ICOSLG (Inducible T Cell Costimulator Ligand), also designated as CD275, is a glycosylated transmembrane protein encoded by the ICOSLG gene located on chromosome 21q22.3 . It belongs to the B7/CD28 superfamily and plays a critical role in modulating T-cell and B-cell responses during adaptive immunity .
T-cell activation: ICOSLG binds to ICOS on activated T cells, enhancing proliferation, differentiation, and cytokine secretion (e.g., IL-4, IL-13) .
B-cell support: Facilitates germinal center formation and antibody class switching .
Endothelial role: Promotes lymphocyte/neutrophil migration across endothelial cells .
A homozygous missense mutation (p.N219K) in ICOSLG causes combined immunodeficiency (CID) characterized by:
ICOSLG’s dual role in antitumor and protumor immunity makes it a therapeutic target:
Tumor Microenvironment: ICOSLG+ TCs and TILs are associated with increased Foxp3+ Tregs and reduced CD4+ T cells, fostering immunosuppression .
Synergy with IL-10: ICOSLG deficiency in mice leads to IL-10 overproduction, exacerbating colitis unless microbiota is controlled (e.g., via metronidazole) .
Data from the Human Protein Atlas reveal widespread ICOSLG expression in both lymphoid and non-lymphoid tissues :
Tissue/Organ | Expression Level | Cell Type |
---|---|---|
Spleen | High | B cells, dendritic cells, macrophages |
Lymph node | High | B cells, T cells, APCs |
Liver | Moderate | Hepatocytes, sinusoidal endothelial cells |
Kidney | Low | Proximal tubular cells |
Brain | Low | Microglial cells |
Inducible T-Cell Costimulator Ligand, also known as ICOSLG, belongs to the B7 family of co-stimulatory molecules, which includes B7-1 and B7-2. This transmembrane glycoprotein possesses extracellular IgV and IgC domains and binds to ICOS on activated T cells, playing a crucial role in their proliferative response through ICOSLG-dependent signaling.
Produced in Sf9 Baculovirus cells, our ICOSLG is a single, glycosylated polypeptide chain comprising 480 amino acids (19-256a.a.). It has a molecular mass of 53.7kDa, although it may appear between 50-70kDa on SDS-PAGE due to glycosylation. The protein is expressed with a C-terminal 239 amino acid hIgG-His-tag and purified using proprietary chromatographic techniques.
The ICOSLG protein solution is provided at a concentration of 1mg/ml in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
The purity of our ICOSLG is determined to be greater than 90.0% by SDS-PAGE analysis.
Inducible T-Cell Costimulator Ligand, B7-Related Protein 1, B7 Homolog 2, B7-Like Protein Gl50, B7 Homologue 2, B7RP-1, ICOSL, B7-H2, B7RP1, B7H2, Transmembrane Protein B7-H2 ICOS Ligand, Inducible T-Cell Co-Stimulator Ligand, CD275 Antigen, ICOS Ligand, KIAA0653, ICOS-L, CD275, LICOS, GL50, ICOS ligand, B7 homolog 2, B7-H2, B7-like protein Gl50, B7-related protein 1, B7RP-1.
Sf9, Baculovirus cells.
ADPDTQEKEV RAMVGSDVEL SCACPEGSRF DLNDVYVYWQ TSESKTVVTY HIPQNSSLEN VDSRYRNRAL MSPAGMLRGD FSLRLFNVTP QDEQKFHCLV LSQSLGFQEV LSVEVTLHVA ANFSVPVVSA PHSPSQDELT FTCTSINGYP RPNVYWINKT DNSLLDQALQ NDTVFLNMRG LYDVVSVLRI ARTPSVNIGC CIENVLLQQN LTVGSQTGND IGERDKITEN PVSTGEKNAA TLEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPGKHHHHHH.
ICOSLG (Inducible T Cell Costimulator Ligand) is a cell surface protein expressed on various cell types, particularly antigen-presenting cells (APCs). Its primary function is to interact with its receptor ICOS (Inducible T Cell Costimulator), which is expressed on activated T cells. This interaction is crucial for generating adaptive immunity through T-B lymphocyte costimulation . ICOSLG-ICOS interaction plays a vital role in regulating T cell activation, differentiation, and effector function, thereby supporting the development of effective immune responses. The cognate interaction between ICOS and ICOSL is necessary for appropriate immune function, and disruption of this pathway can lead to significant immunological abnormalities .
ICOSLG expression is dynamically regulated in different tissues and cell types. In normal human physiology, ICOSLG is constitutively expressed on antigen-presenting cells such as dendritic cells, B cells, and macrophages. Its expression can be further induced or modulated by various inflammatory signals and cytokines. Research has shown that NF-κB signaling pathways play a crucial role in regulating ICOSLG expression, as evidenced by the finding that NF-κB–inducing kinase (NIK) deficiency (due to bi-allelic mutations in MAP3K14) results in loss of induced ICOSLG expression . This regulation ensures appropriate immune responses while preventing excessive immune activation that could lead to autoimmunity.
The ICOS-ICOSLG pathway represents a critical costimulatory system in adaptive immunity. When ICOS is expressed on activated T cells, it interacts with ICOSLG on antigen-presenting cells, providing a second signal necessary for optimal T cell activation, differentiation, and function . This interaction is particularly important for T helper cell differentiation, germinal center formation, and antibody production by B cells. The pathway supports T cell-dependent B cell responses, leading to effective humoral immunity. Notably, through cognate interaction with ICOSLG expressed on various cells, particularly APCs, adaptive immunity is generated . Disruption of this pathway, as seen in ICOS or ICOSLG deficiencies, leads to impaired antibody responses and memory B cell generation, highlighting its essential role in adaptive immunity.
Human ICOSLG deficiency manifests as a combined immunodeficiency (CID) characterized by both adaptive and innate immune abnormalities. The first reported patient with autosomal recessive ICOSLG deficiency presented with recurrent respiratory tract infections and DNA-based viral infections, indicating compromised T cell function . Additional clinical features included hypogammaglobulinemia, reflecting impaired B cell function, and panlymphopenia, indicating broader effects on lymphocyte development or maintenance. Notably, the patient also developed moderate neutropenia but without the typical pyogenic infections usually associated with neutrophil defects . This complex clinical picture suggests that ICOSLG deficiency affects multiple immune cell lineages beyond just the adaptive immune system, placing it firmly in the category of combined immunodeficiencies rather than selective antibody deficiencies.
The N219K mutation in ICOSLG (c.657C>G; p.N219K) represents a missense mutation that significantly impairs protein function through altered cellular localization. Molecular studies revealed that while wild-type ICOSLG is normally expressed at the cell surface where it can interact with ICOS on activated T cells, the mutant ICOSL N219K protein is retained intracellularly in the endoplasmic reticulum/Golgi apparatus . This aberrant localization prevents the protein from reaching its functional site at the cell surface. The retention is predicted to result from deleterious conformational and biochemical changes in the protein structure caused by the amino acid substitution. Without appropriate surface expression, ICOSLG cannot engage with ICOS on T cells, thereby abrogating the critical costimulatory signal necessary for effective T-B cell cooperation and adaptive immune responses .
Patients with ICOSLG deficiency exhibit multiple immunological abnormalities across both lymphoid and myeloid compartments. Laboratory evaluations of the first identified ICOSLG-deficient patient revealed significant reductions in various immune cell populations, as summarized in the following table derived from patient data :
Parameter | Age (yr) | ||
---|---|---|---|
29 | 30–32 | 33–35 | |
ANC | 3,100 | 924–1,862 (↓) | 341–1,000 (↓) |
AMC | 412 | 60–566 | 13–1179 |
ALC | 700 (↓) | 588–849 (↓) | 210–720 (↓) |
CD3 | 525 (↓) | 423–645 (↓) | 259–469 (↓) |
CD4 | 420 | 306–501 (↓) | 190–301 (↓) |
CD8 | 98 (↓) | 112–136 (↓) | 68–154 (↓) |
CD19 | 98 (↓) | 88–114 (↓) | 68–144 (↓) |
NK | 49 (↓) | 47–48 (↓) | 43–90 (↓) |
IgG | 2.8 (↓) | 6.1–15.15 | 10.9–17.6 |
These findings demonstrate panlymphopenia (reduced ALC), affecting T cells (CD3, CD4, CD8), B cells (CD19), and NK cells. Additionally, progressive neutropenia (reduced ANC) developed over time. Hypogammaglobulinemia (reduced IgG) was also observed, consistent with impaired B cell function . Mechanistically, ICOSL N219K was shown to diminish B cell costimulation of T cells, providing a compelling basis for the observed defect in antibody and memory B cell generation. Interestingly, ICOSL N219K also impaired migration of lymphocytes and neutrophils across endothelial cells, which likely contributed to the combined defects in adaptive immunity and neutropenia observed in the patient .
While both ICOS and ICOSLG deficiencies affect the same costimulatory pathway, there are notable differences in their clinical presentations that reflect the broader expression pattern of ICOSLG. ICOS deficiency was initially identified as causing common variable immunodeficiency characterized primarily by hypogammaglobulinemia and recurrent bacterial infections . Subsequent reports expanded this understanding to include T cell dysfunction manifesting as susceptibility to opportunistic infections (e.g., human papillomavirus, Cryptococcus), recategorizing ICOS deficiency as a combined immunodeficiency (CID) .
ICOSLG deficiency presents with a broader phenotype involving both lymphoid and myeloid lineages. Beyond the hypogammaglobulinemia and infections seen in ICOS deficiency, ICOSLG-deficient patients develop neutropenia and more pronounced lymphopenia affecting multiple lineages . This expanded clinical phenotype likely reflects the wider expression pattern of ICOSLG compared to ICOS, including its expression on endothelial cells, which affects immune cell migration. The distinction underscores the importance of considering both molecules when evaluating patients with unexplained combined immunodeficiencies.
ICOSLG demonstrates a ubiquitous expression pattern in oral squamous cell carcinoma (OSCC) microenvironment, being detected in multiple cellular compartments. Research has documented ICOSLG expression in tumor cells (TCs), cancer-associated fibroblasts (CAFs), and tumor-infiltrating lymphocytes (TILs) . This multifaceted expression pattern suggests complex roles for ICOSLG in the tumor microenvironment beyond its canonical function in immune cell interactions. The presence of ICOSLG across diverse cell types in OSCC indicates potential autocrine and paracrine signaling mechanisms that may influence tumor biology through multiple pathways. Importantly, the expression levels of ICOSLG in these different cellular compartments have been associated with distinct clinicopathological features and outcomes, highlighting the context-dependent functions of this molecule in cancer progression .
These associations were not observed for ICOSLG expression in cancer-associated fibroblasts (ICOSLGCAFs). Multivariate analysis revealed that while lymph node metastasis and high expression of ICOSLG in TCs and TILs showed significant differences in OS and MFS, they were not independent prognostic indicators for OSCC . This suggests that ICOSLG expression may contribute to poor outcomes through mechanisms related to tumor progression and metastasis.
ICOSLG expression shows specific associations with clinicopathological features in OSCC, particularly those related to disease progression and metastasis. Research has demonstrated that while ICOSLG expression does not significantly correlate with patient gender, age, or tumor differentiation, high expression of ICOSLG in tumor cells (ICOSLGTCs) and tumor-infiltrating lymphocytes (ICOSLGTILs) is associated with higher risk of lymph node metastasis and advanced TNM stage .
Specifically, immunohistochemistry results showed that high ICOSLGTCs expression correlates with increased lymph node metastasis and advanced TNM stage, while high ICOSLGTILs associates with advanced TNM stage and T stage . Interestingly, analysis of the Tisch2 database revealed that in head and neck squamous cell carcinoma, high ICOSLGTCs expression positively correlates with advanced TNM stage, whereas high ICOSLGTILs expression, particularly in CD4+ T cells, negatively correlates with advanced TNM stage . These findings highlight the complex and context-dependent roles of ICOSLG in different cellular compartments within the tumor microenvironment.
ICOSLG expression significantly influences the composition and distribution of immune cell populations in the tumor microenvironment of OSCC. Research examining the relationship between high ICOSLGTCs expression and immune cell subsets revealed distinct patterns at the invasive front versus the tumor center. At the invasive front, high ICOSLGTCs expression correlates with decreased CD4+ T cells and CD19+ B cells, while Foxp3+ regulatory T cells (Tregs) show an increasing trend . In the tumor center, both CD4+ and CD8+ T cells demonstrate a decreasing trend in patients with high ICOSLGTCs expression .
The ICOSLG-ICOS interaction activates distinct signaling pathways depending on the cellular context, though research in this area is still developing. In T cells, ICOS engagement by ICOSLG triggers PI3K activation and subsequent AKT signaling, which promotes T cell survival, proliferation, and cytokine production. The ICOS-ICOSLG pathway is particularly important for CD4+ T cell differentiation and function, including the development of T follicular helper cells that support germinal center formation and B cell responses.
In B cells and other ICOSLG-expressing cells, reverse signaling through ICOSLG may occur, though this has been less extensively characterized than forward signaling through ICOS. Based on broader research on costimulatory molecules, ICOSLG engagement likely activates NF-κB signaling pathways and potentially MAPK cascades, influencing cellular activation, survival, and cytokine production . In tumor cells expressing ICOSLG, these signaling events may contribute to pro-survival and proliferative advantages, potentially explaining the correlation between high ICOSLG expression and poor clinical outcomes in cancer . The diversity of signaling outcomes in different cellular contexts likely contributes to the pleiotropic effects of this pathway in immunity and disease.
Post-translational modifications (PTMs) represent critical regulatory mechanisms affecting ICOSLG function, though this area requires further investigation. The retention of mutant ICOSL N219K protein in the endoplasmic reticulum/Golgi apparatus suggests that proper protein folding and trafficking are essential for ICOSLG function . This points to the importance of post-translational modifications such as glycosylation in determining ICOSLG cellular localization.
The asparagine residue at position 219 (mutated to lysine in the reported case of ICOSLG deficiency) could potentially be involved in N-linked glycosylation, which is often crucial for proper protein folding and trafficking to the cell surface. Disruption of this site could alter glycosylation patterns, leading to protein misfolding and retention in the ER/Golgi . Additionally, other potential PTMs like phosphorylation and ubiquitination likely regulate ICOSLG stability, turnover, and signaling capacity. Understanding these modifications represents an important frontier for future research, as they may offer opportunities for therapeutic manipulation of the ICOS-ICOSLG pathway in various disease contexts.
The structural basis of ICOSLG-ICOS interaction specificity remains an area requiring deeper investigation, though insights can be gleaned from the reported N219K mutation in ICOSLG. This mutation results in impaired surface localization of ICOSLG, preventing its interaction with ICOS . The fact that a single amino acid substitution can so profoundly affect protein trafficking suggests that proper protein folding is critical for maintaining the structural integrity necessary for ICOS binding.
ICOSLG represents a promising target for cancer immunotherapy due to its roles in immune regulation and direct effects on tumor biology. Several approaches could be developed to therapeutically target the ICOSLG pathway in cancer. Blocking antibodies against ICOSLG could prevent its interaction with ICOS, potentially reversing immunosuppressive effects in the tumor microenvironment. This approach might be particularly beneficial in cancers where high ICOSLG expression correlates with poor outcomes, such as OSCC .
Conversely, ICOS agonists could be developed to stimulate anti-tumor T cell responses. The positive correlation between ICOSLG and multiple immune checkpoints, including PD-L1, CTLA4, and PD1, suggests that combination therapies targeting multiple pathways simultaneously might be more effective than single-agent approaches . Furthermore, given ICOSLG's association with advanced disease stage and metastasis in OSCC, it could serve as a biomarker for patient stratification, identifying those who might benefit from specific immunotherapeutic interventions. Preclinical drug experiments have already demonstrated good clinical application prospects for immunotherapy targeting the ICOS-ICOSLG pathway in various tumors .
For immunodeficiency disorders, genetic testing for ICOSLG mutations could be incorporated into screening panels for patients presenting with combined immunodeficiency phenotypes, particularly those with features of both T and B cell dysfunction plus neutropenia . Additionally, functional assays measuring ICOSLG surface expression on appropriate cell types could provide diagnostic information about pathway integrity. The association between ICOSLG expression and immune checkpoint molecules also suggests potential applications in predicting response to existing immunotherapies, as patients with high ICOSLG expression might respond differently to treatments targeting other immune checkpoints .
Researchers have several methodological options for modulating ICOSLG expression to study its functions in different contexts. Genetic approaches include CRISPR-Cas9-mediated knockout or knockdown of ICOSLG, which can reveal phenotypes associated with complete or partial loss of function. Conversely, overexpression systems using viral vectors can demonstrate effects of increased ICOSLG signaling. Site-directed mutagenesis can be employed to create specific mutations, such as the N219K variant identified in immunodeficiency, to study structure-function relationships .
Pharmacological approaches include using blocking antibodies against ICOSLG to interrupt its interaction with ICOS, or recombinant ICOS-Fc fusion proteins that bind ICOSLG and prevent endogenous ICOS engagement. For studying ICOSLG in tumor contexts, researchers can utilize patient-derived xenografts or syngeneic mouse models with genetic manipulation of ICOSLG expression in tumor cells. In vitro co-culture systems combining ICOSLG-expressing cells with ICOS-expressing T cells can help dissect the bidirectional signaling mechanisms involved in this pathway . These diverse methodological approaches provide researchers with a toolkit for investigating ICOSLG biology across different experimental contexts.
The relationship between ICOSLG and response to existing immunotherapies represents an important frontier in cancer research. ICOSLG expression correlates positively with multiple immune checkpoint molecules, including PD-L1 (CD274), CTLA4, and PD1 (PDCD1), which are targets of approved immunotherapeutic antibodies . This correlation suggests that ICOSLG expression might influence or predict responses to these existing treatments.
In tumors with high ICOSLG expression, the immunosuppressive microenvironment characterized by decreased CD4+ T cells and increased Foxp3+ regulatory T cells might limit the efficacy of single-agent checkpoint inhibitors . Conversely, the positive correlation between ICOSLG and these checkpoints suggests that patients with high ICOSLG might be candidates for combination immunotherapy approaches targeting multiple inhibitory pathways simultaneously. As immune checkpoint therapy enhances anti-tumor effects by regulating T cell function in the tumor microenvironment, understanding how ICOSLG influences this regulation could help optimize treatment strategies . Future clinical studies examining ICOSLG expression in relation to immunotherapy response are needed to clarify these relationships and develop more precise treatment algorithms.
Single-cell technologies offer powerful approaches to dissect the heterogeneous expression and function of ICOSLG across diverse cell types and disease contexts. Single-cell RNA sequencing (scRNA-seq) can reveal cell type-specific expression patterns of ICOSLG and ICOS, identifying previously unrecognized cell populations involved in this signaling axis. This approach could be particularly valuable for understanding the complex cellular interactions in tumor microenvironments, where ICOSLG is expressed across multiple cell types including tumor cells, cancer-associated fibroblasts, and immune cells .
Single-cell ATAC-seq could identify regulatory elements controlling ICOSLG expression in different contexts, while single-cell proteomics approaches like mass cytometry (CyTOF) could simultaneously measure ICOSLG protein expression alongside dozens of other markers to better characterize the phenotypes of ICOSLG-expressing cells. Spatial transcriptomics and multiplexed immunofluorescence would preserve tissue architecture information, allowing researchers to analyze ICOSLG expression in the spatial context of the tumor microenvironment or lymphoid tissues. These approaches could reveal important insights about how ICOSLG-expressing cells interact with ICOS-expressing cells within complex tissue environments .
Animal models provide essential systems for investigating ICOSLG functions in complex physiological and pathological contexts. Genetically engineered mouse models (GEMMs) with germline or conditional knockout of ICOSLG can reveal its roles in immune development and function. Tissue-specific conditional knockout models using Cre-lox systems would be particularly valuable for distinguishing the functions of ICOSLG in different cell types, such as dendritic cells, B cells, or non-hematopoietic cells.
For cancer research, syngeneic mouse models with ICOSLG-expressing tumors can be used to study how tumor-derived ICOSLG affects anti-tumor immunity and response to immunotherapies . Humanized mouse models, where human immune system components are engrafted into immunodeficient mice, could better recapitulate human ICOS-ICOSLG interactions. To model human ICOSLG deficiency, knock-in mice harboring the N219K mutation identified in the human patient could be generated to study the immunological consequences in vivo . These diverse animal models, when used in combination with appropriate functional readouts and therapeutic interventions, can provide insights that are difficult to obtain from human studies alone.
Computational approaches offer powerful tools for predicting novel functions of ICOSLG and generating hypotheses for experimental validation. Network analysis of protein-protein interaction data could identify previously unrecognized binding partners of ICOSLG beyond ICOS, suggesting novel signaling pathways and functions. Gene co-expression analyses across large transcriptomic datasets could reveal genes consistently co-regulated with ICOSLG, pointing to biological processes where it plays important roles.
Structural bioinformatics approaches, including molecular modeling and docking simulations, could predict how mutations like N219K affect ICOSLG protein structure and binding interfaces . Machine learning algorithms applied to multi-omics data from cancer patients could identify patterns associating ICOSLG expression with specific molecular subtypes, clinical outcomes, or treatment responses . Systems biology approaches integrating transcriptomic, proteomic, and metabolomic data could map how ICOSLG perturbations ripple through cellular networks. These computational approaches complement experimental studies by generating testable hypotheses and providing a broader contextual understanding of ICOSLG biology across different physiological and pathological states.
Reconciling contradictory findings about ICOSLG across different disease contexts requires thoughtful experimental design and analytical approaches that account for context-dependent functions. One strategy is to develop integrated experimental models that simultaneously examine multiple cell types and their interactions. Co-culture systems combining tumor cells, immune cells, and stromal elements can help understand how ICOSLG functions differ depending on which cells express it and which cells receive its signals .
Cross-disease comparisons using standardized methodologies could identify context-specific factors that modify ICOSLG function. For instance, comparing ICOSLG effects in different cancer types or between cancer and autoimmunity might reveal important modulatory factors. Meta-analyses of published studies, controlling for methodological differences, could help distinguish robust findings from those that might be context-dependent or methodology-specific. Single-cell approaches are particularly valuable for resolving apparently contradictory bulk tissue findings by revealing distinct effects in specific cell subpopulations .
For example, in head and neck squamous cell carcinoma, high ICOSLGTCs expression positively correlates with advanced TNM stage, while high ICOSLGTILs expression in CD4+ T cells negatively correlates with advanced stage . Such seemingly contradictory findings can be reconciled by recognizing that ICOSLG has different functions depending on the expressing cell type and its microenvironmental context.
Inducible T-cell costimulator ligand (ICOSL), also known as CD275, is a transmembrane protein that plays a crucial role in the immune system. It belongs to the B7 family of proteins and is primarily expressed on antigen-presenting cells (APCs) such as B cells, dendritic cells, and monocytes . The interaction between ICOSL and its receptor, inducible T-cell costimulator (ICOS), is essential for the activation and regulation of T cells .
ICOSL is a member of the B7 family and shares structural similarities with other family members, such as CD80 and CD86 . It is expressed on various cell types, including endothelial cells, fibroblasts, epithelial cells, and several types of tumor cells . The expression of ICOSL is upregulated in response to inflammatory signals, which enhances its interaction with ICOS on activated T cells .
The primary function of ICOSL is to bind to ICOS, a costimulatory receptor expressed on activated T cells . This interaction provides a crucial second signal for T-cell activation, proliferation, and differentiation. ICOSL-ICOS signaling is essential for the development and function of follicular helper T cells (Tfh), which play a vital role in the formation of germinal centers and the production of high-affinity antibodies .
In addition to its role in normal immune responses, ICOSL-ICOS signaling has been implicated in various pathological conditions, including autoimmune diseases and cancer . For instance, the interaction between ICOSL and ICOS can promote the expansion of regulatory T cells (Tregs), which suppress immune responses and contribute to tumor immune evasion .
Given its critical role in immune regulation, ICOSL has emerged as a potential therapeutic target for various diseases. Recombinant ICOSL (human recombinant) has been developed to modulate ICOSL-ICOS signaling in therapeutic applications . For example, activating ICOSL-ICOS signaling using recombinant ICOSL or agonist monoclonal antibodies (mAbs) has shown promise in enhancing antitumor immune responses .
In cancer therapy, combining ICOSL-ICOS activation with other immunotherapies, such as programmed cell death protein 1 (PD-1) blockade, has demonstrated synergistic effects in preclinical models . This combination approach aims to enhance the activation of antitumor T cells and improve the overall efficacy of cancer immunotherapy .