MCAM plays a crucial role in cell adhesion and maintains the cohesion of the endothelial monolayer at intercellular junctions within vascular tissue. Its expression may facilitate melanoma cell interaction with vascular system components, thereby potentially enhancing hematogenous metastasis. MCAM may also function as an adhesion molecule in neural crest cells during embryonic development. Furthermore, it acts as a surface receptor, triggering tyrosine phosphorylation of FYN and PTK2/FAK1, and inducing a transient increase in intracellular calcium concentration.
MCAM (Melanoma Cell Adhesion Molecule)/CD146 is a member of the immunoglobulin superfamily that mediates cell-to-cell and cell-to-matrix interactions. Initially identified as a melanoma antigen, MCAM plays crucial roles in various biological processes including tumor growth, metastasis, angiogenesis, and inflammation. MCAM is expressed on melanoma cells, endothelial cells, certain T cell populations, and mesothelioma cells, but notably absent on normal mesothelial cells . Research significance stems from its involvement in cancer progression, neuroinflammation, and other pathological conditions, making it an important target for therapeutic intervention and biomarker development.
MCAM expression shows distinct patterns across cell types and developmental stages. While absent on normal epidermal melanocytes, MCAM is highly expressed on melanoma cells with considerable intra- and intertumoral heterogeneity . Endothelial cells constitutively express MCAM . Certain CD4+ T cell subsets, particularly Th17 cells, express MCAM during inflammation . Developmental analysis reveals that MCAM is strongly expressed in fetal melanocytes but progressively downregulated during melanocyte maturation, with few neonatal and hardly any adult melanocytes maintaining high MCAM expression . This suggests MCAM expression may represent reactivation of an embryonal transcriptional program in cancer cells.
MCAM antibodies have been instrumental in delineating the role of MCAM in melanoma metastasis through multiple methodological approaches. Time-lapse video microscopy combined with MCAM knockout or antibody blockade has demonstrated that MCAM facilitates interactions between melanoma cells and endothelial cells during metastatic dissemination . In vitro migration assays show that MCAM-deficient melanoma cells display significantly reduced motility on endothelial cell monolayers compared to MCAM-expressing controls, with measurable decreases in migration distance and velocity .
Transwell migration experiments further reveal that MCAM's pro-metastatic function requires direct melanoma-endothelial cell contact, as MCAM knockout specifically impairs migration when endothelial cells are seeded alongside melanoma cells on the transwell membrane . In vivo, MCAM knockout in HCmel12 mouse melanoma cells significantly reduced spontaneous lung metastasis formation following intradermal transplantation in both immunocompetent and immunodeficient mouse models, confirming a tumor cell-intrinsic role for MCAM in metastatic spread .
Analysis of TCGA RNA sequencing data revealed no significant differences in MCAM mRNA expression between primary and metastatic melanomas . Though patients with high MCAM expression (above 90th percentile) showed a trend toward worse melanoma-specific survival compared to those with low expression (below 10th percentile), this difference did not reach statistical significance . These findings suggest that MCAM is abundantly expressed in both primary and metastatic melanomas but may not be a reliable prognostic biomarker for disease progression.
MCAM antibodies provide valuable tools for investigating angiogenesis in cancer research through multiple experimental approaches. Since endothelial cells constitutively express MCAM, antibodies targeting MCAM can directly disrupt tube-like formation by human umbilical vein endothelial cells (HUVECs) in in vitro vessel formation assays . The fully human anti-MCAM antibody ABX-MA1 demonstrated this effect, suggesting that MCAM plays a role in angiogenesis independent of its function in tumor cells .
MCAM antibodies can also help elucidate the crosstalk between tumor cells and the endothelium during angiogenesis. Bioinformatic inference of cellular communication networks revealed that melanoma cells with high MCAM expression more actively engage in signaling crosstalk with endothelial cells . This interaction appears to involve multiple pathways, including matrix metalloproteinase 2 (MMP-2), as ABX-MA1 treatment significantly inhibited MMP-2 promoter and collagenase activity in melanoma cells in vitro, with decreased MMP-2 expression also observed in implanted tumors in vivo .
Immunofluorescence techniques using anti-MCAM antibodies can identify MCAM-positive vessels within tumors, allowing quantification of microvessel density and assessment of anti-angiogenic therapies .
Multiple lines of evidence implicate MCAM in neuroinflammation and CNS infiltration of immune cells. Immunohistochemical studies show that MCAM is upregulated on brain endothelial cells during neuroinflammatory conditions, creating a spatial-temporal association between endothelial MCAM expression and immune cell infiltration into the CNS . In experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, MCAM blockade delayed disease onset by restricting the migration of MCAM-expressing T cells through the choroid plexus (CP) into the CNS .
The laminin 411 component (composed of α4, β1, γ1 chains) has been identified as a major ligand of MCAM and is detected in the endothelial basement membranes of murine CP tissue and human CP endothelial-basement membranes in MS patients' brain tissue . This interaction appears critical for immune cell transmigration, as MCAM blockade reduced in vitro transmigration of MCAM-expressing T cells across both a human fibroblast-derived extracellular matrix layer and a brain-derived endothelial monolayer expressing laminin α4 .
Additionally, MCAM knockout mice showed reduced CD4+ T cell infiltration into the CNS during EAE compared to wild-type controls, with less pronounced disease when induced by adoptive transfer of Th1 and Th17 cells . These findings collectively establish MCAM as a key mediator of pathogenic T cell entry into the CNS during neuroinflammation.
MCAM antibodies can modulate T cell function in neuroinflammatory models through several mechanisms. Functional assays demonstrate that Th1 and Th17 CD4+ T cells show impaired rolling and firm arrest on MCAM-knockout endothelial cells, as well as diminished migration through these cells . This suggests that blocking MCAM interactions impairs the initial adhesion steps required for T cell extravasation into the CNS.
In vitro transmigration assays show that anti-MCAM antibodies reduce the ability of MCAM-expressing T cells to migrate across endothelial barriers expressing laminin α4 . This effect is specific to the MCAM-laminin 411 interaction and represents a distinct mechanism from those targeted by current MS therapies like natalizumab (anti-VLA-4) .
The anti-MCAM antibody PRX003, developed initially for psoriasis, efficiently eliminates MCAM from the Th17 cell surface, thereby presumably interfering with extravasation to target tissue . While this approach did not show clinical benefits in psoriasis according to company reports, the mechanism may still be relevant in multiple sclerosis or other neuroinflammatory conditions by interfering with both endothelial and lymphocytic MCAM simultaneously .
Unlike broad-spectrum immunomodulatory treatments, anti-MCAM antibodies offer potential for targeted intervention restricted to the time and location of early CNS inflammation, potentially providing a more selective therapeutic approach .
Validating MCAM antibody specificity requires a multi-faceted approach combining genetic, biochemical, and functional methods:
Genetic validation:
Biochemical validation:
Western blot analysis should detect a specific band at approximately 140 kDa for human MCAM/CD146 under reducing conditions
Compare staining patterns of multiple anti-MCAM antibodies targeting different epitopes
Include appropriate isotype controls in flow cytometry and immunohistochemistry applications (e.g., MAB002 for mouse antibodies)
Functional validation:
A comprehensive validation approach should demonstrate consistent results across multiple techniques and experimental systems, with appropriate positive and negative controls.
Optimal protocols for MCAM detection vary by experimental system and technique:
Western Blot:
Lyse cells in appropriate buffer (e.g., Immunoblot Buffer Group 1)
Use reducing conditions for standard SDS-PAGE
Load 20-30 μg total protein per lane
Probe membrane with 1 μg/mL of anti-MCAM antibody (e.g., MAB932 for human samples)
Follow with appropriate HRP-conjugated secondary antibody (e.g., HAF018)
Expected band size: approximately 140 kDa for human MCAM/CD146
Flow Cytometry:
Use single-cell suspensions (1×10^6 cells/100 μL)
Stain with anti-MCAM antibody (e.g., MAB932 for human samples, MAB7718 for mouse samples)
Include isotype control antibody in parallel samples
Follow with fluorophore-conjugated secondary antibody (e.g., F0102B)
Analyze on standard flow cytometer with appropriate compensation settings
Immunocytochemistry/Immunofluorescence:
Fix cells with 4% paraformaldehyde or immersion fixation
Use antibody concentration of 10 μg/mL for 3 hours at room temperature
Follow with fluorophore-conjugated secondary antibody (e.g., NL007 or NL013)
Counterstain nuclei with DAPI
Immunohistochemistry:
Use formalin-fixed, paraffin-embedded or frozen tissue sections
Include positive controls (e.g., melanoma tissue) and negative controls (e.g., normal mesothelial tissue)
Quantify expression by considering both staining area and intensity
Interpret results in context of CD31 (endothelial) and SOX10 (melanoma) staining on serial sections
Designing effective MCAM knockdown/knockout experiments requires careful planning and appropriate controls:
Selection of experimental system:
CRISPR/Cas9 knockout design:
siRNA/shRNA knockdown:
Design multiple siRNA/shRNA sequences targeting different regions of MCAM mRNA
Validate knockdown efficiency by qRT-PCR and Western blot
Optimize transfection conditions for each cell type
Include non-targeting controls with similar GC content
Functional validation:
Rescue experiments:
By following these guidelines, researchers can generate reliable data on MCAM function across different experimental systems while minimizing technical and biological artifacts.
Interpreting contradictory findings regarding MCAM's role in different cancer types requires systematic consideration of multiple factors:
Tissue-specific context:
Methodological differences:
Cancer heterogeneity:
Integration framework:
Focus on mechanistic insights rather than correlative data
Prioritize studies with genetic manipulation (knockout/knockdown) over observational studies
Consider whether MCAM functions primarily in tumor cells, stromal cells, or both
Evaluate evidence for homophilic (MCAM-MCAM) versus heterophilic (MCAM-laminin) interactions
To reconcile contradictory findings, researchers should also consider MCAM's developmental context—its expression in fetal melanocytes and subsequent downregulation during maturation suggests it may be part of a broader embryonic program reactivated in cancer . This may explain why its role varies across cancer types depending on their cell of origin and differentiation state.
Analysis of MCAM expression data in patient cohorts requires robust statistical approaches to account for heterogeneity and potential confounding factors:
Data preprocessing and normalization:
Beyond simple correlations:
Multivariate regression models adjusting for known prognostic factors
Propensity score matching to compare patients with similar characteristics but different MCAM expression
Consider MCAM expression as both continuous and categorical variable using appropriate thresholds (e.g., percentile-based cutoffs)
Survival analysis approaches:
Addressing tumor heterogeneity:
Evaluating therapeutic implications:
Interaction tests to identify patient subgroups most likely to benefit from MCAM-targeted therapy
Network analysis to identify co-expressed genes and pathways
Integration with drug sensitivity data to identify potential combination strategies
The TCGA analysis of MCAM in melanoma illustrates these principles, showing that while patients with high versus low MCAM expression showed a trend toward different melanoma-specific survival, this did not reach statistical significance . This highlights the importance of adequately powered studies and careful statistical analysis when evaluating MCAM as a biomarker.
Comparing efficacy of different anti-MCAM antibodies requires systematic evaluation across multiple parameters:
Binding characteristics assessment:
Determine binding affinity (KD) for each antibody using surface plasmon resonance
Map epitopes to identify whether antibodies target distinct MCAM domains
Evaluate cross-reactivity with other proteins and across species
Assess binding to different MCAM glycoforms
Functional assay battery:
Standardized comparison framework:
Use consistent experimental conditions, cell lines, and animal models
Include appropriate controls (isotype-matched non-specific antibodies)
Test multiple antibody concentrations to generate dose-response curves
Normalize data to account for differences in antibody potency
Statistical analysis:
Use appropriate statistical tests for each endpoint
Calculate and compare effect sizes rather than just p-values
Perform power analysis to ensure adequate sample size
Consider multilevel modeling to account for variability across experiments
Translational relevance:
Evaluate pharmacokinetics and biodistribution in relevant animal models
Assess potential immunogenicity for humanized/human antibodies
Consider combination with standard therapies
Identify biomarkers predictive of response
Results from these systematic comparisons can identify the most promising antibody candidates for further development. For example, studies with ABX-MA1 showed significant inhibition of tumor growth and metastasis in melanoma models, suggesting its potential utility as a therapeutic agent . Similar comprehensive assessment of other anti-MCAM antibodies would facilitate informed selection for clinical development.
MCAM-targeting antibodies offer several potential advantages over current immunotherapies:
Dual-action mechanism:
Targeted approach to inflammation:
Potential for combination therapy:
Complementary mechanism to existing immunotherapies and targeted therapies
ABX-MA1 showed efficacy in inhibiting tumor growth and metastasis, suggesting potential as a treatment modality either alone or in combination with conventional chemotherapy or other antitumor agents
Different mode of action from current MS therapies like natalizumab (anti-VLA-4)
Novel applications in difficult-to-treat cancers:
Potential utility in cancers with limited treatment options like mesothelioma
Targeting of MCAM on mesothelioma cells provides tumor specificity as normal mesothelial cells do not express MCAM
Internalizing anti-MCAM antibodies can deliver lethal doses of liposome-encapsulated small molecule drugs specifically to tumor cells
These advantages position MCAM-targeting antibodies as promising candidates for integration into current treatment paradigms, potentially enhancing efficacy while reducing systemic side effects compared to conventional approaches.
Comprehensive evaluation of off-target effects requires a systematic approach:
Cellular specificity profiling:
Immunohistochemical analysis of MCAM expression across normal human tissues
Flow cytometry screening of primary human cells from various tissues
Single-cell RNA sequencing to identify all MCAM-expressing cell populations
Special attention to immune cell subsets, particularly Th17 cells and NK cell populations
Molecular specificity assessment:
Cross-reactivity testing against related proteins
Epitope mapping to identify potential shared epitopes with other proteins
Screening against protein arrays to detect unexpected binding partners
Functional assays in non-target cells:
In vivo toxicology:
Mitigation strategies:
Development of antibodies with tumor-selective binding properties
Consideration of antibody-drug conjugates for enhanced specificity
Dose optimization to maximize therapeutic window
Combination with other agents to allow dose reduction
Understanding MCAM's developmental functions, such as its role in establishing cell autonomous polarity and its high expression in fetal tissues , provides crucial context for anticipating potential off-target effects. The observation that MCAM knockout in mouse models delayed but did not prevent CNS inflammation suggests some redundancy in its physiological functions, potentially reducing the risk of severe adverse effects from therapeutic targeting .
Several innovative delivery approaches can enhance anti-MCAM antibody efficacy:
Antibody-drug conjugates (ADCs):
Leverage MCAM internalization to deliver cytotoxic payloads specifically to MCAM-expressing cells
Particularly valuable for cancer applications, especially mesothelioma where MCAM expression differentiates tumor cells from normal mesothelium
Potential payloads include auristatins, maytansinoids, or DNA-damaging agents
Bispecific antibody platforms:
Dual targeting of MCAM and complementary pathways (e.g., MCAM + immune checkpoint)
Simultaneous targeting of MCAM on both tumor cells and endothelial cells
Recruitment of immune effector cells to MCAM-expressing tumors
Nanoparticle-based delivery:
Local delivery approaches:
Intratumoral administration for enhanced local concentration
Implantable devices for sustained release
For neuroinflammatory applications, delivery strategies to enhance CNS penetration
Combination with blood-brain barrier modulation:
For CNS applications, temporary disruption of the BBB to enhance antibody delivery
Focused ultrasound techniques to enable targeted BBB opening
Shuttle peptides for enhanced transcytosis across the BBB
Evidence supporting these approaches includes successful use of internalizing anti-MCAM antibodies to deliver liposome-encapsulated small molecule drugs to both epithelioid and sarcomatous subtypes of mesothelioma cells , and the demonstration that quantum dot-labeled anti-MCAM single-chain antibodies can effectively target primary mesothelioma cells in tumor fragment spheroids cultured ex vivo . These findings provide proof-of-concept for advanced delivery strategies that could significantly enhance the therapeutic potential of anti-MCAM antibodies.
Several complementary approaches can facilitate identification of novel MCAM ligands and interaction partners:
Proximity labeling proteomics:
BioID or APEX2 fusion proteins to identify proteins in close proximity to MCAM
Crosslinking mass spectrometry to capture transient interactions
Application in multiple cell types to identify context-specific interaction partners
Systematic screening approaches:
Protein microarray screening with recombinant MCAM extracellular domain
Cell-based binding assays with MCAM-Fc fusion proteins
CRISPR activation/interference screens to identify genes affecting MCAM function
Structural biology techniques:
X-ray crystallography or cryo-EM of MCAM complexes with known and putative ligands
Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces
Computational modeling of potential interactions based on structural data
Functional validation strategies:
Targeted disruption of candidate interactions using domain-specific antibodies
Mutational analysis of key residues in MCAM and proposed ligands
Competition assays with soluble MCAM variants
Recent research has identified laminin 411 (composed of α4, β1, γ1 chains) as a major ligand of MCAM in the context of CNS inflammation , and preliminary data suggests matriptase may be another potential binding partner expressed by a small fraction of CD4 memory T cells . Beyond these, both homophilic MCAM-MCAM interactions and heterophilic interactions with unidentified partners appear to play roles in cancer progression and metastasis . Systematic application of these approaches would help complete the picture of MCAM's interaction network, potentially revealing new therapeutic targets.
Exploiting MCAM's developmental regulation offers promising avenues for targeted cancer therapies:
Targeting the developmental reactivation program:
Identify transcription factors driving MCAM re-expression in cancer
Develop inhibitors targeting these upstream regulators
Combine MCAM antibodies with drugs targeting other reactivated embryonic programs
Exploiting developmental context differences:
Identify molecular differences between MCAM signaling in embryonic versus cancer contexts
Develop antibodies specifically recognizing cancer-associated MCAM conformations or modifications
Target cancer-specific MCAM interaction partners
Developmental lineage-based strategies:
Leverage knowledge of normal developmental MCAM expression patterns to predict and target cancer cell vulnerabilities
For melanoma, exploit neural crest-specific pathways co-opted during malignant transformation
Identify synthetic lethal interactions specific to cells expressing the embryonic MCAM program
Combinatorial approaches:
Combine anti-MCAM therapy with differentiation-inducing agents
Target MCAM in conjunction with other developmentally regulated adhesion molecules
Develop therapies forcing cancer cells out of the dedifferentiated, MCAM-expressing state
Bioinformatic analysis has already demonstrated that MCAM is strongly expressed in fetal melanocytes and progressively downregulated during melanocyte maturation, with MCAM showing significant inverse correlation with melanocyte maturation markers . This pattern is evolutionarily conserved, suggesting fundamental importance in developmental processes . The hypothesis that MCAM expression in melanoma reflects reactivation of an embryonic transcriptional program provides a conceptual framework for developing therapies that specifically target this cancer-associated developmental reversion while sparing normal adult tissues.
Innovative biomarker strategies for patient selection include:
Multi-parameter MCAM profiling:
MCAM ligand co-expression analysis:
Functional MCAM assays:
Ex vivo testing of patient-derived cells for response to MCAM blockade
Assessment of MCAM-dependent signaling pathway activation
Evaluation of circulating MCAM-positive cells with metastatic potential
Imaging biomarkers:
Integrated multi-omics approaches:
Combine MCAM expression data with broader gene expression signatures
Integrate with mutational profiles to identify synergistic targeting opportunities
Use AI/machine learning to develop complex biomarker signatures predictive of response
For neuroinflammatory conditions, focal upregulation of endothelial MCAM has been observed in neuroCOVID and may serve as an early biomarker for disease development and/or clinical activity . The potential detection of shed MCAM in cerebrospinal fluid during neuroinflammation offers another promising avenue for biomarker development . In cancer, mapping the association between MCAM expression patterns and response to anti-MCAM therapies could identify patient subgroups most likely to benefit from these approaches.