Anda di halaman 1dari 17

Curr Treat Options Neurol (2018) 20:48

DOI 10.1007/s11940-018-0533-2

Neuro-oncology (R Soffietti, Section Editor)

Novel Systemic Treatments


for Brain Metastases From
Lung Cancer
Bicky Thapa, MD1
Adam Lauko, BS (Medical Student)2
Kunal Desai, MD2
Vyshak Alva Venur, MD3
Manmeet S. Ahluwalia, MD, FACP2,*
Address
1
Fairview Hospital-Cleveland Clinic, Cleveland, OH, USA
*,2
Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute,
Cleveland Clinic, Cleveland, OH, 44195, USA
Email: ahluwam@ccf.org
3
Dana-Farber Cancer Institute and Massachusetts General Hospital, Boston, MA,
USA

* Springer Science+Business Media, LLC, part of Springer Nature 2018

This article is part of the Topical Collection on Neuro-oncology

Keywords Brain metastases I Lung cancer I EGFR I ALK I Targeted therapy I Immunotherapy I Clinical trials

Abstract
Purpose of review Brain metastases are frequent complication of lung cancer and are
associated with poor prognosis. Patients with brain metastases secondary to lung cancer
have traditionally been managed with surgery and radiation with limited role for systemic
chemotherapy. In the past decade, however, this paradigm has shifted largely due to the
advent of targeted therapies and immunotherapies, both of which have demonstrated
efficacy in the treatment of brain metastases and extracranial disease.
Recent findings While patients with brain metastases secondary to lung cancer have
historically been excluded from trials, recent data suggest efficacy of novel targeted
therapies and immunotherapies in these patients. In fact, there are multiple ongoing
trials to further evaluate these therapies in this patient profile.
Summary Targeted therapies and immunotherapies have the potential to improve out-
comes in patients with brain metastases secondary to lung cancer.
48 Page 2 of 17 Curr Treat Options Neurol (2018) 20:48

Introduction
Lung cancer is the most common cause of cancer-related quality of life. The advent of targeted therapies and
death in the USA [1]. Brain metastases (BM) are a fre- immunotherapies has marked an improvement in pro-
quent neurological complication of lung cancer and gression free survival (PFS) and overall survival (OS) in
have historically been associated with a poorer progno- advanced malignancies including lung cancer. Interest-
sis. Lung cancer is the most common cause of brain ingly, the incidence of brain metastases in lung cancer
metastases as 16.3–19.9% of advanced lung cancer pa- patients has increased over the past decade due to im-
tients develop brain metastases [2, 3]. Approximately proved imaging modalities and prolonged survival from
170,000 cancer patients develop brain metastases each novel approved therapies.
year [4]. A recent population-based study showed that Survival in lung cancer patients after development of
lung cancer has the highest incidence of synchronous brain metastases depends on multiple factors, including
brain metastases which ultimately has negative prognos- local and systemic treatments received. Several groups
tic impact on these patients [5]. have proposed prognostic indices to better predict out-
Lung cancer is broadly divided into two categories: comes and guide treatment strategy [11]. A recently
non-small cell lung cancer (NSCLC) and small cell lung published prognostic index known as Disease-Specific
cancer (SCLC). NSCLC accounts for 85% of cases with Graded Prognostic Assessment (DS-GPA) associated re-
three major pathologic subtypes: adenocarcinoma, sponse to therapy with factors such as age, Karnofsky
squamous cell carcinoma, and large cell carcinoma. performance status (KPS), extracranial metastases, and
SCLC accounts for 15% of cases and is a highly malig- number of brain metastases in patients with lung ade-
nant tumor derived from cells exhibiting neuroendo- nocarcinoma [12]. With the emergence of molecular
crine characteristics [6–8]. Approximately 25–40% of markers in clinical practice, the original DS-GPA was
NSCLC patients develop brain metastases during the updated to include epidermal growth factor receptor
course of their disease [9]. Approximately 10% of pa- (EGFR) and anaplastic lymphoma kinase (ALK) muta-
tients with SCLC have brain metastases present at the tional status [13]. Furthermore, there is evidence to sug-
time of diagnosis [10]. Historically, brain metastases gest that the number of brain metastases does not im-
have been treated with surgical resection, whole brain pact overall survival in NSCLC patients with EGFR and
radiation therapy (WBRT), stereotactic radiosurgery ALK mutations due to the efficacy of targeted therapy in
(SRS), and supportive care based on clinical status and these patients [14].

Pathogenesis of brain metastases


Brain metastases are propagated via hematogenous spread and are most com-
monly seen at the cerebral gray-white junction primarily in the territory of the
middle cerebral artery [15]. The most common site of brain metastases is the
cerebrum (80%), followed by the cerebellum (15%) and the brainstem (5%)
[16, 17]. Brain metastases also frequently involve the terminal “watershed”
areas of arterial circulations (i.e., vascular border zones), especially in the
territory between the middle cerebral artery and posterior cerebral artery [18]. In
1989, Stephen Paget hypothesized the “soil and seed” theory which proposed
that tumor cells (i.e., the “seed”) have the capacity to germinate in distant target
tissues. Metastasis itself is a complicated process, whereby tumor cells must
develop the ability to infiltrate the basement membrane, enter the circulatory or
lymphatic systems, extravasate, and ultimately establish themselves within a
new microenvironment before resuming proliferation [19]. Biological hetero-
geneity is a key trait of tumor cells which enables them to be reprogrammed
into diverse karyotypes with differential growth rates and tumor
Curr Treat Options Neurol (2018) 20:48 Page 3 of 17 48

immunogenicity; such a disorderly metabolic process presents a challenge for


therapeutic intervention [20, 21].
Both extrinsic and intrinsic factors drive cancer cell metabolism and metas-
tasis [22]. The brain’s increased demand and subsequently supply of glucose
provides a nutrient-rich environment for tumor cell growth. Tumor cells un-
dergo metabolic reprogramming in order to meet the higher energetic and
biosynthetic demands placed by rapid proliferation [23]. The blood-brain
barrier (BBB) is comprised of continuous, non-fenestrated endothelium with
tight junctions that prevent entry of circulating macromolecules into the brain
parenchyma [24, 25]. The brain is considered an immunologically privileged
site due to the BBB and the lack of a lymphatic system. The integrity of the BBB
however is altered by brain metastases [26, 27]. The BBB also prevents the entry
of most drugs and microorganisms but does not prevent the invasion of
circulating metastatic cells into the brain parenchyma [28, 29]. Astrocytes are
supportive glial cells involved in cerebral homeostasis against neurotoxicity;
however, activated astrocytes are also found to surround and infiltrate brain
metastases which contribute to the protection of tumor cells from chemother-
apeutic agents [21, 30].

Molecular markers
SCLC is considered more aggressive than NSCLC with approximately 15–20%
of newly diagnosed patients presenting with brain metastases [31]. Genomic
alterations in SCLC include mutation of tumor suppressor genes T53 and RB1,
c-Kit overexpression, vascular endothelial growth factor receptor (VEGFR) and
epidermal growth factor receptor (EGFR) mutations, PTEN mutations, and Myc
overexpression [32]. However, none of the currently available targeted thera-
peutic agents have shown efficacy in SCLC. The most common mutations
associated with NSCLC include Kirsten ras oncogene (KRAS), EGFR, and ALK
[33] while less common mutations include BRAF, RIT1, RET, ROS1, and
NTRK1. Identification of molecular markers in lung cancer has driven the
discovery of targeted therapies [34, 35] with corresponding clinical trials sum-
marized in Table 1.

EGFR
Epidermal growth factor receptor (EGFR) is a transmembrane protein receptor
found on epidermal cells that is activated by epidermal growth factor (EGF).
Upon binding of the ligand, the tyrosine kinase component along the inner
cytoplasmic domain is activated, thereby leading to cell growth and prolifera-
tion [46]. Overexpression of mutated EGFR within its intracellular component
has been observed in 43–89% cases of NSCLC [47]. Activating mutations
within the tyrosine kinase domain of the EGFR are seen in 10% of NSCLC
patients in the USA and 35% in East Asia [48, 49]. EGFR mutations in NSCLC
were first described in 2004 [50]. Studies have shown that NSCLC with EGFR
mutations have increased frequency of brain metastases and yet have better
survival with brain metastases [51]. The most common EGFR mutations are
exon 19 deletions and exon 21 L858R point mutations and comprise 90% of all
kinase-activating mutations leading to hyper-activation of pro-survival
48

Table 1. List of important prospective clinical trials in patients with lung cancer brain metastasis
Clinical trial Number of Characteristics Previous treatment Drug used Biomarker Conclusion
patients
Welsh 40 Multi-institutional phase 4 patients had SRS, Erlotinib + WBRT EGFR mutant: Overall response rate (ORR): 86%
et al. [36] II trial of erlotinib with 21 patients were 9 patients (n = 36), Median survival time:
concurrent WBRT for on cisplatin and 11.8 months (95% CI, 7.4 to
patients with BM docetaxel or 19.1); 17 patients with known
Page 4 of 17

from NSCLC. carboplatin and mutational status, median


paclitaxel, 2 patients survival time 19.1 months for
had pemetrexed. EGFR mutation versus
9.3 months for wild-type EGFR.
Iuchi T 41 Phase II trial in Japanese 10 cases received Gefitinib EGFR mutant: 41 patients Response rate was 87.8%,
et al. [37] patients with gefitinib systemic chemotherapy median PFS was 14.5
alone without radiation prior to diagnosis of months (95% CI,
therapy in EGFR-mutant BM; 10.2–18.3 months), the
lung adenocarcinoma. 31 cases received no median OS was 21.9
prior treatment for BM. months (95% CI,
18.5–30.3 months).
Exon 19 deletion was
associated with better
outcome of patients after
treatment with gefitinib
in both PFS (p = 0.003)
and OS (p = 0.025) as
compared to L858R.
Wu YL 48 Phase II, non-randomized, First-line platinum Erlotinib EGFR mutant: ORR: 58.3%, median OS
et al. [38] open-label, multicenter, doublet chemotherapy 8 patients was 18.9 months (95% CI,
single arm clinical trial. 14.4–23.4). The median
Erlotinib as 2nd-line PFS was 10.1 months
therapy for patients (95% CI, 7.1–12.3) for
with asymptomatic BM intracranial progression
in NSCLC. and 9.7 months (95% CI,
2.5–17.8) for intracranial
and systemic progression.
The median PFS for EGFR
mutation versus EGFR wild
type was 15.2 months
and 4.4 months.
Lee SM 40 Two-stage randomized, No previous treatment Erlotinib + WBRT EGFR mutant: Median neurological PFS of
et al. [39] multicenter, phase II 1 patient out of 35 1.6 months in both arms.
double- blind trial, Median OS of 2.9 and
placebo arm (n = 40), 3.4 months in the placebo
erlotinib with concurrent and erlotinib arms.
WBRT arm (n = 40) No statistically significant
neurological PFS or
OS in patients treated
Curr Treat Options Neurol (2018) 20:48

with concurrent erlotinib


and WBRT followed by
maintenance erlotinib
compared to placebo.
Yang JJ 176 Multicenter, open-label, No previous treatment Icotinib EGFR mutant: Median intracranial PFS was
et al. parallel, randomized 91 patients 10.0 months with icotinib
[40••] controlled trial. versus 4.8 months with
Table 1. (Continued)
Clinical trial Number of Characteristics Previous treatment Drug used Biomarker Conclusion
patients
Icotinib versus WBRT in WBRT plus chemotherapy
patients with EGFR- (HR 0.56, 95% CI,
mutant lung cancer BM. 0.36–0.90; p value
Icotinib group (n = 91), of 0·014). Intracranial
WBRT group (n = 85, only ORR in icotinib and WBRT
73 participated in the plus chemotherapy was
clinical trial) 65% versus 37%
respectively with p value
of 0.001. No difference in
OS between 2 groups.
Mok TS 144 The randomized, international, First-line EGFR TKIs Osimertinib EGFR mutant: The median duration of PFS
et al. open-label, phase III trial. 144 patients was longer in osimertinib
[41••] Patient progressed on group (8.5 months) than
Curr Treat Options Neurol (2018) 20:48

first-line EGFR TKIs were the group receiving


randomly assigned in platinum therapy plus
2:1 ratio to receive pemetrexed (4.2 months);
osimertinib or pemetrexed HR, 0.32; 95% CI,
plus carboplatin/cisplatin. 0.21 to 0.49.
Osimertinib group (n = 93),
Platinum-pemetrexed
group (n = 51)
Solomon BJ 39 The randomized, phase III No previous treatment Crizotinib ALK rearrangement: IC-DCR was significantly higher
et al. clinical trial. Randomly 39 patients with crizotinib (85%)
[42••] assigned in 1:1 ratio, versus platinum-pemetrexed
Crizotinib arm (n = 39) (45%) at 12 weeks; p value
versus platinum-pemetrexed of .001 and at 24 weeks,
arm (n = 40) IC-DCR was 56% versus
25% respectively; with
p value of 0.006. PFS
was significantly longer with
crizotinib (9 months) versus
platinum-pemetrexed
(4 months) in both arms,
HR: 0.40; P value of .001.
Crino` 100 Single-arm, open-label, Prior treatment with at Ceritinib ALK rearrangement: For patients with
et al. multicenter, phase II study least one platinum- 100 patients baseline BM (n = 100) ORR
[43••] in patients with ALK- based chemotherapy was 33.0% (95% CI,
rearranged previously treated regimen and crizotinib, 23.9 to 43.1%), the DCR
with chemotherapy and 72 patients had prior was 74.0% (95% CI,
crizotinib radiotherapy 64.3 to 82.3%), the median
DOR was 9.2 months
(95% CI, 5.5 to 11.1 months),
and the median PFS was
5.4 months (95% CI, 4.7
to 7.2 months. For patients
with active target lesions at
baseline (n = 20), ORR was
45.0% (95% CI, 23.1 to 68.5%)
Peters S 122 The randomized, open-label, Previously untreated Crizotinib, ALK rearrangement: A CNS response rate of 59% (95% CI,
Page 5 of 17 48

et al. phase III trial; crizotinib alectinib 122 46 to 71) was observed in
[44••] 38 out of 64 patients in the
48

Table 1. (Continued)
Clinical trial Number of Characteristics Previous treatment Drug used Biomarker Conclusion
patients
group (n = 58), alectinib alectinib group and a CNS
group (n = 64) response rate of 26% (95% CI,
15 to 39) occurred in 15 out
of 58 patients in the crizotinib
group; 29 patients (45%) in the
Page 6 of 17

alectinib group had a complete


CNS response, as compared
with 5 patients (9%) in the
crizotinib group.
Goldberg 18 A single-institution, two-cohort Surgical resection: 2, Pembrolizumab PD-L1 positive: 18 BM response rate of 33% (95% CI,
et al. phase II trial. Patients with WBRT: 6, SRS: 5 14–59), 4 had complete
[45••] NSCLC, untreated or response, 2 had partial response,
progressive BM 2 had stable disease and 6 had
progression of disease.

CNS central nervous system, BM brain metastases, SRS stereotactic radiosurgery, WBRT whole brain radiation therapy, EGFR epidermal growth factor receptor, TKI tyrosine kinase
inhibitor, ALK anaplastic lymphoma kinase, PD-L1 Program death-1 ligand, ORR overall response rate, CI confidence interval, HR hazard ratio, PFS progression free survival, OS overall
survival, IC-DCR intracranial disease control rate, DCR disease control rate, DOR duration of response
Curr Treat Options Neurol (2018) 20:48
Curr Treat Options Neurol (2018) 20:48 Page 7 of 17 48

signaling pathways [52, 53]. EGFR mutations are commonly found in patients
with lung adenocarcinoma who are light or never-smokers and in Asian pop-
ulations [54, 55]. These mutations have served as novel targets for drug devel-
opment and have led to the development of targeted agents such as gefitinib
and erlotinib. Treatment with gefitinib and erlotinib leads to T790M mutation
in EGFR at the exon 20 which encodes the kinase domain. The second-
generation EGFR tyrosine kinase inhibitors such as afatinib irreversibly block
the EGFR receptor with sustained action and are effective in patients who are
resistant to first-generation EGFR inhibitors. Osimertinib is a next-generation
EGFR tyrosine kinase inhibitor with activity in T790M mutant adenocarcinoma
of the lung [41••]. In fact, osimertinib has shown incremental PFS improve-
ment over standard first-line EGFR tyrosine kinase inhibitors [56••].

ALK
ALK is a tyrosine kinase receptor which belongs to the insulin receptor super-
family and is comprised of an extracellular domain, a hydrophobic stretch
corresponding to a single-pass transmembrane region, and an intracellular kinase
domain [57]. The most common ALK alteration is rearrangement with 3–7% of
NSCLC patients noted to have inversion of chromosome 2p leading to aberrant
fusion of the intracellular signaling portion of ALK to protein encoded by the
echinoderm microtubule associated protein like 4 (EML4 gene) [58]. This ALK
alternation is more commonly found in non-smokers and light smokers (G 10
pack years) with NSCLC [59–61]. Crizotinib is a first-generation ALK inhibitor
with efficacy against ALK-mutated NSCLC brain metastases as compared to
standard chemotherapy but has limited intracranial penetration with subsequent
acquisition of resistance to the drug. The second-generation ALK inhibitors such
as ceritinib, brigatinib, and alectinib have better CNS penetration and are effective
in patients who have developed resistance to crizotinib.

KRAS
An activating mutation in the KRAS gene leads to activation of the RAS GTPase
resulting in a sustained proliferative signal within the cell. Currently, no
targeting agents with activity against KRAS mutations have been approved for
NSCLC. Table 2 provides a summary of known mutations in lung
adenocarcinoma.

PD-1/PD-L1
Program cell death-1 (PD-1) receptor is type 1 transmembrane protein from the
CD28 family and is expressed on activated T cells, B cells, and natural killer (Nk)
cells [62]. PD-L1 binds to the PD-1 receptor and downregulates activation of T
cells, thereby leading to suppression of the immune system. Tumor cells inhibit
host innate immunity by various mechanisms including activation of immune
checkpoint pathways such as PD-1/PD-L1 [63]. Inhibition of PD-1/PD-L1 has
been shown to be effective in many advanced malignancies. Immune check-
point inhibitors such as nivolumab and pembrolizumab block PD-1/PDL-1
pathways, thereby resulting in antitumor activity. Currently, various clinical
trials in phases I and II are ongoing to assess the efficacy of the immune
checkpoint inhibitors in patients with brain metastases due to lung cancer
(NCT02085070, NCT02696993, NCT02978404, NCT02858869).
48 Page 8 of 17 Curr Treat Options Neurol (2018) 20:48

Table 2. Summary of mutations in adenocarcinoma of the lung

Known mutations Incidence rate Important targeted agents


EGFR mutation 15 Erlotinib, gefitinib, afatinib, icotinib, osimertinib
ALK translocation 5 Crizotinib, alectinib, ceritinib, brigatinib, lorlatinib
BRAF mutation 2 Vemurafenib, dabrafenib
RET fusions 1 Cabozantinib, vandetinib
ROS1 rearrangement 2 Crizotinib, loratinib
MET alteration 3 Crizotinib
HER2/MEK overexpression 2 Trastuzumab, lapatinib
KRAS mutation 30 None

*No known mutations in the remaining 40% of adenocarcinoma of the lung

Current therapeutic options in lung cancer brain metastases in


the era of molecular markers
Historically, patients with lung cancer brain metastases have been treated with
WBRT, surgery, stereotactic radiosurgery, and systemic chemotherapy for the
primary tumor. Historically, patients with brain metastases have been excluded
from clinical trials. With the advent of novel targeted therapies, there has been
improved survival benefit and revolutionized the use of medical therapy for
patients with brain metastases.

Targeted therapy for EGFR mutations


Erlotinib and gefitinib are the first-generation tyrosine kinase inhibitors (TKIs)
for EFGR mutations and have demonstrated significant PFS benefit compared
to patients without EGFR mutations [38, 64]. These are small molecules which
penetrate the BBB to some extent in patients with brain metastases. Deng Y et al.
found the BBB permeation rate of erlotinib to be 4.4 ± 3.2% in NSCLC patients
with brain metastasis [65]. Clinical studies have demonstrated that brain me-
tastases affect the permeability of the BBB leading to increase in concentration
of TKIs in cerebrospinal fluid (CSF) with good tumor control. Wang et al. found
significant penetration rate of gefitinib in CSF in patients with brain metastases
as compared to patients without brain metastases (1.5% vs 0.9%, p = 0.010)
[66]. However, we face challenges with the acquisition of resistance to standard
EGFR TKIs; third-generation EFGR TKI osimertinib has shown greater efficacy
than first- and second-generation EGFR TKIs in patients with EGFR-mutant
NSCLC [56••].

First-generation EGFR TKIs


Gefitinib and erlotinib were the first TKIs approved by the Food and Drug
Administration (FDA) for advanced NSCLC with EGFR mutations. Both drugs
Curr Treat Options Neurol (2018) 20:48 Page 9 of 17 48

are orally administered, reversible small molecules which inhibit EGFR auto-
phosphorylation by binding to the ATP binding site of the intracellular tyrosine
kinase domain [67]. In a retrospective study by Hotta K et al., gefitinib dem-
onstrated therapeutic efficacy in 14 NSCLC patients with brain metastases, of
which 1 patient showed complete response (CR), 5 patients showed a partial
response (PR), and 8 patients with stable intracranial disease [68]. In another
study of 23 never-smoker patients with lung adenocarcinoma complicated by
brain metastases treated with either gefitinib or erlotinib monotherapy as first-
line treatment demonstrated significant intracranial response in 17 patients
(73.9%), and median progression free survival (PFS) and overall survival (OS)
were 7.1 and 18.8 months respectively [69]. Porta et al. conducted a retrospec-
tive analysis of 69 lung cancer patients with brain metastases who were treated
with erlotinib (of which 17 patients had EGFR mutations); an intracranial
response rate of 82.4% was noted in EGFR-mutant patients versus those with-
out EGFR mutations. In the same study, OS was 12.9 months in EGFR-mutant
patients versus 3.1 months in EGFR-negative patients [70]. In a prospective
phase II trial of 41 Japanese patients with EGFR-mutant lung adenocarcinoma
and brain metastases, those treated with gefitinib demonstrated favorable
response and exon 19 deletions showed better outcome compared to L858R
[37]. This study reported an 87.8% response rate, median PFS of 14.5 months
(95% CI, 10.2–18.3 months) in all patients, and median OS of 21.9 months
(95% CI, 18.5–30.3 months) in patients with exon 19 deletion. There are
several clinical studies which have shown increased efficacy of EGFR TKIs with
radiation therapy. In a phase II clinical trial by Welsh JW et al., 40 patients with
NSCLC brain metastases were enrolled irrespective of EGFR status; patients
received erlotinib 150 mg orally once daily for 1 week, then concurrently with
WBRT, followed by maintenance dose [36]. The study reported an overall
response rate of 86% in a total of 36 patients; only 17 patients had known EGFR
status out of which 9 patients were EGFR mutated. The median overall survival
of 19.1 months was seen compared to 9.3 months in patients without EGFR
mutations. Subsequently, in a phase II clinical trial by Lee et al., 80 patients were
randomized to erlotinib (100 mg, N = 40) and placebo (N = 40) with concur-
rent WBRT and following WBRT, patients were continued either on placebo or
erlotinib (150 mg) until disease progression [39]. Median OS was 3.4 and
2.9 months in the erlotinib and placebo arms respectively; however, only one
patient out of 35 in erlotinib arms was harboring the EGFR mutation. There was
no significant difference in PFS and OS between two arms.
Pulsatile dosing increases CNS concentration via increased permeation
across the BBB, whereas both pulse dosing and low daily dose may delay
EGFR TKI resistance via the acquisition of the T790 mutation [71]. Icotinib is
another first-generation EGFR TKI which is approved in China for EGFR-mu-
tated NSCLC with brain metastases. In a phase II clinical trial that enrolled 20
Chinese patients with NSCLC brain metastases, they received 125 mg orally
three times/day until tumor progression or unacceptable toxicity, concurrently
with WBRT [72]. Out of 18 patients with known EGFR status, the median
survival time (MST) was 22.0 months for patients with an EGFR mutation
versus 7.5 months for those with wild-type EGFR (p = 0.0001). In another phase
III, multicenter, randomized controlled trial in China, icotinib demonstrated
superior efficacy in patients with multiple brain metastases in EGFR-mutant
NSCLC as compared to WBRT plus chemotherapy [40••]. Median intracranial
48 Page 10 of 17 Curr Treat Options Neurol (2018) 20:48

PFS was 10.0 months in the icotinib-treated cohort versus 4.8 months in the
WBRT plus chemotherapy cohort (HR 0.56; 95% CI, 0.36–0.90; p value of
0·014); however, there was no difference in OS between the two cohorts.
Intracranial ORR was 65% versus 37% in the icotinib and WBRT plus chemo-
therapy cohorts respectively with p value of 0.001.

Second-generation EGFR TKIs


Afatinib, dacomitinib, and neratinib are second-generation EGFR TKIs which
irreversibly block EGFR, HER-2, and ERbB4 receptors. Lung adenocarcinoma
patients treated with first-generation EGFR TKIs generally develop resistance
within 12–16 months of treatment [73, 74]. More than 60% of patients acquire
mutation through the T790M missense mutation encoded by exon 20 of EGFR
[75, 76]. Afatinib has also been approved as first-line treatment of advanced
NSCLC in patients with EGFR mutations. LUX-Lung 3 and 6, randomized phase
III studies of afatinib in metastatic lung adenocarcinoma with EGFR mutation,
included patients with asymptomatic brain metastases [77, 78]. Subgroup
analysis showed intracranial activity in afatinib group with significantly higher
median PFS compared to the conventional chemotherapy group in both the
trials (8.2 vs 5.4 months; HR, 0.50; p = 0.0297) [79••].

Third-generation EGFR TKIs


Osimertinib is the third-generation EGFR TKIs that was approved by the FDA;
soon after, preliminary data showed efficacy in previously treated patients with
EGFR-mutated advanced NSCLC and brain metastases [80–82]. Osimertinib is
a potent, oral, irreversible inhibitor of activating EGFR mutations and T790M-
resistant mutations. AURA 3, a randomized phase III clinical study of patients
with T790M-positive advanced NSCLC who had disease progression with first-
line EGFR TKI therapy, showed significant intracranial activity with osimertinib
[41]. In 144 patients with brain metastases, the patients who received
osimertinib had longer median PFS as compared to those who received plati-
num and pemetrexed (8.5 months vs 4.2 months; hazard ratio, 0.32; 95% CI,
0.21 to 0.49). As well, the study showed that patients in the osimertinib-treated
cohort had lower adverse events (23%) as compared to the platinum plus
pemetrexed cohort (47%). In FLAURA, a phase III trial study of osimertinib
versus first-generation TKIs, 53 patients with CNS metastases were treated in the
osimertinib arm compared to 63 CNS metastases in the standard tyrosine
kinase arm. Overall, 17 patients (6%) in the osimertinib-treated cohort had
CNS progression versus 42 patients (15%) in the standard treatment cohort
[56••]. In subgroup analysis of this clinical trial, osimertinib showed better
CNS PFS as compared to erlotinib or gefitinib (HR 0.48; 95% CI, 0.26–0.86)
[83••]. Osimertinib has consistently shown better intracranial activity com-
pared to first-generation EGFR TKIs; however, studies focusing only on brain
metastases are ongoing [84].

ALK inhibitor in lung cancer brain metastases


The ALK gene is located on the short arm of chromosome 2, and it encodes the
ALK receptor tyrosine kinase. ALK gene rearrangement occurs in 3–7% of
NSCLC patients while brain metastases have been reported in about 24% in
ALK gene fusion NSCLC patients at time of diagnosis [85, 86]. Crizotinib is a
Curr Treat Options Neurol (2018) 20:48 Page 11 of 17 48

first-generation ALK inhibitor approved for ALK-mutated NSCLC, and it has


also shown activity against ROS1 rearrangement and MET alteration [87]. In the
randomized phase III prospective clinical trial PROFILE 1014 of ALK-positive
advanced NSCLC patients, crizotinib as first-line treatment demonstrated su-
perior intracranial activity as compared to standard chemotherapy in patients
with treated brain metastases [42••]. Seventy-nine patients with brain metas-
tases were enrolled in this study that showed intracranial activity with crizo-
tinib. Intracranial disease control rate (IC-DCR) was significantly higher (85%)
with crizotinib as compared to 45% with chemotherapy at 12 weeks (p
G 0.001), and IC-DCR was 56% with crizotinib versus 25% with chemotherapy
at 24 weeks (p = 0.006). The median PFS was significantly better among patients
treated with crizotinib (9 months) versus standard chemotherapy (4 months)
with hazard ratio of 0.40 and p G 0.001. The retrospective pooled analysis was
done in patients with brain metastases from the phase II PROFILE 1005 and
phase III PROFILE 1007 trials compared crizotinib to second-line chemother-
apy [88]. Of the 275 patients who had asymptomatic brain metastases, 166 had
undergone prior treatment. In these 166 patients, the IC-DCR was 62% and the
median intracranial TTP was 13.2 months. Among 109 patients with untreated
asymptomatic brain metastases, the IC-DCR was 56% and the median intra-
cranial time to progression (TTP) was 7 months.
Ceritinib and alectinib are second-generation ALK inhibitors with better
CNS penetration with activity in patients resistant to crizotinib [89, 90]. The
phase I multicenter trial of ceritinib demonstrated preliminary evidence of
intracranial activity [91]. In 94 patients with confirmed brain metastases, ICR of
79% was reported in19 patients who had never received any prior ALK inhib-
itors and IC-DCR was 65% in 75 patients who had failed prior ALK inhibitor.
Six of 11 patients with measurable brain lesion achieved a partial intracranial
response. In the ASCEND-2 phase II trial, 100 patients had brain metastases at
enrollment with only 20 of these having measurable target lesions. Among
these 20 patients, the intracranial response rate was 45.0% (95% CI, 23.1 to
68.5%) while the IC-DCR was 80% (n = 20, 95% CI, 56.3–94.3) [43••]. In-
creasingly, clinicians are shifting from crizotinib to ceritinib in patients with
brain metastases.
The AF-002JG phase I/II study of alectinib in patients with crizotinib-
resistant ALK-rearranged NSCLC demonstrated promising intracranial activity
[92]. Of 21 patients with CNS metastases at baseline, 11 had an objective
response, 6 had a complete response, 5 had a partial response, 8 had stable
disease, and the remaining 2 had progressive disease. The J-ALEX phase III trial
showed an objective response rate of 85% in the alectinib group versus 70% in
the crizotinib group; in patients with brain metastases, the hazard ratio for
alectinib versus crizotinib was 0.08 (95% CI, 0.01–0.61) [93]. Furthermore, 14
patients with asymptomatic brain metastases in the alectinib arm showed IC-
DCR of 92.9% and only one of the patients had progressed at the time of data
cutoff. A multicenter, global, ALEX phase III randomized trial of alectinib versus
crizotinib in untreated ALK-positive NSCLC showed efficacy of alectinib in
patients with and without CNS lesions at baseline; 64 patients had CNS lesions
at baseline in alectinib arm and 58 patients in crizotinib arm [44••]. CNS
response was observed in 38 patients in alectinib group compared to 15
patients in crizotinib group with a complete response rate of 45% and 9% in
patients treated with alectinib and crizotinib respectively. This trial also
48 Page 12 of 17 Curr Treat Options Neurol (2018) 20:48

demonstrated that the time to CNS progression was significantly longer with
alectinib than with crizotinib (cause-specific hazard ratio, 0.16, 95% CI, 0.10 to
0.28; the rate of CNS progression was 12% with alectinib and 45% with
crizotinib). Another ALK inhibitor, brigatinib, has demonstrated encouraging
results in CNS metastases. In a multicenter, open-label phase I/II study
(NCT01449461), 6 out of 12 patients with lesions ≥ 10 mm had ≥ 30%
decrease in sum of longest diameters of target lesions and 8 out of 26 patients
with non-measurable lesions had disappearance of all lesions. Median intra-
cranial PFS for these patients was about 97 weeks [94]. Lorlatinib is a third-
generation ALK inhibitor with selective CNS penetration and also has activity
against ROS1; it has been developed as an agent to act against most of the
resistance mutations in patients treated with ALK-targeting agents. Preliminary
results from the phase I of the ongoing phase I/II study NCT01970865 dem-
onstrated an objective intracranial response rate (ICRR) of 44% in targetable
and non-targetable lesions in 16 patients and 60% in the targetable lesion in 12
patients respectively [95].

Immunotherapies
Immune checkpoint inhibitors have shown promising results in many ad-
vanced malignancies, but patients with brain metastases have been historically
excluded from such trials. Nivolumab and pembrolizumab are monoclonal
antibodies against program death-receptor-1 (PD-1) and work to block cyto-
toxic T lymphocyte tolerance. Nivolumab and pembrolizumab have been FDA-
approved for unresectable metastatic melanoma, metastatic NSCLC, renal cell
carcinoma, classical Hodgkin lymphoma, hepatocellular carcinoma, metastatic
colorectal cancer, urothelial carcinoma, and squamous cell carcinoma of the
head and neck. A number of case reports and retrospective studies reported
efficacy of immunotherapies in patients with brain metastases from lung cancer
[96–99]. The only prospective study was a single-institution, phase II trial of
pembrolizumab in which 18 patients with NSCLC brain metastases demon-
strated response rates of 33% (95% CI, 14–59), four had complete response,
two had partial response, 2 had stable disease, and 6 had progression of disease
[45••]. Furthermore, with growing evidence of durable response in various
advanced malignancies, there are ongoing clinical trials such as NCT01454102
(check mate-012), NCT02085070, NCT02681549, and NCT02858869 which
are specifically assessing the efficacy of immune checkpoint inhibitors in pa-
tients with brain metastases in lung cancer and melanoma.

Conclusion
Over the past decade, several actionable mutations that are present in NSCLC
have led to the development of promising novel targeted therapies with im-
proved CNS penetration. Newer targeted therapies against EGFR mutations and
ALK translocations have shown encouraging results with impressive response
rates. Immunotherapy has shown initial promising intracranial activity that is
comparable to the response rates seen extracranially. The future trials will need
to focus on the interactions, toxicity and efficacy of these targeted agents, and
immunotherapy with different forms of radiation. The management of brain
Curr Treat Options Neurol (2018) 20:48 Page 13 of 17 48

metastases should involve a multidisciplinary team of neurosurgeons, radiation


oncologists, and medical and neuro-oncologists to tailor treatment to each
individual patient.

Compliance with Ethical Standards

Conflict of Interest
Dr. Ahluwalia reports grants and personal fees from Elekta, grants and personal fees from Incyte, grants and personal
fees from BMS, grants and personal fees from Astrazeneca, grants from Tracon, grants from Novartis, grants and
personal fees from Novocure, personal fees from Monteris Medical, personal fees from Caris Life Sciences, personal
fees from MRI Solutions, grants and personal fees from Abbvie, personal fees from CBT Pharmaceuticals, personal
fees from Flatiron, personal fees from Varian, personal fees from VBI vaccines, and personal fees and others from
MImivax, outside the submitted work.
Dr. Bicky Thapa, Dr. Kunal Desai, Mr. Adam Lauko, and Dr. Vyshak Alva Venur each declare that they have no
potential conflict of interest.

Human and Animal Rights and Informed Consent


This article does not contain any studies with human or animal subjects performed by any of the authors.

References and Recommended Reading


Papers of particular interest, published recently, have been
highlighted as:
•• Of major importance
1. American Cancer Society, Cancer facts & figures 2017. 7. Kohler B, Ward E, McCarthy B, et al. Annual report to
2. Schouten LJ, Rutten J, Huveneers HA, Twijnstra A. In- the nation on the status of cancer, 1975–2007, featur-
cidence of brain metastases in a cohort of patients with ing tumors of the brain and other nervous system. J
carcinoma of the breast, colon, kidney, and lung and Natl Cancer Inst. 2011;103:1–23.
melanoma. Cancer. 2002;94(10):2698–705. 8. Herbst RS, Heymach JV, Lippman SM. Lung cancer. N
3. Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Engl J Med. 2008;359(13):1367–80.
Lai P, Sawaya RE. Incidence proportions of brain me- 9. Sørense J, Hansen H, Hansen M, Dombernowsky P.
tastases in patients diagnosed (1973 to 2001) in the Brain metastases in adenocarcinoma of the lung: fre-
metropolitan Detroit cancer surveillance system. J Clin quency, risk groups, and prognosis. J Clin Oncol.
Oncol. 2004;22(14):2865–72. https://doi.org/10. 1988;6:1474–80.
1200/jco.2004.12.149. 10. Castrucci W, Knisely J. An update on the treatment of
4. Davey P: Brain metastases: treatment options to im- CNS metastases in small cell lung cancer. Cancer J.
prove outcomes. CNS Drugs 16;2002;325–338. 2008;14:138–46.
5. Kromer, Courtney, Jordan Xu, Quinn T. Ostrom, Haley 11. Venur VA, Ahluwalia MS. Prognostic scores for brain
Gittleman, Carol Kruchko, Raymond Sawaya, and Jill metastasis patients: use in clinical practice and trial
S. Barnholtz-Sloan. Estimating the annual frequency of design. Chin Clin Oncol. 2015;4(2):18. https://doi.
synchronous brain metastasis in the United States org/10.3978/j.issn.2304-3865.2015.06.01.
2010–2013: a population-based study. J Neuro-Oncol 12. SperdutoPW, Chao ST, Sneed PK, LuoX, Suh J, Roberge
134(1);2017:55–64. https://doi.org/10.1007/s11060- D, et al. Diagnosis-specific prognostic factors, indexes,
017-2516-7. and treatment outcomes for patients with newly diag-
6. Howlader N, Noone AM, Krapcho M, et al., editors. nosed brain metastases: a multi-institutional analysis
SEER Cancer Statistics Review, 1975–2008. Bethesda of 4,259 patients. Int J Radiat Oncol Biol Phys.
(MD): National Cancer Institute; 2010. Available at: 2010;77(3):655–61. https://doi.org/10.1016/j.ijrobp.
http://seer.cancer.gov/csr/1975_2008/, based on No- 2009.08.025.
vember 2010 SEER data submission, posted to the 13. Sperduto PW, et al. Estimating survival in patients with
SEER web site, 2011. lung cancer and brain metastases: an update of the
48 Page 14 of 17 Curr Treat Options Neurol (2018) 20:48

graded prognostic assessment for lung cancer using sequestering intracellular calcium through gap junc-
molecular markers (Lung-molGPA). JAMA Oncol. tion channels. Neoplasia. 2010;12:748–54.
2017;3(6):827–31. https://doi.org/10.1001/ 31. Lekic M, Kovac V, Triller N, Knez L, Sadikov A, Cufer T.
jamaoncol.2016.3834. Outcome of small cell lung cancer (SCLC) patients
14. Balasubramanian SK, Alva Venur V, Chao ST, et al. with brain metastases in a routine clinical setting.
Impact of EGFR and ALK mutation on the outcomes of Radiol Oncol. 2012;46:54–9.
non-small cell lung cancer (NSCLC) patients with 32. Sabari JK, Paik PK. Relevance of genetic alterations in
brain metastases. J Clin Oncol. squamous and small cell lung cancer. Ann Transl Med.
2016;34(15_suppl):2005. https://doi.org/10.1200/ 2017;5(18):373. https://doi.org/10.21037/atm.2017.
JCO.2016.34.15_suppl.2005. 06.72.
15. Nonaka H, Akima M, Hatori T, et al. The microvascular of 33. Li T, Kung HJ, Mack PC, Gandara DR. Genotyping and
the cerebral white matter: arteries of the subcortical white genomic profiling of non-small-cell lung cancer: ap-
matter. J Neuropathol Exp Neurol. 2003;62:154–61. plications for current and future therapies. J Clin
16. Porter AT, David M. Palliative care for bone, spinal Oncol. 2013;31(8):1039–49. https://doi.org/10.1200/
cord, brain and liver metastases. In: Gunderson LL, jco.2012.45.3753.
Tepper JE, editors. Clinical radiation oncology. Phila- 34. Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate
delphia: Elsevier; 2007. p. 437–55. AJ, Wistuba II, et al. Using multiplexed assays of on-
17. Narayana A, Liebel SA. Primary and metastatic brain cogenic drivers in lung cancers to select targeted drugs.
tumors. In: Liebel SA, Phillips TL, editors. Textbook of JAMA. (2014);311:1998–2006.
radiation oncology. Philadelphia: Elsevier; 2004. p. 35. Shonka N, Venur VA, Ahluwalia MS. Targeted treat-
463–95. ment of brain metastases. Curr Neurol Neurosci Rep.
18. Hwang TL, Close TP, Grego JM, et al. Predilection of 2017;17(4) https://doi.org/10.1007/s11910-017-
brain metastasis in gray and white matter junction and 0741-2.
vascular border zones. Cancer. 1996;77:1551–5. 36. Welsh JW, Komaki R, Amini A, et al. Phase II trial of
19. Valastyan S, Weinberg RA. Tumor metastasis: molecu- erlotinib plus concurrent whole-brain radiation thera-
lar insights and evolving paradigms. Cell. py for patients with brain metastases from non-small-
2011;147(2):275–92. cell lung cancer. J Clin Oncol. 2013;31:895–902.
20. Ebben D, Johnathan, You M. Brain metastasis in lung 37. Iuchi T, Shingyoji M, Sakaida T, Hatano K, Nagano O,
cancer: building a molecular and systems-level under- Itakura M, et al. Phase II trial of gefitinib alone without
standing to improve outcomes. Int J Biochem Cell Biol. radiaton therapy for Japanese patients with brain me-
2016;78:P14. https://doi.org/10.1016/j.biocel.2016. tastases from EGFR-mutant lung adenocarcinoma. Lung
07.025. cancer (Amsterdam, Netherlands). 2013;82(2):282–7.
21. Fidler IJ. The biology of brain metastasis challenges for https://doi.org/10.1016/j.lungcan.2013.08.016.
therapy. Cancer J. 2015;21(4):284–93. 38. Wu YL, Zhou C, Cheng Y, et al. Erlotinib as second-line
22. Ciminera AK, Jandial R, Termini J. Metabolic advan- treatment in patients with advanced non-small-cell
tages and vulnerabilities in brain metastases. Clin Exp lung cancer and asymptomatic brain metastases: a
Metastasis. 2017;34(6–7):401–10. https://doi.org/10. phase II study (CTONG-0803). Ann Oncol.
1007/s10585-017-9864-8. 2013;24(4):993–9. https://doi.org/10.1093/annonc/
23. Boroughs LK, De Berardinis RJ. Metabolic pathways mds529.
promoting cancer cell survival and growth. Nat Cell 39. Lee SM, Lewanski CR, Counsell N, et al. Randomized
Biol. 2015;17:351–9. trial of erlotinib plus whole-brain radiotherapy for
24. Johansson BB. The physiology of the blood-brain bar- NSCLC patients with multiple brain metastases. J Natl
rier. Adv Exp Med Biol. 1990;274:25–39. Cancer Inst. 2014;106:dju151.
25. Gregoire N. The blood-brain barrier. J Neuroradiol. 40.•• Yang JJ, Zhou C, Huang Y, et al. Icotinib versus whole-
1989;16:238–50. brain irradiation in patients with EGFR-mutant non-
26. Felgenhauer K. The blood-brain barrier redefined. J small-cell lung cancer and multiple brain metastases
Neurol. 1986;233:193–4. (BRAIN): a multicentre, phase 3, open-label, parallel,
randomised controlled trial. Lancet Respir Med.
27. Shapiro WR, Shapiro JR. Principles of brain tumor
2017;5(9):707–16. https://doi.org/10.1016/S2213-
chemotherapy. Semin Oncol. 1986;13:56–69.
2600(17)30262-X.
28. Iannotti F, Fleschi C, Alfano B, et al. Simplified, non- In this phase III, multicenter clinical trial in China, icotinib
invasive PET measurement of blood brain barrier per- showed longer intracranial PFS as compared to WBRT plus
meability. J Comput Assist Tomogr. 1987;11:390–7. chemotherapy in patients with EGFR-mutant NSCLC.
29. Frong D, Israel O, Kohn S, et al. The blood-tissue 41.•• Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR,
barrier of human brain tumors: correlation of scinti- Ramalingam SS, et al. Osimertinib or platinum
graphic and ultrastructural findings (concise commu- pemetrexed in EGFR T790M-positive lung cancer. N
nication). J Nucl Med. 1984;25:461–5. Engl J Med. 2016; https://doi.org/10.1056/
30. Lin Q, Balasubramanian K, Fan D, et al. Reactive as- NEJMoa1612674.
trocytes protect melanoma cells from chemotherapy by In this phase III clinical trial, osimertinib demonstrated better
Curr Treat Options Neurol (2018) 20:48 Page 15 of 17 48

efficacy as compared to standard chemotherapy in patients 50. Pao W, Miller V, Zakowski M, et al. EGF receptor gene
with EGFR-mutant brain metastases in non-small cell lung mutations are common in lung cancers from “never
cancer. smokers” and are associated with sensitivity of tumors
42.•• Solomon BJ, Cappuzzo F, Felip E, et al. Intracranial to gefitinib and erlotinib. Proc Natl Acad Sci U S A.
efficacy of crizotinib versus chemotherapy in patients 2004;101(36):13306–11.
with advanced ALK-positive non-small-cell lung can- 51. Eichler AF, Kahle KT, Wang DL, et al. EGFR mutation
cer: results from PROFILE 1014. J Clin Oncol. status and survival after diagnosis of brain metastasis in
2016;34(24):2858–65. nonsmall cell lung cancer. Neuro Oncol.
In this phase III PROFILE 1014 study, crizotinib showed better 2010;12:1193–9.
intracranial activity in patients with ALK-positive NSCLC brain 52. Ladanyi M, Pao W. Lung adenocarcinoma: guiding
metastases as compared to standard chemotherapy. EGFR-targeted therapy and beyond. Mod Pathol.
43.•• Crino L, Ahn MJ, De Marinis F, Groen HJ, Wakelee H, 2008;21(Suppl 2):S16–22. https://doi.org/10.1038/
Hida T, et al. Multicenter phase II study of whole-body modpathol.3801018.
and intracranial activity with ceritinib in patients with 53. Sordella, et al. Gefitinib-sensitizing EGFR mutations in
ALK-rearranged non-small-cell lung cancer previously lung cancer activate anti-apoptotic pathways. Science.
treated with chemotherapy and crizotinib: results from 2004;305(5687):1163–7.
ASCEND-2. J Clin Oncol. 2016;34(24):2866–73. 54. Pao W, Miller V, Zakowski M, Doherty J, Politi K,
https://doi.org/10.1200/JCO.2015.65.5936. Sarkaria I, et al. EGF receptor gene mutations are com-
This phase II study included patients ALK-positive NSCLC, mon in lung cancers from “never smokers” and are
previously treated with chemotherapy and crizotinib. Ceritinib associated with sensitivity of tumors to gefitinib and
demonstrated durable intracranial response in patients with erlotinib. Proc Natl Acad Sci USA. 2004;101:13306–11.
brain metastases. 55. Rosell R, Moran T, Queralt C, Porta R, Cardenal F,
44.•• Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Camps C, et al. Screening for epidermal growth factor
crizotinib in untreated ALK-positive non-small-cell receptor mutations in lung cancer. N Engl J Med.
lung cancer. N Engl J Med. 2017;377(9):829–38. 2009;361:958–67.
https://doi.org/10.1056/NEJMoa1704795. 56.•• Soria J-C, Ohe Y, Vansteenkiste J, et al. Osimertinib in
In this phase III clinical trial, alectinib demonstrated longer PFS untreated EGFR-mutated advanced non-small-cell lung
as compared to crizotinib in patients with ALK positive NSCLC cancer. N Engl J Med. 2017;NEJMoa1713137. https://
brain metastases. doi.org/10.1056/NEJMoa1713137.
45.•• Goldberg SB, Gettinger SN, Mahajan A, et al. The phase III clinical trial included patients with previously
Pembrolizumab for patients with melanoma or non- untreated, EGFR-mutant advanced NSCLC and compared the
small-cell lung cancer and untreated brain metastases: efficacy of osimertinib with standard EGFR TKIs. Osimertinib
early analysis of a non-randomised, open-label, phase demonstrated better efficacy as compared to standard EGFR
2 trial. Lancet Oncol. 2016;17(7):976–83. https://doi. TKIs.
org/10.1016/S1470-2045(16)30053-5. 57. Nakamichi S, Seike M, Miyanaga A, et al. RT-PCR for
The only prospective trial in patients with lung cancer brain Detecting ALK translocations in cytology samples from
metastases assessing efficacy of immunotherapy. lung cancer patients. Anticancer Res.
Pembrolizumab demonstrated intracranial activity in patients 2017;37(6):3295–9. https://doi.org/10.21873/
with lung cancer brain metastases. anticanres.11696.
46. Sharma SV, Bell DW, Settleman J, Haber DA. Epider- 58. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y,
mal growth factor receptor mutations in lung cancer. Ishikawa S, et al. Identification of the transforming
Nat Rev Cancer. 2007;7(3):169–81. EML4-ALK fusion gene in non-small-cell lung cancer.
47. Gupta R, Dastane AM, Forozan F, Riley-Portuguez A, Nature. 2007;448:561–6.
Chung F, Lopategui J, et al. Evaluation of EGFR ab- 59. Takeuchi K, Choi YL, Soda M, Inamura K, Togashi Y,
normalities in patients with pulmonary adenocarcino- Hatano S, et al. Multiplex reverse transcription-PCR
ma: the need to test neoplasms with more than one screening for EML4-ALK fusion transcripts. Clin Cancer
method. Mod Pathol. 2009;22:128–33. Res. 2008;14:6618–24.
48. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, 60. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR,
Okimoto RA, Brannigan BW, et al. Activating muta- Costa DB, Heist RS, et al. Clinical features and outcome
tions in the epidermal growth factor receptor underly- of patients with non-small-cell lung cancer who harbor
ing responsiveness of non-small cell lung cancer to EML4-ALK. J Clin Oncol. 2009;27:4247–53.
gefitinib. N Engl J Med. 2004;350(21):2129–39. 61. Wong DW-S, Leung EL-H, So KK-T, Tam IY-S, Sihoe
https://doi.org/10.1056/NEJMoa040938. AD-L, Cheng L-C, et al. The EML4-ALK fusion gene is
49. Pao W, Miller VA. Epidermal growth factor receptor involved in various histologic types of lung cancers
mutations, small-molecule kinase inhibitors, and non- from nonsmokers with wild-type EGFR and KRAS.
small-cell lung cancer: current knowledge and future Cancer. 2009;115:1723–33.
directions. J Clin Oncol. 2005;23(11):2556–68. 62. Agata Y, Kawasaki A, Nishimura H, et al. Expression of
https://doi.org/10.1200/jco.2005.07.799. the PD-1 antigen on the surface of stimulated mouse T
and B lymphocytes. Int Immunol. 1996;8(5):765–72.
48 Page 16 of 17 Curr Treat Options Neurol (2018) 20:48

63. Schalper KA, Venur VA, Velcheti V. Programmed death-1/ associated with a second mutation in the EGFR kinase
programmed death-1 ligand axis as a therapeutic target in domain. PLoS Med. 2005;2:e73.
oncology: current insights. J Receptor Ligand Channel 76. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mu-
Res. 2015;8:1–7. https://doi.org/10.2147/JRLCR.S39986. tation and resistance of non-small-cell lung cancer to
64. Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus gefitinib. N Engl J Med. 2005;352:786–92.
cisplatin plus docetaxel in patients with non-small-cell 77. Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V,
lung cancer harbouring mutations of the epidermal Mok T, et al. Phase III study of afatinib or cisplatin plus
growth factor receptor (WJTOG3405): an open label, pemetrexed in patients with metastatic lung adenocar-
randomised phase 3 trial. Lancet Oncol. 2010;11:121–8. cinoma with EGFR mutations. J Clin Oncol.
65. Deng Y, Feng W, Wu J, et al. The concentration of 2013;31(27):3327–34. https://doi.org/10.1200/jco.
erlotinib in the cerebrospinal fluid of patients with 2012.44.2806.
brain metastasis from non-small-cell lung cancer. Mol 78. Wu Y-L, Zhou C, Hu C-P, Feng J, Lu S, Huang Y, et al.
Clin Oncol. 2014;2:116e120. Afatinib versus cisplatin plus gemcitabine for first-line
66. Wang M, Jing Z, Minjiang C. Cerebral penetration of treatment of Asian patients with advanced non-small-
gefitinib in patients with lung adenocarcinoma. J Clin cell lung cancer harbouring EGFR mutations (LUX-
Oncol. 2011;29(15_suppl):7608. https://doi.org/10. lung 6): an open-label, randomised phase 3 trial. Lan-
1200/jco.2011.29.15_suppl.7608. cet Oncol. 2014;15(2):213–22. https://doi.org/10.
67. Jeffrey V. Brower & H. Ian Robins (2016) Erlotinib for 1016/S1470-2045(13)70604-1.
the treatment of brain metastases in non-small cell 79.•• Schuler M, Wu Y-L, Hirsh V, et al. First-line afatinib
lung cancer, Expert Opinion on Pharmacotherapy versus chemotherapy in patients with non-small cell
17(7):1013–1021,DOI: https://doi.org/10.1517/ lung cancer and common epidermal growth factor re-
14656566.2016.1165206. ceptor gene mutations and brain metastases. J Thorac
68. Hotta K, Kiura K, Ueoka H, Tabata M, Fujiwara K, Oncol. 2016;11(3):380–90. https://doi.org/10.1016/j.
Kozuki T,et al. Effect of gefitinib (‘Iressa’, ZD1839) on jtho.2015.11.014.
brain metastases in patients with advanced non-small- The subgroup analyses for both LUX-Lung 3 and LUX-lung 6
cell lung cancer. Lung cancer (Amsterdam, Nether- revealed improved PFS with afatinib versus chemotherapy in
lands). 2004;46(2):255–261. doi:https://doi.org/10. patients with EGFR-mutant NSCLC and asymptomatic brain
1016/j.lungcan.2004.04.036. metastases.
69. Kim JE, Lee DH, Choi Y, et al. Epidermal growth factor 80. Ballard P, Yates JW, Yang Z, Kim DW, Yang JC, Cantarini
receptor tyrosine kinase inhibitors as a first-line thera- M, et al. Preclinical comparison of osimertinib with
py for neversmokers with adenocarcinoma of the lung other EGFRTKIs in EGFR-mutant NSCLC brain metas-
having asymptomatic synchronous brain metastasis. tases models, and early evidence of clinical brain me-
Lung Cancer. 2009;65:351e354. tastases activity. Clin Cancer Res. 2016; https://doi.org/
70. Porta R, Sanchez-Torres JM, Paz-Ares L, Massuti B, 10.1158/1078-0432.ccr-16-0399.
Reguart N, Mayo C, et al. Brain metastases from lung 81. Cross DAE, Ashton SE, Ghiorghiu S, et al. AZD9291, an
cancer responding to erlotinib: the importance of irreversible EGFR TKI, overcomes T790M-mediated re-
EGFR mutation. Eur Respir J. 2011;37(3):624–31. sistance to EGFR inhibitors in lung cancer. Cancer
https://doi.org/10.1183/09031936.00195609. Discov. 2014;4(9):1046–61. https://doi.org/10.1158/
71. Grommes C, Oxnard GR, Kris MG, Miller VA, Pao W, 2159-8290.CD-14-0337.
Holodny AI, et al. “Pulsatile” high-dose weekly erloti- 82. Jänne PA, Yang JC-H, Kim D-W, et al. AZD9291 in
nib for CNS metastases from EGFR mutant non-small EGFR inhibitor-resistant no-small-cell lung cancer. N
cell lung cancer. Neuro-Oncology. 2011;13(12):1364– Engl J Med. 2015;372(18):1689–99. https://doi.org/
9. https://doi.org/10.1093/neuonc/nor121. 10.1056/NEJMoa1411817.
72. Fan Y, Huang Z, Fang L, Miao L, Gong L, Yu H, et al. A 83.•• Vansteenkiste J, Reungwetwattana T, Nakagawa K, et al.
phase II study of icotinib and whole-brain radiothera- LBA5CNS response to osimertinib vs standard of care
py in Chinese patients with brain metastases from (SoC) EGFR-TKI as first-line therapy in patients (pts)
nonsmall cell lung cancer. Cancer Chemother with EGFR-TKI sensitising mutation (EGFRm)-positive
Pharmacol. 2015;76(3):517–23. https://doi.org/10. advanced non-small cell lung cancer (NSCLC): data
1007/s00280-015-2760-5. from the FLAURA study. Ann Oncol.
73. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or 2017;28(suppl_10):mdx729.007. https://doi.org/10.
chemotherapy for non-small-cell lung cancer with 1093/annonc/mdx729.007.
mutated EGFR. N Engl J Med. 2010;362:2380–8. The phase III FLAURA study included patients with previously
74. Janne PA, Wang X, Socinski MA, et al. Randomized untreated EGFR-mutant NSCLC and compared osimertinib
phase II trial of erlotinib alone or with carboplatin and with standard EGFR TKIs (gefitinib or erlotinib). A subgroup
paclitaxel in patients who were never or light former analysis showed better CNS PFS with osimertinib as compared
smokers with advanced lung adenocarcinoma: CALGB to standard EGFR TKIs.
30406 trial. J Clin Oncol. 84. Ahluwalia MS, Becker K, Levy BP. Epidermal growth
75. Pao W, Miller VA, Politi KA, et al. Acquired resistance of factor receptor tyrosine kinase inhibitors for central
lung adenocarcinomas to gefitinib or erlotinib is nervous system metastases from non-small cell lung
Curr Treat Options Neurol (2018) 20:48 Page 17 of 17 48

cancer. Oncology. 2018; https://doi.org/10.1634/ results from the dose-finding portion of a phase 1/2
theoncologist.2017-0572. study. Lancet Oncol. 2014;15:1119–28.
85. Bohn JP, Pall G, Stockhammer G, et al. Targeted ther- 93. Hida T, Nokihara H, Kondo M, et al. Alectinib versus
apies for the treatment of brain metastases in solid crizotinib in patients with ALK-positive non-small-cell
tumors. Target Oncol. 2016. lung cancer (J-ALEX): an open-label, randomised
86. Rangachari D, Yamaguchi N, VanderLaan PA, Folch E, phase 3 trial. Lancet. 2017;390(10089):29–39. https://
Mahadevan A, Floyd SR, et al. Brain metastases in doi.org/10.1016/S0140-6736(17)30565-2.
patients with EGFR-mutated or ALK-rearranged non- 94. Camidge DR, Bazhenova L, Salgia R, et al. Safety and
small-cell lung cancers. Lung Cancer. 2015;88(1):108– efficacy of brigatinib (AP26113) in advanced malig-
11. https://doi.org/10.1016/j.lungcan.2015.01.020. nancies, including ALK1 non-small cell lung cancer
87. The National Comprehensive Cancer Network (NSCLC). J Clin Oncol. 33;2015(suppl; abstr 8062).
(NCCN): Clinical practice guidelines in oncology: non- 95. Solomon B, Bauer T, Felip E, Besse B, James L, Clancy J.
small cell lung cancer. v4.2016. Safety and efficacy of lorlatinib (PF-06463922) from
88. Costa DB, Shaw AT, Ou SHI, et al. Clinical experience the dose-escalation component of a study in patients
with crizotinib in patients with advanced ALK- with advanced ALK+ or ROS1+ non-small cell lung
rearranged non-small-cell lung cancer and brain me- cancer (NSCLC). Abstract 9009 Presented at ASCO
tastases. J Clin Oncol. 2015;33:1881–8. Annual Meeting Proceedings in Chicago (2016).
96. Dudnik E, et al. Intracranial response to nivolumab in
89. Wong A. The emerging role of targeted therapy and
NSCLC patients with untreated or progressing CNS
immunotherapy in the management of brain metasta-
metastases. Lung Cancer (Amsterdam, Netherlands).
ses in non-small cell lung cancer. Front Oncol.
2016;98:114–7. https://doi.org/10.1016/j.lungcan.
2017;7:33. https://doi.org/10.3389/fonc.2017.00033.
2016.05.031.
90 Venur VA, Ahluwalia MS. Targeted therapy in brain 97. Watanabe H, Kubo T, Ninomiya T, et al. The effect of
metastases: ready for primetime? American Society of nivolumab treatment for central nervous system me-
Clinical Oncology educational book. American Society tastases in nonsmall cell lung cancer. J Clin Oncol.
of Clinical Oncology Meeting. 2016;35:e123–30. 2017;35(15).
https://doi.org/10.14694/edbk_100006. 98. Gauvain C, Vauléon E, Chouaid C, et al. Intracerebral
91. Shaw A, Mehra R, Tan DSW, et al. BM-32 Ceritinib efficacy and tolerance of nivolumab in non-small-cell
(LDK378) for treatment of patients with ALK- lung cancer patients with brain metastases. Lung Can-
rearranged (ALK+) non-small cell lung cancer (NSCLC) cer. 2018;116. https://doi.org/10.1016/j.lungcan.
and brain metastases (BM) in the ASCEND-1 trial. 2017.12.008
Neuro Oncol. 2014;16:v39. 99. Thapa B, Ahluwalia M, et al. CMET-01. Efficacy and
92. Gadgeel SM, Gandhi L, Riely GJ, et al. Safety and outcome of anti-PD1 therapy in patients with lung
activity of alectinib against systemic disease and brain cancer brain metastasis. Neuro-Oncol.
metastases in patients with crizotinib-resistant ALK- 2017;19(suppl_6):vi39. https://doi.org/10.1093/
rearranged non-small-cell lung cancer (AF-002JG): neuonc/nox168.151.

Anda mungkin juga menyukai