Abstract
Purpose
Leptomeningeal metastasis (LM) is a severe complication of non-small cell lung cancer (NSCLC). In patients with NSCLC LM harboring epidermal growth factor receptor (EGFR) mutations, osimertinib is favored over alternative EGFR tyrosine kinase inhibitors (TKIs). However, the efficacy of osimertinib relative to other EGFR-TKIs is not well established for patients with LM. We aimed to compare the efficacy of EGFR-TKIs in EGFR-mutated NSCLC LM.
Methods
This systematic review and meta-analysis performed according to PRISMA guidelines included studies of adult patients with EGFR-mutated NSCLC and a diagnosis of LM who received an EGFR-TKI for the treatment of LM. We searched Medline ALL, Embase, Cochrane Central Register of Controlled Trials, Scopus, and Web of Science Core Collection. The evaluation of biases was done by using the Ottawa-Newscastle scale. The hazard ratio was used as the parameter of interest for overall survival (OS) and central nervous system-specific progression-free survival (PFS).
Results
128 publications were included with 243 patients and 282 lines of EGFR-TKI for NSCLC LM that met inclusion criteria. The median PFS in patients receiving any EGFR-TKI was 9.1 months, and the median OS was 14.5 months. In univariate analyses of the entire cohort, osimertinib treatment demonstrated significantly prolonged PFS, but not OS, compared to other EGFR-TKIs. Osimertinib demonstrated significantly prolonged PFS and OS in the subset of patients who were previously treated with EGFR-TKIs, but not in EGFR-TKI naïve patients.
Conclusion
Osimertinib is associated with improved outcomes compared to other EGFR-TKIs, particularly in patients previously treated with EGFR-TKIs. An important limitation is that most patients were derived from retrospective reports. These results highlight the need for prospective studies for this difficult-to-treat patient population.
Keywords: Non-small cell lung cancer, NSCLC, Leptomeningeal, LMD, EGFR, Osimertinib, TKI
1. Introduction
Leptomeningeal metastasis (LM) is a complication in the advanced stages of cancer involving malignant cells metastasizing to the subarachnoid space surrounding the brain and spinal cord [1,2]. Alongside melanoma and breast cancer, lung cancer represents the most common solid adult cancer to metastasize to the leptomeninges [3]. The incidence of LM in non-small cell lung cancer (NSCLC) is approximately 2.8 % overall and up to 9 % in NSCLC patients whose tumor harbors epidermal growth factor receptor (EGFR) mutations [4]. Prior to the era of targeted therapy, patients with LM were treated with systemic chemotherapy or radiotherapy and experienced a median overall survival (mOS) of 2–5 months [[5], [6], [7]].
Current treatment guidelines for NSCLC LM include local and systemic approaches [8]. Whole brain radiotherapy (WBRT) is a frequently employed local therapy given that these patients are generally ineligible for surgery due to the diffuse nature of LM within the subarachnoid space. Systemic approaches for treating NSCLC LM consist of the standard therapies for NSCLC, including chemotherapy, targeted therapy, and immunotherapy [9].
When EGFR mutations are present, EGFR tyrosine kinase inhibitors (EGFR-TKIs) represent important components of the treatment armamentarium against NSCLC LM [10]. EGFR-TKIs approved by the United States Food and Drug Administration for EGFR-mutated NSCLC include the first- and second-generation EGFR-TKIs erlotinib, gefitinib afatinib, and dacomitinib, and the third-generation EGFR-TKI osimertinib [11]. First- and second-generation EGFR-TKIs have been used in various case reports and small-scale cohort studies for NSCLC LM, with prolonged therapeutic responses observed [[12], [13], [14], [15], [16]]. Third-generation EGFR-TKIs are notable for their ability to inhibit mutant EGFR proteins harboring the T790 M mutation, which constitutes the most common resistance mechanism to first- and second-generation EGFR-TKI [[17], [18], [19]]. Furthermore, osimertinib has demonstrated improved survival benefits compared to first-generation EGFR-TKIs for primary EGFR-mutated NSCLC in phase III clinical trials [20,21]. Osimertinib has also shown the ability to reach therapeutic concentrations in the central nervous system (CNS) with improved progression-free survival (PFS) in EGFR-mutated NSCLC patients with CNS parenchymal metastases versus first-generation EGFR-TKIs [9,18,20,22,23].
Given that LM is a relatively uncommon presentation of NSCLC, most studies on EGFR-TKIs for NSCLC have focused on advanced primary disease and CNS parenchymal metastases [[20], [21], [22],24]. There are limited studies specifically evaluating the efficacy of osimertinib for NSCLC LM, predominantly consisting of smaller-scale cohort studies [[25], [26], [27], [28], [29], [30], [31]]. Consequently, the efficacy of osimertinib for NSCLC LM compared to previous-generation EGFR-TKIs remains unclear, with no studies, to our knowledge, directly comparing first-, second-, and third-generation EGFR-TKIs. To address this gap, we conducted a systematic review and meta-analysis of individualized patient data to provide insights into the activity of different generations of EGFR-TKIs in the context of EGFR-mutated NSCLC LM.
2. Methods
2.1. Inclusion and exclusion criteria
Inclusion criteria consisted of the following: published reports of adult EGFR-mutated NSCLC, defined by next generation sequencing, with LM diagnosed by magnetic resonance imaging (MRI) or by cerebrospinal fluid (CSF) cytology, receiving an EGFR-TKI specifically for the treatment of LM. Therapies evaluated included the first-generation EGFR-TKIs erlotinib, gefitinib, and icotinib; the second-generation EGFR-TKIs afatinib and dacomitinib; and the third-generation EGFR-TKIs osimertinib, zorifertinib and furmonertinib (Supplemental Table S1).
2.2. Search strategy
We searched the literature across all databases of Medline (Medline, Medline Epub Ahead of Print, and In-Process & Other Non-Indexed Citations), Embase, Cochrane Central Register of Controlled Trials, Scopus, and the Web of Science Core Collection from January 1964 to December 2021. The complete search strategy is available in Appendix A. Data from published conference abstracts were also included in this meta-analysis. Any additional publications or data sourced by the authors beyond the initial search were integrated into the systematic review where relevant. The study protocol was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and prospectively registered on PROSPERO (ID: CRD42021292539) [[32], [33], [34]].
2.3. Data extraction and management
The Rayyan software was employed for abstract screening. Each abstract was screened by two independent reviewers (www.rayyan.ai). Three reviewers (AML, SMM, MD) resolved conflicts through internal discussions. Patient demographics and outcomes were extracted by the lead author; due to the added complexity of data in some publications screened, a subset (25 %) of abstracts was extracted by a second author (AML) to confirm accuracy. Additionally, Chinese, and Japanese reports with English titles that were included in the original search were translated and extracted by the lead author. Following extraction of all available data from the publications included in the meta-analysis, requests for missing data were sent to the original authors of each publication through two separate email prompts spaced seven days apart.
We categorized the patients included in published reports based on generations of EGFR-TKI into first-, second-, and third-generation EGFR-TKIs, as previously described [11]. Additionally, when patients received multiple EGFR-TKIs simultaneously (N = 4), the patient-line of therapy was assigned to the most recent-generation EGFR-TKI given.
EGFR mutations were classified in the following ways: by mutation location (i.e. exon 18, exon 19, exon 20, and exon 21), by presence or absence of previously described EGFR-TKI on-target resistance mutations (i.e. T790 M, C797X, L718Q, E884K, L747X, G724S, and K754E), and by type of mutation (i.e. insertion, deletion, and point mutation) [35]. We also compared EGFR classical driver mutations (i.e. exon 19 deletion and L858R), and atypical EGFR driver mutations (i.e. G719X, exon 19 insertion, exon 20 insertion, S768I, L861Q, SEPT14-EGFR fusion, and intron 17–28 duplication), which have been previously classified by Robichaux et al. [36].
2.4. Primary outcomes
The co-primary outcomes of interest were CNS-specific PFS, and OS. Both endpoints were calculated from the start of EGFR-TKI therapy for LM. Endpoints were defined as the following: CNS-specific PFS was determined based on CNS-specific disease progression (i.e., MRI or CSF analysis-proven progression), or death of the patient per the authors of the primary paper. Causes of progression were further classified as death of the patient from any cause while on treatment, progression of LM confirmed by MRI or CSF, progression of intraparenchymal brain metastases, and CNS progression of either LM or brain metastases that was not specified by the authors of the primary paper (Supplemental Table S2).
2.5. Quality (risk of bias) assessment
In order to evaluate the methodological quality of the studies in the meta-analysis, we employed a tool previously described adapted from the Newcastle-Ottawa scale, tailored for assessing case reports and case series, allowing to assess the selection and representativeness of cases, as well as the ascertainment of outcomes and exposure [37]. Each parameter evaluated earns one point if the study explicitly reports the relevant information. A study is considered to be of good quality (i.e., low risk of bias) if it meets all five criteria, of moderate quality if it meets four criteria, and of poor quality (high risk of bias) if it meets three or fewer criteria (Supplemental Fig. S1) [38,39].
2.6. Statistical analyses
We conducted one-stage meta-analyses, pooling individual patient data from all studies included. Patient characteristics were then compared between those who received third-generation EGFR-TKI and those who received first- and second-generation EGFR-TKIs with Fisher's Exact test and Pearson's X2. The hazard ratio (HR) served as the primary parameter of interest for OS and CNS-specific PFS. Univariate logistic regression models were utilized to ascertain the HR between the groups of interest along with its corresponding 95 % confidence interval (CI). Multi-level mixed-effects Cox proportional hazards regression models, integrating individual studies as a random effect, were applied.
Multivariate Cox proportional hazards regression models were employed to estimate adjusted CNS-specific PFS and OS (aPFS and aOS), incorporating a multi-level mixed-effects Cox proportional hazards regression model that accounted for individual studies as a random effect. All variables with P < 0.05 in univariate analysis were incorporated into the initial multivariate model. Backward stepwise selection was employed to eliminate variables deemed insignificant. The final model included all variables with P < 0.05. For aPFS, the initial model included sex, prospective versus retrospective studies, previous lines of therapy (0–2 versus 3 or more), previous EGFR-TKI use, concurrent extracranial metastasis, concurrent intracranial metastasis, ECOG status (0–2 versus 3–4), treatment with osimertinib, and treatment with afatinib. In the multivariate model for aPFS, sex and prior EGFR-TKI use were identified as the statistically significant variables. For aOS, the initial multivariate model included year of study (2018 or after versus before 2018), previous lines of therapy (0–2 versus 3 or more), previous EGFR-TKI use, concurrent extracranial metastasis, concurrent intracranial metastasis, ECOG status (0–2 versus 3–4), MRI-only diagnosis, CSF-only diagnosis, and treatment with afatinib. In the multivariate model, following the analysis, ECOG status and prior lines of therapy emerged as the statistically significant variables influencing adjusted aOS.
Correlation analyses between PFS and OS were conducted utilizing linear regression and Pearson's X2. In cases where performance status was indicated by the Karnofsky Performance Status score, it was converted to Eastern Cooperative Oncology Group (ECOG) status employing a conversion scale previously outlined in the literature [40].
Survival curves were depicted and assessed using the Kaplan-Meier method and the log-rank test, which were performed to compare the CNS-specific PFS and OS of osimertinib versus other EGFR-TKIs; afatinib versus other EGFR-TKIs; and presence versus absence of concurrent chemotherapy. Additionally, Kaplan-Meier survival curves and the log-rank test were conducted for subgroup analyses. These survival curves included analysis of CNS-specific PFS and OS on osimertinib compared to other EGFR-TKIs in patients having been treated with prior EGFR-TKIs and those without prior EGFR-TKI treatment; afatinib compared to other EGFR-TKIs in patients having been treated with prior EGFR-TKIs and those without prior EGFR-TKI treatment; and route of administration (intravenous versus intrathecal) of concurrent chemotherapy.
STATA v17 was employed for statistical analyses (StataCorp LLC, College Station, Texas, USA).
3. Results
3.1. Characteristics of included studies and patients
We initially identified 7780 articles potentially eligible for inclusion in the search. Following the initial screening process, which involved removing ineligible articles and supplementing with studies from authors’ files, a total of 128 publications were ultimately included in the meta-analysis (Appendix B and Appendix C). 106 of the publications were case reports [[14], [15], [16], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142]]. 13 of the publications were case series [[143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155]]. 7 of the publications were retrospective studies [13,[25], [26], [27], [28],156,157]. Finally, 2 of the publications were prospective studies [12,29]. This consisted of a total of 243 patients with EGFR-mutated lung cancer LM who received a total of 282 patient-lines of EGFR-TKI therapy for the treatment of LM (Fig. 1). Additionally, following preliminary screening of eligible articles and after contacting authors of all studies with potentially includible data, we were not able to extract data from 55 potentially eligible publications, including 15 prospective studies, 35 retrospective studies, and 5 case series or case reports (Appendix D and Appendix E).
Fig. 1.
Preferred Reporting Items for Systematic Reviews and Meta-Analyses diagram demonstrating search and inclusion of studies for meta-analysis. The flow diagram demonstrates the 20 145 records identified through the initial search of different databases. Repeated searches using the search strategy outlined in Appendix A were performed on Pubmed during the subsequent data extraction phase, identifying an additional 11 records that respected our inclusion criteria. Duplicate removal and further screening had identified 368 eligible articles. 128 articles comprising of 282 patient-lines of therapy were included for the final analysis.
Detailed characteristics of patients receiving third-generation EGFR-TKIs versus those receiving first- or second-generation EGFR-TKIs can be found in Table 1. A risk of bias assessment was conducted for all studies included in the meta-analysis, utilizing a 5-point scale (Supplemental Figure S1).
Table 1.
Individual patient characteristics.
| Variable | Entire Cohort No. (%) | Third Generation EGFR TKI cohort (%) | First and Second Generation EGFR TKI cohort (%) | P (Fisher's exact) | Pearson's χ2 |
|---|---|---|---|---|---|
| Patient-lines of therapy | 282 (100) | 125 (44.3) | 157 (55.7) | ||
| Study characteristics | |||||
| Geographic location | |||||
| North America | 40 (14.2) | 15 (12.0) | 25 (15.9) | 0.394 | 8.045, P = 0.154 |
| Europe | 10 (3.5) | 4 (3.2) | 6 (3.8) | 1 | |
| Asia | 222 (78.7) | 100 (80.0) | 122 (77.7) | 0.77 | |
| Australia | 10 (3.5) | 6 (4.8) | 4 (2.5) | 0.345 | |
| Year of study | |||||
| <2018 | 125 (44.3) | 10 (8.0) | 115 (73.2) | <0.0001 | |
| ≥2018 | 157 (55.7) | 115 (92.0) | 42 (26.8) | ||
| Sample size | |||||
| <5 | 188 (66.7) | 63 (50.4) | 125 (79.6) | <0.0001 | |
| ≥5 | 94 (33.3) | 62 (49.6) | 32 (20.4) | ||
| Risk of bias | |||||
| ≤3 | 113 (40.1) | 50 (40.0) | 63 (40.1) | 1 | |
| 4, 5 | 169 (59.9) | 75 (60.0) | 94 (59.9) | ||
| Study type | |||||
| Retrospective | 258 (91.5) | 112 (89.6) | 146 (93.0) | 0.512 | |
| Prospective | 24 (8.5) | 13 (10.4) | 11 (7.0) | ||
| Patient characteristics | |||||
| Sex | |||||
| Male | 112 (39.7) | 58 (46.4) | 54 (34.4) | 0.006 | |
| Female | 157 (55.7) | 54 (43.2) | 103 (65.6) | ||
| Unknown | 13 (4.6) | 13 (10.4) | 0 (0) | ||
| Age, years | |||||
| <60 | 135 (47.9) | 61 (48.8) | 74 (47.1) | 0.266 | |
| ≥60 | 134 (47.5) | 51 (40.8) | 83 (52.9) | ||
| Unknown | 13 (4.6) | 13 (10.4) | 0 (0) | ||
| Lines of therapy | |||||
| 0–2 | 124 (44.0) | 51 (40.8) | 73 (46.5) | 0.902 | |
| ≥3 | 145 (51.4) | 61 (48.8) | 84 (53.5) | ||
| Unknown | 13 (4.6) | 13 (10.4) | 0 (0) | ||
| Prior EGFR TKI | |||||
| No | 73 (25.9) | 23 (18.4) | 50 (31.8) | 0.014 | |
| Yes | 209 (74.1) | 102 (81.6) | 107 (68.2) | ||
| Concurrent extracranial metastases | |||||
| No | 58 (20.6) | 52 (41.6) | 33 (21.0) | 0.455 | |
| Yes | 190 (67.4) | 94 (75.2) | 96 (61.1) | ||
| Unknown | 34 (12.1) | 6 (4.8) | 28 (17.8) | ||
| Concurrent intracranial metastases | |||||
| No | 91 (32.3) | 46 (36.8) | 45 (28.7) | 0.152 | |
| Yes | 171 (60.6) | 70 (56.0) | 101 (64.3) | ||
| Unknown | 20 (7.1) | 9 (7.2) | 11 (7.0) | ||
| Location of LM | |||||
| Brain | 213 (75.5) | 89 (71.2) | 124 (79.0) | 0.345 | 0.908, P = 0.635 |
| Spinal cord | 6 (2.1) | 3 (2.4) | 3 (1.9) | 0.7 | |
| Both | 13 (4.6) | 7 (5.6) | 6 (3.8) | 0.403 | |
| Unknown | 51 (18.1) | 27 (21.6) | 24 (15.3) | ||
| ECOG status | |||||
| 0–2 | 78 (27.7) | 43 (34.4) | 35 (22.3) | 0.012 | |
| 3 and 4 | 70 (24.8) | 24 (19.2) | 46 (29.3) | ||
| Unknown | 134 (47.5) | 58 (46.4) | 76 (48.4) | ||
| Smoking status | |||||
| Never | 101 (35.8) | 30 (24.0) | 71 (45.2) | 0.05 | |
| Current/Former | 30 (10.6) | 15 (12.0) | 15 (9.6) | ||
| Unknown | 151 (53.5) | 80 (64.0) | 71 (45.2) | ||
| Cancer type | |||||
| EGFR-mutant lung adenocarcinoma | 278 (98.6) | 121 (96.8) | 157 (100.0) | 0.037 | |
| EGFR-mutant lung squamous cell carcinoma | 4 (1.4) | 4 (3.2) | 0 (0) | ||
| Mutational status | |||||
| Specific mutations of interest | |||||
| Exon 19 deletion | 133 (47.2) | 55 (44.0) | 78 (49.7) | 0.197 | |
| L858R point mutation | 109 (38.7) | 52 (41.6) | 57 (36.3) | 0.211 | |
| T790 M point mutation | 50 (17.7) | 39 (31.2) | 11 (8.7) | <0.0001 | |
| Mutational status by exon location | |||||
| Exon 18 mutation | 9 (3.2) | 3 (2.4) | 6 (3.8) | 1 | |
| Exon 19 mutation | 133 (47.2) | 54 (43.2) | 79 (50.3) | 0.377 | |
| Exon 20 mutation | 2 (0.7) | 2 (1.6) | 0 (0) | 0.083 | |
| Exon 21 mutation | 109 (38.7) | 51 (40.8) | 58 (36.9) | 0.31 | |
| Resistance mutations | 61 (21.6) | 40 (32.0) | 21 (13.4) | <0.0001 | |
| Mutational status by classical versus atypical | |||||
| Classical mutations | 237 (84.0) | 103 (82.4) | 134 (85.4) | 0.642 | |
| Atypical mutations | 28 (9.9) | 10 (8.0) | 18 (11.5) | ||
| Mutational status by classical versus atypical | |||||
| Deletion mutations | 133 (47.2) | 55 (44.0) | 78 (49.7) | 0.449 | 1.484, P = 0.476 |
| Insertion mutations | 3 (1.1) | 2 (1.6) | 1 (0.6) | 0.584 | |
| Point mutations | 121 (42.9) | 57 (45.6) | 64 (40.8) | 0.377 | |
| Diagnostic characteristics | |||||
| Method of LM diagnosis | |||||
| Magnetic resonance imaging only | 78 (27.7) | 37 (29.6) | 41 (26.1) | 1 | 3.859, P = 0.145 |
| Cerebrospinal fluid only | 73 (25.9) | 41 (32.8) | 32 (20.4) | 0.098 | |
| Both magnetic resonance imaging and cerebrospinal fluid | 105 (37.2) | 43 (34.4) | 62 (39.5) | 0.098 | |
| Unknown | 27 (9.6) | 4 (3.2) | 23 (14.6) | ||
| Time from primary cancer diagnosis to LM diagnosis, months | |||||
| <12 | 81 (28.7) | 25 (20.0) | 56 (35.7) | 0.136 | |
| ≥12 | 117 (41.5) | 49 (39.2) | 68 (43.3) | ||
| Unknown | 84 (29.8) | 51 (40.8) | 33 (21.0) | ||
| Chemotherapy simultaneously with EGFR TKI | |||||
| No | 210 (74.5) | 95 (76.0) | 115 (732) | 0.259 | |
| Yes | 48 (17.0) | 17 (13.6) | 31 (19.7) | ||
| Unknown | 24 (8.5) | 13 (10.4) | 11 (7.0) | ||
| Route of administration for chemotherapy concurrently to EGFR TKI | |||||
| Intravenous or oral only | 17 (6.0) | 7 (5.6) | 10 (6.4) | 0.547 | 4.693, P = 0.096 |
| Intrathecal only | 23 (8.2) | 5 (4.0) | 18 (11.5) | 0.227 | |
| Intrathecal and intravenous or oral | 8 (2.8) | 5 (4.0) | 3 (1.9) | 0.112 | |
| Radiotherapy for LM | |||||
| Yes | 154 (19.1) | 20 (16.0) | 34 (21.7) | 0.355 | |
| No | 201 (71.3) | 89 (71.2) | 112 (71.3) | ||
| Unknown | 24 (2.8) | 13 (10.4) | 11(7.0) | ||
| Radiotherapy for LM | |||||
| Stereotactic radiosurgery | 5 (1.8) | 3 (2.4) | 2 (1.3) | 0.369 | 1.581, P = 0.209 |
| Whole-brain radiotherapy | 45 (16.0) | 16 (12.8) | 29 (18.5) | 0.195 | |
| Whole-brain radiotherapy and stereotactic | 1 (0.4) | 1 (0.8) | 0 (0) | 0.392 | |
| Received brain radiotherapy, but unknown type | 3 (1.1) | 0 (0) | 3 (1.9) | ||
NOTE. Bold values indicate P < 0.05. Abbreviations: LM, leptomeningeal metastasis; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; ECOG, Eastern Cooperative Oncology Group.
Patients receiving any EGFR-TKI for the treatment of NSCLC LM experienced a CNS-specific median PFS (mPFS) of 9.1 months (Table 2) and a mOS of 14.5 months (Table 3). Among the 243 individual patients, accounting for 282 patient-lines of therapy, 120 lines of therapy consisted of first-generation EGFR-TKI, 37 consisted of second-generation EGFR-TKI, and 125 consisted of third-generation EGFR-TKI (Table 1). In the patient-lines of therapy where CNS-specific PFS was available (N = 272), we observed a significant correlation between CNS-specific PFS and OS (Pearson's R2 = 0.37, P < 0.0001) (Supplemental Fig. S2 A). This significant correlation was maintained when analyses were restricted to patients with documented death, or whose progression cases were not restricted to death (Supplemental Fig. S2 B-D and Supplemental Table S2).
Table 2.
CNS-specific progression-free survival rates associated with clinical variables.
| CNS-SPECIFIC PROGRESSION-FREE SURVIVAL | Patient lines of therapy | Median OS (months) | Univariable Hazard Ratio | Univariable 95 % CI | Univariable P value | Adjusted Hazard Ratio | Multivariable 95 % CI | Adjusted P Value |
|---|---|---|---|---|---|---|---|---|
| Entire Cohort | 272 | 9.1 | ||||||
| Study characteristics | ||||||||
| Geographic location | ||||||||
| America | 40 | 7.6 | 1.361 | 0.832–2.225 | 0.219 | |||
| Europe | 10 | 6 | 1.293 | 0.465–3.303 | 0.669 | |||
| Asia | 212 | 9.4 | 0.788 | 0.507–1.224 | 0.288 | |||
| Australia | 10 | 12 | 0.75 | 0.242–2.335 | 0.621 | |||
| Year of study | ||||||||
| <2018 | 125 | 8 | 0.691 | 0.473–1.008 | 0.055 | |||
| ≥2018 | 147 | 10 | ||||||
| Sample size | ||||||||
| <5 | 187 | 9.3 | 1.282 | 0.796–2.067 | 0.307 | |||
| ≥5 | 85 | 9.1 | ||||||
| Risk of bias | ||||||||
| ≤3 | 113 | 9.1 | 0.773 | 0.510–1.172 | 0.226 | |||
| 4, 5 | 159 | 9.2 | ||||||
| Study type | ||||||||
| Retrospective | 248 | 9.5 | 2.215 | 1.042–4.709 | 0.039 | |||
| Prospective | 24 | 6.5 | ||||||
| Patient characteristics | ||||||||
| Sex | ||||||||
| Female | 153 | 8 | 0.690 | 0.487–0.977 | 0.037 | 0.687 | 0.488–0.968 | 0.032 |
| Male | 106 | 10 | ||||||
| Age, years | ||||||||
| <60 | 131 | 9 | 0.781 | 0.549–1.109 | 0.167 | |||
| ≥60 | 128 | 9.3 | ||||||
| Lines of therapy | ||||||||
| 0–2 | 120 | 12 | 1.723 | 1.209–2.456 | 0.003 | |||
| ≥3 | 139 | 7.5 | ||||||
| Previous EGFR TKI | ||||||||
| No | 71 | 13 | 2.231 | 1.494–3.334 | <0.001 | 2.152 | 1.577–3.134 | <0.001 |
| Yes | 201 | 7.6 | ||||||
| Concurrent extracranial metastasis | ||||||||
| No | 56 | 12 | 1.583 | 1.050–2.386 | 0.028 | |||
| Yes | 182 | 8 | ||||||
| Concurrent intracranial metastasis | ||||||||
| No | 84 | 12 | 1.451 | 1.005–2.094 | 0.047 | |||
| Yes | 168 | 8 | ||||||
| Location of LM | ||||||||
| Brain | 206 | 9 | 1.162 | 0.591–2.284 | 0.663 | |||
| Spinal cord | 6 | 9 | 1.085 | 0.415–2.836 | 0.868 | |||
| Both | 10 | 11 | 0.717 | 0.292–1.763 | 0.496 | |||
| ECOG status | ||||||||
| 0–2 | 74 | 9.5 | 1.817 | 1.110–2.976 | 0.018 | |||
| 3,4 | 65 | 7 | ||||||
| Smoking status | ||||||||
| Never | 100 | 9.6 | 0.822 | 0.453–1.488 | 0.517 | |||
| Current/Former | 30 | 9.4 | ||||||
| Cancer type | ||||||||
| EGFR-mutant lung adenocarcinoma | 268 | 9.1 | 0.663 | 0.147–2.988 | 0.592 | |||
| EGFR-mutant lung squamous cell carcinoma | 4 | 12 | ||||||
| Mutational status | ||||||||
| Specific mutations of interest | ||||||||
| Exon 19 deletion | 128 | 8 | 1.085 | 0.772–1.525 | 0.638 | |||
| L858R point mutation | 103 | 8 | 1.216 | 0.862–1.715 | 0.265 | |||
| T790 M point mutation | 50 | 8 | 1.252 | 0.786–1.995 | 0.344 | |||
| Mutational status by exons | ||||||||
| Exon 18 mutation | 10 | N/A | 0.368 | 0.125–1.082 | 0.069 | |||
| Exon 19 mutation | 129 | 9 | 1.01 | 0.712–1.432 | 0.995 | |||
| Exon 20 mutation | 3 | 8 | 0.95 | 0.205–4.408 | 0.948 | |||
| Exon 21 mutation | 106 | 8 | 1.153 | 0.811–1.639 | 0.428 | |||
| Resistance mutations | 61 | 8 | 1.113 | 0.721–1.717 | 0.63 | |||
| Mutational status by classical versus atypical | ||||||||
| Classical mutations | 228 | 9 | 0.668 | 0.338–1.318 | 0.244 | |||
| Atypical mutations | 19 | 10.3 | ||||||
| Mutational status by mechanism of mutation | ||||||||
| Deletion mutations | 128 | 9 | 0.987 | 0.696–1.400 | 0.942 | |||
| Insertion mutations | 3 | 6 | 2.144 | 0.583–7.889 | 0.251 | |||
| Point mutations | 115 | 9 | 0.982 | 0.693–1.393 | 0.920 | |||
| Diagnostic characteristics | ||||||||
| Method of LM diagnosis | ||||||||
| Magnetic resonance imaging only | 82 | 11 | 0.82 | 0.5786–1.163 | 0.266 | |||
| Cerebrospinal fluid only | 68 | 9.9 | 1.115 | 0.7800–1.594 | 0.55 | |||
| Both magnetic resonance imaging and cerebrospinal fluid | 107 | 9 | 1.039 | 0.750–1.440 | 0.818 | |||
| Time from primary diagnosis to LM diagnosis, months | ||||||||
| <12 months | 81 | 10 | 1.108 | 0.764–1.606 | 0.589 | |||
| ≥12 months | 116 | 9.6 | ||||||
| Treatments | ||||||||
| Type of therapy | ||||||||
| 1st generation EGFR TKI | 119 | 9 | 1.141 | 0.795–1.638 | 0.474 | |||
| 2nd generation EGFR TKI | 37 | 5.6 | 1.922 | 1.160–3.186 | 0.01 | |||
| 3rd generation EGFR TKI | 115 | 11 | 0.63 | 0.432–0.919 | 0.016 | |||
| Regimens including each of the following therapies | ||||||||
| 1st generation EGFR TKI | ||||||||
| Erlotinib | 86 | 9 | 1.179 | 0.804–1.729 | 0.399 | |||
| Gefitinib | 32 | 10 | 1.058 | 0.649–1.726 | 0.821 | |||
| Icotinib | 4 | 12 | 0.383 | 0.0799–1.832 | 0.229 | |||
| 2nd generation EGFR TKI | ||||||||
| Afatinib | 33 | 5.6 | 2.000 | 1.176–3.401 | 0.01 | |||
| Dacomitinib | 4 | 12 | 1.257 | 0.283–5.583 | 0.764 | |||
| 3rd generation EGFR TKI | ||||||||
| Osimertinib | 112 | 11 | 0.620 | 0.424–0.908 | 0.014 | |||
| Zorifertinib | 2 | 3 | 3.800 | 0.488–29.584 | 0.202 | |||
| Furmonertinib | 1 | N/A | N/A | N/A | N/A | |||
| EGFR TKI dosing regimens | ||||||||
| Osimertinib 80 mg daily | 84 | 12 | 1.430 | 0.666–3.068 | 0.359 | |||
| Osimertinib 160 mg daily | 14 | 9 | ||||||
| Chemotherapy for LM simultaneously to EGFR TKI | ||||||||
| No | 201 | 9.1 | 0.699 | 0.451–1.085 | 0.111 | |||
| Yes | 47 | 10 | ||||||
| Route of administration for chemotherapy concurrently to EGFR TKI | ||||||||
| Intravenous or oral only | 17 | 9.6 | 1.35 | 0.624–2.921 | 0.446 | |||
| Intrathecal only | 24 | 12 | 0.614 | 0.272–1.382 | 0.239 | |||
| Intrathecal and intravenous or oral | 7 | 9.7 | 1.287 | 0.441–3.751 | 0.644 | |||
| Concurrent bevacizumab for LM | ||||||||
| No | 248 | 9 | 0.706 | 0.379–1.317 | 0.274 | |||
| Yes | 24 | 11.9 | ||||||
| Radiotherapy for LM | ||||||||
| No | 195 | 9.3 | 0.878 | 0.578–1.334 | 0.543 | |||
| Yes | 53 | 10 | ||||||
| Type of radiotherapy for LM | ||||||||
| Stereotactic radiosurgery | 4 | 6 | 1.442 | 0.337–6.169 | 0.622 | |||
| Whole-brain radiotherapy | 45 | 10 | 0.543 | 0.16–1.840 | 0.327 | |||
| Both stereotactic and whole-brain radiotherapies | 1 | N/A | N/A | N/A | N/A | |||
NOTE. Univariate and multivariate hazard ratios, 95 % CIs, and P-values calculated with a multilevel mixed-effects logistic regression model with article as the random-effects variable. Bold values indicate P < 0.05. Abbreviations: CI, confidence interval; CNS, central nervous system; mPFS, median progression-free survival; LM, leptomeningeal metastasis; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; CSF, cerebrospinal fluid; MRI, magnetic resonance imaging; ECOG, Eastern Cooperative Oncology Group.
Table 3.
Overall survival rates associated with clinical variables.
| OVERALL SURVIVAL | Patient lines of therapy | Median OS (months) | Univariable Hazard Ratio | Univariable 95 % CI | Univariable P value | Adjusted Hazard Ratio | Multivariable 95 % CI | Adjusted P Value |
|---|---|---|---|---|---|---|---|---|
| Entire Cohort | 280 | 14.5 | ||||||
| Study characteristics | ||||||||
| Geographic location | ||||||||
| America | 40 | 15.3 | 1.024 | 0.528–2.112 | 0.946 | |||
| Europe | 10 | 11.5 | 2.033 | 0.622–6.642 | 0.24 | |||
| Asia | 220 | 14.5 | 0.898 | 0.492–1.642 | 0.727 | |||
| Australia | 10 | 21 | 0.62 | 0.130–2.959 | 0.548 | |||
| Year of study | ||||||||
| <2018 | 125 | 11.5 | 0.527 | 0.325–0.854 | 0.009 | |||
| ≥2018 | 155 | 18 | ||||||
| Sample size | ||||||||
| <5 | 186 | 15 | 1.134 | 0.588–2.109 | 0.74 | |||
| ≥5 | 94 | 14.5 | ||||||
| Risk of bias | ||||||||
| ≤3 | 111 | 14 | 0.677 | 0.400–1.148 | 0.148 | |||
| 4, 5 | 169 | 15 | ||||||
| Study type | ||||||||
| Retrospective | 256 | 15.3 | 2.556 | 0.866–7.544 | 0.089 | |||
| Prospective | 24 | 7.5 | ||||||
| Patient characteristics | ||||||||
| Sex | ||||||||
| Female | 157 | 13.67 | 0.709 | 0.478–1.053 | 0.088 | |||
| Male | 110 | 19 | ||||||
| Age, years | ||||||||
| <60 | 134 | 14.5 | 0.817 | 0.549–1.216 | 0.320 | |||
| ≥60 | 133 | 15.8 | ||||||
| Lines of therapy | ||||||||
| 0–2 | 122 | 20 | 2.371 | 1.598–3.519 | <0.001 | 2.413 | 1.551–3.755 | <0.001 |
| ≥3 | 145 | 11 | ||||||
| Previous EGFR TKI | ||||||||
| No | 73 | 21 | 2.412 | 1.533–3.794 | <0.001 | |||
| Yes | 207 | 13.67 | ||||||
| Concurrent extracranial metastasis | ||||||||
| No | 57 | 34 | 1.958 | 1.120–3.425 | 0.018 | |||
| Yes | 189 | 14.5 | ||||||
| Concurrent intracranial metastasis | ||||||||
| No | 90 | 21.5 | 1.965 | 1.254–3.077 | 0.003 | |||
| Yes | 170 | 13.7 | ||||||
| Location of LM | ||||||||
| Brain | 212 | 14.5 | 0.671 | 0.329–1.369 | 0.273 | |||
| Spinal cord | 6 | 14.5 | 1.123 | 0.421–2.999 | 0.817 | |||
| Both | 13 | 11 | 2.229 | 0.788–6.304 | 0.131 | |||
| ECOG status | ||||||||
| 0–2 | 78 | 16.5 | 2.486 | 1.507–4.100 | <0.001 | 2.000 | 1.288–3.105 | 0.002 |
| 3,4 | 69 | 10 | ||||||
| Smoking status | ||||||||
| Never | 101 | 15 | 0.582 | 0.225–1.506 | 0.264 | |||
| Current/Former | 29 | N/A | ||||||
| Cancer type | ||||||||
| EGFR-mutant lung adenocarcinoma | 276 | 14.5 | 0.494 | 0.0574–4.244 | 0.520 | |||
| EGFR-mutant lung squamous cell carcinoma | 4 | N/A | ||||||
| Mutational status | ||||||||
| Specific mutations of interest | ||||||||
| Exon 19 deletion | 131 | 15.1 | 0.937 | 0.639–1.373 | 0.737 | |||
| L858R point mutation | 109 | 14 | 1.310 | 0.890–1.928 | 0.172 | |||
| T790 M point mutation | 50 | 13.7 | 0.522 | 0.696–2.042 | 0.522 | |||
| Mutational status by exons | ||||||||
| Exon 18 mutation | 10 | N/A | 0.474 | 0.130–1.725 | 0.257 | |||
| Exon 19 mutation | 132 | 16.25 | 0.906 | 0.610–1.345 | 0.624 | |||
| Exon 20 mutation | 3 | 13.5 | 0.518 | 0.0566–4.735 | 0.560 | |||
| Exon 21 mutation | 111 | 14 | 1.179 | 0.790–1.758 | 0.420 | |||
| Acquired mutation | 61 | 12 | 1.22 | 0.740–2.012 | 0.436 | |||
| Mutational status by classical versus atypical | ||||||||
| Classical mutations | 235 | 14 | 0.872 | 0.393–1.936 | 0.736 | |||
| Atypical mutations | 20 | 16.5 | ||||||
| Mutational status by mechanism of mutation | ||||||||
| Deletion mutations | 131 | 15.1 | 0.872 | 0.587–1.295 | 0.497 | |||
| Insertion mutations | 3 | 13.5 | 2.006 | 0.291–13.878 | 0.480 | |||
| Point mutations | 121 | 14 | 1.132 | 0.762–1.681 | 0.540 | |||
| Diagnostic characteristics | ||||||||
| Method of LM diagnosis | ||||||||
| Magnetic resonance imaging only | 78 | 17.3 | 0.626 | 0.3935–0.9973 | 0.049 | |||
| Cerebrospinal fluid only | 73 | 13.6 | 1.724 | 1.010–2.942 | 0.046 | |||
| Both magnetic resonance imaging and cerebrospinal fluid | 107 | 17 | 1.058 | 0.657–1.703 | 0.817 | |||
| Time from primary diagnosis to LM diagnosis, months | ||||||||
| <12 months | 80 | 15.1 | 0.974 | 0.566–1.679 | 0.925 | |||
| ≥12 months | 116 | 16.25 | ||||||
| Treatments | ||||||||
| Type of therapy | ||||||||
| 1st generation EGFR TKI | 118 | 14 | 1.017 | 0.657–1.574 | 0.94 | |||
| 2nd generation EGFR TKI | 37 | 9.2 | 2.154 | 1.105–4.199 | 0.024 | |||
| 3rd generation EGFR TKI | 124 | 17 | 0.699 | 0.443–1.103 | 0.124 | |||
| Regimens including each of the following therapies | ||||||||
| 1st generation EGFR TKI | ||||||||
| Erlotinib | 85 | 12 | 1.170 | 0.731–1.871 | 0.513 | |||
| Gefitinib | 32 | 15.8 | 0.894 | 0.507–1.577 | 0.699 | |||
| Icotinib | 4 | N/A | 0.227 | 0.0229–2.247 | 0.205 | |||
| 2nd generation EGFR TKI | ||||||||
| Afatinib | 33 | 9.2 | 2.224 | 1.109–4.458 | 0.024 | |||
| Dacomitinib | 4 | 6 | 1.318 | 0.155–11.237 | 0.801 | |||
| 3rd generation EGFR TKI | ||||||||
| Osimertinib | 121 | 16.8 | 0.720 | 0.455–1.141 | 0.162 | |||
| Zorifertinib | 2 | 24 | 0.629 | 0.0750–5.282 | 0.67 | |||
| Furmonertinib | 1 | N/A | N/A | N/A | N/A | |||
| EGFR TKI dosing regimens | ||||||||
| Osimertinib 80 mg daily | 94 | 17.3 | 1.952 | 0.859–4.436 | 0.110 | |||
| Osimertinib 160 mg daily | 13 | 11 | ||||||
| Chemotherapy for LM simultaneously to EGFR TKI | ||||||||
| No | 208 | 15.3 | 0.733 | 0.411–1.307 | 0.292 | |||
| Yes | 48 | 16.25 | ||||||
| Route of administration for chemotherapy concurrently to EGFR TKI | ||||||||
| Intravenous or oral only | 17 | 16.25 | 1.027 | 0.431–2.446 | 0.952 | |||
| Intrathecal only | 24 | 51.8 | 0.622 | 0.249–1.554 | 0.309 | |||
| Intrathecal and intravenous or oral | 8 | 12.9 | 1.905 | 0.691–5.256 | 0.213 | |||
| Concurrent bevacizumab for LM | ||||||||
| No | 251 | 14 | 0.623 | 0.327–1.188 | 0.151 | |||
| Yes | 29 | 18 | ||||||
| Radiotherapy for LM | ||||||||
| No | 203 | 15.3 | 0.738 | 0.449–1.212 | 0.230 | |||
| Yes | 53 | 17 | ||||||
| Type of radiotherapy for LM | ||||||||
| Stereotactic radiosurgery | 5 | 14.5 | 2.392 | 0.585–9.773 | 0.225 | |||
| Whole-brain radiotherapy | 44 | 21 | 0.449 | 0.131–1.543 | 0.204 | |||
| Whole-brain radiotherapy and spinal radiotherapy | 1 | N/A | N/A | N/A | N/A | |||
NOTE. Univariate and multivariate hazard ratios, 95 % CIs, and P-values calculated with a multilevel mixed-effects logistic regression model with article as the random-effects variable. Bold values indicate P < 0.05. Abbreviations: CI, confidence interval; mOS, median overall survival; LM, leptomeningeal metastasis; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; CSF, cerebrospinal fluid; MRI, magnetic resonance imaging; ECOG, Eastern Cooperative Oncology Group; IV.
3.2. Variables associated with PFS and OS in EGFR-mutated lung cancer LM patients
In univariate analyses, the following variables were associated with shortened CNS-specific PFS and OS: patients who received 3 or more total lines of therapy since initial diagnosis of NSCLC (PFS: HR = 1.72, 95 % CI: 1.21–2.46, P = 0.003; OS: HR = 2.37, 95 % CI: 1.60–3.52, P < 0.001); prior treatment with EGFR-TKIs (PFS: HR = 2.23, 95 % CI: 1.49–3.33, P < 0.001; OS: HR = 2.41, 95 % CI: 1.53–3.79, P < 0.001); presence of concurrent extracranial metastases (PFS: HR = 1.58, 95 % CI: 1.05–2.39, P = 0.028; OS: HR = 1.96, 95 % CI: 1.20–3.43, P = 0.018), presence of concurrent intracranial metastases (PFS: HR = 1.45, 95 % CI: 1.01–2.09, P = 0.047; OS: HR = 1.97, 95 % CI: 1.25–3.08, P = 0.003), and ECOG status of 3–4 (PFS: HR = 1.82, 95 % CI: 1.11–2.99, P = 0.018; OS: HR = 2.49, 95 % CI: 1.51–4.10, P < 0.001) (Tables 2 and 3).
Male sex was associated with prolonged CNS-specific PFS (HR = 0.69, 95%CI: 0.49–0.98, P = 0.037). Patients derived from reports published after 2018 (HR = 0.53, 95%CI: 0.33–0.85, P = 0.009) and having received a diagnosis of LM by MRI-only (HR = 0.63, 95%CI: 0.39–0.997, P = 0.049) experienced prolonged OS. Diagnosis of LM by CSF-only analysis was associated with shortened OS (HR = 1.72, 95%CI: 1.01–2.94, P = 0.046) (Tables 2 and 3).
In multivariate analysis, male sex (HR = 0.69, 95 % CI: 0.49–0.97, P = 0.032) was independently associated with significantly prolonged CNS-specific PFS (Table 2). Total lines of therapy received (3 or more) (HR = 2.41, 95 % CI: 1.55–3.76, P < 0.001) and ECOG status of 3–4 (HR = 2.00, 95 % CI: 1.29–3.11, P = 0.002) remained significantly associated with shortened OS (Table 3). Multivariate analyses also demonstrated that prior EGFR-TKI use (HR = 2.15, 95 % CI: 1.58–3.13, P < 0.001) was associated with significantly shortened OS.
3.3. Osimertinib is associated with improved outcomes compared to alternative EGFR-TKIs, particularly in EGFR-TKI pre-treated patients
Osimertinib treatment was associated with significantly prolonged CNS-specific PFS compared to all previous-generation EGFR-TKIs and showed a non-significant trend towards prolonged OS (PFS: HR = 0.62, 95 % CI: 0.42–0.91, P = 0.014; OS: HR = 0.72, 95 % CI: 0.46–1.14, P = 0.16) (Log rank: P = 0.014 and P = 0.185, respectively) (Tables 2 and 3) (Fig. 2 A-B). CNS-specific PFS and OS were both significantly prolonged when separately comparing all three generations of EGFR-TKIs (Log rank: P = 0.004 and P = 0.043, respectively) (Fig. 2 C-D). Osimertinib was not associated with CNS-specific PFS, or prolonged OS when compared to other EGFR-TKI in LM patients who did not receive prior EGFR-TKIs (Log rank: P = 0.304 and P = 0.744, respectively) (Supplemental Fig. S3 A-B). These findings were similarly observed when comparing all three generations of EGFR-TKIs for CNS-specific PFS and OS (Log rank: P = 0.540 and P = 0.872, respectively) (Supplemental Fig. S3 C-D). However, osimertinib was associated with significantly prolonged CNS-specific PFS (Log rank P = 0.003) and OS (Log rank P = 0.008) in LM patients who were previously treated with EGFR-TKI therapy (Fig. 3 A-B). Significantly different CNS-specific PFS and OS were also observed when comparing all three generations of EGFR-TKIs (Log rank P = 0.0001 and P = 0.0009, respectively) (Fig. 3 C-D).
Fig. 2.
Comparison of osimertinib to alternative EGFR-TKI. (A) CNS-specific PFS and (B) OS of patients who received osimertinib versus other EGFR-TKI. (C) CNS-specific PFS and (D) OS of patients who received first-generation versus second-generation versus third-generation EGFR-TKIs. NOTE. P-values calculated with Log-Rank test. Abbreviations: LM, leptomeningeal metastasis; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; CNS, central nervous system; PFS, progression free survival; OS, overall survival.
Fig. 3.
Comparison of osimertinib to alternative EGFR-TKI in patients who received prior EGFR-TKI. Subgroup analysis of (A) CNS-specific PFS and (B) OS of osimertinib versus other EGFR-TKIs among patients who have previously received EGFR-TKIs. Subgroup analysis of (C) CNS-specific PFS and (D) OS of first-generation versus second-generation versus third-generation EGFR-TKIs among patients who have previously received EGFR-TKIs. NOTE. P-values calculated with Log-Rank test. Abbreviations: LM, leptomeningeal metastasis; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; CNS, central nervous system; PFS, progression free survival; OS, overall survival.
In contrast to osimertinib, patients treated with afatinib experienced shortened CNS-specific PFS and OS compared to all other EGFR-TKIs (PFS: HR = 2.00, 95 % CI: 1.18–3.40, P = 0.01; OS: HR = 2.22, 95 % CI: 1.11–4.46, P = 0.024) (Log rank P = 0.003 and P = 0.045, respectively) (Tables 2 and 3) (Supplemental Fig. S4 A-B). Afatinib treatment demonstrated similar CNS-specific PFS and OS versus other EGFR-TKIs when patients had no prior EGFR-TKI treatment (Log rank P = 0.898 and P = 0.631, respectively), but was associated with shortened CNS-specific PFS and OS in NSCLC LM patients having received prior EGFR-TKI therapy (Log rank P < 0.0001 and P = 0.0001, respectively) (Supplemental Fig. S4 C–F).
3.4. Osimertinib is associated with improved outcomes compared to alternative EGFR-TKIs in patients with EGFR exon 19 deletion mutations, but not EGFR L858R point mutations
Osimertinib was associated with significantly prolonged CNS-specific PFS and OS compared to other EGFR-TKIs in LM patients with EGFR exon 19 deletion mutation (Log rank P = 0.002 and P = 0.0252, respectively) (Supplemental Fig. S5 A-B). In contrast, osimertinib demonstrated no significant difference in CNS-specific PFS and OS compared to other EGFR-TKIs in LM patients with EGFR L858R point mutation (Log rank P = 0.387 and P = 0.609, respectively) (Supplemental Fig. S5 C-D). Moreover, osimertinib demonstrated prolonged CNS-specific PFS, but not OS, in LM patients with the acquired EGFR T790 M point mutation (Log rank P = 0.003 and P = 0.380, respectively) (Supplemental Fig. S5 E-F).
3.5. Concurrent non-EGFR-TKI treatments are not associated with improved outcomes in EGFR-mutated NSCLC LM patients
Concurrent administration of chemotherapy, administered either intrathecally, intravenously, or orally, alongside EGFR-TKI therapy was not associated with differential CNS-specific PFS or OS (PFS: HR = 0.70, 95 % CI: 0.45–1.09, P = 0.11; OS: HR = 0.73, 95 % CI: 0.41–1.31, P = 0.29) (Log rank P = 0.091 and P = 0.404, respectively) (Tables 2 and 3) (Supplemental Fig. S6 A-D). Similarly, concurrent treatment with bevacizumab, as systemic therapy for EGFR-mutated NSCLC LM patients, was not associated with differential outcomes (PFS: HR = 0.71, 95 % CI: 0.38–1.32, P = 0.27; OS: HR = 0.62, 95 % CI: 0.33–1.19, P = 0.15) (Tables 2 and 3). Concurrent radiotherapy, either as WBRT or stereotactic radiosurgery, was also not associated with differential OS or CNS-specific PFS (PFS: HR = 0.88, 95 % CI: 0.58–1.33, P = 0.54; OS: HR = 0.74, 95 % CI: 0.45–1.21, P = 0.23) (Tables 2 and 3).
3.6. Quality assessment
This meta-analysis primarily included patients reported in retrospective studies, potentially introducing higher bias compared to those identified in prospective studies. Within our cohort, a significant difference in CNS-specific PFS was observed between patients identified from retrospective and prospective studies, while no difference was noted in OS (PFS: HR = 2.22, 95 % CI: 1.042–4.71, P = 0.039; OS: HR = 2.56, 95 % CI: 0.87–7.54, P = 0.089) (Tables 2 and 3). Moreover, studies with a high risk of bias (3 or less) showed no significant difference in CNS-specific PFS and OS compared to studies with a low risk of bias (4–5) (PFS: HR = 0.77, 95 % CI: 0.51–1.71, P = 0.23; OS: HR = 0.68, 95 % CI: 0.40–1.15, P = 0.15) (Tables 2 and 3).
4. Discussion
EGFR-TKIs have been used as standard of care for EGFR-mutated lung cancer patients with LM in the absence of published randomized controlled trial data for this patient population. We present the largest meta-analysis of EGFR-TKI-treated NSCLC LM patients to date, comprising 243 patients who received a total of 282 total lines of therapy extracted from 128 individual studies.
Third-generation EGFR-TKIs, notably osimertinib, have been developed with the goal of overcoming the on-target T790 M gatekeeper mutation associated with resistance to first- and second-generation EGFR-TKIs [158]. Osimertinib demonstrated a survival benefit compared to conventional chemotherapy when used as salvage therapy in EGFR T790M-positive NSCLC patients who had progressed on prior EGFR-TKI in the AURA3 trial [18]. The FLAURA trial subsequently demonstrated superior survival outcomes for osimertinib compared to first-generation gefitinib or erlotinib when used as first-line therapy for EGFR-mutated advanced NSCLC [20,21].
Several prospective single-armed studies have demonstrated the potential clinical benefit of osimertinib in NSCLC patients with LM. The single-arm BLOOM study evaluated the efficacy of osimertinib in 41 EGFR-mutated NSCLC patients with LM that had progressed on a previous EGFR-TKI therapy. The patients in the study treated with osimertinib exhibited a mPFS of 8.6 months and a mOS of 11.0 months [159]. Furthermore, Park et al. demonstrated similar results in a phase II single-arm prospective study in lung cancer LM patients treated with osimertinib, with a mOS of 13.3 months and mPFS of 8.0 months [160]. Furthermore, a meta-analysis of the AURA trials similarly revealed mOS of 18.8 months and mPFS of 11.1 in osimertinib-treated patients with stable EGFR T790M-mutated NSCLC LM [31]. As we were unable to extract individual patient data from these prospective studies, they were not included in our dataset (Appendix C).
In our dataset, patients treated with EGFR-TKIs of all generations exhibited a mOS of 14.5 months, similar to results reported in these previous studies [[159], [160], [161], [162]]. We observed a significantly prolonged CNS-specific PFS and a non-significant trend towards a prolonged OS with osimertinib compared to all other EGFR-TKIs in our entire cohort. However, we observe that these differences are strongly dependent upon whether patients received prior EGFR-TKI therapy. Patients whose tumors were refractory to EGFR-TKIs derived significant benefit from osimertinib, while patients who were not previously treated with EGFR-TKI had similar outcomes regardless of whichever EGFR-TKI was used. This suggests that the survival benefit of osimertinib in our cohort is largely due to its ability to overcome resistance to earlier-generation EGFR-TKIs, rather than its reported enhanced CNS activity. In support of this finding, we describe significantly prolonged CNS-specific PFS for osimertinib compared to other EGFR-TKIs in LM patients with EGFR T790 M point mutations.
Furthermore, we observe that afatinib is associated with worse outcomes compared to other EGFR-TKIs. This finding is driven by patients who were previously treated with prior-generation EGFR-TKIs and that afatinib cannot overcome resistance to such EGFR-TKIs. Indeed, afatinib has been previously shown in the LUX-Lung 1 trial to confer no survival benefit compared to placebo in NSCLC patients who were pre-treated with first-generation EGFR-TKIs [163]. Thus, this lack of survival benefit is likely driven by the inability of afatinib to overcome resistance mechanisms to first-generation EGFR-TKIs, such as EGFR T790 M point mutations [164].
Our findings corroborate the results of other smaller cohort studies by Hong et al. and Tamura et al. who similarly did not observe an OS benefit with osimertinib compared to first- and second-generation EGFR-TKIs in patients with LM [165,166]. Together, this suggests that, in the context of LM, the survival benefit associated with osimertinib is limited to its ability to overcome resistance to earlier-generation EGFR-TKI, and not due to increased therapeutic activity in LM.
In preclinical models, osimertinib has demonstrated superior CNS activity compared to other EGFR-TKIs, as demonstrated by increased blood-brain barrier penetration [22,167]. These findings have been congruent with subgroup analyses of both the AURA3 and FLAURA trials; EGFR-mutated NSCLC patients with brain metastases similarly exhibited superior response rates and PFS for osimertinib employed in first- and second-line settings [23,168]. However, these lesions largely represent parenchymal brain metastases that were stable and/or locally treated at the time of patient enrollment. The data from these clinical trials contrasts substantially with the patients included in our dataset. Indeed, leptomeningeal metastases differ from parenchymal brain metastases in several ways. Notably, the blood-brain and blood-CSF barriers are distinct entities with both unique anatomical structures and expression profiles of efflux transporters, cellular permeabilities and interactions with cancer cells [1,2,169,170]. Moreover, the intrinsic biology underlying organotropism to the brain parenchyma and leptomeninges are divergent [2,171]. While dramatic differences in the efficacy of anti-cancer therapies between parenchymal and leptomeningeal metastases have not been observed in current literature, there is a lack of randomized prospective studies properly comparing treatment efficacies in parenchymal and leptomeningeal metastases.
The divergent biology between parenchymal brain metastases and LM can be further highlighted by their differing effective treatment modalities. Parenchymal brain metastases are amenable to local treatments, such as stereotactic or WBRT, which have demonstrated efficacy in parenchymal brain metastases but not LM [172]. In fact, although treatment guidelines for LM include WBRT, there is inconsistent evidence as to whether RT truly provides a significant survival benefit for patients with LM [8,173]. Retrospective studies evaluating the use of radiotherapy for LM in NSCLC patients did not identify a survival benefit, which is consistent with our findings [174,175]. Interestingly, specialized techniques, such as craniospinal irradiation, treating the entire CSF compartment is a treatment modality that has recently demonstrated clinical benefit in a phase II randomized controlled trial of LM patients [176,177].
A growing body of literature has described superior clinical outcomes with osimertinib compared to other EGFR-TKIs in NSCLC patients with EGFR exon 19 deletion mutations and CNS involvement, including LM [166,178]. Meanwhile, these clinical benefits were limited in patients with EGFR L858R point mutations [166,178,179]. Our findings are consistent with these findings, as we similarly describe prolonged survival for osimertinib compared to other EGFR-TKIs in LM patients with EGFR exon 19 deletion mutations, which was not observed in LM patients with EGFR L858R point mutations. These findings further highlight the role of identifying and classifying different EGFR mutational statuses to predict efficacy of specific EGFR-TKIs.
Several studies have attempted to combine additional therapeutic agents with EGFR-TKI for EGFR-mutated NSCLC. The addition of bevacizumab to EGFR-TKIs did not improve OS, data consistent with the results we report herein [180]. Early data from the FLAURA2 study demonstrated that chemotherapy, in addition to osimertinib, is superior to osimertinib alone for advanced NSCLC [181]. However, the findings described herein are not consistent with this observation. We hypothesize that this is because patients with CNS metastases do not derive much benefit from conventional chemotherapy due to the unique anatomy and microenvironment of the blood-brain and blood-CSF barriers that limit chemotherapy penetrance [2].
There are several novel EGFR-TKIs in development which may ultimately impact the care of patients with LM. Zorifertinib is a promising novel third-generation EGFR-TKI, which has shown excellent CNS penetration and antitumor activity in murine models [82,182,183]. An early phase trial conducted by Ahn et al. showed that zorifertinib exhibits CNS penetration in EGFR-mutated NSCLC patients with CNS metastases or LM [184]. Early results from a phase III trial have shown significantly superior mPFS and objective response rates for zorifertinib compared to first-generation EGFR-TKIs in EGFR-mutated NSCLC patients with CNS metastases [185]. In the osimertinib refractory setting, the EGFR-TKI lazertinib and the bispecific EGFR-MET antibody amivantamab has demonstrated encouraging data thus far. Lazertinib is an irreversible third-generation EGFR-TKI with high selectivity for primary and EGFR T790 M mutation [186]. In murine models, lazertinib demonstrated excellent CNS penetration and comparable anti-tumor efficacy to osimertinib, along with decreased toxicity [187]. In the LASER301 phase III clinical trial, lazertinib demonstrated prolonged mPFS compared to gefitinib as first-line treatment in EGFR-mutated NSCLC patients both with and without CNS metastases [188]. Amivantamab is a human bispecific antibody targeting EGFR exon 20 insertions [189]. The PAPILLON phase III clinical trial compared amivantamab plus chemotherapy versus chemotherapy alone in advanced NSCLC patients with primary EGFR exon 20 insertion mutations [190]. Amivantamab plus chemotherapy resulted in prolonged mPFS compared to chemotherapy alone in NSCLC patients both with and without brain metastases. Further investigations comparing the survival benefits of these novel therapies, osimertinib and previous-generation EGFR-TKIs among NSCLC patients with LM are warranted.
Several limitations are associated with our study. Firstly, many patients included were derived from case reports and retrospective case series, potentially introducing selection biases that could inflate survival estimates beyond real-world expectations. Regarding publications from which data couldn't be collected, the challenge lay in the presentation of mOS and mPFS for entire patient cohorts, making it impossible to obtain individualized patient data for analysis. Despite repeated requests to the authors for individualized patient data, no responses were received, further limiting data availability for analysis. Another potential source of methodological bias is the restriction of our database search to English titles and abstracts. Given the higher incidence of EGFR-mutated NSCLC in East Asian countries such as China, South Korea, Japan, and others, studies published in languages other than English from these regions might have been overlooked in our meta-analysis [191]. This could potentially lead to an incomplete representation of available data and affect the generalizability of our findings. While efforts were made to mitigate these biases, including quality assessment, second reviewer extraction for a subset of abstracts, and translation of non-English articles retrieved in our database search, a randomized controlled trial would offer the most suitable approach to directly compare the efficacy of EGFR-TKIs in treating LM. However, as of now, no such studies have been conducted.
This meta-analysis describes the largest cohort of EGFR-mutated NSCLC LM patients treated with EGFR-TKIs compiled to date. Our findings suggest that survival benefits are comparable for first-, second-, and third-generation EGFR-TKIs in the treatment of EGFR-TKI naïve EGFR-mutated NSCLC LM patients. These findings may potentially have clinical implications especially in resource-limited settings, wherein osimertinib may not be available or is cost-ineffective compared to off-patent medications. However, osimertinib demonstrates a survival benefit in LM patients harboring primary EGFR exon 19 deletion mutations, acquired EGFR T790 M point mutations, and experiencing disease progression on previous-generation EGFR-TKI therapy. Concurrent administration of non-EGFR-TKI therapies, such as chemotherapy and other targeted therapies (e.g. bevacizumab), does not confer a survival benefit in this patient population, suggesting that, in the absence of new data, there is no role for these agents outside of a highly treatment refractory setting or clinical trial. Overall, our study serves as an important resource for informing clinical practice in managing lung cancer patients with LM and highlights the need for prospective studies targeting this challenging-to-treat patient population.
Data availability statement
Data available upon request.
Funding
none.
Registration
This study is registered on PROSPERO (ID: CRD42021292539).
CRediT authorship contribution statement
David J.H. Bian: Writing – review & editing, Writing – original draft, Methodology, Investigation, Formal analysis, Conceptualization. Anna-Maria Lazaratos: Writing – review & editing, Writing – original draft, Methodology, Investigation, Formal analysis, Conceptualization. Sarah M. Maritan: Writing – review & editing, Investigation. Andrea Quaiattini: Writing – review & editing, Methodology, Investigation, Conceptualization. Zhimin Zeng: Writing – review & editing, Investigation. Zhengfei Zhu: Writing – review & editing, Investigation. Ugur Sener: Writing – review & editing, Investigation. Rachna Malani: Writing – review & editing, Investigation. Yu Jung Kim: Writing – review & editing, Investigation. Eiki Ichihara: Writing – review & editing, Investigation. Victor Cohen: Writing – review & editing, Writing – original draft. April A.N. Rose: Writing – review & editing, Methodology, Investigation, Conceptualization. Nathaniel Bouganim: Writing – review & editing, Methodology, Investigation, Conceptualization. Matthew Dankner: Writing – review & editing, Writing – original draft, Supervision, Methodology, Investigation, Formal analysis, Conceptualization.
Declaration of competing interest
The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:April A. N. Rose conflicts of interest:
I = Immediate Family Member
Inst = Institutional
Research Funding: Canadian Institutes of Health Research (CIHR), Canadian Cancer Society, Conquer Cancer Foundation of ASCO, TransMedTech Institute, Jewish General Hospital Foundation, Canada Foundation for Innovation, Fonds de Recherche du Québec – Santé, Merck (Inst), AstraZeneca (Inst), Seagen (Inst), Pfizer (Inst)
Consultant: AstraZeneca Canada, Advanced Accelerator Applications/Novartis, Pfizer
Footnotes
Supplementary data to this article can be found online at https://doi.org/10.1016/j.heliyon.2024.e29668.
Appendix ASupplementary data
The following are the Supplementary data to this article:
References
- 1.Dankner M., et al. The underlying biology and therapeutic vulnerabilities of leptomeningeal metastases in adult solid cancers. Cancers. 2021;13(4) doi: 10.3390/cancers13040732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Freret M.E., Boire A. The anatomic basis of leptomeningeal metastasis. J. Exp. Med. 2024;221(4) doi: 10.1084/jem.20212121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wang N., Bertalan M.S., Brastianos P.K. Leptomeningeal metastasis from systemic cancer: review and update on management. Cancer. 2018;124(1):21–35. doi: 10.1002/cncr.30911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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;53:128–137. doi: 10.1016/j.ctrv.2016.12.006. [DOI] [PubMed] [Google Scholar]
- 5.Brower J.V., et al. Management of leptomeningeal metastases: prognostic factors and associated outcomes. J. Clin. Neurosci. 2016;27:130–137. doi: 10.1016/j.jocn.2015.11.012. [DOI] [PubMed] [Google Scholar]
- 6.Kwon J., et al. Impact of multimodality approach for patients with leptomeningeal metastases from solid tumors. J. Kor. Med. Sci. 2014;29(8):1094–1101. doi: 10.3346/jkms.2014.29.8.1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Herrlinger U., et al. Leptomeningeal metastasis: survival and prognostic factors in 155 patients. J. Neurol. Sci. 2004;223(2):167–178. doi: 10.1016/j.jns.2004.05.008. [DOI] [PubMed] [Google Scholar]
- 8.Vogelbaum M.A., et al. Treatment for brain metastases: ASCO-SNO-ASTRO guideline. J. Clin. Oncol. 2021;40(5):492–516. doi: 10.1200/JCO.21.02314. [DOI] [PubMed] [Google Scholar]
- 9.Wu Y.L., et al. Pan-Asian adapted Clinical Practice Guidelines for the management of patients with metastatic non-small-cell lung cancer: a CSCO-ESMO initiative endorsed by JSMO, KSMO, MOS, SSO and TOS. Ann. Oncol. 2019;30(2):171–210. doi: 10.1093/annonc/mdy554. [DOI] [PubMed] [Google Scholar]
- 10.Hendriks L.E., et al. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023;34(4):339–357. doi: 10.1016/j.annonc.2022.12.009. [DOI] [PubMed] [Google Scholar]
- 11.Ayati A., et al. A review on progression of epidermal growth factor receptor (EGFR) inhibitors as an efficient approach in cancer targeted therapy. Bioorg. Chem. 2020;99 doi: 10.1016/j.bioorg.2020.103811. [DOI] [PubMed] [Google Scholar]
- 12.Tamiya A., et al. Cerebrospinal fluid penetration rate and efficacy of afatinib in patients with EGFR mutation-positive non-small cell lung cancer with leptomeningeal carcinomatosis: a multicenter prospective study. Anticancer Res. 2017;37(8):4177–4182. doi: 10.21873/anticanres.11806. [DOI] [PubMed] [Google Scholar]
- 13.Yi H.G., et al. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are effective for leptomeningeal metastasis from non-small cell lung cancer patients with sensitive EGFR mutation or other predictive factors of good response for EGFR TKI. Lung Cancer. 2009;65(1):80–84. doi: 10.1016/j.lungcan.2008.10.016. [DOI] [PubMed] [Google Scholar]
- 14.Clarke J.L., et al. High dose weekly erlotinib achieves therapeutic concentrations in CSF and is effective in leptomeningeal metastases from epidermal growth factor receptor mutant lung cancer. J. Neuro Oncol. 2010;99(2):283–286. doi: 10.1007/s11060-010-0128-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Park J.H., et al. Clinical outcomes of leptomeningeal metastasis in patients with non-small cell lung cancer in the modern chemotherapy era. Lung Cancer. 2012;76(3):387–392. doi: 10.1016/j.lungcan.2011.11.022. [DOI] [PubMed] [Google Scholar]
- 16.Kawaguchi Y., et al. Clinical efficacy of afatinib treatment for a patient with leptomeningeal carcinomatosis. Chemotherapy. 2016;62(3):147–150. doi: 10.1159/000454727. [DOI] [PubMed] [Google Scholar]
- 17.Cross D.A., et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4(9):1046–1061. doi: 10.1158/2159-8290.CD-14-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mok T.S., et al. Osimertinib or platinum–pemetrexed in EGFR T790M–positive lung cancer. N. Engl. J. Med. 2016;376(7):629–640. doi: 10.1056/NEJMoa1612674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ahn M.J., et al. Osimertinib in patients with T790M mutation-positive, advanced non-small cell lung cancer: long-term follow-up from a pooled analysis of 2 phase 2 studies. Cancer. 2019;125(6):892–901. doi: 10.1002/cncr.31891. [DOI] [PubMed] [Google Scholar]
- 20.Soria J.-C., et al. Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer. N. Engl. J. Med. 2017;378(2):113–125. doi: 10.1056/NEJMoa1713137. [DOI] [PubMed] [Google Scholar]
- 21.Ramalingam S.S., et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N. Engl. J. Med. 2019;382(1):41–50. doi: 10.1056/NEJMoa1913662. [DOI] [PubMed] [Google Scholar]
- 22.Ballard P., et al. Preclinical comparison of osimertinib with other EGFR-TKIs in EGFR-mutant NSCLC brain metastases models, and early evidence of clinical brain metastases activity. Clin. Cancer Res. 2016;22(20):5130–5140. doi: 10.1158/1078-0432.CCR-16-0399. [DOI] [PubMed] [Google Scholar]
- 23.Reungwetwattana T., et al. CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-small-cell lung cancer. J. Clin. Oncol. 2018;36(33):3290–3297. doi: 10.1200/JCO.2018.78.3118. [DOI] [PubMed] [Google Scholar]
- 24.Li Y.S., et al. Leptomeningeal metastases in patients with NSCLC with EGFR mutations. J. Thorac. Oncol. 2016;11(11):1962–1969. doi: 10.1016/j.jtho.2016.06.029. [DOI] [PubMed] [Google Scholar]
- 25.Arulananda S., et al. Standard dose osimertinib for erlotinib refractory T790M-negative EGFR-mutant non-small cell lung cancer with leptomeningeal disease. J. Thorac. Dis. 2019;11(5):1756–1764. doi: 10.21037/jtd.2019.05.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hu X., et al. Clinical efficacy analysis of Osimertinib treatment for a patient with leptomeningeal metastasis of EGFR+ non-small cell lung cancer without the T790M mutation. Ann. Palliat. Med. 2019;8(5):525–531. doi: 10.21037/apm.2019.10.13. [DOI] [PubMed] [Google Scholar]
- 27.Ichihara E., et al. Re-administration of osimertinib in osimertinib-acquired resistant non-small-cell lung cancer. Lung Cancer. 2019;132:54–58. doi: 10.1016/j.lungcan.2019.02.021. [DOI] [PubMed] [Google Scholar]
- 28.Yi Y., et al. Potential benefit of osismertinib plus bevacizumab in leptomeningeal metastasis with EGFR mutant non-small-cell lung cancer. J. Transl. Med. 2022;20(1):122. doi: 10.1186/s12967-022-03331-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nanjo S., 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):32–37. doi: 10.1038/bjc.2017.394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yang J.C.H., 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(6):538–547. doi: 10.1200/JCO.19.00457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ahn M.-J., et al. Osimertinib for patients with leptomeningeal metastases associated with EGFR T790M-positive advanced NSCLC: the AURA leptomeningeal metastases analysis. J. Thorac. Oncol. 2020;15(4):637–648. doi: 10.1016/j.jtho.2019.12.113. [DOI] [PubMed] [Google Scholar]
- 32.Moher D., et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann. Intern. Med. 2009;151(4):264–269. doi: 10.7326/0003-4819-151-4-200908180-00135. w64. [DOI] [PubMed] [Google Scholar]
- 33.Moher D., et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7) doi: 10.1371/journal.pmed.1000097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Page M.J., et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. Br. Med. J. 2021;372:n160. doi: 10.1136/bmj.n160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Leonetti A., et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer. 2019;121(9):725–737. doi: 10.1038/s41416-019-0573-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Robichaux J.P., et al. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature. 2021;597(7878):732–737. doi: 10.1038/s41586-021-03898-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Murad M.H., et al. Methodological quality and synthesis of case series and case reports. BMJ Evidence-Based Medicine. 2018;23(2):60. doi: 10.1136/bmjebm-2017-110853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dankner M., et al. Clinical activity of mitogen-activated protein Kinase–targeted therapies in patients with non–V600 BRAF-mutant tumors. JCO Precision Oncology. 2022;(6) doi: 10.1200/PO.22.00107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lazaratos A.-M., et al. Intrathecal trastuzumab versus alternate routes of delivery for HER2-targeted therapies in patients with HER2+ breast cancer leptomeningeal metastases. Breast. 2023;69:451–468. doi: 10.1016/j.breast.2023.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Oken M.M., et al. Toxicity and response criteria of the eastern cooperative Oncology group. Am. J. Clin. Oncol. 1982;5(6):649–655. [PubMed] [Google Scholar]
- 41.Kim M.K., et al. Gefitinib is also active for carcinomatous meningitis in NSCLC. Lung Cancer. 2005;50(2):265–269. doi: 10.1016/j.lungcan.2005.05.022. [DOI] [PubMed] [Google Scholar]
- 42.Sakai M., et al. Carcinomatous meningitis from non-small-cell lung cancer responding to gefitinib. Int. J. Clin. Oncol. 2006;11(3):243–245. doi: 10.1007/s10147-005-0558-x. [DOI] [PubMed] [Google Scholar]
- 43.Choong N.W., et al. Gefitinib response of erlotinib-refractory lung cancer involving meninges—role of EGFR mutation. Nat. Clin. Pract. Oncol. 2006;3(1):50–57. doi: 10.1038/ncponc0400. [DOI] [PubMed] [Google Scholar]
- 44.Flowers A., Kamradt J. 2006. Sequential Intra-Ommaya Chemotherapy and Systemic Tarceva Prolong Survival in a Patient with Brain and Leptomeningeal Metastases from Lung Adenocarcinoma; p. 345. 345. [Google Scholar]
- 45.Jackman D.M., et al. Response and resistance in a non-small-cell lung cancer patient with an epidermal growth factor receptor mutation and leptomeningeal metastases treated with high-dose gefitinib. J. Clin. Oncol. 2006;24(27):4517–4520. doi: 10.1200/JCO.2006.06.6126. [DOI] [PubMed] [Google Scholar]
- 46.Kanaji N., et al. Significance of an epidermal growth factor receptor mutation in cerebrospinal fluid for carcinomatous meningitis. Intern. Med. 2007;46(19):1651–1655. doi: 10.2169/internalmedicine.46.0335. [DOI] [PubMed] [Google Scholar]
- 47.Shinagawa N., et al. Gefitinib administration in a patient with lung cancer undergoing hemodialysis. Lung Cancer. 2007;58(3):422–424. doi: 10.1016/j.lungcan.2007.06.001. [DOI] [PubMed] [Google Scholar]
- 48.Fukuhara T., et al. Successful treatment of carcinomatous meningitis with gefitinib in a patient with lung adenocarcinoma harboring a mutated EGF receptor gene. Tohoku J. Exp. Med. 2008;214(4):359–363. doi: 10.1620/tjem.214.359. [DOI] [PubMed] [Google Scholar]
- 49.Dhruva N., Socinski M.A. Carcinomatous meningitis in non-small-cell lung cancer: response to high-dose erlotinib. J. Clin. Oncol. 2009;27(22):e31–e32. doi: 10.1200/JCO.2008.21.0963. [DOI] [PubMed] [Google Scholar]
- 50.Ruppert A.M., et al. EGFR-TKI and lung adenocarcinoma with CNS relapse: interest of molecular follow-up. Eur. Respir. J. 2009;33(2):436–440. doi: 10.1183/09031936.00162307. [DOI] [PubMed] [Google Scholar]
- 51.Masuda T., et al. Erlotinib efficacy and cerebrospinal fluid concentration in patients with lung adenocarcinoma developing leptomeningeal metastases during gefitinib therapy. Cancer Chemother. Pharmacol. 2011;67(6):1465–1469. doi: 10.1007/s00280-011-1555-6. [DOI] [PubMed] [Google Scholar]
- 52.Nagano T., et al. Long-term outcome after multidisciplinary approach for leptomeningeal carcinomatosis in a non-small cell lung cancer patient with poor performance status. Intern. Med. 2011;50(24):3019–3022. doi: 10.2169/internalmedicine.50.5903. [DOI] [PubMed] [Google Scholar]
- 53.Ohara G., et al. Recovery from carcinomatous meningitis by erlotinib. Onkologie. 2011;34(7):394–395. doi: 10.1159/000329620. [DOI] [PubMed] [Google Scholar]
- 54.Tetsumoto S., et al. Two cases of leptomeningeal metastases from lung adenocarcinoma which progressed during gefitinib therapy but responded to erlotinib. Int. J. Clin. Oncol. 2012;17(2):155–159. doi: 10.1007/s10147-011-0256-9. [DOI] [PubMed] [Google Scholar]
- 55.Weber B., et al. Erlotinib accumulation in brain metastases from non-small cell lung cancer: visualization by positron emission tomography in a patient harboring a mutation in the epidermal growth factor receptor. J. Thorac. Oncol. 2011;6(7):1287–1289. doi: 10.1097/JTO.0b013e318219ab87. [DOI] [PubMed] [Google Scholar]
- 56.Ishikawa R., et al. Successful treatment of carcinomatous meningitis with erlotinib and whole brain radiotherapy. Ann. Cancer Res. Ther. 2012;20(2):58–62. [Google Scholar]
- 57.Togashi Y., et al. Successful erlotinib rechallenge for leptomeningeal metastases of lung adenocarcinoma after erlotinib-induced interstitial lung disease: a case report and review of the literature. Lung Cancer. 2012;77(2):464–468. doi: 10.1016/j.lungcan.2012.04.013. [DOI] [PubMed] [Google Scholar]
- 58.Yuan Y., et al. Activity of pemetrexed and high-dose gefitinib in an EGFR-mutated lung adenocarcinoma with brain and leptomeningeal metastasis after response to gefitinib. World J. Surg. Oncol. 2012;10(1):235. doi: 10.1186/1477-7819-10-235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hollebecque A., et al. First case report of intrathecal panitumumab for treatment of meningeal carcinomatousis in an EGFR mutant lung adenocarcinoma patient. Lung Cancer. 2013;80(1):113–114. doi: 10.1016/j.lungcan.2012.12.016. [DOI] [PubMed] [Google Scholar]
- 60.Nakamichi S., et al. Successful EGFR-TKI rechallenge of leptomeningeal carcinomatosis after gefitinib-induced interstitial lung disease. Jpn. J. Clin. Oncol. 2013;43(4):422–425. doi: 10.1093/jjco/hyt012. [DOI] [PubMed] [Google Scholar]
- 61.Okuda T., et al. Administration of gefitinib via nasogastric tube effectively improved the performance status of a patient with lung adenocarcinoma-derived meningeal carcinomatosis. International Cancer Conference Journal. 2014;3(4):211–214. [Google Scholar]
- 62.Watanabe Y., et al. A case of lung adenocarcinoma which developed carcinomatous meningitis during gefitinib treatment, improving after switching to erlotinib. Haigan. 2013;53(2):114–120. [Google Scholar]
- 63.Avik E.L., Almasri E., Abdulhaq H. D45. CASE REPORTS: TUMORS. American Thoracic Society; 2014. Leptomeningeal disease as initial presentation of lung adenocarcinoma with epidermal growth factor receptor (EGFR) mutation and anaplastic Lymphoma kinase (ALK) gene rearrangement; p. A6026. A6026. [Google Scholar]
- 64.Cessot A., Blanchet B., Goldwasser F. Erlotinib treatment of meningeal carcinomatosis in lung cancer: more is better. Ann. Oncol. 2014;25(10):2093–2094. doi: 10.1093/annonc/mdu261. [DOI] [PubMed] [Google Scholar]
- 65.Lin C.H., et al. Afatinib combined with cetuximab for lung adenocarcinoma with leptomeningeal carcinomatosis. Lung Cancer. 2014;85(3):479–480. doi: 10.1016/j.lungcan.2014.06.002. [DOI] [PubMed] [Google Scholar]
- 66.Oda N., et al. Cerebral venous sinus thrombosis concomitant with leptomeningeal carcinomatosis, in a patient with epidermal growth factor receptor-mutated lung cancer. Oncol. Lett. 2014;8(6):2489–2492. doi: 10.3892/ol.2014.2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ohara S., et al. Brain metastasis effectively treated with erlotinib following the acquisition of resistance to gefitinib: a case report. J. Med. Case Rep. 2014;8:64. doi: 10.1186/1752-1947-8-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Poonia S., et al. EGFR exon 19 deletion mutations and systemic/central nervous system miliary metastasis: clinical correlations and response to therapy. Clin. Lung Cancer. 2014;15(5):387–389. doi: 10.1016/j.cllc.2014.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Xing P., et al. Recurrent response to advanced lung adenocarcinoma with erlotinib developing leptomeningeal metastases during gefitinib therapy and two case reports. Thorac Cancer. 2014;5(1):38–42. doi: 10.1111/1759-7714.12049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yajima T., et al. A case of lung adenocarcinoma with gefitinib-resistant carcinomatous meningitis exhibiting the dose-dependent effects of erlotinib. Haigan. 2014;54(6):806–811. [Google Scholar]
- 71.Yang H., et al. Erlotinib in combination with pemetrexed/cisplatin for leptomeningeal metastases and cerebrospinal fluid drug concentrations in lung adenocarcinoma patients after gefitinib faliure. Targeted Oncol. 2015;10(1):135–140. doi: 10.1007/s11523-014-0326-9. [DOI] [PubMed] [Google Scholar]
- 72.Hoffknecht P., et al. Efficacy of the irreversible ErbB family blocker afatinib in epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-pretreated non-small-cell lung cancer patients with brain metastases or leptomeningeal disease. J. Thorac. Oncol. 2015;10(1):156–163. doi: 10.1097/JTO.0000000000000380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Sekine A., et al. Promising effects of afatinib on leptomeningeal carcinomatosis derived from erlotinib-resistant lung adenocarcinoma. Intern. Med. 2016;55(17):2457–2461. doi: 10.2169/internalmedicine.55.6102. [DOI] [PubMed] [Google Scholar]
- 74.Chan O.S., Leung W.K., Yeung R.M. Sustained response to standard dose osimertinib in a patient with plasma T790M-positive leptomeningeal metastases from primary lung adenocarcinoma. Asia Pac. J. Clin. Oncol. 2017;13(6):428–430. doi: 10.1111/ajco.12673. [DOI] [PubMed] [Google Scholar]
- 75.Chonan M., Narita N., Tominaga T. Total regression of brain metastases in non-small cell lung cancer patients harboring EGFR mutations treated with gefitinib without radiotherapy: two case reports. BMC Res. Notes. 2016;9(1):2. doi: 10.1186/s13104-015-1834-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fan F.S. Rapid response to high-dose, pulsatile erlotinib in afatinib-refractory leptomeningeal carcinomatosis from adenocarcinoma of the lung: a case report. Case Rep. Oncol. 2016;9(3):537–542. doi: 10.1159/000449405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jiang S.Y., et al. Small-cell lung cancer transformation in patients with pulmonary adenocarcinoma: a case report and review of literature. Medicine (Baltim.) 2016;95(6):e2752. doi: 10.1097/MD.0000000000002752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kanemaru R., et al. Successful treatment with weekly high-dose erlotinib against meningeal metastases from epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. Respir Investig. 2016;54(5):372–375. doi: 10.1016/j.resinv.2016.04.001. [DOI] [PubMed] [Google Scholar]
- 79.Lee A.J., et al. Leptomeningeal carcinomatosis in non-small-cell lung cancer: initial response to erlotinib followed by relapse despite continuing radiological resolution of disease. Oxf Med Case Reports. 2016;2016(9):omw069. doi: 10.1093/omcr/omw069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sakaguchi M., et al. Successful treatment of nonsmall cell lung cancer patients with leptomeningeal metastases using whole brain radiotherapy and tyrosine kinase inhibitors. J. Cancer Res. Therapeut. 2020;16(4):930–932. doi: 10.4103/jcrt.JCRT_1343_16. [DOI] [PubMed] [Google Scholar]
- 81.Sakata Y., et al. Erlotinib plus bevacizumab as an effective treatment for leptomeningeal metastases from EGFR mutation-positive non-small cell lung cancer. Lung Cancer. 2016;99:120–122. doi: 10.1016/j.lungcan.2016.07.008. [DOI] [PubMed] [Google Scholar]
- 82.Yang Z., et al. AZD3759, a BBB-penetrating EGFR inhibitor for the treatment of EGFR mutant NSCLC with CNS metastases. Sci. Transl. Med. 2016;8(368):368ra172. doi: 10.1126/scitranslmed.aag0976. [DOI] [PubMed] [Google Scholar]
- 83.Ariyasu R., et al. Efficacy of bevacizumab and erlotinib combination for leptomeningeal carcinomatosis after failure of erlotinib. Anti Cancer Drugs. 2017;28(5):565–567. doi: 10.1097/CAD.0000000000000489. [DOI] [PubMed] [Google Scholar]
- 84.Chalmers A., Jensen L., Akerley W. Durable response to osimertinib in EGFR mutated T790M wildtype non-small cell lung cancer with leptomeningeal metastases: a case report. Lung Cancer. 2017;114:68–69. doi: 10.1016/j.lungcan.2017.10.009. [DOI] [PubMed] [Google Scholar]
- 85.Cordova C., et al. Osimertinib dose escalation induces regression of progressive <em>EGFR</em> T790M–mutant leptomeningeal lung adenocarcinoma. J. Thorac. Oncol. 2017;12(11):e188–e190. doi: 10.1016/j.jtho.2017.07.029. [DOI] [PubMed] [Google Scholar]
- 86.Gortais H., et al. T790M EGFR mutation detection in cerebrospinal fluid and response to osimertinib in a lung cancer patient with meningeal carcinomatosis. J. Thorac. Oncol. 2017;12(9):e138–e139. doi: 10.1016/j.jtho.2017.04.022. [DOI] [PubMed] [Google Scholar]
- 87.Kunimasa K., Mimura C., Kotani Y. Erlotinib is effective for leptomeningeal carcinomatosis due to disease flare after osimertinib treatment failure. J. Thorac. Oncol. 2017;12(7):e93–e94. doi: 10.1016/j.jtho.2017.02.025. [DOI] [PubMed] [Google Scholar]
- 88.Mehta P., Hamid O., Klempner S.J. Pulse dose erlotinib and Zuckerguss improvement in EGFR-mutant NSCLC. J. Thorac. Oncol. 2017;12(12):1857–1858. doi: 10.1016/j.jtho.2017.07.024. [DOI] [PubMed] [Google Scholar]
- 89.Niu H., et al. Treatment of leptomeningeal metastases in a patient with non-small cell lung cancer harboring EGFR T790M mutation. Case Rep. Oncol. 2017;10(3):840–845. doi: 10.1159/000480452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sakai H., et al. Successful osimertinib treatment for leptomeningeal carcinomatosis from lung adenocarcinoma with the T790M mutation of EGFR. ESMO Open. 2017;2(Suppl 1) doi: 10.1136/esmoopen-2016-000104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Takeda T., et al. Osimertinib administration via nasogastric tube in an EGFR-T790M-positive patient with leptomeningeal metastases. Respirol Case Rep. 2017;5(4) doi: 10.1002/rcr2.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Théoleyre S., et al. Treatment of a NSCLC patient with osimertinib based on the detection of the EGFR T790M resistance mutation in cerebrospinal fluid. Lung Cancer. 2017;114:111–112. doi: 10.1016/j.lungcan.2017.10.010. [DOI] [PubMed] [Google Scholar]
- 93.Yamanaka Y., et al. Afatinib therapy for brain metastases aggravated by a reduction in the dose of erlotinib due to the development of hepatotoxicity. Intern. Med. 2017;56(21):2895–2898. doi: 10.2169/internalmedicine.8638-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chen G., et al. Meningeal carcinomatosis: three case-reports. World J. Surg. Oncol. 2018;16(1):78. doi: 10.1186/s12957-018-1376-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chen J., Soudy H. Osimertinib in the treatment of leptomeningeal disease in T790M-negative, epidermal growth factor receptor-mutated non-small cell lung cancer: a case report. Chin. Clin. Oncol. 2019;8(3):29. doi: 10.21037/cco.2019.02.02. [DOI] [PubMed] [Google Scholar]
- 96.Jing W., et al. Great efficacy of bevacizumab plus erlotinib for leptomeningeal metastases from non-small cell lung cancer with initially positive EGFR mutation: a case report. Cancer Biol. Ther. 2018;19(12):1073–1077. doi: 10.1080/15384047.2018.1491504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Li J., Liu X., Yuan C. Treatment response to osimertinib in a patient with leptomeningeal metastasis from lung adenocarcinoma following failure of gefitinib and erlotinib: a case report. Mol Clin Oncol. 2018;9(3):321–324. doi: 10.3892/mco.2018.1666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Senoo S., et al. Osimertinib depletes EGFR T790M in the spinal fluid of patients with carcinomatous meningitis of lung adenocarcinoma harboring de Novo EGFR T790M. J. Thorac. Oncol. 2018;13(8):e140–e142. doi: 10.1016/j.jtho.2018.03.016. [DOI] [PubMed] [Google Scholar]
- 99.Uruga H., et al. Gefitinib successfully administered in a lung cancer patient with leptomeningeal carcinomatosis after erlotinib-induced pneumatosis intestinalis. BMC Cancer. 2018;18(1):825. doi: 10.1186/s12885-018-4743-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wang Y., et al. Non-small cell lung cancer leptomeningeal metastases treated with intrathecal therapy plus osimertinib and temozolomide and whole-brain radiation therapy: a case report. OncoTargets Ther. 2018;11:4733–4738. doi: 10.2147/OTT.S164968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yoshida H., Ooi M., Kim Y.H. Successful treatment with osimertinib and chemotherapy in a non-small cell lung cancer patient with EGFR mutation and meningeal carcinomatosis. J. Thorac. Oncol. 2018;13(11):e219–e220. doi: 10.1016/j.jtho.2018.05.023. [DOI] [PubMed] [Google Scholar]
- 102.Zhang Y., et al. A combined subtype of small cell lung cancer and adenocarcinoma with epidermal growth factor receptor (EGFR) mutation responds to EGFR tyrosine kinase inhibitors. J. Thorac. Dis. 2018;10(10):E739–e743. doi: 10.21037/jtd.2018.09.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Abe M., et al. Erlotinib and bevacizumab combination therapy for afatinib-refractory leptomeningeal carcinomatosis from EGFR-mutated lung cancer. Int Cancer Conf J. 2019;8(2):81–85. doi: 10.1007/s13691-019-00358-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gong L., et al. The evolutionary difference between extracranial lesions and leptomeningeal metastasis in a patient with afatinib-resistant lung cancer. J. Thorac. Oncol. 2019;14(6):e120–e123. doi: 10.1016/j.jtho.2019.01.030. [DOI] [PubMed] [Google Scholar]
- 105.Li H., et al. Treatment response to intrathecal chemotherapy with pemetrexed via an Ommaya reservoir in EGFR-mutated leptomeningeal metastases from non-small cell lung cancer: a case report. Ann. Palliat. Med. 2020;9(4):2341–2346. doi: 10.21037/apm-19-521. [DOI] [PubMed] [Google Scholar]
- 106.Liu J., et al. Afatinib helped overcome subsequent resistance to osimertinib in a patient with NSCLC having leptomeningeal metastasis baring acquired EGFR L718Q mutation: a case report. BMC Cancer. 2019;19(1):702. doi: 10.1186/s12885-019-5915-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Llinás-Quintero N., et al. Response of leptomeningeal metastases in EGFR-mutated non-small-cell lung cancer to afatinib in the absence of radiotherapy. Case Rep Oncol Med. 2019;2019 doi: 10.1155/2019/1939703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sazuka M., et al. An elderly case of leptomeningeal carcinomatosis from non-small cell lung cancer treated effectively with osimertinib and ommaya ReservoirOsimertinibの投与とOmmayaリザーバーからの髄液ドレナージが癌性髄膜炎の治療に有効だった高齢者非小細胞肺癌の1例. Haigan. 2019;59:287–292. [Google Scholar]
- 109.Tsang M.W. Osimertinib 160 mg daily for advanced non-small cell lung cancer with leptomeningeal metastasis: a case report. Asia Pac. J. Clin. Oncol. 2019;15(Suppl 6):5–7. doi: 10.1111/ajco.13246. [DOI] [PubMed] [Google Scholar]
- 110.Wang Z., Wu Y.L. Re-Emerging C797S in trans and rechallenge of osimertinib with erlotinib. J. Thorac. Oncol. 2019;14(4):e81–e82. doi: 10.1016/j.jtho.2018.12.015. [DOI] [PubMed] [Google Scholar]
- 111.Dasgupta A., et al. Focal leptomeningeal disease with perivascular invasion in EGFR-mutant non-small-cell lung cancer. AJNR Am J Neuroradiol. 2020;41(8):1430–1433. doi: 10.3174/ajnr.A6640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Jiang T., et al. Osimertinib in combination with bevacizumab in EGFR-Mutated NSCLC with leptomeningeal metastases. Transl. Lung Cancer Res. 2020;9(6):2514–2517. doi: 10.21037/tlcr-20-984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Li H., et al. Three Novel EGFR Mutations (750_758del, I759S, T751_I759delinsS) in One Patient with Metastatic Non-Small Cell Lung Cancer Responding to Osimertinib: A Case Report. OncoTargets Ther. 2020;13:7941–7948. doi: 10.2147/OTT.S259616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Li H.Y., et al. Durable response to pulsatile icotinib for central nervous system metastases from EGFR-mutated non-small cell lung cancer: a case report. World J Clin Cases. 2020;8(2):370–376. doi: 10.12998/wjcc.v8.i2.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Li Y., et al. Meningeal metastasis patients with EGFR G724S who develop resistance to osimertinib benefit from the addition of afatinib. Transl. Lung Cancer Res. 2020;9(5):2188–2190. doi: 10.21037/tlcr-20-847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ma C., et al. Efficacy of afatinib for pulmonary adenocarcinoma with leptomeningeal metastases harboring an epidermal growth factor receptor complex mutation (exon 19del+K754E): A case report. Medicine (Baltim.) 2020;99(43) doi: 10.1097/MD.0000000000022851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ma C., et al. Effective treatment with afatinib of lung adenocarcinoma with leptomeningeal metastasis harboring the exon 18 p.G719A mutation in the EGFR gene was detected in cerebrospinal fluid: a case report. Front. Oncol. 2020;10:1635. doi: 10.3389/fonc.2020.01635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Mu N., et al. Afatinib successfully treated leptomeningeal metastasis of lung adenocarcinoma in a patient with EGFR G719A mutation in detection of cerebrospinal fluid. Chinese Journal of Contemporary Neurology and Neurosurgery. 2020;20(6):551–555. [Google Scholar]
- 119.Nakamura K., et al. A case of EGFR gene mutation-positive early lung adenocarcinoma that recurred 12 Years postoperatively with meningeal carcinomatosis Alone髄膜癌腫症単独で術後12年目に再発したEGFR遺伝子変異陽性早期肺腺癌の1例. Haigan. 2020;60:192–196. [Google Scholar]
- 120.Okuno T., et al. Efficacy of osimertinib in a patient with leptomeningeal metastasis and EGFR uncommon S768I mutation. Lung Cancer. 2020;143:95–96. doi: 10.1016/j.lungcan.2020.03.016. [DOI] [PubMed] [Google Scholar]
- 121.Zhang Y.J., et al. Detection of meningeal metastasis in the cerebrospinal fluid in lung adenocarcinoma: case report. Clin. Lung Cancer. 2020;21(5):e493–e496. doi: 10.1016/j.cllc.2020.04.010. [DOI] [PubMed] [Google Scholar]
- 122.Chan D. P76.87 efficacy of dacomitinib in EGFR TKI refractory metastatic non-small cell lung cancer (EGFR mutant) with leptomeningeal metastases. J. Thorac. Oncol. 2021;16(3):S627. [Google Scholar]
- 123.Hirokawa E., et al. Durable response to EGFR tyrosine kinase inhibitors in a patient with non-small cell lung cancer harboring an EGFR kinase domain duplication. Thorac Cancer. 2021;12(16):2283–2287. doi: 10.1111/1759-7714.14081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Li S., et al. Next generation sequencing in the management of leptomeningeal metastases of non-small cell lung cancer: a case report and literature review. Recent Pat. Anti-Cancer Drug Discov. 2021;16(1):108–116. doi: 10.2174/1574892815666201127114224. [DOI] [PubMed] [Google Scholar]
- 125.Liu M., et al. Preliminary study on double-dose icotinib successfully treated leptomeningeal metastasis in lung adenocarcinoma patient with EGFR gene sensitive mutation. Chinese Journal of Contemporary Neurology and Neurosurgery. 2021;21(11) [Google Scholar]
- 126.Merkhofer C.M., Baik C.S. Durable response of leptomeningeal disease with osimertinib and pemetrexed in EGFR-mutated metastatic NSCLC: a case report. JTO Clin Res Rep. 2021;2(6) doi: 10.1016/j.jtocrr.2021.100177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Morichika D., et al. Efficacy of multimodal treatment for leptomeningeal metastases in a lung cancer harboring an EGFR mutation. OncoTargets Ther. 2016;9:1753–1758. doi: 10.2147/OTT.S95721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ono Y., et al. First-line osimertinib for leptomeningeal metastasis from lung adenocarcinoma with EGFR mutation as the initial and solitary site of postoperative recurrence. Int Cancer Conf J. 2021;10(1):78–82. doi: 10.1007/s13691-020-00453-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Provenzano L., et al. Carcinomatosis under control by osimertinib in EGFR and TP53 mutated lung adenocarcinoma. Tumori. 2021;107(6):Np136–np140. doi: 10.1177/03008916211052330. [DOI] [PubMed] [Google Scholar]
- 130.Sakurai T., Tuchida A., Nishida H. Significance of an epidermal growth factor receptor mutation in cerebrospinal fluid for leptomeningeal metastasis and successful treatment with osimertinib: a case report and literature review. eNeurologicalSci. 2021;22 doi: 10.1016/j.ensci.2020.100303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shan C.G., et al. A non-small cell lung cancer (NSCLC) patient with leptomeningeal metastasis harboring rare epidermal growth factor receptor (EGFR) mutations G719S and L861Q benefited from doubling dosage of osimertinib: a case report. Ann. Palliat. Med. 2021;10(5):5897–5901. doi: 10.21037/apm-20-2556. [DOI] [PubMed] [Google Scholar]
- 132.Takahashi T., et al. Disease flare of leptomeningeal metastases without radiological and cytological findings after the discontinuation of osimertinib. Lung Cancer. 2021;151:1–4. doi: 10.1016/j.lungcan.2020.11.010. [DOI] [PubMed] [Google Scholar]
- 133.Valet O., et al. Response to the combination of osimertinib, dabrafenib, and trametinib in leptomeningitis from EGFR-mutant NSCLC with acquired braf V600E mutation: a case report. JTO Clin Res Rep. 2021;2(6) doi: 10.1016/j.jtocrr.2021.100192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wang M., et al. Erlotinib combined with bevacizumab and chemotherapy in first line osimertinib-resistant NSCLC patient with leptomeningeal metastasis: a case report. Medicine (Baltim.) 2021;100(44) doi: 10.1097/MD.0000000000027727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Chiba S., et al. Combination treatment with bevacizumab plus erlotinib for meningeal carcinomatosis of afatinib-resistant EGFR mutated lung cancer without T790M mutation: a case report. Ann. Palliat. Med. 2022;11(8):2745–2750. doi: 10.21037/apm-21-1885. [DOI] [PubMed] [Google Scholar]
- 136.Kanbour A., et al. Leptomeningeal metastatic L858R EGFR-mutant lung cancer: prompt response to osimertinib in the absence of T790M-mutation and effective subsequent pulsed erlotinib. OncoTargets Ther. 2022;15:659–667. doi: 10.2147/OTT.S336012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kunimasa K., et al. Dacomitinib overcomes afatinib-refractory carcinomatous meningitis in a lung cancer patient harbouring EGFR Ex.19 deletion and G724S mutation; a case report. Invest. N. Drugs. 2022;40(5):1137–1140. doi: 10.1007/s10637-022-01264-0. [DOI] [PubMed] [Google Scholar]
- 138.Ma Y., et al. Successful treatment using targeted therapy, radiotherapy, and intrathecal chemotherapy in a patient with leptomeningeal metastasis with an epidermal growth factor receptor exon 20 insertion mutation: a case report. Ann. Palliat. Med. 2021;11(4):1533–1541. doi: 10.21037/apm-21-321. [DOI] [PubMed] [Google Scholar]
- 139.Wu K., et al. Salvage therapy of osimertinib plus anlotinib in advanced lung adenocarcinoma with leptomeningeal metastasis: a case report. Respir Med Case Rep. 2022;38 doi: 10.1016/j.rmcr.2022.101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Xu L.Q., et al. Early detection of circulating tumor DNA and successful treatment with osimertinib in thr790met-positive leptomeningeal metastatic lung cancer: a case report. World J Clin Cases. 2022;10(22):7968–7972. doi: 10.12998/wjcc.v10.i22.7968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yokota H., et al. Band-like hyperintensity along the ventral surface of the brain stem on FLAIR and DWI in leptomeningeal carcinomatosis of lung adenocarcinoma. Radiol Case Rep. 2022;17(3):544–548. doi: 10.1016/j.radcr.2021.11.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zheng S., et al. Complete remission in leptomeningeal metastasis of NSCLC with rare EGFR-SEPT14 fusion treated with osimertinib combined with intrathecal chemotherapy with pemetrexed. Anti Cancer Drugs. 2022;33(1):e795–e798. doi: 10.1097/CAD.0000000000001222. [DOI] [PubMed] [Google Scholar]
- 143.Kida Y., et al. Three cases of leptomeningeal carcinomatosis from lung adenocarcinoma responding to EGFR TKIs. Haigan. 2009;49:461–466. [Google Scholar]
- 144.Masuda T., et al. Two cases of lung cancer with carcinomatous meningitis in which erlotinib was effective after gefitinib therapy. Haigan. 2009;49:207–213. [Google Scholar]
- 145.So T., et al. Gefitinib and a ventriculo-peritoneal shunt to manage carcinomatous meningitis from non-small-cell lung cancer: report of two cases. Surg. Today. 2009;39(7):598–602. doi: 10.1007/s00595-008-3909-1. [DOI] [PubMed] [Google Scholar]
- 146.Grommes C., et al. "Pulsatile" high-dose weekly erlotinib for CNS metastases from EGFR mutant non-small cell lung cancer. Neuro Oncol. 2011;13(12):1364–1369. doi: 10.1093/neuonc/nor121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Takenaka T., et al. Long-term survival of a patient with carcinomatous meningitis due to non-small cell lung cancer treated with erlotinib following gefitinib. BMJ Case Rep. 2011:2011. doi: 10.1136/bcr.08.2011.4586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hata A., et al. Erlotinib for whole-brain-radiotherapy-refractory leptomeningeal metastases after gefitinib failure in a lung adenocarcinoma patient. J. Thorac. Oncol. 2012;7(4):770–771. doi: 10.1097/JTO.0b013e31824c96bc. [DOI] [PubMed] [Google Scholar]
- 149.Santhosh-Kumar C.R., Gray D., Fisher D.J. Pulsed erlotinib as sole treatment of leptomeningeal carcinomatosis: can we avoid the use of intrathecal therapy? World J. Oncol. 2014;5(5–6):237–239. doi: 10.14740/wjon859w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Saijo T., Ito T., Ikeda N. Three cases of EGFR mutation-positive lung adenocarcinoma with leptomeningeal carcinomatosis successfully treated with afatinib. Haigan. 2016;56(4):290–296. [Google Scholar]
- 151.Grimsrud K.W., Mrugala M.M. Next-generation epidermal growth factor receptor tyrosine kinase inhibitors for leptomeningeal carcinomatosis: review of 2 cases. Neurol. 2019;24(2):59–61. doi: 10.1097/NRL.0000000000000220. [DOI] [PubMed] [Google Scholar]
- 152.Li H., et al. Treatment response to osimertinib in EGFR-mutated leptomeningeal metastases from non-small cell lung cancer: a case series. OncoTargets Ther. 2019;12:7785–7790. doi: 10.2147/OTT.S199452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sener U., et al. Radiographic appearance of leptomeningeal disease in patients with EGFR-mutated non-small-cell lung carcinoma treated with tyrosine kinase inhibitors: a case series. CNS Oncol. 2019;8(4):Cns42. doi: 10.2217/cns-2019-0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kim H.R., et al. Osimertinib combined with systemic chemotherapy for EGFR mutant, T790M-negative, non-small cell lung cancer patients who develop leptomeningeal metastases with extracranial progression to prior EGFR TKI. Cancer Res Treat. 2023;55(1):344–349. doi: 10.4143/crt.2021.1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Qian C., et al. Case report: rechallenge with EGFR-TKIs after immunotherapy in EGFR-mutated non-small cell lung cancer with leptomeningeal metastasis. Front. Oncol. 2022;12 doi: 10.3389/fonc.2022.957661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Gal O., et al. Tyrosine kinase inhibitors as a treatment of symptomatic CNS metastases in oncogene-driven NSCLC. JAMA Oncol. 2020;2020 doi: 10.1155/2020/1980891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Zhou Y., et al. Survival outcomes and symptomatic central nervous system (CNS) metastasis in EGFR-mutant advanced non-small cell lung cancer without baseline CNS metastasis: osimertinib vs. first-generation EGFR tyrosine kinase inhibitors. Lung Cancer. 2020;150:178–185. doi: 10.1016/j.lungcan.2020.10.018. [DOI] [PubMed] [Google Scholar]
- 158.Kobayashi S., et al. EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 2005;352(8):786–792. doi: 10.1056/NEJMoa044238. [DOI] [PubMed] [Google Scholar]
- 159.Yang J.C.H., 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. 2019;38(6):538–547. doi: 10.1200/JCO.19.00457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Park S., et al. A phase II, multicenter, two cohort study of 160 mg osimertinib in EGFR T790M-positive non-small-cell lung cancer patients with brain metastases or leptomeningeal disease who progressed on prior EGFR TKI therapy. Ann. Oncol. 2020;31(10):1397–1404. doi: 10.1016/j.annonc.2020.06.017. [DOI] [PubMed] [Google Scholar]
- 161.Li N., et al. Survival outcomes of patients with epidermal growth factor receptor mutations in non-small cell lung cancer with leptomeningeal metastasis. Front. Oncol. 2021;11 doi: 10.3389/fonc.2021.723562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Tatineni V., et al. First- versus third-generation EGFR tyrosine kinase inhibitors in EGFR-mutated non-small cell lung cancer patients with brain metastases. Cancers. 2023;15(8) doi: 10.3390/cancers15082382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Miller V.A., et al. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol. 2012;13(5):528–538. doi: 10.1016/S1470-2045(12)70087-6. [DOI] [PubMed] [Google Scholar]
- 164.Tanaka K., et al. Acquisition of the T790M resistance mutation during afatinib treatment in EGFR tyrosine kinase inhibitor-naïve patients with non-small cell lung cancer harboring EGFR mutations. Oncotarget. 2017;8(40):68123–68130. doi: 10.18632/oncotarget.19243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hong Y., et al. Systematic immunological level determined the prognosis of leptomeningeal metastasis in lung cancer. Cancer Manag. Res. 2022;14:1153–1164. doi: 10.2147/CMAR.S347323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Tamura K., et al. Comparison of clinical outcomes of osimertinib and first-generation EGFR-tyrosine kinase inhibitors (TKIs) in TKI-untreated EGFR-mutated non-small-cell lung cancer with leptomeningeal metastases. ESMO Open. 2023;8(4) doi: 10.1016/j.esmoop.2023.101594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Colclough N., et al. 64 - preclinical comparison of the blood brain barrier (BBB) permeability of osimertinib (AZD9291) with other irreversible next generation EGFR TKIs. Eur. J. Cancer. 2016;69:S28. [Google Scholar]
- 168.Wu Y.-L., et al. CNS efficacy of osimertinib in patients with T790M-positive advanced non–small-cell lung cancer: data from a randomized phase III trial (AURA3) J. Clin. Oncol. 2018;36(26):2702–2709. doi: 10.1200/JCO.2018.77.9363. [DOI] [PubMed] [Google Scholar]
- 169.Weller R.O., et al. The meninges as barriers and facilitators for the movement of fluid, cells and pathogens related to the rodent and human CNS. Acta Neuropathol. 2018;135(3):363–385. doi: 10.1007/s00401-018-1809-z. [DOI] [PubMed] [Google Scholar]
- 170.Steeg P.S. The blood-tumour barrier in cancer biology and therapy. Nat. Rev. Clin. Oncol. 2021;18(11):696–714. doi: 10.1038/s41571-021-00529-6. [DOI] [PubMed] [Google Scholar]
- 171.Boire A., et al. Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis. Cell. 2017;168(6):1101–1113.e13. doi: 10.1016/j.cell.2017.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Suh J.H., et al. Current approaches to the management of brain metastases. Nat. Rev. Clin. Oncol. 2020;17(5):279–299. doi: 10.1038/s41571-019-0320-3. [DOI] [PubMed] [Google Scholar]
- 173.Le Rhun E., et al. EANO-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with leptomeningeal metastasis from solid tumours. Ann. Oncol. 2017;28(suppl_4):iv84–iv99. doi: 10.1093/annonc/mdx221. [DOI] [PubMed] [Google Scholar]
- 174.Kuiper J.L., 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(3):255–261. doi: 10.1016/j.lungcan.2015.05.023. [DOI] [PubMed] [Google Scholar]
- 175.Gwak H.-S., et al. Analysis of treatment outcomes of intraventricular chemotherapy in 105 patients for leptomeningeal carcinomatosis from non–small-cell lung cancer. J. Thorac. Oncol. 2013;8(5):599–605. doi: 10.1097/JTO.0b013e318287c943. [DOI] [PubMed] [Google Scholar]
- 176.Yang T.J., et al. Clinical trial of proton craniospinal irradiation for leptomeningeal metastases. Neuro Oncol. 2021;23(1):134–143. doi: 10.1093/neuonc/noaa152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Yang J.T., et al. Randomized phase II trial of proton craniospinal irradiation versus photon involved-field radiotherapy for patients with solid tumor leptomeningeal metastasis. J. Clin. Oncol. 2022;40(33):3858–3867. doi: 10.1200/JCO.22.01148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Gen S., et al. Clinical efficacy of osimertinib in EGFR-mutant non-small cell lung cancer with distant metastasis. BMC Cancer. 2022;22(1):654. doi: 10.1186/s12885-022-09741-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Chan S.-K., Choi H.C.-W., Lee V.H.-F. Overall survival benefits of first-line treatments for Asian patients with advanced EGFR-mutated NSCLC harboring L858R mutation: a systematic review and network meta-analysis. JTO Clinical and Research Reports. 2022;3(5) doi: 10.1016/j.jtocrr.2022.100322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kawashima Y., et al. Bevacizumab plus erlotinib versus erlotinib alone in Japanese patients with advanced, metastatic, EGFR-mutant non-small-cell lung cancer (NEJ026): overall survival analysis of an open-label, randomised, multicentre, phase 3 trial. Lancet Respir. Med. 2022;10(1):72–82. doi: 10.1016/S2213-2600(21)00166-1. [DOI] [PubMed] [Google Scholar]
- 181.Planchard D., et al. Osimertinib plus platinum-pemetrexed in newly diagnosed epidermal growth factor receptor mutation-positive advanced/metastatic non-small-cell lung cancer: safety run-in results from the FLAURA2 study. ESMO Open. 2021;6(5) doi: 10.1016/j.esmoop.2021.100271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zeng Q., et al. Discovery and evaluation of clinical candidate AZD3759, a potent, oral active, central nervous system-penetrant, epidermal growth factor receptor tyrosine kinase inhibitor. J. Med. Chem. 2015;58(20):8200–8215. doi: 10.1021/acs.jmedchem.5b01073. [DOI] [PubMed] [Google Scholar]
- 183.Ozcan G., Singh M., Vredenburgh J.J. Leptomeningeal metastasis from non–small cell lung cancer and current landscape of treatments. Clin. Cancer Res. 2023;29(1):11–29. doi: 10.1158/1078-0432.CCR-22-1585. [DOI] [PubMed] [Google Scholar]
- 184.Ahn M.J., 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(11):891–902. doi: 10.1016/S2213-2600(17)30378-8. [DOI] [PubMed] [Google Scholar]
- 185.Wu Y.-L., et al. Randomized phase 3 study of first-line AZD3759 (zorifertinib) versus gefitinib or erlotinib in EGFR-mutant (EGFRm+) non–small-cell lung cancer (NSCLC) with central nervous system (CNS) metastasis. J. Clin. Oncol. 2023;41(16_suppl):9001. 9001. [Google Scholar]
- 186.Lee J., Hong M.H., Cho B.C. Lazertinib: on the way to its throne. Yonsei Med. J. 2022;63(9):799–805. doi: 10.3349/ymj.2022.63.9.799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Yun J., et al. YH25448, an irreversible EGFR-TKI with potent intracranial activity in EGFR mutant non–small cell lung cancer. Clin. Cancer Res. 2019;25(8):2575–2587. doi: 10.1158/1078-0432.CCR-18-2906. [DOI] [PubMed] [Google Scholar]
- 188.Cho B.C., et al. Lazertinib versus gefitinib as first-line treatment in patients with EGFR-mutated advanced non–small-cell lung cancer: results from LASER301. J. Clin. Oncol. 2023;41(26):4208–4217. doi: 10.1200/JCO.23.00515. [DOI] [PubMed] [Google Scholar]
- 189.Brazel D., Nagasaka M. The development of amivantamab for the treatment of non-small cell lung cancer. Respir. Res. 2023;24(1):256. doi: 10.1186/s12931-023-02558-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Zhou C., et al. Amivantamab plus chemotherapy in NSCLC with EGFR exon 20 insertions. N. Engl. J. Med. 2023;389(22):2039–2051. doi: 10.1056/NEJMoa2306441. [DOI] [PubMed] [Google Scholar]
- 191.Midha A., Dearden S., McCormack R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII) Am. J. Cancer Res. 2015;5(9):2892–2911. [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Data available upon request.



