- Review
- Open access
- Published:
Breaking boundaries: role of the brain barriers in metastatic process
Fluids and Barriers of the CNS volume 22, Article number: 3 (2025)
Abstract
Brain metastases (BMs) are the most common intracranial tumors in adults and occur 3–10 times more frequently than primary brain tumors. Despite intensive multimodal therapies, including resection, radiotherapy, and chemotherapy, BMs are associated with poor prognosis and remain challenging to treat. BMs predominantly originate from primary lung (20–56%), breast (5–20%), and melanoma (7–16%) tumors, although they can arise from other cancer types less frequently. The metastatic cascade is a multistep process involving local invasion, intravasation into the bloodstream or lymphatic system, extravasation into normal tissue, and colonization of the distal site. After reaching the brain, circulating tumor cells (CTCs) breach the blood–brain barrier (BBB).
The selective permeability of the BBB poses a significant challenge for therapeutic compounds, limiting the treatment efficacy of BMs. Understanding the mechanisms of tumor cell interactions with the BBB is crucial for the development of effective treatments. This review provides an in-depth analysis of the brain barriers, including the BBB, blood-spinal cord barrier, blood-meningeal barrier, blood-arachnoid barrier, and blood-cerebrospinal fluid barrier. It explores the molecular and cellular components of these barriers and their roles in brain metastasis, highlighting the importance of this knowledge for identifying druggable targets to prevent or limit BM formation.
Background
Metastasis into the brain poses a challenging task for tumor cells to overcome specialized brain barriers that carefully regulate the flow of substances, such as nutrients, cells, or drugs, between the bloodstream and brain tissue. The mechanisms developed by tumor cells are quite sophisticated but clearly successful because brain metastases (BM) represent the most prevalent intracranial tumors in adults, appearing up to 3–10 times more often than primary brain tumors. [1]. BMs are associated with poor prognosis and are difficult to treat even after intensive multimodal therapy, including resection, radiotherapy, and chemotherapy [2, 3]. They are indicated by symptoms that include seizures, loss of motor and sensory functions, or cranial neuropathies and are usually identified by imaging. One study showed that BMs mainly originate from lung (43.2%), breast (15.7%), and melanoma (16.4%) primary tumors [4] but may appear less frequently due to other cancer types.
Several molecular mechanisms contribute to brain colonization by metastatic tumor cells. In general, the metastatic cascade is a multistep process involving (a) local invasion into nearby tissue, (b) intravasation, that is, entry into the bloodstream or lymphatic system, (c) vascular travel, (d) extravasation, that is, the exit of circulating tumor cells (CTCs) from the bloodstream into normal tissue, and (e) colonization of the distal site [5]. To migrate, cells usually undergo epithelial-mesenchymal transition (EMT), a complex process in which cancer cells shed their epithelial characteristics, become less differentiated, and have more aggressive and stem cell-like characteristics [6, 7]. EMT occurs during local invasion and intravasation. It is regarded as a key biological process in which polarized epithelial cells lose their interaction with the basement membrane due to myriad biochemical changes. These newly formed mesenchymal cells have the capacity to migrate and invade their surroundings and possess the ability to overcome apoptosis [8]. EMT is characterized by decreased expression of E-cadherin, increased expression of N-cadherin [9], and downregulation of β-catenin, a key player in cell junction formation [10]. Once metastatic cells reach the distal site, a reverse process to EMT, termed mesenchymal-to-epithelial transition (MET), occurs [11]. Regardless of the primary cell type [12], MET constitutes an initial stage in the adaptation of CTCs to their new microenvironment during colonization. During the MET process, the expression of genes that were previously attenuated during EMT is reactivated, leading to observable changes in tumor cell morphology [13]. This transient phase of cellular reprogramming is associated with increased coexpression of epithelial markers, such as E-cadherin, and mesenchymal markers, such as vimentin [12, 14]. In the case of BMs, CTCs must extravasate and cross the blood–brain barrier (BBB) by breaching the tight junctions (TJs) between endothelial cells in brain capillaries. This involves the secretion of proteases and the activation of pathways that disrupt the integrity of the BBB.
The BBB is perhaps the major reason for the high incidence of BMs since many therapeutic compounds cannot penetrate the barrier, lowering treatment efficiency [15]. Since BMs represent a great therapeutic challenge, it is crucial to better understand the mechanisms of the interaction of tumor cells with the BBB to find druggable targets to prevent or at least limit BM formation. Hence, in this review, we provide a thorough analysis of intricate brain barriers and the complex mechanisms involved in the metastasis of tumor cells into the brain to understand the complex interplay of brain barriers and the intricate mechanisms governing cancer metastasis, offering potential insights for therapeutic interventions and further research endeavors in the field.
Brain barriers and their permeability
The central nervous system (CNS), comprising the brain and spinal cord, is protected by specialized interfaces that selectively permit the exchange of nutrients, ions, various other molecules, including drugs and contrast agents for brain imaging, and even whole cells between the brain and blood [16]. This allows for the precise control of CNS homeostasis, which is critical for proper neuronal function. These major brain barriers or interfaces, including their structure and role in the migration of metastatic cells into the CNS are discussed. The BBB and blood-cerebrospinal fluid (B-CSF) barrier play a major role in the migration of metastatic cells to the brain and are discussed in detail (Fig. 1A-B; Fig. 4; Fig. 5 and Fig. 7). Much less information is known about the migration of metastatic cells to the CNS via the blood-arachnoid barrier (BAB) (Fig. 6). Other barriers in the CNS include the blood-spinal cord barrier (BSCB), however, the role of this barrier in the spread of metastases remains unknown. The microenvironment of the brain parenchyma is tightly regulated by the BBB, the most selective physical barrier in the CNS. The unique properties of the BBB are manifested by the molecular components (junctional proteins and transporters) of the endothelial cells (ECs) of blood vessels by which the CNS is vascularized. The movement of ions, molecules, and cells between the blood and brain is also facilitated by interactions between ECs and different vascular, immune, and neuronal cells, known as the neurovascular unit (NVU) [17,18,19,20].
Schematic illustration of key central nervous system barriers: Blood–Brain Barrier (BBB) and Blood-Cerebrospinal Fluid Barrier (B-CSF barrier). A Schematic representation of the BBB, illustrating the interaction between brain endothelial cells, pericytes, and astrocytic end-feet that collectively form the barrier. B Illustration of the CP, a vascular structure in the brain ventricles composed of a stromal core and a single-layer cuboidal epithelium forming the B-cerebrospinal fluid (CSF) barrier
The major difference between peripheral metastasis and brain metastasis lies in the unique challenges that metastatic tumor cells face when attempting to enter and survive in the brain environment. The BBB plays a significant role in this difference by acting as a selective barrier, which interacts with tumor cells and factors from the tumor microenvironment in ways that do not occur in peripheral metastasis. In the following section, we will discuss the molecular components, cellular components, and signaling pathways involved in the interaction between metastatic cells and the BBB, highlighting how these mechanisms contribute to the establishment and progression of brain metastasis.
Molecular components of the BBB
The nonfenestrated structure of blood vessels in the brain formed by junctional proteins between ECs results in extremely limited paracellular movement. It allows only hydrophobic molecules of a relatively small size to cross the BBB. Junctional proteins include tight junction (TJ), adherens junction (AJ), and gap junction (GJ) proteins. The structure and function of junctional proteins have been extensively reviewed [21,22,23,24]. Briefly, TJs situated on the apical membrane of ECs consist of transmembrane proteins (claudin and occludin) that are connected to the cytoskeleton by cytoplasmic proteins (zonula occludens proteins; ZO). In addition, the expression profiles of the transporter proteins of ECs are not identical on the luminal and abluminal surfaces, which results in the formation of a highly polarized environment in ECs. Therefore, it allows distinct transport properties, and delivery of essential nutrients across the BBB. The transporters of ECs and their role in brain homeostasis have been well studied [17, 19, 25,26,27,28,29,30].
Cellular components of the neurovascular unit
The NVU comprises ECs, mural cells (vascular smooth muscle cells and pericytes), immune cells, glial cells (astrocytes), and neurons [31]. Mural cells are in immediate contact with ECs on their abluminal side, thus regulating blood flow. Pericytes are embedded in the basement membrane and have the ability to contract and control diameter [17, 26]. The basement membrane, formed by the extracellular matrix, covers the vascular tube from both the luminal and abluminal sides. The luminal side is covered by ECs and pericytes, whereas the abluminal side is wrapped by astroglial endfeet [17, 18]. Pericytes are also involved in maintaining the stability of microcirculation through the angiogenic molecules and growth factors [32]. Even though pericytes are separated from ECs by the basement membrane, pericytes processes communicate with ECs through the synapse-like peg-socket contacts and thereby affect microcirculation [26]. Other important functions of pericytes in maintaining BBB homeostasis include immune or phagocytic functions stem cell potential, angiogenesis as well as participation in the development of the BBB [31, 33, 34].
Glial cells, mainly astrocytes, play significant and complex roles in BBB permeability via polarized cellular processes (endfeet), which ensheathe either neuronal processes or blood vessels. Therefore, a cellular link is formed between the neuronal circuit and blood vessels, enabling blood flow regulation in response to neuronal activity [17]. In addition, the expression of different proteins, such as aquaporin-4 in astrocytes, is critical for regulating the BBB [18].
Blood-spinal cord barrier
The BSCB, a functional equivalent of the BBB, represents the interface between the spinal cord and the systemic circulation, therefore, serves to protect the spinal cord microenvironment [35,36,37]. Structurally, the barrier is composed of ECs joined by TJs and covered by a continuous basement membrane on the basolateral side. Like the BBB, the BSCB is supported by astrocyte endfeet expressing aquaporin 4, a key transporter that mediates water absorption [35]. ECs restrict the passage of molecules from the bloodstream into the spinal cord. Like the BBB, pericytes are crucial for the maintenance of the BSCB. However, fewer pericytes were observed in the BSCB. The different expression profiles of TJ proteins and transporters of ECs between the BSCB and BBB cause a relatively higher permeability of the BSCB. For example, lower expression of occludin and ZO-1, insulin receptor (INSR), low-density lipoprotein receptor-related protein 1 (LRP1), and glucose transporter 1 (GLUT1) in the BSCB has been reported [36,37,38].
Blood-arachnoid barrier
The meninges, consisting of three layers of connective tissue, structurally protect the CNS by securing the brain to the skull, limiting lateral movement, and reducing the risk of injury to the brain and spinal cord. The outermost layer, the dura mater, is formed by two epithelial layers of dense collagen fibers and contains fenestrated vessels, allowing the transport of substances to the dura mater. Different immune cell types, including macrophages, T and B cells, and neutrophils, are present at this site. The leptomeningeal layer, composed of the arachnoid and pia mater, forms the innermost layer where the CSF fills the subarachnoid space. The arachnoid epithelial cells joined by TJs form the BAB, preventing the passage of cells and molecules into the CNS and the passage of CSF from the subarachnoid space into the dura mater. The inner layer of the arachnoid mater is connected to the pia mater by a layer of leptomeningeal cells linked by gap junctions and serves as a semipermeable membrane for solutes [39]. The absence of pericytes and astrocytic endfeet distinguishes BAB from the BBB. The greater expression of intercellular adhesion molecule 1 (ICAM-1) in the BAB than in the BBB makes the barrier more permissive for immune cells to circulate within meningeal spaces even under physiological conditions and as an entry site for lymphocytes and myeloid cells during inflammation [39, 40].
The subarachnoid space contains approximately 80% of the total CSF volume, and the middle layer of the meninges (arachnoid membrane), also called the blood-arachnoid barrier, covers the CSF in this space. The BAB separating the CSF and circulating blood consists of multilayered epithelium with cells joined by TJs. It is characterized by a continuous basal lamina on its inner surface, facing the innermost collagenous part of the arachnoid [41, 42].
The expression of multiple transporters in BAB suggests its importance in eliminating organic anions and neurotoxin cations from the CNS and regulating the passage of molecules between the CSF and fenestrated blood capillaries in the dura mater. It has been shown that transporter abundance in the BAB differs from those in the BBB, which separates the blood and the interstitium of the brain and B-CSF barrier located in the choroid plexus of the brain ventricles [43]. The expression of protein transporters in BAB varies between species [44, 45]. Some of the protein transporters such as OCT3, MCT4 and OATP1A2 have been found in BAB only in humans. On the other hand, efflux transporter MRP3 was found in BAB of pigs and dogs, not in human [44]. The transporter system in porcine BAB contributes to a greater overall transfer of substances compared to the B-CSF barrier. This is probably due to the larger area of the BAB and, thus, higher absolute expression of the transporters than in the B-CSF barrier [45]. For instance, organic anion transporter (OAT)1 and OAT3 and drug-metabolizing enzymes (COMT, GSTP1, and CES1) have been shown to be abundantly expressed in the BAB, while they have been detected at low levels in the BBB and B-CSF barrier [44].
Takeuchi et al. demonstrated that drug transporter expression profiles differ in different regions of the BAB [43]. However, this was not comparable to that at the BSCB. As such, multidrug resistance 1 (MDR1) expressed in both the BAB and BSCB was shown to be lower in the lumbar region at the BSCB than at the BAB. In contrast, it was greater in the cervical region of the BSCB. Moreover, breast cancer resistance protein (BCRP), a restricting transporter of various drugs into the CNS, is expressed at higher levels in the BSCB than in the BAB, with higher expression in the cervical portion [42, 46]. Furthermore, the weaker integrity of either the BAB or BSCB in the lumbar cord was suggested to be due to the leakage of proteins from the blood. Takeuchi et al. reported that although the expression of claudin-11 at the BAB in the cervical and lumbar regions was comparable, claudin-11 expression was lower in the lumbar region at the BSCB.
Blood-cerebrospinal fluid barrier
The choroid plexus (CP), located at the brain ventricles, plays a significant role in the maintenance of CNS homeostasis. The CP comprises highly vascularized stroma, connective tissue, and epithelial cells. The fenestrated capillaries formed by endothelial cells allow for the free entry of cells and molecules into the stroma. However, tightly joined epithelial cells strictly control the passage of substances into the CSF of the brain to form the B-CSF barrier. On the ventricular side of epithelial cells, epiplexus (Kolmer) cells exhibit phagocytic activity [47]. The epithelial cells of the B-CSF barrier express a complex system of junctional and transporter proteins that are fundamental to barrier permeability and integrity. Compared with the BBB, the B-CSF barrier is known to be “leaky,” which is also manifested by the fact that, on brain imaging scans, they become enhanced after the administration of contrast agents. This is believed to be due to the distinct TJ levels between the two barriers [48].
The components of the B-CSF barrier are innervated by the autonomic nervous system, which involves the superior cervical ganglion and parasympathetic fibers of the glossopharyngeal and vagus nerves. Parasympathetic stimulation decreases Na+-K+ adenosine triphosphate-hydrolyzing enzyme (ATPase) activity, which leads to decreased CSF secretion [47]. The wide range of transporters involved in CP activity leads to semipermeable characteristics of the B-CSF barrier. This is critical for the exchange of metabolites and xenobiotics between the blood and CSF [47].
The epithelial cells of the CP are connected via different types of cell junctions, which regulate the permeability and integrity of the B-CSF barrier. One of the most important cell junctions is the tight junctions (TJs) of the apical epithelial cells of the CP, which are responsible for regulating the paracellular diffusion of water-soluble molecules through this barrier. Moreover, TJs maintain electrical resistance across the epithelial layer of the CP. The main transmembrane molecules are occludin and claudin [1,2,3,4,5,6, 9,10,11,12, 19, 22], while the submembrane molecules are zonula occludens (ZO), i.e., ZO-1, ZO-2 and ZO-3. The basolateral epithelial cells of the CP are also connected via AJs, which are linked to cytoskeletal proteins. Among epithelial cells, intracellular protein channels represented by GJs, formed by two hemichannels composed of six connexin proteins, have been identified. GJs mediate intercellular communication and exchange of metabolites and electrolytes [47].
Brain metastases
The generally accepted ‘seed and soil’ hypothesis describes a cancer cell as a ‘seed’ implanted into a suitable environment, i.e., a ‘soil’ [49]. As we mentioned, metastasis is preceded by the EMT process, whereby cancer cells acquire the ability to migrate and behave more aggressively. Metastasis is affected by the qualities of the ‘seed’ as well as the ‘soil’ [50]. Cancer cells may anchor at a distant site, become dormant, and create metastases after a certain period [51]. The state of dormancy is also influenced by microenvironmental conditions in various organs [50]. The metastatic process further depends on the type of molecules [50] and extracellular vesicles (EVs), mostly exosomes, released by tumor cells [52].
Exosomes, which can be found in blood [53], urine [54], saliva [55], or cerebrospinal fluid [56], mediate cell–cell communication [57] and transport numerous types of proteins, lipids, and nucleic acids [58, 59]. Compared with healthy cells, tumor cells release more exosomes [60]. These are known as tumor-derived exosomes (TDEs) and play essential roles in altering the cancer cell microenvironment [61, 62] and all stages of metastasis [63].
Exosomal shaping of the microenvironment
Exosomes can modify the primary microenvironment, primary soil [64], and distant soil [65]. They participate in the activation of EMT by acting as messengers and sending signals from the tumor to recipient cells, thus causing changes in the properties of the recipient cells and their microenvironment [8] (Fig. 2). TDEs also affect the extracellular matrix (ECM) [66]. It has been proposed that matrix metalloproteinases (MMPs) might be integrated into exosomal cargo [67]. Migration and invasion have been linked to MMP2 in lung cancer-derived exosomes [68]. TDEs cause ECM degradation via MMPs [69], thereby stimulating cell motility and influencing the formation of a pre-metastatic niche [70]. Exosomal micro RNAs (miRs) are responsible for the destruction of the BBB integrity. For instance, miR-105 originating in breast cancer cells downregulates ZO-1 tight junctions [71]. Similarly, exosomal miR-181c causes downregulation of phosphoinositide-dependent protein kinase 1, which subsequently leads to inhibition of actinin phosphorylation, cofilin activation, and changes in actin conformation. As a result, tight connections of the BBB are destroyed [72]. Moreover, metastatic cells use exosomal miRs to promote cancer cell proliferation and immunosuppression. Metastatic cell proliferation is enhanced by exosomal miR-122 which increases glucose availability to cancer cells through the downregulation of PKM2 and GLUT1 in niche cells [73]. Following the transfer of metastatic cells through the BBB into the brain tissue, the survival of these cells is supported by exosomal miRs-induced immunosuppression. An example is the conversion of microglia to anti-inflammatory M2 phenotype by exosomal miR-503 [74].
Tumor microenvironment and exosomes in brain metastasis: Molecular mechanisms. Tumor cells and exosomes originating from primary tumors can disrupt the BBB (blood–brain barrier) via their contents. Exosomes carry microRNAs (such as miR-105 and miR-181c), whose involvement leads to increased permeability of the blood–brain barrier (BBB). LncRNA GS1-600G8.5 lowers the expression of ZO-1, Claudin-5, and N-Cadherin. Lnc-Matrix metallopeptidases (MMP)2–2, as part of the exosomal cargo, also disrupt tight junctions, thereby increasing the permeability of the BBB. This promotes intracranial metastasis of tumor cells. Exosomes interact with microglia in the brain and help establish pre-metastatic niches. Furthermore, exosomes promote the proliferation of the BM (brain metastases), alter the immune microenvironment, regulate the stability of tumor cells, and can be used as diagnostic and prognostic biomarkers
TDE cargo also contributes to inflammatory processes; for example, Annexin A2 in TDEs released from breast cancer tissue is responsible for the activation of several signaling pathways, including the p38 mitogen-activated protein kinase (MAPK), nuclear factor κB (NF-κB), and signal transducer and activator of transcription 3 (STAT3) pathways within the cell. This leads to increased concentrations of interleukin (IL)-6 and tumor necrosis factor α (TNF-α), which results in the creation of a pre-metastatic inflammatory microenvironment at distant sites, mainly the lungs and brain [75]. The characteristics of the secondary microenvironment, i.e., the ‘soil’ in distant organs, are thus critical in the development of cancer metastasis [50].
However, tumor cells at the primary site are responsible for the release of exosomes. A classic example is the differentiation of stromal cells into cancer-associated fibroblasts (CAFs), which are triggered by exosomes produced by various tumor cells, such as those of the stomach, colon, and bladder, among others. Transformation into CAFs may involve normal (healthy) fibroblasts in primary tumors, cells in distant metastatic tissues, or other types of cells, such as adipocytes and endothelial or mesenchymal stem cells [76, 77].
ECM of the brain is quite different from other organs in several important ways. In most non-CNS tissues, ECM proteins like collagens, laminins, and fibronectin are widely distributed throughout the tissue. However, in the brain, these proteins are mainly found in association with the vascular basement membranes. In contrast, the major ECM components of normal brain parenchyma include proteoglycans, glycoproteins, and glycosaminoglycans, particularly heparan sulfate proteoglycans (HSPGs) and hyaluronic acid (HA) [78], which may lead to different ECM functions [79]. Stevens et al. demonstrated that the hyaluronan-mediated motility receptor (HMMR) is a key player in the process of lung cancer metastasis, particularly to the brain [80]. HMMR is an ECM receptor that binds to HA, a glycosaminoglycan that is often overproduced in the tumor microenvironment during inflammation and fibrosis. This connection is particularly relevant in lung adenocarcinoma (LUAD), where HMMR expression has been associated with aggressive tumor behavior and poor prognosis [80].
Organotropism and tumor-derived exosomes
Role of exosomes in pre-metastatic niche formation
The mechanism of organ-specific metastasis is complex and involves a variety of mechanisms, including EVs, which may influence the interaction between tumor cells and distant sites [65]. Hoshino et al. showed that the uptake of exosomes in the brain could create a pre-metastatic microenvironment for tumor metastasis and demonstrated that the integrin expression profiles of circulating plasma exosomes isolated from cancer patients could be used as prognostic factors to predict sites of future metastasis [81].
Organ-specific properties and genetic adaptation
In contrast to the BBB, the fenestrated sinusoidal endothelium allows extravasating cells to penetrate more easily [82]. Certain types of metastatic tumor cells have a greater ability to penetrate the BBB than other types do. These are mainly tumor cells from breast cancer, lung cancer, and melanoma, but also renal and colorectal cancer [79]. During the initial stages of BM, metastatic cells operate in a hypoxic environment with a lack of glucose [83,84,85] and thus must express a certain set of genes [79]. Unlike primary tumor cells, tumor cells in brain metastases may harbor mutations in genes such as ERBB2, BRAF, MYC, and BRCA2 [86]. Moreover, BMs may be supported by exosomal miRs that alter the microenvironment to become more favorable for BM formation [71].
Exosomal contribution to tumor progression
The gene composition of the primary tumor is essential in the context of organotropism. In the case of non-small cell lung cancer (NSCLC), mutations in the EGFR or ALK genes lead to enhanced metastasis to the CNS in approximately 50% of patients [87]. Moreover, the gene composition of a primary tumor influences which areas of the brain are affected [88]. In breast cancer, the expression of the NAD+-dependent deacetylase Sirtuin 1 (SIRT1) is decreased, which subsequently leads to increased secretion of exosomes [89]. These exosomes then contribute to tumor growth, the migration of cancer cells, the degradation of the ECM, and higher vascular permeability and metastasis to distant sites. Exosomes derived from lung cancer cells can activate EMT, which promotes migration, invasion, proliferation [90], and angiogenesis [91], hence supporting metastasis. Moreover, they contribute to immune escape, thereby facilitating the progression of lung cancer [92]. In melanoma, exosomes can influence the invasion and motility of cancer cells [93]. Metastasis is also facilitated by exosome-mediated immunosuppression [94] and vascular leakage [72].
Involvement of the immune system
Neuroinflammation precedes BM formation [95]. Pro-inflammatory molecules originating from astrocytes intensified tumor growth in a model of melanoma-derived BM [96]. Similarly, it seems that tumor growth is promoted by glial cells infiltrating BMs by producing pro-inflammatory molecules [79].
The tumor immune microenvironment (TIME) is involved in the entire process of carcinogenesis [97]. It varies slightly from organ to organ and encompasses different types of immune cells, including granulocytes, lymphocytes, monocytes/macrophages, and dendritic cells [98]. Usually, immune cells cooperate to eliminate tumors. However, this ability can be silenced or even reversed so that immune cells can suddenly support tumor progression [97]. Specific mutations in tumor cells are responsible for the secretion of chemokines that draw immune cells to the tumor [99].
Tumor cells communicate with immune cells and dictate their behavior to a certain extent. For example, macrophages, monocytes, neutrophils, and other myeloid populations are able to subdue the cytotoxic characteristics of lymphocytes [97, 100, 101]. Immune cells, mainly macrophages, may stimulate EMT [102]; however, macrophages also work in the opposite manner by encouraging the reverse MET process at the metastatic site [103]. Moreover, experiments with BALB/c mice have shown that NK cells are important for protection from colonization by metastasis or that the loss of NK cells leads to the progression of metastasis [104]. Perforin is a major contributor to NK cell control during tumor metastasis [104]. Higher cytotoxic activity of NK cells or increased expression of NK cell activation receptors indicates a better prognosis for patients at risk of metastatic disease [105].
Hypoxic conditions in solid tumors significantly affect immune cells. Activation of HIF-1α-dependent transcriptional changes in immune cells results in adjustment of their function [106], including immunosuppression in macrophages [107]. Another consequence of a hypoxic state is increased production of monocyte recruitment factors [108], promotion of glycolysis in cancer cells, and the higher production of lactic acid [109].
Metastasis of lung cancer to brain tissue
Lung cancer is one of the most common causes of cancer-related death worldwide and the leading cause of BM [110,111,112]. The frequency of BM is markedly greater in patients with lung cancer than in those with other common cancers [2]. Small cell lung cancer (SCLC) and NSCLC are two main histological cancer subtypes that frequently spread to the brain [111, 113]. Approximately 10% of all SCLC patients develop BM [114]. In contrast, patients with NSCLC, which can be further subdivided into squamous cell carcinoma, adenocarcinoma, and large cell carcinoma types, have 20, 18, and 11% metastasis rates to the brain, respectively [115, 116]. Typically, lung cancer presents with multiple BMs.
Genes involved in BMs derived from lung cancer
In NSCLC, the most common mutations occur in the TP53 gene encoding the tumor suppressor p53 (in almost 50% of cases), and these alterations are associated with a worse prognosis and may be relatively more resistant to chemotherapy and radiation [117]. TP53 regulates vital cellular processes, including DNA repair, cell cycle control, and apoptosis. The involvement of the TP53 gene in lung cancer pathogenesis is underscored by frequent loss of heterozygosity (LOH) at its locus on chromosome 17p13, particularly in SCLC and squamous cell carcinoma [118]. TP53 mutations, which occur early in tumorigenesis, persist throughout tumor progression and metastasis. These mutations, along with increased TP53 expression, are correlated with poor survival and resistance to therapy in NSCLC [119]. Various studies have highlighted TP53 as a prognostic marker, with aberrant TP53 status indicating aggressive disease and shorter survival, particularly in patients with adenocarcinomas [120]. Additionally, the TP53 status influences the response to adjuvant chemotherapy [117], with greater benefits in TP53 + patients. Although TP53 alterations are associated with poor prognosis, definitive evidence supporting their role in individual patient management remains elusive. Nonetheless, understanding the importance of TP53 in NSCLC underscores its potential as a therapeutic target and prognostic indicator [117].
Additional gene mutations implicated in NSCLC include those of EGFR and KRAS, the mutation rates of which depend on ethnicity, histology, and smoking status [121, 122]. EGFR, also known as ErbB-1 or HER1, is a transmembrane glycoprotein that belongs to the ErbB family of tyrosine kinase (TK) receptors, which also includes the homologous receptors HER2, HER3, and HER4 [123, 124]. EGFR and its family members contribute to several complex signaling cascades, including growth, differentiation, adhesion, migration, and survival of cancer cells [123, 125]. Ligand binding triggers hetero or homodimerization, trans-autophosphorylation, and the subsequent activation of various kinase cascades. These include the RAS-RAF-MEK-MAPK, PI3K-AKT-mTOR, PLC-γ-Ca2 + /PKC, and JAK-STAT3 pathways, which collectively promote the expression of genes associated with the progression of BMs [126].
EGFR overexpression is often observed in many solid tumors, such as breast cancer, head and neck cancer, NSCLC, renal cancer, ovarian cancer, and colon cancer [127]. Several studies have reported EGFR mutations in BMs arising from NSCLC [115, 122, 128, 129]. These studies revealed that patients with BM were more likely to have primary tumors with EGFR mutations; in particular, the prevalence of EGFR mutations in East Asian cohorts is high, found in 44–63% of BM cases in this population [115, 130, 131]. In a study by Eichler et al., 93 patients with NSCLC were clinically selected for EGFR mutation screening. Among these patients, 41 (44%) developed BM, indicating the importance of the EGFR mutation status as a prognostic factor [130]. TK domain mutations in EGFR occur mainly in exons 18–21. The most frequently reported mutations include exon 19 deletions and point mutations in exon 21, such as L858R, L861Q, and S768I. Additionally, a case report detailed the G719X mutation in exon 18 [115, 130, 132, 133].Sun et al. reported that copy number variations in EGFR were also associated with BMs. They reported an increase in EGFR copy numbers not only in primary NSCLC (62%) but also in corresponding brain metastases (64%) [124, 131]. Similarly, other studies revealed a gain of chromosome 7p, which could be related to EGFR amplification and, thus, a greater number of BMs [132].
KRAS has emerged as another extensively explored target in lung cancer-associated BMs. The KRAS oncogene encodes a small GTPase transductor protein bearing the same name [134]. Among human cancers, KRAS is one of the most frequently mutated oncogenes, occurring in 95% of pancreatic, 50% of colorectal, and 32% of lung adenocarcinomas [135]. KRAS, along with two family members, neuroblastoma RAS viral oncogene homolog (NRAS) and Harvey rat sarcoma virus (HRAS), functions as a transducer of signals that activate growth factor receptors [135]. Although KRAS comprises six exons, the majority of mutations are concentrated in codons 12, 13, 61, and 146. In NSCLC, G12C is the most prevalent mutation, accounting for almost half of all cases, followed by G12V and G12D [136]. Most studies have indicated that the diagnosis of BM in patients with KRAS mutations is lower than in patients with EGFR mutations. In a study involving 144 patients, Tomasini et al. compared KRAS and EGFR and reported that BMs associated with EGFR mutations were more common (87%) than those associated with KRAS mutations (55%) [137]. In a different study, Vassella et al. reported that 58% of 111 patients with KRAS mutations had brain metastasis. Additionally, they identified 14 patients with alterations in the EGFR pathway, including KRAS alterations associated with BM. The G12C mutation in KRAS was the most frequently detected mutation, occurring in 26% of the patients. Moreover, the KRAS G12C and G13C variants are notably enriched in patients with BM [123].
HER2, a 185 kDa transmembrane protein encoded by HER2/neu, is closely related to EGFR, although it does not bind to identical ligands. When activated, both hetero and homodimers of HER2 initiate phosphorylation events similar to those of EGFR, activating several signaling pathways implicated in breast cancer progression. These pathways include the STAT3, RAS-MAPK, and PI3K pathways, resulting in the inactivation of proteins that trigger apoptosis and the upregulation of genes that promote cell growth (Fig. 3). HER2 mutations have been identified in lung cancer [138, 139]. Approximately 1–4% of NSCLC cases exhibit HER2 mutations [140], and the expression of a HER2 mutant containing a 12 bp insertion at exon 20 (G776YVMA) was found to be more potent in BM than in wild-type HER2. This mutation is linked to activated signal transducers, EGFR phosphorylation, and the promotion of survival, invasiveness, and tumorigenicity [138, 139]. Recent studies have indicated that the high incidence of BM in patients with primary NSCLC is associated with the G776YVMA insertion [141, 142].
Key signaling pathways involved in breast cancer, lung cancer and melanoma Lung cancer, breast cancer, and melanoma are malignancies that often metastasize to the brain, utilizing specific signaling pathways. In lung cancer, the RAS-RAF-MEK-MAPK pathway (marked by the blue arrow) drives protein activity changes and changes in gene expression, while the inhibition of apoptosis and promotion of cell survival depend on the Akt pathway (marked by the orange arrow). Additionally, activation of PLC-γ results in the synthesis of second messengers, such as inositol 1,4,5-triphosphate (IP3) and 1,2-diacylglycerol (DAG), which are crucial for intracellular Ca2 + release (marked by the green arrow). In breast cancer, HER2-positive subtypes often involve ligand binding to HER3, leading to dimerization with HER2 and initiating phosphorylation events similar to those of EGFR, which drive tumor cell proliferation and brain metastasis. In melanoma, activation of EGFR causes phosphorylation of STAT3 (marked by the pink arrow), followed by its dimerization and transport to the nucleus, promoting gene expression that facilitates brain metastasis. Together, these signaling pathways enhance tumor cell proliferation, survival, and invasive potential, ultimately leading to brain metastasis. c-Myc: cellular Myelocytomatosis; COX2: Cyclooxygenase-2; ERK1/2 (Extracellular Signal-Regulated Kinases 1 and 2); HER: Human epidermal growth factor receptor
Cytokines
Cytokines play pivotal roles in the intricate processes by which metastatic lung cancer cells colonize the brain parenchyma. The release of cytokines, chemokines, and tumor-secreted exosomes primes the brain microenvironment, fostering the growth of lung cancer cells within the brain [143]. Several critical factors and interactions significantly contribute to this complex process. Transforming growth factor-beta 1 (TGF-β1) is widely acknowledged for its role in the promotion of EMT. Khan et al. demonstrated that compared with their wild-type counterparts, lung cancer cells pretreated with TGF-β1 in mouse models presented a nearly threefold increase in their ability to metastasize to the brain [144]. TGF-β1 single nucleotide polymorphisms (SNPs), particularly rs1982073, have been associated with reduced BM-free survival in patients with NSCLC who have undergone radiation therapy. This genetic variant appears to serve as a predictive factor for an elevated risk of BM development in the patient population following radiation treatment [145]. Genotype variants within the TGF-β signaling pathway can also act as predictive biomarkers for BM development in NSCLC patients, offering potential insights into disease progression and treatment response [143].
Furthermore, tumor necrosis factor-alpha (TNF-α) is inversely correlated with N-acetyl-aspartate, an indicator of mitochondrial oxidative capacity in the occipital cortex. Even before BM formation, the cerebral metabolic status of patients with lung cancer is altered, characterized by lower levels of glutamate, creatine, and phosphocreatine in the cortex [146].
In NSCLC samples derived from patients with BMs, there was notable upregulation of the expression of the chemokine CXCL12 and its corresponding receptor CXCR4. CXCR4 plays a pivotal role in communication between cancer cells and their microenvironment [147]. In a study by Chen et al. involving 32 patients with BM originating from NSCLC, the expression patterns of CXCR4 were significantly elevated in 90% of primary tumors and 100% of BMs compared with those in NSCLC patients without distant metastases [148]. Similarly, Paratore et al. reported concordant results, indicating that the immunoreactivity of CXCL12 and CXCR4 was significantly greater in NSCLC samples from patients with BMs. Receiver operating characteristic (ROC) analysis was conducted to determine the optimal cut-off values for CXCL12 and CXCR4 immunoreactivity, enabling discrimination between NSCLC patients with and without BMs. This expression profile demonstrated good diagnostic accuracy and adequate predictive power [149]. Furthermore, the downregulation of C-X3-C motif chemokine receptor 1 (CX3CR1) in lung adenocarcinomas is associated with an increased likelihood of metastatic spread to the brain [150].
Another study demonstrated that the frequency of programmed death-ligand 1 (PD-L1) in myeloid cells is correlated with the presence of BM. Compared with controls, patients with lung carcinoma with BMs presented elevated PD-L1 levels in peripheral monocytes [151]. It has also been revealed that tumor-derived IL-6 can induce PD-L1 expression in myeloid cells. Treating lung cancer cells with brain metastasis-conditioned media with anti-IL-6 or anti-IL-6 receptor antibodies reduced PD-L1 expression in patient-derived BM [152].
Other molecules involved in BM
Various other molecules are closely associated with BM formation in primary lung tumors (Table 1). A long non-coding RNA called MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) promotes EMT and BM formation [153]. Not surprisingly, the presence of MALAT1 in primary NSCLC is associated with BM and a poor prognosis [154, 155]. Silencing MALAT1 in invasive lung cancer cells prevents BM formation in mice [156]. In contrast, MALAT1 can have opposing effects on breast cancer, acting as an antimetastatic factor in some cases [157]. In addition, the cell adhesion molecule N-cadherin is overexpressed in the BM of patients with NSCLC [158]. It is a mesenchymal marker belonging to the calcium-dependent adhesion molecule family, which directly mediates cell–cell adhesion and is crucial for cancer progression [159]. In parallel, loss of E-cadherin expression in primary tumors of patients with NSCLC was significantly associated with BM [160]. Moreover, silencing CADM2 in several NSCLC cell lines results in reduced vimentin levels, decreased cell migratory ability, and increased expression of the epithelial marker E-cadherin [161]. Therefore, CADM2 induces EMT and supports the migration of lung cancer cells into the CNS [6]. Tumor necrosis factor-alpha (TNF-α) amplifies the adhesion between CD15 and E-selectin. Studies suggest CD15 facilitates cancer cell adhesion to the brain endothelium, particularly under TNF-α-induced inflammatory conditions [162]. Samah Jassam and colleagues designed experiments using in vitro models to show that blocking CD15 or CD62E significantly reduces cancer cell adhesion to the brain endothelium, highlighting their importance in the early stages of cancer cell extravasation [162].
Additional factors, such as cystatin C, cathepsin L, insulin-like growth factor-binding protein 7 (IGFBP7), and vascular endothelial growth factor (VEGF), are secreted by NSCLC cells that metastasize to the brain [163]. These factors contribute to the impairment of the endothelial glycocalyx, resulting in the upregulation of E-selectin and facilitating the adhesion of metastatic cells to the microvascular endothelium of the brain [143].
CD44, a non-kinase cell surface glycoprotein, constitutes a substantial family of cell adhesion molecules (CAMs) that exhibit diverse functions, such as interactions with ECM components, cytokines, and growth factors secreted by cells within the tumor microenvironment [164]. The entire CD44 family is encoded by a single gene comprising 19 exons [165, 166]. CD44 is present in embryonic stem cells, connective tissue, and bone marrow and has been recognized as a marker for cancer stem cells across a spectrum of tumors [166]. In this respect, a study on 15 patients with BM originating from primary NSCLC tumors demonstrated that a deficiency in CD44 protein expression is associated with increased metastatic potential of NSCLCs [164].
miRs are crucial regulators of BBB permeability, influencing tumor cell extravasation into the brain microenvironment. For instance, in NSCLC, Wu et al. demonstrated that miR-1207-5p targets Erythrocyte Membrane Protein Band 4.1 Like 5 (EPB41L5) [167]. EPB41L5, a member of the band 4.1 protein family, is essential for maintaining the structural integrity and function of the cell membrane [168]. It promotes EMT, disrupts TJs, and increases BBB permeability. By targeting EPB41L5, miR-1207-5p exerts a tumor-suppressive role, helping to maintain BBB integrity and limit metastatic spread to the brain [167, 169]. Similarly, Li et al. found that miR-596-3p targets Yes-associated protein 1 (YAP1) in NSCLC. YAP1 is pivotal in cellular responses to growth signals, and its dysregulation has been linked to various cancers [170]. YAP1 promotes MMP-2 activity, which degrades TJ and impacts BBB permeability [171].
In BM from breast cancer, it was shown that miR-509 targets TNFα and RhoC. RhoC is a member of the Rho family of small GTPases, which are critical regulators of various cellular processes, including cytoskeletal dynamics, cell migration, cell cycle progression, and gene expression [172]. This targeting effectively inhibits RhoC-induced MMP-9 activity, which also disrupts BBB TJ [171].
Metastasis of breast cancer to brain tissue
Breast cancer (BC) is the most common cancer in the female population [173], with 2.26 million diagnosed cases and 685 thousand reported deaths worldwide in 2020. The clinical categorization of BC depends on the expression of estrogen receptor (ER), progesterone receptor (PR), and HER2/Neu, as well as on the value of Ki-67 [174, 175].
HER2 is significantly overexpressed in invasive BC, ranging from 15–35% [176]. In addition to its role in breast cancer, HER2 overexpression has also been observed in other types of cancer [177]. The functional tyrosine kinase domain of HER2 can be activated via interactions with ligand-activated EGFR or HER3. Like EGFR, activated HER2, whether hetero or homodimers, initiates phosphorylation events, activating several signaling pathways implicated in BC progression [176]. These pathways, including the STAT3, RAS-MAPK, and PI3K pathways, result in the inactivation of proteins that trigger apoptosis and the upregulation of genes that promote cell growth, thereby facilitating tumor cell proliferation [176]. Recent research has revealed that HER2 directly interacts with the proapoptotic protein p53 upregulated modulator of apoptosis (PUMA) and phosphorylates PUMA, leading to its degradation and promotion of tumor cell survival [178]. As described below, numerous other factors are involved in BC metastasis to the brain.
STAT3 pathway
The distinctive brain microenvironment inadvertently facilitates the development and expansion of the BM. A subset of reactive astrocytes (RA), activated by STAT3, surrounds BMs [179]. STAT3, a member of the STAT family, plays a role in cell growth, apoptosis, tumor progression, and differentiation [180]. Tumor cells release various cytokines that induce reactivity in astrocytes. The interaction between cytokines and growth factors with their respective receptors activates a critical signaling pathway, the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway [181]. These STAT3 + reactive astrocytes generate cytokines and other substances that impede the body's appropriate response to the tumor, thereby compromising both the innate and adaptive immune systems [179].
Recent investigations have revealed that a significant proportion of astrocytes identified in BMs exhibit phosphorylated (active) STAT3 (pSTAT3), indicating the importance of STAT3 signaling in tumor-associated cells [182]. pSTAT3 + reactive astrocytes hinder the access of CD8 + cytotoxic T cells to cancer cells by increasing the expression of specific immunosuppressive molecules, such as programmed cell death-1 ligand 1 (PD-L1), VEGF-A, lipocalin-2, and tissue inhibitor of metalloproteinases-1 (TIMP-1). In primary brain tumors, tumor-associated astrocytes express STAT3 and PD-L1 at elevated levels, contributing to an immunosuppressive environment by producing additional cytokines such as IL-10 and TGF-b [180, 10972].
RA can also shield cancer cells from chemotherapy by upregulating the expression of specific genes, which is essential for generating gap junctions between the two cell types [183]. The interaction between protocadherin 7 (PCDH7) on cancer cells and Cx43 on astrocytes facilitates the creation of these gap junctions. Through these gap junctions, cancer cells stimulate the cGAS-STING (stimulator of interferon genes) pathway in astrocytes via 2′,3′-cyclic GMP-AMP (cGAMP), increasing the production of potent inflammatory cytokines, such as interferon-α (IFN-α) and tumor necrosis factor [184, 185]. These cytokines can promote BM in cancer cells by activating the STAT1 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling pathways [108].
Moreover, nuclear EGFR, in collaboration with STAT3, has been demonstrated to directly upregulate genes that facilitate the BM process [177]. EGFR activation has additional effects, including direct phosphorylation of the STAT3 transcription factor and its translocation to the nucleus for the regulation of gene expression. EGFR can translocate to the nucleus, influence gene expression, phosphorylate nuclear proteins, and alter their functions. Increased nuclear EGFR expression has been associated with poor clinical outcomes in various cancers. Additionally, EGFR can translocate to the mitochondria to counteract therapy-induced apoptosis [177] (Fig. 3).
BC most frequently metastasizes to three distinct anatomical regions in the brain. The brain parenchyma is the most common site, with 78% of cases presenting with multiple metastases and 14% presenting with solitary metastases. Additionally, 8% of metastases occur in the leptomeninges, and the choroid plexus is considered a sanctuary site for breast cancer metastasis [186].
Xist is a 19 kb long non-coding RNA that originates from the inactive X chromosome and plays a crucial role in X chromosome inactivation in female cells [187]. A study by Xing et al. demonstrated significant downregulation of Xist in metastatic brain tissue, leading to the induction of EMT through c-Met activation [74, 188]. c-Met is a tyrosine kinase receptor that binds to hepatocyte growth factor and subsequently activates various cellular signaling pathways crucial for proliferation, motility, migration, and invasion. Mutations in c-Met are associated with various human cancers [189]. Furthermore, the loss of Xist results in the secretion of exosomal microRNA-503, promoting the transition of microglia, a key component of the brain's innate immune system, from a tumor-suppressive (M1 pro-inflammatory) phenotype to a tumor-promoting (M2 anti-inflammatory) phenotype [74]. This transition allows metastatic tumor cells to overcome the cytotoxic effects of M1 microglia and facilitate their growth.
Microglia pose a significant challenge to overcome, as they are integral to the innate immune system and release a substantial number of cytokines and inflammatory molecules upon activation by inflammation or cancer cells [188]. The primary cytokines associated with the M1 phenotype include interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), IL-2, and IL-12. In contrast, the M2 phenotype is correlated with cytokines such as IL-4, IL-10, IL-13, and TGF-β [190]. Metastatic tumor cells employ two strategies to evade the immune defense mechanism: first, by avoiding the cytotoxic effects of the M1 phenotype, and second, by inducing conversion from the M1 phenotype to the M2 phenotype [188].
Huang et al. investigated the correlation between the PI3K/AKT cellular pathway and Xist, revealing an inverse relationship. Consequently, as the level of Xist decreases in cells, the level of pAKT increases [74]. Activation of the PI3K pathway has been observed in 70% of BC patients with BM [191]. Moreover, downregulation of Xist has been detected in 78% of primary tumors in patients with BC-derived BMs [188].
Cell adhesion molecular markers
As circulating tumor cells (CTCs) traverse the bloodstream, they eventually become trapped in microvessels within the brain, particularly at the branching points of the vasculature, initiating adhesion to endothelial cells (ECs). The interaction between E-selectin on the EC surface and CD44 and MUC1 facilitates shear-resistant adhesion, particularly at branching sections of the vasculature [186]. The expression of E-selectin is provisional and can be released into the circulation in a soluble form, or E-selectin may be internalized after activation. Notably, there have been reports of high levels of soluble E-selectin in the serum of patients with metastatic breast cancer [192]. The overexpression and abnormal glycosylation of MUC1 are commonly observed in various cancers. Physiologically, MUC1 is 5–10 times larger than most membrane proteins because its repeating units are 20 identical amino acid sequences rich in serine and threonine. Abnormal MUC1 with shorter oligosaccharides can efficiently bind E-selectin [193].
Brain metastasis is observed in 10–30% of women diagnosed with stage IV breast cancer [108], with the highest occurrence in the triple-negative subtype (25–27%), followed by the HER2 + subtype (11–20%) [175]. The risk of BM is notably higher in patients with HER2-positive breast cancer or triple-negative breast cancer (TNBC) than in those with luminal BC. The estimated frequency of BMs ranges from 20–30% in patients with HER2-positive breast cancer and TNBC, whereas it is less than 10% in patients with the luminal subtype [108].HER2 promotes the phosphorylation of proteins across multiple signaling pathways, suppresses pro-apoptotic proteins, and enhances the expression of genes related to cell proliferation, processes that contribute to epithelial-mesenchymal transition (EMT) and increase the risk of brain metastasis [194,195,196]. Notably, in aggressive breast cancers, HER2 is often co-expressed with junctional adhesion molecule-A (JAM-A), a cell–cell adhesion protein involved in tight junction formation in epithelial and endothelial cells, further supporting tumor progression and metastatic [197, 198].. HER2-overexpressing BC cells induce the expression of transcription factors such as Snail, Slug, and ZEB1, leading to increased TGF-β production, elevated N-cadherin levels, and reduced E-cadherin and cytokeratin-18 expression [38]. Targeting HER2 has been suggested as a potential therapeutic strategy to inhibit metastasis, as HER2 inhibition has been shown to reduce metastasis not only to the brain but also to the lungs and liver in mouse models.
Another regulator of E-cadherin is SNORA17B, a small nucleolar RNA (snoRNA) involved in ribosomal RNA methylation. SNORA17B is highly expressed in the brain metastases of patients with breast cancer and is associated with a poor prognosis. SNORA17B-transfected human breast cancer cell lines display increased invasive ability and reduced E-cadherin expression, suggesting its involvement in controlling E-cadherin levels [6].
Astrocyte elevated gene-1 (AEG-1) is another molecule that links EMT with BM. AEG-1 overexpression upregulated N-cadherin and reduced E-cadherin and ZO-1 expression. AEG-1 is an endoplasmic reticulum (ER)-associated cytoplasmic RNA-binding protein that interacts with numerous mRNAs encoding secretory, cytosolic, and organelle proteins [199]. Initially discovered in primary astrocytes of the human fetus and mainly localized in the ER, AEG-1 was later found in the cell membranes of breast cancer cells. Its roles in cancer include promoting malignant transformation, resistance to chemotherapy, anoikis, angiogenesis, and metastatic spread [41, 42, 200].
In a study conducted by Wu et al., the roles of MMP1 and cyclooxygenase 2 (COX2) in breast cancer brain metastasis were investigated [201]. This study demonstrated increased expression of MMP1 and COX2 in brain metastatic cancer cells. MMP1 plays a role in degrading claudin and occludin [201], which are crucial transmembrane proteins integral to tight junctions [202]. This study also revealed the upregulation of COX2 in brain metastatic cells. COX2 induces the secretion of prostaglandins, which in turn enhances the expression of MMP1. Moreover, COX2 and prostaglandins can stimulate astrocytes to release CCL7, thereby increasing the self-renewal capacity of tumor cells in the brain. In support of this, when MMP1 and COX2 are knocked down in brain metastatic cells, their ability to metastasize to the brain is reduced [32].
Metastasis of melanoma to brain tissue
Melanoma, a malignancy originating from melanocytes responsible for melanin production and derived from the neural crest [203], presents a notable complication in advanced stages, with BMs affecting nearly half of the patients and contributing up to 54% of melanoma-related mortalities [204]. Among these BMs, approximately 49% are intraparenchymal, 22% are leptomeningeal, and 32% are dural [205, 206].
The development of melanoma BMs (MBMs) is preceded by early changes in the brain microenvironment, including breakdown of the BBB, increased vascular permeability, and reactive astrogliosis. Schwartz et al. revealed the upregulation of pro-inflammatory cytokines such as CXCL10, CCL17, CCL2, IL-6, and IL-1β during MBM [96].
Studies indicate that CXCL10 influences the migration of monocytes, macrophages, T cells, and natural killer (NK) cells to the brain [207]. Patients with advanced melanoma, which is associated with a poor prognosis, have higher levels of CXCL10 [208, 209]. Furthermore, the receptors for CXCL10 and CXCR3 are upregulated in melanoma cells, with a predilection for the brain [210]. Immunokine profiling studies in the CSF of patients with advanced melanoma suggested that increased levels of CXCL10, CCL17, and CCL4 may be correlated with a more aggressive development of BMs [211].
MMP-2 has emerged as a crucial factor, exhibiting heightened expression correlating with increased malignancy, particularly in the most metastatic cell lines, MV3 and BLM. The primary focus is on the mRNA and protein expression patterns of MMP-1, MMP-2, MMP-3, MMP-9, TIMP-1, and TIMP-2, both in vitro and in vivo. Additionally, they revealed that MMP-1, although detectable in all cell lines at the mRNA level, has limited protein expression [212].
In terms of STAT3, MBM cells exhibit increased activity of STAT3, AKT [143, 178, 213], and AKT. STAT3 activity in the MBM triggers a cascade of events that contribute to the potential of the BM, including angiogenesis, cell invasion, MMP-2 secretion, cytokine expression, and immune suppression [178]. In melanoma cell lines, the loss of suppressor of cytokine signaling-1 (SOCS-1) expression is correlated with increased STAT3 signaling and subsequent overexpression of MMP-2, basic fibroblast growth factor (bFGF), and VEGF [214]. This, in turn, increases invasion and angiogenesis in melanoma cells, facilitating the development of MBM [214].
Patel et al. suggested that STAT3 activation is a key contributor to the occurrence of melanoma-derived BMs, as evidenced in a study of 216 autopsied metastatic melanoma specimens [215]. Moreover, brain-metastasizing melanoma cells have the capacity to reprogram astrocytes, leading to the expression of the pro-inflammatory cytokine IL-23, which in turn increases MMP-2 levels and facilitates melanoma cell migration and invasion into the brain parenchyma. Conversely, reduced MMP-2 expression in melanoma cells has been linked to the inhibition of IL-23-induced invasiveness [216].
Brain barriers in relation to brain metastases and metastatic spread
Most BMs are found in the brain parenchyma, dura, and leptomeninges [217, 218] and, to a lesser degree, in the pituitary and pineal glands [217]. One of the main characteristics of primary and brain metastases is the formation of surrounding vasogenic brain edema, which reflects damage to the BBB [219, 220]. The relative frequency of metastases in different brain regions can be influenced by several factors, including brain anatomy and the type of primary tumor.
Generally, metastatic cells can reach brain tissue through the paracellular pathway or through transcellular migration. However, low transcytosis, together with the limited permeability of TJs, makes it challenging for metastatic cells to cross the BBB and reach brain tissue. The paracellular pathway is characterized by the alteration of TJ proteins and the apoptosis of endothelial cells [221, 222]. The extravasation of metastatic cells shares similarities with that of leukocytes, revealing three main steps in the extravasation process: rolling, adhesion, and transmigration (diapedesis).
Brain metastases are most commonly located in the supratentorial region of the brain above the tentorium cerebelli in the cortex and white matter of the brain. In this area, blood flow is slower, allowing metastatic cells to adhere to the capillary wall.
In contrast, the hippocampal region is a preserved part of the brain with a relatively lower incidence of BMs, probably due to a different vessel supply (via the posterior cerebral artery) or due to differences in the BBB related to the unique presence of neural stem cells involved in new memory formation [223]. During whole-brain radiotherapy, the hippocampus is avoided to preserve neurocognitive functions, including memory, due to the relatively low risk of the presence of BM [224].
Blood–brain barrier in relation to brain metastases
Metastatic cells rolling and adhesion to endothelial cells
The rolling step is probably the initial phase of the adhesion of metastatic cells to the brain endothelium, similar to immune cells. The key molecules mediating tumor cell rolling and adhesion are glycoproteins, integrins, galectins, and the Thomsen-Friedenreich antigen, glycoproteins present on the surface of most human cancer cell types [225, 226]. These molecules serve as ligands for the adhesion receptors expressed on the surface of endothelial cells. During the rolling step, E-selectin on the surface of endothelial cells recognizes and binds to glycoproteins, such as HCELL, PSGL-1, CD24, and CEA, which are ligands that bind to L-selectin (Fig. 4). N-cadherin, a member of the calcium-dependent adhesion molecule family localized on the brain endothelium, mediates cell contact via its expression on the metastatic cell surface. Metastatic cells can use leukocytes as bridging cells to promote adhesion through ICAM-1, an adhesion molecule present on the surface of endothelial and tumor cells (Fig. 4). Because metastatic cells, unlike leukocytes, do not express β2-integrins, ligands required to interact with ICAM-1, the adhesion of tumor cells occurs by bridging by immune cells [226, 227]. The mechanisms of adhesion of metastatic cells to the endothelium include the expression of α4, β1, and β7 integrins, which act as ligands for vascular cell adhesion protein 1 (VCAM-1) and fibronectin. As mentioned above, the Thomsen-Friedenreich antigen, represented by MUC1 and CD44v6, and lysosomal membrane-associated glycoproteins (Lamps) 1 and 2 serve as ligands for galectin-3, thus increasing tumor adhesion to endothelial cells [226].
Mechanisms of Metastatic Cell Rolling and Adhesion to Brain Endothelial Cells. This schematic illustrates the endothelial cell layer, pericytes, and astrocytes forming the blood–brain barrier, with metastatic cells depicted in the process of adhering to endothelial cells facilitated by various molecular interactions. Rolling phase: Endothelial cells express E-selectin, which binds to glycoproteins such as HCELL, CD24, PSGL-1, and CEA on the surface of metastatic cells, initiating the rolling process. The adhesion phase includes the following steps: (1) Leukocyte bridging: Metastatic cells use leukocytes (which express β2 integrins) as intermediaries to bind to ICAM-1 on endothelial cells. (2) Direct adhesion: Metastatic cells also express integrins (α4, β1, β7) that interact with VCAM-1 on endothelial cells, facilitating direct adhesion. Additionally, MUC1, CD44v6, and lysosomal-associated membrane proteins (Lamps) 1 and 2 on metastatic cells bind to galectin-3 on endothelial cells, enhancing adhesion. 3) N-cadherin interaction: N-cadherin on metastatic cells interacts with N-cadherin on brain endothelial cells, promoting stable cell contact
Metastatic cell transmigration through endothelial cells
Role of proteolytic enzymes
Metastatic cells squeeze among endothelial cells and disrupt the main TJ transmembrane proteins, such as occludin, claudin-5, and the cytoplasmic protein ZO-1 [228]. The disruption of TJ proteins within endothelial cells and the ECM depends on the accumulation of proteolytic enzymes. The ECM and basement membrane are composed of many different components; therefore, their disintegration, subsequent migration, and invasion of metastatic cells require enzymes with distinct substrate specificities. Increased expression of proteolytic enzymes and other harmful molecules results from specific interactions between metastatic cells and the cellular components of the BBB.
These “harmful” molecules encompass various proteolytic enzymes and signaling molecules that metastatic cells use to breach the BBB. They compromise BBB integrity and can damage surrounding brain tissue, promoting invasion and growth of metastatic cells within the brain. In the following section, we will discuss these molecules in more detail. The enzymatic system triggered by the interaction of these cells catalyzes the proteolysis of TJ proteins and basement membrane components, such as collagen, laminin, or fibronectin proteoglycans, and activates other enzymes, accelerating the penetration of metastatic cells through the BBB [229]. These enzymes include serine proteases, seprases, MMPs, cathepsin S, disintegrin, metalloproteinase domain-containing protein 9 (ADAM9), urokinase-type plasminogen activator (uPA), and the plasminogen/plasmin system [212, 221, 230,231,232,233] (Fig. 5).
Mechanisms of Metastatic Cell Transmigration through Brain Endothelial Cells. This figure represents the transmigration of metastatic cells across the BBB, which is a complex process involving all components of the BBB. Interaction between metastatic cells and BBB components results in an increase in pro-inflammatory cytokines, chemokines, receptors, proteases, and growth factors. These molecules create a suitable microenvironment for metastatic cells to survive and spread. Several proteolytic enzymes, including separase, MMP-2, uPa, dipeptidyl peptidase IV, and membrane-type 1 MMP causing ECM degradation, are highly expressed on the surface of metastatic protrusions called invadopodia. Communication between metastatic and endothelial via the gap junctions contributes to metastatic cell spread, survival and growth
The fibrinolytic enzyme plasmin is generated after the activation of plasminogen-by-uPA and tissue-type plasminogen activator (tPA) and is expressed on the surface of metastatic cells [234]. The conversion of plasminogen into plasmin may be enhanced by membrane-bound melanotransferrin, which stimulates the activation of plasminogen through tPA and uPA [235, 236]. Several proteolytic enzymes, such as seprase associated with α3β1 integrin, dipeptidyl peptidase IV, MMP-2, membrane-type 1 MMP, and uPA, are highly expressed on the metastatic cell surface of cell protrusions associated with sites of ECM degradation, called invadopodia [237, 238]. Separases work synchronously with other proteinases, such as MMPs. This synchronized action leads to the cleavage of partially degraded or denatured ECM components, including collagen I and III [238]. In addition, pericellular proteolysis is potentiated by the formation of supramolecular lytic complexes in the plasma membranes of invading metastatic cells. These supramolecular structures are dependent on both the cytoskeleton and integrins, especially β1-integrin [230]. ECM and BM heparan sulfate proteoglycans are considered storage depots for various cytokines and growth factors. The ECM heparan sulfate proteoglycans contain angiogenic factors, such as basic fibroblast growth factor (bFGF), which are released from the ECM heparan sulfate proteoglycans and can support the induction of neovascularization during the migration of tumor cells through the BBB [239]. Moreover, there is evidence that metastatic cells upregulate the expression of some factors, such as IL-23, in astrocytes, which facilitates the progression of BBB disruption by inducing the secretion of MMP-2 [216]. Activated astrocytes play a direct role in tumor metastasis and BBB disruption through the secretion of MMP-2 and MMP-9 [240]. Astrocytes and microglia are immediately localized in the vicinity of metastatic cells and contribute to the progression of cancer cells through the secretion of MMPs [241].
Extravasation through nonfenestrated brain capillaries is influenced by the expression of COX-2, heparin-binding EGF-like growth factor receptor, and ST6 N-acetylgalactosaminide alpha-2,6-sialyltransferase 5 (ST6GALNAC5) in metastatic cells [242]. ST6GALNAC5, a brain-specific sialyltransferase, catalyzes the addition of sialic acid to gangliosides and glycoproteins. Cell-surface sialylation enhances the adhesion and subsequent transmigration of metastatic cells through the brain endothelium [242, 243].
Role of trophic factors and neuroinflammation
Activated astrocytes produce and release trophic factors, including neurotrophin-3, TGF-β, or bFGF, as well as nerve growth factor (NGF), S100 protein, and lipocortin precursors to β-melanocyte-stimulating hormone. These molecules support the survival of metastatic cells in the CNS [244]. The penetration of metastatic cells into the neurotrophin-rich stromal brain microenvironment is potentiated by NGF, which serves as a chemoattractant for ADAM9 overexpression in metastatic brain cells [232]. Moreover, in response to NGF, metastatic cells express invasive enzymes such as MMP-2, type IV collagenase, and heparinase [245]. The synthesis of neurotrophins from adjacent brain tissue is also potentiated by cytokines, chemokines, and other inflammatory molecules produced by metastatic cells, including VEGF, bFGF, TGF-β, IL-1β, TNF-α, interferon-gamma (IFNγ), CCL2, C-X-C motif chemokine ligand (CXCL) 8, and COX2, which are major players in inflammatory reactions [228, 245]. Increased expression of COX-2, an inducible pro-inflammatory enzyme, and subsequently MMP-1 was associated with decreased expression of the TJ proteins claudin-5 and VE-cadherin [246].
The expression of invasive enzymes such as MMPs and heparinase is potentiated by autocrine activation of EGFR by HB-EGF and EP2 receptors by PGE2 generated by COX-2 [126, 247]. The pro-inflammatory enzyme COX-2 is involved in the generation of prostaglandins, which are involved in tumorigenesis [201]. These prostaglandins activate astrocytes to generate a niche for metastatic cells by increasing the expression of chemokine (C–C motif) ligand 7 (CCL7), which promotes self-renewal of metastatic cells. CCL7 activates G protein-coupled transmembrane receptors and transmits intracellular signals via the MAPK pathway [212]. The subsequent cytoskeletal rearrangement and decreased integrity of the endothelial layer are partially caused by vascular endothelial growth factor (VEGF). Rearranging the cytoskeleton is promoted by VEGF-induced upregulation of cytosolic calcium levels, leading to the phosphorylation and activation of myosin light chain kinase, resulting in the contraction of endothelial cells [248]. In the case of breast cancer, VEGF also activates endothelial cells that increase the secretion of angiopoietin-2 (Ang-2), one of the molecules involved in the initial step of the metastasis process. Increased secretion of Ang-2 leads to decreased expression of TJ proteins, resulting in alterations in the BBB [249].
The overexpression of MMPs and thus invasiveness, as well as the proliferation of metastatic cells, is potentiated by signaling cascades, including the PI-3 K/Akt and Ras/MEK/Erk cascades. Initiation of these pathways results from the amplification of transmembrane tyrosine kinase receptors from the ErbB family, particularly ErbB2, which forms heterodimers with other members, including ErbB1/HER1, ErbB3/HER3, and ErbB4/HER4 [250]. Moreover, ErbB2 induces the upregulation of chemokine receptor 4 (CXCR4) in metastatic cells. SDF-1α (CXCL12), a specific ligand for CXCR4 expressed in various cells, including brain endothelial cells, contributes to the migration and invasion of malignant cells through the BBB [250, 251]. Another specific ligand belonging to the CXC chemokine family, CX3CL1 and CXCL13, may have barrier-compromising effects by affecting BBB integrity in vitro. These chemokines are part of the endothelial inflammatory phenotype, and high levels of CX3CL1 may attract metastatic cells expressing its receptor CX3CR1 [252]. As mentioned above, the activation of downstream cascades via heterodimerization of the human epidermal growth factor ErbB2/HER2 with ErbB3/HER3 is potentiated by neuregulin-1. Increased expression of neuregulin-1 was found in brain endothelial cells; therefore, neuregulin-1-HER2/ErbB2-HER3/ErbB3 signaling likely promotes metastatic cell transmigration through the BBB by increasing the expression of MMPs such as MMP-9 and the adhesive molecule ICAM-1 [253]. Following extravasation, metastatic cells remain in direct contact with abluminal endothelial cells of the brain capillaries. The location of metastatic cells in the perivascular space in a pericyte-like position provides access to the oxygen and other nutrients necessary for successful proliferation [254]. The subsequent migration of metastatic cells is likely promoted by pleckstrin homology domain-containing A5 (PLEKHA5), a protein that is tyrosine-phosphorylated downstream of Met signaling. PLEKHA5 is associated with microtubules and is expressed at the plasma membrane of the leading edge of motile cells, suggesting its role in cell–cell contact and the migration of metastatic cells [255]. Metastatic cell integration into foreign tissue after they leave circulation and perivascular microtumor formation depends on the creation of more favorable conditions in a foreign microenvironment. Thus, the formation of GJs in the Cx43-rich brain endothelium may play an important role. GJ communication between metastatic and endothelial cells mediates the transfer of ATP, small peptides, ions, and small regulatory RNAs, which are crucial for tumor cell survival and growth [256].
3.2. Blood-arachnoid barrier (leptomeninges) in brain metastasis
The dura mater is considered an effective barrier against the spread of metastatic cells to the brain. Dural invasion is a less frequent route by which tumor cells spread to the CNS. However, it may occur secondary to lymphoma and adenocarcinoma of the stomach, breast, or lung [257]. In the literature, metastatic cell migration through the arachnoid and pia mater is referred to as leptomeningeal metastasis. The leptomeningeal system of tissues consists of the arachnoid mater, arachnoid villi, arachnoid vessels, arachnoid granulations, arachnoid trabeculae, cerebrospinal fluid, and pia mater [258]. The pia mater covers tunnels through blood vessels that pass through the CNS, known as perivascular spaces. The perivascular spaces are absent once blood vessels reach the capillary stage at the BBB. This layer is mainly composed of type IV collagen [259]. Ultrastructural studies have shown that the pia mater layer lining the brain vessels is fenestrated. Therefore, metastatic cells can move freely within leptomeninges [260] (Fig. 6). However, neoplastic cells also have the ability to disintegrate the leptomeningeal cell layer. This suggestion is supported by the finding of collagen fiber fragments in the gap between metastatic cells [261]. Metastatic cells use leptomeningeal blood vessels to enter the brain parenchyma. The migration of metastatic cells from the metastatic origin along leptomeningeal microvessels to the surrounding brain tissue contributes to the development of multiple brain lesions [262].
Schematic illustration showing transmigration of metastatic cells through the BAB into the subarachnoid space. Some metastatic cells, such as ALL lymphocytes, use α6 integrin to bind to laminin. After metastatic cells migrate to the meninges, surrounding immune cells release pro-inflammatory cytokines and recruit cytotoxic T cells. Later, metastatic cells begin to induce an immunosuppressive microenvironment including regulatory T cells (Tregs) and inhibitory macrophages. To reach the cerebrospinal fluid, metastatic cells must pass through the epithelial cells of the BAB, which are connected by TJ proteins. Metastatic cells in the subarachnoid space produce proteases such as MMP-2 and MMP-9 that degrade the pial-glial membrane containing type IV collagen and enter the brain tissue
The basement membrane of leptomeningeal vessels is enriched in laminin. During acute lymphoblastic leukemia (ALL), lymphocytes use α6 integrin, a laminin receptor, to enter the leptomeninges and subarachnoid spaces. The arachnoid serves as a barrier that separates the CNS from the dura mater; however, lymphocytes are able to breach this barrier during ALL [263, 264]. Similarly, lymphocytes derived from B-cell lymphoma use the meninges or perivascular spaces to spread to the brain parenchyma. Neoplastic cells that form B-cell lymphomas present as solid meningeal tumors or diffuse tumors that spread in the brain along the leptomeninges.
B cells enter the perivascular space by rolling along the lumen of leptomeningeal blood vessels, followed by conformational changes in integrins, vessel formation, and migration through endothelial cells via diapedesis [265]. After metastatic cells reach the subarachnoid space, to infiltrate the brain, they must destroy the pia-glial membrane, which plays an important role in cerebroprotection. The pia-glial membrane is primarily composed of type IV collagen. It seems that the disintegration of the pial-glial membrane by metastatic cells occurs through type IV collagenases such as MMP-2 and MMP-9 [259].
The leptomeninges are sites of resident immune cells, including B cells, T cells, dendritic cells, and macrophages. When metastatic cells pass through the dura mater, leptomeningeal immune cells release pro-inflammatory cytokines and recruit cytotoxic T cells via adjacent lymphatic drainage. However, in the future, metastatic cells will start to propagate into immunosuppressive tumor microenvironments involving regulatory T cells (Tregs) and inhibitory macrophages. The immunosuppressive nature of metastatic cells may be reversed by immunotherapy, which reactivates the immune response [266].
The circumventricular organs and brain metastasis
Circumventricular organs (CVOs) include secretory and sensory brain structures lacking the BBB. Secretory CVOs include the pineal gland, neurohypophysis (posterior pituitary), and median eminence. Sensory CVO includes the organum vasculosum of the lamina terminalis, the area postrema, and the subfornical organ [267]. Functionally, secretory CVOs that produce hormones and peptides are involved in neurochemical transport and chemoreception [268]. Moreover, sensory CVO controls chemicals from the circulatory system and CSF and conveys this information to neural effector centers to maintain body fluid homeostasis [267, 269]. A common feature of these organs is that they lack the blood–brain barrier (BBB). Therefore, substances in the blood can travel freely between fenestrated capillaries with loosely connected astrocytic endfeet and CVO tissues [267]. The CVO contains tenocytes, specialized ependymal cells with apical processes in contact with the CSF, and basal processes that contact fenestrated capillaries or neurons. These cells serve as gatekeepers; that is, they monitor the composition of the CSF and mediate this information to the CVO. Tanycytes are connected to tight junctions and communicate with each other through connexin 43 gap junctions [270, 271]. Owing to its rare occurrence, we will focus only on the most common sites of metastases in CVO, that is, the pineal and pituitary glands.
Pineal glands in relation to metastasis
Metastatic pineal gland lesions are uncommon. Owing to the absence of the BBB, hematogenous dissemination is the main route of metastasis to the pineal gland. According to autopsy reports, pineal metastasis is found in 0.4–3.8% of patients with solid tumors and in 5% of surgically treated patients with pineal tumors [271, 272]. In the literature, the most common types of primary tumors that metastasize to the pineal gland are lung, breast, kidney, esophagus, stomach, colon carcinoma, malignant melanoma, and myeloma [273, 274]. Metastatic lesions in the pineal gland are associated with leptomeningeal seeding through the CSF in 67% of cases [271].
Metastatic spread in the sellar region
The sellar region, including the hypothalamic-pituitary axis, represents the coordinating center of the endocrine system. Metastases in the pituitary gland were found in 3% to 23% of cancer patients in an autopsy series [275]. The occurrence of clinical symptoms in patients with pituitary gland metastasis is uncommon, and most sellar metastases remain asymptomatic [276]. Lung and breast carcinomas are the most common primary tumors that metastasize to the pituitary gland [277, 278]. The hormonal environment may play a role in the development of pituitary gland metastasis, especially in breast cancer [279]. The absence of the BBB and high vascularity support a favorable niche for metastatic seeding [276]. Therefore, radiation sparing of the hypothalamic-pituitary area, which is highly radiosensitive, might be a treatment option for patients with a limited number of brain metastases not involving the sellar region [280]. This approach may prevent radiation-induced hormonal deficiencies.
Blood-cerebrospinal fluid (B-CSF) barrier in relation to brain metastasis
Choroid plexus metastases (CPMs) are uncommon lesions and have generally been reported separately or in groups [218, 281, 282]. Due to the abundant vascular supply of the CP, the lateral ventricles are the most common sites of intraventricular metastases [283]. Under physiological conditions, the B-CSF barrier allows for immune surveillance of CSF. This concept is based on the permeability of the B-CSF barrier for cellular elements such as fluorescence-labeled T lymphocytes, which are found in the CP stroma and meninges after intravenous injection [284]. Leptomeningeal metastasis can develop from any solid tumor, but it often occurs in patients with lung or breast cancer [285]. The B-CSF barrier, as a potential entry point for leptomeningeal metastases, is often overlooked. A more permeable B-CSF barrier than the BBB predisposes this barrier to a higher rate of metastatic cell migration into the CNS. An in vitro study revealed that breast cancer cell migration across the B-CSF barrier was greater than that across the BBB. The loss of barrier properties in the presence of breast cancer may be associated with proteases secreted by cancer cells into the extracellular environment. This results in the degradation of TJ proteins between endothelial cells and, thus, in the degradation of the B-CSF barrier [286]. Transmigration of cancer cells through the layer of epithelial cells of the B-CSF barrier has been described in neuroblastoma, which can lead to leptomeningeal metastasis. After crossing fenestrated capillaries, neuroblastoma cells undergo paracellular transepithelial migration [287]. The exact mechanism by which cancer cells cross the B-CSF barrier is unknown, but alterations in TJ proteins through rearrangement of the actin cytoskeleton have been proposed (Fig. 7). This action is likely mediated by myosin-induced actin contraction via Rho/ROCK activation, resulting in TJ disassembly [288]. Similarly, leptomeningeal metastases produce C3, a central protein in the complement cascade that can affect the B-CSF barrier via myosin light-chain kinase (MLCK) phosphorylation. The main component of this cascade is C3a, an active molecule generated by the proteolysis of C3-by-C3 convertase. Alterations in the B-CSF barrier induced by C3a potentiate the entry of the EGFR ligand amphiregulin into the CSF, thus promoting the growth of leptomeningeal metastases [289]. While metastasis to the CP is generally rare, there have been multiple case reports documenting metastatic tumors in the CP of the lateral ventricle trigone, originating from a large-cell lung carcinoma [290, 291]. Moreover, an additional study indicated that CP metastasis from primary lung adenocarcinoma carrying the EGFR G719X mutation significantly improved following treatment with EGFR inhibitors [133].
Invasion of metastatic cells through the B-CSF barrier Metastatic cells migrate through fenestrated capillaries into the CP, secrete proteases that degrade TJ proteins, and lead to B-CSF barrier damage. Additionally, myosin light-chain kinase (MLCK) phosphorylation, actin contraction via Rho/ROCK, and activation of the C3a-C3aR signaling cascade are involved in the alteration of TJ proteins. These changes in the B-CSF barrier potentiate the paracellular migration of metastatic cells and the transfer of growth factors into the central nervous system. The attraction and invasion of metastatic cells, especially ALL, is potentiated by VCAM-1 and VLA-4 localized on the luminal side of epithelial cells as well as leukemia-derived exosomes
The B-CSF barrier may play an essential role in regulating lymphocyte entry into the CNS [292,293,294]. Moreover, the selective permeability of the B-CSF barrier plays a role in the trafficking of leukemic cells into the CSF. Primary B-cell precursor (BCP) blasts of acute lymphoblastic leukemia (ALL) frequently infiltrate the CNS by crossing the B-CSF barrier rather than the BBB. Despite the finding that BCP-ALL cells express various chemokines that correspond to chemokine ligands expressed in B-CSF barrier cells, no clear evidence exists for the selection of leukemic cells bearing these receptors [295]. However, some integrins, such as very late antigen-4 (VLA-4) (α4:β1), which drives T-cell attraction to the CNS, are also expressed in the CP. Following migration through fenestrated capillaries and the CP epithelium, ALL cells adhere to VCAM-1 and VLA-4 (α4:β1), located on the apical side of epithelial cells [98, 296]. The invasion of leukemia cells across the B-CSF barrier into the CNS may be facilitated by leukemia-derived exosomes, which form the “pre-metastatic niche” and facilitate the invasion of circulating tumor cells into brain tissue. These small extracellular vesicles are actively produced and released by tumor cells in body fluids and modify healthy cells distant from the primary tumor site. There is some evidence that the invasion of ALL through the B-CSF barrier without destruction of barrier integrity may also be facilitated by these leukemia-derived exosomes [98]. The B-CSF barrier, as a primary transition route for ALL into the CNS, was supported by the finding of leptomeningeal infiltration of leukemic cells without parenchymal involvement [287].
Conclusion
The BBB and B-CSF barrier remain the main brain barriers involved in the metastatic process. We have provided a complex review of many molecules involved in brain metastasis from lung cancer, breast cancer, and melanoma. The pathophysiological cascades involved in the migration of metastatic cells across brain barriers are complex and involve not only cellular but also non-cellular components. The major pathological processes involved in the transfer of metastatic cells through brain barriers include the upregulation of adhesion molecules, proteolytic enzymes, trophic factors, and neuroinflammatory changes.
On the basis of our review, little is known about the pathophysiological cascades and molecular mechanisms involved in the migration of metastatic cells into the CNS through the blood-arachnoid barriers. Interestingly, brain structures that lack the BBB, such as circumventricular organs, do not show metastases very often. This observation suggests the involvement of, protective mechanisms beyond the BBB, potentially including yet-undiscovered molecular components that regulate how tumor cells interact with those regions.
Since metastases are the most common type of brain tumor, a better understanding of how metastatic tumor cells use different mechanisms to cross the brain barrier may be a potential target for prevention and treatment. A deeper understanding of the pathophysiology of brain metastases will enable more frequent administration of targeted immunotherapy in personalized medicine approaches that could prevent or postpone whole-brain radiotherapy.
Availability of data and materials
No datasets were generated or analysed during the current study.
Abbreviations
- ADAM9:
-
A disintegrin and metalloproteinase
- AEG-1:
-
Astrocyte elevated gene-1
- AJ:
-
Adherent junction
- AKT:
-
Ak strain transforming
- ALK:
-
Anaplastic lymphoma kinase
- ALL:
-
Acute lymphoblastic leukemia
- Ang-2:
-
Angiopoietin-2
- ATPase:
-
Adenosine triphosphate hydrolyzing enzymes
- BAB:
-
Blood-arachnoid barrier
- BCRP:
-
Breast cancer resistance protein
- BCP:
-
B-cell precursor
- B-CSF:
-
Blood-cerebrospinal fluid
- BBB:
-
Blood–brain barrier
- BMs:
-
Brain metastases
- BSCB:
-
Blood-spinal cord barrier
- CADM2:
-
Cell adhesion molecule 2
- CAFs:
-
Cancer-associated fibroblasts
- CAMs:
-
Cell adhesion molecules
- CCL7:
-
C–C Motif Chemokine Ligand 7
- cGAMP:
-
Cyclic GMP-AMP
- CES1:
-
Carboxylesterase 1
- CNS:
-
Central nervous system
- COMT:
-
Catechol-O-methyltransferase
- CP:
-
Choroid plexus
- CPM:
-
CP metastases
- CTCs:
-
Circulating tumor cells
- CX3CR1:
-
C-X3-C motif chemokine receptor 1
- CXCR4:
-
CXC chemokine receptor 4
- CVO:
-
Circumventricular organs
- ECs:
-
Endothelial cells
- EGFR:
-
Epidermal growth factor receptor
- EMT:
-
Endothelial-mesenchymal transition
- ER:
-
Estrogen receptor
- ErbB2:
-
V-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2
- EVs:
-
Extracellular vesicles
- FGF:
-
Fibroblast growth factor
- GJ:
-
Gap junction
- GLUT1:
-
Glucose transporter 1
- GSTP1:
-
Glutathione-S-transferase protein 1
- HER:
-
Human epidermal growth factor receptor
- HIF-1α:
-
Hypoxia-inducible factor 1-alpha
- HMMR:
-
Hyaluronan-mediated motility receptor
- HRAS:
-
Harvey Rat sarcoma virus
- ICAM-1:
-
Intercellular adhesion molecule 1
- IFN-γ:
-
Interferon-gamma
- IGFBP7:
-
Insulin-like growth factor-binding protein 7
- IL:
-
Interleukin
- INSR:
-
Insulin receptor
- JAK:
-
Janus kinase
- KRAS:
-
Kirsten rat sarcoma virus
- LOH:
-
Loss of heterozygosity
- LRP1:
-
Lipoprotein receptor-related protein 1
- Lamps:
-
Lysosomal-associated membrane protein 1
- MALAT1:
-
Metastasis-associated lung adenocarcinoma transcript 1
- MAPK:
-
Mitogen-activated protein kinase
- MBM:
-
Melanoma BMs
- MET:
-
Mesenchymal-to-epithelial transition
- MDR1:
-
Multidrug resistance 1
- miR:
-
Micro RNA
- MLCK:
-
Myosin light-chain kinase
- MMPs:
-
Matrix metallopeptidases
- MUC1:
-
Mucin 1
- NGF:
-
Nerve growth factor
- NK:
-
Natural killer
- NRAS:
-
Neuroblastoma RAS viral oncogene homolog
- NSCLC:
-
Non-small cell lung cancer
- NVU:
-
Neurovascular unit
- OAT:
-
Organic anion transporters
- PCDH7:
-
Protocadherin 7
- PET:
-
Positron emission tomography
- PI3K:
-
Phosphatidylinositol-3 kinase
- PLEKHA5:
-
Pleckstrin homology domain-containing A5
- PR:
-
Progesterone receptor
- PUMA:
-
P53 upregulated modulator of apoptosis
- RA:
-
Reactive astrocytes
- ROC:
-
Receiver Operating Characteristics
- SCLC:
-
Small cell lung cancer
- SIRT1:
-
Silent mating type information regulation 2 homolog
- SNPs:
-
Single nucleotide polymorphisms
- SOCS-1:
-
Cytokine signaling-1
- STAT3:
-
Signal transducer and activator of transcription 3
- ST6GALNAC5:
-
ST6 N-Acetylgalactosaminide Alpha-2,6-Sialyltransferase 5
- TDEs:
-
Tumor-derived exosomes
- TGF-β1:
-
Transforming growth factor-beta 1
- TIME:
-
Tumor immune microenvironment
- TIMP-1:
-
Tissue inhibitor of metalloproteinases-1
- TJs:
-
Tight junctions
- TK:
-
Tyrosine kinase
- tPA:
-
Tissue-type plasminogen activator
- TNBC:
-
Triple-negative breast cancer
- TNF-α:
-
Tumor necrosis factor α
- TP53:
-
Tumor protein P53
- VCAM-1:
-
Vascular cell adhesion protein 1
- VEGF:
-
Vascular endothelial growth factor
- VLA-4:
-
Very late antigen-4
- ZO:
-
Zonula occludens proteins
References
Balestrino R, Rudà R, Soffietti R. Brain metastasis from unknown primary tumour: moving from old retrospective studies to clinical trials on targeted agents. Cancers. 2020;12:3350. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers12113350.
Ostrom QT, Wright CH, Barnholtz-Sloan JS. Brain metastases: epidemiology. Amsterdam: Elsevier; 2018.
Preusser M, Capper D, Ilhan-Mutlu A, et al. Brain metastases: pathobiology and emerging targeted therapies. Acta Neuropathol. 2012;123:205–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-011-0933-9.
Berghoff AS, Schur S, Füreder 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/esmoopen-2015-000024.
Franssen LC, Lorenzi T, Burgess AEF, Chaplain MAJ. A mathematical framework for modelling the metastatic spread of cancer. Bull Math Biol. 2019;81:1965–2010. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11538-019-00597-x.
Łazarczyk M, Mickael ME, Skiba D, et al. The journey of cancer cells to the brain: challenges and opportunities. IJMS. 2023;24:3854. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24043854.
Ribatti D, Tamma R, Annese T. Epithelial-mesenchymal transition in cancer: a historical overview. Transl Oncol. 2020;13: 100773. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2020.100773.
Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119:1420–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI39104.
Araki K, Shimura T, Suzuki H, et al. E/N-cadherin switch mediates cancer progression via TGF-β-induced epithelial-to-mesenchymal transition in extrahepatic cholangiocarcinoma. Br J Cancer. 2011;105:1885–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/bjc.2011.452.
Kim WK, Kwon Y, Jang M, et al. β-catenin activation down-regulates cell-cell junction-related genes and induces epithelial-to-mesenchymal transition in colorectal cancers. Sci Rep. 2019;9:18440. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-019-54890-9.
Takaishi M, Tarutani M, Takeda J, Sano S. Mesenchymal to epithelial transition induced by reprogramming factors attenuates the malignancy of cancer cells. PLoS ONE. 2016;11: e0156904. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0156904.
Chao YL, Shepard CR, Wells A. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition. Mol Cancer. 2010;9:179. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1476-4598-9-179.
Das S, Becker BN, Hoffmann FM, Mertz JE. Complete reversal of epithelial to mesenchymal transition requires inhibition of both ZEB expression and the Rho pathway. BMC Cell Biol. 2009;10:94. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2121-10-94.
Palen K, Weber J, Dwinell MB, et al. E-cadherin re-expression shows in vivo evidence for mesenchymal to epithelial transition in clonal metastatic breast tumor cells. Oncotarget. 2016;7:43363–75. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.9715.
Steeg PS, Camphausen KA, Smith QR. Brain metastases as preventive and therapeutic targets. Nat Rev Cancer. 2011;11:352–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrc3053.
Cipolla MJ (2009) Barriers of the CNS. In: The Cerebral Circulation. Morgan & Claypool Life Sciences. NBK53084
Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol. 2015;7: a020412. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a020412.
Daneman R. The blood–brain barrier in health and disease. Ann Neurol. 2012;72:648–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ana.23648.
Abbott NJ, Friedman A. Overview and introduction: the blood–brain barrier in health and disease. Epilepsia. 2012;53:1–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1528-1167.2012.03696.x.
Pandit R, Chen L, Götz J. The blood-brain barrier: physiology and strategies for drug delivery. Adv Drug Deliv Rev. 2020;165–166:1–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2019.11.009.
Hashimoto Y, Campbell M. Tight junction modulation at the blood-brain barrier: current and future perspectives. Biochimica et Biophysica Acta (BBA) - Biomembranes. 2020;1862:183298. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbamem.2020.183298.
Chen Y, Liu L. Modern methods for delivery of drugs across the blood–brain barrier. Adv Drug Deliv Rev. 2012;64:640–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2011.11.010.
Fei Y, Zhu J, Zhao B, et al. XQ-1H regulates Wnt/GSK3β/β-catenin pathway and ameliorates the integrity of blood brain barrier in mice with acute ischemic stroke. Brain Res Bull. 2020;164:269–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainresbull.2020.08.032.
Stamatovic SM, Johnson AM, Keep RF, Andjelkovic AV. Junctional proteins of the blood-brain barrier: new insights into function and dysfunction. Tissue Barriers. 2016;4: e1154641. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/21688370.2016.1154641.
Lajoie JM, Shusta EV. Targeting receptor-mediated transport for delivery of biologics across the blood-brain barrier. Annu Rev Pharmacol Toxicol. 2015;55:613–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-pharmtox-010814-124852.
Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57:178–201. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuron.2008.01.003.
Profaci CP, Munji RN, Pulido RS, Daneman R. The blood–brain barrier in health and disease: important unanswered questions. J Exp Med. 2020;217: e20190062. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20190062.
Cornford EM, Hyman S. Localization of brain endothelial luminal and abluminal transporters with immunogold electron microscopy. Neurotherapeutics. 2005;2:27–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1602/neurorx.2.1.27.
Sweeney MD, Zhao Z, Montagne A, et al. Blood-brain barrier: from physiology to disease and back. Physiol Rev. 2019;99:21–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physrev.00050.2017.
Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12:723–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrn3114.
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS. 2022;19:29. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-022-00312-4.
Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21:193–215. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2011.07.001.
Nakagawa S, Deli MA, Nakao S, Honda M, Hayashi K, Nakaoke R, et al. Pericytes from Brain microvessels strengthen the barrier integrity in primary cultures of rat brain endothelial cells. Cell Mol Neurobiol. 2007;27:687–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10571-007-9195-4.
Chen Y, Li Q, Tang J, Feng H, Zhang JH. The evolving roles of pericyte in early brain injury after subarachnoid hemorrhage. Brain Res. 2015;1623:110.
Chopra N, Menounos S, Choi JP, et al. Blood-spinal cord barrier: its role in spinal disorders and emerging therapeutic strategies. NeuroSci. 2022;3:1–27. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/neurosci3010001.
Echeverry S, Shi XQ, Rivest S, Zhang J. Peripheral nerve injury alters blood-spinal cord barrier functional and molecular integrity through a selective inflammatory pathway. J Neurosci. 2011;31:10819–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.1642-11.2011.
Staines DR, Brenu EW, Marshall-Gradisnik S. Postulated vasoactive neuropeptide immunopathology affecting the blood–brain/blood–spinal barrier in certain neuropsychiatric fatigue-related conditions: a role for phosphodiesterase inhibitors in treatment? Neuropsychiatr Dis Treat. 2009;5:81–9.
Pan W, Zhang L, Liao J, et al. Selective increase in TNFα permeation across the blood–spinal cord barrier after SCI. J Neuroimmunol. 2003;134:111–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0165-5728(02)00426-5.
Rodriguez-Mogeda C, Rodríguez-Lorenzo S, Attia J, et al. Breaching brain barriers: B cell migration in multiple sclerosis. Biomolecules. 2022;12:800. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom12060800.
Kim B, Tabori U, Hawkins C. An update on the CNS manifestations of brain tumor polyposis syndromes. Acta Neuropathol. 2020;139:703–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-020-02124-y.
Gomez-Zepeda D, Taghi M, Scherrmann J-M, et al. ABC transporters at the blood-brain interfaces, their study models, and drug delivery implications in gliomas. Pharmaceutics. 2019;12:E20. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/pharmaceutics12010020.
Ichikawa H, Itoh K. Blood-arachnoid barrier disruption in experimental rat meningitis detected using gadolinium-enhancement ratio imaging. Brain Res. 2011;1390:142–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2011.03.035.
Takeuchi H, Suzuki M, Goto R, et al. Regional differences in the absolute abundance of transporters, receptors and tight junction molecules at the blood-arachnoid barrier and blood-spinal cord barrier among cervical, thoracic and lumbar spines in dogs. Pharm Res. 2022;39:1393–413. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11095-022-03275-1.
Uchida Y, Takeuchi H, Goto R, et al. A human blood-arachnoid barrier atlas of transporters, receptors, enzymes, and tight junction and marker proteins: comparison with dog and pig in absolute abundance. J Neurochem. 2022;161:187–208. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jnc.15599.
Uchida Y, Goto R, Takeuchi H, et al. Abundant expression of OCT2, MATE1, OAT1, OAT3, PEPT2, BCRP, MDR1, and xCT transporters in blood-arachnoid barrier of pig and polarized localizations at CSF- and blood-facing plasma membranes. Drug Metab Dispos. 2020;48:135–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/dmd.119.089516.
Takeuchi H, Suzuki M, Goto R, et al. Regional differences in the absolute abundance of transporters, receptors and tight junction molecules at the blood-arachnoid barrier and blood-spinal cord barrier among cervical, thoracic and lumbar spines in dogs. Pharm Res. 2022;39(7):1393–413. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11095-022-03275-1.
Solár P, Zamani A, Kubíčková L, et al. Choroid plexus and the blood–cerebrospinal fluid barrier in disease. Fluids Barriers CNS. 2020;17:35. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-020-00196-2.
Redzic Z, Segal M. The structure of the choroid plexus and the physiology of the choroid plexus epithelium. Adv Drug Deliv Rev. 2004;56:1695–716. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2004.07.005.
Ewing J. Neoplastic diseases: a treatise on tumors. Am J Med Sci. 1922. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/00000441-192208000-00015.
Liu Q, Zhang H, Jiang X, et al. Factors involved in cancer metastasis: a better understanding to “seed and soil” hypothesis. Mol Cancer. 2017;16:176. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-017-0742-4.
Hüsemann Y, Geigl JB, Schubert F, et al. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13:58–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccr.2007.12.003.
Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Seminars in Cell & Developmental Biol. 2015;40:41–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2015.02.010.
Hornick NI, Huan J, Doron B, et al. Serum exosome MicroRNA as a minimally-invasive early biomarker of AML. Sci Rep. 2015;5:11295. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep11295.
Pisitkun T, Shen R-F, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA. 2004;101:13368–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0403453101.
Zlotogorski-Hurvitz A, Dayan D, Chaushu G, et al. Human saliva-derived exosomes: comparing methods of isolation. J Histochem Cytochem. 2015;63:181–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1369/0022155414564219.
Akers JC, Ramakrishnan V, Kim R, et al. miR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS ONE. 2013;8: e78115. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0078115.
Mallegol J, Van Niel G, Lebreton C, et al. T84-intestinal epithelial exosomes bear MHC Class II/Peptide complexes potentiating antigen presentation by dendritic cells. Gastroenterology. 2007;132:1866–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2007.02.043.
Escola J-M, Kleijmeer MJ, Stoorvogel W, et al. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J Biol Chem. 1998;273:20121–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.273.32.20121.
Hong BS, Cho J-H, Kim H, et al. Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. BMC Genomics. 2009;10:556. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2164-10-556.
Muralidharan-Chari V, Clancy JW, Sedgwick A, D’Souza-Schorey C. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123:1603–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.064386.
Alipoor SD, Mortaz E, Varahram M, et al. The potential biomarkers and immunological effects of tumor-derived exosomes in lung cancer. Front Immunol. 2018;9:819. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.00819.
Janowska-Wieczorek A, Wysoczynski M, Kijowski J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Intl J Cancer. 2005;113:752–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.20657.
Syn N, Wang L, Sethi G, et al. Exosome-mediated metastasis: from epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 2016;37:606–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tips.2016.04.006.
Harris DA, Patel SH, Gucek M, et al. Exosomes released from breast cancer carcinomas stimulate cell movement. PLoS ONE. 2015;10: e0117495. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0117495.
Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17:816–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncb3169.
Li H, Song H, Luo J, et al. Knockdown of glucose-regulated protein 78 decreases the invasion, metalloproteinase expression and ECM degradation in hepatocellular carcinoma cells. J Exp Clin Cancer Res. 2012;31:39. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1756-9966-31-39.
Liang B, Peng P, Chen S, et al. Characterization and proteomic analysis of ovarian cancer-derived exosomes. J Proteomics. 2013;80:171–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jprot.2012.12.029.
Nakamura Y, Suganami A, Fukuda M, et al. Identification of novel candidate compounds targeting TrkB to induce apoptosis in neuroblastoma. Cancer Me. 2014;3:25–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cam4.175.
Lagares-Tena L, García-Monclús S, López-Alemany R, et al. Caveolin-1 promotes Ewing sarcoma metastasis regulating MMP-9 expression through MAPK/ERK pathway. Oncotarget. 2016;7:56889–903. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.10872.
Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–801. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-10-4455.
Zhou W, Fong MY, Min Y, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25:501–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccr.2014.03.007.
Tominaga N, Kosaka N, Ono M, Katsuda T, Yoshioka Y, Tamura K, et al. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood–brain barrier. Nat Commun. 2015;6:6716. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncomms7716.
Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17:183–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncb3094.
Xing F, Liu Y, Wu S-Y, et al. Loss of XIST in breast cancer activates MSN-c-Met and reprograms microglia via exosomal miRNA to promote brain metastasis. Cancer Res. 2018;78:4316–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-18-1102.
Maji S, Chaudhary P, Akopova I, et al. Exosomal annexin II promotes angiogenesis and breast cancer metastasis. Mol Cancer Res. 2017;15:93–105. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1541-7786.MCR-16-0163.
Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev. 2016;99:186–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2015.07.007.
Ringuette Goulet C, Bernard G, Tremblay S, et al. Exosomes induce fibroblast differentiation into cancer-associated fibroblasts through TGFβ signaling. Mol Cancer Res. 2018;16:1196–204. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1541-7786.MCR-17-0784.
Quail DF, Joyce JA. The microenvironmental landscape of brain tumors. Cancer Cell. 2017;31:326–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccell.2017.02.009.
Yuzhalin AE, Yu D. Brain metastasis organotropism. Cold Spring Harb Perspect Med. 2020;10: a037242. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a037242.
Stevens LE, Cheung WKC, Adua SJ, et al. Extracellular matrix receptor expression in subtypes of lung adenocarcinoma potentiates outgrowth of micrometastases. Cancer Res. 2017;77:1905–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-16-1978.
Hoshino A, Costa-Silva B, Shen T-L, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature15756.
Wilhelm I, Molnár J, Fazakas C, et al. Role of the blood-brain barrier in the formation of brain metastases. IJMS. 2013;14:1383–411. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms14011383.
Lowery FJ, Yu D. Brain metastasis: Unique challenges and open opportunities. Biochimica et Biophysica Acta (BBA) - Rev Cancer. 2017;1867:49–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbcan.2016.12.001.
Peters A, Schweiger U, Pellerin L, et al. The selfish brain: competition for energy resources. Neurosci Biobehav Rev. 2004;28:143–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neubiorev.2004.03.002.
Winkler F. The brain metastatic niche. J Mol Med. 2015;93:1213–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00109-015-1357-0.
Brastianos PK, Carter SL, Santagata S, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 2015;5:1164–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.CD-15-0369.
Rangachari D, Yamaguchi N, VanderLaan PA, et al. Brain metastases in patients with EGFR -mutated or ALK -rearranged non-small-cell lung cancers. Lung Cancer. 2015;88:108–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lungcan.2015.01.020.
Takano K, Kinoshita M, Takagaki M, et al. Different spatial distributions of brain metastases from lung cancer by histological subtype and mutation status of epidermal growth factor receptor. Neuro Oncol. 2016;18:716–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuonc/nov266.
Latifkar A, Ling L, Hingorani A, et al. Loss of sirtuin 1 alters the secretome of breast cancer cells by impairing lysosomal integrity. Dev Cell. 2019;49:393-408.e7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2019.03.011.
Ko H, Jeon H, Lee D, et al. Sanguiin H6 suppresses TGF-β induction of the epithelial–mesenchymal transition and inhibits migration and invasion in A549 lung cancer. Bioorg Med Chem Lett. 2015;25:5508–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bmcl.2015.10.067.
Kosaka N, Iguchi H, Yoshioka Y, et al. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285:17442–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M110.107821.
Huang S, Li Y, Zhang J, et al. Epidermal growth factor receptor-containing exosomes induce tumor-specific regulatory T cells. Cancer Invest. 2013;31:330–5. https://doiorg.publicaciones.saludcastillayleon.es/10.3109/07357907.2013.789905.
Guo D, Lui GYL, Lai SL, et al. RAB27A promotes melanoma cell invasion and metastasis via regulation of pro-invasive exosomes. Intl J Cancer. 2019;144:3070–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.32064.
Zhao L, Ma X, Yu J. Exosomes and organ-specific metastasis. Mol Therapy - Methods Clin Dev. 2021;22:133–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.omtm.2021.05.016.
Doron H, Pukrop T, Erez N. A blazing landscape: neuroinflammation shapes brain metastasis. Cancer Res. 2019;79:423–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-18-1805.
Schwartz H, Blacher E, Amer M, et al. Incipient melanoma brain metastases instigate astrogliosis and neuroinflammation. Cancer Res. 2016;76:4359–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-16-0485.
El-Kenawi A, Hänggi K, Ruffell B. The immune microenvironment and cancer metastasis. Cold Spring Harb Perspect Med. 2020;10: a037424. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a037424.
Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21:938–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.3909.
Meylan E, Dooley AL, Feldser DM, et al. Requirement for NF-κB signaling in a mouse model of lung adenocarcinoma. Nature. 2009;462:104–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature08462.
Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018;19:108–19. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41590-017-0022-x.
DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19:369–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-019-0127-6.
Bonde A-K, Tischler V, Kumar S, et al. Intratumoral macrophages contribute to epithelial-mesenchymal transition in solid tumors. BMC Cancer. 2012;12:35. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2407-12-35.
Lee C-C, Lin J-C, Hwang W-L, et al. Macrophage-secreted interleukin-35 regulates cancer cell plasticity to facilitate metastatic colonization. Nat Commun. 2018;9:3763. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-018-06268-0.
Smyth MJ, Thia KY, Cretney E, et al. Perforin is a major contributor to NK cell control of tumor metastasis. J Immunol. 1999;162:6658–62.
López-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK Cells. Cancer Cell. 2017;32:135–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccell.2017.06.009.
Cairns RA, Papandreou I, Sutphin PD, Denko NC. Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy. Proc Natl Acad Sci USA. 2007;104:9445–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0611662104.
Doedens AL, Stockmann C, Rubinstein MP, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70:7465–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-10-1439.
Hosonaga M, Saya H, Arima Y. Molecular and cellular mechanisms underlying brain metastasis of breast cancer. Cancer Metastasis Rev. 2020;39:711–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10555-020-09881-y.
Colegio OR, Chu N-Q, Szabo AL, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature13490.
Dediu M, Ion O, Ion R, et al. Impact of adjuvant chemotherapy in stage IB non-small-cell lung cancer: an analysis of 112 consecutively treated patients. J BUON. 2012;17:317–22.
Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553:446–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature25183.
Thai AA, Solomon BJ, Sequist LV, et al. Lung cancer. The Lancet. 2021;398:535–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(21)00312-3.
Schouten LJ, Rutten J, Huveneers HAM, Twijnstra A. Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma. Cancer. 2002;94:2698–705. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.10541.
Zhu Y, Cui Y, Zheng X, et al. Small-cell lung cancer brain metastasis: From molecular mechanisms to diagnosis and treatment. Biochimica et Biophysica Acta (BBA) Molecular Basis of Disease. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbadis.2022.166557.
Burel-Vandenbos F, Ambrosetti D, Coutts M, Pedeutour F. EGFR mutation status in brain metastases of non-small cell lung carcinoma. J Neurooncol. 2013;111:1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11060-012-0990-5.
Cox JD, Scott CB, Byhardt RW, et al. Addition of chemotherapy to radiation therapy alters failure patterns by cell type within non-small cell carcinoma of lung (NSCCL): analysis of radiation therapy oncology group (RTOG) trials. Int J Radiat Oncol Biol Phys. 1999;43:505–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0360-3016(98)00429-5.
Mogi A, Kuwano H. TP53 mutations in non-small cell lung cancer. J Biomed Biotechnol. 2011;2011:1–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2011/583929.
Baumann M, Zips D, Appold S. Radiotherapy of lung cancer: Technology meets biology meets multidisciplinarity. Radiother Oncol. 2009;91:279–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.radonc.2009.05.001.
Reichel MB, Ohgaki H, Petersen I, Kleihues P. p53 mutations in primary human lung tumors and their metastases. Mol Carcinog. 1994;9:105–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mc.2940090208.
Sozzi G, Miozzo M, Pastorino U, et al. Genetic evidence for an independent origin of multiple preneoplastic and neoplastic lung lesions. Cancer Res. 1995;55:135–40.
Dearden S, Stevens J, Wu Y-L, Blowers D. Mutation incidence and coincidence in non-small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Ann Oncol. 2013;24:2371–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/annonc/mdt205.
Cortot AB, Italiano A, Burel-Vandenbos F, et al. KRAS mutation status in primary non-small cell lung cancer and matched metastases. Cancer. 2010;116:2682–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.25014.
Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol. 2004;59:S21–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijrobp.2003.11.041.
Sun M, Behrens C, Feng L, et al. HER family receptor abnormalities in lung cancer brain metastases and corresponding primary tumors. Clin Cancer Res. 2009;15:4829–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-08-2921.
Seshacharyulu P, Ponnusamy MP, Haridas D, et al. Targeting the EGFR signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012;16:15–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1517/14728222.2011.648617.
Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, et al. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med. 2020;18:1. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-019-02189-8.
Carpenter G, Cohen S. Epidermal growth factor. Annu Rev Biochem. 1979;48:193–216. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.bi.48.070179.001205.
Porta R, Sanchez-Torres JM, Paz-Ares L, et al. Brain metastases from lung cancer responding to erlotinib: the importance of EGFR mutation. Eur Respir J. 2011;37:624–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/09031936.00195609.
Deng Z, Cui L, Li P, et al. Genomic comparison between cerebrospinal fluid and primary tumor revealed the genetic events associated with brain metastasis in lung adenocarcinoma. Cell Death Dis. 2021;12:935. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-04223-4.
Eichler AF, Kahle KT, Wang DL, et al. EGFR mutation status and survival after diagnosis of brain metastasis in non-small cell lung cancer. Neuro Oncol. 2010;12:1193–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuonc/noq076.
Whitsett TG, Inge LJ, Dhruv HD, et al. Molecular determinants of lung cancer metastasis to the central nervous system. Transl Lung Cancer Res. 2013;2:273–83. https://doiorg.publicaciones.saludcastillayleon.es/10.3978/j.issn.2218-6751.2013.03.12.
Ge M, Zhuang Y, Zhou X, et al. High probability and frequency of EGFR mutations in non-small cell lung cancer with brain metastases. J Neurooncol. 2017;135:413–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11060-017-2590-x.
Kong C, Zhou D, Wu N, Bai C. Multiple intraventricular metastases from lung adenocarcinoma with EGFR G719X mutation: a case report. BMC Pulm Med. 2020;20:135. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12890-020-1168-0.
Jancík S, Drábek J, Radzioch D, Hajdúch M. Clinical relevance of KRAS in human cancers. J Biomed Biotechnol. 2010;2010: 150960. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2010/150960.
Rosen N. Finally, effective inhibitors of mutant KRAS. N Engl J Med. 2021;384:2447–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMe2107884.
Salgia R, Pharaon R, Mambetsariev I, et al. The improbable targeted therapy: KRAS as an emerging target in non-small cell lung cancer (NSCLC). Cell Rep Med. 2021;2: 100186. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xcrm.2020.100186.
Tomasini P, Serdjebi C, Khobta N, et al. EGFR and KRAS mutations predict the incidence and outcome of brain metastases in non-small cell lung cancer. IJMS. 2016;17:2132. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms17122132.
Wang SE, Narasanna A, Perez-Torres M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell. 2006;10:25–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccr.2006.05.023.
Li BT, Lee A, O’Toole S, et al. HER2 insertion YVMA mutant lung cancer: Long natural history and response to afatinib. Lung Cancer. 2015;90:617–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lungcan.2015.10.025.
Stephens P, Hunter C, Bignell G, et al. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature. 2004;431(7008):525–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/431525b.
Lin JJ, Gainor JF. Time to tackle the blood-brain barrier in HER2 -mutant lung cancer. Cancer. 2019;125:4363–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.32460.
Yang S, Wang Y, Zhao C, et al. Exon 20 YVMA insertion is associated with high incidence of brain metastasis and inferior outcome of chemotherapy in advanced non-small cell lung cancer patients with HER2 kinase domain mutations. Transl Lung Cancer Res. 2021;10:753–65. https://doiorg.publicaciones.saludcastillayleon.es/10.21037/tlcr-20-559.
Fares J, Cordero A, Kanojia D, Lesniak MS. The network of cytokines in brain metastases. Cancers. 2021;13:142. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13010142.
Khan GJ, Sun L, Abbas M, et al. In-vitro pre-treatment of cancer cells with TGF-β1: a novel approach of tail vein lung cancer metastasis mouse model for anti-metastatic studies. CMP. 2019;12:249–60. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1874467212666190306165703.
Yuan X, Wei Q, Komaki R, et al. TGFβ1 polymorphisms predict distant metastasis-free survival in patients with inoperable non-small-cell lung cancer after definitive radiotherapy. PLoS ONE. 2013;8: e65659. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0065659.
Benveniste H, Zhang S, Reinsel RA, et al. Brain metabolomic profiles of lung cancer patients prior to treatment characterized by proton magnetic resonance spectroscopy. Int J Clin Exp Med. 2012;5:154–64.
Domanska UM, Kruizinga RC, Nagengast WB, et al. A review on CXCR4/CXCL12 axis in oncology: No place to hide. Eur J Cancer. 2013;49:219–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejca.2012.05.005.
Chen G, Wang Z, Liu X, Liu F. High-level CXCR4 expression correlates with brain-specific metastasis of non-small cell lung cancer. World J Surg. 2011;35:56–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00268-010-0784-x.
Paratore S, Banna GL, D’Arrigo M, et al. CXCR4 and CXCL12 immunoreactivities differentiate primary non-small-cell lung cancer with or without brain metastases. CBM. 2012;10:79–89. https://doiorg.publicaciones.saludcastillayleon.es/10.3233/CBM-2011-0232.
Mauri FA, Pinato DJ, Trivedi P, et al. Isogeneic comparison of primary and metastatic lung cancer identifies CX3CR1 as a molecular determinant of site-specific metastatic diffusion. Oncol Rep. 2012;28:647–53. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/or.2012.1818.
Li YD, Lamano JB, Lamano JB, et al. Tumor-induced peripheral immunosuppression promotes brain metastasis in patients with non-small cell lung cancer. Cancer Immunol Immunother. 2019;68:1501–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00262-019-02384-y.
Lamano JB, Lamano JB, Li YD, et al. Glioblastoma-derived IL6 induces immunosuppressive peripheral myeloid cell PD-L1 and promotes tumor growth. Clin Cancer Res. 2019;25:3643–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-18-2402.
Li H, Zhao Q, Chang L, et al. LncRNA MALAT1 modulates ox-LDL induced EndMT through the Wnt/β-catenin signaling pathway. Lipids Health Dis. 2019;18:62. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12944-019-1006-7.
Schmidt LH, Spieker T, Koschmieder S, et al. The long non-coding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. J Thorac Oncol. 2011;6:1984–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/JTO.0b013e3182307eac.
Karimpour M, Ravanbakhsh R, Maydanchi M, et al. Cancer driver gene and non-coding RNA alterations as biomarkers of brain metastasis in lung cancer: a review of the literature. Biomed Pharmacother. 2021;143: 112190. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2021.112190.
Preusser M, Berghoff AS, Berger W, et al. High rate of FGFR1 amplifications in brain metastases of squamous and non-squamous lung cancer. Lung Cancer. 2014;83:83–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lungcan.2013.10.004.
Kim J, Piao H-L, Kim B-J, et al. Long non-coding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet. 2018;50:1705–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41588-018-0252-3.
Grinberg-Rashi H, Ofek E, Perelman M, et al. The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin Cancer Res. 2009;15:1755–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-08-2124.
Mrozik KM, Blaschuk OW, Cheong CM, et al. N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer. 2018;18:939. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-018-4845-0.
Yoo JY, Yang S-H, Lee JE, et al. E-cadherin as a predictive marker of brain metastasis in non-small-cell lung cancer, and its regulation by pioglitazone in a preclinical model. J Neurooncol. 2012;109:219–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11060-012-0890-8.
Dai L, Zhao J, Yin J, et al. Cell adhesion molecule 2 (CADM2) promotes brain metastasis by inducing epithelial-mesenchymal transition (EMT) in human non-small cell lung cancer. Ann Transl Med. 2020;8:465–465. https://doiorg.publicaciones.saludcastillayleon.es/10.21037/atm.2020.03.85.
Jassam SA, Maherally Z, Smith JR, et al. TNF-α enhancement of CD62E mediates adhesion of non-small cell lung cancer cells to brain endothelium via CD15 in lung-brain metastasis. Neuro Oncol. 2016;18:679–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuonc/nov248.
Rai S, Nejadhamzeeigilani Z, Gutowski NJ, Whatmore JL. Loss of the endothelial glycocalyx is associated with increased E-selectin mediated adhesion of lung tumour cells to the brain microvascular endothelium. J Exp Clin Cancer Res. 2015;34:105. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13046-015-0223-9.
Kargi HA, Kuyucuoğlu MF, Alakavuklar M, et al. CD44 expression in metastatic and non-metastatic non-small cell lung cancers. Cancer Lett. 1997;119:27–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0304-3835(97)00254-1.
Morath I, Hartmann TN, Orian-Rousseau V. CD44: More than a mere stem cell marker. Int J Biochem Cell Biol. 2016;81:166–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biocel.2016.09.009.
Wang Y-Y, Vadhan A, Chen P-H, et al. CD44 promotes lung cancer cell metastasis through ERK–ZEB1 signaling. Cancers. 2021;13:4057. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13164057.
Wu D, Deng S, Li L, Liu T, Zhang T, Li J, et al. TGF-β1-mediated exosomal lnc-MMP2-2 increases blood–brain barrier permeability via the miRNA-1207-5p/EPB41L5 axis to promote non-small cell lung cancer brain metastasis. Cell Death Dis. 2021;12:1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-04004-z.
Takakuwa Y. Protein 4.1, a multifunctional protein of the erythrocyte membrane skeleton: structure and functions in erythrocytes and nonerythroid cells. Int J Hematol. 2000;72(3):298–309.
Hudson K, Mondia MW, Zhang Y, Saha S, Gibert MK, Dube C, et al. The role of microRNAs in brain metastasis. J Neurooncol. 2024;166:231–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11060-023-04541-x.
Hansen CG, Moroishi T, Guan K-L. YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol. 2015;25:499–513. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2015.05.002.
Li C, Zheng H, Xiong J, Huang Y, Li H, Jin H, et al. miR-596-3p suppresses brain metastasis of non-small cell lung cancer by modulating YAP1 and IL-8. Cell Death Dis. 2022;13:699. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-022-05062-7.
Lou Y, Jiang Y, Liang Z, Liu B, Li T, Zhang D. Role of RhoC in cancer cell migration. Cancer Cell Int. 2021;21:527. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-021-02234-x.
Łukasiewicz S, Czeczelewski M, Forma A, et al. Breast cancer-epidemiology, risk factors, classification, prognostic markers, and current treatment strategies-an updated review. Cancers (Basel). 2021;13:4287. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13174287.
Inic Z, Zegarac M, Inic M, et al. Difference between luminal a and luminal b subtypes according to Ki-67, tumor size, and progesterone receptor negativity providing prognostic information. Clin Med Insights Oncol. 2014;8:107–11. https://doiorg.publicaciones.saludcastillayleon.es/10.4137/CMO.S18006.
Pedrosa RMSM, Mustafa DA, Soffietti R, Kros JM. Breast cancer brain metastasis: molecular mechanisms and directions for treatment. Neuro Oncol. 2018;20:1439–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuonc/noy044.
Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (her2) in cancers: overexpression and therapeutic implications. Mol Biol Int. 2014;2014:1–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2014/852748.
Lo H-W. EGFR and HER2 signaling in breast cancer brain metastasis. Front Biosci. 2016;8:245–63. https://doiorg.publicaciones.saludcastillayleon.es/10.2741/e765.
Xie T, Huang F-J, Aldape KD, et al. Activation of stat3 in human melanoma promotes brain metastasis. Cancer Res. 2006;66:3188–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-05-2674.
McFarland BC, Benveniste EN. Reactive astrocytes foster brain metastases via STAT3 signaling. Ann Transl Med. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.21037/atm.2019.04.17.
Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer — using tissue repair as a road map. Nat Rev Cancer. 2019;19:82–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41568-018-0090-8.
Tolomeo M, Cascio A. The multifaced role of STAT3 in cancer and its implication for anticancer therapy. IJMS. 2021;22:603. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22020603.
Jablonska PA, Galán N, Barranco J, et al. Presence of activated (Phosphorylated) STAT3 in radiation necrosis following stereotactic radiosurgery for brain metastases. IJMS. 2023;24:14219. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms241814219.
Carney CP, Pandey N, Kapur A, et al. Harnessing nanomedicine for enhanced immunotherapy for breast cancer brain metastases. Drug Deliv and Transl Res. 2021;11:2344–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13346-021-01039-9.
Chen Q, Boire A, Jin X, et al. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533:493–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature18268.
Khoo LT, Chen L. Role of the cGAS–STING pathway in cancer development and oncotherapeutic approaches. EMBO Rep. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.15252/embr.201846935.
Wang Y, Ye F, Liang Y, Yang Q. Breast cancer brain metastasis: insight into molecular mechanisms and therapeutic strategies. Br J Cancer. 2021;125:1056–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41416-021-01424-8.
Huang Y-S, Chang C-C, Lee S-S, et al. Xist reduction in breast cancer upregulates AKT phosphorylation via HDAC3-mediated repression of PHLPP1 expression. Oncotarget. 2016;7:43256–66. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.9673.
Bryan S, Witzel I, Borgmann K, Oliveira-Ferrer L. Molecular mechanisms associated with brain metastases in HER2-positive and triple negative breast cancers. Cancers (Basel). 2021;13:4137. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13164137.
Organ SL, Tsao M-S. An overview of the c-MET signaling pathway. Ther Adv Med Oncol. 2011;3:S7–19. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1758834011422556.
Tu S, Lin X, Qiu J, et al. Crosstalk between tumor-associated microglia/macrophages and CD8-positive T cells plays a key role in glioblastoma. Front Immunol. 2021;12: 650105. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.650105.
Jiang N, Dai Q, Su X, et al. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Mol Biol Rep. 2020;47:4587–629. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11033-020-05435-1.
Kang S-A, Blache CA, Bajana S, et al. The effect of soluble E-selectin on tumor progression and metastasis. BMC Cancer. 2016;16:331. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-016-2366-2.
Geng Y, Yeh K, Takatani T, King MR. Three to tango: MUC1 as a ligand for both E-selectin and ICAM-1 in the breast cancer metastatic cascade. Front Oncol. 2012;2:76. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2012.00076.
Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/35052073.
Nahta R, Esteva FJ. HER2 therapy: molecular mechanisms of trastuzumab resistance. Breast Cancer Res. 2006;8:215. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/bcr1612.
Korkaya H, Kim G-I, Davis A, Malik F, Henry NL, Ithimakin S, et al. Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population. Mol Cell. 2012;47:570–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molcel.2012.06.014.
Osanai M, Murata M, Nishikiori N, Chiba H, Kojima T, Sawada N. Occludin-mediated premature senescence is a fail-safe mechanism against tumorigenesis in breast carcinoma cells. Cancer Sci. 2007;98:1027–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1349-7006.2007.00494.x.
Hoevel T, Macek R, Swisshelm K, Kubbies M. Reexpression of the TJ protein CLDN1 induces apoptosis in breast tumor spheroids. Int J Cancer. 2004;108:374–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.11571.
Hsu JC-C, Reid DW, Hoffman AM, et al. Oncoprotein AEG-1 is an endoplasmic reticulum RNA-binding protein whose interactome is enriched in organelle resident protein-encoding mRNAs. RNA. 2018;24:688–703. https://doiorg.publicaciones.saludcastillayleon.es/10.1261/rna.063313.117.
Taiarol L, Formicola B, Magro RD, et al. An update of nanoparticle-based approaches for glioblastoma multiforme immunotherapy. Nanomedicine. 2020;15:1861–71. https://doiorg.publicaciones.saludcastillayleon.es/10.2217/nnm-2020-0132.
Wu K, Fukuda K, Xing F, et al. Roles of the cyclooxygenase 2 matrix metalloproteinase 1 pathway in brain metastasis of breast cancer. J Biol Chem. 2015;290:9842–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M114.602185.
Pfeiffer F, Schäfer J, Lyck R, et al. Claudin-1 induced sealing of blood-brain barrier tight junctions ameliorates chronic experimental autoimmune encephalomyelitis. Acta Neuropathol. 2011;122:601–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-011-0883-2.
Janavicius M, Lachej N, Anglickiene G, et al. Outcomes of treatment for melanoma brain metastases. J Skin Cancer. 2020;2020:1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2020/7520924.
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA A Cancer J Clinicians. 2017;67:7–30. https://doiorg.publicaciones.saludcastillayleon.es/10.3322/caac.21387.
Thomas DGT, Graham DI. Malignant brain tumours. London, London: Springer; 1995.
Madajewicz S, Karakousis C, West CR, et al. Malignant melanoma brain metastases. review of roswell park memorial institute experience. Cancer. 1984;53:2550–2. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/1097-0142(19840601)53:11%3c2550::AID-CNCR2820531129%3e3.0.CO;2-B.
Metzemaekers M, Vanheule V, Janssens R, et al. Overview of the mechanisms that may contribute to the non-redundant activities of interferon-inducible CXC chemokine receptor 3 ligands. Front Immunol. 2018;8:1970. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2017.01970.
Jiang H, Gebhardt C, Umansky L, et al. Elevated chronic inflammatory factors and myeloid-derived suppressor cells indicate poor prognosis in advanced melanoma patients. Intl J Cancer. 2015;136:2352–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.29297.
Wightman SC, Uppal A, Pitroda SP, et al. Oncogenic CXCL10 signaling drives metastasis development and poor clinical outcome. Br J Cancer. 2015;113:327–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/bjc.2015.193.
Doron H, Amer M, Ershaid N, et al. Inflammatory activation of astrocytes facilitates melanoma brain tropism via the CXCL10-CXCR3 signaling axis. Cell Rep. 2019;28:1785-1798.e6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2019.07.033.
Lok E, Chung AS, Swanson KD, Wong ET. Melanoma brain metastasis globally reconfigures chemokine and cytokine profiles in patient cerebrospinal fluid. Melanoma Res. 2014;24:120–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/CMR.0000000000000045.
Hofmann UB, Westphal JR, Waas ET, et al. Matrix metalloproteinases in human melanoma cell lines and xenografts: increased expression of activated matrix metalloproteinase-2 (MMP-2) correlates with melanoma progression. Br J Cancer. 1999;81:774–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.bjc.6690763.
Othus M, Moon J, Margolin K. Melanoma’s deadly march to the brain: By what route, and can it be stopped? Cancer. 2011;117:1560–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.25716.
Huang F-J, Steeg PS, Price JE, et al. Molecular basis for the critical role of suppressor of cytokine signaling-1 in melanoma brain metastasis. Cancer Res. 2008;68:9634–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-08-1429.
Patel JK, Didolkar MS, Pickren JW, Moore RH. Metastatic pattern of malignant melanoma. Am J Surg. 1978;135:807–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0002-9610(78)90171-X.
Klein A, Schwartz H, Sagi-Assif O, et al. Astrocytes facilitate melanoma brain metastasis via secretion of IL -23. J Pathol. 2015;236:116–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.4509.
Hendrych M, Solar P, Hermanova M, et al. Spinal metastasis in a patient with supratentorial glioblastoma with primitive neuronal component: a case report with clinical and molecular evaluation. Diagnostics (Basel). 2023;13:181. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/diagnostics13020181.
Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97–109. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-007-0243-4.
Solar P, Valekova H, Marcon P, et al. Classification of brain lesions using a machine learning approach with cross-sectional ADC value dynamics. Sci Rep. 2023;13:11459. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-38542-7.
Papadopoulos MC, Saadoun S, Binder DK, et al. Molecular mechanisms of brain tumor edema. Neurosci. 2004;129:1009–18. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2004.05.044.
Fazakas C, Wilhelm I, Nagyőszi P, et al. Transmigration of melanoma cells through the blood-brain barrier: role of endothelial tight junctions and melanoma-released serine proteases. PLoS ONE. 2011;6: e20758. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0020758.
Guo Q, Zhu Q, Miao T, et al. LRP1-upregulated nanoparticles for efficiently conquering the blood-brain barrier and targeted suppressing multifocal and infiltrative brain metastases. J Control Release. 2019;303:117–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jconrel.2019.04.031.
Gondi V, Tomé WA, Mehta MP. Why avoid the hippocampus? a comprehensive review. Radiothern Oncol. 2010;97:370–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.radonc.2010.09.013.
Brown PD, Gondi V, Pugh S, et al. Hippocampal avoidance during whole-brain radiotherapy plus memantine for patients with brain metastases: phase III trial NRG oncology CC001. JCO. 2020;38:1019–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.19.02767.
Hoffmann M, Hayes MR, Pietruszka J, Elling L. Synthesis of the Thomsen-Friedenreich-antigen (TF-antigen) and binding of Galectin-3 to TF-antigen presenting neo-glycoproteins. Glycoconj J. 2020;37:457–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10719-020-09926-y.
Strell C, Entschladen F. Extravasation of leukocytes in comparison to tumor cells. Cell Commun Signal. 2008;6:10. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1478-811X-6-10.
Strell C, Lang K, Niggemann B, et al. Surface molecules regulating rolling and adhesion to the endothelium of neutrophil granulocytes and MDA-MB-468 breast carcinoma cells and their interaction. Cell Mol Life Sci. 2007;64:3306–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-007-7402-6.
Westphal D, Glitza Oliva IC, Niessner H. Molecular insights into melanoma brain metastases. Cancer. 2017;123:2163–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.30594.
Perides G, Zhuge Y, Lin T, et al. The fibrinolytic system facilitates tumor cell migration across the blood-brain barrier in experimental melanoma brain metastasis. BMC Cancer. 2006;6:56. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2407-6-56.
Artym VV. Molecular proximity of seprase and the urokinase-type plasminogen activator receptor on malignant melanoma cell membranes: dependence on beta1 integrins and the cytoskeleton. Carcinogenesis. 2002;23:1593–602. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/carcin/23.10.1593.
Piñeiro-Sánchez ML, Goldstein LA, Dodt J, et al. Identification of the 170-kDa melanoma membrane-bound gelatinase (Seprase) as a serine integral membrane protease. J Biol Chem. 1997;272:7595–601. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.272.12.7595.
Shintani Y, Higashiyama S, Ohta M, et al. Overexpression of ADAM9 in non-small cell lung cancer correlates with brain metastasis. Cancer Res. 2004;64:4190–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-03-3235.
Wrobel JK, Toborek M. Blood-brain barrier remodeling during brain metastasis formation. Mol Med. 2016;22:32–40. https://doiorg.publicaciones.saludcastillayleon.es/10.2119/molmed.2015.00207.
Meissauer A, Kramer MD, Schirrmacher V, Brunner G. Generation of cell surface-bound plasmin by cell-associated urokinase-type or secreted tissue-type plasminogen activator: a key event in melanoma cell invasiveness in vitro. Exp Cell Res. 1992;199:179–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0014-4827(92)90423-6.
Suryo Rahmanto Y, Dunn LL, Richardson DR. The melanoma tumor antigen, melanotransferrin (p97): a 25-year hallmark – from iron metabolism to tumorigenesis. Oncogene. 2007;26:6113–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.onc.1210442.
Rolland Y, Demeule M, Fenart L, Béliveau R. Inhibition of melanoma brain metastasis by targeting melanotransferrin at the cell surface. Pigment Cell Melanoma Res. 2009;22:86–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1755-148X.2008.00525.x.
Monsky WL, Lin CY, Aoyama A, et al. A potential marker protease of invasiveness, seprase, is localized on invadopodia of human malignant melanoma cells. Cancer Res. 1994;54:5702–10.
O’Brien P, O’Connor BF. Seprase: An overview of an important matrix serine protease. Biochim Biophys Acta Proteins Proteom. 2008;1784:1130–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbapap.2008.01.006.
Denkins Y, Reiland J, Roy M, et al. Brain metastases in melanoma: roles of neurotrophins. Neuro Oncol. 2004;6:154–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1215/S115285170300067X.
Wang L, Cossette SM, Rarick KR, et al. Astrocytes directly influence tumor cell invasion and metastasis in vivo. PLoS ONE. 2013;8: e80933. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0080933.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.2353/ajpath.2010.090838.
Bos PD, Zhang XH-F, Nadal C, et al. Genes that mediate breast cancer metastasis to the brain. Nature. 2009;459:1005–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature08021.
Harati R, Mohammad MG, Tlili A, et al. Loss of miR-101-3p promotes transmigration of metastatic breast cancer cells through the brain endothelium by inducing COX-2/MMP1 signaling. Pharmaceuticals. 2020;13:144. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph13070144.
Nicolson GL, Menter DG. Trophic factors and central nervous system metastasis. Cancer Metast Rev. 1995;14:303–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/BF00690600.
Menter DG, Herrmann JL, Marchetti D, Nicolson GL. Involvement of neurotrophins and growth factors in brain metastasis formation. Invas Metastasis. 1994;14:372–84.
Sirkisoon SR, Carpenter RL, Rimkus T, et al. EGFR and HER2 signaling in breast cancer brain metastasis. Front Biosci (Elite Ed). 2016;8:245–63. https://doiorg.publicaciones.saludcastillayleon.es/10.2741/E765.
Lee KY, Kim Y-J, Yoo H, et al. Human brain endothelial cell-derived COX-2 facilitates extravasation of breast cancer cells across the blood-brain barrier. Anticancer Res. 2011;31:4307–13.
Avraham HK, Jiang S, Fu Y, et al. Angiopoietin-2 mediates blood-brain barrier impairment and colonization of triple-negative breast cancer cells in brain: Ang-2 mediates breast cancer colonization in brain. J Pathol. 2014;232:369–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.4304.
Cheng X, Hung M-C. Breast cancer brain metastases. Cancer Metastasis Rev. 2007;26:635–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10555-007-9083-x.
Karin N. The multiple faces of CXCL12 (SDF-1α) in the regulation of immunity during health and disease. J Leukoc Biol. 2010;88:463–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1189/jlb.0909602.
Curtaz CJ, Schmitt C, Herbert S-L, et al. Serum-derived factors of breast cancer patients with brain metastases alter the permeability of a human blood-brain barrier model. Fluids Barriers CNS. 2020;17:31. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-020-00192-6.
Momeny M, Saunus JM, Marturana F, et al. Heregulin-HER3-HER2 signaling promotes matrix metalloproteinase-dependent blood-brain-barrier transendothelial migration of human breast cancer cell lines. Oncotarget. 2015;6:3932–46. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.2846.
Kienast Y, Von Baumgarten L, Fuhrmann M, et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16:116–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.2072.
Jilaveanu LB, Parisi F, Barr ML, et al. PLEKHA5 as a biomarker and potential mediator of melanoma brain metastasis. Clin Cancer Res. 2015;21:2138–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-14-0861.
Stoletov K, Strnadel J, Zardouzian E, et al. Role of connexins in metastatic breast cancer and melanoma brain colonization. J Cell Sci. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.112748.
Sisó S, Jeffrey M, González L. Sensory circumventricular organs in health and disease. Acta Neuropathol. 2010;120:689–705. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-010-0743-5.
Castaldo JE, Bernat JL, Meier FA, Schned AR. Intracranial metastases due to prostatic carcinoma. Cancer. 1983;52:1739–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/1097-0142(19831101)52:9%3c1739::aid-cncr2820520931%3e3.0.co;2-c.
Filippidis AS, Zarogiannis SG, Ioannou M, et al. Permeability of the arachnoid and pia mater. The role of ion channels in the leptomeningeal physiology. Childs Nerv Syst. 2012;28:533–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00381-012-1688-x.
Pollock H, Hutchings M, Weller RO, Zhang E-T. Perivascular spaces in the basal ganglia of the human brain: their relationship to lacunes. J Anat. 1997;191:337–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1469-7580.1997.19130337.x.
Pedersen P-H, Rucklidge GJ, Mork SJ, et al. Leptomeningeal tissue: a barrier against brain tumor cell invasion. JNCI. 1994;86:1593–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jnci/86.21.1593.
Saito N, Hatori T, Murata N, et al. Comparison of metastatic brain tumour models using three different methods: the morphological role of the pia mater. Int J Experimental Path. 2008;89:38–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2613.2007.00563.x.
Simonsen TG, Gaustad J-V, Rofstad EK. Intracranial tumor cell migration and the development of multiple brain metastases in malignant melanoma. Transl Oncol. 2016;9:211–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2016.04.003.
Yao H, Price TT, Cantelli G, et al. Leukaemia hijacks a neural mechanism to invade the central nervous system. Nature. 2018;560:55–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-018-0342-5.
Mastorakos P, McGavern D. The anatomy and immunology of vasculature in the central nervous system. Sci Immunol. 2019;4:0492. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciimmunol.aav0492.
Jain RW, Yong VW. B cells in central nervous system disease: diversity, locations and pathophysiology. Nat Rev Immunol. 2022;22:513–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-021-00652-6.
Cozzi FM, Ampie L, Laws MT, Brown DA. The role of the dura mater in cerebral metastases. Neurosurg Focus. 2023;55:E17. https://doiorg.publicaciones.saludcastillayleon.es/10.3171/2023.5.FOCUS23229.
Horsburgh A, Massoud TF. The circumventricular organs of the brain: conspicuity on clinical 3T MRI and a review of functional anatomy. Surg Radiol Anat. 2013;35:343–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00276-012-1048-2.
Verheggen ICM, De Jong JJ, Van Boxtel MPJ, et al. Permeability of the windows of the brain: feasibility of dynamic contrast-enhanced MRI of the circumventricular organs. Fluids Barriers CNS. 2020;17:66. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-020-00228-x.
Ebling FJP. Tanycytes: Gateway to the hungry brain: An Editorial for "The FGF2-induced tanycyte proliferation involves a connexin 43 hemichannel/purinergic-dependent pathway” on page 182. J Neurochem. 2021;156:143–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jnc.15218.
Recabal A, Fernández P, López S, et al. The FGF2-induced tanycyte proliferation involves a connexin 43 hemichannel/purinergic-dependent pathway. J Neurochem. 2021;156:182–99. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jnc.15188.
Lassman AB, Bruce JN, Fetell MR. Metastases to the pineal gland. Neurology. 2006;67:1303–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1212/01.wnl.0000238516.29603.33.
Fang AS, Meyers SP. Magnetic resonance imaging of pineal region tumours. Insights Imaging. 2013;4:369–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13244-013-0248-6.
Wendel C, Kaech DL, Woodtli M. Primary malignant melanoma in the pineal region: case report and literature review. J Neurol Surg A Cent Eur Neurosurg. 2018;79(4):344–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1055/s-0038-1639504.
Favero G, Bonomini F, Rezzani R. Pineal gland tumors: a review. Cancers. 2021;13:1547. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13071547.
Williams MD, Asa SL, Fuller GN. Medullary thyroid carcinoma metastatic to the pituitary gland: an unusual site of metastasis. Ann Diagn Pathol. 2008;12:199–203. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.anndiagpath.2006.08.007.
Lamorie-Foote K, Rangwala SD, Kammen A, et al. Melanoma metastasis to a nonfunctioning pituitary macroadenoma: illustrative case. J Neurosurg Case Lessons. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3171/CASE2167.
He W, Chen F, Dalm B, et al. Metastatic involvement of the pituitary gland: a systematic review with pooled individual patient data analysis. Pituitary. 2015;18:159–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11102-014-0552-2.
Hong S, Atkinson JL, Erickson D, et al. Treatment outcome of metastasis to the pituitary gland: a case series of 21 patients with pathological diagnosis. Neurosurg Focus. 2023;55:E13. https://doiorg.publicaciones.saludcastillayleon.es/10.3171/2023.5.FOCUS23185.
Fassett DR, Couldwell WT. Metastases to the pituitary gland. FOC. 2004;16:1–4. https://doiorg.publicaciones.saludcastillayleon.es/10.3171/foc.2004.16.4.9.
Janssen S, Mehta P, Bartscht T, et al. Prevalence of metastases within the hypothalamic-pituitary area in patients with brain metastases. Radiat Oncol. 2019;14:152. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13014-019-1337-6.
Nayak L, Lee EQ, Wen PY. Epidemiology of brain metastases. Curr Oncol Rep. 2012;14:48–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11912-011-0203-y.
Siomin V, Lin JL, Marko NF, et al. Stereotactic radiosurgical treatment of brain metastases to the choroid plexus. Int J Radiat Oncol Biol. 2011;80:1134–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijrobp.2010.03.016.
Smith AB, Smirniotopoulos JG, Horkanyne-Szakaly I. From the radiologic pathology archives: intraventricular neoplasms: radiologic-pathologic correlation. Radiographics. 2013;33:21–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1148/rg.331125192.
Frishman-Levy L, Izraeli S. Advances in understanding the pathogenesis of CNS acute lymphoblastic leukaemia and potential for therapy. Br J Haematol. 2017;176:157–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.14411.
Waki F, Ando M, Takashima A, et al. Prognostic factors and clinical outcomes in patients with leptomeningeal metastasis from solid tumors. J Neurooncol. 2009;93:205–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11060-008-9758-3.
Herrera RA, Deshpande K, Martirosian V, et al. Cortisol promotes breast-to-brain metastasis through the blood-cerebrospinal fluid barrier. Cancer Rep (Hoboken). 2022;5: e1351. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cnr2.1351.
Vandenhaute E, Stump-Guthier C, Losada ML, et al. The choroid plexus may be an underestimated site of tumor invasion to the brain: an in vitro study using neuroblastoma cell lines. Cancer Cell Int. 2015;15:102. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-015-0257-2.
Li B, Zhao W-D, Tan Z-M, et al. Involvement of Rho/ROCK signaling in small cell lung cancer migration through human brain microvascular endothelial cells. FEBS Lett. 2006;580:4252–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.febslet.2006.06.056.
Boire A, Zou Y, Shieh J, et al. Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis. Cell. 2017;168:1101-1113.e13. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2017.02.025.
Kart BH, Reddy SC, Rao GR, Poveda H. Choroid plexus metastasis: CT appearance. J Comput Assist Tomogr. 1986;10:537–40.
Tanimoto M, Tatsumi S, Tominaga S, et al. Choroid plexus metastasis of lung carcinoma–case report. Neurol Med Chir (Tokyo). 1991;31:152–5. https://doiorg.publicaciones.saludcastillayleon.es/10.2176/nmc.31.152.
Naumann JA, Gordon PM. In vitro model of leukemia cell migration across the blood–cerebrospinal fluid barrier. Leuk Lymphoma. 2017;58:1747–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/10428194.2016.1254778.
Ghersi-Egea J-F, Strazielle N, Catala M, et al. Molecular anatomy and functions of the choroidal blood-cerebrospinal fluid barrier in health and disease. Acta Neuropathol. 2018;135:337–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-018-1807-1.
Hochman J, Assaf N, Deckert-Schlüter M, et al. Entry routes of malignant lymphoma into the brain and eyes in a mouse model. Cancer Res. 2001;61:5242–7.
Williams MTS, Yousafzai YM, Elder A, et al. The ability to cross the blood–cerebrospinal fluid barrier is a generic property of acute lymphoblastic leukemia blasts. Blood. 2016;127:1998–2006. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2015-08-665034.
Scharff BFSS, Modvig S, Marquart HV, Christensen C. Integrin-mediated adhesion and chemoresistance of acute lymphoblastic leukemia cells residing in the bone marrow or the central nervous system. Front Oncol. 2020;10:775. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2020.00775.
Erb U, Hikel J, Meyer S, et al. The impact of small extracellular vesicles on lymphoblast trafficking across the blood-cerebrospinal fluid barrier In Vitro. IJMS. 2020;21:5491. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21155491.
Acknowledgements
We thank Ms. Marta Lněníčková for her technical support.
Funding
This work was supported by the project SALVAGE (P JAC; reg. no. CZ.02.01.01/00/22_008/0004644) – funded by the European Union and by the State Budget of the Czech Republic, by MH CZ—DRO (MMCI, 00209805), NU20-03-00148, NU22-03-00159, and Masaryk University internal grants MUNI/11/JRG/1143/2023 and MUNI/A/1563/2023.
Author information
Authors and Affiliations
Contributions
NI: conceptualization, writing-original draft preparation; PS: conceptualization, writing-original draft preparation; KH: conceptualization, writing-original draft preparation; AZ: conceptualization, writing-original draft, funding acquisition; LK: writing-original draft preparation; MS: writing-original draft preparation; KM: writing-original draft preparation; MB: writing-review and editing; TK: writing-original draft preparation, and funding acquisition; RH: conceptualization, writing-review, editing, and funding acquisition; MJ: conceptualization, supervision, writing-review, editing, and funding acquisition. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Izadi, N., Solár, P., Hašanová, K. et al. Breaking boundaries: role of the brain barriers in metastatic process. Fluids Barriers CNS 22, 3 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-025-00618-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12987-025-00618-z