MFGE8 Antibody

Shipped with Ice Packs
In Stock

Description

Cancer Research

MFGE8 Antibodies inhibit tumor progression by blocking MFGE8’s pro-survival and pro-angiogenic effects. In triple-negative breast cancer (TNBC) and ovarian carcinoma models, antibodies reduce adhesion (IC50: 0.625–5 µg/mL), migration, and survival of cancer cells . For example:

  • Adhesion Inhibition: Anti-MFGE8 antibodies (e.g., hMc3) block integrin-mediated adhesion to MFGE8-coated surfaces .

  • Survival Suppression: Neutralizing MFGE8 or its receptors (αvβ3/αvβ5) abolishes survival signals in SKOV-3 ovarian cancer cells .

  • Synergy with Chemotherapy: MFGE8 blockade enhances cisplatin efficacy in esophageal squamous cell carcinoma (ESCC) .

Cancer TypeKey FindingAntibodySource
Triple-negative breast82% of TNBC cell lines express MFGE8; antibodies reduce migration/survival 399A12
Ovarian carcinomaAnti-MFGE8 antibodies inhibit tumor growth in xenograft models hMc3
ESCCMFGE8 blockade reverses chemoresistance and angiogenesis Neutralizing

Autoimmune and Inflammatory Diseases

MFGE8 deficiency leads to impaired apoptotic cell clearance, causing inflammation and autoimmunity. Antibodies are used to study this mechanism:

  • Autoimmune Biomarker: MFG-E8-/- mice develop glomerulonephritis due to defective B-cell apoptosis clearance .

  • Inflammatory Bowel Disease: MFGE8 maintains intestinal epithelial homeostasis; its absence exacerbates mucosal damage .

Diagnostic Biomarker

Serum MFGE8 levels correlate with hepatocellular carcinoma (HCC) progression and prognosis:

Clinical ApplicationBiomarker PerformanceAntibody/AssaySource
HCC DiagnosisAUC 0.923 (MFGE8 + DCP model)ELISA
HCC PrognosisLow MFGE8 → Poor survival post-resectionELISA

Key Antibodies and Their Features

AntibodyTarget SpeciesApplicationsEpitopeSource
hMc3HumanAdhesion/migration assaysRGD-containing EGF
MAB27671HumanFlow cytometry, WBFull-length protein
AF2805MouseWB, IHCMouse MFGE8 isoforms
4F6MouseMammary gland involutionC-terminal region

Optimized Protocols

TechniqueConditionsOutcome
Western Blot1:500–1:1000 dilution; Jurkat cells/human placenta lysates Detects 43–51 kDa MFGE8 isoforms
Immunohistochemistry1:50–1:500 dilution; pH 9.0 antigen retrieval for skin cancer tissue Stains epithelial/tumor cells
Flow CytometryPermeabilized cells (e.g., dendritic cells); PE-conjugated secondary Intracellular MFGE8 detection

Cancer Therapy

  • Mechanism: Anti-MFGE8 antibodies disrupt tumor cell survival (via αvβ3/αvβ5) and angiogenesis (via VEGF potentiation) .

  • Preclinical Success: hMc3 + cisplatin synergizes to control ESCC growth in mice .

Autoimmune Disease Therapies

  • Target: Enhancing apoptotic cell clearance to reduce inflammation .

  • Limitations: Off-target effects on vascular integrity (e.g., aortic amyloid deposits linked to MFGE8 fragments) .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timeframes.
Synonyms
BA46 antibody; Breast epithelial antigen BA46 antibody; EDIL1 antibody; HMFG antibody; hP47 antibody; HsT19888 antibody; Lactadherin antibody; Medin antibody; MFG-E8 antibody; MFGE8 antibody; MFGM antibody; MFGM_HUMAN antibody; Milk fat globule EGF factor 8 antibody; Milk fat globule EGF factor 8 protein antibody; Milk fat globule-EGF factor 8 antibody; O acetyl disialoganglioside synthase antibody; OAcGD3S antibody; SED1 antibody; SPAG10 antibody; Sperm associated antigen 10 antibody; Sperm surface protein hP47 antibody
Target Names
Uniprot No.

Target Background

Function
MFG-E8 (Milk Fat Globule-Epidermal Growth Factor 8) plays a pivotal role in maintaining intestinal epithelial homeostasis and promoting mucosal healing. It facilitates VEGF-dependent neovascularization and contributes to the phagocytic removal of apoptotic cells in various tissues. MFG-E8 is a specific ligand for the alpha-v/beta-3 and alpha-v/beta-5 receptors. It also binds to phosphatidylserine-enriched cell surfaces in a receptor-independent manner. MFG-E8 is a zona pellucida-binding protein that may play a role in gamete interaction. Additionally, it is a major constituent of aortic medial amyloid.
Gene References Into Functions
  1. In esophageal cancer patients, high MFG-E8 expression has been associated with poorer overall survival and relapse-free survival, particularly in patients who received preoperative chemotherapy. The high levels of MFG-E8 in esophageal squamous cell carcinoma may negatively impact long-term survival after chemotherapy by influencing T-cell regulation within the tumor microenvironment. PMID: 30156356
  2. Studies in Chinese adults have revealed that MFG-E8 polymorphisms are not associated with metabolic syndrome but are linked to variations in diastolic blood pressure, HDL-C levels, and waist circumference. PMID: 30053459
  3. Research has shown that MFG-E8 expression is impaired in the endometrium of women with endometriosis and infertility during the window of implantation. PMID: 28967712
  4. MFG-E8 and TGFbeta1 have been identified as age-related inflammatory factors, linked to the severity of atherosclerosis. These factors may serve as potential markers for the severity of atherosclerosis. PMID: 29152651
  5. Elevated serum MFG-E8 levels may be associated with cerebrovascular diseases or neuropsychiatric manifestations of systemic lupus erythematosus. PMID: 28266034
  6. Circulating MFG-E8 levels exhibit significant increases during pregnancy and are substantially higher in women with gestational diabetes mellitus compared to those with normal pregnancies. PMID: 28035763
  7. MFG-E8 has emerged as a potential prognostic marker for colorectal cancer. Overexpression of MFG-E8 may be involved in lymph node metastasis and angiogenesis in colorectal cancer. PMID: 28923329
  8. Elevated MFG-E8 levels in individuals with type 2 diabetes (T2D) are suppressed by increased adipose tissue, potentially contributing to higher levels of inflammatory factors. MFG-E8 may serve as a potential biomarker for obesity and T2D in clinical settings. PMID: 28067023
  9. Studies suggest that MFG-E8 plays a role in colorectal cancer progression and may serve as a predictive factor for metastasis and prognosis. PMID: 28653875
  10. Levels of MFG-E8 are reduced in cirrhotic liver tissue compared to healthy controls. PMID: 27956229
  11. An efficient purification method for producing non-aggregated, full-length MFG-E8 has been developed. PMID: 27102803
  12. Lower serum levels of MFG-E8 have been associated with an increased risk of microvascular complications in patients with type 2 diabetes. PMID: 28089751
  13. MFG-E8 has been identified as a protective factor in the pathogenesis of rheumatoid arthritis and subsequent bone loss. PMID: 26391522
  14. Following myocardial infarction, macrophages expressing Mfge8 (and Mertk) work synergistically to clear injured cardiomyocytes. PMID: 26819373
  15. Research using mouse models of inflammatory bowel disease (IBD) has shown that treatment with recombinant human MFG-E8 (rhMFG-E8) significantly attenuates colitis in a dose-dependent manner. PMID: 25751740
  16. MFG-E8 promotes cutaneous wound healing by enhancing angiogenesis. PMID: 24838098
  17. Lactadherin may reduce inflammation by inhibiting secretory phospholipase A2 activity on pre-apoptotic leukemia cells. PMID: 24194865
  18. MFG-E8 has been implicated in promoting tumor progression in oral squamous cell carcinoma. PMID: 25264705
  19. Serum MFG-E8 levels have been negatively associated with high-sensitivity C-reactive protein (hs-CRP) and positively associated with LDL-c. Additionally, serum MFG-E8 levels are negatively correlated with the severity of coronary artery stenosis and the risk of clinical events. PMID: 24561551
  20. Medin, a protein related to MFG-E8, adopts a predominantly beta-sheet conformation with some unstructured elements. PMID: 24602872
  21. MFG-E8 may serve as a biomarker for the diagnosis and monitoring of disease activity in certain patients with systemic lupus erythematosus. PMID: 24554711
  22. Exogenous MFG-E8 inhibits receptor activator NF-kappaB ligand-induced osteoclastogenesis of human osteoclast precursors. PMID: 24958900
  23. Research strongly suggests that MFG-E8 is a promising biomarker for the diagnosis, prognosis, and therapy target of opisthorchiasis-associated cholangiocarcinoma. PMID: 24122204
  24. Blocking MFG-E8 in endometrial tumor cells diminishes trophoblast cell attachment. PMID: 24424369
  25. TNF-alpha up-regulates endometrial epithelial cell migration and MFG-E8 production. PMID: 24262600
  26. MFG-E8-dependent promotion of apoptotic cell clearance is a novel anti-inflammatory mechanism associated with glucocorticoid treatment. PMID: 23832117
  27. Studies indicate that MFGE8 is unlikely involved in the phagocytic clearance of neuronal debris associated with nigrostriatal pathway injury. PMID: 23194669
  28. The NMR solution structure of the C2 domain of MFG-E8 provides insights into its molecular recognition with phosphatidylserine. PMID: 23262193
  29. Milk fat globule-epidermal growth factor 8 (MFG-E8) possesses proapoptotic activity, suggesting a role in endometrial remodeling through an epithelial-stromal cell paracrine effect. PMID: 22921913
  30. MFG-E8 expression in the endometrial epithelium and chorionic villi suggests a potential role in endometrial reorganization during the receptive phase and in events related to normal pregnancy in mammals. PMID: 22770563
  31. Decreased colonic MFGE8 expression in patients with ulcerative colitis may be linked to mucosal inflammatory activity and clinical disease activity through basal cell apoptosis and the prevention of tissue healing in the pathogenesis of ulcerative colitis. PMID: 22204000
  32. MFG-E8 release from apoptotic endothelial cells plays a key role in macrophage reprogramming. PMID: 22558449
  33. The distribution of specific single nucleotide polymorphisms (SNPs) in the MFGE8 gene (rs4945 and rs1878326) was analyzed in patients with "wet" age-related macular degeneration and their age-matched controls. PMID: 22438901
  34. Serum lactadherin levels have been correlated with poor blood glucose control and diabetic vascular complications. PMID: 22018779
  35. In vitro studies have demonstrated that medin amyloid-like fibrils promote the aggregation of protein amyloid A into fibrils. PMID: 22070546
  36. Prolactin has a modulatory role as a stromal/epithelial paracrine factor controlling MFG-E8. This study provides the first report on MFG-E8 protein localization to the human endometrial epithelium and its up-regulation during the window of implantation. PMID: 21177637
  37. MFG-E8 is expressed in triple-negative breast cancers as a target gene of the p63 pathway, but may have a suppressive function in ER(+) and erbB2(+) breast cancers. PMID: 21127199
  38. Overexpression of MFGE8 during bladder tumor development is correlated with the expression of genes involved in cell adhesion or migration and in immune responses. PMID: 20956946
  39. An intronic mutation in the human MFG-E8 gene has been linked to the development of systemic lupus erythematosus (SLE). PMID: 20213738
  40. SED1, a protein related to MFG-E8, is expressed on the surface of acrosome-intact human sperm and in the anterior caput of the human epididymis, similar to observations in mice. PMID: 18990388
  41. A truncated fragment of medin, the hexapeptide NFGSVQ, can form typical amyloid fibrils. PMID: 15478463
  42. MFG-E8 may hold potential for the treatment of prolonged ischemia. PMID: 16115445
  43. Lactadherin (MFG-E8) is expressed in both normal and atherosclerotic human arteries. PMID: 17420351
  44. The trans-activator (TA) isoforms of p63 activate MFGE8 transcription through a p53/p63 motif at -370, as confirmed by a chromatin immunoprecipitation experiment. PMID: 17637751
  45. The last 18-19 amino acid residues have been identified as constituting the amyloid-promoting region of medin. PMID: 17679143
  46. Aggregated medin has been shown to induce death of aortic smooth muscle cells in vitro. Cells incubated with medin exhibit increased production of matrix metalloproteinase-2, a protease that degrades elastin and collagen, potentially weakening the vessel wall. PMID: 17906662
  47. Analysis of membrane-interactive loops of Lact-C2 (a domain of MFG-E8) has been conducted. PMID: 18160406
  48. Some individuals with childhood-onset and adult systemic lupus erythematosus (SLE) have been found to carry significant levels of MFG-E8 in their blood samples. PMID: 18303131
  49. Lactadherin (MFG-E8) promotes the phagocytosis of phosphatidylserine-expressing red blood cells (RBCs) by macrophages in a concentration-dependent manner. PMID: 18647368

Show More

Hide All

Database Links

HGNC: 7036

OMIM: 602281

KEGG: hsa:4240

STRING: 9606.ENSP00000268150

UniGene: Hs.3745

Subcellular Location
Membrane; Peripheral membrane protein. Secreted. Cytoplasmic vesicle, secretory vesicle, acrosome membrane; Peripheral membrane protein.
Tissue Specificity
Mammary epithelial cell surfaces and aortic media. Overexpressed in several carcinomas.

Q&A

What is MFGE8 and what are its primary biological functions?

MFGE8 (Milk Fat Globule-EGF Factor 8) is a secreted glycoprotein that mediates cell-cell interactions through binding to αvβ3 and αvβ5 integrins. It plays critical roles in diverse biological processes including apoptotic cell clearance, angiogenesis, and tissue homeostasis. In cancer biology, MFGE8 has been shown to promote tumor cell survival, invasion, and angiogenesis while contributing to local immune suppression . MFGE8 serves as a bridging molecule between phosphatidylserine on apoptotic cells and integrins on phagocytes, facilitating efferocytosis. This protein is expressed at high levels in diverse tumor types, making it a potential target for cancer therapy .

What types of MFGE8 antibodies are available for research applications?

Several types of MFGE8 antibodies are available for research, including monoclonal and polyclonal antibodies with various specificities. Mouse Anti-Human MFGE8 Monoclonal Antibody (Clone # 278901) is one example designed to detect human MFGE8 . Other antibodies include goat anti-MFGE8 antibodies that have been used in immunodepletion experiments . Multiple antibody clones with different blocking efficiencies have been identified through screening processes, including hMc3, 416H9, and 399A12, which demonstrated high efficacy in inhibiting MFGE8-mediated cellular functions . The selection of an appropriate antibody depends on the specific application and target species of interest.

How is MFGE8 expression detected and quantified in experimental systems?

MFGE8 expression can be detected through multiple complementary techniques. At the mRNA level, RNAScope and RT-qPCR from FACS-sorted cells provide sensitive methods for quantifying expression in specific cell populations . For protein detection, immunohistochemistry (IHC) allows visualization of MFGE8 in tissue sections, while Western blotting enables quantification in tissue or cell lysates . ELISA assays, such as the MFG-E8 quantikine ELISA kit, provide precise quantification of MFGE8 in conditioned media or biological fluids . For cellular localization studies, immunofluorescence microscopy can determine the spatial distribution of MFGE8 within tissues. Each technique offers different advantages in sensitivity, specificity, and contextual information about MFGE8 expression patterns.

How does MFGE8 contribute to tumor progression?

MFGE8 promotes cancer progression through coordinated αvβ3 integrin signaling in both tumor and host cells . It enhances tumor cell survival by activating PI3K/AKT signaling pathways following binding to integrin αvβ3/αvβ5 . This interaction has been confirmed through co-immunoprecipitation experiments demonstrating physical binding between MFGE8 and integrin αV, β3, and β5 subunits . MFGE8 also facilitates tumor cell invasion and migration, as demonstrated in real-time cell analysis systems measuring adhesion and transwell migration . Additionally, MFGE8 contributes to angiogenesis, supporting tumor growth through enhanced blood vessel formation. In the tumor microenvironment, MFGE8 mediates immune suppression, creating a permissive environment for cancer progression by interfering with anti-tumor immune responses .

What experimental models best demonstrate the effects of anti-MFGE8 antibody treatment in cancer?

Several experimental models have demonstrated the efficacy of anti-MFGE8 antibody treatments. In vitro models include adhesion assays using the xCELLigence system to measure real-time cell binding to MFGE8, transwell migration assays to assess MFGE8-directed cell migration, and survival assays under starvation conditions . These complementary approaches allow researchers to evaluate how MFGE8 blockade affects multiple aspects of tumor cell behavior. In vivo models have shown that systemic administration of anti-MFGE8 antibodies cooperates with conventional cancer therapies to control established mouse tumors . Specific examples include ovarian cancer (SKOV-3) and triple-negative breast cancer models, where MFGE8-blocking antibodies impeded adhesion, migration, and survival of cancer cells . The combination of in vitro and in vivo models provides comprehensive insights into the mechanism and therapeutic potential of MFGE8 blockade.

How do anti-MFGE8 antibodies synergize with conventional cancer therapies?

Anti-MFGE8 antibodies demonstrate significant synergy with conventional cancer therapies through multiple mechanisms. When combined with cytotoxic chemotherapy, molecularly targeted therapy, or radiation therapy, MFGE8 blockade enhances tumor cell death and improves treatment outcomes . This synergistic effect occurs through both cell-autonomous and immune-mediated pathways. In cell-autonomous pathways, MFGE8 blockade sensitizes tumor cells to cytotoxic agents by compromising cell viability, as evidenced by increased caspase activation . For immune-mediated effects, anti-MFGE8 antibodies enhance dendritic cell cross-presentation of dying tumor cells to T cells, promoting the generation of potent antitumor effector T cells while inhibiting regulatory T cells (FoxP3+ Tregs) . Experimental data showed that cisplatin combined with MFGE8 antibody treatment significantly reduced tumor volume and mass compared to either treatment alone . This dual-targeting approach—affecting both tumor cells and host immune responses—represents a promising strategy for improving cancer treatment efficacy.

What are the optimal methods for validating MFGE8 antibody specificity?

Validating MFGE8 antibody specificity requires a multi-modal approach combining several complementary techniques. Western blotting using lysates from tissues known to express MFGE8 (such as human milk) provides a primary validation method, with the antibody detecting a specific band at approximately 45 kDa under reducing conditions . For more sensitive detection, Simple Western techniques can identify MFGE8 at approximately 57-58 kDa . Immunoprecipitation followed by mass spectrometry can confirm target interaction specificity. Testing antibody reactivity in MFGE8 knockout samples serves as a gold standard negative control, as demonstrated in studies using MFGE8 KO astrocyte conditioned media compared to wild-type . Cross-reactivity testing against proteins with similar structural domains (like vitronectin and Factor VIII) ensures specificity to MFGE8 rather than shared domains . Finally, immunodepletion experiments can validate antibody functionality by demonstrating effective removal of MFGE8 from biological samples.

What techniques are most effective for studying MFGE8-integrin interactions?

Studying MFGE8-integrin interactions requires specialized techniques that capture both physical binding and functional consequences. Co-immunoprecipitation (CO-IP) experiments provide direct evidence of physical interactions between MFGE8 and integrin αV, β3, and β5 subunits in both cancer cells and endothelial cells (HUVECs) . Immunofluorescence microscopy combined with binding inhibition using neutralizing antibodies against either MFGE8 or integrin αVβ3/αVβ5 can visualize and confirm these interactions in cellular contexts . For functional studies, the xCELLigence system enables real-time measurement of cellular adhesion to MFGE8-coated surfaces, quantifying the strength of integrin-mediated binding . Transwell migration assays using electrode-coated Boyden chambers assess how MFGE8-integrin interactions influence directed cell movement . Signal transduction analysis through Western blotting for phosphorylated downstream effectors (such as PI3K/AKT) provides insights into the molecular consequences of these interactions . This comprehensive approach reveals both the physical nature and functional significance of MFGE8-integrin binding.

How can researchers effectively screen for MFGE8-blocking antibodies?

Screening for effective MFGE8-blocking antibodies requires a systematic approach using complementary functional assays. A successful screening strategy involves initial binding assays to confirm antibody specificity for human MFGE8 while excluding cross-reactivity with related proteins containing common structural domains (like vitronectin and Factor VIII) . Following this initial selection, candidates can be evaluated in a short-term adhesion assay using the xCELLigence system, which measures real-time cell binding to MFGE8-coated surfaces . Promising candidates from this step should be tested in a transwell migration assay to assess their ability to block MFGE8-induced cell migration . Finally, cell survival assays under starvation conditions can determine whether the antibodies inhibit MFGE8's pro-survival effects . This sequential screening approach identified antibodies like 416H9 and 399A12, which demonstrated comparable efficacy to the reference antibody hMc3 in blocking all three MFGE8-mediated functions . The combined use of these complementary assays allows for robust identification of the most effective blocking antibodies from a large candidate pool.

What are the challenges in translating MFGE8 antibody research from preclinical to clinical applications?

Translating MFGE8 antibody research from preclinical models to clinical applications faces several significant challenges. Firstly, species-specific differences in MFGE8 structure and function may limit direct extrapolation from animal models to humans, necessitating humanized antibody development. Secondly, MFGE8 serves important physiological functions in normal tissues, potentially leading to on-target, off-tumor effects that must be carefully evaluated during development. The complex role of MFGE8 in both cell-autonomous and immune-mediated pathways requires sophisticated biomarkers to monitor treatment efficacy across multiple biological processes. Patient stratification presents another challenge, as determining which tumor types and patient subgroups would benefit most from anti-MFGE8 therapy requires extensive biomarker development. Finally, optimizing combination treatment regimens with existing therapies necessitates careful design of clinical trials to identify synergistic rather than antagonistic effects. Addressing these challenges requires coordinated preclinical studies that examine safety profiles, pharmacokinetics, and efficacy in models that closely recapitulate human disease characteristics.

What role does MFGE8 play in neurological processes and disorders?

MFGE8 serves important functions in neurological processes, particularly in microglial synapse elimination. Research has demonstrated that astrocyte-derived MFGE8 facilitates microglial-mediated synapse removal, a process crucial for normal brain development and potentially dysregulated in neurodegenerative conditions . In models of Alzheimer's disease utilizing NL-F KI mice, significantly higher MFGE8-immunoreactivity was observed in the local milieu of bulbous astrocytes compared to bushy astrocytes . Both mRNA and protein levels of MFGE8 were upregulated in the hippocampus of these mice, with increased Mfge8 mRNA expression in astrocytes despite no change in astrocyte numbers . This suggests that enhanced MFGE8 expression coincides with periods when synapses are vulnerable to loss. The mechanistic role of MFGE8 in synaptic pruning represents a potentially targetable pathway in neurodegenerative disorders characterized by aberrant synapse elimination. Researchers investigating neurological applications of MFGE8 antibodies should consider these specific neural functions when designing intervention strategies for neurological disorders.

How do experimental approaches differ when studying MFGE8 in neuroscience versus cancer research?

Experimental approaches for studying MFGE8 differ significantly between neuroscience and cancer research contexts. In neuroscience, studies often focus on cell-cell interactions between astrocytes, microglia, and neurons using co-culture systems or conditioned media approaches . Techniques like RNAScope for cell-specific mRNA detection and fluorescence-activated cell sorting (FACS) of brain cell populations allow precise characterization of MFGE8 expression in neural cell types . Functional readouts in neuroscience typically include synapse density measurements, microglial engulfment assays, and electrophysiological recordings to assess synaptic function. In contrast, cancer research emphasizes tumor cell-autonomous functions and stromal interactions, utilizing adhesion, migration, and survival assays . Cancer studies frequently employ xenograft models and assess tumor volume, metastasis, and immune infiltration as key endpoints . While both fields use antibody-based approaches to block MFGE8 function, the concentration ranges, delivery methods, and timing of intervention may differ substantially based on the biological context. Understanding these methodological differences is crucial for researchers transitioning between these fields or attempting to integrate findings across disciplines.

What are the most promising future applications of MFGE8 antibodies in precision medicine?

The most promising applications of MFGE8 antibodies in precision medicine center on combination therapies and targeted approaches for specific cancer subtypes. For ovarian carcinoma and triple-negative breast carcinoma, MFGE8-blocking antibodies show particular promise as these cancer types express high levels of both MFGE8 and its integrin receptors . Future applications will likely involve combination regimens where anti-MFGE8 antibodies enhance the efficacy of conventional treatments, as demonstrated by the synergistic effects when combined with cisplatin . Patient stratification based on MFGE8 expression levels and integrin receptor status will be crucial for identifying those most likely to benefit from anti-MFGE8 therapy. Beyond cancer, emerging applications in neurodegenerative disorders may target the role of MFGE8 in aberrant microglial synapse elimination . The dual-targeting capability of MFGE8 antibodies—affecting both cell-autonomous processes and host-mediated pathways—positions them uniquely in the precision medicine landscape. Development of companion diagnostics to measure MFGE8 levels in patient samples will be essential for clinical implementation of these promising therapeutic approaches.

What new technological advances might improve MFGE8 antibody development and application?

Several technological advances promise to revolutionize MFGE8 antibody development and application. Single-cell RNA sequencing technologies will provide unprecedented insights into cell-specific MFGE8 expression patterns within complex tissues, enabling more precise targeting strategies. Advanced antibody engineering techniques, including bispecific antibodies that simultaneously target MFGE8 and complementary pathways, may enhance therapeutic efficacy. The development of antibody-drug conjugates (ADCs) using anti-MFGE8 antibodies could deliver cytotoxic payloads specifically to MFGE8-expressing cells while sparing normal tissues. Improved imaging technologies utilizing radiolabeled or fluorescently labeled anti-MFGE8 antibodies could enable both diagnostic applications and monitoring of treatment response. CRISPR-based high-throughput screening for MFGE8 pathway components will identify additional targets for combination therapy. Finally, artificial intelligence and machine learning approaches applied to large datasets of MFGE8 expression and patient outcomes will refine patient selection criteria for clinical applications. These technological advances will collectively accelerate the translation of basic MFGE8 research into clinical applications with improved efficacy and reduced side effects.

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.