Open access peer-reviewed chapter

Non-Small Cell Lung Cancer Brain Metastasis: The Link between Molecular Mechanisms and Novel Therapeutic Approaches

Written By

Shiva Najjary, Dana A.M. Mustafa and Johan M. Kros

Submitted: 08 June 2022 Reviewed: 07 July 2022 Published: 04 October 2022

DOI: 10.5772/intechopen.106385

From the Edited Volume

Cancer Metastasis - Molecular Mechanism and Clinical Therapy

Edited by Yusuf Tutar and Lütfi Tutar

Chapter metrics overview

97 Chapter Downloads

View Full Metrics

Abstract

The prognosis of patients suffering from non-small cell lung carcinomas (NSCLC) worsens significantly when brain metastasis occurs. Seeding to the brain usually happens relatively early in the course of disease and therefore, new therapies anticipating this complication would result in considerable improvement in outcomes. In this review, we address recent molecular data of NSCLC with a focus on the risk of the formation of brain metastasis. Included is new data on the involvement of miRNAs and lncRNAs in the rise of the cerebral seeding of NSCLC. We summarize novel therapeutic approaches developed in the light of these recent molecular discoveries.

Keywords

  • brain metastasis
  • lung cancer
  • blood-brain barrier
  • microRNAs
  • targeted therapy
  • immunotherapy

1. Introduction

Lung carcinoma is among the deadliest cancers and its treatment is an important challenge for oncologists. Approximately 16–20% of patients with lung cancer develop brain metastasis, regarded as the most life-threatening complication of the disease. Population-based incidence proportions for brain metastasis are highest for lung cancer (20% against 9.6% for all common cancers) [1]. The frequency of the diagnosis of lung cancer brain metastasis (LCBM) has increased and the reasons for this are not entirely clear. Certainly, advances in radiology have resulted in increased sensitivity for tracing brain metastatic sites. In addition, metastatic tumor cells behind the blood-brain barrier (BBB) are less vulnerable to chemotherapeutic agents (“pharmacologic sanctuary”). Further, there are effects of the increasing age of the population [2]. The incidence and severity of the cerebral symptoms vary from minimal to severely debilitating. Less than 4% of patients with metastatic NSCLC live longer than five years after the diagnosis [3, 4, 5]. Obviously, protecting patients from developing brain metastases would significantly alleviate the disease burden and improve outcomes. Knowledge of the subsequent steps tumor cells need to take before growing as metastases in the brain is essential. In this review, we will summarize current knowledge on the genes and pathways operative in the development of brain metastasis of NSCL and summarize the application of targeted drugs.

Advertisement

2. Brain metastasis development and the blood-brain-barrier

A significant part of the disease burden and death of cancer is caused by the seeding of tumor cells to the brain [6]. The stages of the development of brain metastasis include the detachment of cancer cells from the primary tumors and their penetration of the BBB followed by extravasation, colonization, and macro-metastatic growth [7]. The detachment of tumor cells from the primary tumor mass depends on cell adhesion molecules (CAMs) including immunoglobulins (IgCAMs), selectins, integrins, and cadherins [8, 9, 10]. The tumor cells require the loss of functional E-cadherin (CDH1) in order to increase their motility, and close relation between reduced E-cadherin expression and poor outcome due to tumor spread in NSCLC exists [11]. CDH1 regulates EGFR activity through receptor tyrosine kinases (RTKs) and provides functions in intracellular signaling. Subsequent events include the epithelial-mesenchymal transition (EMT), a crucial phenomenon in the dissemination and motility of cancer cells [12, 13]. The process of EMT relays on proteases such as secreted matrix metalloproteinases (MMPs) that degrade the extracellular matrix (ECM) components including proteoglycans, collagen, fibronectin, and laminin, and modify the structural and mechanical features of the ECM [14]. MMPs also break down cell-ECM - and cell-cell connections by cleaving CDH1 and CD44. MMP-1, MMP-2, and MMP-9 are particularly associated with metastases of lung cancer [15]. Once detached and motile, the tumor cells enter the circulation to become circulating tumor cells (CTCs). Some CTCs resist the forces of the blood flow and by using surface receptors adhere to the endothelial cells. Subsequently, the cells will migrate through the endothelial layer by the expression of selectins, integrins, and chemokines. This process is accompanied by the creation of a permissive immune microenvironment through the activation of integrins and the release of cytokines such as vascular endothelial growth factor (VEGF) [16]. VEGF is vital in the process of neovascularization and takes part in the creation of high endothelial venules, to increase lymphocyte extravasation and infiltration in the perivascular niches (PVN) at the metastatic sites [17]. The altered microenvironment promotes further migration of CTCs to the brain parenchyma by secreting site-specific chemokines such as CXCR4 and its ligand, CXCL12 [17, 18]. There is high expression of CXCR4/CXCL12 in brain metastases of NSCLC and, together with integrins, CXCR4 enhances further tumor cell invasion. The metastatic cells in the PVNs activate tumor-associated macrophages (TAMs) and microglia. TAMs play a role in the survival of CTCs and induce extravasation and colonization by expressing survival factors such as epidermal growth factor (EGF) [19]. While the TILs try to combat the tumor cells, the microglia switches from the M1 (anti-tumor) phenotype to the M2 (anti-inflammatory) phenotype by factors secreted from tumor cells [20] and display tumor-supporting activity. The M2 microglia counteracts TILs activity via the induction of immunosuppressive factors including programmed cell death protein 1(PD1) /programmed death-ligand 1 (PD-L1) [21]. Also, activated astrocytes promote the proliferation and brain invasion of the tumor cells [22]. Obviously, cell types and pathways that initially are activated to counter-act the metastatic process become collaborators in progressive colonization of the brain later on. So far, therapeutic interventions aimed at the elimination of the tumor cells growing in the brain. Future therapeutic strategies may target any of the preceding events, including CTC trafficking and penetration of the BBB.

Advertisement

3. Genetic alterations in NSCLC associated with brain metastasis

Targeted therapies that successfully combat tumor cells outside the brain may fail to be effective behind the BBB. There are several reasons for this, one of which are differences in genetic alterations between the primary tumors and their metastases [23]. Patients suffering from NSCLC have been classified according to the genetic changes in the primary tumor, which include epidermal growth factor receptor (EGFR), Kirsten rat sarcoma (KRAS), and anaplastic lymphoma kinase (ALK). NSCLC brain metastasis-specific mutations can be detected in the cerebrospinal fluid (CSF) and can also be used to evaluate the presence of disease and response to therapy [24].

3.1 EGFR mutations

EGFR is a receptor for extracellular growth factors such as epithelial growth factor (EGF) and tumor growth factor-α (TGFα). Binding of these factors causes a structural change and activation of the receptor complex, resulting in the activation of signaling pathways that promote cell proliferation, motility, and survival. Dysregulation of the receptor is associated with various human cancers. The prevalence of EGFR mutations is dependent on a variety of factors, including ethnicity, gender, smoking, tumor heterogeneity, and tumor progression. EGFR is often overexpressed in NSCLC and the two most frequent EGFR mutations encountered involve exon 19 (deletions) and 21 (L858R mutations) [25, 26, 27]. There is data supporting that CNS metastases of NSCLC are promoted by EGFR-activated mesenchymal–epithelial transition (MET) through mitogen-activated protein kinases (MAPK) signaling. EGFR activates signal transducer and activator of transcription 3 (STAT3) via the expression of interleukin-6 (IL-6) which would increase the risk of BM [28]. NSCLC patients with EGFR mutations at the time of diagnosis or in the early stages of the disease seem to have two times higher risk of brain metastasis [29, 30, 31]. In a series of 30 primary tumor/metastasis series, there was discordance between EGFR status as measured by IHC of one-third of sample pairs and a little less by FISH [32]. In 14 out of 54 paired samples of lung adenocarcinomas, EGFR alterations of EGFR were restricted to the brain metastases [33]. In a recent paper by Haim et al., the EGFR mutational status of brain metastasis could be predicted with an accuracy of almost 90% by using clinical, radiological, and molecular data for deep learning strategies [34]. Obviously, the presence of CNS metastases leads to poorer outcomes (viz., 11.6 months vs 18.7 months) as shown in a study on 101 EGFR positive metastatic NSCLC previously treated with either combination chemotherapy or oral TKI [35]. The progression of the cerebral lesions is also relatively high during treatment in these patients and there is a connection between the EGFR mutations and EMT-related tumor invasion [36, 37].

3.2 KRAS mutations

The K-Ras protein is encoded by the KRAS gene and is part of the RAS/MAPK pathway, where it transfers signals to proliferate and divide from extracellular into the nucleus. A single substitution of a nucleotide may serve as an activator of the signaling pathway turning tissue hyperplasia into invasive cancers. Although it is believed that KRAS and EGFR mutations are mutually exclusive [38, 39], yet cases of simultaneous occurrence were found [40, 41, 42]. Nearly 15–30% of NSCLCs have activating mutations in the KRAS gene that are associated with adenocarcinoma initiation and clinical aggressiveness [38, 43, 44]. There is a clear connection between KRAS mutations and smoking history [45, 46]. In a study of 482 lung adenocarcinomas (LADC), it was found that KRAS mutations also occur in patients who had never smoked, but the mutations differ from those in the tumors of smokers. For instance, transition mutations (G > A) prevail in those who never smoked while transversion mutations (G > T or G > C) are typical for NSCLCs in smokers [47]. The relation between KRAS mutations in NSCLC and propensity for brain metastasis is still unknown and need to be further studied [36, 42]. Approximately 25% of brain metastatic tumors with KRAS mutations were observed in smokers [44]. Other mutations, including ROS proto-oncogene 1, liver kinase B1 (LKB1), and hepatocyte growth factor receptor (HGFR), were associated with the development of lung carcinoma [48, 49, 50], but their relations with brain metastasis of lung cancer is also unknown. LKB1 is inactivated in nearly 30% of all NSCLCs [46] and its effects are synergistic with those of KRAS mutation on the progression of lung cancer and the development of metastases in general [51, 52]. In a study of 154 patients with NSCLC, Zhao et al. concluded that KRAS mutations in combination with low LKB1 copy numbers (CNs) are related to a 20-fold increase in brain metastasis [53]. So far, therapeutic KRAS targeting has been unsuccessful.

3.3 ALK translocations

ALK gene mutations, copy number changes, or fusion with other genes have oncogenic effects. Similar to EGFR mutations, translocations of ALK are predictive of response to Tyrosine Kinase Inhibitors (TKIs) [54]. ALK testing is mostly recommended for non-squamous cell lung cancers lacking EGFR mutations. The fusion between ALK and EML4 (echinoderm microtubule-associated protein-like 4) produces molecular variants with diverse biological functions and affects various signaling pathways [55, 56]. The incidence of cerebral metastases in NSCLC with ALK mutations is high and ALK translocations of primary tumors and their brain metastases are often similar. Interestingly, the progress of brain metastases of tumors with ALK mutations slows down significantly when treated with targeted therapy: over 45% of patients with BM had overall survival rates of three years [57]. Because nearly 45% of patients with ALK-positive NSCLC have developed BM at death [58], cerebral seeding is an important clinical challenge for developing strategies for personalized care in NSCLC [59].

3.4 MET and RET mutations

The large variety of mutations in EMT: (mesenchymal epithelial transition factor) affects a range of cancers, including NSCLC. The MET gene codes for a tyrosine-kinase receptor that plays role in developmental processes and wound healing. Hepatic growth factor/scatter factor (HGF/SF) and their splice isoforms NK1 and 2 are the only known ligands of the MET receptor. In cancer, abnormal MET activation triggers proliferation, angiogenesis, and metastasis. The MET pathways interfere with the key oncogenic pathways RAS, P13K, STAT3, and beta catenin. In general, mutations consist of duplications of mutant alleles, intronic splice site alterations, and mutations affecting the receptor downstream targets. In NSCLC BM, the majority of MET mutations found at metastatic sites affect the extracellular SEMA: Semaphorin superfamily domain of the receptor [60, 61, 62]. Remarkably, mutations in MET occur more frequently in CNS metastasis from NSCLC than in their primary tumors. RET chromosomal rearrangements have been detected in 1–2% of all patients with NSCLC, particularly in patients with adenocarcinoma. The rearrangements are mutually exclusive with EGFR, ALK, or RAS mutations [63]. Importantly, NSCLC with RET rearrangement is associated with an increased risk of BMs [64].

Advertisement

4. The role of MicroRNAs in NSCLC brain metastasis

MicroRNAs (miRNAs) are conserved short endogenous RNA molecules (21–25 nt) that play critical roles in gene expression patterning by interfering with target mRNAs [65]. MiRNAs regulate cellular functions including cell growth, cell differentiation, and cell death. About half of the miRNAs participate in tumorigenesis [66]. The expression of miRNAs may lead to the rise of tumors by activating the pathways implicated in carcinogenesis. The function of miRNAs in the development of tumor metastasis to brain has recently attracted attention and various studies have addressed the role of miRNAs in the progression of brain metastases of lung cancers in particular (Table 1) [83, 84]. The effects of miRNAs vary widely, depending on the expressional cascades they influence.

miRNATissueTargetTumor suppressor/OncogeneEffectReferences
miR-184, miR-197EGFR-mutant lung tumors[67]
miR-21In vivoSPRY2, TIMP3, CDKN1A, SERPINB5 and PTENOncogeneInitiating cell proliferation promoting brain metastasis-[68]
miR-145-5pBrain and lung tumorsTPD52SuppressorInhibited cell invasion and migration[69]
miR-142-3pTCGA dataTRPA1SuppressorSuppressing NSCLC progression[70]
miR-328Brain and lung tumorsPRKCAOncogeneIncreasing cell migration[71]
miR-375Brain and lung tumorsVEGF and MMP-9Suppressor[72]
miR-590Lung tumorsADAM9SuppressorSuppressing tumorigenesis and invasion[73]
miR-378Brain and lung tumorsMMP-2, MMP-9 and VEGFOncogenePromoting migration, invasion, and angiogenesis[74]
miR-95-3pIn vivoCyclin D1SuppressorInhibiting invasion and proliferation[75]
miR-330-3pLung tumorsGRIA3OncogenePromoting growth, tumor invasion, and migration.[76, 77]
miR-490-3pBrain tissuesPCBP1OncogenePromoting proliferation, invasion, and migration[78]
miR-145Brain and lung tumorsSuppressorInhibiting cell proliferation[79]
miR-423-5pLung tumorsMTSS1OncogenePromoting cell invasion and migration.[80]
miR-15a, miR-210, miR-214Lung tumorPredicting brain metastasis in patients with lung adenocarcinoma[81]
miR-4317Lung tumorsFGF9 and CCND2SuppressorInhibiting proliferation, migration, colony formation, and invasion[82]

Table 1.

MicroRNAs associated with brain metastasis from NSCLC.

TPD52: tumor protein D52; TRPA1: transient receptor potential ankyrin 1; GRIA3: glutamate receptor, ionotropic, AMPA 3; PRKCA: protein kinase C-α; MMP: matrix metalloprotease; ADAM9: a disintegrin and metalloproteinase 9; PCBP1: poly r(C)-binding protein 1; MTSS1: metastasis suppressor protein 1; FGF9: fibroblast growth factor 9; CCND2: cyclin D2; and TCGA: The Cancer Genome Atlas.

In Table 1, a summary of currently known miRNA associations with NSCLC and their brain metastases is presented. MiRNA-184 and miRNA-197 are highly expressed in EGFR-mutant NSCLCs of patients with cerebral metastases and may serve as biomarkers for the risk of cerebral seeding [67]. Expression of miRNA-9 and miRNA-1471 has also been found in lung cancer with brain metastasis. Up-regulation of miRNA-145 inhibits the proliferation of human tumor cells in lung adenocarcinomas via targeting of c-Myc and EGFR [79, 85, 86]. MiRNA-146a is overexpressed in NSCLC and is associated with down-regulation of heterogeneous nuclear ribonucleoprotein (hnRNP) C1/C2 and up-regulation of β-catenin, resulting not only in tumor cell invasion and migration but also in the metastatic potential to brain [87, 88]. Also, MiRNA-95-3p is upregulated in lung adenocarcinoma but overexpression of this MiRNA seems to suppress the formation of brain metastasis via down-regulation of cyclin D1 [75]. MiRNA-378 is overexpressed in NSCLC and their brain metastases and increases tumor growth and metastasis via the upregulation of MMP-7, VEGF, and MMP-9 [74]. Also, MiRNA-328 is overexpressed in NSCLC and allegedly promotes the formation of brain metastases via PRKCA and urokinase-type plasminogen activator (uPA) [71]. PRKCA mediates the expression, resulting in the migration of the cancer cells [89]. Lastly, increased miRNA-21 levels suppress cell death and promote the proliferation and invasion of NSCLC and lung adenocarcinoma cells [68, 90].

Some miRNAs are downregulated in the context of cerebral seeding of lung cancer. MiRNA-768-3p is downregulated in lung cancer cells co-cultured with astrocytes, leading to increased KRAS expression, tumor outgrowth, and propagation of brain metastasis [91]. MiRNA-375 is another miRNA that reportedly is down-regulated in primary NSCLC and reduced levels of miRNA-375 are associated with NSCLC brain metastasis [72]. In tumors in which miRNA-375 was downregulated MMP9 and VEGF were found overexpressed [72]. Reduced miRNA-145 levels also seem to promote brain metastasis in lung adenocarcinoma, while overexpression reduces tumor dissemination [69].

Taken together, miRNAs appear to have great potential for cancer diagnosis, prognosis, and treatment at the molecular level, but the use of miRNAs for the clinical treatment of brain metastases requires further investigation. Many studies focused on the identification of alterered expression patterens of miRNAs after outgrowth in the brain microenvironment, but validation of data in larger groups of tumor samples is needed [31].

Advertisement

5. Role of lncRNAs in NSCLC brain metastasis

Long non-coding RNAs (lncRNAs) are non-coding transcripts comprising > 200 nucleotides that have substantial functions in various physiological and pathological pathways. Similar to miRNAs, lncRNAs also regulate a variety of molecular targets by various mechanisms. Recently, the effective role of lncRNAs in tumorigenesis was shown [92]. lncRNAs are important regulators of lung cancer progression. Some lncRNAs serve different functions in various types of cells [93]. MALAT1 (Metastasis-associated lung adenocarcinoma transcript 1) is a large non-coding RNA gene that is highly conserved in mammals and regulates gene expression via splicing-independent mechanisms in NSCLC metastasis [94]. MALAT1 is located on chromosome 11q13 and increased MALAT1 levels were recently discovered in patients with NSCLC who had developed cerebral metastasis, while not in patients without brain locations [95, 96]. In addition, functional studies revealed that overexpression of MALAT1 leads to overexpression of vimentin in highly invasive metastatic lung cancer cell lines while silencing MALAT1 affects EMT programming and suppresses metastasis of the lung cancer cells [96]. Moreover, RNAi-mediated repression of MALAT1-RNA has a negative impact on the migration and outgrowth of human NSCLC cell lines. Overexpression of MALAT1 in NIH/3T3 fibroblasts significantly enhanced migration [97] and stimulated cell motility via the regulation of related genes [98]. The oncogene c-MYC influences cerebral metastasis of NSCLC by inducing the overexpressing of Non-coding RNA BCYRN1 (brain cytoplasmic RNA 1) in NSCLC cells [99, 100]. c-MYC-activated BCYRN1 induces NSCLC metastasis by the expression of MMP9 and MMP13, members of the matrixin subfamily that behave as ECM-degrading enzymes [101, 102, 103]. HOTAIR (HOX transcript antisense RNA) is a lncRNA that is highly expressed in NSCLCs with brain seeding [104]. In vitro studies have revealed that HOTAIR expression enhances tumor cell migration and outgrowth [105]. At this point, the relationship between MALAT1 and HOTAIR in NSCLC brain metastasis is still unknown [104].

Advertisement

6. Novel therapeutic approaches

For many years, the rise of brain metastases of lung cancer has been considered the final stage of the disease. Patients were treated with standard therapeutic options such as palliative care or whole brain radiotherapy (WBRT). However, since the discovery of new systemic and targeted therapies, additional effective treatments for lung cancer were introduced with the aim to enhance local control and survival [106].

6.1 Targeted systemic therapy

The BBB is an obstacle to enter the brain for many agents and has limited the application of drugs used for systemic therapy [107]. The application of drugs targeting EGFR and ALK has heightened the interest in utilizing systemic agents to treat brain metastases [108, 109, 110, 111]. In Table 2 clinical trials of targeted therapy for NSCLC brain metastases are listed.

Targeted agentTargetPretreatment with radiotherapyProgression-free survival (month)PhaseStatusNCT identifier
Alectinib, bevacizumabALK, VEGFNoNAI/IIRecruitingNCT02521051
AT13387, Crizotinibc-MET, ALK, ROS1, Hsp90NoNAI/IICompletedNCT01712217
MK-3475PD-L1NoNAIICompletedNCT02085070
SunitinibVEGF, KIT, PDGF, FLT-3Yes2.1IICompletedNCT00372775
GRN1005-YesNAIICompletedNCT01497665
DasatinibBCR-ABLYesNAIICompletedNCT00787267
CetuximabEGFRYesNAIICompletedNCT00103207
CertinibALKNoNAIIActive, not recruitingNCT02513667
ErlotinibEGFRYes1.6IIICompletedNCT01887795
AfatinibHER2, EGFR, HER4NoNAIIICompletedNCT02044380
OsimertinibEGFRNoNAIIIb/IVCompletedNCT03790397

Table 2.

Clinical trials of targeted therapy for the treatment.

c-MET: tyrosine-protein kinase Met, hepatocyte growth factor receptor; ROS1: Proto-oncogene tyrosine-protein kinase ROS; Hsp90: heat shock protein 90; KIT: Proto-oncogene c-KIT; PDGF: Platelet-derived growth factor; FLT-3: fms like tyrosine kinase 3, CD135; HER2: human epidermal growth factor receptor 2; and HER4: human epidermal growth factor receptor 4.

6.2 EGFR tyrosine kinase inhibitors

Patients with tumors harboring EGFR mutations are prone to develop brain metastases [112, 113]. Although the efficacy of EGFR-TKIs for NSCLCs with EGFR mutations has been proven, its effectiveness is not clear in patients with brain metastases since they were excluded from controlled clinical trials. In a prospective study, 41 patients with unselected NSCLC brain metastasis were treated with Gefitinib resulting in 10% intracranial partial responses (PR) with an average response period of 13.5 months [114]. However, most information on the efficacy of TKIs in patients with brain metastases was obtained from retrospective studies [110]. Firstly, it appeared that recorded concentrations of Afatinib, Erlotinib, and Gefitinib in cerebrospinal fluid (CSF) clearly exceeded those needed to inhibit the growth of cells with EGFR mutations in vitro. In patients with lung adenocarcinoma, about 70% intracranial tumor response was obtained with Gefitinib or Erlotinib as first-line treatment [115]. Other retrospective clinical studies revealed that patients with brain metastases from EGFR-mutant NSCLC have more favorable responses to WBRT or TKI therapy than patients with brain metastases from EGFR–wild-type NSCLC [116]. The progression periods were 11.7 months for patients with EGFR-mutant NSCLCs treated with Erlotinib and 5.8 months for patients with EGFR-wild-type NSCLCs, respectively [117]. The potent EGFR-TKI, AZD3759 showed significant penetration of the BBB in pre-clinical models for the treatment of EGFR-mutant NSCLC with brain metastasis [118]. Moreover, the third-generation EGFR inhibitors osimertinib and rociletinib targeting the T790M-EGFR resistance mutation in NSCLC appeared effective in treating patients with NSCLCs with these mutations [119, 120]. Unfortunately, a phase 3 trial conducted by RTOG using WBRT plus SRS with Temozolomide and Erlotinib in unselected patients with a maximum of three brain metastases was closed prematurely because of low accrual [121]. No significant benefit of adding Gefitinib to WBRT in phase 2 trials in patients with unselected NSCLC with brain metastasis was recorded [122]. At present, there is not sufficient data to draw conclusions on TKI therapy plus CNS-directed radiation therapy for patients with NSCLCs with EGFR mutations.

6.3 ALK tyrosine kinase inhibitors

ALK rearrangements are found in about 4–8% of NSCLCs, representing a distinct subgroup [110]. ALK-TKIs are active against CNS metastases and novel drugs are effective in treating brain metastases, even in patients with multiple intracranial tumors [123]. Crizotinib (Xalkoric) was the first ALK-TKI for metastatic ALK-positive NSCLCs. In phase 3, randomized clinical trial with a single-arm of crizotinib for patients with NSCLC with cerebral metastases, better intracranial response was obtained for patients who were also treated with RT [124] and median survivals of almost 50 months were recorded for patients with ALK-positive tumors [58]. Second-generation ALK-TKIs including brigatinib, ceritinib (Zykadia), and alectinib (Alecensa) have shown a better BBB penetration and activity against BM in crizotinib-resistant tumors [125]. Ceritinib appeared to be a powerful drug for patients with metastatic ALK-positive NSCLCs in whom treatment with crizotinib was not effective anymore. Ceritinib also showed activity against crizotinib-resistant tumors in the mouse models [126]. Patients with ALK-positive tumors with CNS lesions were treated with alectinib against crizotinib in the ALEX trial. The results showed that patients treated with alectinib had a longer progression-free survival (PFS) rate than patients treated with crizotinib [123]. In addition, nearly half of patients with ALK-positive NSCLCs with cerebral metastasis improved significantly upon treatment with alectinib. Taken together, these results indicate that Alectinib can be used as an effective treatment option for patients with NSCLC-positive ALK with cerebral metastasis [123].

6.4 MET inhibitors

In recent years, several MET inhibitors have been approved and have entered clinical trials. There are limited data available on the role and efficacy of monoclonal antibodies that inhibit MET in brain metastasis [127]. The effectiveness of Sym015, which consists of two monoclonal antibodies targeted to non-overlapping epitopes of MET, was high in inhibiting MET-amplified tumors as compared to emibetuzumab, a humanized monoclonal antibody developed for patients with NSCLC [128]. Among the new small inhibitors, cabozantinib, an inhibitor of MET, RET, and VEGFR2, appeared effective in radiation-resistant MET-mutated BM in renal cell carcinoma [129]. In addition, cabozantinib yields rapid responses in crizotinib-resistant NSCLC harboring a MET exon 14 alteration [130]. Simultaneous activation of the MET receptor and the ALK fusion gene in NSCLC yielded effective responses to crizotinib in patients with brain metastases [131]. The oral administered selective MET inhibitor capmatinib came with controllable toxicity profiles in treatment-naive patients with MET-exon14 positive NSCLC. Preliminary studies in mice that were injected with human BM cells from NSCLC showed that capmatinib is able to cross the BBB and is active in the brain. In in vivo models, the combination of capmatinib and afatinib was found to suppress tumor growth [132]. Recently it was demonstrated that bozitinib, another novel orally administered PLB-1001 compound, better penetrated the BBB as compared to other MET inhibitors in MET-mutated glioblastoma [133]. These preliminary results raise hopes for the effectiveness of PLB-1001 in the treatment of secondary brain lesions from various primary sites.

6.5 RET inhibitors

Cabozantinib and vandetanib are oral multi-kinase, non-selective RET inhibitors that have a modest advantage but significant toxicity. Cabozantinib is effective in RET-rearranged NSCLC and has limited activity against RET, while vandetanib more effectively targets RET. No specific activity against CNS seedings of NSCLC has been reported for these drugs [134, 135]. Selpercatinib and pralsetinib are small highly selective RET inhibitors approved by the FDA for the treatment of NSCLC with RET fusion [136, 137]. Selpercatinib (LOXO-292) is an oral tyrosine-kinase inhibitor specifically targeting the RET kinase domain. Its activity profile and clinical safety were evaluated in phase I/II clinical trial LIBRETTO-001. The study included patients with advanced RET-positive NSCLC who had progressed disease after platinum-based chemotherapy in patients who were treatment naïve. In the phase II trial, 105 patients were pretreated with platinum-based chemotherapy. The ORR was 64% (95% confidence interval (CI): 54% to 73%) with a median duration of response of 17.5 months. A major advantage was observed among the 39 treatment naïve patients, with an ORR of 85% (95% CI: 70% to 94%). Selpercatinib was also designed to have an effect on the CNS. Eleven patients with BM participated and Intracranial responses were observed in 10/11 patients with response rates of 91% (95% CI: 6.7% to NE) [138]. The FDA granted to accelerate the approval of selpercatinib for treating patients with metastatic RET-positive NSCLC, regardless of specific treatment strategy. The RET kinase domain inhibitor Pralsetinib (BLU-667) is currently applied in a multicenter phase I/II ARROW trial. Based on the results of this trial, the FDA approved the efficacy of pralsetinib in patients with RET alteration-positive NSCLC with/without prior therapy. Patients with asymptomatic BM were allowed to be included in this trial. In total, 79 patients participated, and the majority were pretreated primarily with chemotherapy (76%) and immunotherapy (41%). CNS metastasis at the baseline observed in 39% of patients. Efficacy was based on 57 patients, all of whom had at least one follow-up evaluation [139, 140].

6.6 Immune therapy

Although the immune system plays a role in all stages of the development of cerebral metastasis, so far therapeutic interference was limited to the immune response around the tumor cells present in the brain. The inflammatory microenvironment of brain metastases mainly consists of infiltration by tumor-infiltrating lymphocytes (TIL) expressing immunosuppressive factors like programmed death-1 (PD-1) ligand (PD-L1). Immunotherapeutic agents include anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), anti- PD-1, and PD-L1 monoclonal antibodies (mAbs). There are limited data available on the efficacy and safety of immunotherapy for patients with NSCLC brain metastasis. Approximately, 15% of patients participated in studies and all had stable BM or had been treated for BM, while patients with symptomatic BM were excluded from trials [16]. The available data were derived from single-arm phase I/II trials [141, 142, 143], pre-arranged analyses of phase III trials [143, 144, 145], and expanded access programs [146, 147]. In the phase I multi-cohort CheckMate 012 study of the tolerability and safety of nivolumab in patients with NSCLC with BM only twelve patients were included. Their median PFS and OS were 1.6 months and 8.0 months, respectively, and no more than two intracranial responses were observed [143].

In a phase 2 trial, the PD-1 blockade by pembrolizumab was studied in patients with advanced NSCLC with untreated brain metastases. Forty-two patients were treated with Pembrolizumab. The cohort with PD-L1 ≥1% 1, 29.7% of patients had a BM response, while the patients with PD-L1 <1% did not show a response. The median OS and PFS of patients in cohort 1 was 9.9 months and 1.9 months, respectively¸ confirming that pembrolizumab activity in CNS metastasis is limited to NSCLC with higher PD-L1 expressions. Moreover, the PD-L1 expression was associated with long-term OS [142]. In two nivolumab EAP studies conducted in Italy and France, 409 and 130 patients respectively, were included with advanced NSCLC and asymptomatic and stable BM. Part of the patients received corticosteroids and the other part underwent concomitant brain radiotherapy. The OR was 17% in the Italian study and 12% in the French study; the OS was 8.6 and 6.6 months, respectively [146, 147]. In another pooled analysis of larger trials on pembrolizumab monotherapy (KEYNOTE 001, 010, 024, and 042) and pembrolizumab combined with chemotherapy (KEYNOTE 021, 189 and 407), the OS of patients who received pembrolizumab alone or with chemotherapy was better as compared to patients who received chemotherapy alone [144, 145].

Advertisement

7. Conclusions

Brain metastasis of NSCLC is most life-threatening for patients and the treatment is a major challenge. Traditional therapies do not eradicate cerebral cancer cells and recurrent disease is common. A significant obstacle in treating patients with brain metastases is the BBB, which prevents chemotherapeutic agents from entering the brain. Due to this obstacle and the failure of conventional therapies, novel therapeutic approaches are being explored. Despite recent advances in lung cancer treatment, a better understanding of the molecular mechanisms and pathways implicated in lung cancer is essential to identify appropriate targets to prevent brain metastasis. It is undeniable that many factors in the tumor microenvironment contribute to the outgrowth of tumor cells, not only at the primary site but also at the sites of seeding in distant organs. The formation of brain metastases is largely the result of tumor-microenvironment interactions. The brain micro-environment not only contributes to colonization by tumor cells but also affects the results of therapeutic interventions. Obviously, detailing the entire spectrum of genomic alterations and molecular mechanisms involved in lung cancer brain metastasis is important to develop effective treatments. Specifically scrutinizing the mechanisms by which cancer cells cross the BBB is important for establishing preventive brain metastases strategies.

References

  1. 1. Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. Journal of Clinical Oncology. 2004;22(14):2865-2872
  2. 2. Johnson JD, Young B. Demographics of brain metastasis. Neurosurgery Clinics of North America. 1996;7(3):337-344
  3. 3. Meyers CA, Smith JA, Bezjak A, Mehta MP, Liebmann J, Illidge T, et al. Neurocognitive function and progression in patients with brain metastases treated with whole-brain radiation and motexafin gadolinium: Results of a Randomized Phase III Trial. Journal of Clinical Oncology. 2004;22(1):157-165
  4. 4. Bar J, Urban D, Amit U, Appel S, Onn A, Margalit O, et al. Long-term survival of patients with metastatic non-small-cell lung cancer over five decades. Journal of Oncology. 2021;2021:10. DOI: 10.1155/2021/7836264. Article ID 7836264
  5. 5. Walters S, Maringe C, Coleman MP, Peake MD, Butler J, Young N, et al. Lung cancer survival and stage at diagnosis in Australia, Canada, Denmark, Norway, Sweden and the UK: A population-based study, 2004-2007. Thorax. 2013;68(6):551-564
  6. 6. Dillekås H, Rogers MS, Straume O. Are 90% of deaths from cancer caused by metastases? Cancer Medicine. 2019;8(12):5574-5576
  7. 7. Bos PD, Zhang XH-F, Nadal C, Shu W, Gomis RR, Nguyen DX, et al. Genes that mediate breast cancer metastasis to the brain. Nature. 2009;459(7249):1005-1009
  8. 8. Valiente M, Obenauf AC, Jin X, Chen Q, Zhang XH-F, Lee DJ, et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 2014;156(5):1002-1016
  9. 9. Carbonell WS, Ansorge O, Sibson N, Muschel R. The vascular basement membrane as “soil” in brain metastasis. PLoS One. 2009;4(6):e5857
  10. 10. Jeon H-S, Jen JJ. TGF-2 signaling and the role of inhibitory smads in non-small cell. Lung Cancer. 2010;5(4):417-419
  11. 11. Yan B, Zhang W, Jiang L-Y, Qin W-X, Wang XJ. Reduced E-Cadherin expression is a prognostic biomarker of non-small cell lung cancer: A meta-analysis based on 2395 subjects. International Journal of Clinical and Experimental Medicine. 2014;7(11):4352
  12. 12. Pastushenko I, Blanpain CJ. EMT transition states during tumor progression and metastasis. Trends in Cell Biology. 2019;29(3):212-226
  13. 13. Tsoukalas N, Aravantinou-Fatorou E, Tolia M, Giaginis C, Galanopoulos M, Kiakou M, et al. Epithelial–mesenchymal transition in non small-cell lung cancer. Anticancer Research. 2017;37(4):1773-1778
  14. 14. Hanibuchi M, Kim S-J, Fidler IJ, Nishioka YJ. The molecular biology of lung cancer brain metastasis: An overview of current comprehensions and future perspectives. The Journal of Medical Investigation : JMI. 2014;61(3):241-253
  15. 15. Popper HHJC, Reviews M. Progression and metastasis of lung cancer. Cancer Metastasis Reviews. 2016;35(1):75-91
  16. 16. El Rassy E, Botticella A, Kattan J, Le Péchoux C, Besse B, Hendriks LJ. Non-small cell lung cancer brain metastases and the immune system: From brain metastases development to treatment. Cancer Treatment Reviews. 2018;68:69-79
  17. 17. Leibold AT, Monaco GN, Dey MJ. The role of the immune system in brain metastasis. Current Neurobiology. 2019;10(2):33
  18. 18. Chuang HN, van Rossum D, Sieger D, Siam L, Klemm F, Bleckmann A, et al. Carcinoma cells misuse the host tissue damage response to invade the brain. Glia. 2013;61(8):1331-1346
  19. 19. Owyong M, Hosseini-Nassab N, Efe G, Honkala A, van den Bijgaart RJ, Plaks V, et al. Cancer immunotherapy getting brainy: Visualizing the distinctive CNS metastatic niche to illuminate therapeutic resistance. Drug Resistance Updates: Reviews and Commentaries in Antimicrobial and Anticancer Chemotherapy. 2017;33:23-35
  20. 20. Soto MS, Sibson NR. The multifarious role of microglia in brain metastasis. Frontiers in Cellular Neuroscience. 2018;12:414
  21. 21. Berghoff AS, Fuchs E, Ricken G, Mlecnik B, Bindea G, Spanberger T, et al. Density of tumor-infiltrating lymphocytes correlates with extent of brain edema and overall survival time in patients with brain metastases. Oncoimmunology. 2016;5(1):e1057388
  22. 22. Seike T, Fujita K, Yamakawa Y, Kido MA, Takiguchi S, Teramoto N, et al. Interaction between lung cancer cells and astrocytes via specific inflammatory cytokines in the microenvironment of brain metastasis. Clinical & Experimental Metastasis. 2011;28(1):13-25
  23. 23. Brastianos PK, Carter SL, Santagata S, Cahill DP, Taylor-Weiner A, Jones RT, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discovery. 2015;5(11):1164-1177
  24. 24. Preusser M, Winkler F, Valiente M, Manegold C, Moyal E, Widhalm G, et al. Recent advances in the biology and treatment of brain metastases of non-small cell lung cancer: Summary of a multidisciplinary roundtable discussion. ESMO Open. 2018;3(1):e000262
  25. 25. Burel-Vandenbos F, Ambrosetti D, Coutts M, Pedeutour FJ. EGFR mutation status in brain metastases of non-small cell lung carcinoma. Journal of Neuro-Oncology. 2013;111(1):1-10
  26. 26. Jain D, Iqbal S, Walia R, Malik P, Cyriac S, Mathur SR, et al. Evaluation of epidermal growth factor receptor mutations based on mutation specific immunohistochemistry in non-small cell lung cancer: A preliminary study. The Indian Journal of Medical Research. 2016;143(3):308
  27. 27. Li AR, Chitale D, Riely GJ, Pao W, Miller VA, Zakowski MF, et al. EGFR mutations in lung adenocarcinomas: Clinical testing experience and relationship to EGFR gene copy number and immunohistochemical expression. The Journal of Molecular Diagnostics. 2008;10(3):242-248
  28. 28. Li L, Luo S, Lin H, Yang H, Chen H, Liao Z, et al. Correlation between EGFR mutation status and the incidence of brain metastases in patients with non-small cell lung cancer. Journal of Thoracic Disease. 2017;9(8):2510
  29. 29. Bhatt VR, D’Souza SP, Smith LM, Cushman-Vokoun AM, Noronha V, Verma V, et al. Epidermal growth factor receptor mutational status and brain metastases in non–small-cell lung cancer. Journal of Global Oncology. 2017;3(3):208-217
  30. 30. Li Z, Lu J, Zhao Y, Guo HJ. The retrospective analysis of the frequency of EGFR mutations and the efficacy of gefitinib in NSCLC patients with brain metastasis. Journal of Clinical Oncology. 2011;29(15):e18065
  31. 31. Pedrosa RM, Mustafa DA, Aerts JG, Kros JM. Potential molecular signatures predictive of lung cancer brain metastasis. Frontiers in Oncology. 2018;8:159
  32. 32. Italiano A, et al. Comparison of the epidermal growth factor receptor gene and protein in primary non-small-cell-lung cancer and metastatic sites: implications for treatment with EGFR-inhibitors. Annals of Oncology. 2006;17(6):981-985
  33. 33. Vassella E, et al. Mutational profiles of primary pulmonary adenocarcinoma and paired brain metastases disclose the importance of KRAS mutations. European Journal of Cancer. 2021;159:227-236
  34. 34. Haim O, et al. Predicting EGFR mutation status by a deep learning approach in patients with non-small cell lung cancer brain metastases. Journal of Neuro-Oncology. 2022;157(1):63-69
  35. 35. Noronha V, Joshi A, Gokarn A, Sharma V, Patil V, Janu A, et al. The importance of brain metastasis in EGFR mutation positive NSCLC patients. Chemotherapy Research and Practice. Hindawi Publishing Corporation Chemotherapy Research and Practice. 2014;2014:4. DOI: 10.1155/2014/856156. Article ID 856156
  36. 36. Cortot AB, Italiano A, Burel-Vandenbos F, Martel-Planche G, Hainaut PJC. KRAS mutation status in primary nonsmall cell lung cancer and matched metastases. Cancer. 2010;116(11):2682-2687
  37. 37. Buonato JM, Lazzara MJ. ERK1/2 blockade prevents epithelial–mesenchymal transition in lung cancer cells and promotes their sensitivity to EGFR inhibition. Cancer Research. 2014;74(1):309-319
  38. 38. Ahrendt SA, Decker PA, Alawi EA, Zhu Y, Sanchez-Cespedes M, Yang SC, et al. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer. 2001;92(6):1525-1530
  39. 39. Marks JL, Broderick S, Zhou Q, Chitale D, Li AR, Zakowski MF, et al. Prognostic and therapeutic implications of EGFR and KRAS mutations in resected lung adenocarcinoma. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2008;3(2):111-116
  40. 40. Fujimoto N, Wislez M, Zhang J, Iwanaga K, Dackor J, Hanna AE, et al. High expression of ErbB family members and their ligands in lung adenocarcinomas that are sensitive to inhibition of epidermal growth factor receptor. Cancer Research. 2005;65(24):11478-11485
  41. 41. Han S-W, Kim T-Y, Jeon YK, Hwang PG, Im S-A, Lee K-H, et al. Optimization of patient selection for gefitinib in non–small cell lung cancer by combined analysis of epidermal growth factor receptor mutation, K-ras mutation, and Akt phosphorylation. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2006;12(8):2538-2544
  42. 42. Kalikaki A, Koutsopoulos A, Trypaki M, Souglakos J, Stathopoulos E, Georgoulias V, et al. Comparison of EGFR and K-RAS gene status between primary tumours and corresponding metastases in NSCLC. British Journal of Cancer. 2008;99(6):923-929
  43. 43. Rodenhuis S, van de Wetering ML, Mooi WJ, Evers SG, van Zandwijk N, Bos JL. Mutational activation of the K-ras oncogene. The New England Journal of Medicine. 1987;317(15):929-935
  44. 44. Munfus-McCray D, Harada S, Adams C, Askin F, Clark D, Gabrielson E, et al. EGFR and KRAS mutations in metastatic lung adenocarcinomas. Human Pathology. 2011;42(10):1447-1453
  45. 45. Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non–small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. Journal of Clinical Oncology. 2005;23(25):5900-5909
  46. 46. Sanchez-Cespedes M, Parrella P, Esteller M, Nomoto S, Trink B, Engles JM, et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Research. 2002;62(13):3659-3662
  47. 47. Quandt D, Fiedler E, Boettcher D, Marsch WC, Seliger BJ. B7-h4 expression in human melanoma: Its association with patients' survival and antitumor immune response. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2011;17(10):3100-3111
  48. 48. Demuth C, Andersen MN, Jakobsen KR, Madsen AT, Sørensen BSJO. Increased PD-L1 expression in erlotinib-resistant NSCLC cells with MET gene amplification is reversed upon MET-TKI treatment. Oncotarget. 2017;8(40):68221
  49. 49. Ji M, Liu Y, Li Q, Li X-D, Zhao W-Q, Zhang H, et al. PD-1/PD-L1 pathway in non-small-cell lung cancer and its relation with EGFR mutation. Journal of Translational Medicine. 2015;13(1):5
  50. 50. Killock DJN. Lung cancer: Alternative rearrangements—targeting ROS1 in NSCLC. Cancers. 2014;11(11):624
  51. 51. Carretero J, Shimamura T, Rikova K, Jackson AL, Wilkerson MD, Borgman CL, et al. Integrative genomic and proteomic analyses identify targets for Lkb1-deficient metastatic lung tumors. Cancer Cell. 2010;17(6):547
  52. 52. Ji H, Ramsey MR, Hayes DN, Fan C, McNamara K, Kozlowski P, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807
  53. 53. Zhao N, Wilkerson MD, Shah U, Yin X, Wang A, Hayward MC, et al. Alterations of LKB1 and KRAS and risk of brain metastasis: Comprehensive characterization by mutation analysis, copy number, and gene expression in non-small-cell lung carcinoma. Lung Cancer. 2014;86(2):255
  54. 54. Peled N, Zach L, Liran O, Ilouze M, Bunn PA, Hirsch FR. Effective crizotinib schedule for brain metastases in ALK rearrangement metastatic non–small-cell lung cancer. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2013;8(12):e112-e1e3
  55. 55. Shaw AT, Engelman JA. ALK in lung cancer: Past, present, and future. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2013;31(8):1105
  56. 56. Sabir SR, Yeoh S, Jackson G, Bayliss RJC. EML4-ALK variants: Biological and molecular properties, and the implications for patients. Cancers. 2017;9(9):118
  57. 57. Vickers NJ. Animal communication: When I’m calling you, will you answer too? Current Biology. 2017;27(14):R713
  58. 58. Johung KL, Yeh N, Desai NB, Williams TM, Lautenschlaeger T, Arvold ND, et al. Extended survival and prognostic factors for patients with ALK-rearranged non–small-cell lung cancer and brain metastasis. Journal of Clinical Oncology. 2016;34(2):123
  59. 59. Rangachari D, Yamaguchi N, VanderLaan PA, Folch E, Mahadevan A, Floyd SR, et al. Brain metastases in patients with EGFR-mutated or ALK-rearranged non-small-cell lung cancers. Lung Cancer. 2015;88(1):108-111
  60. 60. Stella GM, Senetta R, Inghilleri S, Di Cantogno LV, Mantovani C, Piloni D, et al. MET mutations are associated with aggressive and radioresistant brain metastatic non-small-cell lung cancer. Neuro-Oncology. 2016;18(4):598
  61. 61. Milan M, Benvenuti S, Balderacchi AM, Virzi AR, Gentile A, Senetta R, et al. RON tyrosine kinase mutations in brain metastases from lung cancer. ERJ Open Research. 2018;4(1):598
  62. 62. Gherardi E, Love CA, Esnouf RM, Jones EY. The sema domain. Current Opinion in Structural Biology. 2004;14(6):669-678
  63. 63. Song Z, Yu X, Zhang Y. Clinicopathologic characteristics, genetic variability and therapeutic options of RET rearrangements patients in lung adenocarcinoma. Lung Cancer. 2016;101:16-21
  64. 64. Drilon A, Lin JJ, Filleron T, Ni A, Milia J, Bergagnini I, et al. Frequency of brain metastases and multikinase inhibitor outcomes in patients with RET–rearranged lung cancers. Journal of Thoracic Oncology. 2018;13(10):1595-1601
  65. 65. Markopoulos GS, Roupakia E, Tokamani M, Chavdoula E, Hatziapostolou M, Polytarchou C, et al. A step-by-step microRNA guide to cancer development and metastasis. Cellular Oncology. Dordrecht. 2017;40(4):303-339
  66. 66. Chen P-S, Su J-L, Hung M. Dysregulation of microRNAs in cancer. Journal of Biomedical Science. 2012;19(1):90
  67. 67. Remon J, Alvarez-Berdugo D, Majem M, Moran T, Reguart N, Lianes PJC, et al. miRNA-197 and miRNA-184 are associated with brain metastasis in EGFR-mutant lung cancers. Clinical & Translational Oncology. 2016;18(2):153-159
  68. 68. Singh M, Garg N, Venugopal C, Hallett R, Tokar T, McFarlane N, et al. STAT3 pathway regulates lung-derived brain metastasis initiating cell capacity through miR-21 activation. Oncotarget. 2015;6(29):27461
  69. 69. Donzelli S, Mori F, Bellissimo T, Sacconi A, Casini B, Frixa T, et al. Epigenetic silencing of miR-145-5p contributes to brain metastasis. Oncotarget. 2015;6(34):35183
  70. 70. Berrout J, Kyriakopoulou E, Moparthi L, Hogea AS, Berrout L, Ivan C, et al. TRPA1–FGFR2 binding event is a regulatory oncogenic driver modulated by miRNA-142-3p. Nature Communications. 2017;8:1-15
  71. 71. Arora S, Ranade AR, Tran NL, Nasser S, Sridhar S, Korn RL, et al. MicroRNA-328 is associated with (non-small) cell lung cancer (NSCLC) brain metastasis and mediates NSCLC migration. International Journal of Cancer. 2011;129(11):2621-2631
  72. 72. Chen L-j, Li X-y, Zhao Y-q, Liu W-j, Wu H-j, Liu J, et al. Down-regulated microRNA-375 expression as a predictive biomarker in non-small cell lung cancer brain metastasis and its prognostic significance. Pathology, Research and Practice. 2017;213:882
  73. 73. Wang F-f, Wang S, Xue W, Cheng J. Biochemistry c. microRNA-590 suppresses the tumorigenesis and invasiveness of non-small cell lung cancer cells by targeting ADAM9. Molecular and Cellular Biochemistry. 2016;423(1-2):29-37
  74. 74. L-t C, Xu S-d, Xu H, Zhang J, Ning J, Wang S. MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Medical oncology. Northwood, London, England. 2012;29(3):1673
  75. 75. Hwang SJ, Lee HW, Kim HR, Song HJ, Lee DH, Lee H, et al. Overexpression of microRNA-95-3p suppresses brain metastasis of lung adenocarcinoma through downregulation of cyclin D1. Oncotarget. 2015;6(24):20434
  76. 76. Wei C-H, Wu G, Cai Q, Gao X-C, Tong F, Zhou R, et al. MicroRNA-330-3p promotes cell invasion and metastasis in non-small cell lung cancer through GRIA3 by activating MAPK/ERK signaling pathway. Journal of Hematology & Oncology. 2017;10(1):125
  77. 77. Jiang L-P, Zhu Z-T, Zhang Y, He C-Y. Downregulation of MicroRNA-330 correlates with the radiation sensitivity and prognosis of patients with brain metastasis from lung CancerDownregulation of MicroRNA-330 correlates with the radiation sensitivity and prognosis of patients with brain metastasis from lung Cancer. Biochemistry. 2017;42:2220-2229
  78. 78. Li J, Feng Q, Wei X, Yu YJTB. MicroRNA-490 regulates lung cancer metastasis by targeting poly r (C)-binding protein 1. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine. 2016;37(11):15221-15228
  79. 79. Zhao C, Xu Y, Zhang Y, Tan W, Xue J, Yang Z, et al. Downregulation of miR-145 contributes to lung adenocarcinoma cell growth to form brain metastases. Oncology Reports. 2013;30(5):2027-2034
  80. 80. Sun G, Ding X, Bi N, Wu L, Wang J, Zhang W, et al. MiR-423-5p in brain metastasis: Potential role in diagnostics and molecular biology. Cell Death & Disease. 2018;9(10):1-15
  81. 81. Zhao S, Yu J, Wang LJ. Machine learning based prediction of brain metastasis of patients with IIIA-N2 lung adenocarcinoma by a three-miRNA signature. Translational Oncology. 2018;11(1):157-167
  82. 82. He X, Chen S, Yang Z, Zhang J, Wang W, Liu M, et al. miR-4317 suppresses non-small cell lung cancer (NSCLC) by targeting fibroblast growth factor 9 (FGF9) and cyclin D2 (CCND2). Journal of Experimental & Clinical Cancer Research. 2018;37(1):230
  83. 83. Alsidawi S, Malek E, Driscoll JJ. MicroRNAs in brain metastases: Potential role as diagnostics and therapeutics. Journal of Molecular Medicine. International Journal of Molecular Sciences. 2014;15(6):10508-10526
  84. 84. McDermott R, Gabikian P, Sarvaiya P, Ulasov I, Lesniak MS. MicroRNAs in brain metastases: Big things come in small packages. Journal of Molecular Medicine. 2013;91(1):5-13
  85. 85. Cho WC, Chow AS, Au JS. MiR-145 inhibits cell proliferation of human lung adenocarcinoma by targeting EGFR and NUDT1. RNA Biology. 2011;8(1):125-131
  86. 86. Cho WC, Chow AS, Au JS. Restoration of tumour suppressor hsa-miR-145 inhibits cancer cell growth in lung adenocarcinoma patients with epidermal growth factor receptor mutation. European Journal of Cancer. 2009;45(12):2197-2206
  87. 87. Chen G, Umelo IA, Lv S, Teugels E, Fostier K, Kronenberger P, et al. miR-146a inhibits cell growth, cell migration and induces apoptosis in non-small cell lung cancer cells. PloS One. 2013;8(3):7
  88. 88. Hwang SJ, Seol HJ, Park YM, Kim KH, Gorospe M, Nam D-H, et al. MicroRNA-146a suppresses metastatic activity in brain metastasis. Molecules and Cells. 2012;34(3):329-334
  89. 89. Cheng CY, Hsieh HL, Sun CC, Lin CC, Luo SF, Yang CM. IL-1β induces urokinse-plasminogen activator expression and cell migration through PKCα, JNK1/2, and NF-κB in A549 cells. Journal of Cellular Physiology. 2009;219(1):183-193
  90. 90. Bica-Pop C, Cojocneanu-Petric R, Magdo L, Raduly L, Gulei D, Berindan-Neagoe IJC, et al. Overview upon miR-21 in lung cancer: Focus on NSCLC. Cellular and Molecular life Sciences: CMLS. 2018;75(19):3539
  91. 91. Subramani A, Alsidawi S, Jagannathan S, Sumita K, Sasaki AT, Aronow B, et al. The brain microenvironment negatively regulates miRNA-768-3p to promote K-ras expression and lung cancer metastasis. Scientific Reports. 2013;3:2392
  92. 92. Vitiello M, Tuccoli A, Poliseno LJ. Long non-coding RNAs in cancer: Implications for personalized therapy. Cellular Oncology. 2015;38(1):17-28
  93. 93. Gutschner T, Diederichs SJ. The hallmarks of cancer: A long non-coding RNA point of view. RNA Biology. 2012;9(6):703-719
  94. 94. Wang M, Sun X, Wang H, Xin Y, Jiao WJT. Long non-coding RNAs in non-small cell lung cancer: Functions and distinctions from other malignancies. Translational Cancer Research. 2019;8(7):2636-2653
  95. 95. Zhang X, Hamblin MH, Yin K. The long noncoding RNA Malat1: Its physiological and pathophysiological functions. RNA Biology. 2017;14(12):1705-1714
  96. 96. Shen L, Chen L, Wang Y, Jiang X, Xia H, Zhuang ZJ. Long noncoding RNA MALAT1 promotes brain metastasis by inducing epithelial-mesenchymal transition in lung cancer. Journal of Neuro-Oncology. 2015;121(1):101-108
  97. 97. Peng W, Fan HJB. Long non-coding RNA PANDAR correlates with poor prognosis and promotes tumorigenesis in hepatocellular carcinoma. Pharmacotherapy. 2015;72:113-118
  98. 98. Li X, Wang F, Sun Y, Fan Q, Cui GJ. Expression of long non-coding RNA PANDAR and its prognostic value in colorectal cancer patients. The International Journal of Biological Markers. 2017;32(2):218-223
  99. 99. Hu T, Lu Y. BCYRN1, a c-MYC-activated long non-coding RNA, regulates cell metastasis of non-small-cell lung cancer. Cancer Cell International. 2015;15(1):36
  100. 100. Rapp UR, Korn C, Ceteci F, Karreman C, Luetkenhaus K, Serafin V, et al. MYC is a metastasis gene for non-small-cell lung cancer. Cancer Cell International. 2009;4(6):9
  101. 101. Nabeshima K, Inoue T, Shimao Y, Sameshima TJ. Matrix metalloproteinases in tumor invasion: Role for cell migration. Pathology International. 2002;52(4):255-264
  102. 102. Yu T, Wu Y, Helman JI, Wen Y, Wang C, Li LJ. CXCR4 promotes oral squamous cell carcinoma migration and invasion through inducing expression of MMP-9 and MMP-13 via the ERK signaling pathway. Molecular Cancer Research: MCR. 2011;9(2):161-172
  103. 103. Ghosh S, Basu M, Roy SS. ETS-1 protein regulates vascular endothelial growth factor-induced matrix metalloproteinase-9 and matrix metalloproteinase-13 expression in human ovarian carcinoma cell line SKOV-3. The Journal of Biological Chemistry. 2012;287(18):15001-150015
  104. 104. Nakagawa T, Endo H, Yokoyama M, Abe J, Tamai K, Tanaka N, et al. Large noncoding RNA HOTAIR enhances aggressive biological behavior and is associated with short disease-free survival in human non-small cell lung cancer. Biochemical and Biophysical Research Communications. 2013;436(2):319-324
  105. 105. Wu K, Sharma S, Venkat S, Liu K, Zhou X, Watabe KJ. Non-coding RNAs in cancer brain metastasis. Frontiers in Bioscience. 2016;8:187
  106. 106. Soffietti R, Abacioglu U, Baumert B, Combs SE, Kinhult S, Kros JM, et al. Diagnosis and treatment of brain metastases from solid tumors: Guidelines from the European Association of Neuro-Oncology (EANO). Neuro-Oncology. 2017;19(2):162-174
  107. 107. Churilla TM, Weiss SE. Emerging trends in the management of brain metastases from non-small cell lung cancer. Current Oncology Reports. 2018;20(7):54
  108. 108. Wong AJ. The emerging role of targeted therapy and immunotherapy in the management of brain metastases in non-small cell lung cancer. Frontiers in Oncology. 2017;7:33
  109. 109. Dagogo-Jack I, Gill CM, Cahill DP, Santagata S, Brastianos PK. Treatment of brain metastases in the modern genomic era. Therapeutics. 2017;170:64-72
  110. 110. Chamberlain MC, Baik CS, Gadi VK, Bhatia S, Chow LQ. Systemic therapy of brain metastases: Non–small cell lung cancer, breast cancer, and melanoma. Neuro-Oncology. 2017;19(1):i1-i24
  111. 111. Sekine A, Satoh HJ. Paradigm shift of therapeutic management of brain metastases in EGFR-mutant non-small cell lung cancer in the era of targeted therapy. Medical Oncology. 2017;34(7):121
  112. 112. Eichler AF, Kahle KT, Wang DL, Joshi VA, Willers H, Engelman JA, et al. EGFR mutation status and survival after diagnosis of brain metastasis in nonsmall cell lung cancer. Neuro-Oncology. 2010;12(11):1193-1199
  113. 113. Gainor JF, Ou S-HI, Logan J, Borges LF, Shaw AT. The central nervous system as a sanctuary site in ALK-positive non–small-cell lung cancer. 2013;8(12):1570-1573
  114. 114. Wu C, Li YL, Wang ZM, Li Z, Zhang TX, Wei Z. Gefitinib as palliative therapy for lung adenocarcinoma metastatic to the brain. Lung Cancer. 2007;57(3):359-364
  115. 115. Kim J-E, Lee DH, Choi Y, Yoon DH, Kim S-W, Suh C, et al. Epidermal growth factor receptor tyrosine kinase inhibitors as a first-line therapy for never-smokers with adenocarcinoma of the lung having asymptomatic synchronous brain metastasis. Lung Cancer. 2009;65(3):351-354
  116. 116. Lee H-L, Chung T-S, Ting L-L, Tsai J-T, Chen S-W, Chiou J-F, et al. EGFR mutations are associated with favorable intracranial response and progression-free survival following brain irradiation in non-small cell lung cancer patients with brain metastases. Radiation Oncology. 2012;7(1):181
  117. 117. Porta R, Sanchez-Torres J, Paz-Ares L, Massuti B, Reguart N, Mayo C, et al. Brain metastases from lung cancer responding to erlotinib: The importance of EGFR mutation. The European Respiratory Journal. 2011;37(3):624-631
  118. 118. Kim D-W, Yang JC-H, Chen K, Cheng Z, Yin L, Martin PD, et al. AZD3759, an EGFR inhibitor with blood brain barrier (BBB) penetration for the treatment of non-small cell lung cancer (NSCLC) with brain metastasis (BM): Preclinical evidence and clinical cases. American Society of Clinical Oncology. Science Translational Medicine. 7 Dec 2016;8(368):368ra172. DOI: 10.1126/scitranslmed.aag0976
  119. 119. Jänne PA, Yang JC-H, Kim D-W, Planchard D, Ohe Y, Ramalingam SS, et al. AZD9291 in EGFR inhibitor–resistant non–small-cell lung cancer. 2015;372(18):1689-1699
  120. 120. Xie L, Nagpal S, Wakelee HA, Li G, Soltys SG, Neal JW. Osimertinib for EGFR-mutant lung cancer with brain metastases: Results from a Single-Center Retrospective Study. The Oncologist. 2019;24(6):836-843
  121. 121. Sperduto PW, Wang M, Robins HI, Schell MC, Werner-Wasik M, Komaki R, et al. A phase 3 trial of whole brain radiation therapy and stereotactic radiosurgery alone versus WBRT and SRS with temozolomide or erlotinib for non-small cell lung cancer and 1 to 3 brain metastases: Radiation Therapy Oncology Group 0320. International Journal of Radiation Oncology, Biology, Physics. 2013;85(5):1312-1318
  122. 122. Pesce GA, Klingbiel D, Ribi K, Zouhair A, Von Moos R, Schlaeppi M, et al. Outcome, quality of life and cognitive function of patients with brain metastases from non-small cell lung cancer treated with whole brain radiotherapy combined with gefitinib or temozolomide. A randomised phase II trial of the Swiss Group for Clinical Cancer Research (SAKK 70/03). Journal of Clinical Oncology. 201248;35(3):377-384
  123. 123. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim D-W, et al. Alectinib versus crizotinib in untreated ALK-positive non–small-cell lung cancer. The New England Journal of Medicine. 2017;377(9):829-838
  124. 124. Costa DB, Shaw AT, Ou S-HI, Solomon BJ, Riely GJ, Ahn M-J, et al. Clinical experience with crizotinib in patients with advanced ALK-rearranged non–small-cell lung cancer and brain metastases. Journal of Clinical Oncology. 2015;33(17):1881
  125. 125. Beardslee T, Lawson JJ. Alectinib and Brigatinib: New second-generation ALK inhibitors for the treatment of non–small cell lung cancer. Journal of the Advanced Practitioner in Oncology. 2018;9(1):94
  126. 126. Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, et al. The ALK inhibitor ceritinib overcomes crizotinib resistance in non–small cell lung cancer. Cancer Discovery. 2014;4(6):662-673
  127. 127. Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, et al. Randomized, double-blind, placebo-controlled, multicenter phase II study of onartuzumab plus bevacizumab versus placebo plus bevacizumab in patients with recurrent glioblastoma: Efficacy, safety, and hepatocyte growth factor and O (6)-methylguanine-DNA methyltransferase biomarker analyses. Journal of Clinical Oncology. 2017;35(3):343-351
  128. 128. Poulsen TT, Grandal MM, Skartved NJØ, Hald R, Alifrangis L, Koefoed K, et al. Sym015: A highly efficacious antibody mixture against MET-amplified tumors. Clinical Cancer Research. 2017;23(19):5923-5935
  129. 129. Négrier S, Moriceau G, Attignon V, Haddad V, Pissaloux D, Guerin N, et al. Activity of cabozantinib in radioresistant brain metastases from renal cell carcinoma: Two case reports. Journal of Medical Case Reports. 2018;12(1):1-6
  130. 130. Klempner SJ, Borghei A, Hakimian B, Ali SM, Ou S-HI. Intracranial activity of cabozantinib in MET exon 14–positive NSCLC with brain metastases. Journal of Thoracic Oncology. 2017;12(1):152-156
  131. 131. Wang P, Xiao P, Ye Y, Liu P, Han L, Dong L, et al. Rapid response of brain metastasis to crizotinib in a patient with KLC1-ALK fusion and MET gene amplification positive non-small cell lung cancer: A case report. Cancer Biology & Medicine. 2017;14(2):183
  132. 132. Martinez-Marti A, Felip E, Matito J, Mereu E, Navarro A, Cedrés S, et al. DualMET andERBB inhibition overcomes intratumor plasticity in osimertinib-resistant-advanced non-small-cell lung cancer (NSCLC). Annals of Oncology. 2017;28(10):2451-2457
  133. 133. Hu H, Mu Q, Bao Z, Chen Y, Liu Y, Chen J, et al. Mutational landscape of secondary glioblastoma guides MET-targeted trial in brain tumor. Cells. 2018;175(6):1665-1678
  134. 134. Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Molecular Cancer Therapeutics. 2011;10(12):2298-2308
  135. 135. Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, et al. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Research. 2002;62(24):7284-7290
  136. 136. Subbiah V, Velcheti V, Tuch B, Ebata K, Busaidy N, Cabanillas M, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Annals of Oncology. 2018;29(8):1869-1876
  137. 137. Gautschi O, Milia J, Filleron T, Wolf J, Carbone DP, Owen D, et al. Targeting RET in patients with RET-rearranged lung cancers: Results from the global, multicenter RET registry. Journal of Clinical Oncology. 2017;35(13):1403
  138. 138. Drilon A, Oxnard GR, Tan DS, Loong HH, Johnson M, Gainor J, et al. Efficacy of selpercatinib in RET fusion–positive non–small-cell lung cancer. The New England Journal of Medicine. 2020;383(9):813-824
  139. 139. Gainor JF, Curigliano G, Kim D-W, Lee DH, Besse B, Baik CS, et al. Registrational dataset from the phase I/II ARROW trial of pralsetinib (BLU-667) in patients with advanced RET fusion+ non-small cell lung cancer (NSCLC). Chemotherapy. 2020;71(100):0
  140. 140. Gainor JF, Lee DH, Curigliano G, Doebele RC, Kim D-W, Baik CS, et al. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients with advanced RET-Fusion+ non small cell lung cancer. Journal of Clinical Oncology. 2019;37:9008
  141. 141. Goldberg SB, Gettinger SN, Mahajan A, Chiang AC, Herbst RS, Sznol M, et al. Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: Early analysis of a non-randomised, open-label, phase 2 trial. The Lancet Oncology. 2016;17(7):976-983
  142. 142. Goldberg SB, Schalper KA, Gettinger SN, Mahajan A, Herbst RS, Chiang AC, et al. Pembrolizumab for management of patients with NSCLC and brain metastases: Long-term results and biomarker analysis from a non-randomised, open-label, phase 2 trial. The Lancet Oncology. 2020;21(5):655-663
  143. 143. Goldman JW, Crino L, Vokes EE, Holgado E, Reckamp K, Pluzanski A, et al. P2. 36: Nivolumab (nivo) in patients (pts) with advanced (adv) NSCLC and central nervous system (CNS) metastases (mets): Track: I. Journal of Thoracic Oncology. 2016;11(10):S238-S2S9
  144. 144. Mansfield A, Herbst R, Castro G Jr, Hui R, Peled N, Kim D-W, et al. Outcomes with pembrolizumab (pembro) monotherapy in patients (pts) with PD-L1–positive NSCLC with brain metastases: Pooled analysis of KEYNOTE-001,-010,-024, and-042. Annals of Oncology. 2019;30:v604-v6v6
  145. 145. Powell S, Abreu DR, Langer C, Tafreshi A, Paz-Ares L, Kopp H-G, et al. Pembrolizumab (pembro) plus platinum-based chemotherapy (chemo) in NSCLC with brain metastases: Pooled analysis of KEYNOTE-021, 189, and 407. Annals of Oncology. 2019;30:v606-v6v7
  146. 146. Girard N, Valette CA, Cadranel J, Monnet I, Hureaux J, Hilgers W, et al. IFCT-1502 CLINIVO: Real-life experience with nivolumab in 600 patients (pts) with advanced non-small cell lung cancer (NSCLC): Efficacy and safety of nivolumab and post-nivolumab treatment in the French Expanded Access Program (EAP). Annals of Oncology. 2017;28:v463
  147. 147. Crinò L, Bronte G, Bidoli P, Cravero P, Minenza E, Cortesi E, et al. Nivolumab and brain metastases in patients with advanced non-squamous non-small cell lung cancer. Lung Cancer. 2019;129:35-40

Written By

Shiva Najjary, Dana A.M. Mustafa and Johan M. Kros

Submitted: 08 June 2022 Reviewed: 07 July 2022 Published: 04 October 2022