HLX antibodies are a series of fully humanized or recombinant mAbs developed for treating advanced solid tumors and hematologic malignancies. Notable examples include:
HLX07: Anti-epidermal growth factor receptor (EGFR) antibody .
HLX22: Anti-human epidermal growth factor receptor 2 (HER2) antibody .
These antibodies are distinguished by their unique structural epitopes and mechanisms of action compared to existing therapies.
Targets EGFR, a receptor overexpressed in epithelial cancers.
Inhibits EGFR-mediated signaling, reducing tumor cell proliferation and survival .
Binds PD-1 on T-cells, preventing interaction with PD-L1/PD-L2 on tumor cells.
Restores T-cell activation and enhances anti-tumor immunity .
Structural analysis reveals a unique epitope overlapping with pembrolizumab but with distinct heavy/light chain usage .
Blocks HER2 signaling, critical in HER2-overexpressing cancers (e.g., breast, gastric).
Demonstrates synergistic effects with trastuzumab in preclinical models .
| Parameter | Results (N=19) |
|---|---|
| Dose Range | 50–800 mg/week |
| Most Common TEAEs | Fatigue (68.4%), nausea (47.4%) |
| Serious TEAEs | 11 patients (1 related to treatment) |
| Response | Stable disease in 36.4% of patients |
| PK Profile | Dose-proportional exposure |
HLX07 showed no dose-limiting toxicities and a manageable safety profile up to 800 mg/week .
| Parameter | Results (N=11) |
|---|---|
| Dose Levels | 3, 10, 25 mg/kg every 3 weeks |
| Most Common TEAEs | Lymphopenia (45.5%), hypokalemia (36.4%) |
| MTD | 25 mg/kg |
| Efficacy | Stable disease in 36.4% of patients |
No serious adverse events or deaths were reported, supporting further combination studies .
| Feature | HLX10 | Pembrolizumab | Nivolumab |
|---|---|---|---|
| Epitope Region | BC-loop | BC-loop | C’D-loop |
| Heavy Chain Usage | Unique | Conventional | Conventional |
| Clinical Efficacy | Pending Phase III | Approved | Approved |
HLX10’s structural distinction may offer differentiated pharmacokinetic or safety profiles .
Combination Therapies: HLX antibodies are being tested with chemotherapy, targeted therapies, and other immunotherapies.
Biomarker Development: Identifying predictive markers (e.g., HER2/EGFR expression levels) for patient stratification.
Mechanistic Studies: Further exploration of HLX10’s unique epitope impact on T-cell activation .
HLX (H2.0-like homeobox) is a transcription factor belonging to the homeobox family that plays important roles in developmental processes and immune cell function. Researchers study HLX because of its critical functions in hematopoiesis and immune regulation. The rabbit polyclonal anti-HLX antibody is designed specifically for research applications targeting the human HLX protein . When designing experiments, researchers must consider:
The specific epitope recognized by the antibody
The protein's expression patterns in different tissues and cell types
Potential cross-reactivity with related homeobox proteins
The cellular localization (typically nuclear) of the transcription factor
To properly interpret results, researchers should understand the biological context of HLX expression and function in their experimental system.
Proper validation of any antibody, including HLX antibody, is critical for obtaining reliable and reproducible results. Following the "five pillars" of antibody characterization is recommended :
Genetic strategy validation: Use HLX knockout or knockdown cells/tissues as negative controls
Orthogonal validation: Compare antibody-based detection with antibody-independent methods (e.g., mRNA quantification)
Independent antibody validation: Use multiple antibodies targeting different epitopes of HLX
Expression validation: Test the antibody in systems with induced overexpression of HLX
Immunocapture MS validation: Use mass spectrometry to identify proteins captured by the HLX antibody
At minimum, researchers should demonstrate that the antibody:
Binds to the target HLX protein
Recognizes HLX in complex protein mixtures
Does not exhibit significant cross-reactivity with non-target proteins
Performs consistently under the specific experimental conditions being used
Proper controls are essential for ensuring the validity of results obtained with HLX antibody :
| Control Type | Examples | Purpose |
|---|---|---|
| Positive controls | - Cell lines known to express HLX - Recombinant HLX protein - HLX-transfected cells | Verify antibody can detect the target |
| Negative controls | - Cell lines not expressing HLX - HLX knockout samples - Isotype control antibodies - Primary antibody omission | Assess non-specific binding |
| Procedural controls | - Loading controls (Western blot) - Blocking peptide competition - Secondary antibody-only controls | Validate technique and quantification |
The inclusion of appropriate controls in every experiment is critical for establishing the specificity of the antibody and the reliability of the experimental results .
HLX antibody is validated for several common research applications :
Immunohistochemistry (IHC): For detecting HLX protein in tissue sections, providing information on protein localization and expression patterns.
Immunocytochemistry-Immunofluorescence (ICC-IF): For visualizing HLX in cultured cells, useful for subcellular localization studies.
Western Blotting (WB): For detecting and semi-quantifying HLX protein in cell or tissue lysates, providing information about protein size and relative abundance.
Each application requires specific optimization steps:
For IHC: Optimize fixation, antigen retrieval, antibody concentration, and detection systems
For ICC-IF: Determine optimal fixation/permeabilization conditions and antibody dilutions
For WB: Optimize sample preparation, blocking conditions, antibody concentration, and exposure times
Application-specific validation is necessary as an antibody may perform well in one application but poorly in others .
For multiple bands in Western blotting:
Verify if bands represent isoforms, post-translational modifications, or degradation products
Consult literature for known HLX variants and their molecular weights
Perform peptide competition assays to determine which bands are specific
Test in HLX knockout/knockdown samples to identify specific signals
For unexpected cellular localization:
Consider biological context (HLX may shuttle between nucleus and cytoplasm)
Verify fixation conditions (which can affect epitope accessibility)
Confirm with orthogonal methods (fractionation followed by Western blotting)
Use co-localization studies with organelle markers
For unusual tissue staining patterns:
Compare with known expression patterns from transcriptomic data
Evaluate multiple tissue samples and biological replicates
Use alternative antibodies against different HLX epitopes
Consider the possibility of non-specific binding
Careful documentation of all unexpected results contributes to better understanding of antibody performance and potentially to new biological insights .
Inconsistent Western blotting results with HLX antibody can stem from multiple factors. A methodological troubleshooting approach includes :
Sample preparation issues:
Ensure complete protein denaturation (optimize heating temperature/duration)
Verify protein integrity (check for degradation patterns)
Use fresh protease inhibitors during lysis
Test different lysis buffers to optimize HLX extraction
Antibody-specific considerations:
Titrate antibody concentration (typically 0.5-5 μg/ml for Western blotting)
Extend primary antibody incubation time (overnight at 4°C may improve signal)
Test different blocking reagents (BSA vs. milk can affect HLX detection)
Consider epitope accessibility (N-terminal vs. C-terminal antibodies may give different results)
Detection system optimization:
Compare chemiluminescent vs. fluorescent detection systems
Adjust exposure times to prevent saturation
Use enhanced sensitivity substrates for low-abundance HLX detection
Validation approach:
Compare results with orthogonal methods (qPCR for HLX mRNA)
Test multiple anti-HLX antibodies targeting different epitopes
Include positive and negative control samples in every experiment
Systematic documentation of all variables across experiments can help identify sources of inconsistency .
Quantifying HLX protein expression requires careful selection of methods and rigorous standardization :
Western blotting with densitometry:
Use a standard curve with recombinant HLX protein
Normalize to multiple housekeeping proteins (not just one)
Ensure linear dynamic range of detection
Use technical replicates and biological replicates
Apply statistical analysis to densitometry data
Quantitative immunofluorescence:
Establish standardized image acquisition parameters
Include calibration standards in each experiment
Use automated image analysis software to reduce bias
Correct for background and autofluorescence
Calculate relative fluorescence units or integrated density values
Flow cytometry:
Use antibody titration to determine optimal concentration
Include fluorescence minus one (FMO) controls
Establish gates based on negative populations
Use median fluorescence intensity (MFI) for quantification
Consider using quantitative flow cytometry with calibration beads
Each method has strengths and limitations, and ideally, researchers should validate findings using multiple independent approaches to ensure robust quantification .
Assessing cross-reactivity of HLX antibody with other homeobox proteins is essential for ensuring specificity :
Sequence analysis:
Perform in silico analysis of epitope sequences
Identify homologous regions between HLX and other homeobox proteins
Calculate sequence similarity percentages
Predict potential cross-reactive proteins based on epitope conservation
Experimental validation:
Test the antibody on cells expressing other homeobox proteins but not HLX
Use knockout/knockdown models for HLX and test for residual signal
Perform peptide competition assays with HLX peptides and peptides from related proteins
Express recombinant homeobox proteins and test antibody binding
Advanced analytical approaches:
Use immunoprecipitation followed by mass spectrometry to identify all bound proteins
Perform epitope mapping to precisely define the antibody's binding site
Use surface plasmon resonance (SPR) to measure binding affinities to HLX versus related proteins
Conduct immunodepletion experiments to confirm signal specificity
| Homeobox Family | Sequence Similarity | Cross-Reactivity Risk | Validation Method |
|---|---|---|---|
| HLX subfamily | High (>80%) | High | Knockout controls essential |
| Other H2.0-like | Moderate (50-70%) | Moderate | Peptide competition |
| Distant homeobox | Low (<40%) | Low | Western blot verification |
Thorough cross-reactivity assessment enhances confidence in experimental results and supports accurate interpretation of HLX-specific signals .
Optimizing immunohistochemistry (IHC) protocols for HLX antibody requires attention to several critical parameters :
Tissue preparation and fixation:
Test different fixatives (formalin, paraformaldehyde, alcohol-based)
Optimize fixation time to preserve antigenicity while maintaining morphology
Evaluate embedding methods (paraffin vs. frozen sections)
Consider tissue-specific factors that might affect HLX epitope preservation
Antigen retrieval methods:
Compare heat-induced epitope retrieval (HIER) methods:
Citrate buffer (pH 6.0)
EDTA buffer (pH 8.0-9.0)
Tris-EDTA buffer (pH 9.0)
Test enzymatic retrieval (proteinase K, trypsin) as alternatives
Optimize retrieval times and temperatures
Blocking and antibody conditions:
Evaluate different blocking reagents (normal serum, BSA, commercial blockers)
Titrate primary antibody concentration (typically 1-10 μg/ml for IHC)
Test different incubation times and temperatures
Optimize washing steps to reduce background
Detection system selection:
Compare different detection methods:
DAB chromogenic detection
Fluorescent secondary antibodies
Signal amplification systems (TSA, ABC method)
Consider multiplexing capability if detecting HLX alongside other markers
Systematic optimization of these parameters will help ensure specific and reproducible HLX detection in tissue samples .
Multiplex immunofluorescence allows simultaneous detection of HLX with other proteins of interest :
Panel design considerations:
Select antibodies from different host species when possible
Consider primary antibody directly conjugated to fluorophores
Plan fluorophore selection to minimize spectral overlap
Include nuclear stain and cell type markers
Optimized staining protocols:
Sequential staining approach:
Incubate with first primary antibody
Add corresponding secondary antibody
Block remaining binding sites
Proceed with next primary-secondary pair
Repeat for additional markers
Simultaneous staining approach:
Mix compatible primary antibodies
Incubate simultaneously
Wash thoroughly
Add mixture of secondary antibodies
Controls for multiplex validation:
Single-color controls to assess bleed-through
Fluorescence minus one (FMO) controls
Isotype controls for each species
Absorption controls with competing peptides
Advanced multiplex technologies:
Tyramide signal amplification (TSA):
Allows use of antibodies from same species
Provides signal amplification for low-abundance targets
Requires sequential staining with heat-mediated antibody stripping
Spectral imaging and unmixing:
Captures full emission spectrum at each pixel
Computationally separates overlapping fluorophores
Enables use of more markers simultaneously
Multiplex approaches provide valuable context for understanding HLX expression in relation to other cellular factors and tissue microenvironment .
Chromatin immunoprecipitation (ChIP) with HLX antibody allows researchers to identify genomic binding sites of this transcription factor :
Cell preparation and crosslinking:
Harvest cells expressing HLX protein (1-10 × 10^6 cells per IP)
Crosslink DNA-protein complexes with 1% formaldehyde for 10 minutes at room temperature
Quench with 125 mM glycine for 5 minutes
Wash cells in ice-cold PBS with protease inhibitors
Chromatin preparation:
Lyse cells in appropriate buffers to isolate nuclei
Sonicate chromatin to generate 200-500 bp fragments
Verify sonication efficiency by agarose gel electrophoresis
Pre-clear chromatin with protein A/G beads
Immunoprecipitation with HLX antibody:
Use 2-5 μg of HLX antibody per IP reaction
Include IgG control antibody in parallel reactions
Include input control (non-immunoprecipitated chromatin)
Incubate overnight at 4°C with rotation
Add protein A/G beads and incubate for 1-3 hours
Wash stringently to remove non-specific binding
DNA purification and analysis:
Reverse crosslinks (65°C for 4-12 hours)
Treat with RNase A and Proteinase K
Purify DNA using column or phenol-chloroform extraction
Analyze by qPCR, sequencing, or other methods
Optimizing each step for the specific HLX antibody and cell type will improve ChIP efficiency and specificity .
Flow cytometry analysis of HLX expression requires specific methodological considerations :
Sample preparation for intracellular staining:
Fix cells with 2-4% paraformaldehyde (10-20 minutes)
Permeabilize with appropriate buffer:
0.1-0.5% saponin (reversible, gentle)
0.1-0.3% Triton X-100 (stronger)
Commercial permeabilization buffers
Block with 2-5% normal serum or BSA
Antibody titration and staining:
Perform titration to determine optimal antibody concentration
Include surface markers for cell identification before fixation
Use directly conjugated HLX antibody if available
If using indirect staining, select secondary antibody with appropriate fluorophore
Critical controls:
Unstained cells to assess autofluorescence
Fluorescence minus one (FMO) controls
Isotype controls matched to antibody class and concentration
Positive control (cell type known to express HLX)
Negative control (cells with HLX knockdown/knockout)
Analysis approaches:
Gating strategy:
Exclude debris and doublets
Identify viable cells
Gate on cell populations of interest
Analyze HLX expression within subpopulations
Quantification methods:
Percent positive cells (compared to negative control)
Median fluorescence intensity (MFI)
Integrated MFI (iMFI = % positive × MFI)
These best practices ensure accurate quantification of HLX expression across heterogeneous cell populations .
Quantifying HLX subcellular localization via immunofluorescence requires rigorous methodology :
Sample preparation optimization:
Test different fixation methods:
4% paraformaldehyde (10-20 minutes)
Methanol (-20°C, 10 minutes)
Acetone (-20°C, 5 minutes)
Optimize permeabilization:
0.1-0.5% Triton X-100
0.1-0.5% Saponin
0.01-0.05% SDS
Block with 1-5% BSA or normal serum
Antibody conditions:
Titrate HLX antibody (typically 1-10 μg/ml)
Include nuclear counterstain (DAPI, Hoechst)
Consider co-staining with organelle markers
Lamin B1 (nuclear envelope)
Fibrillarin (nucleolus)
ERGIC-53 (ER-Golgi)
Tom20 (mitochondria)
Quantitative analysis approaches:
Nuclear/cytoplasmic ratio calculation:
Define nuclear and cytoplasmic regions using masks
Calculate mean fluorescence intensity in each compartment
Determine N/C ratio for individual cells
Analyze population distributions
Colocalization analysis:
Calculate Pearson's or Mander's coefficients
Perform object-based colocalization
Test statistical significance of colocalization
Software tools for analysis:
ImageJ/FIJI with appropriate plugins
CellProfiler for high-throughput analysis
Commercial platforms (MetaMorph, Imaris, etc.)
Custom analysis pipelines using Python or R
These approaches provide quantitative data on HLX localization changes in response to experimental conditions or disease states .
Non-specific binding can significantly impact the interpretation of results when using HLX antibody :
Identifying non-specific binding:
Unexpected bands in Western blot that don't match predicted molecular weight
Staining in tissues known not to express HLX
Signal in HLX knockout or knockdown samples
Inconsistent staining patterns across experiments
Optimizing blocking conditions:
Test different blocking agents:
BSA (1-5%)
Non-fat dry milk (3-5%)
Normal serum (2-10%)
Commercial blocking reagents
Extend blocking time (1-2 hours at room temperature or overnight at 4°C)
Include blocking additives:
0.1-0.3% Tween-20
0.1-0.5% Triton X-100
0.1% gelatin
Antibody optimization:
Increase washing stringency:
More wash steps
Higher detergent concentration
Higher salt concentration
Reduce antibody concentration
Preabsorb antibody with knockout tissue lysate
Use affinity-purified antibody fractions
Application-specific approaches:
For Western blot:
Use PVDF membrane instead of nitrocellulose
Apply membrane blocking before antibody incubation
Reduce incubation temperature (4°C)
For IHC/ICC:
Optimize fixation to preserve epitopes
Test different antigen retrieval methods
Block endogenous peroxidase/biotin/avidin
Use directly conjugated primary antibodies
Systematic optimization of these parameters can significantly reduce non-specific binding and improve the signal-to-noise ratio .
The choice between polyclonal and monoclonal HLX antibodies has significant implications for experimental outcomes :
| Characteristic | Polyclonal HLX Antibodies | Monoclonal HLX Antibodies |
|---|---|---|
| Epitope recognition | Multiple epitopes on HLX protein | Single epitope on HLX protein |
| Sensitivity | Generally higher (multiple binding sites) | May be lower (single binding site) |
| Specificity | Variable, batch-dependent | Consistent, highly specific |
| Lot-to-lot variation | Significant | Minimal |
| Applications | More tolerant of protein denaturation | May be sensitive to epitope accessibility |
| Best for | Detection of low-abundance HLX | Precise epitope mapping, consistent results |
| Limitations | Higher risk of cross-reactivity | May miss isoforms or modified forms |
Selection considerations should include:
Experimental goals:
For detecting multiple HLX isoforms: polyclonal may be preferred
For specific domain recognition: monoclonal is often better
For reproducible quantification: monoclonal provides consistency
Validation status:
Review available validation data for each antibody
Check for knockout validation
Examine published literature using the specific antibody
Application compatibility:
Some applications may work better with polyclonal (e.g., IP)
Others may require monoclonal specificity (e.g., therapeutic applications)
Availability and cost:
Consider long-term experimental needs
Evaluate sustainability of supply for research program
The ultimate selection should be based on thorough validation in the specific experimental context .