CDP1 antibody is a recombinant anti-epidermal growth factor receptor (EGFR) human-mouse chimeric monoclonal antibody that has been developed as a biosimilar to cetuximab . The primary structure of CDP1 is identical to cetuximab, with highly similar higher-level structure, physical, and chemical properties . Pre-clinical studies show similar pharmacodynamic activity, pharmacokinetic characteristics, and toxicological reactions to cetuximab .
CDP1 was approved by the China Food and Drug Administration in August 2016 for clinical studies . Clinical trials are planned to compare the pharmacokinetic and clinical effectiveness of CDP1 with the original drug, Erbitux, and to assess its safety and immunogenicity .
CDP1 is currently under evaluation in clinical trials for the treatment of advanced solid tumors and metastatic colorectal cancer (CRC) .
Ongoing Clinical Trials:
Phase I Clinical Trial in Advanced Solid Tumors: This trial aims to evaluate the safety, tolerability, and dose-limiting toxicity of CDP1 in patients with advanced solid tumors to determine the recommended dose for Phase II trials .
Preliminary Clinical Study in Advanced CRC or Head and Neck Squamous Cell Carcinoma (HNSCC): This study is designed to compare the pharmacokinetic characteristics of a single dose of CDP1 with Erbitux in patients with advanced metastatic colorectal cancer . The trial includes single-dose and multi-dose phases to assess pharmacokinetics and safety .
CDP1, similar to cetuximab, targets the epidermal growth factor receptor (EGFR) . EGFR is a receptor tyrosine kinase that plays a critical role in cell growth, proliferation, and survival . By binding to EGFR, CDP1 inhibits the receptor's signaling pathways, thereby suppressing tumor growth and proliferation .
Clinical trials are designed to compare CDP1 with cetuximab (Erbitux) in terms of pharmacokinetics, efficacy, safety, and immunogenicity . The primary objective is to demonstrate that CDP1 is a biosimilar to cetuximab, with comparable clinical outcomes .
SH010: A novel PD-1/CTLA-4 bispecific antibody demonstrating favorable binding activity with both PD-1 and CTLA-4. SH010 has shown excellent blocking capabilities in protein binding between human PD-1 and PD-L1, as well as human CTLA-4 and CD80/CD86 .
CT-011: A humanized IgG1 monoclonal antibody that modulates the immune response through interaction with PD-1, a protein belonging to the B7 receptor family present on lymphocytes. The study showed the antibody to be safe and well tolerated in this patient population .
CUB-domain containing protein 1 (CDCP1) is a cancer associated cell surface protein that amplifies pro-tumorigenic signalling by other receptors including EGFR and HER2. Anti-CDCP1 antibodies inhibit cell migration and survival in vitro, and tumor growth and metastasis in vivo .
A panel of ovarian cancer cell lines was examined for cell surface expression of CDCP1 and loss of expression induced by anti-CDCP1 antibodies 10D7 and 41-2 using flow cytometry and Western blot analysis . The structural features of CDCP1 support its potential as a target for antibody based anti-cancer agents. It is predominantly located on the cell surface and after removal of its 29 residue signal peptide, CDCP1 spans 807 residues including a 637 residue amino-terminal extracellular domain (ECD), a 20 residue transmembrane domain, and a 150 residue carboxyl-terminal intracellular domain .
| Cell Line | CDCP1 Expression Level |
|---|---|
| HEY | Highest |
| CAOV3 | Similar |
| SKOV3 | Similar |
| OVTOKO | Similar |
| OVMZ6 | Non-expressing |
| OVMZ6-CDCP1 | Expressing |
| Antibody | k a (1/Ms) | k d (1/s) | K D (M) |
|---|---|---|---|
| 10D7 | Fast | Slow | 0.44 nM |
| 41-2 | Fast | Slow | 1.2 nM |
CDCP1 is a cell surface oncoreceptor that has emerged as a promising target for cancer diagnostics and therapeutics. The rationale for developing CDCP1-targeted antibodies stems from its differential expression pattern - elevated levels in various cancer types with restricted expression in normal human tissues . This expression profile creates a therapeutic window that allows for selective targeting of malignant cells while minimizing potential off-target effects in healthy tissues.
CDCP1 functions as a transmembrane glycoprotein involved in cell adhesion, migration, and survival signaling pathways, particularly through its interaction with Src family kinases. When developing antibodies against CDCP1, researchers aim to exploit these biological properties to interrupt cancer cell signaling, induce receptor degradation, or deliver therapeutic payloads specifically to cancer cells .
CDCP1 antibodies serve multiple functions in cancer research, with applications spanning from basic laboratory investigations to translational therapeutic development:
Diagnostic imaging agents: CDCP1 antibodies labeled with imaging isotopes such as 89Zirconium can be used for PET imaging to detect and visualize CDCP1-expressing tumors in vivo .
Therapeutic delivery vehicles: When conjugated with cytotoxic payloads like monomethyl auristatin E (MMAE), CDCP1 antibodies can function as antibody-drug conjugates (ADCs) that selectively deliver cytotoxic agents to cancer cells .
Biological research tools: Unconjugated antibodies are valuable for characterizing CDCP1 expression, localization, and signaling in various experimental systems through techniques like Western blotting, immunohistochemistry, and flow cytometry .
Patient stratification biomarkers: CDCP1 expression analysis using antibodies may help identify patients most likely to benefit from CDCP1-targeted therapies, enabling personalized treatment approaches .
The versatility of CDCP1 antibodies as both diagnostic (theranostic) and therapeutic agents makes them particularly valuable in translational cancer research.
Ensuring antibody specificity is critical for obtaining reliable research results. For CDCP1 antibodies, validation should include multiple complementary approaches:
Competitive binding assays: Compare binding of labeled and unlabeled antibodies to CDCP1-expressing cells. As demonstrated with ch10D7 and 10D7 antibodies, competition with equal amounts of unlabeled antibody should reduce binding of fluorescently-labeled antibody by approximately 50% .
Genetic knockdown/knockout controls: Test antibody binding in cell lines with CDCP1 silencing (shRNA) or knockout (CRISPR-Cas9). The TKCC05 PDAC cell line with CDCP1 knockdown showed significantly reduced binding of anti-CDCP1 antibodies compared to control cells .
Western blot analysis: Validate antibody specificity using cell lysates from multiple cell lines with varying CDCP1 expression levels. Look for correlation between signal intensity and known CDCP1 expression patterns .
Cross-reactivity testing: Examine binding to related proteins and in CDCP1-negative tissues to ensure specificity for the target of interest .
Recombinant protein controls: Use purified recombinant CDCP1 protein as a positive control and non-related proteins as negative controls in binding assays .
Proper validation is especially important given the antibody reproducibility crisis, where approximately 50% of commercial antibodies fail to meet basic standards for characterization .
CDCP1 antibody internalization is a complex process critical for the efficacy of antibody-drug conjugates. Research indicates that CDCP1 antibodies like ch10D7 induce rapid internalization upon binding, with significant endocytosis occurring within minutes to hours.
Mechanistically, antibody binding to CDCP1 initiates:
Receptor clustering and signaling: Binding triggers phosphorylation of CDCP1 at Y734 and activation of Src (phosphorylation at Y416) .
Signal-dependent endocytosis: The signaling cascade leads to receptor internalization through clathrin-dependent or independent pathways.
Lysosomal targeting: Internalized antibody-receptor complexes are directed to lysosomes, resulting in degradation of both the antibody and receptor .
Optimization strategies for enhancing internalization include:
Epitope selection: Target epitopes that induce rapid internalization without blocking critical functional domains unless inhibition is desired.
Antibody engineering: Modify antibody properties (affinity, avidity, isotype) to enhance clustering and internalization rates.
Linker chemistry: For ADCs, select linker chemistry compatible with the internalization and processing kinetics of the antibody-receptor complex.
Quantification of internalization can be performed using pH-sensitive dyes conjugated to antibodies, as demonstrated with ch10D7 pH, which shows increased fluorescence upon internalization due to acidification in endosomal/lysosomal compartments .
The relationship between CDCP1 expression and ADC efficacy is a critical consideration for therapeutic development. Analysis of multiple cancer cell lines treated with ch10D7-MMAE revealed a correlation between cell surface CDCP1 expression levels and sensitivity to the ADC.
Key findings from experimental studies include:
Threshold effect: Cell lines with fewer than 5×10⁴ anti-CDCP1 antibodies bound per cell were largely unresponsive to ch10D7-MMAE, suggesting a minimum expression threshold for efficacy .
Expression-response correlation: Higher CDCP1 expression generally correlated with improved ch10D7-MMAE efficacy across multiple cancer types .
Cancer type variations: PDAC (pancreatic ductal adenocarcinoma) cell lines showed particularly high CDCP1 expression, with up to 3×10⁵ antibodies bound per cell .
| Cancer Type | Representative Cell Lines | CDCP1 Antibodies Bound/Cell | ch10D7-MMAE Sensitivity |
|---|---|---|---|
| Pancreatic | TKCC2.1 | ~3×10⁵ | High |
| Ovarian | HEY | Variable | Moderate to High |
| Colorectal | HT29, HCT116 | Variable | Variable |
| Kidney | A498, 786-O | CDCP1-FL only | Low to Moderate |
| Lung | A549, H1650 | Variable | Variable |
| Prostate | DU145 | Mixed CDCP1-FL and CTF | Variable |
Table 1: Relationship between CDCP1 expression and sensitivity to ch10D7-MMAE across cancer types. CDCP1-FL: full-length CDCP1; CTF: C-terminal fragment.
These findings suggest that quantifying CDCP1 expression levels in patient tumors could serve as a predictive biomarker for selecting patients most likely to benefit from CDCP1-targeted ADCs .
The binding kinetics of anti-CDCP1 antibodies significantly influence their therapeutic potential. Engineering efforts to create clinically viable antibodies must maintain optimal binding properties while reducing immunogenicity.
For the chimeric antibody ch10D7, surface plasmon resonance (SPR) spectroscopy revealed:
High affinity binding: ch10D7 displays a KD of 0.28 nM for recombinant CDCP1-ECD, comparable to the parent murine 10D7 (KD of 0.34 nM) .
Association/dissociation rates: Rapid association and slow dissociation kinetics contribute to the high affinity and sustained target engagement.
Binding site conservation: Competition assays demonstrated that ch10D7 and the parent 10D7 bind to the same epitope on CDCP1 .
These kinetic properties impact therapeutic applications in several ways:
Tumor penetration: Very high affinity antibodies may exhibit a "binding site barrier" effect, limiting penetration into solid tumors.
Receptor occupancy: The high affinity ensures sustained receptor occupancy even at lower antibody concentrations.
Internalization dynamics: Binding kinetics influence the rate and extent of antibody-receptor complex internalization, which is critical for ADC efficacy.
Receptor recovery after treatment: After antibody-induced CDCP1 degradation, receptor re-expression begins approximately 24 hours after antibody withdrawal, with levels returning to baseline by 48 hours . This information is valuable for determining optimal dosing schedules for clinical applications.
CDCP1 antibodies have shown potential for multimodal applications in cancer management, functioning as both diagnostic imaging agents and therapeutic delivery vehicles. Optimizing these antibodies requires consideration of several factors:
Conjugation chemistry: Different payloads (imaging isotopes vs. cytotoxic drugs) require distinct conjugation strategies that preserve antibody binding properties while ensuring stability of the conjugate. Successful examples include:
Payload selection: The choice between:
Diagnostic radioisotopes (89Zr, 124I, 68Ga) for PET imaging
Therapeutic isotopes (177Lu, 90Y) for radioligand therapy
Cytotoxic drugs (MMAE, DM1, SN-38) for ADC approaches
Immunomodulatory agents for enhancing anti-tumor immune responses
Format optimization: Considerations include:
Full IgG vs. antibody fragments (Fab, F(ab')2, scFv)
Humanization/chimerization to reduce immunogenicity
Fc engineering to modulate effector functions or half-life
Theranostic pairs: Developing matched diagnostic/therapeutic pairs using the same antibody backbone conjugated with different payloads enables precise patient selection and treatment monitoring .
The theranostic approach is particularly valuable for personalized cancer treatment, as it allows for non-invasive confirmation of target expression before administering targeted therapies, potentially improving response rates and reducing unnecessary treatments.
Working with CDCP1 antibodies presents several potential challenges that researchers should anticipate and address:
CDCP1 isoform heterogeneity: CDCP1 exists in both full-length (135 kDa, CDCP1-FL) and proteolytically cleaved forms (70 kDa C-terminal fragment, CDCP1-CTF). Cell lines may express only CDCP1-FL, only CDCP1-CTF, or a mixture of both . This heterogeneity can complicate interpretation of experimental results if antibodies recognize only specific forms.
Solution: Use antibodies that recognize epitopes present in all relevant CDCP1 forms or combine multiple antibodies targeting different domains.
Antibody-induced receptor downregulation: Anti-CDCP1 antibodies like ch10D7 induce receptor degradation, which can alter experimental outcomes in extended treatments .
Solution: Include time-course analyses to capture dynamic changes in receptor levels and signaling. For long-term experiments, consider the kinetics of receptor recovery after antibody withdrawal.
Variability in CDCP1 expression: CDCP1 expression can vary dramatically between cell lines and may change with culture conditions or cellular stress .
Solution: Regularly quantify CDCP1 expression levels in your experimental system. Consider flow cytometry to assess surface expression specifically.
Antibody characterization inadequacy: The "antibody crisis" affects many research antibodies, with approximately 50% failing to meet basic characterization standards .
Solution: Perform thorough validation of any anti-CDCP1 antibody before use, including specificity testing with appropriate positive and negative controls.
Clone-specific effects: Different anti-CDCP1 antibody clones may have distinct effects on receptor signaling and biology even when targeting similar epitopes .
Solution: When comparing studies using different antibody clones, consider that observed differences might reflect clone-specific properties rather than biological variations.
Accurate quantification of CDCP1 surface expression is essential for predicting response to CDCP1-targeted therapies. Several methodologies can be employed:
Quantitative flow cytometry:
Use calibrated beads with known antibody binding capacity to establish a standard curve
Measure mean fluorescence intensity of cells labeled with fluorescent anti-CDCP1 antibody
Convert to antibodies bound per cell using the standard curve
This approach revealed that cells with <5×10⁴ anti-CDCP1 antibodies bound/cell were generally unresponsive to ch10D7-MMAE treatment
Saturation binding assays:
Incubate cells with increasing concentrations of labeled antibody
Plot binding vs. concentration and determine Bmax (maximum binding capacity)
Calculate receptors per cell based on known antibody:receptor stoichiometry
Immunohistochemistry with digital image analysis:
Stain tissue sections with anti-CDCP1 antibodies
Use digital pathology algorithms to quantify membrane staining intensity
Compare to calibrated standards for relative quantification
Proteomics approaches:
Absolute quantification (AQUA) peptide strategies
Mass spectrometry to determine CDCP1 concentration in membrane preparations
For clinical translation, establishing standardized cutoffs for "CDCP1-high" vs. "CDCP1-low" expression will be critical for patient stratification. The observation that ~5×10⁴ antibodies bound/cell represents a threshold for ADC efficacy provides an important benchmark for such stratification strategies .
Recent advances in antibody engineering have expanded the capabilities of CDCP1-targeted therapeutics:
Humanization and chimerization: The development of chimeric antibodies like ch10D7 (murine variable regions on human IgG1κ backbone) reduces immunogenicity while maintaining high affinity (KD of 0.28 nM) comparable to the parent murine antibody (KD of 0.34 nM) .
Site-specific conjugation technologies: Advances in conjugation chemistry allow for precise control over drug-antibody ratio (DAR) and conjugation sites, improving the homogeneity and stability of antibody-drug conjugates.
Bispecific antibody formats: Engineering bispecific antibodies that simultaneously target CDCP1 and other cancer-associated antigens (e.g., EGFR, HER2) or immune effector cells (e.g., CD3) to enhance therapeutic efficacy.
Enhanced internalization variants: Structure-guided engineering to identify variants with faster internalization kinetics, potentially improving ADC potency.
pH-sensitive binding: Engineering antibodies with pH-dependent binding properties that facilitate payload release in the acidic endosomal/lysosomal environment while maintaining strong binding at the cell surface.
Recombinant antibody production: Moving away from hybridoma-derived antibodies toward recombinant production systems improves batch-to-batch consistency and reduces background binding. This approach aligns with efforts like the Protein Capture Reagent Program (PCRP) and Affinomics to generate well-characterized antibody reagents .
The development of recombinant antibody libraries against CDCP1 epitopes provides researchers with consistent, renewable reagents that avoid the batch-to-batch variability inherent in traditional monoclonal antibody production.
Single-cell technologies offer unprecedented opportunities to understand CDCP1 biology and optimize antibody-based targeting strategies:
Tumor heterogeneity assessment: Single-cell RNA sequencing and mass cytometry can reveal the heterogeneity of CDCP1 expression within tumors, identifying subpopulations that might escape antibody targeting.
Response prediction: Correlating single-cell CDCP1 expression profiles with response to CDCP1-targeted therapies may identify cellular states or marker combinations that predict therapeutic efficacy beyond simple expression level thresholds.
Resistance mechanism identification: Analyzing changes in single-cell profiles before and after treatment can reveal adaptive responses and resistance mechanisms to CDCP1-targeted therapy.
Optimal combination strategies: Single-cell multi-omics approaches can identify pathways co-activated with CDCP1 signaling, suggesting rational combination therapies.
Microenvironment interactions: Examining how CDCP1-expressing tumor cells interact with the tumor microenvironment at single-cell resolution may reveal context-dependent effects of antibody targeting.
The implementation of single-cell approaches represents a frontier in CDCP1 research that could significantly enhance the precision of antibody-based therapeutics by addressing the challenge of intra-tumor heterogeneity in CDCP1 expression.
CDCP1-targeted antibody therapeutics are in early stages of clinical development:
Preclinical validation: Extensive preclinical work has established CDCP1 as a promising target, with antibody-drug conjugates showing efficacy in multiple cancer models including pancreatic, colorectal, and ovarian cancer xenografts .
Clinical trials: A Phase I clinical trial of CDP1 in patients with advanced solid tumors has been initiated to evaluate safety, tolerability, and determine the recommended Phase II dose (RP2D) . The trial objectives include:
Biomarker development: Parallel efforts to develop CDCP1 as a biomarker for patient stratification are ongoing, based on the correlation between CDCP1 expression levels and sensitivity to CDCP1-targeted therapies .
Combination approaches: Strategies combining CDCP1-targeted antibodies with other therapeutic modalities (chemotherapy, immunotherapy, targeted agents) are being explored to enhance efficacy and overcome potential resistance mechanisms.
The advancement of CDCP1 antibodies into clinical testing represents a significant milestone in translating the extensive preclinical research into potential new treatment options for patients with CDCP1-expressing malignancies.