Abstract
Brain metastasis is an important cause of mortality in patients with cancer and represents the majority of all intracranial tumors. A key step during the metastatic journey of the cancer cell to the brain is the invasion through the blood–brain barrier (BBB). Nevertheless, the molecular mechanisms that govern this process remain unknown. The BBB has been blamed for limiting the access of therapeutic drugs to the brain, which provides a safe haven for the cancer cell in the brain and confers poor prognosis for the patient. Here, we explore the genes that control the transmigration of metastatic cancer cells across the BBB, offering new targets for the development of gene and cell therapies against brain metastases.
Keywords: Cancer, brain metastases, transcellular migration, paracellular migration, tight junctions, blood-tumor barrier
THE BLOOD-BRAIN BARRIER: A SELECTIVE GATEKEEPER
Brain metastases constitute the most common mass lesions in the brain. They require invasion of primary cancer cells from lung, breast, or skin, trafficking through the circulatory system, and colonization in the brain parenchyma (see Glossary) [1].
Some metastatic cancer cells possess the unique ability of crossing the blood-brain barrier (BBB), which is a selective gatekeeper composed of endothelial cells, pericytes, and the foot processes of astrocytes. The BBB separates the brain from systemic circulation and protects it from foreign infectious and/or toxic agents, including targeted therapies [2–4]. In fact, the BBB is estimated to block the transport of ~ 98% of molecules in systemic circulation [5].
By default, the network of closely faced endothelial cells and the continuous tight junctions renders the mechanism of metastasis through the BBB to be different from metastasis to other organs (Figure 1). Under physiological conditions, the BBB is so restrictive that not even ions can pass freely from one side to the other, making it challenging to study in experimental settings (Box 1). Nonetheless, >70% of brain metastases have shown leakage from blood vessels, indicating that the BBB is compromised in the setting of brain metastases [6]. Yet, the genetic drivers in cancer cells and in endothelial cells of the BBB that may be exploited to permit crossing the BBB remain unclear.
Figure 1: Anatomy and pathophysiology of the blood-brain barrier.

The metastatic cancer cell faces the BBB on its way to the brain. This barrier consists of multiple layers of different cell types that restrict the entrance of large macromolecules into the brain parenchyma. The innermost layer is formed by endothelial cells, which contain different transporters in their membranes and are connected through tight junctions. Pericytes attach to the abluminal site of the endothelial cells and are embedded in a sheet of extracellular matrix (ECM). The outermost layer consists of the end-feet of astrocytes. While the BBB restricts the entry of many molecules and cells, it is not an impenetrable barrier to transmigration of metastasizing cancer cells (1). Indeed, in this regard, it has been proposed that the brain can function as a sanctuary site for metastatic cells that effectively breach the BBB, where they are subsequently protected from the effects of chemotherapy and other agents that cannot penetrate the brain (2). Eventually, metastasizing cancer cells colonize their new microenvironment and grow into macrometastases (3).
Box 1. Techniques Used to Study Transmigration Across the Blood-Brain Barrier.
In vitro models:
Monoculture models that consist of cerebral endothelial cells lined up on a semipermeable support under static culture conditions [105].
Co-culture models that comprise endothelial cells, astrocytes, and/or pericytes to attempt to form paracellular tight junctions [105].
Stem cell models that differentiate into cerebral endothelial cells that can line up into a BBB model. Neural stem cells can differentiate into neuronal cells and can help in generating a more complex neurovascular unit [106]. Mesenchymal stem cells can also be used to simulate pericytes as both cell types express similar cell surface markers [107, 108].
The Transwell system as a side-by-side vertical diffusion model comprising a microporous semipermeable membrane of seeded cells inundated in culture medium that separates the vascular and parenchymal side compartments [107, 109, 110].
Microfluidic-on-chips as a dynamic 3D model where endothelial cells are cultured in the lumen of fiber tubes inside a closed chamber. The neurovascular unit is seeded in the extraluminal compartment. A variable-speed pulsatile pump generates an intraluminal flow that is physiologically comparable to that observed in capillaries [105].
In vivo models:
Intravenous injection of a compound, after which the analyte concentration is determined at different times in the brain, plasma, and cerebrospinal fluid [111].
Brain perfusion technique allows a direct indication of the permeation of a substance across the BBB [112]. The compound of interest is dissolved and pumped into the vasculature that leads to the brain. At a specific time, perfusion is stopped, the animal is decapitated, and the amount of substance in the brain is determined. Radiolabeled compounds are frequently employed.
Tomographic methods comprise PET and single photon emission-computed tomography (SPECT) that study brain uptake kinetics, cerebral blood flow, BBB integrity, and efflux mechanisms [113].
Microdialysis sampling as a minimally invasive method that allows the monitoring of neurotransmitters in the brain. It can be used to explore the transport and metabolism of drugs and other substances at the BBB [114].
Zebrafish offer an optically transparent model organism with an endothelial BBB that help assess the delivery of natural products, fluorescence dyes and drugs across the BBB [92, 115].
In situ models:
Brain efflux methods complement in vivo models of the BBB by superimposing a vascular perfusion fluid replacement of the circulating blood in the animal [105].
Ex vivo models:
Patient-derived circulating tumor cells or exosomes expanded ex vivo and then reintroduced into animal models to recapitulate metastatic transmigration across the BBB [60].
Multi-organ microfluidic chip model consisting of two bionic organ units - a primary site and a brain with a functional BBB structure [43].
Given the poor prognosis and limited effective therapies [7–9], it is of crucial importance to understand the molecular mechanisms of brain-specific metastasis. The gene and/or protein expression profile of metastatic cells is a key determinant of site-specific metastasis formation. Definite expression formulations might dictate predilection to specific target organs. It is hypothesized that the brain, with its unique physiological barriers, dictates an expression profile from metastatic cells to interact with and/or penetrate the endothelial linings of the BBB.
Genetic alterations in tumor cells drive the metastatic capabilities of cancer cells and their ability to transmigrate through the BBB. This review aims to explore the genetics and biological mechanisms that mediate the transmigration process of the metastatic cancer cells through the BBB. Identifying genetic biomarkers might serve as novel diagnostic and predictive markers for metastasis progression and for development of new molecular targeted therapies against brain metastases.
ROUTES OF TRANSMIGRATION
Transmigration across the BBB is a complex process that is tightly regulated. The endothelial cells of the BBB are closely attached by tight junctions that restrict passive diffusion [10]. Pericytes, also, play a role in stabilizing the brain vasculature, regulating the expression of tight and adherent junctions, restricting transcytosis, synthesis of extracellular matrix components, and initiating neovascularization [11]. The basal membrane, constituted of endothelial cells and pericytes, provides another layer of insulation [11]. Astrocytic foot processes that are in close contact with the basal membrane connect the BBB to the neuronal brain parenchyma. This connection helps to control the diffusion restrictions across the BBB [12].
During the formation of brain metastases, transmigration through the BBB is a rate-limiting step. Two possible routes have been described: the paracellular pathway (between endothelial cells) and the transcellular pathway (across the endothelial cell) [3]. In vitro and in vivo studies have shown that cellular diapedesis can occur either by creating a paracellular gap or by establishing a transcellular pore (Figure 2).
Figure 2: Mechanisms of transmigration across the blood-brain barrier.

Metastatic cells utilize different approaches to cross the multiple cell layers that constitute the blood-brain barrier (BBB). Passage through the paracellular route (left) includes proteolysis of junctional adhesion molecules, such as JAM-B, using proteases and serum factors or intercalation between the layers of endothelial cells and disrupting the integrity of the BBB. The transcellular pathway (right) involves strong adhesion through integrins and the formation of a channel or a pore within the endothelial cell that allows the transmigration of the invading cancer cell into the brain.
The paracellular route across tight junctions allows passive diffusion of water, gases, and lipid-soluble molecules [10]. Physiologically, molecules greater than 400 Da are unable to cross through the BBB, as they are too large for passive diffusion. Charge and chemical (hydrophilic/lipophilic) properties also play a role in the transmigration process [13]. Upon contact with the endothelial lining of the BBB, metastatic cancer cells can disrupt the tight junctions by releasing proteases and/or miRNAs that compromise the integrity of the BBB and increase permeability [3, 14].
Transcellularly, solutes and nutrients can cross by passive diffusion or active transport. Lipid-soluble molecules can cross passively, whereas molecules like glucose and amino acids might necessitate carrier-mediated transporters and metabolic energy to cross the BBB [10].
In addition, the increased tumor heterogeneity of brain metastases and the overexpression/suppression of certain genetic variants can lead to the expression/downregulation of proteins that facilitate the paracellular or transcellular routes [15–19] (Table 1).
Table 1:
Genes and proteins that mediate migration of metastatic cancer cells through the blood-brain barrier
| Gene/Protein | Expression in metastasis | Mechanism of action | Primary tumor | Refs |
|---|---|---|---|---|
| ADAM8 | Overexpression | Upregulated MMP9 | Breast cancer | [41] |
| Adenosine A2A receptor | Downregulation | Inhibition of the A2A receptor downregulates expression of CLDN5, occludin and ZO-1 | NSCLC | [98] |
| AKR1B10 | Overexpression | Affects the expression of MMP-2 and MMP-9 via MEK/ERK signaling | NSCLC | [43] |
| ALCAM | Overexpression | Increases cancer cell adhesion to endothelial cells | NSCLC | [70] |
| Angiopoietin-2 | Overexpression | Changes ZO-1 and claudin-5 tight junction protein structures | Breast cancer | [22] |
| Annexin A1 | Overexpression | Cancer cell expresses high levels of annexin A1 and releases it outside, facilitating entry into brain | SCLC | [83] |
| Cathepsin S | Overexpression | Induces the proteolysis of the junctional adhesion molecule, JAM-B | Breast cancer | [23] |
| CEMIP | Overexpression | Uptake of CEMIP+ exosomes by brain endothelial and microglial cells induce endothelial cell branching and inflammation leading to vascular remodeling and metastasis | Breast cancer | [58] |
| CLDN5 | Downregulation | Untightens endothelial cellular adhesion and destabilize the tight junctions | Lung cancer | [95] |
| COX2 | Overexpression | Facilitates the angiogenic formation of new blood vessels that help tumor cells connect to the circulation | Breast cancer | [71, 72] |
| CXCR4 | Overexpression | Activates the PI3K/AKT signaling | Breast cancer | [80] |
| Desmin+ | Overexpression | - | Breast cancer | [16] |
| FUT4 | Overexpression | Codes for CD15 expression that increases adhesion of metastatic cancer cells to the BBB | Lung cancer | [86] |
| FUT7 | Overexpression | Codes for CD15s expression that increases adhesion of metastatic cancer cells to the BBB | Lung cancer | [86] |
| GBP1 | Overexpression | Induced by the T cells upon interaction with breast cancer cells vamps the metastatic potential | Breast cancer | [82] |
| HBEGF | Overexpression | Activated EGF receptors and their downstream signaling. | Breast cancer | [71, 73] |
| Heparanase | Overexpression | Degrading and compromising ECM structural integrity leading to the release of growth factors, chemokines, and angiogenic factors that are important for the passage of metastatic cancer cells. Promotes Akt signaling and stimulates PI3K- and p38-dependent endothelial cell migration and invasion |
Melanoma | [50–52] |
| Laminin α2 | Downregulation | - | Breast cancer | [16] |
| MFSD2a | Downregulation | Increased the propensity of brain metastasis by reducing DHA transport and altering lipid metabolism | Breast cancer Lung cancer Prostate cancer |
[88] |
| miR-105 | Overexpression | Targets the tight junctional protein ZO-1 | Breast cancer | [55] |
| miR-143–3p | Overexpression | Inhibits VASH1 expression and triggers proteasomal mediated degradation of endothelial junctions | Lung cancer | [57] |
| miR-181c | Overexpression | Delocalize actin by downregulating phosphoinositide-dependent kinase-1 (PDPK1) | Breast cancer | [56] |
| miR-509 | Downregulation | Increases RhoC and TNF-α that control the invasion of cancer cells and the trans-endothelial cell migration through MMP9. | Breast cancer | [94] |
| MLCK | Overexpression | Activated at the adhesion site, leading to regional myosin contraction and, eventually, intravasation of the tumor cell | Breast cancer | [81] |
| MMP1 | Overexpression | ECM degrading capacity and facilitate the angiogenic formation of new blood vessels | Breast cancer | [38] |
| MMP2 | Overexpression | Deregulation of the MAPK pathway | Breast cancer | [40] |
| PLEKHA5 | Overexpression | Adaptor protein that mediates Phosphoinositide 3-kinases (PI3K) signaling | Melanoma | [78] |
| PLGF | Overexpression | Activation of Rho/ROCK followed by activation of ERK1/2 and Ser/Thr phosphorylation of occludin | SCLC Breast cancer Colorectal cancer Gastric cancer |
[44–48] |
| Rab7 | Downregulation | Increases endothelial recycling endocytic transcytosis | Breast cancer | [99] |
| Rho GTPase | Overexpression | Increasing actomyosin contractility | SCLC | [64] |
| S1P3 | Overexpression | Release of IL-6 and CCL2, which causes relaxation of the tight junctions between endothelial cells | Breast cancer | [54] |
| Sdc1 | Overexpression | Increased adhesion to the perivascular regions of the brain and led to eventual trans-endothelial migration | Breast cancer | [85] |
| SEMA4D | Overexpression | Binds to Plexin-B1 receptors in pericytes to promote pro-inflammatory responses that disrupts BBB endothelial tight junctions | Breast cancer | [61] |
| Seprase | Overexpression | Formation of proteinase-rich membrane domains by these enzymes is believed to contribute to the ECM degradation | Melanoma | [24] |
| Serine Protease | Overexpression | Apoptotic induction of the endothelial cells and disruption of tight junctions | Melanoma | [20] |
| Slug | Overexpression | Binds to the promoter region of MMP1 in breast cancer cells, leading to its overexpression | Breast cancer | [39] |
| ST6GALNAC5 | Overexpression | Enhances adhesion of cancer cells to brain endothelial cells | Breast cancer | [71] |
| TNF | Overexpression | TNF receptor-1 pathway | Breast cancer | [21, 22] |
| uPA | Overexpression | Formation of proteinase-rich membrane domains by these enzymes is believed to contribute to the ECM degradation | Melanoma | [24] |
| VASH1 | Downregulation | Triggers proteasomal mediated degradation of endothelial junctions | Lung cancer | [57] |
| Visfatin | Overexpression | Upregulation of the cytokine CCL2, which opens the tight junctions | SCLC | [53] |
| VLA-4 | Overexpression | Induces cells to intercalate into the tight monolayer of endothelial cells causing its destabilization | Melanoma | [69] |
| αB-crystallin | Overexpression | Adhesion occurs via α3β1 integrin-dependent mechanism | Breast cancer | [84] |
| αv-integrin | Overexpression | Influence cancer cell attachment and angiogenesis | Breast cancer Lung cancer Renal cancer Melanoma |
[67] |
GENES MEDIATING BARRIER REMODELING AND PERMEABILITY ALTERATIONS
Brain metastatic cells are known to compromise BBB integrity. Using the paracellular route, they break junctional adhesion molecules while crossing the BBB [20] (Figure 3). Triggering specific signaling pathways can increase leakage and lead to pore formation. For example, in 4T1 models of triple-negative breast cancer (TNBC), the activation of tumor necrosis factor (TNF)-TNF receptor-1 pathway can result in pores of relatively large sizes (11–13nm) in the BBB, which ultimately leads to higher permeabilization [21, 22].
Figure 3: Anatomy of the tight junction.

Tight junctions between endothelial cells are formed by transmembrane proteins, such as junctional adhesion molecules (JAMs), cadherins, claudins, and occludins, and by cytosolic proteins, such as catenins and zonula occludens (ZO) proteins.
Transmigration through Proteolysis and Released Serum Factors
In breast cancer brain metastases, high expression of cathepsin S – a regulator of BBB transmigration produced by tumor cells and macrophages – is significantly associated with decreased survival in patients with breast cancer [23]. In preclinical studies with MDA-MB-231 breast cancer cells, depletion of macrophage and tumor cell reserves of cathepsin S significantly decreased brain metastases [23]. The release of cathepsin S by metastatic cells induces the proteolysis of the junctional adhesion molecule, JAM-B [2]. This in turn allows the metastatic cell to cross the BBB. Furthermore, cathepsin S inhibitors were developed and were successful in significantly reducing brain metastasis in vivo [23]. In malignant melanoma, the release of serine proteases by metastatic melanoma cell lines, A2058 and B16/F10, induced apoptosis of the endothelial cells and disrupted the continuity of tight junctions [20]. This allowed the transmigrating melanoma cells to overcome the BBB. Protease systems, including seprase and urokinase-type plasminogen activator (uPA), have been shown to facilitate transmigration of LOX melanoma cells through brain endothelial cells both in vivo and in vitro [24]. Several studies showed that these enzymes are significantly associated with brain metastasis [25–33]. The formation of proteinase-rich membrane domains by these enzymes is believed to contribute to extracellular matrix (ECM) degradation [24], which subsequently increases the invasive ability of cancer cells across the BBB. Metastatic melanoma cells, for example, have been shown to express several types of proteases including uPA [24], seprase [27], and matrix metalloproteinases (MMP) [34–36] that disintegrate tight junctions. Translational studies revealed that targeting these proteases with the protease inhibitor Pefabloc markedly reduced the metastatic potential of melanoma cells [20].
MMP1 and MMP2 possess ECM degrading capacity and facilitate the angiogenic formation of new blood vessels to help tumor cells connect to the circulation [37, 38]. Targeted knockdown of MMP1 in brain metastatic MDA-MB-231 cells reduces primary tumor growth and significantly inhibits brain metastasis [38]. Furthermore, MMP1 upregulation in resistant metastatic breast cancer cells is controlled by the Slug protein [39]. Slug expression resulted in MMP1 upregulation in MCF-7/ADR cells and was significantly associated with reduced drug sensitivity [39]. However, Slug knockdown reduced MMP1 expression and enhanced the drug sensitivity of breast cancer cells [39]. Slug was shown to bind to the promoter region of MMP1 in breast cancer cells, leading to its overexpression. MMP2, also, plays a role in the invasion and metastasis of breast cancer cells to the brain. Extracellular signal-regulated kinases (ERK)1/2 and mitogen-activated protein kinase (MAPK) have been correlated to MMP2 expression in ENU1564 cells. It was found that MMP2 plays a role in the development of breast cancer brain metastases through the deregulation of the MAPK pathway [40]. Moreover, MMP9 seems central in other works relating to brain metastasis. The metallopeptidase domain 8 (ADAM8) was found to promote early metastatic processes in breast cancer through trans-endothelial migration across the BBB [41]. ADAM8 overexpression upregulated MMP9 in MDA-MB-231 cells and led to the shedding of P-selectin glycoprotein ligand-1 (PSGL-1), a substrate of ADAM8 that promotes adhesion of leukocytes to endothelial cells [41]. Thus, the major effect caused by shedding of PSGL-1 is an increase in mobility and invasiveness into the BBB [42]. As such, MMP9 and ADAM8 are plausible targets for brain metastasis formation. AKR1B10 also affects the expression of MMP-2 and MMP-9 via MEK/ERK signaling [43]; protein expression of AKR1B10 is significantly elevated in brain metastases from non-small cell lung cancer (NSCLC) [43]. Silencing AKR1B10 in brain metastatic PC-9 cells suppressed their transmigration through the BBB [43], suggesting that AKR1B10 could be a prospective therapeutic target for NSCLC brain metastases.
In small cell lung cancer (SCLC), placental growth factor (PLGF) was significantly elevated in the serum of patients with brain metastases and in NCI-H250 cells. PLGF, which belongs to the vascular endothelial growth factor (VEGF) subfamily, binds to its receptor, VEGF receptor-1 (VEGFR-1), on brain endothelial cells [44]. Subsequently, the intracellular Rho/ROCK signaling is activated and followed by ERK1/2 activation [44]. ERK1/2 activation causes Ser/Thr phosphorylation of occludin, a tight junction protein. This results in junctional disassembly and allows for the trans-endothelial migration of SCLC cells [44]. In breast cancer, several studies showed that elevated levels of PLGF were significantly associated with metastasis and mortality [45, 46]. Similar results were also reported in metastatic colorectal and gastric cancers [47, 48], which suggest that PLGF could serve as a possible target for therapeutic regimens.
Neurotrophins are proteins that promote the survival, development, and function of neurons [49]; nevertheless, they can act as mediators of BBB passage for cancer cells. Melanoma cells express receptors that have a high affinity to brain-derived neurotrophins. The binding of neurotrophins to their receptors induces the release of heparanase [50]. Heparanase, an endoglycosidase, is preferentially expressed in brain metastases and its overexpression in tumor cells confers an invasive phenotype in experimental animals [51]. Mechanisms by which heparanase facilitates cancer progression likely involves degrading and compromising ECM structural integrity [52]. This leads to the release of growth factors, chemokines, and angiogenic factors from the ECM that are important for the passage of metastatic cancer cells. In melanoma brain metastases, heparanase is highly expressed [52]. Pre-treating melanoma cells with heparanase leads to a significant increase in the number of cells that invade the brain [52]. These observations and the unexpected identification of a single functional heparanase suggest that the enzyme is a promising target for anti-cancer drug development.
Visfatin, a new pro-inflammatory adipocytokine, is predominantly released by macrophages in visceral fat tissue and is involved in various malignant tumors. In SCLC, visfatin was found to induce the upregulation of the cytokine, CCL2, in NCI-H446 cells [53], which opens tight junctions and promotes metastatic migration. In breast cancer brain metastases, S1P3 is overexpressed as a consequence of the neuroinflammatory reaction induced by the BBB-permeable cancer cells. Astrocytes that overexpress S1P3 release interleukin (IL)-6 and CCL2, which cause relaxation of the tight junctions and increases the BBB permeability [54]. Inhibition of S1P3 leads to the tightening of the BBB both in in vitro and in vivo settings using MDA-MB-231-BR, JIMT-1-BR, and SUM190-BR cells [54], hinting that BBB permeability can be altered via S1P3 regulation.
MiRNAs secreted by cancer cells can mediate crosstalk between metastatic cells and the host microenvironment. In MDA-MB-231 metastatic breast cancer, secreted miR-105 targets the tight junctional protein zonula occludens (ZO)-1, allowing the cancer cells to transmigrate into the brain [55]. Non-metastatic MCF-10A cancer cells exhibit metastatic properties in distant organs when miR-105 is overexpressed, whereas its inhibition removes these abilities [55]. It has been shown that miR-105 can be identified during the pre-metastatic stage, and its levels in the blood and tumor can reflect ZO-1 expression and metastatic progression in early-stage breast cancer [55, 56]. miR-181c, also, has been shown to delocalize actin by downregulating its target gene, phosphoinositide-dependent kinase-1 (PDPK1) [56], which leads to alterations in actin dynamics. In lung cancer brain metastases, miR-143–3p was upregulated when compared to primary cancer tissues [57]. Further investigation revealed that it can increase the invasion capability of an in vitro BBB model by targeting the binding sites of vasohibin-1 (VASH1) to inhibit its expression, triggering proteasomal mediated degradation of endothelial junctions [57]. This implicates a causal role for the miR-143–3p/VASH1 axis in lung cancer brain metastasis. Inhibition of such miRNAs and/or the suppression of their extracellular carriers decrease invasiveness of MDA-MB-231 breast cancer cells through BBB [56].
Tumor-secreted exosomes can also contribute to the transmigration across the BBB. Proteomic analysis identified the protein CEMIP to be increased in exosomes from brain-specific metastases [58]. CEMIP depletion in tumor cells impaired brain metastasis, disrupting invasion and cancer cell attachment to the brain vasculature [58]. The uptake of CEMIP+ exosomes by brain endothelial and microglial cells induce endothelial cell branching and inflammation in the perivascular niche by upregulating the pro-inflammatory cytokines encoded by Ptgs2, TNF and Ccl/Cxcl that are known to promote brain vascular remodeling and metastasis [56, 59]. In patient-derived exosomes and xenografts, CEMIP was elevated. It further predicted brain metastasis progression and overall survival [58].
Circulating tumor cells (CTCs) expanded ex vivo from patients with breast cancer can generate brain metastases in mice. Genome-wide sequencing analyses identified SEMA4D as a regulator of tumor cell transmigration through the BBB [60]. Semaphorins are a large family of secreted or transmembrane proteins that have versatile roles in axonal guidance and the immune system [61]. Most of the effects of SEMA4D are mediated by its interaction with Plexin-B1 receptors in pericytes, which promote pro-inflammatory responses [61]. Studies in CNS diseases revealed that SEMA4D disrupts endothelial tight junctions of the BBB [62, 63], driving transmigration through the paracellular route. Overexpression of the SEMA4D in the primary tumor site was associated with brain-specific metastases. In addition, CTC lines BRx50 and BRx42, which are brain metastatic, exhibited higher SEMA4D expression levels compared to CTC lines with no brain tropism in mice [60].
Physical and Anatomical Alterations of the Tight Junctions
Physical alterations in the BBB increase its permeability. In breast cancer brain metastases, the pericyte protein, desmin, is overexpressed which leads to remodeling of the BBB and increased metastasis of human MDA-MB-231-BR-Her2 and murine 4T1-BR5 cancer cells, suggesting a molecular target that modifies permeability [15]. Elevated desmin levels were also found in human craniotomy specimens of breast cancer brain metastases [16]. The decreases in astrocytic basement membrane component laminin α2 and the CD13 pericyte subpopulation are also a feature of increased BBB permeability in patients with breast cancer brain metastases [16].
Rho GTPases are a family of small signaling G proteins and a subfamily of the Ras superfamily. The members of the Rho GTPase family have been shown to regulate many aspects of intracellular actin dynamics. In SCLC, they are activated during trans-endothelial migration, thereby facilitating the breakdown of intercellular junctions through increasing actomyosin contractility [64]. The inhibition of their effect can prevent the trans-endothelial migration of disseminated SCLC cells across the BBB [65, 66].
GENETIC MEDIATORS OF ADHESION AND EXTRAVASATION
The transcellular route of brain metastasis involves genetic mediators of adhesion that drive the expression of integrins, endothelial receptors, and cellular proteins. These proteins and cellular components enhance the attachment of the metastatic cancer cell to the BBB and promote its migration, passively or actively, through the endothelial cell layer.
Adhesion is often facilitated by integrins that influence cancer cell attachment and angiogenesis and promote invasion and migration of tumor cells. In brain metastases of breast, lung and renal carcinomas and in malignant melanoma, the strong αv-integrin expression – especially of αvβ3 and αvβ8 integrins – in patient-derived samples hints at its involvement in the formation of brain metastasis [67, 68]. As such, αv-integrins may be diagnostic biomarkers and therapeutic targets for patients with metastatic cancers (see Clinician’s Corner). Very Late Antigen-4 (VLA-4) is another integrin that is expressed on the cell surfaces of stem cells, progenitor cells, and other inflammatory cells. It plays an important role in mobilizing leukocytes to the site of inflammation through cellular adhesion. In patient-derived samples, it was found that more than 90% of all human melanoma brain metastases are positive for VLA-4 expression [69]. VLA-4 was found to enhance the adhesion of melanoma hMel-CD49d cancer cells to the endothelial cells and allowed the incorporation of cancer cells in between the endothelial monolayer, causing disruption of the BBB. Inhibiting VLA-4 activity annulled the adhesion of brain metastatic cells with the BBB and prevented extravasation of cancer cells [69]. It was concluded that VLA-4 expression increases the risk of metastasis across the BBB, and VLA-4 was proposed as a therapeutic target for the inhibition of metastatic formation in the brain.
Clinician’s Corner.
Brain metastasis portends poor prognosis as there are limited effective therapies that can cross the blood-brain barrier (BBB).
Understanding the transmigration routes of cancer cells across the BBB is essential for the development of targeted therapies and preventive measures that can target metastatic cancer cells before entering the brain.
Genes and proteins regulating transmigration present an opportunity for developing diagnostic assays that can help in the early detection and prevention efforts against brain metastasis.
Gene therapy that relies on overexpressing genes that repress the transmigration of metastatic cancer cells or downregulating genes that drive transmigration across the BBB is a therapeutic approach that can improve survival rates of patients with brain metastasis
The role of ALCAM in brain metastasis formation has been investigated in NSCLC. CRISPR/Cas9 gene ablation of ALCAM in the NCI-H460 NSCLC cell line showed a significantly decreased cell adhesion capacity to human brain endothelial cells [70]. Intracranial and intracardiac cell injection of NCI-H460 cells in NMRI-Foxn1nu/nu mice revealed that significantly lower tumor cell dissemination in mice injected with ALCAM-Knockout cells in both mouse models and that the number and size of brain metastases were significantly diminished in ALCAM depleted tumors [70]. This suggests that increased ALCAM expression promotes the formation of brain metastases in NSCLC by increasing tumor cell dissemination and interaction with the brain endothelial cells.
In human-derived samples of breast cancer brain metastasis, cyclooxygenase (COX2), heparin-binding EGF (HBEGF), and α2,6-sialyltransferase (ST6GALNAC5) were identified as mediators of cancer cell passage through the BBB [71]. COX2 is known to promote specific extravasation of tumor cells into lung capillaries to seed pulmonary metastasis. When COX2 is expressed in human breast cancer cells, along with epiregulin (EREG) and MMPs, it facilitates angiogenesis [72]. EREG, however, has been shown to increase tumorogenicity and invasion of breast cancer cells to the lungs, but not brain. Still, COX2 knockdown inhibited the transmigration of breast cancer cells across the BBB [71]. This raises the question of whether the passage of cancer cells through different organs can prime them to express genes that mediate their passage through the BBB. HBEGF, which plays an important role in various biological processes such as heart development and maintenance has also been shown to play a role in cellular adhesion to the BBB. Its mode of action involves the activation of EGF receptors (EGFR) and their downstream signaling. In cancer, HBEGF expression has been associated with malignant tumorigenicity, metastasis, and the development of resistance to therapy [73]. In in vitro and in vivo settings, inhibition of HBEGF has been shown to halt tumor growth and progression [71, 73]. Knockdown of HBEGF along with COX2 and EREG has been associated with inhibition of breast cancer cell permeability across the BBB [71].
ST6GALNAC5 is a sialyltransferase that specifically mediates brain metastasis. Normally, it is involved in the synthesis of α-series gangliosides in the nervous tissues. The expression of ST6GALNAC5 is restricted to the brain and may have implications in neurodegenerative diseases [74]. Nevertheless, it has been shown that the expression of ST6GALNAC5 in CN34-BrM2 and MDA231-BrM2 cell lines of breast cancer enhances their adhesion to brain endothelial cells and their passage through the BBB [71]. Conversely, in an in vitro model, the expression of ST6GALNAC5 decreased the interaction of MDA-MB-231 breast cancer cells and the BBB [75]. Further exploration revealed that ST6GALNAC5 increased the expression of GD1α ganglioside at the cell surface of invading breast cancer cells, limiting interaction with the BBB [75]. Furthermore, studies with patient-derived xenografts in TNBC showed that ST6GALNAC5 expression was not related to metastasis location [76]. Nonetheless, in colorectal cancer, ST6GALNAC5 expression was significantly associated with brain metastatic susceptibility in patient-derived samples [77]. Further studies are needed to clarify the role of ST6GALNAC5 as it is possible that the expression of this gene is situational and depends on the stage of the brain metastatic process.
In A375Br melanoma cells, the expression of PLEKHA5 was associated with early development of brain metastases [78]. Inhibition of PLEKHA5 caused a decrease of passage across the BBB. PLEKHA proteins play a role in brain development and are thought to function mostly as adaptor proteins because they lack catalytic subunits [79]. PLEKHA5 clearly has phosphoinositide-binding specificity implicating a role of phosphoinositide 3-kinases (PI3K) in its membrane recruitment. Moreover, phosphoinositide-binding specificities suggest that PLEKHA5 can be a key mediator of cellular signaling, cytoskeleton rearrangement, membrane protein targeting, and vesicular trafficking [78]. As such, the established role of PLEKHA5 in cell migration might facilitate BBB transmigration and promote it as a therapeutic target for brain metastasis.
Expression of chemokine receptors like CXCR4 and CCR7 is significantly associated with metastasis and mortality in breast cancer. In fact, the expression of CXCR4 through NF-κB promoted breast cancer cell migration and metastasis. The interaction of CXCR4 with its ligand, SDF-1α, in MDA-MB-231 and DU4475 breast cancer cells induced activation of the PI3K/AKT signaling pathway and the phosphorylation of focal adhesion kinase (FAK) and forkhead in rhabdomyosarcoma-like 1 (FKHRL1) [80]. This indicates that the activation of the PI3K/AKT signaling pathway by CXCR4/SDF1 is implicated in breast cancer cell migration through the BBB and targeting it may offer therapeutic potential.
In addition to its remodeling capacity (discussed before), heparanase mediates complex interactions between tumor cells and the host tissue environment. Cell surface expression of heparanase elicits a firm cell adhesion, reflecting an involvement in cell–ECM interaction. Heparanase promotes Akt signaling and stimulates PI3K- and p38-dependent endothelial cell migration and invasion [51]. Silencing of the heparanase gene inhibits tumor metastasis and angiogenesis in experimental models.
The visualization of how myosin light chain kinase (MLCK) is modulated during tumor cell intravasation helped elucidate the signaling interaction between metastatic breast cancer cells and endothelial cells lining the BBB [81]. Endothelial MLCK was found to be activated at the adhesion site of MDA-MB-231 cancer cells, leading to regional myosin contraction and, eventually, intravasation of the tumor cell [81]. Blocking endothelial myosin contraction cut the transcellular route of invasion [81], implicating an important role for MLCK in tumor intravasation and highlighting yet another therapeutic target.
The contribution of immune cells to the metastatic process has always been questioned. The co-culturing of T lymphocytes with breast cancer cells increased the metastatic potential of the cancer cells across the BBB. Guanylate-Binding Protein 1 (GBP1) was found to be induced by T cells upon interaction with metastatic MDA-MB-231-BM breast cancer cells [82]. In humans, GBP1 was upregulated in patients with breast cancer brain metastases. Silencing the gene in vitro reduced the invasive ability of breast cancer cells through the BBB [82]. This provides another potential target for intervention to prevent brain metastasis formation.
The brain microvasculature has been implicated as an important player for tumor cell attachment and growth. In SCLC, the endothelial cells in the brain induced NCI-H446 cells to secrete annexin A1 [83]. The release of annexin A1 was found to promote SCLC brain metastasis formation by enhancing cellular adhesion on the BBB [83]. Knockdown of annexin A1 in SCLC cells prevented transcellular migration and metastasis, highlighting the role of annexin A1 as a BBB-scaffolding protein in brain metastasis.
αB-crystallin is a small heat shock protein that in humans is encoded by the CRYAB gene. It prevents protein aggregation, inhibits apoptosis, and contributes to the intracellular architecture. In women with TNBC, αB-crystallin was found to be a regulator of cancer metastasis to the brain. The expression of αB-crystallin enhanced the adhesion of GILM2 and MDA-MB-231 breast cancer cells to the endothelial cells of the BBB and facilitated trans-endothelial migration [84]. Silencing of the CRYAB gene caused binding inhibition with endothelial cells. Further exploration revealed that adhesion occurs via α3β1 integrin-dependent mechanism [84]. As such, CRYAB and αB-crystallin represent an important therapeutic target in TNBC.
Syndecans (Sdcs) are cell surface proteins that function as co-receptors for growth factors, matrix proteins, cytokines, and chemokines. In their role, Sdcs are capable of mediating cell–cell and cell–matrix adhesion. In breast cancer brain metastasis, overexpression of syndecan-1 (Sdc1) was found to increase adhesion to the perivascular regions of the brain and lead to eventual trans-endothelial migration [85]. Silencing expression of Sdc1 in MDA-MB-231 and 4T1 breast cancer cells significantly reduced metastasis to the brain [85]. In addition, loss of Sdc1 also led to changes in breast cancer cell-secreted cytokines/chemokines, which may modulate the BBB.
Fucosylated CD15 and sialyated CD15s are carbohydrate epitopes on cancer cells that aid in adhesion. Overexpression of CD15 or CD15s epitopes in SEBTA-001, NCI-H1299, and COR-L105 lung cancer cell lines led to a significant increase in adhesion of cancer cells to the E-selectin on cerebral endothelial cells [86, 87]. This resulted in the disruption of cerebral endothelial cell monolayers. Knockdown of fucosyltransferases 4 (FUT4) and 7 (FUT7) that code for CD15 and CD15s expression, respectively, in metastatic cancer cells prevented disruption of an in vitro BBB model [86]. In addition, non-metastatic COR-L105 lung cancer cells turn metastatic when forced to overexpress either FUT4 or FUT7 [86]. This hints at the clinical significance of using cancer epitopes as biomarkers for metastasis.
GENE SUPPRESSION OF TRANSMIGRATION
Genes that suppress metastatic transmigration across the BBB are attractive targets for prognostic and therapeutic biomarkers. Comprehension of the biological and genetic mechanisms that govern transmigration suppression can provide insight into the processes that promote metastatic spread.
MFSD2a is a nutritionally regulated gene with important roles in mammalian tissue and organ growth, lipid metabolism and cognitive and motor functions. In the brain, MFSD2a selectively transports docosahexaenoic acid (DHA), an omega-3 fatty acid, across the BBB. Genetic deletion of MFSD2a protein in mice lead to impaired DHA transport and reduced levels of vital lipid metabolites. Using patient-derived xenograft models, the inhibition of MFSD2a increased the propensity of brain metastasis by reducing DHA transport and altering lipid metabolism in the metastatic microenvironment [88]. Deficiencies in DHA transport and metabolism are linked to cognitive deficits and ataxia [89, 90]. This suggests that restoring the DHA levels in the brain tumor microenvironment may be a novel therapeutic strategy to block metastatic cell growth and survival. Furthermore, MFSD2a is a key regulator of BBB function. Genetic ablation of MFSD2a results in a leaky BBB from embryonic stages through to adulthood, but the normal patterning of vascular networks is maintained [91]. Electron microscopy examination reveals a dramatic increase in CNS-endothelial-cell vesicular transcytosis in MFSD2a knockout mice, without obvious tight-junction defects [91]. Zebrafish models further confirmed that MFSD2a inhibits transcytosis, as MFSD2a mutants displayed elevated BBB permeability due to increased transcytosis [92]. Moreover, MFSD2a endothelial expression is regulated by pericytes that facilitate BBB integrity, whereby lipids transported by MFSD2a establish a unique environment that inhibits caveolae vesicle formation in CNS endothelial cells to suppress transcytosis and preserve BBB integrity [93].
MicroRNA profiling analyses revealed that miR-509 is significantly decreased in metastatic MDA-MB-231BrM and CN34BrM breast cancer cells that can cross the BBB [94]. Upon further exploration, miR-509 was capable of modulating the two genes, RhoC and TNF-α that control the invasion of cancer cells and the transendothelial cell migration across the BBB, respectively. Levels of MMP9 induced by TNF-α and RhoC were significantly associated with brain metastasis-free survival in breast cancer patients [94].
Claudin-5 (CLDN5) also plays a role in regulating the permeability of the BBB in lung cancer brain metastasis. Silencing of CLDN5 increased the paracellular permeability of A549 cancer cells and increased the invasion of lung adenocarcinoma [95]. The role that CLDN5 plays in tightening endothelial cellular adhesion and enhancing the function of the tight junctions reduces the formation of lung cancer brain metastasis. In TNBC, angiopoietin-2 was reported to breach CLDN5 junction protein structures and increase metastatic invasion of MDA-MB-BrM2 and 4T1 cells [22]. As such, therapeutic strategies that aim at protecting CLDN5 and targeting angiopoietin-2 should be favored.
Adenosine A2A receptors are widely distributed in the brain [96]. The activation of adenosine A2A receptors inhibit the interaction of SDF-1 and CXCR4, leading to a decrease in the brain metastatic capacity [97]. Adenosine A2A receptor activation inhibited the proliferation and viability of lung cancer PC-9 cells and thus suppressed brain metastasis in vitro [98]. Treatment using A2A receptor agonist and CXCR4 antagonist protected nude mice from brain metastasis in vivo [98]. Further investigation revealed that the activation of A2A receptor increased the expression levels of CLDN5, occludin and ZO-1 dramatically [98]. Conversely, when A2A receptor was blocked by antagonist, these junction proteins were downregulated [98]. This indicates that the activation of the A2A receptor protects the integrity and function of BBB against metastatic transmigration.
Tumor-derived extracellular vesicles can breach the intact BBB. By pretreating nude mice with extracellular vesicles derived from parental MDA-MB-231 tumor cells and then administering an intracardiac injection of tumor cells, the size and incidence of brain metastases increased significantly when compared to non-pretreated mice [99]. Gene knockout studies confirmed that suppressing the brain endothelial expression of Ras-related protein 7 (Rab7) increases the efficiency of the cancer cell transport [99]. Furthermore, high spatiotemporal resolution microscopy demonstrated that the endothelial recycling endocytic pathway is involved in this transcellular transport across the BBB [99].
Pericytes seem to possess protective qualities against metastatic cells. By studying the interaction between pericytes and metastatic A549 and KNS-62 lung cancer cells, it was observed that the expression of tight junction protein was preserved upon co-culture with pericytes [100]. In addition, resistance across the trans-endothelial pathway was left intact. The conditioned medium of pericytes with metastatic cancer cells suppressed the proliferation of the cancer cells significantly. This ultimately revealed that pericytes are major regulators of the resistance of the BBB to lung cancer metastasis to the brain [100]. Further research identified the sub-populations of pericytes that are involved in modulating metastasis in the breast cancer setting [16]. An increase of desmin+ pericytes and a decrease of CD13+ pericytes increased permeability across brain endothelial barriers, which led to the passage of large protein components, osmosis and brain edema [16]. As such, future preventive therapies targeting desmin+ pericytes can be promising in the face of scarce effective therapeutic options.
CONCLUDING REMARKS
The clinical threshold of danger in cancer is metastatic spread. Brain metastasis, per se, portends poor outcomes and low quality of life. Favorable prognosis accounts for diagnosis made prior to the growth of secondary cancerous sites. As such, understanding the various routes of transmigration across the BBB is essential for the development of new targeted therapies and for preventive measures that can target metastatic cancer cells before reaching the brain. Specific mediators of transmigration present an opportunity for novel strategies in facing the challenge raised by brain metastasis. Furthermore, BBB destruction is not limited to the formation of brain metastases. It is also involved in other diseases like stroke, trauma, Alzheimer’s, and primary brain tumors. As such, novel therapeutics focusing on the BBB can have the potential to contribute positively to other diseases as well. Nevertheless, there is a need to acknowledge the limitation of extrapolating the BBB migration strategy from current literature. Although in some cases, the mechanism of migration is clearly identified for some genes, the mechanism is not clear yet for many others. Future research must address this issue by attempting to decipher the exact mechanism of transmigration promoted by different genetic drivers.
It is also becoming clear that metastatic cells heterogeneously affect the integrity of the BBB [1, 3]. Specific subpopulations of pericytes are associated with differential permeability, and correspondingly, only drugs designed to fully penetrate the BBB will be therapeutically efficacious [101]. Moreover, questions have been raised on the role of chemotherapeutic agents in promoting brain metastasis. It has been shown that patients with HER2-positive breast cancer who received taxanes had worse prognosis and higher chance of developing brain metastasis [102, 103]. This raises concern as to whether other chemotherapeutic agents are also involved in promoting metastatic behavior and penetration of the BBB. Furthermore, the role of ionizing radiation in promoting BBB invasion has come into question in light of proteomic analyses showing that metabolic reprograming induced by radiation helps metastatic cancer cells accommodate to the brain [104].
Altogether, these accumulating studies infer that different components of the brain-tumor microenvironment can fundamentally regulate the properties of the BBB, demanding an integrated investigation of these genetic, proteomic and cellular mediators in the future to improve the efficacy of brain-targeted therapies. Moreover, diversifying cell lines and developing better experimental models is necessary to fully elucidate the mechanisms of transmigration of metastatic cells across the BBB. In addition, the development of diagnostic assays that can screen for the expression of these genes/proteins will help in the early detection and prevention efforts in the fight against cancer (see Outstanding Questions).
Outstanding Questions.
What other genes are regulating invasion across the blood-brain barrier (BBB)? Taking into consideration tumor heterogeneity, are these genetic mutations common across all invading tumor cells to the brain?
Knowing that the brain is usually the second most common site of metastasis, after lungs and bone, does the passage of cancer cells through tight tissue cause cellular stress and subsequent genetic alterations that can increase the cancer cell’s potential to invade the BBB?
Knowing that patients with recurring cancer who had received chemotherapy and radiotherapy will have a higher chance of brain metastasis, are such therapies inducing genetic mutations that drive metastatic invasion across the BBB?
If gene therapy is to be adopted to target metastatic cancer cells, what will be the length and extent of such therapeutics? Will it be convenient to translate such therapies to the clinic? Would such therapies be cost effective? Will gene therapy in combination with other therapeutic regimens offer better efficacy?
Supplementary Material
Highlights.
The blood-brain barrier (BBB) is restrictive to the extent that not even ions can pass freely from one side to another.
Most drugs are incapable of permeating through the BBB as transport is tightly regulated by efflux transporters.
Brain metastatic cells are capable of crossing the BBB through the paracellular route or the transcellular pathway.
Genetic alterations in cancer cells drive their metastatic capabilities and their ability to transmigrate through the BBB.
Funding:
This work was supported by: NIH grants R35CA197725 (M.S.L.), R01NS87990 (M.S.L.) and R01NS093903 (M.S.L.).
Glossary
- ADAM8
A gene that encodes a membrane-anchored protein implicated in cell-cell and cell-matrix interactions
- Angiopoietin-2
A biomarker in cancer; involved in assembling and disassembling the endothelial lining of blood vessels
- Annexin A1
A calcium-dependent phospholipid-binding protein located on the cytosolic face of the plasma membrane
- Brain parenchyma
The functional tissue in the brain that is composed of neurons and glial cells
- Cellular diapedesis
The movement of cells out of the circulatory system and towards the site of tissue damage, infection, or tumor
- Efflux pumps
Transport proteins involved in the extrusion of substrates (including from within cells into the external environment
- ERK1/2
Proteins that regulate meiosis, mitosis, and post-mitotic functions in differentiated cells. Disruption of the ERK pathway is common in cancers
- FAK
A protein kinase involved in cellular adhesion and spreading processes
- FKHRL1
Transcription factor that plays important roles in regulating the expression of genes involved in cell growth, proliferation, differentiation, and longevity
- Gangliosides
Components of the cell plasma membrane that modulate cell signal transduction events. They can serve as specific determinants in cellular recognition and cell-to-cell communication
- GBP1
A protein that is induced by interferons and characterized by its ability to specifically bind guanine nucleotides
- MAPK
A protein kinase that regulates cellular proliferation, gene expression, differentiation, mitosis, cell survival, and apoptosis
- miRNAs
A small non-coding RNA molecule that functions in RNA silencing and post-transcriptional regulation of gene expression
- NF-κB
A protein complex that controls transcription of DNA, cytokine production, and cell survival
- PI3K
An enzyme involved in cellular functions such as cell growth, proliferation, differentiation, motility, survival and intracellular trafficking
- Proteases
An enzyme which breaks down proteins and peptides
- Seprase
An integral membrane serine peptidase that promotes cell invasiveness towards the ECM and support tumor growth and proliferation
- Tight junctions
A cell-cell junction found barriers that are impermeable to the majority of soluble molecules between the two sides of the barrier. Major components include claudin and occludin proteins
- Tumor heterogeneity
The observation that different tumor cells within the same tumor can show distinct morphological and phenotypic profiles
- uPA
An enzyme that is made in the kidney and found in the urine. It is used to dissolve blood clots or to prevent them from forming
- VEGF
A potent angiogenic factor
- ZO-1
A scaffold protein that cross-links and anchors Tight Junction strand proteins
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest: None declared
REFERENCES
- 1.Fares J et al. (2020) Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduction and Targeted Therapy 5 (1), 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cheng H and Perez-Soler R (2018) Leptomeningeal metastases in non-small-cell lung cancer. The Lancet Oncology 19 (1), e43–e55. [DOI] [PubMed] [Google Scholar]
- 3.Arvanitis CD et al. (2020) The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nature Reviews Cancer 20 (1), 26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fares J et al. (2020) Landscape of combination therapy trials in breast cancer brain metastasis. Int J Cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pardridge WM (2005) The blood-brain barrier: bottleneck in brain drug development. NeuroRx 2 (1), 3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Langley RR and Fidler IJ (2013) The biology of brain metastasis. Clin Chem 59 (1), 180–9. [DOI] [PubMed] [Google Scholar]
- 7.Fares J et al. (2019) Current state of clinical trials in breast cancer brain metastases. Neuro-oncology practice 6 (5), 392–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fares J et al. (2019) Diagnostic Clinical Trials in Breast Cancer Brain Metastases: Barriers and Innovations. Clinical breast cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fares J et al. (2019) Immune checkpoint inhibitors: Advances and impact in neuro-oncology. Surg Neurol Int 10, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Berghoff AS and Preusser M (2018) Role of the blood–brain barrier in metastatic disease of the central nervous system In Handbook of clinical neurology, pp. 57–66, Elsevier. [DOI] [PubMed] [Google Scholar]
- 11.Obermeier B et al. (2013) Development, maintenance and disruption of the blood-brain barrier. Nat Med 19 (12), 1584–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gee JR and Keller JN (2005) Astrocytes: regulation of brain homeostasis via apolipoprotein E. Int J Biochem Cell Biol 37 (6), 1145–50. [DOI] [PubMed] [Google Scholar]
- 13.Lipinski CA et al. (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46 (1–3), 3–26. [DOI] [PubMed] [Google Scholar]
- 14.Ulasov I et al. (2020) MicroRNA 345 (miR345) regulates KISS1-E-cadherin functional interaction in breast cancer brain metastases. Cancer Lett 481, 24–31. [DOI] [PubMed] [Google Scholar]
- 15.Lockman PR et al. (2010) Heterogeneous blood–tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clinical cancer research 16 (23), 5664–5678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lyle LT et al. (2016) Alterations in Pericyte Subpopulations Are Associated with Elevated Blood-Tumor Barrier Permeability in Experimental Brain Metastasis of Breast Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 22 (21), 5287–5299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Taskar KS et al. (2012) Lapatinib distribution in HER2 overexpressing experimental brain metastases of breast cancer. Pharm Res 29 (3), 770–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Thorsen F et al. (2013) Multimodal imaging enables early detection and characterization of changes in tumor permeability of brain metastases. J Control Release 172 (3), 812–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Adkins CE et al. (2013) P-glycoprotein mediated efflux limits substrate and drug uptake in a preclinical brain metastases of breast cancer model. Front Pharmacol 4, 136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fazakas C et al. (2011) Transmigration of melanoma cells through the blood-brain barrier: role of endothelial tight junctions and melanoma-released serine proteases. PloS one 6 (6), e20758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sarin H et al. (2009) Physiologic upper limit of pore size in the blood-tumor barrier of malignant solid tumors. J Transl Med 7, 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Avraham HK et al. (2014) Angiopoietin-2 mediates blood-brain barrier impairment and colonization of triple-negative breast cancer cells in brain. J Pathol 232 (3), 369–81. [DOI] [PubMed] [Google Scholar]
- 23.Sevenich L et al. (2014) Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S. Nature cell biology 16 (9), 876–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Artym VV et al. (2002) Molecular proximity of seprase and the urokinase-type plasminogen activator receptor on malignant melanoma cell membranes: dependence on β1 integrins and the cytoskeleton. Carcinogenesis 23 (10), 1593–1601. [DOI] [PubMed] [Google Scholar]
- 25.Fodstad O et al. (1988) A new experimental metastasis model in athymic nude mice, the human malignant melanoma LOX. International journal of cancer 41 (3), 442–9. [DOI] [PubMed] [Google Scholar]
- 26.Aoyama A and Chen WT (1990) A 170-kDa membrane-bound protease is associated with the expression of invasiveness by human malignant melanoma cells. Proceedings of the National Academy of Sciences of the United States of America 87 (21), 8296–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Monsky WL et al. (1994) A potential marker protease of invasiveness, seprase, is localized on invadopodia of human malignant melanoma cells. Cancer research 54 (21), 5702–10. [PubMed] [Google Scholar]
- 28.Kelly T et al. (1998) Seprase, a membrane-bound protease, is overexpressed by invasive ductal carcinoma cells of human breast cancers. Modern Pathology 11 (9), 855–863. [PubMed] [Google Scholar]
- 29.HolstHansen C et al. (1996) Urokinase-type plasminogen activation in three human breast cancer cell lines correlates with their in vitro invasiveness. Clinical & Experimental Metastasis 14 (3), 297–307. [DOI] [PubMed] [Google Scholar]
- 30.Duffy MJ (1996) Proteases as prognostic markers in cancer. Clinical Cancer Research 2 (4), 613–618. [PubMed] [Google Scholar]
- 31.Verbovsek U et al. (2015) Complexity of cancer protease biology: Cathepsin K expression and function in cancer progression. Seminars in Cancer Biology 35, 71–84. [DOI] [PubMed] [Google Scholar]
- 32.Zhu LX et al. (2016) Downregulation of ubiquitin-specific protease 14 (USP14) inhibits breast cancer cell proliferation and metastasis, but promotes apoptosis. Journal of Molecular Histology 47 (1), 69–80. [DOI] [PubMed] [Google Scholar]
- 33.Suaifan GARY et al. (2017) Proteinases as Biomarkers in Breast Cancer Prognosis and Diagnosis. Mini-Reviews in Medicinal Chemistry 17 (7), 583–592. [DOI] [PubMed] [Google Scholar]
- 34.Hofmann UB et al. (1999) Matrix metalloproteinases in human melanoma cell lines and xenografts: increased expression of activated matrix metalloproteinase-2 (MMP-2) correlates with melanoma progression. British Journal of Cancer 81 (5), 774–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sounni NE et al. (2002) Expression of membrane type 1 matrix metalloproteinase (MT1-MMP) in A2058 melanoma cells is associated with MMP-2 activation and increased tumor growth and vascularization. International Journal of Cancer 98 (1), 23–28. [DOI] [PubMed] [Google Scholar]
- 36.Tsung AJ et al. (2008) Downregulation of matrix metalloproteinase-2 (MMP-2) utilizing adenovirus-mediated transfer of small interfering RNA (siRNA) in a novel spinal metastatic melanoma model. International Journal of Oncology 32 (3), 557–564. [PMC free article] [PubMed] [Google Scholar]
- 37.Rizzo A et al. (2015) Melanoma Cells Homing to the Brain: An In Vitro Model. Biomed Research International. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu H et al. (2012) The role of MMP-1 in breast cancer growth and metastasis to the brain in a xenograft model. BMC cancer 12, 583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shen C-J et al. (2017) MMP1 expression is activated by Slug and enhances multi-drug resistance (MDR) in breast cancer. PloS one 12 (3), e0174487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mendes O et al. (2007) MMP2 role in breast cancer brain metastasis development and its regulation by TIMP2 and ERK1/2. Clinical & experimental metastasis 24 (5), 341–51. [DOI] [PubMed] [Google Scholar]
- 41.Conrad C et al. (2018) ADAM8 expression in breast cancer derived brain metastases: Functional implications on MMP-9 expression and transendothelial migration in breast cancer cells. International journal of cancer 142 (4), 779–791. [DOI] [PubMed] [Google Scholar]
- 42.Murphy G (2009) Regulation of the proteolytic disintegrin metalloproteinases, the ‘Sheddases’. Semin Cell Dev Biol 20 (2), 138–45. [DOI] [PubMed] [Google Scholar]
- 43.Liu W et al. (2019) AKR1B10 (Aldo-keto reductase family 1 B10) promotes brain metastasis of lung cancer cells in a multi-organ microfluidic chip model. Acta Biomater 91, 195–208. [DOI] [PubMed] [Google Scholar]
- 44.Li B et al. (2013) Elevated PLGF contributes to small-cell lung cancer brain metastasis. Oncogene 32 (24), 2952–62. [DOI] [PubMed] [Google Scholar]
- 45.Parr C et al. (2005) Placenta growth factor is over-expressed and has prognostic value in human breast cancer. European journal of cancer (Oxford, England : 1990) 41 (18), 2819–27. [DOI] [PubMed] [Google Scholar]
- 46.Argaw AT et al. (2012) Astrocyte-derived VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease. J Clin Invest 122 (7), 2454–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wei SC et al. (2005) Placenta growth factor expression is correlated with survival of patients with colorectal cancer. Gut 54 (5), 666–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chen C-N et al. (2004) The significance of placenta growth factor in angiogenesis and clinical outcome of human gastric cancer. Cancer letters 213 (1), 73–82. [DOI] [PubMed] [Google Scholar]
- 49.Kienast Y et al. (2010) Real-time imaging reveals the single steps of brain metastasis formation. Nature medicine 16 (1), 116–22. [DOI] [PubMed] [Google Scholar]
- 50.Denkins Y et al. (2004) Brain metastases in melanoma: roles of neurotrophins. Neuro-oncology 6 (2), 154–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ilan N et al. (2006) Regulation, function and clinical significance of heparanase in cancer metastasis and angiogenesis. The international journal of biochemistry & cell biology 38 (12), 2018–39. [DOI] [PubMed] [Google Scholar]
- 52.Murry BP et al. (2006) Heparanase mechanisms of melanoma metastasis to the brain: Development and use of a brain slice model. Journal of cellular biochemistry 97 (2), 217–25. [DOI] [PubMed] [Google Scholar]
- 53.Liu T et al. (2015) Visfatin Mediates SCLC Cells Migration across Brain Endothelial Cells through Upregulation of CCL2. International journal of molecular sciences 16 (5), 11439–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gril B et al. (2018) Reactive astrocytic S1P3 signaling modulates the blood-tumor barrier in brain metastases. Nature communications 9 (1), 2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhou W et al. (2014) Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer cell 25 (4), 501–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tominaga N et al. (2015) Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nature communications 6, 6716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang H et al. (2019) N6-methyladenosine induced miR-143–3p promotes the brain metastasis of lung cancer via regulation of VASH1. Mol Cancer 18 (1), 181. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 58.Rodrigues G et al. (2019) Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat Cell Biol 21 (11), 1403–1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Doron H et al. (2019) A Blazing Landscape: Neuroinflammation Shapes Brain Metastasis. Cancer Res 79 (3), 423–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Klotz R et al. (2020) Circulating Tumor Cells Exhibit Metastatic Tropism and Reveal Brain Metastasis Drivers. Cancer Discov 10 (1), 86–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Worzfeld T and Offermanns S (2014) Semaphorins and plexins as therapeutic targets. Nature Reviews Drug Discovery 13 (8), 603–621. [DOI] [PubMed] [Google Scholar]
- 62.Smith ES et al. (2015) SEMA4D compromises blood-brain barrier, activates microglia, and inhibits remyelination in neurodegenerative disease. Neurobiol Dis 73, 254–68. [DOI] [PubMed] [Google Scholar]
- 63.Zhou YF et al. (2018) Sema4D/PlexinB1 inhibition ameliorates blood-brain barrier damage and improves outcome after stroke in rats. FASEB J 32 (4), 2181–2196. [DOI] [PubMed] [Google Scholar]
- 64.Yousefi M et al. (2017) Lung cancer-associated brain metastasis: Molecular mechanisms and therapeutic options. Cellular Oncology 40 (5), 419–441. [DOI] [PubMed] [Google Scholar]
- 65.Li B et al. (2006) Involvement of Rho/ROCK signalling in small cell lung cancer migration through human brain microvascular endothelial cells. Febs Letters 580 (17), 4252–4260. [DOI] [PubMed] [Google Scholar]
- 66.Wojciak-Stothard B and Ridley AJ (2002) Rho GTPases and the regulation of endothelial permeability. Vascular Pharmacology 39 (4–5), 187–199. [DOI] [PubMed] [Google Scholar]
- 67.Vogetseder A et al. (2013) αv-Integrin isoform expression in primary human tumors and brain metastases. International journal of cancer 133 (10), 2362–2371. [DOI] [PubMed] [Google Scholar]
- 68.WU YJ et al. (2017) High αv Integrin Level of Cancer Cells Is Associated with Development of Brain Metastasis in Athymic Rats. Anticancer Research 37 (8), 4029–4040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Garcia-Martin AB et al. (2018) VLA-4 mediated adhesion of melanoma cells on the blood-brain barrier is the critical cue for melanoma cell intercalation and barrier disruption. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism, 271678X18775887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Münsterberg J et al. (2020) ALCAM contributes to brain metastasis formation in non-small-cell lung cancer through interaction with the vascular endothelium. Neuro-Oncology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bos PD et al. (2009) Genes that mediate breast cancer metastasis to the brain. Nature 459 (7249), 1005–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Gupta GP et al. (2007) Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446 (7137), 765–70. [DOI] [PubMed] [Google Scholar]
- 73.Miyamoto S et al. (2006) Heparin-binding epidermal growth factor-like growth factor as a novel targeting molecule for cancer therapy. Cancer science 97 (5), 341–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Okajima T et al. (1999) Molecular cloning of brain-specific GD1α synthase (ST6GalNAc V) containing CAG/glutamine repeats. Journal of Biological Chemistry 274 (43), 30557–30562. [DOI] [PubMed] [Google Scholar]
- 75.Drolez A et al. (2016) ST6GALNAC5 Expression Decreases the Interactions between Breast Cancer Cells and the Human Blood-Brain Barrier. International journal of molecular sciences 17 (8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Laimito KR et al. (2016) Characterisation of the triple negative breast cancer phenotype associated with the development of central nervous system metastases. Ecancermedicalscience 10, 632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Stremitzer S et al. (2017) Genetic variants associated with colorectal brain metastases susceptibility and survival. Pharmacogenomics J 17 (1), 29–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jilaveanu LB et al. (2015) PLEKHA5 as a Biomarker and Potential Mediator of Melanoma Brain Metastasis. Clinical cancer research : an official journal of the American Association for Cancer Research 21 (9), 2138–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yamada K et al. (2012) Identification and characterization of splicing variants of PLEKHA5 (Plekha5) during brain development. Gene 492 (1), 270–275. [DOI] [PubMed] [Google Scholar]
- 80.Lee B-C et al. (2004) Involvement of the chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1alpha in breast cancer cell migration through human brain microvascular endothelial cells. Molecular cancer research : MCR 2 (6), 327–38. [PubMed] [Google Scholar]
- 81.Khuon S et al. (2010) Myosin light chain kinase mediates transcellular intravasation of breast cancer cells through the underlying endothelial cells: a three-dimensional FRET study. Journal of cell science 123 (Pt 3), 431–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mustafa DAM et al. (2018) T lymphocytes facilitate brain metastasis of breast cancer by inducing Guanylate-Binding Protein 1 expression. Acta neuropathologica 135 (4), 581–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Liu Y et al. (2015) Brain microvascular endothelium induced-annexin A1 secretion contributes to small cell lung cancer brain metastasis. The international journal of biochemistry & cell biology 66, 11–9. [DOI] [PubMed] [Google Scholar]
- 84.Malin D et al. (2014) αB-crystallin: a novel regulator of breast cancer metastasis to the brain. Clinical Cancer Research 20 (1), 56–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Sayyad MR et al. (2019) Syndecan-1 facilitates breast cancer metastasis to the brain. Breast cancer research and treatment. [DOI] [PubMed] [Google Scholar]
- 86.Jassam SA et al. (2019) Fucosyltransferase 4 and 7 mediates adhesion of non-small cell lung cancer cells to brain-derived endothelial cells and results in modification of the blood–brain-barrier: in vitro investigation of CD15 and CD15s in lung-to-brain metastasis. Journal of Neuro-Oncology 143 (3), 405–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jassam SA et al. (2016) TNF-alpha enhancement of CD62E mediates adhesion of non-small cell lung cancer cells to brain endothelium via CD15 in lung-brain metastasis. Neuro Oncol 18 (5), 679–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Tiwary S et al. (2018) Metastatic Brain Tumors Disrupt the Blood-Brain Barrier and Alter Lipid Metabolism by Inhibiting Expression of the Endothelial Cell Fatty Acid Transporter Mfsd2a. Scientific reports 8 (1), 8267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Alakbarzade V et al. (2015) A partially inactivating mutation in the sodium-dependent lysophosphatidylcholine transporter MFSD2A causes a non-lethal microcephaly syndrome. Nat Genet 47 (7), 814–7. [DOI] [PubMed] [Google Scholar]
- 90.Guemez-Gamboa A et al. (2015) Inactivating mutations in MFSD2A, required for omega-3 fatty acid transport in brain, cause a lethal microcephaly syndrome. Nat Genet 47 (7), 809–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ben-Zvi A et al. (2014) Mfsd2a is critical for the formation and function of the blood–brain barrier. Nature 509 (7501), 507–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.O’Brown NM et al. (2019) Suppression of transcytosis regulates zebrafish blood-brain barrier function. Elife 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Andreone BJ et al. (2017) Blood-Brain Barrier Permeability Is Regulated by Lipid Transport-Dependent Suppression of Caveolae-Mediated Transcytosis. Neuron 94 (3), 581–594.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Xing F et al. (2015) miR-509 suppresses brain metastasis of breast cancer cells by modulating RhoC and TNF-alpha. Oncogene 34 (37), 4890–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ma SC et al. (2017) Claudin-5 regulates blood-brain barrier permeability by modifying brain microvascular endothelial cell proliferation, migration, and adhesion to prevent lung cancer metastasis. CNS Neurosci Ther 23 (12), 947–960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Stockwell J et al. (2017) Adenosine A1 and A2A Receptors in the Brain: Current Research and Their Role in Neurodegeneration. Molecules 22 (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zhou Y et al. (2018) The CXCL12 (SDF-1)/CXCR4 chemokine axis: Oncogenic properties, molecular targeting, and synthetic and natural product CXCR4 inhibitors for cancer therapy. Chinese Journal of Natural Medicines 16 (11), 801–810. [DOI] [PubMed] [Google Scholar]
- 98.Chen L et al. (2020) Adenosine A2A receptor activation reduces brain metastasis via SDF-1/CXCR4 axis and protecting blood-brain barrier. Mol Carcinog 59 (4), 390–398. [DOI] [PubMed] [Google Scholar]
- 99.Morad G et al. (2019) Tumor-Derived Extracellular Vesicles Breach the Intact Blood-Brain Barrier via Transcytosis. ACS Nano 13 (12), 13853–13865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Fujimoto T et al. (2019) Pericytes Suppress Brain Metastasis from Lung Cancer In Vitro. Cell Mol Neurobiol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sprowls SA et al. (2019) Improving CNS Delivery to Brain Metastases by Blood-Tumor Barrier Disruption. Trends Cancer 5 (8), 495–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Altundag K (2018) Taxane use in breast cancer and risk of brain metastases. Medical oncology (Northwood, London, England) 35 (4), 44. [DOI] [PubMed] [Google Scholar]
- 103.Amelot A et al. (2018) Can anticancer chemotherapy promote the progression of brain metastases? Medical oncology (Northwood, London, England) 35 (3), 35. [DOI] [PubMed] [Google Scholar]
- 104.Kalita-de Croft P et al. (2019) Proteomic Analysis of the Breast Cancer Brain Metastasis Microenvironment. International journal of molecular sciences 20 (10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bors LA and Erdő F (2019) Overcoming the Blood–Brain Barrier. Challenges and Tricks for CNS Drug Delivery. Scientia Pharmaceutica 87 (1), 6. [Google Scholar]
- 106.Fares J et al. (2019) Neurogenesis in the adult hippocampus: history, regulation, and prospective roles. Int J Neurosci 129 (6), 598–611. [DOI] [PubMed] [Google Scholar]
- 107.Sivandzade F and Cucullo L (2018) In-vitro blood-brain barrier modeling: A review of modern and fast-advancing technologies. J Cereb Blood Flow Metab 38 (10), 1667–1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Dekmak A et al. (2018) Stem cells and combination therapy for the treatment of traumatic brain injury. Behav Brain Res 340, 49–62. [DOI] [PubMed] [Google Scholar]
- 109.Abbott NJ et al. (2014) In vitro models of CNS barriers In Drug Delivery to the brain, pp. 163–197, Springer. [Google Scholar]
- 110.Naik P and Cucullo L (2012) In vitro blood-brain barrier models: current and perspective technologies. J Pharm Sci 101 (4), 1337–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Smith QR (2003) A review of blood-brain barrier transport techniques. Methods Mol Med 89, 193–208. [DOI] [PubMed] [Google Scholar]
- 112.Kuhnline Sloan CD et al. (2012) Analytical and biological methods for probing the blood-brain barrier. Annu Rev Anal Chem (Palo Alto Calif) 5, 505–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pike VW (2009) PET radiotracers: crossing the blood-brain barrier and surviving metabolism. Trends Pharmacol Sci 30 (8), 431–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Westerink BH and Cremers TI (2007) Handbook of microdialysis: methods, applications and perspectives, Elsevier. [Google Scholar]
- 115.Li Y et al. (2017) Zebrafish: A promising in vivo model for assessing the delivery of natural products, fluorescence dyes and drugs across the blood-brain barrier. Pharmacol Res 125 (Pt B), 246–257. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
