CTLA 4 Antibody

CTLA-4 (CD152), Hamster Anti-Mouse
Shipped with Ice Packs
In Stock

Description

Introduction to CTLA-4 Antibody

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), also known as CD152, is an immune checkpoint receptor that downregulates T-cell activation and maintains immune tolerance . CTLA-4 antibodies are monoclonal antibodies designed to block CTLA-4's interaction with its ligands (CD80/CD86), thereby enhancing antitumor immune responses . These antibodies have revolutionized cancer immunotherapy, particularly in treating metastatic melanoma and other malignancies .

Mechanism of Action

CTLA-4 antibodies inhibit immune suppression through two primary mechanisms:

  1. Competitive Binding: CTLA-4 antibodies bind to CTLA-4 with higher affinity than CD28, preventing CTLA-4 from outcompeting CD28 for CD80/CD86 ligands on antigen-presenting cells (APCs) .

  2. Depletion of Regulatory T Cells (Tregs): Certain CTLA-4 antibodies induce antibody-dependent cellular cytotoxicity (ADCC) to deplete Tregs in the tumor microenvironment (TME), enhancing effector T-cell activity .

Key Pathways Affected:

  • Inhibition of phosphatases (e.g., SHIP2) that attenuate T-cell receptor (TCR) signaling .

  • Upregulation of inducible costimulator (ICOS) on CD4+ T cells, correlating with improved survival .

Clinically Approved CTLA-4 Antibodies

Antibody NameTarget(s)Clinical UseFDA Approval Status
IpilimumabCTLA-4Metastatic melanoma, renal cell carcinomaApproved (2011)
TremelimumabCTLA-4Mesothelioma, hepatocellular carcinomaApproved (2022)

Notable Findings:

  • Ipilimumab (3 mg/kg or 10 mg/kg) increased CD4+ICOS<sup>hi</sup> T cells in bladder cancer patients, correlating with survival .

  • Tremelimumab showed efficacy in hepatocellular carcinoma when combined with durvalumab (anti-PD-L1) .

Bispecific Antibodies (BsAbs)

Examples:

  • MEDI5752: Targets CTLA-4 and PD-1, showing superior tumor growth inhibition in triple-negative breast cancer (TNBC) models .

  • ATOR-1015: Binds CTLA-4 and OX-40, enhancing effector T-cell stimulation .

Table 1: Efficacy of BsAbs in Preclinical Models

BsAb NameTargetsTumor ModelOutcome
KN046CTLA-4/PD-L1TNBCIncreased CD8+ T-cell infiltration
XmAb20717CTLA-4/PD-1MelanomaComplete tumor regression in 40%

Immunotoxins

CTLA-4-targeted immunotoxins (e.g., SS1P, LMB-100) combined with anti-CTLA-4 antibodies induced complete tumor regression in 86% of mammary carcinoma models .

Clinical Challenges and Adverse Effects

Immune-Related Adverse Events (irAEs):

  • Common irAEs: Colitis, dermatitis, hepatitis (grade 1/2 in 70% of patients) .

  • Mechanism: IrAE-prone antibodies (e.g., ipilimumab) trigger lysosomal degradation of CTLA-4, leading to excessive T-cell activation .

Table 2: Antibody Safety Profiles

AntibodyirAE IncidenceCTLA-4 Downregulation
IpilimumabHighYes (10-fold reduction)
HL12 (non-irAE)LowNo

Emerging Applications and Future Directions

  1. Combination Therapies:

    • Anti-CTLA-4 + anti-PD-1 increased CD8+ T-cell infiltration in TNBC and reduced lung metastases .

    • Plant-produced anti-CTLA-4 antibodies demonstrated efficacy comparable to ipilimumab in colon carcinoma models .

  2. Biomarker Development:

    • Sustained CD4+ICOS<sup>hi</sup> T-cell frequency at 12 weeks predicted survival in melanoma .

  3. Next-Generation Constructs:

    • pH-sensitive anti-CTLA-4 antibodies to reduce off-target effects .

Product Specs

Introduction
CTLA-4, belonging to the immunoglobulin superfamily, is a protein responsible for transmitting inhibitory signals to T cells. Structurally, it comprises a V domain, a transmembrane domain, and a cytoplasmic tail. Characterized alternative transcriptional splice variants encode different isoforms of this protein. Functionally, the membrane-bound isoform exists as a homodimer linked by a disulfide bond, whereas the soluble isoform acts as a monomer. Genetic mutations in CTLA-4 have been implicated in a range of autoimmune diseases, including insulin-dependent diabetes mellitus, Graves disease, Hashimoto thyroiditis, celiac disease, systemic lupus erythematosus, and thyroid-associated orbitopathy.
Formulation
The antibody is supplied at a concentration of 1mg/ml in PBS buffer after reconstitution.
Storage Procedures
The antibody is provided lyophilized and should be stored at 4°C. Upon reconstitution, it is recommended to aliquot the antibody and store it at -20°C if not intended for use within a month.
Solubility
For reconstitution, add deionized water to the lyophilized antibody. Ensure gentle mixing, washing the sides of the vial, and allowing 30-60 seconds for complete dissolution before use.
Applications
This antibody is suitable for both blocking and staining applications. Optimal concentrations for blocking CTLA-4 function need to be determined empirically by the investigator.
Available Conjugates
This antibody is exclusively available in its purified form without any conjugates.
Synonyms
GSE, CD152, IDDM12, CELIAC3, CTLA-4.
Purification Method
Ion exchange column.
Type
Hamster Anti Mouse Monoclonal.
Clone
RYNmCTLA-4.
Immunogen
mixture of mouse activated T cells.
Ig Subclass
Hamster IgG.

Q&A

What is the mechanism of action for CTLA-4 in immune regulation?

CTLA-4 (Cytotoxic T-lymphocyte antigen 4) is a membrane glycoprotein expressed by activated effector T cells and regulatory T cells (Tregs). It functions as a critical immune checkpoint by negatively regulating T cell activation, proliferation, cell cycle progression, and cytokine production. CTLA-4 works by competing with the costimulatory receptor CD28 for binding to their shared ligands, CD80 (B7.1) and CD86 (B7.2), on antigen-presenting cells. Due to its higher affinity for these ligands, CTLA-4 effectively outcompetes CD28, thereby impairing T cell activation through modulation of costimulatory signals within the immunologic synapse .

The interaction begins when T cell receptors (TCRs) bind to major histocompatibility complex (MHC) molecules presenting antigens on APCs. For full T cell activation, a co-stimulatory signal involving CD28 binding to B7.1/B7.2 is required. CTLA-4 disrupts this process by preferentially binding to these same ligands, thereby preventing optimal T cell activation .

How do anti-CTLA-4 antibodies enhance anti-tumor immunity?

Anti-CTLA-4 antibodies enhance anti-tumor immunity through multiple mechanisms:

  • Blockade of CTLA-4/B7 interactions: Anti-CTLA-4 antibodies prevent CTLA-4 from binding to CD80/CD86, enabling increased CD28-mediated costimulation and enhanced T cell activation.

  • Selective depletion of intratumoral Tregs: In mouse models, anti-CTLA-4 antibodies selectively eliminate CTLA-4-expressing Tregs within the tumor microenvironment through Fc-gamma receptor (FcγR)-dependent mechanisms, reducing immunosuppression .

  • Enhanced T-cell priming: Anti-CTLA-4 antibodies promote T-cell priming through FcγR-dependent mechanisms, independent of Treg depletion, resulting in increased effector T cell populations .

  • Activation of antigen-presenting cells: Treatment with Fc-enhanced anti-CTLA-4 antibodies leads to activation of antigen-presenting cells, particularly type 1 conventional dendritic cells (cDC1), as evidenced by upregulation of CD40 and other activation markers .

These mechanisms collectively contribute to remodeling of both innate and adaptive immunity in the tumor microenvironment, ultimately leading to enhanced anti-tumor responses.

What distinguishes conventional anti-CTLA-4 antibodies from Fc-enhanced versions?

Conventional anti-CTLA-4 antibodies (like ipilimumab with an unmodified IgG1 Fc region and tremelimumab with an IgG2 Fc region) differ significantly from Fc-enhanced versions in both mechanism and efficacy:

FeatureConventional Anti-CTLA-4Fc-Enhanced Anti-CTLA-4 (e.g., Botensilimab)
Fc RegionUnmodified (ipilimumab: IgG1, tremelimumab: IgG2)Engineered for enhanced FcγR affinity
Primary Effect on TregsLimited intratumoral Treg depletionSuperior Treg depletion in tumor microenvironment
T Cell ExpansionModest expansion of effector T cellsSignificant increase in effector T cells and memory precursor cells
APC ActivationLimited effectEnhanced activation of cDC1 with increased CD40 expression
Efficacy in Poorly Immunogenic TumorsLimitedSuperior efficacy regardless of tumor immunogenicity
Biomarker DependenceEfficacy often correlates with tumor neoantigen burdenEfficacy independent of tumor neoantigen burden but may correlate with FCGR2A and FCGR3A expression

Fc-enhanced anti-CTLA-4 antibodies leverage FcγR-dependent mechanisms to potentiate T-cell responsiveness more effectively than conventional antibodies. This enhancement enables them to treat poorly immunogenic and ICI treatment-refractory cancers more effectively .

What experimental assays should researchers use to evaluate CTLA-4 blockade efficacy?

Researchers have several methodological options for evaluating CTLA-4 blockade efficacy:

  • CTLA-4 Blockade Bioassay: This cell-based assay utilizes two engineered cell lines:

    • CTLA-4 Effector Cells: Jurkat T cells expressing human CTLA-4 and a luciferase reporter driven by a native promoter responsive to TCR/CD28 activation

    • aAPC/Raji Cells: Raji cells expressing an engineered cell surface protein for TCR activation and endogenously expressing CTLA-4 ligands CD80 and CD86

    The assay involves co-culturing these cells with the test antibody, followed by quantification of luciferase activity using a luminometer. This provides a quantitative measurement of CTLA-4 blockade potency .

  • In vivo tumor models: Mouse tumor models (such as CT26 colon carcinoma) allow assessment of anti-CTLA-4 efficacy through measurement of tumor growth inhibition, survival benefit, and immunological changes in the tumor microenvironment. These models can also evaluate combination therapies .

  • Antigen-specific T cell responses: Systems like the staphylococcal enterotoxin B (SEB) challenge, which elicits a specific Vβ8+ TCR T cell response, can assess how anti-CTLA-4 affects expansion of antigen-specific T cell populations, particularly with regard to CD8+ effector T cells, memory precursor effector cells, and activation markers like Ki-67 and granzyme B .

  • Antibody-dependent cellular cytotoxicity (ADCC) assay: For Fc-enhanced anti-CTLA-4 antibodies, researchers can measure ADCC activity by combining ADCC Bioassay Effector cells with CTLA-4 Effector Cells .

How can researchers interpret contradictory results between preclinical models and human clinical outcomes?

Researchers face significant challenges when preclinical and clinical results diverge, particularly with CTLA-4 blockade where animal models typically show greater efficacy than observed in humans (except in melanoma). To address these contradictions:

  • Evaluate model relevance: The immune environment in mouse models often differs from human tumors. Researchers should consider using humanized mouse models or patient-derived xenografts.

  • Consider Fc receptor differences: Variations in FcγR distribution and affinity between mice and humans may explain differing outcomes. Experiments should control for species-specific FcγR interactions.

  • Examine tumor microenvironment differences: Analyze whether mouse models accurately represent the immunosuppressive mechanisms present in human cancers.

  • Assess technical parameters: Experimental dosing, timing, and administration route may not translate directly to clinical settings. Pharmacokinetic/pharmacodynamic modeling can help bridge this gap .

  • Cross-validate mechanisms: When discrepancies arise, researchers should verify whether the proposed mechanism (e.g., Treg depletion) occurs similarly in both preclinical models and human tissues .

The observation that Fc-enhanced anti-CTLA-4 antibodies show superior activity in poorly immunogenic tumors suggests that optimizing FcγR engagement may help bridge the gap between preclinical promise and clinical reality .

How should researchers design experiments to investigate determinants of response to anti-CTLA-4 therapy?

Researchers should implement the following methodological approaches:

  • Biomarker-focused investigations: Design studies to evaluate potential biomarkers such as FCGR2A and FCGR3A expression, which have emerged as potential response biomarkers for Fc-enhanced anti-CTLA-4 antibodies. This contrasts with traditional biomarkers like tumor neoantigen burden, which appears less predictive for newer Fc-enhanced antibodies .

  • Comparative antibody studies: Test multiple anti-CTLA-4 antibodies with differing Fc characteristics (native IgG1, IgG2, Fc-silent variants, and Fc-enhanced variants) across a panel of tumor models with varying immunogenicity profiles.

  • Immune profiling: Implement comprehensive immunophenotyping of tumors before and after treatment, focusing on:

    • T cell subsets (CD8+ effector, memory precursors, Tregs)

    • Dendritic cell activation status (particularly CD103+ and XCR1+ cDC1 populations)

    • FcγR-expressing myeloid populations

  • Mechanistic dissection: Design experiments that can distinguish between CTLA-4 blockade effects and Fc-dependent mechanisms. This can involve engineered antibody variants that retain binding to CTLA-4 but have altered or eliminated Fc functionality.

  • Sequential sampling: Implement longitudinal sampling of both blood and tumor tissue at defined timepoints to track dynamic changes in immune parameters following treatment .

What technical considerations are important when developing CTLA-4 blockade bioassays?

When developing and implementing CTLA-4 blockade bioassays, researchers should address several technical considerations:

  • Cell preparation: Use thaw-and-use cell formats that eliminate the need for continuous cell propagation, thereby reducing variability. The CTLA-4 Effector Cells and aAPC/Raji Cells should be thawed according to strict protocols to maintain viability and functionality .

  • Assay optimization:

    • Plate format selection: The assay can be performed in both 96-well and 384-well formats, with cell densities optimized for each (typically 4 × 10^4/well for CTLA-4 Effector Cells and 2 × 10^4/well for aAPC/Raji Cells in 384-well format) .

    • Incubation time: While 6-hour incubation may be sufficient, a 16-hour incubation provides greater sensitivity.

    • Antibody dilution schemes: Start with appropriate concentrations (e.g., 30μg/ml for control anti-CTLA-4 antibody and 100μg/ml for ipilimumab) and perform threefold serial dilutions to achieve full dose-response curves .

  • Detection method: Use luminescence detection with appropriate reagents (such as Bio-Glo™ Reagent) and ensure the luminometer sensitivity is calibrated for the expected signal range.

  • Controls and validation: Include appropriate positive controls (such as Control Ab, Anti-CTLA-4) and negative controls in each assay to ensure validity and reproducibility.

  • Compatibility with human specimens: Validate the assay performance in the presence of human serum (up to 10%) to ensure it can be used for neutralizing antibody detection in clinical samples .

What mechanisms underlie immune-related adverse events with anti-CTLA-4 therapy?

Immune-related adverse events (irAEs) associated with anti-CTLA-4 therapy arise from several distinct mechanisms:

  • Non-tumor-specific T cell activation: CTLA-4 blockade activates non-tumor-specific T cells, including those with potential reactivity against normal tissues. Unlike tumor-specific immunity, this broad activation contributes to off-target effects in healthy organs .

  • Regulatory T cell disruption: Treatment with anti-CTLA-4 antibodies affects both effector and regulatory T cells. While intratumoral Treg depletion is beneficial for anti-tumor activity, systemic reduction or functional impairment of Tregs can disrupt immune homeostasis in normal tissues .

  • Shared antigen recognition: Some antigens expressed by tumors are also present in healthy tissues. When T cells are activated against these shared antigens, they may attack both tumor and normal tissues expressing the same targets.

  • Pre-existing subclinical autoimmunity: CTLA-4 blockade may unmask or exacerbate underlying autoimmune tendencies that were previously controlled by CTLA-4-mediated immune suppression .

These mechanisms help explain why irAEs with anti-CTLA-4 therapy often manifest as inflammatory conditions in specific organs, including colitis, dermatitis, hepatitis, and endocrinopathies.

How can researchers develop strategies to reduce toxicity while maintaining efficacy?

Researchers can pursue several approaches to improve the therapeutic window of anti-CTLA-4 antibodies:

  • Fc engineering optimization: Design antibodies with Fc regions that preferentially deplete Tregs within the tumor microenvironment while minimizing effects on peripheral Tregs. This approach is exemplified by Fc-enhanced antibodies like botensilimab that maintain efficacy while potentially altering the toxicity profile .

  • Dose and schedule optimization: Experimentally determine the minimum effective dose and optimal treatment schedule through careful dose-finding studies. Lower doses or extended dosing intervals may reduce toxicity while maintaining efficacy.

  • Biomarker-guided patient selection: Identify predictive biomarkers for both efficacy and toxicity to enable targeted treatment of patients most likely to benefit with minimal adverse effects. Expression of FCGR2A and FCGR3A may serve as potential response biomarkers .

  • Local administration: Explore intratumoral or peritumoral delivery of anti-CTLA-4 antibodies to limit systemic exposure while maintaining local efficacy.

  • Combination approaches: Investigate combinations with other immunotherapies that may allow for dose reduction of anti-CTLA-4 while maintaining or enhancing efficacy through synergistic mechanisms.

  • Pre-treatment risk stratification: Develop screening methods to identify patients with heightened risk for specific irAEs based on genetic or immunological parameters.

What combination strategies show the most promise with anti-CTLA-4 antibodies?

Several combination approaches warrant further investigation:

  • Anti-CTLA-4 plus anti-PD-1/PD-L1: This combination has established efficacy in several cancer types by targeting complementary immune checkpoints. Further research should focus on optimizing dosing, sequencing, and patient selection based on molecular profiles .

  • Fc-enhanced anti-CTLA-4 in treatment-refractory settings: Investigate the efficacy of Fc-enhanced antibodies like botensilimab in tumors that have progressed on conventional immunotherapy, including anti-PD-1/PD-L1 or standard anti-CTLA-4 antibodies .

  • Combination with innate immune modulators: Explore combinations with agents targeting innate immunity, such as STING agonists, TLR ligands, or NK cell activators, to enhance both innate and adaptive immune responses.

  • Neoadjuvant applications: Investigate the use of anti-CTLA-4 antibodies in neoadjuvant settings, either alone or in combination with other checkpoint inhibitors, to induce tumor-specific immunity before surgical resection .

  • Combination with targeted therapies: Test the combination of anti-CTLA-4 antibodies with targeted agents that may enhance tumor immunogenicity through mechanisms such as increased neoantigen release or reduced immunosuppressive signals.

How can Fc-enhanced anti-CTLA-4 antibodies be further optimized for clinical application?

Further optimization of Fc-enhanced anti-CTLA-4 antibodies should address:

  • Structure-function relationships: Conduct detailed investigations into how specific Fc modifications influence both efficacy and safety profiles. This includes understanding which FcγRs are most important for efficacy versus toxicity.

  • Biomarker development: Validate and refine biomarkers such as FCGR2A and FCGR3A expression that may predict response to Fc-enhanced antibodies. Determine whether these markers are prognostic or truly predictive of benefit .

  • Resistance mechanisms: Identify mechanisms of primary and acquired resistance to Fc-enhanced anti-CTLA-4 therapy and develop strategies to overcome them.

  • Rational design of next-generation antibodies: Engineer antibodies with multiple enhancements, potentially including:

    • Optimized Fc regions for specific FcγR engagement profiles

    • Modified antigen-binding regions for improved CTLA-4 binding characteristics

    • Bispecific formats targeting CTLA-4 and secondary targets

  • Translational models: Develop improved preclinical models that better recapitulate human immune system interactions with Fc-enhanced antibodies to predict clinical outcomes more accurately.

The successful development of botensilimab demonstrates that Fc-enhanced anti-CTLA-4 antibodies can harness novel mechanisms to overcome limitations of conventional anti-CTLA-4 therapy, effectively treating poorly immunogenic and treatment-refractory cancers . This represents a promising direction for extending the benefits of CTLA-4 blockade to a broader range of cancer patients.

Product Science Overview

Structure and Function

CTLA-4 is a 33 kDa protein that shares significant structural and functional similarities with CD28, another protein receptor found on T cells . Both CTLA-4 and CD28 bind to the same ligands, B7-1 (CD80) and B7-2 (CD86), which are present on antigen-presenting cells (APCs). However, while CD28 provides a costimulatory signal that promotes T cell activation and proliferation, CTLA-4 delivers an inhibitory signal that downregulates immune responses .

The primary function of CTLA-4 is to maintain immune homeostasis and prevent autoimmunity by inhibiting excessive T cell activation. This is achieved through its higher affinity for B7 ligands compared to CD28, allowing it to outcompete CD28 for binding and thus delivering an inhibitory signal to the T cells .

Role in Immunological Tolerance

CTLA-4 is essential for the induction and maintenance of immunological tolerance. It plays a critical role in the regulation of T cell responses, ensuring that the immune system does not overreact to self-antigens, which could lead to autoimmune diseases . CTLA-4 is also involved in the development of regulatory T cells (Tregs), which are crucial for maintaining immune tolerance and preventing autoimmune reactions .

Hamster Anti-Mouse CTLA-4 Antibodies

Hamster anti-mouse CTLA-4 antibodies are monoclonal antibodies derived from hamsters and are used in various immunological research applications. These antibodies are designed to specifically bind to the CTLA-4 protein on mouse cells, allowing researchers to study the function and regulation of CTLA-4 in the immune system .

One commonly used hamster anti-mouse CTLA-4 antibody is the UC10-4B9 clone. This antibody can enhance T cell co-stimulation by blocking CTLA-4 interactions with B7 co-receptors, favoring CD28 interactions . Another example is the 9H10 clone, which inhibits the production of interleukin-2 (IL-2) by Jurkat cells, a type of T cell line, by blocking CTLA-4 interactions .

Applications in Research

Hamster anti-mouse CTLA-4 antibodies are widely used in immunological research to study T cell regulation, immune tolerance, and autoimmune diseases. They are employed in various experimental techniques, including flow cytometry, immunoprecipitation, and in vitro and in vivo blocking assays . These antibodies are also used in the development of immunotherapies for cancer and autoimmune diseases, as they can modulate T cell responses and enhance anti-tumor immunity .

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.