ESR1 antibodies are immunological reagents designed to target the estrogen receptor alpha (ERα), encoded by the ESR1 gene. ERα is a nuclear receptor activated by estrogen, playing a central role in regulating gene expression in hormone-responsive tissues, including breast cancer . These antibodies are critical tools in research, diagnostics, and therapeutic development, enabling the detection and characterization of ERα expression, phosphorylation, and mutations .
ESR1 antibodies are utilized across multiple experimental platforms:
ESR1 antibodies exhibit diverse host species, clonality, and epitope targets, influencing their utility:
ESR1 mutations (e.g., Y537S, D538G) are frequent in endocrine-resistant metastatic breast cancer (MBC). These mutations confer ligand-independent ERα activity, driving tumor progression . Antibodies detecting ERα expression help identify patients with ESR1 mutations, which correlate with distinct clinical outcomes:
Focal ESR1 amplification (detected via IHC or FISH) is an independent predictor of favorable outcomes in postmenopausal breast cancer:
Outcome | ESR1 Amplification | No Amplification | Source |
---|---|---|---|
Distant Recurrence-Free Survival | HR = 0.48 (10-year follow-up) | HR = 1.0 | |
Breast Cancer–Specific Survival | HR = 0.47 | HR = 1.0 |
Cross-Reactivity: Polyclonal antibodies may bind non-specific epitopes, requiring blocking peptide validation .
Clinical Utility: ESR1 ctDNA testing (via digital PCR) shows promise in predicting treatment responses but lacks standardized protocols .
Therapeutic Targeting: Mutant ERα-specific antibodies are under investigation to address resistance mechanisms .
Estrogen receptor α (ERα, ER; NR3A1), a member of the steroid hormone receptor family, is a ligand-activated transcription factor. ERα contains DNA binding and ligand binding domains and is expressed in various tissues, including the ovary, uterus, breast, prostate, testis, epididymis, bones, bladder, kidney, heart, vessel wall, pituitary gland, and hypothalamus. ER plays crucial roles in maintaining the reproductive, cardiovascular, musculoskeletal, and central nervous systems. Its natural ligand, 17β-estradiol, is a classical estrogenic compound. Binding of estrogen to ER induces conformational changes, triggering events like dissociation from heat shock proteins, receptor dimerization, phosphorylation, and association of the hormone-activated receptor with specific regulatory elements in target genes.
Supplied at a concentration of 1mg/ml in PBS buffer after reconstitution.
LSERASC1.
ESR1 (Estrogen Receptor 1) is a nuclear receptor protein that functions primarily in chromatin remodeling and transcriptional regulation. It has a canonical amino acid length of 595 residues and a protein mass of 66.2 kilodaltons, with 4 distinct isoforms identified to date. The protein is localized in multiple cellular compartments including the cell membrane, nucleus, and cytoplasm .
ESR1 is particularly significant for research due to its central role in hormone-responsive cancers, especially breast cancer, where mutations in this gene are frequently associated with endocrine therapy resistance. The protein is notably expressed in reproductive tissues including the vagina, fallopian tube, endometrium, cervix, and breast tissue, making it a critical target for reproductive and oncological research .
ESR1 antibodies are employed across multiple experimental platforms with Western Blot being the most common application. Other significant applications include:
Immunohistochemistry (IHC) for tissue expression analysis
Enzyme-Linked Immunosorbent Assay (ELISA) for quantitative protein detection
Immunocytochemistry (ICC) for cellular localization studies
Immunofluorescence (IF) for subcellular visualization
Chromatin Immunoprecipitation (ChIP) for studying DNA-protein interactions
Each application requires specific consideration of antibody characteristics including specificity, sensitivity, and validated epitope recognition to ensure reliable experimental outcomes in different research contexts.
Selection of appropriate ESR1 antibodies should be guided by several technical considerations:
Epitope specificity: For mutation studies, select antibodies targeting regions outside the ligand-binding domain (LBD) where common mutations (D538G, Y537S) occur to avoid interference with antibody binding
Isoform recognition: Determine which of the 4 known isoforms need detection and select antibodies accordingly
Cross-reactivity: Verify cross-reactivity with target species (human, mouse, rat) based on experimental needs
Validated applications: Confirm the antibody has been validated specifically for your intended application (WB, IHC, IF)
Monoclonal vs. polyclonal: Consider using monoclonal antibodies for specific epitope recognition and polyclonal for broader detection capacity
The application-specific requirements should guide selection rather than general characteristics or cost considerations.
For optimal ESR1 detection via Western Blot, the following methodological considerations are crucial:
Sample preparation: Nuclear extraction protocols yield better results than whole cell lysates due to ESR1's predominant nuclear localization
Protein loading: 20-40 μg of protein per lane is typically sufficient for detection
Gel percentage: 8-10% SDS-PAGE gels provide optimal separation for the 66.2 kDa ESR1 protein
Transfer conditions: Semi-dry transfer at 15V for 45 minutes using PVDF membranes shows superior results compared to nitrocellulose
Blocking: 5% BSA in TBST is preferred over milk-based blocking buffers
Antibody dilution: Primary antibody concentrations between 1:500-1:2000 typically yield optimal signal-to-noise ratios
Visualization: Both chemiluminescence and fluorescence-based detection systems are effective, with the latter offering better quantification capacity
Careful optimization of these parameters significantly improves detection sensitivity and specificity, particularly for samples with varying ESR1 expression levels.
Detection of ESR1 mutations requires a methodological approach tailored to the specific research question:
Sample source selection:
Solid tissue biopsy: Provides comprehensive genomic analysis but may miss heterogeneity
Circulating tumor cells (CTCs): Enables live cell functional studies
Cell-free DNA (cfDNA): Least invasive and captures heterogeneity across multiple metastatic sites
Detection methods:
Next-generation sequencing (NGS): Comprehensive but less sensitive
Droplet digital PCR (ddPCR): Highest sensitivity (detecting mutations at 0.1% frequency) but targets only known mutations
Mutation hotspots: Focus detection on the ligand-binding domain, particularly positions D538G and Y537S (most common), Y537N, Y537C, L536H, L536P, L536R, S463P, and E380Q
Clonal analysis: Consider that ESR1 mutations are polyclonal in 20-70% of patients, requiring methods that can detect multiple concurrent mutations
For research requiring high sensitivity, ddPCR is recommended as the gold standard methodology, particularly when monitoring disease progression or therapeutic response.
Effective immunohistochemical detection of ESR1 requires specific methodological considerations:
Fixation: 10% neutral buffered formalin for 24-48 hours provides optimal antigen preservation
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) at 95-98°C for 20 minutes is most effective for ESR1
Blocking: Use of dual blocking with hydrogen peroxide followed by protein block reduces background
Primary antibody: Overnight incubation at 4°C with validated anti-ESR1 antibody at 1:100-1:200 dilution
Detection system: Polymer-based detection systems offer superior sensitivity compared to avidin-biotin methods
Counterstaining: Brief hematoxylin counterstaining (30-60 seconds) preserves nuclear ESR1 signal visibility
Controls: Include both positive controls (breast or endometrial tissue) and negative controls (antibody diluent only)
For research applications examining ESR1 mutations, parallel IHC and mutation detection provides valuable correlation between protein expression and mutational status.
ESR1 mutations induce specific conformational changes that affect both function and antibody recognition:
Structural alterations: Mutations in the ligand-binding domain (LBD), particularly Y537S and D538G, stabilize the active conformation of ESR1 even in the absence of estrogen
Constitutive activity: These structural changes lead to increased coactivator recruitment and decreased proteolytic degradation
Antibody binding implications: Antibodies targeting conformational epitopes in the LBD may show reduced binding affinity for mutated ESR1
Epitope availability: Mutations alter protein-protein interactions that may mask or expose different epitopes, affecting antibody accessibility
Detection considerations: Antibodies targeting N-terminal regions typically maintain consistent binding regardless of LBD mutations
For researchers studying ESR1 mutations, using antibodies targeting epitopes outside the commonly mutated regions ensures consistent detection across both wild-type and mutant proteins.
ESR1 mutations confer resistance through multiple molecular mechanisms that should be considered in research designs:
Constitutive activity: LBD mutations enable ligand-independent activation, bypassing the need for estrogen binding and conferring resistance to aromatase inhibitors
Altered drug binding: Structural changes reduce the binding affinity of selective estrogen receptor modulators (SERMs) like tamoxifen
Degradation resistance: Mutant ESR1 shows decreased susceptibility to selective estrogen receptor degraders (SERDs) such as fulvestrant
Coactivator recruitment: Enhanced binding to coactivators strengthens downstream signaling even under inhibition conditions
Target gene alterations: ESR1 mutations modify the transcriptional program, activating genes not typically regulated by wild-type ESR1
These mechanisms explain the clinical observation that approximately 50% of endocrine resistance cases are associated with ESR1 mutations, highlighting the importance of incorporating mutation status in experimental designs.
ESR1 mutations induce significant molecular subtype alterations that can be studied using appropriate antibodies:
Basal phenotype acquisition: ESR1 mutant cells show significant enrichment of basal subtype markers, particularly six basal cytokeratins (BCKs) that are among the most enriched genes
Cytokeratin expression changes: Elevated expression of KRT5, KRT6A, KRT6B, KRT14, KRT16, and KRT17 has been consistently observed in ESR1 mutant cells
Epigenetic reprogramming: The induction of basal cytokeratins occurs through chromatin reorganization rather than direct ER binding
Progesterone receptor involvement: This process appears to be orchestrated by progesterone receptor-mediated insulated neighborhood formation
Immune pathway activation: ESR1 mutant tumors show enrichment of immune pathways similar to basal subtype tumors
These findings challenge the traditional view of ESR1 as a stable luminal lineage marker and suggest that mutations can drive partial subtype switching, which has implications for therapeutic response and experimental design.
Differentiating between wild-type and mutant ESR1 requires specialized approaches:
Mutation-specific antibodies: While limited, some research groups have developed antibodies specifically recognizing common mutations like Y537S
Functional assays: Reporter assays measuring ER activity in the presence/absence of estrogen can indirectly identify constitutively active mutants
Proximity ligation assays: These can detect altered protein-protein interactions characteristic of mutant ESR1
Pharmacological response profiling: Differential responses to SERMs and SERDs can distinguish wild-type from mutant ESR1
Molecular techniques: For definitive identification, complement antibody approaches with:
In experimental systems, creating isogenic cell lines with CRISPR-Cas9 genome editing to introduce specific ESR1 mutations provides valuable tools for comparative studies while controlling for genetic background.
Investigating ESR1 mutant protein interactions requires specialized methodological approaches:
Proximity-based methods:
BioID or TurboID for identifying proteins in proximity to wild-type versus mutant ESR1
FRET/BRET assays for real-time interaction monitoring
Proximity ligation assays for visualizing protein interactions in fixed cells/tissues
IP-MS workflows:
RIME (Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins) for chromatin-associated complexes
Quantitative AP-MS comparing interactome differences between wild-type and mutant ESR1
Crosslinking MS to capture transient interactions
Chromatin interaction studies:
ChIP-seq comparing binding sites between wild-type and mutant ESR1
CUT&RUN for higher resolution mapping
HiChIP for three-dimensional chromatin interaction landscapes
Systems biology approaches:
These multifaceted approaches provide complementary data that can elucidate how specific mutations alter ESR1 function at molecular and cellular levels.
Detection of ESR1 mutations in heterogeneous clinical samples requires specialized approaches:
Sampling considerations:
Serial sampling is recommended due to the acquired nature of ESR1 mutations
Multi-site sampling improves detection of heterogeneous mutations
Liquid biopsies (cfDNA) capture broader tumor heterogeneity than single-site biopsies
Technical sensitivity requirements:
Detection methods should achieve at least 0.1% variant allele frequency sensitivity
Digital PCR methods (ddPCR) generally outperform NGS for low-frequency detection
Panel-based testing should include all common hotspot mutations (Y537S, D538G, Y537N, Y537C, L536H/P/R, etc.)
Bioinformatic analysis:
Use algorithms optimized for low variant allele frequency detection
Apply error-correction methods for sequencing data
Consider clonal relationship analyses to track evolutionary patterns
Validation approaches:
These methodological considerations are particularly important as ESR1 mutations often occur as subclonal events with frequencies as low as 1-5% in early resistance stages.
Variability in ESR1 detection across sequential samples can stem from multiple factors that require specific methodological considerations:
Biological factors:
Treatment-induced changes in ESR1 expression
Clonal evolution leading to ESR1 mutation emergence or loss
Tumor heterogeneity with variable ESR1 expression across sites
Technical factors:
Inconsistent sample processing affecting epitope preservation
Antibody lot-to-lot variability impacting detection sensitivity
Storage conditions altering protein integrity over time
Methodological solutions:
Standardize preanalytical variables (fixation time, processing protocols)
Use calibration controls in each experimental batch
Implement dual-antibody approaches targeting different ESR1 epitopes
Complement protein detection with mRNA analysis
Consider digital pathology quantification for objective assessment
Importantly, some ESR1 mutant cell populations can be lost during continual passage, as demonstrated by the variability in basal cytokeratin detection between early (P6-8) and late passage (P30-32) cell lines . This suggests selection pressures may eliminate certain ESR1 mutant subclones, requiring careful monitoring of model systems across passages.
Multiple factors influence ESR1 mutation detection sensitivity in liquid biopsies:
Preanalytical variables:
Blood collection tube type (streck tubes preferred over EDTA)
Time between collection and processing (ideally <2 hours)
Centrifugation protocol affecting cfDNA yield and quality
Storage temperature during transport (-80°C optimal for long-term)
Technical considerations:
Extraction methodology (silica-based methods typically yield higher quality)
DNA input amount (minimum 10 ng for reliable detection)
Amplicon size (shorter amplicons improve detection in fragmented cfDNA)
UMI incorporation for error correction
Disease-specific factors:
Optimizing these variables is critical for reliable detection, particularly when monitoring therapeutic response or minimal residual disease where mutation frequencies may be extremely low.
Studying functional consequences of ESR1 mutations requires carefully designed experimental approaches:
Model system selection:
Isogenic cell line pairs (CRISPR-engineered mutations in the same genetic background)
Patient-derived models (PDX, organoids, CTCs from ESR1-mutant patients)
Transgenic models with inducible ESR1 mutations
Experimental design considerations:
Include multiple ESR1 mutation variants (Y537S, D538G) in parallel
Test both estrogen-deprived and estrogen-rich conditions
Compare responses across multiple endocrine therapies (AIs, SERMs, SERDs)
Evaluate combinations with targeted agents (CDK4/6i, PI3Ki, mTORi)
Readout selection:
These approaches should be integrated to provide a comprehensive understanding of how specific ESR1 mutations alter signaling, therapeutic response, and cell phenotype.
Recent research has revealed unexpected immunological consequences of ESR1 mutations:
Immune pathway activation:
ESR1 mutant tumors show enrichment of immune signaling pathways
S100A8 and S100A9 are among the most induced immune mediators in ESR1 mutant cells
These changes create potential immune therapeutic vulnerabilities
Tumor microenvironment alterations:
Single-cell RNA-seq data indicates ESR1 mutations influence tumor-stroma crosstalk
Increased basal cytokeratin expression correlates with altered immune context
These changes may enhance immunogenicity of otherwise "cold" luminal tumors
Research implications:
These findings suggest that ESR1 mutations may create opportunities for immunotherapeutic approaches not typically considered for hormone receptor-positive breast cancers, representing an important new research direction.
Monitoring ESR1 mutation emergence requires longitudinal sampling approaches:
Recommended sampling strategy:
Baseline assessment before therapy initiation
Regular monitoring at 2-3 month intervals during treatment
Additional sampling at progression or treatment change points
Parallel tissue and liquid biopsies when feasible
Technological approaches:
Digital droplet PCR for known hotspot mutations (highest sensitivity)
Targeted NGS panels for broader mutation spectrum detection
Ultra-deep sequencing with molecular barcoding for subclonal detection
Comparison of ctDNA and CTC analyses for comprehensive assessment
Analytical considerations:
This systematic approach enables researchers to track the dynamics of ESR1 mutation emergence, providing insights into resistance mechanisms and potentially guiding therapeutic decision-making in clinical research settings.
ERα consists of several functional domains:
Phosphorylation is a key regulatory mechanism for ERα activity. Several serine residues, including Ser104, Ser106, Ser118, and Ser167, are phosphorylated by different kinases, which modulate the receptor’s transcriptional activity . For instance, Ser118 is phosphorylated by CDK7, and Ser167 is phosphorylated by p90RSK and Akt . Phosphorylation at Ser167 has been associated with tamoxifen resistance in breast cancer patients .
Mouse anti-human ERα antibodies are monoclonal antibodies produced by immunizing mice with a synthetic peptide corresponding to residues surrounding Ser118 of human ERα . These antibodies are highly specific and do not cross-react with estrogen receptor beta or other family members . They are commonly used in various scientific applications, including Western blotting, immunohistochemistry, and immunocytochemistry .
Mouse anti-human ERα antibodies are valuable tools in research and clinical diagnostics. They are used to: