LAG-3 is a 70-kDa transmembrane glycoprotein belonging to the immunoglobulin superfamily, sharing structural homology with CD4 . Its extracellular domain contains four Ig-like domains (D1-D4), with D1 mediating high-affinity binding to MHC class II molecules (Kd ≈ 1-10 nM) . Unlike CD4, LAG-3 exhibits immunosuppressive functions through:
Key therapeutic mechanisms include:
LAG-3 mAbs serve critical roles in immune monitoring:
| Antibody Clone | Fluorophore | Target Population | Source |
|---|---|---|---|
| 3DS223H | PE-Cy7 | Activated CD8+ TILs | |
| 17B4 | FITC | Th1 cells | |
| EPR20261 | - | Tregs |
Western blot validation shows specific detection at 57 kDa (predicted) and 90 kDa (glycosylated form) . Immunohistochemistry applications demonstrate 85% specificity in tumor stroma analysis .
Current limitations and emerging solutions:
Tumor Microenvironment Complexity
Biomarker Development
Next-Generation Formats
The LAG3 monoclonal antibody is produced through a rigorous process. This process involves immunizing mice with recombinant human LAG3 protein (amino acids 29-450), followed by the isolation of B cells from the mouse spleen. These B cells are then fused with myeloma cells to generate hybridomas. The hybridomas producing LAG3 antibodies are selected and cultured. The LAG3 monoclonal antibody is purified using protein G affinity chromatography from the mouse ascites, achieving a purity exceeding 95%. Its specificity has been validated through various applications, including ELISA, Western blot, immunohistochemistry, and flow cytometry.
LAG3 protein primarily regulates immune responses by inhibiting T-cell activation and promoting T-cell exhaustion. This regulation is achieved by binding to MHC class II molecules on antigen-presenting cells, leading to the suppression of T-cell activation and the promotion of immune tolerance. Notably, LAG3 protein has emerged as a promising immunotherapy target in cancer treatment due to its ability to modulate immune responses.
Lymphocyte activation gene 3 (LAG3) protein is an inhibitory receptor expressed on antigen-activated T cells. It delivers inhibitory signals upon binding to ligands such as FGL1. FGL1 is a major ligand of LAG3, responsible for LAG3's T-cell inhibitory function. Following T cell receptor (TCR) engagement, LAG3 associates with CD3-TCR in the immunological synapse, directly inhibiting T-cell activation.
LAG3 may synergistically inhibit antigen-specific T-cell activation alongside PDCD1/PD-1, potentially functioning as a coreceptor for PDCD1/PD-1. It negatively regulates the proliferation, activation, effector function, and homeostasis of both CD8(+) and CD4(+) T cells.
LAG3 also plays a crucial role in mediating immune tolerance. It is constitutively expressed on a subset of regulatory T cells (Tregs) and contributes to their suppressive function. Additionally, it acts as a negative regulator of plasmacytoid dendritic cell (pDCs) activation.
LAG3 binds to MHC class II (MHC-II); however, the precise role of MHC-II binding is unclear. It may function as a ligand for MHC class II on antigen-presenting cells (APCs), promoting APC activation/maturation and driving Th1 immune responses.
LAG-3 (CD223) is a cell surface inhibitory receptor belonging to the immunoglobulin superfamily that regulates immune homeostasis through multiple biological activities related to T-cell functions. Structurally similar to CD4, LAG-3 binds to MHC-II with considerably higher affinity, transmitting inhibitory signals through its cytoplasmic domain to down-regulate CD4+ T lymphocytes. This interaction negatively regulates T cell antigen-stimulated activation, proliferation, cytokine production, and cytotoxicity .
LAG-3 also interacts with the TCR complex on activated CD4 and CD8 T cells, downregulating TCR signaling. Beyond MHC-II, LAG-3 interacts with additional ligands including LSECtin, Galectin-3 (Gal-3), and Fibrinogen-like protein 1 (FGL1). The FGL1/LAG-3 interaction occurs via the LAG-3 D1 and D2 domains and the FGL1 fibrinogen-like domain in an MHC-II-independent manner .
LAG-3 is expressed on multiple immune cell types, primarily:
CD4+ T cells (especially Th1 cells, with weak or no expression on Th0 and Th2 clones)
CD8+ T cells
Regulatory T cells (Tregs)
Natural Killer (NK) cells
B cells
Dendritic cells
Cytokines regulate LAG-3 expression, with IL-2, IL-7, and IL-12 upregulating its expression. IL-12 provides the strongest stimulus for LAG-3 expression, and LAG-3 expression on activated CD4+ subsets correlates with higher intracellular interferon-gamma production .
LAG-3 expression varies across tumor types and can significantly impact patient prognosis:
These variable outcomes across cancer types highlight the context-dependent role of LAG-3 and the importance of considering tumor-specific microenvironments when designing therapeutic approaches.
LAG-3-targeting therapeutic agents can be classified into three main categories:
Anti-LAG-3 monoclonal antibodies: Single-specificity antibodies that directly target LAG-3, such as TSR-033, which is a high-affinity human IgG4 monoclonal antibody
Anti-LAG-3 bispecific antibodies: Dual-targeting antibodies that simultaneously engage LAG-3 and another immune checkpoint, such as MGD013 (tebotelimab), which is a humanized, hinge-stabilized, IgG4-κ tetravalent bispecific antibody targeting both PD-1 and LAG-3 with high affinity
Soluble LAG-3-Ig fusion proteins: Recombinant proteins consisting of the extracellular domain of LAG-3 fused to an immunoglobulin Fc region
The mechanism of action typically involves disrupting the interaction between LAG-3 and its ligands (primarily MHC-II, but also FGL1 and others), thereby preventing inhibitory signaling and enhancing T cell activation and function.
Anti-LAG-3 monoclonal antibodies function by:
Binding with high affinity to LAG-3 expressed on immune cells, particularly activated T cells
Blocking the interaction between LAG-3 and its ligands (MHC-II, FGL1, Galectin-3, LSECtin)
Preventing the transmission of inhibitory signals through LAG-3's cytoplasmic domain
Relieving LAG-3-mediated suppression of T cell activation, proliferation, and effector functions
Enhancing antitumor immune responses through increased T cell activity
By targeting LAG-3, these antibodies can restore T cell function in the tumor microenvironment, particularly in cases where T cells are dysfunctional or exhausted due to prolonged antigen exposure and inhibitory checkpoint expression.
Bispecific antibodies targeting LAG-3, such as MGD013 (tebotelimab), offer several advantages over conventional monoclonal antibodies:
Dual targeting capability: They simultaneously target LAG-3 and another immune checkpoint (typically PD-1), allowing for concurrent blockade of two inhibitory pathways with a single molecule
Enhanced specificity for dysfunctional T cells: They can specifically target PD-1+ LAG-3+ highly dysfunctional T cells, which represent a particularly suppressed subset of immune cells in the tumor microenvironment
Increased efficacy: PD-1/LAG-3 co-blockade has demonstrated increased cytokine secretion and enhanced T-cell responses compared to single blockade of either PD-1 or LAG-3 alone
Potential to overcome resistance: Bispecific antibodies may help address resistance to single-agent checkpoint inhibitors by targeting multiple inhibitory pathways simultaneously
MGD013, for example, has demonstrated favorable biophysical and manufacturability properties with a prolonged half-life, and preliminary clinical data show encouraging responses and acceptable pharmacokinetics .
For researchers studying LAG-3 expression in tissue samples, consider these methodological approaches:
Immunohistochemistry (IHC):
Use validated anti-LAG-3 antibodies such as clone 3DS223H
Include positive controls (activated T cells or known LAG-3+ tumors)
Quantify expression using digital pathology when possible
Always correlate with other immune markers (CD3, CD4, CD8) for context
Flow cytometry:
Fresh tissue samples should be processed rapidly to maintain cell viability
Use multiple fluorescent markers to identify LAG-3 expression on specific cell subsets
Include FMO (Fluorescence Minus One) controls to accurately determine positive populations
Consider analysis of both surface and intracellular LAG-3 where relevant
Multiplex immunofluorescence:
Enables simultaneous detection of LAG-3 with other immune checkpoint molecules
Provides spatial context for LAG-3 expression within the tumor microenvironment
Useful for analyzing co-expression patterns (LAG-3/PD-1/TIM-3/CTLA-4)
RNA analysis:
qRT-PCR and RNA sequencing provide mRNA expression levels
Important to validate transcriptional findings at protein level due to post-transcriptional regulation
When evaluating LAG-3 expression in clinical samples, it's critical to standardize processing times and preservation methods, as these can significantly affect detection sensitivity .
To evaluate the functional activity of anti-LAG-3 antibodies, researchers should employ multiple complementary assays:
Binding assays:
ELISA to determine binding affinity (KD) to recombinant LAG-3
Flow cytometry to confirm binding to native LAG-3 on cells
Surface plasmon resonance for detailed binding kinetics
Functional T cell assays:
Measure IL-2 and IFN-γ production in presence of anti-LAG-3 antibodies
Assess T cell proliferation using CFSE dilution or Ki-67 staining
Evaluate cytotoxic activity using chromium release or flow-based killing assays
Mixed lymphocyte reactions to assess impact on T cell activation
Competitive binding assays:
Determine if the antibody blocks LAG-3 interaction with MHC-II, FGL1, or other ligands
Compare activity to benchmark antibodies with known mechanisms
In vivo models:
Syngeneic tumor models to assess antitumor efficacy
Humanized mouse models for human-specific antibodies
Monitor changes in tumor-infiltrating lymphocyte (TIL) number and phenotype
Remember that antibody isotype significantly impacts functional outcomes, with IgG4 antibodies typically chosen for therapeutic applications due to reduced Fc-mediated effector functions .
When designing LAG-3 blockade experiments, researchers should consider:
Model selection:
Choose models with documented LAG-3 expression in the tumor microenvironment
Consider models resistant to PD-1 monotherapy to evaluate combination potential
For immunocompetent models, ensure species cross-reactivity of the antibody
Experimental controls:
Include isotype-matched control antibodies
Consider using LAG-3 knockout models as controls
Include PD-1 blockade arms for comparison and combination studies
Dosing and scheduling:
Determine optimal antibody concentration through dose-response studies
Consider different dosing schedules (concurrent vs. sequential for combinations)
Ensure adequate serum levels throughout the experimental period
Comprehensive immune monitoring:
Monitor multiple cell populations (CD4+ T cells, CD8+ T cells, Tregs, NK cells)
Assess changes in both peripheral blood and tumor-infiltrating immune cells
Evaluate systemic and local cytokine production
Potential confounding factors:
Combination therapies with LAG-3 monoclonal antibodies and other checkpoint inhibitors have shown promising results:
LAG-3 and PD-1 co-blockade:
Dual blockade of LAG-3 and PD-1 has demonstrated enhanced efficacy compared to single-agent therapy
In murine models, combination therapy showed strong anti-tumor effects in mice resistant to single antibody treatment without obvious evidence of autoimmunity
Clinical trials confirm these findings, with long-term disease control observed in patients with prior acquired resistance to PD-(L)1 therapy
LAG-3, PD-1, and CTLA-4 combinations:
Bispecific antibodies:
MGD013 (tebotelimab), a PD-1/LAG-3 bispecific antibody, has shown antitumor activity in multiple tumor types
One complete response was observed after single MGD013 administration in a patient who had received chimeric antigen receptor (CAR)-T-cell therapy
Combination of margetuximab (anti-HER2) with PD-1xLAG-3 DART® enhanced lytic activity of immune cells, with an objective response rate of 42.9% in relapsed or refractory HER2+ solid tumors
High baseline LAG-3/PD-1 expression and IFN-γ high gene signatures (CXCL9, CXCL10, CXC11, STAT1) were associated with objective clinical responses to combination therapy .
As of early 2025, the clinical trial landscape for LAG-3-targeting therapies is extensive:
Clinical trial volume and distribution:
97 clinical trials evaluating at least 16 LAG-3-targeting molecules
Two trials have reached phase III (sponsored by BMS and Merck)
Phase III studies in melanoma and colorectal cancer are demonstrating encouraging results
Most trials are testing LAG-3-antagonistic molecules, including combinations with other immune checkpoint inhibitors
Types of investigational agents:
Conventional anti-LAG-3 monoclonal antibodies
Next-generation bispecifics with dual targeting capabilities
Various combination strategies with other immunotherapeutic agents
Clinical outcomes:
LAG-3-targeting cancer immunotherapies have demonstrated good safety profiles and tolerability
Adequate pharmacokinetics and pharmacodynamics have been observed
Promising antitumor efficacy has been reported, particularly in combination settings
Simultaneous co-expression of LAG-3 with other immune checkpoint molecules characterizes highly dysfunctional T cells in cancer patients, making them prime targets for bispecific approaches
The co-blockade of LAG-3 with PD-1 shows particular promise for counteracting resistance to current immunotherapies, though long-term clinical efficacy remains to be fully established.
Several potential biomarkers have been identified for predicting response to LAG-3-targeting therapies:
LAG-3 expression levels:
Co-expression patterns:
Gene expression signatures:
Tumor mutational profile:
The complex interplay between these factors highlights the need for comprehensive biomarker assessment to guide patient selection for LAG-3-targeting therapies.
The tumor microenvironment (TME) significantly impacts LAG-3 expression and function through multiple mechanisms:
Cytokine milieu:
Hypoxia and metabolic factors:
Hypoxic conditions common in solid tumors may alter LAG-3 expression and function
Metabolic competition within the TME can drive T cell exhaustion and upregulate multiple checkpoint molecules including LAG-3
Tumor-specific ligand expression:
Upregulation of FGL1 by tumor cells provides an MHC-II-independent mechanism for LAG-3-mediated immunosuppression
Galectin-3 expression on tumor cells can engage LAG-3 on T cells to suppress their function
LSECtin expressed by tumor cells interacts with LAG-3, inhibiting antitumor T-cell responses by reducing CDK2, CDK4, and CDK6 expression
Cellular cross-talk:
LAG-3+ regulatory T cells within the TME exhibit enhanced suppressive function
LAG-3 expression on NK cells may result in inhibitory feedback signals from surrounding MHC-II+ cells
In gastric cancer, LAG-3+ cell infiltration can associate with immunoevasive contexture featuring decreased IFN-γ+ cells and increased regulatory T cells and M2-like macrophages
Understanding these complex interactions is critical for developing effective therapeutic strategies targeting LAG-3 in different tumor contexts.
Several mechanisms may contribute to resistance to LAG-3-targeted therapies:
Compensatory checkpoint upregulation:
Alternative ligand interactions:
Even with MHC-II binding blocked, LAG-3 may still interact with alternative ligands like FGL1
Complete disruption of all LAG-3 ligand interactions may be necessary for optimal therapeutic effect
T cell-intrinsic dysfunction:
Chronic antigen exposure may lead to epigenetic changes in T cells that persist despite checkpoint blockade
Terminal exhaustion states may be refractory to LAG-3 blockade alone
TME immunosuppressive mechanisms:
Myeloid-derived suppressor cells and regulatory T cells may maintain immunosuppression through LAG-3-independent mechanisms
Metabolic competition and hypoxia can impair T cell function despite LAG-3 blockade
Tumor-intrinsic resistance mechanisms:
These resistance mechanisms underscore the importance of combination approaches targeting multiple aspects of tumor-immune interactions.
LAG-3 variants and polymorphisms can significantly impact antibody binding and function, presenting important considerations for research and therapeutic development:
Structural variants:
Alternative splicing can generate LAG-3 variants with altered domain structure
Deletions in key binding domains (D1-D4) may affect antibody recognition and function
Soluble LAG-3 variants can potentially interfere with antibody binding to membrane-bound LAG-3
Single nucleotide polymorphisms (SNPs):
SNPs in the LAG-3 gene can alter protein structure and expression
Variations in binding epitopes may affect antibody recognition
Polymorphisms in regulatory regions can influence LAG-3 expression levels in response to various stimuli
Species-specific differences:
Significant differences exist between human and murine LAG-3
These differences may limit the translational value of some preclinical models
Species-specific antibodies must be used in appropriate model systems
Post-translational modifications:
Glycosylation patterns can vary and affect antibody binding
Differential processing of LAG-3 in various cell types may generate functionally distinct forms
Proteolytic cleavage of LAG-3 generates soluble forms that may have different immunological properties
For therapeutic development, understanding these variations is crucial as they may contribute to heterogeneous responses observed in clinical trials. Antibody engineering approaches that target conserved epitopes or multiple epitopes simultaneously may help address these challenges .
Beyond conventional antibodies, several innovative approaches to LAG-3 targeting are being explored:
Multifunctional fusion proteins:
LAG-3 blockade combined with targeted cytokine delivery
Tri-specific antibodies targeting LAG-3, PD-1, and tumor-associated antigens
Cell-based therapies:
Genetic modification of CAR-T cells to be resistant to LAG-3 inhibition
Combination of adoptive cell therapy with LAG-3 blockade
Ex vivo expansion of T cells in the presence of LAG-3 blockers
Small molecule inhibitors:
Development of small molecules targeting the LAG-3/MHC-II interaction
Inhibitors of intracellular LAG-3 signaling pathways
LAG-3 ligand targeting:
Antibodies against FGL1 to block its interaction with LAG-3
Strategies targeting Galectin-3 and LSECtin as alternative approaches
Combinations with emerging immunotherapies:
These innovative strategies aim to overcome limitations of current approaches and address resistance mechanisms encountered with existing LAG-3-targeted therapies.
LAG-3 research has potential applications beyond cancer immunotherapy:
Autoimmune diseases:
LAG-3 agonists might suppress pathological immune responses
Targeting LAG-3+ pathogenic T cells could provide selective immunosuppression
Modulation of regulatory T cell function through LAG-3 could restore immune tolerance
Infectious diseases:
LAG-3 blockade might reinvigorate exhausted T cells in chronic viral infections
Potential application in HIV, HBV, and HCV treatment
Role in enhancing vaccine efficacy against challenging pathogens
Neurodegenerative disorders:
LAG-3 is expressed on microglia and neurons in the central nervous system
Modulating neuroinflammation through LAG-3 targeting could impact disease progression
Potential role in Alzheimer's disease and multiple sclerosis pathology
Transplantation medicine:
The expanding understanding of LAG-3 biology continues to reveal potential applications beyond oncology, highlighting its role as a central regulator of immune homeostasis.