LY6G6D Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism of Action

LY6G6D is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein belonging to the LY-6 family of immune-related antigens. It is selectively expressed in CRC, particularly in MSS subtypes, which account for ~85% of CRC cases and show limited responsiveness to checkpoint inhibitors like anti-PD-1 antibodies .

The LY6G6D Recombinant Monoclonal Antibody functions as a bispecific T-cell engager (TcE) or antibody, binding simultaneously to the LY6G6D antigen on tumor cells and the CD3 receptor on T-cells. This dual binding activates cytotoxic T-cells, triggering tumor cell lysis via perforin/granzyme release and cytokine-mediated bystander effects .

Diagnostic and Analytical Uses

  • Flow Cytometry: Detects LY6G6D expression on live CRC cells .

  • Immunohistochemistry: Quantifies LY6G6D prevalence in tumor samples (e.g., 27% positive in a 41-sample cohort) .

  • ELISA: Validates antibody specificity and binding affinity .

Therapeutic Efficacy

In Vitro Studies:

  • Tumor Cell Lysis: LY6G6D/CD3 TcE induces dose-dependent killing of LY6G6D-positive cells (e.g., LS-1034, SK-CO1) .

  • T-cell Activation: Triggers IFNγ and TNFα secretion, enhancing immune infiltration .

In Vivo Models:

  • Tumor Regression: Monotherapy with LY6G6D/CD3 TcE achieves 90% tumor growth inhibition in NSG mice bearing human CRC xenografts .

  • Bystander Killing: Lytic effects extend to LY6G6D-negative cells via cytokine-mediated immune activation .

Key Discoveries

  1. Tumor Selectivity: LY6G6D is absent in normal colon tissues but expressed in 27% of CRC samples, primarily MSS subtypes .

  2. Combination Therapy: LY6G6D-TDB (a bispecific antibody) enhances efficacy when paired with anti-PD-1 blockade .

  3. Mechanistic Insights:

    • Direct Killing: T-cell-mediated cytolysis via CD3 cross-linking .

    • Indirect Effects: IFNγ and TNFα recruit immune cells to the tumor microenvironment .

Challenges and Limitations

  • Heterogeneity: Variable LY6G6D expression within tumors may reduce therapeutic response .

  • Preclinical Focus: Most data derive from murine models; human trials are pending .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The LY6G6D recombinant monoclonal antibody is produced through a four-step process: 1) Immunization and B cell harvesting: Recombinant human LY6G6D protein is used as an immunogen to elicit an immune response, followed by the harvesting of B cells. 2) Gene cloning: Total RNA is extracted from harvested B cells and reverse transcribed into cDNA. LY6G6D antibody genes are then amplified via PCR using primers specific to the antibody constant regions and cloned into an expression vector. 3) Recombinant antibody expression and purification: The expression vector is transfected into host cells for antibody production. The cell culture supernatant is subsequently collected, and the LY6G6D recombinant monoclonal antibody is purified using affinity chromatography. 4) Antibody characterization and validation: The purified antibody has been validated for its ability to recognize and bind human and Macaca fascicularis LY6G6D protein, as demonstrated by ELISA and flow cytometry (FC).

Form
Liquid
Lead Time
Order fulfillment typically takes 1-3 business days. Shipping times may vary depending on the order method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
Lymphocyte antigen 6 complex locus protein G6d; Protein Ly6-D; Megakaryocyte-enhanced gene transcript 1 protein; LY6G6D; C6orf23; G6D; MEGT1; NG25
Target Names
Uniprot No.

Target Background

Gene References Into Functions
  1. Analysis of DRB1, DQA1, DQB1, and TNFA allele frequencies in a West Siberian immigrant population. PMID: 19807019
Database Links

HGNC: 13935

OMIM: 606038

KEGG: hsa:58530

STRING: 9606.ENSP00000364985

UniGene: Hs.591792

Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor. Cell projection, filopodium.
Tissue Specificity
Expressed in the adult lung, and in fetal liver, lung, kidney, brain and spleen.

Q&A

What is LY6G6D and where is it expressed in normal and cancer tissues?

LY6G6D belongs to the Lymphocyte Antigen 6 family and is located in the major histocompatibility complex (MHC) class III region on chromosome 6. It is a GPI-anchored membrane protein whose normal function remains largely unknown, though it has been implicated in regulating tumor growth and immune evasion in colorectal cancer (CRC) . Transcriptomic analysis across multiple datasets (TCGA and GTEx) shows that LY6G6D expression is highly selective for CRC tissues, with minimal expression in normal tissues. Importantly, LY6G6D shows preferential expression in microsatellite stable (MSS) CRC, which represents approximately 85% of CRC cases and typically responds poorly to existing immunotherapies like PD-1 inhibitors .

Immunohistochemistry studies have confirmed that LY6G6D protein is not detected in normal colorectal tissues but is present in approximately 27% of CRC samples, making it a relatively selective tumor antigen with therapeutic potential .

How is LY6G6D expression detected in tumor samples?

Detection of LY6G6D in tumor samples primarily utilizes immunohistochemistry (IHC) with specific anti-LY6G6D antibodies. In the referenced study, researchers developed monoclonal antibody clone 10C1 through mouse immunization with recombinant LY6G6D protein . For IHC protocol:

  • Cut formalin-fixed paraffin-embedded (FFPE) tissue samples into 3μm sections

  • Mount sections on glass slides

  • Stain with anti-LY6G6D clone 10C1 antibody or isotype control

  • Develop using OptiView DAB IHC Detection Kit (Roche Diagnostics)

  • Evaluate by pathologist for:

    • Percentage of stained tumor cells

    • Staining intensity

    • Samples with ≥1% LY6G6D expressing tumor cells are scored as positive

For detection in cell lines and living cells, flow cytometry using antibodies that recognize native LY6G6D (such as clone 2C11A8) is more appropriate than antibodies selected for IHC applications .

What validation methods confirm the specificity of anti-LY6G6D antibodies?

Validating the specificity of anti-LY6G6D antibodies is crucial for research applications. Multiple complementary approaches should be employed:

  • Blocking studies: Specificity of antibody binding can be confirmed by pre-incubation with excess recombinant LY6G6D protein. In the referenced study, staining with clone 10C1 was blocked with 50x recombinant LY6G6D protein, confirming its specificity .

  • Cell line validation: Test antibody binding on cell lines with confirmed positive or negative LY6G6D mRNA expression. The researchers validated antibody binding using both mRNA-positive CRC cell lines (HT55, LS1034, CL-14, NCI-H508) and mRNA-negative lines (SK-CO1, NCM-460) .

  • GPI-anchor verification: Since LY6G6D is GPI-anchored, treatment with phosphatidylinositol-specific phospholipase C (PI-PLC) should reduce antibody binding by cleaving the protein from the cell surface. Researchers observed dose-dependent reduction in LY6G6D detection following PI-PLC treatment, though complete removal was not achieved even at high enzyme concentrations .

  • Functional validation: For antibodies intended for therapeutic applications, functional assays (such as T cell activation assays for TcE constructs) can confirm that antibody binding produces the expected biological effects .

What methods are used to generate and characterize LY6G6D-specific monoclonal antibodies?

Generating high-quality LY6G6D-specific monoclonal antibodies involves several key steps:

  • Immunization strategy: Mice are immunized with human-IgG1-Fc-His6 fusion protein derived from the mature human LY6G6D sequence (amino acids 20-103) .

  • Hybridoma generation:

    • Monitor serum titers by ELISA using His6 fusion protein

    • Pool splenocytes from responsive mice

    • Immortalize cells by PEG-based fusion

    • Select positive clones by ELISA

    • Generate monoclonal hybridoma cells by limited dilution

    • Confirm binding with purified IgG from hybridoma supernatant

  • Application-specific screening:

    • For IHC antibodies: Screen by staining formalin-fixed paraffin-embedded mRNA positive/negative CRC cell lines (resulted in selection of clone 10C1)

    • For TcE applications: Screen by flow cytometry on unfixed, mRNA positive/negative CRC cell lines to ensure recognition of native LY6G6D (resulted in selection of clone 2C11A8)

  • Sequence determination: For antibodies selected for further development, determine nucleotide sequences through V-gene recovery .

  • Affinity analysis: Characterize binding kinetics using surface plasmon resonance (SPR). In the study, researchers used BIAcore T200 device with TcE immobilized on CM5 Chip and recombinant LY6G6D protein injected at concentrations of 0-9000 nM in HBS-EP buffer. Data was analyzed according to the 1:1 Langmuir model .

How can researchers design and optimize T cell engagers (TcE) targeting LY6G6D?

Designing effective LY6G6D-targeting T cell engagers requires careful optimization of multiple parameters:

  • Antibody selection: Choose antibodies that recognize native LY6G6D on the cell surface. For the referenced TcE, researchers selected clone 2C11A8 based on flow cytometry screening against LY6G6D-positive and negative cell lines .

  • Format optimization: The bispecific antibody format must maintain binding to both LY6G6D and CD3 with appropriate affinities. The study used a TcE format that enables simultaneous binding to tumor antigen and CD3 on T cells .

  • Functional testing hierarchy:

    • Initial screening: Use reporter cell lines (e.g., Jurkat WT-NFAT luciferase reporter cells) to assess T cell activation upon target engagement

    • Secondary validation: Test with primary human T cells and measure activation markers (CD25, CD69), proliferation, and cytotoxic markers (CD107, Perforin, Granzyme B)

    • Tertiary validation: Assess target cell killing through LDH release assays with EC50 determination

  • Target density consideration: The efficacy of LY6G6D/CD3 TcE correlates with LY6G6D density on target cells. This can be experimentally modulated using PI-PLC treatment to partially remove LY6G6D from the cell surface, allowing for determination of the minimum target density required for effective T cell activation .

  • Heterogeneity modeling: Since LY6G6D expression in tumors is heterogeneous, testing the TcE in co-cultures of LY6G6D-positive and negative cells provides important insights into potential bystander killing effects in mixed tumor populations .

What experimental models best assess efficacy of LY6G6D-targeting therapeutics?

To comprehensively evaluate LY6G6D-targeting therapeutics, researchers should consider a spectrum of models with increasing complexity:

  • In vitro cell line models:

    • Monocultures of LY6G6D-positive CRC cell lines (HT55, LS1034, CL-14, NCI-H508)

    • Negative control cell lines (SK-CO1, NCM-460)

    • Mixed co-cultures with defined ratios of positive/negative cells to model tumoral heterogeneity

  • 2D and 3D co-culture systems:

    • Standard 2D co-cultures for initial assessments

    • 3D co-cultures for more physiologically relevant tumor architecture

    • Both models can be used to study bystander killing mechanisms

  • Ex vivo patient-derived models:

    • Tumor slice cultures from patient-derived CRC samples

    • These maintained tissue architecture and tumor microenvironment

    • Researchers observed IFNγ secretion in LY6G6D-positive tumor samples treated with the TcE

  • In vivo xenograft models:

    • Human CRC tumor cells engrafted in immunodeficient mice

    • Co-engraftment with human PBMCs to provide effector T cells

    • Allows assessment of tumor regression following LY6G6D/CD3 TcE treatment

    • The referenced study observed tumor regressions in these models with TcE monotherapy

Model TypeAdvantagesLimitationsKey Measurements
Cell line monoculturesSimple, reproducibleLacks heterogeneityT cell activation, cytotoxicity
Mixed co-culturesModels heterogeneityArtificial ratiosDirect vs. bystander killing
Ex vivo tumor slicesMaintains architectureShort viabilityCytokine production, T cell infiltration
In vivo xenograftsSystemic effectsIncomplete immune systemTumor regression, toxicity

What are the primary and secondary mechanisms of LY6G6D/CD3 TcE-mediated tumor killing?

LY6G6D/CD3 TcE mediates tumor cell killing through both direct and indirect mechanisms:

  • Direct killing mechanism:

    • TcE simultaneously binds LY6G6D on tumor cells and CD3 on T cells

    • Cross-linking of the T cell receptor (TCR) complex induces T cell activation

    • Activated T cells release perforin and granzyme B, leading to cytolysis of target-positive cells

    • This direct mechanism is highly potent, with EC50 values ranging from 0.1 to 1 nM in various LY6G6D-positive cell lines

  • Bystander killing mechanism:

    • When LY6G6D-positive and negative cells are co-cultured, TcE treatment also induces killing of nearby LY6G6D-negative cells

    • This effect is mediated by three soluble factors released by activated T cells:

      • Interferon-gamma (IFNγ)

      • Tumor necrosis factor-alpha (TNFα)

      • Fas/FasL interaction

    • Notably, these secondary mechanisms are dispensable for direct killing but crucial for eliminating heterogeneous tumors containing both target-positive and target-negative cells

  • Dose-dependence of bystander effect:

    • The extent of bystander killing correlates with the percentage of LY6G6D-positive cells present

    • When the proportion of target-positive cells decreased from 50% to 20%, researchers observed reduced killing of target-negative cells

This dual killing mechanism is particularly significant given the heterogeneous expression of LY6G6D observed in CRC tumors, where the percentage of positive cells ranged from 1-90% of tumor content .

How does LY6G6D density influence the efficacy of targeted therapies?

Research findings demonstrate a clear correlation between LY6G6D density on cell surfaces and the potency of LY6G6D/CD3 TcE:

  • Experimental approach:

    • Researchers used PI-PLC enzyme treatment to partially cleave the GPI anchor of LY6G6D

    • This created a gradient of LY6G6D densities on the cell surface

    • T cell activation was then measured using Jurkat WT-NFAT luciferase reporter cells

  • Key findings:

    • T cell activation, measured by luciferase signal, decreased proportionally with reduced LY6G6D density

    • Even at the highest PI-PLC concentration tested, some LY6G6D remained on the cell surface

    • This residual expression was sufficient to induce measurable T cell activation, though at reduced levels

  • Implications for research:

    • Researchers should determine the minimum LY6G6D density threshold required for clinical efficacy

    • Semiquantitative IHC scoring may help stratify patients for LY6G6D-targeted therapies

    • Combination therapies might be needed for tumors with low or heterogeneous LY6G6D expression

This density-efficacy relationship provides an important parameter for researchers to consider when designing experiments and interpreting results with LY6G6D-targeting agents.

What factors influence the bystander killing of LY6G6D-negative cells?

The bystander killing effect represents a crucial mechanism for addressing tumoral heterogeneity. Several factors influence this phenomenon:

  • Proportion of target-positive cells:

    • Higher percentages of LY6G6D-positive cells in mixed populations lead to more robust bystander killing

    • Experiments showed decreased killing of LY6G6D-negative cells when the proportion of positive cells was reduced from 50% to 20%

  • Molecular mediators:

    • Three key pathways mediate the bystander effect:

      • IFNγ signaling

      • TNFα signaling

      • Fas/FasL interaction

    • These factors act in combination rather than independently

  • Spatial proximity:

    • Bystander killing requires target-negative cells to be in the vicinity of target-positive cells

    • Both 2D and 3D co-culture models demonstrated this proximity requirement

    • This suggests that the soluble mediators and surface interactions have limited diffusion range

  • Cellular susceptibility:

    • Different cell types may exhibit varying sensitivity to these bystander mechanisms

    • Factors influencing susceptibility include expression levels of:

      • IFNγ and TNFα receptors

      • Fas expression

      • Downstream apoptotic pathway components

Understanding these factors allows researchers to optimize experimental designs for studying bystander effects and potentially enhance this mechanism therapeutically.

What cytotoxicity assays are recommended for evaluating LY6G6D-targeted therapeutics?

To comprehensively evaluate LY6G6D-targeted therapeutics, researchers should employ multiple complementary cytotoxicity assays:

  • LDH release assay:

    • Target cells are co-cultured with T cells and LY6G6D/CD3 TcE at various concentrations

    • After 48-72 hours, LDH release is quantified using commercial kits (e.g., Cytotoxicity detection kit-Plus, Roche)

    • Maximum cell lysis is determined by Triton X-100 treatment (100% lysis)

    • Minimum lysis established by target cells with effector cells alone (0% lysis)

    • Calculate specific cell lysis as: [sample LDH release - spontaneous LDH release]/[maximum LDH release – spontaneous LDH release] x 100

  • Flow cytometry-based cell counting:

    • Label different cell populations with distinct cell tracking dyes (e.g., CellTrace™, Invitrogen)

    • After treatment, quantify absolute numbers of live cells by flow cytometry

    • Include counting beads (e.g., AccuCheck beads, Invitrogen) for precise quantification

    • This approach is particularly valuable for mixed co-cultures with LY6G6D-positive and negative populations

  • T cell activation assessment:

    • Measure surface activation markers (CD25, CD69) by flow cytometry

    • Assess proliferation using cell tracking dyes or Ki-67 staining

    • Quantify cytotoxic effector molecules (CD107a, Perforin, Granzyme B)

    • These measurements provide mechanistic insights beyond simple cell death

  • Reporter cell assays:

    • For high-throughput screening, use Jurkat WT-NFAT luciferase reporter cells

    • Co-culture with target cells and LY6G6D/CD3 TcE

    • Measure luciferase signal as a surrogate for T cell activation

    • This approach is faster than primary T cell assays and offers good reproducibility

How should researchers account for LY6G6D heterogeneity in experimental design?

The heterogeneous expression of LY6G6D in tumors (1-90% positive cells) necessitates careful experimental design:

  • Establishing baseline expression:

    • Characterize LY6G6D expression in experimental models using flow cytometry and IHC

    • Quantify both percentage of positive cells and expression intensity

    • Compare with clinical samples to ensure model relevance

  • Creating defined heterogeneity models:

    • Mix LY6G6D-positive and negative cells at controlled ratios (e.g., 50:50, 20:80)

    • Label each population with different tracking dyes

    • This allows separate quantification of effects on each population

  • 3D culture systems:

    • Develop spheroid or organoid models with mixed cell populations

    • These better recapitulate spatial heterogeneity seen in tumors

    • Assess drug penetration and zone-dependent efficacy

  • Analyzing spatial relationships:

    • In tissue sections, map the distribution of LY6G6D-positive cells

    • Correlate with markers of immune infiltration and activation

    • This helps understand potential bystander effects in vivo

  • Dose-response considerations:

    • Test wider dose ranges than for homogeneous models

    • Determine minimum effective dose required for bystander killing

    • EC50 values will likely differ between direct and bystander killing mechanisms

By systematically addressing heterogeneity in experimental design, researchers can generate more clinically relevant data and better predict therapeutic outcomes.

What patient selection strategies should be considered for LY6G6D-targeted therapies?

Developing effective patient selection strategies for LY6G6D-targeted therapies requires consideration of several factors:

Selection ParameterMeasurementPotential ThresholdRationale
LY6G6D expressionIHC≥1% positive cellsMinimum for study classification
MSS statusPCR or IHC for MMR proteinspMMR/MSSEnriches for LY6G6D expression
T cell infiltrationCD3+ IHCPresence in tumor or marginRequired for TcE mechanism
Target heterogeneitySpatial analysis≥20% positive cellsMinimum for bystander effect

How might resistance to LY6G6D-targeted therapies develop and be addressed?

Although the referenced study doesn't directly address resistance mechanisms, researchers should consider potential resistance pathways based on the TcE mechanism of action:

  • Target downregulation or mutation:

    • Selective pressure may lead to downregulation of LY6G6D expression

    • Mutations affecting the epitope recognized by the antibody could emerge

    • Regular reassessment of LY6G6D expression during treatment may be necessary

  • Immune escape mechanisms:

    • Upregulation of immune checkpoints (PD-L1, TIM-3, LAG-3)

    • Recruitment of immunosuppressive cells (Tregs, MDSCs)

    • These might be addressed through combination with checkpoint inhibitors

  • Impaired bystander killing:

    • Downregulation of death receptors (Fas)

    • Resistance to IFNγ or TNFα signaling

    • Targeting multiple pathways simultaneously could overcome this resistance

  • Research strategies to address resistance:

    • Develop models of acquired resistance through chronic exposure

    • Test combinations with complementary immunotherapeutic approaches

    • Target multiple epitopes of LY6G6D simultaneously

    • Explore combinations with agents that enhance T cell function or infiltration

Understanding and addressing these potential resistance mechanisms will be crucial for maximizing the long-term efficacy of LY6G6D-targeted approaches.

What combination strategies might enhance LY6G6D-targeted immunotherapies?

Based on the mechanism of action and tumor biology, several combination strategies warrant investigation:

  • Immune checkpoint inhibitors:

    • LY6G6D/CD3 TcE induces T cell activation and cytokine release

    • This could upregulate immune checkpoints like PD-1/PD-L1

    • Combining with checkpoint inhibitors might prevent adaptive resistance

    • Particularly relevant for MSS CRC, which typically responds poorly to checkpoint inhibition alone

  • Agents enhancing bystander killing:

    • Fas-activating agents could amplify the bystander effect

    • TNFα or IFNγ pathway modulators might enhance these mechanisms

    • This approach could increase efficacy in tumors with heterogeneous LY6G6D expression

  • Stromal-targeting agents:

    • Dense stroma in CRC may impede T cell infiltration

    • Combinations with agents that normalize tumor vasculature or reduce fibrosis

    • This could improve TcE distribution and T cell access to tumor cells

  • Cytokine support:

    • IL-2, IL-15, or engineered cytokines that support T cell function

    • These could enhance T cell proliferation, survival, and cytotoxic capacity

    • Potentially important for patients with compromised T cell function

  • Conventional therapies:

    • Certain chemotherapies induce immunogenic cell death

    • Radiation can enhance antigen presentation and T cell infiltration

    • These conventional approaches might synergize with LY6G6D-targeted immunotherapy

Systematic investigation of these combinations, beginning with preclinical models and progressing to early-phase clinical trials, will be essential to maximize the therapeutic potential of LY6G6D-targeting approaches.

What advances in antibody engineering might improve LY6G6D-targeted therapeutics?

Future research should explore advanced antibody engineering approaches to enhance LY6G6D-targeted therapeutics:

  • Optimized TcE formats:

    • Varying the distance and flexibility between LY6G6D and CD3 binding domains

    • Testing different antibody isotypes or fragments (Fab, scFv, nanobodies)

    • Adjusting affinities for both targets to optimize T cell activation while minimizing systemic toxicity

  • Multi-specific antibodies:

    • Targeting LY6G6D along with additional tumor antigens to address heterogeneity

    • Incorporating immune checkpoint blockade into the same molecule

    • Adding specificity for components of the tumor microenvironment

  • Conditional activation:

    • Protease-activatable antibodies that become fully active only in the tumor microenvironment

    • pH-sensitive binding that preferentially occurs in the acidic tumor environment

    • These approaches could improve therapeutic window

  • Payload delivery:

    • Using LY6G6D antibodies to deliver cytotoxic payloads (ADCs)

    • Targeted delivery of immunomodulatory agents to the tumor microenvironment

    • Combined approaches leveraging both T cell engagement and direct cytotoxicity

  • Modified pharmacokinetics:

    • Engineering for optimal tissue penetration and residence time

    • Half-life extension or controlled release formulations

    • Balance between tumor exposure and systemic clearance to minimize toxicity

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.