CDA7 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CD7 as a Therapeutic Target

CD7 is a 40 kDa type I transmembrane glycoprotein belonging to the immunoglobulin superfamily (IgSF). It is expressed on T-cells, natural killer (NK) cells, and early hematopoietic progenitors . In oncology, CD7 is overexpressed in >95% of T-cell acute lymphoblastic leukemia (T-ALL) cases and subsets of acute myeloid leukemia (AML) , making it a high-priority target for immunotherapy.

Key Antibody Candidates

Three high-affinity anti-CD7 monoclonal antibodies (mAbs) have been developed:

  • J87: KD = 1.54 × 10⁻¹⁰ M (highest affinity)

  • G73 and A15: Lower internalization efficiency compared to J87

These mAbs were generated using hybridoma technology after immunizing mice with recombinant CD7 extracellular domains .

J87-Dxd

  • Structure: J87 mAb conjugated to deruxtecan (DXd) via a cleavable maleimide-GGFG peptide linker .

  • Mechanism: Internalizes upon binding CD7, releasing the topoisomerase I inhibitor DXd intracellularly .

  • Preclinical Efficacy:

    ParameterJurkat CellsCCRF-CEM Cells
    Apoptosis Induction82%78%
    IC₅₀5.1 nM6.3 nM
    In Vivo Survival (Day 24)80% survival vs. 0% in controls
  • Safety: No significant organ toxicity observed in heart, liver, or kidneys via H&E staining .

CD7-DE-vcMMAE

  • Design: Fc-engineered (S239D/I332E) ADC with monomethyl auristatin E (MMAE) .

  • Activity: EC₅₀ = 5–8 ng/mL in T-ALL cell lines; 70% tumor growth inhibition in xenografts .

Anti-CD7 CAR T-Cell Therapies

Clinical trials targeting CD7 in relapsed/refractory T-ALL show:

Therapy TypeTrial PhasePatients EnrolledCR RateNotable Toxicity
CD7 ADC (J87-Dxd)PreclinicalN/AN/ANone reported
CD7 CAR T-CellsPhase I/II5395.8%Grade 3–4 CRS (11%)

Mechanisms of Resistance and Limitations

  • Fratricide: CD7 expression on CAR T-cells leads to self-elimination. Solutions include CRISPR editing or protein blockers .

  • Antigen Escape: CD7-negative relapse observed in 15% of CAR T-cell recipients .

Comparative Analysis of CD7-Targeted Agents

FeatureADCs (e.g., J87-Dxd)CAR T-Cells (e.g., RD13-01)
Target PopulationCD7⁺ T-ALL/AMLR/R T-ALL/LBL
Response TimeDaysWeeks
ManufacturingScalablePatient-specific
DurabilityTransientLong-term (with HSCT bridge)

Future Directions

  • Combination Therapies: Pairing ADCs with checkpoint inhibitors to enhance cytotoxicity .

  • Bispecific Antibodies: Dual targeting of CD7 and CD3 to engage T-cell responses .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CDA7 antibody; DESC antibody; At4g29600 antibody; T16L4.110Cytidine deaminase 7 antibody; EC 3.5.4.5 antibody
Target Names
CDA7
Uniprot No.

Target Background

Function
CDA7 Antibody is an enzyme that scavenges exogenous and endogenous cytidine and 2'-deoxycytidine for the synthesis of uridine monophosphate (UMP).
Database Links

KEGG: ath:AT4G29600

STRING: 3702.AT4G29600.1

UniGene: At.54546

Protein Families
Cytidine and deoxycytidylate deaminase family

Q&A

What is CD7 and what cellular populations express this marker?

CD7 is a 40 kDa transmembrane, single-chain glycoprotein that belongs to the immunoglobulin gene superfamily. It plays an essential role in T-cell interactions and T-cell/B-cell interaction during early lymphoid development. The expression profile of CD7 includes:

  • Majority of immature and mature T-lymphocytes

  • Natural killer (NK) cells

  • Small subpopulation of normal B-cells

  • Certain malignant B-cells

  • Majority of mature peripheral T-cells

  • Post-thymic T-cells

  • Some myeloid cells

Notably, CD7 expression is conspicuously absent in adult T-cell Leukemia/Lymphoma and Sezary cells, making this an important differential diagnostic marker .

What are the validated applications for CD7 antibodies in laboratory research?

CD7 antibodies have been validated for multiple research applications with specific methodological considerations for each:

ApplicationValidated Cell Types/TissuesTechnical ParametersExpected Results
Western BlotMOLT-4 cell line, HepG2 cell line, peripheral blood lymphocytes0.5 μg/mL antibody concentration, reducing conditions, PVDF membrane35-40 kDa specific band
Flow CytometryPBMCs, T-cell lines (Jurkat, CCRF-CEM)Use with CD3e for co-stainingMembrane-positive staining on T-cells
ImmunocytochemistryFixed PBMCs15 μg/mL concentration, 3 hours at room temperatureCytoplasmic and plasma membrane localization
ImmunohistochemistryFFPE human thymus, tonsil, lymph node3 μg/mL concentration, heat-induced epitope retrievalMembranous staining pattern

These applications provide researchers with multiple approaches to study CD7 expression in various experimental contexts, with each method requiring specific optimization steps .

What is the biological function of CDA and what antibodies are available for its detection?

CDA (Cytidine Deaminase) is an enzyme that scavenges exogenous and endogenous cytidine and 2'-deoxycytidine for UMP synthesis. This enzymatic activity is critical for nucleotide metabolism pathways and has implications for drug metabolism, particularly for nucleoside analogs used in cancer treatment .

CDA antibodies for research include:

Antibody TypeApplicationsSpecies ReactivityImmunogen
Rabbit PolyclonalIHC-PHuman, MouseRecombinant Fragment Protein within Human CDA aa 1 to C-terminus

These antibodies have been validated for immunohistochemical applications, with recommended dilutions of 1:100 for paraffin-embedded tissues, such as gastric carcinoma xenografts .

How do researchers quantitatively assess CD7 antibody affinity and what parameters indicate superior candidates for therapeutic development?

Assessing CD7 antibody affinity requires rigorous biophysical characterization. Researchers typically employ Surface Plasmon Resonance (SPR) techniques such as Biacore assays to determine binding kinetics:

Antibody CloneAssociation Rate (ka)Dissociation Rate (kd)Equilibrium Dissociation Constant (KD)
J87Not specified in dataNot specified in data1.54 × 10⁻¹⁰ M
G73Not specified in dataNot specified in data~10⁻¹⁰ M
A15Not specified in dataNot specified in data~10⁻¹⁰ M

Among the tested anti-CD7 antibodies, J87 demonstrated the highest affinity with a KD of 1.54 × 10⁻¹⁰ M, making it a superior candidate for therapeutic applications. For SPR analysis, the extracellular domain of CD7 (CD7-mFc) is typically immobilized on CM5 chips using amine coupling, followed by injection of antibodies in a 2-fold dilution series across the chip .

The equilibrium dissociation constant (KD) values in the 10⁻¹⁰ M range indicate high-affinity interactions, with lower values representing stronger binding. When developing therapeutic antibodies, researchers should prioritize clones with KD values <10⁻⁹ M and evaluate internalization efficiency in parallel .

What methodologies are most effective for evaluating CD7 antibody internalization kinetics in T-cell leukemia models?

Antibody internalization is a critical parameter for developing effective antibody-drug conjugates (ADCs). The following methodological approach provides a robust assessment of internalization kinetics:

  • Cell preparation:

    • Culture CD7-expressing cell lines (e.g., CCRF-CEM, Jurkat) in appropriate media

    • Harvest cells in exponential growth phase (viability >95%)

    • Prepare cell suspensions at 1 × 10⁶ cells/mL

  • Time-course evaluation:

    • Incubate cells with anti-CD7 antibodies at 0h, 1h, 2h, 4h, and 6h timepoints

    • Maintain consistent temperature (37°C) and antibody concentration

  • Detection methods:

    • Flow cytometry: Stain with FITC-conjugated secondary antibody to quantify remaining surface antibody

    • Confocal microscopy: Use fluorescently-labeled antibodies to visualize intracellular localization

This approach revealed that among three tested anti-CD7 mAbs (J87, G73, and A15), J87 demonstrated superior internalization efficiency, which correlated with its higher binding affinity. This combined profile of high affinity and efficient internalization made J87 the optimal candidate for ADC development .

How can researchers develop and validate a CD7-targeting antibody-drug conjugate for T-cell malignancies?

The development of anti-CD7 ADCs follows a systematic process that combines antibody engineering with drug conjugation technology:

Development StageMethodological ApproachCritical Parameters
Antibody SelectionHybridoma technology; expression and purification of CD7 extracellular domainAffinity (KD), internalization efficiency
Antibody CharacterizationSDS-PAGE analysis, immunofluorescence, Biacore assayPurity, specificity, binding kinetics
Linker-Drug SelectionConjugation of cytotoxic payload (e.g., DXd) via cleavable linkerDrug-to-antibody ratio (DAR), linker stability
In Vitro ValidationCell binding, internalization, cytotoxicity against CD7+ cell linesIC50 values, specificity ratio
In Vivo TestingXenograft models using CD7+ T-ALL cell linesTumor growth inhibition, toxicity profile

A recent study successfully generated J87-Dxd, an anti-CD7 ADC using the topoisomerase I inhibitor Deruxtecan (DXd) conjugated to J87 via a cleavable maleimide-GGFG peptide linker. This ADC demonstrated potent cytotoxicity against CD7-expressing T-ALL cell lines with an IC50 of 6.3 nM against CCRF-CEM cells, highlighting the therapeutic potential of this approach .

What are the optimal conditions for CD7 immunohistochemistry in clinical and research specimens?

Achieving consistent and specific CD7 immunostaining requires careful optimization of multiple parameters:

Protocol ComponentRecommended ConditionsTechnical Considerations
Tissue PreparationFFPE sections, 4-5 μm thicknessConsistent fixation time (12-24h) in 10% neutral buffered formalin
Antigen RetrievalHeat-induced epitope retrieval using Antigen Retrieval Reagent-BasicCritical for optimal staining; typically performed at 95-100°C for 20 minutes
Primary Antibody3 μg/mL for polyclonal or 1:100 dilution for monoclonal (clone LP15)Overnight incubation at 4°C yields optimal signal-to-noise ratio
Detection SystemHRP-DAB Cell & Tissue Staining KitUse with appropriate species-specific secondary antibody
CounterstainHematoxylinLight counterstaining preserves visualization of membranous CD7 staining
ControlsPositive: Human thymus or tonsil; Negative: Isotype controlEssential for validating staining specificity

When performing CD7 IHC, specific staining is primarily localized to the plasma membrane of T-cells. Common tissues used as positive controls include thymus, tonsil, and lymph node, where distinct T-cell zones should show clear membrane positivity .

How should researchers address discrepancies between CD7 detection methods when characterizing novel T-cell populations?

When discrepancies arise between different CD7 detection methods, a systematic troubleshooting approach is essential:

  • Method-specific considerations:

    • Flow cytometry detects native conformation on viable cells

    • IHC visualizes fixed epitopes with spatial context

    • Western blot identifies denatured protein by molecular weight

  • Validation strategy:

    • Employ multiple antibody clones targeting different CD7 epitopes

    • Include known positive controls (MOLT-4, CCRF-CEM) and negative controls (Raji)

    • Compare results across multiple techniques using standardized samples

    • Quantify expression levels rather than relying on binary positive/negative classification

  • Discrepancy resolution:

    • For weak IHC staining with positive flow cytometry: Optimize antigen retrieval conditions

    • For aberrant molecular weight in Western blot: Investigate glycosylation variations (CD7 typically appears at 35-40 kDa)

    • For heterogeneous expression: Increase sample size and evaluate at single-cell resolution

This comprehensive approach allows researchers to reconcile conflicting results and develop a more accurate understanding of CD7 expression in complex cellular populations .

What strategies can overcome common technical challenges when using CDA antibodies for detection in tissue specimens?

Researchers working with CDA antibodies may encounter several technical challenges that require specific optimization approaches:

ChallengeOptimization StrategyTechnical Rationale
Background StainingExtend blocking time (60 min with 5% BSA); Use mouse-on-mouse blocking for mouse tissuesReduces non-specific binding and endogenous biotin interference
Weak SignalTest multiple dilutions (1:50-1:200); Extend primary antibody incubation timeOptimization may be tissue-dependent based on CDA expression levels
Inconsistent ResultsUse positive control (gastric carcinoma tissue) in each experimentServes as internal quality control for staining procedure
Cross-ReactivityValidate with genetic knockdown controlsConfirms specificity of antibody binding

When working with CDA antibody ab137605, immunohistochemical analysis has been successfully performed on paraffin-embedded gastric carcinoma xenografts at a 1:100 dilution. When optimizing new tissue types, researchers should conduct titration experiments with dilutions ranging from 1:50 to 1:200 to determine optimal signal-to-noise ratio for their specific application .

How does CD7 antibody staining complement other T-cell markers in comprehensive immunophenotyping panels?

Comprehensive T-cell immunophenotyping requires strategic combinations of markers to accurately identify cell subsets and activation states:

Cell TypeKey Marker CombinationRole of CD7
T LymphocytesCD3+CD7+CD7 appears earlier than CD3 during T-cell development
Natural Killer CellsCD3-CD7+CD56+CD7 helps distinguish NK cells from other lymphocyte populations
T-ALLCD7+CD3+ variableCD7 is consistently expressed in T-ALL even when other T-cell markers may be lost
ATLLCD7-CD3+CD4+Loss of CD7 is characteristic of adult T-cell leukemia/lymphoma

When designing multicolor flow cytometry panels, CD7 antibodies can be effectively paired with CD3 to provide a comprehensive assessment of T-cell populations. In research settings, including CD7 alongside lineage markers (CD3, CD4, CD8) and activation markers (CD25, CD69) enables more precise characterization of T-cell functionality .

The distinctive expression pattern of CD7 across different T-cell malignancies makes it a valuable component of diagnostic panels, particularly for distinguishing T-ALL (typically CD7+) from other T-cell leukemias where CD7 expression may be downregulated or absent .

What genomic and post-translational modifications influence CD7 expression and antibody recognition in hematologic malignancies?

CD7 expression in hematologic malignancies is regulated through multiple mechanisms that can affect antibody recognition:

  • Transcriptional regulation:

    • Promoter methylation can silence CD7 gene expression in some T-cell malignancies

    • Transcription factor dysregulation in leukemic cells can alter CD7 expression levels

  • Post-translational modifications:

    • Glycosylation variations: CD7 is a glycoprotein with variable glycosylation patterns

    • Phosphorylation status may affect protein conformation and epitope accessibility

    • Proteolytic processing can generate variant forms with altered antibody recognition

  • Methodological implications:

    • Antibodies targeting different epitopes may show discordant results based on post-translational modifications

    • Detection of glycosylated forms results in the 35-40 kDa band observed in Western blot analysis

    • Certain fixation methods may alter glycan structures, affecting antibody binding

Understanding these modifications is crucial when interpreting CD7 detection results, particularly when comparing findings across different methodological platforms or patient samples .

How can CDA antibody-based research contribute to understanding chemotherapeutic resistance mechanisms?

CDA (Cytidine Deaminase) plays a crucial role in nucleoside metabolism and has significant implications for resistance to nucleoside analog drugs:

  • Research applications:

    • Quantification of CDA expression in tumor samples before and after treatment

    • Correlation of CDA levels with response to nucleoside analog drugs (e.g., gemcitabine, cytarabine)

    • Identification of regulatory mechanisms controlling CDA expression in resistant cells

  • Methodological approach:

    • IHC analysis of CDA expression in paired sensitive/resistant tumor samples

    • Western blot quantification of CDA protein in cell line models

    • Integration with functional enzyme activity assays to correlate protein levels with catalytic function

  • Clinical implications:

    • Potential use as a predictive biomarker for response to nucleoside analog therapies

    • Identification of patients who might benefit from CDA inhibitor combination strategies

    • Development of personalized treatment approaches based on CDA expression profile

CDA antibodies provide researchers with tools to investigate one of the key resistance mechanisms for important classes of chemotherapeutic agents. By combining CDA protein detection with functional studies, researchers can develop a more comprehensive understanding of drug resistance pathways and potential therapeutic strategies to overcome them .

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.