Skip to main content
Erschienen in: Radiation Oncology 1/2020

Open Access 01.12.2020 | Research

Whole brain radiotherapy (WBRT) for leptomeningeal metastasis from NSCLC in the era of targeted therapy: a retrospective study

verfasst von: Junjie Zhen, Lei Wen, Mingyao Lai, Zhaoming Zhou, Changguo Shan, Shaoqun Li, Tao Lin, Jie Wu, Wensheng Wang, Shaoqiang Xu, Da Liu, Ming Lu, Dan Zhu, Longhua Chen, Linbo Cai, Cheng Zhou

Erschienen in: Radiation Oncology | Ausgabe 1/2020

Abstract

Background and purpose

Leptomeningeal metastasis (LM) is a rare but detrimental complication in patients with non-small cell lung cancer (NSCLC). Although whole brain radiotherapy (WBRT) is used to eliminating cancer cells or microscopic foci, it is becoming less favorable due to the concerns over neurocognitive toxicity. This study aimed to re-evaluate the role of WBRT in the setting of modern targeted therapy.

Materials and methods

From December 2014 to March 2019, 80 NSCLC patients with cytologically and/or radiologically proven LM diagnosis were retrospectively analyzed.

Results

The median OS (mOS) after diagnosis of LM was 8.0 (95%CI: 4.4 to 11.6) months, and the one-year OS was 39.4%. The mOS for EGFR-mutated LM patients was 12.6 (3.0 to 22.2) months versus only 4.1 (2.8 to 5.4) for patients with wild-type EGFR (P < 0.001). Younger patients (< 53.5 yrs.) appeared to have a better OS than older patients (≥53.5 yrs.) (12.6 vs. 6.1, P = 0.041). No survival benefits were found in EGFR-mutated patients who received WBRT (P = 0.490). In contrast, mOS was significantly prolonged in wild-type EGFR patients with WBRT versus non-WBRT (mOS: 8.0 vs. 2.1, P = 0.002). Multivariate analysis indicated that WBRT (P = 0.025) and younger age (P = 0.048) were independent prognostic factors that predicted prolonged survival for wild-type EGFR LM patients from NSCLC.

Conclusion

Our study demonstrated that WBRT has clear survival advantages for patients with wild-type EGFR, and molecular biological stratification of LM patients for WBRT is highly recommended.
Hinweise
Junjie Zhen and Lei Wen contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALK
Anaplastic lymphoma kinase
CI
Confidence interval
CNS
Central nervous system
CSF
Cerebrospinal fluid
ECOG
Eastern Cooperative Oncology Group
EGFR
Epidermal growth factor receptor
EVD
External ventricular drainage
GCS
Glasgow Coma Scale
Gy
Gray
HR
Hazard ratio
IMRT
Intensity modulated radiation therapy
ITC
Intrathecal chemotherapy
KPS
Karnofsky Performance Status
LCD
Lumbar cistern drainage
LM
Leptomeningeal metastasis
MRI
Magnetic resonance imaging
NSCLC
Non-small-cell lung cancer
OS
Overall survival
PCR
Polymerase chain reaction
PS
Performance status
PD-1
Program death 1
PD-L1
Program death ligand 1
TKI
Tyrosine kinase inhibitors
VP
Ventriculoperitoneal
WBRT
Whole brain radiotherapy
3D-CRT
3-dimensional conformal radiation therapy

Introduction

Leptomeningeal metastasis (LM) is a deleterious complication that occurs in 3–5% of patients with advanced non-small cell lung cancer (NSCLC) [1]. In recent years, the growing incidence of LM is likely due to improved supportive care as well as prolonged survival in patients with targetable mutations, particularly epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations [2, 3]. Nevertheless, LM is associated with a very poor prognosis [46].
The last decade saw considerably changes in management of NSCLC by appreciating the molecular characterization of the disease. These findings have led to biological stratifications of the patients as well as the discovery of a variety of targeted cancer drugs. For instance, the sequential development of tyrosine kinase inhibitors (TKIs) as well as check point inhibitors for program death ligand 1 (PD-L1) were enabled [7, 8]. As a result, the prognosis and quality of life for NSCLC patients has improved. Unfortunately, this is not the case in LM. Many clinical trials in NSCLC excluded patients with LM. There have been a few retrospective studies evaluating the prognosis of LM from NSCLC in the EGFR mutation subgroup. The median overall survival of these studies varies from 3 to 4 months [9, 10] to 9–12 months [11, 12].
Local-regional treatments including whole brain radiotherapy (WBRT), intrathecal chemotherapy (ITC), and ventriculoperitoneal (VP) shunt operations play a critical role in relieving neurological symptoms caused by intracranial hypertension in LM [1315]. Although non-invasive whole brain radiotherapy (WBRT) seems to be effective in palliating neurologic signs and symptoms [13], WBRT has not been widely accepted for treating LM due to its plausible amelioration in survivals at the expense of potentially neurocognitive toxicity [11, 16]. Several important questions have yet to be answered: 1) whether WBRT has an impact on survival outcomes; 2) if so, which sub-population will benefit from WBRT; 3) what are the optimal doses of WBRT for patients with different clinical and biological backgrounds. Herein, we aimed to understand the role of WBRT in treatment of LM from NSCLC based on a retrospective cohort in China. We further evaluated the clinicopathological factors and survival of LM patients who underwent various local or systemic treatments in the era of molecular targeted therapy.

Materials and methods

Patients

Patients diagnosed with LM from NSCLC at our institution were retrospectively analyzed from December 2014 to March 2019. The inclusion criteria included: (1) definitive cases with confirmed malignant cells in CSF, and/or (2) diffuse linear leptomeningeal enhancements following the gyri, sulci or ependymal surface by gadolinium-enhanced magnetic resonance imaging (MRI) scan, and (3) gene tests of EGFR mutation. We excluded patients with ALK translocation (n = 5) and those whose EGFR mutation status was unknown (n = 20). Data on the clinical characteristics of patients as well as disease-related features including radiology, histology, molecular status of EGFR mutation, and treatment regimens were reviewed. This study was approved by the Ethics Committee of Guangdong Sanjiu Brain Hospital.

Treatments

Systemic therapy for patients with LM was administered according to NCCN guidelines for NSCLC including EGFR tyrosine kinase inhibitors (TKIs) for those with EGFR mutations and chemotherapy with or without bevacizumab. WBRT was implemented with either 30 Gy in 15 fractions of 2 Gy, or 36 Gy in 18 fractions of 2 Gy, delivered 5 days a week, by intensity-modulated radiation therapy (IMRT) or 3-dimensional conformal radiation therapy (3D-CRT). Five patients with concurrent brain metastasis were treated by stereotactic radiosurgery (SRS) or fractioned SRS (12–24 Gy/ 1–3 fractions) using the Novalis Tx® system. For patients with significant clinical symptoms of meningeal irritation, surgical interventions including ventriculoperitoneal (VP) shunt, external ventricular drainage (EVD), or lumbar cistern drainage (LCD) were provided especially for those with elevated intracranial pressure prior to radiotherapy. Adverse events were graded according to the National Cancer Institute Common Toxicity Criteria for Adverse Events version 4.0. Adverse events were graded according to the National Cancer Institute Common Toxicity Criteria for Adverse Events version 4.0.

Statistics

The primary endpoint of our study was median overall survival (OS) determined as the time from initial diagnosis of LM to death from any cause or censored at the date of last follow-up unless otherwise specified. The radiological signs of LM are difficult for assessment; therefore, the progression free survival and overall response rates were not calculated in this retrospective cohort. Continuous data were presented as the median (minimum-maximum). Categorical data were presented as quantities and proportions. The overall survival was estimated using the Kaplan-Meier product-limited method with ‘survival’ package [17] in R software; survival curves were compared between groups using a log-rank test. Univariable and multivariable Cox proportional hazards regression models were performed to evaluate the influence of the clinical and pathologic parameters on mortality of NSCLC patients with LM. Continuous data such as age and Karnofsky Performance Status (KPS) score were divided into two subgroups via a cut-off value using the median value of the cohort for univariable and multivariable regression modeling. Statistical analyses used R software version 3.5.1. All statistical assessments were two-sided, and P < 0.05 was considered to be statistically significant.

Results

Patient characteristics

Eighty NSCLC patients with LM were eligible for our study and consisted of 44 males and 36 females. Baseline characteristics of all included patients are listed below (Table 1). The median age at diagnosis of LM was 53.5 (range: 30 to 78 years). Most patients had adenocarcinoma histology (73/80, 91.3%). Malignant cells were found in 55 patients from CSF while the remaining 25 patients were diagnosed with LM according to magnetic resonance imaging (MRI) findings and clinical presentations. Nineteen patients had synchronous LM when diagnosed with NSCLC while 61 patients progressed to LM after at least one line of systematic therapy. The median time from diagnosis of NSCLC to metachronous LM was17.6 month (range: 3.0 to 73.0). Forty-two (52.5%) patients had brain metastasis when diagnosed with LM. Fifty-one (63.8%) patients also had extra-central nervous system (CNS) metastasis mainly involving lung, liver, and lymph nodes (Table 2).
Table 1
Patient characteristics of 80 NSCLC patients with LM
Variable
N (%)
Total
80 (100.0)
Gender
 Male
44 (54.1)
 Female
36 (45.9)
Age, years
 Median (range)
53.5 (30–78)
Histology
 Adenocarcinoma
73 (91.3)
 Squamous
7 (8.8)
Initial stage
 I-III
25 (31.3)
 IV
55 (68.8)
Previous lines of systematic therapy
 0
19 (23.8)
 1
29 (36.3)
 2
19 (23.8)
 ≥ 3
13 (16.3)
Previous EGFR TKI prior to LM
46 (57.5)
EGFR mutation
 EGFR 19del
25 (31.3)
 EGFR L858R
18 (22.5)
 EGFR T790M
5 (6.3)
 EGFR 20INS
1 (1.3)
 Wild-type
31 (38.8)
Abbreviations: EGFR Epidermal growth factor receptor, TKI Tyrosine kinase inhibitors, LM Leptomeningeal metastasis
Table 2
Clinical presentation and treatment of 80 NSCLC patients with LM
Variable
N (%)
Timing of metastasis
 Synchronous
19 (23.8)
 Metachronous
61 (76.3)
Time from diagnosis of lung cancer to LM, months
 Median (range)
12.5 (−0.2 to 73.0)
KPS score
 ≥ 80
21 (26.3%)
 70–50
41 (51.3%)
 ≤ 40
18 (22.5%)
GCS score
 Less than 15
16 (20.0)
With CNS symptoms
80 (100.0)
Modality of LM diagnosis
 MRI
25 (31.3)
 CSF cytology
4 (5.0)
 MRI + CSF cytology
51 (63.8)
Concurrent brain metastasis
42 (52.5)
Extra-CNS metastasis
51 (63.8)
Local treatment for LM
 WBRT
38 (47.5)
 VP shunt
6 (7.5)
 Ventricular external drainage
7 (8.8)
 Lumbar cistern drainage
3 (3.8)
Systematic treatment for LM
 Osimertinib
25 (31.3)
 Gefitinib
12 (15.0)
 Erlotinib
4 (5.0)
 Icotinib
5 (6.3)
 Afatinib
1 (1.3)
 Gefitinib followed by Osimertinib
2 (2.5)
 Bevacizumab
9 (11.3)
 Chemotherapy
36 (45.0)
Abbreviations: CNS Central nervous system, CSF Cerebrospinal fluid, MRI Magnetic resonance imaging, VP Ventriculoperitoneal, KPS Karnofsky performance status, GCS Glasgow coma scale, EGFR Epidermal growth factor receptor, TKI Tyrosine kinase inhibitors, LM Leptomeningeal metastasis, WBRT Whole brain radiotherapy, Gy Gray, HR Hazard ratio, CI Confidence interval
EGFR mutations were determined by gene panel sequencing or polymerase chain reaction (PCR)-based assays on different samples (37 from tumor tissue, 24 from CSF, 9 from both CSF and serum, 6 from serum, 3 from both tumor tissue and serum, and 1 from pleural effusion). Of these, 25 patients had EGFR exon 19 deletion, 18 had EGFR exon L858R mutation, and 1 patient had EGFR exon 20 insertion. The T790M mutations were reported in five patients. The remaining 31 patients were wild-type EGFR. Forty-six (57.5%) patients had received EGFR TKI therapy prior to the diagnosis of LM (Table 1).

Treatment after diagnosis of LM

Thirty-eight (47.5%) patients received WBRT out of a total of 30–36 Gy/15–18 fractions. Stereotactic radiosurgery was implemented in 5 patients for the treatment of concurrent brain metastasis. Sixteen patients underwent surgical interventions including 6 VP shunts, 7 external ventricle drainage cases, and 3 lumbar cistern drainages. With respect to systematic therapy after diagnosis of LM, 25 patients received osimertinib, 22 patients received first- or second-generation EGFR TKI (including gefitinib in 12 patients, erlotinib in 4, icotinib in 5, and afatinib in 1 patients). Two patients received gefitinib followed by osimertinib; 36 patients were given platinum-based chemotherapy, and 9 of them were supplemented with bevacizumab (Table 2).

Survival analysis

The median follow-up time was 8.6 months (range: 1 to 28.4 months); 47 patients died within this period, 29 patients were still alive, and 4 were lost to follow-up. The median OS after diagnosis of LM was 8.0 months (95% confidence interval [CI]: 4.4 to 11.6 months), and one-year OS was 39.4% (Fig. 1a). Subgroup analysis revealed that the median OS for EGFR-mutated patients was 12.6 months (95% CI: 3.0 to 22.2 months), which was significantly longer than those with wild-type EGFR LM patients (4.1 months, 95% CI: 2.8 to 5.4 months, P < 0.001) (Fig. 1b). Patients who received EGFR TKI were identified to have a better OS than non-TKI treatment (11.1 vs. 2.5 months, P < 0.001).
We next asked whether age at diagnosis of LM has an impact on survival and prognosis; subgroup analysis indicated that younger age (< 53.5 yrs.) appeared to gain a favorable OS versus older ones (≥53.5 yrs.) (12.6 vs. 6.1 months, P = 0.041) (Fig. 1c). No significant difference was found in OS with reference to sex, KPS score, Glasgow Coma Scale (GCS) score, concurrent brain metastasis, chemotherapy, and bevacizumab therapy (Table 3). Multivariate analysis showed that EGFR mutations (P = 0.013, HR 0.39, 95% CI: 0.186 to 0.820) and younger age (P = 0.025, HR 0.492, 95% CI: 0.265 to 0.915) were still independent prognostic factors that predicted better survival (Table 3).
Table 3
Univariate and multivariate analysis for survival in 80 NSCLC patients with LM
 
Univariate analysis
Multivariate analysis
HR (95%CI)
P
HR (95%CI)
P
Gender (female vs. male)
0.950 (0.531 to 1.698)
0.862
  
Age (< 53.5 vs. ≥53.5)
0.545 (0.301 to 0.987)
0.043
0.492 (0.265 to 0.915)
0.025
KPS (< 80 vs. ≥80)
1.298 (0.640 to 2.632)
0.470
  
GCS (15 vs. ≤14)
1.126 (0.572 to 2.220)
0.731
  
Concurrent brain metastasis (yes vs. no)
1.039 (0.585 to 1.846)
0.895
  
EGFR mutation (yes vs. no)
0.288 (0.159 to 0.522)
0.000
0.390 (0.186 to 0.820)
0.013
EGFR TKI after LM (yes vs. no)
0.260 (0.138 to 0.487)
0.000
0.549 (0.491 to 1.039)
0.053
WBRT (yes vs. no)
0.565 (0.315 to 1.013)
0.053
0.697 (0.373 to 1.306)
0.260
Chemotherapy (yes vs. no)
0.873 (0.480 to 1.589)
0.657
  
Bevacizumab (yes vs. no)
1.909 (0.677 to 5.386)
0.222
  
Abbreviations: NSCLC non-small-cell lung cancer, KPS Karnofsky Performance Status, GCS Glasgow Coma Scale, EGFR epidermal growth factor receptor, TKI tyrosine kinase inhibitors, LM leptomeningeal metastasis, WBRT whole brain radiotherapy, Gy gray, HR hazard ratio, CI confidence interval

Distinct survival impacts of WBRT

To study the therapeutic effects of WBRT, the OS of patients who received WBRT (WBRT group, n = 38) were evaluated versus those without WBRT (non-WBRT group, n = 42). Although no significant difference in OS was found between two arms (P = 0.051), the median OS of WBRT group was numerically doubled compared to non-WBRT group (11.4 vs. 5.0 months). (Fig. 2a). We next explored the consequences of WBRT in LM patients with different mutation backgrounds via sub-group analysis. For patients harboring EGFR mutations, no survival benefits were observed with WBRT treatment by univariate analysis (WBRT vs. non-WBRT: P = 0.49, HR 0.73, 95%CI 0.297 to 1.794) (Fig. 2b). Interestingly, with respect to patients with wild-type EGFR, the survival advantages of WBRT was exceptionally established (median OS: 8.0 vs. 2.1 months, P = 0.002, HR 0.229, 95%CI 0.083 to 0.632) (Fig. 2c). Multivariate analysis consistently showed that WBRT (P = 0.025) and younger age (P = 0.048) were independent prognostic factors that predicted favorable overall survival in patients with wild-type EGFR (Table 4).
Table 4
Univariate and multivariate analysis for survival in 31 LM patients with wild-type EGFR
 
Univariate analysis
Multivariate analysis
HR (95%CI)
P
HR (95%CI)
P
Gender (female vs. male)
0.777 (0.328 to 1.844)
0.567
  
Age (< 53.5 vs. ≥53.5)
0.397 (0.153 to 1.029)
0.057
0.353 (0.125 to 0.993)
0.048
KPS (< 80 vs. ≥80)
0.654 (0.233 to 1.831)
0.419
  
GCS (15 vs. ≤ 14)
0.687 (0.232 to 2.033)
0.497
  
Concurrent brain metastasis (yes vs. no)
0.891 (0.380 to 2.088)
0.791
  
WBRT (yes vs. no)
0.229 (0.083 to 0.632)
0.004
0.300 (0.105 to 0.858)
0.025
Chemotherapy (yes vs. no)
0.422 (0.176 to 1.014)
0.054
0.487 (0.187 to 1.268)
0.140
Bevacizumab (yes vs. no)
0.034 (0.000 to 23.167)
0.310
  
Abbreviations: KPS Karnofsky performance status, GCS Glasgow coma scale, EGFR Epidermal growth factor receptor, WBRT Whole brain radiotherapy, Gy Gray, HR Hazard ratio, CI Confidence interval

Toxicity of WBRT

Two patients in WBRT group suspended irradiation due to worsening headache, both of them completed irradiation after decompression surgery were conducted. Other WBRT related toxicities including grade 1 to 2 nausea/vomiting in 7 patients, grade 1 transient headache in 5 patients, grade 1 radiation dermatitis in 3 patients and grade 1 hearing impaired in 1 patient.

Discussion

This cohort study shows a relatively long median OS (8.0 months) in patients diagnosed with LM from NSCLC in a single institution, particularly in an EGFR-mutated group (12.6 months) versus previous published data ranging from 3 to 6 months [46, 9, 10]. The most important factor in NSCLC in recent years is an improved understanding of molecular characteristics that leads to precision therapy of metastatic NSCLC [18]. Nonetheless, high-level evidence of targeted therapy for the treatment of LM from NSCLC has been scarce since the Iressa Pan-Asia Study (IPASS) [18] and most subsequent studies have excluded patients with LM from clinical trials. A large-scale Chinese study of 5387 lung cancer patients found that EGFR-mutated subjects have a significantly higher risk for LM versus wild-type subjects (9.4% vs. 1.7%). The OS after LM was remarkably improved in the TKI therapy group versus non-TKI treatment (10.0 vs. 3.3 months) [2]. Another multicenter retrospective analysis from Europe consisting of 92 EGFR-mutated NSCLC patients with LM showed that the median OS from diagnosis of LM was 6.1 months; re-challenging with TKI in TKI-failed patients showed a better prognosis versus patients without further therapy (7.6 vs. 4.2 months) [19].
Besides evidence from retrospective studies, several preliminary prospective studies explored the efficiency of targeted therapy for EGFR-mutated LM patients. Nanjo et al. [20] examined the effects of the third-generation EGFR TKI osimertinib in a prospective pilot study including 13 patients with T790M-positive NSCLC LM patients after failure of first- or second-line EGFR TKI. Six out of eight patients achieved CNS improvement, and extra-CNS improvement was seen in five patients. The median progression-free survival for all 13 patients was 7.2 months. The BLOOM study is a phase I clinical trial to assess the safety and activity of AZD3759 or AZD9291 in patients with EGFR-mutated advanced-stage NSCLC with central nervous system metastasis. AZD3759 [21] showed a tolerable safety profile at a dose of 200 mg twice daily. Of 18 patients with LM pretreated with EGFR TKI, five (28%) patients had a confirmed response, and 14 (78%) achieved confirmed disease control. AZD9291 [22] also showed a manageable safety profile with the investigator-assessed median PFS 8.6 months and median OS 11.0 months in EGFR-mutant NSCLC with LM.
There is also limited progress with respect to wild-type EGFR NSCLC patients with LM. WBRT is an important choice of local treatment in LM patients by eradicating cancer cells or microscopic foci at distant sites within the brain [13]. Owning to the neurologic toxicity, the role of WBRT has long been a controversial issue. There is still no consensus on whether WBRT could improve survival for patients with LM from NSCLC [16]. Suet al. showed that patients who received WBRT had a longer overall survival versus those who did not receive WBRT; WBRT was an independent favorable factor that predicted better survival [9]. Liao et al. reported that WBRT prolonged median OS from 2.4 months to 10.9 months in patients with LM from NSCLC [23]. However, a study from the US suggested that survival was not improved in 56 patients who received WBRT versus 69 patients who received no WBRT [24]. A multicenter study from 11 Dutch hospitals reported that median survival of NSCLC patients with LM was only 3.1 months, and WBRT did not affect survival after LM diagnosis based solely on EGFR-mutant NSCLC LM patients [25].
A probable reason for this discrepancy between the survival impacts of WBRT may be a mixture of patients with a diversity of clinical and molecular biological background. In fact, the therapeutic response to WBRT varies with the number of factors including the Eastern Cooperative Oncology Group (ECOG) performance status (PS) 1, time to leptomeningeal metastasis following NSCLC diagnosis, as well as lack of brain metastasis [26]. Our study reported a doubling of OS with statistical marginal difference between patients who received WBRT and the non-WBRT group (11.4 vs. 5.0 months, P = 0.051). Our subgroup analysis reported that 49 patients with definite EGFR mutation gained almost no benefit from WBRT. This agrees with Yan et al. who found that WBRT did not improve the overall survival of EGFR-mutated patients with LM [11]. We further reported that the overall survival in wild type patients with LM is noticeably enhanced. Su et al. showed that WBRT improved the overall survival in patients mostly (91.3%) had wild-type EGFR or unknown status [9]. As a result, LM patients with wild-type EGFR favor WBRT but EGFR-mutated patients do not — this is a key factor that might explain the various mOS reported from different studies.
Our study further implicated younger age (< 53.5 years) as an independent prognostic factor that predicts better survival. Younger patients tend to have fewer comorbidities and more tolerance to toxicities induced by a variety of treatments. LM commonly presents with increased intracranial hypertension-associated symptoms such as consistent headache, nausea, vomiting, and even disturbed consciousness. These signs may not be fundamentally alleviated by dehydration treatment alone. Surgical intervention such as ventriculoperitoneal shunt (VP shunt) plays an important role in these intractable situations. VP shunting could offer an effective palliative option for symptom relief of severe headache and improved quality of life in LM patients [14, 27]. Intrathecal chemotherapy could be implemented repeatedly and safely via implantation of an ommaya reservoir [28]. In our cohort, a small proportion of patients (16/80, 20%) had undergone surgical interventions including VP shunt, VED, and LCD. Those surgical interventions offer a supportive role for WBRT (data not shown).
Immunotherapy, particularly inhibitors of the programmed death-1 (PD-1)/PD-ligand 1 (PD-L1) pathways—have a greatly modified NSCLC treatment [29]. However, evidence of immunotherapy for the treatment of LM is limited because the tight junctions between ependymal cells in the choroid plexus are less permeable to T cells or anti-PD-1 monoclonal antibodies reaching the leptomeninges and CSF [30]. Few data are available except several case reports on the activity of PD-1/PD-L1 inhibitors for the treatment of LM. Kamath et al. reported a radiologically stable and neurologically intact treatment that lasts for at least 20 months in a woman with reginal bulky LM from NSCLC by combined treatment of stereotactic radiosurgery and pembrolizumab [31]. Gionet al. also reported neurological improvement after nivolumab treatment in a patient with NSCLC and symptomatic LM. The activity of pembrolizumab in LM was also investigated in a phase II study (NCT03091478) [32].
Among potential limitations of this study could be the relatively small number of patients for the subgroup analysis that might reduce the quality of the conclusions. Periodic follow-up on quality of life was also absent in this cohort.
In conclusion, this study revealed that the median overall survival in our cohort is higher than histological experience. EGFR mutations were identified as a prognostic factor that predicts favorable survival in NSCLC patients with LM. Our study also showed that WBRT could significantly improve the survival outcome of LM patients with wild-type EGFR. However, LM patients with EGFR mutations are likely to gain limited benefits from WBRT. Molecular biological stratifications of LM patients for WBRT are therefore recommended for routine clinical practice. Further clinical and mechanistic investigations for optimal radiation dose-fractionation regimens or the development of combined radio-sensitive agents are highly warranted.

Acknowledgements

Not applicable.
This study was approved by the institutional ethical committee.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Remon J, Le Rhun E, Besse B. Leptomeningeal carcinomatosis in non-small cell lung cancer patients: a continuing challenge in the personalized treatment era. Cancer Treat Rev. 2017;53:128–37.CrossRef Remon J, Le Rhun E, Besse B. Leptomeningeal carcinomatosis in non-small cell lung cancer patients: a continuing challenge in the personalized treatment era. Cancer Treat Rev. 2017;53:128–37.CrossRef
2.
Zurück zum Zitat Li YS, Jiang BY, Yang JJ, Tu HY, Zhou Q, Guo WB, et al. Leptomeningeal metastases in patients with NSCLC with EGFR mutations. J Thorac Oncol. 2016;11:1962–9.CrossRef Li YS, Jiang BY, Yang JJ, Tu HY, Zhou Q, Guo WB, et al. Leptomeningeal metastases in patients with NSCLC with EGFR mutations. J Thorac Oncol. 2016;11:1962–9.CrossRef
3.
Zurück zum Zitat Omuro AM, Kris MG, Miller VA, Franceschi E, Shah N, Milton DT, et al. High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib. Cancer. 2005;103:2344–8.CrossRef Omuro AM, Kris MG, Miller VA, Franceschi E, Shah N, Milton DT, et al. High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib. Cancer. 2005;103:2344–8.CrossRef
4.
Zurück zum Zitat Riess JW, Nagpal S, Iv M, Zeineh M, Gubens MA, Ramchandran K, et al. Prolonged survival of patients with non–small-cell lung cancer with leptomeningeal carcinomatosis in the modern treatment era. Clin Lung Cancer. 2014;15:202–6.CrossRef Riess JW, Nagpal S, Iv M, Zeineh M, Gubens MA, Ramchandran K, et al. Prolonged survival of patients with non–small-cell lung cancer with leptomeningeal carcinomatosis in the modern treatment era. Clin Lung Cancer. 2014;15:202–6.CrossRef
5.
Zurück zum Zitat Park JH, Kim YJ, Lee JO, Lee KW, Kim JH, Bang SM, et al. Clinical outcomes of leptomeningeal metastasis in patients with non-small cell lung cancer in the modern chemotherapy era. Lung Cancer. 2012;76:387–92.CrossRef Park JH, Kim YJ, Lee JO, Lee KW, Kim JH, Bang SM, et al. Clinical outcomes of leptomeningeal metastasis in patients with non-small cell lung cancer in the modern chemotherapy era. Lung Cancer. 2012;76:387–92.CrossRef
6.
Zurück zum Zitat Brower JV, Saha S, Rosenberg SA, Hullett CR, Ian RH. Management of leptomeningeal metastases: prognostic factors and associated outcomes. J Clin Neurosci. 2016;27:130–7.CrossRef Brower JV, Saha S, Rosenberg SA, Hullett CR, Ian RH. Management of leptomeningeal metastases: prognostic factors and associated outcomes. J Clin Neurosci. 2016;27:130–7.CrossRef
7.
Zurück zum Zitat Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020;382:41–50.CrossRef Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020;382:41–50.CrossRef
8.
Zurück zum Zitat Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019;37:537–46.CrossRef Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019;37:537–46.CrossRef
9.
Zurück zum Zitat Lee SJ, Lee J-I, Nam D-H, Ahn YC, Han JH, Sun J-M, et al. Leptomeningeal carcinomatosis in non–small-cell lung cancer patients: impact on survival and correlated prognostic factors. J Thorac Oncol. 2013;8:185–91.CrossRef Lee SJ, Lee J-I, Nam D-H, Ahn YC, Han JH, Sun J-M, et al. Leptomeningeal carcinomatosis in non–small-cell lung cancer patients: impact on survival and correlated prognostic factors. J Thorac Oncol. 2013;8:185–91.CrossRef
10.
Zurück zum Zitat Kwon BS, Cho YH, Yoon SK, Lee DH, Kim SW, Kwon DH, et al. Impact of clinicopathologic features on leptomeningeal metastasis from lung adenocarcinoma and treatment efficacy with epidermal growth factor receptor tyrosine kinase inhibitor. Thorac Cancer. 2020;11:436–42.CrossRef Kwon BS, Cho YH, Yoon SK, Lee DH, Kim SW, Kwon DH, et al. Impact of clinicopathologic features on leptomeningeal metastasis from lung adenocarcinoma and treatment efficacy with epidermal growth factor receptor tyrosine kinase inhibitor. Thorac Cancer. 2020;11:436–42.CrossRef
11.
Zurück zum Zitat Yan W, Liu Y, Li J, Han A, Kong L, Yu J, et al. Whole brain radiation therapy does not improve the overall survival of EGFR-mutant NSCLC patients with leptomeningeal metastasis. Radiat Oncol. 2019;14:168.CrossRef Yan W, Liu Y, Li J, Han A, Kong L, Yu J, et al. Whole brain radiation therapy does not improve the overall survival of EGFR-mutant NSCLC patients with leptomeningeal metastasis. Radiat Oncol. 2019;14:168.CrossRef
12.
Zurück zum Zitat Yin K, Li YS, Zheng MM, Jiang BY, Li WF, Yang JJ, et al. A molecular graded prognostic assessment (molGPA) model specific for estimating survival in lung cancer patients with leptomeningeal metastases. Lung Cancer. 2019;131:134–8.CrossRef Yin K, Li YS, Zheng MM, Jiang BY, Li WF, Yang JJ, et al. A molecular graded prognostic assessment (molGPA) model specific for estimating survival in lung cancer patients with leptomeningeal metastases. Lung Cancer. 2019;131:134–8.CrossRef
13.
Zurück zum Zitat Rudnicka H, Niwińska A, Murawska M. Breast cancer leptomeningeal metastasis—the role of multimodality treatment. J Neuro-Oncol. 2007;84:57–62.CrossRef Rudnicka H, Niwińska A, Murawska M. Breast cancer leptomeningeal metastasis—the role of multimodality treatment. J Neuro-Oncol. 2007;84:57–62.CrossRef
14.
Zurück zum Zitat Murakami Y, Ichikawa M, Bakhit M, Jinguji S, Sato T, Fujii M, et al. Palliative shunt surgery for patients with leptomeningeal metastasis. Clin Neurol Neurosurg. 2018;168:175–8.CrossRef Murakami Y, Ichikawa M, Bakhit M, Jinguji S, Sato T, Fujii M, et al. Palliative shunt surgery for patients with leptomeningeal metastasis. Clin Neurol Neurosurg. 2018;168:175–8.CrossRef
15.
Zurück zum Zitat Le Rhun E, Wallet J, Mailliez A, Le Deley MC, Rodrigues I, Boulanger T, et al. Intrathecal liposomal cytarabine plus systemic therapy versus systemic chemotherapy alone for newly diagnosed leptomeningeal metastasis from breast cancer. Neuro Oncol. 2020;22:524-38. Le Rhun E, Wallet J, Mailliez A, Le Deley MC, Rodrigues I, Boulanger T, et al. Intrathecal liposomal cytarabine plus systemic therapy versus systemic chemotherapy alone for newly diagnosed leptomeningeal metastasis from breast cancer. Neuro Oncol. 2020;22:524-38.
16.
Zurück zum Zitat Cheng H, Perez-Soler R. Leptomeningeal metastases in non-small-cell lung cancer. Lancet Oncol. 2018;19:e43–55.CrossRef Cheng H, Perez-Soler R. Leptomeningeal metastases in non-small-cell lung cancer. Lancet Oncol. 2018;19:e43–55.CrossRef
17.
Zurück zum Zitat Therneau TM. A package for survival analysis in S. 2015. version 2.38; 2015. Therneau TM. A package for survival analysis in S. 2015. version 2.38; 2015.
18.
Zurück zum Zitat Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57.CrossRef Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57.CrossRef
19.
Zurück zum Zitat Flippot R, Biondani P, Auclin E, Xiao D, Hendriks L, Le Rhun E, et al. Activity of EGFR tyrosine kinase inhibitors in NSCLC with refractory leptomeningeal metastases. J Thorac Oncol. 2019;14:1400–7.CrossRef Flippot R, Biondani P, Auclin E, Xiao D, Hendriks L, Le Rhun E, et al. Activity of EGFR tyrosine kinase inhibitors in NSCLC with refractory leptomeningeal metastases. J Thorac Oncol. 2019;14:1400–7.CrossRef
20.
Zurück zum Zitat Nanjo S, Hata A, Okuda C, Kaji R, Okada H, Tamura D, et al. Standard-dose osimertinib for refractory leptomeningeal metastases in T790M-positive EGFR-mutant non-small cell lung cancer. Br J Cancer. 2018;118(1):32–7.CrossRef Nanjo S, Hata A, Okuda C, Kaji R, Okada H, Tamura D, et al. Standard-dose osimertinib for refractory leptomeningeal metastases in T790M-positive EGFR-mutant non-small cell lung cancer. Br J Cancer. 2018;118(1):32–7.CrossRef
21.
Zurück zum Zitat Ahn MJ, Kim DW, Cho BC, Kim SW, Lee JS, Ahn JS, et al. Activity and safety of AZD3759 in EGFR-mutant non-small-cell lung cancer with CNS metastases (BLOOM): a phase 1, open-label, dose-escalation and dose-expansion study. Lancet Respir Med. 2017;5:891–902.CrossRef Ahn MJ, Kim DW, Cho BC, Kim SW, Lee JS, Ahn JS, et al. Activity and safety of AZD3759 in EGFR-mutant non-small-cell lung cancer with CNS metastases (BLOOM): a phase 1, open-label, dose-escalation and dose-expansion study. Lancet Respir Med. 2017;5:891–902.CrossRef
22.
Zurück zum Zitat Yang JCH, Kim SW, Kim DW, Lee JS, Cho BC, Ahn JS, et al. Osimertinib in patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer and leptomeningeal metastases: the BLOOM study. J Clin Oncol. 2020;38:538-47. Yang JCH, Kim SW, Kim DW, Lee JS, Cho BC, Ahn JS, et al. Osimertinib in patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer and leptomeningeal metastases: the BLOOM study. J Clin Oncol. 2020;38:538-47.
23.
Zurück zum Zitat Liao BC, Lee JH, Lin CC, Chen YF, Chang CH, Ho CC, et al. Epidermal growth factor receptor tyrosine kinase inhibitors for non-small-cell lung cancer patients with leptomeningeal carcinomatosis. J Thorac Oncol. 2015;10:1754–61.CrossRef Liao BC, Lee JH, Lin CC, Chen YF, Chang CH, Ho CC, et al. Epidermal growth factor receptor tyrosine kinase inhibitors for non-small-cell lung cancer patients with leptomeningeal carcinomatosis. J Thorac Oncol. 2015;10:1754–61.CrossRef
24.
Zurück zum Zitat Morris PG, Reiner AS, Szenberg OR, Clarke JL, Panageas KS, Perez HR, et al. Leptomeningeal metastasis from non-small cell lung cancer: survival and the impact of whole brain radiotherapy. J Thorac Oncol. 2012;7:382–5.CrossRef Morris PG, Reiner AS, Szenberg OR, Clarke JL, Panageas KS, Perez HR, et al. Leptomeningeal metastasis from non-small cell lung cancer: survival and the impact of whole brain radiotherapy. J Thorac Oncol. 2012;7:382–5.CrossRef
25.
Zurück zum Zitat Kuiper JL, Hendriks LE, van der Wekken AJ, de Langen AJ, Bahce I, Thunnissen E, et al. Treatment and survival of patients with EGFR-mutated non-small cell lung cancer and leptomeningeal metastasis: a retrospective cohort analysis. Lung Cancer. 2015;89:255–61.CrossRef Kuiper JL, Hendriks LE, van der Wekken AJ, de Langen AJ, Bahce I, Thunnissen E, et al. Treatment and survival of patients with EGFR-mutated non-small cell lung cancer and leptomeningeal metastasis: a retrospective cohort analysis. Lung Cancer. 2015;89:255–61.CrossRef
26.
Zurück zum Zitat Ozdemir Y, Yildirim BA, Topkan E. Whole brain radiotherapy in management of non-small-cell lung carcinoma associated leptomeningeal carcinomatosis: evaluation of prognostic factors. J Neuro-Oncol. 2016;129:329–35.CrossRef Ozdemir Y, Yildirim BA, Topkan E. Whole brain radiotherapy in management of non-small-cell lung carcinoma associated leptomeningeal carcinomatosis: evaluation of prognostic factors. J Neuro-Oncol. 2016;129:329–35.CrossRef
27.
Zurück zum Zitat Omuro AM, Lallana EC, Bilsky MH, DeAngelis LM. Ventriculoperitoneal shunt in patients with leptomeningeal metastasis. Neurology. 2005;64:1625–7.CrossRef Omuro AM, Lallana EC, Bilsky MH, DeAngelis LM. Ventriculoperitoneal shunt in patients with leptomeningeal metastasis. Neurology. 2005;64:1625–7.CrossRef
28.
Zurück zum Zitat Peyrl A, Chocholous M, Azizi AA, Czech T, Dorfer C, Mitteregger D, et al. Safety of Ommaya reservoirs in children with brain tumors: a 20-year experience with 5472 intraventricular drug administrations in 98 patients. J Neuro-Oncol. 2014;120:139–45.CrossRef Peyrl A, Chocholous M, Azizi AA, Czech T, Dorfer C, Mitteregger D, et al. Safety of Ommaya reservoirs in children with brain tumors: a 20-year experience with 5472 intraventricular drug administrations in 98 patients. J Neuro-Oncol. 2014;120:139–45.CrossRef
29.
Zurück zum Zitat Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.CrossRef Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.CrossRef
30.
Zurück zum Zitat Otsubo K, Seki N, Nakanishi Y, Okamoto I. Development of leptomeningeal carcinomatosis during a marked response of brain metastases to pembrolizumab in a patient with non-small-cell lung cancer. Ann Oncol. 2018;29:780–1.CrossRef Otsubo K, Seki N, Nakanishi Y, Okamoto I. Development of leptomeningeal carcinomatosis during a marked response of brain metastases to pembrolizumab in a patient with non-small-cell lung cancer. Ann Oncol. 2018;29:780–1.CrossRef
31.
Zurück zum Zitat Kamath SD, Kumthekar PU, Kruser TJ, Mohindra NA. Intracranial response to anti-programmed death 1 therapy in a patient with metastatic non-small cell lung cancer with leptomeningeal carcinomatosis. Oncologist. 2018;23:e159–e61.CrossRef Kamath SD, Kumthekar PU, Kruser TJ, Mohindra NA. Intracranial response to anti-programmed death 1 therapy in a patient with metastatic non-small cell lung cancer with leptomeningeal carcinomatosis. Oncologist. 2018;23:e159–e61.CrossRef
32.
Zurück zum Zitat Gion M, Remon J, Caramella C, Soria JC, Besse B. Symptomatic leptomeningeal metastasis improvement with nivolumab in advanced non-small cell lung cancer patient. Lung Cancer. 2017;108:72–4.CrossRef Gion M, Remon J, Caramella C, Soria JC, Besse B. Symptomatic leptomeningeal metastasis improvement with nivolumab in advanced non-small cell lung cancer patient. Lung Cancer. 2017;108:72–4.CrossRef
Metadaten
Titel
Whole brain radiotherapy (WBRT) for leptomeningeal metastasis from NSCLC in the era of targeted therapy: a retrospective study
verfasst von
Junjie Zhen
Lei Wen
Mingyao Lai
Zhaoming Zhou
Changguo Shan
Shaoqun Li
Tao Lin
Jie Wu
Wensheng Wang
Shaoqiang Xu
Da Liu
Ming Lu
Dan Zhu
Longhua Chen
Linbo Cai
Cheng Zhou
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2020
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-020-01627-y

Weitere Artikel der Ausgabe 1/2020

Radiation Oncology 1/2020 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.