Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2020 Jun;10(6):a037051. doi: 10.1101/cshperspect.a037051

Clinical Perspectives in Brain Metastasis

Krutika Deshpande 1,3, Ian Buchanan 1,3, Vahan Martirosian 1,3, Josh Neman 1,2,3
PMCID: PMC7263089  PMID: 31615863

Abstract

Brain metastases (BMs) are responsible for decline in neurological function, reduction in overall quality of life, and mortality from recurrent or untreatable lesions. Advances in diagnostics and imaging have led to increased detection of central nervous system (CNS) metastases in patients with progressive cancers. Improved control of extracranial systemic disease, and the limited ability of current therapeutics to cross the blood–brain barrier (BBB) also contribute to the increase in incidence of brain metastases, as tumor cells seek refuge in the brain. Surgery, chemotherapy, and/or radiation (whole-brain radiation therapy and stereotactic radiation surgery [WBRT/SRS]) are a clinically established treatment paradigm for patients with brain metastases. With the advent of genetic and molecular characterization of tumors and their immune microenvironment, clinical trials seek to include targeted drugs into the therapeutic regimen for eligible patients. Several challenges, like treatment of multiple CNS lesions, superior uptake of chemotherapy into the brain, and trials with multidisciplinary approaches, are now being clinically addressed.


Brain metastases (BMs) are the most common cause of intracranial neoplasms in adults with invasive cancers. Twenty to 45% of cancer patients are diagnosed with BMs in their lifetime (Achrol et al. 2019). Metastatic brain tumors occur at a much higher rate than both adult and pediatric primary brain tumors, and are the major cause of mortality from malignant brain disease (Tabouret et al. 2012; Owonikoko et al. 2014). Intracranial metastasis is associated with worse prognosis (<1 yr), moderate to severe neurodegeneration, and overall reduction in quality of life. At diagnosis, symptomatic patients usually present with headaches, seizures, motor weakness, and dysphasia (Rodin et al. 2016). The incidence and severity of brain metastatic disease varies according to the origin of the primary tumor, and the treatment strategy followed for the patient. Lung, breast, melanoma, colorectal, and renal cancers show most proclivity for the brain, followed more uncommonly by thyroid, gastrointestinal (GI) and prostate cancers (Valiente et al. 2018).

Systemic metastases were usually considered end-stage, and patients were only subjected to palliative therapy. But, because of the advances in control of primary tumors and extracranial metastases, as well as superior methods of early detection, patient survival has increased. This has however led to rising numbers of patients being diagnosed with BMs, with or without concomitant extracranial disease. Tumors originating from different tissues show varying latency to metastasize to the brain. This can be explained by the aggressiveness of the tumor type, modes of dissemination, development of resistance to therapy, or molecular affinity for the neuronal niche. Despite considerable advances in elucidating the cellular and molecular events underway in metastasis, few treatments have been realized and as such prognoses remains poor. The current standard of care for BMs includes surgical excision, whole-brain radiation therapy (WBRT), and stereotactic radiation surgery (SRS) combined with steroids (Lin and DeAngelis 2015). Targeted molecular therapies show little or no effect mainly because of their inability to penetrate the elusive blood–brain barrier (BBB). Thus, with limited options available, malignant tumors take refuge in the brain and escape most forms of intervention, contributing to patient mortality. The management of BMs is an urgent unmet clinical need and warrants immediate attention and investigation. This article summarizes and provides perspective on the current understanding of brain malignancies in the context of their clinical biology, diagnosis, and management. We hope that this knowledge is translated into innovative therapeutic interventions that enhance the quality and expectancy of life for affected patients.

DIAGNOSIS OF BRAIN METASTASES

Sixty to 75% of brain metastases (BMs) are symptomatic, whereas a smaller number of patients harbor central nervous system (CNS) metastases without any neurological signs. Imaging is crucial in the detection and diagnosis of BMs. It is used to confirm previously undiagnosed CNS metastases in patients with neurological symptoms, to confirm brain involvement in a person with systemic metastatic disease, and for staging and monitoring of BMs over the course of therapy. Imaging is also essential before surgery to plan safe excision of the tumor from the brain.

BMs can present as solitary or multiple lesions. Most BMs are solitary with 20% of diagnoses having two or less lesions, although 30% of cases have three or more lesions. Lung and melanoma primarily lead to multiple BMs, whereas breast, renal, colon usually present as single lesions. Contrast enhanced magnetic resonance imaging (MRI) is the preferred method of detection, but nonenhanced computed tomography (NECT) scans are often used for initial screening purposes, especially for naive patients who present with new neurological symptoms. For certain cancers like small cell lung cancers (SCLC), contrast enhanced computerized tomography (CECT) is equivalent to detection by MRI, because no survival benefit is offered by magnetic resonance imaging (MRI) versus computerized tomography (CT). On MRIs, metastases appear hypo-intense on T1 and hyperintense on T2. Gadolinium contrast MRIs are important in detection of small multiple lesions, and their use improves diagnostic confidence (Fink and Fink 2013).

Diagnosis of nonparenchymal BMs can be challenging. Pachymeningeal (dural) metastases can be hard to distinguish from meningiomas. For this, information about the previous history of systemic or CNS metastasis in the patient is key. Detection/diagnosis of leptomeningeal metastases is also difficult by CT, and therefore evaluation of the cerebrospinal fluid (CSF) in suspected cases is essential for confirmation (Sawaya 2001).

CANCER TYPE AND PROPENSITY OF BRAIN METASTASIS

Metastasis is a carefully orchestrated process involving breakage of cells from the primary tumor, gain of invasive properties, and interaction with the microenvironmental niche to establish tumors at distant sites. Clinically, different kinds of tumors show varying proclivities in their ability to successfully metastasize within the CNS. The most common primary sites are lung, breast, and melanoma, in that order. These are followed more uncommonly by thyroid, GI, and prostate cancers. Propensity of occurrence of brain metastasis can also be further ranked by clinical subtypes of solid cancers.

Lung Cancer

Lung cancer is the primary tumor in ∼40%–50% of patients diagnosed with BMs. 50% of lung cancer BMs occur at disease presentation/diagnosis, and sometimes the CNS is the only location of dissemination. This highlights the aggressive nature of the primary tumor, and the short latency period seen for lung-to-brain metastasis. Because of higher incidence, non-small-cell lung cancer (NSCLC) comprises a higher percentage of BMs than small-cell lung cancer (SCLC), but recent studies show that SCLC has a higher biological propensity for CNS spread (Lukas et al. 2017). About 7% of patients with NSCLC present with brain BMs at diagnosis and ∼25%–30% develop BMs over the course of their disease (Owen and Souhami 2014). Surgical resection has been shown to improve disease control in patients with solitary resectable lung-to-brain metastasis (LBM). A combined approach of surgery and WBRT has also improved intracranial disease control for such patients (Patchell et al. 1990; Noordijk et al. 1994; Chi and Komaki 2010), although the efficacy of this combined approach has not yet been unequivocally verified in patients with multiple or advanced LBMs. Targeted therapy is recommended for NSCLC patients with genetic mutations, but in cases of mass effect or impending brain herniation, surgery and subsequent systemic therapy are preferred. BMs are seen at diagnosis in 23.8% of ALK-rearranged tumors, and in 24.4% epidermal growth factor receptor (EGFR)-mutated NSCLC (Bulbul et al. 2018). Patients that harbor mutations in such oncogenic drivers may be treated using targeted therapies. ALK gene fusions have been reported in 2%–7% of patients with NSCLC, but might be higher in select groups (Shaw et al. 2009). Crizotinib was used as a first generation ALK inhibitor for systemic treatment, but patients developed CNS metastases within 1 yr of starting therapy (Costa et al. 2015). Crizotinib also has low CNS penetrance and is a substrate for P-glycoprotein efflux transporters (Petrelli et al. 2018), but showed CNS disease control rates (DCR) comparable to systemic DCR (Bulbul et al. 2018). Current trials are investigating methods to increase CNS availability of crizotinib, ceritinib, alectinib, brigatinib, and lorlatinib are second generation ALK tyrosine kinase inhibitors that have shown improved CNS-efficacy and increased intracranial response rates in ALK-positive NSCLC patients with BMs (Dempke et al. 2015; Bulbul et al. 2018; Petrelli et al. 2018). These drugs are more effective partly because of their increased CNS availability and BBB penetration.

Ten to 20% of patients with lung adenocarcinoma show EGFR mutations. Erlotinib and afatinib are first and second generation EGFR inhibitors that can be used as standalone treatments (without chemotherapy) for EGFR-mutated NSCLC. These drugs have moderate CNS penetrance and have showed substantial intracranial rate of response (RR) (Jamal-Hanjani and Spicer 2012; Dempke et al. 2015; Bulbul et al. 2018; Kelly et al. 2018). But, secondary EGFR T790M mutations have arisen as a mechanism of resistance to these drugs (Joo et al. 2018). Osimertinib is a United States Food and Drug Administration (USFDA)-approved third generation EGFR-mutant inhibitor effective against EGFR T790M (Leonetti et al. 2018). Osimertinib has BBB permeability greater than older EGFR inhibitors (Ballard et al. 2016; Koba et al. 2017), and was shown to be effective in a phase I trial in leptomeningeal disease in EGFR-mutant NSCLC (Yang et al. 2017).

For SCLC patients with BMs, the median overall survival (OS) is 4.9 mo, and 1.9–2.4 mo for leptomeningeal metastases. SCLC usually manifests as multiple brain lesions, and thus surgery is not a common therapeutic avenue for patients. Therapeutic WBRT is the standard of care for SCLC patients with BMs, who have never undergone prophylactic cranial irradiation (PCI). Stereotactic radiosurgery (SRS) is only performed in cases of recurrent BMs, to avoid exacerbation of cognitive decline by repeated WBRT. Systemic chemotherapy is not routinely used for SCLC patients with newly diagnosed or recurrent BMs. Several studies have investigated the use of systemic nontargeted cytotoxic drugs (e.g., cisplatin, temozolomide, etoposide) along with WBRT, and have shown some CNS RR, but OS still remains dismal (Lukas et al. 2017). There are very few indications of SCLC specific targets (Pezzuto et al. 2019) unlike NSCLC, but immunotherapy trials with antibodies targeting programmed cell death (nivolumab) and CTLA4 (ipilimumab) are underway.

Breast Cancer

Up to 30% of all breast cancer patients are diagnosed with BMs within their lifetime. Out of these, patients diagnosed with the triple-negative subtype (TNBC) show increased risk of brain metastasis, followed by the Her2+ and hormone-positive (ER+/PR+) subtypes (Niwinska et al. 2010; Witzel et al. 2016). Successful control of extracranial breast carcinoma, and emergence of advanced diagnostics has increased the incidence and diagnosis of breast-to-brain metastases (BBMs). Breast cancer cells that escape chemotherapy or surgical extraction can persist in the patient's body and eventually lead to BBMs. Breast cancer is unique in its longer latency to forming BMs (Saunus et al. 2011) and this has been associated with the acquisition of neuronal characteristics (Neman et al. 2013, 2014). A recent clinical study also substantiated that BBMs are able to acquire mutations in clinically targetable genes (HER2) and that ∼20% of Her2-negative diagnosed breast tumors showed a switch to Her2-positivity in the brain (Priedigkeit et al. 2017). This highlights the need for molecular characterization of highly invasive breast tumors for patients over the course of their disease, to avoid nonrelevant therapeutic interventions.

Currently, there are no breast-cancer specific treatments for BMs in particular, and a multivariate approach with surgery and radiation is used. For a limited number of BMs lesions (1–4), surgical resection followed by SRS or WBRT is performed. SRS is preferred over WBRT to avoid the associated neurocognitive decline, similar to treatment in other cancers with BMs. For multiple metastatic lesion (>4) or where surgery or SRS is not feasible, WBRT is performed, with a shift toward hippocampal sparing WBRT. This approach is reasonable given the low incidence of BBMs occurring in or around (5 mm margin) the hippocampus (Brosnan and Anders 2018). Currently, systemic drugs such as lapatinib are being investigated as radiosensitizers (Koo and Kim 2016). Recent trials also investigated the inclusion of prophylactic memantine (an oral NMDAR antagonist), which may delay the loss of cognitive capabilities after WBRT. Accordingly, newer regimens with combined use of memantine and hippocampal-sparing WBRT are being tested in patients (Brown et al. 2013).

Systemic targeted therapies especially for Her2+ breast cancer (trastuzumab, lapatinib) have limited CNS permeability, and are not protective against BMs. Lapatinib as monotherapy showed very meager CNS response, but showed improved efficacy in BMs patients when combined with capectabine (Bachelot et al. 2013; Petrelli et al. 2017).Targeted therapies like neratinib and tucatinib are now being investigated for CNS response and efficacy (Freedman et al. 2016, 2019; Murthy et al. 2018). PI3K and cyclin-dependent kinase (CDK) inhibitors in the context of hormone-positive breast cancers are in trials for CNS response in patients with BMs (Liu et al. 2016). For triple-negative breast cancer patients, poly(ADP-ribose) polymerase (PARP) and microtubule inhibitors are currently in trial for treatment of CNS metastases (Pegram et al. 2018).

Melanoma

BMs are a significant complication in patients diagnosed with advanced melanoma. About 20% of these patients present with BMs at diagnosis, and ∼40%–50% develop BMs during the course of the disease (Vosoughi et al. 2018). Melanoma BMs (MBMs) can present as single or multiple intracranial lesions, with better prognostic index for patients presenting with single BMs at melanoma diagnosis. The median survival for melanoma patients with BMs is 3 mo (without treatment) to ∼9 mo (with treatment) (Chukwueke et al. 2016).

Therapy for MBMs has been controversial, and is evolving as more postdiagnostic and -therapeutic data are collected. Surgery is preferred for patients with solitary MBM, and has an overall survival benefit when compared with patients who get radiation alone (Bafaloukos and Gogas 2004). Surgery has an advantage for symptomatic relief, as well as for procurement of tumor tissue for molecular characterization. WBRT has been shown to confer survival benefit to melanoma patients with favorable prognostic profile according to Karnofsky performance score (KPS), age, and number of extracranial metastases. WBRT after surgery or stereotactic radiation (SR) although, does not improve survival, but leads to better intracranial disease control. Because of observed neurocognitive decline after WBRT, hippocampal sparing WBRT, and inclusion of prophylactic memantine (an oral NMDAR antagonist), are in trials for MBM therapy. Stereotactic radiosurgery (SRS) is used to achieve local control in patients with small (<3 cm) and fewer than three brain lesions. SRS use is dependent on a variety of factors including accessibility of lesion, proximity to eloquent brain regions, and suitable candidacy of patient. About 50% of patients with advanced melanoma show BRAF mutations. Inclusion of BRAF-targeted small molecules in metastatic melanoma therapy has shown significant results, and is being tested for intracranial disease (Chukwueke et al. 2016). A retrospective study showed that patients with MBMs who were treated with BRAF inhibitor Vemurafenib showed 71% control of intracranial disease, and 50% of extracranial disease (Dummer et al. 2014). Immunotherapy, especially immune checkpoint inhibitors like CTLA4, PD-1 and PDL-1 antibodies have shown promise in metastatic melanoma. A phase-II trial with ipilimumab in patients with small and asymptomatic MBMs showed that 24% of patients showed partial response or stable disease (Margolin et al. 2012).

BARRIERS TO ENTRY

A highly regulated milieu is the cornerstone for reliable neural signaling within the CNS. The CNS must therefore be shielded from external influence at interfaces where blood and CSF come into close contact with neural elements. Several key zones of restriction have evolved to that end: the BBB, its caudal extension the blood–spinal cord barrier (BSCB) and the blood–cerebrospinal fluid barrier (BCSFB) (Fig. 1) (Abbott et al. 2010). The BBB is a nonfenestrated capillary endothelium ensheathed by a network of pericytes, astrocytic foot processes, and microglia that together constitute the neurovascular unit. This collective has the responsibility of maintaining the CNS as an immunologically and pharmacologically privileged site. Central to this role are the presence of tight junctions (zonulae occludentes), which interconnect endothelial cells and restrict paracellular diffusion between plasma and brain interstitium. In addition, low rates of pinocytosis within the endothelium limit transcellular movement of molecules while a metabolic barrier continually degrades molecules en route to the CNS to achieve their functional exclusion (Serlin et al. 2015).

Figure 1.

Figure 1.

Barriers to entry. The blood–brain barrier, the blood–cerebrospinal fluid barrier, and the blood–spinal cord barrier: (1) A primary tumor originating in the body enters the blood stream in the beginning phases of metastasis. (2–4) Circulating tumors cells in the blood stream arrive at three different locations in the central nervous system (CNS): (2) the blood–brain barrier (BBB), (3) the blood–cerebrospinal fluid barrier (BCSFB), and (4) the blood–spinal cord barrier (BSCB). Each of these barriers comprise of different molecules designed to shield the CNS from external influence. (2) The BBB is composed of tight junctions (zonulae occludens) between endothelial cells, which create a mostly impenetrable barrier. Tight junctions include transmembrane and cytoplasmic proteins consisting of occludin, claudins, and junctional adhesion molecules (JAMs). These are further associated with regulatory proteins including ZO-1, ZO-2, ZO-3, and cingulin. (3) The BCSFB is a structure, which has increased permeability compared with the BBB. BCSFB endothelial cells lack tight junctions and are thus have a leaky choroid plexus tight junction for membrane integrity. (4) The overall framework for the BSCB is similar to the BBB with main difference being in the architecture of the endothelial cells.

At the molecular level, selective permissiveness of the BBB and BSCB is the result of junctional complexes linking endothelial cells through a combination of adherens and tight junctions. Adherens junctions rely on cadherin proteins to form an intercellular conduit that couples the cytoplasm of adjacent cells and provides architectural support. Meanwhile, tight junctions consist of transmembrane and cytoplasmic proteins anchored to actin cytoskeleton; they include: occludin, claudins, and junctional adhesion molecules (JAMs). These are in turn associated with a host of regulatory proteins including cingulin, zona-occludens (ZO)-1, ZO-2, and ZO-3, among others (Abbott et al. 2010). Migration across the BBB and BCSFB under physiologic conditions offers incredible insight into the mechanisms by which tumor cells might gain entry into the CNS parenchyma during brain metastasis. For example, mononuclear cells possess the ability to pierce through the cytoplasm of BBB endothelium in a transcellular fashion, which precludes the need for tight junction rearrangement as is the case with paracellular modes of entry (Engelhardt and Wolburg 2004). Could tumor cells acquire the phenotypic metasignature of mononuclear cells to accomplish transcytoplasmic diapedesis?

Pathologic states are equally informative. Studies using the well-established animal model for multiple sclerosis, experimental allergic encephalomyelitis (EAE), have corroborated a role for transendothelial migration of immune cells, suggesting that this process can be modulated by inflammation (Wolburg et al. 2005). In addition, a causal relationship between loss of tight junction protein claudin 3 and increased BBB permeability (Wolburg et al. 2003) has been established in EAE. Altogether, this implies that up-regulation of both transcellular and paracellular migration occurs during CNS invasion with diseased states. Although the skeletal framework is the same for both BBB and BSCB, specific differences exist in the ultrastructure of the endothelial cells (Bartanusz et al. 2011). This might explain their proclivity to certain pathologies, and avenues for CNS metastasis. Extensive research into immune cell transgression across the BBB and BSCB has unearthed a cornucopia of molecular players, but despite years of active investigation, the precise details of diapedesis across an otherwise impervious barrier remain elusive.

The CSF comes into close contact with blood in two key areas, which formally make up the blood–CSF barrier (BCSFB): (1) where the arachnoid membrane envelopes the subarachnoid space and (2), where choroid plexus (CP) projects into the ventricular system (Johanson et al. 2011). Because CP microvasculature lack tight junctions, they are notorious for being porous to large molecules in contradistinction to brain endothelium. This can be explained by the absence of capillary ensheathment by astrocytic foot processes and the expression of “pore-forming” claudin-1 in choroid plexus, rather than “barrier-forming” claudins 3, 5, and 12 evident in brain. The functional BCSFB is therefore dependent on tight junctions within choroid epithelium, rather than their “leaky” endothelium. The increased permeability of the BCSFB is made evident by measuring the transendothelial resistance (TEER) of the barrier. in vitro TEER measurements of the choroid plexus are ∼150 Ω.cm2, compared with 1500 Ω.cm2 measured in the BBB in vivo (Redzic 2011).

In addition to its barrier properties, the choroid epithelium is responsible for generating CSF. CP endothelial permeability is not a static feature, but can be modulated. Whereas vascular endothelial growth factor (VEGF) secreted by CP epithelial cells has the capacity to diffuse across their basal surface and into the CP interstitium to enhance vessel porosity (Esser et al. 1998), angiopoietin-1 does the opposite (Nourhaghighi et al. 2003). This interface represents a potential point of vulnerability within CNS defenses because plasma-borne solutes, and by extension, pathogenic cells, have unfettered access to the CP interstitium lying between the choroid plexus endothelium and epithelium. Dexamethasone administered during meningitis is theorized to act by stabilizing MMP-3 in the interstitium and in so doing hinders bacterial dissemination into CSF (Tenenbaum et al. 2008). However, it is unclear what role, if any, the BCSFB plays in the spread of malignancy, particularly those prone to dissemination through CSF.

AVENUES FOR DISSEMINATION

The incidence for BMs has grown over the years, owing to increased patient survivability from evolving therapeutics and widespread availability of magnetic resonance imaging. The brain provides a refuge/sanctuary for tumor cells to escape chemotherapy, immune response, and natural clearance. Formation of successful distant metastases requires hematogenous or lymphatic transport of tumor cells. For the CNS, the most commonly known and studied route is transport via the bloodstream. Seeding can occur both in the meninges as well as parenchyma of the brain. This process requires the tumor cells to cross the BBB, according to studies so far. Several new avenues of CNS dissemination are now under consideration, and will be discussed in this section.

Seeding through the BBB

Because the brain is structurally and functionally isolated from the rest of the body by an intricate BBB, it takes longer for tumor cells to successfully invade the CNS, as compared with other organs. Studies have shown that lung cancer cells require 48 h, whereas breast cancer needs 2–7 d, and melanoma takes up to 14 d to cross the BBB (Wrobel and Toborek 2016). These results are also indicative of the intrinsic properties of these primary tumors. The molecular mechanisms of BBB disruption by brain-trophic tumors are not well known, but are an urgent area of investigation. Interference in normal functioning of endothelial cells contributes to CNS spread. Research in patient-derived xenograft (PDX) models showed that BBB is selectively disrupted in BMs through inhibition of Mfsd2a, a fatty acid transporter expressed by endothelial cells. This was accompanied by BBB leakage and loss of transforming growth factor (TGF)β and basic fibroblast growth factor (bFGF) signaling in the BMs endothelium (Tiwary et al. 2018). Exosomal microRNAs (miR-105) secreted by invasive breast cancer cells were shown to target tight junction mRNAs in brain vascular endothelial cells, thereby influencing their barrier properties (Zhou et al. 2014). Exosomal miR-181c from metastatic breast cancer cells also promotes dysregulation of the BBB in vitro through abnormal localization of endothelial N-cadherin and actin via down-regulation target gene PDPK1 (Tominaga et al. 2015).

Radiation therapy for intra- and peri-CNS tumors can also disrupt the integrity of the BBB (van Vulpen et al. 2002). Increase in endothelial and glial ICAM-1 was found after radiation-induced disruption of the BSCB in rats (Nordal and Wong 2004). ICAM-1 is essential for binding and extravasation of immune cells through the CNS vasculature (Ma et al. 2013), and could potentially be used as a gateway by tumor cells for access into the CNS.

Seeding through the CSF and BCSFB

Circulating tumor cells (CTCs) in the CSF and leptomeninges are therapeutic roadblocks, and potential “seeds” of metastasis in the brain and spine. A large number of leptomeningeal CNS metastases come from breast and lung tumors (Corbin and Nagpal 2016). Because the CSF is a nutrient-weak microenvironment, tumor cells modify this niche to their benefit. A recent study showed that invasive tumor cells in the CSF secrete complement C3. This binds to receptor C3aR on the choroid plexus cells, thus disrupting the BSCFB, and allowing unfettered access of nutrients and growth factors into the CSF (Boire et al. 2017). Further research should be conducted to analyze if disruption of the BSCFB can facilitate tumor invasion into the brain parenchyma.

Although BBB disruption is a well-research phenomenon in metastatic cancers, the role of choroid plexus (CP) cells in CNS metastasis is relatively unknown. Rare cases of intraventricular metastases have been reported from renal, lung, GI, breast, and bladder cancers (Tanimoto et al. 1991; Nakabayashi et al. 1994; Della Puppa et al. 2010; Shapira et al. 2014). Some of these have been found to be juxtaposed right alongside CP cells that line the lateral ventricles. Neuroblastoma cells were shown to cross an intact choroid plexus barrier in vitro (Vandenhaute et al. 2015). In mouse models, lymphoma cells have been shown to localize to the brain through the choroid plexus and cranial nerves (Hochman et al. 2001). These studies indicate a role for the BCSFB in CNS-metastatic cancers

Pachymeningeal Seeding Postneurosurgical Resection

Solitary metastatic lesions in the dura are rare in extracranial carcinomas, but have been reported in metastatic breast, lung, prostate cancers, and lymphoma (Fink and Fink 2013; Heo et al. 2017). When they do occur, the origin of spread is mainly hematogenous. The role of therapeutic intervention in facilitating CNS metastasis is an emerging area of research. A recent retrospective study showed that patients whose CNS metastases were treated with a combination of surgical resection and subsequent SRS, versus SRS alone, had an increased probability of developing pachymeningeal (dural/outer arachnoid) lesions away from the SRS site (Cagney et al. 2019). This suggests that individual cells that get dispersed during surgical removal can manifest as detectable tumors outside the targeted lesion region.

THERAPEUTIC PERSPECTIVES IN BRAIN METASTASES

Using conventional therapy to treat brain metastases (BMs) has limited to no efficacy. Chemotherapeutic agents’ entry and access to CNS lesions are blocked or limited by BBB. In addition, the high sensitivity of brain and its significance limit radiotherapy and radiation dosage and constrain resection margins. Therefore, neurosurgical resection of individual BMs using modern microneurosurgical techniques remains the standard of care. Decision making for brain metastasis care is determined based on the histological type of the tumors, number and location of lesions, and the molecular and genetic characteristics of the tumor. Traditionally, WBRT is the first choice after surgical resection of a single lesion or when there are several asymptomatic lesions; however, WBRT is associated with significant risk of cognitive decline. Additionally, the recurrence of the tumor leads to the failure of radiotherapy. Irradiation of the whole brain is associated with hypoxia-inducible factor-1 and stromal cell-derived factor-1 (SDF-1) production, which alternatively increases macrophage recruitment and angiogenesis at the site of the tumor invasion (Lerman et al. 2010; Wang et al. 2013). Overcoming the biological cause of the radiation resistance can allow more effective delivery of radiotherapy. Studies in murine models have shown that inhibition of Chk1 (a DNA-damage checkpoint protein) and c-Met (a receptor tyrosine kinase with downstream oncogenes) can improve the survival and response to radiation (Cullinane et al. 2007; Bhardwaj et al. 2013). Depending on tumor histology and its origin, standard chemotherapy might be implemented following the healing of the surgical site. Personalization of metastatic cancer treatment using molecular profiling to select therapies that benefit the patients while avoiding irrelevant therapies with potential toxicity is limited by poor penetration of most chemotherapeutic agents across the BBB. Mannitol has been used as a nonspecific permeabilizing agent with limited success (Siegal et al. 2000). Since then, attempts have been made to develop selective permeabilization of the BBB at the site of metastasis using intravenous tumor necrosis factor (Connell et al. 2013) and MRI-focused ultrasound combined with microbubbles (Song et al. 2018). Multiple researchers are working on developing novel targeted delivery strategies; therefore, it is likely to have more options available in the future.

Preoperative diagnosis of BMs and inspecting the microscopic edges of the lesions are challenging. Using confocal microscopy as an alternative to light microscopy has become particularly appealing for accurate detecting the edge of the metastases within the resection cavity and residual tumor (Sanai et al. 2011; Mooney et al. 2014). Recent advances have improved the ability of MRI-guided laser interstitial therapy (LITT) technology to resect lesion tissue accurately and safely with higher protection of the nearby brain tissue. Patients with poor response to traditional radiotherapy have shown good result with LITT (Sharma et al. 2016). LITT can be a good option to improve the quality and quantity of life for patients with BMs in deep locations, which are radiation-resistant. However, further studies are required to investigate its effect on neurological morbidity and progression of CNS diseases.

Each stage of brain metastasis process can be a target for personalized intervention or, at least, better prognosis regarding the molecular properties of the metastases. Metastatic tumor cells evolve after establishing themselves within the brain parenchyma. Genomic studies have shown that the molecular profile of BMs is distinctive from the primary tumor. These alterations are clinically critical in patients with inoperative BMs in which primary tumors remain as the only tissue available for genomic analysis for the selection of the proper therapy (Han and Brastianos 2017; Liao et al. 2018). In coming years, further DNA- and RNA-based high-throughput sequencing comparing primary and BMs genomic profiling will reveal additional information on the metastatic process and, subsequently, the potential individual therapies.

The growth, invasion, and colonization of the brain tumors is dependent on several microenvironment-derived signals and on different factor secreted from microglia/macrophages. Therefore, targeting the microglia/macrophage-derived signals has been considered as a potential therapy to inhibit brain tumor growth. For instance, it has been shown that BMs form gap junctions with reactive astrocytes surrounding them (Chen et al. 2016). Metastatic cells use this interaction to clear out the toxic metabolites generated by various stress conditions such as chemotherapy. Targeting gap junctions using BBB permeable drugs has shown promising result supporting the potential of targeting interactions within the microenvironment.

Currently, several immunotherapy approaches are under study for treating the brain tumor patients. Immunotherapy treatment either stimulates immune system or enhances its activity. The immune system promotes tolerance through down-regulating the immune response via the cell surface receptors referred to as immune checkpoints. These receptors are responsible for inhibiting the immune response against cancer cells. Thus, developing the inhibitors of these receptors, known as checkpoint inhibitors, have been widely used for the treatment of certain cancers. A monoclonal antibody, called ipilimumab, targeting the immune checkpoint cytotoxic T-lymphocytes-associated protein 4 (CTLA4) has been approved for treating unresectable and metastatic melanoma. The other checkpoint inhibitor, a monoclonal antibody known as nivolumab, targets immunoglobulin G4 (IgG4) and the programmed cell-death protein1 (PD-1), and is approved to be used in patients with unresectable or metastatic melanoma, previously treated with ipilimumab, and patients with BRAF-mutated melanoma earlier treated with BRAF-inhibitor (Tawbi et al. 2018). Additionally, recent clinical trials showed efficacy of nivolumab and ipilimumab combination against asymptomatic melanoma patients with BMs (Long et al. 2018).

CONCLUDING REMARKS

Brain metastasis has become a common cause of mortality in cancer patients. Around 20%–40% of cancer patients develop brain metastases (BMs) with 60%–75% of the brain metastatic patients become symptomatic. Advances in treating the primary cancers have led to patients living longer and, therefore, the patients are more likely to experience brain metastasis complications. Multiple BMs make their prognosis challenging and worsen the long-term rate of survival.

Surgical resection, radiotherapy, and chemotherapy have remained as available treatments with poor outcome. Personalized therapy through targeting specific tumor molecular pathways is predicted to dominate BM management in the future. Considering the complexity of BMs treatments, a multidisciplinary collaboration between neurosurgeons, medical oncologists, and radiation oncologists is required. Researchers and clinicians have to overcome several obstacles and challenges when designing treatment strategies for cancer patients with BMs. The progress made with targeted therapies in recent years makes us hopeful that continued research for drugs with ability to reach and treat BMs will eventually help patients to have improved quality of life and extended survival.

Footnotes

Editors: Jeffrey W. Pollard and Yibin Kang

Additional Perspectives on Metastasis: Mechanism to Therapy available at www.perspectivesinmedicine.org

REFERENCES

  1. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. 2010. Structure and function of the blood–brain barrier. Neurobiol Dis 37: 13–25. 10.1016/j.nbd.2009.07.030 [DOI] [PubMed] [Google Scholar]
  2. Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, Peters S, Arvold ND, Harsh GR, Steeg PS, et al. 2019. Brain metastases. Nat Rev Dis Primers 5: 5 10.1038/s41572-018-0055-y [DOI] [PubMed] [Google Scholar]
  3. Bachelot T, Romieu G, Campone M, Diéras V, Cropet C, Dalenc F, Jimenez M, Le Rhun E, Pierga JY, Goncalves A, et al. 2013. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol 14: 64–71. 10.1016/S1470-2045(12)70432-1 [DOI] [PubMed] [Google Scholar]
  4. Bafaloukos D, Gogas H. 2004. The treatment of brain metastases in melanoma patients. Cancer Treat Rev 30: 515–520. 10.1016/j.ctrv.2004.05.001 [DOI] [PubMed] [Google Scholar]
  5. Ballard P, Yates JW, Yang Z, Kim DW, Yang JC, Cantarini M, Pickup K, Jordan A, Hickey M, Grist M, et al. 2016. Preclinical comparison of osimertinib with other EGFR-TKIs in EGFR-mutant NSCLC brain metastases models, and early evidence of clinical brain metastases activity. Clin Cancer Res 22: 5130–5140. 10.1158/1078-0432.CCR-16-0399 [DOI] [PubMed] [Google Scholar]
  6. Bartanusz V, Jezova D, Alajajian B, Digicaylioglu M. 2011. The blood–spinal cord barrier: morphology and clinical implications. Ann Neurol 70: 194–206. 10.1002/ana.22421 [DOI] [PubMed] [Google Scholar]
  7. Bhardwaj V, Cascone T, Cortez MA, Amini A, Evans J, Komaki RU, Heymach JV, Welsh JW. 2013. Modulation of c-Met signaling and cellular sensitivity to radiation: potential implications for therapy. Cancer 119: 1768–1775. 10.1002/cncr.27965 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Boire A, Zou Y, Shieh J, Macalinao DG, Pentsova E, Massagué J. 2017. Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis. Cell 168: 1101–1113.e13. 10.1016/j.cell.2017.02.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Brosnan EM, Anders CK. 2018. Understanding patterns of brain metastasis in breast cancer and designing rational therapeutic strategies. Ann Transl Med 6: 163 10.21037/atm.2018.04.35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brown PD, Pugh S, Laack NN, Wefel JS, Khuntia D, Meyers C, Choucair A, Fox S, Suh JH, Roberge D, et al. 2013. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro Oncol 15: 1429–1437. 10.1093/neuonc/not114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bulbul A, Forde PM, Murtuza A, Woodward B, Yang H, Bastian I, Ferguson PK, Lopez-Diaz F, Ettinger DS, Husain H. 2018. Systemic treatment options for brain metastases from non-small-cell lung cancer. Oncology (Williston Park) 32: 156–163. [PubMed] [Google Scholar]
  12. Cagney DN, Lamba N, Sinha S, Catalano PJ, Bi WL, Alexander BM, Aizer AA. 2019. Association of neurosurgical resection with development of pachymeningeal seeding in patients with brain metastases. JAMA Oncol 5: 703–709. 10.1001/jamaoncol.2018.7204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, Jacob LS, Patwa R, Shah H, Xu K, et al. 2016. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533: 493–498. 10.1038/nature18268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chi A, Komaki R. 2010. Treatment of brain metastasis from lung cancer. Cancers (Basel) 2: 2100–2137. 10.3390/cancers2042100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Chukwueke U, Batchelor T, Brastianos P. 2016. Management of brain metastases in patients with melanoma. J Oncol Pract 12: 536–542. 10.1200/JOP.2016.011882 [DOI] [PubMed] [Google Scholar]
  16. Connell JJ, Chatain G, Cornelissen B, Vallis KA, Hamilton A, Seymour L, Anthony DC, Sibson NR. 2013. Selective permeabilization of the blood–brain barrier at sites of metastasis. J Natl Cancer Inst 105: 1634–1643. 10.1093/jnci/djt276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Corbin ZA, Nagpal S. 2016. Leptomeningeal metastases. JAMA oncology patient page. JAMA Oncol 2: 839–839. 10.1001/jamaoncol.2015.3502 [DOI] [PubMed] [Google Scholar]
  18. Costa DB, Shaw AT, Ou SH, Solomon BJ, Riely GJ, Ahn MJ, Zhou C, Shreeve SM, Selaru P, Polli A, et al. 2015. Clinical experience with crizotinib in patients with advanced ALK-rearranged non-small-cell lung cancer and brain metastases. J Clin Oncol 33: 1881–1888. 10.1200/JCO.2014.59.0539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cullinane C, Raleigh J, Anderes K, McArthur G. 2007. Mechanisms of radiation enhancement by the CHK1 inhibitor PF-477736. Cancer Res 67: 5386–5386. [Google Scholar]
  20. Della Puppa A, Dal Pos S, Zovato S, Orvieto E, Ciccarino P, Manara R, Zustovich F, Berti F, Gardiman MP, Scienza R. 2010. Solitary intra-ventricular brain metastasis from a breast carcinoma. J Neurooncol 97: 123–126. 10.1007/s11060-009-9988-z [DOI] [PubMed] [Google Scholar]
  21. Dempke WCM, Edvardsen K, Lu S, Reinmuth N, Reck M, Inoue A. 2015. Brain metastases in NSCL—are TKIs changing the treatment strategy? Anticancer Res 35: 5797–5806. [PubMed] [Google Scholar]
  22. Dummer R, Goldinger SM, Turtschi CP, Eggmann NB, Michielin O, Mitchell L, Veronese L, Hilfiker PR, Felderer L, Rinderknecht JD. 2014. Vemurafenib in patients with BRAFV600 mutation-positive melanoma with symptomatic brain metastases: final results of an open-label pilot study. Eur J Cancer 50: 611–621. 10.1016/j.ejca.2013.11.002 [DOI] [PubMed] [Google Scholar]
  23. Engelhardt B, Wolburg H. 2004. Mini-review: transendothelial migration of leukocytes: through the front door or around the side of the house? Eur J Immunol 34: 2955–2963. 10.1002/eji.200425327 [DOI] [PubMed] [Google Scholar]
  24. Esser S, Wolburg K, Wolburg H, Breier G, Kurzchalia T, Risau W. 1998. Vascular endothelial growth factor induces endothelial fenestrations in vitro. J Cell Biol 140: 947–959. 10.1083/jcb.140.4.947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Fink KR, Fink JR. 2013. Imaging of brain metastases. Surg Neurol Int 4: S209–S219. 10.4103/2152-7806.111298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Freedman RA, Gelman RS, Wefel JS, Melisko ME, Hess KR, Connolly RM, Van Poznak CH, Niravath PA, Puhalla SL, Ibrahim N, et al. 2016. Translational breast cancer research consortium (TBCRC) 022: a phase II trial of neratinib for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J Clin Oncol 34: 945–952. 10.1200/JCO.2015.63.0343 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Freedman RA, Gelman RS, Anders CK, Melisko ME, Parsons HA, Cropp AM, Silvestri K, Cotter CM, Componeschi KP, Marte JM, et al. 2019. TBCRC 022: a phase II trial of neratinib and capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J Clin Oncol 37: 1081–1089. 10.1200/JCO.18.01511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Han CH, Brastianos PK. 2017. Genetic characterization of brain metastases in the era of targeted therapy. Front Oncol 7: 230 10.3389/fonc.2017.00230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Heo MH, Cho YJ, Kim HK, Kim JY, Park YH. 2017. Isolated pachymeningeal metastasis from breast cancer: clinical features and prognostic factors. Breast 35: 109–114. 10.1016/j.breast.2017.07.006 [DOI] [PubMed] [Google Scholar]
  30. Hochman J, Assaf N, Deckert-Schluter M, Wiestler OD, Pe'er J. 2001. Entry routes of malignant lymphoma into the brain and eyes in a mouse model. Cancer Res 61: 5242–5247. [PubMed] [Google Scholar]
  31. Jamal-Hanjani M, Spicer J. 2012. Epidermal growth factor receptor tyrosine kinase inhibitors in the treatment of epidermal growth factor receptor-mutant non-small cell lung cancer metastatic to the brain. Clin Cancer Res 18: 938–944. 10.1158/1078-0432.CCR-11-2529 [DOI] [PubMed] [Google Scholar]
  32. Johanson CE, Stopa EG, McMillan PN. 2011. The blood–cerebrospinal fluid barrier: structure and functional significance. Methods Mol Biol 686: 101–131. 10.1007/978-1-60761-938-3_4 [DOI] [PubMed] [Google Scholar]
  33. Joo JW, Hong MH, Shim HS. 2018. Clinical characteristics of T790M-positive lung adenocarcinoma after resistance to epidermal growth factor receptor-tyrosine kinase inhibitors with an emphasis on brain metastasis and survival. Lung Cancer 121: 12–17. 10.1016/j.lungcan.2018.04.013 [DOI] [PubMed] [Google Scholar]
  34. Kelly WJ, Shah NJ, Subramaniam DS. 2018. Management of brain metastases in epidermal growth factor receptor mutant non-small-cell lung cancer. Front Oncol 8: 208 10.3389/fonc.2018.00208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Koba T, Kijima T, Takimoto T, Hirata H, Naito Y, Hamaguchi M, Otsuka T, Kuroyama M, Nagatomo I, Takeda Y, et al. 2017. Rapid intracranial response to osimertinib, without radiotherapy, in nonsmall cell lung cancer patients harboring the EGFR T790M mutation: two case reports. Medicine 96: e6087 10.1097/MD.0000000000006087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Koo T, Kim IA. 2016. Brain metastasis in human epidermal growth factor receptor 2-positive breast cancer: from biology to treatment. Radiat Oncol J 34: 1–9. 10.3857/roj.2016.34.1.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Leonetti A, Facchinetti F, Tiseo M. 2018. Upfront osimertinib in EGFR-mutated non-small cell lung cancer: is brain still a sanctuary? Ann Transl Med 6: S110 10.21037/atm.2018.11.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Lerman OZ, Greives MR, Singh SP, Thanik VD, Chang CC, Seiser N, Brown DJ, Knobel D, Schneider RJ, Formenti SC, et al. 2010. Low-dose radiation augments vasculogenesis signaling through HIF-1–dependent and -independent SDF-1 induction. Blood 116: 3669–3676. 10.1182/blood-2009-03-213629 [DOI] [PubMed] [Google Scholar]
  39. Liao L, Ji X, Ge M, Zhan Q, Huang R, Liang X, Zhou X. 2018. Characterization of genetic alterations in brain metastases from non-small cell lung cancer. FEBS Open Bio 8: 1544–1552. 10.1002/2211-5463.12501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Lin X, DeAngelis LM. 2015. Treatment of brain metastases. J Clin Oncol 33: 3475–3484. 10.1200/JCO.2015.60.9503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Liu MC, Cortés J, O'Shaughnessy J. 2016. Challenges in the treatment of hormone receptor-positive, HER2-negative metastatic breast cancer with brain metastases. Cancer Metastasis Rev 35: 323–332. 10.1007/s10555-016-9619-z [DOI] [PubMed] [Google Scholar]
  42. Long GV, Atkinson V, Lo S, Sandhu S, Guminski AD, Brown MP, Wilmott JS, Edwards J, Gonzalez M, Scolyer RA, et al. 2018. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol 19: 672–681. 10.1016/S1470-2045(18)30139-6 [DOI] [PubMed] [Google Scholar]
  43. Lukas RV, Gondi V, Kamson DO, Kumthekar P, Salgia R. 2017. State-of-the-art considerations in small cell lung cancer brain metastases. Oncotarget 8: 71223–71233. 10.18632/oncotarget.19333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Ma Q, Chen S, Klebe D, Zhang JH, Tang J. 2013. Adhesion molecules in CNS disorders: biomarker and therapeutic targets. CNS Neurol Disord Drug Targets 12: 392–404. 10.2174/1871527311312030012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Margolin K, Ernstoff MS, Hamid O, Lawrence D, McDermott D, Puzanov I, Wolchok JD, Clark JI, Sznol M, Logan TF, et al. 2012. Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol 13: 459–465. 10.1016/S1470-2045(12)70090-6 [DOI] [PubMed] [Google Scholar]
  46. Mooney MA, Zehri AH, Georges JF, Nakaji P. 2014. Laser scanning confocal endomicroscopy in the neurosurgical operating room: a review and discussion of future applications. Neurosurg Focus 36: E9 10.3171/2013.11.FOCUS13484 [DOI] [PubMed] [Google Scholar]
  47. Murthy R, Borges VF, Conlin A, Chaves J, Chamberlain M, Gray T, Vo A, Hamilton E. 2018. Tucatinib with capecitabine and trastuzumab in advanced HER2-positive metastatic breast cancer with and without brain metastases: a non-randomised, open-label, phase 1b study. Lancet Oncol 19: 880–888. 10.1016/S1470-2045(18)30256-0 [DOI] [PubMed] [Google Scholar]
  48. Nakabayashi H, Murata K, Sakaguchi M, Nakajima K, Katsuyama J. 1994. Choroid plexus metastasis from gastric cancer—case report. Neurol Med Chir (Tokyo) 34: 183–186. 10.2176/nmc.34.183 [DOI] [PubMed] [Google Scholar]
  49. Neman J, Choy C, Kowolik CM, Anderson A, Duenas VJ, Waliany S, Chen BT, Chen MY, Jandial R. 2013. Co-evolution of breast-to-brain metastasis and neural progenitor cells. Clin Exp Metastasis 30: 753–768. 10.1007/s10585-013-9576-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Neman J, Termini J, Wilczynski S, Vaidehi N, Choy C, Kowolik CM, Li H, Hambrecht AC, Roberts E, Jandial R. 2014. Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc Natl Acad Sci 111: 984–989. 10.1073/pnas.1322098111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Niwinska A, Murawska M, Pogoda K. 2010. Breast cancer brain metastases: differences in survival depending on biological subtype, RPA RTOG prognostic class and systemic treatment after whole-brain radiotherapy (WBRT). Ann Oncol 21: 942–948. 10.1093/annonc/mdp407 [DOI] [PubMed] [Google Scholar]
  52. Noordijk EM, Vecht CJ, Haaxma-Reiche H, Padberg GW, Voormolen JH, Hoekstra FH, Tans JT, Lambooij N, Metsaars JA, Wattendorff AR, et al. 1994. The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age. Int J Radiat Oncol Biol Phys 29: 711–717. 10.1016/0360-3016(94)90558-4 [DOI] [PubMed] [Google Scholar]
  53. Nordal RA, Wong CS. 2004. Intercellular adhesion molecule-1 and blood–spinal cord barrier disruption in central nervous system radiation injury. J Neuropathol Exp Neurol 63: 474–483. 10.1093/jnen/63.5.474 [DOI] [PubMed] [Google Scholar]
  54. Nourhaghighi N, Teichert-Kuliszewska K, Davis J, Stewart DJ, Nag S. 2003. Altered expression of angiopoietins during blood–brain barrier breakdown and angiogenesis. Lab Invest 83: 1211–1222. 10.1097/01.LAB.0000082383.40635.FE [DOI] [PubMed] [Google Scholar]
  55. Owen S, Souhami L. 2014. The management of brain metastases in non-small cell lung cancer. Front Oncol 4: 248 10.3389/fonc.2014.00248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Owonikoko TK, Arbiser J, Zelnak A, Shu HK, Shim H, Robin AM, Kalkanis SN, Whitsett TG, Salhia B, Tran NL, et al. 2014. Current approaches to the treatment of metastatic brain tumours. Nat Rev Clin Oncol 11: 203–222. 10.1038/nrclinonc.2014.25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Patchell RA, Tibbs PA, Walsh JW, Dempsey RJ, Maruyama Y, Kryscio RJ, Markesbery WR, Macdonald JS, Young B. 1990. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 322: 494–500. 10.1056/NEJM199002223220802 [DOI] [PubMed] [Google Scholar]
  58. Pegram MD, Zong Y, Yam C, Goetz MP, Moulder SL. 2018. Innovative strategies: targeting subtypes in metastatic breast cancer. Am Soc Clin Oncol Educ Book 38: 65–77. 10.1200/EDBK_200715 [DOI] [PubMed] [Google Scholar]
  59. Petrelli F, Ghidini M, Lonati V, Tomasello G, Borgonovo K, Ghilardi M, Cabiddu M, Barni S. 2017. The efficacy of lapatinib and capecitabine in HER-2 positive breast cancer with brain metastases: a systematic review and pooled analysis. Eur J Cancer 84: 141–148. 10.1016/j.ejca.2017.07.024 [DOI] [PubMed] [Google Scholar]
  60. Petrelli F, Lazzari C, Ardito R, Borgonovo K, Bulotta A, Conti B, Cabiddu M, Capitanio JF, Brighenti M, Ghilardi M, et al. 2018. Efficacy of ALK inhibitors on NSCLC brain metastases: a systematic review and pooled analysis of 21 studies. PLoS ONE 13: e0201425 10.1371/journal.pone.0201425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Pezzuto F, Fortarezza F, Lunardi F, Calabrese F. 2019. Are there any theranostic biomarkers in small cell lung carcinoma? J Thorac Dis 11: S102–S112. 10.21037/jtd.2018.12.14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Priedigkeit N, Hartmaier RJ, Chen Y, Vareslija D, Basudan A, Watters RJ, Thomas R, Leone JP, Lucas PC, Bhargava R, et al. 2017. Intrinsic subtype switching and acquired ERBB2/HER2 amplifications and mutations in breast cancer brain metastases. JAMA Oncol 3: 666–671. 10.1001/jamaoncol.2016.5630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Redzic Z. 2011. Molecular biology of the blood-brain and the blood–cerebrospinal fluid barriers: similarities and differences. Fluids Barriers CNS 8: 3 10.1186/2045-8118-8-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Rodin D, Banihashemi B, Wang L, Lau A, Harris S, Levin W, Dinniwell R, Millar B, Chung C, Laperriere N, et al. 2016. The brain metastases symptom checklist as a novel tool for symptom measurement in patients with brain metastases undergoing whole-brain radiotherapy. Curr Oncol 23: e239–e247. 10.3747/co.23.2936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Sanai N, Eschbacher J, Hattendorf G, Coons SW, Preul MC, Smith KA, Nakaji P, Spetzler RF. 2011. Intraoperative confocal microscopy for brain tumors: a feasibility analysis in humans. Neurosurgery 68: 282–290; discussion 290. [DOI] [PubMed] [Google Scholar]
  66. Saunus JM, Momeny M, Simpson PT, Lakhani SR, Da Silva L. 2011. Molecular aspects of breast cancer metastasis to the brain. Genet Res Int 2011: 219189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Sawaya R. 2001. Considerations in the diagnosis and management of brain metastases. Oncology (Williston Park) 15: 1144–1154; 1157–1158; discussion 1158, 1163–1165. [PubMed] [Google Scholar]
  68. Serlin Y, Shelef I, Knyazer B, Friedman A. 2015. Anatomy and physiology of the blood–brain barrier. Semin Cell Dev Biol 38: 2–6. 10.1016/j.semcdb.2015.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Shapira Y, Hadelsberg UP, Kanner AA, Ram Z, Roth J. 2014. The ventricular system and choroid plexus as a primary site for renal cell carcinoma metastasis. Acta Neurochir (Wien) 156: 1469–1474. 10.1007/s00701-014-2108-7 [DOI] [PubMed] [Google Scholar]
  70. Sharma M, Balasubramanian S, Silva D, Barnett GH, Mohammadi AM. 2016. Laser interstitial thermal therapy in the management of brain metastasis and radiation necrosis after radiosurgery: an overview. Expert Rev Neurother 16: 223–232. 10.1586/14737175.2016.1135736 [DOI] [PubMed] [Google Scholar]
  71. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, et al. 2009. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol 27: 4247–4253. 10.1200/JCO.2009.22.6993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Siegal T, Rubinstein R, Bokstein F, Schwartz A, Lossos A, Shalom E, Chisin R, Gomori JM. 2000. In vivo assessment of the window of barrier opening after osmotic blood–brain barrier disruption in humans. J Neurosurg 92: 599–605. 10.3171/jns.2000.92.4.0599 [DOI] [PubMed] [Google Scholar]
  73. Song KH, Harvey BK, Borden MA. 2018. State-of-the-art of microbubble-assisted blood–brain barrier disruption. Theranostics 8: 4393–4408. 10.7150/thno.26869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Tabouret E, Chinot O, Metellus P, Tallet A, Viens P, Gonçalves A. 2012. Recent trends in epidemiology of brain metastases: an overview. Anticancer Res 32: 4655–4662. [PubMed] [Google Scholar]
  75. Tanimoto M, Tatsumi S, Tominaga S, Kamikawa S, Nagao T, Tamaki N, Matsumoto S. 1991. Choroid plexus metastasis of lung carcinoma–case report. Neurol Med Chir (Tokyo) 31: 152–155. 10.2176/nmc.31.152 [DOI] [PubMed] [Google Scholar]
  76. Tawbi HA, Forsyth PA, Algazi A, Hamid O, Hodi FS, Moschos SJ, Khushalani NI, Lewis K, Lao CD, Postow MA, et al. 2018. Combined nivolumab and ipilimumab in melanoma metastatic to the brain. N Engl J Med 379: 722–730. 10.1056/NEJMoa1805453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Tenenbaum T, Matalon D, Adam R, Seibt A, Wewer C, Schwerk C, Galla HJ, Schroten H. 2008. Dexamethasone prevents alteration of tight junction-associated proteins and barrier function in porcine choroid plexus epithelial cells after infection with Streptococcus suis in vitro. Brain Res 1229: 1–17. 10.1016/j.brainres.2008.06.118 [DOI] [PubMed] [Google Scholar]
  78. Tiwary S, Morales JE, Kwiatkowski SC, Lang FF, Rao G, McCarty JH. 2018. Metastatic brain tumors disrupt the blood–brain barrier and alter lipid metabolism by inhibiting expression of the endothelial cell fatty acid transporter Mfsd2a. Sci Rep 8: 8267 10.1038/s41598-018-26636-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Tominaga N, Kosaka N, Ono M, Katsuda T, Yoshioka Y, Tamura K, Lötvall J, Nakagama H, Ochiya T. 2015. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood–brain barrier. Nat Commun 6: 6716 10.1038/ncomms7716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Valiente M, Ahluwalia MS, Boire A, Brastianos PK, Goldberg SB, Lee EQ, Le Rhun E, Preusser M, Winkler F, Soffietti R. 2018. The evolving landscape of brain metastasis. Trends Cancer 4: 176–196. 10.1016/j.trecan.2018.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Vandenhaute E, Stump-Guthier C, Lasierra Losada M, Tenenbaum T, Rudolph H, Ishikawa H, Schwerk C, Schroten H, Durken M, März M, et al. 2015. 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 15: 102 10.1186/s12935-015-0257-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. van Vulpen M, Kal HB, Taphoorn MJ, El-Sharouni SY. 2002. Changes in blood–brain barrier permeability induced by radiotherapy: implications for timing of chemotherapy? (Review). Oncol Rep 9: 683–688. [PubMed] [Google Scholar]
  83. Vosoughi E, Lee JM, Miller JR, Nosrati M, Minor DR, Abendroth R, Lee JW, Andrews BT, Leng LZ, Wu M, et al. 2018. Survival and clinical outcomes of patients with melanoma brain metastasis in the era of checkpoint inhibitors and targeted therapies. BMC Cancer 18: 490 10.1186/s12885-018-4374-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Wang SC, Yu CF, Hong JH, Tsai CS, Chiang CS. 2013. Radiation therapy-induced tumor invasiveness is associated with SDF-1-regulated macrophage mobilization and vasculogenesis. PLoS ONE 8: e69182 10.1371/journal.pone.0069182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Witzel I, Oliveira-Ferrer L, Pantel K, Müller V, Wikman H. 2016. Breast cancer brain metastases: biology and new clinical perspectives. Breast Cancer Res 18: 8 10.1186/s13058-015-0665-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Wolburg H, Wolburg-Buchholz K, Kraus J, Rascher-Eggstein G, Liebner S, Hamm S, Duffner F, Grote EH, Risau W, Engelhardt B. 2003. Localization of claudin-3 in tight junctions of the blood–brain barrier is selectively lost during experimental autoimmune encephalomyelitis and human glioblastoma multiforme. Acta Neuropathol 105: 586–592. [DOI] [PubMed] [Google Scholar]
  87. Wolburg H, Wolburg-Buchholz K, Engelhardt B. 2005. Diapedesis of mononuclear cells across cerebral venules during experimental autoimmune encephalomyelitis leaves tight junctions intact. Acta Neuropathol 109: 181–190. 10.1007/s00401-004-0928-x [DOI] [PubMed] [Google Scholar]
  88. Wrobel JK, Toborek M. 2016. Blood–brain barrier remodeling during brain metastasis formation. Mol Med 22: 32–40. 10.2119/molmed.2015.00207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Yang JCH, Cho BC, Kim DW, Kim SW, Lee JS, Su WC, John T, Kao SCH, Natale R, Goldman JW, et al. 2017. Osimertinib for patients (pts) with leptomeningeal metastases (LM) from EGFR-mutant non-small cell lung cancer (NSCLC): updated results from the BLOOM study. J Clin Oncol 35: 2020–2020. 10.1200/JCO.2017.35.15_suppl.2020 [DOI] [Google Scholar]
  90. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A, O'Connor ST, Chin AR, et al. 2014. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 25: 501–515. 10.1016/j.ccr.2014.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES