SIGLEC15 is an emerging immune checkpoint molecule widely expressed in various tumors, closely associated with immune evasion, treatment resistance, and poor prognosis. It plays a key role in tumor immunosuppression by inhibiting T cell activity, inducing a non-inflammatory tumor microenvironment, and regulating the polarization of tumor-associated macrophages. In recent years, strategies targeting SIGLEC15, including monoclonal antibodies, bispecific antibodies, CAR-T cells, and gene silencing, have developed rapidly, providing new avenues to overcome the limitations of PD-1/PD-L1 therapy. This article reviews the structure and function of SIGLEC15, systematically analyzes its signaling pathways, disease relevance, and progress in drug development, aiming to provide a theoretical basis for subsequent immunotherapy research and new drug development.
1. The Emergence and Significance of SIGLEC15 Research
2. Biological Characteristics and Expression Pattern of SIGLEC15
3. Mechanism of Action of SIGLEC15 in the Tumor Microenvironment
4. Signal Transduction Pathways of SIGLEC15
5. SIGLEC15 and Related Diseases
The advent of immune checkpoint inhibitors has revolutionized cancer treatment, but a significant number of patients do not respond to PD-1/PD-L1 therapy or develop acquired resistance. Finding new immune regulatory targets has become crucial for advancing immunotherapy. Sialic acid-binding Ig-like lectin 15 (SIGLEC15), a member of the Siglec family, is emerging as a research hotspot for the next generation of immune checkpoints due to its unique immunosuppressive functions and high expression in various tumors [1,2].
Studies have shown that SIGLEC15 is highly expressed in multiple tumor types and is closely associated with poor prognosis. Unlike PD-L1, the upregulation of SIGLEC15 often occurs in PD-L1-negative or immunologically "cold" tumors [1-3], suggesting it may mediate tumor immune escape through independent pathways. Therefore, in-depth analysis of SIGLEC15's mechanism of action and targeting strategies is of great significance for improving responses to tumor immunotherapy.
SIGLEC15 is an important member of the Siglec family, consisting of an extracellular immunoglobulin-like domain, a transmembrane region, and a short intracellular tail. Its extracellular structure recognizes sialylated glycans and is involved in intercellular signaling regulation [4]. Although the intracellular region lacks typical immunoreceptor tyrosine-based inhibitory motifs (ITIMs), it can form complexes with adaptor proteins such as DAP12 via charged residues in its transmembrane segment, thereby mediating downstream signaling [5].
Research indicates that SIGLEC15 is significantly upregulated in various malignancies including hepatocellular carcinoma (HCC) [4], thyroid cancer [6], colon adenocarcinoma [1], glioma [8], bladder cancer [2], and B-cell acute lymphoblastic leukemia (B-ALL) [7]. Its high expression is often associated with reduced CD8+ T cell infiltration, increased immunosuppressive cells, and decreased overall survival. Immunofluorescence studies have found that SIGLEC15 co-localizes with the M2 macrophage marker CD163, suggesting its role in regulating macrophage polarization within the tumor immune microenvironment [8].
SIGLEC15 achieves immunosuppression by inhibiting CD8+ T cell activity, reducing cytotoxicity, and impairing T cell survival. Its high expression is often correlated with decreased levels of chemokines (such as CXCL9/10) and a lower T-cell inflammation score [10]. In B-ALL models, knockout of SIGLEC15 promotes CD8+ T cell expansion and enhances anti-tumor immune responses [7].
SIGLEC15 is highly expressed on M2-type TAMs. It exacerbates local immune tolerance by enhancing the secretion of immunosuppressive factors like TGF-β and IL-10 [11,12]. Simultaneously, it promotes angiogenesis and stroma remodeling, creating a microenvironment conducive to tumor metastasis.
The expression of SIGLEC15 and PD-L1 exhibits complementary or independent patterns across different tumor types [1,4]. High SIGLEC15 levels often accompany low PD-L1 expression and an immunologically cold microenvironment, rendering tumors insensitive to traditional immune checkpoint inhibitors [2]. Combined blockade of both significantly improves the response rate to immunotherapy.
Signal balance in immune cells is co-regulated by activating ITAM (Immunoreceptor Tyrosine-based Activation Motif) and inhibitory ITIM (Immunoreceptor Tyrosine-based Inhibitory Motif) modules. Although SIGLEC15 lacks a typical ITIM structure, its transmembrane region contains positively charged residues that enable binding to the ITAM-containing adaptor protein DAP12 [13]. Upon phosphorylation by Src family kinases (such as Lyn or Fgr), DAP12 recruits Syk kinase, initiating downstream cascade signals including the PI3K/AKT and MAPK/ERK pathways, thereby regulating cell activation and differentiation [14]. This suggests that SIGLEC15 can function as an "activating" regulatory receptor in specific contexts, influencing immune cell responses via the DAP12-Syk axis.
Syk family kinases hold a central position in immune signal transduction. The SIGLEC15-DAP12-Syk pathway can promote functional changes in myeloid cells, enhancing the immunosuppressive properties of monocytes/macrophages. Some studies indicate that Syk activation can upregulate STAT3 and NFκB signaling, driving the expression of immunosuppressive cytokines (e.g., IL-10, TGF-β) [15]. Furthermore, SIGLEC15 may regulate integrin adhesion molecule activity via Src kinases, affecting cell migration and immune synapse formation.
Studies in leukemia and thyroid cancer have found that NFκB activation can induce transcriptional upregulation of SIGLEC15, forming a positive feedback loop [7,16]. Concurrently, SIGLEC15 can directly interact with EGFR, stabilizing its protein structure and enhancing downstream ERK and PI3K/AKT signaling [16]. This mechanism not only promotes tumor cell migration and invasion but also reveals non-immune roles for SIGLEC15 in tumor progression.
Overall, the SIGLEC15 signaling network spans immunosuppressive and tumor-promoting pathways, exerting multi-layered effects on the tumor microenvironment through coordinated regulation via the DAP12-Syk, NFκB, and EGFR axes.
SIGLEC15 is highly expressed in various solid tumors and is an independent indicator of poor prognosis.
In HCC, SIGLEC15 promotes immune evasion by inducing CD8+ T cell apoptosis, and its blockade can restore immune responses [4]. In bladder cancer, its high expression is associated with the formation of an immunologically cold microenvironment, insensitivity to immunotherapy, and an increased risk of hyperprogression [2,10]. In thyroid cancer, high SIGLEC15 expression is closely linked to immune exhaustion and enhanced invasiveness [6].
In glioma, SIGLEC15 levels increase with WHO grade and are closely associated with M2-type TAMs, suggesting its role in remodeling the brain tumor immune microenvironment [8]. Studies in fibrosarcoma show that knocking down SIGLEC15 significantly slows tumor growth and enhances sensitivity to radiotherapy and chemotherapy [9]. Furthermore, its high expression in lung adenocarcinoma spinal metastasis [5] and ovarian cancer [17] has also been confirmed to be closely related to aggressive metastasis and immune evasion.
In B-ALL, NFκB signaling activation directly upregulates SIGLEC15 transcription [7], promoting the secretion of immunosuppressive cytokines and impairing T cell activity. Genetic knockout of SIGLEC15 enhances immune surveillance and improves the leukemia microenvironment. Soluble SIGLEC15 has been detected in patient plasma, suggesting its potential as a biomarker for diagnosis and treatment response prediction.
SIGLEC15 regulates osteoclast differentiation and bone resorption, making it an important factor in bone homeostasis. Research indicates that SIGLEC15 modulates RANKL signaling in the bone microenvironment, affecting osteoclast formation [18]. In breast cancer bone metastasis models, it promotes the formation of bone metastases by inhibiting CD8+ T cell activation [18]. Additionally, SIGLEC15 may be involved in the inflammatory regulation of rheumatoid arthritis, and its association with the DAP12-Syk pathway hints at its role in inflammatory immune regulation [13].
As an emerging immunosuppressive target in the tumor microenvironment, independent of PD-L1 expression, SIGLEC15 offers a new direction to overcome PD-1/PD-L1 resistance. Current research and development are primarily focused on monoclonal antibodies. Among them, NC-318 and PYX-106 have entered Phase II and Phase I clinical trials for non-small cell lung cancer and solid tumors, respectively, while several other candidates are in the preclinical stage. Research in this field is exploring not only their potential as monotherapies but also actively developing combination regimens with existing immunotherapies and expanding into non-oncological areas such as bone diseases. Some investigational candidates are listed in the table below:
| Drug | Mechanism of Action | Drug Type | Indication (Disease Name) | Research Institution | Highest Stage |
|---|---|---|---|---|---|
| NC-318 | Siglec-15 Inhibitor | Monoclonal Antibody | Advanced Non-Small Cell Lung Cancer | NextCure, Inc. | Phase II |
| PYX-106 | Siglec-15 Inhibitor | ADCC | Monoclonal Antibody | Advanced Malignant Solid Tumors | Solid Tumors | Non-Small Cell Lung Cancer | Thyroid Cancer | Pyxis Oncology, Inc. | Biosion (Nanjing) Inc. | Phase I |
| S15-4E6A | Siglec-15 Inhibitor | Biologics | Lung Adenocarcinoma | Nanjing Medical University | Preclinical |
| NP-159 | Siglec-15 Inhibitor | Monoclonal Antibody | Spinal Cord Injury | NextCure, Inc. | Preclinical |
| SHG-8 | Siglec-15 Inhibitor | Small Molecule | Colorectal Cancer | University of Hertfordshire | Preclinical |
| PBI-108 | PD-1 Inhibitor | Siglec-15 Inhibitor | Bispecific Antibody | Solid Tumors | Protheragen, Inc. | Preclinical |
| BCG-008 | Siglec-15 Inhibitor | ADCC | T cell stimulator | Monoclonal Antibody | Solid Tumors | Biocytogen (Beijing) Co., Ltd. | Preclinical |
(Data as of October 31, 2025, sourced from Synapse)
SIGLEC15, as a novel immune checkpoint molecule, plays a central role in tumor immunosuppression. Its signaling pathways span both immune and pro-tumor dimensions, coordinately regulating cell fate through the DAP12-Syk, NFκB, and EGFR axes. Various therapeutic strategies targeting SIGLEC15 (monoclonal antibodies, bispecific antibodies, CAR-T, gene silencing, etc.) are rapidly developing, demonstrating potential to break through PD-1/PD-L1 resistance. CUSABIO provides SIGLEC15 recombinant proteins, antibodies, and ELISA kits to support your related mechanism research and SIGLEC15-targeted drug development.
References
[1] Weixiang Zhan, F. Bai, Yue Cai, Jianwei Zhang, Ge Qin, Yuqian Xie, Yanhong Deng.(2023). Tumor stroma Siglec15 expression is a poor prognosis predictor in colon adenocarcinoma.
[2] Jiao Hu, Anze Yu, Belaydi Othmane, D. Qiu, Huihuang Li, Chao Li, Peihua Liu, Wenbiao Ren, Minfeng Chen, G. Gong, Xi Guo, Huihui Zhang, Jinbo Chen, X. Zu.(2021). Siglec15 shapes a non-inflamed tumor microenvironment and predicts the molecular subtype in bladder cancer.
[3] Huan Lai, Yiyang Liu, Yan Gong, Chuanyu Zong, Wei Zeng, Honglei Chen.(2024). Expression of SIGLEC15 correlates with tumor immune infiltration, molecular subtypes, and breast cancer progression.
[4] Zheng Chen, Mincheng Yu, Bo Zhang, Lei Jin, Qiang Yu, Shuang-Hu Liu, Binghai Zhou, Jiuliang Yan, Wentao Zhang, Xiaoqiang Li, Yongfeng Xu, Yongsheng Xiao, Jian Zhou, Jia Fan, M. Hung, Qinghai Ye, Hui Li, Lei Guo.(2024). SIGLEC15, negatively correlated with PD-L1 in HCC, could induce CD8+ T cell apoptosis to promote immune evasion.
[5] Haifeng Liang, Lei Zhou, Z. Hu, Yuxiang Ge, Tai-Wei Zhang, Qing Chen, Ben Wang, Shunyi Lu, Wang Ding, J. Dong, Fengfeng Xue, Libo Jiang.(2022). Siglec15 Checkpoint Blockade for Simultaneous Immunochemotherapy and Osteolysis Inhibition in Lung Adenocarcinoma Spinal Metastasis via a Hollow Nanoplatform.
[6] Xiaofeng Hou, Chao Chen, X. Lan, Xiao-dong He.(2022). Unveiling the molecular features, relevant immune and clinical characteristics of SIGLEC15 in thyroid cancer.
[7] Claire E Pillsbury, Jodi Dougan, Jennifer L. Rabe, Jairo A Fonseca, Chengjing Zhou, Alyssa N Evans, Hasan Abukharma, Ona Ichoku, Gloria Gonzalez-Flamenco, Sunita I Park, A. Aljudi, D. DeRyckere, S. Castellino, S. Rafiq, S. Langermann, Linda N. Liu, C. Henry, Christopher C Porter.(2023). Siglec-15 Promotes Evasion of Adaptive Immunity in B-cell Acute Lymphoblastic Leukemia.
[8] Jinchao Wang, Lin-gen Xu, Qian Ding, Xiaoru Li, Kai Wang, Shangchen Xu, Bin Liu.(2023). Siglec15 is a prognostic indicator and a potential tumor-related macrophage regulator that is involved in the suppressive immunomicroenvironment in gliomas.
[9] Juan Wang, Baosheng Li.(2025). Siglec15 as a potential therapeutic target for the treatment of fibrosarcoma.
[10] D. Deng, Jiatong Xiao, Jinhui Liu, Huihuang Li, Minghui Hu, Bohan Zhou, Haisu Liang, Benyi Fan, Jinbo Chen, Xiaogen Kuang, Zhenyu Nie, Jiao Hu, Xiongbing Zu.(2024). Evasion of immunosurveillance by the upregulation of Siglec15 in bladder cancer.
[11] Tianjiao Li, Kaifeng Jin, Hao Li, L. Ye, Pengcheng Li, Bruce Jiang, Xuan Lin, Zhen-Yu Liao, Hui-Ru Zhang, Saimeng Shi, Mengxiong Lin, Qinglin Fei, Zhichun Xiao, Hua-Xiang Xu, Liang Liu, Xianjun Yu, Wei Wu.(2022). SIGLEC15 amplifies immunosuppressive properties of tumor-associated macrophages in pancreatic cancer.
[12] R. Takamiya, K. Ohtsubo, S. Takamatsu, N. Taniguchi, T. Angata.(2013). The interaction between Siglec-15 and tumor-associated sialyl-Tn antigen enhances TGF-β secretion from monocytes/macrophages through the DAP12-Syk pathway.
[13] Xianghong Chen, E. Eksioglu, J. Carter, Nicole R. Fortenbery, Sarah S. Donatelli, Jun-min Zhou, Jinhong Liu, Li-li Yang, D. Gilvary, J. Djeu, Sheng Wei.(2015). Inactivation of DAP12 in PMN Inhibits TREM1-Mediated Activation in Rheumatoid Arthritis.
[14] A. Zarbock, C. Abram, M. Hundt, A. Altman, C. Lowell, K. Ley.(2008). PSGL-1 engagement by E-selectin signals through Src kinase Fgr and ITAM adapters DAP12 and FcRγ to induce slow leukocyte rolling.
[15]Frank Fasbender, M. Claus, S. Wingert, M. Sandusky, C. Watzl.(2017). Differential Requirements for Src-Family Kinases in SYK or ZAP70-Mediated SLP-76 Phosphorylation in Lymphocytes.
[16] Sijing Huang, Zhi Ji, Jinqiang Xu, Yue-ning Yang, Bingrui Wu, Qihang Chen, Shuting Geng, Yu Si, Jiayue Chen, Yuanyan Wei, Cong Wang, Zhilong Ai, Jianhai Jiang.(2023). Siglec15 promotes the migration of thyroid carcinoma cells by enhancing the EGFR protein stability.
[17] T. Ochi, Yuko Matsuoka, Tatsuya Konishi, M. Maruta, Y. Miyazaki, K. Tanimoto, Masaki Yasukawa, Masakatsu Yamashita, Katsuto Takenaka.(2023). Development of a New CAR-T-Cell Therapy Targeting an Immune Checkpoint for Effective Treatment of Solid and Liquid Tumors.
[18] Yutong Wu, Hongbo Ai, Yuhang Xi, Jiulin Tan, Ying Qu, Jianzhong Xu, F. Luo, C. Dou.(2023). Osteoclast-derived apoptotic bodies inhibit naive CD8+ T cell activation via Siglec15, promoting breast cancer secondary metastasis.
Comments
Leave a Comment