LGALS3 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

LGALS3 monoclonal antibodies primarily target the carbohydrate-binding domain (CBD) of galectin-3, disrupting its interactions with extracellular matrix components, growth factors, and cell surface receptors. Key mechanisms include:

  • Blocking ligand interactions: Competing with lactose for the CBD, preventing galectin-3 from binding to MUC16, EGFR, or integrins .

  • Modulating signaling pathways: Inhibiting AKT, ERK1/2, and EGFR phosphorylation, which are critical for cancer cell migration and survival .

  • Reducing immune cell infiltration: Mitigating neutrophilia and lymphopenia in autoimmune diseases like systemic sclerosis (SSc) .

Therapeutic Applications

LGALS3 monoclonal antibodies have shown efficacy in diverse disease models:

Disease ModelAntibody CloneKey FindingsSources
Systemic Sclerosis (SSc)D11, E07Reduced skin/lung fibrosis, collagen deposition, and IL-5/IL-6 levels in HOCl-induced SSc mice. E07 normalized gene expression profiles.
Glioblastoma (GBM)EXTH-73Blocked EGFR activation, enhanced temozolomide efficacy, and prolonged survival in orthotopic GBM models.
MUC16-expressing Cancers14D11Inhibited Matrigel invasion, tumor growth, and lung metastasis in breast/ovarian cancer models.
Pancreatic Ductal Adenocarcinoma (PDAC)#67, #84Reduced lung metastasis nodules and improved survival in tail-vein injection models.

Antibody Characteristics

LGALS3 monoclonal antibodies vary in specificity, cross-reactivity, and applications:

AntibodyCloneHostReactivityApplicationsSource
E7B6RE7B6RRabbitHuman, Mouse, RatWB, IP, IHC, IFCell Signaling
OTI21D10OTI21D10MouseHuman, MouseWBAMSBio
CAB11198N/ARabbitHuman, Mouse, RatWB, IHC-P, IPAssayGenie
14D1114D11MouseHuman, MouseIn vitro invasion assays, in vivo tumor models

Fibrosis and Autoimmunity

In a hypochlorous acid (HOCl)-induced SSc mouse model, E07 reduced pulmonary macrophage infiltration and normalized transcriptomic profiles associated with disease severity . The Gal-3 up/down scores correlated with neutrophil-to-lymphocyte ratios, highlighting its role in systemic inflammation .

Oncology

  • GBM: EXTH-73 blocked EGFR-galectin-3 interactions, reducing tumor cell migration and enhancing chemoradiation efficacy .

  • MUC16 Cancers: 14D11 inhibited AKT/ERK signaling and delayed tumor growth in MDA-MB-231 breast cancer models .

  • PDAC: #67/#84 antibodies reduced metastatic nodules and improved survival in orthotopic lung metastasis models .

DNA Damage Response

LGALS3 silencing in HeLa cells increased resistance to ionizing radiation, etoposide, and carboplatin, suggesting a role in DNA repair modulation .

Future Directions

  • Clinical Translation: Humanized versions of murine antibodies (e.g., 14D11, EXTH-73) are needed for clinical trials.

  • Biomarker Development: Identifying Gal-3 interactomes could predict therapeutic responses in SSc or cancers.

  • Combination Therapies: Pairing LGALS3 antibodies with targeted therapies (e.g., EGFR inhibitors) may enhance efficacy.

Product Specs

Buffer
Phosphate Buffered Saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% Glycerol.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchasing method or location. Please contact your local distributors for specific delivery times.
Synonyms
35 kDa lectin antibody; Carbohydrate binding protein 35 antibody; Carbohydrate-binding protein 35 antibody; CBP 35 antibody; CBP35 antibody; Gal-3 antibody; GAL3 antibody; Galactose-specific lectin 3 antibody; Galactoside-binding protein antibody; GALBP antibody; Galectin 3 internal gene,included antibody; Galectin-3 antibody; Galectin3 antibody; GALIG antibody; GBP antibody; IgE binding protein antibody; IgE-binding protein antibody; L 31 antibody; L 34 antibody; L-31 antibody; L-34 galactoside-binding lectin antibody; L31 antibody; Laminin-binding protein antibody; Lectin L-29 antibody; Lectin, galactose binding, soluble 3 antibody; LEG3_HUMAN antibody; LGALS2 antibody; LGALS3 antibody; MAC 2 antigen antibody; Mac-2 antibody; Mac-2 antigen antibody; MAC2 antibody; Macrophage galactose-specific lectin antibody; MGC105387 antibody
Target Names
Uniprot No.

Target Background

Function
Galectin-3 is a galactose-specific lectin that binds to IgE. It may, in conjunction with the alpha-3, beta-1 integrin, mediate the stimulation of endothelial cell migration by CSPG4. In collaboration with DMBT1, it is essential for the terminal differentiation of columnar epithelial cells during early embryogenesis. Within the nucleus, it functions as a pre-mRNA splicing factor. Galectin-3 is involved in acute inflammatory responses, including neutrophil activation and adhesion, chemoattraction of monocytes and macrophages, opsonization of apoptotic neutrophils, and activation of mast cells. Working together with TRIM16, it coordinates the recognition of membrane damage with the mobilization of the core autophagy regulators ATG16L1 and BECN1 in response to damaged endomembranes.
Gene References Into Functions
  1. Research indicates that human resting NK cells express Gal-3 at both the gene and protein levels, and that Gal-3 expression can be modulated upon cytokine stimulation. In the same cells, Gal-3 consistently localizes intracellularly and functionally correlates with the degree of NK cell degranulation. PMID: 29248489
  2. The interaction between galectin-3 and its potential inhibitor, bergenin, has been studied using computational methods. PMID: 30276553
  3. It has been reported that the Galectin-3/NuMA interaction is functionally significant for spindle pole organization; spindle pole cohesion requires glycosylation-mediated localization of NuMA. PMID: 28469279
  4. The Mac-2-binding protein glycosylation isomer (M2BPGi), also known as Wisteria floribunda agglutinin-positive human Mac-2-binding protein, has recently been established as a glyco-biomarker of liver fibrosis in patients with chronic hepatitis C, exhibiting a unique fibrosis-related glycoalteration. PMID: 30128700
  5. Elevated plasma galectin-3 levels have been associated with an increased risk of developing incident chronic kidney disease, particularly among those with hypertension. PMID: 28865675
  6. Pre-interventional plasma Galectin-3 levels are associated with left ventricular reverse remodeling and clinical outcome after percutaneous mitral valve repair. PMID: 29678512
  7. Galectin-3 is significantly associated with functional capacity, cardiac function, and adverse cardiovascular events in adults with congenital heart defects. PMID: 28942393
  8. Gene expression analysis was conducted for miR-1, miR-21, and galectin-3 in hypertensive patients with symptomatic heart failure and left ventricular hypertrophy. PMID: 29905364
  9. Serum expression of miR-1 and miR-21, and the concentration of gal-3 were assessed in systolic heart failure patients with varying degrees of left ventricular dilatation. PMID: 29905365
  10. Serum galectin-3 is correlated with coronary atherosclerosis and obstructive sleep apnoea syndrome (OSAS) severity in OSAS patients. PMID: 28150280
  11. A study demonstrates that mesenchymal stromal cells (MSC)-derived LGALS3 might be crucial for essential biological pathways involved in MSC homeostasis and for regulating AML cell localization and survival within the leukemia microenvironmental niche. PMID: 29655803
  12. Low expression of galectin-3 was observed in all patients with malignant gastrointestinal tumors, regardless of the presence of eosinophilia. PMID: 29926280
  13. These findings highlight a new role for Galectin 3 as a non-canonical RNA-binding protein that regulates MUC4 mRNA post-transcriptionally. PMID: 28262838
  14. Galectin-3 proves to be superior to sST2 in distinguishing HFpEF from controls and HFrEF. PMID: 30039808
  15. Serum Gal-3 levels were significantly higher in breast cancer (BC) patients and did not significantly differ based on the clinical and tumoral characteristics of patients. Moreover, there was no discernible difference in Gal-3 levels between breast patients with and without metastatic disease. PMID: 30249872
  16. Gal3 expression in regional lymph nodes may be associated with the progression of oral squamous cell carcinomas. The increased Gal3 expression in regional lymph nodes of larger tumors underscores the need for immunomodulatory treatment approaches in early-stage oral squamous cell carcinomas. Blocking Gal3 could be a therapeutic option in oral cancer. PMID: 30115022
  17. High Gal-3 serum levels predict fibrosis of the atrial appendage. PMID: 28079145
  18. This study reviews the role of galectin-3 in atrial fibrillation mechanisms and its potential therapeutic implications. PMID: 29587379
  19. Our findings provide evidence that allele C of rs4652 and allele T of rs11125 in the galectin-3 gene may be risk factors for cervical cancer. PMID: 28848207
  20. High GAL-3 expression is associated with salivary gland tumors. PMID: 29580041
  21. Galectin-3 is significantly reduced in patients with cutaneous lupus lesions compared to healthy controls, which may contribute to the recruitment of inflammatory cells in the skin. PMID: 29058991
  22. Progesterone receptor, EGFR, and galectin-3 are expressed differentially in uterine smooth muscle tumors. PMID: 29729689
  23. Galectin-3 belongs to a class of inflammatory mediators that is associated with the degree of myocardial inflammation and fibrosis. It is related to the severity of myocardial ischemia and is negatively correlated with the cardiac ejection fraction. PMID: 29287900
  24. Increased GAL-3 levels are an independent predictor of all-cause mortality in hemodialysis patients. PMID: 26787685
  25. High GAL3 expression is associated with oral squamous cell carcinomas. PMID: 29284429
  26. High expression levels of Galectin-3 and low expression levels of TRAIL were found to be positively correlated with shorter median survival time and overall survival time. PMID: 28925481
  27. Galectin-3 is overexpressed in patients with thyroid nodules and can be used for diagnostic imaging and therapeutic targeting of cancer patients. (Review) PMID: 29393868
  28. With emerging evidence supporting the function and application of galectin3, this review aims to summarize the latest literature regarding the biomarker characteristics and potential therapeutic application of galectin3 in associated diseases. PMID: 29207027
  29. Our results indicate that concomitant stimulation and colocalization of galectin-3 with CD147 are associated with increased gelatinolytic activity in the actively ulcerating human cornea. PMID: 29340650
  30. Galectin-3 can be used as a biomarker for the prognosis evaluation of acute heart failure, and its combined analysis can increase the predictive value of NT-proBNP. PMID: 29077153
  31. Galectin has been identified as a proinflammatory protein and modulates immune responses, either as damage-associated molecular patterns or as pattern recognition receptors. PMID: 28828226
  32. Gal-3 was found to be significantly higher in the stroma of patients with H. pylori infection, mainly on Cag-A positive H. pylori, with chronic gastritis. PMID: 28939284
  33. The present study demonstrates a relationship between galectin-3 levels and total body fat, abdominal fat, body fat distribution, cardiac size and geometry, and an increase in total body fat over 2 years in young children. PMID: 29327139
  34. Results suggest that galectin-3 could help to monitor the risk of short-term mortality in unselected patients with acute heart failure (AHF) attended in the emergency department (ED). PMID: 28406038
  35. This report establishes the functional significance of Gal-3 as a broad-spectrum upstream effector in osteoarthritis (OA). A positive correlation between cartilage degeneration and Gal-3 positivity in chondrocytes and the ECM was found. PMID: 27982117
  36. The secretion of galectin-3 is identified as a novel mechanism for osteoblasts to control osteoclastogenesis and to maintain trabecular bone homeostasis independently of the RANKL/OPG-axis. PMID: 28822790
  37. Gal-3 plasma concentration was significantly higher in pulmonary arterial hypertension patients. PMID: 28826890
  38. Galectin-3 was associated with incident ischemic stroke in younger but not older individuals. PMID: 28872212
  39. As MUC1 and galectin-3 are both commonly overexpressed in most types of epithelial cancers, their interaction and impact on EGFR activation likely contribute significantly to EGFR-associated tumorigenesis and cancer progression. PMID: 28731466
  40. Serum Gal-3 is increased in acute exacerbation of chronic obstructive pulmonary disease patients. PMID: 28947730
  41. High serum levels of galectin-3 were found in patients with schizophrenia compared to controls. PMID: 28698921
  42. N-linked glycoprotein maturation is not required for Gal-3 transport from the cytosol to the extracellular space, but is important for cell surface binding. Additionally, secreted Gal-3 is predominantly free and not packaged into extracellular vesicles. These data support a secretion pathway independent of N-linked glycoproteins and extracellular vesicles. PMID: 28775154
  43. NRG1 and Gal-3 were significantly more elevated in cancer patients than in healthy controls. PMID: 28430337
  44. The discovery of site-specific N-glycosylation changes of LGALS3BP in association with PDAC may provide valuable clues to facilitate cancer detection or phenotype stratification. PMID: 28627758
  45. Our findings establish gal-3 as a molecular regulator of the JAG1/Notch-1 signaling pathway and have direct implications for the development of strategies aimed at controlling tumor angiogenesis. PMID: 28533486
  46. Findings reveal that differential levels of Gal-3 and Gal-8 expression and recruitment to Group A streptococcus (GAS) between epithelial cells and endothelial cells may contribute to the different outcomes of GAS elimination or survival and growth of GAS in these two types of cells. PMID: 28743815
  47. The methylation frequency of the galectin-3 promoter was significantly higher, while galectin-3 mRNA was lower in acute-on-chronic hepatitis B liver failure (ACHBLF) than in chronic hepatitis B and healthy controls. The results suggest that hypermethylation of the galectin-3 promoter might be an early biomarker for predicting disease severity and prognosis in patients with ACHBLF. PMID: 28185839
  48. Serum Gal-3 and Nox4 levels were significantly elevated and correlated in 26 human pulmonary arterial hypertension patients when compared with 14 age- and sex-matched healthy controls. PMID: 28431936
  49. These results demonstrate Tsc2-deficient mesenchymal progenitors cause aberrant morphogenic signals and identify an expression signature, including Lgals3, relevant for human diseases associated with TSC1/TSC2 inactivation and mTORC1 hyperactivity. PMID: 28695825
  50. Galectin-3 is a possible immunological target molecule of the pathogenic auto-antibodies. PMID: 27339072

Show More

Hide All

Database Links

HGNC: 6563

OMIM: 153619

KEGG: hsa:3958

STRING: 9606.ENSP00000254301

UniGene: Hs.531081

Subcellular Location
Cytoplasm. Nucleus. Secreted.
Tissue Specificity
A major expression is found in the colonic epithelium. It is also abundant in the activated macrophages. Expressed in fetal membranes.

Q&A

What is Galectin-3 and what cellular functions does it regulate?

Galectin-3 is a unique member of the chimeric galectins subfamily and the only galectin containing a non-lectin N-terminal region connected to a carbohydrate recognition domain. It functions as a β-galactoside-binding lectin with a molecular weight of approximately 30 kDa . Galectin-3 is involved in numerous cellular processes including cell growth, adhesion, inflammation, mRNA processing, and apoptosis .

In the extracellular space, Galectin-3 mediates cell-cell and cell-matrix interactions through binding to glycoproteins. Intracellularly, it participates in pre-mRNA splicing and associates with proteins involved in DNA damage response such as BARD1 and BRCA1 . Additionally, Galectin-3 plays roles in acute inflammatory responses by activating neutrophils, attracting monocytes/macrophages, and activating mast cells . Recent research also indicates Galectin-3 coordinates with TRIM16 in the recognition of membrane damage and mobilization of core autophagy regulators ATG16L1 and BECN1 .

How do LGALS3 monoclonal antibodies differ from other Galectin-3 inhibitors?

LGALS3 monoclonal antibodies provide highly specific targeting of Galectin-3 compared to small molecule inhibitors or carbohydrate-based antagonists. These antibodies can be designed to target specific epitopes, particularly the carbohydrate-binding domain, enabling precise inhibition of specific Galectin-3 functions . In contrast to other inhibitors that may have off-target effects, monoclonal antibodies offer greater specificity due to their high affinity for Galectin-3.

Research has demonstrated that monoclonal antibodies targeting Galectin-3 can effectively block interactions between Galectin-3 and its binding partners. For example, anti-Galectin-3 antibodies have been shown to block binding between Galectin-3 and EGFR in glioblastoma cells, resulting in decreased cell migration, invasion, and EGFR activation . This targeted approach enables researchers to interrogate specific Galectin-3 functions with greater precision than is possible with broader inhibitors.

What are the standard applications for LGALS3 monoclonal antibodies in research?

LGALS3 monoclonal antibodies are versatile research tools applicable across multiple experimental techniques. Common applications include:

  • Immunohistochemistry-paraffin (IHC-P): Detection of Galectin-3 in fixed tissue samples, typically at dilutions of 1-2 μg/ml .

  • Western blotting: Identification of Galectin-3 protein in cell lysates .

  • Immunofluorescence: Visualization of Galectin-3 localization within cells .

  • Functional inhibition studies: Blocking Galectin-3 interactions to assess effects on cellular processes .

  • Therapeutic targeting: Evaluation of anti-Galectin-3 antibodies as potential treatments for conditions with aberrant Galectin-3 expression .

When using these antibodies, positive controls such as human papillary thyroid carcinoma tissue are recommended for validation . The choice of specific clone and application parameters should be optimized based on the experimental system and research questions being addressed.

How do LGALS3 monoclonal antibodies impact DNA damage response pathways?

Research reveals a complex relationship between Galectin-3 and DNA damage response (DDR) pathways. Studies have identified Galectin-3 in complexes with BARD1 and BRCA1, key proteins involved in DNA damage repair . Interestingly, silencing LGALS3 in HeLa cells resulted in increased resistance to various DNA damaging agents including ionizing radiation, etoposide, carboplatin, and mitomycin C .

Specifically, LGALS3-silenced cells showed up to 60% increased viability compared to control cells after treatment with 20 nM etoposide . This suggests that Galectin-3 may play a regulatory role in cellular sensitivity to DNA damage, potentially through its interactions with BARD1/BRCA1 complexes. When employing LGALS3 monoclonal antibodies in research related to DNA damage, researchers should consider potential effects on:

  • DDR protein complex formation

  • Cell cycle checkpoint activation

  • DNA repair efficiency

  • Cellular sensitivity to genotoxic agents

These findings indicate that LGALS3 monoclonal antibodies may serve as valuable tools for investigating DDR pathways and potentially for sensitizing resistant cancer cells to DNA-damaging therapies.

What is the evidence for using LGALS3 monoclonal antibodies in cancer research models?

Multiple studies have demonstrated the potential utility of LGALS3 monoclonal antibodies in cancer research. In glioblastoma (GBM) models, a novel anti-Galectin-3 antibody showed significant therapeutic effects by:

  • Blocking Galectin-3 binding to EGFR, which is amplified in approximately 40% of GBM patients

  • Decreasing GBM cell migration and invasion in vitro

  • Reducing EGFR activation

  • Decreasing tumor burden in vivo

  • Providing a survival benefit when combined with temozolomide compared to standard treatment alone

In breast cancer models, knockdown of LGALS3 in MDA-MB-231 cells significantly improved survival in mouse xenograft models. Median survival for mice with wild-type MDA-MB-231 cells was 60 days (95% CI, 53.8-66.2), while median survival for mice with LGALS3-knockdown cells was not reached (p = 0.018) . This finding motivated the development of monoclonal antibodies targeting the Galectin-3 carbohydrate-binding domain to inhibit cancer cell invasion and growth .

These studies collectively support the rationale for developing and testing LGALS3 monoclonal antibodies as both research tools and potential therapeutic agents in cancer models, particularly those where Galectin-3 expression correlates with poor outcomes.

How can researchers evaluate the efficacy of LGALS3 monoclonal antibodies in fibrosis models?

Systemic sclerosis (SSc) research provides a framework for evaluating LGALS3 monoclonal antibodies in fibrosis models. A recent study developed Galectin-3 neutralizing monoclonal antibodies (D11 and E07) and assessed their efficacy in a mouse model of hypochlorous acid (HOCl)-induced SSc . The evaluation protocol included multiple parameters:

  • Skin thickness measurements

  • Quantification of collagen deposition in skin and lung tissues

  • Assessment of pulmonary macrophage infiltration

  • Measurement of inflammatory cytokines, particularly IL-5 and IL-6

  • Transcriptomic analysis of Galectin-3-associated gene networks

Results demonstrated that the antibodies reduced pathological skin thickening, lung and skin collagen deposition, pulmonary macrophage content, and plasma IL-5 and IL-6 levels . This suggests that when evaluating LGALS3 monoclonal antibodies in fibrosis models, researchers should employ multiple readouts spanning histological, cellular, molecular, and transcriptomic analyses to comprehensively assess efficacy.

What are the optimal conditions for using LGALS3 monoclonal antibodies in immunohistochemistry?

For optimal immunohistochemistry-paraffin (IHC-P) results with LGALS3 monoclonal antibodies, researchers should consider the following protocol parameters:

  • Antibody concentration: Use at 1-2 μg/ml dilution for most applications

  • Positive control selection: Human papillary thyroid carcinoma tissue is recommended as a reliable positive control

  • Storage conditions: Store at -20°C after aliquoting upon delivery, avoiding freeze/thaw cycles

  • Antibody formulation: Most commercial antibodies are supplied in 10mM Phosphate Buffered Saline with 0.05% BSA and 0.05% Sodium Azide

When optimizing staining protocols, researchers should verify antibody specificity using appropriate controls and may need to adjust concentration based on tissue type and fixation methods. For detection of Galectin-3 in cancer samples, particular attention should be paid to potential heterogeneity of expression within tumors.

How should researchers design experiments to study Galectin-3 interactions with binding partners?

To effectively study Galectin-3 interactions with binding partners such as EGFR, MUC16, or BARD1/BRCA1, researchers should employ a multi-faceted experimental approach:

  • Co-immunoprecipitation (Co-IP): Use LGALS3 monoclonal antibodies to pull down protein complexes, followed by western blotting for suspected binding partners .

  • Protein-protein binding assays: Employ direct binding assays with purified proteins to confirm interactions and determine binding kinetics.

  • Competitive inhibition studies: Test whether LGALS3 monoclonal antibodies can block specific protein-protein interactions, as demonstrated with Galectin-3 and EGFR in GBM cells .

  • Functional consequence assessment: Evaluate downstream signaling pathways affected by blocking interactions; for example, monitoring AKT, EGFR, and ERK1/2 phosphorylation when disrupting Galectin-3/MUC16 interactions .

  • Domain mapping: Use antibodies targeting specific Galectin-3 domains (particularly the carbohydrate-binding domain) to determine which regions mediate specific protein interactions .

This comprehensive approach allows researchers to validate interactions, characterize their specificity, and understand their functional relevance in cellular contexts.

What strategies can optimize the evaluation of LGALS3 monoclonal antibodies in vivo?

When evaluating LGALS3 monoclonal antibodies in vivo, researchers should implement rigorous experimental designs that account for multiple variables:

  • Model selection: Choose disease models with established Galectin-3 involvement, such as the HOCl-induced SSc mouse model for fibrosis or orthotopic GBM models for cancer .

  • Experimental controls: Include appropriate controls such as isotype control antibodies and comparison to established treatments (e.g., temozolomide for GBM) .

  • Dosing optimization: Perform dose-response studies to determine optimal antibody concentrations for efficacy while minimizing off-target effects.

  • Combination studies: Evaluate LGALS3 antibodies alone and in combination with standard treatments, as demonstrated by improved survival when combining anti-Galectin-3 antibody with temozolomide in GBM models .

  • Comprehensive endpoints: Measure multiple parameters including:

    • Survival analysis (e.g., Kaplan-Meier curves)

    • Tissue-specific effects (e.g., skin thickness, collagen deposition)

    • Cellular infiltration (e.g., macrophage content)

    • Molecular biomarkers (e.g., cytokine levels)

    • Pathway activation (e.g., EGFR signaling)

  • Pharmacokinetic assessment: Monitor antibody distribution, half-life, and clearance to optimize dosing schedules.

This systematic approach enables robust evaluation of LGALS3 monoclonal antibodies in preclinical models, laying the foundation for potential translational applications.

How do Galectin-3 expression patterns correlate with disease severity in systemic sclerosis?

Transcriptomic analysis of whole-blood samples from a cross-sectional cohort of 249 systemic sclerosis (SSc) patients revealed that Galectin-3 and its interactants define a strong transcriptomic fingerprint associated with disease severity . Analysis identified two clusters of genes - "Gal-3 up" (21 upregulated genes) and "Gal-3 down" (21 downregulated genes) - whose expression patterns strongly correlated with disease parameters .

Key correlations with Galectin-3 expression patterns included:

  • Disease subtype: Gal-3 up scores were higher in diffuse cutaneous SSc (dcSSc) than in limited cutaneous SSc (lcSSc) and sine scleroderma SSc (ssSSc) patients (p = 0.031). Conversely, Gal-3 down scores were lower in dcSSc patients (p = 0.030) .

  • Organ involvement: Higher Gal-3 up scores were associated with:

    • Pulmonary fibrosis (p = 0.029)

    • Worsening lung function (p = 0.025)

    • Basilar crackles (p = 0.0006)

    • Arrhythmia (p = 6.5 × 10^-6)

  • Immune cell populations: Gal-3 up fingerprint positively correlated with neutrophil counts and inversely correlated with B and T lymphocyte counts. The neutrophil-to-lymphocyte ratio, a marker of systemic inflammation, strongly correlated with the Gal-3 up score .

These findings suggest that monitoring Galectin-3 expression patterns could serve as a biomarker for SSc severity and organ involvement, potentially guiding the application of LGALS3 monoclonal antibodies in personalized treatment approaches.

What evidence supports the therapeutic potential of LGALS3 monoclonal antibodies in glioblastoma?

Glioblastoma (GBM) research has yielded compelling evidence supporting the therapeutic potential of LGALS3 monoclonal antibodies:

  • Correlation with outcomes: Increased Galectin-3 expression levels correlate with lower survival in glioma patients .

  • Treatment resistance: Galectin-3 levels increase following exposure to standard treatments (temozolomide or radiation), suggesting its role in treatment resistance .

  • Molecular mechanism: Galectin-3 directly binds to EGFR, which is amplified in approximately 40% of GBM patients. Anti-Galectin-3 antibodies can block this interaction .

  • In vitro efficacy: GBM cells treated with anti-Galectin-3 antibody showed:

    • Significant decrease in migration and invasion

    • Reduction in EGFR activation

  • In vivo efficacy: In mouse models, anti-Galectin-3 antibody:

    • Decreased tumor burden

    • Provided survival benefit when combined with temozolomide compared to standard treatment alone

These findings provide a strong rationale for further development of LGALS3 monoclonal antibodies as adjunctive therapy for GBM patients, particularly in combination with standard chemoradiation treatments.

How might LGALS3 monoclonal antibodies be integrated into combination therapy strategies?

Based on current research, LGALS3 monoclonal antibodies show promise for integration into combination therapy strategies through several mechanisms:

  • Enhancing standard treatments: In GBM models, anti-Galectin-3 antibodies improved outcomes when combined with temozolomide compared to standard treatment alone . This suggests potential synergistic effects with existing therapies.

  • Targeting treatment resistance: Since Galectin-3 expression increases following exposure to temozolomide or radiation , anti-Galectin-3 antibodies might help overcome acquired resistance to these treatments.

  • Dual-targeting approaches: Combining anti-Galectin-3 antibodies with other targeted therapies could address multiple disease pathways simultaneously. For example:

    • In cancers: Combining with EGFR inhibitors might enhance efficacy by blocking both EGFR and its interaction with Galectin-3 .

    • In fibrotic diseases: Combining with anti-inflammatory agents might address both fibrosis and inflammation components .

  • Sequential therapy protocols: Using anti-Galectin-3 antibodies after standard treatments when Galectin-3 levels are elevated might maximize therapeutic impact .

When designing combination therapy approaches, researchers should consider potential synergistic or antagonistic effects, optimal timing and dosing schedules, and comprehensive monitoring of both efficacy and safety endpoints.

What are the promising targets for next-generation LGALS3 monoclonal antibodies?

Based on current understanding of Galectin-3 biology, several promising targets for next-generation LGALS3 monoclonal antibodies warrant investigation:

  • Carbohydrate recognition domain (CRD): Developing antibodies with higher specificity and affinity for the CRD could more effectively block Galectin-3 interactions with glycoproteins . Research has shown that targeting this domain can inhibit cancer cell invasion and growth .

  • Protein-protein interaction sites: Beyond the CRD, identifying epitopes involved in specific protein interactions (such as with EGFR , BARD1/BRCA1 , or MUC16 ) could enable more selective functional inhibition.

  • Post-translational modification sites: Developing antibodies recognizing specific post-translationally modified forms of Galectin-3 might allow targeting of disease-specific variants.

  • N-terminal domain: The non-lectin N-terminal region of Galectin-3 mediates protein oligomerization and may be involved in some cellular functions distinct from carbohydrate binding .

  • Intracellular vs. extracellular targeting: Engineering antibodies or antibody fragments capable of intracellular delivery could target nuclear and cytoplasmic functions of Galectin-3, such as its role in pre-mRNA splicing .

These targeted approaches could yield more precise tools for both research and therapeutic applications, potentially addressing specific Galectin-3 functions while minimizing off-target effects.

How can transcriptomic data guide the development and application of LGALS3 monoclonal antibodies?

Transcriptomic analyses provide valuable insights that can guide LGALS3 monoclonal antibody development and application:

  • Patient stratification: The identification of Galectin-3 up and down gene signatures in SSc patients demonstrates how transcriptomic data can help identify patient subgroups most likely to benefit from anti-Galectin-3 therapy. Similar approaches could be applied to other diseases.

  • Target validation: Transcriptomic analysis confirming associations between Galectin-3 networks and disease parameters (e.g., organ dysfunction in SSc ) strengthens the rationale for therapeutic targeting.

  • Biomarker discovery: Gene expression patterns associated with Galectin-3 could serve as biomarkers for:

    • Patient selection for clinical trials

    • Monitoring treatment response

    • Early detection of disease progression

  • Pathway identification: Transcriptomic data revealing Galectin-3 associations with immune cell populations (e.g., neutrophilia and lymphopenia in SSc ) can guide investigation of specific cellular targets for antibody therapy.

  • Combination strategy design: Understanding gene networks influenced by Galectin-3 can inform rational design of combination therapies targeting multiple nodes in disease-relevant pathways.

By integrating transcriptomic approaches into LGALS3 monoclonal antibody research, investigators can develop more personalized and effective therapeutic strategies across multiple disease contexts.

What novel delivery systems might enhance the efficacy of LGALS3 monoclonal antibodies in hard-to-reach tissues?

For diseases affecting tissues with limited antibody penetration, such as brain tumors or fibrotic tissues, novel delivery systems could enhance LGALS3 monoclonal antibody efficacy:

  • Blood-brain barrier (BBB) strategies for CNS applications:

    • Bispecific antibodies incorporating transporters that facilitate BBB crossing

    • Nanoparticle encapsulation with BBB-penetrating properties

    • Focused ultrasound to temporarily disrupt the BBB

    • Intranasal delivery routes for brain targeting

  • Tissue-penetrating formats:

    • Antibody fragments (Fab, scFv) with better tissue penetration than full IgG

    • pH-responsive antibody formulations that enhance extravascular diffusion

    • Enzyme-cleavable linkers that release active antibody fragments within target tissues

  • Cell-mediated delivery:

    • Engineered immune cells expressing anti-Galectin-3 antibodies

    • Stem cell vehicles programmed to migrate to disease sites and release antibodies

  • Site-specific activation:

    • Prodrug-like antibody constructs that become fully active only in target tissues

    • Light-activated antibody systems for localized activation

  • Sustained-release formulations:

    • Hydrogel depots for prolonged local antibody release

    • Biodegradable microparticles providing extended antibody delivery

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 2024 Thebiotek. All Rights Reserved.