CCR6 overexpression correlates with tumor angiogenesis and poor prognosis in colorectal cancer . Antibodies targeting CCR6/CCL20 axis reduce regulatory T-cell infiltration and slow tumor growth in murine models .
In a psoriasis model, CCR6 blockade limits Th17 cell recruitment to inflamed skin .
Neutralization of CCL20 (e.g., GSK3050002) reduces CCR6⁺ cell recruitment in human skin blisters, validating the pathway’s role in inflammation .
Specificity: Unlike broad-spectrum immunosuppressants, CCR6 mAbs selectively inhibit pro-inflammatory Th17/Treg trafficking without impairing essential immune responses .
Synergy: Combination with checkpoint inhibitors (e.g., anti-PD-1) may enhance antitumor immunity by modulating the tumor microenvironment .
Species cross-reactivity: Human-specific antibodies (e.g., MSM-R605) lack efficacy in murine models, complicating preclinical validation .
Epitope diversity: Structural studies reveal distinct binding sites (e.g., N-terminal vs. extracellular domains), necessitating tailored antibody engineering .
Optimized engineering:
Bispecific antibodies targeting CCR6 and other chemokine receptors (e.g., CCR5) for enhanced therapeutic breadth.
Biomarker-driven trials:
Stratify patients based on CCR6 expression in tumors or inflammatory tissues to predict response.
Combination therapies:
CCR6 is a G-protein-coupled receptor highly expressed in B lymphocytes, effector and memory T cells, regulatory T cells, and immature dendritic cells. It has gained significant attention as a therapeutic target because:
CCR6 and its ligand CCL20 are upregulated in tissues of patients with chronic inflammatory conditions
It serves as the main chemokine receptor for Th17 cells, which are implicated in many chronic inflammatory conditions
The CCL20/CCR6 axis plays critical roles in cancer, intestinal bowel disease, psoriasis, and autoimmune diseases
Inhibition of CCR6 in preclinical models has shown promising results in reducing disease activity in models of skin inflammation, arthritis, and colitis
Validation of anti-CCR6 antibody specificity involves multiple complementary approaches:
Transfected cell line testing: Testing antibody binding to CCR6-transfected cells (e.g., L1.2, CHO-K1) versus non-transfected parental cell lines
Cross-reactivity assessment: Evaluating binding to CCR6 from different species (human, cynomolgus monkey, mouse) to determine species specificity
Flow cytometry with known CCR6-expressing cells: Testing reactivity with cell populations known to express CCR6, such as B cells and specific T cell subsets
Comparison with isotype controls: Using appropriate isotype control antibodies to confirm specific staining
Functional assays: Demonstrating inhibition of CCL20-mediated effects, such as chemotaxis or calcium flux
For example, one validated antibody (C6Mab-19) demonstrated specific binding to hCCR6-overexpressed CHO-K1 cells with an extremely high affinity (KD of 3.0 × 10^-10 M) .
CCR6 recombinant monoclonal antibodies have diverse research applications:
Anti-CCR6 antibodies vary significantly in their ability to block CCL20-mediated signaling. Key factors affecting blocking efficacy include:
Binding epitope: Antibodies targeting the CCL20 binding region generally show superior blocking activity
Binding affinity: Higher affinity antibodies typically demonstrate more effective blocking
Antibody format: Different isotypes and modifications can affect functional activity
Experimental data demonstrates these differences:
The humanized 6H12 (h6H12) antibody showed high-affinity binding to human CCR6 and effectively blocked CCL20-mediated chemotaxis
MSM-R605, a fully human antibody, demonstrated dose-dependent inhibition of CCL20-induced calcium flux in Chem-1 cells expressing human CCR6, providing quantitative measurement of functional blocking
GSK3050002, a humanized IgG1κ antibody with 48 pM binding affinity to human CCL20, showed dose-dependent decreases in CCR6+ cell recruitment to skin blisters with maximal effects at doses of 5 mg/kg and higher
When evaluating blocking antibodies, researchers should conduct dose-response studies and use appropriate functional readouts relevant to their specific research questions.
Species cross-reactivity represents a significant challenge in CCR6-targeted therapeutic development. Several methodological approaches can address this limitation:
Transgenic animal models: Development of humanized CCR6 transgenic mice (hCCR6-Tg/mCCR6-/-) where mouse CCR6 is replaced with human CCR6, allowing testing of human-specific antibodies in mouse models
Multi-species binding antibodies: Identification and optimization of antibodies that recognize conserved epitopes across species, though this is challenging due to sequence variability
Surrogate antibodies: Development of paired antibodies - one targeting human CCR6 for clinical development and a surrogate antibody targeting the corresponding species CCR6 for preclinical studies
Epitope grafting: Engineering target animals to express the human epitope recognized by the therapeutic antibody
For example, researchers developed hCCR6-Tg/mCCR6-/- mice to test an anti-human CCR6 antibody (h6H12) that did not cross-react with mouse CCR6. This model demonstrated that "following immunization with recombinant mouse myelin oligodendrocyte glycoprotein, a significant increase in hCCR6 expression was observed on CD4+ and CD8+ T cells isolated from lymph nodes and spleen," enabling preclinical testing of the human-specific antibody .
Optimizing experimental design for anti-CCR6 therapeutic studies requires careful consideration of several factors:
Target validation: Confirm CCR6 expression and relevance in the disease model
Analyze CCR6 expression patterns in relevant tissues using flow cytometry and IHC
Compare expression between healthy and disease states
Dosing strategy optimization:
Pharmacokinetic/Pharmacodynamic correlation:
Appropriate controls:
Include isotype control antibodies
Consider Fc-modified variants to distinguish Fc-mediated from target-blocking effects
Multiple readouts:
Clinical scores (e.g., EAE scoring)
Cellular analysis (flow cytometry of infiltrating cells)
Histopathological assessment
Molecular markers of inflammation
A comprehensive example from GSK3050002 clinical studies demonstrates this approach: researchers established a relationship between pharmacokinetics, target engagement, and pharmacodynamics by measuring both serum and blister fluid levels of GSK3050002/CCL20 complex alongside functional effects on CCR6+ cell recruitment .
Developing optimized flow cytometry panels for CCR6+ cell identification requires addressing several technical challenges:
Antibody clone selection:
Panel design considerations:
Technical optimization:
Titrate antibodies to determine optimal concentration
Consider fluorochrome brightness when assigning to markers of different expression levels
Include appropriate FMO (fluorescence minus one) controls
Sample preparation considerations:
Optimize fixation conditions that preserve CCR6 epitope
Minimize time between sample collection and staining as surface receptors may internalize
Consider the impact of enzymatic dissociation on CCR6 epitope integrity
For example, a successful panel used in one study included: "T-cells (CD3+), Th1 (CD3+CD4+CXCR3+CCR6−), Th2 (CD3+CD4+CXCR3−CCR6−), Th17 (CD3+CD4+CXCR3−CCR6+) and Th1/17 (CD3+CD4+CXCR3+CCR6+) T-cells, while monocytes and granulocytes were gated based on forward and side scatter" .
Epitope masking or modulation presents challenges when detecting CCR6 across different experimental contexts. Researchers can implement these methodological solutions:
For flow cytometry applications:
Use multiple anti-CCR6 antibody clones targeting different epitopes
Optimize staining buffer composition (calcium concentration can affect chemokine receptor conformation)
Evaluate the impact of cell activation state on CCR6 detection
Consider using indirect staining approaches for amplification when epitope accessibility is limited
For immunohistochemistry applications:
Optimize antigen retrieval methods (as demonstrated for anti-CCR6 antibody EPR22259, which used "heat mediated antigen retrieval with Tris-EDTA buffer (pH 9.0) for 20 mins")
Test multiple fixation protocols to determine optimal epitope preservation
Consider the impact of tissue processing on epitope accessibility
For Western blotting applications:
Compare reducing vs. non-reducing conditions
Optimize detergent selection for membrane protein extraction
Consider native vs. denatured protein detection approaches
Ligand-induced receptor modulation:
Be aware that CCL20 binding may induce receptor internalization or conformational changes
Design experiments to account for potential ligand-induced epitope masking
Consider time-course experiments to track receptor dynamics after ligand exposure
Validation across methods:
Confirm CCR6 detection using complementary techniques (e.g., flow cytometry, immunohistochemistry, and Western blotting)
Use genetic approaches (knockout/knockdown) to validate antibody specificity
Some antibodies have demonstrated robust performance across multiple applications. For example, anti-CCR6 antibody EPR22259 has been validated for immunoprecipitation, Western blotting, and immunohistochemistry, suggesting reliable epitope recognition across different experimental conditions .
Creating reliable CCR6-expressing cell lines requires careful consideration of expression systems and validation approaches:
Selection of appropriate host cell lines:
Expression vector design considerations:
Codon optimization for host cell line
Selection of appropriate promoters (e.g., CMV for high expression)
Inclusion of selection markers (e.g., antibiotic resistance)
Addition of epitope tags if needed for detection/purification
Validation of CCR6 functionality:
Calcium mobilization assays in response to CCL20
Migration/chemotaxis assays
Receptor internalization studies
Binding assays with labeled CCL20
Quantification of expression levels:
Flow cytometry with anti-CCR6 antibodies
Quantitative Western blotting
Radioligand binding assays for precise quantification
For example, Chem-1 cells expressing human CCR6 were successfully used to demonstrate the inhibitory activity of MSM-R605 on CCL20-induced calcium mobilization, providing a functional readout system for antibody characterization .
CCR6 knockout and transgenic mouse models have provided critical insights into CCR6 biology and therapeutic targeting:
CCR6 knockout mice observations:
Human CCR6 transgenic models (hCCR6-Tg/mCCR6-/- mice):
Enable testing of human-specific anti-CCR6 antibodies in mouse disease models
Allow evaluation of human CCR6 biology in vivo
Provide translational insights for clinical development
Applications in therapeutic development:
Expression pattern insights:
Interestingly, CCR6 expression patterns differ between humans and mice: "all human B cells expressed CCR6, compared with approximately 55% of B cells in mice. A larger proportion of human CD3+ cells expressed the receptor (~14%) compared with mouse CD3+ cells"
These differences highlight the importance of humanized models for translational research
These models have established that "CCR6 plays a critical role in Th17 type inflammatory reactions, and CCR6 inhibition may offer an alternative approach for the treatment of these lesions" .
Evaluating therapeutic efficacy of anti-CCR6 antibodies requires comprehensive assessment strategies:
Disease model selection:
Experimental autoimmune encephalomyelitis (EAE): Well-established model where "hCCR6-Tg/mCCR6–/– mice develop EAE in a manner similar to wild-type C57BL/6 mice"
Skin inflammation models: Relevant for psoriasis research
Colitis models: Important for inflammatory bowel disease studies
Arthritis models: For rheumatoid arthritis applications
Treatment timing strategies:
Comprehensive assessment metrics:
Clinical scoring systems specific to each model
Histopathological analysis of tissue inflammation and damage
Flow cytometric analysis of infiltrating immune cells
Cytokine/chemokine profiling in serum and affected tissues
Functional assessments relevant to the disease model
Mechanistic investigations:
CCR6+ cell trafficking analysis
Examination of lymphoid organ architecture and germinal center formation
Assessment of antigen-specific T and B cell responses
Evaluation of CCL20/CCR6 axis inhibition in target tissues
Translational biomarkers:
In clinical studies, the experimental skin suction blister model has proven valuable for assessing pharmacokinetics, target engagement, and the ability of anti-CCL20 antibody (GSK3050002) to inhibit recruitment of inflammatory CCR6-expressing cells .
Optimizing immunohistochemistry protocols for CCR6 detection in FFPE tissues requires systematic approach:
Antigen retrieval optimization:
Antibody selection and validation:
Signal amplification and detection system selection:
Protocol optimization:
Controls and validation:
Include positive control tissues with known CCR6 expression
Incorporate negative controls (isotype control or secondary antibody only)
Consider dual staining with lineage markers to confirm cell type-specific expression
For troubleshooting purposes, researchers should note that "specific staining was localized to cytoplasm in splenocytes" when using MAB195R in human spleen sections , providing a reference for expected staining patterns.
Ensuring specific CCR6 detection in flow cytometry requires addressing several technical challenges:
Antibody validation strategies:
Test on CCR6-transfected vs. non-transfected cell lines
Include biological positive controls (e.g., B cells) and negative controls
Compare staining patterns with multiple anti-CCR6 clones
Control implementation:
Addressing non-specific binding:
Optimize blocking protocols using serum or FcR blocking reagents
Titrate antibody to minimize background while maintaining specific signal
Consider the impact of dead cell binding (include viability dye)
Sample preparation considerations:
Minimize time between sample collection and staining
Optimize fixation protocols that preserve CCR6 epitope
Evaluate the impact of different buffer compositions on staining quality
Functional validation:
Confirm that CCR6+ sorted cells respond to CCL20 in migration assays
Demonstrate that antibody pretreatment can block CCL20-induced responses
For example, when detecting "CCR6 in CD19+ Human PBMCs by Flow Cytometry," researchers successfully used "Mouse Anti-Human CD19 PE-conjugated Monoclonal Antibody" alongside "Mouse Anti-Human CCR6 Monoclonal Antibody" and compared to isotype control, followed by "Goat anti-Mouse IgG APC-conjugated secondary antibody" .
Evaluating functional inhibition by anti-CCR6 antibodies requires robust assay systems:
Calcium mobilization assays:
Use cells expressing human CCR6 (e.g., Chem-1/hCCR6 cells)
Load cells with calcium-sensitive dyes
Measure fluorescence changes after CCL20 stimulation
As demonstrated with MSM-R605, measure "inhibition of the increase in the intracellular Ca concentration in response to addition of human CCL20 to Chem-1 cells expressing human CCR6"
Generate dose-response curves for the antibody
Chemotaxis/migration assays:
Receptor internalization assays:
Assess CCR6 surface expression after CCL20 stimulation
Determine if antibody prevents receptor internalization
Use flow cytometry or imaging approaches for quantification
Signaling pathway analysis:
Examine downstream pathways (e.g., MAPK, Akt activation)
Use phospho-specific antibodies in Western blot or flow cytometry
Determine IC50 values for inhibition of specific signaling events
Ex vivo cell recruitment models:
These functional assays should include appropriate controls and dose-response analyses to fully characterize the inhibitory potential of anti-CCR6 antibodies.
Quantitative assessment of receptor occupancy and target engagement is critical for therapeutic antibody development:
Direct binding measurement approaches:
Flow cytometry-based methods:
Compare binding of a labeled detection antibody before and after treatment
Use competing antibodies targeting non-overlapping epitopes
Radiolabeled ligand binding studies:
Measure displacement of radiolabeled CCL20
Complex formation quantification:
GSK3050002/CCL20 complex measurement:
Pharmacokinetic/Pharmacodynamic correlation:
Ex vivo functional assays:
Collect patient samples after antibody administration
Test responsiveness to CCL20 stimulation
Correlate functional inhibition with antibody exposure
Biomarker development:
Identification of downstream markers that reflect target engagement
Monitoring changes in CCR6+ cell trafficking patterns
Evaluation of inflammatory markers in affected tissues
For translational development, the GSK3050002 study demonstrated that "levels of GSK3050002/CCL20 complex appeared to increase in a dose-dependent manner and reach maximum levels at doses of 5 mg/kg and higher both in serum and interstitial fluid, suggesting that a saturable target engagement has been achieved at the high doses" .
Development of bispecific antibodies targeting CCR6 and other inflammatory pathways represents an advanced therapeutic strategy:
Target selection considerations:
Complementary inflammatory pathways to CCR6/CCL20:
Th17-related cytokines (IL-17, IL-23)
Other chemokine receptors on CCR6+ cells (e.g., CXCR3 for Th1/17 cells)
Co-stimulatory molecules involved in T cell activation
Consider pathways with demonstrated synergy in disease models
Bispecific antibody format selection:
Functional characterization approaches:
Compare monospecific vs. bispecific effects in:
Calcium flux assays
Chemotaxis inhibition
Cell activation/cytokine production
Test for synergistic vs. additive effects
Evaluate potential antagonism between targeting arms
In vivo testing strategies:
Manufacturing considerations:
Assess expression levels and stability
Implement appropriate quality control testing
Consider strategies to minimize heterogeneity
While no specific examples of CCR6-targeting bispecific antibodies were described in the provided search results, the approach builds on established expertise in CCR6 targeting through monospecific antibodies with demonstrated efficacy in inflammatory disease models.