MAGEA8 Human

Melanoma Antigen Family A, 8 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MAGEA8

MAGEA8 is a 318-amino acid protein with a molecular mass of 37.6 kDa. It is primarily expressed in normal testis and placenta but is aberrantly activated in various cancers, including bladder carcinoma and melanoma . The gene is part of an evolutionarily conserved cluster on the X chromosome, with members sharing 50–80% sequence identity . Recombinant MAGEA8 is produced in Escherichia coli for research applications, featuring a His-tag for purification .

Table 2: Key Interaction Partners

ProteinFunctionInteraction Score
GCACalcium-binding; regulates neutrophil adhesion0.692
BEX5Neuronal differentiation0.627
SMC5DNA double-strand break repair0.494
NSMCE1E3 ubiquitin ligase activity in genome integrity maintenance0.500

Expression and Genetic Variants

  • Tissue Expression: Confined to testis (cytoplasmic/nuclear) and placenta .

  • Variants: 11 public variants reported, including 10 unique DNA changes linked to X-chromosome disorders .

Future Directions

  • Therapeutic Development: Optimize MAGEA8-derived peptides for clinical trials in HLA-A2.1+ cancers.

  • Mechanistic Studies: Clarify MAGEA8’s role in p53/Bax-mediated apoptosis and DNA repair pathways.

  • Biomarker Validation: Explore its utility in monitoring tumor recurrence or treatment response.

Product Specs

Introduction
MAGEA8, also known as Melanoma Antigen Family A 8, is a member of the MAGE gene family. This family comprises twelve known genes, six of which exhibit tumor-specific expression. Initially identified in various tumor types, MAGE genes have garnered significant attention in cancer immunotherapy due to their nearly exclusive expression in adult tumor tissues, making them promising targets for such therapies.
Description
Recombinant human MAGEA8, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 341 amino acids (residues 1-318) with a molecular weight of 37.6 kDa. The protein includes a 23 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution, sterilized by filtration.
Formulation
The MAGEA8 protein is supplied in a solution containing Phosphate Buffered Saline (PBS), 10% glycerol, and 1mM DTT at a concentration of 0.25 mg/ml.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the MAGEA8 protein is determined to be greater than 85% by SDS-PAGE analysis.
Synonyms
Melanoma Antigen Family A8, MAGE-8 Antigen, MAGE8, Cancer/Testis Antigen 1.8, CT1.8, Cancer/Testis Antigen Family 1, Member 8, Cancer/Testis Antigen Family 1, Melanoma-Associated Antigen 8, Melanoma Antigen Family A, 8, Member 8, MAGEA8.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMLLGQKS QRYKAEEGLQ AQGEAPGLMD VQIPTAEEQK AASSSSTLIM GTLEEVTDSG SPSPPQSPEG ASSSLTVTDS TLWSQSDEGS SSNEEEGPST SPDPAHLESL FREALDEKVA ELVRFLLRKY QIKEPVTKAE MLESVIKNYK NHFPDIFSKA SECMQVIFGI DVKEVDPAGH SYILVTCLGL SYDGLLGDDQ STPKTGLLII VLGMILMEGS RAPEEAIWEA LSVMGLYDGR EHSVYWKLRK LLTQEWVQEN YLEYRQAPGS DPVRYEFLWG PRALAETSYV KVLEHVVRVN ARVRISYPSL HEEALGEEKG V.

Q&A

What is MAGEA8 and what are its key molecular characteristics?

MAGEA8 (Melanoma Antigen Family A8) is a member of the MAGE protein family, specifically belonging to the type I MAGE subfamily, which are classified as cancer-testis antigens (CTAs). The human MAGEA8 protein is a single, non-glycosylated polypeptide chain containing 318 amino acids with a molecular mass of approximately 37.6 kDa . The protein contains the characteristic MAGE homology domain (MHD), which is the defining feature of all MAGE family proteins .

From a structural perspective, recombinant MAGEA8 protein produced for research typically contains 341 amino acids when fused with a His-tag at the N-terminus for purification purposes . This recombinant form is commonly used for experimental studies and can be stored as a sterile filtered clear solution containing phosphate-buffered saline, glycerol, and DTT to maintain stability .

How is MAGEA8 classified within the larger MAGE gene family?

MAGEA8 is classified as a type I MAGE family member, specifically part of the MAGE-A subfamily. The MAGE gene family is divided into two major types:

  • Type I MAGEs (including MAGE-A, MAGE-B, and MAGE-C subfamilies) - These are primarily expressed in testicular germ cells and placenta during normal development, but are aberrantly activated in various cancers, earning them the classification as cancer-testis antigens (CTAs) .

  • Type II MAGEs - These display more ubiquitous expression patterns across various tissues and are generally expressed at higher absolute levels than type I genes .

The MAGE-A subfamily, to which MAGEA8 belongs, is located on the X chromosome in a cluster that has undergone various degrees of expansion through duplication or retrotransposition events during evolution . This genomic organization is preserved across diverse mammalian species, although with species-specific variations in gene number and arrangement .

What post-translational modifications have been identified in MAGEA8?

Several post-translational modifications have been documented for MAGEA8, which likely regulate its function, stability, and interactions. The most comprehensively characterized modifications include:

SitePTM TypeSource
K135UbiquitinationPhosphoSitePlus
K144UbiquitinationPhosphoSitePlus
K147UbiquitinationPhosphoSitePlus
S229PhosphorylationPhosphoSitePlus
Y234PhosphorylationPhosphoSitePlus
K288MethylationPhosphoSitePlus
K288UbiquitinationPhosphoSitePlus

These modifications suggest complex regulatory mechanisms controlling MAGEA8 function . Notably, K288 appears to undergo both methylation and ubiquitination, potentially indicating a regulatory switch mechanism. Experimental approaches to study these modifications typically involve mass spectrometry-based proteomics, site-directed mutagenesis, and specific antibodies against the modified forms of the protein.

What is the normal expression pattern of MAGEA8 in human tissues?

MAGEA8, like other type I MAGE family members, exhibits a highly restricted expression pattern in normal adult tissues. It is primarily expressed in the testis, specifically in germ cells, with minimal or undetectable expression in other adult somatic tissues . This restricted expression pattern is characteristic of cancer-testis antigens.

During embryonic development, type I MAGE genes including MAGEA8 show expression in developing testis and ovary, suggesting a role in gametogenesis for both sexes . Some expression has also been detected in placental tissues, although the pattern differs between humans and mice .

The restricted expression in normal tissues contrasts with its aberrant activation in various cancer types, making it valuable as both a cancer biomarker and a potential therapeutic target with limited off-target effects on normal tissues .

What are the optimal experimental methods for studying MAGEA8 function in vitro?

When investigating MAGEA8 function in vitro, researchers should consider the following methodological approaches:

Protein expression and purification:

  • Recombinant MAGEA8 production in E. coli with an N-terminal His-tag (or similar affinity tag) facilitates purification via chromatographic techniques

  • For optimal stability, store purified protein at 4°C if using within 2-4 weeks, or at -20°C for longer periods with a carrier protein (0.1% HSA or BSA) to prevent degradation

  • Avoid multiple freeze-thaw cycles which can compromise protein integrity

Functional assays:

  • Protein interaction studies using pull-down assays, co-immunoprecipitation, or yeast two-hybrid systems to identify binding partners

  • In vitro ubiquitination assays to assess its potential role in protein degradation pathways

  • Phosphorylation assays to evaluate kinase interactions and signaling effects

  • Cell-based assays following ectopic expression or knockdown to assess effects on cellular phenotypes

Expression analysis:

  • RT-qPCR for sensitive detection of MAGEA8 mRNA

  • Western blotting with validated antibodies for protein detection

  • Immunohistochemistry for tissue localization studies

  • Single-cell RNA sequencing for heterogeneity analysis in complex tissues

When interpreting results, it's critical to account for the potentially overlapping functions with other MAGE family members, which may require careful experimental design with appropriate controls to isolate MAGEA8-specific effects.

How does MAGEA8 expression differ between normal tissues and cancer?

MAGEA8 expression demonstrates a striking dichotomy between normal and malignant tissues, consistent with its classification as a cancer-testis antigen:

Normal tissue expression:

  • Primarily restricted to testicular germ cells in adult tissues

  • Some expression in developing gonads (both testis and ovary) during embryonic development

  • Limited or absent expression in other adult somatic tissues

Cancer expression:

  • Aberrantly activated in various tumor types

  • Expression often correlates with advanced disease stages and poorer prognosis

  • The mechanisms of activation typically involve epigenetic changes, particularly DNA hypomethylation of the promoter region

This differential expression pattern is what makes MAGEA8 and other cancer-testis antigens attractive targets for cancer immunotherapy, as they provide relatively cancer-specific targeting opportunities with minimal risk to normal tissues . When designing experiments to assess MAGEA8 expression in clinical samples, researchers should employ multiple detection methods (RT-qPCR, immunohistochemistry, and Western blotting) for confirmation, and include appropriate positive (testicular tissue) and negative controls.

What approaches are recommended for targeting MAGEA8 in cancer immunotherapy?

As a cancer-testis antigen with restricted normal tissue expression, MAGEA8 represents a promising target for cancer immunotherapy. Researchers developing MAGEA8-targeted approaches should consider:

Peptide vaccine development:

  • Identify immunogenic epitopes from MAGEA8 that can elicit strong T-cell responses

  • Validate peptide binding to common HLA molecules through in silico prediction and experimental confirmation

  • Test peptide vaccines in combination with appropriate adjuvants to enhance immunogenicity

Adoptive T-cell therapy:

  • Engineer T cells with T-cell receptors (TCRs) or chimeric antigen receptors (CARs) specific for MAGEA8

  • Perform rigorous cross-reactivity testing to ensure no recognition of essential normal tissues

  • Consider dual-targeting strategies to minimize tumor escape through antigen loss

Antibody-based approaches:

  • Develop antibody-drug conjugates if surface expression can be demonstrated

  • Consider bispecific antibody formats to engage T cells with MAGEA8-expressing tumor cells

Challenges to address:

  • Heterogeneous expression within tumors may lead to treatment escape

  • Potential cross-reactivity with other MAGE family members

  • Limited accessibility of intracellular antigens for certain therapeutic modalities

When designing clinical trials, careful patient selection based on MAGEA8 expression levels is crucial for maximizing therapeutic benefit while monitoring for potential adverse events.

How do epigenetic mechanisms regulate MAGEA8 expression?

The expression of MAGEA8, like other cancer-testis antigens, is primarily regulated through epigenetic mechanisms, particularly DNA methylation:

DNA methylation:

  • In normal somatic tissues, the MAGEA8 promoter is heavily methylated, resulting in transcriptional silencing

  • During germ cell development and in cancer, hypomethylation of the promoter enables expression

  • Experimental approaches using DNA methyltransferase inhibitors (like 5-azacytidine) can induce MAGEA8 expression in cell lines, supporting this regulatory mechanism

Histone modifications:

  • Repressive histone marks (such as H3K9me3 and H3K27me3) are often present at the MAGEA8 locus in non-expressing tissues

  • Activating histone modifications (H3K4me3, H3K9ac) correlate with expression in testis and cancer tissues

  • Histone deacetylase inhibitors can sometimes induce expression, though typically less effectively than DNA demethylating agents

Other regulatory factors:

  • Certain transcription factors may preferentially bind to the unmethylated promoter

  • Long non-coding RNAs and chromatin organization may contribute to regulation

  • X chromosome inactivation mechanisms likely play a role given MAGEA8's location

For experimental studies of MAGEA8 epigenetic regulation, researchers should consider:

  • Chromatin immunoprecipitation (ChIP) assays to profile histone modifications

  • Bisulfite sequencing to analyze DNA methylation patterns at single-nucleotide resolution

  • CRISPR-based epigenetic editing to establish causality between specific epigenetic marks and expression

How is MAGEA8 evolutionarily conserved across species?

MAGEA8 exhibits interesting evolutionary patterns consistent with the broader MAGE gene family:

The MAGE gene family shows significant divergence across mammalian species, with type I MAGEs (including MAGEA8) undergoing species-specific expansion through duplication events . While the MAGE homology domain (MHD) is conserved, the type I MAGE genes have undergone positive selection that has allowed them to diversify or acquire additional functions .

Key evolutionary features of MAGEA8 and related genes include:

  • Type I MAGE genes are present in all mammals but show species-specific patterns of expansion

  • Human and mouse genomes contain different numbers of MAGE subfamily members, with some subfamilies being species-specific

  • Type I MAGE genes reside in syntenic regions on the X chromosome, where testis-expressed genes are overrepresented

  • The rapid expansion of MAGE genes on the X chromosome is thought to be driven by male X chromosome hemizygosity and benefits to male reproductive fitness

For comparative studies, researchers should note that while functional orthologs may exist between species, direct sequence orthologs may not always be present due to the rapid evolution of these genes. This presents challenges for traditional animal model studies but also offers opportunities to understand convergent evolution of reproductive stress protection mechanisms.

How does MAGEA8 functionally compare to other MAGE-A family members?

Within the MAGE-A subfamily, members share the conserved MAGE homology domain (MHD) but exhibit functional specialization:

Structural similarities and differences:

  • All MAGE-A proteins contain the MHD which mediates many protein-protein interactions

  • The N- and C-terminal regions flanking the MHD show greater divergence between paralogs

  • These divergent regions likely contribute to functional specialization

Expression patterns:

  • Most MAGE-A genes show restricted expression in testis and cancer

  • Some MAGE-A genes show broader expression during embryonic development, including in placenta and developing germ cells of both sexes

  • Expression levels and cancer type associations vary among family members

Functional aspects:

  • Several MAGE-A proteins interact with and modulate E3 ubiquitin ligases

  • Some family members directly affect p53 function and apoptotic pathways

  • MAGEA8-specific functions are less well-characterized than those of some other family members (like MAGEA3 or MAGEA11)

For experimental differentiation between MAGE-A family members, researchers should:

  • Use highly specific antibodies or detection methods that can distinguish between closely related family members

  • Consider potential functional redundancy when designing knockdown or knockout experiments

  • Perform rescue experiments with different family members to assess functional equivalence

What are the best methodologies for detecting MAGEA8 expression in clinical samples?

Accurate detection of MAGEA8 in clinical samples requires careful method selection based on the research or diagnostic objective:

RNA-based detection methods:

  • RT-qPCR remains the gold standard for sensitive and specific detection of MAGEA8 mRNA

    • Design primers spanning exon-exon junctions to avoid genomic DNA contamination

    • Include appropriate reference genes for normalization

    • Use testicular tissue as positive control

  • RNA-sequencing provides comprehensive expression data but requires appropriate bioinformatic analysis to distinguish MAGEA8 from other family members

  • RNA in situ hybridization can provide spatial information within tissue sections

Protein-based detection methods:

  • Immunohistochemistry (IHC) on formalin-fixed paraffin-embedded (FFPE) tissues

    • Validate antibodies rigorously for specificity against other MAGE-A family members

    • Use appropriate positive and negative control tissues

    • Implement standardized scoring systems for consistency

  • Western blotting for biochemical validation

    • Include recombinant MAGEA8 protein as positive control

    • Use denaturing conditions that can distinguish MAGEA8 by molecular weight

Considerations for clinical implementation:

  • Pre-analytical variables (fixation time, processing methods) can significantly impact detection sensitivity

  • Heterogeneous expression within tumors may require multiple sampling

  • Quantitative thresholds for "positive" expression should be established based on clinical correlation

  • Combine multiple detection methods for confirmatory testing in research settings

How can researchers effectively design experiments to study MAGEA8's role in cancer progression?

To investigate MAGEA8's specific contribution to cancer biology, researchers should consider the following experimental design strategies:

In vitro approaches:

  • Generate isogenic cell line models with CRISPR-Cas9 knockout or overexpression of MAGEA8

  • Perform comprehensive phenotypic characterization:

    • Proliferation, migration, and invasion assays

    • Resistance to apoptosis and therapy

    • Metabolic profiling

    • Three-dimensional organoid culture to better recapitulate tumor physiology

  • Use inducible expression systems to study dose-dependent and temporal effects

  • Compare effects in multiple cell lines representing different cancer types

In vivo approaches:

  • Xenograft models with MAGEA8-manipulated cell lines

  • Patient-derived xenografts with characterized MAGEA8 expression

  • Humanized mouse models for immunotherapy studies

  • Consider the challenge that mice have a different repertoire of Mage-a genes

Clinical correlation:

  • Analyze MAGEA8 expression in patient cohorts with detailed clinical annotation

  • Perform multi-parameter analysis including other biomarkers

  • Consider temporal changes through analysis of paired primary and metastatic samples

  • Correlate with treatment response data where available

Molecular mechanistic studies:

  • Identify MAGEA8-specific binding partners through techniques like BioID or IP-MS

  • Map signaling pathway interactions using phosphoproteomics

  • Investigate effects on gene expression through RNA-seq

  • Examine potential roles in protein stability through ubiquitination studies

When interpreting results, researchers should carefully distinguish MAGEA8-specific effects from broader MAGE family functions through appropriate controls and validation experiments.

What are the challenges in producing and working with recombinant MAGEA8 protein?

Working with recombinant MAGEA8 presents several technical challenges that researchers should anticipate and address:

Expression and purification challenges:

  • Recombinant MAGEA8 is typically produced in E. coli expression systems with an N-terminal His-tag to facilitate purification

  • The protein may form inclusion bodies requiring denaturing conditions followed by refolding

  • Purification typically employs chromatographic techniques, with specific buffers containing glycerol (10%) and DTT (1mM) to maintain stability

  • The final purified product should achieve >85% purity as determined by SDS-PAGE

Stability considerations:

  • MAGEA8 protein has limited stability in solution, requiring careful storage conditions

  • For short-term use (2-4 weeks), storage at 4°C is recommended

  • For longer periods, storage at -20°C with the addition of a carrier protein (0.1% HSA or BSA) helps maintain stability

  • Multiple freeze-thaw cycles should be avoided as they significantly decrease protein activity

Functional assays:

  • Verifying proper folding and biological activity can be challenging without established functional assays

  • Interaction studies with known binding partners can serve as quality control

  • Structural analysis by circular dichroism or limited proteolysis can verify protein integrity

Antibody specificity issues:

  • Commercial antibodies may cross-react with other MAGE-A family members due to sequence similarity

  • Validation using recombinant proteins and MAGEA8-knockout cells is essential

  • Consider epitope mapping to identify antibodies targeting unique regions

Researchers should include appropriate controls in all experiments using recombinant MAGEA8 and document the specific conditions and protocols to ensure reproducibility.

What bioinformatic approaches are most useful for analyzing MAGEA8 in genomic and proteomic datasets?

Effective analysis of MAGEA8 in large-scale datasets requires specialized bioinformatic approaches:

Genomic data analysis:

  • Sequence alignment tools must account for the high homology between MAGE family members

  • For RNA-seq analysis, use algorithms that handle multi-mapping reads appropriately

  • Consider the following parameters for accurate MAGEA8 quantification:

    • Stringent mapping quality thresholds

    • Unique molecular identifiers (UMIs) to reduce PCR amplification bias

    • Junction-spanning read requirements for improved specificity

Proteomic data analysis:

  • Implement peptide-level filtering to distinguish MAGEA8 from other MAGE proteins

  • Focus on unique peptides that differentiate MAGEA8 from paralogs

  • For post-translational modification analysis, consider:

    • Site localization probability scores

    • Validation of modifications through targeted approaches

    • Integration with phosphoproteomic datasets for pathway analysis

Structural bioinformatics:

  • Homology modeling based on solved structures of other MAGE proteins

  • Molecular dynamics simulations to predict functional domains

  • Protein-protein interaction modeling to predict binding interfaces

Single-cell analysis considerations:

  • Feature selection methods to identify MAGEA8 in heterogeneous samples

  • Trajectory analysis to understand expression dynamics in developmental contexts

  • Integration of epigenomic data (ATAC-seq, ChIP-seq) to identify regulatory mechanisms

These approaches should be complemented with experimental validation to confirm computational predictions and address the challenges of specificity when analyzing highly homologous gene families.

What are the most promising unexplored areas of MAGEA8 research?

Several high-potential research directions remain underexplored for MAGEA8:

Stress response mechanisms:

  • Further investigation into MAGEA8's specific role in protecting germline cells against environmental stressors

  • Comparative studies between MAGEA8 and other MAGE family members in stress response pathways

  • Elucidation of the molecular mechanisms underlying stress protection

  • Potential applications in fertility preservation technologies

Developmental biology:

  • Detailed characterization of MAGEA8 expression during human embryonic development

  • Investigation of its potential roles in gametogenesis in both sexes

  • Studies of MAGEA8 in embryonic stem cells and during cellular differentiation

  • Potential contributions to developmental stress resistance

Novel therapeutic approaches:

  • Development of MAGEA8-specific T cell receptors for adoptive cell therapy

  • Exploration of synthetic lethality approaches targeting MAGEA8-expressing cancers

  • Investigation of combination therapies that enhance immune recognition of MAGEA8

  • Development of proteolysis-targeting chimeras (PROTACs) targeting MAGEA8

Structural biology:

  • Determination of MAGEA8 crystal structure to facilitate drug design

  • Characterization of protein-protein interaction interfaces

  • Investigation of post-translational modification effects on structure and function

  • Comparative structural analysis across MAGE family members

Evolutionary biology:

  • Deeper investigation into the convergent evolution of MAGE genes across species

  • Understanding selective pressures that maintained and expanded the MAGE gene family

  • Comparative studies between human MAGEA8 and functional orthologs in other species

  • Analysis of MAGEA8 polymorphisms across human populations

Researchers entering these areas should consider interdisciplinary approaches and collaborations to address the complex biology of MAGEA8 and its related family members.

How might novel technologies advance our understanding of MAGEA8 function?

Emerging technologies offer exciting opportunities to address longstanding questions about MAGEA8:

CRISPR technologies:

  • Base editing or prime editing for precise modification of MAGEA8 regulatory regions

  • CRISPR activation/inhibition systems for temporal control of expression

  • CRISPR screens to identify synthetic lethal interactions in MAGEA8-expressing cells

  • In vivo CRISPR modeling of MAGEA8 functions in development

Single-cell multiomics:

  • Integrated single-cell RNA-seq and ATAC-seq to correlate MAGEA8 expression with chromatin accessibility

  • Spatial transcriptomics to map MAGEA8 expression in tissues with subcellular resolution

  • Single-cell proteomics to detect low-abundance MAGEA8 protein in heterogeneous samples

  • Lineage tracing to understand MAGEA8 expression dynamics during development

Protein interaction technologies:

  • Proximity labeling approaches (BioID, APEX) to map MAGEA8 interactome in living cells

  • Cross-linking mass spectrometry to capture transient interactions

  • Protein complementation assays for real-time visualization of interactions

  • Microfluidic antibody capture for high-throughput interaction screening

Structural biology advances:

  • Cryo-electron microscopy for structure determination of MAGEA8 complexes

  • Hydrogen-deuterium exchange mass spectrometry for dynamic structural information

  • AlphaFold and related AI approaches for structure prediction and interaction modeling

  • Time-resolved structural studies to capture conformational changes

Organoid and advanced cell culture models:

  • Testicular organoids to study MAGEA8 in its native cellular environment

  • Patient-derived tumor organoids for personalized functional studies

  • Microfluidic organs-on-chips to model tissue-specific functions

  • Bioprinted 3D models incorporating multiple cell types

These technologies, particularly when used in combination, promise to reveal new insights into MAGEA8 biology that have been challenging to address with conventional approaches.

Product Science Overview

Introduction

Melanoma Antigen Family A, 8 (MAGEA8) is a member of the MAGEA gene family, which is part of the larger MAGE (Melanoma Antigen) family. These genes are known for their role in cancer biology, particularly in the context of cancer/testis (CT) antigens. MAGEA8, like other MAGEA genes, is primarily expressed in germline cells and various types of cancer cells, making it a significant target for cancer immunotherapy.

Gene and Protein Structure

The MAGEA8 gene is located on the X chromosome at the Xq28 band . The gene encodes a protein that shares 50 to 80% sequence identity with other members of the MAGEA family . This high degree of similarity suggests that these proteins may have conserved functions, although the variability in their promoters and first exons indicates that they can be regulated differently depending on the cellular context .

Expression and Function

MAGEA8 is expressed in a restricted manner, primarily in immune-privileged tissues such as the placenta and various cancers . This restricted expression pattern makes MAGEA8 an attractive target for cancer immunotherapy, as it minimizes the risk of off-target effects on normal tissues. The protein encoded by MAGEA8 is involved in various cellular processes, including cell cycle regulation and apoptosis, which are critical for cancer cell survival and proliferation .

Role in Cancer

MAGEA8, along with other MAGEA proteins, is associated with resistance to chemotherapy, metastasis, and increased survival potential of cancer cells . These characteristics make MAGEA8 a valuable target for designing specific and effective cancer immunotherapies. Strategies such as cancer vaccination and adoptive T-cell transfer are being explored to target MAGEA8 and other MAGEA proteins .

Clinical Implications

The immune-privileged nature and oncogenic activity of MAGEA8 make it a universal antigen capable of eliciting highly specific immune responses against cancer cells . Ongoing clinical trials and research are focused on developing therapies that target MAGEA8, with the aim of improving treatment outcomes for patients with various types of cancer .

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.