CRK8 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CCR8 Antibodies

CCR8 antibodies are monoclonal antibodies (mAbs) targeting the C-C chemokine receptor type 8 (CCR8), a G protein-coupled receptor (GPCR) predominantly expressed on tumor-resident regulatory T cells (Tregs). These antibodies are designed to modulate Treg activity within the tumor microenvironment (TME), thereby enhancing antitumor immunity .

Mechanism of Action

CCR8 antibodies function through two primary mechanisms:

  1. Antibody-Dependent Cellular Cytotoxicity (ADCC): Engineered mAbs bind to CCR8 on Tregs, recruiting natural killer (NK) cells or macrophages to eliminate immunosuppressive Tregs .

  2. Receptor Blockade: Some antibodies inhibit CCR8 activation by its ligand CCL1, disrupting Treg migration and survival signals .

Preclinical Efficacy

  • S-531011: A humanized anti-CCR8 mAb demonstrated potent ADCC activity, reducing intratumoral Tregs by >90% in xenograft models. This correlated with delayed tumor growth and improved survival .

  • mAb1: Structural studies revealed that this antagonist antibody binds CCR8 extracellular loops, preventing CCL1-induced activation. Cryo-EM data showed unique epitope targeting distinct from chemokine binding sites .

Clinical Relevance

Multi-omics analyses identified CCR8 as overexpressed in cancers such as:

Cancer TypeHazard Ratio (HR)p-Value
Breast Invasive Carcinoma1.290.037
Colon Adenocarcinoma0.390.029
Head and Neck SCC0.620.009

High CCR8 expression correlates with poor prognosis in these cancers, validating its therapeutic targeting .

Engineering and Validation Platforms

Recent advancements include:

  • HEK293-cAMP-biosensor-CCR8: A reporter cell line enabling rapid evaluation of antibody specificity and bioactivity via cAMP signaling dynamics .

  • Jurkat-NFAT-Luc2 reporter systems: Used to quantify ADCC/ADCP (antibody-dependent cellular phagocytosis) activity of CCR8 mAbs like 22H9 .

Therapeutic Synergy

Combining CCR8 antibodies with checkpoint inhibitors (e.g., anti-PD-1) enhances cytotoxic T cell infiltration and tumor regression . For example:

Therapy CombinationTumor Growth Inhibition (%)
Anti-CCR8 (S-531011)65
S-531011 + anti-PD-189

Challenges and Future Directions

  • Specificity: CCR8 shares structural homology with other chemokine receptors, necessitating high-precision antibody engineering .

  • Biomarker Development: Ongoing efforts aim to identify patients most likely to benefit via CCR8 expression profiling .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CRK8 antibody; At4g23160 antibody; F21P8.50Cysteine-rich receptor-like protein kinase 8 antibody; Cysteine-rich RLK8 antibody; EC 2.7.11.- antibody
Target Names
CRK8
Uniprot No.

Target Background

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

Q&A

What is CCR8 and why is it considered a promising immunotherapy target?

CCR8 (C-C motif chemokine receptor 8) is a G protein-coupled receptor that has emerged as a promising drug target for immunotherapy of cancer and autoimmune diseases. It is prominently expressed on tumor-infiltrating regulatory T cells (Tregs), where high CCR8 expression correlates with poor prognosis in several cancer types . The selective depletion of CCR8+ Tregs using anti-CCR8 antibodies has shown potential to restore antitumor immunity without causing systemic autoimmunity. This targeted approach allows for the reduction of immunosuppressive Tregs in the tumor microenvironment while maintaining immune homeostasis elsewhere in the body . Research has demonstrated that anti-CCR8 antibody-based treatments can significantly inhibit tumor growth by regulating tumor-resident Tregs, improving antitumor immunity and potentially enhancing patient survival rates .

How does the CCR8 signaling pathway function in immune regulation?

The CCR8 signaling pathway operates through a classic G protein-coupled receptor mechanism. When CCL1, the sole recognized ligand for CCR8, binds to the receptor, it triggers the recruitment of intracellular Gαi proteins. These Gαi proteins inhibit the activation of adenylate cyclase (AC), resulting in a decrease in intracellular cAMP levels . This signaling cascade influences the function and behavior of CCR8-expressing cells, particularly regulatory T cells in tumor tissues.

The inhibition of CCR8 by specific antibodies disrupts this signaling pathway by preventing CCL1 from binding and activating the receptor. This interference leads to an increase in intracellular cAMP concentrations, which can be measured as an indicator of antibody bioactivity . Understanding this mechanism is crucial for developing therapeutic strategies that target CCR8-expressing cells and for designing assays to evaluate anti-CCR8 antibody efficacy.

What methodologies are available for evaluating anti-CCR8 antibody functions?

Several methodologies have been developed to evaluate anti-CCR8 antibody functions, each with specific advantages:

The engineered cell-based assays offer advantages in terms of time efficiency (completed in approximately 6 hours) and the ability to provide dynamic evaluation of intracellular signaling events .

How can researchers determine the specificity of an anti-CCR8 antibody?

Determining antibody specificity is crucial for research reliability. For anti-CCR8 antibodies, researchers can use the following approaches:

  • Receptor Specificity Testing: Engineered cell lines expressing CCR8 alongside other chemokine receptors can be used to verify that the antibody exclusively recognizes CCR8 among all chemokine receptors . This is critical as many chemokine receptors share structural similarities.

  • Competitive Binding Assays: These assays evaluate whether the antibody can block the binding of CCL1 (the natural ligand) to CCR8. By measuring subsequent changes in cAMP signaling pathway activity, researchers can confirm specific interaction with the target receptor .

  • Cross-Reactivity Assessment: Testing the antibody against a panel of related receptors to ensure it doesn't bind to unintended targets. This is particularly important for therapeutic antibodies to minimize off-target effects.

  • Flow Cytometry Analysis: Using flow cytometry to verify binding to CCR8-expressing cells while showing minimal binding to CCR8-negative cells.

The recently developed HEK293-cAMP-biosensor-CCR8 reporter system shows remarkable sensitivity in detecting antibody specificity, even at low antibody concentrations, making it a valuable tool for specificity assessment .

What are the latest developments in anti-CCR8 antibody research for cancer immunotherapy?

Recent developments in anti-CCR8 antibody research have shown promising results for cancer immunotherapy applications:

  • Novel Humanized Antibodies: Researchers have developed humanized anti-CCR8 monoclonal antibodies, such as S-531011, which demonstrate potent antibody-dependent cell-mediated cytotoxicity (ADCC) activity against CCR8-expressing cells . These antibodies are designed to selectively deplete tumor-infiltrating Tregs.

  • Differentiated Antibody Profiles: Companies like Domain Therapeutics have discovered proprietary collections of anti-CCR8 antibodies with distinct and differentiated profiles. For example, their DT-7012 antibody was selected for its unique ability to achieve 100% complete response as a preclinical monotherapy .

  • Broader Target Recognition: Some newer assets have been designed to target broader forms of the CCR8 receptor compared to other clinical candidates, potentially extending their therapeutic potential .

  • Comprehensive Translational Research: Advanced translational research strategies are being developed that leverage in-depth understanding of CCR8 pharmacology and precise biomarker analysis. These approaches inform the design of first-in-human clinical studies, reducing risk and increasing trial efficiency .

These developments demonstrate that CCR8-targeted immunotherapy continues to be an active area of research with potential for clinical application in cancer treatment.

What cell systems are most effective for evaluating anti-CCR8 antibody functionality?

Several cell systems have proven effective for evaluating anti-CCR8 antibody functionality, each with specific advantages:

The choice of cell system depends on the specific research question, with engineered reporter systems offering advantages in terms of sensitivity, reproducibility, and throughput for initial antibody characterization.

How should researchers design experiments to evaluate antibody-dependent cellular cytotoxicity (ADCC) of anti-CCR8 antibodies?

Designing robust experiments to evaluate ADCC activity of anti-CCR8 antibodies requires careful consideration of several factors:

  • Selection of Target Cells: Use cells that express CCR8 at physiologically relevant levels. The HEK293-cAMP-biosensor-CCR8 reporter cell line has been validated for this purpose .

  • Effector Cell Preparation: Either primary NK cells isolated from human donors or engineered reporter cell lines like Jurkat-NFAT-Luc2-CD16a-V158 can be used as effector cells. The latter provides a standardized system that produces luminescence upon activation of ADCC pathways .

  • Experimental Setup:

    • Include a range of antibody concentrations to generate dose-response curves

    • Maintain appropriate effector-to-target cell ratios (typically 5:1 to 20:1)

    • Include positive controls (known ADCC-inducing antibodies) and negative controls (non-binding antibodies)

    • Incubate for optimal time periods (4-24 hours depending on the readout system)

  • Readout Methods:

    • For reporter systems: measure luminescence intensity, which increases with higher antibody concentrations, allowing detection of biological activity

    • For primary cell systems: assess target cell death using flow cytometry or release assays

  • Data Analysis:

    • Calculate the percentage of specific lysis or fold induction compared to controls

    • Determine EC50 values to compare potency between different antibodies

This experimental design provides a comprehensive evaluation of the ADCC potential of anti-CCR8 antibodies, which is critical for developing effective immunotherapeutic agents.

What controls are essential when evaluating anti-CCR8 antibody specificity and function?

Proper controls are critical for reliable evaluation of anti-CCR8 antibody specificity and function:

Control TypeDescriptionPurpose
Isotype ControlAntibody of same isotype but irrelevant specificityControls for non-specific binding and Fc-mediated effects
Positive ControlWell-characterized anti-CCR8 antibody with known activityValidates assay system and provides benchmark for comparison
Negative ControlCCR8-negative cellsConfirms specificity of antibody binding
Blocking ControlCCL1 (CCR8 ligand) + test antibodyDemonstrates antibody's ability to block ligand-receptor interaction
Competitive BindingUnlabeled antibody competing with labeled antibodyConfirms binding to the same epitope
Forskolin ControlAdenylate cyclase activatorProvides maximum cAMP response for normalization in cAMP-biosensor systems
ADCC/ADCP Positive ControlKnown ADCC/ADCP-inducing antibodyConfirms functionality of effector cells

When using engineered cell lines like HEK293-cAMP-biosensor-CCR8, researchers should include controls that verify the integrity of the cAMP signaling pathway, such as experiments with varying concentrations of CCL1 and Forskolin . Additionally, when evaluating antibody-mediated effector functions, controls that account for spontaneous cytotoxicity or background reporter activity are essential for accurate data interpretation.

How can researchers interpret kinetic data from CCR8 antibody functional assays?

Interpreting kinetic data from CCR8 antibody functional assays requires careful analysis of temporal patterns and quantitative parameters:

  • Response Time Course Analysis: When using systems like the HEK293-cAMP-biosensor-CCR8, observe how the cAMP signal changes over time after antibody addition. Effective CCR8 antibodies show a time-dependent increase in cAMP signal values as they block CCL1 binding . The kinetic profile provides insights into:

    • Onset of action (how quickly the antibody exerts its effect)

    • Duration of effect (how long the inhibition persists)

    • Potential biphasic responses that might indicate complex mechanisms

  • Concentration-Response Relationships: As the concentration of anti-CCR8 antibody increases, the inhibitory effect of CCL1 on the cAMP signaling pathway should decrease, leading to an increase in the induction factor . A dose-dependent relationship confirms specific activity, while the slope of the curve provides information about binding cooperativity.

  • Data Normalization: Express results as fold response or percent of maximum response (with Forskolin often used to determine maximum cAMP response) to enable comparison between experiments and different antibodies.

  • EC50/IC50 Determination: Calculate the half-maximal effective or inhibitory concentration as a measure of antibody potency.

  • Integration with ADCC/ADCP Data: Correlate the binding kinetics with functional outcomes like ADCC/ADCP activity. An antibody with strong binding may not necessarily induce potent effector functions.

The HEK293-cAMP-biosensor-CCR8 system has demonstrated remarkable sensitivity even at low antibody concentrations, making it valuable for detailed kinetic analysis of antibody-receptor interactions .

What approaches can resolve inconsistent results when testing anti-CCR8 antibody efficacy?

When researchers encounter inconsistent results in anti-CCR8 antibody testing, several systematic approaches can help identify and resolve the issues:

  • Validate Reagent Quality and Identity:

    • Confirm antibody concentration, purity, and storage conditions

    • Verify CCR8 expression on target cells using flow cytometry or Western blot

    • Check for potential cross-reactivity with related receptors

  • Assay System Optimization:

    • Titrate key components (cells, antibodies, detection reagents)

    • Optimize incubation times and temperatures

    • Ensure consistent cell culture conditions and passage numbers

    • Consider the impact of cell density on receptor expression levels

  • Complementary Methodologies:

    • When traditional methods like ELISA or Western blot yield inconsistent results, implement newer approaches like the engineered HEK293-cAMP-biosensor-CCR8 system

    • Compare results across multiple functional assays (binding, signaling, ADCC/ADCP)

    • Use primary cells in addition to engineered cell lines to confirm biological relevance

  • Controls and Standardization:

    • Include comprehensive controls in each experiment (as detailed in section 3.3)

    • Develop and use standard operating procedures

    • Implement internal reference standards for normalization between experiments

  • Technical Considerations:

    • Ensure proper instrument calibration

    • Be consistent with data analysis methods

    • Consider biological variability in primary cell samples

When inconsistencies persist, consider that different detection methods measure distinct aspects of antibody function. For example, an antibody might effectively block CCL1 binding but have limited ADCC activity, or vice versa. The integration of multiple complementary assays provides a more complete picture of antibody functionality.

How do different epitope specificities of anti-CCR8 antibodies affect their therapeutic potential?

The epitope specificity of anti-CCR8 antibodies significantly influences their therapeutic potential through several mechanisms:

  • Functional Consequences of Epitope Binding:

    • Antibodies that bind to different regions of CCR8 may have distinct effects on receptor signaling, internalization, and ligand interaction

    • Epitopes within the CCL1 binding site can directly block ligand binding, preventing activation of the receptor

    • Epitopes outside the binding pocket may cause conformational changes that indirectly affect ligand binding or signaling

  • Differential Effects on Effector Functions:

    • Some epitopes may position the antibody's Fc region optimally for interaction with Fc receptors on effector cells, enhancing ADCC or ADCP activity

    • Certain epitopes may be more accessible in the tumor microenvironment, improving targeting of tumor-infiltrating Tregs

  • Receptor Form Recognition:

    • Recent developments have produced antibodies designed to target broader forms of the CCR8 receptor compared to other clinical candidates, potentially extending their therapeutic potential

    • Recognition of different glycosylation patterns or conformational states of CCR8 may affect cell targeting specificity

  • Species Cross-Reactivity:

    • Epitopes conserved between human and mouse CCR8 enable preclinical testing in relevant animal models

    • Species-specific epitopes may limit translation of preclinical findings

  • Resistance Mechanisms:

    • Epitopes less prone to mutation or masking may reduce the development of resistance

    • Multiple antibodies targeting different epitopes could be combined to enhance efficacy and reduce resistance

Understanding these epitope-dependent effects is crucial for antibody selection and engineering. For instance, the humanized anti-CCR8 monoclonal antibody S-531011 was specifically developed to exclusively recognize human CCR8 among all chemokine receptors and demonstrate potent ADCC activity , highlighting the importance of epitope specificity in therapeutic antibody development.

What are the latest technological advances in CCR8 antibody engineering for enhanced function?

Recent technological advances in CCR8 antibody engineering have significantly improved functionality and therapeutic potential:

  • Novel Screening Platforms:

    • The development of CCR8-responsive engineered cell lines, such as HEK293-cAMP-biosensor-CCR8, has enabled rapid and sensitive screening of antibody candidates

    • These platforms provide kinetic detection of antibody function and can evaluate multiple parameters simultaneously

  • Antibody Format Innovations:

    • Bispecific antibodies targeting CCR8 and another tumor-associated antigen to enhance tumor specificity

    • Fc engineering to optimize ADCC/ADCP activity while minimizing unwanted effector functions

    • Development of antibody fragments with improved tumor penetration

  • Humanization and Affinity Maturation:

    • Advanced humanization techniques have produced antibodies like S-531011, a humanized anti-human CCR8 antibody with potent ADCC activity

    • Computational design and directed evolution approaches to optimize binding affinity and specificity

  • Translational Research Integration:

    • Comprehensive translational research strategies that leverage understanding of CCR8 pharmacology and precise biomarker analysis

    • These approaches inform first-in-human clinical study design, reducing risk and increasing trial efficiency

  • Differentiated Antibody Profiles:

    • Development of proprietary collections of anti-CCR8 antibodies with distinct and differentiated profiles

    • Selection criteria focused on functional outcomes, such as DT-7012's ability to achieve 100% complete response as a preclinical monotherapy

  • Target Form Expansion:

    • Design of antibodies targeting broader forms of the CCR8 receptor compared to other clinical candidates

    • This approach potentially extends therapeutic reach to additional CCR8-expressing cell populations

These advances reflect the evolution of CCR8 antibody development from basic research tools to sophisticated therapeutic agents with enhanced specificity, potency, and versatility.

What are the most promising research directions for CCR8 antibodies in cancer immunotherapy?

The field of CCR8 antibody research in cancer immunotherapy is rapidly evolving, with several promising directions:

  • Combination Therapies: Investigating synergistic effects of anti-CCR8 antibodies with established immunotherapies such as PD-1/PD-L1 inhibitors, cancer vaccines, or conventional therapies like chemotherapy and radiation.

  • Biomarker Development: Identifying predictive biomarkers for patient selection, including CCR8 expression levels, regulatory T cell infiltration patterns, and genetic signatures that correlate with response to CCR8-targeted therapy.

  • Expanded Cancer Indications: Building on observations that high CCR8 expression on tumor-infiltrating Tregs correlates with poor prognosis in several cancer types , researchers are exploring efficacy across diverse malignancies.

  • Enhanced Antibody Engineering: Developing next-generation anti-CCR8 antibodies with optimized ADCC/ADCP activity, improved tumor penetration, and reduced immunogenicity.

  • Novel Delivery Approaches: Exploring targeted delivery systems that increase antibody concentration in the tumor microenvironment while reducing systemic exposure.

  • Resistance Mechanism Understanding: Investigating potential resistance mechanisms to CCR8-targeted therapy and developing strategies to overcome them.

  • Translational Research Integration: Implementing comprehensive translational research strategies that leverage understanding of CCR8 pharmacology and precise biomarker analysis to inform clinical study design .

The selective depletion of tumor-infiltrating CCR8+ Tregs represents a promising approach to enhance antitumor immunity without causing systemic autoimmunity, addressing a key challenge in cancer immunotherapy. The continued development of novel antibodies and evaluation systems will accelerate progress in this field and potentially expand therapeutic options for cancer patients.

How might improved understanding of CCR8 biology influence future antibody development?

Advances in our understanding of CCR8 biology are poised to significantly influence future antibody development in several key areas:

  • Receptor Structural Insights: Detailed knowledge of CCR8's three-dimensional structure and conformational states will enable structure-based antibody design targeting specific functional epitopes.

  • Signaling Pathway Complexity: As demonstrated by the CCR8-cAMP signaling studies , deeper understanding of downstream signaling events may reveal additional functional readouts and therapeutic targets.

  • Expression Pattern Refinement: More precise characterization of CCR8 expression across different immune cell subsets and tumor types will guide the development of antibodies with improved specificity profiles.

  • Microenvironmental Factors: Understanding how the tumor microenvironment influences CCR8 expression, function, and accessibility will inform strategies to enhance antibody efficacy in situ.

  • Dynamic Regulation: Insights into how CCR8 expression and function change during disease progression and in response to therapy will help optimize treatment timing and sequencing.

  • Regulatory T Cell Heterogeneity: Recognition that tumor-infiltrating Tregs represent a heterogeneous population may lead to the development of antibodies targeting specific CCR8+ Treg subsets with enhanced immunosuppressive functions.

  • Broader Receptor Forms: Recent developments suggest potential benefits from antibodies designed to target broader forms of the CCR8 receptor compared to current clinical candidates .

  • Engineered Testing Systems: Continued refinement of systems like the HEK293-cAMP-biosensor-CCR8 will accelerate antibody development by providing rapid, sensitive, and physiologically relevant functional readouts.

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.