Phospho-ESR1 (S118) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

The coding sequence for the phospho-ESR1 (S118) monoclonal antibody (isolated by immunizing animals with the synthetic phosphopeptide derived from human ESR1 around the phosphorylation site of Ser 118) was cloned into the plasmids and then transfected into cell lines for in vitro expression. The product underwent affinity-chromatography-mediated purification to get the phospho-ESR1 (S118) recombinant monoclonal antibody. This p-S118-ESR1 antibody is a rabbit IgG. It is suitable for the detection of human ESR1 phosphorylated at Ser 118 residue in ELISA and IHC applications.

ESR1 is a ligand-dependent transcription factor that affects the expression of target genes. Two transcription activation functions, AF-1 and AF-2, act in a promoter- and cell-specific manner to activate gene expression. While estrogen (E2) binding regulates the activity of AF-2, phosphorylation at several sites regulates the activity of AF-1. S118, one of these phosphorylation sites, is of special relevance because its mutation inhibits ESR1 function considerably. S118 has been demonstrated to be phosphorylated by the ERK1/2 mitogen-activated protein kinases (MAPK) and the cyclin-dependent protein kinase Cdk7 in previous research.

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The coding sequence for the phospho-ESR1 (S118) monoclonal antibody, generated by immunizing animals with a synthetic phosphopeptide derived from human ESR1 encompassing the Ser 118 phosphorylation site, was cloned into plasmids and subsequently transfected into cell lines for in vitro expression. The product underwent affinity chromatography-mediated purification to obtain the phospho-ESR1 (S118) recombinant monoclonal antibody. This p-S118-ESR1 antibody, a rabbit IgG, is suitable for detecting human ESR1 phosphorylated at Ser 118 residue in ELISA and IHC applications.

ESR1, a ligand-dependent transcription factor, exerts influence on the expression of target genes. Two transcription activation functions, AF-1 and AF-2, operate in a promoter- and cell-specific manner to activate gene expression. While estrogen (E2) binding regulates the activity of AF-2, phosphorylation at multiple sites governs the activity of AF-1. S118, one of these phosphorylation sites, holds particular significance due to its mutation causing substantial inhibition of ESR1 function. Previous research has demonstrated that S118 undergoes phosphorylation by the ERK1/2 mitogen-activated protein kinases (MAPK) and the cyclin-dependent protein kinase Cdk7.

Form
Liquid
Lead Time
Typically, we can dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
7*/654 isoform antibody; 7*/819 2 isoform antibody; 7*/822 isoform antibody; 8*/901 isoform antibody; 8*/941 isoform antibody; DKFZp686N23123 antibody; ER alpha antibody; ER antibody; ER-alpha antibody; Era antibody; ESR antibody; ESR1 antibody; ESR1_HUMAN antibody; ESRA antibody; Estradiol receptor antibody; Estrogen nuclear receptor alpha antibody; Estrogen receptor 1 antibody; Estrogen receptor alpha 3*,4,5,6,7*/822 isoform antibody; Estrogen receptor alpha antibody; Estrogen receptor alpha delta 3*,4,5,6,7*,8*/941 isoform antibody; Estrogen receptor alpha delta 3*,4,5,6,7*/819 2 isoform antibody; Estrogen receptor alpha delta 4 +49 isoform antibody; Estrogen receptor alpha delta 4*,5,6,7*/654 isoform antibody; Estrogen receptor alpha delta 4*,5,6,7,8*/901 isoform antibody; Estrogen receptor alpha E1 E2 1 2 antibody; Estrogen receptor alpha E1 N2 E2 1 2 antibody; Estrogen receptor antibody; ESTRR antibody; NR3A1 antibody; Nuclear receptor subfamily 3 group A member 1 antibody
Target Names
Uniprot No.

Target Background

Function
ESR1, also known as the estrogen receptor alpha, is a nuclear hormone receptor. Steroid hormones and their receptors play a critical role in regulating eukaryotic gene expression, influencing cellular proliferation and differentiation in target tissues. Ligand-dependent nuclear transactivation involves either direct homodimer binding to a palindromic estrogen response element (ERE) sequence or association with other DNA-binding transcription factors, such as AP-1/c-Jun, c-Fos, ATF-2, Sp1, and Sp3, to mediate ERE-independent signaling. Ligand binding triggers a conformational change, facilitating subsequent or combinatorial association with multiprotein coactivator complexes through LXXLL motifs of their respective components. Mutual transrepression occurs between the estrogen receptor (ER) and NF-kappa-B in a cell-type specific manner. This interaction decreases NF-kappa-B DNA-binding activity and inhibits NF-kappa-B-mediated transcription from the IL6 promoter, displacing RELA/p65 and associated coregulators from the promoter. ESR1 is recruited to the NF-kappa-B response element of the CCL2 and IL8 promoters and can displace CREBBP. It co-localizes with NF-kappa-B components RELA/p65 and NFKB1/p50 on ERE sequences. Additionally, ESR1 can synergistically activate transcription alongside NF-kappa-B involving recruitment to adjacent response elements, a process that includes CREBBP. ESR1 can activate the transcriptional activity of TFF1. It also mediates membrane-initiated estrogen signaling involving various kinase cascades. ESR1 is essential for MTA1-mediated transcriptional regulation of BRCA1 and BCAS3. It participates in the activation of NOS3 and endothelial nitric oxide production. Isoforms lacking one or several functional domains are thought to modulate transcriptional activity by competitive ligand or DNA binding and/or heterodimerization with the full-length receptor. ESR1 binds to ERE and inhibits isoform 1.
Gene References Into Functions
  1. Estrogen-induced miR-191 was identified as a direct upstream regulator of DAB2 in ER-positive breast cancer cells. PMID: 29247596
  2. The comprehensive genomic insights from this study contribute to a deeper understanding of the biological roles of ER1 in breast cancer. PMID: 30301189
  3. A relationship was found between rs2046210 and rs3803662, and the risk of developing breast cancer in Vietnamese women. The A allele is associated with an increased risk for both rs2046210 (OR [95% CI] = 1.43 [1.14 - 1.78], P = 0.0015) and rs3803662 (OR [95% CI] = 1.45 [1.16 - 1.83], P = 0.001). These findings suggest that two polymorphisms, rs2046210 in ESR1 and rs3803662 in TNRC9, are associated with breast cancer risk in the Vietnamese population. PMID: 30078824
  4. This study provides evidence that Oestrogen receptor alpha can enhance the odonto/osteogenic differentiation of stem cells from the apical papilla via ERK and JNK MAPK pathways. PMID: 30069950
  5. No association was found between polymorphisms in genes encoding estrogen receptors (ESR1 and ESR2) and excreted BPA levels in orthodontic patients after bracket bonding. PMID: 29961922
  6. Analysis of genome-wide ER binding sites identified mutant ER unique recruitment mediating the allele-specific transcriptional program. PMID: 29438694
  7. This study identifies RNF8 as a co-activator of ERalpha, enhancing ERalpha stability through a post-transcriptional pathway, providing novel insights into the mechanisms by which RNF8 promotes cell growth in ERalpha-positive breast cancer. PMID: 28216286
  8. Reduced expression of ERbeta1 in female ERalpha-negative papillary thyroid carcinoma patients is associated with increased disease progression. PMID: 29655286
  9. ERbeta exhibits a heterogeneous distribution in deep infiltrating endometriosis. PMID: 29383962
  10. The ER-alpha36/EGFR signaling loop promotes growth in hepatocellular carcinoma cells. PMID: 29481815
  11. This study investigated the presence and localization of estrogen receptors (ERs), progesterone receptors (PRs), and androgen receptors (ARs) in both healthy and varicose vein wall cells, and their relationship with gender. PMID: 30250632
  12. Estrogen receptor-alpha was expressed exclusively in women and showed a positive correlation with the amount of fungi in oral paracoccidioidomycosis, while progesterone receptor was observed in both genders and did not exhibit a correlation with estrogen receptor-alpha or fungi counting. PMID: 29796757
  13. ERalpha upregulates vinculin expression in breast cancer cells; Loss of vinculin promotes amoeboid features of cancer cells. PMID: 28266545
  14. Polymorphisms in ESR1 and ESR2 genes do not predict in vitro fertilization outcome. PMID: 29916276
  15. High ESR1 expression is associated with metastasis in breast cancer. PMID: 29187405
  16. The G/G XbaI genotype of the ESR1 gene is associated with an increased risk of breast cancer. PMID: 29893332
  17. miR-221 may impair the protective effect of estrogen in degenerated cartilaginous endplate cells by targeting estrogen receptor alpha. PMID: 29529124
  18. Results indicated that NAT1 and ESR1 expression were elevated in primary breast tumor samples compared to normal breast tissue samples, and in ER+ primary breast tumors compared to ER- tumors. Furthermore, NAT1 and ESR1 expression appear to share overlapping regulation. PMID: 29901116
  19. All patients without ESR1 mutations identified by molecular barcode next-generation sequencing (MB-NGS) were confirmed to lack mutations upon analysis using ddPCR. These findings suggest that MB-NGS can successfully detect ESR1 mutations in cfDNA with a higher sensitivity (0.1%) than conventional NGS and is considered clinically equivalent to ddPCR. PMID: 28905136
  20. An association was observed between the presence of specific genotypes at three ESR1 polymorphisms (rs2234693, rs6902771, rs7774230) and one ESR2 polymorphism (rs3020449), and the presence of metabolic syndrome in postmenopausal women. PMID: 30049354
  21. Plasma samples exhibited a higher frequency of ESR1 and PIK3CA mutations compared to serum in 33 MBC patients, suggesting that serum samples should not be considered the preferred source of cfDNA. PMID: 29689710
  22. This study suggests that miR-125a-3p can act as a novel tumor suppressor in ER(+) breast cancer by targeting CDK3, potentially offering a novel therapeutic approach for TamR breast cancer therapy. PMID: 28939591
  23. A significant finding of this study is that 20% of patients with breast cancer bone metastases exhibited a receptor discrepancy between the primary tumor and subsequent bone metastases. Loss of hormone receptor (ER and/or PR) expression and gain of HER2 overexpression were the most commonly observed changes. PMID: 28975433
  24. This study reports a pivotal role of IGF-IR in regulating the aggressiveness of ERalpha-positive breast cancer cells and in modulating the expression levels of various extracellular matrix molecules. PMID: 28079144
  25. Associations between PvuII (T>C) and XbaI (A>G) polymorphisms of the estrogen receptor alpha (ESR1) gene with type 2 diabetes mellitus (T2DM) or metabolic syndrome (MetS) are reported. PMID: 29883973
  26. The ERalpha gene appears to play a crucial role in stress urinary incontinence in the premenopausal period. PMID: 29769420
  27. This study reports the first discovery of naturally occurring ESR1 (Y537C) and ESR1 (Y537S) mutations in MCF7 and SUM44 ESR1-positive cell lines following acquired resistance to long-term estrogen deprivation (LTED) and subsequent resistance to fulvestrant (ICIR). Mutations were enriched over time, impacting ESR1 binding to the genome and altering the ESR1 interactome. PMID: 29192207
  28. Concomitant high expression of ERalpha36, GRP78, and GRP94 is associated with aggressive papillary thyroid cancer behavior and may serve as a predictor for extrathyroid extension, lymph node metastasis, and distant metastasis. PMID: 29368272
  29. Estrogen receptor-1 is a key regulator of HIV-1 latency, imposing gender-specific restrictions on the latent reservoir. PMID: 30061382
  30. Down-regulation of ESR1 gene expression was enhanced by the development of breast cancer. PMID: 29543921
  31. This study aimed to assess whether fibrosis markers, estrogen receptor (ER)alpha, and the stromal derived factor (SDF)1/CXC chemokine receptor type 4 (CXCR4) axis are abnormally expressed in the endometrium of women with intrauterine adhesions. PMID: 29568895
  32. The frequency of alleles and genotypes of polymorphisms FSHR(-29G/A) and ESRI (XbaI A/G) in women with normal to poor response did not exhibit a significant correlation. PMID: 29526845
  33. Each estrogen receptor alpha and estrogen receptor beta gene polymorphism might have a differential impact on postmenopausal osteoporosis risk and bone mineral density across various ethnicities. PMID: 29458346
  34. The results suggest that the minor allele A of the ESR1 gene is associated with an increased risk of developing arterial hypertension in men. PMID: 29658078
  35. This study found that tamoxifen treatment induced a decrease in PRMT2 and an increase in ER-alpha36, as well as ER-alpha36-mediated non-genomic effects in the MDA-MB-231 breast cancer cell line. PMID: 29620287
  36. ESR1 mutations are not associated with clinical resistance to fulvestrant in breast cancer patients. PMID: 27174596
  37. Overexpression of COPS5, through its isopeptidase activity, leads to ubiquitination and proteasome-mediated degradation of NCoR, a key corepressor for ERalpha and tamoxifen-mediated suppression of ERalpha target genes. PMID: 27375289
  38. ESR alpha PvuII and XbaI polymorphisms have no association with systemic lupus erythematosus. The combination of the TC/AA and CC/GG genotypes was associated with SLE susceptibility. PMID: 29356461
  39. Estrogen receptor (ER) and progesterone receptor (PR) expression in endometrial carcinoma (EC) were significantly higher than those in the paracarcinoma tissue and control. PMID: 29081408
  40. ESR1 promoter methylation was an independent risk factor and exhibited a high value for predicting 28-day mortality from acute-on-chronic hepatitis B liver failure. PMID: 29082740
  41. By analyzing different estrogen receptor-alpha(ER-a)-positive and ER-a-negative breast cancer cell lines, this study defined the role of CCN5 in the leptin-mediated regulation of growth and invasive capacity. PMID: 29370782
  42. This study identified ESR1 as a direct target of miR-301a-3p. PMID: 29763890
  43. This study reports the first discovery of ESR1 methylation in plasma ctDNA of patients with HGSC. The agreement between ESR1 methylation in primary tumors and paired ctDNA is statistically significant. PMID: 29807696
  44. This study describes the development of a novel class of ERa AF2 inhibitors, which have the potential to effectively inhibit ERa activity through a unique mechanism and to circumvent the issue of mutation-driven resistance in breast cancer. PMID: 29462880
  45. The interaction between P2X7R rs3751143 and ER-alpha PvuII confers a significantly increased susceptibility to osteoporosis in Chinese postmenopausal women. PMID: 28884379
  46. Alcohol consumption may exert differential effects on concordant and discordant receptor subtypes of breast cancer. PMID: 29353824
  47. ERalpha and ERbeta mRNA expression was significantly higher (p < 0.05) in tumor tissues relative to their paired normal mucosa and exhibited an inverse correlation with survival outcome. PMID: 29390981
  48. High ESR1 expression is associated with papillary thyroid carcinoma. PMID: 28124274
  49. Oral administration of RAD140 significantly inhibited the growth of AR/ER(+) breast cancer patient-derived xenografts (PDX). Treatment with RAD140 resulted in AR activation and suppression of the ER pathway, including the ESR1 gene. PMID: 28974548
  50. Polymorphism in the ERalpha gene is associated with an increased risk of advanced Pelvic Organ Prolapse. However, polymorphism in the LAMC1 gene does not appear to be associated with this risk. PMID: 29241914

Show More

Hide All

Database Links

HGNC: 3467

OMIM: 133430

KEGG: hsa:2099

STRING: 9606.ENSP00000206249

UniGene: Hs.208124

Involvement In Disease
Estrogen resistance (ESTRR)
Protein Families
Nuclear hormone receptor family, NR3 subfamily
Subcellular Location
[Isoform 1]: Nucleus. Cytoplasm. Cell membrane; Peripheral membrane protein; Cytoplasmic side.; Nucleus. Golgi apparatus. Cell membrane. Note=Colocalizes with ZDHHC7 and ZDHHC21 in the Golgi apparatus where most probably palmitoylation occurs. Associated with the plasma membrane when palmitoylated.
Tissue Specificity
Widely expressed. Not expressed in the pituitary gland.; [Isoform 3]: Widely expressed, however not expressed in the pituitary gland.

Q&A

What is the biological significance of ESR1 phosphorylation at serine 118?

Phosphorylation of serine 118 on estrogen receptor alpha (ESR1) is a critical post-translational modification that significantly enhances its transcriptional activity, leading to increased expression of estrogen-responsive genes . This phosphorylation represents a key regulatory mechanism in estrogen signaling pathways and affects downstream cellular processes including cell proliferation and differentiation . The S118 site has been demonstrated to be phosphorylated by multiple kinases, including ERK1/2 mitogen-activated protein kinases (MAPK) and the cyclin-dependent protein kinase Cdk7, establishing it as a convergence point for multiple signaling pathways . Mutation of this serine residue substantially inhibits ESR1 function, underscoring its biological importance in normal physiology and pathological conditions .

How do recombinant monoclonal antibodies against phospho-S118 ESR1 differ from polyclonal alternatives?

Recombinant monoclonal antibodies against phospho-S118 ESR1 are produced by cloning the coding sequence for the antibody (initially isolated through animal immunization with synthetic phosphopeptides) into plasmids which are then transfected into cell lines for expression . The resulting antibodies undergo affinity-chromatography-mediated purification to yield a homogeneous antibody population . Unlike polyclonal alternatives, which contain a heterogeneous mixture of antibodies targeting different epitopes, these recombinant monoclonals offer consistent lot-to-lot reproducibility, higher specificity for the phosphorylated S118 site with minimal cross-reactivity to non-phosphorylated ESR1, and reduced background in experimental applications . Polyclonal antibodies, such as those containing rabbit IgG at 1mg/ml in PBS with additives like sodium azide and glycerol, may offer broader epitope recognition but with potential variability between production batches .

What applications are suitable for phospho-S118 ESR1 antibodies in breast cancer research?

Phospho-S118 ESR1 antibodies are valuable tools for multiple applications in breast cancer research, including:

  • Western blotting (WB) to quantify changes in S118 phosphorylation levels in response to treatments or genetic manipulations

  • Immunohistochemistry (IHC) on formalin/PFA-fixed paraffin-embedded tissue sections to evaluate S118 phosphorylation in patient samples and xenograft models

  • Immunocytochemistry (ICC) to visualize subcellular localization of phosphorylated receptor

  • Immunofluorescence (IF) to study co-localization with other signaling molecules

  • ELISA-based transcription factor activity assays to measure functional consequences of S118 phosphorylation on transcriptional output

These applications enable researchers to investigate how phosphorylation at S118 correlates with breast cancer progression, treatment response, and resistance mechanisms .

How does phosphorylation at S118 interact with ESR1 mutations in breast cancer progression and treatment resistance?

The relationship between S118 phosphorylation and ESR1 mutations represents a complex mechanism in breast cancer biology. ESR1 mutations, particularly those in the hormone-binding domain (HBD) such as Y537S and E380D, can lead to ligand-independent activation of the receptor . Research indicates that S118 phosphorylation and certain ESR1 mutations may cooperatively enhance transcriptional activity through distinct but complementary mechanisms .

In the context of treatment resistance, phosphorylation at S118 has been implicated in reduced sensitivity to tamoxifen, particularly when growth factor signaling pathways are active . This parallels findings with the K303R ESR1 mutation, which similarly confers tamoxifen resistance when growth factor signaling is engaged . Evidence suggests that constitutive phosphorylation of S118 may occur in cells expressing certain ESR1 mutations, creating a feed-forward loop that maintains receptor activity even in the presence of endocrine therapies .

Methodologically, investigating these interactions requires combination approaches such as site-directed mutagenesis (to create S118A phospho-null mutants), proximity ligation assays to study protein-protein interactions, and chromatin immunoprecipitation followed by sequencing (ChIP-seq) to map how phosphorylation affects genomic binding patterns in the context of different ESR1 mutations .

What are the technical considerations for validating specificity of phospho-S118 ESR1 antibodies in experimental systems?

Validating the specificity of phospho-S118 ESR1 antibodies requires multiple complementary approaches:

  • Phosphatase treatment controls: Treating one sample set with lambda phosphatase before antibody incubation should abolish signal if the antibody is truly phospho-specific .

  • Peptide competition assays: Pre-incubating the antibody with excess phospho-S118 peptide should eliminate specific binding, while incubation with non-phosphorylated peptide should not affect binding .

  • Genetic controls: Testing the antibody in:

    • S118A mutant cell lines (where serine is replaced with alanine, preventing phosphorylation)

    • ESR1 knockout cell lines (which should show no signal)

    • Cells treated with kinase inhibitors that prevent S118 phosphorylation

  • Cross-reactivity assessment: Confirming the antibody does not detect other phosphorylated proteins or non-phosphorylated ESR1 .

  • Multiple detection methods: Validating findings across different applications (WB, IHC, IF) to ensure consistent results .

Antibody validation protocols should involve both positive controls (such as MCF-7 breast cancer cells treated with estradiol, which induces S118 phosphorylation) and negative controls to establish a robust experimental framework .

How do technical variations in detection methods affect the reported prevalence of ESR1 phosphorylation in breast cancer specimens?

Technical variations in detection methods significantly impact the reported prevalence of ESR1 phosphorylation in breast cancer specimens, creating challenges for data interpretation and cross-study comparisons. The literature reveals substantial discrepancies in detection rates that can be attributed to several methodological factors:

  • Antibody selection: Different commercial antibodies exhibit varying sensitivities and specificities for phospho-S118 ESR1, with recombinant monoclonal antibodies generally providing more consistent results than polyclonal alternatives .

  • Tissue preparation protocols: The phosphorylation state can be affected by:

    • Time from tissue removal to fixation (phosphatases remain active)

    • Fixation method and duration

    • Antigen retrieval techniques (boiling in sodium citrate buffer at pH6.0 for 15 minutes is recommended for optimal epitope exposure)

  • Detection systems: Signal amplification methods vary in sensitivity, with techniques like tyramide signal amplification potentially detecting lower phosphorylation levels than conventional methods .

  • Scoring methods: Threshold selection for considering a specimen "positive" varies between studies, with some using H-score methods and others employing percentage of positive cells or intensity measurements .

  • Control selection: The appropriate phosphorylation-negative controls are essential but inconsistently implemented across studies .

This methodological heterogeneity parallels challenges seen with ESR1 mutation detection, where techniques like fluorescent in situ hybridization (FISH) versus multiplex ligation-dependent probe amplification have yielded contradictory results regarding ESR1 amplification prevalence .

What experimental controls are essential when using phospho-S118 ESR1 antibodies to study treatment response in breast cancer models?

When studying treatment response in breast cancer models using phospho-S118 ESR1 antibodies, several essential controls must be incorporated:

  • Pathway activation controls:

    • Positive control: Stimulate cells with estradiol (E2) to induce S118 phosphorylation

    • Inhibition control: Treat with MEK/ERK inhibitors to block MAPK-mediated phosphorylation of S118

    • Kinase control: Include CDK7 inhibitors to block transcription-associated phosphorylation

  • Antibody specificity controls:

    • Total ESR1 measurement: Parallel detection with non-phospho-specific ESR1 antibody to normalize phosphorylation levels

    • Phosphatase treatment: Lambda phosphatase treatment to verify phospho-specificity

    • Blocking peptide: Competition with phospho-S118 peptide immunogen

  • Genetic controls:

    • ESR1 knockdown/knockout cells (negative control)

    • S118A mutant cells (phospho-null)

    • S118E mutant cells (phosphomimetic positive control)

  • Time-course measurements:

    • Capture both rapid (30 minutes) and sustained (24+ hours) phosphorylation changes

    • Include multiple time points to avoid missing transient effects

  • Biological replicates:

    • Test across multiple cell lines with varying ESR1 mutation status

    • Include patient-derived xenograft models that maintain tumor heterogeneity

This comprehensive control framework ensures that observed changes in S118 phosphorylation can be reliably attributed to the treatment intervention rather than technical artifacts or normal biological fluctuations.

How can researchers optimize immunohistochemical protocols for phospho-S118 ESR1 detection in archived FFPE breast cancer specimens?

Optimizing immunohistochemical protocols for phospho-S118 ESR1 detection in archived formalin-fixed paraffin-embedded (FFPE) breast cancer specimens requires systematic adjustment of multiple parameters:

  • Tissue preparation and antigen retrieval:

    • Use freshly cut sections (4-5 μm thickness)

    • Perform antigen retrieval by boiling in sodium citrate buffer (pH 6.0) for 15 minutes

    • Consider extended retrieval times (up to 20 minutes) for older archived specimens

    • Evaluate alternative retrieval buffers (EDTA, pH 9.0) if standard protocols yield weak signals

  • Blocking and antibody conditions:

    • Block endogenous peroxidase with 3% hydrogen peroxide for 20 minutes

    • Use normal goat serum as blocking buffer at 37°C for 30 minutes

    • Test antibody dilutions ranging from 1:50 to 1:200

    • Extend primary antibody incubation to overnight at 4°C

    • Incorporate phosphatase inhibitors in all buffers to preserve phosphorylation status

  • Detection system optimization:

    • Compare polymer-based versus avidin-biotin complex (ABC) detection systems

    • Consider tyramide signal amplification for older specimens with potential epitope degradation

    • Optimize DAB exposure time through timed development

  • Validation approach:

    • Run parallel sections with total ESR1 antibody for normalization

    • Include known positive controls (e.g., MCF-7 cells treated with E2)

    • Incorporate on-slide negative controls (phosphatase-treated section)

    • Evaluate signal in normal breast tissue adjacent to tumor as internal control

  • Quantification methods:

    • Adopt digital image analysis with consistent threshold settings

    • Consider H-score method (intensity × percentage positive cells) for semi-quantitative assessment

    • Document nuclear versus cytoplasmic staining patterns separately

This optimized protocol should be validated against fresh frozen tissue specimens when available to confirm that phosphorylation status is accurately preserved and detected in the FFPE material.

What approaches can resolve contradictory findings regarding the relationship between phospho-S118 ESR1 status and endocrine therapy response?

Resolving contradictory findings regarding phospho-S118 ESR1 status and endocrine therapy response requires multi-faceted methodological approaches:

  • Standardization of phospho-S118 detection:

    • Establish consensus protocols for tissue handling, fixation, and immunohistochemistry

    • Implement digital pathology with machine learning algorithms for objective quantification

    • Develop calibration standards using cell lines with known S118 phosphorylation levels

  • Contextualization with other biomarkers:

    • Analyze S118 phosphorylation in conjunction with:

      • Other ESR1 phosphorylation sites (S104, S167, S305)

      • Growth factor receptor expression and activation (HER2, EGFR, IGF-1R)

      • ESR1 mutation status, particularly in the hormone-binding domain

      • Progesterone receptor (PR) and Ki-67 status

  • Functional validation studies:

    • Create isogenic cell line panels with wild-type ESR1, S118A (phospho-null), and S118E (phosphomimetic) mutations

    • Perform drug response curves across these models with:

      • Selective estrogen receptor modulators (SERMs, e.g., tamoxifen)

      • Selective estrogen receptor degraders (SERDs, e.g., fulvestrant)

      • Aromatase inhibitors in hormone-supplemented conditions

  • Time-resolved analyses:

    • Implement sequential biopsies (pre-treatment, early-on-treatment, progression)

    • Track dynamic changes in S118 phosphorylation rather than single time points

    • Correlate with clinical outcomes and treatment response markers

  • Computational approaches:

    • Develop multivariate models incorporating:

      • S118 phosphorylation intensity and distribution

      • Patient characteristics (age, menopausal status)

      • Treatment history

      • Tumor molecular subtype

    • Apply machine learning to identify patterns not discernible through conventional statistics

This comprehensive approach acknowledges that S118 phosphorylation exists within a complex signaling network, and its impact on endocrine therapy response likely depends on the broader molecular context of each tumor.

How does phospho-S118 ESR1 analysis complement genomic profiling in personalized breast cancer treatment strategies?

Phospho-S118 ESR1 analysis provides crucial proteomic information that complements genomic profiling in several key ways:

  • Post-translational information beyond genetic alterations:

    • While genomic profiling identifies ESR1 mutations, phospho-S118 analysis reveals functional receptor activation state

    • Patients with wild-type ESR1 genes may still have dysregulated receptor activity due to aberrant phosphorylation

    • Phospho-S118 status may identify actionable pathway activation not evident from genetic profiling alone

  • Predictive biomarker potential:

    • Integrating phospho-S118 ESR1 status with genomic profiles may better predict response to:

      • Endocrine therapies (tamoxifen, aromatase inhibitors)

      • CDK4/6 inhibitors

      • PI3K/AKT/mTOR pathway inhibitors

    • This integration could identify patients likely to benefit from combination therapies targeting both genomic alterations and phosphorylation-mediating pathways

  • Resistance mechanism characterization:

    • Genomic profiling may reveal ESR1 mutations (Y537S, E380D) associated with endocrine resistance

    • Phospho-S118 analysis can determine whether these mutations lead to constitutive phosphorylation

    • Combined analysis clarifies whether resistance emerges through ligand-independent activation or altered cofactor recruitment

  • Temporal dynamics and heterogeneity:

    • Serial phospho-S118 measurement captures dynamic treatment responses not detectable by static genomic analysis

    • Spatial heterogeneity in phosphorylation patterns across tumor regions complements genomic heterogeneity data

    • Single-cell approaches combining genomic and phospho-proteomic analysis may reveal resistant subpopulations

  • Therapeutic targeting opportunities:

    • Identifies patients who might benefit from therapies targeting kinases responsible for S118 phosphorylation

    • Suggests combinatorial approaches targeting both genetic alterations and phosphorylation pathways

    • Enables rational design of clinical trials incorporating both genomic and phosphorylation biomarkers

Methodologically, this integration requires careful correlation of phospho-S118 immunohistochemistry results with next-generation sequencing data from the same specimens, ideally using spatial registration techniques to align specific tumor regions.

What are the challenges and solutions for developing quantitative assays to measure phospho-S118 ESR1 levels in liquid biopsies?

Developing quantitative assays for phospho-S118 ESR1 detection in liquid biopsies presents significant challenges with emerging technical solutions:

Challenges:

  • Phosphoprotein stability issues:

    • Phosphorylation marks are highly labile and susceptible to rapid dephosphorylation by phosphatases in blood

    • Standard blood collection tubes lack phosphatase inhibitors

    • Delay between sample collection and processing causes signal loss

  • Low abundance of target protein:

    • Circulating tumor cells (CTCs) or extracellular vesicles containing phospho-ESR1 are rare events

    • Background from non-tumor cells creates noise in detection systems

    • Traditional antibody-based methods lack sufficient sensitivity

  • Standardization difficulties:

    • Variations in pre-analytical handling significantly impact phosphorylation preservation

    • Lack of calibration standards for absolute quantification

    • Biological variability in baseline phosphorylation levels

Solutions and Emerging Approaches:

  • Optimized sample collection protocols:

    • Specialized blood collection tubes containing phosphatase and protease inhibitor cocktails

    • Immediate sample processing or stabilization within 30 minutes of collection

    • Standardized temperature control during transport (4°C)

  • Enhanced sensitivity detection methods:

    • Digital ELISA platforms (e.g., Simoa technology) with single-molecule detection capability

    • Proximity extension assays combining antibody specificity with PCR amplification

    • Mass spectrometry with phosphopeptide enrichment using titanium dioxide or immobilized metal affinity chromatography

  • CTC-specific approaches:

    • Microfluidic isolation of CTCs followed by on-chip immunocytochemistry for phospho-S118

    • Single-cell phosphoproteomics after CTC isolation

    • Combined genomic and phosphoproteomic analysis of identical CTCs

  • Extracellular vesicle (EV) analysis:

    • EV isolation from plasma followed by phospho-S118 ESR1 quantification

    • Multiparameter EV characterization with phospho-specific antibodies

    • EV subpopulation analysis based on surface markers and phosphoprotein content

  • Artificial intelligence integration:

    • Machine learning algorithms to normalize for pre-analytical variables

    • Pattern recognition across multiple phosphorylation sites

    • Integration of phosphoproteomic data with other liquid biopsy markers

These approaches collectively aim to overcome the significant technical hurdles while providing clinically relevant information on phospho-S118 ESR1 status that could guide treatment decisions without invasive tissue biopsies.

How might phospho-S118 ESR1 analysis contribute to understanding resistance mechanisms in metastatic breast cancer?

Phospho-S118 ESR1 analysis offers crucial insights into resistance mechanisms in metastatic breast cancer through multiple research avenues:

  • Pathway bypass mechanisms:

    • Enhanced S118 phosphorylation despite endocrine therapy suggests activation of alternative kinase pathways

    • Comparative analysis of primary tumors versus metastatic lesions can reveal evolution of phosphorylation patterns

    • Serial biopsies during treatment can identify when phosphorylation status changes precede clinical progression

  • Interaction with ESR1 mutations:

    • Research indicates that hormone-binding domain mutations in ESR1 (Y537S, E380D) may function synergistically with S118 phosphorylation

    • Phospho-S118 analysis can determine whether ESR1 mutations lead to constitutive phosphorylation or altered phosphorylation dynamics

    • Methodologically, this requires simultaneous assessment of mutation status and phosphorylation levels in the same specimens

  • Adaptive response to targeted therapies:

    • CDK4/6 inhibitors, PI3K inhibitors, and mTOR inhibitors can all impact signaling networks affecting S118 phosphorylation

    • Increased phospho-S118 following treatment may indicate compensatory pathway activation

    • Decreased phospho-S118 without clinical response suggests downstream resistance mechanisms

  • Growth factor receptor crosstalk:

    • S118 phosphorylation can be driven by growth factor signaling through MAPK pathways

    • K303R ESR1 mutation research demonstrates how enhanced growth factor receptor-ER crosstalk contributes to tamoxifen resistance

    • Similar mechanisms may operate with other ESR1 mutations or in wild-type receptors with dysregulated phosphorylation

  • Transcriptional reprogramming:

    • Phospho-S118 status affects genomic binding patterns and transcriptional output

    • The K303R mutation alters transcriptional programs with enhanced expression of IGF-1R signaling components

    • Similar analyses with phospho-S118 can reveal how phosphorylation reshapes the transcriptome during resistance development

Research approaches combining phospho-specific antibodies with techniques like ChIP-seq, RNA-seq, and phosphoproteomics in patient-derived xenograft models that faithfully recapitulate resistance mechanisms will be particularly valuable in elucidating these complex interactions.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.