ML2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
ML2 antibody; MEI2 antibody; At2g42890 antibody; F7D19.11Protein MEI2-like 2 antibody; AML2 antibody; MEI2-like protein 2 antibody
Target Names
ML2
Uniprot No.

Target Background

Function
This antibody targets a probable RNA-binding protein implicated in both meiotic and vegetative growth processes.
Database Links

KEGG: ath:AT2G42890

STRING: 3702.AT2G42890.1

UniGene: At.25505

Q&A

What is the ML2 antibody and what target epitope does it engage?

M4-3-ML2 is a glycoengineered humanized antibody targeting MCSP/CSPG4 (Melanoma-associated Chondroitin Sulfate Proteoglycan), specifically binding to a membrane-proximal epitope in the D3 domain. MCSP, also known as NG2 in mice, is a large transmembrane proteoglycan uniformly and abundantly expressed in approximately 60-80% of melanomas. The antibody was generated through mouse immunization with a linear peptide derived from the membrane-proximal D3 domain, followed by boosting with melanoma cells. Unlike antibodies targeting membrane-distal epitopes, M4-3-ML2 was selected for humanization due to its potent induction of antibody-dependent cellular cytotoxicity (ADCC) when constructed as a chimeric antibody .

What is the mechanism of action for the M4-3-ML2 antibody?

M4-3-ML2 primarily functions through antibody-dependent cellular cytotoxicity (ADCC) rather than direct cytotoxicity. The antibody binds to the native epitope on MCSP+ melanoma cells without inducing internalization, enabling it to recruit immune effector cells. When glycoengineered using GlycoMab technology, M4-3-ML2 exhibits increased binding affinity for human FcγRIIIa, resulting in enhanced ADCC potency and improved absolute killing of melanoma cell lines. Importantly, neither wildtype nor glycoengineered M4-3-ML2 (up to 10 μg/mL) induces relevant cytokine release (IL-6, TNF-α, IFN-γ) in human whole blood, supporting that MCSP is not expressed in peripheral blood mononuclear cells (PBMCs) .

What expression systems are optimal for producing ML2 antibodies?

For producing ML2 antibodies, mammalian expression systems using transient gene expression in suspension cultures have proven highly effective. Vector systems like the TGEX™ vector series are specifically designed for rapid antibody expression. For murine Lambda 2 light chain expression, vectors such as TGEX-LC-mL2-Zeo can be employed alongside heavy chain vectors. Using transient transfection technologies in serum-free conditions can yield between 20-200 mg/L of antibody in just a few days. The TGEX-LC-mL2-Zeo vector contains the CMV promoter with TPL, a pelBK leader peptide, and allows for selection using ampicillin (in bacteria) and zeocin (in mammalian cells). Expression of full-length antibodies is achieved by co-transfection with a heavy chain variable region cloned into a complementary vector .

How does glycoengineering enhance the function of ML2 antibodies?

Glycoengineering significantly enhances the therapeutic potential of ML2 antibodies through several key mechanisms:

What methodologies are used to assess ML2 antibody binding characteristics?

Several complementary methodologies are employed to comprehensively characterize ML2 antibody binding properties:

  • Cell binding assays: Flow cytometry to assess specific binding to MCSP+ melanoma cells and evaluate binding patterns.

  • Immunohistochemistry (IHC): Specific staining of MCSP+ cells in formalin-fixed paraffin-embedded tissue (FFPET) samples.

  • Surface Plasmon Resonance (SPR): Determination of binding kinetics and affinity constants. The M4-3-ML2 antibody demonstrates approximately 10 nM monovalent affinity for the human MCSP D3 domain .

  • ELISA-based binding assays: Serial dilutions of antibodies are added to antigen-coated plates, followed by detection with enzyme-conjugated secondary antibodies. This approach is widely used for initial screening and comparative analysis of binding .

  • Competition assays: Evaluation of epitope specificity by competing the binding with known ligands or other antibodies with established binding sites.

How can ML2 antibody efficacy be optimized in disseminated melanoma models?

Optimizing ML2 antibody efficacy in disseminated melanoma models requires a multifaceted approach:

  • Dosage optimization: Studies with glycoengineered M4-3-ML2 have demonstrated efficacy in disseminated melanoma models, but dose-response relationships should be established for each model system.

  • Selection of appropriate models: The hCD16 transgenic Scid mouse model has proven valuable for evaluating ADCC-dependent efficacy, as these mice express functional human high-affinity FcγRIIIa receptors on NK cells.

  • Combination therapy approaches: While not explicitly tested with ML2 antibodies in the available data, combination with other therapeutic modalities (checkpoint inhibitors, targeted therapies, or anti-angiogenic agents) may enhance efficacy.

  • Timing of administration: Both glycoengineered LC007 and M4-3-ML2 have shown efficacy in disseminated models of MV3 and MDA-MB435 melanoma after intravenous injection of tumor cells, suggesting early intervention may be beneficial .

  • Biomarker-guided patient selection: For potential clinical translation, identification of biomarkers beyond MCSP expression that predict response to ML2 antibody therapy would be valuable.

What role does ML2 antibody play in targeting tumor neovasculature?

ML2 antibodies offer unique opportunities for targeting tumor neovasculature due to MCSP/CSPG4 expression patterns:

  • Pericyte-targeted mechanism: MCSP (NG2 in mice) serves as a marker of pericyte recruitment in tumor neovasculature. These pericytes play critical roles in vascular stabilization and maturation.

  • Differential expression pattern: MCSP is present at high levels on pericytes of tumor neovasculature but becomes downregulated as vessels mature, providing a potential therapeutic window targeting immature tumor vessels.

  • Anti-angiogenic potential: By targeting pericytes, ML2 antibodies may disrupt vessel stabilization, potentially leading to vessel collapse or normalization.

  • Monitoring methodologies: Assessment of anti-angiogenic effects can be performed using techniques such as dynamic contrast-enhanced MRI, vessel density quantification by immunohistochemistry, or intravital microscopy in appropriate model systems.

  • Ongoing investigations: Further studies investigating the anti-angiogenic effect of MCSP antibodies via their action on pericytes/vascular smooth muscle cells are currently underway, suggesting this is an active area of research interest .

How do humanized ML2 antibodies compare to chimeric versions in terms of ADCC potency?

The comparison between humanized and chimeric ML2 antibodies reveals important differences in ADCC potency and other functional properties:

What methodologies are most effective for assessing ML2 antibody-mediated cytotoxicity?

Several complementary methodologies can be employed to comprehensively assess ML2 antibody-mediated cytotoxicity:

  • ADCC assays: Using purified NK cells or PBMCs as effector cells and MCSP+ melanoma cell lines as targets. Cytotoxicity can be quantified through chromium release, fluorescent dye release, or real-time cell analysis systems.

  • Whole blood cytokine release assays: Measuring cytokine (IL-6, TNF-α, IFN-γ) release in human whole blood can assess potential off-target effects. Studies with M4-3-ML2 showed no relevant cytokine release up to 10 μg/mL .

  • In vivo efficacy models: The hCD16 transgenic Scid mouse model with disseminated melanoma has proven valuable for evaluating ADCC-dependent in vivo efficacy.

  • Flow cytometry-based killing assays: Multi-parameter flow cytometry can be used to simultaneously assess target cell death, effector cell activation, and antibody binding.

  • Real-time imaging of cytotoxicity: Live-cell imaging techniques can provide insights into the kinetics and mechanisms of antibody-mediated killing.

What approaches enhance reproducibility in therapeutic antibody development?

Ensuring reproducibility in therapeutic antibody development, particularly for complex molecules like ML2 antibodies, requires rigorous methodological approaches:

  • Standardized expression systems: Using well-characterized vector systems like TGEX™ vectors ensures consistent antibody expression. The TGEX-LC-mL2-Zeo vector provides a standardized platform for murine Lambda 2 light chain expression .

  • Quality control metrics: Comprehensive characterization of antibody properties including:

    • Binding affinity determination via SPR

    • Glycan profiling for glycoengineered antibodies

    • Batch-to-batch consistency testing

  • Validated functional assays: Establishing robust assays with appropriate controls for:

    • ADCC activity using standardized effector-to-target ratios

    • Target binding across multiple MCSP+ cell lines

    • Cytokine release in whole blood assays

  • Humanization strategy validation: For humanized antibodies like M4-3-ML2, comparing properties to the parental mouse antibody ensures the humanization process preserves critical functional attributes.

  • Cross-species reactivity testing: Confirming reactivity with both human and non-human primate (e.g., Cynomolgus) targets facilitates translational development .

How can surface plasmon resonance be optimized for ML2 antibody characterization?

Surface plasmon resonance (SPR) is a powerful technique for characterizing antibody-antigen interactions with ML2 antibodies:

  • Experimental setup optimization:

    • Immobilization of purified target protein (e.g., MCSP D3 domain) onto a CM5 chip in 1× PBS buffer (pH 7.4)

    • Testing different concentrations of antibodies in HBS-EP buffer

    • Surface regeneration with 10 mM NaOH or 10 mM glycine-HCl (pH 2.0)

  • Data analysis approach:

    • Fitting sensograms with a 1:1 binding model using BIA Evaluation software

    • Determination of association (ka) and dissociation (kd) rate constants

    • Calculation of equilibrium dissociation constant (KD)

  • Comparative analyses:

    • Direct comparison of binding kinetics between humanized and parental antibodies to confirm consistency

    • Evaluation of binding to different domains or epitopes of the target protein .

What strategies exist for adapting ML2 antibody technology to target emerging viral pathogens?

While the search results primarily discuss ML2 antibodies in the context of melanoma, the antibody development methodologies can be adapted for emerging viral pathogens:

  • B cell isolation techniques: Similar to approaches used for developing MERS-CoV antibodies, B cells from convalescent patients can be isolated and sorted using viral protein probes .

  • Structure-based design: Rational design strategies can be employed, such as using prefusion-stabilized viral surface proteins to enhance antibody generation, as demonstrated with MERS-CoV, SARS-CoV, HIV, and RSV .

  • Transgenic mouse models: Human receptor transgenic mice (such as hDPP4 or hACE2 mice) provide valuable platforms for testing therapeutic efficacy of antibodies against viral pathogens .

  • Binding assay adaptation: ELISA-based binding assays similar to those used for ML2 antibody characterization can be adapted for viral target proteins by coating plates with 0.1 μg/well of viral protein in PBS at 4°C overnight, blocking with 5% skim milk and 2% bovine serum albumin, and detecting binding with HRP-conjugated secondary antibodies .

  • Expression vector utilization: The TGEX™ vector system used for ML2 antibody expression can be adapted for rapid production of antiviral antibodies, enabling yields of 20-200 mg/L in just a few days .

How does ML2 antibody compare with other therapeutic approaches targeting the same pathway?

A comparative analysis of ML2 antibodies with alternative therapeutic approaches targeting MCSP/CSPG4:

What are the key considerations for translating ML2 antibody research to clinical applications?

Translating ML2 antibody research from preclinical studies to clinical applications requires addressing several key considerations:

  • Safety assessment:

    • Evaluation of on-target, off-tumor effects due to low-level MCSP expression in normal tissues

    • Monitoring for potential cytokine release, as has been done in preclinical studies showing minimal cytokine production with M4-3-ML2

  • Manufacturing considerations:

    • Scalable production using systems like the TGEX™ vector platform

    • Ensuring consistency in glycoengineering for antibodies relying on enhanced ADCC

  • Patient selection strategies:

    • Developing companion diagnostics for MCSP/CSPG4 expression

    • Identifying biomarkers predictive of response

  • Clinical trial design:

    • Leveraging experience from other antibody therapeutics targeting surface antigens

    • Consideration of combination approaches with established melanoma therapies

  • Regulatory pathway:

    • Engaging regulatory authorities early regarding novel aspects like glycoengineering

    • Addressing potential questions about target specificity and safety

What emerging technologies might enhance ML2 antibody development and applications?

Several emerging technologies hold promise for advancing ML2 antibody development and expanding applications:

  • Advanced glycoengineering platforms that provide greater control over antibody glycan structures, potentially enhancing ADCC activity while maintaining favorable pharmacokinetic properties.

  • Cell-free expression systems that could complement traditional mammalian cell culture approaches like those using TGEX™ vectors, potentially accelerating early-stage antibody production and screening .

  • AI-driven antibody optimization to improve binding characteristics, stability, and manufacturability of ML2 antibodies.

  • Novel imaging approaches for visualizing antibody biodistribution and target engagement, particularly relevant for assessing effects on tumor neovasculature where MCSP/CSPG4 is expressed on pericytes .

  • Combination approaches with immune checkpoint inhibitors or targeted therapies, which could enhance the therapeutic efficacy of ML2 antibodies against melanoma and potentially expand the range of responsive tumors.

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.