A New Target for Immunotherapy in GBM: Macrophage Immune Checkpoint Siglec-9

Published in Cancer
Like

Share this post

Choose a social network to share with, or copy the URL to share elsewhere

This is a representation of how your post may appear on social media. The actual post will vary between social networks
Glioblastoma multiforme (GBM) is an extremely aggressive tumours with a very short survival period and the treatment options after recurrence are very limited. To date, no immunotherapy for GBM has been approved by the FDA, and immune checkpoint blockade (ICB) agents targeting coinhibitory T-cell signalling, such as anti-PD1/PD-L1/CTLA4 antibodies, have not yet become a first-line treatment. CheckMate-143 failed to show improved overall survival with nivolumab (anti-PD1 monoclonal antibody) monotherapy compared to bevacizumab (anti-angiogenic monoclonal antibody approved by the FDA for recurrent GBM treatment) (Reardon et al., JAMA Oncol 2020). Furthermore, although a subset of patients benefited from neoadjuvant anti-PD-1 therapy in two pilot clinical trials (Cloughesy et al., Nature Med 2019; Schalper et al., Nature Med 2019), most patients still did not respond to ICB therapy.
One of the key reasons for this limited response is the unique immune-suppressive microenvironment in GBM, which includes a large number of tumor-associated macrophages (TAMs). TAMs are the major immune cells in the GBM tumor microenvironment and consist of two distinct cell populations: tissue-resident small glial cells derived from the yolk sac during embryogenesis and border-associated macrophages derived from bone marrow cells that migrate to the tumor through brain vasculature (Kierdorf et al., Nat Rev Neurosci 2019). During the pathological progression of gliomas, TAMs interact with T cells in a unique way, influencing their activation status and functional anti-tumor effects. TAMs can also promote tumor growth processes, including inflammation, angiogenesis, and invasion, thereby generating pro-tumor effects. In tumors, the balance of these functions is disrupted, and the pro-tumor effects prevail. Although strategies targeting TAM regulation have shown the potential to enhance anti-tumor immune responses, existing TAM targets have not yielded satisfactory results in clinical trials (Veillette et al.,  Trends Immunol 2018). As a result, the ongoing search for new TAM-targeted treatment strategies and ideal targets remains active.

In this study, we revealed a novel molecule called Siglec-9 that acts as an immune checkpoint on macrophages in GBM, significantly impacting the tumor immune environment and immunotherapy response. This research provides new clues to address the above-mentioned challenges.

We performed single-cell RNA sequencing and spatial transcriptomic analysis on samples from 24 GBM patients, including primary tumors, recurrent tumors, and patients who received anti-PD-1 antibody as neoadjuvant therapy. We found that in patients who showed ineffective responses to anti-PD-1 therapy, the tumor-associated macrophages (TAMs) with high expression of the Siglec-9 gene were predominant. Furthermore, we discovered that although anti-PD-1 therapy could alter the tumor immune environment in some GBM patients, activating T cells and increasing the expression of certain chemokine and interferon-γ-related genes, it could not overcome the overall immune suppression state of the tumor microenvironment, particularly the immune inhibitory function of Siglec-9 high-expressing macrophages (Siglec-9+ TAMs).

We further identified two distinct subpopulations of Siglec-9+ TAMs: Siglec-9+ MARCO+ TAMs and Siglec-9+SEPP1SIGLEC9+ TAMs. Spatial transcriptomic analysis revealed that these two TAM subpopulations originated from CD14+ monocytes that migrated stepwise into the tumor and underwent differentiation. Siglec-9+MARCO+ TAMs possibly represented a transitional state where circulating monocytes differentiated into Siglec-9+SEPP1+ TAMs. Notably, cross-comparison with published GBM single-cell sequencing data showed that Siglec-9+ TAMs resembled macrophages co-expressing GPNMB/CSTB/MRC1 in GBM patients treated with immune checkpoint blockade (ICB), while they were not enriched in the dataset of recurrent GBM patients (Lee et al., Nat Commun 2021; Antunes et al., Nat Neurosci 2021). Thus, anti-PD-1 therapy might reprogram Siglec-9+ TAMs. This study provides insights into the reasons behind the response and non-response effects produced by neoadjuvant anti-PD-1 therapy.

Siglec-9 belongs to the Siglec family, which consists of immunoglobulin-like lectins expressed on the surface of macrophages and other myeloid immune cells (Duan et al., Annu Rev Immunol 2020). These receptors act as immune checkpoint molecules by delivering immune inhibitory signals when they bind to glycoproteins containing sialic acid residues on the host/cancer cell membrane. Through studies using a mouse model with SiglecE (the murine functional homolog of human Siglec-9) gene knockout in intracranial tumor models, we found that the deletion of SiglecE could inhibit GBM tumor growth, alter the immune-suppressive phenotype of macrophages to a more anti-tumor phenotype, and significantly enhance T cell activation and proliferation. Moreover, we conducted a series of in vivo and in vitro experiments utilizing mouse models and clinical samples from human patients to further validate the role of Siglec-9+/SiglecE+ TAMs in inhibiting tumor growth in Siglece knockout mice. We observed that Siglec-9+/SiglecE+ TAMs directly interact with T cells. Additionally, to gain further insights into the clinical implications of targeting Siglec-9, we evaluated the tutor growth of Siglece knockout mice  upon receiving anti-PD-1 therapy and found significantly prolonged survival. Furthermore, we found the  competitive protein that specifically targeted Siglec-9, could substantially extended the survival of mice bering glioblastoma.

Figure 1: Schematic Illustration of Macrophage Specialization and T Cell Interaction in Human GBM and Mouse Models | Recurrent GBM tumors are accompanied by increased angiogenesis, loss of microglial cells, and infiltration of TAMs derived from monocytes. After neoadjuvant anti-PD-1 therapy, especially in non-responders, Siglec-9+ TAMs accumulated. In a mouse GBM model, targeting Siglec-E (mouse homolog of Siglec-9) on macrophages can preferentially activate T cells by secreting chemokines, interacting with co-stimulatory factors, and upregulating the IFN-γ-related pathway, thereby enhancing the effectiveness of anti-PD-1/PD-L1 therapy. Siglec-9/Siglec-E represents an immune checkpoint on macrophages and serves as a potential target for GBM immunotherapy.

This study provides new insights for the treatment of glioblastoma, suggesting that Siglec-9 is a novel macrophage immune checkpoint molecule that can be targeted to enhance the efficacy of anti-PD-1/PD-L1 therapy for GBM. The study also demonstrates that combining single-cell analysis of human clinical samples with translational research in mouse models can effectively identify novel immune checkpoints (Figure 1).

Please sign in or register for FREE

If you are a registered user on Research Communities by Springer Nature, please sign in