CD40 Human

CD40 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Expression

Structure:

  • Domains:

    • Extracellular domain: 193 amino acids with 22 conserved cysteine residues critical for TNFR family structural integrity .

    • Transmembrane domain: 22 amino acids.

    • Intracellular domain: 62 amino acids in humans (90 in mice) .

Expression:
CD40 is constitutively expressed on antigen-presenting cells (APCs) and other cell types:

Cell TypeExpression LevelKey References
B lymphocytesHigh
Dendritic cellsModerate
MacrophagesModerate
Endothelial cellsInducible
Smooth muscle cellsInducible
Epithelial cellsLow

Biological Functions

Immune Regulation:

  • B Cell Activation: CD40–CD40L interaction induces B cell proliferation, antibody class switching, and germinal center formation .

  • T Cell Costimulation: Enhances antigen presentation by upregulating B7.1/B7.2 on APCs .

  • Cytokine Production: Triggers IL-6, IL-8, TNF-α, and IL-1β release in macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs) .

Non-Immune Roles:

  • Atherogenesis: CD40L–CD40 signaling in ECs and SMCs promotes proinflammatory cytokine release, matrix metalloproteinase activation, and plaque instability .

  • Neuroinflammation: Mediates amyloid-beta-induced microglial activation in Alzheimer’s disease .

Research Findings

In Vitro Studies:

  • Vascular Cells:

    • Human ECs, SMCs, and macrophages express functional CD40L and CD40, enabling autocrine/paracrine cytokine production (e.g., IL-6, IL-8) .

    • Stimulation with IL-1β, TNF-α, or IFN-γ increases CD40L surface expression .

Atherosclerotic Lesions:

  • CD40L and CD40 are co-expressed in ECs, SMCs, and macrophages within plaque shoulder regions but absent in normal arteries .

Recombinant CD40 Ligand (rCD40L):

ParameterSpecificationSource
Molecular Mass16.3 kDa (149 aa monomer)
Purity≥95% by SDS-PAGE
Biological ActivityInduces B7.2 expression on B cells
Endotoxin Level≤0.1 EU/µg

Clinical and Therapeutic Implications

Autoimmune Diseases:

  • Blocking CD40–CD40L interaction with monoclonal antibodies (e.g., anti-CD154) reduces transplant rejection and autoimmune pathology but risks thromboembolic complications .

Cancer Immunotherapy:

  • CD40 agonists enhance antitumor immunity by activating dendritic cells and promoting T cell responses .

  • Preclinical studies show efficacy in pediatric B-cell acute lymphoblastic leukemia (B-ALL) .

Inflammatory Disorders:

  • Targeting CD40 signaling may mitigate atherosclerosis, Alzheimer’s disease, and inflammatory bowel disease .

Challenges and Future Directions

  • Safety: Early anti-CD40L trials faced vascular complications, driving interest in CD40-specific antagonists .

  • Precision Targeting: Developing cell-type-specific modulators to minimize off-target effects .

Product Specs

Introduction
CD40 is a member of the TNF-receptor superfamily and plays a crucial role in various immune and inflammatory responses. These responses include T cell-dependent immunoglobulin class switching, memory B cell development, and the formation of germinal centers. The AT-hook transcription factor AKNA is believed to regulate the expression of both CD40 and its ligand, which is important for interactions between similar cells. The adapter protein TNFR2 interacts with CD40, acting as a mediator for signal transduction. The interaction between CD40 and its ligand is considered essential for microglial activation induced by amyloid-beta, marking it as an early event in the development of Alzheimer's disease.
Description
Recombinant human CD40, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 196 amino acids (21-193a.a) with a molecular weight of 21.6 kDa. It includes a 23 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The CD40 protein solution is provided at a concentration of 0.25 mg/ml and contains 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, 10% glycerol, and 1 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product should be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein such as HSA or BSA (0.1%). Repeated freezing and thawing should be avoided.
Purity
The purity of the CD40 protein is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms
CD40 Molecule, TNF Receptor Superfamily Member 5, TNFRSF5, Tumor Necrosis Factor Receptor Superfamily, Member 5, Bp50, B-Cell Surface Antigen CD40, CD40L Receptor, CDW40, B Cell Surface Antigen CD40, B Cell-Associated Molecule, CD40 Antigen (TNF Receptor Superfamily Member 5), CD40 Type II Isoform, Nerve Growth Factor Receptor-Related B-Lymphocyte Activation Molecule, p50, Tumor Necrosis Factor Receptor Superfamily Member 5, CDw40, CD40 Antigen.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEPPTACR EKQYLINSQC CSLCQPGQKL VSDCTEFTET ECLPCGESEF LDTWNRETHC HQHKYCDPNL GLRVQQKGTS ETDTICTCEE GWHCTSEACE SCVLHRSCSP GFGVKQIATG VSDTICEPCP VGFFSNVSSA FEKCHPWTSC ETKDLVVQQA GTNKTDVVCG PQDRLR

Q&A

What is human CD40 and how does it function in the immune system?

Human CD40 is a 48 kDa transmembrane glycoprotein belonging to the tumor necrosis factor receptor (TNF-R) family. It functions as a critical receptor that initiates multifaceted activation signals in normal B cells and dendritic cells . Upon binding with its natural ligand CD40L (CD154), CD40 triggers signaling cascades that affect multiple immune processes.

The CD40-CD40L interaction exerts profound effects on dendritic cells, promoting their cytokine production, upregulation of costimulatory molecules, and facilitating cross-presentation of antigens . This interaction effectively "licenses" dendritic cells to mature and achieve the necessary characteristics for triggering T-cell activation and differentiation .

In B cells, CD40 signaling promotes germinal center formation, immunoglobulin isotype switching, somatic hypermutation to enhance affinity for antigens, and ultimately the formation of long-lived plasma cells and memory B cells . Additionally, the CD40 pathway is essential for the survival of multiple cell types including germinal center B cells, dendritic cells, and endothelial cells under both normal and inflammatory conditions .

The significance of CD40 in immune regulation is further highlighted by diseases associated with its dysfunction. For instance, X-linked hyper-IgM syndrome (XHIGM) results from mutations in the CD40L gene, leading to defective CD40-CD40L interactions and subsequent impairment in immunoglobulin class switching and somatic hypermutation .

What cell types express CD40 and CD40L in humans?

CD40 shows widespread expression across multiple cell types in the human body. According to the research data:

CD40 is expressed on:

  • All B lymphocytes during various stages of development

  • Follicular dendritic cells

  • Thymic epithelial cells

  • Various carcinoma cell lines

  • Most mature B cell malignancies

  • Some early B cell acute lymphocytic leukemias

  • Human epithelial and mesenchymal tumors, including breast, ovarian, cervical, bladder, non-small cell lung, and squamous epithelial carcinomas

CD40L (CD154) is expressed on:

  • Activated T cells (both CD4+ and CD8+ subsets)

  • Activated B cells

  • Platelets

  • Monocytes (under inflammatory conditions)

  • Both CD45R0+ and CD45RA+ T cell subsets

Northern blot and FACS analysis of peripheral blood mononuclear cells (PBMNC) have confirmed that human CD40L can be detected on T cells but is absent from B cells and monocytes under normal conditions .

Importantly, CD40 expression patterns differ between normal and disease states. While CD40 is selectively expressed on human epithelial and mesenchymal tumors, it is not found on most normal, nonproliferating epithelial tissues . This differential expression pattern makes CD40 an attractive target for cancer therapy approaches.

How is CD40L-CD40 interaction regulated in the human immune system?

The regulation of CD40L-CD40 interaction involves multiple mechanisms at the transcriptional, translational, and post-translational levels:

Cytokine-mediated regulation:
Research has demonstrated that cytokines play a significant role in regulating CD40L expression. IL-4, a known inducer of IgE production, upregulates CD40L mRNA levels, while IFN-γ, an inhibitor of IgE synthesis, reduces the expression of CD40L mRNA . This cytokine-mediated regulation suggests a correlation between human CD40L expression and IgE production.

Age-dependent regulation:
The functionality of CD40L-CD40 interaction shows age-dependent variations. Studies have found that the production of plasmablasts in response to CpG (a CD40-independent B-cell activator) increases with age, mirroring the development of memory B cells . In contrast, the response to CD40L stimulation does not change with age . This suggests different regulatory mechanisms for CD40-dependent and CD40-independent B-cell activation pathways across the human lifespan.

Soluble form generation:
CD40L can exist in both membrane-bound and soluble forms. The soluble form of CD40L is generated by intracellular proteolytic processing of the full-length CD40L . This 17 kDa protein represents the soluble segment of full-length CD40L and encompasses the receptor binding TNF-like domain . The regulation of this proteolytic process provides another layer of control over CD40L-CD40 interactions.

Structural basis of interaction:
Recent crystallographic studies have revealed the structural basis of CD40-antibody interactions, which provides insights into how CD40-CD40L binding is regulated. Agonistic antibodies like dacetuzumab bind to CD40 on the top of cysteine-rich domain 1 (CRD1), the domain most distant from the cell surface, and do not compete with CD40L binding . In contrast, antagonistic antibodies like bleselumab bind to an interface spread between CRD2 and CRD1, overlapping with the binding surface of the ligand . These structural differences explain the functional diversity of CD40-targeted therapeutics.

What are the key signaling pathways downstream of CD40 activation?

CD40 activation initiates complex signaling cascades that ultimately regulate various immune functions:

TRAF-mediated signaling:
Upon CD40 engagement, TRAF (TNF receptor-associated factor) proteins are recruited to the cytoplasmic domain of CD40. Different TRAF proteins (TRAF1, TRAF2, TRAF3, TRAF5, and TRAF6) associate with specific regions of the CD40 cytoplasmic tail, leading to activation of distinct downstream pathways .

NF-κB activation:
One of the primary outcomes of CD40 signaling is the activation of the NF-κB pathway, which regulates the expression of genes involved in immune responses, inflammation, cell survival, and proliferation . The canonical NF-κB pathway involves the degradation of IκB proteins, allowing NF-κB dimers to translocate to the nucleus and regulate gene expression.

MAPK pathways:
CD40 engagement also activates the mitogen-activated protein kinase (MAPK) pathways, including ERK, JNK, and p38 . These kinases regulate various cellular processes including proliferation, differentiation, and apoptosis.

PI3K/Akt pathway:
The phosphoinositide 3-kinase (PI3K)/Akt pathway is another important signaling cascade activated by CD40. This pathway promotes cell survival and proliferation, and is particularly relevant for the anti-apoptotic effects of CD40 signaling in B cells .

Cell-type specific outcomes:
The functional outcomes of CD40 signaling vary depending on the cell type:

  • In B cells: Promotes proliferation, survival, germinal center formation, isotype switching, and memory B cell development

  • In dendritic cells: Enhances maturation, cytokine production, and antigen presentation capabilities

  • In epithelial cancer cells: Can induce either proliferation or growth inhibition depending on the context

This complexity of CD40 signaling allows for diverse and context-dependent immune responses, making it both a fascinating research subject and a challenging therapeutic target.

What experimental methods are used to assess CD40-CD40L interactions and functionality?

Researchers employ multiple complementary approaches to investigate CD40-CD40L interactions:

B-cell activation assays:
A key method involves measuring B-cell activation in response to CD40 stimulation. Commercial anti-CD40 antibodies, in combination with human recombinant IL-21, can be used to induce strong B-cell responses . Functional readouts include:

  • Proliferation (measured by tritiated thymidine incorporation or dye dilution)

  • Surface activation marker expression (CD80, CD86)

  • Differentiation into plasmablasts or plasma cells

  • Antibody production (measured by ELISA or ELISpot)

Dendritic cell maturation assays:
CD40 activation on dendritic cells can be assessed by measuring:

  • Upregulation of costimulatory molecules (CD80, CD86)

  • Cytokine production (particularly IL-12)

  • Antigen presentation capacity and T-cell stimulatory ability

Structural analysis:
X-ray crystallography has been employed to determine the structural basis of CD40-antibody interactions, revealing binding interfaces and providing insights into the mechanisms of agonist versus antagonist antibody actions . These studies show that:

  • Agonistic antibodies (e.g., dacetuzumab) bind to CD40 on cysteine-rich domain 1 (CRD1)

  • Antagonistic antibodies (e.g., bleselumab) bind to an interface between CRD2 and CRD1

In vivo models:
Human CD40 transgenic mice have been developed to study the effects of CD40-targeted therapies in vivo. These models allow the investigation of:

  • Germinal center formation

  • Antibody production

  • Antitumor immune responses

  • Toxicity profiles of CD40-targeted therapeutics

For example, in one study with antagonist anti-human CD40 antibody (ch5D12), investigators measured the ratio of primary over secondary follicles compared to controls, indicating impairment of the germinal center reaction . This impairment was found to be reversible, as recovery animals had normalized ratios unless they maintained residual anti-CD40 antibody levels .

Functional assessment in disease models:
In cancer research, CD40 agonists have been evaluated for their ability to generate antitumor immunity. The human agonistic CD40 antibody ADC-1013 has been shown to:

  • Activate dendritic cells (measured by costimulatory molecule expression and IL-12 production)

  • Induce antigen-specific T-cell proliferation

  • Eradicate bladder tumors in both human xenograft models and syngeneic models in human CD40 transgenic mice

  • Generate long-term tumor-specific immunity

These diverse methodological approaches provide complementary insights into CD40 biology and allow for comprehensive assessment of potential therapeutic interventions targeting this pathway.

How do CD40-targeted therapies differ in their mechanisms and applications?

CD40-targeted therapies can be broadly classified into agonistic and antagonistic approaches, each with distinct mechanisms and clinical applications:

Agonistic CD40 antibodies:

Mechanism:

  • Bind to CD40 and mimic the action of CD40L

  • Primarily activate antigen-presenting cells, particularly dendritic cells

  • Enhance tumor antigen presentation and priming of tumor-specific T cells

  • May have direct effects on CD40-expressing tumor cells

  • Structurally bind to CRD1 domain without competing with CD40L binding

Applications:

  • Cancer immunotherapy, particularly for solid tumors

  • Enhancing vaccine responses

  • Promoting antitumor immunity

Example: ADC-1013 (mitazalimab)
This human agonistic CD40 antibody has demonstrated significant antitumor effects in bladder cancer models through multiple mechanisms:

  • Activation of dendritic cells, resulting in upregulation of costimulatory molecules CD80 and CD86

  • Induction of IL-12 secretion from dendritic cells

  • Stimulation of antigen-specific T-cell proliferation

  • Generation of long-term tumor-specific immunity in vivo

Antagonistic CD40 antibodies:

Mechanism:

  • Block the interaction between CD40 and CD40L

  • Inhibit germinal center formation and antibody production

  • Increase apoptosis in germinal centers

  • Structurally bind to an interface between CRD2 and CRD1, overlapping with CD40L binding site

Applications:

  • Autoimmune diseases

  • Transplant rejection

  • Inflammatory conditions

Example: ch5D12
This chimeric antagonist anti-CD40 mAb has been shown to:

  • Increase the ratio of primary over secondary follicles

  • Impair germinal center reactions (reversibly)

  • Decrease antibody production

  • Increase numbers of apoptotic cells in germinal centers

The antagonistic properties support its potential use in conditions where excessive antibody production or germinal center activity contributes to pathology.

CD40L-based approaches:

Mechanism:

  • Recombinant CD40L proteins activate CD40 signaling

  • CD40L gene therapy approaches (e.g., CD40L-expressing tumor cells)

  • Allow for more restricted or localized CD40 activation

Applications:

  • Cancer vaccines

  • Targeted immune modulation

  • Minimizing systemic inflammatory effects

A significant concern with systemic CD40L-based therapy is the risk of systemic inflammation and autoimmune consequences. To mitigate this risk, approaches using localized CD40L expression within the tumor microenvironment have been developed, such as cancer vaccines composed of autologous cancer cells transduced ex vivo with CD40L .

What research challenges exist in studying CD40 function in human primary immunodeficiencies?

Studying CD40 function in human primary immunodeficiencies presents several unique challenges:

Phenotypic heterogeneity:
Even within specific genetic defects, there can be considerable phenotypic variability among patients. For example, in XHIGM, approximately half of the affected individuals develop neutropenia, either transiently or persistently, but the cause remains unknown . This heterogeneity complicates the interpretation of research findings.

Complex clinical manifestations:
CD40-related immunodeficiencies affect multiple aspects of immunity. Patients with XHIGM or CD40 deficiency have issues with both humoral and cellular immunity, making them susceptible to a wide range of infections and autoimmune complications . Disentangling the specific contribution of CD40 dysfunction to each clinical manifestation requires sophisticated experimental approaches.

Technical challenges in functional assessment:
Assessing CD40 function in patient samples requires specialized techniques. Researchers have developed functional tests using anti-CD40 antibodies combined with IL-21 to evaluate B-cell responses in patients with primary immunodeficiencies . These tests have revealed unexpected findings, such as reduced proliferative response to CD40L in B cells from patients with selective IgA deficiency (SIgAD), despite this condition not being primarily characterized as a CD40 pathway defect .

Translating between mouse models and human disease:
While mouse models have been invaluable for studying CD40 biology, there are important differences between murine and human immune systems. Human CD40 transgenic mice have been developed to bridge this gap, but these models still cannot fully recapitulate the complexity of human immune deficiencies .

Therapeutic implications:
Research challenges extend to therapeutic development. For conditions like XHIGM, where CD40L deficiency is the underlying cause, direct replacement therapy with CD40L would risk thrombotic complications due to CD40L's role in platelet activation. Alternative approaches targeting downstream pathways or cell-specific interventions need to be explored.

How does CD40 function in tumor immunity and cancer immunotherapy?

CD40 plays multifaceted roles in tumor immunity, presenting unique opportunities for cancer immunotherapy:

Direct effects on tumor cells:
CD40 is expressed on various human epithelial and mesenchymal tumors but not on most normal, nonproliferating epithelial tissues . Ligation of CD40 on human breast, ovarian, cervical, bladder, non-small cell lung, and squamous epithelial carcinoma cells produces direct growth-inhibitory effects through:

  • Cell cycle blockage

  • Apoptotic induction

  • No apparent side effects on normal counterparts

This selective expression and response pattern makes CD40 an attractive target for cancer therapy.

Enhancement of antitumor immune responses:
CD40 activation potentiates antitumor immunity through multiple mechanisms:

  • Activation of dendritic cells, enhancing their antigen-presenting capacity

  • Increasing tumor immunogenicity by upregulating costimulatory molecule expression and cytokine production on epithelial cancer cells

  • Generating a "bystander effect" where CD40-negative tumor cells are eliminated by activated tumor-reactive cytotoxic T cells

These immunopotentiating features make CD40 agonists promising candidates for cancer immunotherapy.

Experimental evidence from preclinical models:
The human agonistic CD40 antibody ADC-1013 has shown significant efficacy in preclinical cancer models:

  • In a syngeneic bladder cancer model (negative for human CD40), ADC-1013 induced significant antitumor effects and long-term tumor-specific immunity

  • In human bladder cancer xenografts in immunodeficient NSG mice, ADC-1013 demonstrated significant antitumor effects

  • Mechanistically, ADC-1013 activated dendritic cells to upregulate costimulatory molecules CD80 and CD86, and secrete IL-12

These findings demonstrate that CD40 agonists can induce long-lasting antitumor responses and immunologic memory through CD40 stimulation.

Therapeutic approaches and challenges:
Several approaches to CD40-directed cancer therapy have been developed:

  • Recombinant CD40L protein therapy

  • Agonistic anti-CD40 antibodies

  • CD40L gene therapy (e.g., CD40L-expressing tumor cell vaccines)

  • Localized delivery of CD40 agonists to the tumor microenvironment

  • Inoculating cancer vaccines composed of autologous cancer cells transduced ex vivo with CD40L

  • Engineering CD40 agonists with tumor-targeting domains

Recent clinical trials have explored these approaches, showing promising results while managing toxicity concerns.

What are the structural insights into CD40-antibody interactions and their functional implications?

Recent crystallographic studies have provided crucial insights into the structural basis of CD40-antibody interactions:

Structural determinants of agonistic versus antagonistic activity:
X-ray crystal structures of CD40 in complex with both agonistic (dacetuzumab) and antagonistic (bleselumab) antibodies have revealed distinct binding patterns:

  • Dacetuzumab (agonist) binds to CD40 on the top of cysteine-rich domain 1 (CRD1), which is the domain most distant from the cell surface

  • Importantly, dacetuzumab does not compete with CD40L binding

  • Bleselumab (antagonist) binds to an interface spread between CRD2 and CRD1, overlapping with the binding surface of CD40L

  • This overlap explains bleselumab's antagonistic activity through direct competition with CD40L

These structural differences provide a mechanistic explanation for the divergent functional effects of these antibodies.

Implications for antibody design:
Understanding the structural basis of CD40-antibody interactions has important implications for therapeutic antibody design:

  • Agonistic antibodies can be designed to bind CRD1 without interfering with CD40L binding

  • Antagonistic antibodies should target the CD40L binding interface

  • Engineering antibodies to bind specific epitopes can fine-tune the degree of agonism or antagonism

This structural knowledge allows for rational design of antibodies with desired functional properties, potentially enabling development of more effective and specific CD40-targeted therapeutics.

Relationship to signaling outcomes:
The binding characteristics of antibodies to CD40 influence downstream signaling events:

  • Antibodies that mimic CD40L binding may engage similar signaling pathways

  • Antibodies binding to distinct epitopes may induce conformational changes that preferentially activate certain pathways over others

  • The degree of CD40 receptor clustering induced by antibodies may influence signaling intensity

These structural-functional relationships help explain the diverse biological responses observed with different CD40-targeting agents.

Translational relevance:
The structural insights into CD40-antibody interactions have direct translational relevance:

  • They provide a foundation for developing antibodies with improved therapeutic efficacy

  • They can guide the engineering of antibodies with reduced off-target effects

  • They inform the development of combination therapies targeting multiple epitopes on CD40 or multiple components of the CD40 signaling pathway

As stated in the research literature, "These data can be applied to developing new strategies for designing antibodies with more therapeutic efficacy" .

How can researchers measure CD40-dependent B-cell activation in human samples?

Researchers have developed several methodological approaches to measure CD40-dependent B-cell activation in human samples:

Anti-CD40 antibody stimulation:
Commercial anti-CD40 antibodies, in combination with human recombinant IL-21, can be used to induce strong B-cell responses in vitro . This approach offers several advantages:

  • It allows for controlled stimulation conditions

  • The strength of stimulation can be titrated by adjusting antibody concentrations

  • It enables comparison between different donor samples or patient groups

Proliferation assays:
B-cell proliferation in response to CD40 stimulation can be measured using various techniques:

  • 3H-thymidine incorporation assays measure DNA synthesis

  • CFSE dilution assays track cell division by flow cytometry

  • MTT or other colorimetric assays assess metabolic activity as a proxy for proliferation

Reduced proliferative response to CD40L has been observed in certain conditions, such as selective IgA deficiency, providing insight into disease mechanisms .

Plasma cell differentiation assessment:
CD40 stimulation plays a critical role in B-cell differentiation into antibody-secreting cells. This can be measured by:

  • Flow cytometric quantification of plasma cells (CD27high, CD38high)

  • ELISPOT assays to enumerate antibody-secreting cells

  • Measurement of secreted immunoglobulins in culture supernatants by ELISA

In patients with selective IgA deficiency (SIgAD), researchers have observed that IgA plasma cells are not generated in response to CpG stimulation, consistent with the reduction in switched memory B cells due to the absence of IgA memory B cells .

Comparison with CD40-independent activation:
Comparing CD40-dependent and CD40-independent B-cell activation provides valuable insights:

  • CpG (a TLR9 agonist) can activate B cells independently of CD40

  • The production of plasmablasts in response to CpG increases with age, mirroring the development of memory B cells

  • In contrast, the response to CD40L does not change with age

  • This comparative approach helps dissect age-related changes in B-cell functionality

Memory B-cell analysis:
CD40 signaling is crucial for memory B-cell development and function. Researchers can assess memory B-cell responses to CD40 stimulation by:

  • Separating naive and memory B-cell populations before stimulation

  • Analyzing subset-specific responses (IgM+, IgG+, or IgA+ memory B cells)

  • Evaluating class-switching capabilities after CD40 stimulation

In patients with SIgAD, switched memory B cells are reduced specifically due to the absence of IgA memory B cells, highlighting the importance of analyzing B-cell subsets .

These methodological approaches enable researchers to comprehensively assess CD40-dependent B-cell function in various physiological and pathological conditions, providing valuable insights into both normal immune processes and disease mechanisms.

What are the considerations for designing therapeutic CD40 agonists versus antagonists?

Designing therapeutic CD40 agonists and antagonists requires careful consideration of multiple factors that influence their efficacy, safety, and specificity:

Target binding properties:

For agonists:

  • Binding to domains that don't interfere with CD40L interaction (e.g., CRD1)

  • Ability to induce receptor clustering for effective signaling

  • Fc-dependency considerations (some agonists require Fc receptor cross-linking for activity)

For antagonists:

  • Binding to domains that overlap with CD40L binding site (e.g., interface between CRD1 and CRD2)

  • High affinity to effectively compete with endogenous CD40L

  • Potential for complete or partial antagonism depending on clinical needs

Antibody isotype selection:

The choice of antibody isotype significantly impacts functionality:

  • IgG1 and IgG3 engage Fc receptors effectively, potentially enhancing agonistic activity

  • IgG2 and IgG4 have reduced Fc receptor engagement, which may be preferable for pure antagonists

  • Fc engineering (e.g., ADCC-enhancing or -reducing modifications) can fine-tune effector functions

Tissue distribution and pharmacokinetics:

Considerations for therapeutic delivery include:

  • Systemic versus local administration (local administration may increase efficacy while reducing systemic adverse effects)

  • Half-life optimization through Fc engineering

  • Tissue penetration capabilities, particularly important for solid tumor targeting

Context-dependent effects:

CD40 signaling outcomes vary based on:

  • Cell type (e.g., B cells versus dendritic cells versus tumor cells)

  • Activation state of target cells

  • Presence of other immune stimuli

  • Tissue microenvironment

These variables must be considered in therapeutic design and testing .

Safety considerations:

For agonists:

  • Risk of cytokine release syndrome

  • Potential for autoimmune manifestations

  • Hepatotoxicity concerns

  • Thrombotic complications

For antagonists:

  • Immunosuppression risks

  • Potential for opportunistic infections

  • Balance between efficacy and excessive immune suppression

Clinical development strategies:

Based on the available research:

  • Cancer applications generally utilize agonistic approaches

  • Autoimmune diseases and transplantation typically employ antagonistic strategies

  • Dosing schedules are critical (intermittent versus continuous)

  • Combination therapies may enhance efficacy while reducing toxicity

For example, in cancer therapy, CD40 agonists can be combined with checkpoint inhibitors to enhance antitumor immunity . In contrast, for autoimmune conditions, CD40 antagonists may be combined with other immunosuppressive agents for synergistic effects .

Biomarker development:

Measuring target engagement and biological effects is essential:

  • Pharmacodynamic markers (e.g., changes in B-cell or dendritic cell activation)

  • Biomarkers of clinical response

  • Safety monitoring parameters

These considerations highlight the complexity of developing CD40-targeted therapeutics and the importance of mechanism-based drug design guided by structural and functional insights.

What experimental models are most appropriate for studying CD40 function in human disease contexts?

Researchers employ a spectrum of experimental models to study CD40 function in human disease contexts, each with specific advantages and limitations:

In vitro cellular systems:

Primary human cells:

  • Peripheral blood mononuclear cells (PBMCs)

  • Isolated B cells, T cells, or dendritic cells

  • Advantages: Direct relevance to human biology, accessible from patients with CD40 pathway defects

  • Limitations: Limited lifespan, donor variability, restricted manipulation capabilities

Cell lines:

  • B-cell lines (e.g., Raji, Daudi)

  • Dendritic cell lines

  • Tumor cell lines expressing CD40

  • Advantages: Homogeneous populations, amenable to genetic manipulation, unlimited supply

  • Limitations: May not fully recapitulate primary cell responses, potential artifacts from immortalization

Ex vivo tissue models:

Lymphoid tissue explants:

  • Tonsil or lymph node cultures

  • Advantages: Preserve tissue architecture and cellular interactions

  • Limitations: Short-term viability, limited availability

Organoids:

  • 3D cultures mimicking lymphoid structures

  • Advantages: More physiological than 2D cultures, can incorporate multiple cell types

  • Limitations: Technical complexity, still evolving methodology

In vivo models:

Human CD40 transgenic mice:

  • Mice expressing human CD40 instead of murine CD40

  • Allows testing of human-specific CD40 antibodies

  • Enables study of CD40 function in a living organism

  • Critical for evaluating CD40-targeted therapies before clinical translation

  • Successfully used to demonstrate that ADC-1013 (human agonistic CD40 antibody) induces antitumor effects through DC activation and T-cell stimulation

Humanized mice:

  • Immunodeficient mice reconstituted with human immune system components

  • Advantages: More complete human immune context, allows interaction studies

  • Limitations: Incomplete reconstitution, species barriers still exist

Disease-specific models:

Cancer models:

  • Human tumor xenografts in immunodeficient mice

  • Syngeneic tumor models in human CD40 transgenic mice

  • Patient-derived xenografts

  • These models have demonstrated that CD40 agonists can induce direct growth inhibition of tumor cells and enhance antitumor immune responses

Autoimmune disease models:

  • Collagen-induced arthritis in human CD40 transgenic mice

  • Experimental autoimmune encephalomyelitis

  • These models allow testing of CD40 antagonists for autoimmune indications

Transplantation models:

  • Skin, heart, or islet transplantation in humanized or transgenic mice

  • Enable evaluation of CD40 blockade for preventing graft rejection

Patient-based clinical research:

Natural human mutations:

  • Patients with CD40L deficiency (X-linked hyper-IgM syndrome)

  • Patients with CD40 deficiency

  • These rare conditions provide valuable insights into CD40 pathway function in humans

Clinical trials:

  • Phase I-III studies of CD40-targeted therapeutics

  • Provide definitive human relevance, but limited in mechanistic exploration

The choice of model system depends on the specific research question, with many studies employing multiple complementary approaches. For example, the efficacy of the agonistic CD40 antibody ADC-1013 was demonstrated in both human xenograft tumors in immunodeficient NSG mice and a syngeneic bladder cancer model in human CD40 transgenic mice, providing robust evidence for its antitumor effects .

How do CD40 pathway defects contribute to primary immunodeficiencies?

CD40 pathway defects lead to several well-characterized primary immunodeficiencies with distinct clinical manifestations:

X-linked Hyper IgM Syndrome (XHIGM):

This condition results from mutations in the CD40L gene (also known as CD154):

  • Affected individuals (typically males) have defective CD40-CD40L interactions

  • This leads to impaired class-switch recombination and somatic hypermutation

  • Clinical features include:

    • Normal or elevated IgM levels

    • Severely decreased IgG, IgA, and IgE levels

    • Susceptibility to recurrent bacterial infections

    • Increased risk of opportunistic infections (particularly Pneumocystis jirovecii)

    • Neutropenia (in approximately half of patients)

    • Autoimmune manifestations

    • Increased risk of malignancies, particularly liver tumors

CD40 Deficiency (Autosomal Recessive Hyper IgM Syndrome):

This rarer form results from mutations in the CD40 gene itself:

  • Clinically similar to XHIGM but affects both males and females

  • Can be suspected if a patient has XHIGM characteristics but is female, or is male with normal CD40L expression on activated T cells

  • Diagnosis is confirmed by genetic analysis of the CD40 gene

Other Autosomal Recessive Forms:

Several other genetic defects can disrupt the CD40 pathway:

  • Mutations in activation-induced cytidine deaminase (AID)

  • Mutations in uracil-DNA glycosylase (UNG)

  • These defects specifically impair class-switch recombination and somatic hypermutation downstream of CD40 signaling

  • Patients typically have high IgM levels but low IgG, IgA, and IgE

  • Autoimmune manifestations are common in these forms

Neutropenia in CD40 Pathway Defects:

A notable feature of XHIGM and CD40 deficiency is neutropenia:

  • Occurs in approximately half of affected individuals

  • Can be transient or persistent

  • The exact cause remains unknown

  • Most individuals respond to treatment with granulocyte colony-stimulating factor (G-CSF)

  • Severe neutropenia often associates with oral ulcers, inflammation, ulceration of the rectum (proctitis), and skin infections

Autoimmune Complications:

CD40 pathway defects predispose to various autoimmune manifestations:

  • Chronic arthritis

  • Thrombocytopenia

  • Hemolytic anemia

  • Hypothyroidism

  • Kidney disease

  • These are particularly common in patients with AID or UNG defects

The study of these primary immunodeficiencies has provided valuable insights into CD40 biology and highlighted its critical role in both humoral and cellular immunity. The clinical phenotypes demonstrate that CD40-CD40L interactions are essential not only for antibody diversification but also for broader immune homeostasis.

What are the current approaches for targeting CD40 in cancer immunotherapy?

CD40-targeted cancer immunotherapy encompasses several strategic approaches, each with unique mechanisms and potential advantages:

Agonistic CD40 antibodies:

These antibodies activate CD40 signaling to enhance antitumor immunity:

  • Examples include dacetuzumab, ADC-1013 (mitazalimab), and CP-870,893

  • Mechanisms of action include:

    • Activation of dendritic cells, enhancing their antigen-presenting capacity

    • Upregulation of costimulatory molecules (CD80, CD86) on antigen-presenting cells

    • Induction of IL-12 secretion, promoting Th1 responses

    • Direct growth inhibition in CD40-expressing tumor cells

    • Generation of tumor-specific T-cell responses

ADC-1013 has demonstrated significant efficacy in preclinical models:

  • Eradication of bladder cancer in both human xenograft and syngeneic models

  • Induction of long-term tumor-specific immunity

  • Activation of dendritic cells evidenced by upregulation of CD80/CD86 and IL-12 production

Recombinant CD40L therapy:

Direct application of the natural CD40 ligand:

  • Recombinant soluble CD40L protein

  • CD40L-expressing cellular therapies

  • Advantages include physiological receptor activation

  • Concerns include potential thrombotic complications due to CD40L interaction with platelets

CD40L gene therapy approaches:

These approaches aim to deliver localized CD40L expression:

  • Tumor cells transduced ex vivo with CD40L for vaccination

  • Viral vectors encoding CD40L for intratumoral delivery

  • Benefits include restricted CD40L expression within the tumor microenvironment

  • This localization minimizes systemic inflammatory and autoimmune consequences

  • Clinical trials have demonstrated feasibility and preliminary efficacy

Combination strategies:

CD40 agonists are increasingly being evaluated in combination with:

  • Immune checkpoint inhibitors (anti-PD-1, anti-CTLA-4)

  • Chemotherapy (potentially enhancing immunogenic cell death)

  • Radiation therapy (enhancing tumor antigen release)

  • Cancer vaccines (providing defined antigens for presentation)

These combinations aim to address multiple aspects of the cancer-immunity cycle, potentially overcoming resistance mechanisms.

Delivery considerations:

The route of administration significantly impacts efficacy and toxicity:

  • Systemic administration provides broad coverage but increases toxicity risk

  • Intratumoral delivery concentrates effect at the tumor site

  • Locoregional delivery (e.g., intranodal) targets relevant immune compartments

Toxicity management:

Managing the inflammatory consequences of CD40 activation is crucial:

  • Dose titration and scheduling (intermittent versus continuous)

  • Prophylactic anti-inflammatory treatments

  • Careful patient selection based on comorbidities

  • Biomarker-guided dosing

These approaches have advanced significantly, with multiple CD40-targeting agents in clinical development for various cancer types, offering promise for enhancing cancer immunotherapy strategies.

How is CD40 function assessed in patients with suspected immunodeficiencies?

Comprehensive assessment of CD40 function in patients with suspected immunodeficiencies involves multiple complementary approaches:

Clinical evaluation:

Initial assessment includes:

  • Infection history (pattern, severity, causative organisms)

  • Family history (X-linked inheritance pattern for CD40L deficiency)

  • Physical examination (lymphoid tissue development, signs of chronic infections)

  • Associated features (neutropenia, oral ulcers, autoimmune manifestations)

Immunoglobulin profiling:

Characteristic patterns include:

  • Normal or elevated IgM levels

  • Severely decreased IgG, IgA, and IgE levels

  • This pattern is typical of hyper-IgM syndromes including CD40L deficiency and CD40 deficiency

  • Specific antibody responses to vaccines are typically poor

Flow cytometric analysis:

Direct assessment of CD40 and CD40L expression:

  • CD40 expression on B cells, monocytes, and dendritic cells

  • CD40L (CD154) expression on activated T cells

  • For CD40L assessment, T cells must be activated (typically with PMA/ionomycin)

  • Normal CD40L expression on activated T cells excludes XHIGM but not CD40 deficiency

Functional B-cell assays:

B-cell activation with CD40L provides functional assessment:

  • Proliferation in response to CD40L stimulation

  • Surface activation marker expression (CD80, CD86)

  • Class-switch recombination capacity

  • In vitro antibody production

Interestingly, patients with selective IgA deficiency (SIgAD) show reduced proliferative response to CD40L, even though this condition is not primarily characterized as a CD40 pathway defect .

Memory B-cell analysis:

Assessment of memory B-cell subsets:

  • Total memory B cells (CD27+)

  • Switched memory B cells (IgD-CD27+)

  • IgM, IgG, and IgA memory B-cell subsets

In patients with SIgAD, switched memory B cells are reduced specifically due to the absence of IgA memory B cells .

Plasmablast generation assays:

B-cell differentiation capacity can be assessed by:

  • Stimulation with CD40L+IL-21 or CpG

  • Measurement of plasmablast generation

  • Isotype-specific plasma cell enumeration

In SIgAD patients, IgA plasma cells are not generated in response to CpG stimulation .

Genetic testing:

Definitive diagnosis requires genetic analysis:

  • CD40L gene sequencing for suspected XHIGM

  • CD40 gene sequencing for suspected CD40 deficiency

  • Next-generation sequencing panels for primary immunodeficiencies

  • Whole exome/genome sequencing for undiagnosed cases

These diagnostic approaches help differentiate CD40/CD40L defects from other immunodeficiencies with similar clinical presentations, allowing for appropriate management strategies including immunoglobulin replacement therapy, prophylactic antibiotics, and consideration for hematopoietic stem cell transplantation in severe cases.

What emerging technologies are advancing our understanding of CD40 biology?

Several cutting-edge technologies are transforming CD40 research, opening new avenues for understanding its biology and therapeutic applications:

Single-cell technologies:

Single-cell RNA sequencing (scRNA-seq) is revolutionizing our understanding of:

  • Cell-specific CD40 expression patterns across tissues

  • Heterogeneity in CD40 signaling responses

  • Temporal dynamics of CD40-induced gene expression

  • Identification of novel CD40-responsive cell populations

These approaches can reveal how CD40 activation impacts different cell subsets within complex tissues, providing unprecedented resolution of CD40 biology.

Advanced structural biology techniques:

Cryo-electron microscopy and advanced crystallography have enabled:

  • High-resolution structures of CD40-CD40L complexes

  • Visualization of CD40-antibody interactions

  • Insights into conformational changes upon receptor engagement

  • Structural basis for agonist versus antagonist activity

Recent crystallographic studies revealed that agonistic antibodies like dacetuzumab bind to CD40 on CRD1 without competing with CD40L binding, while antagonistic antibodies like bleselumab bind to an interface between CRD2 and CRD1, overlapping with the CD40L binding site .

CRISPR-based genetic screening:

CRISPR/Cas9 technology enables:

  • Genome-wide screens for regulators of CD40 signaling

  • Precise engineering of CD40 pathway components

  • Generation of improved animal models

  • Identification of synthetic lethal interactions with CD40 signaling

These approaches can uncover new therapeutic targets and resistance mechanisms in CD40-targeted therapy.

Spatial transcriptomics and proteomics:

These technologies provide spatial context to CD40 signaling:

  • Visualization of CD40-expressing cells within tissue microenvironments

  • Mapping of downstream signaling events with spatial resolution

  • Understanding CD40-mediated cell-cell interactions in situ

  • Correlation of CD40 activity with clinical outcomes in patient samples

Advanced in vivo imaging:

Intravital microscopy and molecular imaging allow:

  • Real-time visualization of CD40-dependent interactions

  • Tracking of CD40-activated cell migration and behavior

  • Monitoring of treatment responses to CD40-targeted therapies

  • Assessment of tumor infiltration by immune cells after CD40 agonist treatment

Humanized and transgenic animal models:

Improved animal models facilitate translational research:

  • Human CD40 transgenic mice for testing human-specific therapeutics

  • Tissue-specific CD40 expression or deletion

  • Inducible systems for temporal control of CD40 signaling

  • Patient-derived xenograft models incorporating human immune components

These models have been crucial for evaluating CD40-targeted therapies, as demonstrated by studies with the agonistic CD40 antibody ADC-1013 in human CD40 transgenic mice .

Systems biology approaches:

Integrative analysis enhances understanding of CD40 network effects:

  • Multi-omics integration (genomics, transcriptomics, proteomics, metabolomics)

  • Computational modeling of CD40 signaling networks

  • Prediction of combination therapy strategies

  • Patient stratification for CD40-targeted therapies

These emerging technologies collectively provide unprecedented insights into CD40 biology, accelerating both fundamental research and therapeutic development targeting this critical immune pathway.

What are the key unresolved questions in CD40 research?

Despite significant advances in CD40 research, several critical questions remain unresolved, representing important areas for future investigation:

Context-dependent signaling outcomes:

  • What determines whether CD40 signaling leads to cell survival, proliferation, or apoptosis in different cell types?

  • How do the microenvironment and concurrent signals integrate with CD40 pathways to determine functional outcomes?

  • What explains the paradoxical effects of CD40 stimulation in certain cancer cells, where it can either promote growth or induce growth arrest?

Receptor clustering and signaling initiation:

  • What is the precise stoichiometry of CD40-CD40L interactions required for optimal signaling?

  • How does membrane organization influence CD40 signaling capacity?

  • What role do lipid rafts and the cytoskeleton play in CD40 signal transduction?

  • How do different modes of CD40 activation (soluble versus membrane-bound CD40L, different antibodies) influence signaling quality?

Pathway cross-talk:

  • How does CD40 signaling integrate with other immune pathways (TLRs, cytokine receptors, BCR, TCR)?

  • What explains the synergistic effects observed between CD40 and other immune stimuli?

  • How is CD40 signaling modulated by regulatory mechanisms like immune checkpoints?

Neutropenia in CD40 pathway defects:

  • Why do approximately half of patients with XHIGM or CD40 deficiency develop neutropenia?

  • What mechanisms link CD40-CD40L interactions to neutrophil homeostasis?

  • Why do these patients respond to G-CSF treatment?

Therapeutic targeting optimization:

  • What biomarkers can predict response to CD40-targeted therapies?

  • How can we minimize toxicity while maximizing efficacy of CD40 agonists?

  • What is the optimal scheduling and dosing of CD40-targeted agents?

  • Which combination strategies will yield the best outcomes in cancer immunotherapy?

Germinal center dynamics:

  • What is the exact role of CD40 in different stages of germinal center reactions?

  • How does CD40 signaling influence the selection of high-affinity B cell clones?

  • What determines the fate decision between memory B cells and plasma cells following CD40 stimulation?

In antagonist anti-CD40 studies, researchers observed increased ratios of primary over secondary follicles and increased numbers of apoptotic cells in germinal centers, but the precise mechanisms require further investigation .

Species differences:

  • What are the key differences between human and murine CD40 signaling?

  • How can these differences be addressed in preclinical models?

  • To what extent do findings in mouse models translate to human patients?

Epigenetic regulation:

  • How does CD40 signaling influence the epigenetic landscape of responding cells?

  • What is the role of epigenetic mechanisms in establishing memory after CD40 stimulation?

  • How do epigenetic changes modify long-term responses to CD40 activation?

Addressing these unresolved questions will require integrative approaches combining advanced technologies, diverse experimental models, and translational research. The answers will not only advance our fundamental understanding of CD40 biology but also inform the development of more effective CD40-targeted therapeutics for cancer, autoimmune diseases, and immunodeficiencies.

Product Science Overview

Structure and Function

CD40 is a 277 amino acid protein with a molecular weight of approximately 30 kDa. It consists of an extracellular domain, a single transmembrane domain, and a cytoplasmic tail. The extracellular domain is responsible for binding to its natural ligand, CD40L (CD154), which is predominantly expressed on activated CD4+ T cells .

The interaction between CD40 and CD40L is crucial for various immune responses. This interaction leads to the activation and proliferation of B cells, differentiation of dendritic cells, and induction of cytokine production in peripheral blood monocytes and T cells . Additionally, CD40-CD40L signaling plays a significant role in T cell-APC interactions, B cell differentiation and proliferation, isotype class-switching, and protection of B cells from apoptosis .

Recombinant Human CD40

Recombinant human CD40 (rhCD40) is a form of CD40 that is produced using recombinant DNA technology. This involves inserting the gene encoding CD40 into a suitable expression system, such as Escherichia coli (E. coli), to produce the protein in large quantities . Recombinant CD40 is used in various research applications, including cell culture, differentiation studies, and functional assays .

Applications

Recombinant human CD40 has several applications in scientific research:

  1. Stimulation of B Cell Activation and Proliferation: CD40-CD40L interaction is essential for B cell activation and proliferation. Recombinant CD40 can be used to study these processes in vitro .
  2. Dendritic Cell Maturation: CD40 signaling is crucial for the maturation of dendritic cells, which are key players in the immune response .
  3. Cytokine Production: CD40-CD40L interaction induces cytokine production in peripheral blood monocytes and T cells, which can be studied using recombinant CD40 .
  4. Cancer Immunotherapy: CD40-activated B cells have been studied as potent antigen-presenting cells that can potentially be used for cancer immunotherapy .

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 2024 Thebiotek. All Rights Reserved.