LGALS3BP Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Profile

LGALS3BP antibody binds to Galectin-3-binding protein (LGALS3BP), a secreted protein encoded by the LGALS3BP gene on chromosome 17q25. The protein interacts with integrins (α1β1, α5β1, αvβ1, α6β1) and galectins, influencing cellular adhesion, immune response, and tumor microenvironment (TME) dynamics . Elevated LGALS3BP levels correlate with poor prognosis in cancers and HIV infection .

Mechanism of Action

LGALS3BP antibodies interfere with oncogenic signaling by blocking integrin-mediated pathways. Key mechanisms include:

Target PathwayEffect of Antibody InterventionOutcome
Integrin α5β1 bindingInhibits Akt/JNK/Ras-ERK signalingReduces survival and proliferation
Galectin-3 interactionDisrupts TME crosstalkImpedes angiogenesis and metastasis
Extracellular vesicle (EV) bindingBlocks EV-mediated communicationLimits tumor progression

For example, the SP2 antibody inhibits LGALS3BP-driven integrin activation, suppressing motility and migration without compromising cell viability .

Therapeutic Applications

LGALS3BP antibodies are leveraged in antibody-drug conjugates (ADCs) for precision oncology:

Key ADC Candidates:

  • 1959-sss/DM3: Targets LGALS3BP on neuroblastoma-derived EVs, achieving an 80% reduction in lung metastases in murine models .

  • DM4-conjugated ADC: Demonstrates potent cytotoxicity in oral squamous cell carcinoma (OSCC) with an IC~50~ of 0.3 nM .

Efficacy Data:

Cancer TypeModelResponse
NeuroblastomaOrthotopic PDX modelsTumor eradication in 60% of cases
OSCCHOC621/CAL27 cell lines95% growth inhibition at 1 nM DM4

Diagnostic and Prognostic Utility

LGALS3BP antibodies aid in biomarker detection:

  • OSCC Diagnosis: Cytoplasmic LGALS3BP overexpression correlates with aggressive histology (p < 0.01) .

  • Serum Monitoring: Elevated serum LGALS3BP levels predict metastatic relapse in breast and lung cancers .

Research Advancements

Recent studies highlight:

  • EV Targeting: LGALS3BP antibodies selectively bind cancer-derived EVs, enabling TME-specific drug delivery .

  • Immune Modulation: Antibody intervention enhances NK/LAK cell cytotoxicity by disrupting LGALS3BP-Mac-2 interactions .

Clinical Challenges

  • Heterogeneous Expression: LGALS3BP levels vary across cancer subtypes, requiring patient stratification .

  • Off-Target Effects: Limited data exist on antibody safety in non-malignant tissues expressing low LGALS3BP .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on your location and purchasing method. For specific delivery times, please contact your local distributor.
Synonyms
90K antibody; 90K MAC 2 BP antibody; Basement membrane autoantigen p105 antibody; BTBD17B antibody; Galectin 3 binding protein antibody; Galectin-3-binding protein antibody; L3 antigen antibody; Lectin galactoside binding soluble 3 binding protein antibody; Lectin galactoside-binding soluble 3-binding protein antibody; LG3BP_HUMAN antibody; LGALS3BP antibody; M2BP antibody; Mac 2 binding protein antibody; Mac 2 BP antibody; Mac-2 BP antibody; Mac-2-binding protein antibody; Mac2 BP antibody; MAC2BP antibody; Serum protein 90K antibody; TANGO10B antibody; Transport and golgi organization 10 homolog B antibody; Tumor associated antigen 90K antibody; Tumor-associated antigen 90K antibody
Target Names
LGALS3BP
Uniprot No.

Target Background

Function
LGALS3BP (Galectin-3 Binding Protein) promotes integrin-mediated cell adhesion and may contribute to host defense mechanisms against viruses and tumor cells.
Gene References Into Functions
  1. Serum M2BP (Mac-2 binding protein) might be involved in the inflammatory processes associated with systemic lupus erythematosus. PMID: 29233037
  2. M2BP inhibits both HIV-1 Env processing and virion production. M2BP interacts with HIV-1 Gag and directs it to vimentin filaments, preventing Gag from reaching the plasma membrane and inhibiting virion production. PMID: 27604950
  3. Studies have demonstrated that 90K (Mac-2 binding protein) destabilizes E-cadherin, impacting cell adhesion and invasion in subconfluent cancer cells by disrupting the E-cadherin-p120-catenin complex. This suggests a role for 90K in promoting the early stages of cancer metastasis. PMID: 29207493
  4. Low LGALS3BP expression has been associated with HIV infections. PMID: 29743357
  5. Research indicates that serum levels of RBP4 (retinol binding protein 4) and LGAL3BP are elevated after menopause, particularly when complicated by NAFLD (non-alcoholic fatty liver disease). These proteins may serve as potential biomarkers for NAFLD in postmenopausal women. PMID: 29679552
  6. Elevated plasma M2BP levels have been linked to unstable plaque and are independently associated with poor cardiovascular outcomes in patients experiencing acute coronary syndromes. PMID: 28731888
  7. Serum Mac-2-binding protein expression has been shown to predict disease severity in chronic hepatitis C patients. PMID: 28811008
  8. A higher pre-treatment WFA+-M2BP level was associated with an increased risk of HCC (hepatocellular carcinoma) development in patients with undetectable HBV DNA under NA therapy. PMID: 28537900
  9. Targeting the LGALS3BP-mediated specific survival signaling pathway in resistant cells could be a novel therapeutic strategy for cancer treatment. This has been demonstrated in studies where the combination of 17-AAG and PI3K/Akt inhibitor effectively suppressed acquired resistance to 17-AAG. PMID: 28336809
  10. Elevated serum M2BP levels may reflect silent atherosclerosis in individuals seemingly healthy. PMID: 27344370
  11. The serum M2BP-adiponectin complex is elevated in men with coronary artery disease. PMID: 27588936
  12. WFA+-M2BP from hepatic stellate cells induces Mac-2 expression in Kupffer cells, which in turn activates hepatic stellate cells, leading to fibrosis. PMID: 28008658
  13. Mac-2BP may regulate the extracellular spreading and storage of Wnt proteins, thereby modulating their bioavailability and stability. PMID: 28756229
  14. Research has highlighted the role of 90K (Mac-2 binding protein) in promoting the degradation of mutant beta-catenin lacking phosphorylation sites in the N-terminus. PMID: 27668402
  15. Serum M2BP may serve as a valuable biomarker for the diagnosis of pancreatic ductal adenocarcinoma and the prediction of disease progression. This is due to its potential reflection of altered pro-oncologic glycosylation enzymes. PMID: 27665173
  16. Studies analyzing sLex expression alongside two glycoproteins (BST-2 and LGALS3BP) identified a subgroup of patients with ER-negative tumors exhibiting higher risks of liver and brain metastasis and a significantly reduced survival rate. PMID: 27176937
  17. Research suggests that galectin-9 and galectin-3BP are induced in dengue patients during the acute febrile phase compared to healthy controls. This indicates their potential role as significant inflammatory mediators in acute dengue virus infection. PMID: 27240351
  18. 90K/Mac-2BP has been correlated with the size of colorectal cancer. PMID: 26448934
  19. Evidence suggests that CALU and LGALS3BP may act as potential negative regulators in the virus-triggered induction of type I interferons. PMID: 26124285
  20. Low expression of LGALS3BP has been linked to the malignant progression of colorectal cancer. PMID: 26219351
  21. Post-Tx WFA+-M2BP levels greater than 2.0 COI (cut-off index) are associated with the risk of HCC development among patients who have achieved SVR (sustained virological response). These values may serve as a novel predictor for HCC after SVR. PMID: 26070204
  22. Serum Mac-2 binding protein levels have emerged as a potential diagnostic biomarker for predicting disease severity and the presence of nonalcoholic steatohepatitis. PMID: 23775887
  23. Studies have helped elucidate the molecular mechanism behind the tumor-suppressive effect achieved through the down-regulation of LGALS3BP by miR-596 in Oral Squamous Cell Carcinoma. PMID: 26502662
  24. Research has examined galectin-3 binding protein levels in milk from HIV-infected Zambian mothers. Higher levels were observed in mothers who transmitted HIV through breastfeeding. PMID: 23899964
  25. LGALS3BP-mediated integrin activation leads to signal transmission via Akt, JNK, and the Ras cascade through the Raf-ERK axis while maintaining p38 activity at baseline levels. PMID: 24362527
  26. Gal-3BP levels in patients with hemorrhagic fever with renal syndrome correlated with increased complement activation and clinical variables reflecting the severity of acute hantavirus infection. PMID: 25013204
  27. Research suggests a novel immunoinhibitory function for LGALS3BP, which may play a critical role in the immune evasion of tumor cells during cancer progression. PMID: 25320078
  28. 90K (Mac-2 binding protein) has been identified as a novel antiviral factor that reduces HIV-1 particle infectivity by interfering with the maturation and incorporation of HIV-1 Env molecules into virions. PMID: 24156545
  29. LGALS3BP is enriched in human ovarian carcinoma exosomes. PMID: 24302979
  30. Recombinant 90K exhibits an apparent molecular weight of approximately 78kDa, which is smaller than the natural 90K (approximately 97kDa). This suggests that recombinant 90K has smaller N-glycans with about half the molecular weight of N-glycans in natural 90K. PMID: 23830458
  31. Serum Mac-2 binding protein expression has emerged as a potential therapeutic target and biomarker for lung cancer. PMID: 23184915
  32. LGALS3BP has been shown to suppress the assembly of centriolar substructures. PMID: 23443559
  33. LGALS3BP induces vascular endothelial growth factor in human breast cancer cells and promotes angiogenesis. PMID: 22864925
  34. MIR596 is frequently observed in oral squamous cell carcinoma and regulates the expression of LGAL3BP. PMID: 23233740
  35. Breast cancer cells express Mac-2BP as a novel E-selectin ligand, potentially identifying a new prognostic and therapeutic target for breast cancer. PMID: 22970241
  36. Single nucleotide polymorphisms (SNPs) of the LGALS3BP gene, as found in the NCBI database, are not characteristic of papillary thyroid cancer, follicular adenomas, or nodular goiter. PMID: 17091456
  37. Research indicates that NF-kappaB regulates the expression of G3BP (Galectin-3 Binding Protein) and that G3BP enhances the adhesion of T47D breast cancer cells to fibronectin. PMID: 22447108
  38. Adeno-associated virus type 6 interacts with G3BP in human and dog sera but not in macaque serum. PMID: 22496229
  39. There is no difference in Gal-3 expression in peripheral blood lymphocytes in patients with papillary thyroid cancer. PMID: 17091455
  40. The LGALS3BP gene is expressed by neuroblastoma cell lines and neuroblasts from patients. PMID: 21660451
  41. Serum changes in three glycoproteins, galectin-3 binding protein, insulin-like growth factor binding protein 3, and thrombospondin 1, have been associated with the development of hepatocellular carcinoma. PMID: 21474793
  42. Mac-2BP has been identified as a predominant DC-SIGN ligand expressed on some primary colorectal cancer tissues. PMID: 21515679
  43. Research suggests that galectin-3 binding protein could be a potential therapeutic target, at least for colon cancer patients with high expression of this protein. PMID: 21094132
  44. Serum Mac-2BP does not appear to originate in the prostate and is unlikely to be useful for differentiating prostate cancer from benign prostatic hyperplasia. PMID: 20583127
  45. Initial assessments have shown that serum Mac-2BP is significantly elevated in patients with NETs (neuroendocrine tumors) and is expressed by the majority of NET tissues. PMID: 20019050
  46. Research has focused on the structural and functional properties of M2BP. PMID: 11867635
  47. Expression of 90K (Mac-2 BP) correlates with distant metastasis and predicts survival in stage I non-small cell lung cancer patients. PMID: 11980646
  48. Reviews have highlighted the role of LGALS3BP in tumor progression and metastasis. PMID: 14758079
  49. 90K (Mac-2 binding protein) plays a crucial role in maintaining an appropriate level of immune response. PMID: 15231701
  50. Elevated Mac-2 binding protein is associated with distant metastasis and higher tumor stage in gastric cancer. PMID: 17131321

Show More

Hide All

Database Links

HGNC: 6564

OMIM: 600626

KEGG: hsa:3959

STRING: 9606.ENSP00000262776

UniGene: Hs.514535

Subcellular Location
Secreted. Secreted, extracellular space, extracellular matrix.
Tissue Specificity
Ubiquitous. Detected in body fluids such as semen, milk, serum, tears, saliva and urine. Expressed by keratinocytes and fibroblasts.

Q&A

What is LGALS3BP and what cellular functions does it have?

LGALS3BP is a glycosylated protein with a calculated molecular weight of 65 kDa (585 amino acids), though it typically appears as 65-90 kDa in Western blots due to extensive post-translational modifications . Functionally, LGALS3BP serves as a negative regulator of NF-κB activation and proinflammatory cytokine production . Research demonstrates that it interacts with transforming growth factor β-activated kinase 1 (TAK1), inhibiting its phosphorylation and suppressing its kinase activity . In cancer biology, LGALS3BP is involved in various tumoral progression mechanisms including invasion, migration, immune evasion, and metastatic dissemination .

When designing experiments to study LGALS3BP function, researchers should consider its role both as a cellular protein and as a secreted factor that can act through autocrine and paracrine mechanisms. Immunoprecipitation approaches targeting TAK1 can help confirm the interaction with LGALS3BP in your experimental system.

What applications are LGALS3BP antibodies validated for?

LGALS3BP antibodies have been validated for multiple experimental applications including:

ApplicationValidation Details
Western Blot (WB)Validated in HeLa, COLO 320, HEK-293, HepG2, A549 cells, fetal human brain, human milk, and plasma
Immunoprecipitation (IP)Successfully used in HepG2 and HEK-293 cells
Immunohistochemistry (IHC)Validated in human esophageal, breast, and colon cancer tissues
Immunofluorescence (IF/ICC)Confirmed in HepG2 cells
Co-Immunoprecipitation (CoIP)Used to study protein-protein interactions
ELISAEffective for quantitative analysis

When selecting an antibody for your specific application, consider whether the antibody has been validated in similar sample types. The 10281-1-AP antibody, for example, has been cited in numerous publications spanning diverse applications .

What tissue/cell types show high LGALS3BP expression?

LGALS3BP expression varies across tissues and is particularly elevated in cancer contexts. Expression patterns include:

Normal tissues/cells:

  • Human fetal brain tissue

  • Human milk

  • Human plasma

  • Various cell lines including HeLa, COLO 320, HEK-293, HepG2, and A549

Cancer tissues:

  • Glioblastoma multiforme (GBM), with significantly higher expression than normal brain tissue

  • Adenoid cystic carcinoma (ACC), showing heterogeneous expression patterns

  • Melanoma and neuroblastoma, with elevated expression

  • Human esophageal, breast, and colon cancer tissues

When studying LGALS3BP expression, it's important to include appropriate positive controls from this list. Immunohistochemical analysis has revealed that expression can be heterogeneous within tumor samples, suggesting the need for comprehensive tissue assessment rather than limited sampling .

How should I optimize antibody dilutions for detecting LGALS3BP?

Optimizing antibody dilutions is critical for achieving specific signal while minimizing background. For LGALS3BP detection, recommended dilution ranges are:

ApplicationRecommended Dilution Range
Western Blot (WB)1:1000-1:4000
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg of total protein lysate
Immunohistochemistry (IHC)1:200-1:800
Immunofluorescence (IF/ICC)1:200-1:800

Methodological considerations for optimization:

  • Perform a titration series around the recommended range (e.g., 1:500, 1:1000, 1:2000, 1:4000, 1:8000 for WB)

  • Include both positive controls (e.g., HepG2 cells) and negative controls

  • For IHC, test both suggested antigen retrieval methods: TE buffer pH 9.0 (preferred) or citrate buffer pH 6.0

  • Evaluate signal-to-noise ratio at each dilution rather than simply selecting the strongest signal

  • For challenging samples, consider extending incubation times rather than increasing antibody concentration to improve signal quality

Remember that optimal dilution is sample-dependent; titration in each experimental system is essential for obtaining reliable results .

What is the molecular weight range for LGALS3BP detection on Western blots?

When performing Western blot analysis of LGALS3BP, researchers should be aware of the following molecular weight considerations:

  • Calculated molecular weight: 65 kDa (585 amino acids)

  • Observed molecular weight range: 65-90 kDa

  • The discrepancy between calculated and observed weight is primarily due to extensive glycosylation

Methodological recommendations:

  • Use molecular weight markers that span 50-100 kDa range

  • Run gels with sufficient resolution in the 65-90 kDa region (8-10% polyacrylamide gels are suitable)

  • Be prepared for potential variation in apparent molecular weight across different sample types due to differential glycosylation patterns

  • For cleaner detection, consider longer wash steps after primary and secondary antibody incubations

  • If multiple bands appear, enzymatic deglycosylation (e.g., with PNGase F) can help confirm the specific LGALS3BP band

These considerations ensure accurate identification of LGALS3BP in Western blot applications.

How does LGALS3BP expression differ between normal and cancer tissues?

LGALS3BP shows significant expression differences between normal and cancer tissues, making it a protein of interest for cancer diagnostics and therapeutics:

  • Cancer overexpression: LGALS3BP is "largely overexpressed in tumor tissue compared to the non-neoplastic counterpart," as demonstrated across multiple cancer types .

  • Specific cancer findings:

    • Glioblastoma multiforme (GBM): Immunohistochemical analysis shows high LGALS3BP expression compared to normal brain tissue

    • Adenoid cystic carcinoma (ACC): Heterogeneous expression observed in both PDX models and patient tumor samples

    • Multiple other cancers including melanoma and neuroblastoma show elevated expression

  • Methodological approaches for comparative analysis:

    • Paired analysis of tumor and adjacent normal tissue from the same patient

    • Tissue microarray (TMA) analysis across multiple patients

    • Quantitative image analysis of immunohistochemistry staining intensity

    • Correlation of protein expression with patient outcomes and clinical parameters

  • Vesicular vs. total protein: In GBM patients, "the amount of vesicular but not total circulating protein is increased" compared to healthy donors , suggesting the importance of analyzing both total and vesicle-associated LGALS3BP.

This differential expression pattern provides both diagnostic opportunities and therapeutic rationale for targeting LGALS3BP in cancer.

What are the best sample preparation methods for LGALS3BP detection in different applications?

Optimal sample preparation is crucial for reliable LGALS3BP detection across different experimental applications:

For Western Blot analysis:

  • Lysis buffer: RIPA buffer containing protease inhibitors is suitable for most applications

  • For phosphorylation studies (e.g., TAK1 interaction), include phosphatase inhibitors

  • Process samples quickly and maintain cold temperatures to prevent protein degradation

  • For secreted LGALS3BP, concentrate cell culture supernatants using centrifugal filters or TCA precipitation

For Immunohistochemistry (IHC):

  • Fixation: 10% neutral buffered formalin fixation for 24-48 hours

  • Antigen retrieval: TE buffer pH 9.0 is recommended as the primary method, with citrate buffer pH 6.0 as an alternative

  • Blocking: Use 3-5% normal serum or BSA to reduce background

  • For multiplexed staining, carefully select antibodies from different host species

For Immunoprecipitation (IP):

  • Gentler lysis buffers (e.g., NP-40 or CHAPS-based) help preserve protein interactions

  • Pre-clear lysates with protein A/G beads to reduce non-specific binding

  • Use 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate

  • For co-IP studies targeting TAK1 interactions, consider crosslinking approaches

For Extracellular Vesicle (EV) analysis:

  • Isolation methods: Differential ultracentrifugation, size-exclusion chromatography, or precipitation methods

  • Validation: Confirm EV isolation using markers like CD63, CD9, and CD81

  • Characterization: Nanoparticle tracking analysis and electron microscopy

  • Controls: Include detergent controls to confirm vesicular nature of the signal

These optimized methods will enhance detection sensitivity and specificity across applications.

How can I validate the specificity of a LGALS3BP antibody?

Validating antibody specificity is essential for generating reliable data. For LGALS3BP antibodies, a comprehensive validation approach should include:

  • Genetic validation approaches:

    • LGALS3BP knockout cells/tissues as negative controls

    • CRISPR-Cas9 generated LGALS3BP-knockout mice have been reported and provide excellent validation models

    • siRNA or shRNA knockdown of LGALS3BP shows dose-dependent reduction in signal

  • Biochemical validation methods:

    • Peptide competition assays where pre-incubation with immunizing peptide abolishes specific signal

    • Immunoprecipitation followed by mass spectrometry to confirm identity

    • Deglycosylation experiments to confirm glycoprotein identity based on mobility shift

  • Multiple antibody approach:

    • Compare results using antibodies targeting different LGALS3BP epitopes

    • Consistent results with different antibodies increase confidence in specificity

  • Expression pattern validation:

    • Confirm detection in known positive control samples (HeLa, HepG2 cells)

    • Verify molecular weight range (65-90 kDa) in Western blots

    • Correlate protein expression with mRNA levels using qPCR

  • Application-specific controls:

    • For IHC/IF: Include isotype controls and primary antibody omission controls

    • For WB: Include positive control lysates from cells with confirmed expression

    • For IP: Perform parallel IPs with non-specific IgG

Implementing these validation strategies will establish confidence in antibody specificity and experimental results.

What controls should I use when studying LGALS3BP expression?

Proper experimental controls are essential for accurate interpretation of LGALS3BP expression studies:

Positive Controls:

  • Cell lines: HeLa, COLO 320, HEK-293, HepG2, and A549 cells all express LGALS3BP and serve as reliable positive controls

  • Tissues: Human fetal brain, milk, and plasma samples have confirmed LGALS3BP expression

  • Cancer tissues: Esophageal, breast, and colon cancer tissues show robust expression

Negative Controls:

  • LGALS3BP knockout models: CRISPR-Cas9 generated LGALS3BP-knockout mice provide ideal negative controls

  • Antibody controls: Isotype-matched IgG or pre-immune serum in place of primary antibody

  • Peptide competition: Primary antibody pre-incubated with immunizing peptide

Expression Modulation Controls:

  • Overexpression: Transient transfection with LGALS3BP expression vectors

  • Knockdown: siRNA or shRNA targeting LGALS3BP with scrambled sequences as controls

  • LPS stimulation: For NF-κB pathway studies, compare with and without LPS treatment

Technical Controls:

  • Loading controls: β-actin or GAPDH for Western blots

  • Microscopy controls: DAPI nuclear counterstain for IHC/IF

  • Quantification controls: Standard curves for ELISA-based quantification

These multi-layered controls ensure reliable interpretation of LGALS3BP expression data across experimental contexts.

How does LGALS3BP regulation relate to NF-κB signaling pathways?

LGALS3BP functions as a significant negative regulator of NF-κB signaling through specific molecular mechanisms:

  • TAK1 interaction mechanism:

    • LGALS3BP directly interacts with transforming growth factor β-activated kinase 1 (TAK1)

    • This interaction inhibits TAK1 phosphorylation, suppressing its kinase activity

    • Reduced TAK1 activity leads to decreased protein stability and dampened downstream signaling

  • Impact on inflammatory signaling:

    • LGALS3BP inhibits NF-κB activation in response to lipopolysaccharide (LPS) stimulation

    • This results in decreased production of proinflammatory cytokines including IL-6, TNF-α, and IL-1β

    • Both mRNA expression and protein secretion of these cytokines are affected

  • Experimental evidence:

    • LGALS3BP-deficient mice show enhanced sensitivity to LPS-induced endotoxin shock

    • These mice exhibit shorter survival time and increased levels of proinflammatory cytokines

    • Overexpression of LGALS3BP in both wild-type and knockout MEFs suppresses NF-κB luciferase activity

    • Similar effects were observed in RAW264.7 macrophage cells

  • Experimental approaches to study this pathway:

    • NF-κB reporter assays (luciferase) to measure activation

    • Immunoblotting for phosphorylated TAK1 and downstream components

    • Co-immunoprecipitation to confirm LGALS3BP-TAK1 interaction

    • Cytokine ELISAs and qPCR to measure inflammatory outputs

This regulatory role suggests LGALS3BP may have therapeutic potential for inflammatory conditions beyond its applications in cancer.

What are the challenges in targeting LGALS3BP in therapeutic applications?

Developing LGALS3BP-targeted therapeutics presents several complex challenges that researchers must address:

  • Expression heterogeneity:

    • LGALS3BP shows heterogeneous expression across patients, as documented in adenoid cystic carcinoma

    • Variability may limit therapeutic efficacy in low-expressing subpopulations

    • Patient stratification based on expression levels may be necessary

  • Protein characteristics challenges:

    • As a secreted glycoprotein rather than a membrane-bound receptor, targeting strategies differ from traditional approaches

    • Extensive glycosylation (causing observed 65-90 kDa molecular weight variation) may affect antibody recognition

    • Glycosylation patterns may vary between patients and cancer types

  • Physiological function considerations:

    • LGALS3BP's role in negative regulation of NF-κB signaling suggests potential immune-related adverse effects

    • Long-term inhibition might increase inflammatory responses

    • Differential targeting of tumor-associated vs. normal LGALS3BP may be necessary

  • Technical and development challenges:

    • For antibody-drug conjugates, controlled conjugation chemistry and optimal drug-to-antibody ratio are critical

    • "Generation of stable cell line producing the engineered 1959-sss antibody as well as scale-up of the site-specific linker-less conjugation process" presents manufacturing challenges

    • For brain tumors, blood-brain barrier penetration must be considered

  • Preclinical model limitations:

    • Limited number of PDX models for efficacy testing, as noted in ACC research where "only one PDX model was used for therapeutic studies"

    • Translating preclinical efficacy to clinical outcomes remains uncertain

Despite these challenges, promising results have been obtained with anti-LGALS3BP ADCs, showing "long-lasting response... in 100% of treated animals" in an ACC model and "potent and dose-dependent antitumor activity" in GBM models .

How can LGALS3BP antibodies be used to study extracellular vesicle biology?

LGALS3BP antibodies provide valuable tools for investigating extracellular vesicle (EV) biology, particularly in cancer research:

  • Vesicular LGALS3BP as cancer biomarker:

    • "Compared with healthy donors, the amount of vesicular but not total circulating protein is increased" in glioblastoma patients

    • "Plasma vesicular LGALS3BP levels correlate with glioma tumour grade, making this protein a potential biomarker for early detection"

    • Research has shown that "LGALS3BP can be used for liquid biopsy as a marker of disease" in GBM models

  • Methodological approaches for EV isolation and analysis:

    • Differential ultracentrifugation: Sequential centrifugation steps (300g, 2000g, 10,000g, 100,000g)

    • Size-exclusion chromatography: Separate EVs based on size using specialized columns

    • Density gradient separation: Distinguish EV subpopulations with different densities

    • Characterization: Nanoparticle tracking analysis, electron microscopy, and Western blotting

  • LGALS3BP detection in EVs:

    • Western blotting of EV lysates using anti-LGALS3BP antibodies

    • ELISA-based quantification for high-throughput analysis

    • Immunoelectron microscopy for visualization of LGALS3BP on individual EVs

    • Flow cytometry using bead-based capture systems for EV analysis

  • Experimental design considerations:

    • Include EV markers (CD9, CD63, CD81) to confirm vesicle isolation

    • Use negative markers (GM130, calnexin) to exclude cellular contamination

    • Compare multiple isolation methods to ensure consistent results

    • Match isolation protocols between patient samples and controls

This approach allows researchers to leverage LGALS3BP as a biomarker for minimally invasive liquid biopsy applications, potentially improving early cancer detection and monitoring.

What experimental approaches can assess LGALS3BP's role in immune evasion?

Investigating LGALS3BP's potential role in cancer immune evasion requires specialized experimental approaches:

  • Tumor-immune cell interaction studies:

    • Co-culture systems: Compare wild-type vs. LGALS3BP-knockout tumor cells with immune cells

    • Functional assays: Measure T cell proliferation, cytotoxicity, and cytokine production

    • Migration assays: Assess immune cell recruitment in response to LGALS3BP-containing conditioned media

  • NF-κB pathway analysis in immune contexts:

    • Given LGALS3BP's role as a negative regulator of NF-κB activation , examine effects on immune cell activation

    • Measure TAK1 phosphorylation in immune cells exposed to recombinant LGALS3BP

    • Compare cytokine production in immune cells from wild-type vs. LGALS3BP-knockout mice

  • In vivo immune profiling:

    • Utilize LGALS3BP-knockout mice to assess immune responses to tumor challenge

    • Perform immunophenotyping of tumor-infiltrating lymphocytes in presence/absence of LGALS3BP

    • Evaluate combination approaches with immune checkpoint inhibitors

  • Vesicular LGALS3BP immunomodulation:

    • Isolate LGALS3BP-containing EVs from tumor cells

    • Assess effects on dendritic cell maturation and T cell activation

    • Compare immunomodulatory properties of EVs from LGALS3BP-knockout vs. wild-type cells

  • Molecular and cellular analysis techniques:

    • Multiplex cytokine analysis focused on IL-6, TNF-α, and IL-1β

    • Flow cytometry to assess immune cell activation markers

    • Single-cell RNA sequencing to identify transcriptional changes in immune populations

    • Spatial transcriptomics to map LGALS3BP expression relative to immune infiltration

These approaches can help elucidate whether LGALS3BP contributes to tumor immune evasion, potentially opening new avenues for immunotherapy combinations.

How do post-translational modifications affect LGALS3BP antibody recognition?

Post-translational modifications (PTMs), particularly glycosylation, significantly impact LGALS3BP antibody recognition and must be carefully considered:

  • Molecular weight variability due to glycosylation:

    • LGALS3BP has a calculated molecular weight of 65 kDa but is observed at 65-90 kDa in Western blots

    • This substantial variation (~25 kDa) indicates extensive glycosylation

    • Different cell types and tissues may show different apparent molecular weights

  • Experimental approaches to address PTM variability:

    • Enzymatic deglycosylation: Treat samples with PNGase F to remove N-linked glycans

    • Compare native vs. deglycosylated forms on Western blots

    • Use multiple antibodies targeting different epitopes to ensure comprehensive detection

    • Include positive control samples with known glycosylation patterns

  • Technical considerations for antibody selection:

    • Antibodies targeting glycosylated epitopes may show variable recognition

    • The 10281-1-AP antibody was generated against a LGALS3BP fusion protein (Ag0294) , which may affect epitope presentation

    • For consistent detection, select antibodies targeting protein regions less affected by glycosylation

  • Methodological recommendations for consistent detection:

    • Run gradient gels to improve resolution of different glycoforms

    • Consider lectin blotting in parallel to assess glycosylation patterns

    • For quantitative analysis, compare results from antibodies targeting different regions

    • Document the specific molecular weight observed in your experimental system

  • Therapeutic implications:

    • For antibody-drug conjugates like 1959-sss/DM4, PTM variability could affect targeting efficiency

    • Selection of antibodies for therapeutic use should prioritize epitopes consistently presented despite PTM variations

Understanding these PTM effects is crucial for reliable detection and effective therapeutic targeting of LGALS3BP.

What are the best methodologies for developing LGALS3BP-targeted antibody-drug conjugates?

Development of effective LGALS3BP-targeted antibody-drug conjugates (ADCs) requires sophisticated methodological approaches, as exemplified by the 1959-sss/DM4 ADC described in the literature:

  • Antibody engineering considerations:

    • The 1959-sss antibody is described as an "engineered humanized monoclonal antibody"

    • Humanization reduces immunogenicity for clinical applications

    • Site-specific engineering enables controlled conjugation chemistry

  • Target validation methodologies:

    • Comprehensive immunohistochemical analysis across normal and cancer tissues

    • Quantitative assessment of expression levels in target cancers

    • Heterogeneity analysis within tumor samples to predict response rates

    • Confirmation that LGALS3BP is "largely overexpressed in the tumor tissue compared to the non-neoplastic counterpart"

  • Strategic payload selection:

    • The 1959-sss/DM4 ADC utilizes SH-DM4, "a highly lipophilic analogue of maytansine"

    • Lipophilic properties enable diffusion into cells and potential bystander effect

    • This is particularly important for LGALS3BP as a secreted rather than membrane-bound target

  • Conjugation technology optimization:

    • "Site-specific linker-less conjugation process" provides more homogeneous ADC products

    • Optimizing drug-to-antibody ratio for maximum efficacy while maintaining stability

    • Analytical characterization of ADC homogeneity and stability

  • Preclinical evaluation approaches:

    • In vivo imaging using "[89Zr]Zr-DFO-1959" radioimmunoconjugate to confirm tumor targeting

    • Patient-derived xenograft (PDX) models that maintain tumor heterogeneity

    • Efficacy studies demonstrating "durable tumor growth inhibition (TGI) in 100% of animals without observed toxicity"

    • Pharmacokinetic and biodistribution analysis

The 1959-sss/DM4 ADC has demonstrated impressive preclinical results across multiple cancer types including "potent and durable antitumor activity in melanoma, neuroblastoma and glioblastoma" and promising activity in adenoid cystic carcinoma , warranting further clinical development.

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.