ARGAH2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ARGAH2 Antibody

C0021158 is a high-affinity human monoclonal antibody designed to inhibit ARG2, an enzyme overexpressed in cancers such as pancreatic ductal adenocarcinoma and acute myeloid leukemia . ARG2 depletes extracellular L-arginine, creating an immunosuppressive tumor microenvironment that suppresses T-cell function . This antibody represents a novel immunotherapeutic strategy to restore anti-tumor immunity.

Mechanism of Action

C0021158 employs a noncompetitive allosteric inhibition mechanism:

  • Epitope Binding: Targets a regulatory site distant from ARG2's substrate-binding cleft .

  • Structural Remodeling: Induces conformational changes in ARG2, including:

    • Rearrangement of a surface loop into a short helix .

    • Reorientation of Arg39, sterically blocking L-arginine binding .

  • Catalytic Disruption: Alters the pKA of catalytic His160, reducing enzymatic activity .

Table 1: Key Functional Properties of C0021158

PropertyValueSource
Binding Affinity (KD)173 pM for ARG2
IC50 (Enzyme Inhibition)18.5 ± 5.1 nM (IgG form)
SpecificityNo binding to ARG1
T-cell ProliferationFully restored in vitro

Development and Affinity Maturation

C0021158 was optimized using an unbiased ribosome display strategy:

  • Library Design: Combined chain shuffling and staggered extension PCR (Shuffle/ShuffleStEP) to explore mutations across all six CDRs .

  • Pool Maturation: Seven leads were optimized simultaneously, achieving a 50-fold affinity improvement over the parent antibody (C0020187) .

Table 2: Affinity Maturation Outcomes

MetricParent (C0020187)Matured (C0021158)
Binding Affinity (KD)8.6 nM173 pM
Inhibitory CooperativityNegativeNeutral
Epitope Surface Area750 Ų1,100 Ų

Preclinical Efficacy

  • Immune Restoration: Reversed ARG2-mediated suppression of T-cell proliferation in vitro .

  • Synergy Potential: Enhanced efficacy observed in combination with chemotherapy (e.g., gemcitabine) .

Table 3: In Vitro Efficacy Data

Assay TypeResult
ARG2 Inhibition (100 nM)100% enzyme activity suppression
T-cell ProliferationRestored to baseline levels

Comparative Analysis with AGR2 Antibodies

While C0021158 targets ARG2, other antibodies (e.g., P3A5, P1G4) focus on Anterior Gradient 2 (AGR2), a distinct tumor-associated antigen .

Table 4: Target-Specific Antibody Comparison

AntibodyTargetMechanismClinical Stage
C0021158ARG2Allosteric enzyme inhibitionPreclinical
P3A5AGR2Cell surface bindingPreclinical

Current Status and Future Directions

C0021158 is in preclinical development, with plans to:

  • Partner with pharmaceutical companies for clinical trials .

  • Explore combinations with checkpoint inhibitors or chemotherapy .

  • Investigate applications in non-oncology indications (e.g., chronic infections) .

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
ARGAH2 antibody; At4g08870 antibody; T3H13.10Arginase 2 antibody; chloroplastic/mitochondrial antibody; EC 3.5.3.1 antibody; Agmatinase ARGAH2 antibody; EC 3.5.3.11 antibody; Arginine amidohydrolase 2 antibody
Target Names
ARGAH2
Uniprot No.

Target Background

Function
ARGAH2 catalyzes the hydrolysis of L-arginine, producing urea and L-ornithine. L-ornithine can be utilized in the urea cycle or as a precursor for the synthesis of polyamines and proline. Additionally, ARGAH2 possesses agmatinase activity, catalyzing the formation of putrescine from agmatine.
Gene References Into Functions
  1. AtARGAHs, including AtARGAH2, negatively regulate various abiotic stress tolerances. This regulation is, at least in part, attributed to their roles in modulating arginine metabolism and reactive oxygen species accumulation. PMID: 23378380
  2. Gene mutations within ARGAH1 or ARGAH2 are linked to increased nitric oxide signaling during root development. PMID: 18567826
Database Links

KEGG: ath:AT4G08870

STRING: 3702.AT4G08870.1

UniGene: At.4188

Protein Families
Arginase family
Subcellular Location
Mitochondrion. Plastid, chloroplast.
Tissue Specificity
Expressed in vasculature of roots, root tips, leaves and cotyledons.

Q&A

What is Arginase 2 (ARG2) and why is it a significant target for inhibitory antibody development?

Arginase 2 (ARG2) is a binuclear manganese metalloenzyme that catalyzes the hydrolysis of L-arginine into L-ornithine and urea. Its significance as a therapeutic antibody target stems from its role in creating immunosuppressive tumor microenvironments. ARG2 is overexpressed in various cancer types, including pancreatic ductal adenocarcinoma, bowel cancer, and acute myeloid leukemia . In these contexts, excess ARG2 depletes L-arginine around tumors, critically impairing T-cell function since L-arginine is essential for immune cell activity . This depletion creates an immunosuppressive local environment that allows tumors to grow unchecked, effectively hiding from the immune system . Inhibitory antibodies targeting ARG2 can potentially restore immune function in the tumor microenvironment by preventing L-arginine depletion, making ARG2 a promising target for cancer immunotherapy approaches .

How do experimental methodologies differ when evaluating ARG2 antibody specificity versus inhibitory potency?

When evaluating ARG2 antibody specificity, researchers typically employ:

  • Epitope competition (EC) assays to measure how well an antibody competes with the parental antibody for the epitope, serving as a surrogate measure for binding affinity

  • Cross-reactivity testing against related proteins (particularly ARG1)

  • Immunohistochemistry with appropriate controls to confirm specific binding

For assessing inhibitory potency, complementary approaches include:

  • Enzyme inhibition assays (EIA) to directly measure the antibody's ability to inhibit ARG2 enzymatic activity

  • T-cell proliferation assays in arginine-depleted conditions

  • Functional immune restoration assays to demonstrate the antibody's ability to reverse ARG2-mediated immunosuppression

The most robust experimental designs employ both specificity and potency assessments, as high binding affinity does not necessarily correlate with optimal enzyme inhibition . Using these complementary approaches provides a comprehensive characterization of ARG2 antibody functionality.

What are the key structural considerations when developing ARG2 inhibitory antibodies?

Structural analyses of ARG2 antibodies have revealed several critical considerations for effective inhibitor development:

Structural AspectSignificanceMethodological Implications
Binding paratope orientationDetermines contact surface and shape complementarityCrystal structures reveal striking reorientations in optimized antibodies
Epitope locationAffects mechanism of inhibitionConsider targeting beyond the active site for novel inhibitory mechanisms
CDR optimizationAll six CDRs can contribute to bindingUnbiased approach to CDR optimization yields superior results
Conformational changesCan affect ARG2 enzyme activityStructural analysis necessary to understand inhibition mechanism

Crystal structures comparing parent and affinity-matured ARG2 antibodies have demonstrated that extensive sequence changes can translate into extraordinary structural modifications . These include large reorientations of the binding paratope that facilitate increased contact surface and improved shape complementarity to ARG2 . Such comprehensive structural evolution is unlikely to be achieved through conventional affinity maturation methods that focus only on limited regions of the antibody .

What innovative affinity maturation techniques have proven most effective for ARG2 inhibitory antibodies?

Conventional affinity maturation strategies are often restrictive, focusing on small regions of the antibody. For ARG2 inhibitory antibodies, innovative approaches have demonstrated superior results:

The Shuffle/ShuffleStEP method combines antibody chain shuffling with a staggered-extension process to produce unbiased libraries . This approach:

  • Recombines beneficial mutations from all six complementarity-determining regions (CDRs) simultaneously

  • Utilizes ribosome display's vast display capacity to accommodate the extensive sequence space required

  • Achieves comprehensive optimization across the entire antibody binding interface

Additional innovations include pool maturation, which optimizes multiple lead candidates simultaneously . In one study, optimization of CDRH1 and CDRH2 regions produced the highest hit rates (43% and 54% respectively), while CDRH3 optimization resulted in lower success rates (12%) . This contrasts with conventional approaches that often focus primarily on CDRH3.

These advanced techniques have yielded antibodies with substantial improvements in binding properties and inhibition potency . The extent of sequence and structural evolution achieved through these unbiased approaches would be extremely difficult to obtain through conventional methods .

How can researchers effectively assess the inhibitory function of ARG2 antibodies in experimental settings?

Effective assessment of ARG2 antibody inhibitory function requires multi-faceted experimental approaches:

  • Biochemical inhibition assays:

    • Enzyme inhibition assays (EIA) directly measure the antibody's ability to prevent ARG2 from hydrolyzing L-arginine

    • Determination of inhibition constants (Ki) and IC50 values provides quantitative measures of potency

    • Kinetic analysis can reveal inhibition mechanisms (competitive, non-competitive, etc.)

  • Functional cellular assays:

    • T-cell proliferation assays in ARG2-conditioned media can demonstrate restoration of immune cell function

    • Co-culture systems with ARG2-expressing cells and T-cells model the tumor microenvironment

    • Cytokine production assays (IL-2, IFN-γ) measure functional T-cell outputs

  • Combined screening approaches:

    • Parallel screening with both epitope competition (EC) and enzyme inhibition assays (EIA) provides complementary data on binding and function

    • Comparing hit rates across different optimization strategies helps identify the most productive approaches for further development

When implementing these methods, researchers should consider that binding affinity does not always correlate directly with inhibitory potency . A comprehensive assessment requires both binding and functional assays to identify antibodies with optimal therapeutic potential.

How do ARG2 antibodies interact with other components of the tumor immune microenvironment?

ARG2 antibodies interact with multiple components of the tumor immune microenvironment in ways that extend beyond simple enzyme inhibition:

  • Effects on regulatory T cells (Tregs):
    Research indicates that ARG2-specific T cells can recognize and react to activated Tregs with high ARG2 expression . This suggests that ARG2 antibodies may indirectly modulate Treg activity in the tumor microenvironment by affecting ARG2 function in these immunosuppressive cells .

  • Impact on myeloid cells:
    ARG2 expression has been observed in immunosuppressive dendritic cells (DCs) and tumor-associated macrophages (TAMs) . ARG2 antibodies may alter the function of these myeloid cells, potentially shifting them from immunosuppressive to immunostimulatory phenotypes.

  • Restoration of T-cell function:
    By inhibiting ARG2 and preventing L-arginine depletion, ARG2 antibodies can restore T-cell proliferation and function . This effect extends to both pre-existing tumor-infiltrating lymphocytes and newly recruited T cells.

  • Interactions with cancer-associated fibroblasts:
    Some studies indicate ARG2 expression in cancer-associated fibroblasts , suggesting that ARG2 antibodies may affect stromal components of the tumor microenvironment.

When designing experiments to study these interactions, researchers should employ multiparametric analyses to capture the complex effects of ARG2 inhibition on different cell populations within the tumor ecosystem.

What mechanisms explain the structural and functional improvements seen in affinity-matured ARG2 antibodies?

Crystal structures comparing parent and affinity-matured ARG2 antibodies have revealed striking structural changes that explain their enhanced functional properties:

  • Binding interface reorientation:
    Affinity-matured antibodies exhibit a large reorientation of the binding paratope . This structural reorganization facilitates substantial increases in contact surface area and improved shape complementarity to ARG2 .

  • Epitope expansion:
    The structural changes enable the optimized antibodies to interact with an expanded epitope on ARG2 . This expanded interaction surface contributes to improved binding affinity and potentially more effective enzyme inhibition.

  • Relief from negative cooperativity:
    Some optimized ARG2 antibodies show improvement in binding properties resulting from an apparent relief from negative cooperativity of binding . This may enhance their ability to maintain inhibition under physiological conditions.

  • Comprehensive sequence evolution:
    The nature and magnitude of these structural changes are extraordinary and unlikely to have been produced through conventional affinity maturation methods . The unbiased approach to optimizing all six CDRs simultaneously allows for coordinated sequence changes that work together to improve binding and function.

These insights demonstrate the value of comprehensive structural analysis in understanding the molecular basis of improved antibody function, rather than relying solely on binding or inhibition assays.

What screening strategies are most effective for identifying high-potency ARG2 inhibitory antibodies?

Effective screening strategies for ARG2 inhibitory antibodies combine multiple assays to identify candidates with optimal therapeutic properties:

Researchers should implement comprehensive screening strategies that balance throughput with detailed functional characterization to identify antibodies with the most promising therapeutic potential.

How can researchers investigate the relationship between ARG2 antibodies and ARG2-specific T cells?

The relationship between ARG2 antibodies and ARG2-specific T cells represents an intriguing area of investigation with potential therapeutic implications:

  • Complementary targeting mechanisms:
    While ARG2 antibodies directly inhibit enzyme function, ARG2-specific T cells recognize and react to cells expressing ARG2 . These complementary mechanisms suggest potential synergistic approaches targeting ARG2-mediated immunosuppression.

  • Experimental approaches:
    Researchers can investigate these relationships through:

    • Co-culture systems with ARG2-expressing cells, ARG2-specific T cells, and ARG2 antibodies

    • Analysis of epitope overlap between antibody binding sites and T-cell recognition domains

    • In vivo models combining ARG2 vaccination and ARG2 antibody administration

  • Targeting regulatory immune cells:
    ARG2-specific T cells can recognize activated Tregs with high ARG2 expression . This suggests that ARG2 antibodies might modulate interactions between effector T cells and regulatory T cells within the tumor microenvironment.

  • Monitoring immune responses:
    ARG2 vaccination in murine tumor models has demonstrated tumor growth suppression and antitumorigenic immunomodulation . Researchers could investigate whether ARG2 antibodies enhance these effects by temporarily blocking ARG2 function while the adaptive immune response develops.

This area represents a promising frontier in ARG2-targeted therapy research, potentially combining passive immunotherapy (antibodies) with active immunomodulation (T-cell responses).

What are the most promising strategies for optimizing the therapeutic potential of ARG2 antibodies?

Several strategic approaches show particular promise for enhancing the therapeutic utility of ARG2 antibodies:

  • Combination therapies:
    ARG2 antibodies may synergize with other immunotherapeutic approaches, such as:

    • Immune checkpoint inhibitors that target complementary immunosuppressive pathways

    • Therapeutic vaccines that induce ARG2-specific T-cell responses

    • Other metabolic modulators that affect amino acid availability in the tumor microenvironment

  • Antibody engineering beyond affinity:
    Advanced engineering approaches can optimize:

    • Tissue penetration and distribution within solid tumors

    • Half-life and stability under physiological conditions

    • Fc-mediated effector functions that might contribute to therapeutic activity

  • Targeting multiple ARG2-expressing cell types:
    Comprehensive approaches addressing ARG2 in diverse cellular sources:

    • Cancer cells with endogenous ARG2 expression

    • Regulatory T cells with high ARG2 expression

    • Immunosuppressive myeloid populations (TAMs, MDSCs)

    • Cancer-associated fibroblasts expressing ARG2

  • Biomarker-guided therapy:
    Development of companion diagnostics to identify:

    • Tumors with high ARG2 expression

    • Patients likely to benefit from ARG2 inhibition

    • Early response indicators for ARG2 antibody therapy

These multifaceted approaches will likely be necessary to maximize the impact of ARG2 antibodies in clinical settings, addressing both the direct effects of ARG2 inhibition and the broader immunomodulatory consequences in the tumor microenvironment.

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.