SLAMF6 Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

SLAMF6 antibodies modulate immune responses by altering receptor clustering, signaling, or interactions with other immune cells:

Blocking SLAMF6

  • Inhibits Homophilic Interactions: SLAMF6 forms “head-to-head” complexes via IgV domains . Antibodies prevent these interactions, disrupting T cell-B cell adhesion and cytotoxic synapse formation .

  • Enhances Antitumor Activity: In melanoma models, SLAMF6-blocking antibodies improved CD8+ T-cell polyfunctionality and tumor regression by reducing inhibitory signaling .

Enhancing SLAMF6 Signaling

  • Augments T Cell Activation: Bispecific antibodies (e.g., anti-CD3/SLAMF6) cluster SLAMF6 with TCR, promoting adhesion and cytotoxicity .

  • Antiviral Applications: SLAMF6 clustering enhances CD8+ T cell killing of HIV-1-infected targets, suggesting therapeutic potential for viral infections .

Tumor Models

ModelOutcomeSource
Melanoma (Pmel-1)SLAMF6 -/- T cells showed improved tumor eradication and LAG-3 upregulation, synergizing with LAG-3 blockade
CLL (TCL1-192)Anti-SLAMF6 + ibrutinib eliminated leukemic cells in spleen/bone marrow
Lymphoma (LMP2A/λMyc)Anti-SLAMF6 reduced tumor burden in SCID mice

Functional Insights

  • T Cell Exhaustion: Anti-SLAMF6 restored effector functions in exhausted CD8+ T cells, increasing IFN-γ production and degranulation .

  • Cytotoxic Synapse Formation: SLAMF6 antibodies disrupted CD8+ T cell-CD4+ T cell adhesion, critical for HIV-1-specific cytotoxicity .

  • Combination Therapies: Synergy observed with LAG-3 blockade (melanoma) and BTK inhibitors (CLL) .

Cancer Immunotherapy

  • SLAMF6 Expression in Tumors: High SLAMF6 correlates with favorable prognosis in breast cancer and melanoma, associated with TCF7 (T-bet) and effector T cell signatures .

  • Therapeutic Targeting: Anti-SLAMF6 antibodies may complement checkpoint inhibitors (e.g., anti-PD-1) by reinvigorating exhausted T cells .

Viral Infections

  • HIV-1: SLAMF6 downregulation by viral proteins (e.g., Vpu) impairs cytotoxic responses. Antibodies that enhance SLAMF6 clustering could restore antiviral immunity .

Challenges and Future Directions

  • Context-Dependent Effects: SLAMF6’s role varies between tumor microenvironments (inhibitory in cancer, activating in viral responses) .

  • Optimizing Antibody Design: Bispecific antibodies targeting SLAMF6 and co-receptors (e.g., CD3) may enhance specificity and efficacy .

  • Synergistic Combinations: Trials investigating anti-SLAMF6 with ibrutinib or LAG-3 inhibitors are warranted .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery time may vary depending on the purchase method and location. For specific delivery times, please consult your local distributors.
Synonyms
Activating NK receptor antibody; CD352 antibody; KALI antibody; KALIb antibody; LY108 antibody; MGC104953 antibody; Natural killer T and B cell antigen antibody; NK T B antigen antibody; NK-; T-; and B-cell antigen antibody; NK-T-B-antigen antibody; NTB A antibody; NTB-A antibody; NTBA antibody; NTBA receptor antibody; SF2000 antibody; SLAF6_HUMAN antibody; SLAM family member 6 antibody; SLAM family member 6 precursor antibody; SLAM Family Receptor antibody; Slamf6 antibody
Target Names
SLAMF6
Uniprot No.

Target Background

Function
SLAMF6, a self-ligand receptor belonging to the signaling lymphocytic activation molecule (SLAM) family, plays a pivotal role in modulating immune cell activation and differentiation. SLAM receptors, triggered by homo- or heterotypic cell-cell interactions, regulate both innate and adaptive immune responses. Their activities are influenced by the presence or absence of small cytoplasmic adapter proteins, SH2D1A/SAP and/or SH2D1B/EAT-2.

SLAMF6 specifically triggers cytolytic activity only in natural killer (NK) cells expressing high surface densities of natural cytotoxicity receptors. Positive signaling in NK cells involves phosphorylation of VAV1, with NK cell activation seemingly dependent on SH2D1B rather than SH2D1A.

In conjunction with SLAMF1, SLAMF6 controls the transition between positive selection and the subsequent expansion and differentiation of the thymocytic natural killer T (NKT) cell lineage.

SLAMF6 promotes T-cell differentiation into a helper T-cell Th17 phenotype, leading to increased IL-17 secretion. This costimulatory activity requires SH2D1A and involves the recruitment of RORC to the IL-17 promoter.

Together with SLAMF1 and CD84/SLAMF5, SLAMF6 may act as a negative regulator of the humoral immune response.

In the absence of SH2D1A/SAP, SLAMF6 can transmit negative signals to CD4(+) T-cells and NKT cells. It negatively regulates germinal center formation by inhibiting T-cell:B-cell adhesion. This function likely involves increased association with PTPN6/SHP-1 via ITSMs in the absence of SH2D1A/SAP. However, SLAMF6 has also been implicated in maintaining B-cell tolerance in germinal centers and preventing autoimmunity.
Gene References Into Functions
  1. Research has shown that the NTB-A/SAP pathway regulates T-cell activation and restimulation-induced cell death during human tuberculosis. PMID: 28546549
  2. Beyond its established role in Invariant NKT(iNKT) cell ontogeny, Ly108 regulates iNKT cell function in both mice and humans. PMID: 28373584
  3. In addition to their role in NK cell activation by hematopoietic cells, the SLAM-SAP-SHP1 pathways influence responsiveness toward nonhematopoietic targets, employing a mechanism similar to NK cell 'education'. PMID: 26878112
  4. Our findings reveal how SAP nucleates a previously unknown signaling complex involving NTB-A and LCK, amplifying restimulation-induced cell death of activated human T cells. PMID: 24688028
  5. Our data suggest that the reduction of NTB-A from the cell surface is associated with the Vpu-mediated effect on the glycosylation pattern of newly synthesized NTB-A molecules. PMID: 23528733
  6. Data indicate that the dominance of the SLAMF3/SLAMF6 pathway in inducing IL-17A production can be attributed to an increased nuclear abundance and recruitment of RORgammat to the IL17A promoter. PMID: 22989874
  7. SLAMF3 and SLAMF6 T cell surface expression and IL-17 levels significantly correlate with disease activity in systemic lupus erythematosus patients. PMID: 22184727
  8. While the expression of SLAMF6 on the surface of T cells from patients with systemic lupus erythematosus (SLE) T cells is comparable to that on normal T cells, engagement of SLAMF6 results in significantly reduced Th1 and IL-2 cytokine production. PMID: 21231893
  9. Vpu downmodulation of NTB-A protects the infected cell from lysis by NK cells. PMID: 21075351
  10. Regulation of interferon-gamma secretion, and not interleukin-4 in vitro, as well as inhibition of Th1 cell-induced isotype switching and attenuation of experimental allergic encephalomyelitis identifies NTB-A as a regulator of T cell response. PMID: 14988414
  11. NTB-A is an interlymphocyte signaling molecule, which serves to orchestrate the activities of immune cells. PMID: 15153464
  12. NTB-A-mediated IFN-gamma production was greatly reduced in the absence of SLAM-associated protein (SAP), demonstrating that cytokine production and cytotoxicity are differentially dependent on SAP and possibly EAT-2. PMID: 16920955
  13. The 3.0 A crystal structure of the complete NTB-A ectodomain revealed a rod-like monomer that self-associates to form a highly kinked dimer spanning an end-to-end distance of approximately 100 A. PMID: 17045824

Show More

Hide All

Database Links

HGNC: 21392

OMIM: 606446

KEGG: hsa:114836

STRING: 9606.ENSP00000357036

UniGene: Hs.492348

Subcellular Location
Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed by all (resting and activated) natural killer cells (NK), T- and B-lymphocytes. Increased surface expression on T-cells of systemic lupus erythematosus (SLE) patients.

Q&A

What is SLAMF6 and what signaling pathways does it regulate in immune cells?

SLAMF6 (Signaling Lymphocytic Activation Molecule Family member 6) is a homotypic receptor of the Ig-superfamily expressed on hematopoietic cells. It functions as an important regulator of T cell activation, with both its ectodomain and endodomain contributing differentially to T cell functions .

Key signaling pathways affected by SLAMF6:

  • SLAMF6 enhances T cell function by increasing T cell adhesiveness through activation of the small GTPase Rap1

  • The receptor depends on its association with SAP and other SH2 domain-containing proteins for signaling

  • Tyrosine 308 in the cytoplasmic domain is crucial for SLAMF6's ability to enhance T cell function, as it serves as a docking site for SAP association

Research demonstrates that SLAMF6 can function as either a stimulatory or inhibitory co-receptor depending on its localization and interaction with other receptors in the immunological synapse .

What methods are available for detecting and analyzing SLAMF6 expression in experimental settings?

Detection methods for SLAMF6:

MethodApplicationTypical DilutionNotes
Western BlottingProtein expression1:1000Detects 55-70 kDa band
ImmunoprecipitationProtein interactions1:100Useful for co-IP studies
Flow Cytometry (Live)Surface expression1:50 - 1:200Enables quantification on intact cells
Flow Cytometry (Fixed)Total expression1:50 - 1:200Allows intracellular detection
ImmunofluorescenceLocalization studies1:200 - 1:800Visualizes receptor distribution

When analyzing SLAMF6 expression, researchers should be aware that this receptor is constitutively expressed on T cells regardless of their activation status, which distinguishes it from typical exhaustion markers . For knockout validation, flow cytometry remains the gold standard method .

How can SLAMF6 knockout or knockdown models be generated for functional studies?

Several approaches have been successfully used to modulate SLAMF6 expression:

CRISPR-Cas9 knockout:

  • SLAMF6 can be knocked out in Jurkat T cells using lentiCRISPR v2 plasmids containing guide RNAs

  • Dual transduction with different guide RNA constructs improves knockout efficiency

  • Selection with puromycin followed by flow cytometry validation is recommended

shRNA knockdown:

  • Stable knockdown can be achieved using Mission shRNA plasmids

  • Lentiviral particles generated by transfecting HEK293T cells with pMD2G, psPAX2, and shRNA plasmid

  • Transduction by centrifugation followed by puromycin selection

For in vivo studies, breeding strategies have successfully generated SLAMF6-/- mice, which can be further crossed with TCR transgenic mice (e.g., Pmel-1) to create antigen-specific T cells lacking SLAMF6 .

What antibody formats are available for SLAMF6 research applications?

Available SLAMF6 antibody formats:

FormatApplicationAdvantages
Monoclonal antibodiesBlocking/stimulationHigh specificity, consistent results
Fab fragmentsFunctional studiesNo Fc-mediated effects, allows separation of clustering effects
Bispecific antibodiesEnhanced T cell activationForces co-localization with CD3, increases T cell function
Recombinant antibodiesMultiple applicationsSuperior lot-to-lot consistency, continuous supply

Over 544 commercially available SLAMF6 antibodies exist across different suppliers, with applications ranging from fundamental research to therapeutic development . When selecting an antibody, consider species cross-reactivity, as many antibodies are species-specific (particularly mouse vs. human) .

How does SLAMF6 compartmentalization affect T cell activation and function?

SLAMF6 compartmentalization represents a critical regulatory mechanism for T cell function. Research demonstrates that:

  • Co-localization with CD3: When SLAMF6 clusters with the TCR complex in the immunological synapse, it provides strong synergistic activation of T cells

  • Segregation from CD3: Physically separating SLAMF6 from CD3 reduces its co-stimulatory function and can even result in inhibitory effects

Experimental evidence:

  • T cells stimulated with beads conjugated with both anti-CD3 and anti-SLAMF6 antibodies show enhanced IL-2 production compared to anti-CD3 alone

  • In contrast, stimulation with separate anti-CD3 and anti-SLAMF6 beads fails to augment T cell activity

  • Similar results were observed using antibody cross-linking experiments, where clustered SLAMF6 and CD3 enhanced T cell function, while separated receptors did not

This spatial regulation mechanism suggests that targeted modulation of SLAMF6 localization, such as through bispecific antibodies, represents a promising therapeutic approach .

What are the contradictory findings regarding SLAMF6's role in T cell exhaustion and tumor immunity?

The literature reveals interesting contradictions regarding SLAMF6's role in T cell exhaustion and anti-tumor immunity:

Evidence for inhibitory role:

  • SLAMF6-deficient T cells show improved polyfunctionality and stronger tumor cytolysis

  • Adoptive transfer of SLAMF6-/- T cells to melanoma-bearing mice results in lasting tumor regression compared to temporary responses with wild-type T cells

  • Blocking SLAMF6 using antibodies can correct T cell dysfunction in exhausted T cells

Evidence for stimulatory role:

  • Anti-SLAMF6 antibodies can augment TCR-mediated responses in primary T cells

  • High expression of SLAMF6 in tumors correlates with better patient survival and elevated immune activity in breast cancer and melanoma

  • SLAMF6 enhances T cell function by increasing T cell adhesiveness through Rap1 activation

These contradictions might be explained by:

  • Context-dependent effects based on receptor compartmentalization

  • Different experimental systems and timepoints examined

  • Compensatory mechanisms (e.g., upregulation of LAG-3 in SLAMF6-/- T cells)

How can bispecific antibodies targeting SLAMF6 and CD3 be designed and validated?

Designing bispecific antibodies targeting SLAMF6 and CD3 involves several critical steps:

Design and production:

  • Bispecific antibodies can be generated using "knobs-into-holes" technology

  • Co-expression of monovalent OKT3-IgG-hole (anti-CD3) and monovalent anti-SLAMF6-IgG-knob constructs in 293 cells

  • Purification typically involves protein A chromatography followed by size exclusion chromatography

Validation methods:

  • ELISA validation:

    • Use SLAMF6-expressing cell lysate and SLAMF6 KO cell lysate as immobilized antigens

    • Confirm binding specificity to both targets

  • Functional validation:

    • Compare T cell activation (IL-2, IFN-γ secretion) between bispecific and individual antibodies

    • Assess proliferation using CFSE dilution assays

    • Evaluate cytotoxicity against target cells in co-culture systems

Experimental considerations:

  • Include appropriate controls (monospecific antibodies, isotype controls)

  • Test different concentrations to establish dose-response relationships

  • Compare soluble vs. immobilized antibody formats

Bispecific antibodies targeting SLAMF6 and CD3 have shown enhanced T cell activation compared to individual antibodies or their combinations, highlighting their potential therapeutic value .

What molecular mechanisms underlie SLAMF6-mediated enhancement of T cell function?

SLAMF6 augments T cell function through several molecular mechanisms:

1. Signaling through tyrosine phosphorylation:

  • The cytoplasmic tail of SLAMF6 contains immunoreceptor tyrosine-based switch motifs (ITSMs)

  • Tyrosine 308 is particularly crucial for SLAMF6's ability to enhance T cell function

  • Mutation of Y308F completely abolishes the effects of SLAMF6 engagement

2. Adaptor protein recruitment:

  • SLAMF6 associates with SAP (SLAM-associated protein) through its second ITSM centered around tyrosine 308

  • This association is critical for downstream signaling events

  • In the absence of functional SAP, SLAMF6 may act primarily as an inhibitory receptor

3. Regulation of T cell adhesion:

  • SLAMF6 activates the small GTPase Rap1, a master regulator of T cell adhesion

  • This activation increases integrin-mediated adhesion

  • Enhanced adhesion strengthens immunological synapse formation and TCR signaling

4. Transcriptional programming:

  • SLAMF6-deficient T cells show increased T-bet expression, driving an effector-memory phenotype

  • T-bet-mediated transcriptional events contribute to the enhanced cytotoxicity of SLAMF6-/- T cells

Understanding these mechanisms provides insights for rational design of SLAMF6-targeted immunotherapies.

How does SLAMF6 expression in the tumor microenvironment correlate with clinical outcomes?

Recent analyses reveal important correlations between SLAMF6 expression in the tumor microenvironment and clinical outcomes:

  • High SLAMF6 expression in tumors correlates with better patient survival in breast cancer and melanoma

  • SLAMF6 expression is associated with increased immune activity in the tumor microenvironment

  • Single-cell profiling demonstrates that SLAMF6 is exclusively expressed in immune cells (T cells, NK cells, and B cells) but not in cancerous cells within the tumor microenvironment

Mechanistic insights:

  • High SLAMF6 expression is associated with expression of TCF7 (encoding T-cell factor 1)

  • SLAMF6 correlates with increased gene signatures of anti-tumor immunity

  • The favorable prognostic impact may be linked to SLAMF6's role in enhancing T cell function when properly localized with TCR signaling complexes

These findings suggest that SLAMF6 expression could serve as a biomarker for predicting response to immunotherapy, although further validation in clinical settings is needed .

What are the key considerations for experimental design when studying SLAMF6 function?

When designing experiments to study SLAMF6 function, researchers should consider:

1. Receptor clustering and localization:

  • The functional outcome of SLAMF6 engagement depends on its proximity to CD3/TCR

  • Compare clustered vs. segregated experimental designs (e.g., co-immobilized antibodies vs. soluble antibodies)

  • Consider using imaging techniques to visualize receptor compartmentalization

2. Antibody format selection:

  • Full antibodies vs. Fab fragments (different clustering effects)

  • Soluble vs. immobilized antibodies (different compartmentalization)

  • Bispecific antibodies (forced co-localization with CD3)

3. Time-course considerations:

  • Short-term vs. long-term activation (different functional outcomes)

  • SLAMF6 effects may differ between acute activation and chronic stimulation/exhaustion models

4. Functional readouts:

  • Cytokine production (IL-2, IFN-γ)

  • Proliferation (CFSE dilution, cell counting)

  • Cytotoxicity assays against relevant targets

  • Signaling studies (phosphorylation of ERK, ZAP70, SRC)

5. Control experiments:

  • Include SLAMF6-deficient cells as controls

  • Consider compensatory mechanisms (e.g., LAG-3 upregulation)

  • Include appropriate isotype controls for antibody experiments

Careful attention to these factors will help generate more consistent and interpretable data regarding SLAMF6 function.

How can SLAMF6-targeted approaches be optimized for cancer immunotherapy?

Several SLAMF6-targeted approaches show promise for cancer immunotherapy:

1. Anti-SLAMF6 antibodies:

  • Direct anti-SLAMF6 antibodies can reduce leukemic burden in CLL models and suppress B16 melanoma growth

  • Mechanism involves correction of CD8+ T-cell dysfunction and direct effects on tumor progression

  • Optimization requires careful selection of epitopes and antibody isotypes

2. SLAMF6-deficient adoptive T cell therapy:

  • SLAMF6-/- T cells show enhanced anti-tumor activity in melanoma models

  • Combined with LAG-3 blockade, SLAMF6-deficient T cells induced faster and more complete tumor regression

  • Can be generated through CRISPR-Cas9 editing of patient-derived T cells

3. Bispecific antibodies:

  • Anti-CD3/SLAMF6 bispecific antibodies enhance T cell activation by forcing co-localization

  • Optimization involves determining optimal binding affinities for each arm

  • Format selection (e.g., diabody, tandem scFv, IgG-like) affects pharmacokinetics and efficacy

4. Combination therapies:

  • SLAMF6 blockade synergizes with LAG-3 inhibition

  • Could be combined with other checkpoint inhibitors (PD-1, CTLA-4)

  • Biomarker-guided patient selection based on SLAMF6 expression patterns may improve outcomes

Each approach requires careful optimization of dosing, schedule, and combination strategies to maximize therapeutic efficacy while minimizing toxicity.

What are the limitations and challenges in using anti-SLAMF6 antibodies for research and therapy?

Despite promising results, several challenges exist in developing anti-SLAMF6 approaches:

Technical challenges:

  • Variability in antibody specificity and functional effects across different clones

  • Difficulty in translating murine findings to human systems due to species differences

  • Limited understanding of how antibody epitope selection affects functional outcomes

Biological complexities:

  • Dual role of SLAMF6 as both stimulatory and inhibitory receptor depending on context

  • Compensatory upregulation of other immune checkpoints (e.g., LAG-3) when SLAMF6 is blocked

  • Potential off-target effects due to SLAMF6 expression on multiple immune cell types

Therapeutic considerations:

  • Optimal dosing and scheduling remain unclear

  • Patient selection strategies need refinement

  • Potential for adverse immune-related effects due to broad expression pattern

  • Competition with established checkpoint inhibitors in the clinical landscape

Future directions:

  • Development of humanized antibodies for clinical translation

  • Better understanding of predictive biomarkers for response

  • Optimization of combination strategies

  • Exploration of alternative formats (e.g., antibody-drug conjugates, CAR-T cells with SLAMF6-targeting domains)

Addressing these challenges will be critical for successful translation of SLAMF6-targeted approaches to the clinic.

How do mutations in SLAMF6 tyrosine residues affect its signaling capabilities?

SLAMF6 contains immunoreceptor tyrosine-based switch motifs (ITSMs) in its cytoplasmic domain that are critical for signaling:

Key tyrosine residues:

  • Tyrosine 308 is crucial for SLAMF6's ability to enhance T cell function

  • Tyrosine 284 is another potential phosphorylation site

Mutational studies reveal:

  • Y308F mutation: Completely abolishes the effects of SLAMF6 engagement on TCR activation

  • Y284F mutation: Has less dramatic effects compared to Y308F mutation

Mechanistic impact:

  • Y308 serves as a docking site for SAP (SLAM-associated protein)

  • SAP associates preferentially with ITSMs that contain the homology to TIYxxV/I/L/T

  • Mutation prevents this association, disrupting downstream signaling

  • Loss of Y308 phosphorylation affects Rap1 activation and subsequent integrin-mediated adhesion

These findings highlight the importance of site-specific phosphorylation in determining SLAMF6 function and provide targets for developing more selective therapeutic approaches.

What is known about SLAMF6 homotypic interactions and their structural basis?

SLAMF6 is a homotypic receptor, meaning it primarily binds to identical molecules on opposing cells:

Structural characteristics:

  • SLAMF6 belongs to the immunoglobulin superfamily with an extracellular portion containing Ig-like domains

  • The ectodomain is required for its function, but interestingly, not for its recruitment to the immunological synapse

  • Homotypic binding occurs through interaction between the N-terminal IgV domains

Functional implications:

  • Homotypic interactions between SLAMF6 molecules on interacting cells trigger signaling

  • This homotypic nature allows SLAMF6 to mediate interactions between similar immune cells (e.g., T-T, B-B, or NK-NK cell interactions)

  • Such interactions contribute to immune cell communication and regulation

Research approaches:

  • Soluble SLAMF6 ectodomains can be used to disrupt homotypic interactions

  • ΔSLAMF6-GFP constructs (lacking the ectodomain) help dissect the role of homotypic binding

  • Crystal structures of SLAMF family members provide insights into binding interfaces

Understanding the structural basis of SLAMF6 homotypic interactions could facilitate the development of novel therapeutics that either block or enhance these interactions for immunomodulatory purposes.

What emerging technologies could advance our understanding of SLAMF6 biology?

Several cutting-edge technologies offer promising approaches to deepen our understanding of SLAMF6 biology:

1. Advanced imaging techniques:

  • Super-resolution microscopy to visualize SLAMF6 nanoclusters and their dynamic rearrangement during T cell activation

  • Live-cell imaging with fluorescent reporters to track SLAMF6 recruitment to the immunological synapse in real-time

  • Proximity labeling approaches (BioID, APEX) to map SLAMF6 protein interaction networks

2. Single-cell technologies:

  • Single-cell RNA-seq to identify transcriptional signatures associated with SLAMF6 expression across immune cell subsets

  • CyTOF/spectral cytometry for high-dimensional analysis of SLAMF6 expression and co-expression with other receptors

  • Single-cell ATAC-seq to map changes in chromatin accessibility in SLAMF6-deficient cells

3. CRISPR-based approaches:

  • CRISPR activation/repression systems to modulate SLAMF6 expression

  • CRISPR screens to identify modulators of SLAMF6 signaling

  • Base editing to introduce specific mutations in endogenous SLAMF6

4. Protein engineering:

  • Novel bispecific formats beyond traditional antibodies

  • Optogenetic control of SLAMF6 clustering to dissect spatial requirements

  • Engineered receptors with modified signaling domains to selectively activate specific pathways

These technologies would significantly advance our understanding of SLAMF6 biology and accelerate the development of therapeutic applications .

How might understanding SLAMF6 signaling inform the development of next-generation immunotherapies?

Insights into SLAMF6 signaling pathways provide several opportunities for developing innovative immunotherapies:

1. Enhanced adoptive cell therapies:

  • Engineering CAR-T cells with SLAMF6 mutations or deletions to enhance anti-tumor activity

  • Developing ex-vivo expansion protocols using anti-SLAMF6 antibodies to generate more potent T cells

  • Creating SLAMF6-deficient NK cells with enhanced cytotoxic potential

2. Novel antibody-based approaches:

  • Next-generation bispecific antibodies targeting SLAMF6 and tumor antigens

  • Antibody-drug conjugates exploiting SLAMF6 expression on malignant B cells

  • Conditional activation systems where SLAMF6 signaling is modulated only in specific tissue contexts

3. Combinatorial immunotherapy strategies:

  • Rational combinations with existing checkpoint inhibitors based on mechanistic understanding

  • Sequential therapy approaches targeting different phases of T cell activation

  • Personalized treatment selection based on SLAMF6 expression patterns in individual patients

4. Biomarker development:

  • SLAMF6 expression as a predictive biomarker for immunotherapy response

  • Monitoring changes in SLAMF6+ T cell populations during treatment

  • Developing assays to assess SLAMF6 compartmentalization in patient samples

These approaches leverage our understanding of SLAMF6 biology to create more effective and precisely targeted immunotherapies with the potential to overcome resistance to current treatments .

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.