CEACAM5 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Structure

CEACAM5 recombinant mAbs are immunoglobulin G (IgG)-based molecules produced by cloning antigen-binding regions into stable cell lines. They target distinct epitopes on CEACAM5, a glycosylphosphatidylinositol (GPI)-anchored protein with six immunoglobulin-like domains (N, A1–A3, B1–B3). CEACAM5 facilitates tumor progression by inhibiting anoikis, promoting metastasis, and modulating immune evasion .

  • CC4: Demonstrated 50–70% suppression of colorectal tumor growth in xenograft models and enhanced NK cell cytotoxicity by blocking CEACAM5-mediated immune evasion .

  • 061: Validated for distinguishing pulmonary adenocarcinoma (60–70% CEACAM5+) from mesothelioma (rarely CEACAM5+) .

  • BLR198J: Shows no cross-reactivity with CEACAM1 or CEACAM6 in lung and bladder carcinoma samples .

Mechanisms of Action

  • Tumor Growth Inhibition:

    • CC4 blocks CEACAM5 homophilic/heterophilic interactions, reducing cell adhesion, migration, and proliferation (e.g., 25 µg/mL CC4 decreased LS174T colorectal cell migration by 60% ).

  • Immune Modulation:

    • CEACAM5 binds CEACAM1 on NK cells, inhibiting cytotoxicity. CC4 reverses this suppression, restoring NK-mediated tumor cell killing by 40–50% .

  • Targeted Drug Delivery:

    • Bispecific antibodies like NILK-2401 (anti-CEACAM5/CD47) avoid anemia/thrombocytopenia by sparing CD47 on erythrocytes .

    • Tusamitamab (SAR408701), an antibody-drug conjugate (ADC), delivers DM4 toxin to CEACAM5+ NSCLC cells, showing 30–40% objective response rates in Phase II trials .

Epitope Mapping and Specificity

  • CC4: Binds the N-terminal IgV-like domain (aa 42–61), critical for CEACAM5-CEACAM1 interactions .

  • Tusamitamab: Targets a conformational epitope on A3-B3 domains (aa 400–511), avoiding cross-reactivity with CEACAM1/6/8 .

  • Structural Insights: Cryo-EM resolved tusamitamab’s paratope interactions with CEACAM5 A3-B3 domains at 2.8 Å resolution, informing ADC design .

Clinical and Diagnostic Utility

  • Diagnostics:

    • CEACAM5 mAbs differentiate adenocarcinomas (e.g., 90% GI/pancreatic tumors) from benign tissues .

  • Therapeutics:

    • SAR408701: Phase III trials in CEACAM5+ NSCLC (NCT04394624) show median progression-free survival of 5.4 months .

    • NILK-2401: Reduces tumor volume by 70% in colorectal PDX models without RBC/platelet toxicity .

Challenges and Future Directions

  • Tumor Heterogeneity: CEACAM5 expression varies spatially within tumors, requiring companion diagnostics for ADC eligibility .

  • Novel Formats: Bispecific antibodies (e.g., CEACAM5xCD3 T-cell engagers) and glycoengineered mAbs are in preclinical testing to improve efficacy .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method or location. Please contact your local distributor for specific delivery time estimates.
Synonyms
Carcinoembryonic antigen antibody; Carcinoembryonic antigen-related cell adhesion molecule 5 antibody; CD66e antibody; CEA antibody; Ceacam5 antibody; CEAM5_HUMAN antibody; DKFZp781M2392 antibody; Meconium antigen 100 antibody; OTTHUMP00000199032 antibody; OTTHUMP00000199033 antibody; OTTHUMP00000199034 antibody
Target Names
Uniprot No.

Target Background

Function
CEACAM5 is a cell surface glycoprotein that plays a crucial role in cell adhesion, intracellular signaling, and tumor progression. It mediates both homophilic and heterophilic cell adhesion with other carcinoembryonic antigen-related cell adhesion molecules, such as CEACAM6. CEACAM5 acts as an oncogene by promoting tumor progression and inducing resistance to anoikis in colorectal carcinoma cells. In the context of microbial infections, CEACAM5 serves as a receptor for *E. coli* Dr adhesins. Binding of these adhesins leads to the dissociation of the CEACAM5 homodimer.
Gene References Into Functions
  1. Positive CEA mRNA expression in low rectal cancer is a factor associated with an elevated risk of overall recurrence, particularly local recurrence. PMID: 28291565
  2. Elevated carcinoembryonic antigen levels are correlated with recurrence in rectal cancer. PMID: 29774483
  3. CEA and CA19-9 are cancer antigens that serve as late markers of carcinogenesis. Significantly elevated serum concentrations of these antigens are observed in cases of colon cancer with already developed metastases. Older patient age groups exhibit significantly elevated levels of both antigens. Cancer prevalence was found to be twice as high in men compared to women. PMID: 25568506
  4. High CEA expression is associated with breast cancer metastasis. PMID: 29433529
  5. Pretreatment serum CEA levels exceeding 30.02 ng/mL are indicative of worse tumor characteristics, unfavorable tumor behavior, and a poor prognosis for gastric cancer patients, with a nearly doubled risk of mortality. PMID: 29358864
  6. Elevation of CEA levels is an independent risk factor for poor prognosis in patients with early gastric cancer. PMID: 29121872
  7. Serum CEA level elevation was independent of other tumor markers in hypohidrotic conditions characterized by acquired idiopathic generalized anhidrosis. PMID: 28295553
  8. Elevated CEA levels during targeted therapy might be a more sensitive predictor of explosive lung adenocarcinoma progression in patients harboring mutant EGFRs compared to conventional imaging methods. PMID: 28705152
  9. Increased CEA expression is associated with colorectal cancer. PMID: 28128739
  10. The combined detection of TK1 with cytokeratin-19 fragment (CYFRA21-1), CEA, or NSE enhanced the diagnostic value of TK1 for lung squamous cell carcinoma, adenocarcinoma, and small cell lung cancer, respectively. PMID: 29247745
  11. In patients with non-small-cell lung cancer receiving nivolumab therapy, worse pretreatment performance status and higher carcinoembryonic antigen levels were linked to inferior progression-free survival. PMID: 29277824
  12. To investigate the relationship between carcinoembryonic antigen (CEA) response and tumor response and survival in patients with (K)RAS wild-type metastatic colorectal cancer undergoing first-line chemotherapy in the FIRE-3 trial comparing FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab. PMID: 27234640
  13. This population-based study of a large cohort of exclusively stage I rectal cancer patients provides strong evidence that elevated preoperative CEA levels are a significant predictor of worse overall and cancer-specific survival. PMID: 27067235
  14. Elevated systemic IFNgamma and IL-6 levels indicated that CEACAM5-specific T cells had undergone immune activation in vivo, but only in patients receiving high-intensity pre-conditioning. PMID: 28660319
  15. The combination of the two tumor markers, CEA and CYFRA21-1, serves as a predictive and prognostic marker for docetaxel monotherapy in previously treated NSCLC patients. PMID: 28870944
  16. Patients with CEA-High stage I NSCLC have a higher risk of regional or systemic relapse and require careful follow-up. PMID: 28870949
  17. Postoperative CEA levels greater than 2.5 ng/ml are a predictor of distant metastasis and a negative prognostic factor for survival in rectal cancer patients who undergo preoperative chemoradiotherapy and curative surgery. PMID: 27553616
  18. Data indicate that triple-negative breast cancer (TNBC) patients with pre-therapeutic serum high levels of both carcinoembryonic antigen (CEA) and cancer antigen 15-3 (CA15-3) experienced shorter overall survival (OS) and disease-free survival (DFS) rates compared to those in the low-level groups. PMID: 27561099
  19. Serum CEA levels are not influenced by the presence of benign dominant biliary stricture or superimposed bacterial cholangitis in patients with primary sclerosing cholangitis. PMID: 27943017
  20. Preoperative-CEA was elevated (≥ 5.0 ng/mL) in 73.6% of the colorectal cancer patients and remained elevated after surgery in 32.7% of the patients. Elevated postoperative-CA 19-9 (≥ 50 U/mL) was observed in 9.5% of the patients. Neither elevated pre-CEA nor elevated pre-CA 19-9 was associated with relapse-free survival (RFS), but both elevated post-CEA and elevated post-CA 19-9 were associated with reduced RFS. PMID: 27664887
  21. The diagnostic sensitivity and specificity of serum reactive oxygen species modulator 1 were only 41.38% and 86.21%, respectively, with a cutoff value of 27.22 ng/mL. The sensitivity and specificity of pleural fluid carcinoembryonic antigen were 69.23% and 88.00%, respectively, at the cutoff value of 3.05 ng/mL, while serum carcinoembryonic antigen were 80.77% and 72.00% at the cutoff value of 2.60 ng/mL. PMID: 28459208
  22. Collectively, these findings identify CEACAM5 as a novel cell surface binding target of Middle East respiratory syndrome coronavirus, facilitating infection by enhancing viral attachment to the host cell surface. PMID: 27489282
  23. Data suggest that carcinoembryonic antigen (CEA) modestly differentiated between mucinous and nonmucinous lesions, and amylase did not distinguish intraductal papillary mucinous neoplasms (IPMNs) from mucinous cystadenomas (MCAs). PMID: 26646270
  24. In this study, the tumor marker carcinoembryonic antigen (CEA) was detected using the QD-LFTS system, enabling quantitative analysis within the range of 1-100 ng/mL with an ideal detection limit of 0.049 ng/mL. Therefore, the system is suitable for detecting CEA within the clinically accepted range. Additionally, 70 positive and 30 negative serum samples were analyzed using the Handing system, demonstrating good specificity and sensitivity. PMID: 27825889
  25. As a proof-of-concept study, the constructed platform exhibits good specificity for CEA, achieving a detection limit as low as 8 pg/mL (45 fM) with a wide linear range from 0.01 to 60 ng/mL in both cases. PMID: 27886601
  26. Under optimal conditions, the proposed immunosensor was used for the detection of CEA with a wide dynamic range from 5 fg/mL to 50 ng/mL, achieving a low detection limit of 2 fg/mL (S/N=3). PMID: 27871047
  27. The relative CL intensity of the all-in-one dual-aptasensor, operated with the competitive reaction of CEA and hemin in the presence of the dual aptamer, exhibited an exponential decrease with increasing CEA concentration in human serum. PMID: 27875751
  28. Data reveal that serum tumor markers, particularly S-CYFRA 21-1 (cytokeratin 19 fragment), S-SCCA, and S-CEA, at higher levels compared to lower levels, were associated with significantly shorter 3-year progression-free survival (PFS). PMID: 26432331
  29. Multivariate logistic regression analysis identified TC and DeltaCEA as independent predictors of TRG. TC demonstrated a sensitivity of 62.79%, specificity of 91.49%, Youden index of 0.543, cutoff value of 5.52, and an AUC of 0.800, compared to DeltaCEA (sensitivity 76.74%, specificity 65.96%, Youden index 0.427, and AUC 0.761). PMID: 26531721
  30. Findings suggest that the combined detection of receptor-binding cancer antigen expressed on SiSo cells (RCAS1) and carcinoembryonic antigen (CEA) can enhance diagnostic sensitivity and specificity. PMID: 26438059
  31. High CEA levels are associated with Oral Squamous Cell Carcinoma. PMID: 27165212
  32. High CEA expression is associated with metastasis and recurrence in endometrial cancer. PMID: 26779635
  33. ZKSCAN3 appears to promote colorectal tumor progression and invasion. ZKSCAN3 may facilitate hepatic metastasis of CRC associated with CEA, particularly in cases involving CEA-producing tumors. PMID: 27127149
  34. This study evaluated the clinical performance of LOCItrademark-based tumor marker assays CEA, CA19-9, CA15-3, CA125, and AFP in patients with gastrointestinal cancer, demonstrating their high diagnostic power. PMID: 28011514
  35. High carcinoembryonic Antigen expression is associated with gastric cancer. PMID: 25124614
  36. High CEA expression is associated with Squamous cell carcinoma of the skin. PMID: 27039776
  37. Preparation of Au-polydopamine functionalized carbon encapsulated FeO magnetic nanocomposites and their application for ultrasensitive detection of carcino-embryonic antigen. PMID: 26868035
  38. Elevated Carcinoembryonic Antigen Levels are associated with Colon Cancer. PMID: 26759308
  39. Baseline serum CEA levels can serve as predictive factors for the treatment of EGFR-TKI in non-small cell lung cancer patients harboring EGFR mutations. PMID: 27072247
  40. BALF and serum NSE, CEA, and CYFRA21.1 levels were elevated in lung cancer, demonstrating their diagnostic value, particularly for BALF. PMID: 27072263
  41. Significant levels of CEA, CYFRA 21-1, NSE, and TSGF were detected in the serum. The amounts found were useful for diagnosing non-small cell lung cancer (NSCLC) patients, contributing to the limited biomarker development in this field. PMID: 27072222
  42. These findings suggest that patients with high preoperative serum CEA levels should undergo more intensive follow-up to detect synchronous liver metastasis. PMID: 26756614
  43. Elevated CEA expression is associated with Gastric Cancer. PMID: 26620645
  44. CEA, NSE, CA125, and pro-GRP could serve as biomarkers for SCLC, and CEA and CYFRA21-1 could serve as biomarkers for NSCLC. Pro-GRP, CA125, and CEA were related to the clinical stages of lung cancer. PMID: 26560853
  45. In this phase I/II study, 14 high-risk disease-free ovarian (OC) and breast cancer (BC) patients after completion of standard therapies were vaccinated with MUC1, ErbB2, and carcinoembryonic antigen (CEA) HLA-A2+-restricted peptides and Montanide. PMID: 26892612
  46. Combining pre-chemoradiotherapy CEA and post-chemoradiotherapy CEA levels enables more accurate prediction of rectal adenocarcinoma prognosis. PMID: 26962798
  47. ESCC patients with lower Cyfra21-1 and CEA, higher miR-7, and severe myelosuppression were more sensitive to CRT. PMID: 26708917
  48. Cyst fluid CEA levels have a clinically suboptimal accuracy level in differentiating pancreatic mucinous cystic neoplasms from pancreatic nonmucinous cystic neoplasms. PMID: 26077458
  49. High carcinoembryonic antigen expression is associated with Colon Adenomas. PMID: 27100181
  50. Serum CYFRA21-1 and CEA can be used as prognostic factors for NSCLC patients. The combined detection of these two indices provides a more reliable assessment. PMID: 26333429

Show More

Hide All

Database Links

HGNC: 1817

OMIM: 114890

KEGG: hsa:1048

STRING: 9606.ENSP00000221992

UniGene: Hs.709196

Protein Families
Immunoglobulin superfamily, CEA family
Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor. Apical cell membrane. Cell surface.
Tissue Specificity
Expressed in columnar epithelial and goblet cells of the colon (at protein level). Found in adenocarcinomas of endodermally derived digestive system epithelium and fetal colon.

Q&A

What is CEACAM5 and why is it a significant target for cancer research?

CEACAM5, also known as CD66e or CEA, is the founding member of the carcinoembryonic antigen (CEA) family of proteins within the immunoglobulin (Ig) superfamily. Its significance stems from several key characteristics:

  • CEACAM5 is synthesized during fetal gut development and is re-expressed in increased amounts in various malignancies, particularly intestinal carcinomas

  • The extracellular domains of CEACAM5 function as:

    • Homophilic and heterophilic intercellular adhesion molecules

    • Human pathogen receptors

  • CEACAM5 expression has been documented in multiple solid tumors including:

    • Colorectal adenocarcinoma (primary expression site)

    • Lung adenocarcinoma (60-70% are CEACAM5+)

    • Gastric adenocarcinoma

    • Esophageal adenocarcinoma

    • Pancreatic carcinoma

    • Gallbladder adenocarcinoma

    • Urachal carcinoma

    • Salivary gland tumors

    • Ovarian and endocervical adenocarcinomas

This expression profile makes CEACAM5 a valuable diagnostic marker and therapeutic target, particularly for differentiating adenocarcinomas from other malignancies like pleural mesotheliomas (which rarely or weakly express CEACAM5) .

How do recombinant monoclonal anti-CEACAM5 antibodies differ from conventional antibodies in research applications?

Recombinant monoclonal anti-CEACAM5 antibodies offer several advantages over conventional (hybridoma-derived) antibodies:

  • Improved consistency: Recombinant antibodies show higher repeatability and batch-to-batch consistency

  • Increased sensitivity: Enhanced detection capabilities for low-abundance CEACAM5 expression

  • Confirmed specificity: Better discrimination between CEACAM5 and other family members like CEACAM1, CEACAM6, and CEACAM8

  • Production flexibility: Can be produced in various expression systems (e.g., HEK293 cells)

  • Sequence-defined paratopes: Enable detailed epitope mapping studies for therapeutic development

For example, the CC4 anti-CEACAM5 monoclonal antibody demonstrates excellent specificity for CEACAM5 in flow cytometry, immunofluorescence, and immunohistochemistry applications, with minimal cross-reactivity to other family members .

What are the optimal protocols for using anti-CEACAM5 antibodies in immunohistochemistry?

For optimal immunohistochemical staining with anti-CEACAM5 antibodies, researchers should consider:

Sample preparation:

  • For formalin-fixed paraffin-embedded (FFPE) tissues:

    • Some antibodies (e.g., COL-1 clone) require antigen retrieval by boiling tissue sections in 10mM Tris Buffer, pH 8.5-9.0 for 10-20 minutes followed by cooling at room temperature for 20 minutes

    • Typical section thickness: 4-6 μm

Protocol recommendations:

  • Working dilution: 0.5-2 μg/mL for most commercial anti-CEACAM5 antibodies

  • Incubation: 30 minutes at room temperature

  • Detection system: Polymer-based detection systems are preferred for enhanced sensitivity

  • Controls:

    • Positive control: Colorectal adenocarcinoma tissue

    • Negative control: Non-epithelial tissues (confirm antibody specificity)

Expected results:

  • Positive staining patterns: Cytoplasmic and luminal membrane localization in adenocarcinoma cells

  • Negative tissues: Most benign glands, stroma, and non-epithelial malignancies should show no staining

The staining pattern can help evaluate tumor differentiation and heterogeneity, with stronger membranous staining typically observed in well-differentiated adenocarcinomas.

How can researchers effectively employ anti-CEACAM5 antibodies in flow cytometry applications?

For effective flow cytometry applications with anti-CEACAM5 antibodies:

Protocol optimization:

  • Cell preparation:

    • Single-cell suspension preparation is critical (enzymatic dissociation may affect some epitopes)

    • Fixation: If needed, use 1-4% paraformaldehyde for 10-15 minutes at room temperature

  • Antibody concentration: 0.5-1 μg per 10^6 cells

  • Buffer composition: PBS with 0.5-2% BSA and 0.05-0.1% sodium azide

  • Incubation conditions: 30 minutes at 4°C

Gating strategy:

  • Forward/side scatter to exclude debris and select intact cells

  • Live/dead discrimination using appropriate viability dyes

  • Isotype controls to set negative populations

Validation approach:

  • Transfected vs. non-transfected cells as controls

    • As demonstrated with anti-CEACAM5 (DM120) on Expi293 cells transfected with human CEACAM5 versus Expi293 cells transfected with irrelevant protein

  • Comparison with known CEACAM5-positive cell lines (e.g., LS174T, SW1116)

Data analysis considerations:

  • Mean fluorescence intensity (MFI) for quantitative comparison

  • Percent positive cells for heterogeneous populations

  • Compensation for spectral overlap when using multiple fluorophores

This approach enables accurate detection and quantification of CEACAM5 expression levels across different cell populations.

What methodology should be used to validate the specificity of anti-CEACAM5 antibodies?

Validating antibody specificity is crucial for reliable research outcomes. For anti-CEACAM5 antibodies, a comprehensive validation approach includes:

1. Molecular characterization:

  • Western blot analysis under reduced and non-reduced conditions

    • Example: CC4 mAb shows differential binding patterns under these conditions, confirming conformation-dependent epitope recognition

  • Immunoprecipitation followed by mass spectrometry

    • As demonstrated with CC4 mAb, which identified four CEACAM5-specific peptides

2. Cell-based validation:

  • Transfection studies:

    • Overexpression of CEACAM5 in non-expressing cell lines

    • Example: Bladder cancer T2-4 cells transfected with CEACAM5 expression plasmids showed positive staining with CC4 antibody

  • Knockout/knockdown approaches to confirm signal loss

3. Cross-reactivity assessment:

  • Testing against other CEACAM family members

    • Important for verifying that antibodies don't recognize CEACAM1, CEACAM6, or CEACAM8

  • Epitope mapping studies:

    • Using recombinant CEACAM5 fragments and deletion mutants

    • Example: CC4 epitope mapped to aa42-61 region

4. Tissue validation:

  • Immunohistochemistry on known positive and negative tissues

  • Comparison of staining patterns with established CEACAM5 expression profiles

5. Functional validation:

  • Confirming biological effects align with CEACAM5 mechanisms

    • Example: CC4 mAb suppresses colorectal tumor cell proliferation, migration, and aggregation in a dose-dependent manner

This multi-platform validation approach ensures confident interpretation of experimental results with anti-CEACAM5 antibodies.

What are the critical structural features of CEACAM5 that influence antibody binding and specificity?

The structural features of CEACAM5 that impact antibody binding include:

Domain architecture:

Key binding regions:

  • The N-terminal domain (aa35-155) contains epitopes for many antibodies

    • CC4 antibody specifically binds to a narrow region (aa42-61) in this domain

  • The A3-B3 domains (aa499-685) harbor epitopes for other antibodies

    • Tusamitamab specifically targets this region and doesn't bind to other CEACAM family members

Glycosylation patterns:

  • CEACAM5 contains multiple N-linked glycosylation sites that influence antibody recognition

  • High-resolution structural studies have included these glycans in epitope mapping

Conformational epitopes:

  • Some antibodies recognize conformational epitopes dependent on proper protein folding

    • Evidenced by differential binding under reduced versus non-reduced conditions

Unique binding determinants:

  • Specific amino acid sequences distinguish CEACAM5 from other family members

    • For example, the region aa42-61 in CEACAM5 differs from the corresponding region in CEACAM1

These structural characteristics enable the development of highly specific antibodies that can discriminate between CEACAM5 and closely related family members.

How do anti-CEACAM5 monoclonal antibodies like CC4 suppress tumor growth mechanistically?

Anti-CEACAM5 antibodies suppress tumor growth through multiple mechanisms, as demonstrated with the CC4 antibody:

1. Direct cellular effects:

  • Inhibition of proliferation:

    • CC4 treatment reduces cell division in colorectal cancer cell lines (LS174T and Lovo)

    • Demonstrated using CFSE staining in dose-dependent manner

  • Suppression of cell migration:

    • CC4 inhibits cell motility in chamber assays

    • Dose-dependent effect observed in multiple colorectal cancer cell lines (LS174T, Lovo, Colo-205)

  • Disruption of cell aggregation:

    • CC4 prevents cancer cell aggregation in a calcium-dependent manner

    • At 50 μg/ml, approximately 80% of LS174T cells remained single, compared to only 40% with control IgG

2. Immune modulation effects:

  • Enhancement of NK cell-mediated cytotoxicity:

    • CC4 disrupts the inhibitory CEACAM5-CEACAM1 interaction between tumor cells and NK cells

    • This prevents the immune evasion mechanism normally triggered by this interaction

    • Demonstrated in experiments with both CD56+CD16+ and CD56+CD16- NK cells

  • Blockade of CEACAM1 inhibitory signaling:

    • CEACAM1 contains ITIM (immunoreceptor tyrosine-based inhibition motif) sequences

    • When engaged by CEACAM5, these motifs trigger inhibitory signaling in NK cells

    • CC4 prevents this engagement, restoring NK cell activity against tumor cells

3. In vivo tumor suppression:

  • Xenograft studies show CC4 specifically accumulates at tumor sites

  • Remarkable repression of colorectal tumor growth observed in these models

This multi-modal mechanism of action makes anti-CEACAM5 antibodies promising therapeutic candidates for colorectal and other CEACAM5-expressing cancers.

What methodologies are used for epitope mapping of anti-CEACAM5 antibodies?

Epitope mapping of anti-CEACAM5 antibodies employs multiple complementary approaches:

1. Recombinant domain/fragment analysis:

  • Domain-level mapping:

    • Expression of individual CEACAM5 domains (N, A1, B1, A2, B2, A3, B3)

    • Example: CC4 antibody epitope was initially localized to Domain1 (aa35-155)

  • Progressive truncation strategy:

    • Creation of fragments with systematic truncations

    • CC4 epitope was further narrowed to aa42-61 through testing fragments F42-273, F62-273, F80-273, and F101-273

2. Alanine scanning mutagenesis:

  • Systematic amino acid substitution:

    • Key residues within epitopes are individually replaced with alanine

    • Example: For tusamitamab, five variants in the heavy chain (Asp33Ala, Phe101Ala, Gly102Ala, Gly105Ala, and Pro106Ala) and two in the light chain (Tyr32Ala and Asn50Ala) exhibited complete loss of binding to CEACAM5

3. Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

  • Measures the rate of hydrogen-deuterium exchange in peptide backbones

  • Regions protected from exchange upon antibody binding indicate epitope locations

  • Provides detailed insights into conformational epitopes

4. Structural biology approaches:

  • Cryo-electron microscopy (cryo-EM):

    • High-resolution structure determination of antibody-antigen complexes

    • Example: Structure of tusamitamab bound to the A3-B3 domains of CEACAM5

  • X-ray crystallography:

    • Atomic-level resolution of antibody-antigen interfaces

    • Particularly useful for rigid epitopes

5. Competitive binding assays:

  • Evaluates whether different antibodies compete for the same epitope region

  • Useful for comparing novel antibodies to those with known epitopes

These methods collectively provide comprehensive mapping of epitope-paratope interactions, informing rational design of improved CEACAM5-targeting therapeutics.

How can researchers leverage anti-CEACAM5 antibodies to study NK cell-mediated tumor immunity?

Anti-CEACAM5 antibodies offer unique tools for investigating NK cell-tumor interactions:

1. Experimental models for studying CEACAM5-CEACAM1 interactions:

  • Cell line system development:

    • Transfection of CEACAM5-negative cancer cells (e.g., HCT-15) with CEACAM5 or CEACAM1 expression vectors

    • Creation of stable cell lines (HCT-15/CEACAM5 and HCT-15/CEACAM1)

    • Verification of expression using flow cytometry

  • NK cell preparation:

    • Isolation of CD56+CD16+ and CD56+CD16- NK cell subsets

    • Confirmation of CEACAM1 expression on NK cells

2. NK cytotoxicity assay methodologies:

  • Tumor target cell preparation:

    • CEACAM5-expressing colorectal cancer cells (endogenous or transfected)

  • Effector:target ratio optimization:

    • Testing multiple E:T ratios to determine optimal assay sensitivity

    • Example study used varying ratios to demonstrate CEACAM5-dependent effects

  • Cytotoxicity measurement:

    • 51Cr-release assays or flow cytometry-based methods

  • Antibody blockade experiments:

    • Addition of anti-CEACAM5 antibodies (e.g., CC4) at defined concentrations

    • Inclusion of isotype controls for comparison

3. Signaling pathway analysis:

  • ITIM-dependent signaling investigation:

    • Analysis of tyrosine phosphorylation in CEACAM1's cytoplasmic tail

    • Evaluation of downstream inhibitory signaling cascade components

  • Molecular intervention approaches:

    • CEACAM1 mutants lacking ITIM motifs

    • SHP-1/SHP-2 phosphatase inhibitors

4. In vivo models:

  • Humanized mouse systems:

    • Mice engrafted with human NK cells and CEACAM5+ tumors

    • Treatment with anti-CEACAM5 antibodies

    • Monitoring tumor growth and NK cell infiltration

This research framework has revealed that CEACAM5-CEACAM1 interactions represent an immune evasion mechanism in colorectal cancer, with CEACAM5 on tumor cells engaging CEACAM1 on NK cells to suppress cytotoxicity. Anti-CEACAM5 antibodies like CC4 can disrupt this interaction, potentially restoring anti-tumor immunity .

What approaches can address cross-reactivity issues with other CEACAM family members?

Cross-reactivity with related CEACAM family members presents a significant challenge in developing specific anti-CEACAM5 antibodies. Researchers can employ several strategies to address this issue:

1. Epitope-focused antibody design:

  • Unique epitope identification:

    • Comparative sequence analysis of CEACAM family members

    • Targeting regions with lowest homology (e.g., aa42-61 region for CC4)

  • Structure-guided approach:

    • Utilizing high-resolution structures of CEACAM5-specific domains

    • Example: A3-B3 domains targeted by tusamitamab show unique structural features compared to other family members

2. Comprehensive screening methodologies:

  • Multi-platform validation:

    • Testing candidate antibodies against cells expressing different CEACAM family members

    • Example: Flow cytometry analysis using transfected cells expressing individual CEACAM proteins

  • Sequential absorption:

    • Pre-absorbing antibodies with recombinant non-target CEACAM proteins

    • Testing residual binding to CEACAM5

3. Mutation and chimeric protein approaches:

  • Domain swapping experiments:

    • Creating chimeric proteins with domains exchanged between CEACAM family members

    • Identifying which domains confer specificity

  • Site-directed mutagenesis:

    • Converting specific amino acids in CEACAM5 to corresponding residues in other family members

    • Example: Mutation studies comparing region aa42-61 of CEACAM1 and CEACAM5

4. Negative selection strategies:

  • Counter-screening:

    • Eliminating antibody candidates showing binding to CEACAM1, CEACAM6, or CEACAM8

    • Prioritizing antibodies with >100-fold selectivity for CEACAM5

These approaches have successfully yielded highly specific antibodies like CC4 and tusamitamab that discriminate between CEACAM5 and other family members, enabling both precise research applications and targeted therapeutic development.

How can researchers optimize storage and handling of anti-CEACAM5 recombinant antibodies to maintain functionality?

Proper storage and handling are critical for maintaining antibody functionality. For anti-CEACAM5 recombinant antibodies:

1. Formulation considerations:

  • Buffer composition:

    • Typically supplied in phosphate-buffered saline (PBS), pH 7.4

    • Some formulations include 5-8% trehalose as a cryoprotectant

  • Protein stabilizers:

    • Addition of 0.1% human serum albumin (HSA) recommended for long-term storage after reconstitution

  • Preservatives:

    • Sodium azide (0.05-0.1%) may be included for prevention of microbial growth

    • Note: Azide can interfere with some applications (e.g., cell culture)

2. Storage conditions:

  • Temperature requirements:

    • Lyophilized form: Stable for 1 week at room temperature; store at -20°C to -80°C for 12 months

    • Reconstituted: Store at 4°C for 2-7 days or below -20°C for up to 3 months

  • Freeze-thaw considerations:

    • Limit freeze-thaw cycles to 3-5 maximum

    • Aliquot antibodies before freezing to avoid repeated freeze-thaw cycles

3. Reconstitution protocols:

  • Recommended procedure:

    • Reconstitute lyophilized antibody with sterile water to 100 μg/ml

    • Solubilize at room temperature for 30 minutes with occasional gentle mixing

  • Critical precautions:

    • Avoid vigorous vortexing (can cause denaturation)

    • Centrifuge briefly after reconstitution to collect solution

4. Quality control approaches:

  • Functionality testing:

    • Validate activity periodically using established applications (ELISA, flow cytometry)

    • Example: ELISA binding to recombinant CEACAM5 protein in the 0.1-50 ng/ml range

  • Appearance monitoring:

    • Check for visible precipitation or cloudiness before use

    • Filter if necessary using low protein-binding filters

These practices help ensure consistent performance of anti-CEACAM5 recombinant antibodies across experimental applications, enabling reliable research outcomes.

What are the emerging applications of anti-CEACAM5 antibodies in targeted cancer therapeutics?

Anti-CEACAM5 antibodies are being explored in multiple innovative therapeutic approaches:

1. Antibody-drug conjugates (ADCs):

  • Current development:

    • Tusamitamab ravtansine (SAR408701) combines anti-CEACAM5 antibody with a cytotoxic payload

    • Investigated for treatment of advanced nonsquamous non-small cell lung cancer and other solid tumors

  • Mechanistic considerations:

    • The epitope location on A3-B3 domains is important for invagination of the cell membrane, which affects ADC internalization and efficacy

2. Bispecific antibodies:

  • Design strategies:

    • Combining anti-CEACAM5 binding with immune effector cell engagement (T cells, NK cells)

    • Potential to simultaneously block inhibitory CEACAM5-CEACAM1 interactions while recruiting immune cells

3. CAR-T cell therapy:

  • Engineered T-cell approaches:

    • Development of chimeric antigen receptors targeting CEACAM5-specific epitopes

    • Challenges include addressing on-target/off-tumor effects due to CEACAM5 expression in normal tissues

4. Radioimmunotherapy:

  • Targeted radiation delivery:

    • Conjugation of anti-CEACAM5 antibodies with radioisotopes

    • Potential for treating microscopic disease and overcoming resistance mechanisms

5. Structure-guided antibody engineering:

  • Affinity optimization:

    • Using high-resolution structural data of epitope-paratope interfaces

    • Rational design of improved binding properties

  • Novel binding domains:

    • Development of alternative binding scaffolds (nanobodies, affibodies) targeting CEACAM5

These emerging applications leverage the specificity of anti-CEACAM5 antibodies while addressing the complex challenges of targeting tumor antigens that may also be expressed at lower levels in normal tissues.

How might advances in antibody engineering enhance the utility of anti-CEACAM5 recombinant antibodies?

Advanced antibody engineering approaches offer significant potential for enhancing anti-CEACAM5 antibodies:

1. Affinity maturation strategies:

  • Directed evolution approaches:

    • Phage display with error-prone PCR to generate variant libraries

    • Selection under increasingly stringent conditions

  • Computational design:

    • In silico modeling of antibody-antigen interactions based on structural data

    • Prediction of beneficial mutations to enhance binding

2. Format diversification:

  • Fragment-based formats:

    • Single-chain variable fragments (scFvs)

    • Fab fragments for improved tissue penetration

  • Multispecific designs:

    • Bispecific antibodies targeting CEACAM5 and immune checkpoint molecules

    • Dual-targeting of CEACAM5 and other tumor-associated antigens

3. Fc engineering:

  • Enhanced effector functions:

    • Afucosylation to increase antibody-dependent cellular cytotoxicity

    • Point mutations to modulate complement activation

  • Extended half-life:

    • Fc mutations that enhance FcRn binding

    • PEGylation or fusion to albumin-binding domains

4. Payload development for ADCs:

  • Novel linker chemistry:

    • Stimulus-responsive linkers (pH, protease-sensitive)

    • Increasing drug-to-antibody ratio while maintaining stability

  • Alternative payloads:

    • Exploring immunomodulatory agents beyond traditional cytotoxics

    • Site-specific conjugation approaches

5. Glycoengineering:

  • Controlled glycan profiles:

    • Uniform glycosylation patterns for consistent function

    • Removal of immunogenic glycan structures

  • Strategic glycan modification:

    • Addition of sialic acids to reduce immunogenicity

    • Afucosylation to enhance NK cell activation

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.