AXL antibodies are immunotherapeutics designed to inhibit the AXL receptor, a member of the TAM (TYRO3/AXL/MERTK) family implicated in cancer progression, immune evasion, and viral entry . AXL is overexpressed in aggressive malignancies and contributes to:
Tumor cell proliferation, metastasis, and epithelial-mesenchymal transition (EMT)
Resistance to chemotherapy, radiation, and immune checkpoint inhibitors
Immunosuppression via dendritic cell inhibition and M2 macrophage polarization
The following table summarizes leading AXL-targeting antibodies under investigation:
AXL antibodies exert therapeutic effects through:
Ligand blockade: CDX-0168 inhibits Gas6-dependent AXL phosphorylation (IC₅₀ <1 nM)
Receptor internalization: D9/E8 antibodies downregulate AXL expression by 70% within 24 hrs
Downstream pathway inhibition:
M2 macrophage repolarization: AXL02-MMAE decreases CSF-1 production by 90%, enhancing CD8+ T-cell infiltration
Dendritic cell activation: CDX-0168 increases IL-12 secretion by 300% in primary human DCs
PD-L1 synergy: Bispecific AXL/PD-L1 antibodies show 2.5x greater T-cell activation vs. monotherapies
ADCT-601 Phase 1b (NCT05389462): Evaluating safety in sarcoma patients, with preliminary data showing:
While AXL antibodies show promise, key considerations include:
Biomarker development: AXL overexpression (IHC 3+ in 76% of PDACs ) correlates with poor survival but requires standardized detection assays
Resistance mechanisms: Compensatory upregulation of c-MET and EGFR observed in 30% of treated tumors
Delivery optimization: ADC therapies like ADCT-601 face pharmacokinetic challenges due to AXL's broad normal tissue expression
Emerging strategies include bispecific antibodies targeting AXL/PD-L1 and CAR-T cells engineered with AXL-targeting scFvs .
AXL receptor tyrosine kinase belongs to the TAM (Tyro3/Axl/MerTK) family of receptor tyrosine kinases. It functions as a negative regulator of innate immunity and plays critical roles in multiple cellular processes. In cancer biology, AXL has significant relevance as its activation through its ligand Gas6 drives tumor growth, metastasis, and is associated with acquired resistance to targeted therapies, radiotherapy, and chemotherapy . AXL protein expression has been demonstrated in 55-70% of pancreatic cancers and is associated with aggressive tumor behavior, higher frequency of distant metastases, and poor survival . Structurally, the protein has a molecular weight of approximately 98.3 kilodaltons, though in Western blotting applications it often appears as a band of approximately 138 kDa in cell lysates due to post-translational modifications .
Researchers should employ specific antibodies that recognize either the phosphorylated form (p-AXL) or total AXL protein. When conducting studies examining AXL activation, it's crucial to analyze both forms simultaneously:
For phosphorylated AXL detection:
Use phospho-specific antibodies that recognize specific phosphorylation sites
Include positive controls such as Gas6-stimulated cells to validate phosphorylation state
Consider phosphatase inhibitors in lysate preparation
For total AXL detection:
When analyzing both forms, use quantitative analysis to determine the ratio of phosphorylated to total AXL as a measure of activation. Studies have demonstrated correlation between p-AXL and total AXL expression levels in various cancer specimens, particularly in triple-negative breast cancer where they positively correlate with KLF5 expression .
Optimal sample preparation for AXL detection varies by experimental approach:
For Western blotting:
Use 25-40 μg of total cell lysate for optimal AXL and phospho-AXL detection
Include protease and phosphatase inhibitors in lysis buffers
Employ denaturating conditions with proper reducing agents
For membrane proteins like AXL, consider detergent selection carefully
For immunohistochemistry:
Appropriate fixation is critical; formalin-fixed paraffin-embedded tissues have been successfully used to detect AXL expression patterns in tumor specimens
Antigen retrieval steps are essential for exposing the epitope
Signal amplification may be necessary for detecting lower expression levels
Optimal antibody dilution should be determined empirically for each application
In clinical specimens, validation with multiple antibodies may be necessary to confirm specificity of staining patterns, as demonstrated in studies of pancreatic adenocarcinoma where AXL was detected in 76% of cases with stronger staining observed in invasive cells .
Anti-AXL antibodies have been validated for multiple experimental applications, each requiring specific optimization:
Western Blotting:
Immunohistochemistry:
Immunofluorescence:
Flow Cytometry:
Quantifies AXL expression levels on cell surfaces
Useful for sorting AXL-positive cellular populations
Immunoprecipitation:
The choice of application should be guided by the specific research question, with appropriate validation controls included in experimental design.
Validation of anti-AXL antibody specificity is critical for reliable experimental results and should include:
Molecular weight verification:
Positive and negative controls:
Peptide competition assays:
Pre-incubate antibody with purified AXL protein or peptide
Observe elimination of specific signal
Cross-validation with multiple antibodies:
Use different antibodies targeting distinct epitopes of AXL
Compare staining/detection patterns for consistency
Functional validation:
Proper validation increases confidence in experimental results and prevents misinterpretation of data due to non-specific antibody binding.
When investigating AXL-mediated signaling pathways, essential controls include:
Stimulation controls:
Compare unstimulated vs. Gas6-stimulated samples to confirm pathway activation
Include time-course analysis to capture optimal activation windows
Inhibition controls:
Knockdown/knockout validation:
Downstream marker analysis:
Cross-pathway controls:
Include markers for related pathways to assess specificity and cross-talk
Consider potential redundancy with other TAM family receptors (Tyro3, MerTK)
Using these controls ensures that observed effects are specifically related to AXL signaling and not due to off-target effects or experimental artifacts.
Anti-AXL antibodies have demonstrated significant effects on epithelial-mesenchymal transition (EMT), a process strongly linked to AXL activity:
Mechanism of action:
AXL has been demonstrated to be required for EMT of malignant cells induced by various stimuli, including H-RASV12 and overexpression of SLUG
Anti-AXL antibodies can interfere with this process by:
a) Blocking Gas6-mediated AXL activation
b) Inducing AXL internalization and downregulation
c) Inhibiting downstream signaling pathways essential for EMT maintenance
Experimental evidence:
Studies have shown that anti-AXL mAbs (D9 and E8) induce down-expression of AXL by internalization within 1.5 hours of exposure
This internalization leads to inhibition of AXL phosphorylation by Gas6, disrupting EMT-supporting signaling
In pancreatic cancer cell lines, anti-AXL antibodies reduced cell migration, a key EMT-associated phenotype
Impact on EMT markers:
Anti-AXL antibody treatment can restore epithelial marker expression (E-cadherin)
Treatment reduces mesenchymal markers (N-cadherin, vimentin)
Affects EMT-inducing transcription factors (Snail, Slug, ZEB1)
Relevance to cancer progression:
Understanding these mechanisms allows researchers to effectively utilize anti-AXL antibodies to study and potentially target EMT in cancer progression.
Anti-AXL antibodies serve as valuable tools for investigating therapy resistance mechanisms across multiple cancer types:
AXL overexpression in resistant populations:
AXL overexpression is associated with resistance to standard chemotherapy and tyrosine kinase inhibitors in multiple cancers
Documented resistance mechanisms include:
a) Acute and chronic myeloid leukemia resistance to standard therapy
b) Gastrointestinal stromal tumor resistance to TKIs
c) Breast, ovarian, and lung cancer therapy resistance
Methodological approaches:
Use anti-AXL antibodies to screen resistant vs. sensitive cell populations
Compare AXL expression levels before and after exposure to therapeutic agents
Monitor AXL phosphorylation status during resistance development
Example: ERL-resistant lung cancer cell lines showed increased AXL expression and phosphorylation compared to parental cell lines
Intervention studies:
Anti-AXL antibodies can be used to determine if AXL inhibition resensitizes resistant cells
Combined blockade approaches (e.g., AXL with PD-L1) show promise for overcoming resistance
A humanized IgG1 Axl-targeting monoclonal antibody (CDX-0168) inhibits Gas6-dependent Axl phosphorylation and signaling, potentially addressing resistance mechanisms
Downstream signaling analysis:
Anti-AXL antibodies enable tracking of altered signaling in resistant cells
Common pathways involved include PI3K/AKT and MAPK/ERK cascades
By systematically applying these approaches, researchers can elucidate the specific mechanisms by which AXL contributes to therapy resistance and develop strategies to overcome them.
Anti-AXL antibodies demonstrate significant immunomodulatory effects in the tumor microenvironment through multiple mechanisms:
AXL's role in immune suppression:
Antibody-mediated immune activation:
Humanized IgG1 anti-AXL antibodies (e.g., CDX-0168) induce potent release of pro-inflammatory cytokines and chemokines from:
a) Dendritic cells
b) Monocytes
c) Macrophages
This effect occurs through an Fc receptor-dependent mechanism
Treatment enhances T cell activation in mixed lymphocyte reactions
Combination strategies:
Antibody-dependent cellular cytotoxicity (ADCC):
These immunomodulatory effects represent an important dimension of anti-AXL antibody function, supporting their potential application in cancer immunotherapy approaches.
Successful Western blotting with anti-AXL antibodies requires attention to several critical parameters:
Sample preparation:
Gel electrophoresis considerations:
Transfer optimization:
Use wet transfer methods for large proteins like AXL
Extend transfer times or reduce current for better transfer efficiency
Consider transfer buffers optimized for high molecular weight proteins
Antibody selection and dilution:
Common issues and solutions:
Weak signals: Increase protein loading, optimize antibody concentration, extend exposure times
Multiple bands: Validate specificity using knockdown controls, test different antibody clones
Background issues: Increase blocking time, optimize washing steps, reduce antibody concentration
Following these guidelines will help ensure reproducible and reliable detection of AXL in Western blotting applications.
Optimizing immunohistochemical detection of AXL in tissue specimens requires attention to several key parameters:
Tissue preparation and fixation:
Antigen retrieval methods:
Heat-induced epitope retrieval (HIER) is typically necessary for AXL detection
Compare citrate-based (pH 6.0) versus EDTA-based (pH 9.0) retrieval buffers
Optimize retrieval times and temperatures for specific tissue types
Antibody selection and validation:
Test multiple anti-AXL antibodies targeting different epitopes
Validate specificity using positive and negative control tissues
Determine optimal antibody dilutions through titration experiments
Signal development considerations:
Compare DAB versus other chromogens for optimal visualization
Consider signal amplification methods for low-expression samples
Implement standardized development times for consistency across specimens
Scoring and evaluation methods:
Define clear criteria for positive staining (membrane, cytoplasmic, or both)
Consider intensity scoring scales (e.g., mild positive (+), moderate positive (++), strong positive (+++)) as used in previous studies
Evaluate specific patterns such as stronger staining in invasive cells at tumor periphery
Studies have successfully used these approaches to demonstrate AXL expression in 76% of pancreatic ductal adenocarcinoma cases, with 66% of AXL-positive tumors showing stronger staining in invasive cells .
Antibody internalization experiments are crucial for understanding the mechanisms of anti-AXL antibody action. Effective troubleshooting requires:
Temperature control protocols:
Compare internalization at 37°C (active internalization) versus 4°C (surface localization only)
Previous studies have shown that anti-AXL mAbs (D9 and E8) are internalized in cancer cells following incubation at 37°C for 1 hour but remain localized at the cell surface after incubation at 4°C
Maintain strict temperature control throughout experiments
Time-course optimization:
Visualization techniques:
Immunofluorescence microscopy can effectively track antibody localization
Consider:
a) Direct fluorophore conjugation to anti-AXL antibodies
b) Secondary antibody detection systems
c) Co-localization with endosomal/lysosomal markers
Quantification approaches:
Flow cytometry to measure surface versus internalized antibody levels
Image analysis software for quantifying intracellular fluorescence
Western blotting to assess total AXL protein levels after internalization
Common challenges and solutions:
Poor internalization: Confirm antibody binding to correct epitope, verify cell viability
High background: Optimize washing procedures, adjust antibody concentrations
Cell type variation: Different cell lines may show varying internalization kinetics
Proper implementation of these approaches allows researchers to accurately characterize the internalization dynamics of anti-AXL antibodies, which is critical for understanding their mechanisms of action in downregulating AXL signaling.
Bispecific antibodies targeting AXL represent an innovative frontier in cancer immunotherapy research:
Mechanism and design considerations:
Tetravalent bispecific AXL x PD-L1 antibodies combine anti-AXL with anti-PD-L1 mAbs in an IgG-scFv format
This approach simultaneously targets:
a) AXL-mediated tumor growth and metastasis
b) PD-L1-mediated immune checkpoint blockade
CDX-0168 (anti-AXL) combined with 9H9 (anti-PD-L1) has demonstrated promising results in preclinical studies
Enhanced immune activation:
Advantages over combination therapy:
Simplified administration of a single agent
Potential synergistic effects from co-targeting within the same molecule
Consistent ratio of targeting moieties
Current research status:
The continued development of bispecific antibodies targeting AXL in combination with immune checkpoint inhibitors could potentially address multiple cancer hallmarks simultaneously, improving therapeutic outcomes.
Assessing the impact of anti-AXL antibodies on cancer stem cell (CSC) populations requires specialized methodological approaches:
In vitro analysis techniques:
Sphere formation assays to measure self-renewal capacity
Flow cytometry for CSC marker expression (CD44, CD133, ALDH activity)
Serial limiting dilution assays to quantify stem cell frequency
Gene expression analysis of stemness-associated pathways
In vivo tumorigenicity assays:
Limited dilution transplantation studies after antibody treatment
Research has demonstrated that AXL inhibition (using DCC-2036) affected CSC frequency in 4T1 cells as determined by in vivo tumorigenicity assays
Transplantation of varying cell numbers (e.g., 200,000, 20,000, or 2,000 cells) to calculate CSC frequency using ELDA (Extreme Limiting Dilution Analysis)
Molecular mechanism investigation:
Resistance and recurrence models:
Development of resistant cell populations after serial antibody treatment
Analysis of AXL expression in tumor recurrence models
Comparison of primary and metastatic lesions for AXL and CSC marker expression
These methodological approaches provide comprehensive assessment of how anti-AXL antibodies affect cancer stem cell populations, potentially addressing a key mechanism of therapy resistance and tumor recurrence.
Effective combination of anti-AXL antibodies with other targeted therapies requires systematic experimental design:
Rational selection of combination partners:
AXL upregulation occurs in resistance to various therapies including:
a) Tyrosine kinase inhibitors in myeloid leukemia and gastrointestinal stromal tumors
b) Conventional chemotherapy in breast, ovarian, and lung cancers
Select complementary targets based on:
a) Pathway analysis (e.g., combining AXL inhibition with downstream effector blockade)
b) Resistance mechanisms (e.g., combining with the primary therapy causing AXL-mediated resistance)
c) Immune modulatory potential (e.g., combining with immune checkpoint inhibitors)
Synergy assessment methodologies:
Sequence-dependent effects:
Compare different treatment sequences:
a) Concurrent administration
b) Sequential administration (anti-AXL antibody first followed by partner therapy)
c) Alternating schedules
Document temporal dynamics of pathway inhibition
Multidimensional endpoint analysis:
Assess combination effects on:
a) Cell proliferation and survival
b) Migration and invasion capabilities
c) Cancer stem cell properties
d) Immune system activation
e) In vivo tumor growth and metastasis formation
Translation to in vivo models:
Use both subcutaneous and orthotopic xenograft models
Consider syngeneic models for immune-related combinations
Implement patient-derived xenografts for clinical relevance
The systematic implementation of these approaches enables researchers to identify and characterize the most promising combination strategies involving anti-AXL antibodies, potentially leading to more effective therapeutic interventions.