Abstract
Brain metastasis is an important cause of morbidity and mortality in cancer patients. Hence, the need to develop improved therapies to prevent and treat metastasis to the brain is becoming urgent. Recent studies in this area are bringing about some advanced progress on brain metastasis. It was concluded that the occurrence and poor prognosis of brain metastasis have been mostly attributed to the exclusion of anticancer drugs from the brain by the blood‐brain barrier. And several highly potent new generation targeted drugs with enhanced CNS distribution have been developed constantly. However, the noted “seed and soil” hypothesis also suggests that the outcome of metastasis depends on the relationship between unique tumor cells and the specific organ microenvironment. Moreover, increasing studies in multiple tumor types demonstrated that brain metastasis has great molecular differences between primary tumors and extracranial metastasis to a large extent. Here, the authors summarized the most common malignancies that could lead to brain metastasis—lung cancer, breast cancer and melanoma and their related mutated factors. Only by comprehending a deeper understanding of the molecular mechanisms, more effective brain‐specific therapies will be developed for brain metastasis.
Keywords: anaplastic lymphoma kinase, blood‐brain barrier, BRAF, brain metastasis, vascular endothelial growth factor
1. INTRODUCTION
Brain metastasis (BM) has been becoming a concerned and urgent public health problem,1, 2 of which the annual incidence is between 8.3 and 14.3 per 100 000 population.3 However, datum above collected prior to the advent of modern imaging techniques, the extent of BM is rather likely to be underestimated. In 2009, over 250 000 patients were diagnosed with BM in the United States.4 Once diagnosed with BM, the median survival of untreated patients is shorter than 2 months, while patients who were treated with surgery, chemotherapy and radiotherapy could be extended to 4‐6 months.5, 6 Even so, the prognosis for patients with BM is still dismal. BM is becoming a main threatening factor for cancer patient survival while extracranial cancer has been controlled to some extent.
Lung cancer, breast cancer, and melanoma are the most common causes of BM and they account for 67%‐80% BM clinical cases.7 Once these primary tumors become metastatic, risks of BM will increase rapidly. About 30%‐50% of lung cancer, particularly nonsmall cell lung cancer (NSCLC), will develop BM during the course of their diseases.8 Among metastatic breast cancer patients, approximately 10%‐16% patients develop symptomatic BM and another 10% patients are found to have asymptomatic brain involvement in postmortem.9 In addition, the melanoma which ranked third is calculated that more than half of patients with metastatic melanoma will develop BM since the rapidly changing systemic treatment in this disease.10 BM possesses distinct pathological patterns due to its different sources of tissues. The high incidence and lethality of BM makes it urgent to explore the mechanisms of BM and to find predictable drugs which are becoming the raring direction to research.
2. THE HYPOTHESIS ABOUT THE ORIGIN OF BM
In 1889, Stephen Paget proposed the “seed and soil” hypothesis. He concluded that the nonrandom pattern of metastasis was not accidental case, indeed. Certain tumor cells (namely the “seed”) had a specific affinity for the microenvironment of certain organs (namely the “soil”). In other words, metastasis resulted only when the seed and soil were appropriate.11 Based on the hypothesis, the relationship between “seed and soil” hypothesis and BM are indispensable. The study of organ‐specific metastasis to the brain has been gradually gaining recognition nowadays. Nevertheless, a lot of brain‐derived factors are developed in recent studies, including secreted proteins and microRNA‐containing exosomes which alter the brain microenvironment to facilitate the survival and growth of BM.12, 13 Extracellular vesicles (EVs) including exosomes, mediate cell to cell communication with the delivery of their contents and then adjust multiple factors of malignancy in cancer cells.14, 15 The EVs which released from brain metastatic cancer cells could induce tight junction proteins like N‐cadherin or actin filaments located by mistake, and that may lead to the destruction of the cell to cell connection. Hence, secreted factors would be messenger to maintain the long‐distance communication and help metastatic cancers affect alterations in distant sites to build the premetastatic niches.
3. THE BBB PENETRATION AND BM
The brain microenvironment has highly selective blood‐brain barrier (BBB), high‐energy consumption, and high‐nutrition demands. All of these specific characteristics contributed to its unique physiological status.16 The BBB is a protective network consisting of endothelial cells and supporting components which balance the central nervous system (CNS) microenvironment frequently. Due to the features of brain microvessels endothelium, BBB owns continuous tight junctions, decreased pinocytosis activity, and overexpressed efflux pumps.17 The BBB could enhance the abilities of surrounding extracellular matrix (ECM), basal membrane, astrocyte, and pericytes end‐foot, in order to guard effectively against the free exchange of substances between the interstitial fluid of the brain and the blood.18 To research the distinction of BBB, some findings report the method to establish in vitro BBB model using primary rat's astrocytes and microvascular endothelia cells, and through measuring trans‐endothelial electrical resistance (TEER) value, which show more closely to the characteristics of the BBB in vivo to identify the model.19
Blood‐brain barrier is a lipid membrane, so as we know only small lipid‐soluble molecules whose diameter is <1.8 nm and molecular weights <400 Da may permeate brain microvessels normally.20 Therefore, the BBB limits the access of large molecules from the blood to the brain, especially several chemotherapeutic agents because of the tight structure.21 The impermeable nature of the BBB may become an obstacle during treatment. In addition, the BBB anchors various ATP binding cassette efflux transporters including P‐glycoprotein (P‐gp), breast cancer resistance protein (BCRP) and other cancer resistance proteins, which bind to structurally diverse drugs and make them ineffective.22 These structures exist biochemically, morphologically and functionally heterogeneous in disparate regions of the brain and they always lead to the failure of BM treatment with chemotherapeutic drugs,23 so further mechanism still remains to be studied.
However, growing evidences have shown that BM could disrupt the BBB integrity. Sodium fluorescein, a hydrosoluble molecule and excluded from the brain with an intact BBB 24 was found in brain once the diameter of brain metastasis exceeded 0.5 mm.25 Additionally, the permeability of the BBB ambient the tumor area increased in a time‐dependent manner and positively related with tumor size.26 Tumor cells in the perivascular space could render endothelial altered, and lead to the leakage through the BBB.27 Some ultrastructural studies concluded that brain tumors destroy adjacent endothelium.28 Several clinical studies also support the disruption of the BBB by BM. For example, leaky blood vessels would be found through electron microscopy,29 and increased blood vessel permeability would be detected by positron emission tomography.30
When BBB was disrupted, significant responses to chemotherapy are reported. That is to say, the disruption of BBB may enable the delivery of drugs. Rosner studied 100 breast cancer patients with symptomatic BM which treated with multifarious chemotherapies, its brain‐specific objective response rate surprisingly rises to 50%.31 Meanwhile, radiation is also known to disrupt the BBB,32 but finally this way could not achieve good prognosis. At this point, the combined therapy was taken into consideration. Trastuzumab, like most other monoclonal antibodies, could not cross the intact BBB,33 and was actually 421 times lower in cerebrospinal fluid (CSF) than that in serum before any local therapy.34 However after radiotherapy, the ratio dramatically increased to 79/1. So the blood‐tumor barrier (BTB) is leakier than the intact BBB. If chemotherapy and radiation therapy were combined, it could allow delivery in brain lesions especially at advanced stages of disease.35 Nowadays, numerous techniques are developed to improve the delivery of therapeutics across the BBB, just like chemical modification of the drug,36 temporary disruption of the BBB 37 and so on. Fortunately, strategies to reinforce the delivery of therapeutics into the CNS have been popularly pursued and are initiated to undergo clinical evaluation.
4. THE MECHANISM OF BRAIN METASTASIS
4.1. Angiogenesis and brain metastasis
4.1.1. Vascular endothelial growth factor pathway
Vascular endothelial growth factor signaling plays an important role in angiogenesis and vascular permeability.38 In fact, angiogenesis is essential for efficient colonization and growth of cancer cells in the brain. As reported, brain metastatic growth of brain‐tropic tumor cells would decrease when the activity of VEGF receptor was inhibited.39 When VEGF expression in colon cancer cells and lung adenocarcinoma cells were inhibited, the incidence of BM and developed blood vessels significantly decreased.40 Furthermore, Transfection of melanoma cells with antisense VEGF coding DNA (cDNA) could reduce the formation of BM.41 Overexpression of VEGF in melanoma cells accelerated the progress of BM.42 Angiogenic pathways, such as phosphatidylinositol 3‐kinase (PI3K) and mammalian target of rapamycin (mTOR) signaling pathway which mediated by VEGF also play an important role in BM.43, 44 The higher level of phosphorylated Akt (p‐Akt), and lower level of the pathway negative regulator phosphatase and tensin homolog deleted on chromosome 10 (PTEN) were determined among clinical melanoma brain metastasis (MBM) patients.45 Relatively, the same phenomenon has not found among cancer patients that have other distant organ metastasis like lung and liver. The secretion of VEGF could be mediated by hypoxia inducible factor 1‐alpha (HIF‐1α), and that plays a critical role in neovascularization.46 As known, once Akt was phosphorylated, several downstream pathways that strongly related to tumor metastasis would be activated. The gene Snail could be upregulated in metastatic cells, and activated by a number of pathways, including HIF‐1, Notch and nuclear factor kappa B (NF‐κB). Meanwhile, Snail regulates the transcription and expression of E‐cadherin, and further promote Epithelial‐mesenchymal transition (EMT) and cell invasion.47 During the tumor metastasis, nutrients and oxygen are mainly supported by the generation of blood vessels. The increase of HIF‐1α can promote the transcription of VEGF and accelerate the process of BM.48 Hence, these results informed that VEGF expression is necessary for the production of BM.
Clinical researchers found that patients with BM will benefit from the employ of VEGFR kinase inhibitor like vatalanib, cediranib or VEGF antibody like bevacizumab. Several phase I and II studies related to bevacizumab are in various stages of development. Whether using bevacizumab alone or combined with other antineoplastic agents are both tested in BM from breast cancer and melanoma. Meanwhile, other drugs like lapatinib and pazopanib are able to prevent the formation of metastasis by brain‐tropism breast cancer cells.49, 50 Certainly, the cross‐talk between the human epidermal growth factor receptor‐2 (HER‐2) and VEGF pathways also attracted some researchers’ attention. The dual combination of anti‐VEGF therapy and HER‐2 inhibition, such as trastuzumab accompany with lapatinib, showed the best efficacy in preclinical models of breast cancer brain metastasis (BCBM).51 Sunitinib is a small molecule and tyrosine kinase inhibitor (TKI), which targets the VEGF receptors 1‐3 and the platelet‐derived growth factor (PDGF) receptors A and B. Attributed to its excellent BBB penetration, well prognosis would be achieved.52 Other antiangiogenic agents are undergoing experiments in clinical patients, and some of tested drugs may further expand the function of VEGF inhibitors in BM therapy.
4.1.2. Epidermal growth factor receptor pathway
The epidermal growth factor receptor (EGFR) is closely related to the HER‐2 receptor, and both are belong to the ErbB family. As known, EGFR mutations, being deemed to be a biological marker of NSCLC in recent decades, account for 10%‐25% of NSCLC.53 As a result of its constitutive activation of EGFR signaling and oncogenic transformation, EGFR was confirmed to be an independent risk factor and served as a crucial role.54 So, it is meaningful to evaluate metastatic characteristics in patients with EGFR mutation during clinical screening and treatment.55
Epidermal growth factor receptor is also a vital member of receptor tyrosine kinase (RTKs). Therefore, it certainly has cross‐talk with numerous biological effects induced by VEGF. A clinical research involving 52 BCBMs patients and 12 matched primary breast cancers indicates that the expression of p‐Akt, p‐S6, and lack of PTEN was observed as 75%, 69%, and 25% separately for BCBMs and as 67%, 58%, 83% for primary breast cancers.56 Both EGFR and PTEN alterations were closely associated with primary triple‐negative breast cancer (TNBC) and high risk of brain relapse.57 On the other hand, RAS/Raf/ERK is also related to EGFR. More than 60% of brain metastatic melanoma patients have BRAF mutations accompanied by the activation of mitogen‐activated protein kinase (MAPK) pathway. In addition, the growth of MBM cells could be more effectively inhibited in vitro if combined treatment of MAPK (BRAF) inhibitor vemurafenib and mTOR inhibitor temsirolimus.58 In EGFR‐mutated NSCLC, EGFR could induce MET phosphorylation through the RAS/ERK/p38MAPK pathway, and then enhance NSCLC invasion and even metastasis to brain.59 In summary, multi‐target combination therapy focus on tumor angiogenesis will be better for BM therapy in a manner. The first‐generation EGFR TKIs, gefitinib and erlotinib exhibit variability and short‐term effect after long‐term clinical practice. Poor capability to penetrate the BBB may be the dominating cause.60, 61 The afatinib, second‐generation EGFR TKI, was accessed to a phase II study of BCBM, but failed to show more benefits during the course of treatment.62 According to the above results, lots of researchers put forward the subtle relationship between BBB permeability and tumor‐resistance protein that could remove toxins, drugs, or chemotherapies from the CNS. It would be a primary reason of failure to some extent.63 Besides, the EGFR T790M mutation could be another important mechanism for resistance to EGFR TKIs.64 Repeated biopsy showed that it may be responsible for half of acquired resistance cases.65 Several third‐generation EGFR TKIs have been developed particularly target the T790M mutation, including HM61713, EGF816, and ASP8273, with response rates ranging from 31% to 54%.66 The objective response rate (ORR) of osimertinib is over 60%, which has been confirmed in a phase I study and two phase II studies. Its median progression‐free survival (PFS) is 11 months for T790M‐positive NSCLC. Therefore, it is promising to take efforts to develop specific brain penetrant EGFR inhibitors. Whole‐brain radiation therapy (WBRT) combined with EGFR TKIs appears to be a safe way. However it should be adequately studied.67 Figure 1 provides angiogenesis and brain metastasis, including classic pathway and representative drugs.
Figure 1.
The Molecular Mechanism of Angiogenesis Regulating Brain Metastasis and its Representative Molecules
4.2. Signaling kinase and brain metastasis
4.2.1. Anaplastic lymphoma kinase (ALK) rearranged
Approximately 5% of NSCLC patients had the rearrangement in the anaplastic lymphoma kinase (ALK) gene.68 The incidence of BM in patients with ALK+ NSCLC ranges from 20% to 30%, which could be compared with those observed in EGFR‐mutated NSCLC patients.69, 70 Furthermore, ALK‐rearranged NSCLC patients who have not treated with ALK therapy, exhibited a high incidence of CNS metastasis from approximately 45%‐70%, implying that BM is the most common pattern in ALK+ NSCLC with therapy failure.71, 72 The production of anaplastic lymphoma kinase with the echinoderm microtubule‐associated protein‐like 4 (EML4‐ALK) fusion tyrosine kinase is the most common changes.73 The promoter of EML4 is located upstream of the intracellular tyrosine kinase of ALK, resulting in activation of the fusion gene and expressing the EML4‐ALK fusion protein. ALK experiences autophosphorylation in the absence of ligand, and then activates downstream cell signaling pathways leading to malignant transformation of cells.74
The EML4‐ALK fusion gene can directly phosphorylate signal transducer and activator of transcription 3 (STAT3) or activate janus‐family tyrosine kinase 3 (JAK3), while resulting in the activation of STAT3 indirectly. Through upregulating anti‐apoptotic molecules such as b‐cell leukemia 2 protein (BCL‐2) and b‐cell leukemia XL protein (BCL‐XL), STAT3 regulates cell cycle and inhibits cell apoptosis.75 The ALK fusion protein also plays a role of linker molecule which interacts with downstream molecules during signal transduction. Its specific amino acid residues respectively bind to intracytoplasmic insulin receptor substrate 1 (IRS1), v‐src sarcoma [Schmidt‐Ruppin A‐2] viral oncogene homolog [avian] (SRC) and SRC homology 2 domain‐containing (SHC), and sequentially activate Ras/ERK pathway, simultaneously activate mTOR and its downstream ribosomal protein S6 kinase (p70S6K) and S6 ribosomal protein (S6RP), in final stimulate gene transcription and promote ribosome formation.76 Besides STAT3 and extracellular signal‐regulated kinase (ERK), PI3K also took part in regulation of ALK+ NSCLC survival and anti‐apoptosis. Activated Akt1/2 could phosphorylate forkhead box O3 (FOXO3), so that the apoptotic gene was inhibited. And this cascade reaction would promote cell survival and accelerate cell cycle from G1 to S phase through upregulating cyclin D2 at the same time. In addition, phosphorylation of eukaryotic initiation factor 4E (eIF4E) and other transcription factors can upregulate the expression of anti‐apoptosis‐related genes to promote cell survival.77
Crizotinib, the first generation TKI, was approved by the food and drug administration (FDA) for treating NSCLC patients who have the ALK gene rearrangement. The drug could induce rapid tumor regression and the majority of patients’ ORR up to 53%.78 However, after long‐time therapy, most of patients develop resistance in <1 year.79
Considering the limited activities of early generations of ALK TKIs, the FDA approved ceritinib, a second‐generation ALK TKI in 2014, especially for the patients who have experienced treatment with or who are intolerant of crizotinib.80 Ceritinib is known to be an inhibitor of ALK and insulin‐like growth factor‐1 (IGF‐1). Statistical evaluation of 124 patients with BM in Phase I clinical study, showed the overall response rate was 69% and that is about 19% higher than using ALK inhibitor alone.81 When it comes to the efficacy of ceritinib for intracranial metastasis, its brain‐to‐blood exposure ratio is about 15% according to the preclinical rat model.82 Alectinib is also a highly selective, second‐generation ALK inhibitor. Preclinical experiments have demonstrated that it is able to block mutated forms of ALK.83 In addition, two phase II studies about alectinib containing 50 cases suffering CNS disease showed a response rate of 57%‐69%.84 This potent antineoplastic activity of alectinib is probably due to its high penetration into the brain, and more importantly alectinib was assured not to be transported by P‐gp.85 Of course, other ALK‐targeting drugs are in various stages of development like AP26113 and PF‐06463922. They have been specifically designed to have outstanding CNS penetration, and are anticipated to be applied in future.86, 87 Figure 2 provides ALK and brain metastasis, including occurrence, development, and some representative drugs.
Figure 2.
The Molecular Illustration of ALK Controlling Brain Metastasis
4.2.2. Mutation in BRAF leading to the MAPK pathway
As stated above, the relationship between MAPK pathway and BM is inextricably connected. Numerous studies have demonstrated that about half patients with advanced melanoma have BRAF mutation, and in some studies the mutation rate even reaches as high as 60%.88 Actually, the occurrence of BRAF mutations increases the risk of BM at first diagnosis of metastatic disease.89 It is well‐known that V600E is the most common mutation in BRAF which leads to the MAPK signaling pathway aberrantly activated.90 At the same time, the activation of many bypass pathways and transcription factors enable the progression of BM becoming more rapid and uncontrollable.
As the “seed and soil” hypothesis mentioned before, brain‐derived signals promote the adhesion of melanoma cells to intracranial blood vessels, and foster melanoma metastasis formation. During this process, the role of chemokines and their cognate receptors cannot be ignored.91 Izraely discovered that brain‐metastasizing melanoma cells expressed really higher level of C‐C motif chemokine receptor 4 (CCR4). Moreover, “brain‐derived soluble factors” could upregulate CCR4 expression in melanoma cells and facilitate the migration of brain‐metastasizing melanoma cells specifically.92 Recently, a crucial relationship between several altered C‐C motif receptor 4 (CCR4) ligands, including C‐C motif ligand 4/17/22 (CCL4/17/22) and poor clinical outcomes have been observed. The changes may influence the establishment of MBM through adjusting cytokine and receptor signaling.93
The BBB presents a powerful shield that tumor cells must cross to construct residence in the brain. The presence of heparanase (HPSE) could increase melanoma cells invading into brain tissues.94 Suppressing HPSE RNA expression has been shown to inhibit melanoma migration, invasion, and adhesion.95 Moreover, astrocytes lately were confirmed its significant bidirectional relationship to melanoma cell. Brain‐metastasizing melanoma cells would stimulate astrocytes to express the pro‐inflammatory interleukin 23 (IL‐23) cytokine which in turn stimulate the secretion of matrix metalloproteinase‐2 (MMP‐2).96 Nevertheless, STAT3 regulates the expression of MMP‐2, both human brain metastatic melanoma cells and tissue biopsies show increased STAT3 activity compared to cutaneous melanoma cells.97 Therefore, increased MMP‐2 secretion by IL‐23 signaling can be mediated through STAT3 to mediate the degradation of extracellular matrix and facilitate extravasation. Metastasizing melanoma cells obtain blood supply in two ways generally, one is keeping close contact to microvessels and another is perivascular growth by vessel co‐option.98 For example, the activation of STAT3 would stimulate vascular remodeling and promote BM through increased expression of basic fibroblast growth factor (bFGF), VEGF, and MMP‐2.97 Connexin 26 (Cx26) is also involved in vessel co‐option during MBM.99
Last but not least, the function of PI3K/Akt must be mentioned in the process of MBM. Analysis of patients with melanoma of BRAFV600E or NRAS mutated showed the loss of PTEN would suppress MBM and reduce overall survival (OS) of patients.100 In general, the PI3K/Akt pathway is closely related to several key steps in MBM and significantly regulates cell adhesion, extravasation, degradation of extracellular matrix proteins and angiogenesis. These mechanisms also contain the cross‐talk of CCR4, HSPE, VEGF, STAT3, and Cx26/43. On account of the V600E‐mutated of BRAF, vemurafenib and dabrafenib, as two BRAFV600E inhibitors are currently approved for clinical use. Vemurafenib is a specific inhibitor of BRAFV600E mutated protein, which get 70% response rate with improved PFS and OS in BRAFV600E mutated metastatic melanoma patients.101 Similar to some other anticancer agents, treatment of BRAFV600E positive metastatic melanoma with vemurafenib showed good clinical responses at initial stage. However, most of the patients ultimately relapsed because of acquired resistance.102 The mean ratio of CSF/plasma vemurafenib concentration is only 0.98%±0.84%, indicating the poor ability to penetrate BBB.103 Under the circumstances, combined stereotactic radiosurgery (SRS) with BRAF inhibitors therapy were proposed and get increased overall survival of patients indeed.104
Another BRAFV600E inhibitor, Dabrafenib, has also shown curative effect for melanoma patients with BM. BRAF inhibitors dabrafenib combined with mitogen‐activated extracellular signal‐regulated kinase (MEK) inhibitors like trametinib could increase anti‐tumor activity and reduce side‐effect.105, 106 In conclusion, targeted therapy such as small molecule kinase inhibitors have achieved outstanding development, but still needs to pay more attention and take more effort on investigation and preferable application in clinic. Figure 3 provides BRAF mutation and brain metastasis, including Mutagenic factors and therapeutic drugs.
Figure 3.
The Role of BRAF Mutation in Brain Metastasis
4.2.3. The cyclin‐dependent kinase 4/6 (CDK‐4/6) pathway
Serin/threonin kinases CDKs, especially their activating regulatory subunits cyclins, control cell cycle.107 Promitotic signals induce increased expression of D‐type cyclins and cause CDK4 and CDK6 complex activated, and then the cyclin D1‐CDK4/6 complex could phosphorylate retinoblastoma protein (pRb), p130, and p107 which decrease their inhibition of E2F transcription factor family, finally allowing transcription of genes that control the cell cycle.108 Recent studies focused on breast cancer brain metastasis (BCBM) and found its increased E2F expression often activates Wnt or NF‐κB pathways to promote EMT. Moreover, increasing PI3K/Akt/mTOR activity, modulating apoptosis, altering Rho/Rac pathway would promote angiogenesis finally.109, 110 The CDK4/6‐DUB3 axis may act as an important regulatory mechanism of BCBM. Deubiquitinase 3 (DUB3) is a novel target of CDK4/6, so that CDK4/6‐mediated activation is crucial for the deubiquitination or stabilization of Snail1. The axis may regulate the possibility of BCBM to some extent.111
The first generation of CDK inhibitors showed modest clinical activity but considerable toxicity. Through constant technical improvement, selective small molecule CDK inhibitors have come out. Three compounds have reached the clinical stage: abemaciclib, ribociclib, and palbociclib. Recently, these drugs have been exploring the potential role in patients with estrogen receptor (ER) positive BCBM.112 A phase II study is evaluating the safety and activity of abemaciclib in hormone receptor (HR) positive BCBM and lung cancer or melanoma with BM.113 Vimentin and Snail, known as the EMT markers, could be downregulated with palbociclib treatment,114 supporting its inhibition of migration and invasion of breast cancer cells. Ribociclib is also being developed along a similar pathway to palbociclib, so FDA named it”breakthrough therapy” based on lots of experiments results. Therefore, the existing reactions give us courage to do more researches in HR+/HER2− advanced breast cancer sequentially.115 However, CDK4/6 inhibitors seem to need a more intact pRb pathway as a mechanism of action, and sometimes that may potentially limit the use in advanced breast cancers. Given the complexity of the cell cycle regulating pathways, more efforts should be devoted to confirm the role of CDK4/6 inhibitors in the treatment of BCBM patients furthermore. Figure 4 provides CDK‐4/6 and brain metastasis, including a series of mutagenic factors and therapeutic drugs.
Figure 4.
The Interaction Between CDK‐4/6 and Brain Metastasis
4.3. Immunity and metastasis
Nowadays, immune checkpoint inhibitors have already been used successfully in a wide variety of malignancies. FDA currently approved it to apply in metastatic melanoma, NSCLC and renal cell carcinoma. Brain was traditionally considered as an immunologically privileged site. Nevertheless, investigators found activated T‐cells can dramatically cross the BBB and patrol the CNS.116, 117 These observations inspired us to take advantage of the characteristic of T‐cell and to study more about immunotherapies. In conclusion, immunotherapies consists of programmed cell death‐1 (PD‐1), programmed death‐ligand 1 (PD‐L1) and monoclonal antibodies primarily against the epitopes of cytotoxic T‐lymphocyte‐associated protein 4 (CTLA‐4).
An anti‐CTLA‐4 antibody named ipilimumab was approved by the FDA for treating patients with advanced melanoma in 2011.118 When ipilimumab cooperated with SRS, a median survival was increased from 4.9 to 21.3 months, along with a 2‐year survival rate from 19.7% to 47.2%.119 Intracranial disease control rates are reported as 10% and 24% in patients with stable BM and those with asymptomatic BM respectively.120 Nivolumab and pembrolizumab, both of them against PD‐1, have gained durable clinical response in patients with advanced melanoma and metastatic NSCLC. In fact, PD‐L1 possess 52% high expression in BM, and it is in accordance with 32% in matched primary tumor tissue. In other words, BM indeed closely correlates with high expression of PD‐L1.121 Therefore, it was hypothesized that CTLA‐4 and PD‐1 could play complementary or synergistic role in the enhancement of immune function. Fortunately, recent studies indicated that combined treatment of ipilimumab and nivolumab achieved more rapid and deeper clinical responses compared with previous experiences using either agent alone in phase I study.122 Although the primary clinical effect has been proved, the CNS antitumor activity of PD‐1 inhibitors needs to be further explored deeply. Figure 5 provides CDK‐4/6 and brain metastasis, including PD‐1/PD‐L1 immunotherapy.
Figure 5.
Immunity and Brain Metastasis. The role of immune check point inhibitors PD‐1/PD‐L1 in cancer cells results in new therapies for brain metastasis
4.4. Targeting the genes strongly related to BM
4.4.1. ST6GALNAC5
ST6GALNAC5 is a 2,6‐sialyltransferase that modify cell surface glycoproteins and gangliosides. The ST6GALNAC5 gene is specifically expressed in mouse brain tissues, primarily in the forebrain and cerebellum.123 It was identified as one of the overexpressed genes in breast cancer cells which tend to develop BM in previous reports. Small interfering RNA (siRNA) directed against ST6GALNAC5 could decreased the adhesion of tumor cells to brain endothelial cells and impaired their ability to transmigrate the BBB in vitro. Above all, statistical analysis showed that ST6GALNAC5 only closely connected with BM but not with lung metastasis or bone metastasis.124 Utilizing the established CD34+ derived human BBB model in vitro, they found ST6GALNAC5 cDNA expression leads to a decrease of the interaction between MDA‐MB‐231 and the CD34+ derived human BBB model.125 In consequence, ST6GALNAC5 does not seem to be a mediator which promotes breast cancer cell interaction with the human BBB. Therefore, considering the tight relation between ST6GALNAC5 and BM, I think more detailed mechanism should be searched furthermore and the better targeted drugs will be engineered.
4.4.2. SERPINS
In recent studies, a new small gene whose expression is closely related to brain metastatic phenotypes was discovered both in lung and breast cancer models.126 SERPIN I1, encoding the plasminogen activator (PA) inhibitor neuroserpin (NS), is commonly expressed in the brain. The PA could degrade the thrombus through activating the fibrinolytic enzyme, and take part in other neuro‐matrix reactions at the same time.127 Under normal circumstances, neurons will overexpress NS to resist the adverse reactions that derived from over secretion of plasma enzymes. The brain microenvironment would maintain integrant balance via this manner.
However, the balance sometimes be disrupted when disease occurred. The overexpression of anti‐PA serpins in brain metastatic cells from lung cancer or breast cancer often induces plasmin generation and presents high possibility to metastasis conclusively. Therefore, the anti‐PA serpins provide a common mechanism for the initiation of BM in lung and breast cancer to some extent, and it will keep cancer cells away from death signals and facilitate vascular co‐option. Most of all, the serpins are also specifically associated with BM, not other metastatic organs. As we thought, the incidence of BM was significantly reduced after interfering with serpins in tumor cells, while the transfer rate of other organs was not affected.126
4.4.3. COX2
Cyclo‐oxygen‐ase 2 (COX2) is a tumor‐associated gene and closely related to the development of several tumors. The brain metastatic activity of brain metastatic derivative 2 (BrM2) cells was experimentally decreased by RNA interference (RNAi)‐mediated knockdown of COX2 expression.124 Therefore, COX2 was probably indicated to be a mediator in brain and lung metastasis.
In the research of effect of 21 matrix metalloproteinases on brain metastasis‐free survival of breast cancer, only matrix metalloproteinase‐1(MMP‐1) is significantly correlated with BM.128 MMP‐1 has highly expressed in brain metastatic cells and is able to degrade claudin and occludin but not ZO‐1, which are critical factors of BBB. Moreover, COX2 overexpresses in many aggressive cancer cells, and its product prostaglandin could directly upregulate the expression of MMP‐1.129 And prostaglandin is indeed capable to increase permeability of BBB due to the upregulation of MMP‐1. So a COX2 inhibitor (NS398) could effectively block both MMP‐1 expression and BBB permeability simultaneously.128 Thus, the critical role of COX2‐PGs‐MMP1 axis is essential in BCBM, and a COX2 inhibitor could be used for preventing BM.
5. CONCLUSIONS
Brain metastasis is a complicated process because of the heterogeneity between cancer cells and the microenvironment. Meanwhile, the regulation of signal pathways is also diverse and interactive. In conclusion, VEGF or EGFR plays an important role in regulating the pivotal switch of BM in tumor angiogenesis. In addition, lung cancer, melanoma, and breast cancer with high BM probability have respective molecular mechanisms, such as ALK rearrangement, BRAF mutation, and D1‐CDK4/6 complex formation. And the tumor immunotherapy has also gradually been applied to the treatment of BM. Besides, several target genes specifically associated with BM have been reported recently. Indeed, the need for the clinical treatment of BM is strongly supported by a growing literature demonstrating many unique molecular features.
However, CNS disease progression always escapes the extracranial disease control. And the more important is that our present knowledge on brain is not so clear that molecular mechanism of BM cannot be elucidated actually. In our opinion, further clinical trials should be thought about to combine targeted therapies with radiation therapy such as WBRT and SRS, or with immunotherapeutic agents. With the advanced understanding of concrete molecular mechanism of BM, we are eagerly expecting to find a brand‐new therapy that specifically targets BM.
CONFLICT OF INTEREST
None declared.
ACKNOWLEDGMENTS
The work was supported by National Natural Science Foundation of China (No. 81673648, 81673725), Natural Science Foundation of Higher School of Jiangsu Province (17KJA360003), the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD) and the Preliminary Foundation for National Natural Science (JKLPSE201609).
Ni W, Chen W, Lu Y. Emerging findings into molecular mechanism of brain metastasis. Cancer Med. 2018;7:3820–3833. 10.1002/cam4.1667
Contributor Information
Wenxing Chen, Email: chenwx@njucm.edu.cn.
Yin Lu, Email: profyinlu@163.com.
REFERENCES
- 1. Fox BD, Cheung VJ, Patel AJ, et al. Epidemiology of metastatic brain tumors. Neurosurg Clin N Am. 2011;22(1):1‐6. [DOI] [PubMed] [Google Scholar]
- 2. Percy AK. Neoplasms of the central nervous system: epidemiologic considerations. Neurology. 1972;22(1):40‐48. [DOI] [PubMed] [Google Scholar]
- 3. Walker AE, Robins M, Weinfeld FD. Epidemiology of brain tumors: the national survey of intracranial neoplasms. Neurology. 1985;35(2):219‐226. [DOI] [PubMed] [Google Scholar]
- 4. Nayak L, Lee EQ, Wen PY. Epidemiology of brain metastasis. Curr Oncol Rep. 2012;14(1):48‐54. [DOI] [PubMed] [Google Scholar]
- 5. Mcwilliams RR, Rao RD, Brown PD, et al. Treatment options for brain metastasis from melanoma. Expert Rev Anticancer Ther. 2005;5(5):809‐820. [DOI] [PubMed] [Google Scholar]
- 6. Sawaya R, Wildrick DM, Mesfin FB. Metastatic brain tumors: viewpoint—surgery. Principle Pract Stereot Radiosurg. 2015;2:233‐240. [Google Scholar]
- 7. Barnholtz‐Sloan JS, Sloan AE, Davis FG, et al. Incidence proportions of brain metastasis in patients diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J Clin Oncol. 2004;22(14):2865‐2872. [DOI] [PubMed] [Google Scholar]
- 8. Arrieta O, Villarreal‐Garza C, Zamora J, et al. Long‐term survival in patients with non‐small cell lung cancer and synchronous brain metastasis treated with whole‐brain radiotherapy and thoracic chemoradiation. Radiat Oncol. 2011;6(1):166‐172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lin NU, Amirikordestani L, Palmieri D, et al. CNS metastasis in breast cancer: old challenge, new frontiers. Clin Cancer Res. 2013;19(23):6404‐6418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Davies MA, Liu P, Mcintyre S, et al. Prognostic factors for survival in melanoma patients with brain metastases. Cancer. 2011;117(8):1687‐1696. [DOI] [PubMed] [Google Scholar]
- 11. Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989;133(3421):98‐101. [PubMed] [Google Scholar]
- 12. Suchorska WM, Lach MS. The role of exosomes in tumor progression and metastasis (Review). Oncol Rep. 2016;35(3):1237‐1244. [DOI] [PubMed] [Google Scholar]
- 13. Liu CM, Hsieh CL, Shen CN, et al. Exosomes from the tumor microenvironment as reciprocal regulators that enhance prostate cancer progression. Int J Urol. 2016;23(9):734‐744. [DOI] [PubMed] [Google Scholar]
- 14. Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro‐metastatic phenotype through MET. Nat Med. 2012;18(6):883‐891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Valadi H, Ekström K, Bossios A, et al. Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654‐659. [DOI] [PubMed] [Google Scholar]
- 16. Zhang C, Yu D. Microenvironment determinants of brain metastasis. Cell Biosci. 2011;1(1):1‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Fokas E, Steinbach JP, Rödel C. Biology of brain metastasis and novel targeted therapies: time to translate the research. Biochim Biophys Acta. 2013;1835(1):61‐75. [DOI] [PubMed] [Google Scholar]
- 18. Abbott N, Ronnback L, Hansson E. Astrocyte‐endothelial interactions at the blood‐brain barrier. Nat Rev Neurosci. 2006;7(1):41‐53. [DOI] [PubMed] [Google Scholar]
- 19. Chen IC, Hsiao IL, Lin HC, et al. Influence of silver and titanium dioxide nanoparticles on in vitro blood‐brain barrier permeability. Environ Toxicol Pharmacol. 2016;47:108‐118. [DOI] [PubMed] [Google Scholar]
- 20. Pardridge WM. Blood‐brain barrier drug targeting: the future of brain drug development. Mol Interventions. 2003;3(2):90‐105. [DOI] [PubMed] [Google Scholar]
- 21. Shapiro WR, Shapiro JR. Principles of brain tumor chemotherapy. Semin Oncol. 1986;13(1):56‐69. [PubMed] [Google Scholar]
- 22. de Vries NA, Zhao J, Kroon E, et al. P‐Glycoprotein and breast cancer resistance protein: two dominant transporters working together in limiting the brain penetration of Topotecan. Clin Cancer Res. 2007;13(21):6440‐6449. [DOI] [PubMed] [Google Scholar]
- 23. Fenstermacher J, Gross P, Sposito N, et al. Structural and functional variations in capillary systems within the brain. Ann N Y Acad Sci. 1988;529(1):21‐30. [DOI] [PubMed] [Google Scholar]
- 24. Kawamura S, Schürer L, Goetz A, et al. An improved closed cranial window technique for investigation of blood‐brain barrier function and cerebral vasomotor control in the rat. Int J Microcir Clin Exp. 1990;9(4):369‐383. [PubMed] [Google Scholar]
- 25. Zhang RD, Price JE, Fujimaki T, et al. Differential permeability of the blood‐brain barrier in experimental brain metastasis produced by human neoplasms implanted into nude mice. Am J Pathol. 1992;141(5):1115‐1124. [PMC free article] [PubMed] [Google Scholar]
- 26. Zagzag D, Goldenberg M, Brem S. Angiogenesis and blood‐brain barrier breakdown modulate CT contrast enhancement: an experimental study in a rabbit brain‐tumor model. AJR Am J Roentgenol. 1989;153(1):141‐146. [DOI] [PubMed] [Google Scholar]
- 27. Jr BW, Schimpff RD. An experimental model for cerebral metastasis: preliminary light and ultrastructural studies. J Neuropathol Exp Neurol. 1979;38(1):19‐34. [DOI] [PubMed] [Google Scholar]
- 28. Pardridge WM, Oldendorf WH, Cancilla P, et al. Blood‐brain barrier: interface between internal medicine and the brain. Ann Intern Med. 1986;105(1):82‐95. [DOI] [PubMed] [Google Scholar]
- 29. Long DM. Capillary ultrastructure in human metastatic brain tumors. J Neurosurg. 1979;51(1):53‐58. [DOI] [PubMed] [Google Scholar]
- 30. Pozzilli C, Bernardi S, Mansi L, et al. Quantitative assessment of blood‐brain barrier permeability in multiple sclerosis using 68‐Ga‐EDTA and positron emission tomography. J Neurol Neurosurg Psychiatry. 1988;51(8):1058‐1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Rosner D, Nemoto T, Lane WW. Chemotherapy induces regression of brain metastasis in breast carcinoma. Cancer. 1986;58(4):832‐839. [DOI] [PubMed] [Google Scholar]
- 32. Brown WR, Thore CR, Moody DM, et al. Vascular damage after fractionated whole‐brain irradiation in rats. Radiat Res. 2005;164(5):662‐668. [DOI] [PubMed] [Google Scholar]
- 33. Pestalozzi BC, Brignoli S. Trastuzumab in CSF. J Clin Oncol. 2000;18(11):2349‐2351. [DOI] [PubMed] [Google Scholar]
- 34. Colozza M, Minenza E, Gori S, et al. Extended survival of a HER‐2‐positive metastatic breast cancer patient with brain metastasis also treated with intrathecal trastuzumab. Cancer Chemother Pharmacol. 2009;63(6):1157‐1159. [DOI] [PubMed] [Google Scholar]
- 35. Murrell DH, Foster PJ, Chambers AF. Brain metastasis from breast cancer: lessons from experimental magnetic resonance imaging studies and clinical implications. J Mol Med. 2014;92(1):5‐12. [DOI] [PubMed] [Google Scholar]
- 36. Mazel M, Clair P, Rousselle C, et al. Doxorubicin‐peptide conjugates overcome multidrug resistance. Anticancer Drugs. 2001;12(2):107‐116. [DOI] [PubMed] [Google Scholar]
- 37. Dahlborg SA, Henner WD, Crossen JR, et al. Non‐AIDS primary CNS lymphoma: first example of a durable response in a primary brain tumor using enhanced chemotherapy delivery without cognitive loss and without radiotherapy. Cancer J Sci Am. 1996;2(3):166‐174. [PubMed] [Google Scholar]
- 38. Ferrara N, Gerber HP, Lecouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669‐676. [DOI] [PubMed] [Google Scholar]
- 39. Kim LS, Huang S, Lu W, et al. Vascular endothelial growth factor expression promotes the growth of breast cancer brain metastasis in nude mice. Clin Exp Metas. 2004;21(2):107‐118. [DOI] [PubMed] [Google Scholar]
- 40. Yano S, Shinohara H, Herbst RS, et al. Expression of vascular endothelial growth factor is necessary but not sufficient for production and growth of brain metastasis. Can Res. 2000;60(17):4959‐4967. [PubMed] [Google Scholar]
- 41. Claffey KP, Brown LF, del Aguila LF, et al. Expression of vascular permeability factor/vascular endothelial growth factor by melanoma cells increases tumor growth, angiogenesis, and experimental metastasis. Can Res. 1996;56(1):172‐181. [PubMed] [Google Scholar]
- 42. Küsters B, Leenders WP, Wesseling P, et al. Vascular endothelial growth factor‐A (165) induces progression of melanoma brain metastasis without induction of sprouting angiogenesis. Can Res. 2002;62(2):341‐345. [PubMed] [Google Scholar]
- 43. Crespo S, Kind M, Arcaro A. The role of the PI3K/Akt/mTOR pathway in brain tumor metastasis. J Cancer Metastasis Treat. 2016;2(3):80‐89. [Google Scholar]
- 44. Criscuoli ML, Nguyen M, Eliceiri BP. Tumor metastasis but not tumor growth is dependent on Src‐mediated vascular permeability. Blood. 2005;105(4):1508‐1514. [DOI] [PubMed] [Google Scholar]
- 45. Davies MA, Stemkehale K, Lin E, et al. Integrated molecular and clinical analysis of AKT activation in metastatic melanoma. Clin Cancer Res. 2009;15(24):7538‐7546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Xia C, Meng Q, Cao Z, et al. Regulation of angiogenesis and tumor growth by p110 alpha and AKT1 via VEGF expression. J Cell Physiol. 2006;209(1):56‐66. [DOI] [PubMed] [Google Scholar]
- 47. Boutet A, Esteban MA, Maxwell PH, et al. Reactivation of Snail genes in renal fibrosis and carcinomas: a process of reversed embryogenesis? Cell Cycle. 2007;6(6):638‐642. [DOI] [PubMed] [Google Scholar]
- 48. Schmid MC, Varner JA. Myeloid cell trafficking and tumor angiogenesis. Cancer Lett. 2007;250(1):1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Gril B, Palmieri D, Bronder JL, et al. Effect of lapatinib on the outgrowth of metastatic breast cancer cells to the brain. J Natl Cancer Inst. 2008;100(15):1092‐1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Gril B, Palmieri D, Qian Y, et al. Pazopanib reveals a role for tumor Cell B‐Raf in the prevention of HER2 + breast cancer brain metastasis. Clin Cancer Res. 2011;17(1):142‐153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Kodack DP, Chung E, Yamashita H, et al. Combined targeting of HER2 and VEGFR2 for effective treatment of HER2‐amplified breast cancer brain metastasis. Proc Natl Acad Sci USA. 2012;109(45):3119‐3127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Patyna S, Peng G. 56 POSTER Distribution of sunitinib and its active metabolite in brain and spinal cord tissue following oral or intravenous administration in rodents and monkeys. Eur J Cancer Suppl. 2006;4(12):21. [Google Scholar]
- 53. Chen AM, Jahan TM, Jablons DM, Garcia J, Larson DA. Risk of cerebral metastasis and neurological death after pathological complete response to neoadjuvant therapy for locally advanced non‐small‐cell lung cancer. Cancer. 2007;109(8):1668‐1675. [DOI] [PubMed] [Google Scholar]
- 54. Shin DY, Na II, Kim CH, et al. EGFR mutation and brain metastasis in pulmonary adenocarcinomas. J Thorac Oncol. 2014;9(2):195‐199. [DOI] [PubMed] [Google Scholar]
- 55. Enomoto Y, Takada K, Hagiwara E, et al. Distinct features of distant metastasis and lymph node stage in lung adenocarcinoma patients with epidermal growth factor receptor gene mutations. Respir Investig. 2013;51(3):153‐157. [DOI] [PubMed] [Google Scholar]
- 56. Adamo B, Deal AM, Burrows E, et al. Phosphatidylinositol 3‐kinase pathway activation in breast cancer brain metastasis. Breast Cancer Res. 2011;13(6):125‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Hohensee I, Lamszus K, Riethdorf S, et al. Frequent Genetic Alterations in EGFR‐ and HER2‐Driven Pathways in Breast Cancer Brain Metastasis. Am J Pathol. 2013;183(1):83‐95. [DOI] [PubMed] [Google Scholar]
- 58. Daphu I, Horn S, Stieber D, et al. In vitro treatment of melanoma brain metastasis by simultaneously targeting the MAPK and PI3K signaling pathways. Int J Mol Sci. 2014;15(5):8773‐8794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Breindel JL, Haskins JW, Cowell EP, et al. EGF receptor activates MET through MAPK to enhance non‐small cell lung carcinoma invasion and brain metastasis. Can Res. 2013;73(16):5053‐5065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Clarke JL, Pao W, Wu N, et al. High dose weekly erlotinib achieves therapeutic concentrations in CSF and is effective in leptomeningeal metastasis from epidermal growth factor receptor mutant lung cancer. J Neurooncol. 2010;99(2):283‐286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jackman DM, Cioffredi LA, Jacobs L, et al. A phase I trial of high dose gefitinib for patients with leptomeningeal metastasis from non‐small cell lung cancer. Oncotarget. 2015;6(6):4527‐4536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Bono P, Joensuu H. Afatinib alone or afatinib plus vinorelbine versus investiga. Lancet Oncol. 2015;16(16):1700‐1710. [DOI] [PubMed] [Google Scholar]
- 63. Vries NAD, Buckle T, Zhao J, et al. Restricted brain penetration of the tyrosine kinase inhibitor erlotinib due to the drug transporters P‐gp and BCRP. Invest New Drugs. 2012;30(2):443‐449. [DOI] [PubMed] [Google Scholar]
- 64. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non‐small‐cell lung cancer to gefitinib. N Engl J Med. 2005;352(8):786‐792. [DOI] [PubMed] [Google Scholar]
- 65. Sun JM, Ahn MJ, Choi YL, et al. Clinical implications of T790M mutation in patients with acquired resistance to EGFR tyrosine kinase inhibitors. Lung Cancer. 2013;82(2):294‐298. [DOI] [PubMed] [Google Scholar]
- 66. Wang S, Cang S, Liu D. Third‐generation inhibitors targeting EGFR T790M mutation in advanced non‐small cell lung cancer. J Hematol Oncol. 2016;9(1):1‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Chinnaiyan P, Huang S, Vallabhaneni G, et al. Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (Tarceva). Can Res. 2005;65(8):3328‐3335. [DOI] [PubMed] [Google Scholar]
- 68. Gainor JF, Varghese AM, Ou SHI, et al. ALK Rearrangements Are Mutually Exclusive with Mutations in EGFR or KRAS: An Analysis of 1,683 Patients with Non‐Small Cell Lung Cancer. Clin Cancer Res. 2013;19(15):4273‐4281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Doebele RC, Lu X, Sumey C, et al. Oncogene status predicts patterns of metastatic spread in treatment‐naive non‐small cell lung cancer. Cancer. 2012;118(18):4502‐4511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Rangachari D, Yamaguchi N, Vanderlaan PA, et al. Brain metastasis in patients with EGFR‐mutated or ALK‐rearranged non‐small‐cell lung cancers. Lung Cancer. 2015;88(1):108‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Hida T, Nokihara H, Kondo M, et al. Alectinib versus crizotinib in patients with ALK‐positive non‐small‐cell lung cancer (J‐ALEX): an open‐label, randomised phase 3 trial. Lancet. 2017;390(10089):29‐39. [DOI] [PubMed] [Google Scholar]
- 72. Seto T, Kiura K, Nishio M, et al. CH5424802 (RO5424802) for patients with ALK‐rearranged advanced non‐small‐cell lung cancer (AF‐001JP study): a single‐arm, open‐label, phase 1‐2 study. Lancet Oncol. 2013;14(7):590‐598. [DOI] [PubMed] [Google Scholar]
- 73. Enomoto M. Identification of the transforming EML4‐ALK fusion gene in non‐small‐cell lung cancer. Nature. 2007;448(7153):561‐566. [DOI] [PubMed] [Google Scholar]
- 74. Matsumura I, Mizuki M, Kanakura Y. Roles for deregulated receptor tyrosine kinases and their downstream signaling molecules in hematologic malignancies. Cancer Sci. 2008;99(3):479‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Howes J. Anaplastic lymphoma kinase (ALK) activates Stat3 and protects hematopoietic cells from cell death. Oncogene. 2002;21(7):1038‐1047. [DOI] [PubMed] [Google Scholar]
- 76. Fujimoto J, Shiota M, Iwahara T, et al. Characterization of the transforming activity of p80, a hyperphosphorylated protein in a Ki‐1 lymphoma cell line with chromosomal translocation t (2;5). Proc Natl Acad Sci USA. 1996;93(9):4181‐4186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Slupianek A, Skorski T. NPM/ALK downregulates p27 Kip1, in a PI‐3K‐dependent manner. Exp Hematol. 2004;32(12):1265‐1271. [DOI] [PubMed] [Google Scholar]
- 78. Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK‐positive lung cancer. N Engl J Med. 2013;368(25):2385‐2394. [DOI] [PubMed] [Google Scholar]
- 79. Sasaki T, Koivunen J, Ogino A, et al. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Can Res. 2011;71(18):6051‐6060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Shaw AT, Kim DW, Mehra R, et al. Ceritinib in ALK‐rearranged non–small‐cell lung cancer. N Engl J Med. 2014;370(13):1189‐1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Shaw AT, Mehra R, Tan DSW, et al. 1293pevaluation of ceritinib‐treated patients (pts) with anaplastic lymphoma kinase rearranged (alk+) non‐small cell lung cancer (nsclc) and brain metastases in the ascend‐1 study. Mol Biol Cell. 2014;18(4):1385‐1396. [Google Scholar]
- 82. Dhillon S, Clark M. Ceritinib: first global approval. Drugs. 2014;74(11):1285‐1291. [DOI] [PubMed] [Google Scholar]
- 83. Kodama T, Tsukaguchi T, Yoshida M, et al. Selective ALK inhibitor alectinib with potent antitumor activity in models of crizotinib resistance. Cancer Lett. 2014;351(2):215‐221. [DOI] [PubMed] [Google Scholar]
- 84. Shaw AT, Gandhi L, Gadgeel S, et al. Phase 2 prospective analysis of alectinib in ALK‐positive, crizotinib‐resistant non‐small‐cell lung cancer. Lancet Oncol. 2016;17(2):234‐242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Bartels AL, Kortekaas R, Bart J, et al. Blood‐brain barrier P‐glycoprotein function decreases in specific brain regions with aging: a possible role in progressive neurodegeneration. Neurobiol Aging. 2009;30(11):1818‐1824. [DOI] [PubMed] [Google Scholar]
- 86. Gettinger SN, Bazhenova L, Salgia R, et al. Updated efficacy and safety of the ALK inhibitor AP26113 in patients (pts) with advanced malignancies, including ALK+ non‐small cell lung cancer (NSCLC). Ann Oncol. 2014;63(10):4166‐4169. [Google Scholar]
- 87. Zou HY, Friboulet L, Kodack DP, et al. PF‐06463922, an ALK/ROS1 inhibitor, overcomes resistance to first and second generation ALK inhibitors in preclinical models. Cancer Cell. 2015;28(1):70‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949‐954. [DOI] [PubMed] [Google Scholar]
- 89. Jakob JA, Jr Bassett RL, Ng CS, et al. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer. 2012;118(16):4014‐4023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Ueda M, Toji E, Nunobiki O, et al. Mutational analysis of the BRAF gene in human tumor cells. Hum Cell. 2008;21(2):13‐17. [DOI] [PubMed] [Google Scholar]
- 91. Klein A, Sagi‐Assif O, Meshel T, et al. CCR4 is a determinant of melanoma brain metastasis. Oncotarget. 2017;8(19):31079‐31091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Izraely S, Klein A, Sagiassif O, et al. Chemokine–chemokine receptor axes in melanoma brain metastasis. Immunol Lett. 2010;130(1):107‐114. [DOI] [PubMed] [Google Scholar]
- 93. Lok E, Chung AS, Swanson KD, et al. Melanoma brain metastasis globally reconfigures chemokine and cytokine profiles in patient cerebrospinal fluid. Melanoma Res. 2014;24(2):120‐130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Murry BP, Blust BE, Singh A, et al. Heparanase mechanisms of melanoma metastasis to the brain: Development and use of a brain slice model. J Cell Biochem. 2006;97(2):217‐225. [DOI] [PubMed] [Google Scholar]
- 95. Liu X, Fang H, Chen H, et al. An artificial miRNA against HPSE suppresses melanoma invasion properties, correlating with a down‐regulation of chemokines and MAPK phosphorylation. PLoS ONE. 2012;7(6):38659‐38670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Klein A, Schwartz H, Sagi‐Assif O, et al. Astrocytes facilitate melanoma brain metastasis via secretion of IL‐23. J Pathol. 2015;236(1):116‐127. [DOI] [PubMed] [Google Scholar]
- 97. Aldape KD, Kang SH, Gershenwald JE, et al. Activation of stat3 in human melanoma promotes brain metastasis. Can Res. 2006;66(6):3188‐3196. [DOI] [PubMed] [Google Scholar]
- 98. Kienast Y, Von BL, Fuhrmann M, et al. Real‐time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16(1):116‐122. [DOI] [PubMed] [Google Scholar]
- 99. Stoletov K, Strnadel J, Zardouzian E, et al. Role of connexins in metastatic breast cancer and melanoma brain colonization. J Cell Sci. 2013;126(4):904‐913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Bucheit AD, Chen G, Siroy A, et al. Complete loss of PTEN protein expression correlates with shorter time to brain metastasis and survival in stage IIIB/C melanoma patients with BRAFV600 mutations. Clin Cancer Res. 2014;20(21):5527‐5536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Poulikakos PI. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N Engl J Med. 2011;364(26):2507‐2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Sullivan RJ, Flaherty KT. Resistance to BRAF‐targeted therapy in melanoma. Eur J Cancer. 2013;49(6):1297‐1304. [DOI] [PubMed] [Google Scholar]
- 103. Sakjidupr L, Le RE, Templier C, et al. Cerebrospinal fluid concentrations of vemurafenib in patients treated for brain metastatic BRAF‐V600 mutated melanoma. Melanoma Res. 2015;25(4):302‐305. [DOI] [PubMed] [Google Scholar]
- 104. Wolf A, Zia S, Verma R, et al. Impact on overall survival of the combination of BRAF inhibitors and stereotactic radiosurgery in patients with melanoma brain metastasis. J Neurooncol. 2016;127(3):607‐615. [DOI] [PubMed] [Google Scholar]
- 105. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30‐39. [DOI] [PubMed] [Google Scholar]
- 106. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367(18):1694‐1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Ma Z, Wu Y, Guo AY, et al. Phylogenetic analysis reveals the evolution and diversification of cyclins in eukaryotes. Mol Phylogenet Evol. 2013;66(3):1002‐1010. [DOI] [PubMed] [Google Scholar]
- 108. Chen HZ, Tsai SY, Leone G. Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nat Rev Cancer. 2009;9(11):785‐797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Yamamoto‐Ibusuki M, Arnedos M, André F. Targeted therapies for ER+/HER2‐ metastatic breast cancer. BMC Med. 2015;13(1):137‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Owsley J, Jimeno A, Diamond JR. Palbociclib: CDK4/6 inhibition in the treatment of ER‐positive breast cancer. Drugs Today. 2016;52(2):119‐129. [DOI] [PubMed] [Google Scholar]
- 111. Liu T, Yu J, Deng M, et al. CDK4/6‐dependent activation of DUB3 regulates cancer metastasis through SNAIL1. Nat Commun. 2017;8:13923‐13934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Hamilton E, Infante JR. Targeting CDK4/6 in patients with cancer. Cancer Treat Rev. 2016;45:129‐138. [DOI] [PubMed] [Google Scholar]
- 113. Stemmler HJ, Heinemann V. Central nervous system metastasis in HER‐2‐overexpressing metastatic breast cancer: a treatment challenge. Oncologist. 2008;13(7):739‐750. [DOI] [PubMed] [Google Scholar]
- 114. Qin G, Xu F, Qin T, et al. Palbociclib inhibits epithelial‐mesenchymal transition and metastasis in breast cancer via c‐Jun/COX‐2 signaling pathway. Oncotarget. 2015;6(39):41794‐41808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Shah AN, Cristofanilli M. The growing role of CDK4/6 inhibitors in treating hormone receptor‐positive advanced breast cancer. Curr Treat Options Oncol. 2017;18(1):6‐21. [DOI] [PubMed] [Google Scholar]
- 116. Engelhardt B. The blood‐central nervous system barriers actively control immune cell entry into the central nervous system. Curr Pharm Des. 2008;14(16):1555‐1565. [DOI] [PubMed] [Google Scholar]
- 117. Prins RM, Vo DD, Khanfarooqi H, et al. NK and CD4 cells collaborate to protect against melanoma tumor formation in the brain. J Immunol. 2006;177(12):8448‐8455. [DOI] [PubMed] [Google Scholar]
- 118. Hodi FS, O'Day SJ, Mcdermott DF, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med. 2010;363(8):711‐723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Knisely JPS, Yu JB, Flanigan J, et al. Radiosurgery for melanoma brain metastasis in the ipilimumab era and the possibility of longer survival. J Neurosurg. 2012;117(2):227‐233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Queirolo P, Spagnolo F, Ascierto PA, et al. Efficacy and safety of ipilimumab in patients with advanced melanoma and brain metastasis. J Neurooncol. 2014;118(1):109‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Berghoff AS, Ricken G, Wilhelm D, et al. Tumor infiltrating lymphocytes and PD‐L1 expression in brain metastasis of small cell lung cancer (SCLC). J Neurooncol. 2016;130(1):19‐29. [DOI] [PubMed] [Google Scholar]
- 122. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122‐133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Ikehara Y, Shimizu N, Kono M, et al. A novel glycosyltransferase with a polyglutamine repeat: a new candidate for GD1alpha synthase (ST6GalNAc V). FEBS Lett. 1999;463(1–2):92‐96. [DOI] [PubMed] [Google Scholar]
- 124. Bos PD, Zhang XHF, Nadal C, et al. Genes that mediate breast cancer metastasis to the brain. Nature. 2009;459(7249):1005‐1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Drolez A, Vandenhaute E, Delannoy CP, et al. ST6GALNAC5 expression decreases the interactions between breast cancer cells and the human blood‐brain barrier. Int J Mol Sci. 2016;17(8):1309‐1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Valiente M, Obenauf A, Jin X, et al. Serpins Promote Cancer Cell Survival and Vascular Co‐Option in Brain Metastasis. Cell. 2014;156(5):1002‐1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119(1):7‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Wu K, Fukuda K, Xing F, et al. Roles of the cyclooxygenase 2 matrix metalloproteinase 1 pathway in brain metastasis of breast cancer. J Biol Chem. 2015;290(15):9842‐9854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Lee KY, Kim YJ, Yoo H, et al. Human brain endothelial cell‐derived COX‐2 facilitates extravasation of breast cancer cells across the blood‐brain barrier. Anticancer Res. 2011;31(12):4307‐4313. [PubMed] [Google Scholar]