Crosstalk of methylation and tamoxifen in breast cancer (Review)
- Authors:
- Published online on: August 9, 2024 https://doi.org/10.3892/mmr.2024.13304
- Article Number: 180
-
Copyright: © Shen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
As the most common female malignant tumor in the world, breast cancer (BC) has become a major health problem for women due to its high mortality and morbidity rates, and 5-year survival rate of <30% (1). The treatment of BC includes surgery, radiotherapy, chemotherapy, endocrine therapy and targeted therapy (1,2). According to different conditions and needs, it is necessary to develop personalized treatment plans to achieve the best treatment effect (3). Surgery is the main treatment for BC, including radical mastectomy breast-conserving surgery and breast reconstruction (1). Surgical intervention can effectively remove the tumor and reduce the risk of recurrence (4). After surgery, adjuvant therapy (such as radiotherapy and chemotherapy) may be required according to tumor stage, tumor grade, molecular typing and patient status (3). Radiation therapy refers to the use of high-energy rays to kill cancer cells and is often used as an adjunct treatment after surgery to reduce the risk of recurrence (4). For several patients who are inoperable or at high risk of surgery, radiation therapy can also be used as the primary treatment (5). Chemotherapy is a method of killing cancer cells with drugs and can be used to shrink tumors before surgery (6). Endocrine therapy is mainly aimed at patients with hormone receptor-positive BC, which inhibits hormones to block cancer cell growth (3,7). Endocrine therapy drugs include anti-estrogen drugs and aromatase inhibitors (3,7). Targeted therapies are treatments that target specific cancer cells, such as trastuzumab for HER2-positive BC (2), and compared with traditional therapy, has higher pertinence and lower side effects (2).
Endocrine therapy for BC is a long-term treatment (7). Postoperative adjuvant therapy for patients with early BC generally occurs over 5 years, but in some cases may extend to 10 years (8). Risk stratification is clinically performed for early patients with hormone receptor-positive BC. Low-risk patients only need to be treated with a single drug for 5 years, while high-risk patients need intensive treatment (8). On the one hand, endocrine therapy for high-risk patients is supplemented by intensive treatment with ovarian function inhibition or CDK4/6 inhibitors (3). On the other hand, the duration of treatment for high-risk patients was extended to 10 years (7). For medium-risk patients, it is necessary to further determine whether endocrine therapy should last for 5 or 10 years and whether a combination regimen or a single drug regimen is selected according to the clinical characteristics of patients (9). Given that the sources of hormones in premenopausal and postmenopausal women are different, the choice of therapeutic drugs needs to be tailored to different populations (9). For patients with BC with a relatively low risk of recurrence after surgery, selective estrogen receptor modulator (SERM) therapy is often given, of which tamoxifen is a commonly used drug (10).
Tamoxifen is a synthetic non-steroidal anti-estrogen drug, widely used in patients with ER-positive BC, which prolongs the survival of patients (11). Although tamoxifen has made important clinical advances in endocrine therapy, primary and acquired drug resistance has limited its clinical efficacy (12). Downregulation of ERα expression, up-regulation of ERβ expression, the activation of signaling pathways (such as the PI3K/AKT/mTOR signaling pathway), and the activation of certain key proteins and RNA can all lead to tamoxifen resistance in patients with BC (12–14).
Methylation is a common chemical modification in organisms, which affects the expression of DNA, RNA and proteins (15). Enzymes that catalyze DNA methylation are termed DNA methyltransferases (DNMTs) (16). DNMTs are highly expressed in tamoxifen-resistant patients and are important factors for tamoxifen resistance in BC (17). In addition, Jahangiri et al (18) found that promoters of DNMTs are demethylated, which leads to overexpression of DNMTs, promoting tamoxifen resistance and the recurrence of BC. Therefore, the promoters of tamoxifen-resistant cell lines have higher methylation levels, resulting in reduced expression of genes including nuclear receptor-interacting protein 1, human homolog of the Drosophila headcase and the mitochondrial fission protein 1 (19).
In the present review, methylation is introduced in detail, including DNA methylation, RNA methylation and protein methylation. After which, tamoxifen is described and divided into its clinical use and tamoxifen resistance-related mechanisms. In addition, the effect of methylation on tamoxifen resistance, and clustering on the ER and PI3K/AKT/mTOR signaling pathways is described. Furthermore, tamoxifen-induced methylation is also elucidated. Finally, the clinical applications of methylation are also described, with a focus on prognostic analysis and potential clinical drug discovery.
Methylation
Epigenetic modifications mainly include chemical modifications that occur on DNA, RNA and proteins (20). In general, chemical modifications that occur on RNA are termed post-transcriptional modifications and those that occur on proteins are termed post-translational modifications (21). These epigenetic modifications do not change the genetic code of genes, but they have an important effect on gene expression (21). DNA modification is broadly divided into DNA methylation, which silences the expression of genes, and DNA phosphorylation, which affects the structure and function of DNA (22). RNA modifications include N6-methyladenosine (m6A), N6,2′-O-dimethyladenosine (m6Am) and N1-methyladenosine (23). Among them, m6A, the methylation of the N-6 adenosine base, is the most widely studied, affecting splicing, output, stability, degradation and translation, and thus affecting gene expression (24). As for protein modifications, phosphorylation is one of the most abundant post-translational modifications, which is involved in the regulation of cell signal transduction, protein-protein interaction and gene transcription (25). Ubiquitination, acetylation and methylation can also affect protein-protein interactions, protein stability, subcellular localization or enzymatic activity (26).
Methylation refers to the process of transferring methyl groups from active methyl compounds to other compounds, which results in the formation of various methyl compounds, or chemical modifications in proteins or nucleic acids to form methylated products (27,28). Based on the substrate to which the methyl group binds, methylation is mainly divided into DNA methylation, RNA methylation and protein methylation (Fig. 1) (28). Methyl donors are mainly derived from one-carbon metabolism (29), which is a one-carbon unit metabolic process, including folate cycle, the methionine cycle and trans-vulcanization pathway, wherein the methionine cycle is the main pathway to produce methyl donors (29). In the methionine cycle, one carbon unit can be used to remethylate homocysteine to produce methionine (30). Methionine produces s-adenosylmethionine (SAM) with the help of methionine adenylyl transferase, and transfers methyl groups to biomolecules, including proteins, nucleic acids and lipids (30). The change of methylation status is caused by the difference in the activity of methyltransferase and demethylase (31), with SAM as the main methyl donor of these enzymes (31). Alterations in the methylation of proteins, nucleotides and metabolites can lead to the occurrence of cancer (31).
DNA methylation
DNA methylation is a chemical modification process, in which the cytosine of vertebrate CpG dinucleotides is catalyzed by DNMTs and acquires a methyl group using SAM as the methyl donor (27). DNA methylation occurs on the N-6 of adenine, N-7 of guanine, and C-5 of cytosine (31). However, DNA methylation primarily occurs on the cytosine of 5′-CpG-3′, resulting in the formation of 5-methylcytosine (27). A portion of CpG dinucleotides is dispersed throughout the genome, while another portion occurs in dense clusters of CpG islands (32). In normal tissues, most of the dispersed CpG is methylated, while CpG islands tend to be unmethylated (32). Normally, CpG dinucleotides in relatively useless or unfavorable genomes are rare and are always in a methylated state (33). By contrast, CpG islands rich in CpG dinucleotides with a size of 100-1,000 bp in the genome are always unmethylated, and CpG islands are often located near the transcriptional regulatory region and are associated with more than half of the genes encoded (33). Therefore, it is necessary to study the methylation of CpG islands in the transcription region.
The hypermethylation of CpG sites in enhancers or promoters leads to transcriptional silencing, whereas the hypomethylation of CpG sites usually leads to transcriptional activation, so methylation regulates gene expression through gene transcription (27). DNA methylation is mainly dependent on the DNMT family, which has five members including DNMT1, DNMT2, DNMT3A, DNMT3B and DNMT3L (27). DNMT1 is primarily involved in DNA methylation, which is required to silence tumor suppressor genes (16). DNMT2 is an RNA methyltransferase that modifies cytosine residues in certain tRNA anticodon rings (16). The key role of DNMT3A and DNMT3B is de novo methylation (34) Although DNMT3L is a member of the DNMT3 family, it does not have methyltransferase activity (34), however, DNMT3L can assist DNMT3A/B-mediated de novo methylation (35). In addition, DNMT3A includes two isoforms, while DNMT3B contains >30 isoforms, all of which have methylation activity and conserved C-terminal domains (35). In conclusion, DNMT1 participates in DNA methylation, whereas DNMT3A and DNMT3B, whose primary role is de novo methylation, could also be involved in DNA methylation (16).
DNA methylation has been detected at a very early stage, and it is also a common epigenetic phenomenon, which plays an important role in the maintenance of the stability of the genome and the regulation of normal physiological functions (36). In the process of tumorigenesis, there is a decrease in the DNA methylation level of most genes and an increase in the methylation level of CpG islands of some genes, including DNA repair genes, cell cycle genes and apoptosis genes (36). Low levels of DNA methylation not only lead to reduced genomic stability and mutation rates, but also abnormally activate the expression of multiple oncogenes, such as chromatin modifiers and transcription factors (37). By contrast, high levels of DNA methylation indirectly induce malignant tumors by decreasing the transcriptional activity of tumor suppressor genes and then affecting the expression (37).
RNA methylation
In addition to DNA methylation, RNA can also be methylated to participate in the regulation of gene expression (38). RNA methylation is a post-transcriptional modification that transfers methyl groups from methyl donors to RNA bases with the help of RNA methyltransferase, thereby regulating stability, splicing, localization and translation (38,39). RNA methylation modification is reversible and occurs in different types of RNA, including microRNA (miRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), small nuclear RNA (snRNA) and small nucleolar RNA (38,40).
RNA methylation occurs at the m6A methylation modification at adenylate N6, which is the most abundant RNA modification found in eukaryotes (41). The m6A methylation has its methylase (writer), demethylase (eraser) and methylation recognition protein (reader), which synergistically mediate RNA methylation (41). The m6A methylation in mRNA is mainly catalyzed by the METTL3/METTL14 complex (40). METTL3 primarily acts as a catalyst, while METTL14 is an allosteric activator that helps bind to the target RNA (40). METTL3 is mainly located in the nucleus, but METTL3 is present in the cytoplasm in several cells (42). The majority of m6A methylation occurs in rRNA, mediated by the N6-adenosine-methyltransferase zinc finger CCHC-type containing 4 and methyltransferase-like 5/tRNA methyltransferase activator subunit 11-2 complexes (41). In addition, m6A methylation of snRNA U6 is catalyzed by METTL16, which also catalyzes small amounts of mRNA and other non-coding RNA, in particular microRNAs and lncRNAs (43). The m6A demethylase is an enzyme that converts m6A to adenylate, thereby removing m6A, and this process is catalyzed by obesity-associated protein (FTO) and AlkB homolog 5 (ALKBH5) (43). FTO catalyzes the demethylation of 3-methyluridine, m6A and m6Am, but which RNA is mainly targeted is still controversial (44). Furthermore, m6Am methylation in snRNA may be a specific target of FTO, and its mechanism remains to be further revealed (44). Although ALKBH5 is a demethylase, it has no effects on m6Am (45). As for m6A recognition proteins, it is usually recruited by m6A methylation, which affects mRNA functions such as localization (46). The m6A methylation recognition proteins include the YTH domain family (YTHDF1/2/3 and YTHDC1/2), hnRNP family (hnRNPC, hnRNPG and hnRNPA2B1) and insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) (46). The YTH domain can directly bind to m6A, and YTHDF2 can recruit complexes to promote the degradation of m6A-modified mRNA (47). By contrast, IGF2BPs can enhance the stability and translation efficiency of m6A-modified mRNA (48). HnRNPC selectively recognizes m6A-induced splicing in the secondary structure of mRNA (49).
Protein methylation
Protein methylation refers to the methylation of arginine or lysine in the protein (50). Arginine can be methylated once or twice by arginine methyltransferase, whereas lysine can be methylated 1-3 times by lysine methyltransferase (51). Among them, arginine methyltransferase transfers two methyl groups to the same nitrogen atom of the arginine polypeptide to form asymmetric dimethylarginine, and one methyl group is added to each nitrogen terminal to form symmetric dimethylarginine (51).
Among the protein methylation, the methylation of histones has been studied most extensively (51). Histone methylation is the methylation of lysine or arginine residues on the N terminal of the H3 or H4 histones (51,52). The effects of histone methylation are mainly reflected in heterochromatin formation, gene imprinting, X chromosome inactivation and transcriptional regulation (53). This process is catalyzed by histone methyltransferase (HMT), which can be divided into histone lysine methyltransferase and protein arginine methyltransferase (PRMT) (53). H3 lysine (H3K) 4, 9, 27, 36, 79 and H4 lysine (H4K) 20 can be methylated, with histones H3K4 and H3K9 being the two most common modification sites (54). Likewise, the histone lysine residues can undergo single, double or trimethylation, while the arginine residues undergo only mono-methylation and di-methylation (54). The methylation of different sites of histone H3 and H4 and the amount of methylation have different influences on transcriptional regulation. In most cases, H3K9me3, H3K27me3 and H4K20me2/3 mediate transcriptional suppression, while H3K4me1/3, H3K9me1, H3K27me1, H3K36me1/3 and H3K79me1/3 mediate transcriptional activation (54).
In addition to HMT, there are also histone demethylases. Histone demethylases can be divided into two families: i) The lysine-specific demethylase (LSD) family; and ii) the Jumonji C domain-containing (JMJD) family (55). LSD can specifically remove the mono-methylation and di-methylation of histones H3K4 and H3K9, while JMJD can remove the tri-methylation of lysine (55). Therefore, methylase and demethylase together promote the stability and dynamics of histone methylation.
Tamoxifen
Tamoxifen in a clinical setting
In 1978, the FDA approved tamoxifen for the treatment of patients with advanced BC (56). The 2010 ASCO guidelines recommend tamoxifen as the standard for premenopausal women, and at present has become an important drug for adjuvant endocrine therapy of BC (57).
Currently, tamoxifen is mainly used in patients with hormone receptor-positive BC, and it is mostly used in premenopausal patients or postmenopausal patients who cannot tolerate aromatase inhibitors (58). In addition, tamoxifen can also be used in the following situations: i) To treat recurrent metastatic BC and ovarian cancer (59); ii) for adjuvant treatment of lymph node-negative BC after breast surgery, and radiotherapy and chemotherapy (60); iii) it is used for the adjuvant treatment of postmenopausal breast surgery and lymph node-positive BC after radiotherapy and chemotherapy (60); and iv) for patients with ductal carcinoma in situ after breast surgery and radiation as it may reduce the risk of invasive BC (61). Moreover, clinical studies suggest that for women with a family history of BC, the use of tamoxifen can reduce their risk of BC by more than one-third (62–65). Therefore, tamoxifen is also used for BC prevention. The conventional dose of tamoxifen for BC is 20 mg, but it can also be increased to a maximum of 40 mg/day (66), and can be taken by mouth as a single dose or in two equal doses (66). The conventional endocrine treatment cycle is 5 years, and the dose of the drug and the treatment drug can also be changed after 2 years of tamoxifen treatment (67). However, long-term use of tamoxifen leads to a series of adverse reactions, including secondary estrogen effects, gastrointestinal reactions, neuropsychiatric symptoms and bone marrow suppression (68). Secondary antiestrogenic effects include facial flushing, vulvar pruritus, menstrual disorders, amenorrhea, increased leucorrhea and vaginal bleeding (68,69). Gastrointestinal reactions include loss of appetite, nausea, vomiting and diarrhea (69). Neuropsychiatric symptoms include headache, dizziness and depression (68). Furthermore, several patients may suffer from transient leukopenia and thrombocytopenia (69). In addition, large doses and long-term application can lead to visual impairment, rash, hair loss, weight gain and liver dysfunction (70). However, most patients experience relatively mild symptoms, which can be overcome and will be relieved after stopping the drug in later stages (70). However, it must be noted that the use of tamoxifen may be associated with more serious adverse effects including venous thrombosis and endometrial cancer (EC), but this is less likely to occur (71). Therefore, patients should have a gynecological examination once a year during the use of tamoxifen (71).
Tamoxifen is an estrogen receptor modulator, and its main target is ERα (72). ERα promotes intracellular signaling primarily through estrogen/ERα-mediated nucleus-initiated steroid signaling (genomic signaling) (72). The process can be divided into three steps. First, estrogen enters the cell through diffusion or in situ synthesis. Second, estrogens bind to ERα in the nucleus, which activates and forms ERα homodimers or heterodimers (73). Finally, the activated ERα binds to the DNA enhancer estrogen response element (ERE), so that the ERα-ERE complex promotes the formation of the transcription initiation complex and induces transcription (74). In addition to the ERE mechanism, ERα binds to other transcription factors and then binds to the activating protein 1 at the activating region of the target genes to regulate gene transcription (73). By binding with ERα in BC, tamoxifen blocks the binding of estrogen to ERα, making estrogen inactive, either blocking stimulation of transcription or weakening its effect, thus inhibiting the occurrence and development of BC (75). In addition, tamoxifen can up-regulate the production of transforming growth factor β, which makes tamoxifen suitable for patients with osteoporosis (76).
Tamoxifen resistance
After long-term use of tamoxifen, some patients will develop tamoxifen resistance, and its specific molecular mechanism is complex and diverse. It includes downregulation of ERα, up-regulation of ERβ, the emergence of BC stem cells (BCSCs) and activation of the signaling pathway (Fig. 2) (12).
A number of studies indicate that the inhibition of ERα expression may be the main cause of resistance to endocrine therapy (12,14). Given that the mechanism of tamoxifen is mainly mediated by ERα and the expression of ERα is a good predictor of response to tamoxifen, loss of expression of ERα has been widely recognized as a major factor in tamoxifen resistance (12). Loss of ERα expression may be associated with methylation of CpG islands and increased histone deacetylation, resulting in a more compact nucleosome structure that restricts ERα transcription and thus limits the efficacy of tamoxifen (77). Moreover, loss of ERα has been associated with tumor invasion and suggests a poor prognosis. In addition, a study showed that re-expression of ERα can reverse tamoxifen resistance in MCF-7 cells, suggesting that loss of ERα expression may be an important mechanism of tamoxifen resistance (12). Similarly, ERα protein point mutations (such as K303R) enhance ERα-mediated cell growth, which is caused by increased estrogen sensitivity and alters various cellular signaling pathways (78). Since these signaling pathways normally downregulate ERα signaling, the ERα signaling pathway is inhibited and patients become resistant to tamoxifen (78). There are also multiple ERα mutations in the ligand-binding region of ERα, and in the absence of ligands, these mutations promote ERα conformational changes that lead to the proliferation of hormone-independent tumor cells and resistance to tamoxifen (12). ERβ is a product of ESR2 located at chromosome 14q.21 (79). ERβ and ERα have 96% homology in the DNA binding region and 59% homology in the ligand binding region (79). Interestingly, the activation of ERβ expression enhances tamoxifen resistance, which is mainly determined by the physiological function of ERβ (12). ERβ is highly homologous to ERα and binds to estrogen with a similar affinity to ERα (80). ERα and ERβ respond in different ways depending on the ligands and the responding elements (81). ERα up-regulates the expression of genes related to cell growth (81). However, ERβ is more abundant than ERα expression in normal breast cells (72). In ERβ knockout mice, the proliferation of mammary cells accelerated, whereas in ERα knockout mice, the mammary cells shrank (72). ERβ has various effects on ERα-related regulatory genes, which include regulating the expression of the ERα gene and enhancing or weakening the effect of ERα (82). Overall, ERβ inhibited ~70% of ERα-regulated genes (82). In contrast to tamoxifen-sensitive tumors, ERβ was ~2 times higher in tamoxifen-resistant tumors than ERα (12). Thus, ERβ expression activated by ERβ-selective agonists, such as LY500307, may promote tamoxifen resistance by inhibiting ERα (72). However, studies have also shown that some ERβ splicing variants can lead to the development of tamoxifen resistance, which is associated with a poorer prognosis in advanced BC (79,83–85). In addition, the expression of ERβ in tumor infiltrating leukocytes was much higher than that of ERα in the tumor microenvironment, suggesting that ERβ may alter the tamoxifen response by influencing immune cells (72).
Except for ER, abnormal activation of signaling pathways can also lead to tamoxifen resistance. The related signaling pathways include the PI3K/AKT/mTOR signaling pathway, the NF-κB signaling pathway and the Hedgehog signaling pathway (75,78,86). The most important of these is the PI3K/AKT/mTOR signaling pathway (87). The PI3K/AKT/mTOR signaling pathway regulates multiple biological processes, including cell proliferation, apoptosis and metabolism (87). This signaling pathway is a complex regulatory network involving multiple components including upstream regulation, internal regulation and downstream regulation. Its upstream regulation is receptor tyrosine kinase (RTK) (88). When the upstream protein is activated by external stimuli, such as growth factors, the RTK undergoes a conformational change that activates its tyrosine kinase activity (88). The activated RTK binds directly to p85, the subunit of PI3K, causing p85 to release its inhibition on p110, thereby activating PI3K (89). Activated PI3K converts phosphatidylinositol diphosphate (PIP2) to phosphatidylinositol triphosphate (PIP3) (89). After which, PIP3 attracts PDK1 and AKT, enabling PDK1 to phosphorylate AKT at Thr308, thereby activating AKT (90,91). In addition to AKT, the inhibitory factor of internal regulation is PTEN (92). PTEN can dephosphorylate PIP3 to PIP2, which limits the activation of the signaling pathway (92). Therefore, the absence or abnormal function of PTEN leads to overactivation of the PI3K/AKT/mTOR signaling pathway (93). Activated AKT phosphorylates multiple downstream target proteins and thus participates in cellular physiological processes. Phosphorylated proteins targeted by AKT include mTOR, FOXO, GSK3, Bcl-2 and NF-κB (88,94–96). Activation of the PI3K/AKT/mTOR signaling pathway was found to cause tamoxifen-resistant cells to become resistant to DNA-damaged chemotherapy by up-regulating BARD1 and BRCA1, suggesting that the PI3K/AKT/mTOR signaling pathway is important in the treatment of BC (97). In addition, high expression of phosphorylated AKT is associated with poor prognosis, and the inhibition of AKT expression is conducive to activation of drug-resistant cells (87). While multiple drugs targeting the PI3K/AKT/mTOR signaling pathway have been used to overcome tamoxifen resistance, inhibiting this pathway will activate the compensatory mechanism due to the complexity of the PI3K/AKT/mTOR pathway, which limits the effects of inhibitors (87).
The cancer stem cell model is another important factor in BC resistance to tamoxifen (89). BCSCs refer to a small subset of BC cytoplasmic cells that have the ability to self-renew, differentiate and perform tumorigenesis (98). Clinically, BCSCs are considered to be relatively resistant to radiotherapy, chemotherapy and molecularly targeted therapy, leading to the development of drug resistance and cancer recurrence (99). CD44+/CD24− and ALDH+ are the most common molecular markers of BCSCs, and other surface proteins are also considered markers of BCSCs, such as CD133, CD61, C-X-C chemokine receptor type 4 (CXCR4) and microsatellite instability (100,101). Ectopic expression of ERα mutations (such as Y537S, Y537N and D538G) enriches CD44+/CD24− cells, increases the formation of mammospheres, and upregulates a variety of stemness genes such as octamer-binding protein-4, SRY-box transcription factor 2, SRY-box transcription factor 9 (SOX9) and B-cell-specific Moloney murine leukemia virus integration site 1, thereby promoting BCSC enrichment and leading to endocrine resistance (102). Furthermore, PI3K/AKT/mTOR, Notch, Wnt and Hippo signaling pathways can also promote the enrichment of BCSCs, thus leading to the generation of endocrine therapy resistance (103). Based on the study of these BCSCs models, a number of therapeutic options are being gradually introduced to the clinic, including multi-drug chemotherapy and molecular targeted therapy (100). For example, the γ-secretase inhibitor MK-0752 in combination with tamoxifen or letrozole in patients with early-stage BC, and the γ-secretase inhibitor RO4929097 in combination with exemestane in patients with advanced BC are already in clinical trials (104–107).
In addition to the aforementioned three mechanisms, other proteins and RNAs play a crucial role in the complex network of tamoxifen. For example, SOX9, HDAC1, SIRT1 and HIF-1αcan promote BC resistance to tamoxifen (108–111). Furthermore, several miRNAs (such as miR-342 and miR-375) and LncRNA can also alter the BC response to tamoxifen through multiple mechanisms (112–114).
Effects of methylation on the tamoxifen response
Estrogen receptor
In the majority of cases, methylation affects tamoxifen sensitivity and resistance by altering transcription of the ESR1 gene and ERα protein-mediated transcription (115–118). In general, up-regulation of ERα protein or enhanced ERα-mediated transcriptional regulation promotes tamoxifen sensitivity (Table I; Fig. 3) (12). By contrast, inhibition of ERα protein expression or reduction of ERα-mediated transcriptional regulation may promote tamoxifen resistance (Table I; Fig. 3) (12).
Firstly, the methylation of the ESR1 promoter affects ESR1 transcription and ERα protein expression, thus playing an important role in BC response to tamoxifen. DNA methyltransferase-mediated hypermethylation of the ESR1 promoter is associated with poor prognosis and indicates the development of tamoxifen resistance (115). The methylation of H3 is also involved in ESR1 transcription and thus affects the tamoxifen response. MLL3 and SET1A-mediated methylation of histone H3K4 enhance ESR1 transcription, thereby promoting sensitivity to hormone therapy (116). In addition, ZEB1 and IL-1β also promote tamoxifen resistance by promoting the methylation of ESR1 (117,118). ZEB1 induces hypermethylation of the ESR1 promoter and silencing of ERα by forming the ZEB1/DNMT3B/HDAC1 complex on the ESR1 promoter, leading to resistance to anti-estrogenic drugs (117). IL-1β induces EMT by activating the IL-1β/IL-1RI/β-catenin pathway, thereby enabling TWIST1 to induce methylation of the ESR1 promoter, which leads to reduced expression of ERα and thus increased tamoxifen resistance (118). Similarly, loss of demethylase also mediates tamoxifen resistance. Deletion of the DNA demethylase TET2 promotes the methylation of ESR1, thereby downregulating the expression of ERα and thus promoting tamoxifen resistance (119). Interestingly, ERβ showed the exact opposite effect to ERα in response to tamoxifen. In tamoxifen-resistant BC, ERβ is hypomethylated, suggesting that ERβ hypermethylation harms tamoxifen resistance (120).
Secondly, the methylation-mediated enhanced transcription of ERα-targeted genes can promote tamoxifen sensitivity, and vice versa. For example, ANCCA mediates the recruitment of MLL1 HMT at the promoters of ESR1 target genes for H3K4 methylation associated with gene activation, which may effectively induce tamoxifen sensitivity (121). Similarly, SETD1A is also involved in H3K4 methylation, subsequent ERα recruitment, and transcription of ERα target genes (122). Conversely, inhibition of ESR1 transcription or ERα-mediated transcription promotes BC resistance to tamoxifen. Hypermethylation of estrogen response enhancers leads to reduced binding to ERα and downregulation of key regulators of ERα activity, resulting in weakened endocrine responses (123). By contrast, the hypomethylation of enhancers plays a role in the transformation of normal breast cells into endocrine-reactive BC (123). Overall, the methylation of the ESR1 enhancer has potential in endocrine stratification therapy (123). Moreover, EZH2-mediated trimethylation of H3K27 promotes BC susceptibility (124). The single nucleotide polymorphism of EZH2 further promotes the methylation of H3K27, which inhibits the transcription, thereby reducing overall survival (OS) and progression-free survival in patients that are ER-positive/tamoxifen-treated (124). EZH2 may also promote the methylation of the GREB1 promoter through DNMT1 and DNMT3B, thereby silencing GREB1 and inhibiting ERα transcription, thus promoting tamoxifen resistance (125). Moreover, TET1 and TET3 promote demethylation of the UCHL1 promoter, thereby promoting UCHL1 transcription, which further downregulates ERα expression via the UCHL1-KLF5 axis, leading to tamoxifen resistance (126).
Finally, the methylation can also respond to tamoxifen through promoter methylation of ERα-associated genes. Among them, the upstream genes of ERα include p21 (127), WT1 (128) ID4 (129) and miR-27b (130). Activation of p21 activates ERα transcription in ER-negative BC, thereby activating the ERα signaling pathway (129). LncRNA UCA1 interacts with the enhancer of EZH2, which inhibits p21 expression through H3K27 histone methylation on the p21 promoter, thereby promoting tamoxifen resistance (131). In the development of tamoxifen resistance, WT1 is involved in the expression of ERα (128). Through RNA-seq and TCGA databases, Ren et al (132) found that WT1 was hypermethylated and upregulated in all molecular subtypes of BC, which was closely related to the efficacy of tamoxifen in patients with BC. By contrast, ID4 inhibits ERα expression and regulates estrogen biosynthesis (129). In BC resistant to tamoxifen, ID4 is hypomethylated, suggesting that it may be the key to identifying drug resistance (133). However, there was no difference between ID4 hypermethylation and ID4 hypomethylation on the risk of disease progression (P=0.287) (133). As for miR-27b, it targets ERα to exert its anti-proliferation and anti-metastasis effects (130). The methylation of the miR-27b promoter promotes activation of HMGB3, leading to tamoxifen resistance (134). The downstream genes of ERα are relatively rich, including NAT1 (135), ELOVL2 (136) and PRA (137). High expression of ERα enhances the expression of NAT1, ELOVL2 and PRA, so that the expression level of ERα is positively associated with NAT1, ELOVL2 and PRA (135–137). The increased methylation of these gene promoters leads to decreased expression, which may lead to the dysfunction of ERα and induce tamoxifen resistance (138–143). Using methylation-specific PCR and bisulfite genomic sequencing, Kim et al (138) found that the methylation of NAT1 was significantly enhanced in tamoxifen-resistant BC, while the methylation of COMT, CYP1A1, CYP2D6 and SULT1A1 was not significantly altered compared with the control group. Therefore, hypermethylation of NAT1 may influence the initiation of tamoxifen resistance (138). LncRNA H19 mediates methylation of the NAT1 promoter to downregulate NAT1 expression, which leads to tamoxifen resistance in BC (139). Similarly, the hypermethylation of the ELOVL2 promoter and the PRA promoter downregulate ELOVL2 and PRA to drive tamoxifen resistance (140,141). Notably, the majority (74%) of patients with PRA in PRA-negative BC did not exhibit methylation status (141). However, hypermethylation of downstream genes negatively associated with ERα expression also promotes tamoxifen resistance, such as PAX2 (142) and E-cadherin (143). Estrogen receptors promote methylation of the PAX2 promoter, thereby downregulating its expression (142). In patients with tamoxifen-resistant BC, abnormally elevated methylation of PAX2 promoter leads to decreased expression of PAX2 mRNA (144). The estrogen/ERα signaling pathway downregulates E-cadherin to promote BC (143). The methylation of E-cadherin leads to the downregulation of E-cadherin expression, promoting the upregulation of Twist and the occurrence of EMT, thus resulting in tamoxifen resistance (145). In addition, although ERα upregulates the expression of MMP-1, the hypomethylation of the MMP1 promoter leads to its overexpression, thereby inducing tamoxifen resistance in BC (146,147). As for ERβ, estrogen inhibits the expression of PTPRO through ERβ, thus inducing the occurrence of BC (148). The hypomethylation of the PTPRO promoter elevates its expression, thus promoting the sensitivity of tamoxifen (149).
PI3K/AKT/mTOR
In addition to ERα, the methylation-mediated PI3K/AKT/mTOR signaling pathway and methylation on the PI3K/AKT/mTOR signaling pathway also influence tamoxifen resistance (12,14). In short, methylation-induced activation of the PI3K/AKT/mTOR signaling pathway contributes to tamoxifen resistance (Table II; Fig. 4) (14). By contrast, methylation-induced inactivation of the PI3K/AKT/mTOR signaling pathway promotes tamoxifen sensitivity (Table II; Fig. 4) (14).
Above all, the key proteins in the PI3K/AKT/mTOR signaling pathway (mainly including PTEN and AKT) can be methylated to reduce their expression, resulting in an altered response to tamoxifen. Low expression of PTEN due to hypermethylation of the PTEN promoter (−819 to −787 bp) predicts poor disease-free survival (DFS) and OS in patients with hormone-receptor-positive BC treated with tamoxifen (150). More precisely, in the −819 to −787 bp region of the promoter, only the hypermethylation of −796 CpG islands, but not the hypermethylation of the remaining four CpG islands, predicted shorter DFS and OS (150). A total of two sites in the PTEN promoter can be methylated by DNMT1, thereby downregulating the expression of PTEN and increasing phosphorylation of AKT, leading to tamoxifen resistance (151). By contrast, miR-146b reduced NF-κB-mediated MAT2A expression, which inhibited SAM-mediated methylation of the PTEN promoter to restore PTEN expression, thereby reversing tamoxifen resistance (152). Furthermore, SETDB1 regulates the expression of ER and AKT target genes by mediating the methylation and phosphorylation of AKT through interaction with PELP1, thus promoting tamoxifen resistance (153).
Next, the methylation of genes involved in inhibiting the PI3K/AKT/mTOR signaling pathway activates the PI3K/AKT/mTOR signaling pathway, thereby inducing tamoxifen resistance. Firstly, as for the AKT protein in PI3K/AKT/mTOR signaling pathway, ERRFI1 (154), PITX2 (155) and DOK7 (156) all can downregulate the expression of phosphorylated AKT, thereby inhibiting the PI3K/AKT/mTOR signaling pathway. The downregulated expression of ERRFI1 (157), PITX2 (158) and DOK7 (159) due to the hypermethylation of their promoters leads to impaired inhibition of the PI3K/AKT/mTOR signaling pathways, thereby inducing tamoxifen resistance in BC. Moreover, the silence of AK4 can downregulate the expression of p-AKT (160). METTL3-mediated increased methylation at multiple m6A sites of the 5′-UTR of AK4 mRNA stabilizes AK4 mRNA and thus increases ROS and p38 levels, leading to tamoxifen resistance (161). Secondly, inhibition of PTEN can also activate the PI3K/AKT/mTOR signaling pathway. In tamoxifen-resistant BCs, the SALL2 promoter is hypermethylated, which inhibits SALL2 expression and leads to SALL2-mediated transcription suppression of ESR1 and PTEN, thus promoting the AKT/mTOR signaling pathway and leading to tamoxifen resistance (162). Thirdly, the activation of mTOR also ensures the function of PI3K/AKT/mTOR. The methylation of the ZDHHC22 promoter leads to the low expression of ZDHHC22 in ER-negative BC, which leads to the activation of the mTOR, resulting in tamoxifen resistance (163). In addition, the reduction of p16 (164) and GDF15 (165) inhibits the occurrence and development of cancer through the inhibition of PI3K/AKT/mTOR signaling pathway. EZH2 downregulates p16 by promoting methylation of p16, which regulates the cell cycle and leads to tamoxifen resistance (166). In tamoxifen-resistant BC, the promoter of GDF15 is hypermethylated, which leads to low expression and tamoxifen resistance (167).
After which, the hypomethylation of upstream promoter genes of the PI3K/AKT/mTOR signaling pathway promotes tamoxifen methylation via the PI3K/AKT/mTOR pathway. For example, ATF3 mediates the PI3K/AKT/mTOR signaling pathway by activating AKT phosphorylation, thereby increasing radiation resistance in BC (168). In BC, low expression of YTHDF2 leads to reduced hypomethylation of the 5′-UTR of ATF3 mRNA, resulting in the stabilization of ATF3 mRNA, which stimulates the expression of ABCB1 and leads to tamoxifen resistance (169). Similarly, the hypermethylation of downstream genes of the PI3K/AKT/mTOR signaling pathway also promotes tamoxifen resistance. The PI3K/AKT/mTOR signaling pathway regulates phagocytosis in macrophages through Beclin1 (170). LncRNA H19 binds and inhibits S-adenosylhomocysteine hydrolase, which inhibits DNMT3B binding to the Beclin1 promoter and reduces Beclin1 methylation, thus leading to autophagy dysregulation and tamoxifen resistance in BC (171).
Ultimately, the hypermethylation of upstream promoter genes of the PI3K/AKT/mTOR signaling pathway leads to tamoxifen resistance, which may be mediated by pathways other than the PI3K/AKT/mTOR signaling pathway. For instance, TROP2 promotes cell proliferation and migration through the PI3K/AKT/mTOR signaling pathway (172). However, in tamoxifen-resistant BC cells, the promoter of TROP2 is methylated and silenced, which is meditated by DNMT1 (173).
Others
Except for ERα protein and PI3K/AKT/mTOR signaling pathways, the methylation of several other genes can influence BC sensitivity and resistance to tamoxifen. This can be divided into two components: i) Hypermethylation-mediated tamoxifen resistance (or hypomethylation-mediated tamoxifen sensitivity); and ii) hypomethylation-mediated tamoxifen resistance (or hypermethylation-mediated tamoxifen sensitivity).
For the first part, hypermethylation-mediated tamoxifen resistance mainly includes VGLL4 (174), DPYD (175), ZNF350 (176) and MAGED1 (177). HAGLR inhibits VGLL4 expression by promoting DNMT1-mediated DNA hypermethylation, thereby promoting tamoxifen resistance (174). In tamoxifen-resistant BC cells, methylation of the DPYD promoter region leads to a decrease in DPYD mRNA (175). In addition, when the methylation inhibitor was applied in tamoxifen-resistant BC, the methylation of ZNF350 and MAGED1 promoters was significantly reduced, suggesting that ZNF350 and MAGED1 may play a role in tamoxifen resistance (176).
For the second part, hypomethylation-mediated tamoxifen resistance mainly includes TSTD1 (177), LDH (178), PAST1 (179) and GNB4 (180). The hypomethylation of the TSTD1 promoter leads to upregulation of TSTD1, which is associated with adverse reactions to tamoxifen therapy in patients with BC (177). In tamoxifen-treated cells, less methylation of the LDH promoter led to increased LDH expression, suggesting that LDH expression could promote tamoxifen resistance (178). Similarly, PSAT1 mRNA expression inhibited by hypermethylation of the PSAT1 promoter predicts a good prognosis after tamoxifen treatment (179). Moreover, DNMT3B-mediated methylation of GNB4 leads to silencing of GNB4, which promotes tamoxifen sensitivity (180).
Effects of tamoxifen on methylation
The degree of methylation is also altered after treatment with tamoxifen. However, there are relatively few studies on this area and they are relatively less comprehensive. Overall, tamoxifen reduces the level of methylation (109,181–183). Treatment of male rat embryos with tamoxifen resulted in increased methylation of multiple imprinted genes (such as Grb10, Igf2r, and Kcnq1), leading to downregulation of the expression of these imprinted genes (181). In BC, tamoxifen increases CXCL12 expression through reducing the methylation of CXCL12 promoters, thus making cells less susceptible to exogenous CXCL12 attraction to metastatic sites (178). In addition, tamoxifen altered the methylation of the ESR1 promoter in patients with BC (182). Tamoxifen induces PRMT5 to translocate to nucleus, where it methylates the ERα protein, which causes corepressor proteins such as SMRT and HDAC1 to bind to the target gene promoter of ERα, thereby inhibiting transcription and cell proliferation (109). By contrast, in tamoxifen-resistant cell lines, PRMT5 is predominantly localized in the cytoplasm, suggesting that PRMT5 in the nucleus is a biomarker of tamoxifen sensitivity (109). For the embryonic development of sperm, the methylation of IGF2-H19 ICR in sperm is reduced after tamoxifen treatment, triggering the sperm to acquire paternal imprints and ensuring embryonic development (183). However, there was no significant increase or decrease in the overall methylation level of the rats (183).
It has been reported that after long-term tamoxifen treatment, the body may induce the formation of second tumors, especially EC, which may be related to tamoxifen-induced methylation impairment (Table III) (184–186). For example, the application of tamoxifen leads to the hypomethylation of the promoters of CXCR4 and CXCL12 by promoting the formation of DNA methyltransferase 3B4 splice variant, thereby up-regulating the expression of CXCR4 and CXCL12 in EC, thus promoting cell proliferation and metastasis (185). Moreover, tamoxifen induces hypomethylation of the PAX2 promoter, thereby activating PAX2 expression, which induces EC (186). In addition to this, the use of tamoxifen can also contribute to the development of liver cancer through methylation (187). Tryndyak et al (187) reported that the application of tamoxifen reduces the expression of DNMT1, DNMT3a and DNMT3b, which leads to the decrease of liver DNA methylation, thus inducing liver cancer.
Clinical application
Given the complex crosstalk between methylation and tamoxifen, it has multiple guidelines for clinical applications. Specifically, this can be divided into two parts, namely, the prediction of prognosis and recurrence of patients with BC treated with tamoxifen, and the exploration of potential clinical drugs.
Simply put, the higher degree of methylation, the worse prognosis of patients and the greater risk of relapse. Using the Illumina HumanMethylation450 BeadChip, Williams et al (176) found that tamoxifen-resistant cell lines share 3,000 hypermethylated and 200 hypomethylated CpG islands. For example, the proportion of patients with PITX2 hypomethylation who did not have metastases after 10 years of tamoxifen treatment was higher than that of patients with PITX2 hypermethylation (158). Similarly, DOK7 CpG is hypermethylated in leukocytes of patients which are tamoxifen-resistant, therefore the degree of DOK7 methylation is important for early diagnosis of tamoxifen resistance and prevention of cancer recurrence (159). Compared with the inactivation of genes due to promoter hypermethylation, the activation of growth-promoting genes due to promoter hypomethylation was also observed in tamoxifen-resistant cells (188). PSAT1 mRNA expression inhibited by hypermethylation of the PSAT1 promoter predicts a good prognosis after tamoxifen treatment (179).
In addition, methylation of different promoters of some genes can result in different prognoses. The methylation of promoters in the U region of GR is associated with poorer OS, while methylation of promoters in the C region of GR is associated with improved OS (189). Thus, methylation of promoters in specific regions of GR can suggest a poor prognosis in patients who do not receive tamoxifen (189).
As for potential clinical drugs, the majority promote BC sensitivity to tamoxifen by affecting DNMT. The combination of resveratrol and astragalus leads to the downregulation of DNMT activity and decreases the methylation of the ESR1 promoter, thus promoting the expression of ERα, which promotes tamoxifen sensitivity (190). Likewise, arsenic trioxide (191) and 5-aza-CdR (192) also inhibit DNMT1-mediated methylation of ESR1, thereby promoting ERα expression and thus tamoxifen sensitivity. However, sodium arsenate induces the recruitment of DNMT1, which increases CpG hypermethylation of ESR1 and BRCA1, leading to tamoxifen resistance (193). In addition, procainamide promotes the expression of ERβ by inhibiting the methylation of the ERβ promoter, thereby inhibiting the signaling of ERα, thus inhibiting tamoxifen sensitivity (83). However, lycorine inhibits DNMT1-mediated methylation of VGLL4, thereby promoting VGLL4 expression and reversing tamoxifen resistance (174). In terms of the PI3K/AKT/mTOR signaling pathway, luteolin-induced MLL3 increases the mono-methylation of H3K4 on the Ras enhancer and promoter, thereby inhibiting Ras expression, which inhibits the activation of the PI3K/AKT/mTOR signaling pathway, thereby promoting tamoxifen sensitivity (194).
Discussion and conclusion
In summary, methylation promotes the development of tamoxifen resistance (115–118). The downregulation of ERα expression and abnormal activation of the PI3K/AKT/mTOR signaling pathway are the main causes of tamoxifen resistance by DNA methylation (10–14). By contrast, inhibition of methylation promotes BC sensitivity to tamoxifen (115–118). In addition, elevated methylation levels can be used as a predictive indicator of tamoxifen resistance (188–190). Moreover, a methylation inhibitor combined with tamoxifen is expected to improve the efficacy of tamoxifen (83,192–194).
Current research on methylation-induced changes in the efficacy of tamoxifen has focused on enhanced DNA methylation, which leads to the downregulation of the gene, resulting in tamoxifen resistance (18,144,173,195). However, methylation is not only limited to DNA methylation, but also m6A methylation, which is extremely important in RNA methylation and even protein methylation (196). In addition, methylation promotes tamoxifen resistance mainly by downregulating ERα and up-regulating the PI3K/AKT/mTOR signaling pathway (118,131,145,194). However, the ERα and PI3K/AKT/mTOR pathways are not isolated. Therefore, the crosstalk between them may have a further impact on tamoxifen resistance. ERα activates the PI3K/AKT/mTOR signaling pathway by downregulating PTEN expression and activating PTEN phosphorylation (197). The PI3K/AKT/mTOR signaling pathway acts as a bridge between growth factor and ERα signal transduction (198). However, the specific effect of tamoxifen, whether it increases resistance or sensitivity, needs more research.
Although drugs that target DNA methylation are gradually being used in the clinic and may provide new therapeutic approaches for improving tamoxifen response and cancer treatment, they are still at an early stage and there are numerous challenges to overcome. The precise interaction between methylation and tamoxifen resistance needs to be further elucidated, which will affect the accuracy of clinical applications. This includes controlling the selective effects of methyltransferases on target cells and the complex association between target gene methylation and the development of tamoxifen resistance. Although new generation sequencing can detect high-throughput methylation sites and accurately identify various DNA/RNA methylation patterns, it does not directly detect DNA methylation in body fluids, but relies more on DNA extraction and PCR techniques (199,200). In addition, inhibitors or potential drugs that inhibit the function or activity of DNMT also face challenges. Given that DNA methylation is present not only in tumor cells, but also in normal cells, it is a challenge to precisely target tumor cells during treatment with inhibitors or potential drugs targeting DNMT without affecting the epigenetic legacy modification of normal cells (201–203). Therefore, it is urgent to develop DNMT inhibitors that are selective for tumor cells and, more importantly, tamoxifen-resistant BC cells. Moreover, the combination of methylation-targeted drugs with endocrine therapy drugs such as tamoxifen is also an aspect worth considering. Compared with monotherapy, combination therapy can effectively reduce drug resistance (199). However, whether the combination therapy has a synergistic effect and whether it can minimize adverse reactions needs to be confirmed by further studies.
Furthermore, the present review only describes the crosstalk between methylation and tamoxifen. However, numerous other post-translational modifications can also affect tamoxifen resistance (184,204,205). Ubiquitination, acetylation and phosphorylation can also directly affect the ERα protein and the PI3K/AKT/mTOR signaling pathway, which may mediate the response to tamoxifen (184,204,205). Therefore, other post-translational modifications are worth investigation. In addition, the various post-translational modifications are not isolated, and they also have crosstalk with each other (206,207). For example, the methylation of histone is regulated by histone ubiquitination or by enzymes that catalyze ubiquitination (208). These modifications and their crosstalk play important roles in gene expression, genome stability, heterochromatin formation and cancer development (208). Furthermore, HMTs and demethylases are phosphorylated, suggesting that phosphorylation can control the initiation and extent of histone methylation (209). Therefore, revealing the association between various modifications can not only improve the crosstalk between methylation and tamoxifen resistance, but also further improve the mechanism of tamoxifen resistance, providing a theoretical basis for improving the efficacy of tamoxifen.
Finally, tamoxifen is only one type of SERMs, and other endocrine drugs that are classified as SERMs have a similar mechanism to tamoxifen. Toremifene, raloxifene, opemifene, lasoxifene and bardoxifene are members of the SERM family (184). They are all able to interact with ER in specific tissues, resulting in conformational changes in the receptor, thus affecting the ERα-mediated signaling pathway (184). In theory, methylation-mediated downregulation of ERα could also promote resistance to these drugs. Unfortunately, this field of research is relatively rare and incomplete. Therefore, the research in related fields needs to be further explored. In addition, another class of drugs also targets the estrogen receptor, termed selective estrogen receptor degraders, and its representative drug is fulvestrant (210). Fulvestrant inhibits the binding of estrogen to ERα, but it promotes the ubiquitination degradation of ERα to inhibit ERα signaling (184). Similar to tamoxifen, loss of ESR1 methylation leads to expression of ESR1, which restores sensitivity to fulvestrant (211). Although AKT inhibitors or PI3K inhibitors combined with fulvestrant are effective in ER-positive BC with palbociclib-resistance, it is not clear whether the PI3K/AKT/mTOR signaling pathway is one of the mechanisms of fulvestrant resistance (212). In addition, whether the activation of the PI3K/AKT/mTOR pathway or other signaling pathways promoted by methylation also leads to the development of resistance to fulvestrant remains to be further investigated. Furthermore, the present study that aimed to discuss the effect of methylation on tamoxifen resistance, mainly focused on preclinical studies, with phase I, II and III clinical trials practically absent. Therefore, future research may consider advancing the preclinical research into clinical practical application.
Acknowledgements
Not applicable.
Funding
This research was funded by The General Project of Hunan Provincial Chinese Medicine Research Program (grant no. D2022012).
Availability of data and materials
Not applicable.
Authors' contributions
HC conceived the study; JS and HC wrote the manuscript; JS, YH and SL collated the data; and HC revised and edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Katsura C, Ogunmwonyi I, Kankam HKN and Saha S: Breast cancer: Presentation, investigation and management. Br J Hosp Med (Lond). 83:1–7. 2022. View Article : Google Scholar : PubMed/NCBI | |
Singh H: Role of molecular targeted therapeutic drugs in treatment of breast cancer: A review article. Global Med Genet. 10:79–86. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhai J, Wu Y, Ma F, Kaklamani V and Xu B: Advances in medical treatment of breast cancer in 2022. Cancer Innov. 2:1–17. 2023. View Article : Google Scholar : PubMed/NCBI | |
Chatterjee A: Long term effects of modern breast cancer surgery. Gland Surg. 7:366–370. 2018. View Article : Google Scholar : PubMed/NCBI | |
Haque W, Butler EB and Teh BS: Personalized radiation therapy for breast cancer. Curr Oncol. 31:1588–1599. 2024. View Article : Google Scholar : PubMed/NCBI | |
Bartsch R and Bergen E: SABCS 2017: Update on chemotherapy, targeted therapy, and immunotherapy. Memo. 11:204–207. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shi L, Chen Z and Wang X: ASCO 2018: Recent advances in endocrine therapy for advanced breast cancer. Chin J Clin Oncol. 45:919–921. 2018. | |
Bhave MA and Henry NL: Extended endocrine therapy: Is 5 years enough? Curr Oncol Rep. 19:162017. View Article : Google Scholar : PubMed/NCBI | |
Barroso-Sousa R, Silva DDAFR, Alessi JVM and Mano MS: Neoadjuvant endocrine therapy in breast cancer: Current role and future perspectives. Ecancermedicalscience. 10:6092016. View Article : Google Scholar : PubMed/NCBI | |
Jordan VC and O'Malley BW: Selective estrogen-receptor modulators and antihormonal resistance in breast cancer. J Clin Oncol. 25:5815–5824. 2007. View Article : Google Scholar : PubMed/NCBI | |
Zhang W, Xu J, Shi Y, Sun Q, Zhang Q and Guan X: The novel role of miRNAs for tamoxifen resistance in human breast cancer. Cell Mol Life Sci. 72:2575–2584. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ring A and Dowsett M: Mechanisms of tamoxifen resistance. Endocr Relat Cancer. 11:643–658. 2004. View Article : Google Scholar : PubMed/NCBI | |
Yao J, Deng K, Huang J, Zeng R and Zuo J: Progress in the understanding of the mechanism of tamoxifen resistance in breast cancer. Front Pharmacol. 11:5929122020. View Article : Google Scholar : PubMed/NCBI | |
Chang M: Tamoxifen resistance in breast cancer. Biomol Ther (Seoul). 20:256–267. 2012. View Article : Google Scholar : PubMed/NCBI | |
Chen Y: Recent advances in methylation: A guide for selecting methylation reagents. Chemistry. 25:3405–3439. 2019. View Article : Google Scholar : PubMed/NCBI | |
Castillo-Aguilera O, Depreux P, Halby L, Arimondo PB and Goossens L: DNA methylation targeting: The DNMT/HMT crosstalk challenge. Biomolecules. 7:32017. View Article : Google Scholar : PubMed/NCBI | |
Jahangiri R, Jamialahmadi K, Gharib M, Emami Razavi A and Mosaffa F: Expression and clinicopathological significance of DNA methyltransferase 1, 3A and 3B in tamoxifen-treated breast cancer patients. Gene. 685:24–31. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jahangiri R, Mosaffa F, Emami Razavi A, Teimoori-Toolabi L and Jamialahmadi K: Altered DNA methyltransferases promoter methylation and mRNA expression are associated with tamoxifen response in breast tumors. J Cell Physiol. 233:7305–7319. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lin X, Li J, Yin G, Zhao Q, Elias D, Lykkesfeldt AE, Stenvang J, Brünner N, Wang J, Yang H, et al: Integrative analyses of gene expression and DNA methylation profiles in breast cancer cell line models of tamoxifen-resistance indicate a potential role of cells with stem-like properties. Breast Cancer Res. 15:R1192013. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Hong T, Wang S, Mo J, Tian T and Zhou X: Epigenetic modification of nucleic acids: From basic studies to medical applications. Chem Soc Rev. 46:2844–2872. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yu L, Davis IJ and Liu P: Regulation of EWSR1-FLI1 function by Post-Transcriptional and Post-Translational modifications. Cancers (Basel). 15:3822023. View Article : Google Scholar : PubMed/NCBI | |
Zaib S, Rana N and Khan I: Histone modifications and their role in epigenetics of cancer. Curr Med Chem. 29:2399–2411. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM and Wang H: RNA modifications in cellular metabolism: Implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther. 9:702024. View Article : Google Scholar : PubMed/NCBI | |
Song T, Lv S, Li N, Zhao X, Ma X, Yan Y, Wang W and Sun L: Versatile functions of RNA m6A machinery on chromatin. J Mol Cell Biol. 14:mjac0112022. View Article : Google Scholar : PubMed/NCBI | |
Singh V, Ram M, Kumar R, Prasad R, Roy BK and Singh KK: Phosphorylation: Implications in Cancer. Protein J. 36:1–6. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wu X, Xu M, Geng M, Chen S, Little PJ, Xu S and Weng J: Targeting protein modifications in metabolic diseases: Molecular mechanisms and targeted therapies. Signal Transduct Target Ther. 8:2202023. View Article : Google Scholar : PubMed/NCBI | |
Moore LD, Le T and Fan G: DNA methylation and its basic function. Neuropsychopharmacology. 38:23–38. 2013. View Article : Google Scholar : PubMed/NCBI | |
Małecki JM, Davydova E and Falnes P: Protein methylation in mitochondria. J Biol Chem. 298:1017912022. View Article : Google Scholar : PubMed/NCBI | |
Newman AC and Maddocks ODK: One-carbon metabolism in cancer. Br J Cancer. 116:1499–1504. 2017. View Article : Google Scholar : PubMed/NCBI | |
Froese DS, Fowler B and Baumgartner MR: Vitamin B12, folate, and the methionine remethylation cycle-biochemistry, pathways, and regulation. J Inherit Metab Dis. 42:673–685. 2019. View Article : Google Scholar : PubMed/NCBI | |
Raghubeer S and Matsha TE: Methylenetetrahydrofolate (MTHFR), the One-Carbon Cycle, and Cardiovascular Risks. Nutrients. 13:45622021. View Article : Google Scholar : PubMed/NCBI | |
Nishiyama A and Nakanishi M: Navigating the DNA methylation landscape of cancer. Trends Genet. 37:1012–1027. 2021. View Article : Google Scholar : PubMed/NCBI | |
Jones PA: Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI | |
Gros C, Fahy J, Halby L, Dufau I, Erdmann A, Gregoire JM, Ausseil F, Vispé S and Arimondo PB: DNA methylation inhibitors in cancer: Recent and future approaches. Biochimie. 94:2280–2296. 2012. View Article : Google Scholar : PubMed/NCBI | |
Lyko F: The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nat Rev Genet. 19:81–92. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kulis M and Esteller M: DNA methylation and cancer. Adv Genet. 70:27–56. 2010. View Article : Google Scholar : PubMed/NCBI | |
Delpu Y, Cordelier P, Cho WC and Torrisani J: DNA methylation and cancer diagnosis. Int J Mol Sci. 14:15029–15058. 2013. View Article : Google Scholar : PubMed/NCBI | |
Yang B, Wang JQ, Tan Y, Yuan R, Chen ZS and Zou C: RNA methylation and cancer treatment. Pharmacol Res. 174:1059372021. View Article : Google Scholar : PubMed/NCBI | |
Bai Y, Zhao H, Liu H, Wang W, Dong H and Zhao C: RNA methylation, homologous recombination repair and therapeutic resistance. Biomed Pharmacother. 166:1154092023. View Article : Google Scholar : PubMed/NCBI | |
An Y and Duan H: The role of m6A RNA methylation in cancer metabolism. Mol Cancer. 21:142022. View Article : Google Scholar : PubMed/NCBI | |
Chen XY, Zhang J and Zhu JS: The role of m6A RNA methylation in human cancer. Mol Cancer. 18:1032019. View Article : Google Scholar : PubMed/NCBI | |
He PC and He C: m6 A RNA methylation: From mechanisms to therapeutic potential. EMBO J. 40:e1059772021. View Article : Google Scholar : PubMed/NCBI | |
Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C and Chen Y: The role of m6A modification in the biological functions and diseases. Signal Transduct Target Ther. 6:742021. View Article : Google Scholar : PubMed/NCBI | |
Azzam SK, Alsafar H and Sajini AA: FTO m6A demethylase in obesity and cancer: Implications and underlying molecular mechanisms. Int J Mol Sci. 23:38002022. View Article : Google Scholar : PubMed/NCBI | |
Qu J, Yan H, Hou Y, Cao W, Liu Y, Zhang E, He J and Cai Z: RNA demethylase ALKBH5 in cancer: From mechanisms to therapeutic potential. J Hematol Oncol. 15:82022. View Article : Google Scholar : PubMed/NCBI | |
Liu ZX, Li LM, Sun HL and Liu SM: Link between m6A modification and cancers. Front Bioeng Biotechnol. 6:892018. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Zhou X and Wang X: m6A binding protein YTHDF2 in cancer. Exp Hematol Oncol. 11:212022. View Article : Google Scholar : PubMed/NCBI | |
Ramesh-Kumar D and Guil S: The IGF2BP family of RNA binding proteins links epitranscriptomics to cancer. Semin Cancer Biol. 86:18–31. 2022. View Article : Google Scholar : PubMed/NCBI | |
Bi Z, Liu Y, Zhao Y, Yao Y, Wu R, Liu Q, Wang Y and Wang X: A dynamic reversible RNA N6-methyladenosine modification: Current status and perspectives. J Cell Physiol. 234:7948–7956. 2019. View Article : Google Scholar : PubMed/NCBI | |
Malbeteau L, Pham HT, Eve L, Stallcup MR, Poulard C and Le Romancer M: How protein methylation regulates steroid receptor function. Endocr Rev. 43:160–197. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zheng K, Chen S, Ren Z and Wang Y: Protein arginine methylation in viral infection and antiviral immunity. Int J Biol Sci. 19:5292–5318. 2023. View Article : Google Scholar : PubMed/NCBI | |
Black JC, Van Rechem C and Whetstine JR: Histone lysine methylation dynamics: Establishment, regulation, and biological impact. Mol Cell. 48:491–507. 2012. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Chen X and Lu C: The interplay between DNA and histone methylation: Molecular mechanisms and disease implications. EMBO Rep. 22:e518032021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Sun Z, Jia J, Du T, Zhang N, Tang Y, Fang Y and Fang D: Overview of histone modification. Adv Exp Med Biol. 1283:1–16. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Guo B and Guo X: Histone demethylases in neurodevelopment and neurodegenerative diseases. Int J Neurosci. 1–11. 2023.doi: 10.1080/00207454.2023.2276656. View Article : Google Scholar | |
Harvey HA, Kimura M and Hajba A: Toremifene: An evaluation of its safety profile. Breast. 15:142–157. 2006. View Article : Google Scholar : PubMed/NCBI | |
Burstein HJ, Temin S, Anderson H, Buchholz TA, Davidson NE, Gelmon KE, Giordano SH, Hudis CA, Rowden D, Solky AJ, et al: Adjuvant endocrine therapy for women with hormone receptor-positive breast cancer: American society of clinical oncology clinical practice guideline focused update. J Clin Oncol. 32:2255–2269. 2014. View Article : Google Scholar : PubMed/NCBI | |
Montagna E, Cancello G and Colleoni M: The aromatase inhibitors (plus ovarian function suppression) in premenopausal breast cancer patients: Ready for prime time? Cancer Treat Rev. 39:886–890. 2013. View Article : Google Scholar : PubMed/NCBI | |
Crump M, Sawka CA, DeBoer G, Buchanan RB, Ingle JN, Forbes J, Meakin JW, Shelley W and Pritchard KI: An individual patient-based meta-analysis of tamoxifen versus ovarian ablation as first line endocrine therapy for premenopausal women with metastatic breast cancer. Breast Cancer Res Treat. 44:201–210. 1997. View Article : Google Scholar : PubMed/NCBI | |
Tsoutsou PG, Koukourakis MI, Azria D and Belkacemi Y: Optimal timing for adjuvant radiation therapy in breast cancer A comprehensive review and perspectives. Crit Rev Oncol Hematol. 71:102–116. 2009. View Article : Google Scholar : PubMed/NCBI | |
Nakhlis F, Lazarus L, Hou NJ, Acharya S, Khan SA, Staradub VL, Rademaker AW and Morrow M: Tamoxifen use in patients with ductal carcinoma in situ and T1a/b N0 invasive carcinoma. J Am Coll Surg. 201:688–694. 2005. View Article : Google Scholar : PubMed/NCBI | |
Nichols HB, DeRoo LA, Scharf DR and Sandler DP: Risk-Benefit profiles of women using tamoxifen for chemoprevention. J Natl Cancer Inst. 107:3542014.PubMed/NCBI | |
Anderson C, Nichols HB, House M and Sandler DP: Risk versus benefit of chemoprevention among raloxifene and tamoxifen users with a family history of breast cancer. Cancer Prev Res. 12:801–808. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chlebowski RT: Current concepts in breast cancer chemoprevention. Pol Arch Med Wewn. 124:191–199. 2014. View Article : Google Scholar : PubMed/NCBI | |
Nichols HB, Sturmer T, Lee VS, Anderson C, Lee JS, Roh JM, Visvanathan K, Muss H and Kushi LH: Breast cancer chemoprevention in an integrated health care setting. JCO Clin Cancer Inform. 1:1–12. 2017. View Article : Google Scholar : PubMed/NCBI | |
Friedl A and Jordan VC: What do we know and what don't we know about tamoxifen in the human uterus. Breast Cancer Res Treat. 31:27–39. 1994. View Article : Google Scholar : PubMed/NCBI | |
DeCensi A, Johansson H, Helland T, Puntoni M, Macis D, Aristarco V, Caviglia S, Webber TB, Briata IM, D'Amico M, et al: Association of CYP2D6 genotype and tamoxifen metabolites with breast cancer recurrence in a low-dose trial. NPJ Breast Cancer. 7:342021. View Article : Google Scholar : PubMed/NCBI | |
Buijs SM, Koolen SLW, Mathijssen RHJ and Jager A: Tamoxifen dose De-escalation: An effective strategy for reducing adverse effects? Drugs. 84:385–401. 2024. View Article : Google Scholar : PubMed/NCBI | |
Reinhorn D, Yerushalmi R, Moore A, Desnoyers A, Saleh RR, Amir E and Goldvaser H: Evolution in the risk of adverse events of adjuvant endocrine therapy in postmenopausal women with early-stage breast cancer. Breast Cancer Res Treat. 182:259–266. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wibowo E, Pollock PA, Hollis N and Wassersug RJ: Tamoxifen in men: A review of adverse events. Andrology. 4:776–788. 2016. View Article : Google Scholar : PubMed/NCBI | |
Sestak I and Cuzick J: Endometrial cancer risk in postmenopausal breast cancer patients treated with tamoxifen or aromatase inhibitors. Expert Rev Endocrinol Metab. 11:425–432. 2016. View Article : Google Scholar : PubMed/NCBI | |
Huang B, Warner M and Gustafsson JA: Estrogen receptors in breast carcinogenesis and endocrine therapy. Mol Cell Endocrinol. 418:240–244. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ye H, Dudley SZ and Shaw IC: Intimate estrogen receptor-α/ligand relationships signal biological activity. Toxicology. 408:80–87. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ge Y, Ni X, Li J, Ye M and Jin X: Roles of estrogen receptor α in endometrial carcinoma (Review). Oncol Lett. 26:5302023. View Article : Google Scholar : PubMed/NCBI | |
Nass N and Kalinski T: Tamoxifen resistance: From cell culture experiments towards novel biomarkers. Pathol Res Pract. 211:189–197. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ramchand SK, Cheung YM, Yeo B and Grossmann M: The effects of adjuvant endocrine therapy on bone health in women with breast cancer. J Endocrinol. 241:R111–R124. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mansouri S, Farahmand L, Hosseinzade A, Eslami SZ and Majidzadeh AK: Estrogen can restore Tamoxifen sensitivity in breast cancer cells amidst the complex network of resistance. Biomed Pharmacother. 93:1320–1325. 2017. View Article : Google Scholar : PubMed/NCBI | |
BachmannMoisson N, BarberiHeyob M and Merlin JL: Molecular mechanisms of tamoxifen resistance. Bull Du Cancer. 84:69–75. 1997. | |
Kulkoyluoglu E and Madak-Erdogan Z: Nuclear and extranuclear-initiated estrogen receptor signaling crosstalk and endocrine resistance in breast cancer. Steroids. 114:41–47. 2016. View Article : Google Scholar : PubMed/NCBI | |
Balfe PJ, McCann AH, Welch HM and Kerin MJ: Estrogen receptor beta and breast cancer. Eur J Surg Oncol. 30:1043–1050. 2004. View Article : Google Scholar : PubMed/NCBI | |
Khalid AB and Krum SA: Estrogen receptors alpha and beta in bone. Bone. 87:130–135. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hartman J, Stroem A and Gustafsson JA: Current concepts and significance of estrogen receptor β in prostate cancer. Steroids. 77:1262–1266. 2012. View Article : Google Scholar : PubMed/NCBI | |
Altundag O, Altundag K and Gunduz M: DNA methylation inhibitor, procainamide, may decrease the tamoxifen resistance by inducing overexpression of the estrogen receptor beta in breast cancer patients. Med Hypotheses. 63:684–687. 2004. View Article : Google Scholar : PubMed/NCBI | |
Levy N, Paruthiyil S, Zhao X, Vivar OI, Saunier EF, Griffin C, Tagliaferri M, Cohen I, Speed TP and Leitman DC: Unliganded estrogen receptor-β regulation of genes is inhibited by tamoxifen. Mol Cell Endocrinol. 315:201–207. 2010. View Article : Google Scholar : PubMed/NCBI | |
Tee MK, Rogatsky I, Tzagarakis-Foster C, Cvoro A, An J, Christy RJ, Yamamoto KR and Leitman DC: Estradiol and selective estrogen receptor modulators differentially regulate target genes with estrogen receptors alpha and beta. Mole Biol Cell. 15:1262–1272. 2004. View Article : Google Scholar : PubMed/NCBI | |
Mansouri S, Feizi N, Mahdi A, Majidzadeh AK and Farahmand L: A review on the role of VEGF in tamoxifen resistance. Anticancer Agents Med Chem. 18:2006–2009. 2018. View Article : Google Scholar : PubMed/NCBI | |
Provenzano A, Kurian S and Abraham J: Overcoming endocrine resistance in breast cancer: Role of the PI3K and the mTOR pathways. Expert Rev Anticancer Ther. 13:143–147. 2013. View Article : Google Scholar : PubMed/NCBI | |
Mohite R and Doshi G: Elucidation of the role of the epigenetic regulatory mechanisms of PI3K/Akt/mTOR signaling pathway in human malignancies. Curr Cancer Drug Targets. 24:231–244. 2024. View Article : Google Scholar : PubMed/NCBI | |
Luo Q, Du R, Liu W, Huang G, Dong Z and Li X: PI3K/Akt/mTOR signaling pathway: Role in esophageal squamous cell carcinoma, regulatory mechanisms and opportunities for targeted therapy. Front Oncol. 12:8523832022. View Article : Google Scholar : PubMed/NCBI | |
Su H, Peng C and Liu Y: Regulation of ferroptosis by PI3K/Akt signaling pathway: A promising therapeutic axis in cancer. Front Cell Dev Biol. 12:13723302024. View Article : Google Scholar : PubMed/NCBI | |
Bertacchini J, Heidari N, Mediani L, Capitani S, Shahjahani M, Ahmadzadeh A and Saki N: Targeting PI3K/AKT/mTOR network for treatment of leukemia. Cell Mol Life Sci. 72:2337–2347. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hashemi M, Etemad S, Rezaei S, Ziaolhagh S, Rajabi R, Rahmanian P, Abdi S, Koohpar ZK, Rafiei R, Raei B, et al: Progress in targeting PTEN/PI3K/Akt axis in glioblastoma therapy: Revisiting molecular interactions. Biomed Pharmacother. 158:1142042023. View Article : Google Scholar : PubMed/NCBI | |
Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Bahonar A, Nakhaee Z, Entezari M, Beig Goharrizi MAS, Salimimoghadam S, Ren J, et al: New emerging targets in osteosarcoma therapy: PTEN and PI3K/Akt crosstalk in carcinogenesis. Pathol Res Pract. 251:1549022023. View Article : Google Scholar : PubMed/NCBI | |
Xiang Y, Yang Y, Liu J and Yang X: Functional role of MicroRNA/PI3K/AKT axis in osteosarcoma. Front Oncol. 13:12192112023. View Article : Google Scholar : PubMed/NCBI | |
Huang C, Li J and Ma TT: PI3K/Akt signaling pathway and liver fibrosis. Zhongguo Yaolixue Tongbao. 27:1037–1041. 2011. | |
Li L, Han C and Liu X: Regulation of PI3K-Akt-mTORC1 signal pathway in the cell growth and proliferation. J Biology. 31:75–77. 2014. | |
Minami A, Nakanishi A, Ogura Y, Kitagishi Y and Matsuda S: Connection between tumor suppressor BRCA1 and PTEN in damaged DNA repair. Front Oncol. 4:318. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Chen Q, Zou Y, Chen H, Qi L and Chen Y: Stem cells and cellular origins of breast cancer: Updates in the rationale, controversies, and therapeutic implications. Front Oncol. 9:8202019. View Article : Google Scholar : PubMed/NCBI | |
Zheng Q, Zhang M, Zhou F, Zhang L and Meng X: The breast cancer stem cells traits and drug resistance. Front Pharmacol. 11:5999652021. View Article : Google Scholar : PubMed/NCBI | |
Telang N: Stem cell models for cancer therapy. Int J Mol Sci. 23:70552022. View Article : Google Scholar : PubMed/NCBI | |
Haiduk TS, Sicking M, Brücksken KA, Espinoza-Sánchez NA, Eder KM, Kemper B, Eich HT, Götte M, Greve B and Troschel FM: Dysregulated stem cell markers Musashi-1 and Musashi-2 are associated with therapy resistance in inflammatory breast cancer. Arch Med Res. 54:1028552023. View Article : Google Scholar : PubMed/NCBI | |
Gelsomino L, Panza S, Giordano C, Barone I, Gu G, Spina E, Catalano S, Fuqua S and Andò S: Mutations in the estrogen receptor alpha hormone binding domain promote stem cell phenotype through notch activation in breast cancer cell lines. Cancer Lett. 428:12–20. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ghasemi F, Sarabi PZ, Athari SS and Esmaeilzadeh A: Therapeutics strategies against cancer stem cell in breast ca ncer. Int J Biochem Cell Biol. 109:76–81. 2019. View Article : Google Scholar : PubMed/NCBI | |
Albain KS, Czerlanis C, Zlobin A, Covington KR, Rajan P, Godellas C, Bova D, Lo SS, Robinson P, Sarker S, et al: S1-5: Modulation of cancer and stem cell biomarkers by the notch inhibitor MK-0752 added to endocrine therapy for early stage ER + breast cancer. Cancer Res. 71 (Suppl 24):S1–S5. 2011. View Article : Google Scholar | |
Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT, et al: Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 19:1512–1524. 2013. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Xu J, Liu B, He X, Zhou L, Hu X, Qiao F, Zhang A, Xu X, Zhang H, et al: IL6 blockade potentiates the anti-tumor effects of γ-secretase inhibitors in Notch3-expressing breast cancer. Cell Death Differ. 25:330–339. 2018. View Article : Google Scholar : PubMed/NCBI | |
Means-Powell JA, Mayer IA, Ismail-Khan R, Del Valle L, Tonetti D, Abramson VG, Sanders MS, Lush RM, Sorrentino C, Majumder S and Miele L: A Phase ib dose escalation trial of RO4929097 (a γ-secretase inhibitor) in combination with exemestane in patients with er plus metastatic breast cancer (MBC). Clin Breast Cancer. 22:103–114. 2022. View Article : Google Scholar : PubMed/NCBI | |
Jeselsohn R, Cornwell M, Pun M, Buchwalter G, Nguyen M, Bango C, Huang Y, Kuang Y, Paweletz C, Fu X, et al: Embryonic transcription factor SOX9 drives breast cancer endocrine resistance. Proc Natl Acad Sci USA. 114:E4482–E4491. 2017. View Article : Google Scholar : PubMed/NCBI | |
Poulard C, Pham TH, Drouet Y, Jacquemetton J, Surmielova A, Kassem L, Mery B, Lasset C, Reboulet J, Treilleux I, et al: Nuclear PRMT5 is a biomarker of sensitivity to tamoxifen in ERα+ breast cancer. Embo Mol Med. 15:e172482023. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Xu M, Le K, Ming J, Guo H, Ruan S and Huang T: SRC promotes tamoxifen resistance in breast cancer via Up-Regulating SIRT1. Onco Targets Ther. 13:4635–4647. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jogi A, Ehinger A, Hartman L and Alkner S: Expression of HIF-1α is related to a poor prognosis and tamoxifen resistance in contralateral breast cancer. PLoS One. 14:e02261502019. View Article : Google Scholar : PubMed/NCBI | |
Ward A, Balwierz A, Zhang JD, Küblbeck M, Pawitan Y, Hielscher T, Wiemann S and Sahin Ö: Re-expression of microRNA-375 reverses both tamoxifen resistance and accompanying EMT-like properties in breast cancer. Oncogene. 32:1173–1182. 2013. View Article : Google Scholar : PubMed/NCBI | |
Cittelly DM, Das PM, Spoelstra NS, Edgerton SM, Richer JK, Thor AD and Jones FE: Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol Cancer. 9:3172010. View Article : Google Scholar : PubMed/NCBI | |
Xu E, Hu M, Ge R, Tong D, Fan Y, Ren X and Liu Y: LncRNA-42060 regulates tamoxifen sensitivity and tumor development via regulating the miR-204-5p/SOX4 axis in canine mammary gland tumor cells. Front Vet Sci. 8:6546942021. View Article : Google Scholar : PubMed/NCBI | |
Sukocheva OA, Lukina E, Friedemann M, Menschikowski M, Hagelgans A and Aliev G: The crucial role of epigenetic regulation in breast cancer anti-estrogen resistance: Current findings and future perspectives. Semin Cancer Biol. 82:35–59. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kim SS, Lee MH and Lee MO: Histone methyltransferases regulate the transcriptional expression of ERα and the proliferation of tamoxifen-resistant breast cancer cells. Breast Cancer Res Treat. 180:45–54. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Zhou C, Jiang H, Liang L, Shi W, Zhang Q, Sun P, Xiang R, Wang Y and Yang S: ZEB1 induces ER-α promoter hypermethylation and confers antiestrogen resistance in breast cancer. Cell Death Dis. 8:e27322017. View Article : Google Scholar : PubMed/NCBI | |
Jiménez-Garduño AM, Mendoza-Rodríguez MG, Urrutia-Cabrera D, Domínguez-Robles MC, Pérez-Yépez EA, Ayala-Sumuano JT and Meza I: IL-1β induced methylation of the estrogen receptor ERα gene correlates with EMT and chemoresistance in breast cancer cells. Biochem Biophys Res Commun. 490:780–785. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kim MR, Wu MJ, Zhang Y, Yang JY and Chang CJ: TET2 directs mammary luminal cell differentiation and endocrine response. Nat Commun. 11:46422020. View Article : Google Scholar : PubMed/NCBI | |
Chang HG, Kim SJ, Chung KW, Noh DY, Kwon Y, Lee ES and Kang HS: Tamoxifen-resistant breast cancers show less frequent methylation of the estrogen receptor beta but not the estrogen receptor alpha gene. J Mol Med (Berl). 83:132–139. 2005. View Article : Google Scholar : PubMed/NCBI | |
Zou JX, Duan Z, Wang J, Sokolov A, Xu J, Chen CZ, Li JJ and Chen HW: Kinesin family deregulation coordinated by bromodomain protein ANCCA and histone methyltransferase MLL for breast cancer cell growth, survival, and tamoxifen resistance. Mol Cancer Res. 12:539–549. 2014. View Article : Google Scholar : PubMed/NCBI | |
Jin ML, Kim YW, Jin HL, Kang H, Lee EK, Stallcup MR and Jeong KW: Aberrant expression of SETD1A promotes survival and migration of estrogen receptor α-positive breast cancer cells. Int J Cancer. 143:2871–2883. 2018. View Article : Google Scholar : PubMed/NCBI | |
Stone A, Zotenko E, Locke WJ, Korbie D, Millar EK, Pidsley R, Stirzaker C, Graham P, Trau M, Musgrove EA, et al: DNA methylation of oestrogen-regulated enhancers defines endocrine sensitivity in breast cancer. Nat Commun. 6:77582015. View Article : Google Scholar : PubMed/NCBI | |
Gautam N, Verma H, Choudhary S, Kaur S and Silakari O: Functional relationship of SNP (Ala490Thr) of an epigenetic gene EZH2 results in the progression and poor survival of ER+/tamoxifen treated breast cancer patients. J Genet. 100:862021. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Zhang Z, Cenciarini ME, Proietti CJ, Amasino M, Hong T, Yang M, Liao Y, Chiang HC, Kaklamani VG, et al: Tamoxifen resistance in breast cancer is regulated by the EZH2-ERα-GREB1 transcriptional axis. Cancer Res. 78:671–684. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li J, Liang Y, Zhou S, Chen J and Wu C: UCHL1 contributes to insensitivity to endocrine therapy in triple-negative breast cancer by deubiquitinating and stabilizing KLF5. Breast Cancer Res. 26:442024. View Article : Google Scholar : PubMed/NCBI | |
Redeuilh G, Attia A, Mester J and Sabbah M: Transcriptional activation by the oestrogen receptor α is modulated through inhibition of cyclin-dependent kinases. Oncogene. 21:5773–5782. 2002. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Zhang X and Wang ZY: The Wilms' tumor suppressor WT1 regulates expression of members of the epidermal growth factor receptor (EGFR) and estrogen receptor in acquired tamoxifen resistance. Anticancer Res. 30:3637–3642. 2010.PubMed/NCBI | |
Best SA, Hutt KJ, Fu NY, Vaillant F, Liew SH, Hartley L, Scott CL, Lindeman GJ and Visvader JE: Dual roles for Id4 in the regulation of estrogen signaling in the mammary gland and ovary. Development. 141:3159–3164. 2014. View Article : Google Scholar : PubMed/NCBI | |
Mobini K, Tamaddon G, Fardid R, Keshavarzi M and Mohammadi-Bardbori A: Aryl hydrocarbon-estrogen alpha receptor-dependent expression of miR-206, miR-27b, and miR-133a suppress cell proliferation and migration in MCF-7 cells. J Biochem Mol Toxicol. 33:e223042019. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Yu D, Li H, Lv Y and Li S: Long non-coding RNA UCA1 confers tamoxifen resistance in breast cancer endocrinotherapy through regulation of the EZH2/p21 axis and the PI3K/AKT signaling pathway. Int J Oncol. 54:1033–1042. 2019.PubMed/NCBI | |
Ren C, Tang X and Lan H: Comprehensive analysis based on DNA methylation and RNA-seq reveals hypermethylation of the up-regulated WT1 gene with potential mechanisms in PAM50 subtypes of breast cancer. PeerJ. 9:e113772021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Zhang B, Fang J and Cao X: Hypomethylation of DNA-binding inhibitor 4 serves as a potential biomarker in distinguishing acquired tamoxifen-refractory breast cancer. Int J Clin Exp Pathol. 8:9500–9505. 2015.PubMed/NCBI | |
Li X, Wu Y, Liu A and Tang X: MiR-27b is epigenetically downregulated in tamoxifen resistant breast cancer cells due to promoter methylation and regulates tamoxifen sensitivity by targeting HMGB3. Biochem Biophys Res Commun. 477:768–773. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Carlisle SM, Doll MA, Martin RCG, States JC, Klinge CM and Hein DW: High N-Acetyltransferase 1 expression is associated with estrogen receptor expression in breast tumors, but is not Under Direct Regulation by Estradiol, 5α-androstane-3β, 17β-Diol, or dihydrotestosterone in breast cancer cells. J Pharmacol Exp Ther. 365:84–93. 2018. View Article : Google Scholar : PubMed/NCBI | |
González-Bengtsson A, Asadi A, Gao H, Dahlman-Wright K and Jacobsson A: Estrogen enhances the expression of the polyunsaturated fatty acid elongase Elovl2 via ERα in breast cancer cells. PLoS One. 11:e01642412016. View Article : Google Scholar : PubMed/NCBI | |
Rickard DJ, Waters KM, Ruesink TJ, Khosla S, Katzenellenbogen JA, Katzenellenbogen BS, Riggs BL and Spelsberg TC: Estrogen receptor isoform-specific induction of progesterone receptors in human osteoblasts. J Bone Miner Res. 17:580–592. 2002. View Article : Google Scholar : PubMed/NCBI | |
Kim SJ, Kang HS, Jung SY, Min SY, Lee S, Kim SW, Kwon Y, Lee KS, Shin KH and Ro J: Methylation patterns of genes coding for drug-metabolizing enzymes in tamoxifen-resistant breast cancer tissues. J Mol Med (Berl). 88:1123–1131. 2010. View Article : Google Scholar : PubMed/NCBI | |
Sun H, Wang G, Cai J, Wei X, Zeng Y, Peng Y and Zhuang J: Long non-coding RNA H19 mediates N-acetyltransferase 1 gene methylation in the development of tamoxifen resistance in breast cancer. Exp Ther Med. 23:122022. View Article : Google Scholar : PubMed/NCBI | |
Kim HW, Baek M, Jung S, Jang S, Lee H, Yang SH, Kwak BS and Kim SJ: ELOVL2-AS1 suppresses tamoxifen resistance by sponging miR-1233-3p in breast cancer. Epigenetics. 18:22763842023. View Article : Google Scholar : PubMed/NCBI | |
Pathiraja TN, Shetty PB, Jelinek J, He R, Hartmaier R, Margossian AL, Hilsenbeck SG, Issa JP and Oesterreich S: Progesterone receptor isoform-specific promoter methylation: Association of PRA promoter methylation with worse outcome in breast cancer patients. Clin Cancer Res. 17:4177–4186. 2011. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Li L, Liu H, Qin P, Chen R, Liu S, Xiong H, Li Y, Yang Z, Xie M, et al: PAX2 is regulated by estrogen/progesterone through promoter methylation in endometrioid adenocarcinoma and has an important role in carcinogenesis via the AKT/mTOR signaling pathway. J Pathol. 262:467–479. 2024. View Article : Google Scholar : PubMed/NCBI | |
Oesterreich S, Deng W, Jiang S, Cui X, Ivanova M, Schiff R, Kang K, Hadsell DL, Behrens J and Lee AV: Estrogen-mediated down-regulation of E-cadherin in breast cancer cells. Cancer Res. 63:5203–5208. 2003.PubMed/NCBI | |
Jahangiri R, Mosaffa F, Emami Razavi A, Teimoori-Toolabi L and Jamialahmadi K: PAX2 promoter methylation and AIB1 overexpression promote tamoxifen resistance in breast carcinoma patients. J Oncol Pharm Pract. 28:310–325. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Gun M and Hong XY: Induced tamoxifen resistance is mediated by increased methylation of E-Cadherin in estrogen receptor-expressing breast cancer cells. Sci Rep. 9:141402019. View Article : Google Scholar : PubMed/NCBI | |
Kim HW, Park JE, Baek M, Kim H, Ji HW, Yun SH, Jeong D, Ham J, Park S, Lu X, et al: Matrix Metalloproteinase-1 (MMP1) upregulation through promoter hypomethylation enhances tamoxifen resistance in breast cancer. Cancers (Basel). 14:12322022. View Article : Google Scholar : PubMed/NCBI | |
Jung HH, Park YH, Jun HJ, Kong J, Kim JH, Kim JA, Yun J, Sun JM, Won YW, Lee S, et al: Matrix Metalloproteinase-1 expression can be upregulated through mitogen-activated protein kinase pathway under the influence of human epidermal growth factor Receptor 2 synergized with estrogen receptor. Mol Cancer Res. 8:1037–1047. 2010. View Article : Google Scholar : PubMed/NCBI | |
Ren W, Yi H, Bao Y, Liu Y and Gao X: Oestrogen inhibits PTPRO to prevent the apoptosis of renal podocytes. Exp Ther Med. 17:2373–2380. 2019.PubMed/NCBI | |
Ramaswamy B, Majumder S, Roy S, Ghoshal K, Kutay H, Datta J, Younes M, Shapiro CL, Motiwala T and Jacob ST: Estrogen-mediated suppression of the gene encoding protein tyrosine phosphatase PTPRO in human breast cancer: Mechanism and role in tamoxifen sensitivity. Mol Endocrinol. 23:176–187. 2009. View Article : Google Scholar : PubMed/NCBI | |
Fan Y, Xie G, Wang Z, Wang Y, Wang Y, Zheng H and Zhong X: PTEN promoter methylation predicts 10-year prognosis in hormone receptor-positive early breast cancer patients who received adjuvant tamoxifen endocrine therapy. Breast Cancer Res Treat. 192:33–42. 2022. View Article : Google Scholar : PubMed/NCBI | |
Phuong NT, Kim SK, Lim SC, Kim HS, Kim TH, Lee KY, Ahn SG, Yoon JH and Kang KW: Role of PTEN promoter methylation in tamoxifen-resistant breast cancer cells. Breast Cancer Res Treat. 130:73–83. 2011. View Article : Google Scholar : PubMed/NCBI | |
Phuong NT, Kim SK, Im JH, Yang JW, Choi MC, Lim SC, Lee KY, Kim YM, Yoon JH and Kang KW: Induction of methionine adenosyltransferase 2A in tamoxifen-resistant breast cancer cells. Oncotarget. 7:13902–13916. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Liu J, Ebrahimi B, Pratap UP, He Y, Altwegg KA, Tang W, Li X, Lai Z, Chen Y, et al: SETDB1 interactions with PELP1 contributes to breast cancer endocrine therapy resistance. Breast Cancer Res. 24:262022. View Article : Google Scholar : PubMed/NCBI | |
Schomann T, Mirzakhani K, Kallenbach J, Lu J, Rasa SMM, Neri F and Baniahmad A: Androgen-Induced MIG6 regulates phosphorylation of retinoblastoma protein and AKT to counteract Non-Genomic AR signaling in prostate cancer cells. Biomolecules. 12:10482022. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Li J, Wu W, Shen R, Jiang H, Qian Y, Tang Y, Bai T, Wu S, Wei L, et al: Smad4-dependent suppressor pituitary homeobox 2 promotes PPP2R2A-mediated inhibition of Akt pathway in pancreatic cancer. Oncotarget. 7:11208–11222. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yue C, Bai Y, Piao Y and Liu H: DOK7 inhibits cell proliferation, migration, and invasion of breast cancer via the PI3K/PTEN/AKT pathway. J Oncol. 2021:40352572021. View Article : Google Scholar : PubMed/NCBI | |
Barkovskaya A, Seip K, Prasmickaite L, Mills IG, Moestue SA and Itkonen HM: Inhibition of O-GlcNAc transferase activates tumor-suppressor gene expression in tamoxifen-resistant breast cancer cells. Sci Rep. 10:169922020. View Article : Google Scholar : PubMed/NCBI | |
Maier S, Nimmrich I, Koenig T, Eppenberger-Castori S, Bohlmann I, Paradiso A, Spyratos F, Thomssen C, Mueller V, Nährig J, et al: DNA-methylation of the homeodomain transcription factor PITX2 reliably predicts risk of distant disease recurrence in tamoxifen-treated, node-negative breast cancer patients-Technical and clinical validation in a multi-centre setting in collaboration with the European Organisation for Research and Treatment of Cancer (EORTC) PathoBiology group. Eur J Cancer. 43:1679–1686. 2007. View Article : Google Scholar : PubMed/NCBI | |
Gowdini E, Aleyasin SA, Ramezani N, Nafisi N and Tutuni M: DOK7 CpG hypermethylation in blood leukocytes as an epigenetic biomarker for acquired tamoxifen resistant in breast cancer. J Hum Genet. 68:33–38. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wujak M, Veith C, Wu CY, Wilke T, Kanbagli ZI, Novoyatleva T, Guenther A, Seeger W, Grimminger F, Sommer N, et al: Adenylate Kinase 4-A key regulator of proliferation and metabolic shift in human pulmonary arterial smooth muscle cells via Akt and HIF-1α signaling pathways. Int J Mol Sci. 22:103712021. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Gonzalez G, Dai X, Miao W, Yuan J, Huang M, Bade D, Li L, Sun Y and Wang Y: Adenylate Kinase 4 modulates the resistance of breast cancer cells to tamoxifen through an m6A-Based epitranscriptomic mechanism. Mol Ther. 28:2593–2604. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ye L, Lin C, Wang X, Li Q, Li Y, Wang M, Zhao Z, Wu X, Shi D, Xiao Y, et al: Epigenetic silencing of SALL2 confers tamoxifen resistance in breast cancer. EMBO Mol Med. 11:e106382019. View Article : Google Scholar : PubMed/NCBI | |
Huang J, Li J, Tang J, Wu Y, Dai F, Yi Z, Wang Y, Li Y, Wu Y, Ren G and Xiang T: ZDHHC22-mediated mTOR palmitoylation restrains breast cancer growth and endocrine therapy resistance. Int J Biol Sci. 18:2833–2850. 2022. View Article : Google Scholar : PubMed/NCBI | |
Al-Ansari MM, Hendrayani SF, Tulbah A, Al-Tweigeri T, Shehata AI and Aboussekhra A: p16INK4A represses breast stromal fibroblasts migration/invasion and their VEGF-A-dependent promotion of angiogenesis through Akt inhibition. Neoplasia. 14:1269–1277. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhou J and Chen C: Suppression of malignant melanoma by knocking down growth differentiation factor-15 via inhibiting PTEN/PI3K/AKT signaling pathway. J Cancer. 15:1115–1123. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Yao F, Xiao Q, Liu Q, Yang Y, Li X, Jiang G, Kuno T and Fang Y: EZH2 inhibition sensitizes tamoxifen-resistant breast cancer cells through cell cycle regulation. Mol Med Rep. 17:2642–2650. 2018.PubMed/NCBI | |
Stone A, Valdés-Mora F, Gee JM, Farrow L, McClelland RA, Fiegl H, Dutkowski C, McCloy RA, Sutherland RL, Musgrove EA and Nicholson RI: Tamoxifen-induced epigenetic silencing of oestrogen-regulated genes in anti-hormone resistant breast cancer. PLoS One. 7:e404662012. View Article : Google Scholar : PubMed/NCBI | |
Zhao W, Sun M, Li S, Chen Z and Geng D: Transcription factor ATF3 mediates the radioresistance of breast cancer. J Cell Mol Med. 22:4664–4675. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Yuan J, Zhang X, Li L, Dai X, Chen Q and Wang Y: ATF3 modulates the resistance of breast cancer cells to tamoxifen through an N6-Methyladenosine-Based epitranscriptomic mechanism. Chem Res Toxicol. 34:1814–1821. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lv Y, Fang L, Ding P and Liu R: PI3K/Akt-Beclin1 signaling pathway positively regulates phagocytosis and negatively mediates NF-κB-dependent inflammation in Staphylococcus aureus-infected macrophages. Biochem Biophys Res Commun. 510:284–289. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Xie S, Yang J, Xiong H, Jia Y, Zhou Y, Chen Y, Ying X, Chen C, Ye C, et al: The long noncoding RNA H19 promotes tamoxifen resistance in breast cancer via autophagy. J Hematol Oncol. 12:812019. View Article : Google Scholar : PubMed/NCBI | |
Gu Q-Z, Nijiati A, Gao X, Tao KL, Li CD, Fan XP and Tian Z: TROP2 promotes cell proliferation and migration in osteosarcoma through PI3K/AKT signaling. Mol Med Rep. 8:1782–1788. 2018.PubMed/NCBI | |
Zimmers SM, Browne EP, Williams KE, Jawale RM, Otis CN, Schneider SS and Arcaro KF: TROP2 methylation and expression in tamoxifen-resistant breast cancer. Cancer Cell Int. 18:942018. View Article : Google Scholar : PubMed/NCBI | |
Zhai J, Jiang JF and Shi L: Lycorine weakens tamoxifen resistance of breast cancer via abrogating HAGLR-mediated epigenetic suppression on VGLL4 by DNMT1. Kaohsiung J Med Sci. 39:278–289. 2023. View Article : Google Scholar : PubMed/NCBI | |
Watanabe T, Oba T, Tanimoto K, Shibata T, Kamijo S and Ito KI: Tamoxifen resistance alters sensitivity to 5-fluorouracil in a subset of estrogen receptor-positive breast cancer. PLoS One. 16:e02528222021. View Article : Google Scholar : PubMed/NCBI | |
Williams KE, Anderton DL, Lee MP, Pentecost BT and Arcaro KF: High-density array analysis of DNA methylation in Tamoxifen-resistant breast cancer cell lines. Epigenetics. 9:297–307. 2014. View Article : Google Scholar : PubMed/NCBI | |
Ansar M, Thu LTA, Hung CS, Su CM, Huang MH, Liao LM, Chung YM and Lin RK: Promoter hypomethylation and overexpression of TSTD1 mediate poor treatment response in breast cancer. Front Oncol. 12:10042612022. View Article : Google Scholar : PubMed/NCBI | |
Pietkiewicz PP, Lutkowska A, Lianeri M and Jagodzinski PP: Tamoxifen epigenetically modulates CXCL12 expression in MCF-7 breast cancer cells. Biomed Pharmacother. 64:54–57. 2010. View Article : Google Scholar : PubMed/NCBI | |
Martens JW, Nimmrich I, Koenig T, Look MP, Harbeck N, Model F, Kluth A, Bolt-de Vries J, Sieuwerts AM, Portengen H, et al: Association of DNA methylation of phosphoserine aminotransferase with response to endocrine therapy in patients with recurrent breast cancer. Cancer Res. 65:4101–4117. 2005. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Li D, Rodriguez-Juarez R, Farfus A, Storozynsky Q, Malach M, Carpenter E, Filkowski J, Lykkesfeldt AE and Kovalchuk O: A suppressive role of guanine nucleotide-binding protein subunit beta-4 inhibited by DNA methylation in the growth of anti-estrogen resistant breast cancer cells. BMC cancer. 18:8172018. View Article : Google Scholar : PubMed/NCBI | |
Kedia-Mokashi NA, Kadam L, Ankolkar M, Dumasia K and Balasinor NH: Aberrant methylation of multiple imprinted genes in embryos of tamoxifen-treated male rats. Reproduction. 146:155–168. 2013. View Article : Google Scholar : PubMed/NCBI | |
Liggett TE, Melnikov AA, Marks JR and Levenson VV: Methylation patterns in cell-free plasma DNA reflect removal of the primary tumor and drug treatment of breast cancer patients. Int J Cancer. 128:492–499. 2011. View Article : Google Scholar : PubMed/NCBI | |
Pathak S, Kedia-Mokashi N, Saxena M, D'Souza R, Maitra A, Parte P, Gill-Sharma M and Balasinor N: Effect of tamoxifen treatment on global and insulin-like growth factor 2-H19 locus-specific DNA methylation in rat spermatozoa and its association with embryo loss. Fertil Steril. 91:2253–2263. 2009. View Article : Google Scholar : PubMed/NCBI | |
Ge Y, Zhan Z, Ye M and Jin X: The crosstalk between ubiquitination and endocrine therapy. J Mol Med (Berl). 101:461–486. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kubarek Ł and Jagodzinski PP: Epigenetic up-regulation of CXCR4 and CXCL12 expression by 17 beta-estradiol and tamoxifen is associated with formation of DNA methyltransferase 3B4 splice variant in Ishikawa endometrial adenocarcinoma cells. FEBS Lett. 581:1441–1448. 2007. View Article : Google Scholar : PubMed/NCBI | |
Wu H, Chen Y, Liang J, Shi B, Wu G, Zhang Y, Wang D, Li R, Yi X, Zhang H, et al: Hypomethylation-linked activation of PAX2 mediates tamoxifen-stimulated endometrial carcinogenesis. Nature. 438:981–987. 2005. View Article : Google Scholar : PubMed/NCBI | |
Tryndyak VP, Muskhelishvili L, Kovalchuk O, Rodriguez-Juarez R, Montgomery B, Churchwell MI, Ross SA, Beland FA and Pogribny IP: Effect of long-term tamoxifen exposure on genotoxic and epigenetic changes in rat liver: Implications for tamoxifen-induced hepatocarcinogenesis. Carcinogenesis. 27:1713–1720. 2006. View Article : Google Scholar : PubMed/NCBI | |
Fan M, Yan PS, Hartman-Frey C, Chen L, Paik H, Oyer SL, Salisbury JD, Cheng AS, Li L, Abbosh PH, et al: Diverse gene expression and DNA methylation profiles correlate with differential adaptation of breast cancer cells to the antiestrogens tamoxifen and fulvestrant. Cancer Res. 66:11954–11966. 2006. View Article : Google Scholar : PubMed/NCBI | |
Snider H, Villavarajan B, Peng Y, Shepherd LE, Robinson AC and Mueller CR: Region-specific glucocorticoid receptor promoter methylation has both positive and negative prognostic value in patients with estrogen receptor-positive breast cancer. Clin Epigenetics. 11:1552019. View Article : Google Scholar : PubMed/NCBI | |
Kala R and Tollefsbol TO: A novel combinatorial epigenetic therapy using resveratrol and pterostilbene for restoring estrogen Receptor-α (ERα) expression in ERα-Negative breast cancer cells. PLoS One. 11:e01550572016. View Article : Google Scholar : PubMed/NCBI | |
Zhang WJ, Xu DF, Fan QX, Wu XA, Wang F, Wang R and Wang LX: Arsenic trioxide restores ERα expression in ERα-negative human breast cancer cells and its treatment efficacy in combination with tamoxifen in xenografts in nude mice. Zhonghua Zhong Liu Za Zhi. 34:645–651. 2012.(In Chinese). PubMed/NCBI | |
Tang B, Peng ZH and Jiang J: The synergistic inhibitory effect of 5-aza-2-deoxycytidine and Tamoxifen on estrogen receptor alpha negative breast cancer cell lines in vitro. Zhonghua Wai Ke Za Zhi. 43:1545–1549. 2005.(In Chinese). PubMed/NCBI | |
Selmin OI, Donovan MG, Skovan B, Paine-Murieta GD and Romagnolo DF: Arsenic-induced BRCA1 CpG promoter methylation is associated with the downregulation of ERα and resistance to tamoxifen in MCF7 breast cancer cells and mouse mammary tumor xenografts. Int J Oncol. 54:869–878. 2019.PubMed/NCBI | |
Wu HT, Liu YE, Hsu KW, Wang YF, Chan YC, Chen Y and Chen DR: MLL3 induced by luteolin causes apoptosis in Tamoxifen-Resistant breast cancer cells through H3K4 monomethylation and suppression of the PI3K/AKT/mTOR Pathway. Am J Chin Med. 48:1221–1241. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jin K, Lan H, Zhang J, Lv J, Chen Y, Yu K and Wang W: UBASH3B promotes tamoxifen resistance and could be negatively regulated by ESR1. Oncotarget. 9:8326–8333. 2018. View Article : Google Scholar : PubMed/NCBI | |
Jayasree PJ, Dutta S, Karemore P and Khandelia P: Crosstalk between m6A RNA methylation and miRNA biogenesis in cancer: An unholy nexus. Mol Biotechnol. Oct 13–2023.doi: 10.1007/s12033-023-00921-w. View Article : Google Scholar | |
Noh EM, Lee YR, Chay KO, Chung EY, Jung SH, Kim JS and Youn HJ: Estrogen receptor α induces down-regulation of PTEN through PI3-kinase activation in breast cancer cells. Mol Med Rep. 4:215–219. 2011.PubMed/NCBI | |
Khatpe AS, Adebayo AK, Herodotou CA, Kumar B and Nakshatri H: Nexus between PI3K/AKT and estrogen receptor signaling in breast cancer. Cancers (Basel). 13:3692021. View Article : Google Scholar : PubMed/NCBI | |
Zhu D, Zeng S, Su C, Li J, Xuan Y, Lin Y, Xu E and Fan Q: The interaction between DNA methylation and tumor immune microenvironment: From the laboratory to clinical applications. Clin Epigenetics. 16:242024. View Article : Google Scholar : PubMed/NCBI | |
Sina AA, Carrascosa LG and Trau M: DNA Methylation-based point-of-care cancer detection: Challenges and possibilities. Trends Mol Med. 25:955–966. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Wang M, Lin Y, Zhao J, Gu H and Li X: Circulating tumor DNA methylation: A promising clinical tool for cancer diagnosis and management. Clin Chem Lab Med. Mar 7–2024.doi: 10.1515/cclm-2023-1327. View Article : Google Scholar | |
Garcia-Ortiz MV, Cano-Ramirez P, Toledano-Fonseca M, Aranda E and Rodriguez-Ariza A: Diagnosing and monitoring pancreatic cancer through cell-free DNA methylation: Progress and prospects. Biomark Res. 11:882023. View Article : Google Scholar : PubMed/NCBI | |
Fan S and Chi W: Methods for genome-wide DNA methylation analysis in human cancer. Brief Funct Genomics. 15:432–442. 2016.PubMed/NCBI | |
Liu B, Wang T, Wang H, Zhang L, Xu F, Fang R, Li L, Cai X, Wu Y, Zhang W and Ye L: Oncoprotein HBXIP enhances HOXB13 acetylation and co-activates HOXB13 to confer tamoxifen resistance in breast cancer. J Hematol Oncol. 11:262018. View Article : Google Scholar : PubMed/NCBI | |
Geter PA, Ernlund AW, Bakogianni S, Alard A, Arju R, Giashuddin S, Gadi A, Bromberg J and Schneider RJ: Hyperactive mTOR and MNK1 phosphorylation of eIF4E confer tamoxifen resistance and estrogen independence through selective mRNA translation reprogramming. Genes Dev. 31:2235–2249. 2017. View Article : Google Scholar : PubMed/NCBI | |
Filipčík P, Curry JR and Mace PD: When Worlds Collide-mechanisms at the interface between phosphorylation and ubiquitination. J Mol Biol. 429:1097–1113. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bailey LT, Northall SJ and Schalch T: Breakers and amplifiers in chromatin circuitry: Acetylation and ubiquitination control the heterochromatin machinery. Curr Opin Struct Biol. 71:156–163. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shukla A, Chaurasia P and Bhaumik SR: Histone methylation and ubiquitination with their cross-talk and roles in gene expression and stability. Cell Mol Life Sci. 66:1419–1433. 2009. View Article : Google Scholar : PubMed/NCBI | |
Separovich RJ, Pang CNI and Wilkins MR: Controlling the controllers: Regulation of histone methylation by phosphosignalling. Trends Biochem Sci. 45:1035–1048. 2020. View Article : Google Scholar : PubMed/NCBI | |
Blackburn SA, Parks RM and Cheung KL: Fulvestrant for the treatment of advanced breast cancer. Expert Rev Anticancer Ther. 18:619–628. 2018. View Article : Google Scholar : PubMed/NCBI | |
Intabli H, Gee JM, Oesterreich S, Yeoman MS, Allen MC, Qattan A and Flint MS: Glucocorticoid induced loss of oestrogen receptor alpha gene methylation and restoration of sensitivity to fulvestrant in triple negative breast cancer. Gene. 851:1470222023. View Article : Google Scholar : PubMed/NCBI | |
Hopcroft L, Wigmore EM, Williamson SC, Ros S, Eberlein C, Moss JI, Urosevic J, Carnevalli LS, Talbot S, Bradshaw L, et al: Combining the AKT inhibitor capivasertib and SERD fulvestrant is effective in palbociclib-resistant ER+ breast cancer preclinical models. NPJ Breast Cancer. 9:642023. View Article : Google Scholar : PubMed/NCBI |