Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Feb 28.
Published in final edited form as: Clin Cancer Res. 2021 Jun 3;27(15):4422–4434. doi: 10.1158/1078-0432.CCR-21-1549

Aging and CNS Myeloid Cell Depletion Attenuate Breast Cancer Brain Metastasis

Alex Man Lai Wu 1, Selamawit Gossa 2, Ramakrishna Samala 3, Monika A Chung 1, Brunilde Gril 1, Howard H Yang 4, Helen R Thorsheim 3, Andy D Tran 4,5, Debbie Wei 1, Esra Taner 1, Kristine Isanogle 6, Yuan Yang 4, Emma L Dolan 1, Christina Robinson 6, Simone Difilippantonio 6, Maxwell P Lee 4, Imran Khan 1, Quentin R Smith 3, Dorian B McGavern 2, Lalage M Wakefield 4, Patricia S Steeg 1
PMCID: PMC9974011  NIHMSID: NIHMS1870285  PMID: 34083229

Abstract

Purpose:

Breast cancer diagnosed in young patients is often aggressive. Because primary breast tumors from young and older patients have similar mutational patterns, we hypothesized that the young host microenvironment promotes more aggressive metastatic disease.

Experimental Design:

Triple-negative or luminal B breast cancer cell lines were injected into young and older mice side-by-side to quantify lung, liver, and brain metastases. Young and older mouse brains, metastatic and naïve, were analyzed by flow cytometry. Immune populations were depleted using antibodies or a colony-stimulating factor-1 receptor (CSF-1R) inhibitor, and brain metastasis assays were conducted. Effects on myeloid populations, astrogliosis, and the neuroinflammatory response were determined.

Results:

Brain metastases were 2- to 4-fold higher in young as compared with older mouse hosts in four models of triple-negative or luminal B breast cancer; no age effect was observed on liver or lung metastases. Aged brains, naïve or metastatic, contained fewer resident CNS myeloid cells. Use of a CSF-1R inhibitor to deplete myeloid cells, including both microglia and infiltrating macrophages, preferentially reduced brain metastasis burden in young mice. Downstream effects of CSF-1R inhibition in young mice resembled that of an aged brain in terms of myeloid numbers, induction of astrogliosis, and Semaphorin 3A secretion within the neuroinflammatory response.

Conclusions:

Host microenvironmental factors contribute to the aggressiveness of triple-negative and luminal B breast cancer brain metastasis. CSF-1R inhibitors may hold promise for young brain metastasis patients.

Introduction

An unsolved problem in breast cancer is its aggressive disease course when diagnosed in young patients. Twelve multivariate analyses of patients with breast cancer concluded that a young age at diagnosis is a significant correlate of poor outcome when adjustments were made for tumor grade and histopathology, patient socioeconomic status, patient race and ethnicity, year of diagnosis, molecular subtype of cancer, TNM status, treatment, laterality, detection mode, marital status, and family history; case���control studies came to the same conclusion (Supplementary Table S1; refs. 1, 2). Of the major subtypes of breast cancer at least two, triple negative (estrogen and progesterone receptor negative, HER2 normal, including basal cancers; refs. 35) and luminal B (hormone receptor positive, often with HER2 overexpression; refs. 2, 4, 6), have been associated with aggressive cancer in young patients. Young breast cancer patients are often treated aggressively and endure the side effects without superior outcomes (79).

The root causes of the aggressiveness of breast cancer in young patients are unknown. No consensus molecular mutations or epigenetic alterations have been identified and functionally validated in tumors from young patients (10). Because tumor cell-intrinsic characteristics have not provided clear mechanistic insight, here we hypothesized that the microenvironment of the young “host” may be a key contributor to the aggressive disease course in young women, using animal models of tumor metastasis as an endpoint. Herein, we addressed three aspects of age-related aggressiveness: (i) the potential difference in metastasis development between young and older animals using multiple models of triple-negative and luminal B disease, (ii) changes in host inflammatory and immune components in young and older animals bearing brain metastases, and (iii) myeloid cell contributions to age-related brain metastasis. The data define a role for host physiology in brain metastasis development in young patients and could lead to therapeutic strategies for this high-risk patient population.

Materials and Methods

Cell lines

Stock vials of cells were tested to be free of infectious microbes (e.g., mycoplasma) by PCR (Animal Diagnostic Laboratory Services, NCI). Cells for injections were used within three passages after thawing or within 15 passages for in vitro experiments. Human and mouse cell lines were authenticated by short tandem repeat analysis, which was performed by Laragen Inc. (October 2016) and ATCC (September/October 2020), respectively.

MDA-MB-231BR-eGFP (231-BR) and 4T1-BR cells were cultured in DMEM supplemented with 10% FBS. MDA-MB-231T, a subline of MDA-MB-231 breast cancer cells were obtained from Dr. Danny Welch (University of Kansas Medical Center). 4T1-Luc2 were purchased from PerkinElmer and maintained in DMEM supplemented with 10% FBS. E0771, 6DT1, and MVT1 cells were cultured as described previously (11). The brain-selected E0771 subline was generated by first injecting 1 × 105 parental E0771 into the left cardiac ventricle of 2-month-old female mice. Cells recovered from the brains of these mice were re-injected into a new cohort of mice. This was repeated five times to generate E0771-BR5 cells. 99LN-BrM cells that were derived from one round of in vivo selection to the brain was generously provided by Drs. Johanna Joyce and Florian Klemm (University of Lausanne). These cells were maintained in DMEM/F12 supplemented with 10% FBS and were selected in vivo three more times to generate 99LN-BrM4 cells. EOC2 cells were a gift from Dr. Jing Wu (Neuro-Oncology Branch, NCI) and cultured in DMEM supplemented with 10% FBS and 20% LADMAC conditioned media.

Animals

All animal experiments were approved by the Institutional Animal Care and Use Committees of the NCI (NCI-Bethesda and NCI-Frederick campuses), National Institute of Neurological Disorders and Stroke, Texas Tech University of Health Sciences Center, and Use Review Office of the United States Army Medical Research and Materiel Command. Unless specified otherwise, for aging studies in immunocompetent mice (BALB/C, C57BL/6, and FVB), young female mice were 4 to 6 months old and older female mice were 18 to 24 months old at the time of cancer cell injection. The older cohorts were obtained as retired female breeders and housed to the desired age. The young cohorts underwent one full cycle of pregnancy-lactation and were used >1 month after weaning of pups. Athymic nu/nu virgin mice were used at 2 to 3 months (young) and 12 to 15 months (older). Two-month-old female virgin mice were used for baseline immunophenotyping studies comparing naïve/healthy brains to metastatic brains and for immunodepletion experiments.

Brain and lung metastasis models

For brain metastasis models, cancer cells suspended in 100 μL sterile PBS were injected with aid of ultrasound into the left cardiac ventricle of mice under isoflurane anesthesia. 4T1-BR (5 × 104) and 231-BR (1.75 × 105) cells were injected into BALB/C and nu/nu mice, respectively. E0771-BR5 (1 × 105 or 2 × 105) and 99LN-BrM4 (5 × 105) were injected into C57BL/6 mice. For lung metastasis models, MDA-MB-231T (7.5 × 105) were suspended in 100 mL sterile PBS and injected into the tail-vein of nu/nu mice. 4T1-Luc2 (5 × 105), E0771 (1 × 105), MVT1 (2 × 105), and 6DT1 (3.5 × 104) cells were suspended in 50 mL sterile PBS and injected orthotopically into the #4 mammary fat pad (MFP) of syngeneic mice as described previously (11). For the 6DT1 model, tumors were resected at day 14. Primary tumor size was monitored twice a week and weighed after tissue collection.

Brain metastatic tumor burden was quantified from five (4T1-BR, MDA-MB-231BR, 99LN-BrM4) or ten (E0771-BR5) 8-μm-thick hematoxylin and eosin (H&E)-stained step-sections spaced 300 to 500 μm apart through the whole brain or one hemisphere. A metastatic cluster was defined as a group of four or more closely spaced metastatic deposits or one large lesion more than 300 μm in both dimensions. Total numbers of lung or liver metastases were quantified from one representative 5-μm-thick H&E-stained tissue section with all lung lobes or liver lobes visible.

Flow cytometry

Mice anaesthetized with isoflurane or heavily sedated with a terminal dose of chloral hydrate were transcardially perfused with saline. Single cell preparations were blocked with anti-CD16/32 and mouse IgG on ice for 10 minutes and then stained with a cocktail of fluorescently labelled antibodies for 30 minutes on ice. All samples were analyzed on a BD LSRFortessa or BD FACSymphony cell analyzer. Cell counts were determined using CountBright Absolute Counting Beads (Thermo Fischer Scientific, Catalog No. C36950). Data were analyzed using FlowJo version 10 (see Supplementary Materials and Methods).

Immune-depletion

To deplete CD4+ or CD8+ T cells, BALB/C were injected intraperitoneally with 250 μg of depleting antibodies on day -2, and then maintenance doses of 100 μg at day 2, 6, and 10. To deplete monocytic and granulocytic myeloid cells, BALB/C mice were injected intraperitoneally with 250 μg of anti-GR1 antibodies on day -1 and -2, followed by 100 μg doses at day 5 and 8. Control mice were injected with isotype rat IgG2b. See Supplementary Materials and Methods for antibody identifications. Mouse 4T1-BR breast cancer cells were injected into the left cardiac ventricle on day 0.

To deplete CNS myeloid cells in 2-month-old mice, mice were given PLX3397 (SelleckChem, Catalog No. S7818) formulated at 300 mg/kg in NIH-31 Open Diet (Envigo) ad libitum for 28 days prior to the injection of cancer cells. Mice remained on this diet until study termination. Control mice were given standard NIH-31 Open diet. To deplete resident CNS myeloid cells in a young versus older experiment, mice were given PLX3397 at 100 mg/kg body weight by daily oral gavage for 14 days prior to injection of cancer cells and then continuously thereafter (see Supplementary Materials and Methods). Control mice were given vehicle with an equivalent dose of DMSO.

Immunofluorescence staining of tissue sections

See Supplementary Materials and Methods for tissue acquisition, antibodies used, and image analysis. Frozen brain tissue sections (8- to 10-μm thick) were fixed or permeabilized with methanol for 5 minutes at −20°C, washed with PBS, blocked with 5% normal goat serum (Vector Labs) or 5% normal donkey serum (Jackson ImmunoResearch, Catalog No. 017–000–121) in PBS with 0.05% (v/v) Tween-20 (PBST), and then incubated overnight at 4°C with primary antibodies diluted in the appropriate blocking buffer. After washing slides with PBS, tissue sections were incubated with DAPI (2 mg/mL) and secondary antibodies diluted to 1:250 to 1:500 in 5% serum/PBST for 1 hour at room temperature. See Supplementary Materials and Methods for a list of primary and secondary antibodies used. Slides were coverslipped with Fluorescence Mounting Medium (Dako). Images of metastatic lesions and of uninvolved regions were taken using a Zeiss Axioskop with a 10×, 20×, or 40× objective. Metastatic lesions were identified on the basis of cytokeratin staining or dense clusters of DAPI. Uninvolved regions were defined as areas within the cerebral cortex without metastatic lesions and located at least 1 mm away from a lesion. Images of at least three distinct visual views of uninvolved brain regions were captured for each single tissue section.

In vitro assays

Boyden chamber motility and cell viability assays are described in Supplementary Materials and Methods.

Statistical analysis

Statistical tests were performed using GraphPad Prism 7, with P values < 0.05 considered significant. The Mann–Whitney U test and Wilcoxon rank test was used for comparing unpaired or pair-matched data, respectively. The Kruskal–Wallis test followed by Dunn test was used for multiple comparisons. All data presented, except for frequency data, are box-whisker plots or bar graphs depicting median ± interquartile range.

Refer to Supplementary Materials and Methods for blood–tumor barrier permeability assays, intracranial tumor model, microglia conditioned medium experiment, detailed methods of flow cytometry and immunofluorescence staining protocols including full list of antibodies used, tissue collection and preservation protocols, image analysis, RNA in situ hybridization (RNAscope), and methods used to analyze publicly available datasets.

Results

Breast tumors from young and older patients have a similar mutational spectrum

In further support of previously published findings that primary breast cancers from young patients have few if any defining genetic alterations (12), we compared the mutational status of primary breast tumors from young (≤40 years) and older patients (>40 years) in two online datasets, TCGA (n = 1,083) and METABRIC (n = 1,980; Supplementary Figs. S1 and S2). Approximately 8% and 6% of patients in the TCGA and METABRIC databases were young, respectively. No consistent mutations were observed between the two datasets, which may be partly due to differences in patient characteristics, for instance a higher proportion of younger women have TNBC in the METABRIC dataset compared with TCGA (Supplementary Fig. S1C). Similar conclusions were obtained when varying the age cutoff (Supplementary Figs. S3S6). Given the lack of somatic mutations consistently enriched in young tumors, we hypothesized instead that extrinsic microenvironmental factors contribute to age-related differences in breast cancer aggressiveness.

Young age promotes breast cancer metastasis specifically in the brain

Previous epidemiologic studies have demonstrated a strong association between young age at breast cancer diagnosis and risk of brain metastasis (1315). To test whether the younger brain is more permissive for breast cancer metastasis, brain-tropic breast cancer cell lines (-BR) were injected into young and older mice side-by-side via the left cardiac ventricle and metastatic clusters were enumerated. The definition of young and older mice varied by strain as immunodeficient mice do not survive as long (see Materials and Methods). In general, ages of the young (2–6 months) and older (>12 months) cohorts were chosen to model human age less than or greater than 40 years old, respectively. Nulliparous versus parous status was held constant within each experiment.

For the triple-negative subtype 4T1 breast cancer model, young parous BALB/C mice developed 4.5-fold more 4T1-BR brain metastatic clusters (Fig. 1A). Young parous C57BL/6 mice similarly developed more brain metastatic clusters when injected with E0771-BR5 cells (Fig. 1B), a subline of the mouse triple-negative E0771 breast cancer cells that we selected in vivo for increased capacity to form metastases (Supplementary Fig. S7AS7C). Young immunocompromised nulliparous nude mice injected with human MDA-MB-231-BR triple-negative breast cancer cells (231-BR) developed 2.3-fold more metastatic clusters (P < 0.05) compared with older mice (Fig. 1C). To test whether this phenomenon was restricted to the triple-negative subtype, a published 99LN-BrM luminal B model system (16) was subjected to three additional rounds of in vivo selection to produce 99LN-BrM4 cells with increased brain metastatic efficiency (Supplementary Fig. S7D and S7E). Young parous C57BL/6 mice injected with 99LN-BrM4 cells developed 3.5-fold more brain metastatic clusters compared with their older counterparts (Fig. 1D, top). Collectively, data from four models encompassing two molecular subtypes of breast cancer indicate that young age promotes breast cancer metastasis in the brain, or that older age protects against it.

Figure 1.

Figure 1.

A young age promotes breast cancer metastasis in the brain. Breast cancer cells were injected intracardially into young and older mice and metastatic tumor burden was quantified. Metastatic clusters, a group of four or more closely spaced metastatic deposits, or one large lesion > 300 μm in both dimensions, was used as the histological endpoint. A, Brain metastatic tumor burden in young (n = 18) and older (n = 13) BALB/C mice 13 days after inoculation with 4T1-BR cells. B, Brain metastatic tumor burden in young (n= 17) and older (n= 11) C57BL/6 mice 13 days after inoculation with E0771-BR5 cells. C, Brain metastatic tumor burden in young (n = 22) and older (n = 16) nude mice 30 days after inoculation with MDA-MB-231-BR (231-BR) cells. D, Metastatic tumor burden in the brain (top) and liver (bottom) of young (n = 23) and older (n = 15) C57BL/6 mice 29 to 35 days after inoculation with 99LN-BrM4 cells. Metastatic tumor burden reported is the average number of metastatic clusters per brain tissue section, or number of liver lesions in one representative tissue section. H&E tissue section scale bar, 500 μm. Each data point is one mouse. One independent experiment per model (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001, Mann–Whitney test).

We also determined the effect of age on metastasis to other sites. Many mice injected with 99LN-BrM4 cells simultaneously developed liver metastases but, unlike the brain, there was a trend towards reduced incidence and number of liver metastases in younger mice (Fig. 1D, bottom). Liver metastases were not reproducibly detected in the other brain-tropic model systems.

Breast cancer lung metastasis models were tested, including four spontaneous metastasis assays involving mammary fat pad injection with (Fig. 2A and B) or without (Fig. 2C and D) tumor resection, and one experimental metastasis assay where cells were injected into the tail vein (Fig. 2E). In most cases, lung metastasis did not vary by age, with the exception of 4T1-Luc2 where metastases were higher in older animals.

Figure 2.

Figure 2.

No effect of age on breast cancer metastasis in lung. A, Resected tumor weight and number of lung metastases in young (n = 18) and older (n = 8) BALB/C mice inoculated with 4T1-Luc2 cells into MFP. B, Resected tumor weight and number of lung metastases inyoung (n = 12) and older (n = 10) FVB mice inoculated with 6DT1 cells into MFP. C, Primary tumor weight and number of lung metastases in young (n = 18) and older (n= 15) C57BL/6 mice inoculated with parental E0771 cells into MFP. D, Primary tumor weight and number of lung metastases in young (n = 19) and older (n = 19) FVB mice inoculated with MVT1 cells into MFP. E, Number of lung metastases inyoung (n = 16) and older (n = 6) nude mice injected via tail-veinwith MDA-MB-231 cells. Each data point is one mouse. One independent experiment per model (*, P < 0.05, Mann–Whitney test).

The young brain confers a survival advantage to breast cancer cells

We next ruled out several potential underlying causes of the increased brain metastasis in young mice. Breast cancer cells must traverse the blood–brain barrier (BBB) to establish a hematogenous metastasis. At the experimental endpoint of a 4T1-BR metastasis assay, the permeability of the blood–tumor barrier, the remnants of the BBB once a metastasis has established, was comparable between young and older mice (Supplementary Fig. S8A). To specifically test whether the effect of age is exerted on tumor cells at the extravasation step, 4T1-BR cells were directly injected into the brains of young and older BALB/C mice to bypass the BBB. At 8 days postinjection, young mice injected intracranially with cancer cells still developed larger tumors compared with older mice (Supplementary Fig. S8B).

High levels of estrogen can promote growth of triple-negative breast cancer cells in brain by inducing reactive glial fibrillary acidic protein (GFAP)-expressing astrocytes in the neuroinflammatory response to secrete metastasis-promoting factors (17). Serum samples from young and older mice with 4T1-BR and 231-BR brain metastases had comparable corticosterone and testosterone levels; no consistent differences in estradiol and progesterone concentrations were observed with age, possibly reflecting the fact that, unlike humans, mice do not undergo a true menopause (Supplementary Fig. S8C and S8D; ref. 18). Consistent with this finding, the astrocytic neuroinflammatory response of mice harboring 4T1-BR or 231-BR metastases did not differ with age (Supplementary Fig. S9A and S9B). In other experiments, tumor cell Ki67 and cleaved caspase 3 measurements of proliferation and apoptosis, respectively, were comparable between age groups (Supplementary Fig. S9CS9F). These data suggest that initiation of metastatic colonization may vary with age, but once cancer cells have adapted to grow in either the young or older brain, they are equally able to thrive.

Brains with metastases have elevated numbers of lymphoid and myeloid cells

Aging is accompanied by prominent alterations to the immune system (19). We next examined whether immune cells contribute to this age effect on brain metastasis. In preliminary experiments, young mice injected with either 4T1-BR or E0771-BR5 brain colonizing cells were compared with sham injections (Supplementary Figs. S10S13). The normal brain had few intravasated immune cells in agreement with the literature (20). Multiple alterations were observed in BALB/C brains with 4T1-BR metastases including significant increases in CD4+ and CD8+ T cells, Ly6G+ neutrophils, and Ly6C+F4/80 monocytes. There was also an increase in regulatory T cells (Treg). No changes in numbers of Ly6C+F4/80+ macrophages and resident CNS myeloid cell numbers were observed as a result of metastasis. Similar trends were also observed in a second immunocompetent model, E0771-BR5, with the addition of a significant increase in macrophages and CNS resident myeloid cells. Immune cell invasion trends into the metastatic brain differed quantitatively from blood.

Young mice have more CNS myeloid cells compared with older mice

Aging in the CNS is accompanied by alterations to the brain immune landscape (21) and thus a changed immune contexture stood as a candidate mechanism to affect brain metastatic aggressiveness. We examined the effect of age on brain immune profile, both naïve and metastatic, in the 4T1-BR and 231-BR models (Fig. 3). CD4+ and CD8+ T cells did not significantly differ in metastatic mouse brains with age. CD3+ T cells that were FOXP3+ (Tregs) were scarce in metastatic lesions, with 17/27 and 21/26 4T1-BR metastatic clusters negative in young and older mice, respectively. For the Treg+ lesions, the number of Tregs was unchanged with age when normalized for lesion size (P = 0.2863; Supplementary Fig. S14).

Figure 3.

Figure 3.

Effect of age and metastatic status on brain immune composition. A and B, Number of brain immune cells in young (5–6 months) and older (18–20 months) BALB/C mice with (A) or without (B) 4T1-BR brain metastases. Results shown in A are combined data from two independent experiments, terminated at day 13 and 14 after injection with cancer cells and in B, are one independent experiment. A and B, Immune subsets were defined as follows: CD4+ T cells (CD3+CD4+), CD8+ T cells (CD3+CD8α+), neutrophils (CD11b+Ly6G+), monocytes (CD11b+Ly6C+Ly6GF4/80), macrophages(CD11b+Ly6C+Ly6GF4/80+), and resident myeloid/microglia (CD45LoCD11b+CD39+CD3Ly6CLy6G). C and D, Number of brain immune cells in young (2–3 months) and older (15 months) athymic nude mice with (C) or without (D) 231-BRbrain metastases. C,Mice were euthanized at day28 after intracardial injection with cancer cells. C and D, Immune subsets were defined as follows: neutrophils (CD11b+Ly6G+), monocytes (CD11b+Ly6C+Ly6GF4/80), macrophages (CD11b+Ly6C+Ly6GF4/80+), and resident myeloid/microglia (CD45.2+CD11b+Thy1.2Ly6CLy6G). E, Number of Tmem119+ myeloid cells (CD45+CD11b+Tmem119+) in young (2 months) and older (15–17 months) athymic nude mice. Each data point is one mouse. One independent experiment (*, P < 0.05; **, P < 0.01, Mann–Whitney test).

Microglia, the resident myeloid immune cells of the brain, constituted the majority of brain immune cells in all groups. Young brains with 4T1-BR metastases had 1.5-fold more brain resident CNS myeloid cells compared with older brains with metastases (Fig. 3A). Microglia were accompanied by infiltrating blood-borne monocytes and macrophages, also higher in young brains.

To determine whether these age-related differences were inherent or induced by the brain metastases, brains from naïve young and older BALB/C mice were compared. Naïve brains from young BALB/C mice also contained 2-fold more CNS resident microglia compared with that in older BALB/C mice (Fig. 3B), indicating an inherent difference in myeloid numbers with age. No significant increase in infiltrating macrophage or monocyte populations were observed. Analysis of the 231-BR model system confirmed significantly increased microglia in young naïve and metastatic brains, with inconsistent trends in infiltrating myeloid cells (Fig. 3C and D). Increased microglia were also observed in naïve young nude mice when standard FACS gating or microglial-specific Tmem119 was used (Fig. 3D and E). The majority of immune cells in blood did not significantly differ with age in either model system (Supplementary Fig. S15). The data identify a prominent age-associated change in the resident CNS immune repertoire that could underlie differences in metastatic propensity.

CNS myeloid cells promote brain metastasis in young mice

Although the data nominated resident CNS myeloid cells as the most plausible age-associated determinants of brain metastatic ability, it remained possible that more subtle differences in other immune components actively recruited to the metastatic brain could also contribute. To eliminate this possibility, peripherally derived CD4+ T cells, CD8+ T cells, and GR1+ neutrophils and monocytes were depleted from young BALB/C mice by extended treatment with depleting antibodies. Depletion of these immune subsets at experimental termination was confirmed by flow cytometry (Supplementary Fig. S16). Loss of CD4+ T cells (Fig. 4A), CD8+ T cells (Fig. 4B), or GR1+ neutrophils and monocytes (Fig. 4C) did not significantly affect 4T1-BR metastasis burden in the young brain.

Figure 4.

Figure 4.

Depletion of immune subsets reveals a contributory role for CNS myeloid cells in brain metastasis. A–C, Frequency of immune subsets in terminal blood draws and number of 4T1-BR brain metastatic clusters quantified from BALB/C mice (2 months) injected with anti-CD4 (A), anti-CD8α (B), or anti-GR1 (C) antibodies or isotype IgG controls. Each data point is one mouse (n = 6–15 per experimental group). One independent experiment per depletion strategy (*, P < 0.05; **, P < 0.01, Mann–Whitney test). D–H, BALB/C mice (2 months) were given control diet (Ctrl, n = 11) or diet supplemented with 300 mg/kg PLX3397, a CSF-1R inhibitor (PLX, n = 11) for 28 days prior to injection of 4T1-BR cancer cells (day 0) and continuously thereafter until study termination at day 13. D, Schematic representation of the experiment and average number of metastatic clusters per brain tissue section. Each data point is one mouse. One independent experiment (***, P < 0.001, Mann–Whitney test). E, Representative immunofluorescence images of uninvolved cortical brain tissue and brain metastases in control and PLX3397-treated mice. Images of uninvolved tissue and brain metastases are from the same animal. Scale bar, 50 μm. Inset scale bar, 10 μm. F, Quantification of percent of region-of-interest (ROI) covered by Iba1. G, Staining intensity [arbitrary units (AU)] for Iba1, CD68, and P2y12 within the Iba1+ regions. H, Quantification of percent of ROI covered by P2y12. I, Staining intensity for Iba1 and CD68 within the P2y12+ regions. F–I, The Wilcoxon matched-pairs test was used to compare differences in percent coverage and staining intensity between uninvolved (Uninv.) brain regions and brain metastases (Met.) within each treatment group (*, P < 0.05; **, P < 0.01, significant difference from uninvolved brain). The Mann–Whitney test was used to compare control versus PLX3397 within each tissue compartment (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001, n.s., not significant). ROI for uninvolved brain is the whole field whereas ROI for metastases is area covered by brain metastases.

Resident microglia depend on colony-stimulating factor 1 receptor (CSF-1R) signaling for survival (22, 23). RNAscope analysis detected mRNA for CSF-1 or IL34, two ligands for CSF-1R, in some CNS myeloid cells, other cells of the brain microenvironment, and 4T1-BR cancer cells (Supplementary Figs. S17 and S18). To study whether disruption ofCSF-1R signaling, and thus survival of CNS myeloid cells, affected metastasis formation, BALB/C mice were fed chow containing the CSF-1R inhibitor PLX3397 to deplete myeloid cells. CNS myeloid cell depletion resulted in a highly significant 2.1-fold (P = 0.0001) reduction in brain metastatic burden compared with mice given control diet (Fig. 4D).

Additional markers were investigated to determine the effects of PLX3397 on brain myeloid subpopulations, in both the uninvolved brain away from metastases and the neuro-inflammatory response surrounding them (Fig. 4EI). Iba1 is a dual microglia/macrophage marker that increases in staining intensity on cellular activation (24, 25). Iba1+ cells were elevated in the metastatic neuroinflammatory response as compared with the uninvolved brain (Fig. 4F). PLX3397 reduced the number of Iba1+ cells in the uninvolved brain and metastatic microenvironment by 33% and 31% over control, respectively, with no effect on Iba1 intensity, the latter indicative of activation (Fig. 4G). These Iba1+ metastasis-associated myeloid cells also expressed comparable intensities of the CD68 phagocytic activation marker, with or without PLX3397. The P2y12 marker binds a purinergic receptor that is specific for microglia and loses staining intensity upon chemotactic activation (26, 27). P2y12+ microglia were present in higher numbers in the metastatic neuroinflammatory response than in the uninvolved brain and were depleted in both compartments by PLX3397 (Fig. 4H). Both Iba1 and CD68 intensity in P2y12+ microglia, indicative of activation, were elevated in the metastatic microenvironment but unaffected by PLX3397 (Fig. 4I) confirming a depletion of microglia without altering activation status. Infiltrating border associated macrophages, identified by CD206 staining (28), were primarily present in the leptomeninges and covered a median of 1.4% of brain metastatic capillaries (Supplementary Fig. S19AS19C). To the extent that they were present, CD206+ cells were reduced by PLX3397 (Supplementary Fig. S19B and S19C). In summary, treatment with the CSF-1R inhibitor PLX3397 demonstrated that resident CNS myeloid cells functionally support or promote brain metastasis in young mice, and that the drug works by reducing resident CNS myeloid cell numbers without affecting their activation status.

The CSF-1R inhibitor PLX3397 implicates CNS myeloid cells in metastasis promotion in young and older mice

Microglia undergo significant alterations with age (29). Two hypotheses could explain the contribution of CNS myeloid cells to age-related changes in brain metastasis: either young cells promoted, or older cells inhibited brain metastasis. Young and older BALB/C mice were given PLX3397 to deplete resident CNS myeloid cells (Fig. 5A). As previously, immunostaining for Iba1 (macrophage or microglia), P2y12 (microglia), or CD206 (border associated macrophage) markers showed substantial (>60%) loss of CNS myeloid cells following treatment with PLX3397 (Fig. 5B and C; Supplementary Fig. S19DS19F). Young mice treated with PLX3397 showed a strong trend towards reduced numbers of brain metastatic burden (Fig. 5D), consistent with our earlier results (Fig. 4D). Few brain metastases were observed in older mice, but there was also a trend towards reduction in metastatic burden. Using the median number of metastatic clusters in a combined dataset of untreated BALB/C mice to establish an age-specific cut-off for high vs. low metastatic tumor burden status (Supplementary Fig. S20), 30.8% of young animals had a high brain metastatic burden in the vehicle arm. This percentage was reduced to zero in the PLX3397 arm (Fig. 5E; P = 0.035). No statistically significant effect was observed in older animals (P = 0.25). The data demonstrate that CNS myeloid cells promote brain metastasis at all ages, but that this effect is more pronounced in young animals.

Figure 5.

Figure 5.

Effect of resident CNS myeloid cell depletion on brain metastasis development in young and older mice. Young (4.5 months) and older (18 months) BALB/C mice were given daily oral gavages of PLX3397 (PLX; 100 mg/kg) or vehicle control (Ctrl) and injected with 4T1-BR cancer cells. Mice were euthanized at day 12 after injection of cancer cells. Young-Ctrl (n = 13), Young-PLX (n = 15), Older-Ctrl (n = 7), Older-PLX (n = 10), one independent experiment. A, Schematic representation of the experiment. B and C, Percent reduction in Iba1+ myeloid (B) and P2y12+ microglia (C) in PLX3397-treated mice relative to control mice within the same tissue compartment (***, P < 0.001, Mann–Whitney test). Too few aged mice developed brain metastases therefore no statistical test could be performed. D, Average number of metastatic clusters per brain tissue section. Each data point is one mouse. The Mann–Whitney test was used to test statistical difference between treatment arms within one age group. E, Frequency of mice within each age group defined as having high and low brain metastatic tumor burden using age-specific cut-offs (*, P < 0.05, Fischer exact test). F, Percent of ROI in uninvolved brain regions covered with GFAP+ astrocytes (*, P < 0.05; **, P < 0.0001 Kruskal–Wallis test, followed by Dunn post hoc pairwise comparison). ROI for uninvolved brain is the whole field. G, Cellular viability of 4T1-BR cells treated with increasing concentrations of recombinant mouse SEMA3A Fc chimera protein were assessed at 48 hours (mean ± SEM, n = 8–16 wells). Representative of two independent experiments (****, P < 0.0001 compared with untreated control, one-way ANOVA, Dunnett post hoc comparison). H, Results from Boyden chamber motility assay used to test the migration of 4T1-BR cells towards recombinant SEMA3A Fc chimera protein (100 μg/mL), control IgG Fc, and diluent PBS/BSA (0.1%). Pooled results from three independent experiments (n = 6–22 wells). I, Representative immunofluorescent image of an Iba1þ myeloid cell expressing SEMA3A. J, Representative immunofluorescent images of SEMA3A in 4T1-BR brain metastases of young (n = 6) and older (n = 5) mice. Scale bar = 50 μm. K, Quantitation of SEMA3A expression in 4T1-BR brain metastases. Each data point is a metastatic cluster. Statistical test, Mann–Whitney test.

CNS myeloid cells promote distinct age-related alterations in the uninvolved brain and metastatic microenvironment

The myeloid contribution to brain metastasis is likely multifactorial. Younger mouse brains contain greater numbers of resident myeloid/microglial cells which could contribute to the age-dependent difference in metastatic burden; myeloid cells could also vary in function with age. In addition, distinct age-related differences might be seen in the normal, uninvolved brain versus the metastatic neuroinflammatory response.

Known functions of CNS resident myeloid cells were investigated in both the uninvolved brain and the metastasis-associated neuroinflammatory response. Traditional markers of activation and resting state did not vary with CSF-1R inhibition in either location (Supplementary Fig. S21). Microglia are known to interact with and activate CSF-1R-negative astrocytes to either promote or inhibit a variety of brain pathologies (30, 31). Astrocytic activation has also been reported with normal brain aging (32), as was observed in our mouse cohorts (Fig. 5F). PLX3397 depletion increased astrocyte activation (GFAP positivity) in the uninvolved brain of young mice to levels observed in older mice (Fig. 5F; Supplementary Fig. S22A). PLX3397 did not affect the astrocytic neuroinflammatory response in either age group (Supplementary Fig. S22B), suggesting that any age-dependent differences in astrogliosis could only affect metastasis at the very earliest steps of colonization before development of a neuroinflammatory response.

We then asked whether CNS myeloid cells augmented metastatic colonization at later stages when a neuroinflammatory response was apparent. Conditioned medium from cultured EOC2 microglial cells was capable of stimulating proliferation of 4T1-BR tumors cells (Supplementary Fig. S23), suggesting that a component of the microglial secretome might promote metastasis. We therefore queried published RNA sequencing (RNA-seq) data on Ribotag-isolated microglia from young (3 months) and older (24 months) mice (29), with particular attention to secreted factors that could mediate myeloid–tumor crosstalk, and found that Semaphorin 3A (Sema3a; Log2FC 0.271, FDR = 0.0241) was significantly upregulated in younger mice. Sema3a is a member of semaphorin family that provides a repellant cue in axonal growth guidance during development. It binds the Plexin-A1/neuropilin1 co-receptors and is produced by microglia and other brain cells (33). Sema3a has been linked to diverse CNS diseasessuch astraumatic brain injury (34) and multiple sclerosis(33). It also contributes to aspects of cancer progression (35, 36). Similar to microglial conditioned medium, recombinant Sema3a stimulated tumor cell proliferation and motility in vitro (Fig. 5G and H). Immunofluorescence confirmed that Sema3a was produced by Iba1+ myeloid cells in vivo (Fig. 5I). Importantly, expression of Sema3a in the neuroinflammatory response of young metastatic brains showed a strong trend toward higher expression than in older brains (Fig. 5J and K). This effect of age was not observed in uninvolved brain (Supplementary Fig. S24). Not all myeloid products showed the same expression trend with age (Supplementary Fig. S25), demonstrating specificity in this pathway. Thus, factors secreted from young CNS myeloid cells in the developing tumor microenvironment change with age and can influence aspects of metastatic colonization.

Discussion

The twin conundrums of why young patients with breast cancer have aggressive disease and how to best treat them have, to date, been addressed by epidemiological studies and limited molecular analyses of primary tumor cohorts. Here we have harnessed the power of mouse model systems to investigate the causal underpinnings of this phenomenon by specifically addressing the effect of host age on metastasis, the most lethal step in cancer progression. Use of mouse models enabled the tumor cell intrinsic properties to be kept constant, with the key variable being the effect of age on the host tissues comprising the tumor microenvironment. In four breast cancer models (three triple-negative and one luminal B subtype), young animals developed a significantly higher brain metastatic burden than their aged counterparts, in agreement with reported higher rates of brain metastases in young patients (1315). In striking contrast, age did not significantly affect breast cancer metastasis burden in lung and liver. Overall, our preclinical data demonstrate that, independent of intrinsic properties of the cancer cells, age-associated changes to the microenvironment prominently affects breast cancer metastasis in brain.

Age is a major risk factor for cancer (37). Essentially all studies to date support the idea that the aged microenvironment is tumor promoting (38). For example, meticulous work has concluded that for melanoma, the fibroblast component of an aged microenvironment promotes metastasis through multiple mechanisms including effects on the immune contexture (39). Our results show that for breast cancer brain metastasis, the reverse is true, and the aged microenvironment is less supportive of metastasis development than the young one

We explored a potential role for immune cells in age-related differences in breast cancer brain metastasis. To our knowledge, this analysis of immune subsets and their functional relevance in the metastatic mouse brain is among the few reported to date for breast cancer (4042). Although multiple immune subsets infiltrate the metastatic brain, depletion of CD4+ T cells, CD8+ T cells, and GR1+ monocytes and neutrophils did not affect metastatic efficiency in young mice. In contrast, depletion of resident CNS myeloid cells, which include microglia and other resident macrophages, strongly suppressed brain metastasis development in young mice, and a trend was seen in older mice. These data eliminated the possibility that the major role of aged resident myeloid cells is to actively protect against metastasis, and instead pointed towards a metastasis-stimulating effect of resident CNS myeloid cells. Importantly, PLX3397 only reduced the proportion of young mice with high numbers of brain metastases. This may be of clinical interest, as a limited number of brain metastases are treatable with local therapy (surgery, stereotactic radiotherapy), but patients with high metastatic burdens have fewer and more toxic options (whole brain radiotherapy).

CNS myeloid cells, primarily microglia, serve multiple functions in the normal brain such as synaptic pruning, regulation of neuronal plasticity, provision of innate immunity, and phagocytosis (reviewed in ref. 43). Myeloid cells are a major immune population activated around brain metastases in patients (44), and limited preclinical evidence exists for their potentiation of this disease in vivo (42, 45, 46). In our mouse models, myeloid cells were found both throughout the normal brain (uninvolved) and in the reactive neuroinflammatory metastatic microenvironment. The mechanism of action of CNS myeloid cells in limiting metastasis in older brains is likely complex (Fig. 6). Older animals contain fewer CNS myeloid cells/microglia in both naïve or metastatic brains, thus fewer metastasis-promoting cells are present. However, our data suggested that CNS myeloid cells/microglia also differ qualitatively with age. Young CNS myeloid cells in the normal uninvolved brain inhibit the activation of astrocytes; PLX3397-treated young brains resembled aged brains in terms of astrogliosis. We hypothesize that these activated astrocytes are inhibitory to the initial steps in metastatic colonization, consistent with the literature (47). This effect of age and CSF-1R inhibition was not seen in the neuroinflammatory response as it is replete with activated microglia and astrocytes. Because the neuroinflammatory response has a dense myeloid and astrocytic cell presence with an activated status, we hypothesized that it may contribute to myeloid age-dependent promotion of metastatic colonization in a manner distinct from astrocyte activation. A literature search identified Sema3a as downregulated in aged microglia (29); in our model systems, Sema3a is a myeloid/microglial product in the brain metastatic neuroinflammatory response that declines in expression with age. In vitro experiments demonstrated that Sema3a can potently stimulate tumor cell proliferation and motility, making it a plausible contributor to the pro-metastatic effect of the young CNS myeloid cells. It is provocative to consider that the efficacy of CSF-1R inhibition in young mice may result from its ability to partially phenocopy an “aged” inflamed environment in the brain, through reduction in myeloid/microglial numbers, alterations in their secretome, and/or induction of astrocyte activation. Our data suggest that both prevalent microglia and less abundant infiltrating macrophages may participate.

Figure 6.

Figure 6.

Schema of brain metastasis in young and older mice. A, The young normal brain microenvironment features capillaries enclosed by the blood–brain barrier, neurons, astrocytes, myeloid cells (primarily microglia), and other cells. B, With age, the normal brain has fewer myeloid cells/microglia, less Semaphorin 3a (Sema3a) production, and more activated astrocytes, defined by GFAP expression. C, The young metastatic brain contains a relatively high metastatic burden and shows alterations in two areas: in uninvolved (noncancer containing) regions, activated astrocytes are observed and may be inhibitory to initiation of colonization. Prominent alterations are observed in the neuroinflammatory response that forms around a metastatic lesion. These include high numbers of neuroinflammatory microglia and high levels of Sema3a, which may potentiate a developing metastatic lesion. D, With age, the metastatic brain contains fewer metastases and decreased myeloid cells overall. In the uninvolved brain, greater GFAP+ activated astrocytes occur. In the neuroinflammatory response, less Sema3a is produced. Many of these same features (fewer microglia, activated astrocytes) are also observed in young metastatic brains treated with a CSF-1R inhibitor, suggesting that CSF-1R inhibition phenocopies aspects of aging.

Strengths of this study include the generality of the conclusions over multiple models, different parity states, and different immune status. However, this work has several potential limitations. Although we attempted to test most hypotheses using adequate animal numbers from at least two model systems at different ages, the inevitable attrition of the aged mice cohorts limited our ability to do so in all cases and may have contributed to some conclusions being just statistical trends. Other host characteristics may also contribute to increased brain metastasis at young ages, such as response to therapy. Finally, it will be interesting to determine whether the age-related effects of microglia on metastasis described herein apply only to breast cancer or whether they extend to other cancer types that metastasize to the brain.

Our data importantly suggest that CSF-1R inhibitors may hold promise for management of brain metastases of breast cancer, particularly in young patients. Several CSF-1R inhibitors are available and must be compared for brain penetration, off-target effects, adverse effects, and prevention of colonization versus shrinkage of established lesions. To the extent that CSF-1R inhibitors “age” the brain, it will be important to examine neurocognitive effects of such treatment. Our data so far support a preventive effect on metastasis. Brain metastasis primary prevention trials are difficult to conduct, but secondary prevention trials are ongoing, such as metronomic temozolomide in HER2+ disease (NCT03190967). It will be of interest to determine if efficacy of CSF-1R inhibition is linked to patient age in a similarly designed clinical trial.

Supplementary Material

34083229_PatriciaSteeg_SuppleTable
34083229_PatriciaSteeg_SuppleMaterial1
34083229_PatriciaSteeg_SuppleMaterial2

Translational Relevance.

Multivariate analyses in epidemiologic studies indicate that a young age at breast cancer diagnosis, typically ≤40 years old, is associated with an aggressive disease course, including increased brain metastasis. Analyses of primary tumor cohorts has failed to date to identify a specific causal mutation in tumors from young patients. Metastasis assays in young and older mice demonstrated that, in four triple-negative or luminal B model systems, brain metastases were quantitatively higher in young mice. Lung and liver metastases were unaffected by host age. Brains from both naïve and metastatic young animals contained higher numbers of myeloid cells, both microglia and infiltrating macrophages. Inhibition of myeloid cells using a colony stimulating factor-1 receptor (CSF-1R) inhibitor reduced brain metastatic burden; in only young animals CSF-1R inhibition eliminated a high burden of brain metastases. The data support the hypothesis that CSF-1R inhibitors can limit brain metastasis, particularly in younger patients.

Acknowledgments

This research was supported by the NIH Intramural Research Program through an NCI FLEX award to P.S. Steeg and L.M. Wakefield and by the Intramural program of the NCI, the National Institute of Neurological Disorders & Stroke, and the U.S. Department of Defense Breast Cancer Research Program, grant no.: W81 XWH-062-003.3. This project has been funded in whole or in part with Federal funds from the NCI, NIH, under contract no. HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. NCI-Frederick is accredited by AAALAC International and follows the Public Health Service Policy for the Care and Use of Laboratory Animals. Animal care was provided in accordance with the procedures outlined in the “Guide for Care and Use of Laboratory Animals” (National Research Council, 2011; National Academy Press, Washington, D.C.). The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core is supported by the Eunice Kennedy Shriver NICHD/NIH Grant No. R24HD102061.

Footnotes

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

Authors’ Disclosures

No disclosures were reported.

References

  • 1.Anders CK, Hsu DS, Broadwater G, Acharya CR, Foekens JA, Zhang Y, et al. Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol 2008;26:3324–30. [DOI] [PubMed] [Google Scholar]
  • 2.Partridge AH, Hughes ME, Warner ET, Ottesen RA, Wong YN, Edge SB, et al. Subtype-dependent relationship between young age at diagnosis and breast cancer survival. J Clin Oncol 2016;34:3308–14. [DOI] [PubMed] [Google Scholar]
  • 3.van der Hage JA, Mieog JSD, van de Velde CJH, Putter H, Bartelink H, van de Vijver MJ. Impact of established prognostic factors and molecular subtype in very young breast cancer patients: pooled analysis of four EORTC randomized controlled trials. Breast Cancer Res 2011;13:R68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cancello G, Maisonneuve P, Rotmensz N, Viale G, Mastropasqua MG, Pruneri G, et al. Prognosis and adjuvant treatment effects in selected breast cancer subtypes of very young women (<35 years) with operable breast cancer. Ann Oncol 2010;21:1974–81. [DOI] [PubMed] [Google Scholar]
  • 5.Anders CK, Fan C, Parker JS, Carey LA, Blackwell KL, Klauber-DeMore N, et al. Breast carcinomas arising at a young age: unique biology or a surrogate for aggressive intrinsic subtypes? J Clin Oncol 2011;29:E18–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Azim HA, Michiels S, Bedard PL, Singhal SK, Criscitiello C, Ignatiadis M, et al. Elucidating prognosis and biology of breast cancer arising in young women using gene expression profiling. Clin Cancer Res 2012;18:1341–51. [DOI] [PubMed] [Google Scholar]
  • 7.Liedtke C, Hess KR, Karn T, Rody A, Kiesel L, Hortobagyi GN, et al. The prognostic impact of age in patients with triple-negative breast cancer. Breast Cancer Res Treat 2013;138:591–9. [DOI] [PubMed] [Google Scholar]
  • 8.Zimmer AS, Zhu KM, Steeg PS, Wu A, Gatti-Mays ME, Soltani S, et al. Analysis of breast cancer in young women in the Department of Defense (DOD) database. Breast Cancer Res Treat 2018;168:501–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gregorowitsch ML, Ghedri A, Young-Afat DA, Bijlsma R, Baas IO, van Schaik-van de Mheen C, et al. The effect of chemotherapy on subjective cognitive function in younger early-stage breast cancer survivors treated with chemotherapy compared to older patients. Breast Cancer Res Treat 2019;175:429–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Azim HA Jr, Partridge AH. Biology of breast cancer in young women. Breast Cancer Res 2014;16:427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yang Y, Yang HH, Hu Y, Watson PH, Liu H, Geiger TR, et al. Immunocompetent mouse allograft models for development of therapies to target breast cancer metastasis. Oncotarget 2017;8:30621–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Azim HA, Nguyen B, Brohee S, Zoppoli G, Sotiriou C. Genomic aberrations in young and elderly breast cancer patients. BMC Med 2015;13:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Evans AJ, James JJ, Cornford EJ, Chan SY, Burrell HC, Pinder SE, et al. Brain metastases from breast cancer: identification of a high-risk group. Clin Oncol (R Coll Radiol) 2004;16:345–9. [DOI] [PubMed] [Google Scholar]
  • 14.Hung MH, Liu CY, Shiau CY, Hsu CY, Tsai YF, Wang YL, et al. Effect of age and biological subtype on the risk and timing of brain metastasis in breast cancer patients. PLoS One 2014;9:e89389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J Clin Oncol 2004;22:2865–72. [DOI] [PubMed] [Google Scholar]
  • 16.Bowman RL, Klemm F, Akkari L, Pyonteck SM, Sevenich L, Quail DF, et al. Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies. Cell Rep 2016;17:2445–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sartorius CA, Hanna CT, Gril B, Cruz H, Serkova NJ, Huber KM, et al. Estrogen promotes the brain metastatic colonization of triple negative breast cancer cells via an astrocyte-mediated paracrine mechanism. Oncogene 2016;35:2881–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Koebele SV, Bimonte-Nelson HA. Modeling menopause: the utility of rodents in translational behavioral endocrinology research. Maturitas 2016;87:5–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nikolich-Zugich J The twilight of immunity: emerging concepts in aging of the immune system. Nat Immunol 2018;19:10–9. [DOI] [PubMed] [Google Scholar]
  • 20.Mastorakos P, McGavern D. The anatomy and immunology of vasculature in the central nervous system. Sci Immnol 2019;4:eaav0492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 2018;48:380–95. [DOI] [PubMed] [Google Scholar]
  • 22.Elmore MRP, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014;82:380–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Van Hove H, Martens L, Scheyltjens I, De Vlaminck K, Pombo Antunes AR, De Prijck S, et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat Neurosci 2019;22:1021–35. [DOI] [PubMed] [Google Scholar]
  • 24.Ohsawa K, Imai Y, Kanazawa H, Sasaki Y, Kohsaka S. Involvement of Iba1 in membrane ruffling and phagocytosis of macrophages/microglia. J Cell Sci 2000;113:3073–84. [DOI] [PubMed] [Google Scholar]
  • 25.Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. Microglia-specific localisation of a novel calcium binding protein, Iba1. Brain Res Mol Brain Res 1998;57:1–9. [DOI] [PubMed] [Google Scholar]
  • 26.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, et al. The P2Y(12) receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci 2006;9:1512–9. [DOI] [PubMed] [Google Scholar]
  • 27.Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, et al. Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nat Neurosci 2014;17:131–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kierdorf K, Masuda T, Jordao MJC, Prinz M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat Rev Neurosci 2019;20:547–62. [DOI] [PubMed] [Google Scholar]
  • 29.Kang SS, Ebbert MTW, Baker KE, Cook C, Wang X, Sens JP, et al. Microglial translational profiling reveals a convergent APOE pathway from aging, amyloid, and tau. J Exp Med 2018;215:2235–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Liddelow SA, Barres BA. Reactive astrocytes: production, function, and therapeutic potential. Immunity 2017;46:957–67. [DOI] [PubMed] [Google Scholar]
  • 31.Erblich B, Zhu LY, Etgen AM, Dobrenis K, Pollard JW. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS One 2011;6:e26317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Palmer AL, Ousman SS. Astrocytes and aging. Front Aging Neurosci 2018;10:337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Costa C, Martinez-Saez E, Gutierrez-Franco A, Eixarch H, Castro Z, Ortega-Aznar A, et al. Expression of semaphorin 3A, semaphorin 7A and their receptors in multiple sclerosis lesions. Mult Scler J 2015;21:1632–43. [DOI] [PubMed] [Google Scholar]
  • 34.Yang MC, Wang XX, Fan YS, Chen YQ, Sun DD, Xu X, et al. Semaphorin 3A contributes to secondary blood-brain barrier damage after traumatic brain injury. Front Cell Neurosci 2019;13:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tominaga K, Minato H, Murayama T, Sasahara A, Nishimura T, Kiyokawa E, et al. Semaphorin signaling via MICAL3 induces symmetric cell division to expand breast cancer stem-like cells. Proc Natl Acad Sci U S A 2019;116:625–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hu ZQ, Zhou SL, Zhou ZJ, Luo CB, Chen EB, Zhan H, et al. Overexpression of semaphorin 3A promotes tumor progression and predicts poor prognosis in hepatocellular carcinoma after curative resection. Oncotarget 2016;7:51733–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.White MC, Holman DM, Boehm JE, Peipins LA, Grossman M, Henley SJ. Age and cancer risk: a potentially modifiable relationship. Am J Prev Med 2014;46:S7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Aging Campisi J., cellular senescence, and cancer. Annu Rev Physiol 2013;75:685–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kaur A, Ecker BL, Douglass SM, Kugel CH 3rd, Webster MR, Almeida FV, et al. Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility. Cancer Discov 2019;9:64–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rippaus N, Taggart D, Williams J, Andreou T, Wurdak H, Wronski K, et al. Metastatic site-specific polarization of macrophages in intracranial breast cancer metastases. Oncotarget 2016;7:41473–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Andreou T, Rippaus N, Wronski K, Williams J, Taggart D, Cherqui S, et al. Hematopoietic stem cell gene therapy for brain metastases using myeloid cell-specific gene promoters. J Natl Cancer Inst 2020;112:617–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Guldner IH, Wang Q, Yang L, Golomb SM, Zhao Z, Lopez JA, et al. CNS-native myeloid cells drive immune suppression in the brain metastatic niche through Cxcl10. Cell 2020;183:1234–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med 2017;23:1018–27. [DOI] [PubMed] [Google Scholar]
  • 44.Soto MS, Sibson NR. The multifarious role of microglia in brain metastasis. Front Cell Neurosci 2018;12:414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Qiao S, Qian Y, Xu GQ, Luo QM, Zhang ZH. Long-term characterization of activated microglia/macrophages facilitating the development of experimental brain metastasis through intravital microscopic imaging. J Neuroinflammation 2019;16:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Andreou KE, Soto MS, Allen D, Economopoulos V, de Bernardi A, Larkin JR, et al. Anti-inflammatory microglia/macrophages as a potential therapeutic target in brain metastasis. Front Oncol 2017;7:251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Priego N, Valiente M. The potential of astrocytes as immune modulators in brain tumors. Front Immunol 2019;10:1314. [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

34083229_PatriciaSteeg_SuppleTable
34083229_PatriciaSteeg_SuppleMaterial1
34083229_PatriciaSteeg_SuppleMaterial2

RESOURCES