LTBR Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can dispatch the products within 1-3 working days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time information.
Synonyms
CD18 antibody; D12S370 antibody; LT beta R antibody; LTBETAR antibody; Ltbr antibody; Lymphotoxin B receptor antibody; Lymphotoxin beta receptor (TNFR superfamily; member 3) antibody; Lymphotoxin beta receptor antibody; Lymphotoxin-beta receptor antibody; TNF R III antibody; TNF-RIII antibody; TNFCR antibody; TNFR RP antibody; TNFR superfamily member 3 antibody; TNFR-III antibody; TNFR2 RP antibody; TNFR2RP antibody; TNFR3 antibody; TNFRII antibody; TNFRRP antibody; TNFRSF 3 antibody; TNFRSF3 antibody; TNR3_HUMAN antibody; Tumor necrosis factor C receptor antibody; Tumor necrosis factor receptor 2 related protein antibody; Tumor necrosis factor receptor 2-related protein antibody; Tumor necrosis factor receptor superfamily member 3 antibody; Tumor necrosis factor receptor superfamily member 3 precursor antibody; Tumor necrosis factor receptor type III antibody
Target Names
Uniprot No.

Target Background

Function
The lymphotoxin beta receptor (LTβR) is a receptor for the heterotrimeric lymphotoxin containing LTA and LTB, and for TNFS14/LIGHT. It promotes apoptosis through TRAF3 and TRAF5. LTβR may play a role in the development of lymphoid organs.
Gene References Into Functions
  1. The lymphotoxin beta receptor (LTβR) elicits the rapid release of NF-kappaB inducing kinase (NIK) from the receptor complex, leading to non-canonical NF-kappaB signaling. PMID: 29329668
  2. These results suggest that the LTBR rs12354 polymorphism might be associated with the spontaneous resolution of hepatitis B virus infection. PMID: 28575727
  3. LTBR and BCLAF1 exhibited higher DNA methylation percentages in the marsupialized OKCs, but this difference did not affect gene expression (P > .05). PMID: 28864293
  4. Membrane-bound lymphotoxin-beta receptor (LTβR) and CXC chemokine receptor 2 (CXCR2) are involved in type B EAE development. PMID: 27820602
  5. The interaction between LIGHT and LTβR enhances the survival and proliferation of human bone marrow-derived mesenchymal stem cells. This suggests that LIGHT might play a crucial role in stem cell therapy. PMID: 27835685
  6. We found that LTβR polymorphisms were associated with severe BPH. Therefore, LTβR might contribute to the risk of BPH development. PMID: 26782510
  7. Activation of LTβR enhances the LPS-induced expression of IL-8 through NF-kappaB and IRF-1. PMID: 25887375
  8. LIGHT, via LTβR signaling, may contribute to the exacerbation of airway neutrophilic inflammation through cytokine and chemokine production by bronchial epithelial cells. PMID: 25501580
  9. Relative expression of HVEM and LTβR modulates canonical NF-kappaB and pro-apoptotic signals stimulated by LIGHT. PMID: 24980868
  10. LTβR is a lymphotoxin-B-specific receptor. PMID: 24563505
  11. Dimerization of LTβR by LTalpha1beta2 is necessary and sufficient for signal transduction. PMID: 24248355
  12. Results suggest that blockade of lymphotoxin-beta receptor (LTBR) pathways may hold therapeutic potential for the treatment of Sjogren's syndrome. PMID: 22044682
  13. LTBR gene polymorphisms may be associated with the risk of IgA nephropathy in Korean children. PMID: 22417320
  14. Research demonstrates that a cognate interaction between LTalphabeta on CD4(+) helper T cells and LTβ receptor on DCs results in unique signals that are essential for optimal CD8(+) T-cell expansion via a type I IFN-dependent mechanism. PMID: 21245292
  15. These findings indicate that AdipoR1 interacted with LTBR and mediated the inhibition of LTBR-activated NF-kappaB pathway. PMID: 21195057
  16. LTβR levels are independently associated with atherosclerosis in multiple vascular beds. PMID: 20599198
  17. Increased potential for LTβ receptor signaling, coupled with increased bioavailability due to lower decoy receptor-3 (DcR3) avidity, provides a mechanism for polymorphic variants in LIGHT to contribute to the pathogenesis of inflammatory diseases. PMID: 20592286
  18. The LTβR modifies the ubiquitin:NIK E3 ligase and also acts as an allosteric regulator of the ubiquitin:TRAF E3 ligase. PMID: 20348096
  19. Modulation of cellular TRAF3 levels may contribute to the regulation of NFkappaB-dependent gene expression by LTBR by affecting the balance of LTBR-dependent activation of canonical and non-canonical NFkappaB pathways. PMID: 20185819
  20. Lymphotoxin beta receptor induces interleukin 8 gene expression via NF-kappaB and AP-1 activation. PMID: 12169272
  21. The NF-kappa B activation in lymphotoxin beta receptor signaling depends on the phosphorylation of p65 at serine 536. PMID: 12419817
  22. Data show that mRNA encoding LIGHT and its receptors [HVEM, LTβR, and TR6 (DcR3)] are present in placentas and cytotrophoblast cells at term. PMID: 12466117
  23. GA-binding protein (GABP) and Sp1 and retinoid receptors are required to mediate retinoic acid responsiveness of CD18 transcriptional activation in myeloid cells. PMID: 12485937
  24. ASK1 is one of the factors involved in the caspase-independent pathway of LTβR-induced cell death. PMID: 12566458
  25. TRAF2, TRAF3, cIAP1, Smac, and lymphotoxin beta receptor associate and are involved in apoptosis. PMID: 12571250
  26. LTβR, CD40, and TANK interact with TRAF3 at sites that promote molecular interactions driving specific signaling. PMID: 14517219
  27. Both LTβR and HVEM can discriminatively mediate the expression of different genes in cultured human umbilical vein endothelial cells, including LIGHT, a proinflammatory cytokine. PMID: 15917993
  28. Signaling through the lymphotoxin beta receptor (LTβR) expressed on mature hepatocytes induces massive hepatomegaly; LTβR and its ligands define a new pathway in supporting liver regeneration. PMID: 16002734
  29. Blockade of TNFSF14 signaling caused a substantial reduction in the expression of lymphotoxin beta receptor (LTβR)-controlled migration factors within the islets and disrupts organization of tertiary structures, leading to the prevention of diabetes. PMID: 16934497
  30. The membrane-bound receptor that mediates apoptosis (LTβR) was present in syncytiotrophoblast and cytotrophoblast cells in all samples but were detected in placental stromal cells only at week 8 and thereafter. PMID: 17010447
  31. Evidence in this review indicates that double positive-thymocytes regulate the differentiation of early thymocyte progenitors and gamma delta T cells through a mechanism dependent on LT beta receptor. PMID: 17336158
  32. These results define a model for the study of inflammatory lymphangiogenesis in the thyroid and implicate lymphotoxin beta receptor signaling in this process. PMID: 17360402
  33. This review discusses recent studies indicating that LT beta R signaling is required for liver regeneration, hepatitis, and hepatic lipid metabolism. PMID: 17896993
  34. Lymphotoxin beta receptor ligation directly activates gene expression in human vascular endothelial cells via both noncanonical and classical NF-kappa B pathways. PMID: 18292573
  35. LTbeta-R constitutively induces NF-kappaB activation, and this event may be associated with autonomous growth of melanoma cells. PMID: 18347013
  36. Lymphotoxin beta receptor (LTβR) was detected in Rheumatoid arthritis fibroblast-like synoviocytes. PMID: 18412315

Show More

Hide All

Database Links

HGNC: 6718

OMIM: 600979

KEGG: hsa:4055

STRING: 9606.ENSP00000228918

UniGene: Hs.1116

Subcellular Location
Membrane; Single-pass type I membrane protein.

Q&A

What is the molecular structure of LTBR and how does it affect antibody selection?

LTBR (also known as TNFRSF3) is a type I single-pass transmembrane protein with a molecular weight of 47 kDa (unglycosylated) or 61 kDa (fully glycosylated) . The protein structure includes:

  • Extracellular domain: Contains four cysteine-rich motifs characteristic of the TNF receptor superfamily, responsible for ligand binding

  • Transmembrane domain: Anchors the protein to the cell membrane

  • Intracellular domain: Consists of 175 amino acids with a proline-rich region near the cell membrane that interacts with TRAF proteins

When selecting antibodies, researchers should consider:

  • The target epitope location (extracellular vs. intracellular domains)

  • Whether the antibody recognizes glycosylated forms

  • Cross-reactivity with other TNF receptor family members

For functional studies, antibodies targeting the extracellular domain are preferred, while structural studies may benefit from antibodies recognizing conserved intracellular regions .

What are the primary applications of LTBR antibodies in immunological research?

LTBR antibodies serve multiple research purposes across different experimental platforms:

ApplicationMethodologyKey Considerations
Western BlotProtein detection in cell/tissue lysatesUse reducing conditions; expected bands at 47 kDa (unglycosylated) or 61 kDa (glycosylated)
ImmunoprecipitationProtein complex isolationEffective for studying LTBR interactions with TRAFs and signaling complexes
Flow CytometryCell-surface expression analysisPrimarily detects LTBR on monocytes, fibroblasts, epithelial cells; negative on T and B cells
ImmunohistochemistryTissue localization studiesWorks best on paraffin-embedded sections with antigen retrieval
ImmunofluorescenceSubcellular localizationShows predominant Golgi apparatus localization
Functional assaysSignaling pathway activationAgonistic antibodies can induce NF-κB activation and apoptosis (with IFN-γ)

For comprehensive pathway analysis, researchers often combine these techniques to correlate protein expression with functional outcomes in cell-specific contexts .

How should researchers interpret LTBR expression patterns across different cell types?

LTBR shows distinct expression patterns that are critical for experimental design and data interpretation:

  • High expression: Lung, liver, kidney, epithelial cells, fibroblasts, myeloid cells

  • Moderate expression: Heart, testes

  • Low expression: Brain, thymus, spleen, lymph nodes

  • Negative expression: T cells, B cells, NK cells

When analyzing LTBR expression:

  • Include appropriate positive controls (e.g., HeLa, HepG2 cell lines)

  • Use negative controls (lymphocytes) to establish specificity

  • Consider tissue-specific glycosylation differences affecting antibody recognition

  • Note that LTBR expression changes in inflammatory conditions and tumor microenvironments

Recent single-cell RNA-seq analysis has revealed that LTBR is dominantly expressed in myeloid cells, particularly tumor-associated macrophages (TAMs), which has significant implications for cancer immunotherapy research .

How can LTBR antibodies be utilized to study tertiary lymphoid structures (TLS) in the tumor microenvironment?

Tertiary lymphoid structures (TLS) in tumors strongly correlate with improved prognosis and treatment outcomes. LTBR signaling is essential for their development:

Methodological Approach:

  • TLS Identification: Use multiplex immunofluorescence with LTBR antibodies combined with markers for high endothelial venules (HEVs), B cells, T cells, and dendritic cells

  • Functional Analysis: Apply agonistic LTBR antibodies to in vitro co-cultures of stromal and immune cells to assess TLS formation capacity

  • In vivo Evaluation: Utilize surrogate bispecific antibodies in murine tumor models to measure:

    • TLS formation (histological assessment)

    • Immune cell infiltration (flow cytometry)

    • Tumor growth kinetics

    • Response to immunotherapy (e.g., anti-PD-L1)

Research findings show that LTBR agonism leads to robust formation of high endothelial venules (HEVs) and increased infiltration of T and B cells into tumors . This approach has demonstrated significant therapeutic potential, with LTBR agonist antibodies showing monotherapy activity and enhanced efficacy in combination with anti-PD-L1 therapy in breast cancer models .

What is the role of LTBR in tumor-associated macrophages (TAMs) and how can antibodies help investigate this?

Recent research has identified LTBR as a novel immune checkpoint specifically expressed on tumor-associated macrophages (TAMs):

Research Methodology:

  • Expression Analysis: Use flow cytometry with LTBR antibodies to quantify expression on different immune cell populations in tumor tissues

  • Functional Assessment: Apply agonistic or blocking LTBR antibodies to isolated TAMs to evaluate:

    • Expression of immunosuppressive molecules (PDL1, ARG2, COX2, IL10, TGFβ)

    • Effect on CD8+ T cell activation and proliferation

    • Signaling pathway activation (noncanonical NF-κB, Wnt/β-catenin)

  • Clinical Correlation: Analyze LTBR+ TAM infiltration in patient samples and correlate with:

    • Clinical stage and prognosis

    • Response to immunotherapy

    • Immune cell composition in the tumor microenvironment

Data show that LTBR+ TAMs correlate with lung adenocarcinoma stages, immunotherapy resistance, and poor prognosis. LTBR activation enhances TAM-mediated immunosuppression of CD8+ T cells by upregulating immunosuppressive molecules like PDL1 and ARG2, while disruption of LTBR in TAMs enhances the therapeutic effect of cancer immunotherapy .

How can bispecific antibodies targeting LTBR be developed and evaluated for cancer therapy?

The development of conditionally active LTBR-targeting bispecific antibodies represents an advanced research direction:

Development and Evaluation Protocol:

  • Design Strategy: Create bispecific antibodies combining:

    • LTBR binding arm: Targets the receptor for agonistic activity

    • FAP (Fibroblast Activation Protein) binding arm: Provides tumor microenvironment specificity

    • Engineered Fc with minimal FcγR binding

  • In Vitro Characterization:

    • Binding specificity and affinity assessment

    • Conditional activation testing with FAP+ and FAP- cells

    • Signaling pathway analysis (NF-κB activation)

    • Cytokine/chemokine production profiling

  • In Vivo Evaluation:

    • Pharmacokinetics and biodistribution

    • Tumor growth inhibition in syngeneic models

    • Combination studies with checkpoint inhibitors

    • Immune cell infiltration and TLS formation assessment

Recent research demonstrated that bispecific antibodies conditionally activated LTBR in the presence of FAP-expressing cells while showing no activity in their absence. These antibodies led to tumor regressions in combination with anti-PDL1 therapy in an EMT6 mouse model of breast cancer .

How should researchers select the appropriate LTBR antibody for specific experimental applications?

Selecting the optimal LTBR antibody requires careful consideration of multiple factors:

ApplicationRecommended Antibody TypeImportant Selection Criteria
Western BlotPolyclonal or monoclonalValidate with positive controls (HeLa, HepG2); expect 47-61 kDa bands
Flow CytometryFluorophore-conjugated monoclonal (e.g., PE, APC)Clone selection crucial; clones 31G4D8 and 71319 well-validated
IHC/IFPurified monoclonal or polyclonalEpitope accessibility after fixation; test on known positive tissues
Functional StudiesAgonistic monoclonalSelect clones with demonstrated biological activity (e.g., 31G4D8)
Blocking ExperimentsNeutralizing monoclonalValidated ability to inhibit ligand binding

For advanced applications:

  • Validate antibody specificity using LTBR knockout or knockdown controls

  • Perform epitope mapping to ensure targeting of functional domains

  • Consider cross-reactivity with mouse LTBR for translational research

  • Test multiple clones when developing therapeutic applications

What are the key methodological considerations for analyzing LTBR in single-cell and spatial transcriptomic studies?

Advanced transcriptomic approaches provide valuable insights into LTBR biology:

Methodological Framework:

  • Single-Cell RNA-seq Analysis:

    • Include all major immune and stromal cell populations in tumor samples

    • Apply dimensionality reduction (tSNE/UMAP) to identify distinct LTBR+ populations

    • Perform differential gene expression analysis between LTBR+ and LTBR- cells within the same lineage

    • Conduct trajectory analysis to study LTBR+ cell differentiation and plasticity

  • Spatial Transcriptomics:

    • Combine with multiplex immunofluorescence for protein-level validation

    • Map LTBR expression relative to tertiary lymphoid structures and tumor regions

    • Correlate with ligand expression (LTα1β2, LIGHT) to identify potential paracrine signaling

  • Data Integration:

    • Integrate single-cell and spatial data to understand contextual LTBR signaling

    • Correlate with clinical parameters and treatment responses

    • Develop computational models of LTBR-mediated intercellular communication

Recent studies using this approach discovered that LTBR is specifically expressed in tumor-associated macrophages rather than other tumor-infiltrated immune cells or even macrophages in normal lung tissues, with its ligand LTα1β2 mainly expressed by lymphoid cells (T, B, and NK cells) .

What experimental approaches can be used to study LTBR signaling pathways in different cell types?

LTBR activates diverse signaling pathways that can be studied using complementary approaches:

Comprehensive Signaling Analysis Protocol:

  • Pathway Activation Assessment:

    • Phosphorylation analysis of key signaling proteins (Western blot, phospho-flow cytometry)

    • Nuclear translocation of NF-κB subunits (immunofluorescence, nuclear fractionation)

    • Canonical vs. noncanonical NF-κB pathway activation (p100 processing)

  • Transcriptional Response Analysis:

    • RNA-seq after LTBR stimulation at multiple time points

    • ChIP-seq for NF-κB binding sites and epigenetic modifications

    • Reporter assays for pathway-specific transcriptional activity

  • Protein-Protein Interaction Studies:

    • Co-immunoprecipitation of LTBR with TRAF proteins

    • Proximity ligation assays for in situ interaction detection

    • FRET/BRET for real-time interaction dynamics

  • Functional Outcome Assessment:

    • Cell type-specific readouts (e.g., cytokine production, apoptosis, differentiation)

    • Genetic perturbation using CRISPR/Cas9 for pathway component validation

    • Pharmacological inhibitors to dissect pathway dependencies

Research has shown that LTBR maintains TAM immunosuppressive activity and M2 phenotype by activating noncanonical NF-κB signaling and Wnt/β-catenin signaling . Different cell types may exhibit distinct signaling outcomes following LTBR engagement, emphasizing the importance of cell-specific analyses.

How can researchers address common issues with LTBR detection in Western blotting?

Western blotting for LTBR can present several challenges requiring specific troubleshooting strategies:

Problem: Multiple bands observed

  • Solution: LTBR exhibits variable glycosylation (47-61 kDa). Treat samples with glycosidases to confirm identity of bands

  • Method: Incubate lysates with PNGase F before SDS-PAGE to remove N-linked glycans

Problem: Weak or no signal

  • Solution: Optimize lysis conditions; NETN buffer has been validated for LTBR extraction

  • Method: Prepare lysates using NETN lysis buffer (20 mM Tris-HCl pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.5% NP-40) with protease inhibitors

Problem: Non-specific bands

  • Solution: Validate specificity with blocking peptide and negative control cells

  • Method: Pre-incubate antibody with immunizing peptide (as demonstrated with HeLa cell extracts)

Problem: Inconsistent results between samples

  • Solution: Standardize protein extraction and loading

  • Method: Use 50 μg of total protein per lane for cell lines with moderate LTBR expression (e.g., HeLa, HEK-293T)

For optimal results, researchers should use 1:500-1:1000 dilution of primary antibody and include both positive (HeLa, HepG2) and negative (lymphocyte) control lysates .

What approaches can help optimize LTBR immunostaining in different tissue types?

Immunohistochemical detection of LTBR requires optimization for different tissue contexts:

Tissue-Specific Optimization Protocol:

  • Fixation and Processing:

    • FFPE tissues: Use standard 10% neutral buffered formalin fixation (12-24 hours)

    • Frozen sections: Brief 4% paraformaldehyde fixation (10 minutes)

    • Critical step: Avoid overfixation which can mask LTBR epitopes

  • Antigen Retrieval Methods:

    • Heat-induced epitope retrieval: Citrate buffer (pH 6.0) for 20 minutes

    • For challenging tissues: Try Tris-EDTA (pH 9.0) or enzymatic retrieval

    • Optimize time and temperature based on tissue type

  • Background Reduction:

    • Implement dual blocking: Serum block (5% normal serum) followed by protein block

    • For high-background tissues: Add avidin-biotin blocking step

    • Consider tissue-specific autofluorescence quenching for IF

  • Signal Amplification Options:

    • Low expression tissues: Employ tyramide signal amplification

    • Multiplex detection: Use fluorophore-conjugated secondary antibodies

    • Chromogenic detection: HRP-polymer systems preferred over ABC method

Research has shown that LTBR is primarily localized in the Golgi apparatus in cancer cells, which may require additional permeabilization steps for optimal detection .

How can researchers validate the specificity and functionality of LTBR antibodies?

Comprehensive validation is essential for reliable LTBR antibody-based research:

Multi-level Validation Strategy:

  • Expression Level Validation:

    • Compare detection in cells with known LTBR expression levels (high: HeLa, HepG2; negative: T and B lymphocytes)

    • Use siRNA knockdown to confirm specificity of signal reduction

    • For therapeutic antibodies, test multiple cell lines representing target tissues

  • Functional Validation for Agonistic Antibodies:

    • NF-κB reporter assays with dose-response curves

    • Assessment of downstream gene expression (qPCR for target genes)

    • Cell-type specific functional readouts (e.g., apoptosis with IFN-γ co-treatment)

  • Epitope-Specific Validation:

    • Competitive binding assays with known ligands (LTα1β2, LIGHT)

    • Epitope mapping using overlapping peptides or mutagenesis

    • Cross-reactivity testing with related TNFR family members

  • In vivo Validation:

    • Use surrogate antibodies binding to mouse LTBR for murine models

    • Compare effects with LTBR knockout models

    • Evaluate pharmacodynamic markers of target engagement

For therapeutic development, conditional activation testing with tumor microenvironment markers (e.g., FAP) is crucial to confirm the specificity of targeted activation .

How might LTBR antibodies contribute to developing immunotherapy resistance biomarkers?

The emerging role of LTBR in immunotherapy resistance presents opportunities for biomarker development:

Research Approach:

  • Patient Cohort Analysis:

    • Analyze LTBR+ TAM infiltration in pre- and post-treatment biopsies

    • Correlate with response to immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1)

    • Perform multiparametric analysis combining LTBR with other immune markers

  • Mechanistic Studies:

    • Investigate how LTBR+ TAMs influence T cell exclusion and dysfunction

    • Examine relationships between LTBR expression and other resistance mechanisms

    • Develop in vitro co-culture systems to model LTBR-mediated resistance

  • Biomarker Development Pipeline:

    • Establish standardized LTBR immunohistochemistry protocols

    • Develop multiplexed imaging panels for contextual LTBR assessment

    • Create computational algorithms integrating LTBR with established biomarkers

Research has shown that LTBR+ TAMs are significantly increased in immunotherapy non-responders compared to responders in lung adenocarcinoma, suggesting potential utility as a predictive biomarker . Analysis of clinical trial cohorts (OAK and POPLAR) revealed that non-responders to atezolizumab had higher LTBR expression than responders, and patients with higher LTBR expression showed worse prognosis .

What are the potential applications of LTBR antibodies in combined immunotherapy approaches?

LTBR targeting offers unique opportunities for combination immunotherapy strategies:

Combination Strategy Framework:

  • Rationale-Based Combinations:

    • LTBR agonists + immune checkpoint inhibitors: Enhance TLS formation while blocking T cell inhibition

    • LTBR antagonists for TAMs + conventional immunotherapy: Reduce immunosuppression in the tumor microenvironment

    • Sequential approaches: LTBR modulation followed by checkpoint inhibition

  • Preclinical Evaluation Models:

    • Syngeneic mouse models with varying immunogenicity

    • Humanized mouse models for human-specific antibodies

    • Ex vivo tumor slice cultures for rapid screening

  • Monitoring Parameters:

    • Dynamic changes in immune cell composition

    • Tertiary lymphoid structure formation and function

    • Tumor-specific T cell responses

    • Changes in TAM phenotype and function

Preclinical research has demonstrated that LTBR agonist bispecific antibodies led to tumor regressions when combined with anti-PD-L1 therapy in breast cancer models . Conversely, disruption of LTBR in TAMs enhanced the therapeutic effect of cancer immunotherapy by reducing immunosuppression .

How can advanced molecular engineering approaches improve LTBR-targeting antibodies for research and therapy?

Next-generation LTBR antibody development leverages sophisticated engineering strategies:

Advanced Engineering Approaches:

  • Conditional Activation Technologies:

    • Tumor microenvironment-activated bispecific antibodies (e.g., LTBR x FAP)

    • Protease-activated antibodies responsive to tumor-associated enzymes

    • pH-sensitive antibodies exploiting tumor acidity

  • Format Optimization:

    • Fragment-based approaches (Fab, scFv) for improved tissue penetration

    • Multi-specific formats targeting complementary pathways

    • Antibody-drug conjugates for targeted cytotoxicity

  • Enhanced Functional Properties:

    • Fc engineering for optimal effector functions or extended half-life

    • Affinity maturation for improved binding characteristics

    • Stability engineering for better manufacturing and in vivo performance

  • Novel Screening Methodologies:

    • Phenotypic screening in complex co-culture systems

    • In vivo screening using surrogate antibody libraries

    • AI-assisted antibody design and optimization

Research on conditionally active therapeutic LTBR bispecific antibodies has demonstrated their ability to specifically activate LTBR signaling in the tumor microenvironment while minimizing systemic effects, representing a significant advancement in targeted immunotherapy approaches .

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.