Brain metastases are more common than primary CNS tumors and confer grave prognosis on patients, as existing treatments have very limited efficacy. The tumor microenvironment has a central role in facilitating tumorigenesis and metastasis. In recent years, there has been much progress in our understanding of the functional role of the brain metastatic microenvironment. In this review, we discuss the latest advances in brain metastasis research, with special emphasis on the role of the brain microenvironment and neuroinflammation, integrating insights from comparable findings in neuropathologies and primary CNS tumors. In addition, we overview findings on the formation of a hospitable metastatic niche and point out the major gaps in knowledge toward developing new therapeutics that will cotarget the stromal compartment in an effort to improve the treatment and prevention of brain metastases.

Brain metastases are one of the deadliest forms of tumor metastasis. Arising in 10%–30% of adult patients with systemic malignancies (1), brain metastases confer dismal prognosis, with a median survival of less than one year (Fig. 1A; refs. 2, 3). The main cancer types that frequently metastasize to the brain are lung, breast, melanoma, renal, and colorectal cancers (2). Different studies suggest that the incidence of brain metastasis is twice to 10 times higher than primary central nervous system (CNS) malignancies (e.g., glioma; refs. 4, 5). Interestingly, postmortem studies suggest higher incidence of brain metastases compared with clinically diagnosed incidence (6). Moreover, the incidence of brain metastasis appears to be on the rise (7). Possible explanations for this apparent increase include better diagnosis of smaller, asymptomatic brain metastasis by MRI, and improved control of extracranial disease by systemic therapy, enabling the emergence of otherwise not clinically manifested metastasis (1, 6).

Figure 1.

A, Epidemiology of brain metastases. Summary of main epidemiologic findings from various primary tumor types that metastasize to brain (percentage of brain metastasis cases diagnosed; m, months; refs. 1, 2, 171–173). B, The brain microenvironment. Illustration of different cell types in normal brain. The physiologic roles of different brain cells are detailed in Box 1.

Figure 1.

A, Epidemiology of brain metastases. Summary of main epidemiologic findings from various primary tumor types that metastasize to brain (percentage of brain metastasis cases diagnosed; m, months; refs. 1, 2, 171–173). B, The brain microenvironment. Illustration of different cell types in normal brain. The physiologic roles of different brain cells are detailed in Box 1.

Close modal

When discussing brain metastases, it is important to keep in mind that they are not a single clinical entity: major differences in the diagnosis, treatment, and prognosis depend on various parameters, including the primary tumor from which metastases developed, suitability for targeted therapies, number of metastases, stage of extracranial disease, etc. Brain metastasis from different primary tumors can occur early in the clinical course of the disease, at the time of initial diagnosis (synchronous), or sometimes months or years after surgical removal of the primary tumor (metachronous). The currently used diagnosis-specific graded prognostic assessment (DS-GPA) of patients with brain metastases are associated with tumor-specific parameters: The DS-GPA for non–small cell lung carcinoma (NSCLC), breast cancer, and malignant melanoma includes molecular predictive markers, such as EGFR, Her2, and B-Raf, respectively, to identify subgroups with a significantly improved overall survival (OS; refs. 8–10). For example, patients with the best melanoma molecular markers (mol-GPA) score have an estimated OS of 34.1 months compared with 7.1 months in the past (8). In addition to molecular subgroups, the OS of patients with brain metastasis also correlates with the infiltration pattern of macrometastases at the brain parenchyma/metastatic interface: while metastases of renal cell cancer are mainly noninfiltrative and are additionally protected by a highly vascularized collagen capsule, the majority of NSCLC brain metastases infiltrate into the adjacent brain parenchyma with tumor cell cohorts, and malignant melanoma cells favor an angio-cooptive infiltration (11). In general, an infiltrative phenotype is associated with a poor prognostic outcome (11). However, the underlying mechanisms that differentiate patterns of brain metastatic infiltration are poorly understood.

In brain metastasis, earlier studies identified gene signatures in primary breast cancer cells that were associated with brain tropism (12), while a more recent study demonstrated branched evolution that distinguishes the mutation landscape in the primary tumor from its brain metastases (13). Thus, additional coevolution with the brain microenvironment may be required to enable brain colonization by disseminated metastatic cells. There is a growing understanding that the metastatic microenvironment plays a crucial role in enabling brain tropism and colonization of disseminated tumor cells. Herein, we summarize the main findings of recent preclinical studies focused on the biology of the brain metastatic microenvironment, with emphasis on the role of neuroinflammation.

Reciprocal interactions between cancer cells and the microenvironment were shown to contribute to tumor progression and to organ-specific metastasis (14–17). The brain harbors a unique microenvironment: the macroglia cell population, which is composed of 75.6% oligodendrocytes and 17.3% astrocytes and the microglia that represent 6.5% of the nonneuronal cells (18). The brain microenvironment also includes endothelial cells, pericytes, ependymal cells, resident and infiltrating immune cells, and other cell types, which play a crucial role in maintaining physiologic homeostasis by communications with neurons (Box 1; Fig. 1B; ref. 18).

Similarly to other organs of the body, cells in the microenvironment of the brain can sense and respond to danger signals induced by pathogen invasion or tissue damage, resulting in instigation of neuroinflammation (19). This physiologic response in the brain microenvironment is reminiscent to changes that occur in the brain metastatic niche, as will be discussed herein. Notably, preclinical studies of brain metastasis and of the interactions of metastatic tumor cells with the microenvironment mostly rely on murine models. While all models entail certain limitations (Box 2), they nevertheless provide important insights on metastatic extravasation, invasion into the brain parenchyma, growth in the brain, interactions with the microenvironment, and therapeutic responses.

Box 1: Physiologic roles of the different brain cells
  • Neurons are the excitable component of the CNS, their axons conduct electrical current. The development and specialization of neurons during embryogenesis is a very complex and temporally regulated process (20).

  • Oligodendrocytes produce myelin, which is essential for proper electric conduction of neuronal signaling (21, 22).

  • Astrocytes have many functional roles in maintaining brain homeostasis: they regulate potassium and pH levels, modulate synaptic transmission by glutamate and GABA uptake, and control the vascular tone and cerebral blood flow (23). Two main classes of astrocytes include protoplasmic astrocytes residing within the gray matter and fibrous astrocytes located in the white matter (21).

  • Microglia are specialized resident macrophages in the brain, which originate in the yolk sac. Microglia secrete growth factors and inflammatory mediators, scavenge cell debris, and is involved in the CNS homeostasis (24).

  • Endothelial cells and their extracellular matrix form the blood vessels of the brain. Endothelial cells express efflux pumps and establish adjacent tight junctions, both of which are necessary for selective permeability of the blood–brain barrier (BBB; ref. 25).

  • Pericytes are mesenchymal cells, which constitute an integral part of the neovascular unit of the BBB. Pericytes occur throughout the vasculature of the body, but are most dense in brain capillaries. Pericytes regulate cerebral blood flow (26) and produce proteins of the basal lamina (27).

  • Ependymal cells line the ventricles of the brain and are part of the blood–cerebrospinal fluid (CSF) barrier (BCSFB; refs. 28, 29).

  • Choroid plexus is a specialized organ within the brain that produces the CSF and is also part of the BCSFB (28).

  • Post-capillary venules: an important part of the BBB. Composed of at least seven layers: 1, endothelial cells connected by tight junctions; 2, inner basement membrane; 3, media, 4, outer basement membrane; 5, Virchow–Robin space with pericytes, macrophages, and other cell types; 6, a third basement membrane; 7, the glia limitans, consistent of astrocyte end-feet (See Fig. 1B).

  • Blood–leptomeningeal barrier (BLMB): an important immunologic barrier, typically activated during meningitis. Activation switches the cellular and molecular composition of the CSF from an immunosuppressive body fluid to a highly immune active one.

  • Infiltrating immune cells can be recruited to the brain via the BCSFB to the CSF or via the blood capillaries, mainly via post-capillary venules (29, 30). T cells and monocyte-derived macrophages contribute to neuronal functions and are central to neuronal repair (31, 32) and during the brain response to pathogen invasion and inflammation (33).

Box 2: Murine models of brain metastasis to study the brain microenvironment and neuroinflammation
  • Models of spontaneous brain metastasis

    By definition, a spontaneous model of brain metastasis is a model in which cancerous cells from the primary tumor are allowed to detach, invade, circulate, extravasate, and colonize the brain. The main advantage of utilizing such models is that they allow investigation of the different metastatic stages, as well as studying early stages of brain metastasis in a clinically relevant setting. However, most spontaneous models are time consuming and do not always provide high percentages of brain metastasis, making large cohorts of mice necessary.

    • The primary tumors in spontaneous models are either orthotopically inoculated or arise in a genetically engineered mouse model (GEMM) of a certain cancer type. Brain metastasis in GEMMs was reviewed elsewhere (34, 35).

    • Resection of the primary tumor, when feasible, allows better modeling of the clinical settings, and usually entails a period of latency before metastatic relapse, thus enabling a time window for preclinical preventative studies (36).

    • Spontaneous models of brain metastasis can be established in immunocompetent or in immunodeficient mice, depending on the source of the cells to be studied.

Summary of murine models of spontaneous brain metastasis

  • Melanoma:

    • RMS: orthotopic (subdermal), immunocompetent, primary tumor removal. Micrometastases, 50%; macrometastases, 23% (37).

    • YDFR.CB3: orthotopic (subdermal), immunodeficient, micrometastases 75% (38).

    • 131/4-5B2: orthotopic (subdermal), immunodeficient, primary tumor removal. Micrometastases, 66%; macrometastases, 20% (39).

  • Lung

    • A549: orthotopic (intrathoracic), immunodeficient. Macrometastases, 61% (40).

  • Breast

    • 4T1: orthotopic (intraductal), immunocompetent, primary tumor removal. Micrometastasis, 67% (41).

    • Fgf4-MCF7: orthotopic (intraductal), immunodeficient. Macrometastases, 35% (42).

    • MDA-MB-231 and CN34BrM: orthotopic (bilateral intraductal), immunodeficient, NSG mice. Macrometastases: MDA-MB-231, 100%; CN34BrM, 90% (43).

  • Models of experimental brain metastasis

    Experimental metastasis models include the direct inoculation of tumor cells into the circulation and provide a model for hematogenous dissemination of cancer cells to generate brain metastases.

  • Intracardiac injection: inoculation of tumor cells into the left ventricle of the heart.

  • Intracarotid injection: inoculation of tumor cells directly into the cerebral circulation.

    Experimental models are widely used, as they achieve a high penetrance of brain metastasis in relatively small cohorts of mice and are a valuable tool for studying macrometastases, especially in preclinical testing of therapeutic intervention.Disadvantages of these models include the technically challenging nature of injection techniques, and the fact that the high load of injected cells may not accurately mimic the clinical setting in which metastasis occurs.

  • Models of intracranial tumor cell injectionsTumor cell growth in the brain can be achieved by intracranial injection: the direct inoculation of tumor cells from various cancer types directly into the brain tissue.Intracranial models of brain metastasis can be established in immunocompetent or in immunodeficient mice (including patient-derived xenografts; ref. 44), depending on the source of the cells to be studied.Intracranial injections yield rapidly growing brain lesions and are an important tool to assess the growth pattern of tumor cells in brain, the various infiltration types and dissemination into the brain parenchyma, and to test the efficacy of novel therapeutics in a preclinical setting of full blown macrometastasis. However, the injection of cells into the parenchyma may initially cause trauma to the brain, thus invoking astrogliosis and neuroinflammation. Therefore, when employing such models, it is important to design suitable controls.

Neuroinflammation in the CNS is instigated in response to damage signals. Cells in the brain parenchyma can sense and respond to danger signals resulting from pathogen invasion (pathogen-associated molecular patterns, PAMP) or tissue damage (damage-associated molecular pattern, DAMP). In the first line of response, DAMP/PAMP signals rapidly activate the adjacent astrocytes and microglia (45). This locally circumscribed first line of glial response is very potent in the defense from pathogenic intruders or in repairing tissue damage without recruiting other cell types. Thus, astrocytes and microglia are central in regulating the CNS homeostasis and tissue repair. However, if this first line of defense is not effective or the damage/affected area is too large, then full blown neuroinflammation develops. This is characterized by enhanced secretion of cytokines and chemokines, persistent activation of astrocytes and microglia, increased blood vessel permeability, and recruitment of immune cells (46, 47). Knowledge on the instigation and regulation of neuroinflammation mainly originated in studies of neurologic diseases and CNS injuries (19, 48). Dysregulation of these pathways leads to uncontrolled damage response, which contributes to the pathology of neurodegenerative diseases (49), exacerbates vascular injury following stroke (50), drives autoimmune diseases (51), and facilitates primary CNS tumors (52).

Many of the changes described in the brain metastatic microenvironment are reminiscent of these dysregulated tissue damage responses, including permeability of the BBB (53), instigation of astrogliosis and neuroinflammation (54), and recruitment of leukocytes (55), which in turn collectively facilitate metastatic growth of invading cancer cells (25, 56). Therefore, findings from these fields may help us understand the roles of cellular brain components at various phases of brain metastasis. These parallels suggest that the physiologic tissue damage response in the brain is hijacked during colonization by metastatic cells, and neuroinflammation is recently emerging as a promising direction to uncover processes associated with the development of brain metastases (57–59).

Astrocytes in neuroinflammation

Activation of astrocytes in response to tissue damage is termed “astrogliosis.” It is characterized by upregulation of the cytoskeletal glial fibrillary acidic protein (GFAP), upregulation of proinflammatory genes (60), and identified by the formation of a “gliotic scar”: reactive astrocytes with elongated processes that form a physical and functional wall around demarcated brain lesions (61). Astrogliosis has neuroprotective functions that support neuronal recovery during the acute phase, but may entail detrimental inflammatory responses that induce further tissue damage (62). Astrogliosis is also evident in brain metastases: astrocytes surround and infiltrate brain metastases in mice, and are recruited to the brain–metastases interface in human brain metastases (57, 59, 63), and therefore the complex interactions between astrocytes and tumor cells are a topic of intense research.

Proinflammatory signaling in astrocytes was shown to be activated by lung cancer cell–derived factors such as macrophage migration inhibitory factor (MIF), IL8, and plasminogen activator inhibitor-1 (PAI-1) in coculture experiments (64). Once activated, astrocytes secrete TNFα, IL6, and IL1β. Such proinflammatory activation of astrocytes stimulated in vitro proliferation of lung cancer cells (64). While the molecular players may vary, inflammatory activation of astrocytes seems to be a general feature of brain metastasis: melanoma cell–secreted factors induced IL23 expression in astrocytes, leading to enhanced transendothelial migration of melanoma cells in vitro (65). Notably, astrocytes also express IL23 during autoimmune diseases, such as multiple sclerosis (MS; ref. 66). In breast cancer, tumor cell–derived IL1β activated JAG1 signaling in astrocytes, which in turn promoted cancer stem cell (CSC) self-renewal in vitro and experimental brain metastasis in vivo. Brain metastasis-free survival in patients with breast cancer correlated with lower expression of IL1β, suggesting clinical relevance of these findings (67). Similarly, secretion of MMP-1 and COX-2 by brain-metastasizing breast cancer cells induced expression of CCL7 by activated astrocytes, which in turn promoted BBB permeability and experimental brain metastasis in vivo (68). These findings implicate the importance of proinflammatory signaling by astrocytes to promote multiple aspects of brain metastasis. However, the functional role of specific cytokines and chemokines that were identified in coculture studies should be further evaluated in vivo.

Induction of neuroinflammation was shown to precede macrometastases: brains bearing spontaneous melanoma micrometastases already express higher levels of the inflammatory mediators CCL17, CCL2, and CXCL10 (37). Notably, CXCL10 was also shown to be expressed by astrocytes in Alzheimer's disease in association with senile plaques (69), and in response to LPS exposure and stroke (60). These findings implicate stromal cell–derived CXCL10 in neuroinflammation, as well as in the formation of brain metastasis. Similarly, CCL17, shown to be expressed by microglia in Alzheimer's disease (70), is upregulated in brains of mice inoculated with vemurafenib-resistant melanoma cells (71). This may implicate stromal cell–derived CCL17 in supporting chemoresistance to BRAF inhibitors. The parallels of inflammatory mediators in brain pathologies and in brain metastasis suggest that canonical neuroinflammatory pathways are hijacked by tumor cells to promote metastases formation and growth.

The mechanisms by which astrocytes are activated by tumor cells are still largely unresolved. The close proximity between astrocytes and tumor cells, resulting from astrogliosis, implicates reciprocal paracrine signaling as a main communication route (37, 65, 67, 72–75). Assembly of carcinoma–astrocyte gap junctions composed of connexin 43 (Cx43), resulting in proinflammatory activation of astrocytes and enhanced resistance of tumor cells to chemotherapy in vitro and in vivo (76–78). This interaction may represent another aspect of cancer-induced astrogliosis and gliotic scar formation, physically “bridging” between astrocytes and tumor cells. Once recruited and activated by brain-metastasizing cells, astrocytes induce growth-promoting signaling in tumor cells (76, 79).

The reciprocal interactions of tumor cells and astrocytes were also shown to contribute to chemoresistance and tumor cell survival due to upregulation of GASTA5, BCL2L1, and TWIST1 expression in cancer cells in vitro (77). Other studies demonstrated that bidirectional interactions between astrocytes and breast cancer cells support chemoresistance via activation of the endothelin axis (80). Blocking this pathway with an antagonist of the endothelin receptor was beneficial in the treatment of breast and lung cancer experimental brain metastases (81). Interestingly, the initial response of astrocytes to brain invasion by tumor cells was suggested to be antitumorigenic: astrocytes were shown to induce tumor cell apoptosis via plasmin activator (PA) secretion. Secretion of Serpins (PA inhibitors) by tumor cells prevented apoptosis and increased vascular cooption in experimental brain metastasis (73). Thus, while the early response of astrocytes to brain invasion by tumor cells may be antitumorigenic, growth-promoting mechanisms eventually prevail and enable metastatic growth.

Microglia in neuroinflammation

Microglia are important mediators of the brain response to tissue damage (82), and are similarly activated at the tumor–brain interface in brain metastasis (83, 84). IHC analysis of microglia in human brain metastases of NSCLC, breast cancer, and melanoma indicated intense microglia activation with evident peritumoral accumulation and intratumoral infiltration (85). Thus, the localization of microglia is reminiscent of the gliosis response to traumatic brain injury sites, at the interface between normal and damaged brain tissue (86).

Proinflammatory signaling in microglia was shown to be instigated following lipopolysaccharide (LPS) injection, including expression of TNFα, iNOS, and IL6 (87). Moreover, targeting of activated microglia/macrophages was shown to reduce tumor growth in a model of intracranial injection of breast cancer cells (84). Microglia were also shown to facilitate invasion of metastatic breast carcinoma cells in an organotypic ex vivo brain slices model (88). This proinvasive function of microglia could be inhibited by microglia depletion, by the WNT inhibitor Dickkopf-2, by LPS treatment, or by CXCR4 inhibition, indicating the molecular pathways involved (82, 88, 89). Moreover, similar to the role of the PI3Kγ pathway in mediating immune suppression by tumor-associated macrophages (TAM) in primary breast tumors (90), a recent study demonstrated the activation of PI3K in patients with breast cancer brain metastases, and identifies its function as a master regulator of the metastasis-promoting function of microglia (91).

Findings from primary brain malignancies and from brain metastasis indicate that the metastasis-promoting functions of microglia phenotype are induced by tumor cells (92–94). Interestingly, astrocytes were also shown to modulate the recruitment of microglia to brain metastasis in vivo (95), adding another dimension to the complex neuroinflammatory networks that facilitate metastasis. However, while in neurodegenerative pathologies, astrocytes and microglia were demonstrate to interact reciprocally in mediating inflammatory responses that contribute to disease progression (96, 97), surprisingly little is known about the molecular factors that underlie the role of microglia in brain metastasis, and their crosstalk with astrocytes.

Recruited immune cells

Recruited macrophages and T cells are a prominent feature of neuroinflammation and brain metastasis. However, in most studies, there is no clear distinction between resident microglia and recruited macrophages regarding identification and function. A study utilizing multitransgenic mouse models that enable ontology tracing, demonstrated that bone marrow–derived macrophages are recruited into primary brain tumors (glioma) and to experimental brain metastases, where they are reprogrammed to express distinct inflammatory gene signatures (92). These findings are supported by a study that demonstrated in vitro functional differences between microglia and monocyte-derived macrophages in facilitating tumor cell invasion, evident by their response to CSF-1R blockade (89).

Macrophages were shown to infiltrate into experimental breast cancer brain metastases, and to enhance tumor cell invasion and colonization via secretion of Cathepsin S (98). Intracranially implanted breast cancer cells were demonstrated to express high levels of lymphotoxin β, which propelled the polarization of infiltrating macrophages toward a tumor-promoting M2-like phenotype (99), suggesting that metastases-recruited macrophages may function in promoting an immunosuppressive brain microenvironment. Macrophages were also shown to induce breast cancer cell invasion into the brain via CSF-1 secretion (89). Interestingly, targeting macrophages in a model of primary CNS malignancy (glioma) resulted in blockade of tumor progression (93).

T cells were also implicated in brain colonization. In intracranial injections of melanoma, breast, and colon cancer cells into mice, regulatory T cells (Tregs) were shown to infiltrate cancerous lesions and to mediate immunosuppression (100). In human brain metastasis, T cells are found mainly at the interface of the brain parenchyma/brain metastases tissue and in the metastatic stroma. Several studies suggested that CD8+ T cells may play a functional role in the microenvironment of brain metastasis. Moreover, while the density of regulatory T cells is not correlated with prognosis, high density of CD3+ or CD8+ T cells was shown to be correlated with better OS (57, 101). A study of experimental melanoma brain metastasis in mice reported a similar infiltration pattern, where most of the CD3+ T cells were CD4+ rather than CD8+ lymphocytes (85). Interestingly, the infiltration of the effector T cells was suggested to be facilitated by the presence of extracerebral tumor lesions, which activated the endothelial cells of the BBB to enhance leukocyte trafficking into brain metastases (102). Taken together, these findings implicate the adaptive immune system in brain metastasis and suggest that immunotherapy could be a promising therapeutic avenue for the treatment of brain metastasis, as discussed below. However, the recruitment, trafficking, functions, and activation of T cells in the microenvironment of brain metastases are still largely unresolved.

Thus, activation of brain stromal cells and recruited immune cells orchestrates neuroinflammation and facilitates metastatic growth. These observations support cotargeting of both tumor cells and the inflammatory stroma in brain metastases.

Brain-metastasizing cancer cells initially encounter the BBB, a highly specialized vascular structure that selectively controls the blood flow and its contents into the brain (53). The BBB is composed of endothelial cells connected by tight junctions, pericytes and the end-feet of astrocytes, which surround the basement membrane of blood vessels, effectively limiting passive paracellular permeability. In addition to this physical barrier, the BBB contains active molecular transport systems (103), which impair CNS drug delivery. Importantly, the BBB is also central in regulating the recruitment and trafficking of peripheral immune cells into the brain. For example, brain endothelial cells express abluminaly CXCL12, the ligand for CXCR4, to prevent CXCR4-expressing leukocytes from infiltrating the brain parenchyma under normal conditions (104). Similarly, the astrocyte end-feet express the death ligand CD95L, which, under physiologic conditions, leads to T-cell apoptosis (105). Therefore, under normal conditions, the BBB also functions as an immunologic barrier. During active neuroinflammation, T cells and myeloid cells enter the brain mainly via post-capillary venules and their infiltration requires not only transmigrating the endothelial cells, but also overcoming the glia limitans (see Box 1; Fig. 1B). This is enabled during neuroinflammation by activation of brain endothelial cells and downregulation of immunologic barrier molecules on perivascular cells, to permit the entry of peripheral immune cells to the brain parenchyma. However, very little is known about the regulation of immune cell infiltration via the BBB in the context of brain metastases.

Following extravasation in the brain, tumor cells may outgrow, or remain dormant. Findings from mouse models of experimental brain metastasis of breast cancer (106) and melanoma (107) demonstrated that tumor cells were predominately positioned along the abluminal surfaces of microvessels in the perivascular space, suggesting that cooption of blood vessels in the brain and initial growth on the inner basement membrane is a central mechanism during the early growth of brain disseminated cells. Notably, the initial interactions with the brain perivascular niche may also affect dormancy, as suggested by preclinical imaging studies in mice, that detected brain-metastasizing cancer cells (from lung, breast cancer, and melanoma) in a dormant state at the perivascular niche in the brain (108–110). The specific mechanisms of vessel interactions and vascular cooption depend on various molecules, and may be fostered via adhesion with integrin beta-1 (106), and expression of L1CAM by brain-metastasizing cancer cells (73, 107). Clinical evidence from patients with melanoma brain metastasis suggested that initial metastatic growth along blood vessels is operative also in human brain metastasis (106). Interestingly, astrocytes and perivascular macrophages, which are part of the BBB, may be involved in this process: a study using various breast cancer cell lines showed that astrocyte-derived HGF induced c-MET activation in tumor cells, which in turn enhanced adhesion to endothelial cells (111). Taken together, these findings link astrocytes and microglia as part of the BBB, with tumor cell extravasation, dormancy, and immune cell trafficking (Fig. 1B).

The brain–tumor barrier

The brain–tumor barrier (BTB) is the vascular system around and within CNS tumors and brain metastases. Permeability of the BTB is a subject of debate; some studies suggested that impaired BBB and permeable BTB are hallmarks of brain metastasis (53). Conversely, other studies in various mouse models of brain metastasis (including breast cancer and melanoma), reported that the permeability of the BTB is vastly heterogeneous, and is not well correlated with metastatic growth rate (112–115). Interestingly, quantitative fluorescence microscopy in preclinical tumor models of glioma compared with brain metastasis of breast cancer, reported that glioma-associated vasculature is more permeable than the vasculature in brain metastases (116).

Increased BBB permeability was shown to be an early event, associated with the formation of spontaneous brain micrometastases in mice (37). Increased permeability may also be part of the instigated neuroinflammation in the metastatic microenvironment. CCL2 upregulation in cancer cells resulted in increased transendothelial migration of small-cell lung carcinoma (117).

Moreover, endothelial cells are active mediators of proinflammatory signaling that facilitates increased BBB permeability and enhanced invasion capacity of tumor cells: secretion of TNFα by brain endothelial cells induced by tumor cell–secreted substance P, augmented breakdown of the BBB and colonization of breast cancer cells in brain by modifying the localization and distribution of tight junctions on brain endothelial cells (118). A study on breast cancer brain metastasis showed that VEGF secretion from breast cancer cells induced activation of inflammatory STAT3 signaling in endothelial cells. STAT3 inhibition resulted in decreased invasion of breast cancer cells to brain, and suppression of angiogenesis and metastasis (119). These studies suggest that proinflammatory signaling contributes to increased BBB permeability thus enhancing tumor cell invasion into the brain.

In this context, it is important to ask whether modifying the brain vessel permeability has therapeutic potential. While at early stages, enhanced permeability may contribute to metastatic invasion, selectively increasing the BTB permeability in overt brain metastasis was suggested to improve efficacy of therapeutic targeting in a mouse model of brain metastasis (120). In clinical studies, whole-brain radiotherapy (WBRT) for brain metastasis was reported to result in increased BBB permeability in patients with brain metastases from NSCLC (121). However, future studies are required to better elucidate the interplay between neuroinflammation, BTB permeability, and improved therapeutic delivery and efficacy of drugs to treat brain metastases.

Most chemotherapies, targeted therapeutics, and immunotherapy that are routinely used systemically to treat metastatic disease are often less effective in treating brain metastases from solid tumors. The brain microenvironment is an important determinant in the response to systemic therapeutics, and was shown to actively mediate resistance to chemotherapy in multiple in vitro and in vivo mouse studies (77, 80, 122).

Preclinical testing of microenvironment-targeted therapeutics resulted in some promising outcomes. For example, targeting of Cathepsin S, secreted by both tumor cells and macrophages reduced experimental brain metastasis of breast cancer (97) and cotargeting of tumor cells and VEGF receptor-2 inhibited growth of breast cancer xenografts in brain (123). In clinical trials, targeting VEGF-A with bevacizumab was tested in combination with WBRT in patients with brain metastases from various solid tumors, but did not prove to be effective (124), and a first-in-human clinical trial with BLZ945, a drug targeting CSF-1R, is ongoing for metastatic solid tumors (ClinicalTrials.gov). Preclinical studies also showed that cotargeting astrocytes using macitentan, an antagonist of the endothelin receptors, was beneficial in experimental brain metastasis of lung and breast cancers (81, 125). The results from preclinical studies emphasize the potential advantages of therapeutic combinations that include targeting of the brain microenvironment (Table 1). Nevertheless, data on the benefits of microenvironment-targeted therapeutics in brain metastases is very limited, as most clinical trials of targeted therapeutics are focused on molecules expressed by cancer cells (126, 127).

Table 1.

Concise summary of main findings and targetable molecules in neuroinflammation instigated in brain metastasis

Neuroinflammatory pathways or functionNeuroinflammatory pathways
found in brain metastasisSystemTargeted?Referencesor function in CNS pathologyReferences
Proliferation of metastatic cells is mediated by: In vitro No (64) C/EBP-β (148) 
Tumor cell secreted: MIF, IL8, and PAI-1    NFκB (149, 150) 
Astrocyte secreted: IL1β, TNFα, and IL6      
Tumor cell invasion to the brain is regulated by:      
Astrocyte-derived: In vitro     
 IL23  Anti-IL23 Ab (65) Antigen presentation (66) 
 CCL7  shCCL7 (68) MAPK, NFκB (151) 
 MMP2  Anti-MMP2 Ab, (153)   
 MMP9  Anti-MMP9 Ab,    
  ONO-4817a Leukocyte transmigration (152) 
  Marimastata   
  Batimastata    
Tumor-derived: In vivo     
 MMP2  NO (65)   
 COX-2  shCOX2, (68)   
 MMP1  shMMP1, GM6001b    
CSC self-renewal is mediated by:      
JAG1–Notch–HES5 signaling in astrocytes In vitro shHES5, shJAG1 (67) Notch (154) 
IL1β secreted from tumor cells In vivo shIL1β, Compound Ec    
Activated astrocytes express gliosis related gene In vitro, NO (37) STAT3, NFκB (155) 
signature: Cxcl10, Lcn2, Timp1, Serpine1 In vivo     
Astrocytes induce Gsta5, Bcl2l1, Twist1 expression In vitro siRNA of survival genes (77) Gap-junction intercellular (156) 
in tumor cells → chemoresistance    communication  
Astrocytes activate the Endothelin axis in tumor In vitro Carbenoxolone (CBX)d(80, 81)   
cells, providing chemoprotection  BQ123e, BQ788e    
 In vivo Macitentane+Paclitaxel (125) ET-1 is a potent vasoconstrictor (157) 
Gap junctions allow communication between In vitro, CBX, (76) Gap-junction intercellular (156) 
tumor cells and astrocytes In vivo shcGAS  communication  
Initial anti-tumorigenic responses of astrocytes to      
metastasizing cells mediated via:   (73)   
FasL, plasminogen activator from astrocytes In vitro anti-FasL  Tissue damage, Apoptosis (158) 
Serpins from reactive stroma  α2-antiplasmin    
Tumor cells expressing L1CAM In vivo shL1CAM,    
  shSerpins    
Microglia facilitate transport of cancer cells into In vitro WNT-inhibitor (88) Wnt/βcatenin (159) 
the brain  Dickkopf-2, Clodronatef    
      
Microglia defense response is hijacked by In vitro Anti-CSF-1 Ab 5A1, (89) Microglia survival and growth (160) 
carcinoma cells  Anti-CD34 Ab    
Depletion of the anti-inflammatory microglia/ In vivo Clodronatef (84) M1 and M2 phenotypes of (161) 
macrophage cell population attenuate metastatic    microglia/macrophage  
colonization      
Different phenotypes of metastasis-associated In vivo NO (99) Macrophages are involved in (162) 
macrophages in dural vs. parenchymal metastasis    neuroinflammation  
NK cells and CD8+ T cells are required for efficient In vivo PD-1/CTLA-4 combined (102) Immune checkpoints are possible (163) 
anti-PD-1/anti-CTLA-4 intracranial tumor response  blockade  therapeutic targets in AD  
Adhesion and angiogenesis are mediated via:    HGF attenuates autoimmunity in (164) 
Tumor cell secreted IL1b, CXCL1, IL18 In vitro, Pterostilbeneg (111) experimental autoimmune  
Astrocyte-derived HGF In vivo   encephalitis (EAE)  
Endothelial cells expressing CXCR1      
STAT3 activation induces cancer cell-directed VEGF In vitro, WP1066h (119) STAT3 (165) 
secretion → STAT3 activation in endothelial cells In vivo     
→ upregulated VEGFR2 in endothelial cells      
Neuroinflammatory pathways or functionNeuroinflammatory pathways
found in brain metastasisSystemTargeted?Referencesor function in CNS pathologyReferences
Proliferation of metastatic cells is mediated by: In vitro No (64) C/EBP-β (148) 
Tumor cell secreted: MIF, IL8, and PAI-1    NFκB (149, 150) 
Astrocyte secreted: IL1β, TNFα, and IL6      
Tumor cell invasion to the brain is regulated by:      
Astrocyte-derived: In vitro     
 IL23  Anti-IL23 Ab (65) Antigen presentation (66) 
 CCL7  shCCL7 (68) MAPK, NFκB (151) 
 MMP2  Anti-MMP2 Ab, (153)   
 MMP9  Anti-MMP9 Ab,    
  ONO-4817a Leukocyte transmigration (152) 
  Marimastata   
  Batimastata    
Tumor-derived: In vivo     
 MMP2  NO (65)   
 COX-2  shCOX2, (68)   
 MMP1  shMMP1, GM6001b    
CSC self-renewal is mediated by:      
JAG1–Notch–HES5 signaling in astrocytes In vitro shHES5, shJAG1 (67) Notch (154) 
IL1β secreted from tumor cells In vivo shIL1β, Compound Ec    
Activated astrocytes express gliosis related gene In vitro, NO (37) STAT3, NFκB (155) 
signature: Cxcl10, Lcn2, Timp1, Serpine1 In vivo     
Astrocytes induce Gsta5, Bcl2l1, Twist1 expression In vitro siRNA of survival genes (77) Gap-junction intercellular (156) 
in tumor cells → chemoresistance    communication  
Astrocytes activate the Endothelin axis in tumor In vitro Carbenoxolone (CBX)d(80, 81)   
cells, providing chemoprotection  BQ123e, BQ788e    
 In vivo Macitentane+Paclitaxel (125) ET-1 is a potent vasoconstrictor (157) 
Gap junctions allow communication between In vitro, CBX, (76) Gap-junction intercellular (156) 
tumor cells and astrocytes In vivo shcGAS  communication  
Initial anti-tumorigenic responses of astrocytes to      
metastasizing cells mediated via:   (73)   
FasL, plasminogen activator from astrocytes In vitro anti-FasL  Tissue damage, Apoptosis (158) 
Serpins from reactive stroma  α2-antiplasmin    
Tumor cells expressing L1CAM In vivo shL1CAM,    
  shSerpins    
Microglia facilitate transport of cancer cells into In vitro WNT-inhibitor (88) Wnt/βcatenin (159) 
the brain  Dickkopf-2, Clodronatef    
      
Microglia defense response is hijacked by In vitro Anti-CSF-1 Ab 5A1, (89) Microglia survival and growth (160) 
carcinoma cells  Anti-CD34 Ab    
Depletion of the anti-inflammatory microglia/ In vivo Clodronatef (84) M1 and M2 phenotypes of (161) 
macrophage cell population attenuate metastatic    microglia/macrophage  
colonization      
Different phenotypes of metastasis-associated In vivo NO (99) Macrophages are involved in (162) 
macrophages in dural vs. parenchymal metastasis    neuroinflammation  
NK cells and CD8+ T cells are required for efficient In vivo PD-1/CTLA-4 combined (102) Immune checkpoints are possible (163) 
anti-PD-1/anti-CTLA-4 intracranial tumor response  blockade  therapeutic targets in AD  
Adhesion and angiogenesis are mediated via:    HGF attenuates autoimmunity in (164) 
Tumor cell secreted IL1b, CXCL1, IL18 In vitro, Pterostilbeneg (111) experimental autoimmune  
Astrocyte-derived HGF In vivo   encephalitis (EAE)  
Endothelial cells expressing CXCR1      
STAT3 activation induces cancer cell-directed VEGF In vitro, WP1066h (119) STAT3 (165) 
secretion → STAT3 activation in endothelial cells In vivo     
→ upregulated VEGFR2 in endothelial cells      

NOTE: Findings from preclinical studies in which targeting of neuroinflammation showed efficacy in the treatment of brain metastases, as well as targets that may be promising.

aMMP inhibitors.

bBBB-permeable metalloproteinase inhibitor.

cA potent BBB-permeable g-secretase inhibitor.

dGap junction inhibitor.

eAntagonists of ETAR and ETBR.

fBisphosphonate.

gc-MET inhibitor.

hSTAT3 inhibitor.

Immunotherapy for treatment of brain metastasis

The approval of immunotherapeutic strategies and immune checkpoint blockade dramatically changed the landscape of treatment strategies for several cancer types, in particular melanoma, NSCLC, and renal cell carcinoma (128), which have a high rate of brain metastasis. Brain metastases originating in tumors with a high mutagenic load (such as melanoma and lung cancer) are likely to be better candidates for immune checkpoint therapy. Indeed, immune checkpoint inhibition with mAbs targeting the CTLA4 (Ipilimumab, anti-CTLA-4) and antibodies that target the programmed cell death protein 1 (nivolumab, pembrolizumab, anti-PD-1) have revolutionized the treatment of metastatic melanoma (129).

Findings of T-cell composition and infiltration in patients with brain metastasis suggest that these considerations may be relevant also to brain metastatic relapse: while some patients have very little TILs in brain, high density of CD3+ or CD8+ T cells correlated with better OS (101, 130). Notably, expression of the PD-1 ligand (PD-L1) was demonstrated in human specimens of brain metastases from melanoma (131) and breast cancer (130), and correlated with higher density of tumor-infiltrating lymphocytes expressing PD-1, suggesting that upregulation of immune checkpoints may be important for the ability of brain metastases to evade the immune system and enhance immunosuppression.

In this context, it is also important to consider the role of myeloid derived suppressor cells (MDSC), immature myeloid cells with suppressive activity on T cells via immune checkpoint signaling. While not much is known about MDSCs in brain metastasis, they were implicated in immune suppression in neuroinflammatory diseases (132) and their infiltration to human glioblastoma tumors was associated with reduced TILs in brain lesions (133). Moreover, blockade of MDSCs was suggested to be beneficial in combination with immune checkpoint treatment in mouse models of glioblastoma (134) and in prevention of brain metastasis (135), suggesting that MDSC blockade should also be considered in immunotherapy of human brain metastasis.

Patients with brain metastasis were previously excluded from clinical trials with immune checkpoint inhibitors, limiting the available knowledge on the efficacy of immunotherapeutics for brain metastasis. However, several recent clinical studies performed in patients with melanoma or in NSCLC with active brain metastases that were treated in monotherapy or in combination of both checkpoint inhibitors, demonstrated benefit on OS of double checkpoint blockade (136–141), and multiple other studies are ongoing (142). Notably, the beneficial effects of anti-CTLA-4 and anti-PD1 on brain metastasis may be mediated, in part, by sustained systemic effects on activated T cells that infiltrate the brain and engage in antitumor responses. This is supported by a recent preclinical study demonstrating that efficient immune checkpoint inhibition for the treatment of intracranial melanoma depended on systemic activation of CD8+ T cells and enhancement of their recruitment to the brain (102). Additional clinical findings, as well as preclinical mechanistic studies, are required to investigate the efficacy, and provide the rationale for immune checkpoint blockade in the treatment of brain metastasis.

Radiotherapy, a standard therapy approach for CNS tumors and brain metastasis, was shown to induce immunostimulatory effects that may be therapeutically beneficial, including downregulation of immunosuppressive cytokines, enhanced immune cell recruitment, and increased CTL efficacy (143). These effects on activation of the immune response result, at least partially, from radiation-induced tissue damage and necrosis. However, radiotherapy was also demonstrated to enhance the expression of PD-L1 in tumor cells and in antigen-presenting cells in a mouse model of breast cancer, suggesting that it may also have immunosuppressive effects (144). Importantly, recent preclinical and clinical studies, which assessed the combination of radiotherapy with immunotherapy, found potential synergism between these two treatment modalities (143, 145). While these results still need to be validated in larger studies, the data suggest that combining radiotherapy with immune checkpoint blockade may prove to be beneficial in targeting the metastatic microenvironment to combat brain metastases.

The incidence of brain metastasis is rising and survival remains very poor. Therefore, there is an acute need to define mechanisms and test novel therapeutic approaches for brain metastasis. The accumulated knowledge gained in past years from the studies discussed here clearly indicate that reciprocal interactions between tumor cells and brain stromal cells are a driving force of brain metastasis. This is mediated partially by hijacking of physiologic tissue damage response and immune cell trafficking pathways in the brain microenvironment, and subsequent induction of local neuroinflammation around the metastatic lesion (Fig. 2).

Figure 2.

Reciprocal interactions between brain-metastasizing tumor cells and the brain microenvironment facilitate metastatic growth. Main findings of neuroinflammatory networks operative in brain metastasis.

Figure 2.

Reciprocal interactions between brain-metastasizing tumor cells and the brain microenvironment facilitate metastatic growth. Main findings of neuroinflammatory networks operative in brain metastasis.

Close modal

Studying the metastatic niche and mechanisms that sustain dormancy are promising approaches toward uncovering the earliest stages of metastasis. Understanding the early events that precede the formation of brain macrometastases will greatly advance our ability to design more efficient therapeutics. Future studies aimed to promote this characterization should focus on elucidating the tumor-derived and stromal cell–derived factors that govern the instigation of neuroinflammation and vascular changes at the premetastatic niche. To facilitate this research direction, development of novel preclinical models of brain metastasis that will include genetic tools for specific targeting of candidate factors is required (some available experimental and genetic tools are summarized in Tables 1 and 2, respectively). Moreover, because recent studies suggest that the operative mechanisms in micrometastases may be different than the ones operative in full-blown brain macrometastases (146), experimental tools that enable this distinction are also needed.

Table 2.

Genetic tools to study the brain microenvironment and neuroinflammation in murine models

Available strainsEnables the study ofPotential applications
GFAP-Cre/ER (166) Astrocytes Knockdown of astrocyte-specific genes 
GFAP-GFP (167)   
ALDH1L1-GFP (60)  Isolating metastasis-associated astrocytes by FACS 
CX3CR1-CRE (168) Microglia Knockdown of microglia-specific genes 
CX3CR1-CRE/ER (168)   
CX3CR1-GFP (168)  Isolating metastasis-associated microglia by FACS 
PDGFRb-Cre/ER (169) Pericytes Knockdown of pericyte-specific genes 
Cre-LoxP System to Measure EV Uptake (170) Tumor-derived exosomes Studying exosomes in premetastatic niche 
Available strainsEnables the study ofPotential applications
GFAP-Cre/ER (166) Astrocytes Knockdown of astrocyte-specific genes 
GFAP-GFP (167)   
ALDH1L1-GFP (60)  Isolating metastasis-associated astrocytes by FACS 
CX3CR1-CRE (168) Microglia Knockdown of microglia-specific genes 
CX3CR1-CRE/ER (168)   
CX3CR1-GFP (168)  Isolating metastasis-associated microglia by FACS 
PDGFRb-Cre/ER (169) Pericytes Knockdown of pericyte-specific genes 
Cre-LoxP System to Measure EV Uptake (170) Tumor-derived exosomes Studying exosomes in premetastatic niche 

NOTE: Genetic tools are valuable in studying the reciprocal interactions between cancer cells and cells of the microenvironment. Combining these tools to create unique orthotopic spontaneous brain metastasis models of brain metastasis will facilitate the attainment of innovative functional experiments on mechanisms of premetastatic niche, screening of potential drugs, validation of diagnostic techniques, etc.

Clinical targeting of brain metastases formation at early stages will only be feasible if diagnostic tools for patient stratification are available. This may include identification of predictive biomarkers that will reflect changes in the brain microenvironment (e.g., circulating inflammatory proteins, circulating DNA, and tumor-derived extracellular vesicles), or the presence of disseminated tumor cells. Such markers could potentially be identified in liquid biopsies of blood and/or CSF and allow personalized design of treatment approaches, following removal of primary tumors, to monitor progression to systemic disease.

Another major limitation of developing more efficient tools for early, preventive targeting of brain metastasis is the efficiency of imaging modalities (147). Insights from preclinical models of the early stages can be applicable to human disease only if available imaging methods will enable earlier diagnosis, in patients with yet asymptomatic brain metastatic lesions. Further studies are also needed to thoroughly characterize cancer type–specific responses of the brain metastatic microenvironment, as well as unifying mechanisms that are common to the brain response during brain metastasis originating from different tumors, to test their applicability as therapeutic targets.

While knowledge from preclinical studies on the brain metastatic microenvironment is emerging, data from clinical studies on microenvironment targeting in the treatment of brain metastasis therapy is still limited. Hopefully, in the coming years, we will see integration of the preclinical findings described in this review into the design of novel clinical strategies aimed at prevention and better treatment of brain metastasis.

No potential conflicts of interest were disclosed.

The authors would like to thank Malak Amer and Dr. Raquel Blazquez for critical reading of the manuscript. N. Erez is supported by funding from the European Research Council (ERC) under the European Union's Horizon 2020 Research and Innovation Programme (grant agreement no. 637069 MetCAF), The Israel Cancer Research Fund (ICRF), and the Israel Cancer Association (ICA). T. Pukrop and N. Erez are supported by The German Research Foundation (DFG PU 355/4-1). T. Pukrop is supported by the German Research Foundation (DFG FOR2127 PU 355/5-1).

1.
Loeffler
JS
.
Epidemiology, clinical manifestations, and diagnosis of brain metastases
.
Wen
PY
,
Eichler
AF
, ed.
Waltham, MA
:
UpToDate Inc.
http://www.uptodate.com (Accessed on November 20, 2016).
2.
Berghoff
AS
,
Schur
S
,
Fureder
LM
,
Gatterbauer
B
,
Dieckmann
K
,
Widhalm
G
, et al
Descriptive statistical analysis of a real life cohort of 2419 patients with brain metastases of solid cancers
.
ESMO Open
2016
;
1
:
e000024
.
3.
Cagney
DN
,
Martin
AM
,
Catalano
PJ
,
Redig
AJ
,
Lin
NU
,
Lee
EQ
, et al
Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: a population-based study
.
Neuro Oncol
2017
;
19
:
1511
21
.
4.
Ekici
K
,
Temelli
O
,
Dikilitas
M
,
Halil Dursun
I
,
Bozdag Kaplan
N
,
Kekilli
E
. 
Survival and prognostic factors in patients with brain metastasis: single center experience
.
J BUON
2016
;
21
:
958
63
.
5.
Gavrilovic
IT
,
Posner
JB
. 
Brain metastases: epidemiology and pathophysiology
.
J Neurooncol
2005
;
75
:
5
14
.
6.
Nayak
L
,
Lee
EQ
,
Wen
PY
. 
Epidemiology of brain metastases
.
Curr Oncol Rep
2012
;
14
:
48
54
.
7.
Takei
H
,
Rouah
E
,
Ishida
Y
. 
Brain metastasis: clinical characteristics, pathological findings and molecular subtyping for therapeutic implications
.
Brain Tumor Pathol
2016
;
33
:
1
12
.
8.
Sperduto
PW
,
Jiang
W
,
Brown
PD
,
Braunstein
S
,
Sneed
P
,
Wattson
DA
, et al
Estimating survival in melanoma patients with brain metastases: an update of the graded prognostic assessment for melanoma using molecular markers (Melanoma-molGPA)
.
Int J Radiat Oncol Biol Phys
2017
;
99
:
812
16
.
9.
Sperduto
PW
,
Yang
TJ
,
Beal
K
,
Pan
H
,
Brown
PD
,
Bangdiwala
A
, et al
Estimating survival in patients with lung cancer and brain metastases: an update of the graded prognostic assessment for lung cancer using molecular markers (Lung-molGPA)
.
JAMA Oncol
2017
;
3
:
827
31
.
10.
Sperduto
PW
,
Chao
ST
,
Sneed
PK
,
Luo
X
,
Suh
J
,
Roberge
D
, et al
Diagnosis-specific prognostic factors, indexes, and treatment outcomes for patients with newly diagnosed brain metastases: a multi-institutional analysis of 4,259 patients
.
Int J Radiat Oncol Biol Phys
2010
;
77
:
655
61
.
11.
Siam
L
,
Bleckmann
A
,
Chaung
HN
,
Mohr
A
,
Klemm
F
,
Barrantes-Freer
A
, et al
The metastatic infiltration at the metastasis/brain parenchyma-interface is very heterogeneous and has a significant impact on survival in a prospective study
.
Oncotarget
2015
;
6
:
29254
67
.
12.
Bos
PD
,
Zhang
XH
,
Nadal
C
,
Shu
W
,
Gomis
RR
,
Nguyen
DX
, et al
Genes that mediate breast cancer metastasis to the brain
.
Nature
2009
;
459
:
1005
9
.
13.
Brastianos
PK
,
Carter
SL
,
Santagata
S
,
Cahill
DP
,
Taylor-Weiner
A
,
Jones
RT
, et al
Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets
.
Cancer Discov
2015
;
5
:
1164
77
.
14.
Erez
N
,
Coussens
LM
. 
Leukocytes as paracrine regulators of metastasis and determinants of organ-specific colonization
.
Int J Cancer
2011
;
128
:
2536
44
.
15.
Hanahan
D
,
Coussens
LM
. 
Accessories to the crime: functions of cells recruited to the tumor microenvironment
.
Cancer Cell
2012
;
21
:
309
22
.
16.
Joyce
JA
,
Pollard
JW
. 
Microenvironmental regulation of metastasis
.
Nat Rev Cancer
2009
;
9
:
239
52
.
17.
Nguyen
DX
,
Bos
PD
,
Massague
J
. 
Metastasis: from dissemination to organ-specific colonization
.
Nat Rev Cancer
2009
;
9
:
274
84
.
18.
Pelvig
DP
,
Pakkenberg
H
,
Stark
AK
,
Pakkenberg
B
. 
Neocortical glial cell numbers in human brains
.
Neurobiol Aging
2008
;
29
:
1754
62
.
19.
Fitch
MT
,
Silver
J
. 
CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure
.
Exp Neurol
2008
;
209
:
294
301
.
20.
Stiles
J
,
Jernigan
TL
. 
The basics of brain development
.
Neuropsychol Rev
2010
;
20
:
327
48
.
21.
Barres
BA
. 
The mystery and magic of glia: a perspective on their roles in health and disease
.
Neuron
2008
;
60
:
430
40
.
22.
Bradl
M
,
Lassmann
H
. 
Oligodendrocytes: biology and pathology
.
Acta Neuropathol
2010
;
119
:
37
53
.
23.
Kimelberg
HK
,
Nedergaard
M
. 
Functions of astrocytes and their potential as therapeutic targets
.
Neurotherapeutics
2010
;
7
:
338
53
.
24.
Tay
TL
,
Savage
J
,
Hui
CW
,
Bisht
K
,
Tremblay
ME
. 
Microglia across the lifespan: from origin to function in brain development, plasticity and cognition
.
J Physiol
2016
;
595
:
1929
45
.
25.
Steeg
PS
,
Camphausen
KA
,
Smith
QR
. 
Brain metastases as preventive and therapeutic targets
.
Nat Rev Cancer
2011
;
11
:
352
63
.
26.
Hall
CN
,
Reynell
C
,
Gesslein
B
,
Hamilton
NB
,
Mishra
A
,
Sutherland
BA
, et al
Capillary pericytes regulate cerebral blood flow in health and disease
.
Nature
2014
;
508
:
55
60
.
27.
Dore-Duffy
P
. 
Pericytes: pluripotent cells of the blood brain barrier
.
Curr Pharm Des
2008
;
14
:
1581
93
.
28.
Del Bigio
MR
. 
Ependymal cells: biology and pathology
.
Acta Neuropathol
2010
;
119
:
55
73
.
29.
Shechter
R
,
London
A
,
Schwartz
M
. 
Orchestrated leukocyte recruitment to immune-privileged sites: absolute barriers versus educational gates
.
Nat Rev Immunol
2013
;
13
:
206
18
.
30.
Wilson
EH
,
Weninger
W
,
Hunter
CA
. 
Trafficking of immune cells in the central nervous system
.
J Clin Invest
2010
;
120
:
1368
79
.
31.
Raposo
C
,
Graubardt
N
,
Cohen
M
,
Eitan
C
,
London
A
,
Berkutzki
T
, et al
CNS repair requires both effector and regulatory T cells with distinct temporal and spatial profiles
.
J Neurosci
2014
;
34
:
10141
55
.
32.
Schwartz
M
,
Raposo
C
. 
Protective autoimmunity: a unifying model for the immune network involved in CNS Repair
.
Neuroscientist
2014
;
20
:
343
58
.
33.
Pedemonte
E
,
Mancardi
G
,
Giunti
D
,
Corcione
A
,
Benvenuto
F
,
Pistoia
V
, et al
Mechanisms of the adaptive immune response inside the central nervous system during inflammatory and autoimmune diseases
.
Pharmacol Ther
2006
;
111
:
555
66
.
34.
Daphu
I
,
Sundstrom
T
,
Horn
S
,
Huszthy
PC
,
Niclou
SP
,
Sakariassen
PO
, et al
In vivo animal models for studying brain metastasis: value and limitations
.
Clin Exp Metastasis
2013
;
30
:
695
710
.
35.
Damsky
WE
 Jr
,
Bosenberg
M
. 
Mouse melanoma models and cell lines
.
Pigment Cell Melanoma Res
2010
;
23
:
853
9
.
36.
Gengenbacher
N
,
Singhal
M
,
Augustin
HG
. 
Preclinical mouse solid tumour models: status quo, challenges and perspectives
.
Nat Rev Cancer
2017
;
17
:
751
65
.
37.
Schwartz
H
,
Blacher
E
,
Amer
M
,
Livneh
N
,
Abramovitz
L
,
Klein
A
, et al
Incipient melanoma brain metastases instigate astrogliosis and neuroinflammation
.
Cancer Res
2016
;
76
:
4359
71
.
38.
Izraely
S
,
Sagi-Assif
O
,
Klein
A
,
Meshel
T
,
Tsarfaty
G
,
Pasmanik-Chor
M
, et al
The metastatic microenvironment: brain-residing melanoma metastasis and dormant micrometastasis
.
Int J Cancer
2012
;
131
:
1071
82
.
39.
Cruz-Munoz
W
,
Man
S
,
Xu
P
,
Kerbel
RS
. 
Development of a preclinical model of spontaneous human melanoma central nervous system metastasis
.
Cancer Res
2008
;
68
:
4500
5
.
40.
Mathieu
A
,
Remmelink
M
,
D'Haene
N
,
Penant
S
,
Gaussin
JF
,
Van Ginckel
R
, et al
Development of a chemoresistant orthotopic human nonsmall cell lung carcinoma model in nude mice: analyses of tumor heterogenity in relation to the immunohistochemical levels of expression of cyclooxygenase-2, ornithine decarboxylase, lung-related resistance protein, prostaglandin E synthetase, and glutathione-S-transferase-alpha (GST)-alpha, GST-mu, and GST-pi
.
Cancer
2004
;
101
:
1908
18
.
41.
Pulaski
BA
,
Ostrand-Rosenberg
S
. 
Reduction of established spontaneous mammary carcinoma metastases following immunotherapy with major histocompatibility complex class II and B7.1 cell-based tumor vaccines
.
Cancer Res
1998
;
58
:
1486
93
.
42.
Kurebayashi
J
,
McLeskey
SW
,
Johnson
MD
,
Lippman
ME
,
Dickson
RB
,
Kern
FG
. 
Quantitative demonstration of spontaneous metastasis by MCF-7 human breast cancer cells cotransfected with fibroblast growth factor 4 and LacZ
.
Cancer Res
1993
;
53
:
2178
87
.
43.
Puchalapalli
M
,
Zeng
X
,
Mu
L
,
Anderson
A
,
Hix Glickman
L
,
Zhang
M
, et al
NSG mice provide a better spontaneous model of breast cancer metastasis than athymic (nude) mice
.
PLoS One
2016
;
11
:
e0163521
.
44.
Ni
J
,
Ramkissoon
SH
,
Xie
S
,
Goel
S
,
Stover
DG
,
Guo
H
, et al
Combination inhibition of PI3K and mTORC1 yields durable remissions in mice bearing orthotopic patient-derived xenografts of HER2-positive breast cancer brain metastases
.
Nat Med
2016
;
22
:
723
6
.
45.
Hanisch
UK
,
Kettenmann
H
. 
Microglia: active sensor and versatile effector cells in the normal and pathologic brain
.
Nat Neurosci
2007
;
10
:
1387
94
.
46.
O'Callaghan
JP
,
Sriram
K
,
Miller
DB
. 
Defining "neuroinflammation"
.
Ann N Y Acad Sci
2008
;
1139
:
318
30
.
47.
Gyoneva
S
,
Ransohoff
RM
. 
Inflammatory reaction after traumatic brain injury: therapeutic potential of targeting cell-cell communication by chemokines
.
Trends Pharmacol Sci
2015
;
36
:
471
80
.
48.
Bajetto
A
,
Bonavia
R
,
Barbero
S
,
Schettini
G
. 
Characterization of chemokines and their receptors in the central nervous system: physiopathological implications
.
J Neurochem
2002
;
82
:
1311
29
.
49.
Kisler
K
,
Nelson
AR
,
Montagne
A
,
Zlokovic
BV
. 
Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease
.
Nat Rev Neurosci
2017
;
18
:
419
34
.
50.
Guo
S
,
Lo
EH
. 
Dysfunctional cell-cell signaling in the neurovascular unit as a paradigm for central nervous system disease
.
Stroke
2009
;
40
:
S4
7
.
51.
Dendrou
CA
,
Fugger
L
,
Friese
MA
. 
Immunopathology of multiple sclerosis
.
Nat Rev Immunol
2015
;
15
:
545
58
.
52.
Quail
DF
,
Joyce
JA
. 
The microenvironmental landscape of brain tumors
.
Cancer Cell
2017
;
31
:
326
41
.
53.
Wilhelm
I
,
Molnar
J
,
Fazakas
C
,
Hasko
J
,
Krizbai
IA
. 
Role of the blood-brain barrier in the formation of brain metastases
.
Int J Mol Sci
2013
;
14
:
1383
411
.
54.
Placone
AL
,
Quinones-Hinojosa
A
,
Searson
PC
. 
The role of astrocytes in the progression of brain cancer: complicating the picture of the tumor microenvironment
.
Tumour Biol
2016
;
37
:
61
9
.
55.
Soto
MS
,
Serres
S
,
Anthony
DC
,
Sibson
NR
. 
Functional role of endothelial adhesion molecules in the early stages of brain metastasis
.
Neuro-Oncol
2014
;
16
:
540
51
.
56.
Steeg
PS
. 
Targeting metastasis
.
Nat Rev Cancer
2016
;
16
:
201
18
.
57.
Berghoff
AS
,
Lassmann
H
,
Preusser
M
,
Hoftberger
R
. 
Characterization of the inflammatory response to solid cancer metastases in the human brain
.
Clin Exp Metastasis
2013
;
30
:
69
81
.
58.
Berghoff
AS
,
Preusser
M
. 
The inflammatory microenvironment in brain metastases: potential treatment target?
Chin Clin Oncol
2015
;
4
:
21
.
59.
Fidler
IJ
,
Balasubramanian
K
,
Lin
Q
,
Kim
SW
,
Kim
SJ
. 
The brain microenvironment and cancer metastasis
.
Mol Cells
2010
;
30
:
93
8
.
60.
Zamanian
JL
,
Xu
L
,
Foo
LC
,
Nouri
N
,
Zhou
L
,
Giffard
RG
, et al
Genomic analysis of reactive astrogliosis
.
J Neurosci
2012
;
32
:
6391
410
.
61.
Burda
JE
,
Sofroniew
MV
. 
Reactive gliosis and the multicellular response to CNS damage and disease
.
Neuron
2014
;
81
:
229
48
.
62.
Liu
Z
,
Chopp
M
. 
Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke
.
Prog Neurobiol
2016
;
144
:
103
20
.
63.
Lorger
M
. 
Tumor microenvironment in the brain
.
Cancers
2012
;
4
:
218
43
.
64.
Seike
T
,
Fujita
K
,
Yamakawa
Y
,
Kido
MA
,
Takiguchi
S
,
Teramoto
N
, et al
Interaction between lung cancer cells and astrocytes via specific inflammatory cytokines in the microenvironment of brain metastasis
.
Clin Exp Metastasis
2011
;
28
:
13
25
.
65.
Klein
A
,
Schwartz
H
,
Sagi-Assif
O
,
Meshel
T
,
Izraely
S
,
Ben Menachem
S
, et al
Astrocytes facilitate melanoma brain metastasis via secretion of IL-23
.
J Pathol
2015
;
236
:
116
27
.
66.
Constantinescu
CS
,
Tani
M
,
Ransohoff
RM
,
Wysocka
M
,
Hilliard
B
,
Fujioka
T
, et al
Astrocytes as antigen-presenting cells: expression of IL-12/IL-23
.
J Neurochem
2005
;
95
:
331
40
.
67.
Xing
F
,
Kobayashi
A
,
Okuda
H
,
Watabe
M
,
Pai
SK
,
Pandey
PR
, et al
Reactive astrocytes promote the metastatic growth of breast cancer stem-like cells by activating Notch signalling in brain
.
EMBO Mol Med
2013
;
5
:
384
96
.
68.
Wu
K
,
Fukuda
K
,
Xing
F
,
Zhang
Y
,
Sharma
S
,
Liu
Y
, et al
Roles of the cyclooxygenase 2 matrix metalloproteinase 1 pathway in brain metastasis of breast cancer
.
J Biol Chem
2015
;
290
:
9842
54
.
69.
Xia
MQ
,
Bacskai
BJ
,
Knowles
RB
,
Qin
SX
,
Hyman
BT
. 
Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: in vitro ERK1/2 activation and role in Alzheimer's disease
.
J Neuroimmunol
2000
;
108
:
227
35
.
70.
Walker
DG
,
Link
J
,
Lue
LF
,
Dalsing-Hernandez
JE
,
Boyes
BE
. 
Gene expression changes by amyloid beta peptide-stimulated human postmortem brain microglia identify activation of multiple inflammatory processes
.
J Leukoc Biol
2006
;
79
:
596
610
.
71.
Zubrilov
I
,
Sagi-Assif
O
,
Izraely
S
,
Meshel
T
,
Ben-Menahem
S
,
Ginat
R
, et al
Vemurafenib resistance selects for highly malignant brain and lung-metastasizing melanoma cells
.
Cancer Lett
2015
;
361
:
86
96
.
72.
Neman
J
,
Choy
C
,
Kowolik
CM
,
Anderson
A
,
Duenas
VJ
,
Waliany
S
, et al
Co-evolution of breast-to-brain metastasis and neural progenitor cells
.
Clin Exp Metastasis
2013
;
30
:
753
68
.
73.
Valiente
M
,
Obenauf
AC
,
Jin
X
,
Chen
Q
,
Zhang
XH
,
Lee
DJ
, et al
Serpins promote cancer cell survival and vascular co-option in brain metastasis
.
Cell
2014
;
156
:
1002
16
.
74.
Fitzgerald
DP
,
Palmieri
D
,
Hua
E
,
Hargrave
E
,
Herring
JM
,
Qian
Y
, et al
Reactive glia are recruited by highly proliferative brain metastases of breast cancer and promote tumor cell colonization
.
Clin Exp Metastasis
2008
;
25
:
799
810
.
75.
Lorger
M
,
Felding-Habermann
B
. 
Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis
.
Am J Pathol
2010
;
176
:
2958
71
.
76.
Chen
Q
,
Boire
A
,
Jin
X
,
Valiente
M
,
Er
EE
,
Lopez-Soto
A
, et al
Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer
.
Nature
2016
;
533
:
493
8
.
77.
Kim
SJ
,
Kim
JS
,
Park
ES
,
Lee
JS
,
Lin
Q
,
Langley
RR
, et al
Astrocytes upregulate survival genes in tumor cells and induce protection from chemotherapy
.
Neoplasia
2011
;
13
:
286
98
.
78.
Stoletov
K
,
Strnadel
J
,
Zardouzian
E
,
Momiyama
M
,
Park
FD
,
Kelber
JA
, et al
Role of connexins in metastatic breast cancer and melanoma brain colonization
.
J Cell Sci
2013
;
126
:
904
13
.
79.
Jandial
R
,
Choy
C
,
Levy
DM
,
Chen
MY
,
Ansari
KI
. 
Astrocyte-induced Reelin expression drives proliferation of Her2+ breast cancer metastases
.
Clin Exp Metastasis
2017
;
34
:
185
96
.
80.
Kim
SW
,
Choi
HJ
,
Lee
HJ
,
He
J
,
Wu
Q
,
Langley
RR
, et al
Role of the endothelin axis in astrocyte- and endothelial cell-mediated chemoprotection of cancer cells
.
Neuro Oncol
2014
;
16
:
1585
98
.
81.
Lee
HJ
,
Hanibuchi
M
,
Kim
SJ
,
Yu
H
,
Kim
MS
,
He
J
, et al
Treatment of experimental human breast cancer and lung cancer brain metastases in mice by macitentan, a dual antagonist of endothelin receptors, combined with paclitaxel
.
Neuro Oncol
2016
;
18
:
486
96
.
82.
Chuang
HN
,
van Rossum
D
,
Sieger
D
,
Siam
L
,
Klemm
F
,
Bleckmann
A
, et al
Carcinoma cells misuse the host tissue damage response to invade the brain
.
Glia
2013
;
61
:
1331
46
.
83.
He
BP
,
Wang
JJ
,
Zhang
X
,
Wu
Y
,
Wang
M
,
Bay
BH
, et al
Differential reactions of microglia to brain metastasis of lung cancer
.
Mol Med
2006
;
12
:
161
70
.
84.
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
.
85.
Amit
M
,
Laider-Trejo
L
,
Shalom
V
,
Shabtay-Orbach
A
,
Krelin
Y
,
Gil
Z
. 
Characterization of the melanoma brain metastatic niche in mice and humans
.
Cancer Med
2013
;
2
:
155
63
.
86.
Yousuf
MA
,
Tan
C
,
Torres-Altoro
MI
,
Lu
FM
,
Plautz
E
,
Zhang
S
, et al
Involvement of aberrant cyclin-dependent kinase 5/p25 activity in experimental traumatic brain injury
.
J Neurochem
2016
;
138
:
317
27
.
87.
Song
GJ
,
Jung
M
,
Kim
JH
,
Park
H
,
Rahman
MH
,
Zhang
S
, et al
A novel role for protein tyrosine phosphatase 1B as a positive regulator of neuroinflammation
.
J Neuroinflammation
2016
;
13
:
86
.
88.
Pukrop
T
,
Dehghani
F
,
Chuang
HN
,
Lohaus
R
,
Bayanga
K
,
Heermann
S
, et al
Microglia promote colonization of brain tissue by breast cancer cells in a Wnt-dependent way
.
Glia
2010
;
58
:
1477
89
.
89.
Rietkotter
E
,
Bleckmann
A
,
Bayerlova
M
,
Menck
K
,
Chuang
HN
,
Wenske
B
, et al
Anti-CSF-1 treatment is effective to prevent carcinoma invasion induced by monocyte-derived cells but scarcely by microglia
.
Oncotarget
2015
;
6
:
15482
93
.
90.
Kaneda
MM
,
Messer
KS
,
Ralainirina
N
,
Li
H
,
Leem
CJ
,
Gorjestani
S
, et al
PI3Kgamma is a molecular switch that controls immune suppression
.
Nature
2016
;
539
:
437
42
.
91.
Blazquez
R
,
Wlochowitz
D
,
Wolff
A
,
Seitz
S
,
Wachter
A
,
Perera-Bel
J
, et al
PI3K: A master regulator of brain metastasis-promoting macrophages/microglia
.
Glia
2018
;
66
:
2438
55
.
92.
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
.
93.
Pyonteck
SM
,
Akkari
L
,
Schuhmacher
AJ
,
Bowman
RL
,
Sevenich
L
,
Quail
DF
, et al
CSF-1R inhibition alters macrophage polarization and blocks glioma progression
.
Nat Med
2013
;
19
:
1264
72
.
94.
Moshe
A
,
Izraely
S
,
Sagi-Assif
O
,
Prakash
R
,
Telerman
A
,
Meshel
T
, et al
Cystatin C takes part in melanoma-microglia cross-talk: possible implications for brain metastasis
.
Clin Exp Metastasis
2018
;
35
:
369
78
.
95.
Zhang
L
,
Zhang
S
,
Yao
J
,
Lowery
FJ
,
Zhang
Q
,
Huang
WC
, et al
Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth
.
Nature
2015
;
527
:
100
4
.
96.
von Bernhardi
R
,
Ramirez
G
. 
Microglia-astrocyte interaction in Alzheimer's disease: friends or foes for the nervous system?
Biol Res
2001
;
34
:
123
8
.
97.
Kavanagh
E
,
Burguillos
MA
,
Carrillo-Jimenez
A
,
Oliva-Martin
MJ
,
Santiago
M
,
Rodhe
J
, et al
Deletion of caspase-8 in mouse myeloid cells blocks microglia pro-inflammatory activation and confers protection in MPTP neurodegeneration model
.
Aging
2015
;
7
:
673
89
.
98.
Sevenich
L
,
Bowman
RL
,
Mason
SD
,
Quail
DF
,
Rapaport
F
,
Elie
BT
, et al
Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S
.
Nat Cell Biol
2014
;
16
:
876
88
.
99.
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
.
100.
Sugihara
AQ
,
Rolle
CE
,
Lesniak
MS
. 
Regulatory T cells actively infiltrate metastatic brain tumors
.
Int J Oncol
2009
;
34
:
1533
40
.
101.
Berghoff
AS
,
Fuchs
E
,
Ricken
G
,
Mlecnik
B
,
Bindea
G
,
Spanberger
T
, et al
Density of tumor-infiltrating lymphocytes correlates with extent of brain edema and overall survival time in patients with brain metastases
.
Oncoimmunology
2016
;
5
:
e1057388
.
102.
Taggart
D
,
Andreou
T
,
Scott
KJ
,
Williams
J
,
Rippaus
N
,
Brownlie
RJ
, et al
Anti-PD-1/anti-CTLA-4 efficacy in melanoma brain metastases depends on extracranial disease and augmentation of CD8(+) T cell trafficking
.
Proc Natl Acad Sci U S A
2018
;
115
:
E1540
E49
.
103.
Engelhardt
B
,
Sorokin
L
. 
The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction
.
Semin Immunopathol
2009
;
31
:
497
511
.
104.
Shechter
R
,
Miller
O
,
Yovel
G
,
Rosenzweig
N
,
London
A
,
Ruckh
J
, et al
Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus
.
Immunity
2013
;
38
:
555
69
.
105.
Bechmann
I
,
Steiner
B
,
Gimsa
U
,
Mor
G
,
Wolf
S
,
Beyer
M
, et al
Astrocyte-induced T cell elimination is CD95 ligand dependent
.
J Neuroimmunol
2002
;
132
:
60
5
.
106.
Carbonell
WS
,
Ansorge
O
,
Sibson
N
,
Muschel
R
. 
The vascular basement membrane as "soil" in brain metastasis
.
PLoS One
2009
;
4
:
e5857
.
107.
Bentolila
LA
,
Prakash
R
,
Mihic-Probst
D
,
Wadehra
M
,
Kleinman
HK
,
Carmichael
TS
, et al
Imaging of angiotropism/vascular co-option in a murine model of brain melanoma: implications for melanoma progression along extravascular pathways
.
Sci Rep
2016
;
6
:
23834
.
108.
Kienast
Y
,
von Baumgarten
L
,
Fuhrmann
M
,
Klinkert
WE
,
Goldbrunner
R
,
Herms
J
, et al
Real-time imaging reveals the single steps of brain metastasis formation
.
Nat Med
2010
;
16
:
116
22
.
109.
Murrell
DH
,
Foster
PJ
,
Chambers
AF
. 
Brain metastases from breast cancer: lessons from experimental magnetic resonance imaging studies and clinical implications
.
J Mol Med
2014
;
92
:
5
12
.
110.
Winkler
F
. 
Hostile takeover: how tumours hijack existing vascular environments to thrive
.
J Pathol
2017
;
242
:
267
72
.
111.
Xing
F
,
Liu
Y
,
Sharma
S
,
Wu
K
,
Chan
MD
,
Lo
HW
, et al
Activation of the c-Met pathway mobilizes an inflammatory network in the brain microenvironment to promote brain metastasis of breast cancer
.
Cancer Res
2016
;
76
:
4970
80
.
112.
Lockman
PR
,
Mittapalli
RK
,
Taskar
KS
,
Rudraraju
V
,
Gril
B
,
Bohn
KA
, et al
Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer
.
Clin Cancer Res
2010
;
16
:
5664
78
.
113.
Murrell
DH
,
Hamilton
AM
,
Mallett
CL
,
van Gorkum
R
,
Chambers
AF
,
Foster
PJ
. 
Understanding heterogeneity and permeability of brain metastases in murine models of HER2-positive breast cancer through magnetic resonance imaging: implications for detection and therapy
.
Transl Oncol
2015
;
8
:
176
84
.
114.
Osswald
M
,
Blaes
J
,
Liao
Y
,
Solecki
G
,
Gommel
M
,
Berghoff
AS
, et al
Impact of blood-brain barrier integrity on tumor growth and therapy response in brain metastases
.
Clin Cancer Res
2016
;
22
:
6078
87
.
115.
Lyle
LT
,
Lockman
PR
,
Adkins
CE
,
Mohammad
AS
,
Sechrest
E
,
Hua
E
, et al
Alterations in pericyte subpopulations are associated with elevated blood-tumor barrier permeability in experimental brain metastasis of breast cancer
.
Clin Cancer Res
2016
;
22
:
5287
99
.
116.
Mittapalli
RK
,
Adkins
CE
,
Bohn
KA
,
Mohammad
AS
,
Lockman
JA
,
Lockman
PR
. 
Quantitative fluorescence microscopy measures vascular pore size in primary and metastatic brain tumors
.
Cancer Res
2017
;
77
:
238
46
.
117.
Liu
T
,
Miao
Z
,
Jiang
J
,
Yuan
S
,
Fang
W
,
Li
B
, et al
Visfatin mediates SCLC cells migration across brain endothelial cells through upregulation of CCL2
.
Int J Mol Sci
2015
;
16
:
11439
51
.
118.
Rodriguez
PL
,
Jiang
S
,
Fu
Y
,
Avraham
S
,
Avraham
HK
. 
The proinflammatory peptide substance P promotes blood-brain barrier breaching by breast cancer cells through changes in microvascular endothelial cell tight junctions
.
Int J Cancer
2014
;
134
:
1034
44
.
119.
Lee
HT
,
Xue
J
,
Chou
PC
,
Zhou
A
,
Yang
P
,
Conrad
CA
, et al
Stat3 orchestrates interaction between endothelial and tumor cells and inhibition of Stat3 suppresses brain metastasis of breast cancer cells
.
Oncotarget
2015
;
6
:
10016
29
.
120.
Connell
JJ
,
Chatain
G
,
Cornelissen
B
,
Vallis
KA
,
Hamilton
A
,
Seymour
L
, et al
Selective permeabilization of the blood-brain barrier at sites of metastasis
.
J Natl Cancer Inst
2013
;
105
:
1634
43
.
121.
Zeng
YD
,
Liao
H
,
Qin
T
,
Zhang
L
,
Wei
WD
,
Liang
JZ
, et al
Blood-brain barrier permeability of gefitinib in patients with brain metastases from non-small-cell lung cancer before and during whole brain radiation therapy
.
Oncotarget
2015
;
6
:
8366
76
.
122.
Lin
Q
,
Balasubramanian
K
,
Fan
D
,
Kim
SJ
,
Guo
L
,
Wang
H
, et al
Reactive astrocytes protect melanoma cells from chemotherapy by sequestering intracellular calcium through gap junction communication channels
.
Neoplasia
2010
;
12
:
748
54
.
123.
Kodack
DP
,
Chung
E
,
Yamashita
H
,
Incio
J
,
Duyverman
AM
,
Song
Y
, et al
Combined targeting of HER2 and VEGFR2 for effective treatment of HER2-amplified breast cancer brain metastases
.
Proc Natl Acad Sci USA
2012
;
109
:
E3119
27
.
124.
Levy
C
,
Allouache
D
,
Lacroix
J
,
Dugue
AE
,
Supiot
S
,
Campone
M
, et al
REBECA: a phase I study of bevacizumab and whole-brain radiation therapy for the treatment of brain metastasis from solid tumours
.
Ann Oncol
2014
;
25
:
2351
6
.
125.
Kim
SJ
,
Lee
HJ
,
Kim
MS
,
Choi
HJ
,
He
J
,
Wu
Q
, et al
Macitentan, a dual endothelin receptor antagonist, in combination with temozolomide leads to glioblastoma regression and long-term survival in mice
.
Clin Cancer Res
2015
;
21
:
4630
41
.
126.
Owonikoko
TK
,
Arbiser
J
,
Zelnak
A
,
Shu
HK
,
Shim
H
,
Robin
AM
, et al
Current approaches to the treatment of metastatic brain tumours
.
Nat Rev Clin Oncol
2014
;
11
:
203
22
.
127.
Kodack
DP
,
Askoxylakis
V
,
Ferraro
GB
,
Fukumura
D
,
Jain
RK
. 
Emerging strategies for treating brain metastases from breast cancer
.
Cancer Cell
2015
;
27
:
163
75
.
128.
Sharma
P
,
Allison
JP
. 
The future of immune checkpoint therapy
.
Science
2015
;
348
:
56
61
.
129.
Redman
JM
,
Gibney
GT
,
Atkins
MB
. 
Advances in immunotherapy for melanoma
.
BMC Med
2016
;
14
:
20
.
130.
Duchnowska
R
,
Peksa
R
,
Radecka
B
,
Mandat
T
,
Trojanowski
T
,
Jarosz
B
, et al
Immune response in breast cancer brain metastases and their microenvironment: the role of the PD-1/PD-L axis
.
Breast Cancer Res
2016
;
18
:
43
.
131.
Berghoff
AS
,
Ricken
G
,
Widhalm
G
,
Rajky
O
,
Dieckmann
K
,
Birner
P
, et al
Tumour-infiltrating lymphocytes and expression of programmed death ligand 1 (PD-L1) in melanoma brain metastases
.
Histopathology
2015
;
66
:
289
99
.
132.
Melero-Jerez
C
,
Ortega
MC
,
Moline-Velazquez
V
,
Clemente
D
. 
Myeloid derived suppressor cells in inflammatory conditions of the central nervous system
.
Biochim Biophys Acta
2016
;
1862
:
368
80
.
133.
Raychaudhuri
B
,
Rayman
P
,
Huang
P
,
Grabowski
M
,
Hambardzumyan
D
,
Finke
JH
, et al
Myeloid derived suppressor cell infiltration of murine and human gliomas is associated with reduction of tumor infiltrating lymphocytes
.
J Neurooncol
2015
;
122
:
293
301
.
134.
Kamran
N
,
Candolfi
M
,
Baker
GJ
,
Ayala
MM
,
Dzaman
M
,
Lowenstein
PR
, et al
Gene therapy for the treatment of neurological disorders: central nervous system neoplasms
.
Methods Mol Biol
2016
;
1382
:
467
82
.
135.
Liu
Y
,
Kosaka
A
,
Ikeura
M
,
Kohanbash
G
,
Fellows-Mayle
W
,
Snyder
LA
, et al
Premetastatic soil and prevention of breast cancer brain metastasis
.
Neuro Oncol
2013
;
15
:
891
903
.
136.
Margolin
K
,
Ernstoff
MS
,
Hamid
O
,
Lawrence
D
,
McDermott
D
,
Puzanov
I
, et al
Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial
.
Lancet Oncol
2012
;
13
:
459
65
.
137.
Ajithkumar
T
,
Parkinson
C
,
Fife
K
,
Corrie
P
,
Jefferies
S
. 
Evolving treatment options for melanoma brain metastases
.
Lancet Oncol
2015
;
16
:
e486
97
.
138.
Goldberg
SB
,
Gettinger
SN
,
Mahajan
A
,
Chiang
AC
,
Herbst
RS
,
Sznol
M
, et al
Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
976
83
.
139.
Long
GV
,
Atkinson
V
,
Lo
S
,
Sandhu
S
,
Guminski
AD
,
Brown
MP
, et al
Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study
.
Lancet Oncol
2018
;
19
:
672
81
.
140.
Queirolo
P
,
Spagnolo
F
,
Ascierto
PA
,
Simeone
E
,
Marchetti
P
,
Scoppola
A
, et al
Efficacy and safety of ipilimumab in patients with advanced melanoma and brain metastases
.
J Neurooncol
2014
;
118
:
109
16
.
141.
Tawbi
HA
,
Forsyth
PA
,
Algazi
A
,
Hamid
O
,
Hodi
FS
,
Moschos
SJ
, et al
Combined nivolumab and ipilimumab in melanoma metastatic to the brain
.
N Engl J Med
2018
;
379
:
722
30
.
142.
Tan
AC
,
Heimberger
AB
,
Menzies
AM
,
Pavlakis
N
,
Khasraw
M
. 
Immune checkpoint inhibitors for brain metastases
.
Curr Oncol Rep
2017
;
19
:
38
.
143.
D'Souza
NM
,
Fang
P
,
Logan
J
,
Yang
J
,
Jiang
W
,
Li
J
. 
Combining radiation therapy with immune checkpoint blockade for central nervous system malignancies
.
Front Oncol
2016
;
6
:
212
.
144.
Deng
L
,
Liang
H
,
Burnette
B
,
Beckett
M
,
Darga
T
,
Weichselbaum
RR
, et al
Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice
.
J Clin Invest
2014
;
124
:
687
95
.
145.
Twyman-Saint\sVictor
C
,
Rech
AJ
,
Maity
A
,
Rengan
R
,
Pauken
KE
,
Stelekati
E
, et al
Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer
.
Nature
2015
;
520
:
373
7
.
146.
Oskarsson
T
,
Batlle
E
,
Massague
J
. 
Metastatic stem cells: sources, niches, and vital pathways
.
Cell Stem Cell
2014
;
14
:
306
21
.
147.
Zakaria
R
,
Das
K
,
Bhojak
M
,
Radon
M
,
Walker
C
,
Jenkinson
MD
. 
The role of magnetic resonance imaging in the management of brain metastases: diagnosis to prognosis
.
Cancer Imaging
2014
;
14
:
8
.
148.
Cox
GM
,
Kithcart
AP
,
Pitt
D
,
Guan
Z
,
Alexander
J
,
Williams
JL
, et al
Macrophage migration inhibitory factor potentiates autoimmune-mediated neuroinflammation
.
J Immunol
2013
;
191
:
1043
54
.
149.
Shabab
T
,
Khanabdali
R
,
Moghadamtousi
SZ
,
Kadir
HA
,
Mohan
G
. 
Neuroinflammation pathways: a general review
.
Int J Neurosci
2017
;
127
:
624
33
.
150.
Mehra
A
,
Ali
C
,
Parcq
J
,
Vivien
D
,
Docagne
F
. 
The plasminogen activation system in neuroinflammation
.
Biochim Biophys Acta
2016
;
1862
:
395
402
.
151.
Thompson
WL
,
Van Eldik
LJ
. 
Inflammatory cytokines stimulate the chemokines CCL2/MCP-1 and CCL7/MCP-3 through NFkB and MAPK dependent pathways in rat astrocytes [corrected]
.
Brain research
2009
;
1287
:
47
57
.
152.
Konnecke
H
,
Bechmann
I
. 
The role of microglia and matrix metalloproteinases involvement in neuroinflammation and gliomas
.
Clin Dev Immunol
2013
;
2013
:
914104
.
153.
Wang
L
,
Cossette
SM
,
Rarick
KR
,
Gershan
J
,
Dwinell
MB
,
Harder
DR
, et al
Astrocytes directly influence tumor cell invasion and metastasis in vivo
.
PLoS One
2013
;
8
:
e80933
.
154.
Cai
Z
,
Zhao
B
,
Deng
Y
,
Shangguan
S
,
Zhou
F
,
Zhou
W
, et al
Notch signaling in cerebrovascular diseases (review)
.
Mol Med Rep
2016
;
14
:
2883
98
.
155.
Lee
S
,
Kim
JH
,
Kim
JH
,
Seo
JW
,
Han
HS
,
Lee
WH
, et al
Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration
.
J Biol Chem
2011
;
286
:
43855
70
.
156.
Kielian
T
. 
Glial connexins and gap junctions in CNS inflammation and disease
.
J Neurochem
2008
;
106
:
1000
16
.
157.
D'Haeseleer
M
,
Beelen
R
,
Fierens
Y
,
Cambron
M
,
Vanbinst
AM
,
Verborgh
C
, et al
Cerebral hypoperfusion in multiple sclerosis is reversible and mediated by endothelin-1
.
Proc Nat Acad Sci USA
2013
;
110
:
5654
8
.
158.
Glass
CK
,
Saijo
K
,
Winner
B
,
Marchetto
MC
,
Gage
FH
. 
Mechanisms underlying inflammation in neurodegeneration
.
Cell
2010
;
140
:
918
34
.
159.
Vallee
A
,
Vallee
JN
,
Guillevin
R
,
Lecarpentier
Y
. 
Interactions Between the canonical WNT/beta-catenin pathway and PPAR gamma on neuroinflammation, demyelination, and remyelination in multiple sclerosis
.
Cell Mol Neurobiol
2018
;
38
:
783
95
.
160.
Walker
DG
,
Tang
TM
,
Lue
LF
. 
Studies on colony stimulating factor receptor-1 and ligands colony stimulating factor-1 and interleukin-34 in Alzheimer's disease brains and human microglia
.
Front Aging Neurosci
2017
;
9
:
244
.
161.
Cherry
JD
,
Olschowka
JA
,
O'Banion
MK
. 
Neuroinflammation and M2 microglia: the good, the bad, and the inflamed
.
J Neuroinflam
2014
;
11
:
98
.
162.
Mammana
S
,
Fagone
P
,
Cavalli
E
,
Basile
MS
,
Petralia
MC
,
Nicoletti
F
, et al
The role of macrophages in neuroinflammatory and neurodegenerative pathways of Alzheimer's disease, amyotrophic lateral sclerosis, and multiple sclerosis: pathogenetic cellular effectors and potential therapeutic targets
.
Int J Mol Sci
2018
;
19
:
pii:E831
.
163.
Baruch
K
,
Deczkowska
A
,
Rosenzweig
N
,
Tsitsou-Kampeli
A
,
Sharif
AM
,
Matcovitch-Natan
O
, et al
PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer's disease
.
Nat Med
2016
;
22
:
135
7
.
164.
Benkhoucha
M
,
Santiago-Raber
ML
,
Schneiter
G
,
Chofflon
M
,
Funakoshi
H
,
Nakamura
T
, et al
Hepatocyte growth factor inhibits CNS autoimmunity by inducing tolerogenic dendritic cells and CD25+Foxp3+ regulatory T cells
.
Proc Nat Acad Sci USA
2010
;
107
:
6424
9
.
165.
Chen
E
,
Xu
D
,
Lan
X
,
Jia
B
,
Sun
L
,
Zheng
JC
, et al
A novel role of the STAT3 pathway in brain inflammation-induced human neural progenitor cell differentiation
.
Curr Mol Med
2013
;
13
:
1474
84
.
166.
Garcia
AD
,
Doan
NB
,
Imura
T
,
Bush
TG
,
Sofroniew
MV
. 
GFAP-expressing progenitors are the principal source of constitutive neurogenesis in adult mouse forebrain
.
Nat Neurosci
2004
;
7
:
1233
41
.
167.
Kuzmanovic
M
,
Dudley
VJ
,
Sarthy
VP
. 
GFAP promoter drives Muller cell-specific expression in transgenic mice
.
Invest Ophthalmol Visual Sci
2003
;
44
:
3606
13
.
168.
Yona
S
,
Kim
KW
,
Wolf
Y
,
Mildner
A
,
Varol
D
,
Breker
M
, et al
Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis
.
Immunity
2013
;
38
:
79
91
.
169.
Gerl
K
,
Miquerol
L
,
Todorov
VT
,
Hugo
CP
,
Adams
RH
,
Kurtz
A
, et al
Inducible glomerular erythropoietin production in the adult kidney
.
Kidney Int
2015
;
88
:
1345
55
.
170.
Zomer
A
,
Maynard
C
,
Verweij
FJ
,
Kamermans
A
,
Schafer
R
,
Beerling
E
, et al
In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior
.
Cell
2015
;
161
:
1046
57
.
171.
Christensen
TD
,
Spindler
KL
,
Palshof
JA
,
Nielsen
DL
. 
Systematic review: brain metastases from colorectal cancer–Incidence and patient characteristics
.
BMC Cancer
2016
;
16
:
260
.
172.
Goulart
CR
,
Mattei
TA
,
Ramina
R
. 
Cerebral melanoma metastases: a critical review on diagnostic methods and therapeutic options
.
ISRN Surg
2011
;
2011
:
276908
.
173.
Wyler
L
,
Napoli
CU
,
Ingold
B
,
Sulser
T
,
Heikenwalder
M
,
Schraml
P
, et al
Brain metastasis in renal cancer patients: metastatic pattern, tumour-associated macrophages and chemokine/chemoreceptor expression
.
Br J Cancer
2014
;
110
:
686
94
.