCA1 expression may also lead to BRCA1 pathway dysfunction, and subsequently reduce the efficiency of HR repair [38]. BRCA1-defective human breast cancer, HCC1937 cells (a BRCA1 mutation with an insertion of C at nucleotide 5382, and a negative expression of HER2/neu, and estrogen receptor α (ERα)) were significantly more sensitive to cisplatin than BRCA1 reconstituted cells with a full-length cDNA transfection [39]. In a similar situation, BRCA1-deficient mouse embryonic stem cells displayed defective DNA repair and a 100-fold increased sensitivity to the alkylating agent mitomycin C and cisplatin compared to those containing wild-type BRCA1 [40,41]. This sensitivity was reversed upon correction of the BRCA1 mutation in mouse embryonic fibroblast cells with disrupted BRCA1 [42]. Reconstitution of BRCA1 in the cells via transfection meant that BRCA1 functions were regained, and resulted in a reduced level of cancer cell death, following treatment with cisplatin or other DNA damaging agents [34].
As described earlier, BRCA1 contains a C-terminal transactivation domain [4346]. The transactivation domain is mapped to the region of the protein encoded by exon 21–24 using deletion constructs of BRCA1 fused to the GAL4 DNA binding domain. The BRCA1-BRCT domain has been implicated in the regulation of transcription of several genes responsible for DNA damage. The ability of BRCA1 to act as either a co-activator or a co-repressor of transcription may involve its ability to recruit basal transcription machinery and other proteins that have been implicated in chromatin remodeling [47]. BRCA1 was shown to interact with the RNA polymerase II holoenzyme [11], and is capable of activating the p21 promoter [48]. It has been reported that BRCA1 participated in stabilizing p53 in response to DNA damage, and served as a co-activator for p53 [49]. The interaction of BRCA1 and p53 potentially resulted in the redirection of a p53-mediated transactivation from apoptotic target genes involved in DNA repair and cell cycle arrest [49]. In addition, it has been reported that Smad3, a component of the transforming growth factor β (TGF-β) signaling pathway, which is a potent regulator of growth and apoptosis, also for invasiveness of tumor cells, forms a complex with BRCA1 in vitro and in vivo. The interaction is mediated by the MH1 domain of Smad3 and the C-terminal part of BRCA1. However, Smad3 counteracted the BRCA1-dependent repair of DNA double-strand breaks in human breast epithelial cells, as evaluated by formation of BRCA1 nuclear foci. Smad3, therefore, suppresses BRCA1-dependent DNA repair in response to a DNA damaging agent [50]. In addition, BRCA1 was shown to repress the transcription of ERα and its downstream estrogen responsive genes [51]. The transcriptional repression activity of BRCA1 was found to abolish its ability to inhibit ERα activity that occurs by the association of the N-terminus of BRCA1 (residues 1–300) with the C-terminal activation function (AF-2) of ERα. As described above, BRCA1 and BARD1 interact through their RING domain to form a heterodimer with E3 ubiquitin ligase activity. The BRCA1 E3 ligase activity has been found to be inactivated by a breast cancer-derived mutation and platinum-based drugs [3,52]. In addition, cancer-predisposing mutations in BRCA1 have been observed to abrogate ERα ubiquitination [53].

2. Estrogen, Estrogen Receptor and Breast Cancer

Estrogen (E2) is important in women for a variety of physiological processes. It affects growth, differentiation, and the function of tissues in the reproductive system, including the mammary glands, uterus, vagina, and ovaries [54,55]. Estrogen action is primarily mediated through binding with nuclear proteins called estrogen receptors (ER). Estrogen receptors are members of nuclear hormone receptors, a family of hormone activated transcription factors that can initiate or enhance the transcription of genes containing specific hormone response elements [56,57]. The ERα protein is encoded from the estrogen receptor 1 gene (ESR1 gene). It consists of 595 amino acids with a molecular weight of 66 kDa that has been separated into six different functional domains (A–F) (Figure 3) [58,59].
The N-terminal A/B domain is involved in activation of ligand-dependent transcription. The DNA binding-domain (DBD) on the C-region mediates ERα binding to specific DNA enhancer sequences, called the estrogen responsive element (ERE) with the consensus sequence of 5′-GGTCANNNTGACC-3′ [60,61]. Several three-dimensional structures are known for ERα DBD alone and in complex with DNA [55,6163]. The topology of ERα DBD is characterized by a zinc finger-like motif with eight cyteines that constitute the tetrahedral coordination of the two zinc ions. The E-region is the ligand-binding domain (LBD) which is responsible for the high affinity for binding of estrogen. The DBD and LBD are connected by the hinge region (D-domain). The F-domain is the C-terminal extension region of LBD [64].
The mechanism of estrogen action via ERα is a very complex process (Figure 4). In the non-stimulated receptor, the heat shock protein 90 (HSP90) is positioned in such a way that prevents dimerization and strong binding to the ERE in DNA. The addition of estrogen (E2) is followed by the separation of HSP90, which is accompanied by dimerization, with a strong interaction with the LBD, and a weaker interaction with the DBD. The resulting dimer binds to the ERE and activates transcription through the intervention of the ligand-inducible transcription-activating function 2 (TAF-2: recently called AF2) and the constitutive transcription activation 1 (TAF1: recently called AF1), located at the LBD and the region A/B, respectively [65].

3. Association between BRCA1 Expression and Response to Antiestrogen Treatment

Breast tumorigenesis and breast cancer progression involves the deregulation or hyper- activation of intracellular signaling proteins that leads to uncontrolled cellular proliferation, invasion and metastasis. The estrogen receptor and transforming growth factor β (TGF-β) signaling pathways especially, change during breast tumorigenesis and breast cancer progression, through the downstream mediator, Smad3. Several studies have reported that ERα suppresses the expression of Smad3 induced by estrogen [66]. Reversal of the suppression of Smad3 activity by ERα/E2 decreased, when induced by the antiestrogen tamoxifen, which indicates that this effect is mediated directly from the ERα activity. These findings are consistent with the reported therapeutic effects of antiestrogens such as tamoxifen, through local boosting of TGF-β signaling [67]. Previous studies have reported that induction of TGF-β in a breast cancer cell line revealed that the response to antiestrogen was confined to ER-positive (MCF-7) cells and not ER-negative (MDA-MB-231) cells. ER-positive (MCF-7) cells responded to antiestrogen, tamoxifen, but ER-negative MDA-MB-231 cells did not [68].
The estrogen receptor status is useful in predicting the benefit obtained from endocrine therapy. It may also help predict which patients benefit from advances in adjuvant chemotherapy [69]. In patients with hormone-sensitive tumors, tamoxifen reduces the risk of recurrence and death. Furthermore, treatment with the aromatase inhibitor alone or consecutively with tamoxifen replaces or further reduces the risk of recurrence in post-menopausal women with estrogen receptor-positive tumors [70]. Endocrine therapy with selective estrogen receptor modulators (SERMs) has been the mainstay of breast cancer prevention trials to date. It is also known that tamoxifen exerts pure antagonism on genes that require only the AF-2 domain for ERα-mediated transcriptional activity. In contrast, in genes for which ERα AF-2 is not required, transcription is then driven only by AF-1 and tamoxifen can function as a partial agonist [71,72].
Germline mutations in the BRCA1 gene confer a genetic predisposition to breast and ovarian cancers. BRCA1-mutant breast cancers exhibit a distinct pathologic phenotype and lack of ERα [73]. BRCA1 has been shown to inhibit ERα signaling, which results in negative regulation of expression of downstream genes [74], as well as regulation of estrogen biosynthesis through transcriptional inhibition of the aromatase encoding genes [75]. Recently, an alternative pathway for breast cancer treatment was described using pure antiestrogen. The effect of BRCA1 expression on the response of breast cancer cells to a pure steroidal ERα antagonist, fulvestrant, has been investigated [76,77] (Figure 5).
Unlike the selective estrogen receptor modulator tamoxifen, the primary mechanism of the action of fulvestrant is through downregulation of ERα (Figure 6). Fulvestrant is a steroidal analogue of 17β-estradiol, which competitively binds to ERα with a high affinity [78,79]. It acts as an antiestrogen chemical by reducing the half-life of ERα [80], resulting in a decrease in expression of ERα. Formation of the drug-receptor complex results in stabilization of the receptor, which is then degraded by an ubiquitin-proteasome complex [8183].
Fulvestrant has been shown to inhibit the growth of cells that were transfected with siRNA [58]. This indicates that the wild-type BRCA1 is required for fulvestrant to inhibit the growth of breast tumor cells, and loss of BRCA1-mediated transcriptional activation of the expression of ERα results in an increased resistance to ERα antagonists [58]. Due to its unique action in the downregulation of the ERα, the mechanism of fulvestrant is described by the acronym “SERD” (Selective ER downregulator). In addition, binding of fulvestrant to the ERα, inhibits ERα dimerization and the uptake of the drug-receptor complex by the nucleus (Figure 6) [79]. Fulvestrant also causes inactivation of two regions of the ERα, the activating function-1 (AF-1) and the activating function-2 (AF-2), that normally recruit coactivator and corepressor proteins required for expression of ERα-regulated genes [80,84]. Considerable data has demonstrated the efficacy of fulvestrant in postmenopausal women with ER-positive advanced breast cancer, both for the first-line setting and following disease progression after tamoxifen or aromatase inhibitors. Recently, fulvestrant was reported to provide improved benefits with alternative dosing strategies. Considering all administration schedules, fulvestrant has an excellent safety profile with no significant adverse effects [85]. More recently, the aromatase inhibitors, that inhibit the final chemical conversion of androgens to estrogens, have shown an increased disease-free survival benefit over tamoxifen in patients with primary hormone receptor-positive breast cancer, as well as reducing the risk of developing contralateral breast cancers [86].

4. “Triple-Negative” Breast Cancer and Treatment

Breast cancer is a heterogeneous disease, and gene expression profiling has shown that it is possible to classify and identify five major biologically distinct intrinsic subtypes: luminal A, luminal B, human epidermal growth factor receptor 2 (HER2) overexpression, basal-like, and normal-like [8789]. These molecular subtypes have prognostic and predictive values as HER2-overexpressing and basal-like breast cancers have poor outcomes. Follow-up studies have shown that these subtypes are conserved across diverse patient series and array platforms [90,91], and have shown that different gene expression-based predictors are a good way for tracking a similar, common set of biological subtypes, with significant agreement in predicting patient outcomes [92]. A subtype of breast cancer, characterized by the lack of expression of estrogen receptor (ER), progesterone (PR), and human epidermal growth factor 2 (HER2) is called a ‘triple-negative’ breast cancer (TNBCs) [9397]. This subgroup accounts for about 15% of all breast cancers and for a higher percentage of breast cancers detected in African and African-American women who are premenopausal [94]. TNBC has important clinical implications, because it is typicality high grade, has a ductal histology, and exhibits a high rate of proliferation. In general, compared with other subtypes of breast cancer, TNBC has a less favorable clinical outcome in terms of the nature and likelihood of progression, availability of various treatment options, and survival. Although a cure is likely if TNBC is diagnosed early and responds well to treatment, the highly aggressive nature of this disease has contributed to poorer outcomes, overall [95]. TNBC patients are known to have a greater pathological complete response (pCR) rate when compared with non-TNBC patients [98].
Currently, there is no preferred standard form of chemotherapy for TNBC, and treatment should be selected as it is for other cancer subtypes. In the adjuvant setting, anthracyclines and taxanes remain the standard of care for TNBC patients in node-positive breast cancer [99]. Retrospective analysis indicates that the addition of docetaxel or paclitaxel to anthracycline containing adjuvant regimens may be of greater benefit for the treatment of ER-negative and HER2-negative cancers, which are much more common [100,101]. More experimental neoadjuvant regimens including platinum drugs paired with taxane have been shown to achieve high pCR rates in TNBC [98,102]. Newer treatment approaches to the use of platinum agents, cisplatin and carboplatin to treat TNBC are currently being assessed in clinical trials; on the basis that the dysfunction of BRCA1 and its various pathways is associated with a specific DNA-repair defect that sensitizes cells to these agents in an animal model [103]. In addition, single-agent cisplatin induced a response in TNBC patients. Decreased BRCA1 expression in TNBC sensitizes patients to cisplatin [104]. Therefore, cisplatin-based chemotherapy has improved outcomes for treating TNBC patients [105]. The molecular pathway of cisplatin-induced cell death for TNBC has been discovered using triple-negative cells (HCC1937 cells that express only mutationally inactivated BRCA1 and MDA-MB-468 cells that express wild-type BRCA1) [106]. The mediator signal of this pathway is p63/p73. The expression of these genes appears to reflect a functional pathway shared by BRCA1-associated tumors [106]. Inactivation of this pathway increases the IC50 of breast cancer cells for cisplatin by 10 to 100 fold. In addition, enhancement of p73 protein expression was also observed in MDA-MB-231, MDA-MB-468 and HCC1937 cells following the treatment of cells with a cisplatin-rapamycin combination [107]. BRCA1 mutant cells that exhibit sensitivity to a gemcitabine and cisplatin combination treatment, appear to be mediated by sustained DNA damage and inefficient DNA repair that triggers p63/p73 mediated apoptosis [108,109]. The p63/p73 proteins are expressed in about 30%–50% of TNBC patients and this might be a biomarker for clinical sensitivity to cisplatin [97,106,110]. Initial findings indicate that neoadjuvant use of cisplatin results in high rates of complete pathological response in patients with breast cancer who have BRCA1 mutations [104,111,112]. This anticancer platinum drug produces a better response in TNBC than in non-TNBC diseases in both the neoadjuvant and adjuvant settings [113].
TNBC tumors are strongly associated with germline mutations in the BRCA1 gene [114], although much about this relationship remains to be defined [115]. The present data indicates that a defective BRCA1 function could be more specifically targeted by poly (ADP-ribose) polymerase (PARP) inhibitors. PARP has an important role in base excision repair of single-strand DNA breaks. Inhibition of PARP leads to accumulation of single-stranded DNA breaks that can cause the formation of double-stranded DNA breaks, after stalling the progressing DNA replication forks. These double-stranded DNA breaks cannot be accurately repaired in tumors with homologous recombination deficiency [12]. The inhibition of PARP using synthetic killing agents has therefore been advanced as a novel targeted therapy for cancers harboring BRCA1 mutations [116]. Preclinical data on the mechanisms of PARP inhibitors are at a different stage of clinical development for the targeted treatment of BRCA1-deficient breast cancer and TNBC. PARP inhibitors include olaparib (AZD2281, KU-0059436), iniparib (BSI-201), and veliparib (ABT888). Olaparib has been evaluated in a phase I study. In vitro data has shown that inhibition of PARP leads to a highly selective apoptosis of BRCA1 null cells [117]. The DNA repair defects, that are the characteristics of BRCA1-deficient cells, confer sensitivity to PARP inhibition, which could be relevant to the treatment of TNBC [118]. PARP inhibitors have recently shown very encouraging clinical activity in early trials of tumors arising in BRCA1 mutation [119]. A recent multi-center proof-of-concept phase II trial demonstrated a positive result with a single olaparib treatment in a BRCA1-mutated breast cancer [120]. In addition, olaparib has been tested in combination with cisplatin in TNBC, and with gemcitabine in solid tumors [121,122]. However, iniparib failed to improve survival in TNBC patients in phase III clinical trials in combination with chemotherapy [123]. Nevertheless, it is optimistic that future development of this class of compounds remains necessary to determine the effectiveness of PARP inhibitors in the treatments of breast cancer patients with BRCA1-associated mutations [124].

5. Conclusions

The goal of all cancer therapies is to selectively eradicate the cancer while sparing normal tissues. The cellular responses to DNA damage, especially related to repair or tolerance of the damage are critical issues in determining the efficacy of most cancer chemotherapy. The selective sensitivity of cancer cells relative to normal cells should improve the therapeutic ratio for cancer chemotherapy. Cancer cells with defective DNA repair pathways, such as loss of BRCA1, inactivation of the BRCA1 DNA repair pathway, and synthetic lethality, alter their sensitivity to DNA-damaging agents or chemotherapeutic drugs. BRCA1 represses the transcription of ERα and its downstream estrogen responsive genes. Wild-type BRCA1 is required for the inhibition of growth of breast tumor cells in response to the pure steroidal ERα antagonist fulvestrant. Also a loss of BRCA1-mediated transcriptional activation of ERα expression results in increased resistance to ERα antagonists. Platinum-based drugs, poly (ADP-ribose) polymerase (PARP) inhibitors, and their combination are currently included in chemotherapy regimens for breast cancer. Although widely used as anticancer therapeutics, the clinical applications of the anticancer platinum drugs are limited due to their adverse side effects and the cancer cells can also develop resistance to the drugs. Therefore, rationally designed drugs that exert their anticancer activities on both estrogenic activity and synthetic lethality could lead to the discovery of new opportunities for the development of targeted breast cancer therapies.

Acknowledgements

The author would like to thank the National Research Council of Thailand (PHA570058S-1) and Prince of Songkla University for financial support (PHA550314S), Khwanjira Hongthong is for the preparation of the figures, and Brian Hodgson for assistance with the English.

References

  1. Huen, M.S.; Sy, M.H.S.; Chen, J. BRCA1 and its toolbox for the maintenance of genome integrity. Nat. Rev. Mol. Cell Biol 2010, 11, 138–148. [Google Scholar]
  2. Rosen, E.M.; Fan, S.; Pestell, R.G.; Goldberg, I.D. BRCA1 gene in breast cancer. J. Cell. Physiol 2003, 196, 19–41. [Google Scholar]
  3. Hashizume, R.; Fukuda, M.; Maeda, I.; Nishikawa, H.; Oyake, D.; Yabuki, Y.; Ogata, H.; Ohta, T. The RING heterodomer BRCA1-BARD1 is ubiquitin ligase inactivated by a breast cancer-derived mutation. J. Biol. Chem 2001, 276, 14537–14540. [Google Scholar]
  4. Jin, Y.; Xu, X.L.; Yang, M.C.; Wei, F.; Ayi, T.C.; Bowcock, A.M.; Baer, R. Cell cycle-dependent colocalization of BARD1 and BRCA1 proteins in discrete nuclear domains. Proc. Natl. Acad. Sci. USA 1997, 94, 12075–12080. [Google Scholar]
  5. Xia, Y.; Pao, G.M.; Chen, H.W.; Verma, I.M.; Hunter, T. Enhancement of BRCA1 E3 ubiquitin ligase activity through direct interaction with the BARD1 protein. J. Biol. Chem 2003, 278, 5255–5263. [Google Scholar]
  6. Mark, W.Y.; Liao, J.C.; Lu, Y.; Ayed, A.; Laister, R.; Szymczyna, B.; Chakrabartty, A.; Arrowsmith, C.H. Characterization of segments from the central region of BRCA1: An intrinsically disordered scaffold for multiple protein-protein and protein-DNA interactions? J. Mol. Biol 2005, 345, 275–285. [Google Scholar]
  7. Rosen, E.M.; Fan, S.; Pestell, R.G.; Goldberg, I.D. BRCA1 in hormone-responsive cancer. Trends Endocrinol. Metab 2003, 14, 378–385. [Google Scholar]
  8. Watts, F.Z.; Brissett, N.C. Linking up and interacting with BRCT domains. DNA Repair (Amst.) 2010, 9, 103–108. [Google Scholar]
  9. Liu, W.; Zong, W.; Wu, G.; Fujita, T.; Li, W.; Wu, J.; Wan, Y. Turnover of BRCA1 involves in radiation-induced apoptosis. PLoS One 2010, 12, e14484. [Google Scholar]
  10. Starita, L.M.; Parvin, J. The multiple nuclear functions of BRCA1: Transcription ubiquitination and DNA repair. Curr. Opin. Cell Biol 2003, 13, 345–350. [Google Scholar]
  11. Scully, R.; Chen, J.; Plug, A.; Xiao, Y.; Weaver, D.; Feunteun, J.; Ashley, T.; Livingston, D.M. Association of BRCA1 with RAD51 in mitotic and meiotic cell. Cell 1997, 88, 265–275. [Google Scholar]
  12. Ashworth, A. A synthetic lethal therapeutic approach poly (ADP) ribose polymeraseinhibitors for the treatment of cancers deficient in DNA double-strand break repair. J. Clin. Oncol 2008, 26, 3785–3790. [Google Scholar]
  13. Ohta, T.; Wu, W.; Koike, A.; Asakawa, H.; Koizumi, H.; Fukuda, M. Contemplating chemosensitivity of basal-like breast cancer based on BRCA1 dysfunction. Breast Cancer 2009, 16, 268–274. [Google Scholar]
  14. Bassing, C.H.; Suh, H.; Ferguson, D.O.; Chua, K.F.; Manis, J.; Eckersdorff, M.; Gleason, M.; Bronson, R.; Lee, C.; Alt, F.W. Histone H2AX: A dosage-dependent suppressor of oncogenic translocations and tumors. Cell 2003, 114, 359–370. [Google Scholar]
  15. Celeste, A.; Fernandez-Capetillo, O.; Kruhlak, M.J.; Pilch, D.R.; Staudt, D.W.; Lee, A.; Bonner, R.F.; Bonner, W.M.; Nussenzweig, A. Histone H2AX phosphorylation is dispensable for the initial recognition of DNA breaks. Nat. Cell Biol 2003, 5, 675–679. [Google Scholar]
  16. Lou, Z.; Minter-Dykhoise, K.; Franco, S.; Gostissa, M.; Rivera, M.A.; Celeste, A.; Manis, J.P.; van Deursen, J.; Nussenzweig, A.; Paull, T.T.; et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Mol. Cell 2006, 21, 187–200. [Google Scholar]
  17. Stucki, M.; Clapperton, J.A.; Moharmmad, D.; Yaffe, M.B.; Smerdon, S.J.; Jackson, S.P. MDC1 directly binds phosphorylated histone H2AX to regulate cellular response to DNA double-strand breaks. Cell 2005, 123, 1213–1226. [Google Scholar]
  18. Stucki, M.; Jackson, S.P. gammaH2AX and MDC1: Anchoring the DNA-damage-response machinery to broken chromosomes. DNA Repair (Amst.) 2006, 5, 534–543. [Google Scholar]
  19. Huen, M.S.; Grant, R.; Manke, I.; Minn, K.; Yu, X.; Yaffe, M.B.; Chen, J. RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell 2007, 131, 901–914. [Google Scholar]
  20. Kolas, N.K.; Chapman, J.R.; Nakada, S.; Ylanko, J.; Chahwan, R.; Sweeney, F.D.; Panier, S.; Mendez, M.; Wildenhain, J.; Thomson, T.M.; et al. Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science 2007, 318, 1637–1640. [Google Scholar]
  21. Mailand, N.; Bekker-Jensen, S.; Faustrup, H.; Melander, F.; Bartek, J.; Lukas, C.; Lukas, J. RNF8 ubiquitylate histones at DNA double-strand breaks and promotes assembly of repair proteins. Cell 2007, 131, 887–900. [Google Scholar]
  22. Plans, V.; Scheper, J.; Soler, M.; Loukili, N.; Okano, Y.; Thomson, T.M. The RING finger protein RNF8 recruits UBC13 for lysine 63-based self polyubiquitylation. J. Cell. Biochem 2006, 97, 572–582. [Google Scholar]
  23. Kim, H.; Chen, J.; Yu, X. Ubiquitin-binding protein RAP80 mediates BRCA1-dependent DNA damage response. Science 2007, 316, 1202–1205. [Google Scholar]
  24. Sobhian, B.; Shao, G.; Lilli, D.R.; Culhance, A.C.; Moreau, L.A.; Xia, B.; Livingston, D.M.; Greenberg, R.A. RAP80 targets BRCA1 to specific ubiquitin structures at DNA damage sites. Science 2007, 316, 1198–1202. [Google Scholar]
  25. Wang, B.; Matsuoka, S.; Ballif, B.A.; Zhang, D.; Smogirzewska, A.; Gygi, S.P.; Elledge, S.J. Abraxas and RAP80 form a BRCA1 protein complex required for the DNA damage response. Science 2007, 316, 1194–1198. [Google Scholar]
  26. Greenberg, R.A.; Sobhian, B.; Pathania, S.; Cantor, S.B.; Nakatani, Y.; Livingston, D.M. Multifactorial contributions to an acute DNA damage response by BRCA1/BARD1-containing complexes. Genes Dev 2006, 20, 34–46. [Google Scholar]
  27. Ransburgh, D.J.; Chiba, N.; Ishioka, C.; Toland, A.E.; Parvin, J.D. Identification of breast tumor mutations in BRCA1 that abolish its function in homologous DNA recombination. Cancer Res 2010, 70, 988–995. [Google Scholar]
  28. Yu, D.S.; Sonoda, E.; Takeda, S.; Huang, C.L.; Pellegrini, L.; Blundell, T.L.; Venkitaraman, A.R. Dynamic control of RAd51 recombinase by self-association and interaction with BRCA2. Mol. Cell 2003, 12, 1029–1041. [Google Scholar]
  29. Morris, J.R.; Boutell, C.; Keppler, M.; Densham, R.; Weekes, D.; Alamshah, A.; Butler, L.; Pangon, L.; Kiuchi, T.; Ng, T.; et al. The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress. Nature 2009, 462, 886–890. [Google Scholar]
  30. Morris, J.R.; Pangon, L.; Boutell, C.; Katagiri, T.; Keep, N.H.; Solomon, E. Genetic analysis of BRCA1 ubiquitin ligase activity and its relationship to breast cancer susceptibility. Hum. Mol. Genet 2006, 15, 599–606. [Google Scholar]
  31. Snouwaert, J.N.; Gowen, L.C.; Latour, A.M.; Mohn, A.R.; Xiao, A.; DiBiase, L.; Koller, B.H. BRCA1 deficient embryonic stem cell displays a decreased homologous recombination frequency and an increased frequency of non-homologous recombination that is corrected by expression of Brac1 transgene. Oncogene 1999, 18, 7900–7907. [Google Scholar]
  32. Husain, A.; He, G.; Venkatraman, E.S.; Spriggs, D.R. BRCA1 up-regulation is associated with repair-mediated resistance to cis-diaminedichloroplatinum (II). Cancer Res 1998, 58, 1120–1123. [Google Scholar]
  33. Lafarge, S.; Sylvain, V.; Ferrara, M.; Bignon, Y.J. Inhibition of BRCA1 leads to increased chemoresistance to microtubule-interfering agents, and effects that involves the JNK pathway. Oncogene 2001, 20, 6597–6606. [Google Scholar]
  34. Quinn, J.E.; Kennedy, R.E.; Mullan, P.B.; Gilmore, P.M.; Carty, M.; Johnston, P.G.; Harkin, D.P. BRCA1 functions as a differential modulator of chemotherapy-induced apoptosis. Cancer Res 2003, 63, 6221–6228. [Google Scholar]
  35. James, C.R.; Quinn, J.E.; Mullan, P.B.; Johnston, P.G.; Harkin, D.P. BRCA1, a potential predictive biomarker in the treatment of breast cancer. Oncologist 2007, 12, 142–150. [Google Scholar]
  36. Quinn, J.E.; Carser, J.E.; James, C.R.; Kennedy, R.D.; Harkin, D.P. BRCA1 and implications for response to chemotherapy in ovarian cancer. Gynecol. Oncol 2009, 113, 134–142. [Google Scholar]
  37. Turner, N.C.; Reis-Filho, J.S.; Russell, A.M.; Springall, R.J.; Ryder, K.; Steele, D.; Savage, K.; Gillett, C.E.; Schmitt, F.C.; Ashworth, A.; et al. BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 2007, 26, 2126–2132. [Google Scholar]
  38. Moskwa, P.; Buffa, F.M.; Pan, Y.; Panchakshari, R.; Gottipati, P.; Muschel, R.J.; Beech, J.; Kulshrestha, R.; Abdelmohsen, K.; Weinstock, D.M.; et al. miR-182-mediated downregulation of BRCA1 impacts DNA repair and sensitivity to PARP inhibitors. Mol. Cell 2011, 41, 210–220. [Google Scholar]
  39. Tassone, P.; Tagliaferri, P.; Pericelli, P.; Blotta, S.; Quaresima, B.; Martelli, M.L.; Goel, A.; Barbieri, V.; Costanzo, F.; Boland, C.R.; et al. BRCA1 expression modulates chemosensitivity of BRCA1-defective HCC1937 human breast cancer cells. Br. J. Cancer 2003, 88, 1285–1291. [Google Scholar]
  40. Bhattacharyya, A.; Ear, U.S.; Koller, B.H.; Weicheselbaum, R.R.; Bishop, D.K. The breast cancer susceptibility gene BRCA1 is required for subnuclear assembly of Rad51 and survival following treatment with the DNA cross-linking agent cisplatin. J. Biol. Chem 2000, 275, 23899–23903. [Google Scholar]
  41. Moynahan, M.E.; Cui, T.Y.; Jasin, M. Homology-induced DNA repair, mitomycin C resistance, and chromosome stability is restored with correction of a BRCA1 mutation. Cancer Res 2001, 61, 4842–4850. [Google Scholar]
  42. Fedier, A.; Steiner, R.A.; Schwarz, V.A.; Lenherr, L.; Haller, U.; Fink, D. The effect of loss of BRCA1 on the sensitivity to anticancer agents in p53 deficient cells. Int. J. Oncol 2003, 22, 1169–1173. [Google Scholar]
  43. Chapman, M.S.; Verma, I.M. Transcription activation by BRCA1. Nature 1996, 382, 678–679. [Google Scholar]
  44. Monteiro, A.N.; August, A.; Hanafusa, H. Evidence for a transcriptional activation function of BRCA1 C-terminal region. Proc. Natl. Acad. Sci. USA 1996, 93, 13595–13599. [Google Scholar]
  45. Ratanaphan, A. A DNA Repair Protein BRCA1 as a Potentially Molecular Target for the Anticancer Platinum Drug Cisplatin, DNA Repair; Kruman, I., Ed.; InTech Open Access Publisher: Rijeka, Croatia, 2011. Available online: http://www.intechopen.com/books/dna-repair/a-dna-repair-protein-brca1-as-a-potentially-molecular-target-for-the-anticancer-platinum-drug-cispla accessed on 12 November 2012.
  46. Ratanaphan, A.; Wasiksiri, S.; Canyuk, B.; Prasertsan, P. Cisplatin-damaged BRCA1 exhibits altered thermostability and transcriptional transactivation. Cancer Biol. Ther 2009, 8, 890–898. [Google Scholar]
  47. Mullan, P.B.; Quinn, J.E.; Harkin, D.P. The role of BRCA1 in transcriptional regulation and cell cycle control. Oncogene 2006, 25, 5854–5863. [Google Scholar]
  48. Somasundaram, K.; Zhang, H.; Zeng, Y.X.; Houvras, Y.; Peng, Y.; Zhang, H.; Wu, G.S.; Licht, J.D.; Weber, B.L.; El-Deiry, W.S. Arrest of the cell cycle by the tumor-suppressor BRCA1 requires the CDK-inhibitor p21WAF1/CiP1. Nature 1997, 389, 187–190. [Google Scholar]
  49. Zhang, H.; Somasundaram, K.; Peng, Y.; Tian, H.; Bi, D.; Weber, B.L.; El-Deiry, W.S. BRCA1 physically associates with p53 and stimulates its transcriptional activity. Oncogene 1998, 16, 1713–1721. [Google Scholar]
  50. Dubrovska, A.; Kanamoto, T.; Lomnyyska, M.; Heldin, C.H.; Volodko, N.; Souchelnytskyi, S. TGF beta1/Smad3 counteracts BRCA1-dependent repair of DNA damage. Oncogene 2005, 14, 2289–2297. [Google Scholar]
  51. Fan, S.; Wang, J.; Yuan, R.; Ma, Y.; Meng, Q.; Erdos, M.R.; Pestell, R.G.; Yuan, F.; Auborn, K.S.; Goldberg, I.D.; et al. BRCA1 inhibition of estrogen receptor signaling in transfected cells. Science 1999, 284, 1354–1356. [Google Scholar]
  52. Atipairin, A.; Canyuk, B.; Ratanaphan, A. The RING heterodimer BRCA1-BARD1 is a ubiquitin ligase inactivated by the platinum-based anticancer drugs. Breast Cancer Res. Treat 2011, 126, 203–209. [Google Scholar]
  53. Eakin, C.M.; Maccoss, M.J.; Finney, G.L.; Klevit, R.E. Estrogen receptor α is putative substrate for the BRCA1 ubiquitin ligase. Proc. Natl. Acad. Sci. USA 2007, 104, 5794–5799. [Google Scholar]
  54. Osborne, C.K.; Zhao, H.; Fuqua, S.A.W. Selective estrogen receptor modulators: Structure, function, and clinical use. J. Clin. Oncol 2000, 18, 3172–3186. [Google Scholar]
  55. Schwabe, J.W.; Chapman, L.; Finch, J.T.; Rhodes, D.; Neuhaus, D. DNA recognition by the oestrogen receptor: from solution to the crystal. Structure 1993, 1, 187–204. [Google Scholar]
  56. Chen, X.; Danes, C.; Lowe, M.; Herliczek, T.W.; Keyomarsi, K. Activation of the estrogen-signalling pathway by p21WAF1/CIP1 in estrogen receptor-negative breast cancer cells. J. Natl. Cancer Inst 2000, 92, 1403–1431. [Google Scholar]
  57. Osborne, C.K. Steroid hormone receptors in breast cancer management. Breast Cancer Res. Treat 1998, 51, 227–238. [Google Scholar]
  58. Hosey, A.M.; Gorski, J.J.; Murray, M.M.; Quinn, J.E.; Chung, W.Y.; Gail, E.C.; Colin, R.S.; Susan, M.J.; Jude, M.F.; Alistair, N.M.; et al. Molecular basis for estrogen receptor α deficiency in BRCA1-linked breast cancer. J. Natl. Cancer Inst 2007, 99, 1683–1694. [Google Scholar]
  59. Macgregor, J.; Jordan, C. Basic guide to the mechanisms of antiestrogen action. Pharm. Rev 1998, 50, 152–187. [Google Scholar]
  60. Klinge, C.M. Estrogen receptor interaction with estrogen response elements. Nucleic Acid Res 2001, 29, 2905–2919. [Google Scholar]
  61. Schwabe, J.W.; Chapman, L.; Finch, J.T.; Rhodes, D. The crystal structure of the estrogen receptor DNA-binding domain bound to DNA: how receptors discriminate between their response elements. Cell 1993b, 75, 567–578. [Google Scholar]
  62. Schwabe, J.W.; Chapman, L.; Rhodes, D. The oestrogen receptor recognizes an imperfectly palindromic response element through an alternative side-chain conformation. Structure 1995, 3, 201–213. [Google Scholar]
  63. Schwabe, J.W.; Neuhaus, D.; Rhodes, D. Solution structure of the DNA-binding domain of the oestrogen receptor. Nature 1990, 348, 458–461. [Google Scholar]
  64. Ruff, M.; Gangloff, M.; Wurtz, J.M.; Moras, D. Estrogen receptor transcription and transactivation structure function relationship in DNA and ligand binding domains of estrogen receptor. Breast Cancer Res 2000, 2, 353–359. [Google Scholar]
  65. Cano, A.; Hermenegildo, C. Modulation of the oestrogen receptor: A process with distinct susceptible steps. Hum. Reprod. Update 2000, 6, 207–211. [Google Scholar]
  66. Cherlet, T.; Murphy, L.C. Estrogen receptors inhibit Smad3 transcriptional activity throughAp-1 transcription factors. Mol. Cell Biochem 2007, 306, 33–42. [Google Scholar]
  67. Matsuda, T.; Yamamoto, T.; Muraguchi, A.; Saatcioglu, F. Cross-talk between transforming growth factor-beta and estrogen receptor signaling through Smad3. J. Biol. Chem 2001, 276, 42908–42914. [Google Scholar]
  68. Herman, M.E.; Katzenellenbogen, B.S. Response-specific antiestrogen resistance in a newly characterized MCF-7 human breast cancer cell line resulting from long-term exposure to trans-hydroxytamoxifen. J. Steroid Biochem. Mol. Biol 1996, 59, 121–134. [Google Scholar]
  69. Berry, D.A.; Cirrincione, C.; Citron, M.L.; Budman, D.R.; Goldstein, L.J.; Martino, S.; Perez, E.A.; Muss, H.B.; Norton, L.; Hudis, C.; et al. Estrogen-receptor status and outcomes of modern chemotherapy for patients with node-positive breast cancer. JAMA 2006, 295, 1658–1667. [Google Scholar]
  70. Gross, P.E.; Ingle, J.N.; Martino, S.; Robert, N.J.; Muss, H.B.; Piccart, M.J.; Castigliome, M.; Tu, D.; Shepherd, L.E.; Pritchard, K.I.; et al. A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. N. Engl. J. Med. 2003, 3491, 1793–1802. [Google Scholar]
  71. McDonnell, D.P.; Clemm, D.L.; Hermann, T.; Goldman, M.E.; Pike, J.W. Analysis of estrogen receptor function in vitro reveals three distinct classes of antiestrogens. Mol. Endocrinol 1995, 9, 659–669. [Google Scholar]
  72. Tzukerman, M.T.; Esty, A.; Santiso, M.D.; Danielian, P.; Parker, M.G.; Stein, R.B.; Pike, J.W.; McDonnell, D.P. Human estrogen receptor transactivational capacity is determined by both cellular and promoter context and mediated by two functionally distinct intramolecular regions. Mol. Endocrinol 1994, 8, 21–30. [Google Scholar]
  73. Foulkes, W.D.; Metcalfe, K.; Sun, P.; Hanna, W.M.; Lynch, H.T.; Ghadirian, P.; Tung, N.; Olopade, O.I.; Weber, B.L.; McLennan, J.; et al. Estrogen receptor status in BRCA1- and BRCA2-related breast cancer: the influence of age, grade, and histological type. Clin. Cancer Res 2004, 10, 2029–2034. [Google Scholar]
  74. Rosen, E.M.; Fan, S.; Isaacs, C. BRCA1 in hormonal carcinogenesis: basis and clinical research. Endocr. Relat. Cancer 2005, 12, 533–548. [Google Scholar]
  75. Hu, Y.; Ghosh, S.; Amleh, A.; Yue, W.; Lu, Y.; Katz, A.; Li, R. Modulation of aromatase expression by BRCA1: A possible link to tissue-specific tumor suppression. Oncogene 2005, 24, 8343–8348. [Google Scholar]
  76. Howell, A.; Osborne, C.K.; Morris, C.; Wakeling, A.E. ICI 182,780 (Faslodex): Development of a novel, “pure” antiestrogen. Cancer 2000, 89, 817–825. [Google Scholar]
  77. Vendrell, J.A.; Magnino, F.; Danis, E.; Duchesne, M.J.; Pinloche, S.; Pons, M.; Birnbaum, D.; Nguyen, C.; Theillet, C.; Cohen, P.A. Estrogen regulation in human breast cancer cells of new downstream gene targets involved in estrogen metabolism, cell proliferation and cell transformation. J. Mol. Endocrinol 2004, 32, 397–414. [Google Scholar]
  78. Wakeling, A.E. Similarities and distinctions in the mode of action of different classes of antiestrogens. Endocr. Relat. Cancer 2000, 7, 17–28. [Google Scholar]
  79. Walkeling, A.E.; Bowler, J. Steroidal pure antioestrogens. J. Endocrinol 1987, 112, R7–R10. [Google Scholar]
  80. Wijayaratne, A.L.; McDonnell, D.P. The human estrogen receptor-alpha is a ubiquitinated protein whose stability is affected differentially by agonists, antagonists, and selective estrogen receptor modulators. J. Biol. Chem 2001, 276, 35684–35692. [Google Scholar]
  81. Alarid, E.T.; Bakopoulos, N.; Solodin, N. Proteasome-mediated proteolysis of estrogen receptor: A novel component in autologous down-regulation. Mol. Endocrinol 1999, 13, 1522–1534. [Google Scholar]
  82. Lonard, D.M.; Nawaz, Z.; Smith, C.L.; O’Malley, B.W. The 26S proteasome is required for estrogen receptor-alpha and coactivator turnover and for efficient estrogen-alpha transcription. Mol. Cell 2000, 5, 939–948. [Google Scholar]
  83. Nawaz, Z.; Lonard, D.M.; Dennis, A.P.; Smith, C.L.; O’Malley, B.W. Proteasome-dependent degradation of the human estrogen receptor. Proc. Natl. Acad. Sci. USA 1999, 96, 1858–1862. [Google Scholar]
  84. McKenna, N.J.; Lanz, R.B.; O’Malley, B.W. Nuclear receptor coregulators: cellular and molecular biology. Endocr. Rev 1999, 20, 321–344. [Google Scholar]
  85. Scott, S.M.; Brown, M.; Come, S.E. Emerging data on the efficacy and safety of fulvestrant, a unique antiestrogen therapy for advanced breast cancer. Expert Opin. Drug Saf 2011, 10, 819–826. [Google Scholar]
  86. Litton, J.K.; Arun, B.K.; Brown, P.H.; Hortobagyi, G.N. Aromatase inhibitors and breast cancer prevention. Expert. Opin. Pharmacother 2012, 13, 325–331. [Google Scholar]
  87. Perou, C.M.; Sorlie, T.; Eisen, M.B.; van de Rijin, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumors. Nature 2000, 406, 747–752. [Google Scholar]
  88. Sorlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; van de Rijin, M.; Jeffrey, S.S.; et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar]
  89. Sorlie, T.; Tibshirani, R.; Parker, J.; Hastie, T.; Marron, J.S.; Nobel, A.; Deng, S.; Johnsen, H.; Pesich, R.; Geisler, S.; et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl. Acad. Sci. USA 2003, 100, 8418–8423. [Google Scholar]
  90. Hu, Z.; Fan, C.; Oh, D.S.; Marron, J.S.; He, X.; Qaqish, B.F.; Livasy, C.; Carey, L.A.; Reynolds, E.; Dressler, L.; et al. The molecular portraits of breast tumors are conserved across microarray platform. BMC Genomics 2006, 7, 96. [Google Scholar]
  91. Sorlie, T.; Wang, Y.; Xiao, C.; Johnsen, H.; Naume, B.; Samaha, R.R.; Borresen-Dale, A.L. Distinct molecular mechanisms underlying clinically relevant subtypes of breast cancer: Gene expression analyses across three different platforms. BMC Genomics 2006, 7, 127. [Google Scholar]
  92. Fan, C.; Oh, D.S.; Wessels, L.; Weigelt, B.; Nuyten, D.S.; Nobel, A.B.; van’t Veer, L.J.; Perou, C.M. Concordance among gene-expression-based predictors for breast cancer. N. Eng. J. Med 2006, 355, 560–569. [Google Scholar]
  93. Bauer, K.R.; Brown, M.; Cress, R.D.; Parise, C.A.; Caggiano, V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: A population-based study from the California Cancer Registry. Cancer 2007, 109, 1721–1728. [Google Scholar]
  94. Cleator, S.; Heller, W.; Coombes, R.C. Triple-negative breast cancer: Therapeutic options. Lancet Oncol 2007, 8, 235–244. [Google Scholar]
  95. Cleere, D.W. Triple-negative breast cancer: A clinical update. Commun. Oncol 2010, 7, 203–211. [Google Scholar]
  96. Irvin, W.J., Jr; Carey, L.A. What is triple-negative breast cancer? Eur. J. Cancer 2008, 44, 2799–2805. [Google Scholar]
  97. Isakoff, S.J. Triple-negative breast cancer: Role of specific chemotherapy agents. Cancer J 2010, 16, 53–61. [Google Scholar]
  98. Liedtke, C.; Mazouni, C.; Hess, K.R.; André, F.; Tordai, A.; Mejia, J.A.; Symmans, W.F.; Gonzalez-Angulo, A.M.; Hennessy, B.; Green, M.; et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol 2008, 26, 1275–1281. [Google Scholar]
  99. Hugh, J.; Hanson, J.; Cheang, M.C.U.; Nielsen, T.O.; Perou, C.M.; Dumontet, C.; Reed, J.; Krajewska, M.; Treilleux, I.; Rupin, M.; et al. Breast cancer subtypes and response to docetaxel in node-positive breast cancer: Use of immunohistochemical definition in the BCIRG 001 trial. J. Clin. Oncol 2009, 27, 1168–1176. [Google Scholar]
  100. Ellis, P.; Barrett-Lee, P.; Johnson, L.; Cameron, D.; Wardley, A.; O’Reilly, S.; Verrli, M.; Smith, I.; Yarnold, J.; Coleman, R.; et al. The TACT trial management group and the TACT trialists. Sequential docetaxel as adjuvant chemotherapy for early breast cancer (TACT): An open-label, phase III, randomized controlled trial. Lancet 2009, 373, 1681–1692. [Google Scholar]
  101. Hayes, D.; Thor, A.D.; Dressler, L.G.; Weaver, D.; Ederton, S.; Cowan, D.; Broadwater, G.; Goldstein, L.J.; Martino, S.; Ingle, J.; et al. The cancer and leukemia group B (CALGB) investigator. HER2 and response to paclitaxel in node-positive breast cancer. N. Engl. J. Med 2007, 357, 1496–1506. [Google Scholar]
  102. Sikov, W.M.; Dizon, D.S.; Strenger, R.; Legare, R.D.; Theall, K.P.; Graves, T.A.; Gass, J.S. Frequent pathologic complete responses in aggressive stage II to III breast cancers with every-4-week-carboplatin and weekly paclitaxel with or without trastuzumab: A Brown University Oncology Group study. J. Clin. Oncol 2008, 927, 4693–4700. [Google Scholar]
  103. Foulkes, W.D. Traffic control for BRCA1. N. Engl. J. Med 2010, 326, 755–756. [Google Scholar]
  104. Silver, D.P.; Richardson, A.L.; Eklund, A.C.; Wang, Z.C.; Szallasi, Z.; Li, Q.; Juul, N.; Leong, C.O.; Calogrias, D.; Buraimoh, A.; et al. Efficacy of neoadjuvant cisplatin in triple-negative breast cancer. J. Clin. Oncol 2010, 28, 1145–1153. [Google Scholar]
  105. Koshy, N.; Quispe, D.; Shi, R.; Mansour, R.; Burton, G.V. Cisplatin-gemcitabine therapy in metastatic breast cancer: Improved outcome in triple-negative breast cancer patients compared to non-triple negative patients. Breast 2010, 19, 246–248. [Google Scholar]
  106. Leong, C.O.; Vidnovic, N.; DeYoung, M.P.; Sgroi, D.; Ellisen, L.W. The p63/p73 network mediates chemosensitivity to cisplatin in a biologically defined subset of primary breast cancers. J. Clin. Invest 2007, 117, 1370–1380. [Google Scholar]
  107. Wong, S.W.; Tiong, K.H.; Kong, W.Y.; Yue, Y.C.; Chua, C.H.; Lim, J.Y.; Lee, C.Y.; Quah, S.I.; Fow, C.; Chung, C.; et al. Rapamycin synergizes cisplatin sensitivity in basal-like breast cancer through up-regulation of p73. Breast Cancer Res. Treat 2011, 128, 301–313. [Google Scholar]
  108. Gong, J.G.; Costanzo, A.; Yang, H.-Q.; Melinos, G.; Kaelin, W.G.; Levrero, M.; Wang, J.Y.J. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature 1999, 399, 806–809. [Google Scholar]
  109. Hastak, K.; Elizabeth, A.; Ford, J.M. Synergistic chemosensitivity of triple-negative breast cancer cell line to poly (ADP-ribose) polymerase inhibition, gemcitabine, and cisplatin. Cancer Res 2010, 70, 7970–7980. [Google Scholar]
  110. Santana-Davila, R.; Perez, E.A. Treatment options for patients with triple-negative breast cancer. J. Hematol. Oncol 2010, 3, 42. [Google Scholar]
  111. Byrski, T.; Huzarski, T.; Dent, R.; Gronwald, J.; Zuziak, D.; Cybulski, C.; Kladny, J.; Gorski, B.; Lubinski, J.; Narod, S.A. Response to neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res. Treat 2009, 115, 359–363. [Google Scholar]
  112. Caray, L.; Winer, E.; Viale, G.; Cameron, D.; Gianni, L. Triple-negative breast cancer: Disease entity or title of convenience? Nat. Rev. Clin. Oncol 2010, 7, 683–692. [Google Scholar]
  113. Frasci, G.; Comella, P.; Rinaldo, M.; Iodice, G.; Di Bonito, M.; D’Aiuto, M.; Lastoria, S.; Sini, C.; Comella, G.; D’Aiuto, G. Preoperative weekly cisplatin-epirubicin-paclitaxel with G-CSF support in triple-negative large operable breast cancer. Ann. Oncol 2009, 20, 1185–1192. [Google Scholar]
  114. Anders, C.K.; Carey, L.A. Biology, metastatic patterns, and treatment of patients with triple-negative breast cancer. Clin. Breast Cancer 2009, 9, S73–S81. [Google Scholar]
  115. Nofech-Mozes, S.; Trudeau, M.; Kahn, H.K.; Dent, R.; Rawlinson, E.; Sun, P.; Narod, S.A.; Hanna, W.M. Patterns of recurrence in the basal and non-basal subtypes of triple-negative breast cancers. Breast Cancer Res. Treat 2009, 118, 131–137. [Google Scholar]
  116. Dedes, K.J.; Wilkerson, P.M.; Wetterskog, D.; Weigelt, B.; Ashworth, A.; Reis-Filho, J.S. Synthetic lethality of PARP inhibition in cancers lacking BRCA1 and BRCA2 mutations. Cell Cycle 2011, 10, 1192–1199. [Google Scholar]
  117. Farmer, H.; McCabe, N.; Lord, C.J.; Tutt, A.N.; Johnson, D.A.; Richardson, T.B.; Santarosa, M.; Dillon, K.J.; Hickson, I.; Knights, C.; et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434, 917–921. [Google Scholar]
  118. Rottenberg, S.; Jaspers, J.E.; Kersbergen, A.; van der Burg, E.; Nygren, A.O.; Zander, S.A.; Derksen, P.W.; de Bruin, M.; Zevenhoven, J.; Lau, A.; et al. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and combination with platinum drugs. Proc. Natl. Acad. Sci. USA 2008, 105, 17079–17089. [Google Scholar]
  119. Fong, P.C.; Boss, D.S.; Yap, T.A.; Tutt, A.; Wu, P.; Mergui-Roelvink, M.; Mortimer, P.; Swaisland, H.; Lau, A.; O’Connor, M.J.; et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med 2009, 361, 123–134. [Google Scholar]
  120. Tutt, A.; Robson, M.; Garber, J.E.; Domchek, S.M.; Audeh, M.W.; Weitzel, J.N.; Friedlander, M.; Arun, B.; Loman, N.; Schmutzler, R.K.; et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: A proof-of-concept trial. Lancet 2010, 376, 235–244. [Google Scholar]
  121. Rajan, A.; Carter, C.A.; Kelly, R.J.; Gutierrez, M.; Kummar, S.; Szabo, E.; Yancey, M.A.; Ji, J.; Mannargudi, B.; Woo, S.; et al. A phase I combination study of olaparib with cisplatin and gemcitabine in adults with solid tumors. Clin. Cancer Res 2012, 18, 2344–2351. [Google Scholar]
  122. Rouleau, M.; Patel, A.; Hendzel, M.J.; Kaufmann, S.H.; Poirier, G.G. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer 2010, 10, 429–301. [Google Scholar]
  123. Goncalves, A. PARP inhibitors and breast cancer: Update and perspectives. Bull. Cancer 2012, 99, 441–451. [Google Scholar]
  124. Comen, E.A.; Robson, M. Poly(ADP-ribose) polymerase inhibitors in triple-negative breast cancer. Cancer J 2010, 16, 48–52. [Google Scholar]
Figure 1. BRCA1 interacting proteins.
Figure 1. BRCA1 interacting proteins.
Ijms 13 14898f1
Figure 2. BRCA1-mediated homologous recombination (HR) repair. DNA double-strand break (the most destructive and cytotoxic DNA lesion is induced by irradiation or anticancer agents) activates the protein kinase ATM. The MRE11-RAD50-NSB1 (MRN) complex acts as a DSB sensor, recognizes DSB, and recruits ATM to the site of the DNA damage. ATM phophorylates the histone variant H2AX (γ-H2AX) that can directly recruit MDC1. ATM further phosphorylates MDC1, then recruits an E3 ubiquitin ligase, RNF8, that catalyzes polyubiquitin chains at the sites of DNA damage. The ubiquitin polymer next recruits the BRCA1-Abraxas-RAP80 complex through the RAP80 component. BRCA1 forms RING heterodimer E3 ligase activity with BARD1, which is required for recruitment of BRCA2 and RAD51 to damaged sites for HR repair through sister chromatid exchange. Resolvases restore Holliday junctions, and error-free DNA molecules are finally produced.
Figure 2. BRCA1-mediated homologous recombination (HR) repair. DNA double-strand break (the most destructive and cytotoxic DNA lesion is induced by irradiation or anticancer agents) activates the protein kinase ATM. The MRE11-RAD50-NSB1 (MRN) complex acts as a DSB sensor, recognizes DSB, and recruits ATM to the site of the DNA damage. ATM phophorylates the histone variant H2AX (γ-H2AX) that can directly recruit MDC1. ATM further phosphorylates MDC1, then recruits an E3 ubiquitin ligase, RNF8, that catalyzes polyubiquitin chains at the sites of DNA damage. The ubiquitin polymer next recruits the BRCA1-Abraxas-RAP80 complex through the RAP80 component. BRCA1 forms RING heterodimer E3 ligase activity with BARD1, which is required for recruitment of BRCA2 and RAD51 to damaged sites for HR repair through sister chromatid exchange. Resolvases restore Holliday junctions, and error-free DNA molecules are finally produced.
Ijms 13 14898f2
Figure 3. The functional domain of ERα.
Figure 3. The functional domain of ERα.
Ijms 13 14898f3
Figure 4. The model for the mechanism of action of estrogen. An unoccupied estrogen receptor (ER) binds to HSP90 (I). Estrogen binds to the region E ligand binding domain (LBD). Antiestrogen tamoxifen is capable of inducing dimerization and DNA-binding, but does not activate the transcription-activation function 2 (TAF-2). ERE is an estrogen responsive DNA element.
Figure 4. The model for the mechanism of action of estrogen. An unoccupied estrogen receptor (ER) binds to HSP90 (I). Estrogen binds to the region E ligand binding domain (LBD). Antiestrogen tamoxifen is capable of inducing dimerization and DNA-binding, but does not activate the transcription-activation function 2 (TAF-2). ERE is an estrogen responsive DNA element.
Ijms 13 14898f4
Figure 5. Chemical structure of fulvestrant.
Figure 5. Chemical structure of fulvestrant.
Ijms 13 14898f5
Figure 6. The molecular mechanism for the pure steroidal ERα antagonist, fulvestrant. Fulvestrant binds competitively to ERα with a high affinity. It acts as an antiestrogen chemical by reducing the half-life of ERα, resulting in a decrease in expression of ERα. Formation of the drug-receptor complex leads to stabilization of the receptor, which is degraded by an ubiquitin-proteasome complex.
Figure 6. The molecular mechanism for the pure steroidal ERα antagonist, fulvestrant. Fulvestrant binds competitively to ERα with a high affinity. It acts as an antiestrogen chemical by reducing the half-life of ERα, resulting in a decrease in expression of ERα. Formation of the drug-receptor complex leads to stabilization of the receptor, which is degraded by an ubiquitin-proteasome complex.
Ijms 13 14898f6

Share and Cite

MDPI and ACS Style

Ratanaphan, A. A DNA Repair BRCA1 Estrogen Receptor and Targeted Therapy in Breast Cancer. Int. J. Mol. Sci. 2012, 13, 14898-14916. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms131114898

AMA Style

Ratanaphan A. A DNA Repair BRCA1 Estrogen Receptor and Targeted Therapy in Breast Cancer. International Journal of Molecular Sciences. 2012; 13(11):14898-14916. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms131114898

Chicago/Turabian Style

Ratanaphan, Adisorn. 2012. "A DNA Repair BRCA1 Estrogen Receptor and Targeted Therapy in Breast Cancer" International Journal of Molecular Sciences 13, no. 11: 14898-14916. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms131114898

Article Metrics

Back to TopTop