Recombinant Human B- and T-lymphocyte attenuator (BTLA), partial

Shipped with Ice Packs
In Stock

Description

Introduction

Recombinant Human B- and T-lymphocyte Attenuator (BTLA), partial, also known as CD272, is a co-inhibitory receptor belonging to the immunoglobulin superfamily . It is crucial in regulating immune responses by negatively modulating the activity of T and B lymphocytes . BTLA is expressed on immune cells including CD4+ and CD8+ T cells, B cells, natural killer (NK) cells, and antigen-presenting cells (APCs) like dendritic cells and macrophages . This widespread expression highlights its significance in maintaining immune homeostasis and preventing excessive immune activation, which could lead to autoimmunity or tissue damage .

Production of Recombinant Human BTLA Protein

Producing recombinant human BTLA protein involves several steps:

  1. Gene cloning

  2. Plasmid assembly

  3. Protein expression

  4. Purification

  5. Evaluation

Specific primers amplify the gene fragment encoding amino acids 31-150 of the human BTLA, which is ligated into a plasmid carrying the C-terminal hFc-Myc-tag gene . Mammalian cells are transfected with the recombinant plasmid using a transfection reagent, followed by the addition of a selective antibiotic to screen the successfully transfected cells . The recombinant BTLA protein is released by lysing the cells and is purified from the supernatant using affinity chromatography . Its purity, confirmed via SDS-PAGE, exceeds 90%, and the LAL assay shows its endotoxin levels below 1.0 EU/μg . Functional ELISA confirms that this active human BTLA protein binds the biotinylated human TNFRSF14 with an EC50 of 137.8-233.4 ng/mL .

BTLA and Immune Checkpoints in Cancer

BTLA's role in cancer has been explored, with studies showing its involvement in immune cell infiltration and patient prognosis in various cancers .

BTLA and TLR Signaling

BTLA inhibits TLR4 signaling in dendritic cells (DCs) . Cytokine responses against LPS were significantly enhanced in BTLA−/− DCs . BTLA inhibits both MyD88- and TRIF-dependent pathways on LPS stimulation in DCs, potentially by inducing the recruitment of SHP-2 into lipid rafts .

BTLA's Impact on TLR Signaling

TLR LigandLocationCytokine Response in BTLA−/− DCs
LPS (TLR4)Cell SurfaceSignificantly Enhanced
Pam3CSK4 (TLR2/1)Cell SurfaceSignificantly Enhanced
poly(I:C) (TLR3)EndosomeNormal
CpG (TLR9)EndosomeNormal

BTLA and Autoimmunity

Defective BTLA functionality in autoimmune diseases like lupus can be rescued by restoring lipid metabolism . When the TCR and BTLA are co-engaged, CD4+ T cell proliferation is inhibited . BTLA recruitment to TCR clusters is significantly lower in SLE patients compared to healthy controls .

BTLA Promoter Hypomethylation

BTLA promoter hypomethylation leads to increased BTLA mRNA and protein expression and higher immune cell infiltration .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary by purchasing method and location. Consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested and pre-arranged. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt; aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us for preferential development.
Synonyms
B and T lymphocyte associated protein; B and T lymphocyte attenuator; B and T lymphocyte associated; B- and T-lymphocyte attenuator; B- and T-lymphocyte-associated protein; BTLA; BTLA_HUMAN; BTLA1; CD272; CD272 antigen; FLJ16065; MGC129743
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
BTLA (B- and T-lymphocyte attenuator) is an inhibitory receptor on lymphocytes that negatively regulates antigen receptor signaling through PTPN6/SHP-1 and PTPN11/SHP-2. It may interact in cis (on the same cell) or in trans (on other cells) with TNFRSF14. Cis interactions appear to play an immunoregulatory role, inhibiting trans interactions in naive T cells to maintain a resting state. Trans interactions may predominate during the adaptive immune response, providing survival signals to effector T cells.
Gene References Into Functions
  1. Studies in B, T, and natural killer cell lines suggest that HVEM and UL144 (human herpes virus entry mediator and human herpesvirus 5 membrane glycoprotein UL144, respectively) bind a common epitope of BTLA, regardless of cis or trans engagement. (PMID: 29061848)
  2. The BTLA/HVEM pathway contributes to peripheral T cell suppression in hepatocellular carcinoma patients. (PMID: 30116751)
  3. Elevated BTLA levels predict poor prognosis in diffuse large B-cell lymphoma (DLBCL), suggesting that BTLA blockade, combined with other checkpoint inhibitors, could be a novel immunotherapy strategy. (PMID: 29353075)
  4. Genetic variants of rs76844316 in BTLA influence the susceptibility to severe chronic hepatitis B and may offer protection against disease progression. (PMID: 29558758)
  5. Impaired Bregs and CD19+/BTLA+ cells may contribute to the pathogenesis of multiple sclerosis (MS). (PMID: 27412504)
  6. BTLA uniquely provides both costimulatory and coinhibitory signals to activated CD8+ T cells, promoting survival, enhanced tumor control, and a functional recall response. (PMID: 28754817)
  7. The rs1982809 polymorphism in BTLA may be a low-penetrance risk factor for renal cell carcinoma (RCC), requiring further validation. (PMID: 27234378)
  8. Higher circulating BTLA+CD4+ lymphocytes are observed in patients with severe community-acquired pneumonia. (PMID: 28164546)
  9. Elevated plasma soluble BTLA levels early in sepsis/septic shock correlate with disease severity; baseline levels above 21 ng/mL predict poor prognosis. (PMID: 28056053)
  10. In chronic hepatitis B virus patients, inefficient interferon-gamma-producing antigen-specific CD8+ T cells recruited to the liver exhibit high BTLA expression. (PMID: 27743606)
  11. BTLA expression is associated with positive, rather than negative, regulation of CD11c antigen-presenting cell stimulatory capacity. (PMID: 27717503)
  12. The rs1982809 BTLA gene polymorphism is linked to mRNA expression levels and may be a low-penetrance risk factor for chronic lymphocytic leukemia. (PMID: 27933341)
  13. Decreased BTLA expression in ocular Behçet's disease suggests its involvement in disease development and recurrence. (PMID: 26841832)
  14. BTLA expression declines on B cells with age and is associated with reduced responsiveness to the trivalent influenza vaccine. (PMID: 26277622)
  15. CD200/BTLA deletions are recurrent genetic lesions in BCP-ALL (B-cell precursor acute lymphoblastic leukemia). (PMID: 26137961)
  16. Focal BTLA deletions are associated with B-cell precursor acute lymphoblastic leukemia. (PMID: 25261097)
  17. High BTLA expression in gastric cancer (identified by IHC) is an independent prognostic biomarker for poor outcome. (PMID: 25334051)
  18. Lung function and the expression of BTLA and Treg cells are lower in rheumatism patients compared to healthy controls. (PMID: 24909289)
  19. BTLA negatively regulates human Vγ9Vδ2 T-cell proliferation and contributes to immune escape in lymphoma cells. (PMID: 23692853)
  20. BTLA and HVEM may play roles in kidney transplant rejection. (PMID: 23375291)
  21. BTLA regulates human B cell responses, with implications for therapies targeting B cells. (PMID: 22903545)
  22. BTLA expression in rheumatoid arthritis (RA) synovial tissue suggests its involvement in regulating local T cell activation and RA pathogenesis. (PMID: 22691359)
  23. BTLA and/or HVEM may serve as diagnostic markers for innate immune response and therapeutic targets for sepsis. (PMID: 22459947)
  24. HVEM/BTLA may regulate joint inflammation in rheumatoid arthritis. (PMID: 22179929)
  25. BTLA-HVEM interactions impair minor histocompatibility antigen-specific T cell function, suggesting BTLA blockade as a post-stem cell transplantation therapy. (PMID: 22634623)
  26. BTLA blockade, along with PD-1 and Tim-3 blockade, enhances the expansion, proliferation, and cytokine production of NY-ESO-1-specific CD8+ T cells. (PMID: 22205715)
  27. The HVEM-BTLA cis complex intrinsically regulates T cells, silencing signals from the microenvironment. (PMID: 21920726)
  28. A mutagenesis study suggests that the CD160 and BTLA binding regions on HVEM overlap but are slightly different. (PMID: 21959263)
  29. The 590C SNP in BTLA is associated with rheumatoid arthritis susceptibility. (PMID: 21403914)
  30. BTLA expression on CD4+ and CD8+ T cells decreases with HIV-1 infection progression. (PMID: 21592997)
  31. BTLA pathway blockade enhances allogeneic and cytomegalovirus-specific CD8+ T cell proliferation, improving cytotoxic T lymphocyte function in viral infections. (PMID: 20693422)
  32. BTLA gene polymorphisms may affect sporadic breast cancer risk and prognosis in Chinese women. (PMID: 19585237)
  33. HVEM binding to BTLA attenuates T cell activation, identifying them as a coinhibitory receptor pair. (PMID: 15647361)
  34. Different herpesviruses target the HVEM-BTLA cosignaling pathway, highlighting its importance in regulating T cell activation during host defense. (PMID: 16131544)
  35. The 2.8-Å crystal structure of the BTLA-HVEM complex reveals that BTLA binds the N-terminal cysteine-rich domain of HVEM using a unique binding surface. (PMID: 16169851)
  36. BTLA is constitutively expressed on most CD4+ and CD8+ T cells and its expression decreases upon T cell activation. (PMID: 16643847)
  37. BTLA cross-linking suppresses T cell proliferation, downregulates CD25, and inhibits cytokine production. (PMID: 17257317)
  38. BTLA is an inhibitory BCR coreceptor, attenuating B cell activation by targeting Syk and BLNK. (PMID: 19155498)
  39. A study found no association between BTLA gene polymorphisms and the development of type 1 diabetes (T1D) or systemic lupus erythematosus (SLE) in the Japanese population. (PMID: 19207938)
  40. Downregulation of the BTLA-HVEM pathway may be involved in germinal center B-cell activation. (PMID: 19762537)
  41. The HVEM-BTLA cis-complex competitively inhibits HVEM activation, helping maintain T cells in a naive state. (PMID: 19915044)
  42. HVEM binds to BTLA, inhibiting T cell proliferation. (PMID: 15568026)
Database Links

HGNC: 21087

OMIM: 607925

KEGG: hsa:151888

STRING: 9606.ENSP00000333919

UniGene: Hs.445162

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

Q&A

What is BTLA and what is its primary function in the immune system?

BTLA is an inhibitory immune checkpoint receptor belonging to the CD28 family that plays a crucial role in maintaining immune homeostasis. When BTLA engages with its ligand HVEM, it induces phosphorylation of immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in its cytoplasmic tail, leading to recruitment of tyrosine phosphatases SHP1 and SHP2 . These phosphatases dephosphorylate key signaling molecules in T cell activation pathways, resulting in suppression of T cell proliferation, cytokine production, and effector functions . BTLA shares functional and structural similarity with other checkpoint receptors such as CTLA-4 and PD-1, delivering inhibitory signals that reduce cellular activation and growth .

How does BTLA interact with HVEM at the molecular level?

The crystal structure of the BTLA/HVEM complex (Protein Data Bank ID 2AW2) reveals that BTLA interacts primarily with the first cysteine-rich domain (CRD1) of HVEM . The binding interface involves a β-hairpin structure formed by residues 23-39 of HVEM's CRD1 domain, which creates an anti-parallel inter-molecular β-sheet with the G° strand of BTLA . Key residues in this interaction include Tyr23 and Val36 of HVEM, which serve as hot-spots for binding . The stability of this interaction is maintained through multiple hydrogen bonds that remain largely preserved during molecular dynamics simulations . The HVEM CRD1 domain contains four short β-strands connected by three disulfide bonds formed between cysteine residues 4-15, 16-29, and 19-37, contributing to the structural stability of the interaction interface .

What cell types express BTLA and how is its expression regulated?

BTLA is predominantly expressed on various immune cells, including:

Cell TypeBTLA Expression PatternDisease Context Variations
T cells (CD4+)Increased on activated cellsElevated in HCC and CLL; high expression correlates with shorter time to treatment in CLL
T cells (CD8+)Variable depending on activation stateIncreased in tumors but no correlation with disease progression in CLL
B cellsConstitutive expressionAlterations in malignancy contexts
Dendritic cellsModerate expressionIncreased after Hem/CLP (hemorrhage/cecal ligation and puncture)
Natural killer cellsLow to moderate expressionVaries by activation status

In cancer contexts such as hepatocellular carcinoma (HCC), BTLA expression is elevated on tumor-infiltrating T cells, with over 85% of BTLA+ CD4+ T cells co-expressing PD-1 . The regulation of BTLA expression involves both transcriptional and post-translational mechanisms influenced by cytokine signaling, T cell receptor engagement, and interactions with other immune checkpoint pathways .

What are the optimal methods to quantify BTLA expression on immune cells?

Several complementary methodological approaches can be employed to accurately quantify BTLA expression:

  • Flow Cytometry: The gold standard approach involves fluorescence-activated cell sorting (FACS) using fluorochrome-conjugated anti-BTLA monoclonal antibodies. Multi-parameter flow cytometry allows simultaneous detection of BTLA along with other surface markers to identify specific cell subsets .

  • Magnetic Cell Separation (MACS): For isolation of BTLA-expressing cells, magnetic cell separation using anti-BTLA antibodies coupled to magnetic beads can be employed, followed by flow cytometric analysis to confirm purity .

  • Immunohistochemistry/Immunofluorescence: These techniques can assess BTLA expression in tissue sections, providing spatial context for expression patterns.

  • Quantitative PCR: For analysis of BTLA mRNA expression, qPCR can be used, although this does not provide information about protein localization or expression on specific cell subsets unless combined with cell sorting.

When analyzing BTLA expression in disease contexts, it is essential to include appropriate controls and to consider co-expression with other immune checkpoints like PD-1, as co-expression patterns can provide valuable insights into the functional state of immune cells .

How can recombinant BTLA proteins be used in experimental settings?

Recombinant BTLA proteins serve as valuable tools in various experimental applications:

ApplicationMethodologyKey Considerations
Binding AssaysELISA, surface plasmon resonance, or biolayer interferometryWhen recombinant mouse HVEM/TNFRSF14 Fc chimera is coated at 0.5 µg/mL, recombinant BTLA binds with an ED50 of 0.1-0.5 µg/mL
Functional StudiesPlate-bound or soluble protein stimulationPurified recombinant BTLA can stimulate HVEM-expressing cells to study reverse signaling
Structural StudiesCrystallography, NMR, cryo-EMRequires high-purity, properly folded protein
Detection Reagent DevelopmentAntibody generation and validationConsider epitope selection for specificity
Therapeutic DevelopmentInitial screening and epitope mappingImportant for validating binding specificity

When working with recombinant BTLA proteins, researchers should consider the presence of tags (e.g., His-tag), the expression system used for production, and potential post-translational modifications that might affect function or interaction properties .

What experimental approaches can be used to study BTLA-HVEM interactions?

Multiple experimental approaches provide complementary insights into BTLA-HVEM interactions:

  • Molecular Modeling and Simulations: Using the crystal structure of BTLA/HVEM complex (PDB ID 2AW2), molecular dynamics simulations with GROMACS and the CHARMM22 force field can assess interaction stability and dynamics over time .

  • ELISA-Based Binding Assays: Direct binding assays can be performed by coating plates with recombinant BTLA-Fc protein (400 ng/well) and then adding titrated amounts of HVEM-Fc protein. The interaction can be detected using appropriately labeled secondary antibodies .

  • Competition Assays: To identify regions important for binding or to screen potential inhibitors, competition assays can test peptides or small molecules for their ability to disrupt the BTLA-HVEM interaction .

  • Ellman's Assay: For investigating the role of free thiols in the interaction, Ellman's assay using DTNB can measure free thiol content in protein samples, providing insights into the biochemical nature of the interaction .

  • Surface Plasmon Resonance (SPR): SPR enables real-time kinetic analysis of protein-protein interactions, determining association and dissociation rates as well as binding affinities.

These approaches provide structural, biochemical, and functional information about BTLA-HVEM interactions, essential for comprehensive understanding of this important immune regulatory axis .

How does BTLA expression correlate with disease progression in cancer?

BTLA expression demonstrates significant correlations with disease progression and outcomes across several cancer types:

Cancer TypeBTLA Expression PatternClinical Correlation
Hepatocellular Carcinoma (HCC)Increased BTLA+PD-1+ CD4+ T cellsSignificantly increased in advanced stage HCC (stage III/IV vs. I/II, p=0.0097)
Chronic Lymphocytic Leukemia (CLL)High BTLA on CD4+ T cellsCorrelates with diminished time to treatment
Other CancersBTLA/HVEM axis dysregulationLinked to poor outcomes in pancreatic adenocarcinoma, non-small-cell lung cancer, and follicular lymphoma

The relationship between BTLA expression and disease progression appears related to its role in promoting T cell exhaustion and impairing antitumor immunity . The co-expression of BTLA with other inhibitory checkpoints like PD-1 further contributes to the functional impairment of tumor-infiltrating lymphocytes, facilitating immune evasion and tumor progression .

What are the differences between BTLA and PD-1 in regulating T cell responses?

BTLA and PD-1 exhibit important differences in their mechanisms of action and effects on T cell responses:

  • Signaling Mechanisms: While both BTLA and PD-1 recruit SHP1 and SHP2 phosphatases, they affect downstream signaling pathways differently. BTLA potently inhibits the phosphorylation of both TCR (CD3ζ) and CD28, whereas PD-1 may have more selective effects on specific signaling pathways .

  • Expression Patterns: In hepatocellular carcinoma, about 83 ± 6.5% of BTLA-expressing tumor CD4+ T cells are PD-1+, whereas only 54 ± 7.9% of PD-1-expressing tumor CD4+ T cells are BTLA+, suggesting they mark partially overlapping but distinct T cell populations .

  • Functional Effects: BTLA engagement results in a more robust inhibition of IL-2 production and CD28 phosphorylation compared to PD-1 engagement in some experimental systems . BTLA can suppress T cell signaling through mechanisms independent of both SHP1 and SHP2 .

  • Response to Blockade: The effects of blocking BTLA versus PD-1 vary depending on the disease context. In some cases, dual BTLA/PD-1 blockade shows enhanced efficacy compared to monotherapies, suggesting non-redundant roles .

  • Ligand Interactions: Unlike PD-1, which interacts with PD-L1 and PD-L2, BTLA interacts with HVEM, a member of the TNF receptor superfamily. This cross-family interaction adds complexity not present in the PD-1 pathway .

These differences are crucial for developing targeted therapeutic approaches and predicting the effects of blocking these pathways in different disease contexts .

How does BTLA contribute to immune cell exhaustion in chronic diseases?

BTLA contributes to immune cell exhaustion through several interconnected mechanisms:

  • Sustained Inhibitory Signaling: Persistent engagement of BTLA with HVEM in chronic disease settings leads to continuous inhibitory signaling, suppressing T cell activation, proliferation, and effector functions over time .

  • Altered Cytokine Production: BTLA activation significantly reduces the production of key cytokines required for effective immune responses. In CLL, BTLA activation leads to decreased percentages of:

    • IL-2+ CD3+ T cells: 33.81 ± 5.57% vs. 21.71 ± 6.93% (p=0.01)

    • IL-2+ CD4+ T cells: 37.03 ± 5.52% vs. 23.87 ± 8.05% (p=0.01)

    • IL-2+ CD8+ T cells: 18.05 ± 4.02% vs. 10.35 ± 3.05% (p=0.01)

    • Similar reductions in IFN-γ across all T cell subsets

  • Impact on TCR Signaling Pathways: BTLA engagement suppresses key T cell signaling pathways, including MAPK, NF-κB, and NFAT activation, which are essential for T cell function . This broad inhibition of signaling contributes to the progressive functional impairment characteristic of exhausted T cells.

  • Co-expression with Other Inhibitory Receptors: In chronic diseases, BTLA is often co-expressed with other inhibitory receptors like PD-1, creating a multi-layered suppressive environment that enhances T cell exhaustion .

BTLA blockade has shown promise in reversing some aspects of T cell exhaustion. For example, BTLA blockade enhances IFN-γ production, particularly in CD8+ T cells, and can boost cytotoxic responses against tumor cells, suggesting that BTLA contributes to the reversible component of T cell exhaustion .

What is the relationship between BTLA expression and response to immunotherapy?

The relationship between BTLA expression and immunotherapy response is complex and context-dependent:

  • Predictive Biomarker Potential: BTLA expression on T cells may serve as a predictive biomarker for response to immunotherapy. In some cancers, high BTLA expression is associated with poor outcomes and may indicate patients who could benefit from targeted approaches .

  • Impact on Checkpoint Inhibitor Efficacy: BTLA on tumor-infiltrating lymphocytes may limit the efficacy of other checkpoint inhibitors, such as PD-1/PD-L1 blockade. Studies have shown that BTLA+ cells identify highly dysfunctional PD-1-expressing CD4+ T cell subsets in HCC .

  • Differential Effects of Blockade: BTLA blockade enhances IFN-γ production by T cells, particularly CD8+ T cells, but may have limited effects on IL-2 production. This suggests that BTLA blockade may selectively enhance certain aspects of T cell function .

  • Combination Approaches: Emerging evidence suggests that targeting BTLA in combination with other immunotherapeutic approaches provides enhanced efficacy. In CLL models, the combination of BTLA blockade with bispecific anti-CD3/anti-CD19 antibodies significantly boosted CD8+ T cell anti-leukemic activity .

  • Emerging Clinical Data: Clinical trials with novel anti-BTLA monoclonal blocking antibodies, such as icatolimab, are showing promising preliminary results in patients with advanced solid tumors .

Understanding this complex interplay is crucial for optimizing immunotherapeutic approaches and identifying patients most likely to benefit from BTLA-targeted interventions .

What approaches can be used to develop therapeutics targeting BTLA-HVEM interactions?

Several sophisticated approaches can be employed to develop therapeutics targeting BTLA-HVEM interactions:

  • Monoclonal Antibodies: Developing antibodies that block the BTLA-HVEM interaction is a primary approach. Icatolimab, a first-in-class anti-BTLA monoclonal blocking antibody, has shown promising preliminary results in clinical trials for advanced solid tumors .

  • Engineered HVEM Variants: Computational design methods like ProtLID (Protein Ligand Interface Design) can generate residue-based pharmacophores over the binding interfaces of HVEM. Single mutations like H86I and D100R on HVEM reduce HVEM binding to LIGHT, while double mutants like D100R with M103K achieve BTLA-selective binding .

  • Peptide Inhibitors: Peptides that mimic key interaction regions can provide competitive inhibitors of BTLA-HVEM binding. Molecular dynamics simulations show that the HVEM(23-39) fragment can stably interact with BTLA, suggesting it as a potential starting point for peptide inhibitor design .

  • Structural-Based Design: The crystal structure of BTLA/HVEM complex (PDB ID 2AW2) provides a foundation for structure-based drug design approaches to develop molecules that interfere with this interaction .

  • Bispecific Antibodies: Developing bispecific antibodies that simultaneously target BTLA and another relevant molecule can provide more specific modulation of immune responses in the tumor microenvironment.

These approaches provide various strategies for therapeutic intervention in the BTLA-HVEM pathway, each with distinct advantages and challenges in terms of specificity, efficacy, and clinical translation .

How does BTLA blockade affect different immune cell subsets in various disease contexts?

BTLA blockade has differential effects on immune cell subsets depending on the disease context:

Cell TypeCancer ContextSepsis/Inflammation ContextTransplantation Context
CD8+ T CellsEnhanced cytotoxicity and IFN-γ production (27.1 ± 6.01% to 37.32 ± 7.82%, p<0.05) Potentially exacerbated inflammation May promote rejection
CD4+ T CellsPartial restoration of cytokine production Increased pro-inflammatory cytokines Increased allograft rejection
Natural Killer CellsEnhanced cytotoxicity against tumor cells Enhanced recruitment and activation Limited data available
Myeloid CellsLimited direct effectsIncreased recruitment of neutrophils, macrophages, and dendritic cells Limited data available

In cancer models like CLL, BTLA blockade enhances CD8+ T cell cytotoxicity against leukemic cells, particularly when combined with bispecific anti-CD3/anti-CD19 antibodies . In contrast, in sepsis models, anti-BTLA antibody treatment increased cytokine/chemokine production and inflammatory cell recruitment, exacerbating organ injury and increasing mortality .

In renal transplantation, BTLA plays a protective role by suppressing acute rejection. Overexpression of BTLA in rat models significantly inhibited acute rejection and prolonged allograft survival by suppressing IL-2 and IFN-γ production while increasing IL-4 and IL-10 production .

These diverse effects highlight the importance of understanding BTLA's specific role in different disease contexts when developing therapeutic strategies targeting this pathway .

What are potential combination approaches for targeting BTLA along with other immune checkpoints?

Optimizing combination approaches targeting BTLA and other immune checkpoints requires several strategic considerations:

  • Checkpoint Co-expression Analysis: Detailed profiling of BTLA co-expression with other immune checkpoints guides rational combination strategies. In HCC, BTLA+ cells represent a subset of PD-1+ CD4+ T cells with heightened dysfunction, suggesting potential benefits of dual targeting .

  • BTLA and PD-1 Dual Blockade: Studies suggest that dual BTLA/PD-1 blockade results in heightened IFN-γ levels and improved outcomes compared to monotherapies in some cancer models .

  • Integration with Targeted Therapies: In CLL, the combination of BTLA blockade with ibrutinib (a BTK inhibitor) significantly increased leukemic cell depletion without affecting T cell numbers, suggesting beneficial interaction between these approaches .

  • Biomarker-Guided Selection: Patients with high co-expression of BTLA and PD-1 on tumor-infiltrating lymphocytes might be more suitable candidates for dual blockade approaches .

  • Sequence Optimization: Determining whether sequential or simultaneous blockade of multiple checkpoints provides optimal efficacy is crucial, as priming with one checkpoint inhibitor may alter the expression or function of others.

Combination ApproachExperimental FindingsDisease Context
BTLA + PD-1 blockadeEnhanced IFN-γ production and improved survivalMurine glioblastoma models
BTLA blockade + bispecific antibodiesBoosted CD8+ T cell anti-leukemic activityChronic lymphocytic leukemia
BTLA blockade + BTK inhibitorsIncreased leukemic cell depletion without affecting T cell numbersChronic lymphocytic leukemia

By addressing these considerations, combination approaches targeting BTLA along with other immune checkpoints can potentially extend immunotherapy benefits to a broader range of patients .

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.