MET2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to MET2 Antibody

The term "MET2 Antibody" refers to therapeutic or research-grade antibodies targeting the MET receptor tyrosine kinase (RTK), a proto-oncoprotein critical in cancer cell growth, invasion, and metastasis. MET (mesenchymal-epithelial transition factor) is activated by its ligand, hepatocyte growth factor (HGF), and dysregulation of this pathway drives tumor progression in various cancers. MET2 antibodies are designed to modulate MET signaling through mechanisms such as receptor degradation, inhibition of ligand binding, or disruption of downstream signaling pathways. These antibodies are classified based on their epitope specificity, engineering (e.g., biparatopic designs), and functional outcomes (e.g., promoting lysosomal degradation) .

Mechanism of Action

MET2 antibodies employ distinct mechanisms to inhibit MET-driven oncogenesis:

  • Biparatopic Binding: A biparatopic MET×MET antibody binds two non-overlapping epitopes in the MET Sema domain, inducing irreversible receptor clustering, lysosomal degradation, and sustained inhibition of downstream signaling (e.g., ERK, AKT) .

  • Receptor Internalization: Antibodies like SAIT301 accelerate MET internalization and degradation, blocking HGF-mediated activation .

  • Clustering-Induced Downregulation: Antibody-induced MET clustering on the plasma membrane leads to transient signaling followed by rapid receptor removal and degradation .

Key Mechanistic Findings:

  • Biparatopic antibodies reduce MET recycling by 70% compared to parental antibodies, enhancing tumor suppression .

  • MET2 antibodies inhibit MET-dependent biological responses (e.g., cell migration, proliferation) by disrupting receptor stability .

Preclinical Studies

Antibody TypeModel SystemEfficacy OutcomeReference
Biparatopic MET×METMET-driven tumor models80% reduction in tumor growth vs. parental antibodies; 60% longer progression-free survival
Anti-MET (SAIT301)Colorectal cancer (CRC)Partial response in 9.1% of patients; stable disease in 36.4%
Anti-MET (APC-conjugated)Glioblastoma cellsInduces MET clustering within 5 minutes; 90% receptor degradation within 24 hours

Clinical Trials

A phase I trial of SAIT301 (anti-MET monoclonal antibody) in MET-overexpressed solid tumors demonstrated:

  • Safety: Dose-limiting toxicities included hypophosphatemia (25%) and elevated alkaline phosphatase (18.8%) .

  • Efficacy: 1/11 patients achieved partial response; 4/11 had stable disease at doses ≤3.69 mg/kg .

Applications in Therapy

MET2 antibodies are being explored for:

  • MET-Amplified Cancers: Tumors with MET exon 14 skipping mutations or amplification (e.g., glioblastoma, NSCLC) .

  • Combination Therapies: Synergy with tyrosine kinase inhibitors (TKIs) or immune checkpoint inhibitors .

  • Resistance Mitigation: Biparatopic antibodies prevent compensatory signaling seen with single-target therapies .

Comparative Analysis with Other MET-Targeting Agents

FeatureBiparatopic MET×METMonovalent MET AntibodiesSmall-Molecule TKIs
Receptor DegradationYes (lysosomal)PartialNo
Recycling Inhibition70% reduction≤30% reductionNot applicable
Clinical Response80% tumor growth inhibition30–50% inhibitionVariable (resistance common)
ToxicityLow (transient signaling)ModerateHigh (off-target effects)

Data derived from .

Challenges and Future Directions

  • Biomarker Identification: Improved patient stratification using MET overexpression or mutation status .

  • Resistance Mechanisms: Addressing compensatory pathways (e.g., AXL, EGFR) through combinatorial approaches .

  • Next-Generation Engineering: Bispecific antibodies targeting MET and immune checkpoints (e.g., PD-1/PD-L1) .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
MET2 antibody; METIIb antibody; At4g08990 antibody; F23J3.20DNA antibody; cytosine-5)-methyltransferase 2 antibody; EC 2.1.1.37 antibody; DNA methyltransferase 2 antibody; DNA methyltransferase IIb antibody
Target Names
MET2
Uniprot No.

Target Background

Function
MET2 Antibody maintains chromatin CpG methylation, a critical process in genomic imprinting, embryogenesis, and seed viability. It is essential for establishing correct CG DNA methylation patterns in dividing cells.
Database Links

KEGG: ath:AT4G08990

STRING: 3702.AT4G08990.1

UniGene: At.54221

Protein Families
Class I-like SAM-binding methyltransferase superfamily, C5-methyltransferase family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed at low levels in vegetative and floral organs.

Q&A

What is the Ma2 antibody and what is its clinical significance?

Ma2 antibodies are IgG biomarkers found in patients with paraneoplastic encephalitis (limbic encephalitis or brainstem encephalitis) or cerebellar ataxia. They serve as critical diagnostic indicators for autoimmune central nervous system disorders. Methodologically, testing for Ma2 antibodies involves enzyme-linked immunosorbent assay (ELISA) of serum samples. The presence of these antibodies strongly suggests an autoimmune etiology and potentially a paraneoplastic basis for neurological symptoms, particularly when patients present with encephalopathy, dementia, seizures, brainstem dysfunction, or cerebellar ataxia .

How do Ma2 antibodies relate to associated cancer types?

Ma2 antibody seropositivity shows distinct cancer association patterns. When only Ma2 antibodies are detected (without Ma1), there is a strong association with testicular germinoma (both testicular and extra-testicular). When both Ma1 and Ma2 antibodies are present simultaneously, the cancer associations become more diverse. This differential pattern necessitates targeted cancer screening approaches based on the specific antibody profile detected. Methodologically, clinicians should prioritize thorough testicular examination and imaging in Ma2-positive male patients .

What is the relationship between Ma1 and Ma2 antibodies?

Antibodies to Ma antigens demonstrate a specific pattern: they can be found directed at Ma2 alone, or at both Ma1 and Ma2 simultaneously, but never at Ma1 alone. This unique relationship serves as an important diagnostic consideration. The pattern of positivity (Ma2-only versus Ma1+Ma2) has significant implications for predicting cancer types and treatment responses. Methodologically, comprehensive testing for both antibodies provides more valuable clinical information than testing for either in isolation .

What is the detailed mechanism of Ma2 antibody detection via ELISA?

The Ma2 autoantibody ELISA utilizes an indirect ELISA methodology where:

  • Ma2 autoantibodies in patient serum bind to PNMA2 (the antigenic protein) coated on ELISA plate wells

  • An anti-human secondary antibody specifically recognizes the Fc region of the bound autoantibody

  • The secondary antibody is conjugated with alkaline phosphatase that enzymatically hydrolyzes a substrate

  • This hydrolysis produces a yellow-colored end product measurable at 405 nm wavelength

The precise protocol involves:

  • Adding controls and diluted samples in duplicate to coated plate wells

  • Incubating for 2 hours followed by extensive washing

  • Adding conjugated secondary antibody and incubating for 1 hour

  • Washing again before adding p-nitrophenyl phosphate substrate

  • Final 1-hour incubation before measuring absorbance at 405 nm

How do treatment responses differ between Ma2-only positive patients versus Ma1+Ma2 positive patients?

Neurological improvement following cancer treatment or immunotherapy shows significant variation based on antibody profile. Patients seropositive for Ma2 only demonstrate more frequent and substantial neurological improvement compared to those with both Ma1 and Ma2 antibodies. This differential response pattern has important implications for prognosis discussions and treatment planning. Methodologically, longitudinal tracking of neurological outcomes in patients with different antibody profiles is essential for building a more complete understanding of these response patterns .

What are the limitations and interpretation challenges of Ma2 antibody testing?

A negative Ma2 antibody test result does not definitively exclude autoimmune neurological disease or underlying cancer. This critical limitation requires clinicians to maintain clinical suspicion despite negative serological results when symptoms are suggestive. Methodologically, Ma2 antibody testing should be considered one component of a comprehensive diagnostic approach rather than a standalone definitive test. Additional investigations such as CSF analysis, neuroimaging, and comprehensive cancer screening remain essential components of the evaluation process .

How can modern antibody design techniques enhance diagnostic antibody development?

Advanced computational approaches have transformed antibody engineering. Modern AI-based protocols like IsAb2.0 use AlphaFold-Multimer (2.3/3.0) to construct accurate 3D structures of antibody-antigen complexes without requiring templates. These approaches facilitate:

  • More precise modeling of antibody-antigen binding interactions

  • Prediction of antibody hotspots for targeted optimization

  • Identification of specific mutations that can enhance binding affinity

Methodologically, this represents a significant advance over previous approaches that required extensive templates and complex procedures. The application of these techniques could potentially improve the sensitivity and specificity of antibodies used in diagnostic assays for neurological conditions .

What considerations are important when evaluating universal binding potential of therapeutic antibodies?

Research on M2e-specific monoclonal antibodies demonstrates the importance of evaluating binding across diverse strains and variants. Effective methodological approaches include:

  • Testing binding against synthetic peptides representing the target epitope

  • Confirming binding to cells expressing the target protein

  • Verifying binding to whole virus particles

  • Assessing binding to infected cells expressing different viral strains

This comprehensive evaluation is critical for antibodies intended to have broad therapeutic potential. Similar approaches could be valuable when developing antibodies for detecting diverse variants of neurological antigens .

How can point mutation analysis improve antibody affinity and specificity?

Strategic point mutations can significantly enhance antibody performance. Advanced methodologies like FlexddG provide precise in silico antibody optimization by:

  • Identifying critical binding residues through alanine scanning

  • Systematically testing all possible amino acid substitutions at key positions

  • Predicting energy changes to identify mutations that enhance binding affinity

In practical applications, this approach successfully improved the binding affinity of a humanized nanobody (HuJ3) to its target (HIV-1 gp120) through the E44R mutation. Similar approaches could potentially enhance Ma2 antibodies for improved diagnostic sensitivity .

What are the key considerations for translating laboratory antibodies to clinical applications?

Translating antibodies from laboratory research to clinical applications requires addressing several challenges:

  • Humanization to reduce immunogenicity while preserving binding properties

  • Optimization of affinity to enhance sensitivity while maintaining specificity

  • Validation across diverse patient populations and clinical scenarios

  • Development of standardized detection protocols suitable for clinical laboratories

The experience with antibodies like TCN-032 (M2e-specific) demonstrates that even promising antibodies may show limited clinical efficacy. This highlights the importance of robust validation at multiple stages of development .

How do different antibody types compare in research and diagnostic applications?

Antibody TypeTargetAssociated ConditionsResearch ApplicationsClinical Utility
Ma2 AntibodyPNMA2 proteinParaneoplastic encephalitis, cerebellar ataxiaBiomarker research, autoimmune mechanism studiesDiagnosis of autoimmune CNS disorders, cancer screening guidance
M2e-MAbsMatrix Protein 2 extracellular domainInfluenza A infectionUniversal influenza treatment research, viral mechanismsPotential therapeutic applications
Humanized antibodiesVariousVariousReduced immunogenicity in human applicationsImproved therapeutic safety profile
NanobodiesVariousVariousEnhanced tissue penetration, stability studiesNovel diagnostic and therapeutic applications

This comparative analysis demonstrates how different antibody types serve specific research and clinical purposes, with methodological approaches tailored to their unique properties and applications .

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.