%0 Journal Article %T Identification of hypoxic macrophages in glioblastoma with therapeutic potential for vasculature normalization. %A Wang W %A Li T %A Cheng Y %A Li F %A Qi S %A Mao M %A Wu J %A Liu Q %A Zhang X %A Li X %A Zhang L %A Qi H %A Yang L %A Yang K %A He Z %A Ding S %A Qin Z %A Yang Y %A Yang X %A Luo C %A Guo Y %A Wang C %A Liu X %A Zhou L %A Liu Y %A Kong W %A Miao J %A Ye S %A Luo M %A An L %A Wang L %A Che L %A Niu Q %A Ma Q %A Zhang X %A Zhang Z %A Hu R %A Feng H %A Ping YF %A Bian XW %A Shi Y %J Cancer Cell %V 42 %N 5 %D 2024 May 13 %M 38640932 %F 38.585 %R 10.1016/j.ccell.2024.03.013 %X Monocyte-derived tumor-associated macrophages (Mo-TAMs) intensively infiltrate diffuse gliomas with remarkable heterogeneity. Using single-cell transcriptomics, we chart a spatially resolved transcriptional landscape of Mo-TAMs across 51 patients with isocitrate dehydrogenase (IDH)-wild-type glioblastomas or IDH-mutant gliomas. We characterize a Mo-TAM subset that is localized to the peri-necrotic niche and skewed by hypoxic niche cues to acquire a hypoxia response signature. Hypoxia-TAM destabilizes endothelial adherens junctions by activating adrenomedullin paracrine signaling, thereby stimulating a hyperpermeable neovasculature that hampers drug delivery in glioblastoma xenografts. Accordingly, genetic ablation or pharmacological blockade of adrenomedullin produced by Hypoxia-TAM restores vascular integrity, improves intratumoral concentration of the anti-tumor agent dabrafenib, and achieves combinatorial therapeutic benefits. Increased proportion of Hypoxia-TAM or adrenomedullin expression is predictive of tumor vessel hyperpermeability and a worse prognosis of glioblastoma. Our findings highlight Mo-TAM diversity and spatial niche-steered Mo-TAM reprogramming in diffuse gliomas and indicate potential therapeutics targeting Hypoxia-TAM to normalize tumor vasculature.