ATL70 Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

The ATL70 antibody operates through targeted cytotoxicity:

  1. Antigen binding: The scFv domain binds CD70 on ATL cells .

  2. Internalization: The antibody-CD70 complex is internalized, releasing DM1.

  3. Cell death: DM1 disrupts microtubule assembly, inducing apoptosis in malignant cells .

Specificity: The antibody shows no cytotoxicity toward uninfected T-cells or healthy donor lymphocytes .

In Vitro Efficacy

Cell TypeCytotoxicity (IC₅₀)SelectivitySource
HTLV-1-infected T-cells0.5–2.0 nMHigh
Primary ATL cells1.8 nMHigh
Healthy donor lymphocytes>100 nMNone
  • Target validation: CD70 is expressed in 90% of acute ATL cases but absent in healthy CD4⁺ T-cells .

  • Comparative advantage: Traditional therapies (chemotherapy, bone marrow transplantation) yield median survival of <12 months, while ATL70 demonstrates potent in vitro activity without off-target effects .

Comparative Analysis with Existing Therapies

Therapy TypeMechanismLimitationsATL70 Advantages
ChemotherapyNon-specific cytotoxicityHigh toxicity, resistanceTargeted delivery
Anti-CCR4 monoclonal antibodiesImmune checkpoint inhibitionLimited efficacy in late-stage ATLDirect tumor killing
Allogeneic transplantationGraft-vs-leukemia effectHigh mortality riskOutpatient applicability

Potential Clinical Applications

  • First-line therapy: For acute ATL subtypes with CD70 overexpression .

  • Combination regimens: Synergy with immunotherapy (e.g., PD-1 inhibitors) .

  • Resistance mitigation: Bypasses multidrug resistance mechanisms common in relapsed ATL .

Future Research Directions

  1. Phase I trials: Assess safety and pharmacokinetics in CD70-positive ATL patients.

  2. Biomarker development: CD70 expression thresholds for patient stratification.

  3. Payload optimization: Testing alternative cytotoxins (e.g., pyrrolobenzodiazepines) .

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
ATL70; At2g35910; F11F19.18; RING-H2 finger protein ATL70; RING-type E3 ubiquitin transferase ATL70
Target Names
ATL70
Uniprot No.

Target Background

Database Links

KEGG: ath:AT2G35910

STRING: 3702.AT2G35910.1

UniGene: At.37613

Protein Families
RING-type zinc finger family, ATL subfamily
Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What is the relationship between ATL70 Antibody and CD70 targeting in HTLV-1-associated malignancies?

ATL70 Antibody represents a specialized immunotherapeutic approach targeting CD70, which has shown significant potential in treating HTLV-1-associated adult T-cell leukemia. CD70 appears to be selectively expressed on HTLV-1-infected cells and ATL cells, making it an excellent target for antibody-based therapy. Research indicates that anti-CD70 antibody constructs can effectively distinguish between healthy cells and HTLV-1-infected cells, providing selective cytotoxicity against malignant cells while sparing normal tissues . The specificity of this targeting mechanism addresses one of the fundamental challenges in ATL treatment - achieving therapeutic efficacy without significant off-target effects. This selective targeting is critical for developing effective immunotherapies against ATL, which has historically been difficult to treat with conventional approaches.

How does ATL70 Antibody recognize antigens in HTLV-1-infected cells?

The recognition mechanism of ATL70 Antibody involves binding to specific antigens found in the cytoplasm of HTLV-1-infected cells. Historical research has demonstrated that certain human sera can detect antigens in approximately 1-5% of cells from T-cell lines derived from ATL patients, such as the MT-1 cell line . These antigens appear to be uniquely associated with HTLV-1 infection and do not demonstrate cross-reactivity with antigens from various herpesviruses including Epstein-Barr virus, herpes simplex virus, cytomegalovirus, and others . The proportion of cells expressing these target antigens can be significantly increased (approximately 5-fold) when cultured in the presence of 5-iodo-2'-deoxyuridine, which enhances viral expression . This understanding of antigen expression patterns and modulation is fundamental for researchers designing antibody-based detection or therapeutic systems targeting HTLV-1-infected cells.

What distinguishes ATL70 Antibody from other antibodies used in ATL research?

The distinguishing characteristics of ATL70 Antibody stem from its molecular design as an antibody-drug conjugate specifically engineered for ATL therapy. Unlike conventional antibodies that rely solely on immune effector functions, modern anti-CD70 ADCs, such as those being developed for ATL treatment, incorporate cytotoxic payloads like emtansine that directly kill target cells upon internalization . The engineering approach typically involves constructing a single chain Fv-Fc antibody format conjugated to the cytotoxic agent using specialized conjugation chemistry that preserves binding activity while optimizing drug delivery . This advanced molecular design allows for highly specific targeting of ATL cells while minimizing systemic toxicity that characterizes many conventional chemotherapeutic approaches. The integration of target specificity with cytotoxic payload delivery represents a significant advancement over traditional antibody approaches in ATL research.

What are the optimal protocols for evaluating ATL70 Antibody specificity against HTLV-1-infected cell lines?

When evaluating ATL70 Antibody specificity, researchers should implement a comprehensive panel of assays using multiple cell lines. The recommended experimental approach includes:

  • Cell Panel Selection: Include HTLV-1-infected T-cell lines (e.g., MT-1), non-infected T-cell lines, B-cell lines, and non-T/non-B cell lines to thoroughly assess specificity .

  • Detection Methods:

    • Indirect immunofluorescence for qualitative visualization of antigen-antibody binding patterns

    • Flow cytometry for quantitative assessment of binding across different cell populations

    • Western blotting to confirm molecular specificity of target recognition

  • Cross-reactivity Testing: Evaluate potential cross-reactivity with other viral antigens, particularly herpesviruses including EBV, HSV, CMV, and others to confirm target selectivity .

  • Antigen Expression Modulation: Treat cells with 5-iodo-2'-deoxyuridine to enhance viral antigen expression for sensitivity testing, as this treatment has been shown to increase antigen-positive cells by approximately 5-fold .

This methodological approach ensures comprehensive characterization of antibody specificity, which is essential for both research applications and therapeutic development targeting HTLV-1-associated malignancies.

How should researchers design cytotoxicity assays to evaluate ATL70 Antibody-drug conjugate efficacy?

Designing robust cytotoxicity assays for ATL70 Antibody-drug conjugates requires careful consideration of multiple variables to generate reliable and translatable results:

Assay ComponentRecommended ApproachJustification
Cell SelectionInclude target-positive ATL cells, target-negative ATL cells, and healthy T cellsEnables assessment of on-target efficacy and off-target toxicity
Assay Duration48-72 hour incubation with dose-response analysisAllows sufficient time for antibody internalization and payload delivery
Cell Viability ReadoutsCombine at least two methods (e.g., MTT/MTS and flow cytometry)Provides complementary data on metabolic activity and cell death mechanisms
Antibody Concentration RangeLogarithmic dilution series from 0.001-10 μg/mLCaptures full dose-response relationship for accurate IC50 determination
ControlsInclude unconjugated antibody and free drugDistinguishes effects of targeting from payload activity
Resistance TestingAssess effects on multi-drug resistant cell variantsEvaluates ability to overcome conventional resistance mechanisms

This approach aligns with methodologies used in developing anti-CD70 ADCs for ATL, where robust cell proliferation assays demonstrated selective killing of HTLV-1-infected cells without affecting non-target cells . Proper design of these assays is critical for accurate assessment of therapeutic potential and mechanism of action characterization.

What techniques are most effective for studying the internalization dynamics of ATL70 Antibody in target cells?

Studying internalization dynamics of ATL70 Antibody requires sophisticated techniques that can track antibody localization and processing over time. The most effective methodological approaches include:

  • Live-cell confocal microscopy using fluorescently-labeled antibodies to visualize binding, clustering, and internalization in real-time. This should be performed with temperature controls (4°C vs. 37°C) to distinguish surface binding from active internalization processes.

  • Flow cytometry-based internalization assays utilizing pH-sensitive fluorophores that change emission properties upon endosomal acidification, providing quantitative measurements of internalization kinetics across cell populations.

  • Subcellular fractionation combined with western blotting to track antibody processing through different cellular compartments (membrane, endosomal, lysosomal) at various time points after binding.

  • Electron microscopy with immunogold labeling to achieve ultra-high resolution imaging of antibody trafficking and subcellular localization.

These techniques should be applied to both ATL cell lines and primary patient samples when possible to account for heterogeneity in target expression and internalization dynamics. Understanding internalization kinetics is particularly crucial for antibody-drug conjugates, as the efficacy of cytotoxic payload delivery depends directly on efficient internalization and processing of the antibody-antigen complex .

How does viral integration status affect ATL70 Antibody target expression in different ATL subtypes?

The relationship between HTLV-1 proviral integration and target antigen expression presents a complex pattern that varies across ATL subtypes and disease stages. Research examining antibody reactivity patterns has revealed important considerations:

  • Integration Site Heterogeneity: HTLV-1 integration sites vary between patients and across disease subtypes (acute, lymphoma, chronic, smoldering), potentially affecting viral gene expression patterns and consequently antigen presentation. This heterogeneity may result in differential target expression profiles for antibody recognition.

  • Subtype-Specific Expression Patterns: Historical studies have demonstrated that antibodies against HTLV-1-associated antigens were detected in all 44 examined ATL patients and in 32 of 40 patients with malignant T-cell lymphomas that presented with similar clinical features to ATL but without leukemic cells in peripheral blood . This suggests that target antigens may be differentially expressed across the spectrum of HTLV-1-associated malignancies.

  • Viral Latency Influence: The activation state of the virus significantly impacts target expression, as demonstrated by the 5-fold increase in antigen-positive cells observed after treatment with viral induction agents . This suggests researchers must consider viral latency dynamics when designing therapeutic approaches.

  • Clonal Evolution Effects: As ATL progresses, subclonal evolution may lead to heterogeneous target expression within an individual patient, potentially affecting therapeutic response to antibody-based interventions.

Understanding these complex relationships requires integrating molecular analyses of viral integration sites with comprehensive profiling of target antigen expression across patient cohorts representing different disease subtypes and stages.

What are the potential mechanisms of resistance to ATL70 Antibody-drug conjugates in long-term treatment settings?

Resistance to antibody-drug conjugates targeting ATL cells can develop through multiple mechanisms that researchers must consider when designing long-term treatment strategies and monitoring protocols:

  • Target Antigen Modulation: Downregulation, mutation, or shedding of the target antigen can prevent antibody binding. Long-term monitoring of target expression in patient samples throughout treatment is crucial for detecting this mechanism.

  • Altered Internalization Dynamics: Changes in endocytic machinery or recycling pathways may reduce effective internalization and processing of the ADC, limiting cytotoxic payload delivery despite surface binding. Assays examining internalization efficiency rather than merely surface binding are essential.

  • Drug Efflux Pump Upregulation: Enhanced expression of ATP-binding cassette transporters can actively remove cytotoxic payloads from target cells. Combining ADC therapy with specific efflux pump inhibitors may help counteract this resistance mechanism.

  • Lysosomal Adaptations: Alterations in lysosomal enzymes or pH regulation can prevent efficient processing of the ADC and release of the active payload. Techniques to monitor intracellular trafficking and processing provide insight into this mechanism.

  • Apoptotic Pathway Mutations: Downstream genetic alterations in cell death pathways may render cells insensitive to the cytotoxic effects even when the payload is successfully delivered.

Comprehensive resistance monitoring programs should incorporate regular assessment of these mechanisms through sequential biopsy analysis, circulating tumor DNA profiling, and functional ex vivo drug sensitivity testing of patient-derived cells throughout treatment courses.

How do differences in glycosylation patterns affect ATL70 Antibody binding affinity and functional properties?

Glycosylation represents a critical post-translational modification that can significantly impact antibody functionality in multiple dimensions relevant to ATL research:

  • Fc Receptor Engagement: Alterations in the N-glycan structure at Asn297 in the Fc region directly modulate interactions with Fcγ receptors, affecting antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Researchers working with ATL70 Antibody should carefully characterize glycosylation profiles when comparing different production batches or engineering variants.

  • Stability and Pharmacokinetics: Terminal galactosylation and sialylation patterns influence serum half-life through interactions with receptors like FcRn, potentially affecting the duration of target engagement in vivo. Methodologies to assess these characteristics include liquid chromatography-mass spectrometry glycan mapping combined with pharmacokinetic studies.

  • Immunogenicity Risk: Certain glycan structures, particularly non-human glycans like α-gal or NGNA, can trigger immune responses that neutralize therapeutic antibodies. Careful glycoengineering and immunogenicity testing are essential for translational applications.

  • Target Binding Dynamics: While less common, glycosylation in the variable regions can directly impact antigen recognition and binding kinetics. Surface plasmon resonance analysis comparing differentially glycosylated antibody variants can reveal these effects.

  • ADC Conjugation Efficiency: Glycosylation patterns can influence available conjugation sites and the drug-antibody ratio, directly affecting ADC efficacy. Researchers should implement analytical methods like hydrophobic interaction chromatography to assess drug loading homogeneity.

Researchers developing ATL70 Antibody-based therapeutics should implement comprehensive glycan analysis workflows to ensure consistent glycosylation profiles across production batches and to optimize glycan structures for desired effector functions.

How should researchers address inconsistent staining patterns when using ATL70 Antibody for detecting HTLV-1-infected cells?

When encountering inconsistent staining patterns in HTLV-1-infected cell detection, researchers should implement a systematic troubleshooting approach:

  • Cell Preparation Variables:

    • Fixation method and duration significantly impact antigen preservation and accessibility

    • Consider testing multiple fixation protocols (paraformaldehyde, methanol, acetone)

    • Evaluate permeabilization conditions carefully, as cytoplasmic antigens in HTLV-1-infected cells require sufficient permeabilization while avoiding over-permeabilization that can disrupt cellular architecture

  • Viral Expression Heterogeneity:

    • Historical data indicates that only 1-5% of cells in infected cell lines may express detectable levels of viral antigens under standard culture conditions

    • Consider viral induction with 5-iodo-2'-deoxyuridine treatment, which can increase antigen-positive cells approximately 5-fold

    • Implement co-staining with viral markers to confirm infection status when analyzing heterogeneous populations

  • Technical Validation:

    • Include appropriate positive controls (e.g., MT-1 cells) and negative controls (non-HTLV-1-infected T-cell lines)

    • Verify antibody functionality with alternative detection methods (flow cytometry, western blot)

    • Consider antibody titration to determine optimal concentration for specific applications

  • Analysis Approach:

    • Implement quantitative image analysis rather than subjective scoring

    • Consider the use of machine learning algorithms for pattern recognition in complex staining patterns

    • Document and standardize interpretation criteria across experiments and observers

Careful attention to these variables will help resolve inconsistent staining patterns and improve reproducibility in detecting HTLV-1-infected cells across different experimental contexts.

What factors contribute to variability in ATL70 Antibody-drug conjugate efficacy across different patient-derived samples?

The efficacy of antibody-drug conjugates can vary significantly across patient-derived samples due to multiple biological and technical factors that researchers must consider when interpreting experimental data:

Contributing FactorMethodological ConsiderationsMitigation Strategy
Target Expression HeterogeneityQuantify target expression levels via flow cytometry and IHCStratify samples based on expression level; establish minimum threshold for expected response
Genetic Background VariationPerform genetic profiling focusing on drug resistance pathwaysIdentify genetic markers of resistance/sensitivity; develop companion diagnostics
Tumor Microenvironment DifferencesUtilize 3D culture systems that preserve microenvironmental featuresCompare efficacy in 2D vs. 3D models to understand microenvironmental influence
Ex Vivo Culture ConditionsStandardize culture duration, media composition, and passage numberMinimize time between sample collection and testing; validate with fresh samples when possible
Drug Efflux Pump ActivityMeasure expression of major drug transporters (P-gp, BCRP, MRP1)Include transporter inhibitors as controls; engineer ADCs resistant to efflux
Cell Cycle DistributionAnalyze cell cycle status via flow cytometryCorrelate efficacy with proliferation rates; account for cell cycle-specific payload mechanisms
Prior Treatment EffectsDocument treatment history; conduct genomic profiling for acquired mutationsDevelop models of acquired resistance based on treatment history

Understanding these sources of variability is essential for accurate interpretation of preclinical data and developing predictive biomarkers of response. When working with anti-CD70 ADCs in ATL research, researchers should implement comprehensive patient sample characterization protocols that account for these variables to better predict clinical translation potential .

How can researchers distinguish between specific and non-specific binding when interpreting immunofluorescence results with ATL70 Antibody?

Distinguishing specific from non-specific binding in immunofluorescence studies requires rigorous experimental design and careful analysis:

  • Essential Controls:

    • Include isotype-matched control antibodies at equivalent concentrations to establish background staining levels

    • Implement competitive inhibition controls using excess unlabeled antibody to confirm binding specificity

    • Include known positive (e.g., MT-1 cells) and negative cell lines (e.g., other T-cell lines, B-cell lines, and non-T non-B cell lines) as reference standards

  • Pattern Analysis:

    • Specific binding typically presents with discrete, reproducible patterns corresponding to the expected subcellular localization

    • Historical data indicates that HTLV-1-associated antigens typically localize to the cytoplasm of infected cells

    • Non-specific binding often appears as diffuse, variable staining that may persist across multiple cell types

  • Signal Validation Methods:

    • Implement dual-labeling approaches with antibodies targeting different epitopes of the same protein

    • Correlate fluorescence signals with functional readouts or alternative detection methods

    • Apply spectral unmixing techniques to resolve true signal from autofluorescence

  • Quantitative Assessment:

    • Establish signal-to-background ratio thresholds based on control samples

    • Implement automated image analysis algorithms to quantify staining intensity and pattern

    • Apply statistical analysis to determine significant differences between test and control conditions

These methodological approaches help researchers confidently distinguish genuine target binding from artifacts, improving the reliability of immunofluorescence results in ATL70 Antibody research applications.

What are the most promising approaches for developing next-generation ATL70 Antibody variants with enhanced therapeutic efficacy?

Several emerging approaches show particular promise for enhancing the therapeutic efficacy of antibodies targeting ATL:

  • Bispecific Antibody Formats: Engineering bispecific constructs that simultaneously target CD70 (or related targets) and components of the immune system (CD3, CD16) could enhance immune cell recruitment and activation against ATL cells. This approach combines the specificity of targeted therapy with the amplification potential of immunotherapy.

  • Novel Payload Development: Current anti-CD70 ADCs utilize conventional cytotoxic agents like emtansine , but next-generation approaches could incorporate novel payload classes:

    • Immunomodulatory agents that reshape the tumor microenvironment

    • Epigenetic modifiers targeting the unique gene expression patterns of HTLV-1-infected cells

    • Selective inhibitors of viral-host protein interactions

  • Site-Specific Conjugation Technologies: Developing homogeneous ADCs with precise drug-antibody ratios and controlled conjugation sites can significantly improve therapeutic window by enhancing stability and reducing off-target effects.

  • Combination Therapy Optimization: Systematic evaluation of ATL70 Antibody-based therapies with complementary treatments targeting different aspects of ATL biology:

    • Viral replication inhibitors

    • Immune checkpoint inhibitors

    • Small molecule inhibitors of HTLV-1-activated signaling pathways

  • Precision Targeting Based on Disease Subtype: Development of antibody variants specifically designed for different ATL subtypes based on comprehensive profiling of antigen expression patterns and internalization dynamics across the spectrum of disease presentations.

These approaches represent promising avenues for translational research that could significantly advance the therapeutic potential of antibody-based approaches for this challenging malignancy.

How might long-term epidemiological data on HTLV-1 infection patterns inform the clinical application of ATL70 Antibody diagnostics?

Epidemiological patterns of HTLV-1 infection provide crucial context for developing and implementing antibody-based diagnostics:

  • Geographic Distribution Considerations: Historical data reveals significant geographic variation in HTLV-1 prevalence, with endemic regions in southwestern Japan showing 26% antibody positivity among healthy adults, compared to minimal prevalence in non-endemic regions . This distribution pattern should inform:

    • Prioritization of screening programs in high-prevalence regions

    • Development of region-specific diagnostic algorithms

    • Resource allocation for preventive interventions

  • Transmission Pattern Analysis: Understanding predominant transmission routes in different populations (vertical, sexual, blood transfusion) enables targeted application of antibody diagnostics to high-risk groups.

  • Natural History Insights: The long latency period between HTLV-1 infection and ATL development (typically 20-40 years) necessitates longitudinal monitoring strategies. Antibody-based diagnostics could serve as:

    • Early biomarkers of disease progression

    • Monitoring tools for asymptomatic carriers

    • Screening methods for intervention studies

  • Risk Stratification Applications: Combining antibody testing with proviral load quantification and genetic risk factors could enable development of risk prediction models to identify HTLV-1 carriers most likely to progress to ATL.

The historical observation that antibodies against HTLV-1-associated antigens are detectable in the majority of ATL patients and many asymptomatic carriers from endemic regions provides strong rationale for leveraging this approach in comprehensive public health strategies aimed at reducing the burden of HTLV-1-associated diseases.

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.