ATL39 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Targeting in ATL Treatment

ATL therapies frequently employ monoclonal antibodies (mAbs) targeting surface markers on malignant T-cells. Key targets include:

  • CD25 (IL-2Rα): Targeted by anti-Tac antibodies

  • CCR4: Targeted by mogamulizumab

  • CD2: Targeted by MEDI-507

  • CD39: Targeted by TTX-030

TargetAntibody NameMechanismClinical Status
CD25Anti-Tac (HAT)Blocks IL-2 signalingPhase II
CCR4MogamulizumabAntibody-dependent cellular cytotoxicity (ADCC)Approved in Japan
CD2MEDI-507ADCC via FcγRIII engagementPreclinical
CD39TTX-030Inhibits adenosine production in tumor microenvironmentPhase Ib

MEDI-507 (Anti-CD2)

  • Prolonged survival in murine ATL models compared to anti-CD25 therapy

  • Requires FcγRIII on neutrophils/monocytes for tumor killing

Mogamulizumab (Anti-CCR4)

  • Reduces ATL cells via NK cell-mediated ADCC

  • Risk of severe graft-versus-host disease in transplant recipients

TTX-030 (Anti-CD39)

  • Combined with chemoimmunotherapy in gastric/GEJ cancer trials

  • Modulates extracellular ATP/adenosine balance to enhance antitumor immunity

Challenges in ATL Antibody Development

  • Immune evasion: ATL cells upregulate PD-L1 and HLA mutations to resist immune responses

  • Tumor heterogeneity: Variability in Tax protein expression limits vaccine efficacy

  • Toxicity: Off-target effects (e.g., GVHD with mogamulizumab)

Emerging Strategies

  • Combination therapies: Anti-CD39 + anti-PD-1 (e.g., TTX-030 + budigalimab)

  • Vaccines: Tax-targeted dendritic cell vaccines to boost CTL 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
14-16 week lead time (made-to-order)
Synonyms
ATL39; At4g09100; F23J3.130; T8A17.4; RING-H2 finger protein ATL39; RING-type E3 ubiquitin transferase ATL39
Target Names
ATL39
Uniprot No.

Target Background

Database Links

KEGG: ath:AT4G09100

STRING: 3702.AT4G09100.1

UniGene: At.54223

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

Q&A

What is the relationship between HTLV-1 and ATL?

Human T-cell leukemia virus type 1 (HTLV-1) causes adult T-cell leukemia/lymphoma (ATL) in approximately 5% of infected individuals after a decades-long latency period . HTLV-1 also causes HTLV-1–associated myelopathy, a chronic progressive inflammatory disease of the central nervous system, in about 0.3% to 4% of infected people . The virus primarily infects CD4+ T cells, with about 5% of the proviral load present in CD8+ T cells . HTLV-1 has been present in human populations for more than 20,000 years and is estimated to infect at least 10 million people globally, though the true number is likely higher due to incomplete epidemiological surveillance in endemic areas . Understanding this relationship is fundamental for researchers designing antibody-based detection and therapeutic strategies.

How do researchers detect HTLV-1 in clinical samples?

Researchers typically detect HTLV-1 in clinical samples through indirect immunofluorescence (IF) testing for Adult T-cell leukemia associated antibody (ATLA-Ab) . When testing lymphocytes from ATLA-Ab positive individuals, researchers collect lymphocytes from concentrated red blood cells (CRC) and culture them in vitro with and without phytohemagglutinin (PHA) for approximately 10 days . The expression of ATL virus (ATLV) positive lymphocytes during in vitro culture is then analyzed using IF assay with specific antibodies such as mouse monoclonal antibody ATL-19, which is reactive to p19 core protein of ATLV .

Research findings demonstrate that 97% of ATLA-Ab positive CRC samples show ATLV positive lymphocytes after being cultured for more than 10 days with PHA, whereas ATLA-Ab negative samples do not . The viability and detectability of these lymphocytes decrease with sample storage time - samples stored for 2-7 days maintain full detection capacity, while only 70% of samples stored for 14 days and merely 10% of samples stored for 20 days yield positive results . This methodological approach allows researchers to reliably detect HTLV-1 infected cells in clinical specimens.

What are the key surface markers targeted by antibodies in ATL research?

In ATL research, several surface markers are commonly targeted by therapeutic and research antibodies. CD52 is targeted by alemtuzumab (CAMPATH-1H), a humanized monoclonal antibody that has shown efficacy in clinical trials . CD25 (IL-2R alpha, Tac) is another important marker expressed on chronic, acute, and lymphomatous ATL subtypes, targeted by the antibody daclizumab . CD2 is targeted by siplizumab (MEDI-507) .

For research inclusion criteria, patients typically need to have serum antibodies directed to HTLV-1 confirmed by Western blot and more than 10% of malignant cells expressing CD52 and CD25 as determined by flow cytometry or immunohistochemical staining . These surface markers represent critical research targets for both diagnostic applications and potential therapeutic interventions, allowing for specific identification and targeting of ATL cells.

How do antibody-based treatments compare in efficacy for different ATL subtypes?

Clinical research shows varying efficacy of antibody-based treatments across the different ATL subtypes (acute, chronic, and lymphomatous). In a Phase II clinical trial of alemtuzumab (CAMPATH-1H), the antibody induced responses primarily in patients with acute HTLV-1-associated ATL . The study enrolled 29 patients (15 with acute ATL, 11 with lymphoma type, and 3 with chronic ATL) . While the treatment showed acceptable toxicity, the duration of responses was short, suggesting that alemtuzumab alone is insufficient as a monotherapy .

The differential response across subtypes likely reflects the biological heterogeneity of ATL. Preclinical research using xenograft murine models of human ATL has demonstrated efficacy with antibodies targeting different surface markers: the CD2-directed monoclonal antibody siplizumab, the anti-CD25 antibody daclizumab, and the anti-CD52 antibody alemtuzumab . These findings suggest that ATL subtypes may benefit from different antibody approaches or combination strategies, highlighting the need for personalized treatment approaches based on disease subtype and molecular characteristics.

What methodological challenges exist in developing antibodies for ATL-specific antigens?

Developing antibodies for ATL-specific antigens presents several methodological challenges. One major challenge is the design of antibodies that specifically target ATL cells while sparing normal T cells. This requires identifying unique or differentially expressed antigens on ATL cells. Currently, many antibodies target antigens that are overexpressed on ATL cells (such as CD25) but are also present on normal activated T cells, leading to potential off-target effects .

Another significant challenge is designing antibodies that can effectively bind to their targets in the three-dimensional context of the cell surface. Recent advances in computational approaches, such as diffusion probabilistic models and equivariant neural networks, aim to address this by jointly modeling sequences and structures of antibody complementarity-determining regions (CDRs) . These models need to consider not only the position but also the orientation of amino acids, as interactions between amino acids are mainly determined by side-chains stretching out from the protein backbone .

Furthermore, optimizing antibodies for increased binding affinity while maintaining specificity requires sophisticated methodologies. The development of deep learning models that are explicitly conditional on the 3D structures of the antigen and generate CDRs that fit the antigen structure in 3D space represents a promising approach for ATL-specific antibody development .

How can researchers optimize antibody persistence and efficacy in ATL treatment?

Optimizing antibody persistence and efficacy in ATL treatment involves several research strategies. First, antibody engineering techniques can be employed to modify the Fc region, potentially extending the half-life and enhancing immune effector functions . Second, combination approaches with other therapeutics may overcome the limited duration of response observed with single-agent antibody therapy, as seen in the alemtuzumab clinical trial where responses were of short duration .

Research models for antibody optimization include both computational approaches and experimental validation. Computational methods such as DiffAb can perform antibody sequence-structure co-design, antibody sequence design based on existing backbones, and antibody optimization . These approaches can generate antibodies that explicitly target specific antigen structures and consider both position and orientation of amino acids to enhance binding affinity .

Experimental validation requires rigorous testing in preclinical models before clinical application. The MET-1 in vivo model of ATL has been valuable for demonstrating the efficacy of various monoclonal antibodies before translation to clinical trials . Additionally, pharmacokinetic and pharmacodynamic studies are essential to understand the distribution, metabolism, and clearance of antibodies in ATL patients, factors that significantly impact persistence and efficacy.

What are the optimal protocols for validating antibody specificity in ATL research?

Validating antibody specificity in ATL research requires a multi-faceted approach. Western blot analysis with appropriate controls is a fundamental validation method. For example, when validating antibodies like the Anti-Leptin Receptor antibody, researchers compare antibody reactivity in different tissue samples (such as brain membranes, dorsal root ganglion lysate) with and without a blocking peptide . The absence of signal when the antibody is pre-incubated with its specific blocking peptide confirms specificity .

Immunohistochemical staining provides another validation method, particularly valuable for confirming tissue-specific expression patterns. This approach involves staining tissue sections with the antibody followed by fluorescent secondary antibodies, with parallel experiments using blocking peptides as controls . The suppression of staining upon pre-incubation with a specific blocking peptide confirms specificity .

For ATL-specific research, additional validation steps include:

  • Testing antibody reactivity against HTLV-1 positive versus negative cell lines

  • Conducting competitive binding assays with known ligands

  • Using genetic approaches (knockdown/knockout) to confirm target specificity

  • Flow cytometry analysis to confirm cell surface binding patterns

These comprehensive validation approaches ensure that antibodies used in ATL research accurately detect their intended targets, critical for reliable research outcomes and potential clinical applications.

How can researchers interpret conflicting antibody binding data in ATL studies?

Interpreting conflicting antibody binding data in ATL studies requires systematic troubleshooting and methodological considerations. First, researchers should examine experimental conditions that might affect antibody binding, including fixation methods, epitope masking, and antigen retrieval techniques. For instance, the detection of HTLV-1 bearing lymphocytes can be significantly affected by sample storage conditions - samples stored for more than 14 days show reduced ATLV-positive lymphocyte detection .

Second, antibody characteristics need careful consideration. Different antibodies targeting the same protein but recognizing different epitopes may yield discrepant results. For example, antibodies targeting intracellular versus extracellular domains might perform differently depending on sample preparation . Additionally, the format of the antibody (monoclonal vs. polyclonal) and its species of origin can influence binding patterns and cross-reactivity.

Third, biological variability in ATL samples can lead to apparently conflicting results. ATL is biologically heterogeneous, with variations in antigen expression across subtypes (acute, chronic, and lymphomatous) . Patient-specific factors, including prior treatments and genetic background, may also contribute to variable antibody binding.

To resolve such conflicts, researchers should:

  • Employ multiple antibodies targeting different epitopes of the same protein

  • Use complementary detection methods (Western blot, immunohistochemistry, flow cytometry)

  • Include appropriate positive and negative controls

  • Consider patient-specific factors in data interpretation

  • Validate findings using functional assays to confirm biological relevance

What advanced imaging techniques maximize the utility of antibodies in ATL research?

Advanced imaging techniques significantly enhance the utility of antibodies in ATL research by providing spatial, temporal, and quantitative information about target antigens. Confocal microscopy with fluorescently labeled antibodies allows high-resolution visualization of antigen localization within cells and tissues. This approach has been used successfully to visualize receptor distribution, as demonstrated in studies of Leptin Receptor expression in mouse brain sections using Anti-Leptin Receptor antibodies followed by AlexaFluor-488 secondary antibodies .

Super-resolution microscopy techniques such as Stimulated Emission Depletion (STED), Structured Illumination Microscopy (SIM), and Photoactivated Localization Microscopy (PALM) overcome the diffraction limit of conventional microscopy, enabling visualization of antigen distribution at nanometer resolution. These techniques are particularly valuable for studying the subcellular localization of viral proteins in HTLV-1 infected cells.

Multiplexed imaging approaches allow simultaneous detection of multiple antigens in the same sample. Techniques such as multiplexed immunofluorescence, imaging mass cytometry, and cyclic immunofluorescence enable comprehensive phenotyping of ATL cells and their microenvironment. These methods are especially valuable for studying the complex interactions between ATL cells and immune cells in the tumor microenvironment.

Live-cell imaging with fluorescently labeled antibody fragments or nanobodies facilitates real-time visualization of dynamic cellular processes. This approach can be used to study virus-host interactions, cell-to-cell spread via the virological synapse, and the dynamics of receptor expression and internalization in response to therapeutic interventions.

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.