MCAM Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% sodium azide, 50% glycerol, pH 7.3. Stored at -20°C. Avoid freeze-thaw cycles.
Lead Time
Product dispatch occurs within 1-3 business days of order receipt. Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
A32 antigen antibody; CD 146 antibody; CD146 antibody; CD146 antigen antibody; Cell surface glycoprotein MUC18 antibody; Cell surface glycoprotein P1H12 antibody; Gicerin antibody; Mcam antibody; Melanoma adhesion molecule antibody; Melanoma associated antigen A32 antibody; Melanoma associated antigen MUC18 antibody; Melanoma associated glycoprotein MUC18 antibody; Melanoma cell adhesion molecule antibody; Melanoma-associated antigen A32 antibody; Melanoma-associated antigen MUC18 antibody; MelCAM antibody; MUC 18 antibody; MUC18 antibody; MUC18_HUMAN antibody; S endo 1 antibody; S endo 1 endothelial associated antigen antibody; S-endo 1 endothelial-associated antigen antibody
Target Names
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
MCAM/CD146 Research Summary: The following publications highlight the diverse roles of MCAM (also known as CD146) across various biological processes and disease contexts. Further research is ongoing to fully elucidate its multifaceted functions.
  • MCAM's role in coordinating apical-basal and planar cell polarity provides insights into morphogenesis mechanisms. PMID: 28589943
  • CD146 suppresses breast cancer progression as a downstream target of CD44 signaling. PMID: 29121955
  • Analysis of cultured adipose-derived stem cells (ASCs) revealed low levels of CD146 mRNA, expressed in two splicing variants: a predominantly long form and a smaller amount of short form. Protein expression mirrored this pattern. PMID: 28549249
  • MUC18/Muc18 may regulate airway inflammation and mucus overproduction in asthma, suggesting it as a potential therapeutic target. PMID: 28451734
  • CD146 promoter polymorphisms showed no association with clear cell renal carcinoma risk in a Chinese population, but rs3923594 predicted recurrence in localized cases. PMID: 28626293
  • Two distinct cancer-associated fibroblast (CAF) subtypes, defined by CD146 expression, exist in ER+ breast cancers. CD146-negative CAFs suppress ER expression, decrease estrogen sensitivity, and increase tamoxifen resistance in tumor cells. PMID: 27702820
  • CD146 suppresses tumorigenesis and cancer stemness in colorectal cancer by inactivating the Wnt/β-catenin pathway. PMID: 27302922
  • A novel class of committed myogenic progenitors in human postnatal skeletal muscle, distinct from satellite cells, was identified within the subendothelial cells of the microvascular compartment. PMID: 29186180
  • CD146 acts as a retention signal for macrophages in artery walls, representing a potential therapeutic target in atherosclerosis. PMID: 28084332
  • CD146 is expressed in all Philadelphia chromosome-positive B-cell acute lymphoblastic leukemia cases and most T-cell acute lymphoblastic leukemia cases. PMID: 26102234
  • MCAM may serve as a therapeutic target to overcome chemoresistance in small cell lung cancer. PMID: 28646020
  • KDM3A regulates MCAM expression both directly (modifying H3K9 methylation at the MCAM promoter) and indirectly (via the Ets1 transcription factor). PMID: 28319067
  • Increased CD146 expression in cultured annulus fibrosus cells indicates a shift towards a contractile phenotype, enhanced by transforming growth factor beta1. PMID: 27273299
  • CD146 positivity in malignant rhabdoid tumor (MRT) samples correlates with poor patient outcomes, suggesting it as a therapeutic target. PMID: 27041577
  • Promoter methylation of MCAM, ERα, and ERβ may serve as biomarkers for early prostate cancer detection. PMID: 28147335
  • MCAM promotes tamoxifen resistance by suppressing ERα expression and activating the AKT pathway, leading to epithelial-mesenchymal transition. PMID: 27838413
  • High MCAM expression is associated with lung metastasis in malignant melanoma. PMID: 27151304
  • MUC18 promotes viral infections in vivo and in vitro. PMID: 27701461
  • Soluble CD146 is released from the peripheral vasculature in response to venous stretch and may indicate systemic congestion in chronic heart failure. PMID: 28062630
  • Radiolabeled OI-3 antibodies can target CD146 in vivo. PMID: 27776176
  • Decreased CD146 expression in cancer-associated fibroblasts promotes pancreatic cancer progression. PMID: 26373617
  • METCAM/MUC18 positively promotes tumorigenesis of SK-BR-3 breast cancer cells by increasing survival and proliferation signaling. PMID: 27125403
  • Elevated sCD146 levels are observed in systemic sclerosis, but decreased levels are found in patients with pulmonary arterial hypertension. PMID: 27726047
  • Nestin and CD146 are expressed in highly aggressive breast cancer cells, potentially contributing to relapse via epithelial-mesenchymal transition and neovascularization. PMID: 28347241
  • A specific anti-MUC18 scFv antibody inhibited MUC18-positive cell line migration (76%) and invasion (67%), suggesting its potential for breast cancer immunotherapy. PMID: 27565656
  • Pro-angiogenic genes PECAM1, PTGS1, FGD5, and MCAM play a role in psoriatic dermal angiogenesis. PMID: 26748901
  • Increased METCAM/MUC18 expression in SK-OV-3 ovarian cancer cells suppressed tumorigenesis and ascites formation, indicating a suppressor role in ovarian cancer progression. PMID: 26906545
  • CD146 defines a subpopulation of human mesenchymal stem cells capable of bone formation and trans-endothelial migration. PMID: 26753846
  • CD146 promotes hepatocellular carcinoma (HCC) cell metastasis and predicts poor prognosis, potentially through IL-8 upregulation and STAT1 downregulation. PMID: 26928402
  • CD146 is an effective marker for enriching tumor-propagating cells in primary sarcomas. PMID: 26517673
  • MUC18 is an independent prognostic factor for clear cell renal cell carcinoma. PMID: 26617818
  • CD146 predicts senescence in human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), with potential applications in quality control for hUCB-MSC-based therapies. PMID: 26941359
  • ZBTB7A directly represses MCAM expression by binding to its promoter. PMID: 25995384
  • CD146 and HIF1α expression correlates with EGFR and CD31 expression, respectively, in salivary gland adenoid cystic carcinoma. PMID: 25997612
  • CD166 regulates MCAM through PI3K/AKT and c-Raf/MEK/ERK signaling, inhibiting betaTrCP and Smurf1 E3 ligases. PMID: 26004137
  • Combined EpCAM/MCAM CellSearch enrichment improves circulating tumor cell (CTC) detection rates. PMID: 25552367
  • MCAM is a novel YAP target in hepatocellular carcinoma (HCC), with elevated serum levels suggesting its potential as a diagnostic marker. PMID: 25728681
  • CD146, p53, and Ki-67 are overexpressed in uterine sarcoma. PMID: 26293576
  • CDCP1 expression identifies a subset of marrow fibroblasts distinct from CD146+ fibroblasts. PMID: 25275584
  • CD146 expression in gastric neoplasm cells correlates with lymph node metastasis and epithelial-mesenchymal transition markers. PMID: 22754372
  • MCAM, a major galectin-1 ligand, is largely dependent on melanoma malignancy. PMID: 25756799
  • MCAM is expressed by effector CD8+ T lymphocytes and is upregulated during multiple sclerosis relapses. MCAM blockade restricts CD8+ T lymphocyte transmigration across the blood-brain barrier. PMID: 25869475
  • HuMETCAM/MUC18 levels are significantly higher in ovarian carcinomas and metastatic lesions than in normal tissues and cystadenomas. PMID: 25510693
  • MUC18's pro-angiogenic potency may contribute to collateral angiogenesis in peripheral stenotic arteriosclerotic disease, but not in dilatative aortic diseases. PMID: 25729916
  • Endothelial CD146 is proposed as a target for specific drug delivery in hepatocellular carcinoma. PMID: 25238265
  • Sema 3A regulates PTEN, FOXO3a, and MelCAM to suppress breast cancer growth and angiogenesis. PMID: 24727891
  • Ets transcription factors upregulate MCAM in an Akt and ERK/MEK-dependent manner, suggesting potential therapeutic targets in melanoma. PMID: 24743054
  • MCAM overexpression indicates melanoma progression. PMID: 24902661
  • CD146+ chondroprogenitors in advanced osteoarthritis cartilage exhibit migratory and survival capabilities, suggesting their potential for cartilage regeneration. PMID: 25266708
  • Functional characterization of N-acetylglucosaminyltransferases III and V in human melanoma cells. PMID: 24726881
Database Links

HGNC: 6934

OMIM: 155735

KEGG: hsa:4162

STRING: 9606.ENSP00000264036

UniGene: Hs.599039

Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Detected in endothelial cells in vascular tissue throughout the body. May appear at the surface of neural crest cells during their embryonic migration. Appears to be limited to vascular smooth muscle in normal adult tissues. Associated with tumor progress

Q&A

What is MCAM/CD146 and why is it significant in research?

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.

What cell types express MCAM and how does expression vary during 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.

How do MCAM antibodies help elucidate mechanisms of melanoma metastasis?

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 .

What is the controversy surrounding MCAM as a biomarker for melanoma progression?

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.

How can MCAM antibodies be used to study angiogenesis in cancer research?

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 .

What evidence suggests MCAM is involved in neuroinflammation and CNS infiltration?

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.

How do MCAM antibodies affect T cell function in neuroinflammatory models?

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 .

What are the optimal methods for validating MCAM antibody specificity?

Validating MCAM antibody specificity requires a multi-faceted approach combining genetic, biochemical, and functional methods:

  • Genetic validation:

    • Use MCAM knockout cell lines or tissues as negative controls in all applications

    • Confirm knockout by next-generation sequencing of the targeted locus

    • Validate absence of protein expression by Western blot analysis

  • 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:

    • Perform knockdown/knockout phenotype rescue experiments with MCAM expression constructs

    • Demonstrate that antibody-mediated neutralization reproduces knockout phenotypes

    • Test antibody effects on known MCAM-dependent processes such as cell adhesion or migration

A comprehensive validation approach should demonstrate consistent results across multiple techniques and experimental systems, with appropriate positive and negative controls.

What are the recommended protocols for MCAM detection in different experimental systems?

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

  • Expected staining pattern: cell membranes and cytoplasm

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

How can researchers design effective MCAM knockdown/knockout experiments?

Designing effective MCAM knockdown/knockout experiments requires careful planning and appropriate controls:

  • Selection of experimental system:

    • Choose cell lines with confirmed MCAM expression

    • For cancer studies, consider using cell lines with metastatic potential (e.g., A375SM, WM2664 for melanoma)

    • For endothelial studies, use primary HUVECs or established lines like bEND

  • CRISPR/Cas9 knockout design:

    • Target early exons to ensure complete protein disruption

    • Design multiple guide RNAs to increase efficiency

    • Validate knockout by sequencing the targeted locus and confirming absence of protein by Western blot

    • Generate and validate multiple knockout clones to account for clonal variation

  • 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:

    • Compare MCAM knockout/knockdown cells with control cells for proliferation rates to rule out non-specific effects

    • Test functional phenotypes in context-appropriate assays (e.g., migration on endothelial monolayers for metastasis studies)

    • Consider both in vitro and in vivo validation when possible

  • Rescue experiments:

    • Reintroduce wild-type or mutant MCAM to knockout cells to confirm specificity

    • For domain-specific studies, use constructs with specific deletions (e.g., endocytosis motif)

    • Ensure appropriate expression levels to avoid overexpression artifacts

By following these guidelines, researchers can generate reliable data on MCAM function across different experimental systems while minimizing technical and biological artifacts.

How should researchers interpret contradictory findings regarding MCAM's role in different cancer types?

Interpreting contradictory findings regarding MCAM's role in different cancer types requires systematic consideration of multiple factors:

  • Tissue-specific context:

    • MCAM may have different binding partners and downstream signaling pathways depending on the tissue microenvironment

    • Expression of MCAM ligands (e.g., laminin 411) varies across tissues

    • The functional consequence of MCAM expression may depend on co-expressed molecules within specific cancer types

  • Methodological differences:

    • Studies use varied methodologies (IHC, RNA-seq, functional assays) with different sensitivities

    • Antibodies targeting different MCAM epitopes may yield inconsistent results

    • Thresholds for defining "high" versus "low" expression differ between studies

  • Cancer heterogeneity:

    • MCAM shows high intra- and inter-tumoral heterogeneity

    • Different cancer subtypes within the same organ may show opposing MCAM functions

    • Stage-dependent effects may explain apparent contradictions

  • 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.

What statistical approaches are recommended for analyzing MCAM expression data in patient cohorts?

Analysis of MCAM expression data in patient cohorts requires robust statistical approaches to account for heterogeneity and potential confounding factors:

  • Data preprocessing and normalization:

    • For RNA-seq data, appropriate normalization methods (e.g., TPM, RPKM, or variance stabilizing transformation)

    • For immunohistochemistry, standardized scoring systems accounting for both staining intensity and percentage of positive cells

    • Batch correction when combining data from multiple sources

  • 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:

    • Kaplan-Meier analysis with log-rank test for initial assessment of prognostic value

    • Cox proportional hazards models for multivariate analysis

    • Competing risk analysis when appropriate

    • Consider time-dependent effects of MCAM expression

  • Addressing tumor heterogeneity:

    • Stratification by molecular subtypes

    • Analysis of MCAM expression in specific cell populations within the tumor microenvironment

    • Single-cell analysis when available to assess MCAM expression patterns at cellular resolution

  • 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.

How can researchers effectively compare the efficacy of different anti-MCAM antibodies in preclinical models?

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:

    • In vitro assays: cell migration, invasion, tube formation, spheroid disruption

    • Ex vivo assays: transmigration across endothelial monolayers, binding to tumor fragment spheroids

    • In vivo models: tumor growth inhibition, metastasis suppression, immune cell infiltration

  • 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.

What are the potential advantages of MCAM-targeting antibodies compared to current immunotherapies?

MCAM-targeting antibodies offer several potential advantages over current immunotherapies:

  • Dual-action mechanism:

    • Simultaneously target both tumor cells and tumor vasculature, as MCAM is expressed on both cancer cells and endothelial cells

    • Disrupt both homotypic interactions (MCAM-MCAM) and heterotypic interactions (MCAM-laminin)

    • Inhibit multiple processes including tumor growth, angiogenesis, and metastasis

  • Targeted approach to inflammation:

    • More selective than broad immunosuppressive therapies

    • Specifically block pathogenic T cell migration into the CNS without global immune suppression

    • Effects restricted to the time and locus of inflammation rather than systemic effects

  • 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.

How can researchers evaluate potential off-target effects of MCAM-targeting therapeutic antibodies?

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:

    • Effects on normal endothelial cell function and vascular integrity

    • Impact on immune cell subsets not directly targeted

    • Assessment of key developmental processes where MCAM plays a role (e.g., myogenesis)

  • In vivo toxicology:

    • Comprehensive toxicology studies in relevant animal models

    • Evaluation of vascular integrity in multiple organs

    • Assessment of wound healing and other physiological processes requiring cell migration

    • Special focus on the CNS given MCAM's role in the blood-brain barrier

  • 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 .

What novel delivery approaches can enhance the efficacy of anti-MCAM antibodies?

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:

    • Liposomal encapsulation of anti-MCAM antibodies for improved pharmacokinetics

    • Co-delivery of anti-MCAM antibodies with small molecule drugs

    • Targeted nanoparticles functionalized with anti-MCAM antibodies for delivery of nucleic acid therapeutics

  • 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.

What are the most promising approaches for identifying additional MCAM ligands and interaction partners?

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.

How might researchers exploit the developmental regulation of MCAM to develop more targeted cancer therapies?

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.

What novel biomarker strategies could enhance patient selection for MCAM-targeted therapies?

Innovative biomarker strategies for patient selection include:

  • Multi-parameter MCAM profiling:

    • Quantify both MCAM expression levels and activation state

    • Assess MCAM cellular localization (membrane vs. cytoplasmic)

    • Evaluate MCAM shedding into circulation as a potential liquid biopsy marker

    • Characterize MCAM glycosylation patterns, which may affect function

  • MCAM ligand co-expression analysis:

    • Assess expression of identified MCAM ligands (e.g., laminin 411)

    • Evaluate potential for functional MCAM signaling based on presence of necessary interaction partners

    • Develop multiplexed IHC or RNA-based assays for MCAM and its ligands

  • 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:

    • Development of MCAM-targeted PET imaging agents

    • MRI with anti-MCAM-coupled contrast agents

    • Use of labeled anti-MCAM antibodies in SPECT studies, as demonstrated in mesothelioma organotypic xenografts

  • 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.

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.