ARGOS Antibody

Shipped with Ice Packs
In Stock

Description

Overview of AGS-009 (ARGOS Antibody)

AGS-009, developed by Argos Therapeutics (now part of Precision BioSciences), is a humanized immunoglobulin G4 (IgG4) monoclonal antibody (mAb) designed to neutralize interferon-alpha (IFN-α), a cytokine implicated in autoimmune diseases such as systemic lupus erythematosus (SLE) . Unlike traditional immunotherapies, AGS-009 specifically targets IFN-α, a driver of inflammatory pathways in lupus, aiming to restore immune balance .

Mechanism of Action

Structure and Function:

  • IgG4 subclass: Engineered to minimize immune activation while retaining high specificity for IFN-α .

  • Target binding: Binds and neutralizes IFN-α, inhibiting its interaction with cell-surface receptors and downstream pro-inflammatory signaling .

  • Therapeutic rationale: Overexpression of IFN-α in SLE correlates with disease severity, making it a critical therapeutic target .

Phase 1 Trial (NCT01242018)

A double-blind, placebo-controlled, single-dose escalation study evaluated safety and pharmacokinetics in 24 mild-to-moderate SLE patients :

ParameterDetails
Design6 cohorts (3:1 randomization of AGS-009 vs. placebo per cohort)
Dose RangeSingle intravenous doses escalated across cohorts
Primary EndpointSafety and tolerability (dose-limiting toxicities)
Secondary EndpointsPharmacokinetics, pharmacodynamics, anti-drug antibodies
Key FindingsNo dose-limiting toxicities reported; trial progressed to higher doses

Funding: Supported by a $1 million grant from the Alliance for Lupus Research (2005) .

Comparative Analysis of Argos Therapeutics’ Pipeline

While AGS-009 targeted autoimmune disease, Argos’ broader pipeline included:

CompoundTargetIndicationPhaseKey Outcome
AGS-004HIV-specific antigensHIV eradicationPhase 2bInduced memory T-cell responses but missed primary endpoint
AGS-003Tumor antigensMetastatic renal cell carcinomaPhase 330-month median survival in Phase 2; Phase 3 ongoing

Challenges and Future Directions

  • AGS-009: Limited subsequent data since the Phase 1 trial (2010) suggests potential discontinuation or strategic prioritization of other assets like AGS-003 .

  • Scientific relevance: IFN-α remains a validated target, with competing therapies like anifrolumab (anti-IFNAR mAb) achieving FDA approval for SLE in 2021 .

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
ARGOS antibody; At3g59900 antibody; F24G16.170Protein AUXIN-REGULATED GENE INVOLVED IN ORGAN SIZE antibody; AtARGOS antibody
Target Names
ARGOS
Uniprot No.

Target Background

Function
This antibody promotes cell proliferation-dependent organ growth. It plays a role in the AXR1-dependent auxin signaling pathway, which requires ANT during organogenesis.
Gene References Into Functions
  1. Overexpression of maize ARGOS1 (ZmARGOS1), ZmARGOS8, Arabidopsis ARGOS homolog ORGAN SIZE RELATED1 (AtOSR1), and AtOSR2 reduced plant sensitivity to ethylene, leading to enhanced drought tolerance. PMID: 26220950
  2. Research suggests that ARL functions downstream of BRI1 and partially mediates brassinosteroid-related cell expansion signals during organ growth. [ARGOS-LIKE] [ARL] PMID: 16824178
Database Links

KEGG: ath:AT3G59900

STRING: 3702.AT3G59900.1

UniGene: At.34537

Protein Families
Plant organ size related (OSR) protein family
Subcellular Location
Membrane; Multi-pass membrane protein. Nucleus. Cytoplasm. Endoplasmic reticulum.
Tissue Specificity
Mostly expressed in flowers, inflorescence stems, leaf primordia and young leaves, and, to a lower extent, in siliques, cotyledon vascular bundles, roots (pericycle and root tips) and mature leaves.

Q&A

What is the Arcelis® Technology Platform?

The Arcelis® technology platform is a fully personalized immunotherapy technology developed by Argos Therapeutics that captures mutated and variant antigens specific to each patient's disease. This platform is designed to overcome immunosuppression by producing durable memory T-cell responses without adjuvants that might be associated with toxicity . The technology serves as the foundation for several therapeutic candidates, including AGS-003 for cancers and AGS-004 for HIV treatment, both utilizing patient-specific immune mechanisms to target disease manifestations . The platform's distinctive feature lies in its ability to customize immune responses to each patient's unique disease variants, which has been recognized through patent protection granted by the U.S. Patent and Trademark Office . This personalized approach represents a significant advancement in immunotherapy, as it addresses the heterogeneity of diseases like cancer and HIV, where genetic variations between patients can significantly impact treatment efficacy.

How Are Dendritic Cells Utilized in ARGOS Immunotherapies?

In ARGOS immunotherapies, particularly AGS-004, dendritic cells serve as the foundation for creating personalized treatments through a sophisticated manufacturing process. These dendritic cells are first obtained from patients through leukapheresis, then processed using the Arcelis® platform to develop an individualized therapy . The manufacturing process involves co-electroporating these cells with in vitro transcribed RNA encoding autologous HIV antigens derived from the patient's viremic plasma collected before antiretroviral therapy initiation . Additionally, the cells are co-electroporated with CD40L, which induces secretion of IL-12, a cytokine essential for generating CD8+ memory T-cell responses . This process effectively transforms the patient's own dendritic cells into specialized antigen-presenting cells capable of stimulating specific immune responses against their unique viral variants. The personalized nature of this approach is particularly valuable for addressing diseases like HIV, where the virus rapidly mutates and creates a heterogeneous population of variants within each patient, requiring tailored therapeutic approaches rather than one-size-fits-all solutions.

What Is the Mechanism of Action for AGS-009 in Lupus Treatment?

AGS-009 is a humanized immunoglobulin G4 (IgG4) monoclonal antibody developed specifically for treating systemic lupus erythematosus (SLE) through a targeted immunomodulatory approach. The antibody functions by binding and neutralizing human interferon alpha (IFN-alpha), a cytokine implicated in the pathogenesis of SLE . This mechanism addresses a critical disease pathway, as interferon alpha is often overexpressed in lupus patients and contributes to the aberrant immune activation characteristic of the disease. In clinical development, AGS-009 has progressed to Phase 1 clinical trials, structured as a double-blind, placebo-controlled, single dose escalation study in patients with mild to moderate SLE . The trial design includes six planned single dose levels, with careful monitoring for safety, pharmacokinetics, pharmacodynamics, and preliminary bioactivity indicators over a 12-week period following administration . The primary endpoint focuses on dose-limiting toxicities, which helps determine the appropriate progression through escalating dose levels while balancing efficacy with safety concerns. This targeted approach to neutralizing IFN-alpha represents a promising strategy for addressing the underlying immune dysregulation in lupus, potentially offering advantages over broader immunosuppressive therapies.

How Can Researchers Assess Immunogenicity of ARGOS Antibody Therapies?

Researchers can implement a multi-faceted approach to assess the immunogenicity of ARGOS antibody therapies, focusing on quantifiable immune cell responses and functional outcomes. The primary methodology involves measuring multi-functional HIV-1 specific effector/memory T-cells using multi-color flow cytometry, as demonstrated in the AGS-004 clinical trials . This technique allows for precise quantification of immune responses, with a positive response defined as greater than 2-fold increase from baseline in the number of multi-functional HIV-1 specific effector/memory T-cells . Additionally, researchers should implement comprehensive immune profiling using specialized assays such as Adaptive Biotechnologies' immunoSEQ platform, which provides high-throughput immune receptor repertoire characterization and advanced bioinformatics analysis . This approach enables detailed characterization of the transfer of specific genetic signatures from the patient's disease through the therapeutic to the resulting immune response . For monitoring therapeutic effects on viral reservoirs, researchers should quantify integrated viral DNA in target cells, as this measurement provides insights into the therapy's impact on persistent infection reservoirs, which is particularly relevant for HIV treatments . Taken together, these methodologies offer a robust framework for assessing both the magnitude and quality of immune responses induced by ARGOS antibody therapies.

What Methodologies Are Used to Evaluate T-cell Responses to AGS-004 Therapy?

Evaluation of T-cell responses to AGS-004 therapy employs sophisticated immunological techniques that capture both quantitative and qualitative aspects of the immune response. Multi-color flow cytometry serves as the cornerstone methodology, allowing researchers to identify and enumerate multi-functional HIV-1 specific effector/memory T-cells with great precision . This technique enables simultaneous assessment of multiple cellular markers and cytokine production, providing insights into the functional quality of the induced immune response beyond simple cell counting. The analysis typically focuses on establishing a baseline measurement before treatment initiation, followed by sequential assessments after 3-4 doses of AGS-004 to track the evolution of the immune response over time . Advanced bioinformatic tools complement these laboratory techniques by helping characterize the immune response patterns and identify correlations with clinical outcomes . Additionally, researchers employ methods to measure the latent viral reservoir by quantifying CD4+ T-cells with integrated HIV DNA, which allows for correlation between T-cell responses and potential impacts on the viral reservoir . This comprehensive approach not only determines whether an immune response has been generated but also characterizes its functional quality and potential clinical relevance, providing crucial insights for therapy optimization and patient stratification in clinical trials.

How Can Researchers Analyze the Impact of AGS-004 on the Latent HIV Reservoir?

Analyzing AGS-004's impact on the latent HIV reservoir requires a comprehensive approach combining molecular, cellular, and clinical methodologies. Researchers should quantify CD4+ T-cells with integrated HIV DNA using PCR-based techniques before and after treatment to directly measure changes in the viral reservoir size . This quantification should be correlated with the development of antiviral memory T-cell responses, as demonstrated in clinical studies where patients with positive antiviral memory T-cell responses showed significantly fewer CD4+ T-cells with integrated HIV DNA compared to non-responders . Clinical researchers can implement analytical treatment interruption (ATI) protocols under careful monitoring to assess the functional impact on viral control, measuring time to viral rebound and viral load set points after interruption . The median time to viral rebound (approximately 29 days in previous studies) serves as a quantifiable metric for comparing reservoir control between treatment and control groups . Additionally, researchers should employ Adaptive Biotechnologies' immunological sequencing technologies to characterize "the transfer of specific genetic signatures from each patient's HIV, through AGS-004, to a specific immune response against each patient's unique virus population" . This multidimensional analysis provides a comprehensive understanding of how AGS-004 therapy impacts not only the size but also the reactivation potential of the latent HIV reservoir, offering insights into mechanisms of action and potential optimization strategies.

What Factors Contribute to the Physicochemical Properties of ARGOS Antibodies?

The physicochemical properties of ARGOS antibodies are determined by multiple structural and biochemical factors that collectively influence their developability and therapeutic efficacy. The antibody's structure directly affects key attributes including homogeneity, stability, solubility, and specificity—all essential determinants of developability-related features such as appearance, expression level, yield, and purity . For AGS-009, its humanized immunoglobulin G4 (IgG4) structure was specifically selected to optimize binding and neutralization of human interferon alpha while potentially minimizing unwanted effector functions that could lead to adverse events . Post-translational modifications, particularly glycosylation patterns, significantly impact antibody function and stability, requiring careful characterization during development . The high mutation rate of HIV presents unique challenges for AGS-004, necessitating Argos's strain-independent amplification technology to produce large amounts of variant antigens from small patient samples, enabling the production of immunotherapies matched to each patient's viral "swarm" . Computational tools, including artificial intelligence-aided prediction of protein structures and features, are increasingly employed to predict, screen, and optimize antibody candidates' developability, substantially reducing the risk of advancing suboptimal candidates to development stages . Additionally, the expected clinical indication and planned dosage and administration route must be considered when establishing developability criteria, as these factors influence the relative importance of specific physicochemical properties in different therapeutic contexts .

What Experimental Design Considerations Are Important When Testing ARGOS Therapies in Clinical Trials?

When designing clinical trials for ARGOS therapies, researchers must implement rigorous methodological approaches that address the unique aspects of personalized immunotherapies. For monoclonal antibody therapies like AGS-009, a dose-escalation design with careful safety monitoring is essential, as exemplified by the Phase 1a trial that employed a double-blind, placebo-controlled approach with six planned single dose levels and a 3:1 randomization ratio of active treatment to placebo . This design allows for systematic assessment of dose-limiting toxicities while gathering preliminary efficacy data. For dendritic cell-based therapies like AGS-004, incorporating appropriate immunological endpoints is crucial, with positive immune response typically defined as a greater than 2-fold increase from baseline in multi-functional HIV-specific effector/memory T-cells . Clinical trials must also include adequate follow-up periods to capture both immediate immune responses and longer-term impacts on disease parameters, with AGS-004 HIV trials implementing 12-week monitoring periods to assess virologic control after treatment interruption . The incorporation of analytical treatment interruption (ATI) protocols, while ethically complex, provides valuable information about the therapy's ability to control viral rebound in the absence of antiretroviral therapy, serving as a critical functional endpoint . Additionally, researchers should consider stratifying patients based on disease characteristics, particularly for HIV studies where viral diversity, reservoir size, and immune status significantly impact treatment response. This comprehensive approach to clinical trial design enables both safety assessment and mechanistic insights into how ARGOS therapies modulate immune responses in different patient populations.

How Can Researchers Optimize the Production of Personalized ARGOS Therapies?

Optimizing the production of personalized ARGOS therapies requires addressing the complex challenges associated with patient-specific manufacturing while maintaining consistent quality and efficacy. Researchers should implement standardized collection protocols for patient materials, with particular attention to timing for conditions like HIV where capturing the diverse viral quasispecies prior to antiretroviral therapy initiation is crucial . The leukapheresis procedure for collecting dendritic cell precursors must be optimized to yield sufficient high-quality monocytes while minimizing patient discomfort, typically requiring processing of 1.5-2 blood volumes to obtain adequate cell numbers . The electroporation parameters for RNA loading must be carefully calibrated to achieve efficient transfection without compromising cell viability, with optimization studies determining the optimal voltage, pulse duration, and cell concentration for each product . Quality control measures should include assessment of dendritic cell phenotype (typically by flow cytometry), RNA incorporation efficiency, and functional assays to confirm the cells' ability to stimulate appropriate T-cell responses in vitro before administration . Researchers should also explore innovations in cryopreservation and storage to extend product shelf-life while maintaining cellular functionality, potentially enabling more flexible treatment schedules. Additionally, implementation of advanced bioinformatic tools for antigen selection can enhance therapeutic potency by identifying the most immunogenic epitopes from patient-derived sequences, particularly important for highly variable pathogens like HIV . These methodological refinements collectively contribute to more consistent, potent, and clinically practical personalized immunotherapies.

How Can Computational Tools Enhance ARGOS Antibody Development?

Computational tools offer transformative potential for enhancing ARGOS antibody development through multiple sophisticated applications that span the discovery-to-development continuum. With the advancement of artificial intelligence-aided prediction of protein structures and features, researchers can now predict, screen, and optimize the developability of antibody candidates, substantially reducing the risk of advancing suboptimal candidates to the development stage . These computational approaches enable high-throughput virtual screening of antibody candidates based on their predicted physicochemical properties, including homogeneity, stability, solubility, and specificity—key attributes that determine developability-related features such as appearance, expression level, yield, and purity . For highly variable pathogens like HIV, bioinformatic tools can analyze patient-derived viral sequences to identify conserved epitopes or recurrent mutations across the viral quasispecies, informing more effective antigen selection for personalized immunotherapies . Companies like Adaptive Biotechnologies provide specialized immune profiling capabilities and bioinformatic tools that characterize the immune response patterns following ARGOS therapy administration, offering insights into efficacy mechanisms and potential optimization strategies . These tools can track "the transfer of specific genetic signatures from each patient's HIV, through AGS-004, to a specific immune response against each patient's unique virus population," enabling unprecedented clarity in understanding the precision of immunotherapeutic approaches . Furthermore, machine learning algorithms can integrate complex datasets from clinical trials to identify biomarkers predictive of treatment response, potentially enabling patient stratification and personalized treatment protocols that maximize therapeutic benefits while minimizing adverse effects.

What Are the Implications of ARGOS Technology for Treating Other Diseases?

The ARGOS technology platform, particularly the Arcelis® approach, offers profound implications for treating various diseases beyond its current applications in HIV and lupus. The platform's fundamental ability to capture mutated and variant antigens specific to each patient's disease creates opportunities for personalized interventions across a spectrum of conditions characterized by heterogeneity and immune dysregulation . For autoimmune disorders beyond lupus, the success of AGS-009 in targeting interferon alpha suggests similar approaches could be developed for conditions like rheumatoid arthritis, multiple sclerosis, or psoriasis, where specific cytokine pathways drive pathogenesis . The dendritic cell-based technology used in AGS-004 has significant potential for treating persistent viral infections beyond HIV, such as hepatitis B, hepatitis C, or human papillomavirus, where viral mutation and immune evasion present major therapeutic challenges . Cancer immunotherapy represents another promising application area, with ongoing development of AGS-003 leveraging the platform's capacity to induce tumor-specific immune responses based on each patient's unique tumor antigens . The integration of Argos's dendritic cell expertise with monoclonal antibody technology could enable novel combination approaches that simultaneously target multiple aspects of disease pathophysiology . Furthermore, the strain-independent amplification technology developed for HIV applications could be adapted to address other highly variable pathogens, including influenza, dengue, or emerging viral threats where rapid mutation complicates vaccine and therapeutic development . These diverse applications highlight how the fundamental principles of ARGOS technology—personalization, immune modulation, and precision targeting—could transform treatment paradigms across multiple disease areas, potentially addressing conditions where conventional approaches have shown limited efficacy.

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.