ATL40 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of Anti-CD40 Antibodies

Anti-CD40 antibodies are monoclonal antibodies (mAbs) targeting CD40, a tumor necrosis factor receptor (TNFR) superfamily member expressed on antigen-presenting cells (APCs), B cells, and certain tumor cells . These antibodies are classified into two functional categories:

  • Agonists: Activate CD40 signaling to enhance anti-tumor immunity .

  • Antagonists: Block CD40/CD40 ligand (CD40L) interaction to suppress autoimmune responses .

Agonistic Anti-CD40 Antibodies

  • Immune Activation: Mimic CD40L to license dendritic cells (DCs), promoting cytotoxic T lymphocyte (CTL) responses .

  • Direct Tumor Effects: Induce apoptosis in CD40-expressing malignancies (e.g., lymphoma, carcinoma) .

  • FcγR Dependency: Most require Fcγ receptor (FcγR) binding for optimal agonism, though hIgG2 isotype enables FcγR-independent activity .

Antagonistic Anti-CD40 Antibodies

  • Immune Suppression: Block CD40L interaction, inhibiting B cell activation and germinal center formation .

  • Fc Engineering: Fc-silent variants (e.g., IgG4) avoid FcγR-mediated adverse effects (e.g., thrombocytopenia) .

Table 1: Functional Comparison of Anti-CD40 Antibodies

PropertyAgonistic (e.g., ChiLob7/4)Antagonistic (e.g., KPL-404)
IsotypeIgG1, hIgG2IgG4, Fc-silent mutants
Primary UseCancer immunotherapyAutoimmune diseases
Key MechanismAPC activation, tumor apoptosisCD40/CD40L blockade
FcγR InteractionRequired (except hIgG2)Avoided
Clinical PhasePhase I/II Phase II

Cancer Immunotherapy

  • ChiLob7/4 (Agonist):

    • Phase I Trial: Doses up to 200 mg (MTD) showed B cell depletion, NK cell activation, and disease stabilization in 15/29 patients .

    • Combination Therapy: Synergizes with anti-CTLA-4 or chemotherapy .

Autoimmune Diseases

  • KPL-404 (Antagonist):

    • Inhibits B cell activation in Sjögren’s syndrome (SjS) and systemic lupus erythematosus (SLE) without internalization or cytokine release .

    • No depletion of CD40⁺ cells due to Fc silencing .

Isotype Switching

  • hIgG2 Conversion: Antagonists (e.g., 341G2) transformed into super-agonists via hIgG2 hinge engineering, enabling potent tumor control without FcγR .

  • Mechanistic Insight: Disulfide bonding in hIgG2 facilitates antibody clustering, enhancing CD40 agonism .

Biomarker Correlations

  • B Cell Counts: Agonists reduce peripheral CD19⁺ B cells by >90% at therapeutic doses .

  • Cytokine Release: MIP-1β and IL-12 elevation correlates with dose-dependent immune activation .

Future Directions

  • Next-Gen Agonists: FcγR-independent hIgG2 variants to minimize systemic toxicity .

  • Combination Regimens: Pairing with checkpoint inhibitors (e.g., anti-PD-1) or vaccines .

  • Biomarker-Driven Trials: Stratification by CD40 expression or tumor microenvironment signatures .

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
ATL40; At2g42350; MHK10.7; RING-H2 finger protein ATL40; RING-type E3 ubiquitin transferase ATL40
Target Names
ATL40
Uniprot No.

Target Background

Database Links

KEGG: ath:AT2G42350

STRING: 3702.AT2G42350.1

UniGene: At.42716

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

Q&A

What is ABvac40 and how does it target Alzheimer's disease pathology?

ABvac40 is an active vaccine specifically designed to target the C-terminal end of Aβ40 in Alzheimer's disease (AD) patients. This approach builds on research demonstrating that specific anti-Aβ40 antibodies label intra- and extra-neuronal neurofibrillary tangles (NFTs) in critical brain regions affected by AD, particularly the entorhinal cortex and hippocampus. Importantly, these antibodies identify degenerating neuronal populations containing C-terminal fragments of Aβx-40 that do not co-localize with tau NFTs, suggesting they target distinct pathological features in AD brains . The mechanistic hypothesis centers on the ability of ABvac40 to induce the production of antibodies that specifically recognize and potentially clear these pathological Aβ40 forms.

How is immunogenicity measured for ABvac40 in clinical trials?

Immunogenicity of ABvac40 is assessed through a rigorous methodology that quantifies specific anti-Aβ40 antibodies in plasma. The procedure involves comparing baseline plasma samples with post-treatment samples using enzyme-linked immunosorbent assay (ELISA) techniques. Patients are classified as positive responders if their plasma samples at a 1:10 dilution show signal increments of ≥3 standard deviations compared to baseline, and if this increment is reduced by ≥50% in pre-adsorbed samples. Additionally, researchers express biological activity quantitatively through antibody titration, defined as the inverse of the maximal plasma sample dilution showing an increase in mean optical density (MOD) ≥3 standard deviations relative to baseline .

What experimental validation confirms the specificity of ABvac40-induced antibodies?

The specificity of antibodies induced by ABvac40 vaccination is validated through multiple experimental approaches. Researchers assess the reactivity of plasma samples with amyloid plaques through immunohistochemistry using brain sections from both APP/PS1-transgenic mice and AD patients. This involves using plasma samples (diluted 1:500 in 0.5% Triton X-100 PBS) as primary antibodies. Additionally, the ability of ABvac40-raised antibodies to target different forms of Aβ is analyzed using immunoblotting techniques . These complementary approaches verify that the immune response generated is indeed specific to the pathological forms of Aβ40 that are relevant to Alzheimer's disease mechanisms.

How should dose-escalation protocols be implemented for novel antibody therapies like ABvac40?

For novel antibody therapies like ABvac40, a stepped recruiting protocol with careful dose escalation represents best practice for first-in-human studies. The ABvac40 phase I trial exemplifies this approach: the first group of patients received half-doses administered sequentially on consecutive days, with careful safety monitoring after the second injection before proceeding. Only after this initial group completed safety assessments did researchers proceed with full-dose administration in subsequent groups. This phased approach minimizes risk while allowing for interim analysis of safety and immunogenicity data . Researchers implementing similar protocols should build in predefined safety checkpoints and decision trees for dose escalation based on observed adverse events and preliminary efficacy signals.

What protocol amendments may become necessary during antibody therapy trials?

Based on the ABvac40 trial experience, researchers should anticipate potential protocol amendments following interim analyses. In the ABvac40 study, after the first eight patients completed safety assessments following their second dose, an interim analysis of the immune response led to a protocol amendment introducing a third immunization . This highlights the importance of building flexibility into trial designs, particularly for novel antibody therapies where optimal dosing regimens may not be fully established pre-trial. Potential amendments may include: (1) adjusting the number of immunizations based on antibody titer responses; (2) modifying dosing intervals; and (3) extending follow-up periods to capture longer-term immune responses. All amendments should be implemented while maintaining study blinding and statistical integrity.

How should antibody panel design be optimized for monitoring immune responses to therapies like ABvac40?

Optimizing antibody panel design for monitoring immune responses requires a systematic approach based on specific research questions. First, researchers must clearly define their biological hypothesis and identify which cell populations need to be characterized. For ABvac40 immune monitoring, panels should be designed to detect both T and B cell responses to the vaccine. Start by selecting markers based on their expression levels (low vs. high) and match them appropriately with fluorophores - pairing low-expressed antigens with bright fluorophores and high-expressed antigens with dimmer fluorophores . Avoid selecting similar fluorophores for co-expressed markers to prevent spectral overlap issues. Consider instrument limitations and autofluorescence characteristics of the cells being studied, particularly important when analyzing elderly AD patient samples which may have higher autofluorescence backgrounds.

What strategies can improve detection sensitivity when measuring antibody responses in clinical samples?

To improve detection sensitivity when measuring antibody responses to therapies like ABvac40, implement a multi-faceted approach focusing on both experimental design and technical optimization. Begin by establishing a precise gating strategy that includes: size/shape discrimination (FSC vs. SSC), doublet exclusion (Area vs. Height), dead cell exclusion, and sequential marker identification (e.g., CD45+ → CD3hi → CD4+/CD8+) . To maximize detection sensitivity, select fluorophores based on their staining index (a measurement of brightness) and ensure optimal antibody concentrations through titration experiments. When measuring rare antibody-producing cells, use fluorophores with minimal spectral overlap to reduce background and improve signal-to-noise ratios. Finally, implement standardized controls for each experimental run, including fluorescence-minus-one (FMO) controls to establish accurate gating boundaries for weakly expressed markers.

How can researchers develop custom antibody libraries for rapid detection of specific targets?

Researchers can develop custom antibody libraries using the DT40 cell line approach, which offers advantages for rapid antibody generation against specific targets like Aβ40. The DT40 chicken lymphoma cell line naturally produces IgM-type antibodies and undergoes gene conversion and somatic mutations in the variable region of the immunoglobulin gene during culture, creating a natural antibody library . To control this process, researchers should consider generating an AID-inducible DT40 cell line where the endogenous activation-induced cytidine deaminase (AID) gene is knocked out using CRISPR/Cas9 and replaced with an inducible AID gene using systems like Tet-Off . This allows precise regulation of hypermutation: activating it to increase antibody diversity during selection phases and suppressing it to fix high-affinity antibodies once identified. For Aβ40-specific libraries, researchers should immunize with the C-terminal epitope of Aβ40 prior to B cell isolation to enrich the starting material before library creation.

What criteria define a positive immunological response to ABvac40 therapy?

A positive immunological response to ABvac40 therapy follows strictly defined criteria established in clinical protocols. According to the phase I trial methodology, patients are classified as positive responders if their plasma samples at a 1:10 dilution demonstrate a signal increment at any post-treatment visit that is ≥3 standard deviations above baseline values. Additionally, this increment must be reduced by ≥50% in pre-adsorbed samples, confirming specificity of the response . Beyond this binary classification, researchers quantify response magnitude through antibody titration, defining titers as the inverse of the maximal plasma sample dilution showing an increase in mean optical density ≥3 standard deviations relative to baseline. These standardized criteria ensure consistent interpretation across different patients and research centers, enabling reliable assessment of immunogenicity.

How should researchers interpret heterogeneity in antibody responses among clinical trial participants?

When interpreting heterogeneity in antibody responses to therapies like ABvac40, researchers should conduct multi-parameter analyses considering demographic, genetic, and clinical factors. First, analyze demographic variables including age, sex, concomitant medications, and disease severity (such as MMSE scores in Alzheimer's studies) to identify potential correlation patterns . Genetic factors, particularly APOE genotype, should be examined as potential modulators of antibody response magnitude. Longitudinal analysis of antibody titers can reveal different response patterns - including rapid responders, delayed responders, and non-responders - which may have distinct clinical implications. When comparing treatment groups, researchers should employ appropriate statistical methods for repeated measures while accounting for baseline characteristics. Finally, correlate antibody response patterns with any available biomarker data (CSF Aβ40/Aβ42 ratios, neuroimaging) to assess potential relationships between immunological response and disease-modifying effects.

What analytical approaches can distinguish antibody-specific effects from placebo responses?

To distinguish antibody-specific effects from placebo responses in trials like the ABvac40 study, researchers should implement a comprehensive analytical framework. Begin with a robust randomization procedure and maintain double-blinding throughout the study, as demonstrated in the ABvac40 trial where "patients, carers and investigators were blind to treatment allocation" . Statistically, compare immunological parameters between treatment and placebo groups using both intention-to-treat (ITT) and per-protocol (PP) populations to ensure consistency of findings. For laboratory assessments of antibody specificity, implement pre-adsorption tests that can confirm target binding is truly specific to Aβ40. Additionally, conduct immunohistochemistry validation using brain sections from both animal models and human AD patients to verify that antibodies from treated patients, but not placebo recipients, recognize pathological features . Temporal analysis comparing the emergence of antibody responses with any clinical changes can further strengthen causality arguments.

How might experimental design evolve in Phase II trials of ABvac40?

Building on phase I results demonstrating that "ABvac40 showed a favourable safety and tolerability profile while eliciting a consistent and specific immune response" , phase II experimental design should evolve to incorporate several key methodological advancements. First, implement a stratified randomization approach accounting for APOE genotype and baseline disease severity to ensure balanced distribution of these potentially important variables. Extend the treatment duration to assess long-term antibody persistence and potential tachyphylaxis effects. Incorporate comprehensive biomarker assessments including PET amyloid imaging, CSF Aβ40/Aβ42 ratios, and markers of neurodegeneration to establish potential surrogate endpoints. Introduce cognitive assessment tools with greater sensitivity to detect subtle changes in early disease stages. Finally, consider adaptive design elements that allow for dose adjustments based on interim analyses of antibody titers and preliminary efficacy signals, while maintaining study blinding and statistical integrity.

What methodological approaches could enhance the production of therapeutic antibodies against Aβ40?

Advanced methodological approaches to enhance therapeutic antibody production against Aβ40 could leverage innovations in cell line engineering and selection techniques. The DT40 cell line system represents a promising platform due to its natural capacity for gene conversion and somatic hypermutation in the immunoglobulin variable region . Researchers could generate specialized DT40 cell lines with regulated activation-induced cytidine deaminase (AID) expression using inducible systems like Tet-Off, allowing precise control over hypermutation processes . This approach enables both diversification of the antibody repertoire and fixation of high-affinity clones once identified. For clinical applications, selected antibody sequences from this system could be reformatted into humanized or fully human constructs with optimized effector functions. Additionally, researchers should explore bispecific antibody formats that simultaneously target Aβ40 and facilitate blood-brain barrier penetration to enhance central nervous system exposure and efficacy.

How can flow cytometry panel design principles improve immunological monitoring in ABvac40 trials?

Implementing advanced flow cytometry panel design principles can significantly enhance immunological monitoring in ABvac40 clinical trials. Begin by clearly defining the research question and biological hypothesis, then identify all relevant cell populations requiring characterization . For ABvac40 specifically, panels should be designed to comprehensively assess both B cell (antibody-producing) and T cell (helper) responses to vaccination. Select markers based on their expression levels, matching low-expressed antigens with bright fluorophores and high-expressed antigens with dimmer fluorophores . Develop optimized multi-parameter panels by avoiding similar fluorophores on co-expressed markers and considering the autofluorescence characteristics of patient samples. Implement standardized quality control procedures including fluorescence compensation controls, FMO controls, and assay standardization across multiple timepoints and clinical sites. Finally, employ advanced computational analysis approaches including high-dimensional clustering algorithms to identify potentially relevant cell subpopulations that may correlate with antibody response magnitude or clinical outcomes.

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.