[go: up one dir, main page]
More Web Proxy on the site http://driver.im/ Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Nov 1.
Published in final edited form as: Cell. 2020 May 28;181(7):1643–1660.e17. doi: 10.1016/j.cell.2020.05.007

Interrogation of the Microenvironmental Landscape in Brain Tumors Reveals Disease-Specific Alterations of Immune Cells

Florian Klemm 1,2, Roeltje R Maas 1,2,3,4, Robert L Bowman 5, Mara Kornete 1,2, Klara Soukup 1,2, Sina Nassiri 1,2,6, Jean-Philippe Brouland 7, Christine A Iacobuzio-Donahue 8, Cameron Brennan 9, Viviane Tabar 9, Philip H Gutin 9, Roy T Daniel 4, Monika E Hegi 3,4, Johanna A Joyce 1,2,10,#
PMCID: PMC8558904  NIHMSID: NIHMS1740100  PMID: 32470396

Summary

Brain malignancies encompass a range of primary and metastatic cancers including low-grade and high-grade gliomas, and brain metastases (BrMs) originating from diverse extracranial tumors. Our understanding of the brain tumor microenvironment (TME) remains limited and it is unknown whether it is sculpted differentially by primary versus metastatic disease. We therefore comprehensively analyzed the brain TME landscape via flow cytometry, RNA sequencing, protein arrays, culture assays, and spatial tissue characterization. This revealed disease-specific enrichment of immune cells with pronounced differences in proportional abundance of tissue-resident microglia, infiltrating monocyte-derived macrophages, neutrophils and T cells. These integrated analyses also uncovered multifaceted immune cell activation within brain malignancies entailing converging transcriptional trajectories while maintaining disease- and cell type-specific programs. Given the interest in developing TME-targeted therapies for brain malignancies this comprehensive resource of the immune landscape offers insights into possible strategies to overcome tumor-supporting TME properties and instead harness the TME to fight cancer.

Keywords: glioma, glioblastoma, brain metastasis, tumor microenvironment, cancer immunology, microglia, macrophages, monocyte-derived macrophages, tumor-associated macrophages, neutrophils, T cells

Graphical Abstract

graphic file with name nihms-1740100-f0001.jpg

Introduction

Brain malignancies include tumors that arise within the brain, such as low-grade gliomas and glioblastomas, and brain metastases (BrMs) which originate from extracranial primary tumors, including melanoma, breast, and lung cancers (Cagney et al., 2017). Gliomas mutant for the metabolic enzymes isocitrate dehydrogenase 1 and 2 (IDH mut) are generally low-grade (II/III) and have a significantly better prognosis than IDH wildtype (wt) tumors which are typically grade IV glioblastomas. Despite standard of care treatment comprising surgery followed by radiation and temozolomide (Stupp et al., 2005), median survival rates for glioblastoma patients remain stubbornly low (Aldape et al., 2019). Patient survival following BrM diagnosis can be even lower, with rates typically measured in months (Cagney et al., 2017; Ceccarelli et al., 2016), and among all adult brain tumors the incidence of BrMs significantly exceeds that of gliomas.

Given the current limited treatment options for these patients, a key question to address is whether a deep comprehensive understanding of how primary and metastatic cancers develop within the brain tumor microenvironment (TME) could reveal promising new targets for therapeutic intervention. While diverse TME cell types can critically regulate cancer progression and response to therapy across a broad range of extracranial tumors (Klemm and Joyce, 2015), we cannot simply extrapolate findings from these cancers to the singular brain TME given its unique cell types including astrocytes, neurons and microglia (MG); the immune-suppressive environment of this organ; and the challenges presented for cells and drugs to cross the blood-brain barrier (BBB) (Quail and Joyce, 2017).

Immune checkpoint blockade (ICB), adoptive cell therapy and vaccines represent treatments targeted against immune cells both within the TME and systemically. The success of immunotherapies in certain extracranial cancers has led to a clear motivation for their evaluation in brain malignancies. However, while showing some clinical efficacy in a subset of BrM patients (Hendriks et al., 2019; Long et al., 2018; Tawbi et al., 2018), ICB has only resulted in responses in isolated cases of primary gliomas to date (Lim et al., 2018; Schalper et al., 2019). Beyond tumor cell-intrinsic effects, this may be attributed in part to immune-suppressive components of the brain TME, including tumor-associated macrophages (TAMs), which have emerged as prominent players in brain cancers (Gutmann and Kettenmann, 2019; Quail and Joyce, 2017).

Lineage-tracing experiments in mice revealed that brain TAMs can originate from either tissue-resident MG or monocyte-derived macrophages (MDM) recruited from the peripheral circulation (Bowman et al., 2016; Chen et al., 2017). TAMs are highly plastic cells that integrate input from cytokines, growth factors and other stimuli resulting in diverse activation states and cellular phenotypes, including the promotion of invasion, angiogenesis, metastasis and immune suppression (Mantovani et al., 2017; Noy and Pollard, 2014). This plasticity and their position at the nexus between malignant cells and tumor-infiltrating T cells makes TAMs a promising target of TME-directed therapies in different cancers. Indeed, studies in mice showed that phenotypic alteration of TAMs resulted in anti-tumor efficacy in glioblastoma (Pyonteck et al., 2013; Quail et al., 2016; Yan et al., 2017), while TAM depletion prevented BrM outgrowth (Qiao et al., 2019).

Despite these preclinical studies, the precise contribution of the two ontogenetically distinct TAM cell types in human brain malignancies is unclear, which hinders clinical translation. For example, previous studies interrogating the role of TAMs in patient brain tumors did not distinguish between MG and MDM based on use of lineage tracing-derived markers (Gabrusiewicz et al., 2016; Sankowski et al., 2019; Szulzewsky et al., 2016) or focused solely on gliomas (Muller et al., 2017; Venteicher et al., 2017). We therefore interrogated the TME landscape in both gliomas and BrMs, with an emphasis on exploring TAMs, while also investigating their relation to other immune cells and structures in the TME. We leveraged this multimodal resource to address a number of questions including: Do tumors arising within the brain shape their TME differently to cancers that metastasize from extracranial sites? Does IDH mutation status impact the TME? How do distinct TME compositions potentially modulate the activation states of immune cells? By integrating the answers to these questions herein we provide insights into potential strategies to harness the brain TME in the fight against these deadly diseases.

Results

The immune composition of brain malignancies

We first determined the broad immune cell abundance in the brain TME by analyzing the pan-leukocyte marker CD45 through immunofluorescence (IF) staining of whole tissue sections and flow cytometry (FCM) analyses of non-tumor brain tissue, IDH mut low-grade and IDH wt high-grade gliomas, and BrMs originating from different primaries including breast cancer, lung cancer and melanoma (Figures 1A, 1B and S1A). This showed a leukocyte abundance from ~20–40% across the cancer samples. Stratification of CD45+ cells into myeloid and lymphoid lineages revealed a significant increase in myeloid cells in IDH mut and IDH wt gliomas, and of lymphocytes in IDH wt tumors and BrMs compared to non-tumor tissue (Figure 1B, p<0.05, one-sided Student’s t-test). We used multicolor fluorescence-activated cell sorting (FACS) to analyze 14 major immune cell populations across 100 clinical samples (Figure S1A, Tables S1 and S2), and collected cells for RNA sequencing (RNA-seq) from 48 patients (Table S3, full clinical annotation).

Figure 1. The immune cell composition of brain malignancies.

Figure 1.

(A) Quantification of immunofluorescence (IF) staining of non-immune (CD45−) and immune cells (CD45+) in sections of non-tumor brain tissue (n=6), gliomas (nIDHmut=16, nIDHwt=16), and brain metastases (BrM, nbreast=12, nlung=5, nmelanoma=7). (B) Flow cytometry (FCM) quantification of non-immune cells (CD45−), myeloid cells (CD45+, CD11B+) and lymphocytes (CD45+, CD11B−) in non-tumor tissue (n=6), gliomas (nIDHmut=17, nIDHwt=40) and BrM (nbreast=13, nlung=16, nmelanoma=8). (C) Gene set variation analysis (GSVA) normalized enrichment score (NES) of MG and MDM ontogeny-specific core gene signatures in CD49Dlow MG and CD49Dhigh MDM from non-tumor and tumor tissues. (D) Heatmap of immune cell proportions in relation to all CD45+ cells (MG=microglia, MDM=monocyte-derived macrophages, CD14low/CD16+=CD14low/CD16+ monocytes, CD14+/CD16+=CD14+/CD16+ monocytes, CD16− Gran.=CD16− granulocytes, iMC=immature myeloid cells, DC=dendritic cells, Treg=regulatory T cells, DNT=double negative T cells) across the cohort (nnon-tumor=6, nglioma=57, nBrM=37). Cluster assignment, disease type, IDH mutation status, and BrM primary tumor annotated per column (clinical information, Table S1). (E) Principal component (PC) biplot of FCM data with sample scores and top 5 loadings of the first two PCs (n=100 clinical samples, proportion of variance shown on PC axes). (F) Mean of immune cell populations in non-tumor tissue (n=6), gliomas (nIDHmut=17, nIDHwt=40), and BrMs (nbreast=13, nlung=16, nmelanoma=8) as percentage of CD45+ cells.

See also Figure S1 and Table S1 and S2.

By incorporating cell lineage tracing and mouse models of high-grade gliomas and BrM, we previously identified the cell surface marker integrin alpha 4, ITGA4/CD49D, as a means to discriminate tumor-associated MG (T-MG) from tumor-associated MDM (T-MDM) (Bowman et al., 2016), which we integrated into clinical sample analyses herein. This enabled sorting of CD45− non-immune cells, CD49Dlow MG, CD49Dhigh MDM, neutrophils, CD4+ and CD8+ T cells (Figure S1A, Tables S2 and S3A) for transcriptome analysis by RNA-seq. We assessed sorting fidelity by FCM re-analysis of the sorted CD49Dlow and CD49Dhigh TAM populations (purity 98.4–99.8%), and by investigating the frequency of the canonical IDH codon 132 missense mutation in the RNA-seq reads from CD45− cells, CD49Dlow and CD49Dhigh TAM populations. While we observed a mean mutated allele frequency of 0.43 in CD45− cells from IDH mut gliomas (range 0.3 – 0.61), this was very rare in TAMs (mean = 0.01, range 0.0 – 0.09), indicating a reliable separation of cell populations. In a t-distributed stochastic neighbor embedding (t-SNE) visualization of sorted populations, samples clustered mostly by cell type (Figure S1B) with gliomas and BrMs discernible as separate groups in the CD45− population.

In this global expression analysis within the context of the other major brain TME components, CD49Dlow and CD49Dhigh TAM populations clustered closely, suggesting a broad transcriptomic similarity. We thus further interrogated the utility of CD49D to differentiate between TAM populations by analyzing association of MG- and MDM-specific ontogeny core gene sets, previously identified from lineage-tracing studies (Bowman et al., 2016), in human CD49Dlow and CD49Dhigh cells sorted from non-malignant and brain cancer tissues. This revealed an enrichment of ontogeny core gene sets in the corresponding cell type (Figure 1C), demonstrating our ability to accurately distinguish MG and MDM in human samples across different disease entities. Interestingly, these core signatures were influenced within certain tumor types, with T-MDM showing an increased MG core gene set signal in IDH mut gliomas, and T-MG acquiring MDM features in BrMs, suggesting a tissue-dependent transcriptional programming of these cells, as further interrogated below.

We next assessed the landscape of intratumoral immune cell populations (Figure S1A and Table S2) using clustering analysis to identify patterns of cellular abundance (Figure 1D, Chi-square test for independence, p value < 0.0001). This revealed three major clusters: (1) non-tumor samples and IDH mut gliomas characterized by a dominance of MG with low numbers of other immune cells; (2) IDH wt gliomas, and several BrMs, with an influx of MDM and to some extent neutrophils into the tumor, while mostly excluding lymphocytes; (3) predominantly BrMs, and few IDH wt gliomas, exhibiting the most diverse immune cell landscape with a substantial infiltration of T cells and neutrophils. Certain tumors contained CD14low/CD16+ non-classical monocytes, CD14+/CD16+ intermediate monocytes, CD16 granulocytes, dendritic cells (DC), or immature myeloid cells. Across all samples the lymphocyte compartment was mostly composed of T cells with fewer NK cells and B cells.

Principal component analysis (PCA) of the relative abundance of all investigated populations confirmed that MG, MDM, neutrophils, CD4+ and CD8+ T cells are the major immune cell determinants of the brain TME landscape (Figure 1E). PC1 separated non-tumor tissue and IDH mut gliomas from IDH wt gliomas and BrMs, while PC2 distinguished IDH wt gliomas and BrMs. Further analysis stratifying for IDH status in gliomas and the primary tumor site in BrMs verified a substantially higher proportion of lymphocytes in BrMs (Figure 1F, meanlymphocytes %CD45+ = 46.23 %, SEM = 4.15, t-test, p < 0.0001). Melanoma-BrMs exhibited the most abundant lymphocyte infiltrate with a sizeable CD8+ T cell fraction (meanCD8+ %CD45+ = 33.01 %, SEM = 5.82, one-way ANOVA, p < 0.01). Regulatory T cells (Tregs) were detected in certain BrMs (meanTreg %CD45+=1.2 %, SEM = 0.36), while rare in gliomas (meanTreg %CD45+=0.25 %, SEM = 0.05, t-test, p < 0.05).

Because of the prominence of T-MG and T-MDM in the myeloid compartment of brain malignancies we used IF staining and deconvolution analyses to independently validate their presence. Commonly employed MG markers such as P2RY12, TMEM119, and SALL1, and MDM-associated genes such as AHR and VDR, showed varying RNA expression levels across different brain malignancies while maintaining their cell type-specificity (Figure S2A), in a similar manner as observed for the ontogeny core gene sets (Figure 1C). An equivalent pattern was observed at the protein level (Figure S2B), where P2RY12 showed the highest expression in non-tumor tissue and CD68 was most abundant in BrM TAM populations. This necessitated the use of both markers, complemented with CD49D, to reliably identify MG and MDM in IF analyses (Figure S2C). We used this strategy to interrogate a cohort of non-tumor, glioma and BrM samples by whole-section quantification, confirming MDM accumulation in IDH wt gliomas and BrMs (Figure 2AC). Furthermore, comparison of tissue processed independently for IF and FCM from the same individual samples demonstrated significant concordance (Figure S2D).

Figure 2. Analysis of MG and MDM abundance.

Figure 2.

(A) Representative IF images and (B) corresponding cell type identification of MG (CD45+, P2RY12+/CD68+, CD49D−), MDM (CD45+, P2RY12+/CD68+, CD49D+), non-immune (CD45−) and non-TAM-immune cells (CD45+, P2RY12−/CD68−, CD49D−/+) in non-tumor brain tissue, IDH mut, IDH wt gliomas and BrMs. Scale bars = 100µm, insets show quantification per field of view (FOV). (C) IF quantification of MG and MDM abundance in non-tumor brain tissue (n=6), IDH mut (n=16), IDH wt (n=16) gliomas, and BrMs (n=24). (D) EPIC deconvolution of merged GTEX and TCGA glioma datasets showing relative abundance of MG, MDM and non-TAMs (“other cells”) in healthy frontal cortex, IDH mut and IDH wt gliomas. Wilcoxon rank-sum test used for statistical analysis: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

See also Figure S2.

We queried the sorted cell populations for T-MG- and T-MDM-specific differentially expressed genes (DEG) that separate these two populations from the most abundant other cell types, i.e. CD45− cells, neutrophils and T cells (Figure S2E). Several of the genes highly expressed in T-MG are well-established MG markers (P2RY12, TMEM119, TAL1), while genes highly expressed in T-MDM include markers of alternative macrophage polarization (FCGR2B, CLEC10A) and DC-like phenotypes (CD1C, CD1B, CD207) with increased phagocytic- and antigen cross-presentation ability (CD209). These gene sets also allowed us to utilize a publicly available integrated dataset (Vivian et al., 2017) containing bulk expression data of healthy cortical brain tissue from the Genotype-Tissue Expression project (GTEx, The GTEx Consortium, 2013), and low- and high-grade glioma samples from The Cancer Genome Atlas (TCGA, Ceccarelli et al., 2016) in a bulk tissue transcriptome deconvolution approach (Racle et al., 2017). The estimates obtained of MG and MDM proportions in this external dataset (n=711 samples) verified the prevalence of MG in IDH mut gliomas and MDM enrichment in IDH wt gliomas (Figure 2D).

MG and MDM exhibit a multifaceted polarization phenotype in brain malignancies

We next employed PCA to specifically focus on TAMs and analyze genes whose expression was influenced by tissue type (i.e. reference MDMs, non-tumor brain, gliomas and BrMs) and cell type (i.e. MG and MDM) (Figure 3A). Within the first two PCs, MG and MDM projected into different spaces, with in vitro differentiated MDMs distinct from tissue-derived samples. We observed a gradient across PC1 with non-tumor brain tissue at one end, traversing IDH mut and IDH wt gliomas, and ending with BrMs. Thus, TAM transcriptomic changes are influenced by the brain TME per se and also by the specific type of malignancy.

Figure 3. MG and MDM exhibit a multifaceted polarization phenotype in brain malignancies.

Figure 3.

(A) PC biplot of MG and MDM transcriptome data from non-tumor brain tissue, IDH mut and IDH wt gliomas, and BrMs (clinical information, Table S3A, reference = in vitro generated MDMs, proportion of variance shown on PC axes). (B) Visualization of intersects of the conserved sets of significantly upregulated genes in MG and MDM. Intersects between sets are shown in the combination matrix. ngenes found uniquely in a gene set or intersect indicated above individual bars. (C) Stimulus-specific macrophage gene expression modules overrepresented (within conserved differentially expressed genes (DEGs) vs. respective references) in tumor associated-MG (T-MG) and -MDM (T-MDM). Bar heights and color indicate significance level. (GC = glucocorticoid, IFNγ = interferon gamma, LA = lauric acid, LiA = linoleic acid, OA = oleic acid, PA = palmitic acid, PGE2 = prostaglandin E2, sLPS = standard lipopolysaccharide, TNFα = tumor necrosis factor alpha, TPP = TNFα+PGE2+Pam3CysSerLys4, IL10 = interleukin 10). (D) Heatmap of gene ontology overrepresentation analysis of leading edge metagenes (LEM) in MG and MDM from gliomas and BrMs. Tile fill indicates significance (hypergeometric test,-log10(adjusted p value), terms were filtered by significance). (E) IF quantification of proportion of proliferating Ki67+ MG and MDM in non-tumor tissue (n=5), IDH mut (n=10) and IDH wt gliomas (n=9) and BrMs (n=8). Means compared with one-tailed t test: *p < 0.05. (F) qRT-PCR of Type I IFN-LEM marker genes from Group 2 (see Figure S3F) in in vitro generated MDMs stimulated with the indicated tumor microenvironment culture-conditioned media (TME-CM). Fold-changes calculated relative to colony stimulating factor-1 (CSF-1)-treated MDM baseline (one-way ANOVA p value < 0.1, nMDM = 4–11).

See also Figure S3 and Table S4.

We contrasted T-MG and T-MDM from BrMs or gliomas (regardless of IDH mutation status) to MG from non-tumor brain or in vitro differentiated MDM from healthy donors respectively (Figure S3A, Table S3A, Table S4). This revealed profound expression changes in both populations, with T-MDM exhibiting a higher magnitude in their transcriptional response compared to T-MG (Figure S3A). The intersect of DEGs in gliomas and BrMs was highest in T-MDM (Figure S3B), potentially reflecting the greater changes experienced by these cells upon entering the completely foreign environment of a brain tumor. This was also evident when focusing on genes upregulated in both glioma and BrMs that are private to either T-MG or T-MDM (Figure 3B). In both T-MG and T-MDM the number of shared genes was higher across different diseases than between these two cell populations within the same tumor type. Consequently, only a small number of genes (n=137) showed concordant upregulation across a comparison of all diseases and TAM types (Figure 3B).

To explore the underlying biological processes conserved in both gliomas and BrMs we examined the intersect of upregulated genes (Figure S3B) in T-MG or T-MDM using gene set over-representation analysis (ORA). In the Molecular Signature Database (MSigDB, Liberzon et al., 2015) “hallmark” collection of major biological categories both T-MG and T-MDM showed pathway enrichment in (i) modeling of the TME (“Angiogenesis”, “Hypoxia”), (ii) inflammation (“Inflammatory Response”, “Allograft Rejection”), and (iii) immune cell activation states (“TNFα Signaling via NFκB”, “Interferon α Response”, “Interferon γ Response”, “IL2 STAT5 Signaling” and “IL6 JAK STAT3 Signaling”) (Figure S3C).

We also assessed the M1 and M2 polarization status of T-MG and T-MDM using a panel of marker genes (Murray et al., 2014). However, no evident pattern emerged of a defined M1 vs. M2 phenotype in either glioma- or BrM T-MG or T-MDM (Figure S3D). To further explore the activation state of T-MG and T-MDM, we subjected their respective upregulated genes to ORA of macrophage stimulus-specific programs (Xue et al., 2014). This revealed a multifaceted response (Figure 3C) incorporating both canonical M1 (IFNγ) and M2 polarization (IL4), including expression changes associated with chronic inflammatory stimuli (TNFα + prostaglandin E (PGE2), and TNFα + PGE2 + Pam3CysSerLys4 (TPP)) and exposure to free fatty acids (oleic acid (OA), palmitic acid (PA)), which have been implicated in modulating myeloid cell function (Thapa and Lee, 2019). This indicates a diverse transcriptional programming of T-MG and T-MDM in gliomas and BrMs extending beyond a simple M1 vs. M2 polarization.

To understand which processes are linked to, and potentially driving, these responses we identified the gene set enrichment (GSEA, Subramanian et al., 2005) leading edge genes in T-MG and T-MDM in gliomas and BrMs, and clustered them into leading edge metagenes (LEMs) with non-negative matrix factorization (Godec et al., 2016). This identified up to 5 distinct LEMs per cell type and comparison that were tested for significant overlap in a pairwise fashion (Figure S3E), and annotated using Gene Ontology (GO) terms (Figure 3D). LEMs associated with mitosis and cell proliferation were present in both T-MG and T-MDM in gliomas and BrMs (Figure 3D, Group 1). The biological validity of these LEMs were verified by staining for Ki-67, a marker of cell proliferation, in non-tumor, glioma and BrM tissue sections (Figure 3E), showing increased proliferation in both T-MG and T-MDM in IDH wt gliomas and BrMs, and in T-MG in IDH mut gliomas.

Interestingly, LEMs enriched for Type I Interferon (IFN) signaling were detected in glioma and BrM T-MDM and in BrM T-MG, but not in glioma T-MG (Figure 3D, Group 2). Sustained Type I IFN signaling has been implicated in mediating immune suppression and ICB resistance (Benci et al., 2016). The stringency of these Group 2 LEMs was validated by building a protein-protein interaction (PPI) network of the shared LEM genes (Figure S3F). Beyond their role in antiviral responses, the genes highlighted at the center of the PPI network (Figure S3F, red nodes) have been implicated in a variety of both tumor promoting and suppressing roles (Benci et al., 2016). Similarly, the more peripheral network nodes IL15 and TNFSF10 are potentially able to modulate an effective immunological anti-tumor response or induce apoptosis in cancer cells respectively (Bouralexis et al., 2005; Santana Carrero et al., 2019). We asked if these genes were directly induced by secreted factors in the brain TME, and established cell-based assays to expose MDMs to TME-conditioned medium (CM) generated from single cell suspensions of freshly-isolated glioma or BrM samples in culture. All genes analyzed were upregulated by BrM-TME-CM, and to a lesser extent by Glioma-TME-CM (Figure 3F). We also detected induction of Inflammation- and NFκB-signaling-associated LEMs in BrM-MG, glioma-MDM and BrM-MDM (Figure 3D, Group 3). LEMs that point towards a Th17 response (Group 4), and the recruitment of immune cells and interactions between different immune cell compartments were exclusively detected in MDM (Group 5). Collectively, these analyses reveal an acquisition of a multifaceted activation state of MG and MDM upon their integration into the TME of brain malignancies.

IDH mutation status associated with changes in glioma TAM activation

We next asked whether MG and MDM occupy distinct regions within the TME of IDH wt gliomas. Spatial analysis of tissue sections showed significant enrichment of both populations in the perivascular niche (Figures 4A and S4A). Analysis of their distribution relative to CD31+ vascular structures showed a closer proximity of T-MDM compared to T-MG (Figures 4B and S4A). Interrogation of anatomical transcriptome data from the Ivy GAP study (Puchalski et al., 2018) also demonstrated an enrichment of T-MDM in the microvascular compartment (Figure S4B). This enrichment coincided with CD4+ and CD8+ T cells, indicating further spatial TME organization in IDH wt gliomas.

Figure 4. IDH mutation status shapes TAM activation in gliomas.

Figure 4.

(A) Number of MG and MDM per mm2 inside the perivascular niche (PVN) or distant from the PVN (non-PVN) within IDH wt gliomas by IF staining. Means compared with Wilcoxon signed-rank test: ***p < 0.001. (B) Distance of MG and MDM to nearest vessel in IDH wt gliomas (nsamples=14, nMG=88781 and nMDM=92969 cells counted). (C) Box plot of HLA-DR geometric mean fluorescence intensity measured by FCM in MG and MDM in IDH mut and IDH wt gliomas. MG and MDM from the same samples are connected by lines (nIDHmut=17, nIDHwt=39, Wilcoxon signed-rank test: ***p < 0.001, ****p < 0.0001). (D) GSVA of antigen-processing and -presentation pathways from Molecular Signatures Database (MSigDB) “Canonical Pathways” collection with significant differential enrichment between MG and MDM in IDH wt tumors, and in MG and MDM across IDH mut and IDH wt samples. Columns ordered by IDH mutation status and cell type, rows (z-score) hierarchically clustered. (E) Expression heatmap of T-MG and (F) T-MDM DEGs (compared to T-MG in IDH mut gliomas) in IDH mut and IDH wt glioma samples. Columns and rows (z-score) hierarchically clustered. (G) Normalized counts of selected genes in MG and MDM in gliomas stratified by IDH status. (H) Relative expression in CD45−, MG and MDM cells of ligands and receptors upregulated in CD45− cells in IDH wt vs. IDH mut samples and their matching counterparts. Variance-stabilized expression values were scaled to the expression range. (I) Kaplan-Meier estimator of survival in TCGA glioma cohort based on enrichment for MDM IDH wt signature assessed by GSVA in IDH mut and IDH wt gliomas from combined TCGA cohort. GSVA scores were separated into tertiles across combined IDH mut and IDH wt sample set. Pairwise p values calculated using log-rank test. (J) Hazard ratios of multivariate cox proportional hazards model with transcriptomic subtype (TCGA annotation), IDH status (TCGA annotation) and T-MDM IDH wt GSVA score as covariates for overall survival within TCGA glioma cohort (PN = proneural, NE = neural, CL = classical, and ME = mesenchymal subtype).

See also Figure S4 and Table S5.

We assessed if the distinct T-MG and T-MDM distribution and cell number is paralleled by their activation state. In the LEM analysis we had detected a Type I IFN response in glioma MDM but not MG (Figure 3D); we therefore queried the FCM data to analyze levels of MHC class II HLA-DR expression. This showed significantly increased HLA-DR in T-MDM compared to T-MG in both IDH mut and IDH wt tumors (Figure 4C). We screened the associated RNA-seq data for antigen processing and presentation pathway gene sets using GSEA and gene set variation analysis (GSVA) (Figure 4D). Interestingly, we found evidence for increased expression of MHC class II antigen presentation gene sets in IDH wt glioma MDM, and also antigen processing-associated pathways (Figure S4C), and MHC class I presentation gene sets (Figure 4D). While these findings suggest the potential of TAMs, particularly T-MDM, to initiate an immune response, this potential is generally not realized in the glioma TME - based on the current status of ICB trials in this disease, and we thus asked if there was also evidence of pro-tumor states in these cell populations.

We compared T-MG and T-MDM from IDH wt gliomas to T-MG from IDH mut gliomas, as they constitute the most abundant TME cell types in these tumors respectively (Figure 1F and 2C, Table S5). This revealed 489 DEGs in T-MG (Figure 4E, Table S5, 406 up-, 83 down-regulated), and 1478 DEGs in T-MDM (Figure 4F, Table S5, 903 up-, 575 down-regulated). While these gene lists were generated by comparing T-MDM from IDH wt gliomas vs. T-MG from IDH mut gliomas, they similarly separated T-MDMs in IDH mut vs. IDH wt disease in a clustering analysis (Figure 4F), indicating they indeed reflect T-MDM alterations based on the IDH status of the tumor. 421 genes exhibit a similar pattern across both TAM cell types (343 up-, 78 down-regulated), suggesting that T-MG and T-MDM can also acquire a common transcriptional pattern in IDH wt tumors. Among the shared genes were several encoding extracellular matrix (ECM) proteins (Figure 4G, FN1 and VCAN) and ECM-associated matricellular proteins (THBS1, TGFBI, LGALS3 and ANGPTL4), that regulate the availability of ECM-sequestered ligands, angiogenesis, and tumor immunity (Mushtaq et al., 2018). This suggests that TAMs help shape the composition and effector functions of ECM proteins in IDH wt tumors. We also found the anti-inflammatory molecules ANXA1 and GPNMB (Figure 4G), previously implicated in pro-tumorigenic macrophage polarization and inhibition of T cell activation (Kobayashi et al., 2019; Ripoll et al., 2007), upregulated in T-MG and T-MDM.

We next investigated inflammation mediators within the CD45− population of IDH wt tumors in parallel with their corresponding receptors in TAMs. TGFB2 expression was elevated compared to IDH mut CD45− cells and the accessory TGFβ receptor ENG was highly expressed in IDH wt TAMs (Figure 4H). TGFB2 has pleiotropic effects in inflammation and tissue remodeling during wound healing, and has been implicated in an autocrine signaling loop in glioblastoma cells (Rodon et al., 2014). The neuroinflammatory cytokine MDK, which modulates TAM polarization to a M2-like phenotype in glioma (Meng et al., 2019), was upregulated in CD45− cells from IDH wt tumors, and its receptors SDC4 and ITGA4/CD49D were differentially expressed in T-MDM versus T-MG (Figure 4H), suggesting cell type-specific effects of this inferred signaling loop.

We asked if a T-MDM-specific gene set generated from IDH wt gliomas was associated with a survival difference in patients. By logistic regression we derived a representative signature, consisting of 36 genes (Figure S4D), from the total number of genes upregulated in TAMs in brain malignancies (Figure 3B). This included the macrophage marker RUNX3, the atypical chemokine receptor ACKR3 which can regulate CXCL12-CXCR4 signaling, the ER-stress protein HERPUD1 and the inhibitory Fc receptor FCGR2B, which can modulate macrophage activation (Bournazos et al., 2016; Li et al., 2018), and the cytokine IL19 that affects angiogenesis and macrophage polarization (Richards et al., 2015). The signature was used to classify patients in a merged TCGA dataset of low- and high-grade gliomas (Figure 4I, Figure S4E). In IDH mut patients a decrease in median overall survival was associated with enrichment of the T-MDM IDH wt signature, while IDH wt patients with a low enrichment score showed increased survival. This was confirmed in a multivariate Cox proportional hazard model that included the transcriptomic glioma subtypes (as annotated in the TCGA dataset) and IDH status (Figure 4J). To verify this effect did not simply reflect changes in T-MDM number, we classified the TCGA cohort based on the enrichment of the T-MDM-specific gene set used for deconvolution, which showed low impact on survival (Figure S4F).

In light of disappointing outcomes from PD1/PDL1 ICB trials in glioblastoma to date, we queried whether the abundant T-MG and T-MDM could contribute to the limited therapeutic efficacy. We performed ORA of a panel of 20 gene sets previously associated with “innate anti-PD1 resistance” (IPRES, Hugo et al., 2016) in the TAM DEGs of IDH wt gliomas, and found a sizeable fraction to be upregulated in both T-MG and T-MDM (Figure S4G). We then included the CD45− population and interrogated enrichment of IPRES gene sets on the single sample level by GSVA (Figure S4H). This yielded a diverse picture, with both tumor cells and TAMs enriched for IPRES gene sets to varying degrees. Therefore, both TAMs and CD45− cells from IDH wt gliomas may contribute to mediating innate ICB resistance.

The immune contexture influences the tumor microenvironment on a global level

Through the integrated analysis of protein and gene expression data we next explored the impact of immune cell infiltration. From 200 inflammation-associated proteins assessed, 55 were differentially detected in our sample cohort (clinical information, Table S3B). Unsupervised clustering analysis revealed distinct clusters with abundant inflammatory proteins in tumors (Figure 5A). The profile of IDH wt gliomas and BrMs showed a sizeable overlap (protein cluster 1), encompassing angiogenic factors (VEGFA, ANG), growth factors (PDGFA, TGFB1, SPP1 and GDF15), several proteases and protease inhibitors (SERPINE1, CTSS, TIMP1), the proteolysis cascade regulator PLAUR, and the cytokines CCL2 and CCL5 (Figure 5A, Figure S5A). However, we also found distinct protein patterns between gliomas and BrMs. The neurotrophic growth factor FGF2 and neuronal cell adhesion molecules including ALCAM, which regulates immune cell infiltration during neuroinflammation (Lecuyer et al., 2017), were highly expressed in non-tumor brain, IDH mut and IDH wt samples (protein cluster 3, Figure S5A). Conversely, BrM samples had abundant immune-regulatory molecules affecting myeloid and lymphocytic cells and their heterotypic signaling (protein cluster 2, Figure S5A), such as CD40L, IL6R, INHBA and AREG (Morianos et al., 2019; Zaiss et al., 2015) possibly reflecting the greater immune cell diversity in BrMs. This orthogonal dataset reinforces the RNAseq analyses showing inflammatory signaling pathways are highly enriched in brain tumors.

Figure 5. The immune contexture influences the tumor microenvironment on a global level.

Figure 5.

(A) Inflammation-associated bulk tissue protein concentration heatmap, subset on 55 proteins with significantly different concentrations between non-tumor brain, gliomas and BrMs in an ANOVA (p value < 0.1, nnon-tumor=3, nglioma=14, nBrM=12, concentrations were log10 transformed and z-scored). Rows and columns are hierarchically clustered. Clinical data is annotated per row, column annotation reflects the major protein clusters (further information in Table S3B). (B) Self-organizing map (SOM) of RNA expression data of major cell populations in glioma and BrM samples. SOM-spots are highlighted, numbered with roman numerals and annotated with their cell-type association. (C) Overlap of individual proteins and SOM spot metagenes, tile color fill reflects protein cluster membership from Figure 5A. (D) RNA-seq counts (normalized, scaled to expression range) of proteins from Figure 5A across major cell types in IDH mut and IDH wt gliomas and BrMs. SOM spot membership of individual genes indicated per row.

See also Figure S5.

We integrated the cell type-specific RNA-seq data and bulk protein data to distinguish proteins with a more restricted expression versus those that are expressed across a range of cell types. Transcriptome data from CD45− cells, TAMs, neutrophils, CD4+ and CD8+ T cells from all tumor samples was clustered using a self-organizing map (SOM). This yielded 6 SOM spots, i.e. metagenes of co-expressed genes (Figure 5B), which recapitulated the respective cell lineages (Figure S5B). The CD45− populations were assigned to three distinct spots which were associated with more aggressive IDH wt gliomas and BrMs (spot VI), or reflected the brain-intrinsic or -extrinsic tumor origin (spots I and V). These cell type-associated SOM spots overlapped considerably with the protein data (30/55 proteins, Fisher’s exact test, p < 0.0001, Figure 5C). While VEGFA, ANG and TGFB1 were expressed by diverse cell types in gliomas and BrMs, other genes such as GDF15 and IGFBP2 showed a more CD45− cell-restricted expression (Figure 5D). The significant contribution of TAMs to production of key inflammatory proteins including SPP1 and IHNBA is reflected by TAM SOM spot III constituting the largest group of proteins with cell type-specific expression (Figures 5C and 5D).

Myeloid cells show a distinct phenotype in BrMs

Our global analysis juxtaposing the expression patterns of TAMs in gliomas (regardless of IDH status) to BrMs showed disease- and cell type-specific transcriptomic changes. We thus explored BrM-specific alterations by focusing on genes upregulated only in relation to both the corresponding reference and to IDH wt gliomas (Figure S6A and S6B, Table S6). Various cytokines, chemokines and pro-inflammatory molecules were elevated in both BrM-MG and BrM-MDM (Figure 6A), including potent mediators of autoimmune neuroinflammation CSF2 and IL23A (Zhao et al., 2017), and the pattern-recognition receptor MARCO. Intriguingly, antibody-mediated MARCO-targeting in extracranial tumors increases M1-like polarization of TAMs and enhances ICB efficacy (Georgoudaki et al., 2016). These effects relied on interaction of the antibody with FCGR2B, which is also part of the T-MDM IDH wt signature (Figures S2E and S4C). Finally, RETN, which is involved in systemic inflammatory disorders (Filkova et al., 2009), was upregulated in BrM-TAMs (Figure 6A).

Figure 6. Myeloid cells show distinct transcriptional changes in BrMs.

Figure 6.

(A) Normalized counts of indicated genes in MG and MDM in non-tumor/reference, IDH wt gliomas and BrM. (B) Expression heatmap of ECM-associated genes differentially expressed between MG and MDM in BrMs. Rows z-scored and manually sorted, columns ordered by cell type. (C) Expression of indicated BrM-specific genes in neutrophils from unmatched healthy blood, IDH wt gliomas and BrMs.

See also Figure S6 and Table S6.

Analysis of individual BrM-TAM populations uncovered distinct expression patterns. BrM-MG showed restricted upregulation of IL6 (Figure 6A), which exerts immunosuppressive effects on T cell function in cancer and mediates ICB resistance (Tsukamoto et al., 2018), and the receptor TREM1 which modulates pro-inflammatory responses both in MG and systemically in myeloid cells during neuroinflammation (Liu et al., 2019; Xu et al., 2019). Among upregulated chemokines, we found increases in both TAM cell types (CCL23), and either BrM-MG- (CXCL5, CXCL8) or BrM-MDM-restricted increases (CCL8, CCL13, CCL17 and CCL18) (Figure 6A). These results reveal distinct contributions of TAM populations to the inflammatory TME milieu in a disease-specific manner.

GSEA identified additional cell type-specific enrichment patterns: BrM-MG showed evidence of IL6 pathway activity (Figure S6C), and in BrM-MDM, the “Naba core matrisome” gene set was significantly enriched (Figure S6D). This prompted us to assess expression of genes encoding ECM and matricellular proteins in BrM-MDM vs. BrM-MG, which revealed genes encoding matrix proteins including type III and IV collagens, FN1, the proteoglycans LUM and OGN and the matricellular proteins ECM1, SPARC, and SPARCL1 as highly expressed in BrM-MDM (Figure 6B). While ECM remodeling has been implicated in tumor progression, LUM, OGN, SPARCL and SPARCL1 exhibit both pro- and anti-metastatic properties which underscores the complex context-dependent role of the ECM (Kai et al., 2019). We also found high expression of cathepsin proteases CTSB and CTSW in BrM-MDM (Figure 6B), which participate in multiple tumor-promoting processes including invasion and metastasis (Olson and Joyce, 2015). The hyaluronan receptor HMMR, involved in macrophage chemotaxis and fibrosis in lung injury (Cui et al., 2019), was also higher in BrM-MDM (Figure 6B). Together these data suggest that the ECM is not only shaped by macrophages at the primary site (Afik et al., 2016), but that T-MDM may also play a pivotal role in ECM niche construction in BrM that is distinct from IDH wt gliomas (Figure 4G).

Given the upregulation of CXCL8, a key neutrophil chemoattractant, by BrM-MG (Figure 6A) we explored the TME contribution to recruitment of neutrophils, which were highly abundant in BrM (Figure 1F). Analysis of major neutrophil-recruiting chemokines and their receptors showed broad expression across all interrogated myeloid cells (Figure S6E). To explore the phenotype of BrM-associated neutrophils we queried the RNA-seq data which revealed BrM-specific upregulation of ITGA3 (Figure 6C), which is involved in neutrophil tissue infiltration in sepsis, and CXCL17 previously implicated in neutrophil- and macrophage recruitment in cancer (Li et al., 2014). We also observed upregulation of the adenosine receptor ADORA2A (Figure 6C), that attenuates the phenotype of pro-inflammatory neutrophils (Barletta et al., 2012). Furthermore, we found increased expression of CD177 (Figure 6C), a cell surface receptor that modulates neutrophil migration and activation and serves as a marker for PR3-positive neutrophils that in turn negatively affect T cell proliferation (Yang et al., 2018). Notably, MET, which has been linked to recruitment of immunosuppressive neutrophils in cancer (Glodde et al., 2017), was upregulated in neutrophils in a BrM-specific manner (Figure 6C). In sum, we have uncovered multiple disease-specific alterations of myeloid cells extending beyond BrM-TAMs to neutrophils, which has potential implications for the recruitment and activation of other cell types within the TME, including T cells.

TAMs are poised towards an immunomodulatory phenotype in BrMs

While we found a significant accumulation of CD4+ and CD8+ T cells in BrM vs. IDH wt gliomas by FCM, this analysis of dissociated tissue samples lacks structural information. We thus performed neighborhood analysis of IF-phenotyped IDH wt and BrM tissue sections to elucidate whether there is a spatial relationship between TAMs and CD3+ T cells in BrM. In IDH wt gliomas, both T-MG and T-MDM mostly neighbored homotypic cells while lacking T cells in their close vicinity (Figure 7AB, S7A), possibly reflecting the general T cell sparseness in these tumors. By contrast, both TAM populations neighbored T cells far more frequently in BrM, indicating the potential for interaction (Figure 7AB, S7A).

Figure 7. TAMs exert a wide range of immunomodulatory functions in BrMs.

Figure 7.

(A) Representative IF images and corresponding cell type identification of non-immune cells (CD45−), MG (CD45+, P2RY12/CD68+, CD49D−), MDM (CD45+, P2RY12/CD68+, CD49D+), CD3+ (CD45+, P2RY12/CD68−, CD49D−/+, CD3+) and CD45+ other cells (CD45+, P2RY12/CD68−, CD49D−/+, CD3−) in IDH wt gliomas and BrMs. Scale bars = 50µm, insets show quantifications per FOV. (B) Neighborhood analyses of IDH wt glioma and BrM IF tissue sections. Rows show mean proportion of each neighboring cell type per frequency of observed nneighbors within the vicinity of either MG or MDM (nIDHwt=9, nBrM=13). (C) Gene set enrichment analysis (GSEA) of a T cell anergy gene set in CD4+ T cells and (D) a T cell exhaustion gene set in CD8+ T cells from the MSigDB “C2” collection. (E) Gene expression heatmap of antigen presenting cell (APC) and T-cell activating and inhibitory signaling mediators (left panels, scaled to expression range of variance-stabilized counts across all cell types in IDH wt glioma and BrMs) and corresponding fold changes (right panels, BrMs vs. non-tumor/reference and IDH wt glioma vs BrMs, absolute log2(fc) > 1, p.adj < 0.05) in CD45−, MG, MDM, CD4+ and CD8+ T cells in IDH wt gliomas and BrMs. Grey tiles indicate expression below threshold (normalized counts < 10), white tiles correspond to non-significant fold change. (F) Scatter plot of module membership (correlation of expression to module eigengene) and gene significance (correlation of expression to CD4+ T cell abundance) of genes from the BrM MDM-related gene co-expression network. Highly connected genes with immunomodulatory functions are annotated. (G) Expression of indicated genes in matched bulk primary breast cancer and BrM tissues using Vareslija et al., 2019 dataset (Wilcoxon signed-rank test: ***p < 0.001, ****p < 0.0001).

See also Figure S7.

We thus investigated the T cell activation state in BrMs in relation to unmatched healthy donor blood and also juxtaposed them to the corresponding populations from IDH wt gliomas. Compared to controls, CD4+ T cells from BrM showed evidence of a hyporesponsive, anergic phenotype (Figure 7C) while CD8+ T cells exhibited an exhaustion signature (Figure 7D) which usually occurs upon chronic activation, resulting in upregulation of inhibitory receptors. These defective T cell states can be caused by aberrant activation or T cell inhibition by both tumor cells and antigen-presenting cells in the TME and constitute a major obstacle in treating cancers,.

To delineate putative mechanisms in the BrM TME that may drive these alterations, we probed the RNAseq data from CD45− cells, TAMs, and T cells (Figure 7E) for expression of activating and inhibitory immunomodulatory signals (Wei et al., 2018). This revealed upregulation of various canonical T cell activators and co-activators but also mediators of inhibition in T cells (PDCD1/PD1, CD28, CTLA4), while T cell-inhibiting and activating signals were detected in both TAM populations (CD274/PD-L1, PDCD1LG2/PD-L2). Notably, we found upregulated CD80, which has diverse roles in T cell activation as it heterodimerizes with CD274, provides co-stimulatory signals to T cells via CD28, and exerts inhibitory effects via interaction with CTLA4 (Zhao et al., 2019), in both TAM populations when compared to their normal references and IDH wt tumor populations (Figure 7E). The potential contribution of TAMs to metabolic immune evasion is also suggested by high expression of IDO1 and IDO2 (Zhai et al., 2018) in BrM (Figure 7E).

We investigated additional immunomodulatory mediators using weighted gene correlation network analysis (WGCNA, Langfelder and Horvath, 2008), and correlated the resulting expression patterns with paired FCM abundance of CD4+ and CD8+ cells in a disease and cell type-specific manner. We identified 15 unique co-expression modules showing significant correlation (p < 0.05) of their eigengenes (i.e. the first PC of the module expression data) with any of the provided sample traits (Figure S7B). Among these the “brown” WGCNA module correlated with both a specific BrM-MDM annotation and CD4+ T cell abundance. ORA of this module revealed signals for pathways such as coagulation and ECM modulation (Figure S7C) that impact the availability and activity of growth factors and cytokines within the TME (Mohan et al., 2020). We ranked genes by module membership strength, and correlation with CD4+ T cell abundance, which identified several factors with opposing immunomodulatory functions (Figure 7F). While the receptors CD300E and BST1 promote monocyte motility and survival (Isobe et al., 2018; Ortolan et al., 2019), we also detected effectors of immunosuppression such as actin-associated regulatory protein CNN2 which negatively regulates macrophage motility and phagocytic activity (Huang et al., 2008). The leukocyte immunoglobulin-like receptor subfamily B members LILRB2 and LILRB3, which attenuate myeloid cell activation (van der Touw et al., 2017), are also highly ranked genes within this module. Interestingly, LILRB2 was identified as a novel myeloid immune checkpoint that limits antitumor immunity (Chen et al., 2018). We also found evidence for effects on T cells as CD52, which in its soluble form inhibits T cell function, was among the BrM-MDM module genes. The notion that BrM-MDM undergo disease-specific alterations distinct from the primary extracranial tumor is supported by the upregulation of these genes (Figure 7G) in our analysis of an external cohort of BrM samples compared to their matched primary tumor tissue (Vareslija et al., 2019).

Discussion

Brain tumors, including glioblastoma and BrM, confer some of the poorest prognoses for patients with cancer, with survival rates often measured in just months. Given the current dearth of effective therapeutic options for these patients, and the modest effects of the various immunotherapies evaluated to date, it is of critical urgency to identify novel targets for future clinical evaluation. One potentially rich source of therapeutic targets is the TME. However, while the TME is now widely accepted as an important regulator of cancer progression and therapeutic response, our knowledge of the brain TME is restricted to individual brain tumor types or cellular compartments and lacks a comprehensive and integrative analysis.

In this study we have leveraged a diverse panel of analyses to deeply interrogate the immune landscape of primary and metastatic brain cancers. Through integration of multiparameter FCM analyses, RNA-seq data, TME cell culture assays, protein arrays and spatial tissue characterization, we have uncovered critical insights into the composition and transcriptomes of the most abundant immune cell populations in patient samples from IDH mut and wt gliomas, and BrMs originating from distinct extracranial primary tumors.

By exploring the broad immune landscape we uncovered several pronounced differences between gliomas and BrMs when directly compared side by side. In brain tumors, TAMs are composed of both tissue-resident MG and recruited MDM, and we found a significant shift in the ratio of MG to MDM between IDH mut and IDH wt gliomas. Additionally, gliomas contain an abundance of TAMs while T cells were much fewer, particularly in IDH mut tumors. This confirms the notion that gliomas are immunologically cold tumors (Jackson et al., 2019). While T cell sequestration in the bone marrow has been observed in glioma mouse models and following intracranial implantation of brain-extrinsic tumors (Chongsathidkiet et al., 2018), our clinical BrM samples showed pronounced accumulation of both lymphocytes and neutrophils. This indicates that tumors which arise within the brain indeed shape their TME differently to cancers that metastasize from extracranial sites. Moreover, when exploring BrM that originate from distinct primary tumors, there were additional differences; for example, in melanoma-BrM samples the combined abundance of CD4+ and CD8+ T cells represented the major immune compartment, while breast-BrM samples showed the highest neutrophil infiltration. These key differences in the TME landscape, which are evident only when directly juxtaposing different brain malignancies, mirror the efficacy of immunotherapies that show promising efficacy in melanoma patients for controlling BrM, but with very modest effects to date in treating T cell-excluded glioblastoma (Schalper et al., 2019).

We have also uncovered complex multifaceted phenotypes for TAMs across different brain tumors that extends beyond their numerical abundance. T-MG and T-MDM showed both distinct transcriptomic profiles and shared expression signatures, which are additionally influenced by the underlying disease type (IDH mut vs. IDH wt glioma vs. BrMs). A T-MDM signature derived from IDH wt gliomas, consisting of macrophage activation markers, chemokine receptors and cytokines, proved to also be a predictor of patient survival in IDH mut gliomas. Moreover, analyses of T-MDM indicated that while these recruited cells have the potential to process and present antigens, and can be located proximally to T cells in BrM, this potential is evidently not being sufficiently utilized within the brain TME. Orthogonal analyses from the diverse panel of experimental assays used in this study reveal additional insights into potential mechanisms of immune suppression. These included our findings that different TAM populations produced pro-inflammatory molecules, negative regulators of myeloid cell activation, factors associated with innate anti-PD1 resistance, IDO1 and IDO2 immune checkpoint inhibitors, and specific ECM components and proteases that may collectively help sculpt an immune suppressive niche. As such, therapeutic strategies that alter the multifaceted phenotypes of TAMs (Kowal et al., 2019), rather than aiming to simply deplete all of these cells with potentially opposing functions, should be considerably more effective.

Looking beyond TAMs, it will also be critical to assess the roles of neutrophils, particularly in BrMs where we found they are highly abundant, as they can act as potent immune suppressive cells as indicated from studies in other organs (Coffelt et al., 2016). Given the highly complex and multifaceted immune landscape of brain cancers revealed in this study, it is clear that rational combinations of TME-targeted agents will be critical to avoid the emergence of adaptive resistance, incorporating preclinical studies to help determine the optimal combinations (Quail et al., 2016). In sum, this rich resource is available for further interrogation by the research community so that we can work collectively to uncover novel therapeutic strategies that unleash the potential of diverse cells in the TME to combat different brain malignancies.

STAR Methods:

Resource availability:

Lead contact

Further information and requests for resources should be directed to the Lead Contact, Johanna Joyce (johanna.joyce@unil.ch).

Materials availability

This study did not generate new unique reagents.

Data and code availability

RNA-seq count expression data generated during this study can be visualized and downloaded at https://joycelab.shinyapps.io/braintime/. Due to patient privacy protection, the raw RNA-seq data will be made available upon request.

Experimental model and subject details

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed consent was obtained from all individual participants included in this study. The collection of non-tumor and tumor tissue samples at the Centre Hospitalier Universitaire Vaudois (CHUV, Lausanne, Switzerland) was approved by the Commission cantonale d’éthique de la recherche sur l’être humain (CER-VD, protocol PB 2017–00240, F25 / 99). Sample collection at Memorial Sloan Kettering Cancer Center (MSKCC, New York, NY, USA) was approved by the institutional review board (IRB, protocols #IRB #06–107, #14–230). Non-tumor samples of cerebral cortex tissues were collected at CHUV during medically indicated surgical treatment of refractory epilepsy patients, and at MSKCC in normal brain distant from the tumor in patients with low-grade glioma or from post-mortem samples collected through the rapid autopsy program with no history of brain malignancy.

Tissue specimens were immediately collected from the operating room and processed as described below. All patient-related data and unique identifiers were removed so that human samples were anonymized before any further processing.

Pathological review and molecular analysis of tumor samples was performed as part of standard clinical care at the respective locations (CHUV or MSKCC). In all glioma samples subjected to RNA sequencing, the IDH1 and IDH2 mutation status was verified by inspection of the reads from the CD45− population aligning to the IDH1 and IDH2 loci with the Integrative Genomics Viewer (IGV, Robinson et al., 2017). For immunofluorescence sections the tumor diagnosis was confirmed independently, for all non-tumor samples, the absence of malignancy was equally confirmed by a pathologist.

Peripheral blood and buffy coats were obtained from the Transfusion Interrégionale, Croix-Rouge Suisse (Epalinges/Lausanne, Switzerland), the New York Blood Center (New York, NY, USA), and healthy donors.

Method Details

Clinical sample processing, flow cytometry (FCM) and fluorescence activated cell sorting (FACS)

Tissue specimens were washed in HBSS and macro-dissected under sterile conditions. Parts of the tissue were either immediately frozen by submerging the sample in liquid nitrogen-cooled 2-methyl butane (Sigma-Aldrich) or OCT-embedded (Tissue-Tek) before freezing for subsequent sectioning and immunofluorescence staining. OCT embedding was performed by placing the sample in a freezing mold filled with OCT and then submerging the mold in 2-methyl butane cooled with dry ice.

The remaining tissue was further processed with either the Brain Tumor Dissociation Kit (Miltenyi) for non-tumor tissue and gliomas, or the Tumor Dissociation Kit for BrMs (Miltenyi) using the gentleMACS Octo Dissociator (Miltenyi). Myelin debris in cell suspensions from non-tumor and glioma tissues was removed by incubating the cells with Myelin Removal Beads (Miltenyi) and magnetic-activated cell sorting (MACS) using LS columns (Miltenyi) according to the manufacturer’s instructions. All tissue suspensions were filtered through a 40 μm filter and underwent red blood cell lysis (BioLegend). Single cell suspensions were stained with a fixable live-dead stain (Zombie NIR, BioLegend), FC-blocked for 10 min (Human TruStain FcX, BioLegend) and then incubated with direct fluorophore-conjugated antibodies for 20 min at 4ºC. All FCM antibodies were titrated in a lot-specific manner. Antibody details are listed in the Key Resources table. Cells were washed with PBS +2% fetal bovine serum (FBS) +0.5 mM EDTA and stored at 4ºC in the dark until FAC-sorting.

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
FCM: AF700 mouse monoclonal anti-human CD45 (clone HI30) BioLegend Cat#304024
FCM: BV421 rat monoclonal anti-mouse/human CD11B (clone M1/70) BioLegend Cat#101251
FCM: PE mouse monoclonal anti-human CD66B (clone G10F5) BioLegend Cat#305106
FCM: AF488 mouse monoclonal anti-human CD14 (clone HCD14) BioLegend Cat#325610
FCM: BUV737 mouse monoclonal anti-human CD16 (clone 3G8) BD Cat#612786
FCM: APC mouse monoclonal anti-human CD49D (clone 9F10) BioLegend Cat#304308
FCM: BV605 mouse monoclonal anti-human CD11C (clone 3.9) BioLegend Cat#301636
FCM: BV711 mouse monoclonal anti-human anti HLA-DR (clone L243) BioLegend Cat#307644
FCM: PerCP/Cy5.5 mouse monoclonal anti-human CD3 (clone HIT3a) BioLegend Cat#300328
FCM: BV 650 mouse monoclonal anti-human anti CD4 (clone OKT4) BioLegend Cat#317436
FCM: PE mouse monoclonal anti-human CD25 (clone BC96) BioLegend Cat#302606
FCM: BV510 mouse monoclonal anti-human CD127 (clone A019D5) BioLegend Cat#351332
FCM: PE/Cy7 mouse monoclonal anti-human CD8A (clone HIT8a) BioLegend Cat#300914
FCM: BUV563 mouse monoclonal anti-human CD20 (clone 2H7) BD Cat#748456
FCM: BUV563 mouse monoclonal anti-human CD19 (clone SJ25C1) BD Cat#612916
FCM: PE/Dazzle mouse monoclonal anti-human CD56 (clone HDC56) BioLegend Cat#318348
FCM: PE mouse monoclonal anti-human P2RY12 (clone S16001E) BioLegend Cat#392103
FCM: PE/Cy7 Mouse monoclonal anti-human CD68 (clone Y1/82A) BioLegend Cat#333816
IF: Mouse monoclonal anti-human CD68 (clone KP1), 1:100 dilution Abcam Cat#ab955
IF: Rat monoclonal anti-human CD49D (clone PS/2), 1:100 dilution Abcam Cat#ab25247
IF: Rabbit polyclonal anti-human P2RY12, 1:600 dilution Sigma-Aldrich Cat#HPA014518
IF: Goat polyclonal anti-human CD45, 1:100 dilution LSBio Cat#LS-B14248–300
IF: AF488 mouse monoclonal anti-human CD45 (clone HI30), 1:100 dilution BioLegend Cat#304019
IF: AF488 mouse monoclonal anti-human CD3 (clone UCHT1), 1:100 dilution BioLegend Cat#300406
IF: Sheep polyclonal anti-human CD31, 1:200 dilution R&D Cat#AF806
IF: APC rat monoclonal anti Ki-67 (clone SolA15), 1:100 dilution Thermo Fisher Scientific Cat#17–5698–82
IF: AF555 donkey anti-rabbit IgG 1:1000 dilution Thermo Fisher Scientific Cat#A31572
IF: AF555 donkey anti-mouse IgG, 1:500 dilution Thermo Fisher Scientific Cat#A32773
IF: AF488 donkey anti-rat IgG, 1:500 dilution Thermo Fisher Scientific Cat#A21208
IF: AF647 donkey anti-rat IgG, 1:500 dilution abcam Cat#ab150155
IF: DyLight755 donkey anti-goat IgG, 1:500 dilution Thermo Fisher Scientific Cat# SA5–10091
IF: AF555 donkey anti-sheep IgG, 1:500 dilution Thermo Fisher Scientific Cat#A21436
Bacterial and Virus Strains
N/A N/A N/A
Biological Samples
Non-tumor, glioma and brain metastasis tissue Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland N/A
Non-tumor, glioma and brain metastasis tissue Memorial Sloan Kettering Cancer Center, New York, NY, USA N/A
Healthy donor blood Transfusion Interrégionale Croix-Rouge Suisse, Epalinges, Switzerland N/A
Healthy donor blood New York Blood Bank, New York, NY, USA N/A
Chemicals, Peptides, and Recombinant Proteins
DMEM-F12 (1:1), GlutaMAX Gibco Cat#31331028
DMEM, high glucose, GlutaMAX, pyruvate Gibco Cat#31966021
Penicillin/Streptomycin Gibco Cat#15140122
Human recombinant CSF-1 R&D Systems Cat#216-MC-025
Ficoll-Paque Premium GE Cat#17–5442–02
Trizol Thermo Fisher Scientific Cat#15596018
Trizol LS Thermo Fisher Scientific Cat#10296028
Tween 20 Applied Chemicals Cat#A4974
Triton X-100 Applied Chemicals Cat#A4975
TNB Blocking Reagent Perkin Elmer Cat#FP1020
Fluorescence Mounting Medium Dako Cat#S302380
Critical Commercial Assays
Brain Tumor Dissociation Kit (P) Miltenyi Cat#130–095–942
Tumor Dissociation Kit, human Miltenyi Cat#130–095–929
Myelin Removal Beads Miltenyi Cat#130–096–733
CD14 MicroBeads, human Miltenyi Cat#130–050–201
Human TruStain FcX BioLegend Cat#422302
ZombieNIR Fixable Viability Kit BioLegend Cat#423106
High Capacity cDNA Reverse Transcription Kit Applied Biosystems Cat#4368814
TaqMan Universal PCR Master Mix Applied Biosystems Cat#4304437
Quantibody Array Q4000 ELISA Raybiotech Cat#QAH-CAA-4000–1
Deposited Data
Raw data This paper https://joycelab.shinyapps.io/braintime/
Human reference genome, hg38 Genomics Data Common https://gdc.cancer.gov/about-data/data-harmonization-and-generation/gdc-reference-files
TCGA LGG and GBM datasets Genomics Data Common https://portal.gdc.cancer.gov/
TOIL TGCA TARGET GTEx datasets Vivian et al., 2017 https://xenabrowser.net/datapages/
Ivy Glioblastoma Atlas Project RNA sequencing fata Puchalski et al., 2018 https://glioblastoma.alleninstitute.org/static/download.html
STRING Protein-Protein-Interaction database, version 10.5 Szklarczyk et al., 2017 https://version-10-5.string-db.org/cgi/download.pl
Molecular Signatures Database gene set collection Liberzon et al., 2015; Subramanian et al., 2005 http://software.broadinstitute.org/gsea/msigdb/
RNA sequencing count matrix from matched breast cancer primaries and brain metastases Vareslija et al., 2019 https://github.com/npriedig
Experimental Models: Cell Lines
N/A N/A N/A
Experimental Models: Organisms/Strains
N/A N/A N/A
Oligonucleotides
See Table S7.
Recombinant DNA
N/A N/A N/A
Software and Algorithms
FlowJo, version 10.4 BD https://www.flowjo.com/
BBDuk, version 38.12 Joint Genome Institute https://jgi.doe.gov/data-and-tools/bbtools/
STAR aligner, version 2.5.2b Dobin et al., 2013 https://github.com/alexdobin/STAR
R environment, version 3.5.0 R Core Team, 2018 https://www.r-project.org/
VIS Image Analysis, version 2019.7 Visiopharm https://www.visiopharm.com/
Other
gentleMACS Octo Dissociator Miltenyi Cat#130–095–937
gentleMACS C Tubes Miltenyi Cat#130–096–334
LS Columns Miltenyi Cat#130–042–401
SepMate-50 StemCell Cat#85450
PermaLife Cell Culture Bags OriGen Biomedical Cat#PL30–2G
LSR II flow cytometer BD N/A
Fortessa flow cytometer BD N/A
FACSAria III, flow cytometer & cell sorter BD N/A
Axio Scan.Z1 slide scanner Zeiss N/A
QuantStudio 6 Flex Applied Biosystems N/A
Omni Tissue Homogenizer (TH) Omni International Cat#TH220

All FCM acquisition was completed on either a BD Fortessa or a BD LSR II device (BD), and cell sorting was performed on a FACSAria III (BD) using FACSDiva (BD). Cells were sorted directly into Trizol LS (Thermo Fisher Scientific) and immediately snap frozen with liquid nitrogen. Analysis of FCM data was performed with FlowJo (BD).

Tumor microenvironment-conditioned medium (TME-CM) generation

Single cell suspensions from whole tumor samples were resuspended in DMEM-F12 (1:1) +Glutamax (Gibco) +10% FBS +1% penicillin/streptomycin (P/S, Gibco) and adjusted to a concentration of 2 × 106 cells/ml with 2ml plated into each well of a 6-well plate (TPP). The supernatant of these tissue cultures, containing cancer cells, immune cells etc. from the complex brain TME, was harvested at 24 hours after initial seeding, spun down to remove debris (300 g, 10 min) and stored at −80ºC until further use.

In vitro generation of monocyte-derived macrophages (MDM) and TME-CM stimulation

Peripheral blood mononuclear cells were isolated from buffy coats of healthy donors with a Ficoll (GE) gradient using SepMate tubes (StemCell) and monocytes selected by MACsorting with CD14 MicroBeads (Miltenyi). Monocytes were differentiated into macrophages by culture in Teflon-coated bags (OriGen) for 7 days in DMEM +GlutaMAX (Gibco) +10% FBS +1% P/S with the addition of 10 ng/ml recombinant human CSF-1 (R&D Systems).

Differentiated MDMs were plated at a density of 1 × 106 cells/well of a 6-well plate in DMEM +10% FBS +1% P/S +10ng/ml CSF-1. After cell attachment, MDMs were cultured in serum free medium for 6 hours before stimulation with TME-CM for 24 hours.

RNA isolation, cDNA synthesis and quantitative real-time PCR

TME-CM-stimulated MDMs were lysed with Trizol (Thermo Fisher Scientific), RNA was purified with Direct-zol columns (Zymo Research), DNase treated and 1.0 μg of RNA was used for cDNA synthesis using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems). An amount of cDNA equivalent to 5 ng total RNA was used for real-time PCR. For primer and probe details see Supplementary Table 6. Assays were run in triplicate on a QuantStudio 6 Flex instrument (Applied Biosystems) using the TaqMan Universal PCR Master Mix (Applied Biosystems) and expression was normalized to the average expression of Ubiquitin C (UBC) and Ribosomal Protein L19 (RPL19) for each sample.

Immunofluorescence staining and microscopy image acquisition

10 μm cryostat sections were thawed, air dried and fixed with ice-cold 100% methanol for 5 minutes. After rehydration with PBS, sections were washed twice in PBS +0.2% Tween 20 (Applied Chemicals), permeabilized with PBS +0.2% Triton X-100 (Applied Chemicals) for 3 hours and washed again with PBS +0.2% Tween 20. Blocking was performed with PBS +0.5% Tween 20 +1% TNB Blocking Reagent (Perkin Elmer), followed by incubation with primary antibody in the same buffer overnight at 4ºC. Primary antibody information and dilutions are listed in the Key Resources table. Sections were washed with PBS +0.2% Tween 20 before incubation with fluorophore-conjugated secondary antibodies at a dilution of 1:500 in PBS +0.5% Tween 20 +1% TSA Blocking Reagent +1 μg/ml DAPI at room temperature. Directly-conjugated primary antibodies were employed where indicated after an initial round of primary and secondary antibody staining, to avoid potential for cross reactivity. Finally, sections were washed with PBS +0.2% Tween and mounted with Fluorescence Mounting Medium (Dako).

Stained tissue sections were imaged with an Axio Scan.Z1 slide scanner (Zeiss) equipped with a Colibri 7 LED light source (Zeiss) using a Plan-Apochromat 20x/0.8 DIC M27 cover slip-corrected objective (Zeiss). All slides from the same staining panel were digitalized using identical acquisition settings.

Image analysis and cell type identification

Image quantification was performed using the VIS Image Analysis software (Visiopharm). For each staining panel a specific application was created using the software’s authoring module. The tissue outline was detected after applying a 21 pixel mean filter. The edges of the derived regions of interest were smoothened with the built-in function “close” and holes in the mask were filled using the “fill holes” command. Aberrant signals resulting from e.g. dust particles, tissue folds or air bubbles were manually excluded from these regions of interest. Nuclear classification was based on the watershed signal of the DAPI staining and filtered by area to exclude incomplete nuclei. The obtained nuclear label was expanded by 5 pixels to capture both nuclear and adjacent cytoplasmic fluorescent signal. Cell types were identified using a hierarchical decision tree with manually set thresholds. Finally, a representation of the cytoplasm was created using the inbuilt growth algorithm with a maximum distance of 15 pixels from the nucleus. Vessel segmentation was performed by creating a separate classifier based on pixel intensity of the CD31 signal. Nuclear classifiers were excluded a priori and incorporated in the vessel label only when exceeding the threshold for CD31. Perivascular niches (PVNs) were established by generating an ROI around vessels at a distance of 20μm. All object-based phenotyping result tables were exported as csv files for further analysis within the R environment.

Protein isolation and enzyme-linked immunosorbent assay (ELISA)

Frozen tissues were weighed and homogenized on ice with an Omni Tissue Homogenizer (Omni International) in 10 μl of RIPA lysis buffer (Thermo Fisher Scientific) +cOmplete Protease Inhibitor (Roche Diagnostics) per mg of tissue. The homogenate was gently agitated on ice for 10 minutes, centrifuged at 10.000g for 5 minutes at 4ºC and the supernatant collected. The protein concentration was determined using a Bradford assay (Bio-Rad) and adjusted to 1 μg/μl. Samples were shipped to Raybiotech (Peachtree Corners, GA, USA) for quantitative analysis with the multiplexed Quantibody Array Q4000 ELISA.

RNA sequencing (RNA-seq)

RNA was isolated by chloroform extraction and isopropanol precipitation. RNA sequencing libraries were generated with the SMART-Seq preparation kit (CloneTech) and fragmented with the Nextera XT kit (Illumina). Paired end, 100 or 150 base pair and single end 100 base pair, sequencing was performed by Genewiz (South Plainfield, New Jersey, USA) on an Illumina HiSeq 2500 (Illumina).

Reads were adapter trimmed and quality clipped using BBDuk (version 38.12, sourceforge.net/projects/bbmap/). Trimmed reads were mapped to the Genomic Data Commons (GDC) GRCh38.d1.vd1 reference sequence using the STAR aligner (version 2.5.2b, Dobin et al., 2013) in two-pass mode with parameters corresponding to the GDC RNA-seq alignment workflow. Transcript abundance was estimated using the corresponding GDC reference gtf file. A raw count matrix was produced and differential gene expression was assessed with DESeq2 using an absolute log2 fold change of 1 and a false discovery rate of 0.01 when contrasting to reference samples, and 0.05 for within tumor contrasts (Love et al., 2014).

Bioinformatic analysis environment

All bioinformatic analyses were performed within the R environment (version 3.5.0, R Core Team 2018).

Gene set-centered analyses

The Molecular Signatures Database (MSigDB, version 6.1, Liberzon et al., 2015; Subramanian et al., 2005) was used as the main source for gene set-based analyses.

Over-representation was assessed with the goseq R package (Young et al., 2010) for differentially expressed genes to correct for gene length bias, otherwise the hypergeometric test was employed. For individual samples, gene set enrichment was estimated with the Gene Set Variation Analysis R package (GSVA, Hanzelmann et al., 2013) using the “gsva” function. Gene set enrichment analysis (GSEA) was evaluated with the R package fgsea (https://github.com/ctlab/fgsea) using the maximum likelihood log fold changes determined by DESeq2 as the ranking metric.

Deconvolution of Toil-RNA sequencing data

Toil-processed (Vivian et al., 2017), DESeq2-standardized gene expression data and matching phenotype data from the TCGA and Genotype-Tissue Expression Project (GTEx) databases were downloaded from the UCSC Xena platform and filtered to include only low-grade glioma “TCGA-LGG” and high-grade TCGA-GBM” and “frontal cortex” GTEx samples to integrate bulk glioma expression data with unmatched non-tumor samples. MG- and MDM-specific marker genes were derived by identifying differentially expressed genes in these two populations vs. all other sorted populations in a pairwise fashion, determining the intersect and ranking the resulting genes by their fold change vs. the CD45− population. The 20 highest ranked genes were then used as cell type-specific marker genes. Deconvolution of MG- and MDM-proportions in tumor and non-tumor sample expression data was done with the EPIC R package (Racle et al., 2017) using these marker genes and providing the expression data from the sorted populations as reference profiles. As the exact amount of RNA within the estimated cell types is not known, this parameter was set to 1 when running the deconvolution.

Leading edge metagene (LEM) analysis

To capture biologically meaningful patterns of gene expression within the differentially expressed genes the LEM approach (Godec et al., 2016) was employed: (a) GSEA was performed using the MSigDB C7 collection as described above, (b) the leading edge genes of the significant gene sets were arranged into a genes by gene sets matrix with the shrunken fold changes as the entries, (c) this matrix was clustered using non-negative matrix factorization with the R package NMF (Gaujoux and Seoighe, 2010), (d) genes with a small coefficient in each metagene were filtered based on the 95th quantile of a fitted exponential distribution of the coefficients and (e) each gene with a coefficient above the threshold was assigned to the metagene where it had the highest coefficient.

Protein-Protein-Interaction network building

Version 11.0 of the STRING database (Szklarczyk et al., 2017) was downloaded from the consortium’s website and gene identifiers from RNA-seq were mapped to Ensembl Protein IDs using the provided accessory data. The resulting interaction data was filtered to contain only interactions with a high confidence STRING combined score (i.e. > 700). For network layout calculation the combined score was used as an edge weight.

Nearest neighbor distance measurements and neighborhood analysis of IF data

Nearest neighbor distances from MG and MDM to vessels in IDH wt glioma samples were calculated using the spatstat R package (Baddeley et al., 2015). Statistical significance was assessed by fitting a mixed effects model with the cell type as the fixed effect, and the clinical sample ID as the random effect using the R package lme4 (Bates et al., 2015).

Neighbors for each individual cell were determined based on their occurrence within a range of 5 μm outside of the radius of the cell (calculated based on the area). This was used to tabulate the number of neighbors and their cell type for each cell within the tissue section.

Cell type abundance estimation in spatial Ivy Glioblastoma Atlas Project (GAP) data

The micro-dissected Ivy GAP (Puchalski et al., 2018) RNA-seq RSEM count data and sample annotation containing anatomical location were downloaded from the Ivy GAP website (https://glioblastoma.alleninstitute.org/static/download.html) and normalized using DESeq2. The relative abundance of cell types was estimated by deriving marker genes through a multinomial logistic regression model on the normalized expression data of the FAC-sorted cell types of interest in IDH wt tumors and then computing the GSVA enrichment scores in the Ivy GAP samples.

Survival analysis of the IDH wt MDM-specific gene signature in gliomas

The harmonized TCGA low-grade and high-grade HTSeq hg38 count data and clinical data was accessed from the GDC repository using the TCGAbiolinks R package (Colaprico et al., 2016). Datasets were pre-processed to remove outliers and normalized using the functions provided by TCGAbiolink before merging. Subsequent analyses were performed including only samples where an annotation of the IDH mutation status was available. Cell type-specific gene signatures were derived by training a multinomial logistic regression model with an elastic-net penalty to separate between MG and MDMs along IDH status with the “glmnet” R package (Friedman et al., 2010). A mean-centered expression matrix of all MG and MDMs expression data in gliomas and BrMs, subset by genes that were upregulated in tumors vs non-tumor tissue or healthy controls, served as the input matrix. The strength of the penalty was determined by a 10-fold cross-validation of the λ parameter. For survival analysis, GSVA enrichment scores of these cell type-specific gene signatures were estimated and used to divide samples into tertiles. Kaplan-Meier survival curves were computed using the “survfit” function. Survival curves were compared with a log-rank test between the individual levels and multivariate Cox regression analysis was performed with the “coxph” function.

Self-organizing map (SOM) clustering

Variance stabilized counts from sorted populations of interest from IDH mut, IDH wt glioma and BrM samples were filtered with the R package HTSFilter (Rau et al., 2013) to ensure removal of genes with a low, constant expression. The resulting matrix of genes and samples was used as input for the SOM neural network building, which was performed with the oposSOM R package (Loffler-Wirth et al., 2015) with a map space of 50 × 50. To investigate associations between the sample phenotype and the SOM metagenes, the tumor type and cell type were provided as group labels.

Weighted gene correlation network analysis (WGCNA)

The WGCNA (Langfelder and Horvath, 2008) R package was used to identify co-regulated genes associated with a MG- or MDM-BrM phenotype. A variance stabilized, batch-corrected count matrix of MG and MDM samples was filtered with the R package HTSFilter (Rau et al., 2013) yielding input expression data with 15826 genes and 56 samples. WGCNA standard parameters were changed as follows: the soft-thresholding power was raised to 7, the minModuleSize was increased to 50, “bicor” was used to calculate the correlation, the network type was set to “signed hybrid” and a dendrogram cut height of 0.25 was used for module merging. This yielded 20 modules whose eigengene, i.e. the first principal component, was tested for correlation to the provided sample information, i.e. tumor- and cell-type and abundance as determined by FCM.

Expression analysis of external dataset of matched primary breast cancer and BrMs

RNA-seq raw count data from patient-matched primary breast tumors and corresponding BrMs (Vareslija et al., 2019) were downloaded (https://github.com/npriedig/jnci_2018) and transformed using DESeq2. The statistical significance of gene expression changes between primary tumors and BrMs was assessed with a two-tailed Wilcoxon signed-rank test on the variance-stabilized counts.

Plotting and graph generation

Plots were created using the ggplot2 R package (Wickham, 2016) and the ggpubr (https://cran.r-project.org/web/packages/ggpubr/), survminer (https://cran.r-project.org/web/packages/survminer/), ggraph (https://cran.r-project.org/web/packages/ggraph/) and ggcyto extensions (Van et al., 2018). Annotated heatmaps were drawn with the pheatmap R package (https://cran.r-project.org/web/packages/pheatmap/).

Quantification and statistical analysis

Summary data are presented as mean ± standard error of the mean (SEM) or Tukey boxplots using “ggplot2”. Numerical data was analyzed using the statistical tests noted within the corresponding sections of the manuscript. Hierarchical clustering was performed using Ward’s method with 1-Pearson correlation coefficient as the distance metric unless noted otherwise. P values were annotated as follows: * <0.05, ** <0.01, *** <0.001, **** <0.0001, ns >0.05.

Supplementary Material

1. Figure S1. Fluorescence-activated cell sorting (FACS) of cell populations and RNA-sequencing, related to Figure 1.

(A) Flow cytometry (FCM) plots illustrating the gating strategy employed during FAC-sorting of immune cell populations in non-tumor and tumor tissue (for cell type markers, see Table S2). (B) tSNE plot of gene expression data (500 most variable genes) from all sorted cell populations (n=226) across the complete clinical cohort (MG = microglia, MDM = monocyte-derived macrophages, reference = unmatched healthy blood and in vitro generated MDMs).

See also Table S2.

2. Figure S2. MG and MDM marker expression, related to Figure 2.

(A) Normalized counts (log10 transformed) of MG and MDM marker genes in sorted CD49Dlow MG and CD49Dhigh MDM populations across both non-tumor and tumor tissues (reference = healthy donor in vitro generated MDMs). (B) Percentage of CD49Dlow MG and CD49Dhigh MDMs positive for P2RY12 and CD68 as determined by FCM in relation to the total number of MG/MDMs in non-tumor (n=8) and tumor tissue (nIDHmut=6, nIDHwt=6, nBrM=9). (C) Single channel and merged immunofluorescence (IF) images of CD45, CD68, P2RY12 and CD49D stainings which were employed to delineate MG and MDMs. The last column shows the resulting Visiopharm cell type assignments for quantitative analyses (MG (CD45+, P2RY12+/CD68+, CD49D−), MDM (CD45+, P2RY12+/CD68+, CD49D+), non-immune cells (CD45−) and non-TAM-immune cells (CD45+, P2RY12−/CD68−, CD49D−/+). Scale bars represent 100µm. (D) Scatter plots of the abundance of MG and MDMs as determined by IF vs. FCM in non-tumor (n=4) and tumor tissues (nIDHmut=13, nIDHwt=14, nBrM=18) processed independently from the same individual samples. Pearson’s correlation coefficient and significance are indicated at the top of each plot. (E) Heatmap of human MG- and MDM-specific gene set expression used for deconvolution across FAC-sorted population samples from all disease types.

3. Figure S3. Analysis of differentially expressed genes and TAM activation patterns, related to Figure 3.

(A) Summary of contrasts applied when performing differential gene expression analysis in MG and MDMs in gliomas (regardless of IDH status) and BrMs (from all primaries) in comparison to normal controls (non-tumor brain MG and in vitro differentiated MDMs respectively) with the corresponding log2(fold-change) vs. -log10(adjusted p value) volcano plots. (B) Euler plot of the number of differentially expressed genes (DEG, log2(fc)>1, p.adj<0.01) that overlap in MG and MDMs as shown in (A). (C) Molecular Signatures Database (MSigDB) “Hallmark” gene set collection overrepresentation analysis (ORA) in genes upregulated in both gliomas and BrMs vs. non-tumor brain tissue or healthy donors in MDMs and MG in MDMs and MG. Dot sizes reflect the fraction of gene set members found within the analyzed DEGs, and dot color indicates cell type. (D) Heatmap of fold changes of macrophage M1 and M2 polarization marker genes (absolute log2(fc)>1, p.adj<0.05) in MDMs and MG in gliomas and BrMs. Blank tiles indicate the lack of significant fold change. Genes are annotated with their canonical stimuli and the associated polarization phenotype. (GC = glucocorticoid, Ic = immune complexes, IFNγ = Interferon gamma, IL10 = interleukin 10, IL4 = interleukin 4, LPS = lipopolysaccharide, TGFβ = transforming growth factor beta). (E) Overlap between leading edge metagenes (LEMs) in MG and MDMs in gliomas and BrMs. Tile fill color indicates significance of overlap determined by hypergeometric testing (-log10(p.adj)). (F) String-DB protein-protein-interaction network of the intersect from IFN Type-1 group 2 modules from LEMs “BrM MG 1”, “Glioma MDM 1” and “BrM MDM 4”. Genes selected for validation through qRT-PCR are highlighted in red (corresponding data shown in Figure 3E). Node size indicates the centrality, while edge width corresponds to the String-DB interaction score (only scores >700, i.e. with a high degree of confidence have been included).

8. Supplementary Table 1. Summary of clinical information for FCM analyses, related to Figure 1.

BrM = brain metastasis, ER = estrogen receptor, HER2 = Erb-B2 Receptor Tyrosine Kinase 2, HR = hormone receptor, NOS = not otherwise specified, NSCLC = non-small-cell lung cancer, P = primary tumor, SCLC = small-cell lung cancer

5. Figure S5. Protein concentration in bulk tumor tissues and relation to cell type-associated SOM spots, related to Figure 5.

(A) Bulk tissue protein concentrations of indicated proteins in non-tumor brain (n=3), gliomas (n=14) and BrMs (n=12). Color indicates disease type and IDH status. (B) Heatmap of self-organizing map (SOM) spot metagene expression across the analyzed samples. Rows were z-scored and have been hierarchically clustered, columns were ordered by cell type, disease type and IDH mutation status.

4. Figure S4. IDH wt specific alterations in TAMs, related to Figure 4.

(A) Representative IF image and cell type quantification below of non-immune cells (CD45−), non-TAM immune cells (CD45+, P2RY12/CD68−, CD49D+/−), MG (CD45+, P2RY12/CD68+, CD49D) and MDM (CD45+, P2RY12/CD68+, CD49D+) and vessels (CD31+) in IDH wt glioma. Dashed line indicates the border of the perivascular niche (PVN), scale bar represents 100µm. (B) Heatmap of cell-type GSVA enrichment scores of micro-dissected Ivy GAP glioblastoma samples (dataset from Puchalski et al., 2018). Columns are ordered by anatomical location, rows have been z-scored. (C) Gene set enrichment analysis (GSEA) results of MSigDB “C2” antigen processing and cross-presentation associated pathways in T-MDMs vs. T-MG in IDH wt glioma. (D) Heatmap of MDM IDH wt gene set expression in sorted MG and MDMs from IDH mut and wt glioma samples. Columns are ordered by IDH status and cell type, expression values have been z-scored. (E) Plot of z-scored MDM IDH wt signature scores in the TCGA glioma dataset. Subjects are ranked by their enrichment score (small amount of random variation added for readability) and the IDH status is indicated by color. (F) Kaplan-Meier estimator of survival in the combined TCGA glioma cohort based on the enrichment for a cell type-specific T-MDM signature (see Figure S2E). (G) ORA of “innate anti-PD-1 resistance” (IPRES) signatures within DEG from MG- and MDMs in IDH wt gliomas DEGs (vs. MG from IDH mut tumors) with tile fill indicating the -log10 of the adjusted p value. (H) Gene set variation analysis (GSVA) of IPRES signatures in CD45− cells, MG, and MDMs from IDH mut and IDH wt gliomas. Columns are ordered by cell type, rows (z-score) have been hierarchically clustered.

6. Figure S6. Gene expression analysis in BrM-TAMs, related to Figure 6.

(A) Overlap of the number of differentially expressed genes (DEG, log2(fc) > 1, p.adj < 0.05) in MG and (B) MDMs in the indicated comparisons. BrM-specific genes sets are highlighted in grey within each cell type. The intersect of highlighted BrM-MG and BrM-MDM sets contains 87 genes. (C) GSEA of the “Biocarta IL-6 pathway” in BrM-MG vs. -MDM and the (D) “Naba core matrisome” gene set from the MSigDB “C2” collection in BrM-MDM vs. -MG. (E) Expression (log10-transformed normalized counts) of neutrophil-recruiting chemokines and receptors in sorted MG, MDMs and neutrophil populations from IDH wt and BrM samples.

9

Supplementary Table 2. Flow cytometry markers and immune cell definitions, related to Figure 1 and Figure S1.

10. Supplementary Table 3. Clinical characteristics (tumor type, grade, histology, IDH status, previous therapies) of patients included in the (A) RNA sequencing analysis and (B) protein array analysis.

BC = breast cancer, BrM = brain metastasis, ER = estrogen receptor, HER2 = Erb-B2 Receptor Tyrosine Kinase 2, HR = hormone receptor, NOS = not otherwise specified, NSCLC = non-small-cell lung cancer, P = primary tumor.

7. Figure S7. Correlation of WGCNA modules to external traits and module pathway ORA, related to Figure 7.

(A) Representative immunofluorescence images in IDH wt gliomas and BrMs. Scale bars = 100µm, boxed area is shown in higher magnification in Figure 7A. (B) Heatmap of the weighted gene correlation network analysis (WGCNA) module eigengene (= first principal component of expression data, columns, module columns are labelled with a color code) correlation to the traits (rows, cell type and disease, abundance of CD4+ or CD8+ T-cells in % of CD45+). Values inside the cells state Pearson’s r and the associated p value. (C) “Brown” BrM-MDM module MSigDB “C2CP” ORA results (p value < 0.01) enrichment map network visualization. Node size represents p-value, edge thickness reflects overlap of genes between gene sets.

12

Supplementary Table 5. RNA-seq fold changes in MG and MDMs in IDH wt gliomas vs. IDH mut MG, related to Figure 4.

14

Supplementary Table 7. qRT PCR TaqMan assays, related to Key Resources Table.

11

Supplementary Table 4. RNA-seq fold changes in MG and MDMs in gliomas and BrMs vs. non-tumor MG and in vitro generated MDMs respectively, related to Figure 3.

13

Supplementary Table 6. RNA-seq fold changes in MG and MDMs in BrMs vs. IDH wt gliomas, related to Figure 6.

Acknowledgements:

We thank Prof. Ron Stoop, Dr. Nathalie Piazzon, the Neurosurgery/Neuro-oncology clinical and nursing teams at CHUV and MSKCC for excellent infrastructural support, Joyce lab members for insightful discussion, Hegi lab members for technical help during sample processing, and Vladimir Wischnewski for critical manuscript review. We thank UNIL and MSKCC Flow Cytometry Core Facilities for exceptional technical assistance, especially Romain Bedel. Finally, we convey our immense gratitude to all patients who volunteered to participate in this study. Research in the Joyce lab is supported by Swiss Cancer League, Swiss Bridge Award, Ludwig Institute for Cancer Research, University of Lausanne, Breast Cancer Research Foundation, Cancer Research UK. FK was supported in part by the German Research Foundation (KL2491/1–1) and Fondation Medic, and KS by the Austrian FWF (J4343-B28).

The results shown here are in part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga.

Footnotes

Declaration of Interests:

The authors declare no competing interests.

References

  1. Afik R, Zigmond E, Vugman M, Klepfish M, Shimshoni E, Pasmanik-Chor M, Shenoy A, Bassat E, Halpern Z, Geiger T, et al. (2016). Tumor macrophages are pivotal constructors of tumor collagenous matrix. J Exp Med 213, 2315–2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, Gottardo N, Gutmann DH, Hargrave D, Holland EC, et al. (2019). Challenges to curing primary brain tumours. Nature Reviews Clinical Oncology 16, 509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baddeley AD, Eysenck MW, and Anderson MC (2015). Spatial Point Patterns: Methodology and Applications with R (London: Chapman and Hall/CRC Press; ). [Google Scholar]
  4. Barletta KE, Ley K, and Mehrad B (2012). Regulation of neutrophil function by adenosine. Arterioscler Thromb Vasc Biol 32, 856–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bates D, Machler M, Bolker BM, and Walker SC (2015). Fitting Linear Mixed-Effects Models Using lme4. Journal of Statistical Software 67, 1–48. [Google Scholar]
  6. Benci JL, Xu BH, Qiu Y, Wu TJ, Dada H, Twyman-Saint Victor C, Cucolo L, Lee DSM, Pauken KE, Huang AC, et al. (2016). Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell 167, 1540–1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bouralexis S, Findlay DM, and Evdokiou A (2005). Death to the bad guys: targeting cancer via Apo2L/TRAIL. Apoptosis 10, 35–51. [DOI] [PubMed] [Google Scholar]
  8. Bournazos S, Wang TT, and Ravetch JV (2016). The Role and Function of Fcgamma Receptors on Myeloid Cells. Microbiol Spectr 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bowman RL, Klemm F, Akkari L, Pyonteck SM, Sevenich L, Quail DF, Dhara S, Simpson K, Gardner EE, Iacobuzio-Donahue CA, et al. (2016). Macrophage Ontogeny Underlies Differences in Tumor-Specific Education in Brain Malignancies. Cell Rep 17, 2445–2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cagney DN, Martin AM, Catalano PJ, Redig AJ, Lin NU, Lee EQ, Wen PY, Dunn IF, Bi WL, Weiss SE, et al. (2017). Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: a population-based study. Neuro Oncol 19, 1511–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray BA, Morozova O, Newton Y, Radenbaugh A, Pagnotta SM, et al. (2016). Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 164, 550–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chen HM, van der Touw W, Wang YS, Kang K, Mai S, Zhang J, Alsina-Beauchamp D, Duty JA, Mungamuri SK, Zhang B, et al. (2018). Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest 128, 5647–5662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chen Z, Feng X, Herting CJ, Garcia VA, Nie K, Pong WW, Rasmussen R, Dwivedi B, Seby S, Wolf SA, et al. (2017). Cellular and Molecular Identity of Tumor-Associated Macrophages in Glioblastoma. Cancer Res 77, 2266–2278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber SH, Woroniecka K, Elsamadicy AA, Dechant CA, Kemeny HR, et al. (2018). Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med 24, 1459–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Coffelt SB, Wellenstein MD, and de Visser KE (2016). Neutrophils in cancer: neutral no more. Nat Rev Cancer 16, 431–446. [DOI] [PubMed] [Google Scholar]
  16. Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, et al. (2016). TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res 44, e71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cui Z, Liao J, Cheong N, Longoria C, Cao G, DeLisser HM, and Savani RC (2019). The Receptor for Hyaluronan-Mediated Motility (CD168) promotes inflammation and fibrosis after acute lung injury. Matrix Biol 78–79, 255–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Filkova M, Haluzik M, Gay S, and Senolt L (2009). The role of resistin as a regulator of inflammation: Implications for various human pathologies. Clin Immunol 133, 157–170. [DOI] [PubMed] [Google Scholar]
  20. Friedman J, Hastie T, and Tibshirani R (2010). Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw 33, 1–22. [PMC free article] [PubMed] [Google Scholar]
  21. Gabrusiewicz K, Rodriguez B, Wei J, Hashimoto Y, Healy LM, Maiti SN, Thomas G, Zhou S, Wang Q, Elakkad A, et al. (2016). Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype. JCI Insight 1(2), e85841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Gaujoux R, and Seoighe C (2010). A flexible R package for nonnegative matrix factorization. BMC Bioinformatics 11, 367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J, Dahan R, Harris RA, Rantalainen M, Klevebring D, et al. (2016). Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep 15, 2000–2011. [DOI] [PubMed] [Google Scholar]
  24. Glodde N, Bald T, van den Boorn-Konijnenberg D, Nakamura K, O’Donnell JS, Szczepanski S, Brandes M, Eickhoff S, Das I, Shridhar N, et al. (2017). Reactive Neutrophil Responses Dependent on the Receptor Tyrosine Kinase c-MET Limit Cancer Immunotherapy. Immunity 47, 789–802. [DOI] [PubMed] [Google Scholar]
  25. Godec J, Tan Y, Liberzon A, Tamayo P, Bhattacharya S, Butte AJ, Mesirov JP, and Haining WN (2016). Compendium of Immune Signatures Identifies Conserved and Species-Specific Biology in Response to Inflammation. Immunity 44, 194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Gutmann DH, and Kettenmann H (2019). Microglia/Brain Macrophages as Central Drivers of Brain Tumor Pathobiology. Neuron 104, 442–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Hanzelmann S, Castelo R, and Guinney J (2013). GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Hendriks LEL, Henon C, Auclin E, Mezquita L, Ferrara R, Audigier-Valette C, Mazieres J, Lefebvre C, Rabeau A, Le Moulec S, et al. (2019). Outcome of Patients with Non-Small Cell Lung Cancer and Brain Metastases Treated with Checkpoint Inhibitors. J Thorac Oncol 14, 1244–1254. [DOI] [PubMed] [Google Scholar]
  29. Huang QQ, Hossain MM, Wu K, Parai K, Pope RM, and Jin JP (2008). Role of H2-calponin in regulating macrophage motility and phagocytosis. J Biol Chem 283, 25887–25899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, et al. (2016). Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165, 35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Isobe M, Izawa K, Sugiuchi M, Sakanishi T, Kaitani A, Takamori A, Maehara A, Matsukawa T, Takahashi M, Yamanishi Y, et al. (2018). The CD300e molecule in mice is an immune-activating receptor. J Biol Chem 293, 3793–3805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Jackson CM, Choi J, and Lim M (2019). Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol 20, 1100–1109. [DOI] [PubMed] [Google Scholar]
  33. Kai F, Drain AP, and Weaver VM (2019). The Extracellular Matrix Modulates the Metastatic Journey. Dev Cell 49, 332–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Klemm F, and Joyce JA (2015). Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 25, 198–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Kobayashi M, Chung JS, Beg M, Arriaga Y, Verma U, Courtney K, Mansour J, Haley B, Khan S, Horiuchi Y, et al. (2019). Blocking Monocytic Myeloid-Derived Suppressor Cell Function via Anti-DC-HIL/GPNMB Antibody Restores the In Vitro Integrity of T Cells from Cancer Patients. Clin Cancer Res 25, 828–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Kowal J, Kornete M, and Joyce JA (2019). Re-education of macrophages as a therapeutic strategy in cancer. Immunotherapy 11, 677–689. [DOI] [PubMed] [Google Scholar]
  37. Langfelder P, and Horvath S (2008). WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Lecuyer MA, Saint-Laurent O, Bourbonniere L, Larouche S, Larochelle C, Michel L, Charabati M, Abadier M, Zandee S, Haghayegh Jahromi N, et al. (2017). Dual role of ALCAM in neuroinflammation and blood-brain barrier homeostasis. Proc Natl Acad Sci U S A 114, E524–E533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Li L, Yan J, Xu J, Liu CQ, Zhen ZJ, Chen HW, Ji Y, Wu ZP, Hu JY, Zheng L, et al. (2014). CXCL17 expression predicts poor prognosis and correlates with adverse immune infiltration in hepatocellular carcinoma. PLoS One 9, e110064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Li Y, Xie Y, Hao J, Liu J, Ning Y, Tang Q, Ma M, Zhou H, Guan S, Zhou Q, et al. (2018). ER-localized protein-Herpud1 is a new mediator of IL-4-induced macrophage polarization and migration. Exp Cell Res 368, 167–173. [DOI] [PubMed] [Google Scholar]
  41. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, and Tamayo P (2015). The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 1, 417–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Lim M, Xia Y, Bettegowda C, and Weller M (2018). Current state of immunotherapy for glioblastoma. Nat Rev Clin Oncol 15, 422–442. [DOI] [PubMed] [Google Scholar]
  43. Liu Q, Johnson EM, Lam RK, Wang Q, Bo Ye H, Wilson EN, Minhas PS, Liu L, Swarovski MS, Tran S, et al. (2019). Peripheral TREM1 responses to brain and intestinal immunogens amplify stroke severity. Nat Immunol 20, 1023–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Loffler-Wirth H, Kalcher M, and Binder H (2015). oposSOM: R-package for high-dimensional portraying of genome-wide expression landscapes on bioconductor. Bioinformatics 31, 3225–3227. [DOI] [PubMed] [Google Scholar]
  45. Long GV, Atkinson V, Lo S, Sandhu S, Guminski AD, Brown MP, Wilmott JS, Edwards J, Gonzalez M, Scolyer RA, et al. (2018). Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol 19, 672–681. [DOI] [PubMed] [Google Scholar]
  46. Love MI, Huber W, and Anders S (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Mantovani A, Marchesi F, Malesci A, Laghi L, and Allavena P (2017). Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14, 399–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Meng X, Duan C, Pang H, Chen Q, Han B, Zha C, Dinislam M, Wu P, Li Z, Zhao S, et al. (2019). DNA damage repair alterations modulate M2 polarization of microglia to remodel the tumor microenvironment via the p53-mediated MDK expression in glioma. EBioMedicine 41, 185–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Mohan V, Das A, and Sagi I (2020). Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol 62, 192–200. [DOI] [PubMed] [Google Scholar]
  50. Morianos I, Papadopoulou G, Semitekolou M, and Xanthou G (2019). Activin-A in the regulation of immunity in health and disease. J Autoimmun 104, 102314. [DOI] [PubMed] [Google Scholar]
  51. Muller S, Kohanbash G, Liu SJ, Alvarado B, Carrera D, Bhaduri A, Watchmaker PB, Yagnik G, Di Lullo E, Malatesta M, et al. (2017). Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol 18, 234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al. (2014). Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Mushtaq MU, Papadas A, Pagenkopf A, Flietner E, Morrow Z, Chaudhary SG, and Asimakopoulos F (2018). Tumor matrix remodeling and novel immunotherapies: the promise of matrix-derived immune biomarkers. J Immunother Cancer 6, 65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Noy R, and Pollard JW (2014). Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Olson OC, and Joyce JA (2015). Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer 15, 712–729. [DOI] [PubMed] [Google Scholar]
  56. Ortolan E, Augeri S, Fissolo G, Musso I, and Funaro A (2019). CD157: From immunoregulatory protein to potential therapeutic target. Immunol Lett 205, 59–64. [DOI] [PubMed] [Google Scholar]
  57. Puchalski RB, Shah N, Miller J, Dalley R, Nomura SR, Yoon JG, Smith KA, Lankerovich M, Bertagnolli D, Bickley K, et al. (2018). An anatomic transcriptional atlas of human glioblastoma. Science 360, 660–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V, et al. (2013). CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 19, 1264–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Qiao S, Qian Y, Xu G, Luo Q, and Zhang Z (2019). Long-term characterization of activated microglia/macrophages facilitating the development of experimental brain metastasis through intravital microscopic imaging. J Neuroinflammation 16, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Quail DF, Bowman RL, Akkari L, Quick ML, Schuhmacher AJ, Huse JT, Holland EC, Sutton JC, and Joyce JA (2016). The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science 352, 952–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Quail DF, and Joyce JA (2017). The Microenvironmental Landscape of Brain Tumors. Cancer Cell 31, 326–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. R Core Team (2018). R: A Language and Environment for Statistical Computing.
  63. Racle J, de Jonge K, Baumgaertner P, Speiser DE, and Gfeller D (2017). Simultaneous enumeration of cancer and immune cell types from bulk tumor gene expression data. Elife 6, e26476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Rau A, Gallopin M, Celeux G, and Jaffrezic F (2013). Data-based filtering for replicated high-throughput transcriptome sequencing experiments. Bioinformatics 29, 2146–2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Richards J, Gabunia K, Kelemen SE, Kako F, Choi ET, and Autieri MV (2015). Interleukin-19 increases angiogenesis in ischemic hind limbs by direct effects on both endothelial cells and macrophage polarization. J Mol Cell Cardiol 79, 21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Ripoll VM, Irvine KM, Ravasi T, Sweet MJ, and Hume DA (2007). Gpnmb is induced in macrophages by IFN-gamma and lipopolysaccharide and acts as a feedback regulator of proinflammatory responses. J Immunol 178, 6557–6566. [DOI] [PubMed] [Google Scholar]
  67. Robinson JT, Thorvaldsdottir H, Wenger AM, Zehir A, and Mesirov JP (2017). Variant Review with the Integrative Genomics Viewer. Cancer Res 77, e31–e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Rodon L, Gonzalez-Junca A, Inda Mdel M, Sala-Hojman A, Martinez-Saez E, and Seoane J (2014). Active CREB1 promotes a malignant TGFbeta2 autocrine loop in glioblastoma. Cancer Discov 4, 1230–1241. [DOI] [PubMed] [Google Scholar]
  69. Sankowski R, Bottcher C, Masuda T, Geirsdottir L, Sagar, Sindram E, Seredenina T, Muhs A, Scheiwe C, Shah MJ, et al. (2019). Mapping microglia states in the human brain through the integration of high-dimensional techniques. Nat Neurosci 22, 2098–2110. [DOI] [PubMed] [Google Scholar]
  70. Santana Carrero RM, Beceren-Braun F, Rivas SC, Hegde SM, Gangadharan A, Plote D, Pham G, Anthony SM, and Schluns KS (2019). IL-15 is a component of the inflammatory milieu in the tumor microenvironment promoting antitumor responses. Proc Natl Acad Sci U S A 116, 599–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, Lopez-Janeiro A, Porciuncula A, Idoate MA, Inoges S, de Andrea C, Lopez-Diaz de Cerio A, Tejada S, et al. (2019). Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med 25, 470–476. [DOI] [PubMed] [Google Scholar]
  72. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. (2005). Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996. [DOI] [PubMed] [Google Scholar]
  73. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al. (2017). The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res 45, D362–D368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Szulzewsky F, Arora S, de Witte L, Ulas T, Markovic D, Schultze JL, Holland EC, Synowitz M, Wolf SA, and Kettenmann H (2016). Human glioblastoma-associated microglia/monocytes express a distinct RNA profile compared to human control and murine samples. Glia 64, 1416–1436. [DOI] [PubMed] [Google Scholar]
  76. Tawbi HA, Forsyth PA, Algazi A, Hamid O, Hodi FS, Moschos SJ, Khushalani NI, Lewis K, Lao CD, Postow MA, et al. (2018). Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. N Engl J Med 379, 722–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Thapa B, and Lee K (2019). Metabolic influence on macrophage polarization and pathogenesis. BMB Rep 52, 360–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. The GTEx Consortium (2013). The Genotype-Tissue Expression (GTEx) project. Nat Genet 45, 580–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, Senju S, Ihn H, Nishimura Y, and Oshiumi H (2018). Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res 78, 5011–5022. [DOI] [PubMed] [Google Scholar]
  80. van der Touw W, Chen HM, Pan PY, and Chen SH (2017). LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother 66, 1079–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Van P, Jiang W, Gottardo R, and Finak G (2018). ggCyto: next generation open-source visualization software for cytometry. Bioinformatics 34, 3951–3953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Vareslija D, Priedigkeit N, Fagan A, Purcell S, Cosgrove N, O’Halloran PJ, Ward E, Cocchiglia S, Hartmaier R, Castro CA, et al. (2019). Transcriptome Characterization of Matched Primary Breast and Brain Metastatic Tumors to Detect Novel Actionable Targets. J Natl Cancer Inst 111, 388–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Venteicher AS, Tirosh I, Hebert C, Yizhak K, Neftel C, Filbin MG, Hovestadt V, Escalante LE, Shaw ML, Rodman C, et al. (2017). Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science 355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, Pfeil J, Narkizian J, Deran AD, Musselman-Brown A, et al. (2017). Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol 35, 314–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Wei SC, Duffy CR, and Allison JP (2018). Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 8, 1069–1086. [DOI] [PubMed] [Google Scholar]
  86. Wickham H (2016). ggplot2: Elegant Graphics for Data Analysis (New York: Springer-Verlag; ). [Google Scholar]
  87. Xu P, Zhang X, Liu Q, Xie Y, Shi X, Chen J, Li Y, Guo H, Sun R, Hong Y, et al. (2019). Microglial TREM-1 receptor mediates neuroinflammatory injury via interaction with SYK in experimental ischemic stroke. Cell Death Dis 10, 555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, et al. (2014). Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40, 274–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Yan D, Kowal J, Akkari L, Schuhmacher AJ, Huse JT, West BL, and Joyce JA (2017). Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene 36, 6049–6058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Yang TH, St John LS, Garber HR, Kerros C, Ruisaard KE, Clise-Dwyer K, Alatrash G, Ma Q, and Molldrem JJ (2018). Membrane-Associated Proteinase 3 on Granulocytes and Acute Myeloid Leukemia Inhibits T Cell Proliferation. J Immunol 201, 1389–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Young MD, Wakefield MJ, Smyth GK, and Oshlack A (2010). Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol 11, R14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Zaiss DMW, Gause WC, Osborne LC, and Artis D (2015). Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair. Immunity 42, 216–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Zhai L, Ladomersky E, Lenzen A, Nguyen B, Patel R, Lauing KL, Wu M, and Wainwright DA (2018). IDO1 in cancer: a Gemini of immune checkpoints. Cell Mol Immunol 15, 447–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Zhao J, Sun L, and Li X (2017). Commanding CNS Invasion: GM-CSF. Immunity 46, 165–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, Xiao C, Bonorino C, Lu LF, Bui JD, et al. (2019). PD-L1:CD80 Cis-Heterodimer Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-4 Pathways. Immunity 51, 1059–1073 e1059. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1. Figure S1. Fluorescence-activated cell sorting (FACS) of cell populations and RNA-sequencing, related to Figure 1.

(A) Flow cytometry (FCM) plots illustrating the gating strategy employed during FAC-sorting of immune cell populations in non-tumor and tumor tissue (for cell type markers, see Table S2). (B) tSNE plot of gene expression data (500 most variable genes) from all sorted cell populations (n=226) across the complete clinical cohort (MG = microglia, MDM = monocyte-derived macrophages, reference = unmatched healthy blood and in vitro generated MDMs).

See also Table S2.

2. Figure S2. MG and MDM marker expression, related to Figure 2.

(A) Normalized counts (log10 transformed) of MG and MDM marker genes in sorted CD49Dlow MG and CD49Dhigh MDM populations across both non-tumor and tumor tissues (reference = healthy donor in vitro generated MDMs). (B) Percentage of CD49Dlow MG and CD49Dhigh MDMs positive for P2RY12 and CD68 as determined by FCM in relation to the total number of MG/MDMs in non-tumor (n=8) and tumor tissue (nIDHmut=6, nIDHwt=6, nBrM=9). (C) Single channel and merged immunofluorescence (IF) images of CD45, CD68, P2RY12 and CD49D stainings which were employed to delineate MG and MDMs. The last column shows the resulting Visiopharm cell type assignments for quantitative analyses (MG (CD45+, P2RY12+/CD68+, CD49D−), MDM (CD45+, P2RY12+/CD68+, CD49D+), non-immune cells (CD45−) and non-TAM-immune cells (CD45+, P2RY12−/CD68−, CD49D−/+). Scale bars represent 100µm. (D) Scatter plots of the abundance of MG and MDMs as determined by IF vs. FCM in non-tumor (n=4) and tumor tissues (nIDHmut=13, nIDHwt=14, nBrM=18) processed independently from the same individual samples. Pearson’s correlation coefficient and significance are indicated at the top of each plot. (E) Heatmap of human MG- and MDM-specific gene set expression used for deconvolution across FAC-sorted population samples from all disease types.

3. Figure S3. Analysis of differentially expressed genes and TAM activation patterns, related to Figure 3.

(A) Summary of contrasts applied when performing differential gene expression analysis in MG and MDMs in gliomas (regardless of IDH status) and BrMs (from all primaries) in comparison to normal controls (non-tumor brain MG and in vitro differentiated MDMs respectively) with the corresponding log2(fold-change) vs. -log10(adjusted p value) volcano plots. (B) Euler plot of the number of differentially expressed genes (DEG, log2(fc)>1, p.adj<0.01) that overlap in MG and MDMs as shown in (A). (C) Molecular Signatures Database (MSigDB) “Hallmark” gene set collection overrepresentation analysis (ORA) in genes upregulated in both gliomas and BrMs vs. non-tumor brain tissue or healthy donors in MDMs and MG in MDMs and MG. Dot sizes reflect the fraction of gene set members found within the analyzed DEGs, and dot color indicates cell type. (D) Heatmap of fold changes of macrophage M1 and M2 polarization marker genes (absolute log2(fc)>1, p.adj<0.05) in MDMs and MG in gliomas and BrMs. Blank tiles indicate the lack of significant fold change. Genes are annotated with their canonical stimuli and the associated polarization phenotype. (GC = glucocorticoid, Ic = immune complexes, IFNγ = Interferon gamma, IL10 = interleukin 10, IL4 = interleukin 4, LPS = lipopolysaccharide, TGFβ = transforming growth factor beta). (E) Overlap between leading edge metagenes (LEMs) in MG and MDMs in gliomas and BrMs. Tile fill color indicates significance of overlap determined by hypergeometric testing (-log10(p.adj)). (F) String-DB protein-protein-interaction network of the intersect from IFN Type-1 group 2 modules from LEMs “BrM MG 1”, “Glioma MDM 1” and “BrM MDM 4”. Genes selected for validation through qRT-PCR are highlighted in red (corresponding data shown in Figure 3E). Node size indicates the centrality, while edge width corresponds to the String-DB interaction score (only scores >700, i.e. with a high degree of confidence have been included).

8. Supplementary Table 1. Summary of clinical information for FCM analyses, related to Figure 1.

BrM = brain metastasis, ER = estrogen receptor, HER2 = Erb-B2 Receptor Tyrosine Kinase 2, HR = hormone receptor, NOS = not otherwise specified, NSCLC = non-small-cell lung cancer, P = primary tumor, SCLC = small-cell lung cancer

5. Figure S5. Protein concentration in bulk tumor tissues and relation to cell type-associated SOM spots, related to Figure 5.

(A) Bulk tissue protein concentrations of indicated proteins in non-tumor brain (n=3), gliomas (n=14) and BrMs (n=12). Color indicates disease type and IDH status. (B) Heatmap of self-organizing map (SOM) spot metagene expression across the analyzed samples. Rows were z-scored and have been hierarchically clustered, columns were ordered by cell type, disease type and IDH mutation status.

4. Figure S4. IDH wt specific alterations in TAMs, related to Figure 4.

(A) Representative IF image and cell type quantification below of non-immune cells (CD45−), non-TAM immune cells (CD45+, P2RY12/CD68−, CD49D+/−), MG (CD45+, P2RY12/CD68+, CD49D) and MDM (CD45+, P2RY12/CD68+, CD49D+) and vessels (CD31+) in IDH wt glioma. Dashed line indicates the border of the perivascular niche (PVN), scale bar represents 100µm. (B) Heatmap of cell-type GSVA enrichment scores of micro-dissected Ivy GAP glioblastoma samples (dataset from Puchalski et al., 2018). Columns are ordered by anatomical location, rows have been z-scored. (C) Gene set enrichment analysis (GSEA) results of MSigDB “C2” antigen processing and cross-presentation associated pathways in T-MDMs vs. T-MG in IDH wt glioma. (D) Heatmap of MDM IDH wt gene set expression in sorted MG and MDMs from IDH mut and wt glioma samples. Columns are ordered by IDH status and cell type, expression values have been z-scored. (E) Plot of z-scored MDM IDH wt signature scores in the TCGA glioma dataset. Subjects are ranked by their enrichment score (small amount of random variation added for readability) and the IDH status is indicated by color. (F) Kaplan-Meier estimator of survival in the combined TCGA glioma cohort based on the enrichment for a cell type-specific T-MDM signature (see Figure S2E). (G) ORA of “innate anti-PD-1 resistance” (IPRES) signatures within DEG from MG- and MDMs in IDH wt gliomas DEGs (vs. MG from IDH mut tumors) with tile fill indicating the -log10 of the adjusted p value. (H) Gene set variation analysis (GSVA) of IPRES signatures in CD45− cells, MG, and MDMs from IDH mut and IDH wt gliomas. Columns are ordered by cell type, rows (z-score) have been hierarchically clustered.

6. Figure S6. Gene expression analysis in BrM-TAMs, related to Figure 6.

(A) Overlap of the number of differentially expressed genes (DEG, log2(fc) > 1, p.adj < 0.05) in MG and (B) MDMs in the indicated comparisons. BrM-specific genes sets are highlighted in grey within each cell type. The intersect of highlighted BrM-MG and BrM-MDM sets contains 87 genes. (C) GSEA of the “Biocarta IL-6 pathway” in BrM-MG vs. -MDM and the (D) “Naba core matrisome” gene set from the MSigDB “C2” collection in BrM-MDM vs. -MG. (E) Expression (log10-transformed normalized counts) of neutrophil-recruiting chemokines and receptors in sorted MG, MDMs and neutrophil populations from IDH wt and BrM samples.

9

Supplementary Table 2. Flow cytometry markers and immune cell definitions, related to Figure 1 and Figure S1.

10. Supplementary Table 3. Clinical characteristics (tumor type, grade, histology, IDH status, previous therapies) of patients included in the (A) RNA sequencing analysis and (B) protein array analysis.

BC = breast cancer, BrM = brain metastasis, ER = estrogen receptor, HER2 = Erb-B2 Receptor Tyrosine Kinase 2, HR = hormone receptor, NOS = not otherwise specified, NSCLC = non-small-cell lung cancer, P = primary tumor.

7. Figure S7. Correlation of WGCNA modules to external traits and module pathway ORA, related to Figure 7.

(A) Representative immunofluorescence images in IDH wt gliomas and BrMs. Scale bars = 100µm, boxed area is shown in higher magnification in Figure 7A. (B) Heatmap of the weighted gene correlation network analysis (WGCNA) module eigengene (= first principal component of expression data, columns, module columns are labelled with a color code) correlation to the traits (rows, cell type and disease, abundance of CD4+ or CD8+ T-cells in % of CD45+). Values inside the cells state Pearson’s r and the associated p value. (C) “Brown” BrM-MDM module MSigDB “C2CP” ORA results (p value < 0.01) enrichment map network visualization. Node size represents p-value, edge thickness reflects overlap of genes between gene sets.

12

Supplementary Table 5. RNA-seq fold changes in MG and MDMs in IDH wt gliomas vs. IDH mut MG, related to Figure 4.

14

Supplementary Table 7. qRT PCR TaqMan assays, related to Key Resources Table.

11

Supplementary Table 4. RNA-seq fold changes in MG and MDMs in gliomas and BrMs vs. non-tumor MG and in vitro generated MDMs respectively, related to Figure 3.

13

Supplementary Table 6. RNA-seq fold changes in MG and MDMs in BrMs vs. IDH wt gliomas, related to Figure 6.

Data Availability Statement

RNA-seq count expression data generated during this study can be visualized and downloaded at https://joycelab.shinyapps.io/braintime/. Due to patient privacy protection, the raw RNA-seq data will be made available upon request.

RESOURCES