CRK40 Antibody

Shipped with Ice Packs
In Stock

Description

Potential Explanations for Terminology Confusion

Hypothesis 1: The term may involve a typographical error. "CRKL" (v-crk sarcoma virus CT10 oncogene homolog-like) is a documented adaptor protein with phosphorylation sites studied in hematologic malignancies (e.g., Tyr207 phosphorylation analyzed using Phospho-CrkL Antibody #3181 and PE Mouse anti-CrkL (pY207) ).

Hypothesis 2: The query may conflate terminology for anti-CD40 antibodies, which are extensively studied immunotherapies. Key examples include:

Antibody NameIsotypeMechanismClinical PhaseKey Findings
CP-870,893IgG2AgonistPhase I/II20% PR in advanced melanoma at 0.2 mg/kg
Selicrelumab (2141-V11)IgG1FcγRIIB-enhanced agonistPhase IImproved tumor control via myeloid cell activation
Chi Lob 7/4IgG1AgonistPhase IActivated B/NK cells with 37-month disease stabilization in 15/29 patients

Mechanism of Action

  • Agonistic anti-CD40 mAbs activate antigen-presenting cells through TNF receptor superfamily signaling, enhancing T-cell priming .

  • Antagonistic anti-CD40 mAbs inhibit CD40/CD40L interaction to suppress autoimmunity .

Emerging Technologies

Novel bispecific CD40 agonists (e.g., BiA9*2_HF) demonstrate:

  • 5.1 nM affinity for human CD40

  • 3-fold reduction in hepatotoxicity vs. selicrelumab

  • Modular antigen delivery capability via antibody-peptide conjugation

Recommended Actions

  1. Verify antibody nomenclature (potential CD40 vs. CRKL/CrkL confusion)

  2. Explore CD40-targeting clinical candidates:

    • APX005M: IgG1 agonist in Phase II NSCLC trials (NCT03123783)

    • ADC-1013: IgG1 with 87% DC activation at 0.3 mg/kg

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CRK40 antibody; At4g04570 antibody; F4H6.9Cysteine-rich receptor-like protein kinase 40 antibody; Cysteine-rich RLK40 antibody; EC 2.7.11.- antibody
Target Names
CRK40
Uniprot No.

Target Background

Database Links

KEGG: ath:AT4G04570

STRING: 3702.AT4G04570.1

UniGene: At.33996

Protein Families
Protein kinase superfamily, Ser/Thr protein kinase family, CRK subfamily
Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What is an anti-CD40 antibody and what role does it play in cancer immunotherapy?

Anti-CD40 antibodies are monoclonal antibodies designed to target and bind to CD40, a costimulatory protein found on antigen-presenting cells and various tumor cells. In cancer immunotherapy, agonistic anti-CD40 antibodies function by activating CD40 signaling pathways, which stimulates both innate and adaptive immune responses against tumors. These antibodies have demonstrated impressive antitumor effects in preclinical models when combined with chemotherapy, radiotherapy, or other immunotherapies . Unlike checkpoint inhibitors that block inhibitory signals, agonistic anti-CD40 antibodies actively trigger immune activation, necessitating careful dosing to balance efficacy and toxicity .

What are the main mechanisms of action for agonistic anti-CD40 antibodies?

Agonistic anti-CD40 antibodies operate through multiple mechanisms:

  • T-cell dependent antitumor immunity: CD40 ligation enhances antigen presentation and costimulatory molecule expression on dendritic cells, promoting effective T-cell priming and activation .

  • T-cell independent mechanisms: These include reprogramming of tumor-associated macrophages into antitumor phenotypes, affecting the tumor microenvironment independent of T-cell function .

  • Direct cytotoxic effects: In CD40-expressing tumors, these antibodies can directly trigger apoptotic pathways .

  • Cytokine release induction: CD40 stimulation leads to production of inflammatory cytokines that support antitumor immune responses, including IL-12 and MIP-1β .

How do researchers evaluate the efficacy of anti-CD40 antibodies in experimental models?

Researchers employ several methodological approaches to assess anti-CD40 antibody efficacy:

  • Tumor growth monitoring: Measurement of tumor volume changes in murine models following antibody administration, particularly in combination with other therapies.

  • Flow cytometry analysis: Quantification of immune cell populations and activation status. Key markers include B-cell depletion (CD19+), NK cell activation (CD54 expression), and T-cell responses .

  • Cytokine profiling: Measurement of inflammatory cytokines in plasma using technologies like Luminex to assess systemic immune activation .

  • Histopathological assessment: Evaluation of tissue sections for necrosis, inflammatory infiltrates, and thromboses .

  • Liver enzyme monitoring: Assessment of ALT levels to evaluate hepatotoxicity as a dose-limiting adverse effect .

What are the common adverse effects observed with anti-CD40 antibody administration?

Based on preclinical and clinical studies, researchers should anticipate and monitor:

  • Liver toxicity: The most significant dose-limiting toxicity, characterized by necroinflammatory liver disease, elevated transaminases, and in severe cases, extensive hepatic necrosis .

  • Cytokine release syndrome: Systemic inflammatory response with elevated pro-inflammatory cytokines .

  • Infusion reactions: Generally mild to moderate (Grade 1-2) at clinical doses above 16 mg, which can be mitigated with corticosteroid premedication .

  • Hematological changes: Transient reductions in B-cell and NK cell populations in peripheral blood .

  • Endothelial activation: Progressive activation of endothelial cells with expression of adhesion molecules like VCAM1, potentially contributing to thrombotic events .

How do Kupffer cells drive anti-CD40 antibody-induced liver inflammatoxicity?

Kupffer cells (liver-resident macrophages) have been identified as the central initiators of anti-CD40 antibody-induced liver toxicity through a specific sequence of events:

  • Initial Kupffer cell activation: Time-resolved single-cell RNA-sequencing reveals that Kupffer cells are the first responders to anti-CD40 stimulation, occurring within 7 hours of antibody administration .

  • Inflammatory transformation: Following activation, Kupffer cells undergo phenotypic changes characterized by upregulation of chemokines (CXCL9, CXCL10) and activation of interferon signaling pathways .

  • Monocyte recruitment: Activated Kupffer cells produce CCL2, which recruits CCR2+ inflammatory monocytes to the liver, observed at 14 hours post-treatment .

  • Neutrophil infiltration: By 22 hours post-treatment, substantial S100a8+ neutrophil recruitment occurs, further exacerbating inflammatory damage .

  • Endothelial activation: Progressive activation of liver endothelial cells with VCAM1 expression contributes to microvascular thrombosis and tissue damage .

This sequential cascade was confirmed using conditional knockout models, where Clec4f-Cre CD40 flox/flox mice (with CD40 deletion restricted to Kupffer cells) showed significant protection against liver damage while maintaining systemic inflammatory responses, demonstrating the non-redundant role of Kupffer cells in this toxicity .

What strategies can researchers employ to mitigate anti-CD40 antibody-induced liver toxicity without compromising antitumor activity?

Research has identified several innovative approaches to address this challenge:

  • Antibody-directed erythrophagocyte reprogramming: Administration of Ter119 antibody targeting red blood cells (RBCs) induces controlled erythrophagocytosis specifically in liver macrophages, reprogramming them to an anti-inflammatory phenotype (Hmox1^high/Marco^high/MHCII^low) that protects against subsequent anti-CD40-induced damage .

  • Selective Kupffer cell targeting: Since conditional deletion of CD40 in Kupffer cells prevents liver toxicity while preserving systemic inflammation, developing Kupffer cell-specific delivery systems for anti-inflammatory agents represents a promising strategy .

  • Dose optimization protocols: Clinical studies have established that dose escalation to achieve optimal pharmacokinetics (maintaining trough levels above 25 μg/mL) while monitoring liver function can maximize therapeutic window .

  • Corticosteroid premedication: For infusion reactions, single-dose corticosteroid premedication has been shown effective without significantly compromising antitumor activity .

  • Modified antibody engineering: Developing anti-CD40 antibodies with altered Fc regions or binding characteristics that maintain antitumor efficacy with reduced hepatotoxicity potential.

How does time-resolved single-cell RNA-sequencing inform our understanding of anti-CD40 antibody immunobiology?

Time-resolved scRNA-seq has provided unprecedented insights into the cellular dynamics following anti-CD40 antibody administration:

Table 1: Temporal Sequence of Cellular Events After Anti-CD40 mAb Administration

Time PointPrimary Cellular EventsKey Molecular SignaturesPathway Activation
BaselineResident Clec4f+ Kupffer cells and endothelial cellsHomeostatic markersBaseline metabolism
7 hoursInflammatory transformation of Kupffer cellsUpregulation of CXCL9, CXCL10IFN-α and IFN-γ pathways
14 hoursRecruitment of Ly6c2+ monocytesCCR2 expression on monocytesChemotaxis pathways
22 hoursInfiltration of S100a8+ neutrophils, progressive endothelial activationVCAM1 on endothelial cellsAdhesion and inflammatory pathways

This sequential analysis revealed:

  • Cellular hierarchy in inflammation: Identified Kupffer cells as the initiators rather than just participants in the inflammatory cascade .

  • Precise timing of interventions: Suggested optimal timing for prophylactic interventions before monocyte and neutrophil recruitment phases .

  • Molecular targets for intervention: Identified specific chemokine pathways (CCL2-CCR2 axis) and adhesion molecules (VCAM1) as potential therapeutic targets .

  • Endothelial activation dynamics: Revealed progressive endothelial activation as a contributor to microvascular thrombosis .

This approach demonstrates how high-dimensional single-cell analysis can dissect complex immunological processes with temporal resolution, informing rational design of combination immunotherapy protocols.

What are the optimal dosing strategies and pharmacodynamic markers for anti-CD40 antibody therapy in clinical settings?

Clinical investigations have established several key parameters for anti-CD40 antibody administration:

  • Dose range and MTD: For the chimeric anti-CD40 antibody ChiLob7/4, the maximum tolerated dose was established at 200 mg (2.1-3.3 mg/kg) administered weekly for 4 doses, with dose-limiting liver transaminase elevations occurring at 240 mg .

  • Pharmacokinetic targets: Maintaining trough levels above 25 μg/mL before subsequent doses appears optimal for biological activity .

  • Pharmacodynamic markers for efficacy monitoring:

    • Reduction of peripheral blood CD19+ B cells to ≤10% of baseline

    • NK cell activation (increased CD54 expression)

    • Plasma cytokine elevations (particularly MIP-1β and IL-12)

    • B-cell phenotypic changes (reduced CD21 expression)

  • Immunogenicity monitoring: Human anti-chimeric antibody (HACA) responses were observed at doses between 1.6-50 mg but not at higher doses, suggesting high-dose therapy may overcome immunogenicity concerns .

  • Clinical response indicators: Disease stabilization was achieved in approximately 50% of treatments (15/29), with duration ranging from 6 to 37 months, providing benchmarks for future efficacy studies .

How do conditional gene knockout models contribute to our understanding of cell-specific roles in anti-CD40 antibody mechanisms?

Conditional knockout models have been instrumental in delineating cell-specific contributions to both therapeutic efficacy and toxicity of anti-CD40 antibodies:

  • Kupffer cell-specific targeting: Clec4f-Cre CD40 flox/flox mice (with CD40 deletion restricted to Kupffer cells) demonstrated:

    • Near-complete protection against liver necrosis and ALT elevation

    • Minimal impact on systemic inflammatory cytokine production

    • Preservation of extrahepatic immune activation

  • Pan-macrophage targeting: LysM-Cre CD40 flox/flox mice (with CD40 deletion in most macrophage populations) showed:

    • Complete protection against liver toxicity

    • Significant reduction in systemic inflammatory cytokine production

    • Potential compromise of therapeutic efficacy

These models provided critical evidence that:

  • Kupffer cells are the non-redundant trigger of anti-CD40 mAb-induced liver toxicity

  • CD40 signaling in other macrophage populations contributes to systemic inflammation and potentially antitumor activity

  • Selective targeting of Kupffer cells represents a viable strategy to uncouple hepatic toxicity from beneficial antitumor immunity

This cell-specific approach exemplifies how conditional genetic models can guide the development of more precise immunotherapeutic interventions with improved therapeutic windows.

What experimental techniques are most effective for evaluating anti-CD40 antibody-induced immune activation in preclinical models?

Researchers should consider implementing a multi-modal assessment approach:

  • Flow cytometry panels for comprehensive immune profiling:

    • B-cell depletion and activation (CD19, CD21)

    • T-cell subsets and activation status (CD4, CD8, CD25, CD69)

    • Myeloid cell recruitment and polarization

    • NK cell activation (CD54 expression)

  • Single-cell RNA sequencing with temporal resolution:

    • Cell suspension preparation: CD45-enriched liver cell suspensions

    • Cell barcoding for temporal analysis

    • Bioinformatic analyses: UMAP visualization, clustering, and pathway analysis

    • Marker gene identification for cell population tracking

  • Cytokine and chemokine profiling:

    • Multiplex assays (Luminex technology)

    • Key analytes: MIP-1β, IL-12, TNF-α, IFN-γ

    • Hierarchical clustering analysis for pattern recognition

  • Histological and immunohistochemical assessment:

    • H&E staining for necrosis evaluation

    • Immunofluorescence for immune cell infiltration

    • Digital pathology quantification (QuPath)

  • Liver function monitoring:

    • ALT measurement in plasma

    • Timing: 30 hours post-administration for optimal detection

How can researchers effectively design combination therapies involving anti-CD40 antibodies?

Effective combination therapy design requires strategic consideration of:

  • Sequencing of therapeutic agents:

    • Improper administration of anti-CD40 before chemotherapy can result in lethal hepatotoxicity in mouse models

    • Optimal sequencing may involve chemotherapy/radiotherapy first, followed by anti-CD40 therapy

  • Prophylactic strategies for toxicity management:

    • Erythrophagocyte reprogramming protocol: administration of Ter119 antibody prior to anti-CD40 treatment

    • Single-dose corticosteroid premedication for infusion reactions

  • Dose optimization:

    • Sub-toxic doses in combination may yield synergistic effects

    • Monitoring of pharmacodynamic markers to confirm biological activity

  • Selection of complementary mechanisms:

    • Combining T-cell-directed therapies (checkpoint inhibitors) with innate immune activators (anti-CD40)

    • Pairing direct tumor-targeting agents with immune-enhancing antibodies

  • Comprehensive toxicity monitoring:

    • Liver function tests

    • Cytokine profiles

    • Complete blood counts

    • Coagulation parameters

What analytical approaches should be used to interpret contradictory data in anti-CD40 antibody research?

When facing contradictory results, researchers should employ these analytical strategies:

  • Cellular heterogeneity assessment:

    • Determine if apparent contradictions result from effects on different cell populations

    • Apply single-cell technologies to resolve population-level variations

  • Temporal dynamics consideration:

    • Evaluate if contradictions reflect different time points in a dynamic process

    • Implement time-course experiments to resolve temporal discrepancies

  • Dose-response relationship analysis:

    • Examine if contradictions stem from different dosing regimens

    • Conduct detailed dose-response studies spanning therapeutic and toxic ranges

  • Model system differences evaluation:

    • Compare results across different preclinical models

    • Assess translation between mouse models and human clinical data

  • Antibody structural and functional comparison:

    • Different anti-CD40 antibodies vary in isotype, epitope targeting, and agonist potency

    • Characterize the specific antibody properties that might explain divergent results

How do findings from murine models of anti-CD40 antibody therapy translate to human clinical applications?

Translational aspects to consider when moving from murine models to human applications:

  • Toxicity profiles:

    • Murine models: High-dose (5-20 mg/kg) intravenous administration causes severe necroinflammatory liver injury

    • Human trials: Even low-dose (0.1-0.2 mg/kg) administration results in mild to moderate transaminase elevations

  • Effective dosing:

    • Maximum tolerated dose in humans: 200 mg for ChiLob7/4 (equivalent to 2.1-3.3 mg/kg)

    • Effective antitumor doses in mice often exceed human-equivalent doses

  • Pharmacodynamic markers:

    • Common across species: B-cell depletion, NK cell activation, and cytokine elevations

    • These markers can guide clinical dose selection and timing

  • Response predictors:

    • Tumor CD40 expression

    • Baseline immune infiltration

    • Patient-specific factors affecting liver susceptibility to inflammation

  • Combination strategies:

    • Erythrophagocyte reprogramming approaches show promise in mice and warrant clinical exploration

    • Timing of combination therapies appears critical in both species

What are the current challenges in developing more selective anti-CD40 antibodies with improved therapeutic windows?

Key challenges and potential solutions include:

  • Tissue-selective targeting:

    • Challenge: Systemic administration affects CD40+ cells throughout the body

    • Approach: Development of antibody-drug conjugates or bispecific antibodies targeting CD40+ cells in tumor microenvironment

  • Fc engineering optimization:

    • Challenge: Fc-mediated effects contribute to both efficacy and toxicity

    • Approach: Modifying Fc regions to selectively engage desired Fc receptor subtypes

  • Epitope-specific agonism:

    • Challenge: Different epitopes on CD40 may trigger distinct signaling cascades

    • Approach: Epitope mapping to identify binding sites that favor antitumor immunity over inflammatory toxicity

  • Dosing and administration optimization:

    • Challenge: Narrow therapeutic window

    • Approach: Development of novel dosing schedules and routes of administration

  • Biomarker-guided patient selection:

    • Challenge: Heterogeneous patient responses

    • Approach: Identification of predictive biomarkers for efficacy and toxicity risk stratification

How can anti-CD40 antibody-induced erythrophagocytosis be harnessed beyond cancer immunotherapy?

The concept of antibody-induced erythrophagocyte reprogramming of Kupffer cells has broader applications:

  • Autoimmune liver diseases:

    • Potential application in autoimmune hepatitis, primary biliary cholangitis

    • The anti-inflammatory reprogramming could dampen pathological immune responses

  • Metabolic liver diseases:

    • Non-alcoholic steatohepatitis (NASH)

    • Modulation of Kupffer cell phenotype could reduce inflammation-driven fibrosis

  • Transplant immunology:

    • Prevention of liver transplant rejection

    • Creating tolerogenic environment through macrophage reprogramming

  • Infectious disease:

    • Modulation of excessive inflammatory responses in viral hepatitis

    • Potential application in sepsis-associated liver injury

  • Combination with other immunotherapies:

    • Prevention of liver toxicity associated with other immunotherapeutic agents

    • Checkpoint inhibitor-induced hepatitis

The liver-selective nature of this approach makes it particularly valuable for targeting hepatic inflammation while preserving systemic immune function .

What emerging technologies might enhance our understanding of anti-CD40 antibody mechanisms and applications?

Several cutting-edge technologies hold promise for advancing anti-CD40 antibody research:

  • Spatial transcriptomics:

    • Mapping the spatial distribution of immune cell activation within the liver and tumor microenvironment

    • Correlating spatial patterns with efficacy and toxicity

  • Multi-omics integration:

    • Combining transcriptomics, proteomics, and metabolomics to comprehensively characterize anti-CD40 responses

    • Identifying novel biomarkers and therapeutic targets

  • Organoid models:

    • Development of liver and tumor organoids with integrated immune components

    • Testing antibody effects in more physiologically relevant systems

  • In vivo imaging:

    • Real-time visualization of immune cell trafficking and activation

    • Non-invasive monitoring of treatment responses

  • AI-driven predictive modeling:

    • Predicting patient-specific responses and optimal dosing strategies

    • Identifying novel combination approaches through computational modeling

What are the most promising combination strategies involving anti-CD40 antibodies currently under investigation?

Current research suggests several promising combination approaches:

  • Anti-CD40 + checkpoint inhibitors:

    • Combines innate immune activation with removal of T-cell inhibition

    • Potential for synergistic effects through complementary mechanisms

    • Erythrophagocyte reprogramming may mitigate combined liver toxicity

  • Anti-CD40 + chemotherapy/radiotherapy:

    • Enhanced immunogenic cell death and antigen release

    • Optimal sequencing critical to avoid lethal hepatotoxicity

  • Anti-CD40 + cancer vaccines:

    • Improved antigen presentation and T-cell priming

    • Enhanced epitope spreading and broader antitumor immunity

  • Anti-CD40 + CAR-T cell therapy:

    • Anti-CD40 conditioning of the tumor microenvironment may enhance CAR-T cell infiltration and function

    • Potential to overcome resistance mechanisms in solid tumors

  • Anti-CD40 + targeted therapies:

    • Combining with BRAF inhibitors, PARP inhibitors, or other molecularly targeted agents

    • May enhance immunogenicity of dying tumor cells

How might genomic and proteomic profiling enhance patient selection for anti-CD40 antibody therapy?

Advanced profiling approaches could improve therapeutic outcomes through:

  • Tumor CD40 expression profiling:

    • Quantitative assessment of CD40 expression levels and distribution

    • Correlation with response likelihood

  • Liver risk stratification markers:

    • Genetic polymorphisms affecting inflammatory responses

    • Baseline liver macrophage activation status

  • Immune microenvironment characterization:

    • T-cell infiltration and activation state

    • Myeloid cell composition and polarization state

    • Presence of immunosuppressive factors

  • Circulating biomarkers:

    • Soluble CD40/CD40L levels

    • Baseline cytokine profiles predictive of response or toxicity

    • Liquid biopsy approaches for real-time monitoring

  • Integrative multi-omics approaches:

    • Combining genomic, transcriptomic, and proteomic data

    • Developing composite biomarker signatures for patient selection

    • Adaptive trial designs based on biomarker-driven selection

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.