CDA6 Antibody

Shipped with Ice Packs
In Stock

Description

CD6 Antibody: Definition and Target

CD6 antibodies are monoclonal antibodies (mAbs) that bind to CD6, a type I transmembrane protein expressed on T cells and natural killer (NK) cells. CD6 modulates immune synapse formation, T-cell activation, and adhesion through interactions with its ligands, including CD166 (ALCAM) and CD318 .

Mechanisms of Action

CD6 antibodies exhibit dual roles depending on epitope and context:

  • Antagonistic Effects: Block CD6-CD166 interactions, reducing T-cell adhesion and migration (e.g., itolizumab in autoimmune diseases) .

  • Agonistic Effects: Trigger CD6 signaling, enhancing T-cell activation (e.g., UMCD6 in cancer immunotherapy) .

  • Cytotoxic Effects: CD6-ADC (antibody-drug conjugate) delivers monomethyl auristatin E (MMAE) to proliferating T cells, eliminating pathogenic cells in autoimmune uveitis and graft-versus-host disease (GVHD) .

Autoimmune Diseases

  • Multiple Sclerosis (MS): CD6 knockout mice show reduced Th1/Th17 cells and disease severity. UMCD6 suppressed experimental autoimmune encephalomyelitis (EAE) without T-cell depletion .

  • Rheumatoid Arthritis (RA): Itolizumab reduced joint inflammation in preclinical models by inhibiting pathogenic T-cell subsets .

  • Psoriasis: Anti-CD6 mAbs (e.g., MEM-98) mitigate inflammation by blocking CD6-CD166 interactions .

Cancer Immunotherapy

  • UMCD6 internalizes CD6 on NK and CD8+ T cells, upregulating NKG2D and granzyme B, enhancing tumor cell killing .

  • CD6-ADC eliminated malignant T cells in lymphoma models and reduced tumor size in breast cancer xenografts .

Clinical Trials

Clinical Trial IDPhaseConditionInterventionOutcome
NCT04128579IbSystemic lupus erythematosusItolizumabReduced nephritis activity
NCT03763318I/IIbAcute GVHDItolizumab + steroidsImproved survival
NCT05263999IIIAcute GVHDItolizumab + steroidsPending results

Source:

Challenges and Future Directions

  • Selectivity: CD6 is expressed on all T cells, but CD6-ADC spares resting T cells, targeting only proliferating pathogenic subsets .

  • Dual Roles: CD6 antibodies must balance immunosuppression (in autoimmunity) and immune activation (in cancer) .

  • Safety: Early trials show manageable toxicity, but long-term effects require monitoring .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
CDA6 antibody; DESD antibody; At4g29610 antibody; T16L4.120Cytidine deaminase 6 antibody; EC 3.5.4.5 antibody
Target Names
CDA6
Uniprot No.

Target Background

Function
This enzyme scavenges both exogenous and endogenous cytidine and 2'-deoxycytidine to facilitate uridine monophosphate (UMP) synthesis.
Database Links

KEGG: ath:AT4G29610

STRING: 3702.AT4G29610.1

UniGene: At.54547

Protein Families
Cytidine and deoxycytidylate deaminase family

Q&A

What is CD6 and what is its role in T-cell biology?

CD6 is a type I T-cell surface receptor that modulates antigen receptor signaling. Its activity is regulated through binding of its membrane proximal domain (domain 3) to cell surface ligands, with CD166 being a primary binding partner. CD6 plays a complex role in immune cell function, with evidence showing it can both restrain and promote activation of immune cells depending on the context. This dual role makes CD6 particularly interesting as a therapeutic target, as it acts as an immunomodulatory molecule that influences T-cell development, activation, and differentiation processes .

What are the major domains of CD6 and which domains do different antibodies target?

CD6 contains three extracellular SRCR (scavenger receptor cysteine-rich) domains, typically referred to as domains 1, 2, and 3. Most therapeutic and research antibodies target either domain 1 (the membrane distal domain) or domain 3 (the membrane proximal domain). Domain 1 antibodies include itolizumab, MEM98, UMCD6, MT605, and T12.1, while OX126 is an example of a domain 3 antibody. Domain specificity is crucial because each domain plays a different role in CD6 function - domain 3 primarily mediates binding to CD166, while domain 1 antibodies can modulate CD6 function through mechanisms that may not directly interfere with ligand binding .

How can I determine which epitope a CD6 antibody recognizes?

Epitope mapping of CD6 antibodies can be performed using point mutants of CD6 based on the crystal structure. For example, research has identified that the R77 residue is crucial for MT605 and T12.1 binding, while E63 is critical for itolizumab and MEM98 binding. Surface plasmon resonance (SPR) experiments with these mutants can effectively distinguish between antibodies targeting different epitopes. Additionally, competitive binding assays using flow cytometry with sequential antibody staining can determine if antibodies recognize overlapping epitopes. This was demonstrated when pre-incubation of cells with UMCD6 or MEM98 blocked binding of itolizumab, indicating overlapping specificities .

How should CD6 antibodies be used to study T-cell activation in vitro?

When designing experiments to study T-cell activation with CD6 antibodies, researchers should consider several methodological approaches:

  • Pre-incubation protocols: Isolate T cells and pre-incubate with the CD6 antibody before adding stimuli like anti-CD3/CD28. Include appropriate isotype controls to distinguish CD6-specific effects.

  • Activation readouts: Measure multiple parameters including cytokine production (especially IL-2), proliferation, surface activation markers, and signaling events.

  • Time course considerations: CD6 antibodies can have different effects at early versus late timepoints of T-cell activation, so include multiple timepoints in your experimental design.

  • Concentration titration: Different CD6 antibodies have varying affinities (e.g., itolizumab has lower affinity compared to other CD6 domain 1 mAbs), so determining optimal concentrations through titration is essential .

  • Context specificity: Test the antibody in both resting and pre-activated T-cell populations, as effects may differ substantially.

What methods can be used to measure the binding kinetics of CD6 antibodies?

Binding kinetics of CD6 antibodies can be measured using several techniques:

  • Surface Plasmon Resonance (SPR): This is the gold standard for measuring binding kinetics. Immobilize CD6 on a sensor chip and inject varying concentrations of antibody to determine association (ka) and dissociation (kd) rate constants, as well as equilibrium dissociation constant (KD). This approach revealed that itolizumab has lower affinity compared to other CD6 domain 1 mAbs .

  • Bio-Layer Interferometry (BLI): Similar to SPR but uses optical interference patterns instead of SPR, providing real-time measurement of binding kinetics.

  • Flow cytometry-based approaches: While less quantitative for kinetics, these can provide valuable information about binding to cell-surface CD6 under physiological conditions.

  • Isothermal Titration Calorimetry (ITC): Provides thermodynamic parameters in addition to binding constants.

When reporting binding kinetics, include the experimental temperature, buffer conditions, and whether the antibody was used as a whole IgG or as a fragment (Fab, scFv), as these factors can influence the measured parameters.

How do CD6 domain 1 versus domain 3 antibodies differ in their functional effects?

Domain 1 and domain 3 antibodies produce distinct functional effects that should be carefully considered when designing experiments:

ParameterDomain 1 Antibodies (e.g., itolizumab, MEM98)Domain 3 Antibodies (e.g., OX126)
CD166 BindingIndirectly hinder binding of immobilized CD166Directly block CD166 binding
IL-2 ProductionEffective at triggering IL-2 production in CD6-CAR expressing cellsSimilarly effective at triggering IL-2 production
T-cell SignalingModulate signaling without directly blocking ligand bindingDirectly interfere with CD6-CD166 interactions
Autoimmunity EffectsSuppress pathogenic Th1/Th17 responsesSimilar suppressive effects
Cancer ImmunotherapyEnhance killing by CD8+ T cells and NK cellsLess well-characterized for cancer applications

Both domain types can suppress strong immune responses, but through potentially different mechanisms. Domain 1 antibodies may be inferior to domain 3 antibodies in blocking multivalent CD166 interactions but can still effectively modulate CD6 function through alternative mechanisms .

How does the anti-CD6 antibody UMCD6 enhance cancer cell killing by immune cells?

UMCD6 enhances cancer cell killing through multiple distinct mechanisms affecting both CD8+ T cells and NK cells:

  • Receptor modulation: UMCD6 upregulates the expression of the activating receptor NKG2D and downregulates expression of the inhibitory receptor NKG2A on both NK cells and CD8+ T cells .

  • Enhanced cytotoxic machinery: Treatment with UMCD6 leads to concurrent increases in perforin and granzyme B production in cytotoxic lymphocytes .

  • CD6 internalization: Upon binding, UMCD6 causes rapid internalization of CD6, creating CD3+CD6- T cells that cannot bind CD6 ligands expressed on cancer cells. This internalization appears to be crucial for the enhanced cytotoxicity .

  • Cancer specificity: UMCD6 has been shown to augment killing of multiple cancer types, including breast, lung, and prostate cancer cells, more robustly than checkpoint inhibitors targeting the PD-1/PD-L1 axis in vitro .

  • In vivo efficacy: UMCD6 also enhances in vivo killing by human peripheral blood lymphocytes of human breast cancer xenotransplants in immunodeficient mice .

Importantly, these effects occur without requiring CD4+ T cell help, distinguishing the mechanism from some current checkpoint inhibitor therapies.

How can CD6 antibodies be used in models of autoimmune disease?

CD6 antibodies have shown significant therapeutic potential in autoimmune disease models through several mechanisms:

  • Disease model selection: CD6 antibodies have demonstrated efficacy in mouse models of multiple sclerosis, uveitis, and rheumatoid arthritis . When designing experiments, consider models where pathogenic Th1/Th17 responses are prominent.

  • Use of CD6-humanized mice: Since many therapeutic CD6 antibodies (like UMCD6 and itolizumab) target human CD6, researchers often use CD6-humanized mice where mouse CD6 is replaced with human CD6 .

  • Assessment parameters: When evaluating efficacy, measure:

    • Clinical disease scores

    • Histological assessment of target organ inflammation

    • Th1/Th17 cytokine production (IL-17, IFN-γ, TNF-α)

    • Inflammatory cell infiltration into target organs

    • T cell activation markers

  • Dosing considerations: CD6 antibodies do not typically cause lymphopenia, as CD6 is rapidly internalized upon antibody binding. Consider this when planning dosing schedules .

  • Combination approaches: Because CD6 antibodies have distinct mechanisms from other immunomodulatory agents, combination strategies may yield synergistic benefits.

How can CD6 antibodies be developed into antibody-drug conjugates (ADCs) for selective targeting of pathogenic T cells?

Development of CD6-targeted antibody-drug conjugates (CD6-ADCs) represents an innovative approach for selectively eliminating pathogenic T cells while sparing normal T cells:

  • Conjugation strategy: A proven approach involves conjugating an inactive form of monomethyl auristatin E (MMAE), a potent mitotic toxin, onto a monoclonal antibody against CD6. The key insight is that while CD6 is present on all T cells, only actively dividing (pathogenic) T cells are susceptible to the antimitotic MMAE-mediated killing .

  • Selectivity testing: To confirm selective killing, test the CD6-ADC against:

    • Activated vs. resting T cells in vitro

    • Antigen-specific T cells

    • Normal T cells in naive CD6-humanized mice

  • Disease model validation: CD6-ADCs have shown efficacy in preclinical models of autoimmune uveitis and graft versus host disease without significant detrimental effects on normal T cells .

  • Technical considerations:

    • Drug-to-antibody ratio (DAR) optimization

    • Linker stability in circulation

    • Internalization kinetics (CD6 undergoes rapid internalization)

    • Off-target toxicity assessment

This approach leverages the unique biology of CD6 and differences between resting and pathogenic T cells to achieve selective therapeutic targeting.

What computational approaches can be used to design improved CD6 antibodies?

Modern computational pipelines can significantly enhance the discovery and design of therapeutic CD6 antibodies:

  • Integrated approaches: Effective pipelines incorporate both physics-based and AI-based methods for the generation, assessment, and validation of developable candidate antibodies. This orthogonal approach can lead to more promising designs with fewer experimental iterations .

  • Key design tasks:

    • Traversing sequence landscapes to identify highly dissimilar antibodies that retain binding

    • Rescuing binding from escape mutations

    • Improving developability characteristics while maintaining binding properties

  • Few-shot experimental screening: Computational methods enable efficient screening of smaller candidate pools, potentially showing up to 54% of designs gaining binding affinity to new variants .

  • Structural considerations: Crystal structures of CD6 domains provide valuable templates for computational design. For domain 1 antibodies, key residues like R77 and E63 should be considered in the design process .

  • Validation protocols: Experimentally characterize binding against different antigen targets, developability profiles, and ideally obtain structural data (e.g., cryo-EM) of designed antibodies bound to CD6 .

How do the mechanisms of action differ between CD6 antibodies used for autoimmunity versus cancer immunotherapy?

CD6 antibodies exhibit distinct mechanisms of action in autoimmunity versus cancer immunotherapy contexts:

ParameterAutoimmunity ApplicationsCancer Immunotherapy Applications
Primary Target CellsCD4+ T cellsCD8+ T cells and NK cells
Desired OutcomeSuppression of pathogenic Th1/Th17 responsesEnhancement of cytotoxic responses
Key MechanismAmelioration of T cell infiltration into target organsUpregulation of activating receptors (NKG2D) and downregulation of inhibitory receptors (NKG2A)
CD6 Ligand RelevanceBlocking CD6-CD166 interactionsTargeting CD6-CD318 axis (CD318 is associated with cancer aggressiveness)
Cellular EffectAttenuation of inflammatory responsesIncreased perforin and granzyme B production
Combination PotentialWith conventional immunosuppressantsWith checkpoint inhibitors (potential synergy with PD-1/PD-L1 blockers)

This dual capability of anti-CD6 antibodies to control autoimmunity while enhancing anti-cancer responses represents a unique profile among immunomodulatory agents. Unlike many cancer immunotherapies that can trigger autoimmune side effects, CD6-targeted approaches may suppress rather than instigate autoimmunity while still promoting anti-tumor immunity .

What factors should be considered when using CD6 antibodies for flow cytometry?

When using CD6 antibodies for flow cytometry, researchers should consider several important factors:

  • Internalization dynamics: CD6 antibodies like UMCD6 cause rapid internalization of CD6, resulting in apparent loss of CD6 expression. This is not due to cell depletion but rather internalization of the CD6-antibody complex. To accurately assess CD6 expression:

    • Include time-course experiments

    • Use alternative antibody clones recognizing different epitopes

    • Consider evaluating both surface and intracellular CD6

  • Panel design considerations:

    • CD6 is highly expressed on T cells but also present on some B cell and NK cell subsets

    • Include markers to properly identify cell populations (CD3, CD4, CD8, CD56)

    • When studying CD6 modulation, include activation markers (CD25, CD69) and functional markers (NKG2D, NKG2A)

  • Fluorochrome selection:

    • Choose bright fluorochromes for detecting potentially low-level expression

    • Consider including a secondary antibody approach for signal amplification if needed

  • Controls and validation:

    • Include isotype controls matched to the CD6 antibody

    • Validate with CD6 knockout or CD6-humanized systems if available

    • Use multiple antibody clones to confirm findings

How can contradictory results with different CD6 antibodies be reconciled in research studies?

Contradictory results with different CD6 antibodies are common and can be reconciled through careful experimental design and interpretation:

  • Epitope-specific effects: Different antibodies target distinct epitopes on CD6 (e.g., domain 1 vs. domain 3, or different faces of domain 1). Map the epitope specificity of antibodies using mutants like R77A and E63A to understand potential functional differences .

  • Affinity considerations: Antibodies have different binding affinities (e.g., itolizumab has lower affinity than other domain 1 mAbs), which can impact functional outcomes especially at lower concentrations .

  • Format effects: The antibody format (whole IgG, F(ab')2, Fab) affects:

    • Valency (bivalent vs. monovalent binding)

    • Fc-mediated effects

    • Tissue penetration

    • Half-life

  • Experimental context: Results may differ between:

    • In vitro vs. in vivo experiments

    • Different cell types (primary T cells vs. cell lines)

    • Activation state of target cells

    • Presence of CD6 ligands in the experimental system

  • Reconciliation approach: When facing contradictory results:

    • Directly compare antibodies side-by-side under identical conditions

    • Perform dose-response experiments

    • Test multiple readouts (not just a single parameter)

    • Consider kinetic differences in responses

The pleiotropic effects of CD6 can lead to more than one interpretation of the effects of a CD6 mAb in a particular assay, making careful experimental design crucial .

What are promising combination approaches for CD6 antibodies in cancer immunotherapy?

Several promising combination approaches for CD6 antibodies in cancer immunotherapy warrant investigation:

  • Combination with checkpoint inhibitors: Since UMCD6 has demonstrated superior stimulation of cancer killing by lymphocytes in vitro compared to pembrolizumab or nivolumab, combination strategies targeting both CD6 and PD-1/PD-L1 may yield additive or synergistic effects due to their distinct mechanisms of action .

  • Integration with ADC technology: Combining the targeted approach of CD6 antibodies with potent cytotoxic payloads (as in CD6-ADCs) offers a promising direction, particularly for selectively eliminating pathogenic T cells in settings where T cells themselves can be malignant .

  • CD6 and NKG2D/NKG2A axis modulation: Since CD6 antibodies like UMCD6 upregulate NKG2D and downregulate NKG2A on both NK cells and CD8+ T cells, combining CD6 targeting with agents specifically targeting these receptors might amplify anti-tumor responses .

  • Bispecific approaches: Development of bispecific antibodies targeting CD6 and tumor-associated antigens could enhance the specificity of the immune response against cancer cells.

  • CAR-T cell engineering: Incorporating CD6-targeting domains into CAR-T cell designs might enhance their functionality and persistence in solid tumors.

These approaches should be systematically evaluated in preclinical models before advancing to clinical testing, with careful assessment of both efficacy and potential autoimmune complications.

How might computational approaches improve the next generation of CD6 antibodies?

Computational approaches offer several avenues to improve next-generation CD6 antibodies:

  • Epitope-focused optimization: Using the crystal structure of CD6, computational methods can design antibodies targeting specific epitopes (like R77 or E63 on domain 1) with optimized binding properties .

  • Developability improvements: Computational pipelines can identify and address potential developability issues while maintaining or enhancing therapeutic properties. This includes:

    • Reducing aggregation propensity

    • Optimizing thermal stability

    • Minimizing oxidation-sensitive residues

    • Improving manufacturability

  • Affinity maturation: In silico affinity maturation can enhance binding properties without introducing developability issues that often arise with traditional experimental approaches.

  • Cross-reactivity prediction: Computational methods can predict and minimize potential cross-reactivity with off-target proteins, enhancing safety profiles.

  • Species cross-reactivity engineering: For preclinical studies, designing antibodies that bind both human and mouse CD6 could facilitate translation from animal models to clinical applications.

  • Fc engineering: Computational approaches can optimize Fc domains for specific functions (enhanced ADCC, extended half-life, reduced immunogenicity) depending on the therapeutic goal.

These computational methods can significantly reduce the need for large-scale experimental screening, making the development process more efficient and potentially leading to antibodies with superior properties .

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 2025 TheBiotek. All Rights Reserved.