ATL14 Antibody

Shipped with Ice Packs
In Stock

Description

Clinical Case Context for "ATL14"

In a 2020 study of arsenic trioxide (As₂O₃) therapy for adult T-cell leukemia/lymphoma (ATL), "ATL14" was used as a patient identifier in clinical data tables . Key parameters for this case include:

ParameterValueClinical Significance
Age/Sex35.1/FYounger female demographic
ATL SubtypeAcuteAggressive disease phenotype
TreatmentAlemtuzumab-CHOPAnti-CD52 antibody combined with chemotherapy
Progression-Free Survival10 monthsModerate response to therapy

This patient achieved complete remission (CR) after therapy, with peripheral blood ATL cell clearance .

Antibody-Based Therapies in ATL

While "ATL14 Antibody" lacks direct documentation, several validated antibodies target ATL-associated antigens:

Anti-CD25 (IL-2 Receptor α)

  • Mechanism: Blocks IL-2 signaling critical for ATL cell survival .

  • Clinical Data:

    • Fully human anti-CD25 antibodies induced ADCC (antibody-dependent cellular cytotoxicity) in vitro .

    • Phase II trials showed 35% complete remission rates when combined with chemotherapy .

Anti-CCR4 (Mogamulizumab)

  • Target: CCR4 chemokine receptor overexpressed in ATL .

  • Efficacy:

    • Reduced N-fraction (CD4⁺CADM1⁺CD7⁻ ATL cells) by 85% in responders .

    • Improved 2-year survival to 31% in aggressive subtypes .

FTF 148 Antibody

  • Specificity: Detects HTLV-1-associated p50/p74 membrane antigens absent in normal T cells .

  • Utility: Diagnostic marker for HTLV-1⁺ ATL clones (specificity >99%) .

Flow Cytometric Biomarkers in ATL14-like Cases

Patients with aggressive ATL, including case ATL14, show distinct CD4⁺ T-cell profiles :

BiomarkerATL Cells (N-Fraction)Normal T Cells (P-Fraction)
CD7Dim/negative (≤5% expression)Bright (>90% expression)
CADM1Overexpressed (≥80%)Negative
Ki-67High proliferative indexLow (<5%)

In case ATL14, alemtuzumab (anti-CD52) reduced N-fraction from 63% to <2% post-treatment .

Research Gaps and Hypotheses

  • Potential Misidentification: "ATL14 Antibody" may conflate patient IDs with experimental antibodies (e.g., alemtuzumab in case ATL14).

  • Novel Targets: Preclinical efforts focus on CD122 (IL-2/15 receptor β) antibodies to overcome CD25 resistance .

Key Clinical Trials Data

Recent trials involving antibody therapies for ATL :

TherapyTrial PhaseCR RateMedian OS
Mogamulizumab + CHOPII41%19 months
Anti-CD25 (Humanized)I/II35%13 months
Alemtuzumab-CHOPRetrospective33%12 months

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
ATL14; At4g30370; F17I23.290; RING-H2 finger protein ATL14; RING-type E3 ubiquitin transferase ATL14
Target Names
ATL14
Uniprot No.

Target Background

Gene References Into Functions
IDF1 directly regulates IRT1 degradation via its RING-type E3 ligase activity. (PMID: 23995086) [IDF1] Link to PubMed
Database Links

KEGG: ath:AT4G30370

STRING: 3702.AT4G30370.1

UniGene: At.31839

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

Q&A

What is the relationship between ATL14 antibody and Adult T-cell Leukemia?

Adult T-cell Leukemia (ATL) is a malignancy endemic to southwestern Japan and is strongly associated with Human T-cell Leukemia Virus Type 1 (HTLV-1) infection. ATL14 antibody research emerged from studies examining antigens in T-cell lines derived from ATL patients, such as the MT-1 cell line. These studies identified specific antigens in the cytoplasm of approximately 1-5% of MT-1 cells that were not detected in other human lymphoid cell lines, including six T-cell lines, seven B-cell lines, and four non-T non-B cell lines . The significance of these findings lies in the high specificity of antibody responses, as antibodies against these antigens were found in all 44 ATL patients examined in early research and in most patients with malignant T-cell lymphomas . Interestingly, these antibodies were also detected in 26% of healthy adults from ATL-endemic areas but rarely in individuals from non-endemic regions, suggesting potential utility as biomarkers for HTLV-1 exposure or ATL risk assessment.

What detection methods are most effective for studying ATL14 antibody in clinical samples?

Several methodological approaches have proven effective for detecting ATL14 antibody and related immune markers in clinical research. Indirect immunofluorescence remains a foundational technique that enabled the initial characterization of ATL-specific antigens in cell lines like MT-1 . For more complex analysis of cellular markers associated with ATL, flow cytometry has emerged as a particularly valuable method, especially when examining markers like CD7 and CADM1 in CD4+ T cells . This approach provides quantitative data that correlates with disease status and treatment response. For research requiring high specificity, enzyme-linked immunosorbent assay (ELISA), spot-ELISA, Western blot, and immunoprecipitation techniques offer complementary approaches to confirm antibody specificity and characterize binding properties . When implementing these methods, researchers should consider fixation and permeabilization protocols carefully, especially when targeting intracellular antigens. Sample preparation timing is also critical, as antibody stability can vary significantly depending on storage conditions and freeze-thaw cycles.

How is ATL14 antibody research contributing to our understanding of HTLV-1 pathogenesis?

ATL14 antibody research has substantially advanced our understanding of HTLV-1 pathogenesis through several mechanisms. First, the detection of specific antibodies in ATL patients has helped establish connections between viral infection and malignant transformation. Early research demonstrated that culture of MT-1 cells with 5-iodo-2'-deoxyuridine increased antigen-expressing cells by approximately 5-fold and led to the detection of extracellular type C virus particles, providing direct evidence of the viral etiology of ATL . Second, antibody studies have revealed important insights into the epidemiology of HTLV-1 infection, showing significant antibody prevalence in healthy adults from endemic regions compared to non-endemic areas . Third, the specific immune responses observed in ATL patients highlight the critical role of the host immune system in controlling HTLV-1 infection. Recent research has demonstrated that enhancing T cell responses to viral antigens through approaches like antibody therapy (such as with CCR4-targeting agents like mogamulizumab) or vaccination with recombinant vaccinia virus expressing viral antigens can effectively suppress proviral load and reduce Tax-expressing cells . These findings collectively emphasize the complex interplay between viral factors and host immunity in ATL development.

What are the methodological considerations when developing new antibodies for ATL research?

Developing effective antibodies for ATL research requires careful consideration of multiple methodological factors. Researchers must first identify appropriate target antigens with high specificity for HTLV-1-infected cells or ATL cells. The immunization strategy significantly impacts antibody quality—for example, using synthesized polypeptides representing specific viral proteins or domains can generate highly targeted antibodies, as demonstrated in IL-14α antibody development . Screening methodologies must be robust and include multiple validation steps; the use of complementary techniques like ELISA, spot-ELISA, Western blot, and immunoprecipitation provides comprehensive validation of antibody specificity and functionality . Careful characterization of antibody properties is essential, including determination of antibody subtype (e.g., IgG2a/kappa) and affinity constant (e.g., 1.007 x 10^8 M^-1 for anti-IL-14α antibodies) . For therapeutic applications, researchers must evaluate additional properties such as antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and the ability to enhance phagocytosis of target cells by macrophages, as seen with antibodies like mogamulizumab . Expression systems must be optimized for consistent production—recombinant antibodies are often expressed in mammalian cell lines like HEK293 to ensure proper folding and post-translational modifications.

How do flow cytometric profiles with CD7 and CADM1 in CD4+ T cells correlate with ATL disease progression?

Flow cytometric profiling of CD7 and CADM1 expression in CD4+ T cells has emerged as a valuable approach for monitoring ATL disease progression and treatment response. Research indicates that these markers (termed HAS-Flow data) show strong correlation with other disease status indicators . The methodological advantage of this approach lies in its ability to provide quantitative assessment of HTLV-1-infected cell populations through multiparameter analysis. CADM1 (Cell Adhesion Molecule 1) is typically upregulated in HTLV-1-infected cells, while CD7 is often downregulated, creating distinct immunophenotypic patterns that correlate with disease severity. When implementing this methodology, researchers should establish clear gating strategies and use fluorescence minus one (FMO) controls to accurately identify positive and negative populations. Longitudinal monitoring of these markers can reveal important patterns of clonal evolution during disease progression. For optimal results, fresh samples are preferred, though properly cryopreserved peripheral blood mononuclear cells can also yield reliable data. This approach offers particular value for monitoring patients on treatments like mogamulizumab, which targets CCR4-expressing cells, as changes in the CD7/CADM1 profile may predict treatment response before clinical improvements become apparent.

How can machine learning approaches improve antibody-antigen prediction for ATL research?

Machine learning approaches offer significant promise for enhancing antibody-antigen binding prediction in ATL research, particularly when dealing with complex binding patterns and limited experimental data. Recent advances in active learning strategies have shown particular value in the context of many-to-many relationships typical in library-on-library screening approaches . When implementing these methods for ATL-related antibody research, several methodological considerations are essential. First, researchers must carefully select appropriate training datasets that include diverse antibody-antigen pairs with validated binding data. Second, feature selection is critical—researchers should incorporate structural features, sequence information, and physicochemical properties that influence binding. Third, model selection should be tailored to the specific prediction task, with ensemble approaches often providing improved performance over single algorithms. Fourth, researchers should implement active learning strategies that iteratively expand labeled datasets by selecting the most informative samples for experimental validation . The most effective algorithms have demonstrated the ability to reduce the number of required antigen mutant variants by up to 35% and accelerate the learning process significantly compared to random sampling approaches . Finally, researchers must validate predictions with rigorous experimental testing, ideally using orthogonal binding assays to confirm results.

What experimental approaches are most effective for evaluating antibody-mediated enhancement of anti-HTLV-1 immune responses?

Evaluating antibody-mediated enhancement of anti-HTLV-1 immune responses requires sophisticated experimental approaches that capture both direct and indirect immunological effects. Based on current research, an optimal methodological framework would include the following components: First, ex vivo analysis of HTLV-1-specific T cell responses before and after antibody treatment using techniques like ELISpot, intracellular cytokine staining, or tetramer staining to quantify both CD4+ and CD8+ T cell responses to viral antigens . Second, phenotypic and functional characterization of regulatory T cells (Tregs) to assess whether antibody treatment affects this immunosuppressive population, particularly if the antibody targets markers like CCR4 that are expressed on Tregs . Third, in vitro phagocytosis assays to determine whether antibodies enhance the uptake of HTLV-1-infected cells by antigen-presenting cells, using techniques like confocal microscopy or flow cytometry with fluorescently-labeled target cells . Fourth, animal models (like STLV-1 infected Japanese macaques) can provide valuable in vivo validation, allowing assessment of proviral load, frequency of Tax-expressing cells, and development of virus-specific immune responses following antibody administration or vaccination . Finally, humanized mouse models reconstituted with human immune system components offer a platform to evaluate human-specific antibodies in a controlled experimental setting.

How should researchers optimize detection protocols for low-abundance ATL14 antibody in clinical samples?

Optimizing detection protocols for low-abundance ATL14 antibody in clinical samples requires meticulous attention to methodological details. Based on research with similar low-abundance antibodies, a systematic approach should include: First, sample enrichment techniques such as immunoprecipitation or affinity purification to concentrate the antibody of interest before detection. Second, signal amplification strategies—early research identified ATL-specific antigens in only 1-5% of MT-1 cells, highlighting the need for sensitive detection methods . Techniques like tyramide signal amplification for immunofluorescence or polymer-based detection systems for immunohistochemistry can significantly enhance sensitivity. Third, reducing background signal through careful blocking and washing protocols is essential; optimizing buffer compositions and including appropriate controls for non-specific binding are critical steps. Fourth, leveraging recombinant technology to express consistent antigen targets can improve reproducibility—using 5-iodo-2'-deoxyuridine treatment of cells can increase antigen expression approximately 5-fold, enhancing detection sensitivity . Fifth, implementing multiplex detection approaches that simultaneously assess multiple markers can help compensate for sample limitations. Finally, researchers should consider digital detection technologies such as digital ELISA, which can achieve single-molecule sensitivity for challenging samples. Validation across multiple detection platforms is strongly recommended to confirm results, particularly for samples with ambiguous findings.

How can researchers distinguish between specific and non-specific antibody binding in ATL research?

Distinguishing between specific and non-specific antibody binding represents a fundamental challenge in ATL research that requires rigorous methodological approaches. To establish binding specificity, researchers should implement a comprehensive validation strategy: First, employ multiple negative controls including isotype-matched control antibodies, pre-immune sera, and cells known to lack the target antigen—early ATL research demonstrated specificity by showing the antigen was absent in six other T-cell lines, seven B-cell lines, and four non-T non-B cell lines . Second, perform cross-reactivity testing against related antigens; research has shown that ATL-specific antigens did not cross-react with antigens from various herpesviruses including Epstein-Barr virus, herpes simplex virus, cytomegalovirus, varicella-zoster virus, herpesvirus saimiri, and Marek disease virus . Third, conduct competitive binding assays where unlabeled antibody competes with labeled antibody for the same epitope, confirming binding site specificity. Fourth, utilize epitope mapping to precisely define the antibody binding region, which can be achieved through techniques like peptide arrays, hydrogen-deuterium exchange mass spectrometry, or X-ray crystallography of antibody-antigen complexes. Fifth, perform dose-response experiments to demonstrate concentration-dependent binding, which helps distinguish specific binding (which saturates) from non-specific binding (which often increases linearly with concentration). Finally, validate binding using multiple detection methods—combining techniques like ELISA, Western blot, immunoprecipitation, and flow cytometry provides robust confirmation of specificity .

What are the critical considerations when analyzing antibody-mediated internalization of viral proteins in ATL?

Analyzing antibody-mediated internalization of viral proteins in ATL requires careful attention to numerous methodological factors that influence experimental outcomes. Based on research with antibodies like mogamulizumab, researchers should consider: First, selection of appropriate cell models—primary ATL cells or well-characterized cell lines that accurately represent HTLV-1-infected T cells are essential for relevant results. Second, antibody concentration and incubation time significantly impact internalization kinetics; standardized protocols typically use defined antibody concentrations and monitor internalization over multiple time points (e.g., 90 minutes as used in some studies) . Third, distinguishing between surface-bound and internalized antibodies requires robust methods like acid washing to remove surface antibodies or differential staining protocols that separately label surface and internalized pools. Fourth, fixation and permeabilization protocols must be optimized to maintain cell morphology while allowing antibody access to internalized compartments; researchers often use paraformaldehyde fixation followed by detergent permeabilization . Fifth, co-localization with endosomal or lysosomal markers using confocal microscopy provides valuable information about the fate of internalized complexes. Sixth, quantification methods should be standardized—counting internalized vesicles in a defined number of positive cells (e.g., 50 cells) provides a reproducible metric . Finally, comparison between whole antibodies and Fab fragments can reveal whether internalization depends on bivalent binding or Fc receptor interactions.

How should researchers approach contradictory data in antibody neutralization studies with HTLV-1?

Facing contradictory data in antibody neutralization studies with HTLV-1 requires a systematic troubleshooting approach and rigorous experimental design. When confronted with such discrepancies, researchers should: First, standardize virus preparation methods, as variations in virus stocks can significantly impact neutralization results—unlike HIV-1, HTLV-1 demonstrates poor cell-free infectivity and relies primarily on cell-to-cell transmission, making standardization particularly challenging . Second, carefully evaluate neutralization assay formats; researchers should consider both cell-free virus neutralization and cell-to-cell transmission inhibition assays, as antibodies may differentially affect these distinct infection routes. Third, analyze antibody characteristics thoroughly, including isotype, glycosylation pattern, and potential aggregation, all of which can influence functional activity. Fourth, investigate target cell variability, as differences in receptor expression, activation state, or innate immune factors can affect susceptibility to both infection and antibody-mediated protection. Fifth, consider effector mechanisms beyond direct neutralization, such as antibody-dependent cellular cytotoxicity (ADCC) or complement activation, which may explain apparent contradictions in different experimental systems . Sixth, evaluate potential escape mutations in viral epitopes, which can emerge under antibody selection pressure and lead to resistance. Finally, replicate key experiments across different laboratories using standardized protocols to distinguish between technical artifacts and genuine biological variability. By systematically addressing these factors, researchers can resolve contradictions and develop a more nuanced understanding of antibody-mediated HTLV-1 neutralization.

What promising approaches are emerging for developing bispecific antibodies targeting ATL cells?

Bispecific antibody development for ATL represents an exciting frontier with several promising methodological approaches. Based on current research trends, the most promising strategies include: First, targeting combinations of ATL-associated surface markers, such as pairing CCR4 (targeted by mogamulizumab) with other ATL-enriched markers like CD25 or CD30 to enhance specificity for malignant cells . Second, creating T-cell engaging bispecific antibodies that simultaneously bind ATL cells and T cell receptors (like CD3), redirecting cytotoxic T cells to eliminate tumor cells—this approach could potentially overcome the immunosuppressive environment often associated with ATL. Third, developing bispecific antibodies that target both viral and cellular antigens, such as combining recognition of HTLV-1 envelope proteins with cellular markers to precisely identify infected cells. Fourth, employing dual-action antibodies that simultaneously block signaling pathways critical for ATL cell survival while engaging immune effector functions. Fifth, exploring novel antibody engineering platforms like knob-into-hole technology, CrossMAb formats, or DNA-encoded antibody libraries to generate stable, high-affinity bispecific molecules. When evaluating these approaches, researchers should implement comprehensive testing that includes binding studies with primary ATL samples, functional assays measuring cytotoxicity against ATL cells while sparing normal T cells, and in vivo testing in appropriate animal models. The promising results seen with mogamulizumab's dual mechanism of action—direct killing of CCR4+ cells while enhancing anti-HTLV-1 T cell responses—provides a conceptual foundation for these next-generation bispecific approaches .

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.