CUSABIO employed a meticulous procedure to produce the DDR1 recombinant monoclonal antibody. Initially, B cells were isolated from the immunized animal's spleen, utilizing recombinant human DDR1 protein as the immunogen. Subsequently, RNA was extracted from the B cells and converted into cDNA through reverse transcription. Using the cDNA as a template, the gene encoding the DDR1 antibody was extended using a degenerate primer through PCR and inserted into a vector. The recombinant vector was then introduced into host cells through transfection, enabling efficient antibody expression. The DDR1 recombinant monoclonal antibodies were harvested from the cell culture supernatant and subjected to purification via affinity chromatography. Rigorous validation, including ELISA testing, was conducted to confirm this antibody's specific reactivity with human DDR1 protein, ensuring its reliability and suitability for a range of applications.
CUSABIO meticulously developed the DDR1 recombinant monoclonal antibody. Initially, B cells were isolated from the immunized animal's spleen using recombinant human DDR1 protein as the immunogen. Subsequently, RNA was extracted from the B cells and converted into cDNA via reverse transcription. Employing this cDNA as a template, the gene encoding the DDR1 antibody was amplified using degenerate primers through PCR and inserted into a vector. Transfection of host cells with the recombinant vector facilitated efficient antibody expression. The DDR1 recombinant monoclonal antibodies were harvested from the cell culture supernatant and purified using affinity chromatography. Rigorous validation, including ELISA testing, confirmed the antibody's specific reactivity with human DDR1 protein, ensuring its reliability and suitability for a range of applications.
DDR1, a tyrosine kinase, functions as a cell surface receptor for fibrillar collagen. It plays a crucial role in regulating various cellular processes, including cell attachment to the extracellular matrix, remodeling of the extracellular matrix, cell migration, differentiation, survival, and cell proliferation. Collagen binding triggers a signaling pathway involving SRC, ultimately leading to the activation of MAP kinases. DDR1 regulates extracellular matrix remodeling by up-regulating the matrix metalloproteinases MMP2, MMP7, and MMP9, thus facilitating cell migration and wound healing. It is essential for normal blastocyst implantation during pregnancy, normal mammary gland differentiation, and normal lactation. Additionally, DDR1 is required for normal ear morphology and hearing. It promotes smooth muscle cell migration, contributing to arterial wound healing. DDR1 also plays a role in tumor cell invasion. Notably, it phosphorylates PTPN11.
DDR1 is a 120-140 kDa type I transmembrane glycoprotein belonging to the discoidin-like domain containing subfamily of receptor tyrosine kinases. Also known as CAK, CD167a, RTK6, and TrkE, DDR1 serves as a receptor for collagen and is predominantly expressed on epithelial tissues, activated monocytes, neutrophils, and in several cancer types . DDR1 has been identified as a cancer-associated receptor tyrosine kinase that is highly expressed in several malignancies relative to normal tissues, including lung, breast, esophagus, ovary, and colon cancers . Immunohistochemical analysis of tissue microarrays containing 100 colon cancer specimens revealed that DDR1 was highly expressed in 81% of tumor tissues, with high expression levels associated with poor survival outcomes in patients .
Research methodology: To investigate DDR1's role in cancer, researchers typically perform immunohistochemistry on tissue microarrays, assess expression levels through flow cytometry, and conduct survival analyses to correlate expression with clinical outcomes. Western blotting and RT-PCR may be employed to examine protein and transcript levels, respectively.
The production of DDR1 recombinant monoclonal antibodies follows a multi-step process:
Immunization and B cell isolation: B cells are isolated from the spleen of animals immunized with recombinant human DDR1 protein (specifically the extracellular domain, amino acids 21-417) .
RNA extraction and reverse transcription: RNA is extracted from the B cells and converted into cDNA through reverse transcription .
Gene amplification and vector insertion: Using the cDNA as a template, the gene encoding the DDR1 antibody is amplified using a degenerate primer through PCR and inserted into an expression vector .
Transfection and expression: The recombinant vector is introduced into host cells (typically HEK293F cells) through transfection, enabling efficient antibody expression .
Harvesting and purification: The DDR1 recombinant monoclonal antibodies are harvested from the cell culture supernatant and subjected to purification via affinity chromatography, typically using protein G .
Validation: Rigorous validation, including ELISA testing, is conducted to confirm the antibody's specific reactivity with human DDR1 protein .
Alternative approaches include the mouse hybridoma method, where harvested spleen cells are fused with SP2/0 cells, and hybridoma supernatants are screened by ELISA against the immunogen, followed by flow cytometry to assess binding and internalization abilities .
DDR1 antibodies serve multiple research applications:
Diagnostic marker: DDR1 antibodies can identify DDR1 expression in tumor samples through immunohistochemistry, flow cytometry, and western blotting. This is particularly valuable as DDR1 is overexpressed in various cancer types including colon (81% of specimens), lung, breast, esophagus, and ovarian cancers .
Therapeutic development: DDR1 antibodies can be engineered into antibody-drug conjugates (ADCs) for targeted cancer therapy. For example, T 4H 11-DM4, an ADC targeting DDR1 carrying the tubulin inhibitor payload DM4, has demonstrated potent anti-proliferative activity in colon cancer cell lines with IC50 values in the nanomolar range .
Mechanism studies: These antibodies help elucidate DDR1-mediated signaling pathways in tumor progression, including those activated upon collagen binding that lead to sustained receptor phosphorylation .
Biomarker investigations: DDR1 expression levels can serve as prognostic biomarkers, as high expression correlates with poor survival in colon cancer patients .
When performing these applications, researchers should validate antibody specificity against the target using positive and negative control cell lines with known DDR1 expression levels.
Engineering DDR1 antibodies into effective antibody-drug conjugates involves several critical steps:
Antibody selection: Choose high-affinity anti-DDR1 monoclonal antibodies (such as clone T 4H 11) that demonstrate specific binding to the extracellular domain and efficient internalization upon binding .
Linker-payload selection: The linking chemistry is crucial for stability and payload release. For example, the T 4H 11-DM4 conjugate utilizes SPDB-DM4 (a disulfide-containing linker with the maytansinoid DM4 payload) .
Conjugation procedure:
Mix the anti-DDR1 mAb with a 10-fold molar excess of the linker-payload (e.g., SPDB-DM4) in conjugation buffer at a concentration of 5 mg·mL⁻¹
Perform coupling reactions overnight at 25°C
Separate the reaction mixtures by chromatography using a desalting column to yield the conjugates
Functional validation:
Assess binding affinity to DDR1-expressing cells via flow cytometry
Evaluate internalization efficiency using fluorescently labeled antibodies
Measure cytotoxicity in DDR1-expressing cancer cell lines
Confirm selectivity by testing in DDR1-negative cell lines
In the case of T 4H 11-DM4, this approach produced an ADC that exhibited potent anti-proliferative activity in vitro and achieved complete tumor regression at doses of 5 and 10 mg·kg⁻¹ in HT-29 and HCT116 tumor models in vivo .
Comprehensive validation of DDR1 antibodies requires multiple complementary approaches:
Binding specificity assessment:
ELISA: Using recombinant DDR1 protein (extracellular domain, amino acids 21-417) to confirm binding specificity
Western blotting: Testing against cell lysates with varying DDR1 expression levels
Flow cytometry: Analyzing binding to DDR1-expressing cells versus negative controls
Immunoprecipitation: Confirming ability to pull down DDR1 protein
Cross-reactivity testing:
Test against closely related proteins (particularly DDR2)
Assess binding to different DDR1 isoforms (DDR1a, DDR1b, etc.)
Functional validation:
Collagen binding inhibition assays: Determine if the antibody blocks DDR1-collagen interaction
Phosphorylation studies: Assess impact on DDR1 autophosphorylation using phospho-specific antibodies
Signaling pathway analysis: Examine effects on downstream pathways
Internalization studies:
In vivo validation:
Biodistribution studies using labeled antibodies
Target engagement assessment in tumor models
For therapeutic applications, additional functional assays such as antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) may be relevant depending on the antibody isotype and intended mechanism of action.
DDR1 expression patterns have significant implications for treatment response and patient stratification:
Expression correlation with outcomes:
Predictive value for targeted therapies:
Resistance mechanisms:
Patient stratification methodology:
IHC scoring system: Develop standardized scoring for DDR1 expression levels in tumor samples
Flow cytometry: Quantify cell surface DDR1 levels on tumor cells
Threshold determination: Establish expression thresholds that predict therapeutic response
For optimal patient stratification, researchers should:
Validate DDR1 expression assessment methods across multiple tumor types
Correlate expression with response to various therapeutic modalities
Consider heterogeneity of expression within tumors
Evaluate DDR1 isoform distribution, as different isoforms may influence therapeutic response
Integrating DDR1 antibodies with immunotherapies requires careful consideration of several factors:
Rationale for combination:
DDR1 inhibition may modulate the tumor microenvironment, potentially enhancing immunotherapy efficacy
Anti-PD-1/PD-L1 agents could complement DDR1-targeted therapies by removing immune suppression while DDR1 antibodies directly target tumor cells
Preclinical evidence:
While specific data on DDR1 antibody combinations with immunotherapy is limited in the provided search results, similar combination approaches have shown promise
For example, the addition of anti-PD-1 antibodies to oncolytic virus therapy (MG1) overcame resistance in advanced melanoma models with "immunologically colder" tumor microenvironments
Sequencing considerations:
Determine optimal timing (concurrent vs. sequential administration)
Assess whether DDR1 antibody treatment alters immune cell infiltration or activation
Monitoring parameters:
Track changes in immune cell populations following DDR1 antibody treatment
Assess alterations in cytokine profiles and immune checkpoint expression
Safety assessment:
Monitor for potential enhanced immune-related adverse events
Evaluate overlapping toxicity profiles
A methodological approach would include in vitro co-culture systems with tumor cells and immune components, followed by syngeneic mouse models (if cross-reactive antibodies are available) or humanized mouse models to assess combination efficacy before clinical translation.
Optimized flow cytometry protocols for DDR1 expression assessment in tumor samples should include:
Sample preparation:
Staining procedure:
Controls:
Isotype control antibody at the same concentration
FMO (Fluorescence Minus One) controls
Positive control (cell line with known high DDR1 expression)
Negative control (cell line with no DDR1 expression)
Multi-parameter considerations:
Include markers for specific cell populations (e.g., EpCAM for epithelial tumor cells)
Consider intracellular staining for phosphorylated DDR1 following permeabilization
Use compensation controls when multiple fluorophores are employed
Analysis:
This protocol can be adapted for analyzing patient-derived xenografts, clinical samples, or experimental models to ensure consistent and reliable DDR1 quantification across different research applications.
Optimizing DDR1 antibody-based immunohistochemistry requires attention to multiple technical parameters:
Tissue preparation:
Fixation: 10% neutral buffered formalin for 24-48 hours
Processing: Standard paraffin embedding
Sectioning: 4-5 μm thick sections
Mounting: Positively charged slides to prevent tissue loss
Antigen retrieval optimization:
Test multiple methods: Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Duration: Typically 15-20 minutes in pressure cooker or 30 minutes in water bath at 95-98°C
Cool gradually to room temperature before proceeding
Blocking and antibody parameters:
Endogenous peroxidase blocking: 3% hydrogen peroxide for 10 minutes
Protein blocking: 5-10% normal serum (species matched to secondary antibody)
Primary antibody concentration: Titrate between 1-10 μg/mL
Incubation time: Test both short (1-2 hours at room temperature) and overnight (4°C) incubations
Detection system: HRP-polymer based for increased sensitivity
Validation controls:
Scoring system development:
Establish a 0-3 intensity scale (0=negative, 1=weak, 2=moderate, 3=strong)
Calculate H-score (0-300) by multiplying intensity (0-3) by percentage of positive cells
Define thresholds for "high" vs "low" expression based on clinical correlation
Automation considerations:
Program standardized protocols on automated platforms
Include controls in each run
Implement regular quality control assessments
For multiplexing applications, consider fluorescent-based approaches to co-localize DDR1 with other markers of interest, which may provide additional insights into the tumor microenvironment and DDR1's relationship with other signaling pathways.
The efficacy of DDR1-targeted ADCs depends on multiple optimization variables:
Target expression levels:
Internalization kinetics:
Linker-payload selection:
The SPDB-DM4 linker system used with T 4H 11 proved effective
Key considerations:
Linker stability in circulation
Appropriate payload release mechanism inside target cells
Potency of payload against target tumor type
Optimization approach: Compare multiple linker-payload combinations (disulfide, peptide, non-cleavable)
Drug-antibody ratio (DAR):
Higher DAR increases potency but may affect pharmacokinetics
Analytical method: Confirm DAR via LC-MS
Optimization range: Test DAR ranges from 2-8 to identify optimal balance
Dose and schedule optimization:
T 4H 11-DM4 achieved complete tumor regression at doses of 5 and 10 mg·kg⁻¹ in colon cancer models
Approach: Test multiple dose levels and schedules (single vs. multiple dosing)
Safety assessment: Monitor for toxicities at higher doses (T 4H 11-DM4 exhibited no overt toxicities up to 50 mg·kg⁻¹ in BALB/c mice)
Resistance mechanisms:
When optimizing DDR1-targeted ADCs, researchers should consider the differential expression of DDR1 isoforms, as these may affect antibody binding, internalization rates, and therapeutic efficacy.
Addressing variability in DDR1 antibody performance requires systematic troubleshooting:
Antibody storage and handling:
Store according to manufacturer recommendations (typically at -20°C or -80°C)
Avoid repeated freeze-thaw cycles (aliquot upon receipt)
Maintain cold chain during experiments
Validate activity after prolonged storage
Epitope considerations:
DDR1 has five isoforms with variations in structure, though the extracellular domains are highly conserved
Solution: Use antibodies targeting conserved regions of the extracellular domain (amino acids 21-417)
Verification: Confirm which isoforms are expressed in your experimental system via PCR or western blotting
Post-translational modifications:
DDR1 is a glycoprotein with variable glycosylation patterns
Approach: For western blotting, consider deglycosylation treatments
Control: Include recombinant DDR1 protein as standard
Sample preparation impact:
Expression level detection limits:
Low expression may require signal amplification
Solutions:
For IHC: Use polymer-based detection systems
For flow cytometry: Consider indirect staining or higher antibody concentrations
For western blotting: Increase protein loading or use enhanced chemiluminescence
Validation across platforms:
Create a validation panel of cell lines with known DDR1 expression levels
Compare antibody performance across techniques (IHC, flow cytometry, western blotting)
Establish standardized protocols for each application
Batch-to-batch variability:
Request certificate of analysis with each lot
Test new lots alongside previous lots
Consider recombinant antibodies for improved consistency
By systematically addressing these variables, researchers can establish reliable protocols that yield consistent results across different experimental systems and applications.
Developing effective in vivo imaging applications with DDR1 antibodies requires attention to several technical and biological factors:
Conjugation chemistry:
Biodistribution optimization:
Antibody fragments may provide better tumor penetration and faster clearance
Options to consider:
Full IgG (longest half-life, slower tumor penetration)
F(ab')2 fragments (intermediate properties)
Fab fragments (rapid penetration, faster clearance)
Assessment approach: Compare biodistribution profiles of different formats at various time points
Target validation considerations:
Imaging parameters:
Timing: Determine optimal imaging window based on pharmacokinetics
Initial dynamic imaging to capture perfusion phase
Delayed imaging (24-72 hours post-injection) for optimal tumor-to-background ratio
Quantification methods:
Region of interest (ROI) analysis
Standardized uptake values (SUVs) for PET imaging
Tumor-to-background ratios
Correlation with treatment response:
Serial imaging to monitor changes in DDR1 expression during treatment
Correlation with ex vivo analysis (IHC, flow cytometry) to validate imaging findings
Integration with other imaging modalities (e.g., FDG-PET, MRI) for comprehensive assessment
Technical challenges and solutions:
Autofluorescence: Use near-infrared fluorophores (>700 nm) and spectral unmixing
Tissue attenuation: Consider photoacoustic imaging for deeper tissue penetration
Signal-to-noise ratio: Optimize antibody dose and imaging time point
When developing these applications, researchers should consider the correlation between DDR1 expression levels and therapeutic response to DDR1-targeted therapies, as this could provide valuable imaging biomarkers for patient selection and treatment monitoring.