[en] Chemokines and their receptors form a complex interaction network, crucial for precise leukocyte positioning and trafficking. In cancer, they promote malignant cell proliferation and survival but are also critical for immune cell infiltration in the tumor microenvironment. Glioblastoma (GBM) is the most common and lethal brain tumor, characterized by an immunosuppressive TME, with restricted immune cell infiltration. A better understanding of chemokine-receptor interactions is therefore essential for improving tumor immunogenicity. In this study, we assessed the expression of all human chemokines in adult-type diffuse gliomas, with particular focus on GBM, based on patient-derived samples. Publicly available bulk RNA sequencing datasets allowed us to identify the chemokines most abundantly expressed in GBM, with regard to disease severity and across different tumor subregions. To gain insight into the chemokines–receptor network at the single cell resolution, we explored GBmap, a curated resource integrating multiple scRNAseq datasets from different published studies. Our study constitutes the first patient–based handbook highlighting the relevant chemokine–receptor crosstalks, which are of significant interest in the perspective of a therapeutic modulation of the TME in GBM.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
D'Uonnolo, Giulia ✱; Luxembourg Institute of Health > Department of Infection and Immunity > Immuno-Pharmacology and interactomics
Isci, Damla ✱; Université de Liège - ULiège > GIGA
Nosirov, Bakhtiyor; Luxembourg Institute Health > Department of Neuro-Oncology > NORLUX Neuro-Oncology Laboratory
Wantz, May; Luxembourg Institute of Health > Department of Infection and Immunity > Immuno-Pharmacology and Interactomics
Nazarov, Petr; Luxembourg Institute of Health > Department of Cancer Research > Multiomics Data Science Research Group
Golebiewska, Anna; Luxembourg Institute of Health > Department of Cancer Research > NORLUX Neuro-Oncology Laboratory
Rogister, Bernard ; Université de Liège - ULiège > GIGA > GIGA Neurosciences - Nervous system disorders and therapy ; Centre Hospitalier Universitaire de Liège - CHU > > Service de neurologie
Chevigné, Andy ✱; Luxembourg Institute of Health > Department of Infection and Immunity > Immuno-Pharmacology and Interactomics
Louis, D.N., et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 23 (2021), 1231–1251.
Louis, D.N., et al. The 2016 world Health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 131 (2016), 803–820.
Zhang, Y., et al. Prospective genomically guided identification of “early/evolving” and “undersampled” IDH-wildtype glioblastoma leads to improved clinical outcomes. Neuro Oncol. 24 (2022), 1749–1762.
Guo, X., et al. Histological and molecular glioblastoma, IDH-wildtype: a real-world landscape using the 2021 WHO classification of central nervous system tumors. Front. Oncol., 13, 2023, 1200815.
Miller, J.J., et al. Isocitrate dehydrogenase (IDH) mutant gliomas: a Society for Neuro-Oncology (SNO) consensus review on diagnosis, management, and future directions. Neuro Oncol. 25 (2023), 4–25.
Stupp, R., et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352 (2005), 987–996.
Pinson, H., et al. Epidemiology and survival of adult-type diffuse glioma in Belgium during the molecular era. Neuro Oncol., 2023, 10.1093/NEUONC/NOAD158.
Rapp, M., et al. Recurrence pattern analysis of primary glioblastoma. World Neurosurg 103 (2017), 733–740.
Wen, P.Y., Kesari, S., Malignant gliomas in adults. N. Engl. J. Med. 359 (2008), 492–507.
Patel, A.P., et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science, 344, 2014, 1396.
Ou, A., Alfred Yung, W.K., Majd, N., Molecular mechanisms of treatment resistance in glioblastoma. Int. J. Mol. Sci. 22 (2021), 1–24.
Neftel, C., et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178 (2019), 835–849.e21.
Dirkse, A., et al. Stem cell-associated heterogeneity in Glioblastoma results from intrinsic tumor plasticity shaped by the microenvironment. Nat. Commun., 10, 2019.
Yabo, Y.A., Niclou, S.P., Golebiewska, A., Cancer cell heterogeneity and plasticity: a paradigm shift in glioblastoma. Neuro Oncol., 2021, 10.1093/NEUONC/NOAB269.
Jacob, F., et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity. Cell 180 (2020), 188–204.e22.
Osswald, M., et al. Brain tumour cells interconnect to a functional and resistant network. Nature 528 (2015), 93–98.
Gargini, R., Segura-Collar, B., Sánchez-Gómez, P., Cellular plasticity and tumor microenvironment in gliomas: the struggle to hit a moving target. Cancers 12 (2020), 1–24.
Hambardzumyan, D., Gutmann, D.H., Kettenmann, H., The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci., 19, 2016, 20.
Catalano, M., D'Alessandro, G., Trettel, F., Limatola, C., Role of infiltrating microglia/macrophages in glioma. Adv. Exp. Med. Biol. 1202 (2020), 281–298.
Gutmann, D.H., Kettenmann, H., Microglia/brain macrophages as central drivers of brain tumor pathobiology. Neuron, 104, 2019, 442.
Gielen, P.R., et al. Elevated levels of polymorphonuclear myeloid-derived suppressor cells in patients with glioblastoma highly express S100A8/9 and arginase and suppress T cell function. Neuro Oncol., 18, 2016, 1253.
Alban, T.J., et al. Global immune fingerprinting in glioblastoma patient peripheral blood reveals immune-suppression signatures associated with prognosis. JCI Insight, 3, 2018.
Maddison, K., et al. Low tumour-infiltrating lymphocyte density in primary and recurrent glioblastoma. Oncotarget 12 (2021), 2177–2187.
White, K., et al. Identification, validation and biological characterisation of novel glioblastoma tumour microenvironment subtypes: implications for precision immunotherapy. Ann. Oncol. 34 (2023), 300–314.
Yeo, A.T., et al. Single-cell RNA sequencing reveals evolution of immune landscape during glioblastoma progression. Nat. Immunol. 23:6 23 (2022), 971–984 2022.
Kohli, K., Pillarisetty, V.G., Kim, T.S., Key chemokines direct migration of immune cells in solid tumors. Cancer Gene Ther. 29:1 29 (2021), 10–21 2021.
Vilgelm, A.E., Richmond, A., Chemokines modulate immune surveillance in tumorigenesis, metastasis, and response to immunotherapy. Front. Immunol., 10, 2019.
Franciszkiewicz, K., Boissonnas, A., Boutet, M., Combadière, C., Mami-Chouaib, F., Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res. 72 (2012), 6325–6332.
Zlotnik, A., Yoshie, O., The chemokine superfamily revisited. Immunity, 36, 2012, 705.
Bonecchi, R., Graham, G.J., Atypical chemokine receptors and their roles in the resolution of the inflammatory response. Front. Immunol., 7, 2016, 224.
Bachelerie, F., et al. New nomenclature for atypical chemokine receptors. Nat. Immunol. 15 (2014), 207–208.
Mantovani, A., Bonecchi, R., Locati, M., Tuning inflammation and immunity by chemokine sequestration: decoys and more. Nat. Rev. Immunol. 6:12 6 (2006), 907–918 2006.
Chen, K., et al. Chemokines in homeostasis and diseases. Cell. Mol. Immunol. 15 (2018), 324–334.
Blanchet, X., Langer, M., Weber, C., Koenen, R., von Hundelshausen, P., Touch of chemokines. Front. Immunol., 3, 2012.
Groblewska, M., Litman-Zawadzka, A., Mroczko, B., The role of selected chemokines and their receptors in the development of gliomas. Int. J. Mol. Sci., 21, 2020.
Takacs, G.P., Flores-Toro, J.A., Harrison, J.K., Modulation of the chemokine/chemokine receptor axis as a novel approach for glioma therapy. Pharmacol. Ther., 222, 2021.
Zhou, Y., Larsen, P.H., Hao, C., Yong, V.W., CXCR4 is a major chemokine receptor on glioma cells and mediates their survival. J. Biol. Chem. 277 (2002), 49481–49487.
Isci, D., et al. Patient-oriented perspective on chemokine receptor expression and function in glioma. Cancers, 14, 2022, 130.
Ceccarelli, M., et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164 (2016), 550–563.
Tang, Z., Kang, B., Li, C., Chen, T., Zhang, Z., GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47 (2019), W556–W560.
Puchalski, R.B., et al. An anatomic transcriptional atlas of human glioblastoma. Science 360 (2018), 660–663.
Ruiz-Moreno, C., et al. Harmonized Single-Cell Landscape, Intercellular Crosstalk and Tumor Architecture of Glioblastoma. 2022, 10.1101/2022.08.27.505439.
GitHub - sqjin/CellChat: R toolkit for inference, visualization and analysis of cell-cell communication from single-cell data. https://github.com/sqjin/CellChat.
Wickham, H., ggpolt2 elegant graphics for data analysis. Use R! series, 2016, 211.
Arranging plots in a grid • cowplot. https://wilkelab.org/cowplot/articles/plot_grid.html.
Authors and Citation • patchwork. https://patchwork.data-imaginist.com/authors.html#citation.
Darmanis, S., et al. Single-cell RNA-seq analysis of infiltrating neoplastic cells at the migrating front of human glioblastoma. Cell Rep., 21, 2017, 1399.
Erreni, M., et al. Human glioblastoma tumours and neural cancer stem cells express the chemokine CX3CL1 and its receptor CX3CR1. Eur. J. Cancer 46 (2010), 3383–3392.
Desbaillets, I., et al. Human astrocytomas and glioblastomas express monocyte chemoattractant protein-1 (MCP-1) in vivo and in vitro. Int. J. Cancer 58 (1994), 240–247.
Khandani, B.K., et al. The intricate expression of CC chemokines in glial tumors: evidence for involvement of CCL2 and CCL5 but not CCL11. Acta Med. Iran. 53 (2015), 770–777.
Kokovay, E., et al. Adult SVZ lineage cells home to and leave the vascular niche via differential responses to SDF1/CXCR4 signaling. Cell Stem Cell 7 (2010), 163–173.
Goffart, N., et al. Adult mouse subventricular zones stimulate glioblastoma stem cells specific invasion through CXCL12/CXCR4 signaling. Neuro Oncol., 17, 2015, 81.
Gatti, M., et al. Inhibition of CXCL12/CXCR4 autocrine/paracrine loop reduces viability of human glioblastoma stem-like cells affecting self-renewal activity. Toxicology 314 (2013), 209–220.
Lee, E.Q., et al. Phase I and biomarker study of plerixafor and bevacizumab in recurrent high-grade glioma. Clin. Cancer Res. 24 (2018), 4643–4649.
Thomas, R.P., et al. Macrophage exclusion after radiation therapy (MERT): a first in human phase I/II trial using a CXCR4 inhibitor in glioblastoma. Clin. Cancer Res. 25 (2019), 6948–6957.
K, H., et al. The chemokine receptor CXCR7 is highly expressed in human glioma cells and mediates antiapoptotic effects. Cancer Res. 70 (2010), 3299–3308.
Neves, M., et al. The role of ACKR3 in breast, lung, and brain cancer. Mol. Pharmacol. 96 (2019), 819–825.
Salazar, N., et al. A chimeric antibody against ACKR3/CXCR7 in combination with TMZ activates immune responses and extends survival in mouse GBM models. Mol. Ther., 26, 2018, 1354.
Jacobs, S.M., et al. CXCR4 expression in glioblastoma tissue and the potential for PET imaging and treatment with [68Ga]Ga-Pentixafor/[177Lu]Lu-Pentixather. Eur. J. Nucl. Med. Mol. Imag. 49 (2022), 481–491.
Adamski, V., et al. Entry and exit of chemotherapeutically-promoted cellular dormancy in glioblastoma cells is differentially affected by the chemokines CXCL12, CXCL16, and CX3CL1. Oncogene 39:22 39 (2020), 4421–4435 2020.
Nitta, T., Allegretta, M., Okumura, K., Sato, K., Steinman, L., Neoplastic and reactive human astrocytes express interleukin-8 gene. Neurosurg. Rev. 15 (1992), 203–207.
Sharma, I., Singh, A., Siraj, F., Saxena, S., IL-8/CXCR1/2 signalling promotes tumor cell proliferation, invasion and vascular mimicry in glioblastoma. J. Biomed. Sci., 25, 2018.
Infanger, D.W., et al. Glioblastoma stem cells are regulated by interleukin-8 signaling in a tumoral perivascular niche. Cancer Res. 73 (2013), 7079–7089.
Kumar, A., et al. CXCL14 promotes a robust brain tumor-associated immune response in glioma. Clin. Cancer Res. 28 (2022), 2898–2910.
Kranjc, M.K., Novak, M., Pestell, R.G., Lah, T.T., Cytokine CCL5 and receptor CCR5 Axis in glioblastoma multiforme. Radiol. Oncol., 53, 2019, 397.
Wang, Y., et al. Hypoxia and macrophages promote glioblastoma invasion by the CCL4-CCR5 axis. Oncol. Rep. 36 (2016), 3522–3528.
Zhang, X., et al. CCL8 secreted by tumor-associated macrophages promotes invasion and stemness of glioblastoma cells via ERK1/2 signaling. Lab. Invest. 100 (2020), 619–629.
Hu, J., et al. Regulation of tumor immune suppression and cancer cell survival by CXCL1/2 elevation in glioblastoma multiforme. Sci. Adv., 7, 2021.
Alafate, W., et al. Elevation of CXCL1 indicates poor prognosis and radioresistance by inducing mesenchymal transition in glioblastoma. CNS Neurosci. Ther. 26 (2020), 475–485.
Wang, L., et al. Overexpression of CCL20 and its receptor CCR6 predicts poor clinical prognosis in human gliomas. Med. Oncol. 29 (2012), 3491–3497.
Shono, K., et al. Downregulation of the CCL2/CCR2 and CXCL10/CXCR3 axes contributes to antitumor effects in a mouse model of malignant glioma. Sci. Rep., 10, 2020.
Wang, G., et al. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol. Ther., 2022, 10.1016/J.YMTHE.2022.08.021.
Wang, Z., et al. The CXCL family contributes to immunosuppressive microenvironment in gliomas and assists in gliomas chemotherapy. Front. Immunol., 12, 2021.
Chan, T.Y.H., Wong, J.S.Y., Kiang, K.M.-Y., Sun, C.W.Y., Leung, G.K.-K., The duality of CXCR3 in glioblastoma: unveiling autocrine and paracrine mechanisms for novel therapeutic approaches. Cell Death Dis. 14:12 14 (2023), 1–13 2023.
Chui, R., Dorovini-Zis, K., Regulation of CCL2 and CCL3 expression in human brain endothelial cells by cytokines and lipopolysaccharide. J. Neuroinflammation 7 (2010), 1–12.
Subileau, E.A., et al. Expression of chemokines and their receptors by human brain endothelium: implications for multiple sclerosis. J. Neuropathol. Exp. Neurol. 68 (2009), 227–240.
Xie, Y., et al. Key molecular alterations in endothelial cells in human glioblastoma uncovered through single-cell RNA sequencing. JCI Insight, 6, 2021.
Liao, Y.Y., et al. CCL3 promotes angiogenesis by dysregulation of miR-374b/VEGF-A axis in human osteosarcoma cells. Oncotarget, 7, 2016, 4310.
Bajetto, A., et al. Expression of CXC chemokine receptors 1-5 and their ligands in human glioma tissues: role of CXCR4 and SDF1 in glioma cell proliferation and migration. Neurochem. Int. 49 (2006), 423–432.
Zagzag, D., et al. Hypoxia- and vascular endothelial growth factor-induced stromal cell-derived factor-1α/CXCR4 expression in glioblastomas: one plausible explanation of scherer's structures. Am. J. Pathol., 173, 2008, 545.
Identification, Localization of the Cytokine SDF1 and Its Receptor, CXC Chemokine Receptor 4, to Regions of Necrosis and Angiogenesis in Human Glioblastoma |. Clin. Cancer Res. 6 (2000), 102–111.
Rao, S., et al. CXCL12 mediates trophic interactions between endothelial and tumor cells in glioblastoma. PLoS One, 7, 2012.
Salmaggi, A., et al. CXCL12 in malignant glial tumors: a possible role in angiogenesis and cross-talk between endothelial and tumoral cells. J. Neuro Oncol. 67 (2004), 305–317.
Dai, D., et al. miR-24 regulates angiogenesis in gliomas. Mol. Med. Rep. 18 (2018), 358–368.
Guo, X., Jiao, H., Cao, L., Meng, F., Biological implications and clinical potential of invasion and migration related miRNAs in glioma. Front. Integr. Neurosci., 16, 2022.
Oishi, T., Koizumi, S., Kurozumi, K., Molecular mechanisms and clinical challenges of glioma invasion. Brain Sci., 12, 2022.
So, J.S., Kim, H., Han, K.S., Mechanisms of invasion in glioblastoma: extracellular matrix, Ca2+ signaling, and glutamate. Front. Cell. Neurosci., 15, 2021.
Li, M., Ransohoff, R.M., Multiple roles of chemokine CXCL12 in the central nervous system: a migration from immunology to neurobiology. Prog Neurobiol 84 (2008), 116–131.
Mithal, D.S., Banisadr, G., Miller, R.J., CXCL12 signaling in the development of the nervous system. J. Neuroimmune Pharmacol., 7, 2012, 820.
Dar, A., Kollet, O., Lapidot, T., Mutual, reciprocal SDF-1/CXCR4 interactions between hematopoietic and bone marrow stromal cells regulate human stem cell migration and development in NOD/SCID chimeric mice. Exp. Hematol. 34 (2006), 967–975.
Park, J.H., Lee, H.K., Current understanding of hypoxia in glioblastoma multiforme and its response to immunotherapy. Cancers, 14, 2022.
Domènech, M., Hernández, A., Plaja, A., Martínez‐balibrea, E., Balañà, C., Hypoxia: the cornerstone of glioblastoma. Int. J. Mol. Sci., 22, 2021.
Ahn, S.H., et al. Necrotic cells influence migration and invasion of glioblastoma via NF-κB/AP-1-mediated IL-8 regulation. Sci. Rep. 6:1 6 (2016), 1–12 2016.
Brat, D.J., Bellail, A.C., Van Meir, E.G., The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro Oncol. 7 (2005), 122–133.
Urbantat, R.M., et al. The CXCL2/IL8/CXCR2 pathway is relevant for brain tumor malignancy and endothelial cell function. Int. J. Mol. Sci. 22 (2021), 1–19.
Conroy, S., Kruyt, F.A.E., Wagemakers, M., Bhat, K.P.L., den Dunnen, W.F.A., IL-8 associates with a pro-angiogenic and mesenchymal subtype in glioblastoma. Oncotarget, 9, 2018, 15721.
Jung, Y., et al. MCP-1 and MIP-3α secreted from necrotic cell-treated glioblastoma cells promote migration/infiltration of microglia. Cell. Physiol. Biochem. 48 (2018), 1332–1346.
Jin, P., et al. Astrocyte-derived CCL20 reinforces HIF-1-mediated hypoxic responses in glioblastoma by stimulating the CCR6-NF-κB signaling pathway. Oncogene 37:23 37 (2018), 3070–3087 2018.
Chang, A.L., et al. CCL2 produced by the glioma microenvironment is essential for the recruitment of regulatory T cells and myeloid-derived suppressor cells. Cancer Res. 76 (2016), 5671–5682.
Böttcher, J.P., et al. NK cells stimulate recruitment of cDC 1 into the tumor microenvironment promoting cancer immune control. Cell 172 (2018), 1022–1037.e14.
Chow, M.T., Luster, A.D., Chemokines in cancer. Cancer Immunol. Res. 2 (2014), 1125–1131.
Ozga, A.J., Chow, M.T., Luster, A.D., Chemokines and the immune response to cancer. Immunity 54 (2021), 859–874.
Nagarsheth, N., Wicha, M.S., Zou, W., Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 17:9 17 (2017), 559–572 2017.
Yu, K., et al. Surveying brain tumor heterogeneity by single-cell RNA-sequencing of multi-sector biopsies. Natl. Sci. Rev. 7 (2020), 1306–1318.
Lepore, F., et al. CXCL16/CXCR6 axis drives microglia/macrophages phenotype in physiological conditions and plays a crucial role in glioma. Front. Immunol., 9, 2018, 2750.
Kollis, P.M., et al. Characterising distinct migratory profiles of infiltrating T-cell subsets in human glioblastoma. Front. Immunol., 13, 2022.
Arrieta, V.A., et al. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J. Clin. Invest., 133, 2023.
Karimi, E., et al. Single-cell spatial immune landscapes of primary and metastatic brain tumours. Nature 614:7948 614 (2023), 555–563 2023.
Wischnewski, V., et al. Phenotypic diversity of T cells in human primary and metastatic brain tumors revealed by multiomic interrogation. Nature Cancer 4:6 4 (2023), 908–924 2023.
Yang, S., et al. Relationship between chemokine/chemokine receptor and glioma prognosis and outcomes: systematic review and meta-analysis. Int. Immunopharm., 133, 2024, 112047.