ADT1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ADT1 Antibody

ADT1 Antibody is a primary antibody designed to detect arogenate dehydratase/prephenate dehydratase 1 (ADT1), a plastid-localized enzyme critical for phenylalanine biosynthesis in plants. It is primarily used in plant biology research to study enzymatic pathways, gene expression, and metabolic regulation in model organisms like Arabidopsis thaliana.

Biological Context of ADT1

ADT1 is part of a family of six genes (ADT1–ADT6) in Arabidopsis thaliana responsible for converting arogenate to phenylalanine, a precursor for lignin, flavonoids, and aromatic amino acids. ADT1 is localized to plastids, where it catalyzes the final step in phenylalanine biosynthesis, making it indispensable for plant growth and secondary metabolism .

Research Applications

ADT1 Antibody is utilized in:

  • Enzyme localization studies: Identifying ADT1 distribution in plastids and tissues.

  • Gene expression profiling: Quantifying ADT1 protein levels in developmental stages or stress conditions.

  • Metabolic pathway analysis: Studying interactions between ADT1 and other enzymes in phenylalanine biosynthesis.

  • Functional genomics: Validating knockdown/knockout phenotypes in mutants lacking ADT1 .

Technical Considerations

  • Handling: Reconstitute in sterile PBS or Tris-buffered saline (TBS). Avoid contamination to prevent nonspecific binding.

  • Limitations:

    • Currently out of stock; availability requires direct inquiry to manufacturers .

    • Potential challenges in distinguishing ADT1 from paralogs (ADT2–ADT6) due to sequence similarities.

  • Optimal Use: Pair with Western blotting or immunolocalization techniques for precise detection in Arabidopsis samples.

Research Findings and Insights

While specific studies using ADT1 Antibody are not detailed in publicly available literature, its development aligns with broader trends in plant metabolic research:

  1. Phenylalanine Biosynthesis: ADT1’s role in converting arogenate to phenylalanine positions it as a key regulatory point. Antibody-based detection aids in mapping pathogen-induced stress responses or nutrient deprivation effects on aromatic amino acid production .

  2. Gene Family Complexity: The presence of six ADT isoforms in Arabidopsis suggests functional redundancy or tissue-specific roles. ADT1 Antibody may help elucidate isoform-specific contributions to phenylalanine synthesis.

Future Directions

  • Cross-species applications: Testing ADT1 Antibody in other plant models (e.g., rice, maize) to explore conserved roles in phenylalanine metabolism.

  • Omics integration: Combining ADT1 detection with metabolomic or transcriptomic data to link enzyme activity to phenotypic traits.

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
ADT1 antibody; PDT1 antibody; At1g11790 antibody; F25C20.4 antibody; Arogenate dehydratase/prephenate dehydratase 1 antibody; chloroplastic antibody; AtADT1 antibody; AtPDT1 antibody; EC 4.2.1.51 antibody; EC 4.2.1.91 antibody
Target Names
ADT1
Uniprot No.

Target Background

Function
This antibody targets ADT1, an enzyme that catalyzes the conversion of prephenate (a product of the shikimate-chorismate pathway) into phenylalanine.
Gene References Into Functions
ADT1 functions as a prephenate dehydratase, utilizing prephenate as a substrate. (PMID: 21388819)
Database Links

KEGG: ath:AT1G11790

STRING: 3702.AT1G11790.1

UniGene: At.11172

Subcellular Location
Plastid, chloroplast stroma.
Tissue Specificity
Expressed in roots, leaves, stems, flowers and siliques.

Q&A

What is the mechanism of action for ADT-010 antibody in cancer immunotherapy?

ADT-010 is a first-in-class, fully human monoclonal antibody that selectively recognizes and binds to the Vδ1 TCR, leading to Vδ1 T cell activation. Unlike conventional immunotherapies, ADT-010 harnesses the specific anti-tumor properties of Vδ1 T cells, which combine features of both innate and adaptive immunity. These cells recognize and kill transformed cells through direct recognition of stress-associated ligands expressed on tumor cell surfaces, independent of MHC-restricted antigen presentation .

The activation mechanism has several key components:

  • ADT-010 mediates activation of Vδ1 T cells specifically in the presence of target tumor cells

  • Activation is demonstrated through TCR downregulation and CD107a/CD25 upregulation

  • The antibody enhances anti-tumor cytotoxicity while sparing healthy non-diseased cells

  • Vδ1 T cells activated by ADT-010 can potentially orchestrate potent and selective anti-tumor responses

This mechanism is particularly valuable for addressing "cold tumors" with decreased or impaired tumor antigen presentation, as γδ T cells can recognize tumor cells without relying on classical antigen presentation pathways.

How does ADT-010 differ from conventional T cell-targeting antibody therapies?

ADT-010 represents a fundamentally different approach compared to conventional T cell-based immunotherapies in several critical ways:

FeatureADT-010 ApproachConventional T Cell Therapies
Target cell populationVδ1 γδ T cellsPredominantly αβ T cells
Antigen recognitionMHC-independent recognition of stress ligandsMHC-dependent antigen recognition
Tissue distributionEnriched in epithelial tissues and tumor sitesPredominantly circulating in blood
Tumor infiltrationForm significant proportion of TILs in solid tumorsOften limited infiltration in "cold" tumors
Specificity mechanismDirect recognition of transformed cellsTypically requires antigen presentation

ADT-010 specifically targets Vδ1 T cells, which are abundant in epithelial tissues including gut and skin and form a significant proportion of tumor-infiltrating lymphocytes (TILs) in multiple solid tumor indications . This tissue-resident nature gives them advantageous positioning for tumor surveillance and response compared to predominantly circulating conventional T cells.

What are the optimal methods for evaluating ADT-010 binding specificity and functional activity?

Researchers investigating ADT-010 or similar immunomodulatory antibodies should implement a multi-step approach to comprehensively evaluate binding specificity and functional activity:

Binding Specificity Assessment:

  • High-stringency antigen binding protocols: ADT-010 was selected via high-stringency antigen binding using a human phage display library to ensure selectivity for the Vδ1 TCR .

  • Cross-reactivity testing: Evaluate binding to other TCR types (e.g., Vδ2, αβ TCRs) to confirm specificity.

  • Affinity measurements: Determine binding kinetics using surface plasmon resonance or similar techniques.

Functional Activity Evaluation:

  • Source appropriate T cells: Use expanded skin-derived Vδ1 T cells for initial testing .

  • Assess T cell activation markers:

    • Measure TCR downregulation following antibody engagement

    • Evaluate CD107a and CD25 upregulation as markers of activation

    • Assess cytokine production profiles (IFN-γ, TNF-α, etc.)

  • Cytotoxicity assays:

    • Perform in vitro tumor cell killing assays with and without ADT-010

    • Include healthy cell controls to confirm tumor specificity

    • Measure cytotoxicity against multiple tumor cell lines

For optimal sensitivity in multimodal analyses, researchers should consider manipulating antibody concentration, cell number, and staining volume to optimize signal-to-noise ratios .

How should researchers optimize antibody concentration for maximum signal detection in analytical assays?

Optimizing antibody concentration is critical for achieving reliable results in analytical assays. Based on empirical studies with oligonucleotide-conjugated antibodies, researchers should consider:

  • Systematic titration approach:

    • Begin with serial four-fold dilutions of antibody concentrations to identify optimal ranges

    • Dilution factors should span from manufacturer-recommended concentrations to significantly lower concentrations

    • Quantify signal using UMI (Unique Molecular Identifier) counts within cell-containing droplets

  • Epitope abundance considerations:

    • Antibodies targeting highly abundant epitopes (like CD31, CD44, CD45) often require lower concentrations to avoid signal saturation and reagent waste

    • Antibodies targeting less abundant epitopes may require higher concentrations for adequate detection

  • Cell density optimization:

    • For highly expressed epitopes, reducing cell number during staining (e.g., from 1×10^6 to 0.2×10^6 cells) while maintaining antibody concentration can significantly improve signal detection

    • This approach is particularly effective for antibodies used at low concentration targeting highly expressed epitopes

  • Panel balancing:

    • Aim for similar UMI counts per positive cell across antibodies in a panel

    • Consider the background signal for each antibody when determining optimal concentration

    • Markers with low background typically show low UMI cutoff and better dynamic range

What strategies can mitigate background signal and improve sensitivity for ADT-010 detection?

Researchers frequently encounter challenges with background signal when working with antibodies. Several evidence-based approaches can improve signal-to-noise ratio:

How can researchers assess and address post-translational modifications that may affect ADT-010 performance?

Post-translational modifications (PTMs) can significantly impact antibody stability, efficacy, and immunogenicity. A systematic approach to identify and mitigate PTM risks includes:

  • In silico analysis:

    • Perform computational analysis of antibody sequences to identify potential PTM hotspots

    • Mark unusual residues at particular positions, deletions, additions, or sequence anomalies

    • Identify high-risk motifs including NG/NS/DG motifs, extra Cys residues, and N-glycosylation motifs on variable regions

  • Experimental verification:

    • Test antibody candidates under stress conditions to evaluate stability

    • Monitor appearance, expression level, and purity throughout development

    • Conduct thermal stability assays to identify potential structural vulnerabilities

  • Engineering solutions:

    • Remove high-risk PTM hotspots through site-directed mutagenesis

    • If PTM hotspots cannot be removed without altering antigen binding, perform forced stress studies to evaluate potential risk

    • After sequence optimization, reassess all developability properties

  • Formulation strategies:

    • For antibodies with visible particles or precipitates, test different formulation buffers

    • Screen various buffers (acetate, citrate, phosphate) at different pH values to identify optimal solubility conditions

    • Consider adding suitable excipients (e.g., sucrose, polysorbate 80) to improve stability

How might ADT-010 be combined with other immunotherapeutic approaches for enhanced efficacy?

The complex nature of tumor microenvironments suggests that combinatorial approaches may yield superior results. Several strategic combinations warrant investigation:

  • Combination with checkpoint inhibitors:

    • ADT-010's activation of Vδ1 T cells could complement PD-1/PD-L1 or CTLA-4 blockade

    • Checkpoint inhibitors may relieve immunosuppression while ADT-010 specifically activates tumor-resident Vδ1 T cells

    • This dual approach might address both tumor immune evasion and direct cytotoxicity

  • Multi-target antibody approaches:

    • Similar to approaches in Alzheimer's disease research, targeting multiple mechanisms simultaneously may improve efficacy

    • Consider combinations targeting different aspects of tumor biology alongside immune activation

    • For example, combining ADT-010 with antibodies targeting tumor vasculature or tumor-specific antigens

  • Experimental design considerations:

    • Evaluate potential synergistic, additive, or antagonistic effects through systematic in vitro studies

    • Design factorial experiments to test different dose combinations and timing strategies

    • Consider sequential versus simultaneous administration protocols

  • Addressing cold tumor microenvironments:

    • ADT-010's ability to activate tissue-resident T cells may help convert "cold" tumors to "hot" immunologically responsive tumors

    • This conversion could enhance efficacy of other immunotherapies that depend on immune cell infiltration

    • Measure changes in tumor immune cell composition and cytokine profiles following combination treatments

What parameters should be considered when developing ADT-010 for clinical applications?

Successful translation of ADT-010 from research to clinical applications requires careful consideration of several developability parameters:

  • Early-stage developability assessment:

    • Evaluate homogeneity, stability, solubility, and specificity during discovery phase

    • Conduct in silico analysis to identify potential manufacturability issues

    • Perform targeted experiments to assess critical quality attributes before advancing candidates

  • Manufacturing considerations:

    • Develop stable cell lines for consistent antibody expression

    • Establish purification protocols that maintain antibody functionality

    • Consider stability under various storage and handling conditions

  • Toxicology planning:

    • Design studies to establish dose-response relationships, toxic dose thresholds, and NOAEL (no observed adverse effect level)

    • Include sufficiently high doses to guide clinical usage while monitoring safety parameters

    • Consider species-specific differences in γδ T cell biology when interpreting preclinical toxicology data

  • Immunogenicity risk assessment:

    • ADCs (antibody-drug conjugates) pose potentially greater immunogenicity risk than conventional monoclonal antibodies

    • Evaluate anti-drug antibody development in preclinical models

    • Consider structural features that might increase immunogenicity risk

  • Formulation development:

    • Screen different buffer compositions at various pH values

    • Identify excipients that enhance stability and solubility

    • Monitor appearance, concentration, and SEC profiles over time under various storage conditions

How can researchers evaluate ADT-010 efficacy across different tumor types with varying immune infiltration profiles?

Evaluating ADT-010 across diverse tumor types requires systematic approaches to account for heterogeneity in immune infiltration and γδ T cell prevalence:

  • Tumor microenvironment profiling:

    • Characterize baseline Vδ1 T cell infiltration across tumor types

    • Assess expression patterns of stress-associated ligands recognized by γδ T cells

    • Map spatial distribution of immune populations within tumor vs. stroma

  • Predictive biomarker identification:

    • Correlate response to ADT-010 with specific tumor or immune signatures

    • Develop multiplexed imaging approaches to characterize tumor-immune cell interactions

    • Identify tumor-specific factors that modulate Vδ1 T cell activation or recruitment

  • Ex vivo tumor explant models:

    • Utilize fresh tumor explants to maintain native immune cell composition

    • Evaluate ADT-010-mediated Vδ1 T cell activation in this context

    • Compare efficacy between "hot" (immune-infiltrated) vs. "cold" tumors

  • Comparative efficacy studies:

    • Test activity against tumor types with impaired antigen presentation

    • Evaluate efficacy in models resistant to conventional immunotherapies

    • Quantify tumor-specific Vδ1 T cell expansion and activation following ADT-010 treatment

These approaches should help identify which tumor types are most likely to benefit from ADT-010 therapy and inform rational combination strategies for tumors with different immune infiltration profiles.

What next-generation modifications might enhance ADT-010's therapeutic potential?

Building on the first-in-class foundation of ADT-010, several engineering approaches could potentially enhance its therapeutic profile:

  • Affinity optimization:

    • Affinity maturation to enhance binding properties without compromising specificity

    • Fine-tuning association/dissociation kinetics to optimize T cell activation

    • Structure-guided modifications to increase stability while maintaining function

  • Bispecific adaptations:

    • Developing bispecific antibodies that simultaneously engage Vδ1 TCR and tumor-specific antigens

    • Creating formats that bring Vδ1 T cells into proximity with tumor cells

    • Engineering trispecific constructs to engage multiple immune cell populations

  • Antibody-drug conjugate approaches:

    • Leveraging ADT-010's binding specificity to deliver payloads to the tumor microenvironment

    • Selecting payloads that might enhance Vδ1 T cell function or target immunosuppressive factors

    • Optimizing drug-to-antibody ratio and linker chemistry for stability and efficacy

  • Alternative formats:

    • Exploring fragment-based approaches for enhanced tumor penetration

    • Investigating alternative scaffolds with improved pharmacokinetic properties

    • Developing multimerization strategies to enhance avidity and functional activity

Each of these approaches would require systematic developability assessment to ensure the modified constructs maintain favorable stability, solubility, and manufacturing characteristics while enhancing therapeutic potential.

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.