ARR20 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ARR20 antibody; At3g62670 antibody; F26K9_100Putative two-component response regulator ARR20 antibody
Target Names
ARR20
Uniprot No.

Target Background

Function
ARR20 Antibody targets a putative transcriptional activator that binds specifically to the DNA sequence 5'-[AG]GATT-3'. This protein functions as a response regulator involved in the His-to-Asp phosphorelay signal transduction system. Phosphorylation of the Asp residue within the receiver domain activates the protein, enabling it to promote the transcription of target genes. Notably, ARR20 Antibody could directly activate certain type-A response regulators in response to cytokinins.
Database Links
Protein Families
ARR family, Type-B subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Predominantly expressed in mature pistil tip. Also detected in the shoot apical meristem as well as vascular tissue and hydathodes of the leaves.

Q&A

What is mAb-AR20.5 and what target does it recognize?

mAb-AR20.5 is a monoclonal antibody that targets MUC1, a transmembrane glycoprotein that is overexpressed and aberrantly glycosylated in various cancers, particularly pancreatic cancer. This antibody is designed to recognize specific epitopes on the MUC1 antigen, allowing for targeted immunotherapy approaches. Mechanistically, mAb-AR20.5 works by opsonizing tumor cells expressing MUC1, effectively marking them for recognition by the immune system .

How does mAb-AR20.5 contribute to anti-tumor immunity?

mAb-AR20.5 contributes to anti-tumor immunity through antigen opsonization, where the antibody binds to MUC1 on tumor cells, enhancing their recognition and processing by antigen-presenting cells. This process facilitates the generation of MUC1-specific T cell responses. Research has demonstrated that mAb-AR20.5 can prime tumor-bearing hosts with their own antigen, initiating an immunological cascade that, when combined with other immunomodulatory agents, leads to effective anti-tumor responses .

What experimental models are appropriate for testing mAb-AR20.5?

Human MUC1 transgenic (MUC.Tg) mice have been established as an appropriate model for testing mAb-AR20.5, as these mice express human MUC1 and can develop MUC1-expressing tumors. This model allows researchers to evaluate both the efficacy and specificity of mAb-AR20.5-based therapies in a controlled setting. The model is particularly valuable for testing combinatorial approaches and for investigating mechanisms of tumor rejection and immune memory formation .

What is the rationale behind combining mAb-AR20.5 with immune checkpoint inhibitors?

The combination of mAb-AR20.5 with immune checkpoint inhibitors, particularly anti-PD-L1, addresses multiple aspects of tumor-induced immunosuppression. While mAb-AR20.5 enhances tumor antigen presentation and initiates immune responses, checkpoint inhibitors prevent the exhaustion and suppression of activated T cells. Research has shown that this combination can overcome the immunosuppressive tumor microenvironment that typically renders MUC1-specific immune responses ineffective in cancer patients. Specifically, studies have demonstrated that the mAb-AR20.5 + anti-PD-L1 + PolyICLC combination induced rejection of human MUC1-expressing tumors and provided long-lasting, MUC1-specific cellular immune responses .

How can researchers evaluate the MUC1-specific immune response generated by mAb-AR20.5 combinations?

Evaluating MUC1-specific immune responses requires multiple complementary approaches:

  • Adoptive transfer studies: Transferring immune cells from treated to naive animals to demonstrate protection against tumor challenge

  • Antibody depletion studies: Selectively depleting specific immune cell subsets (e.g., CD8+ T cells) to identify effector mechanisms

  • Flow cytometry analysis: Tracking changes in circulating activated immune cells over time

  • Tumor rejection assays: Monitoring tumor growth in treated versus control animals

Research has demonstrated that CD8+ T cells are critical effectors for the MUC1-specific immune response generated by the mAb-AR20.5 + anti-PD-L1 + PolyICLC combination. Multichromatic flow cytometry has revealed a significant increase in circulating, activated CD8+ T cells over time in treated subjects .

What are the methodological considerations for studying durable immunity induced by mAb-AR20.5 combinations?

When studying durable immunity induced by mAb-AR20.5 combinations, researchers should consider:

  • Long-term follow-up: Extending study duration to evaluate persistence of immunity

  • Recall response assessment: Challenging cured animals with secondary tumors to test immune memory

  • Immune phenotyping: Characterizing memory T cell populations (effector memory vs. central memory)

  • Mechanistic analyses: Investigating the molecular basis of sustained immune responses

Research has shown that mAb-AR20.5 in combination with anti-PD-L1 and PolyICLC can provide long-lasting, MUC1-specific cellular immune responses that can be adoptively transferred, suggesting the development of durable immunological memory .

What are the optimal dosing strategies for mAb-AR20.5 in combination therapies?

Determining optimal dosing strategies requires consideration of:

  • Dose-response relationships: Testing multiple dose levels to identify minimum effective dose

  • Timing of administration: Evaluating sequential versus concurrent administration of combination agents

  • Route of administration: Comparing intravenous, intraperitoneal, or other routes

  • Treatment schedule: Determining frequency and duration of dosing

Research suggests that timing may be critical - administering mAb-AR20.5 to opsonize tumor cells, followed by immune checkpoint inhibition and immunostimulation with PolyICLC, may provide optimal sequencing for generating robust anti-tumor immunity .

How can researchers overcome technical challenges in antibody production and quality control?

Production and quality control of research-grade mAb-AR20.5 involve several critical steps:

  • Hybridoma cell culture optimization: Controlling serum conditions, cell density, and media composition

  • Purification process development: Optimizing protein A/G affinity chromatography and polishing steps

  • Functional validation: Testing binding specificity, affinity, and biological activity

  • Stability assessment: Evaluating thermal stability, aggregation propensity, and freeze-thaw resistance

For experimental reproducibility, researchers should characterize each antibody batch using techniques such as ELISA, flow cytometry, and Western blotting to confirm target binding before use in critical experiments.

How does mAb-AR20.5 compare to other anti-MUC1 antibodies in research applications?

When comparing mAb-AR20.5 to other anti-MUC1 antibodies, researchers should consider:

  • Epitope specificity: Different antibodies recognize distinct domains or glycoforms of MUC1

  • Binding affinity: Variation in KD values affects target engagement

  • Effector functions: Differences in Fc-mediated activities (ADCC, CDC, ADCP)

  • In vivo efficacy: Comparative tumor growth inhibition in relevant models

While limited comparative data is available, mAb-AR20.5 has demonstrated the ability to effectively opsonize MUC1-expressing tumor cells and, in combination with immune checkpoint inhibitors and immune stimulants, generate robust anti-tumor immunity .

What methodology is recommended for comparing different antibody combinations in preclinical models?

When comparing antibody combinations, researchers should employ:

  • Factorial experimental design: Testing all possible combinations systematically

  • Standardized endpoints: Using consistent metrics across experiments (tumor volume, survival, immune markers)

  • Pharmacokinetic/pharmacodynamic analyses: Understanding exposure-response relationships

  • Biomarker evaluation: Identifying predictive factors for response

Research has demonstrated that the specific combination of mAb-AR20.5 + anti-PD-L1 + PolyICLC provides superior efficacy compared to single or dual agent approaches, highlighting the importance of systematic combination testing .

What biomarkers are relevant for monitoring mAb-AR20.5 therapy response?

Key biomarkers for monitoring mAb-AR20.5 therapy response include:

  • MUC1 expression levels: Assessing target availability in tumor tissue

  • CD8+ T cell infiltration: Quantifying effector cells in the tumor microenvironment

  • T cell activation markers: Monitoring CD69, CD25, HLA-DR on circulating and tumor-infiltrating T cells

  • Cytokine profiles: Measuring changes in Th1/Th2 cytokine balance

  • Immune checkpoint expression: Evaluating adaptive resistance mechanisms

Multichromatic flow cytometry analysis has demonstrated significant increases in circulating, activated CD8+ T cells over time in subjects responding to mAb-AR20.5 combination therapy .

What are the key considerations for translating mAb-AR20.5 combinations from preclinical models to clinical studies?

Translating mAb-AR20.5 combinations to clinical studies requires addressing:

  • Species differences: Human versus mouse MUC1 biology and immune system variations

  • Patient selection strategies: Identifying appropriate biomarkers for patient stratification

  • Safety profile characterization: Evaluating potential autoimmune adverse events

  • Combination rationale: Providing mechanistic evidence for each component

  • Manufacturing considerations: Ensuring consistent antibody quality for human use

The proof-of-principle established in human MUC1 transgenic mice provides a strong rationale for clinical investigation, demonstrating that effective and long-lasting anti-tumor cellular immunity can be achieved in pancreatic tumor-bearing hosts through this combination approach .

How can researchers address variability in immune responses to mAb-AR20.5 combinations?

To address variability in immune responses, researchers should:

  • Control for genetic background: Using genetically defined mouse strains

  • Standardize tumor models: Controlling for passage number, inoculation technique

  • Monitor microbiome effects: Considering housing conditions and potential probiotic interventions

  • Account for age and sex differences: Including balanced experimental groups

  • Validate antibody batches: Testing each batch for consistent activity

Research has shown that CD8+ T cells are critical effectors for mAb-AR20.5 combination therapy, suggesting that baseline CD8+ T cell functionality might be an important variable to control in experiments .

What strategies can address potential immunogenicity issues with repeated mAb-AR20.5 administration?

Strategies to address potential immunogenicity include:

  • Antibody humanization: Replacing murine sequences with human counterparts

  • Anti-idiotype monitoring: Measuring host antibody responses against mAb-AR20.5

  • Alternative dosing schedules: Testing intermittent versus continuous administration

  • Combination with immunosuppressive agents: Evaluating co-administration with low-dose cyclophosphamide

  • Engineering antibody variants: Modifying immunogenic epitopes

Long-term studies in MUC.Tg mice have demonstrated the feasibility of generating durable immune responses without prohibitive immunogenicity issues when using appropriate dosing strategies .

How might mAb-AR20.5 combinations be applied to other MUC1-expressing cancers beyond pancreatic cancer?

Applications to other MUC1-expressing cancers could involve:

  • Cancer-specific optimization: Adjusting combination components based on immune context

  • Tumor microenvironment modulation: Adding agents targeting cancer-specific immunosuppressive mechanisms

  • Combining with standard therapies: Integrating with chemotherapy, radiation, or targeted therapies

  • Biomarker-guided approaches: Selecting patients based on MUC1 expression patterns and immune profiles

The mechanistic insights from pancreatic cancer studies, particularly regarding CD8+ T cell activation and durable immunity, provide a framework for investigating applications in breast, lung, colon, and ovarian cancers that also overexpress MUC1 .

What novel technologies could enhance the research applications of mAb-AR20.5?

Emerging technologies that could enhance mAb-AR20.5 research include:

  • Bispecific antibody engineering: Creating molecules targeting both MUC1 and immune effector cells

  • Antibody-drug conjugates: Coupling mAb-AR20.5 with cytotoxic payloads

  • CRISPR-based screening: Identifying resistance mechanisms and synergistic targets

  • Single-cell sequencing: Characterizing heterogeneous immune responses at resolution

  • In vivo imaging techniques: Tracking antibody biodistribution and immune cell trafficking

These approaches could build upon the established efficacy of mAb-AR20.5 + anti-PD-L1 + PolyICLC combinations while addressing remaining challenges in achieving universal and durable responses .

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.