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.
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.
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 .
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 .
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 .
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 .
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:
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 .
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:
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 .
When studying treatment response in breast cancer models using phospho-S118 ESR1 antibodies, several essential controls must be incorporated:
Pathway activation controls:
Antibody specificity controls:
Genetic controls:
Time-course measurements:
Biological replicates:
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.
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:
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.
Resolving contradictory findings regarding phospho-S118 ESR1 status and endocrine therapy response requires multi-faceted methodological approaches:
Standardization of phospho-S118 detection:
Contextualization with other biomarkers:
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:
Time-resolved analyses:
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.
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.
Developing quantitative assays for phospho-S118 ESR1 detection in liquid biopsies presents significant challenges with emerging technical solutions:
Challenges:
Phosphoprotein stability issues:
Low abundance of target protein:
Standardization difficulties:
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.
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:
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.