Leptomeningeal metastasis (LM), also known as leptomeningeal carcinomatosis (LC), is a devastating complication of metastatic cancer that occurs when neoplastic cells invade the meningeal space. Diagnosis of LM remains challenging given the heterogeneous signs and symptoms at presentation and requires thorough neurological examination, cerebrospinal fluid (CSF) analysis, and MRI of the brain and spine with gadolinium. Detecting neoplastic cells in the CSF is the gold standard for diagnosing leptomeningeal metastases; however, it has low sensitivity and may require multiple CSF samples. New emerging technologies, such as liquid biopsy of CSF, have increased sensitivity and specificity for detecting circulating tumor cells in CSF. The management of LM in patients with NSCLC requires an individualized multidisciplinary approach. Treatment options include surgery for ventricular shunt placement, radiation therapy to bulky or symptomatic disease sites, systemic or intrathecal chemotherapy, molecularly targeted agents, and, more recently, immunotherapy. Targeting actionable mutations in LM from NSCLC, such as EGFR tyrosine kinase inhibitors or anaplastic lymphoma kinase gene rearrangement inhibitors, has shown encouraging results in terms of disease control and survival. Although there are limited data regarding the use of immunotherapy in LM, immunotherapy has produced promising results in several case reports. In this review, we focused on the epidemiology, pathophysiology, clinical presentation, diagnosis, and current treatment strategies, with a special emphasis on novel agents, including targeted therapies and immunotherapy of LM in patients with NSCLC.

Leptomeningeal metastasis (LM) is a devastating complication of metastatic cancer, which occurs when neoplastic cells invade the meningeal space. LM is found in approximately 5% of patients with malignant tumors and most commonly occurs in patients with lung cancer, breast carcinoma, and melanoma (1). The postmortem series showed an incidence of LM of approximately 20% or more with many solid tumors (2–5), suggesting that LM might often be underdiagnosed. Diagnosis of LM remains challenging given the heterogeneous presenting signs and symptoms, and requires a thorough neurological examination, cerebrospinal fluid (CSF) analysis, and MRI of the brain and spine with gadolinium.

In the past, the presence of LM often represented a failure of treatments for the metastatic disease, which sometimes represents the final event for the patient and the almost total absence of effective treatments before molecular-targeted therapies and immunotherapy. The management of LM in patients with non—small cell lung cancer (NSCLC) requires a multidisciplinary approach. Treatment options include surgery for ventricular shunt placement, radiation therapy to bulky or symptomatic disease sites, systemic or intrathecal chemotherapy, molecular-targeted therapy, and immunotherapy. The recent development of molecular-targeted therapy and immunotherapy with better CNS penetration is changing the landscape of LM management, but the prognosis of LM remains dismal. This review focuses on the epidemiology, pathophysiology, clinical presentation, diagnosis, and current treatment strategies, with a particular emphasis on novel agents, including targeted therapies, for leptomeningeal disease in patients with NSCLC.

LM is found in approximately 5% of patients with malignant tumors and most commonly occurs in patients with lung cancer, breast carcinoma, and melanoma (1). The incidence of LM in patients with NSCLC was 3.4% in molecularly unselected patients and higher in ALK-rearranged (10.3%) and EGFR-mutant subgroups (9.4%; refs. 6, 7). NSCLC with mutant EGFR and altered anaplastic lymphoma kinase (ALK) genes is more likely to relapse with LM (8). Concomitant brain metastases affect one-third of the patients. The median overall survival time of patients with NSCLC with LM remains grim and ranges from 3.6 to 11 months (9, 10), mostly related to the use of novel therapies (11). The European Association of Neuro-Oncology (EANO) class IIA patients (LM with a typical linear enhancement of meninges in MRI and typical neurological symptoms, negative/inconclusive CSF cytology) had the longest overall survival (OS), whereas type I LM (LM with positive CSF cytology) patients had the shortest OS (12). A retrospective study including 149 patients with NSCLC with LM reported that treatment with EGFR-TKIs, normal CSF flow, lack of fixed neurological impairments, and good performance status were all associated with a better outcome, whereas poor performance status, high CSF protein level, and high initial CSF WBC count were associated with poor prognosis (13). Yin and colleagues developed a molecular graded prognostic assessment (molGPA) model specific for estimating survival in lung cancer patients with leptomeningeal metastases (14). In this model, positive EGFR/ALK, Karnofsky performance score (KPS) of 60 and more, and the lack of extracranial metastasis (ECM) were associated with better overall survival rate. The molGPA model stratified the patients in three groups: high, moderate, and low risk. In the training set, the median OS for high, moderate, and low risk LM patients was 0.3, 3.5, and 15.9 months, respectively (P < 0.001; ref. 14).

The brain and spinal cord are covered by meninges consisting of three layers known as the dura mater, arachnoid, and pia mater, from superficial to deep, respectively. The meningeal system can also be divided into pachymeninges, which only contains the dura mater, and leptomeninges, which contains the arachnoid and pia mater. The space between the arachnoid and the pia mater is called the subarachnoid space and is filled with CSF. Tumoral involvement of the dura mater (pachymeninges) should be differentiated from the tumoral involvement of the leptomeninges as the tumor spreads to the leptomeninges, which enables tumor cells to spread through the CSF.

Invasion of malignant cells into the leptomeningeal system is thought to occur through a variety of routes, including direct seeding from the brain parenchyma, hematogenous seeding (especially in hematologic cancers), and dura, bone, and endoneurial/perineural invasion. Cancer cells in CSF encounter physiologic challenges such as inflammation and limited micronutrient. Chi and colleagues designed a study investigating the cancer cell mechanism to overcome this effect (15). They reported that cancer cells appear to outcompete macrophages for iron, allowing them to thrive in the CSF. Chi and colleagues discovered that the iron-binding protein lipocalin-2 (LCN2) and its receptor SCL22A17 were by cancer cells but not macrophages in the CSF. These macrophages have been shown to produce inflammatory cytokines that stimulate LCN2 expression in cancer cells, but they do not produce LCN2. The LCN2/SLC22A17 system promotes cancer cell proliferation in LM animal models, whereas iron chelation treatment inhibits it (15). Boire and colleagues discovered that component 3 (C3) was upregulated in the leptomeningeal cancer cells and shown to be necessary for cancer cell growth in CSF (16). Boire and colleagues reported that the C3a receptor in the choroid plexus epithelium is activated by cancer cell-derived C3, which disrupts the blood–CSF barrier, which enables plasma components such as amphiregulin and other mitogens to enter the CSF and support cancer cell overcome mitogen-poor microenvironment of CSF (16). There is also literature suggesting an increased risk of developing LM in patients who have undergone neurosurgic metastatic resection compared with patients who have not undergone neurosurgery, suggesting iatrogenic spread (17–19).

The initial clinical presentation of LM is often subtle. Multifocal neurologic involvement in a patient with known cancer should raise clinical suspicion of LM. The initial manifestations may include cranial nerve palsies, headaches, back pain, visual disturbances, diplopia, hearing deficits, changes in cognition, radiculopathies, myelopathies, or spinal cord syndromes such as cauda equina syndrome (3). In a series of 150 patients with solid tumor LM from Memorial Sloan Kettering Cancer Center (MSKCC) who were followed up between 2002 and 2004, the most common presenting signs and symptoms were headache (39%), nausea and vomiting (25%), leg weakness (21%), cerebellar signs (17%), altered mental status (16%), diplopia (14%), facial weakness (13%), back pain (12%), leg numbness (12%), facial weakness (8%), and facial numbness (6%; refs. 1, 20). In the same cohort (20), dizziness, fatigue, gait difficulty, aphasia, vision loss, hearing loss, dysarthria, meningeal irritation, arm pain, leg pain, and bowel/bladder dysfunction were reported at presentation in ≤5% of the patients.

The differential diagnoses include subacute to chronic meningitis, skull base/dural/parenchymal metastases, primary leptomeningeal melanomatosis, and metabolic and toxic encephalopathies (21).

The diagnosis of LM is based on neurological examination, CSF analysis, and radiographic findings. Positive CSF cytology is considered the gold standard for the diagnosis of LM, although multiple CSF samplings may be necessary. The sensitivity can be increased up to 75% and 85% with a second CSF analysis (22) compared with the sensitivity of the initial CSF analysis, which is reported to be as low as 50%. For the diagnosis, treatment, and follow-up of patients with LM from solid tumors, the European Association of Neuro-Oncology-European Society of Medical Oncology (EANO-ESMO) group has developed a diagnostic flowchart that contains neurologic symptoms, imaging, and CSF cytology (23). On the basis of the combination of these three factors, LM can be classified as type I (positive CSF cytology) or type II (probable/possible), with typical MRI features and neurological symptoms. MRI findings had been classified as linear enhancement of meninges (subtype A), nodular enhancement of meninges (subtype B), both (subtype C), or hydrocephalus (subtype D). A retrospectively involving 254 patients with LM from solid tumors using the EANO-ESMO guidelines showed shorter OS in patients with type I compared with type II LM, whereas systemic or intrathecal treatment is linked to better OS in type I LM, but not in type II LM (24), albeit this has to be investigated further in larger datasets and prospective studies. Diagnostic tests and imaging techniques are discussed below.

Diagnostic tests

The definitive diagnosis of LM is based on direct visualization of tumor cells in the CSF by cytology. However, positive CSF cytology has high specificity and sensitivity, between 80% and 95%, which might require repeated LP (25). Obtaining large CSF volumes (>10 mL), processing CSF specimens promptly, and obtaining CSF from the closest site for symptoms or radiologic involvement have been shown to increase the sensitivity of CSF cytology results (25, 26). Notably, 20% of individuals with clinically or radiographically unambiguous LM were reported to have a negative CSF evaluation (1, 25), in which cases the diagnosis can be made in the clinical context supported by neuroimaging findings alone (27). High protein content, low glucose concentration, lymphocytic pleocytosis, and positive cytology for malignant cells are all characteristic CSF findings of LM. Although most individuals do not have all of these characteristics, it is unusual to have a completely normal CSF examination (3, 25, 26). LP is a relatively safe procedure, but adverse events related to LP include hemorrhage, post-LP headache, bleeding, cerebral herniation, minor neurologic symptoms such as radicular pain or numbness, and back pain (28). EANO-ESMO clinical practice guidelines recommend through physical exam, CSF cytology, and cerebrospinal MRI as part of diagnostic workup for any patients with cancer who presented with concerning symptoms for LM (23). CSF cytology is also used classification of LM (Type 1: confirmed with CSF cytology, Type 2: unequivocal/negative CSF cytology), which can be used in prognostication (29). EANO-ESMO clinical practice guidelines also recommend repeating LP, if initial CSF analysis unequivocal/negative CSF in patients with suspected LM (23).

Measuring serum and CSF tumor markers in a clinically appropriate context can aid in the diagnosis of LM, as several studies have shown elevated tumor markers (e.g., CEA, PSA, CA-15-3, CA-125, MART-1, and MAGE-3 in melanoma) in CSF compared with serum, suggesting LM even with negative CSF cytology (30–34).

Emerging diagnostic techniques such as liquid biopsy can provide helpful information for LM diagnosis and monitoring. CTCs and circulating cell-free tumor DNA (ct-DNA) in the CSF are the two most well-developed biomarkers that can be detected by liquid biopsy. Several studies have found that the CellSearch approach, which uses immunomagnetic selection, identification, and quantification of CTCs in the CSF, is more sensitive than traditional cytology and MRI in detecting leptomeningeal metastases (35–37). A few studies reported that CTC in CSF could be predictive of survival in LM (38, 39); however, given the small sample size of studies, larger and prospective cohorts will be useful in determining the cut-off value (40). CTCs liquid biopsy can also isolate single CTCs to screen for genetic abnormalities common to solid tumors. One study reported a highly concordant molecular profile (89.5%) among CTCs from the CSF of patients with LM from EGFR-mutated or ALK-rearranged NSCLC and their primary tumors (39). Liquid biopsy for detecting CTCs in the CSF has been shown to increase the sensitivity and specificity of diagnosing LM secondary to epithelial tumors in the right clinical context (36, 41). One study included 81 patients with a clinical suspicion of LM but an unequivocal MRI to compare the performance of CTC in CSF and cytology at diagnosing LM; the sensitivity of CTC assay was 94% and specificity was 100%, whereas the sensitivity of cytology was reported to be 76% (42). Another study showed that a cutoff of ≥1 CSF-CTC/mL had superior sensitivity and specificity compared with integrated clinical diagnosis (clinical suspicion with positive CSF cytology or unequivocal neuroimaging findings; ref. 41). Furthermore, CSF ct-DNA analysis has been demonstrated to be effective in enhancing LM diagnosis (43). Using ct-DNA from CSF for detecting genetic alterations in brain tumors has substantially better sensitivity than using plasma (43). One study involving 26 patients with LM secondary to EGFR showed that driver genes were found in 100% (26/26) of CSF cell-free DNA (cfDNA), 84.6% (22/26), and 73.1% (19/26) of CSF precipitates and plasma samples, respectively (44). The study also reported that CSF ct-DNA was superior to plasma in detecting loss of heterozygosity of TP53 (73% vs. 7.7%, P < 0.001; ref. 44). Next-generation sequencing of CSF to risk-stratify patients with brain metastasis secondary to lung adenocarcinoma was demonstrated in research comprising 94 patients with brain metastasis secondary to lung adenocarcinoma (45). The researchers identified five molecular subtypes associated with different overall survival. They also reported that EGFR mutations in conjunction with CDK4, CDK6, MYC, and MET were related to poor outcomes (45). Zheng and colleagues compared the CSF gene sequencing of two cohorts of people. In the first cohort, patients with LM secondary to EGFR mutated NSCLC who underwent CSF and plasma genotyping before initiation of the first dose of Osimertinib, patients with an EGFR exon 19 deletions exhibited a longer median intracranial progression-free survival (iPFS) than those with an EGFR exon 21 L858R mutation (11.9% vs. 2.8%; ref. 46). The second cohort involved patients with EGFR-mutated advanced NSCLC who developed LM while on Osimertinib therapy. In cohort 2, patients with T790M loss in the CSF showed a lower median iPFS than those with T790M reserved (7.4 months vs. 13.6 months, P = 0.01; ref. 46).

Overall, liquid biopsy from CSF can be useful for diagnosis, detection of genetic mutations, and monitoring therapy responses in LM, but further studies are needed to establish clear cut-offs and standardize different techniques. There is a necessity to update the guidelines in the light of new advancements.

Imaging findings

MRI of the brain and full spine with gadolinium enhancement is the gold standard radiologic modality for LM imaging and has been shown to be superior to CT imaging (47, 48). Sing and colleagues (49) reported that standard contrast-enhanced T1-weighted MR sequences are more sensitive than contrast-enhanced fast fluid attenuation inversion recovery (FLAIR) sequences in detecting intracranial neoplastic leptomeningeal disease. Optimally, imaging studies should be completed before lumbar puncture (LP) to prevent false-positive leptomeningeal enhancement secondary to inflammation from the LP (50). Approximately 20% to 30% of patients with LM report having a normal MRI at the time of diagnosis (51). MRI findings consistent with or suggestive of LM include leptomeningeal, subependymal, dural, or cranial nerve enhancement; superficial cerebral lesions; and communicating hydrocephalus (1, 5, 42). Ko and colleagues conducted a retrospective study to investigate MRI findings in 283 patients with LM from NSCLC and reported that positive MRI findings were suggestive of a heavier disease burden than negative imaging findings in patients with LM who died from CNS causes (52). An example of MRI involvement in LM is shown in Figs. 1 and 2.

Figure 1.

Lumbosacral leptomeningeal carcinomatosis from NSCLC.

Figure 1.

Lumbosacral leptomeningeal carcinomatosis from NSCLC.

Close modal
Figure 2.

MRI T1 sequence showing leptomeningeal carcinomatosis from NSCLC.

Figure 2.

MRI T1 sequence showing leptomeningeal carcinomatosis from NSCLC.

Close modal

The sensitivity of CT scan is reported to be 23% and 38%; hence, it should only be used for patients who are unable to undergo MRI (53). Radionuclide ventriculography studies using technetium-99m-DTPA (Tc-99) or 111Indium-DTPA are not indicated for diagnosis but can be useful in evaluating patency of the ventricular system before intrathecal chemotherapy administration. Abnormalities in radionuclide CSF studies have been shown to be associated with poor outcomes and increased treatment-related toxicities (54–56).

Current treatment for LM requires a multidisciplinary approach. The goals of treatment are to stabilize neurologic symptoms, improve quality of life, and prolong survival with minimal toxicity. Patients with solid tumors and leptomeningeal metastases are divided into two categories according to the US National Comprehensive Cancer Network recommendations: good risk and poor risk. According to the NCCN guidelines (57), best supportive care is recommended for patients in the poor risk category, which includes limited performance status, significant and major neurologic impairments, broad systemic disease with few therapeutic choices, bulky CNS disease, and encephalopathy, and intensive systemic treatment is indicated for individuals in the good risk category (characterized by satisfactory performance status, no substantial neurological abnormalities, limited systemic disease, and reasonable systemic treatment alternatives if needed). This classification may not apply to individuals with NSCLC who have actionable mutations, as some of the most recent molecular treatments have demonstrated high blood–brain barrier penetration and promising antitumor efficacy. The formulation of new guidelines for this group of patients is warranted in light of these promising action (58, 59).

Radiotherapy (RT)

Information to guide treatment decisions is scarce because LM accounts for a small percentage of CNS metastases (11%–20%; ref. 60). RT is commonly used for symptom relief, CSF flow correction, and debulking during preparation for chemotherapy (61). Focal RT may be indicated for clinically symptomatic sites and bulky disease. Yan and colleagues analyzed 51 EGFR-mutated patients with NSCLC with LM and reported that WBRT did not improve the objective response rate (ORR) or intracranial disease control rates (DCR; ref. 62). Li and colleagues analyzed 184 patients with LM secondary to NSCLC and reported OS benefits in patients who received TKI therapy (10 months vs. 3.3 months, P < 0.001), but 42 patients who received WBRT did not have a longer OS than those who did not receive WBRT, and a combination of WBRT and TKIs had no additional survival benefit (7). A retrospective study reported that WBRT was associated with better prognosis in patients with NSCLC with LM (13). A systematic review of eight studies involving 389 patients with NSCLC and LM found no conclusive evidence that WBRT prolonged survival (63).

There is no universally accepted plan for palliative WBRT dose fractionation, although common variations include 20 Gy/5 or 30 Gy/10 administered daily, 5 days a week (64, 65). In certain institutions, lengthier schedules (such as 37.5 Gy/15 and 40 Gy/20) may occasionally be employed (64). Acute adverse effects related to WBRT include cerebral edema, nausea, vomiting, headache, anorexia, seizures, dermatitis, and malaise, whereas late AEs include chronic fatigue, neurocognitive impairment, cerebrovascular damage, and pituitary malfunction. These side effects might be limiting factors to the utility of the treatment in practice. Ongoing clinical trials are investigating the role of WBRT, including a phase one trial for WBRT combined with avelumab in patients with solid tumors (NCT03719768), and a phase III trial for WBRT combined with veliparib among NSCLC (NCT01657799).

There are also limited data available on the role of craniospinal irradiation (CSI) in LM. A retrospective study involving 25 patients with LM who received craniospinal radiation reported stabilization of neurological symptoms in 40% of the cohort, but the grade 3 myelosuppression rate was 32% (66). A recent phase-1 clinical trial involving 24 patients treated with proton CSI in LM secondary to solid tumors reported a median CNS PFS of 7 months and OS of 8 months (67). In this study, 2 of 24 patients developed Grade 4 thrombocytopenia and lymphopenia and/or grade 3 fatigue (67). An ongoing phase I trial (NCT03520504) is investigating proton radiation to the brain and spinal cord among patients with LM from solid tumors.

Systemic and intrathecal chemotherapy

For patients with LM from NSCLC with systemic metastasis who do not have actionable mutations, systemic chemotherapy is the treatment of choice because it has been shown to be an independent predictor of survival (9, 51). There is no consensus regarding the standard of chemotherapy, and promising results with agents such as bevacizumab and pemetrexed have been reported (68, 69). Targeted therapies are discussed in a separate section.

A pooled analysis showed that intrathecal chemotherapy is an effective treatment for individuals with LM from NSCLC (69); however, the best drug, dose, and regimen have yet to be determined. Methotrexate, cytarabine, and thiotepa are the most regularly utilized intrathecal chemotherapeutic drugs (5, 51), but none of the intrathecal chemotherapy (IT) regimens have been shown to be superior (5). In a phase I study of 13 patients with LM and NSCLC who received weekly intrathecal pemetrexed in addition to twice-weekly systemic pemetrexed, the ORR was 31% and 54%, respectively (70). A phase II clinical trial involving 30 patients with LM secondary to EGFR-mutated NSCLC who progressed on TKI therapy treated with IT pemetrexed combined with dexamethasone showed an 84.6% of clinical response rate (22/26), and median OS was 9 months (71, 72). Most frequent reported adverse effect was myelosuppression (30%) and recommended pemetrexed dose was 50 mg (71). A randomized clinical trial involving 34 patients with LM secondary to solid tumors (21/34 NSCLC) investigated the role of IT pemetrexed (10 mg) followed by involved-field RT for 3 days and showed a clinical response rate of 68%, median OS of 5.5 months; however, 53% of patients developed adverse events (myelosuppression, liver injury, radiculitis) including 6 patients with Grade 3, and 1 patient with Grade 4 adverse events (AE; ref. 73).

IT chemotherapy can be administered via injection during a LP or by placing an Ommaya catheter, which is a small implantable device placed under the scalp that drains into the lateral ventricle through a tube. Compared with LP, Ommaya catheter offers multiple benefits, including less painful administration of IT and more homogenous drug delivery; however, Ommaya catheter has its own drawbacks, including the necessity of surgery for placement of the catheter, complications such as infections, intracranial bleeding, and symptomatic leukoencephalopathy (74). One study showed an OS benefit with intraventricular chemotherapy via the Ommaya catheter compared with LP (9.2 months vs. 4 months, P = 0.0006; ref. 75). Glantz and colleagues (76) conducted a randomized controlled trial and found that intraventricular methotrexate was associated with a higher rate of progression-free survival than intrathecal methotrexate (43 days vs. 19 days, P = 0.048).

Molecular-targeted agents

In select patients with NSCLC with CNS involvement, including LM, systemic application of molecularly targeted treatments has shown clinical benefits (77). Multiple successful studies have led to the approval of molecularly targeted medicines with increased CNS penetration (61). Current strategies for targeted therapies are as follows:

Role of EGFR tyrosine kinase inhibitors in LM from NSCLC

LM occurs in approximately 9% of individuals with NSCLC with EFGR mutations (78). A single-center retrospective analysis including 136 patients with LM from EGFR-mutated NSCLC reported longer OS in patients treated with TKIs compared with those who did not receive TKIs (10.0 m vs. 3.3 m, P < 0.001). The analysis also demonstrated that WBRT did not provide further OS benefit (79). Selected studies regarding EGFR TKI treatment for patients with EGFR mutated NSCLC listed in Table 1.

Table 1.

Selected studies on EGFR TKIs in patients with LM from EGFR mutated NSCLC.

PublicationType of studyNo. of patientsPatient characteristicsPrevious chemotherapyPrior TKI therapyTreatment regimenResponse to therapy
Yi et al. (126Retrospective (2022) 27 EGFR positive NSCLC with LM 4/27 patients had cytoreductive chemotherapy, and 4/27 patients had WBRT prior to LM diagnosis 24/27 patients had prior TKI exposure Arm 1: osimertinib plus bevacizumab group (n = 16) Median OS:
  • -Arm 1: 18 months

  • -Arm 2: 13.7 months (P = 0.046)

 
      Arm 2: osimertinib alone (n = 11) iPFS:
  • -Arm 1: 10.6 months

  • -Arm 2: 5.5 months

 
Zhang et al. (127Case report (2022)  EGFR-positive (exon 21 mutation) NSCLC with LM, which progressed on osimertinib Patient has not received only resection for lung cancer before development of LM. After developing LM, patient received osimertinib with bevacizumab Osimertinib Osimertinib (80 mg daily), bevacizumab (7.5 mg/kg q3w) PFS: 6 months OS: 13 months 
Zhang et al. (128Retrospective (2021) 78 EGFR-positive NSCLC with LM (44/78 patients had T790M mutation) 66.0% patients received cytotoxic chemotherapy, and 20.8% had undergone WBRT prior to developing LM 53/78 patients had prior TKI exposure Arm 1: first-/second- generation TKI Median OS:
  • -Arm 1: 3 months

  • -Arm 2: 13.15 months

 
      Arm 2: osimertinib PFS
  • -Arm 1: 1.50 months

  • -Arm 2: 9.50 months

 
       Regardless of T790M status, the osimertinib arm had longer OS 
Mizusaki et al. (129Case report (2021) NSCLC with EGFR mutation—exon 19 deletion Carboplatin + pemetrexed Afatinib Dacomitinib 30 mg daily Symptomatic improvement in 3 weeks and radiologic improvement in LM in 9 weeks 
Xu et al. (130Case series EGFR positive NSCLC with LM: 2/3 patients had multiple lines of cytotoxic chemotherapy and had WBRT prior to developing LM All patients had exposure to gefitinib and/or erlotinib Nimotuzumab (200 mg/m2) weekly + erlotinib (150 mg/day) 2/3 patients reported a radiologic response in within 6–8 weeks therapy 
   1/3 EGFR 19del without T790M     
   2/3 EGFR mutation (exon 19 deletion)     
Lee et al. (131Retrospective (2020) 351 EGFR-positive NSCLC with LM (87/351 with T790 mutation) Osimertinib arm: 45% received IT, 34% received WBRT 343/351 patients had prior TKI exposure prior to developing LM Osimertinib vs. control Median OS 17 months in patients treated with osimertinib vs. 5.5 months in patients who did not receive osimertinib 
    Control arm: 21% received cytotoxic chemotherapy, 53% received other EGFR TKIs, and <1% received immunotherapy   No difference in median OS according to T790M mutational status. Osimertinib had OS benefit regardless of T790M mutational status 
Yang et al. (88)
BLOOM study 
Phase I clinical trial (2020) 41 EGFR positive NSCLC with LM: 21 unselected, 20 with T790M mutation 35 patients received prior chemotherapy All had prior EGFR therapy (31 on gefitinib, 7 on erlotinib, 2 on afatinib, 1 on dacomitinib) Osimertinib 160 mg daily PFS was 8.6 months (95% CI, 5.4–13.7 months); median OS was 11.0 months. (95% CI, 8.0–18.0 months). CSF tumor cell clearance was confirmed in 11 (28%; 95% CI, 15%–44%) of 40 patients. Neurologic function was improved in 12 (57%) of 21 patients with an abnormal assessment at baseline. 
Park et al. (132Phase II (2020) 40 EGFR T790M-positive patients with non–small cell lung cancer with brain metastases or leptomeningeal disease who progressed on prior EGFR TKI therapy. Not specified All patients had been treated with TKIs priorly, including standard dose osimertinib 80 mg daily Osimertinib 160 mg daily In LM group: intracranial DCR, 92.5%; CR, 12.5%
Median OS: 13.3 months
Median PFS was 8.0 months 
   Arm 1: brain metastasis without LM     
   Arm 2: LM with and without brain metastasis     
Ahn et al. (89Retrospective analysis (2020) 22 EGFR T790M-positive patient with non–small cell lung cancer with LM who had been treated with EGFR-TKI from AuRA-3, AURA-17, AURA extensions, and AURA-2 trials 64% of patients had cytotoxic chemotherapy, while 41% of patients had WBRT in prior study All patients had EGFR-TKI exposure in prior Osimertinib 80 mg daily ORR 55% 
       Median OS: 18.8 months 
Sakaguchi et al. (133Case report (2020) NSCLC with EGFR mutation—exon 19 deletion Carboplatin+ paclitaxel N/A Gefitinib (250 mg/day) + WBRT No recurrence after 43 months 
       Switched to erlotinib due to resistance 
Saboundji et al. (134Retrospective (2018) 20 NSCLC with EGFR positive LM:
  • -Exons 18 (2/10),

  • -Exon 19 (7/20)

  • -Exon 21 (11/20)

 
Patients had received a mean of 2.3 treatment lines prior to osimertinib therapy All patients had EGFR-TKI exposure in prior Osimertinib 80 mg daily Median PFS: 17.2 months 
       Median OS: 18 months 
Cho et al. (135Prospective 18 NSCLC with EGFR positive LM who progressed on EGFR-TKIs Not mentioned All patients had EGFR-TKI exposure in prior Arm 1: AZD3759 200 mg daily 5/18 patients had radiologic response. 3/18 patients had clearance of tumor cells in CSF after 2 consecutive assessments. 
      Arm 2: AZD3759 300 mg daily  
Nanjo et al. (136Retrospective (2018) 13 10/13 patients: EGFR exon 19 mutation, None of the patients had IT chemotherapy. 3/13 patients had prior WBRT. All patients had prior first- or second-generation TKI exposure Osimertinib 80 mg daily PFS: 2 months,
OS: 3.8 months 
   3/13 patients: EGFR exon 21 mutation     
Tamiya et al. (85Prospective trial (2017) 11 Confirmed EGFR mutation positive NSCLC with LMC:
  • 5 patients - exon 19 deletion

  • 3 patients - p.L858R point mutation

  • 3 patients - exon 18 mutation

 
1–3 lines of chemotherapy in all patients 9/11 patients had prior TKI therapy Afatinib 40 mg daily PFS: 2 months, OS: 3.8 months 
Gong et al. (137Retrospective review (2015) 21 10 patients: EGFR exon 21-point mutation Not reported 5/21 prior icotinib exposure Icotinib (125 mg, 3 times daily) Median survival: 10.2 months 
   11 patients: EGFR exon 19 deletion   Icotinib (250 mg, 3 times daily for patients with prior icotinib exposure)  
Liao et al. (9Retrospective review (2015) 212 EGFR mutation positive NSCLC with LM:
  • 68 patients - exon 19 deletion or p.L858R point mutation

  • 26 patients - wild type (exon 20 insertion, exon 21 mutation, exon 18 mutation)

 
78/212 patients received CT 129/212 patients had prior TKI exposure WBRT, EGFR TKIs (gefitinib or erlotinib or afatinib), WBRT TKI therapy and cytotoxic chemotherapy after diagnosis of LM remained the independent factors predictive of extended survival in the multivariate analysis 
Jackman et al. (138Phase I (2015) 5 patients: EGFR exon 19 deletions 2/7 patients had prior systemic therapy (not specified), and 6/7 patients had WBRT prior to LM diagnosis All patients had prior TKI exposure (erlotinib, gefitinib, vandetanib) 2 weeks of high-dose gefitinib (750–1,000 mg/day) and 2 weeks of 500 mg/day Median OS: 3.5 months
Median PFS: 2.3 months 
   1 patient:
  • EGFR L858R mutation

  • 1 patient had insufficient tissue for mutation analysis

 
    
Yang et al. (139Retrospective study (2015) NSCLC with EGFR positive LM with gefitinib resistance All patients had received cytotoxic chemotherapy prior All patients had prior gefitinib exposure with initial response, but all of them developed resistance Pemetrexed +cisplatin +erlotinib Median OS: 9 months
  • CR: 1/6

  • PR: 2/6

 
Kawamura et al. (80Retrospective study (2015) 35 NSCLC with EGFR positive LM who progressed on standard-dose EGFR-TKIs All patients had received cytotoxic chemotherapy All patients had prior standard dose TKI exposure and developed resistance later Arm 1: high-dose erlotinib (200–600 mg/day every 2–4 days) Median OS:
  • -Arm 1: 6.2 months

  • -Arm 2: 5.9 months

 
      Arm 2: standard dose erlotinib (150 mg/day)  
Lee et al. (140Retrospective review (2013) 25 NSCLC with LM:
  • 9 patients: EGFR exon 21-point mutation

  • 8 patients: EGFR Exon 19 deletion

 
Intrathecal methotrexate in all patients 9 /25 patients had prior TKI exposure Arm 1: gefitinib (250 mg/day) Better radiologic response in erlotinib arm (9/14 patients) when compared with gefitinib arm (1/11 patients) 
      Arm 2: erlotinib (150 mg/day)  
Grommes et al. (141Retrospective study (2011) NSCLC with EGFR exon 3/9 patients had various lines of chemotherapy All patients developed progression on regular daily dose of erlotinib or other TKIs Pulsatile high-dose erlotinib (1,500 mg weekly) Radiological response in 6/9 patients (66.7%) 
       Median OS: 12 months 
Yi et al. (82Retrospective study (2009) 11 EGFR-mutated NSCLC with LM 8/11 patients had prior chemotherapy 6/11 patients had prior EGFR-TKI exposure Standard dose erlotinib (150 mg/day) n = 9 or High dose gefitinib (n = 2) followed by erlotinib 9/11 patients showed clinical improvement 
So et al. (81Case series (2009) EGFR-mutated NSCLC with LM Both patients had two lines of chemotherapy before the diagnosis of LC No prior EGFR_TKI therapy Gefitinib and VP shunt placement Neurologic symptom relief seen in both patients. OS was 5 months in first patient and 15 months in second patient. 
PublicationType of studyNo. of patientsPatient characteristicsPrevious chemotherapyPrior TKI therapyTreatment regimenResponse to therapy
Yi et al. (126Retrospective (2022) 27 EGFR positive NSCLC with LM 4/27 patients had cytoreductive chemotherapy, and 4/27 patients had WBRT prior to LM diagnosis 24/27 patients had prior TKI exposure Arm 1: osimertinib plus bevacizumab group (n = 16) Median OS:
  • -Arm 1: 18 months

  • -Arm 2: 13.7 months (P = 0.046)

 
      Arm 2: osimertinib alone (n = 11) iPFS:
  • -Arm 1: 10.6 months

  • -Arm 2: 5.5 months

 
Zhang et al. (127Case report (2022)  EGFR-positive (exon 21 mutation) NSCLC with LM, which progressed on osimertinib Patient has not received only resection for lung cancer before development of LM. After developing LM, patient received osimertinib with bevacizumab Osimertinib Osimertinib (80 mg daily), bevacizumab (7.5 mg/kg q3w) PFS: 6 months OS: 13 months 
Zhang et al. (128Retrospective (2021) 78 EGFR-positive NSCLC with LM (44/78 patients had T790M mutation) 66.0% patients received cytotoxic chemotherapy, and 20.8% had undergone WBRT prior to developing LM 53/78 patients had prior TKI exposure Arm 1: first-/second- generation TKI Median OS:
  • -Arm 1: 3 months

  • -Arm 2: 13.15 months

 
      Arm 2: osimertinib PFS
  • -Arm 1: 1.50 months

  • -Arm 2: 9.50 months

 
       Regardless of T790M status, the osimertinib arm had longer OS 
Mizusaki et al. (129Case report (2021) NSCLC with EGFR mutation—exon 19 deletion Carboplatin + pemetrexed Afatinib Dacomitinib 30 mg daily Symptomatic improvement in 3 weeks and radiologic improvement in LM in 9 weeks 
Xu et al. (130Case series EGFR positive NSCLC with LM: 2/3 patients had multiple lines of cytotoxic chemotherapy and had WBRT prior to developing LM All patients had exposure to gefitinib and/or erlotinib Nimotuzumab (200 mg/m2) weekly + erlotinib (150 mg/day) 2/3 patients reported a radiologic response in within 6–8 weeks therapy 
   1/3 EGFR 19del without T790M     
   2/3 EGFR mutation (exon 19 deletion)     
Lee et al. (131Retrospective (2020) 351 EGFR-positive NSCLC with LM (87/351 with T790 mutation) Osimertinib arm: 45% received IT, 34% received WBRT 343/351 patients had prior TKI exposure prior to developing LM Osimertinib vs. control Median OS 17 months in patients treated with osimertinib vs. 5.5 months in patients who did not receive osimertinib 
    Control arm: 21% received cytotoxic chemotherapy, 53% received other EGFR TKIs, and <1% received immunotherapy   No difference in median OS according to T790M mutational status. Osimertinib had OS benefit regardless of T790M mutational status 
Yang et al. (88)
BLOOM study 
Phase I clinical trial (2020) 41 EGFR positive NSCLC with LM: 21 unselected, 20 with T790M mutation 35 patients received prior chemotherapy All had prior EGFR therapy (31 on gefitinib, 7 on erlotinib, 2 on afatinib, 1 on dacomitinib) Osimertinib 160 mg daily PFS was 8.6 months (95% CI, 5.4–13.7 months); median OS was 11.0 months. (95% CI, 8.0–18.0 months). CSF tumor cell clearance was confirmed in 11 (28%; 95% CI, 15%–44%) of 40 patients. Neurologic function was improved in 12 (57%) of 21 patients with an abnormal assessment at baseline. 
Park et al. (132Phase II (2020) 40 EGFR T790M-positive patients with non–small cell lung cancer with brain metastases or leptomeningeal disease who progressed on prior EGFR TKI therapy. Not specified All patients had been treated with TKIs priorly, including standard dose osimertinib 80 mg daily Osimertinib 160 mg daily In LM group: intracranial DCR, 92.5%; CR, 12.5%
Median OS: 13.3 months
Median PFS was 8.0 months 
   Arm 1: brain metastasis without LM     
   Arm 2: LM with and without brain metastasis     
Ahn et al. (89Retrospective analysis (2020) 22 EGFR T790M-positive patient with non–small cell lung cancer with LM who had been treated with EGFR-TKI from AuRA-3, AURA-17, AURA extensions, and AURA-2 trials 64% of patients had cytotoxic chemotherapy, while 41% of patients had WBRT in prior study All patients had EGFR-TKI exposure in prior Osimertinib 80 mg daily ORR 55% 
       Median OS: 18.8 months 
Sakaguchi et al. (133Case report (2020) NSCLC with EGFR mutation—exon 19 deletion Carboplatin+ paclitaxel N/A Gefitinib (250 mg/day) + WBRT No recurrence after 43 months 
       Switched to erlotinib due to resistance 
Saboundji et al. (134Retrospective (2018) 20 NSCLC with EGFR positive LM:
  • -Exons 18 (2/10),

  • -Exon 19 (7/20)

  • -Exon 21 (11/20)

 
Patients had received a mean of 2.3 treatment lines prior to osimertinib therapy All patients had EGFR-TKI exposure in prior Osimertinib 80 mg daily Median PFS: 17.2 months 
       Median OS: 18 months 
Cho et al. (135Prospective 18 NSCLC with EGFR positive LM who progressed on EGFR-TKIs Not mentioned All patients had EGFR-TKI exposure in prior Arm 1: AZD3759 200 mg daily 5/18 patients had radiologic response. 3/18 patients had clearance of tumor cells in CSF after 2 consecutive assessments. 
      Arm 2: AZD3759 300 mg daily  
Nanjo et al. (136Retrospective (2018) 13 10/13 patients: EGFR exon 19 mutation, None of the patients had IT chemotherapy. 3/13 patients had prior WBRT. All patients had prior first- or second-generation TKI exposure Osimertinib 80 mg daily PFS: 2 months,
OS: 3.8 months 
   3/13 patients: EGFR exon 21 mutation     
Tamiya et al. (85Prospective trial (2017) 11 Confirmed EGFR mutation positive NSCLC with LMC:
  • 5 patients - exon 19 deletion

  • 3 patients - p.L858R point mutation

  • 3 patients - exon 18 mutation

 
1–3 lines of chemotherapy in all patients 9/11 patients had prior TKI therapy Afatinib 40 mg daily PFS: 2 months, OS: 3.8 months 
Gong et al. (137Retrospective review (2015) 21 10 patients: EGFR exon 21-point mutation Not reported 5/21 prior icotinib exposure Icotinib (125 mg, 3 times daily) Median survival: 10.2 months 
   11 patients: EGFR exon 19 deletion   Icotinib (250 mg, 3 times daily for patients with prior icotinib exposure)  
Liao et al. (9Retrospective review (2015) 212 EGFR mutation positive NSCLC with LM:
  • 68 patients - exon 19 deletion or p.L858R point mutation

  • 26 patients - wild type (exon 20 insertion, exon 21 mutation, exon 18 mutation)

 
78/212 patients received CT 129/212 patients had prior TKI exposure WBRT, EGFR TKIs (gefitinib or erlotinib or afatinib), WBRT TKI therapy and cytotoxic chemotherapy after diagnosis of LM remained the independent factors predictive of extended survival in the multivariate analysis 
Jackman et al. (138Phase I (2015) 5 patients: EGFR exon 19 deletions 2/7 patients had prior systemic therapy (not specified), and 6/7 patients had WBRT prior to LM diagnosis All patients had prior TKI exposure (erlotinib, gefitinib, vandetanib) 2 weeks of high-dose gefitinib (750–1,000 mg/day) and 2 weeks of 500 mg/day Median OS: 3.5 months
Median PFS: 2.3 months 
   1 patient:
  • EGFR L858R mutation

  • 1 patient had insufficient tissue for mutation analysis

 
    
Yang et al. (139Retrospective study (2015) NSCLC with EGFR positive LM with gefitinib resistance All patients had received cytotoxic chemotherapy prior All patients had prior gefitinib exposure with initial response, but all of them developed resistance Pemetrexed +cisplatin +erlotinib Median OS: 9 months
  • CR: 1/6

  • PR: 2/6

 
Kawamura et al. (80Retrospective study (2015) 35 NSCLC with EGFR positive LM who progressed on standard-dose EGFR-TKIs All patients had received cytotoxic chemotherapy All patients had prior standard dose TKI exposure and developed resistance later Arm 1: high-dose erlotinib (200–600 mg/day every 2–4 days) Median OS:
  • -Arm 1: 6.2 months

  • -Arm 2: 5.9 months

 
      Arm 2: standard dose erlotinib (150 mg/day)  
Lee et al. (140Retrospective review (2013) 25 NSCLC with LM:
  • 9 patients: EGFR exon 21-point mutation

  • 8 patients: EGFR Exon 19 deletion

 
Intrathecal methotrexate in all patients 9 /25 patients had prior TKI exposure Arm 1: gefitinib (250 mg/day) Better radiologic response in erlotinib arm (9/14 patients) when compared with gefitinib arm (1/11 patients) 
      Arm 2: erlotinib (150 mg/day)  
Grommes et al. (141Retrospective study (2011) NSCLC with EGFR exon 3/9 patients had various lines of chemotherapy All patients developed progression on regular daily dose of erlotinib or other TKIs Pulsatile high-dose erlotinib (1,500 mg weekly) Radiological response in 6/9 patients (66.7%) 
       Median OS: 12 months 
Yi et al. (82Retrospective study (2009) 11 EGFR-mutated NSCLC with LM 8/11 patients had prior chemotherapy 6/11 patients had prior EGFR-TKI exposure Standard dose erlotinib (150 mg/day) n = 9 or High dose gefitinib (n = 2) followed by erlotinib 9/11 patients showed clinical improvement 
So et al. (81Case series (2009) EGFR-mutated NSCLC with LM Both patients had two lines of chemotherapy before the diagnosis of LC No prior EGFR_TKI therapy Gefitinib and VP shunt placement Neurologic symptom relief seen in both patients. OS was 5 months in first patient and 15 months in second patient. 

Abbreviations: CR, complete response; IT, intrathecal chemotherapy; PR, partial response; WBRT, whole-brain radiotherapy.

First-generation EGFR TKIs

Multiple case reports have described clinical and radiologic improvement with gefitinib therapy, a first-generation EGFR inhibitor, at standard doses (80, 81) or high doses (82) among patients with LM from lung adenocarcinoma. A retrospective study including 35 patients with LM from EGFR-mutated NSCLC who experienced disease progression after failure of standard-dose EGFR-TKIs showed that high-dose erlotinib (various dosages and regimens of high-dose erlotinib were used: 200 mg on alternate days, 300 mg on alternate days, 300 mg every 3 days, 450 mg every 3 days, and 600 mg every 4 days) showed a radiologic response in 30% of patients, and symptomatic improvement in neurologic symptoms in 50% of patients. The median survival time from the diagnosis of LM in patients treated with high-dose erlotinib and those not treated with erlotinib was not statistically different (6.2. months in the erlotinib arm vs. 5.9 in the control arm, P = 0.94; ref. 80). A retrospective analysis involving 22 patients with LM from EGFR-mutant NSCLC showed that OS was longer in erlotinib-treated patients than in gefitinib-treated patients (6.6 months vs. 2.1 months, P = 0.07; ref. 83). Nanjo and colleagues reported that gefitinib resistance of PC-9/LMC-GR cells was associated with MET copy-number growth with MET activation, despite EGFR-T790M being negative in the acquired gefitinib-resistant mouse model (84). Furthermore, combined use of EGFR TKI with crizotinib, which has MET activity, led to a regression in LM with an acquired EGFR-TKI–resistant mouse model (84). In certain patients with LM related to NSCLC and prior TKI failure and overamplification of MEK, Cheng and colleagues proposed using a MEK inhibitor with a MEK inhibitor in addition to an EGFR-TKI (58).

Afatinib

Afatinib is a second-generation EGFR TKI, has been shown to be effective in patients with LM caused by EGFR-mutant NSCLC (85, 86), including at least one report of a patient with LM responding to afatinib following disease progression with osimertinib (87).

Osimertinib

Osimertinib is a third-generation EGFR TKI with considerable intracranial action. A multicenter phase I trial study (BLOOM; NCT02228369) involving 41 patients with LM from EGFR-mutated NSCLC who had disease progression on prior EGFR-TKI therapy showed an ORR of 62%, PFS of 8.6 months, and median OS of 11.0 months, and reported CSF clearance in 11/40 patients (88) with osimertinib 160 mg daily. The AURA-LM analysis examined the clinical efficacy of osimertinib 80 mg daily as a second-line treatment for EGFR T790M-NSCLC patients and demonstrated an ORR of 55%, CR of 27%, median PFS of 11.1, and median OS of 18.8 months (89). The phase III FLAURA trial showed overall prolonged OS with osimertinib compared with first-generation TKI in patients with previously untreated advanced EGFR-mutated NSCLC (38.6 m vs. 31.8 m, P = 0.0462; ref. 89). Four of the five patients in the osimertinib arm who had baseline radiologic evidence of LM obtained a full radiologic response, compared with one of the 2 patients with suspected LM in the comparator arm who exhibited a full radiologic response (89). Zheng and colleagues reported that median iPFS was significantly longer in patients with T790M-positive CSF genotyping (15.6 months) than in T790M-negative CSF (7.0 months; P = 0.04; refs. 46, 90).

AZD3759

AZD3759 is a novel, potent, oral EGFR TKI with an impressive CNS penetration (91). Yang and colleagues reported that free concentration of drug is equal in blood, CSF, or brain tissue and showed antitumor activity (92). A phase one clinical trial exhibited that AZD3759 at 200 mg twice daily showed a tolerable safety profile and good CNS penetration in EGFR-mutated NSCLC with brain or LM without any EGFR-TKI exposure or patients with LM who are pretreated with EGFR-TKIs (93). Grade 3 AE at the dose of 200 mg twice daily were skin and gastrointestinal (17%), hepatobiliary and renal (13%), and nutrition disorders (4%). No Grade 4 AE was reported.

Role of ALK inhibitors in LM from ALK mutated NSCLC

In individuals with NSCLC with ALK arrangements, LM develops in approximately 10% of cases (6). At the time of diagnosis, up to 40% of individuals with ALK-positive lung cancer were found to have CNS metastasis (94). The ALK inhibitors crizotinib, ceritinib, alectinib, brigatinib, and lorlatinib have been approved by the FDA for the treatment of patients with NSCLC with ALK rearrangement. Crizotinib is a first-generation ALK inhibitor with limited CNS penetration (95–97).

Second-generation ALK inhibitors include alectinib, brigatinib, and ceritinib, all of which have important clinical activities in patients with brain metastases, including those with LM. In the phase III ALEX trial (98), alectinib was compared with crizotinib among patients with treatment-naive ALK-positive NSCLC and showed prolonged investigator-assessed median PFS with alectinib (34.8 months vs. 10.9 months). Although OS data remain immature, the alectinib arm showed a higher 5-year OS rate compared with crizotinib (62.5% vs. 45.5%), and an OS benefit was observed in patients with or without brain metastasis. A case report demonstrated a sustained clinical and radiologic response to alectinib (>15 months) in a patient who developed symptomatic leptomeningeal disease while on crizotinib (99). A case series of four patients with ALK-positive NSCLC and LM who had progressed on crizotinib and ceritinib demonstrated radiographic and clinical improvement with alectinib (100). A few LM cases have demonstrated that switching from one ALK inhibitor to another or dose escalation can be effective in overcoming crizotinib resistance (101–103). Zou and colleagues conducted a retrospective study involving 65 patients with ALK-rearrangement NSCLC with brain metastasis or LM reported that need of mannitol or steroid decreased significantly in patients treated with alectinib, and 8/9 patients who had LM with or without BM reported improvement in CNS-related symptoms (104).

In early trials, brigatinib, a newer second-generation ALK inhibitor, demonstrated a reduction in brain metastases in patients previously treated with crizotinib and ALK inhibitor-naïve patients (101, 105). CNS PFS exceeded 14 months with modestly improved outcomes at 180 mg/day over 90 mg/day.

Lorlatinib is a third-generation ALK inhibitor with high CNS penetration. In an ongoing phase II study (NCT01970865) involving 197 patients with ALK-positive lung cancer whose disease progressed on prior ALK-TKI therapy, lorlatinib showed substantial intracranial activity in patients with ALK-positive NSCLC whose disease progressed on first- or second-generation ALK TKIs, regardless of baseline CNS involvement (106). Lorlatinib was active in two individuals with LM in this study, one of whom had a full and continued response at 22 months (106). In an interim analysis of the CROWN clinical trial (NCT03052608) involving 296 patients with previously untreated advanced ALK-positive lung cancer, patients who received lorlatinib showed longer PFS at 12 months (78% vs. 39%) and a higher frequency of intracranial response (83% vs. 23%) than those who received crizotinib (101, 107). A recent phase II clinical trial investigating the efficacy of Lorlatinib in patients with LM secondary to ALK-rearrangement NSCLC presented with CNS-only progression on the second-generation reported control of intracranial disease control in 12 weeks in 95% of patients, the intracranial objective response rate of 59%, and median PFS of 24.6 months (108). Patients who progressed only in CNC on the prior first- and second-generation of ALK inhibitor have a better response to Lorlatinib compared with patients with CNS and concomitant extracranial progression (108). Selected studies regarding ALK inhibitor treatment for patients with ALK rearranged NSCLC listed in Table 2.

Table 2.

Studies on ALK inhibitors in patients with LM from ALK-rearranged NSCLC.

StudyType of studyALK inhibitor usedNo. of patientsPrior chemotherapyPrior therapy with ALK inhibitorTreatment regimenResponse to therapy
Dagogo-Jack et al. (108Prospective Lorlatinib 23 patients with LM 35% of patients had prior systemic therapy All patients had CNS only progression on prior first (4%) or second generation ALK inhibitor (96%) Lorlatinib 100 mg daily Intracranial DCR in 12 weeks: 95% 
 Phase II (2022)      The intracranial objective response rate: 59% 
       Median PFS: 24.6 months 
Zou et al. (104Retrospective study (2022) Alectinib 65 patients with ALK-mutated lung cancer with brain metastasis or LM (9/65 LM) Not available 5/9 patients with LM were ALK-TKI naïve, 2/9 patients were crizotinib-resistant, and 3/9 patients had other second generation ALK-TKI exposure Alectinib 8/9 patients with LM reported improvement in CNS-related symptoms 
Chow et al. (142Prospective Ceritinib 18 LM 15/18 patients had prior chemotherapy 88% of patients had treated and progressed on crizotinib Ceritinib 750 mg once daily Whole-body ORR was 16.7% (95% CI, 3.6–41.4) and DCR was 66.7% (95% CI, 41.0–86.7) 
 Phase II (2022)       
Frost et al. (143Prospective (2020) Lorlatinib 9 LM All patients had at least one line of systemic chemotherapy All patients had prior ALK inhibitor exposure: crizotinib, ceritinib, alectinib, brigatinib Lorlatinib 100 mg daily Median duration of treatment: 10.4 months 
   36 brain metastases without LM    PFS: 8.0 months 
       Intracranial response rate: 54% 
Pellerino et al. (144Case report (2020) Lorlatinib Cisplatin and paclitaxel Crizotinib and ceritinib Lorlatinib 100 mg once daily Complete radiologic and neurologic response in total of 12 months 
Gaye et al. (103Case report (2019) Brigatinib Bevacizumab Yes, crizotinib and ceritinib with progression Brigatinib 180 mg once daily Sustained intracranial response for 14 months 
Gainor et al. (145Case series (2016) Alectinib One patient received chemo with carboplatin and pemetrexed Yes. One patient received crizotinib and ceritinib, the other patient only received crizotinib. Alectinib 900 mg twice daily There was symptomatic improvement for 6 months in one patient and 3 months in the other patient 
Metro et al. (146Case series (2016) Alectinib 11 patients with CNS Median number of prior lines of chemotherapy was 1 10/11 patients had crizotinib treatment prior Alectinib 600 twice daily Median CNS PFS: 8 months 
       Median OS: 13 months 
Ou et al. (99Case report (2015) Alectinib Carboplatin, pemetrexed, bevacizumab Yes, treated with crizotinib with progression Alectinib 600–750 mg twice daily Radiologic improvement in 6 weeks with sustained response for 15 months on going therapy 
Gainor et al. (147Case series (2015) Alectinib No Yes, treated with crizotinib and ceritinib prior with progression Alectinib 600 mg twice daily Radiologic and neurologic improvement in 3/4 patients (75%) 
       The 4th patient had stable intracranial disease for 4 months 
Dudnik et al. (148Case series (2015) Ceritinib No Crizotinib WBRT plus ceritinib 500 mg/daily PFS: 7 months in 2 patients and 18 months in the third patient 
Arrondeau et al. (149Case report (2014) Ceritinib Cisplatin and gemcitabine carboplatin, pemetrexed, and bevacizumab No Ceritinib 750 mg daily Radiologic improvement in 5 weeks and no progression seen at 5.5 months 
Ahn et al. (150Case series (2012) Crizotinib Patient 1: paclitaxel, carboplatinerlotinibpemetrexedvinorelbinedocetaxel,etoposide with cisplatin No Intrathecal MTX plus crizotinib 250 mg twice daily PFS of 10 and 6 months in the 2 patients 
    Patient 2: cisplatin plus pemetrexed    
Costa et al. (95Case report (2011) Crizotinib Cisplatin plus pemetrexed and second line of erlotinib No WBRT plus crizotinib 250 mg twice daily OS: 3 months 
StudyType of studyALK inhibitor usedNo. of patientsPrior chemotherapyPrior therapy with ALK inhibitorTreatment regimenResponse to therapy
Dagogo-Jack et al. (108Prospective Lorlatinib 23 patients with LM 35% of patients had prior systemic therapy All patients had CNS only progression on prior first (4%) or second generation ALK inhibitor (96%) Lorlatinib 100 mg daily Intracranial DCR in 12 weeks: 95% 
 Phase II (2022)      The intracranial objective response rate: 59% 
       Median PFS: 24.6 months 
Zou et al. (104Retrospective study (2022) Alectinib 65 patients with ALK-mutated lung cancer with brain metastasis or LM (9/65 LM) Not available 5/9 patients with LM were ALK-TKI naïve, 2/9 patients were crizotinib-resistant, and 3/9 patients had other second generation ALK-TKI exposure Alectinib 8/9 patients with LM reported improvement in CNS-related symptoms 
Chow et al. (142Prospective Ceritinib 18 LM 15/18 patients had prior chemotherapy 88% of patients had treated and progressed on crizotinib Ceritinib 750 mg once daily Whole-body ORR was 16.7% (95% CI, 3.6–41.4) and DCR was 66.7% (95% CI, 41.0–86.7) 
 Phase II (2022)       
Frost et al. (143Prospective (2020) Lorlatinib 9 LM All patients had at least one line of systemic chemotherapy All patients had prior ALK inhibitor exposure: crizotinib, ceritinib, alectinib, brigatinib Lorlatinib 100 mg daily Median duration of treatment: 10.4 months 
   36 brain metastases without LM    PFS: 8.0 months 
       Intracranial response rate: 54% 
Pellerino et al. (144Case report (2020) Lorlatinib Cisplatin and paclitaxel Crizotinib and ceritinib Lorlatinib 100 mg once daily Complete radiologic and neurologic response in total of 12 months 
Gaye et al. (103Case report (2019) Brigatinib Bevacizumab Yes, crizotinib and ceritinib with progression Brigatinib 180 mg once daily Sustained intracranial response for 14 months 
Gainor et al. (145Case series (2016) Alectinib One patient received chemo with carboplatin and pemetrexed Yes. One patient received crizotinib and ceritinib, the other patient only received crizotinib. Alectinib 900 mg twice daily There was symptomatic improvement for 6 months in one patient and 3 months in the other patient 
Metro et al. (146Case series (2016) Alectinib 11 patients with CNS Median number of prior lines of chemotherapy was 1 10/11 patients had crizotinib treatment prior Alectinib 600 twice daily Median CNS PFS: 8 months 
       Median OS: 13 months 
Ou et al. (99Case report (2015) Alectinib Carboplatin, pemetrexed, bevacizumab Yes, treated with crizotinib with progression Alectinib 600–750 mg twice daily Radiologic improvement in 6 weeks with sustained response for 15 months on going therapy 
Gainor et al. (147Case series (2015) Alectinib No Yes, treated with crizotinib and ceritinib prior with progression Alectinib 600 mg twice daily Radiologic and neurologic improvement in 3/4 patients (75%) 
       The 4th patient had stable intracranial disease for 4 months 
Dudnik et al. (148Case series (2015) Ceritinib No Crizotinib WBRT plus ceritinib 500 mg/daily PFS: 7 months in 2 patients and 18 months in the third patient 
Arrondeau et al. (149Case report (2014) Ceritinib Cisplatin and gemcitabine carboplatin, pemetrexed, and bevacizumab No Ceritinib 750 mg daily Radiologic improvement in 5 weeks and no progression seen at 5.5 months 
Ahn et al. (150Case series (2012) Crizotinib Patient 1: paclitaxel, carboplatinerlotinibpemetrexedvinorelbinedocetaxel,etoposide with cisplatin No Intrathecal MTX plus crizotinib 250 mg twice daily PFS of 10 and 6 months in the 2 patients 
    Patient 2: cisplatin plus pemetrexed    
Costa et al. (95Case report (2011) Crizotinib Cisplatin plus pemetrexed and second line of erlotinib No WBRT plus crizotinib 250 mg twice daily OS: 3 months 

Abbreviations: CR, complete response; LM, leptomeningeal disease; MTX, methotrexate; PR, partial response; WBRT, whole-brain radiotherapy; ORR, overall response rate.

Other driver mutations

Other oncogenes, such as ErbB2, KRAS, BRAF, PI3K, RET, PDGFR, ROS, MET exon 14 (METex14) skipping mutations, and MEK1 and HER2 have been found to be mutated, translocated, or amplified in NSCLCs (109), and might respond to targeted therapy. A case report showed that LOXO-292 demonstrated efficacy in a patient with LM from NSCLC with a RET mutation (110). Vemurafenib is presently being used in patients with lung cancer with BRAFV600E mutations, with one case report indicating that the drug provided 6 months of clinical and radiologic control in a patient with LM (111). Another case report of a patient with a V600E mutation who had progressed on dabrafenib/trametinib reported a quick response in the brain and leptomeninges to encorafenib and binimetinib (112). However, randomized trials focusing on LM in NSCLC with these rare mutations are needed.

Immunotherapy for LM secondary to NSCLC

Immune checkpoint inhibitors (ICI) have changed the current landscape of NSCLC management. There are several studies showing the promising effect of ICI in LM from NSCLC (113, 114). Hendriks and colleagues conducted a prospective study including 19 patients with LM from NSCLC who were treated with ICIs (13 with nivolumab and 6 with pembrolizumab), and showed a median PFS of 3.7 months, and a 6- and 12-months OS of 36.8% and 21.1%, respectively (113). A single-arm phase II clinical trial investigating the efficacy of pembrolizumab for LM in solid tumors (NCT02886585) reached its primary endpoint, and 60% of patients were alive 3 months after enrollment (115). A phase II clinical trial (116) that investigated the activity of pembrolizumab among patients with LM from solid tumors, including 23% of participants with NSCLC, showed an overall CNS response rate of 38% in 12 weeks, with an acceptable safety profile. Another phase II clinical trial involving 18 patients with LM secondary to solid tumors receiving combined ipilimumab and nivolumab showed promising activity and reported that 8 of 18 patients reached the primary endpoint of 3 months survival whereas one third of patients experienced grade 3 or 4 AE (117). Zheng and colleagues conducted a study including 32 patients with LM secondary to NSCLC who received ICI (nivolumab 21/32, pembrolizumab 9/32, atezolizumab 2/32) and reported that 62.5% of patients had neurologic symptom improvement, median PFS was 2 months, and OS was 4 months in patients who received single-agent ICI; median PFS and OS in patients who received other therapies with ICI was 3 and 5.4 months, respectively. In this study, all patients who received ICI as a single agent had cranial radiotherapy prior and received ICI as second-line therapy (118). Zheng and colleagues reported that patients with better Eastern Cooperative Oncology Group Performance Status (ECOG-PS) score had significantly longer PFS (P = 0.04), but there was no significant OS difference among patients who received single-agent ICI versus combined therapy (118). A retrospective study reported that incidence of LMD among patients who received postoperative stereotactic radiosurgery (SRS) with immunotherapy (either nivolumab or pembrolizumab) was less than among patients who received SRS alone (6% vs. 22%, P = 0.007; ref. 119). Prakadan and colleagues compiled the patients from two different clinical trials for patients with LM who received ICU and showed that ICI treatment alters the tumor microenvironment in patients with LM from any histology by conducting single-cell RNA and cell-free DNA profiling from CSF. Prakadan and colleagues reported that active immune response in CSF after intravenous ICI was correlated with OS (120). In both trials, there were increased overall levels of IFN-signaling, and cytotoxicity in CD8+ T cells posttreatment. These findings imply that intravenous ICI treatment modifies the immunologic milieu in the CSF of a subgroup of patients participating in these clinical trials and that this may have one of the mechanisms of the therapeutic benefit of ICI treatment (120). Another recent study used immune cell profiling of CSF using single-cell RNA sequencing in patients with brain metastasis showed that matching T-cell receptor clonotypes of CD8 T cells in CSF and brain tumor, which is very promising to use immune profiling of CSF developing cell-therapies for brain metastasis or exploring tumor microenvironment (121). The studies using immunotherapy in LM secondary to NSCLC demonstrated in Table 3.

Table 3.

Publications using immunotherapy in LM secondary to NSCLC.

Immunotherapy treatment usedPublicationNumber of patients observedDuration of follow-upOutcomes measuredSymptomatic or radiographic improvement
Pembrolizumab or combined ipilimumab and nivolumab Prakadan et al. (120)
  • -combined analysis of two clinical trials (NCT02886585 and NCT02939300)

 
19 patients with LM secondary to any histology (n = 10 patients with PD-1 inhibitor treatment, n = 9 patients with PD-1 and CTLA-4 inhibitor treatment) NA CD8 cells in CSF post-ICI treatment In both trials, there were overall higher levels of IFN- signaling and cytotoxicity in CD8+ T cells posttreatment 
    IFNγ response and antigen presentation Active immune response in CSF after intravenous ICI was correlated with OS 
Nivolumab (n = 21), pembrolizumab (n = 9), or atezolizumab (n = 2) Zheng et al. (118)
∼metanalysis 
32 patients with LM secondary to NSCLC who received ICI therapy NA In single agent ICI group:
PFS: 2 months
OS: 4 months 
62.5% of patients had neurologic symptom improvement 
  18.8% patients had PD-L1 expression >80%   ECOG-PS score was associated with longer PFS (P = 0.04) 
  37.5% patients had druggable mutations (EGFR/ALK/BRAF/MET/ERBB2)   No difference seen in OS between monotherapy with ICI vs. combined therapy 
    In combined group:  
    PFS: 3 months OS: 5.4 months  
Combined therapy with ipilimumab and nivolumab Brastianos et al. (117)
∼phase II clinical trial 
18 patients with LM secondary to solid tumors 8 months (median follow-up for patients who were alive) Primary end point: 3-month OS 8/18 patients reached the primary endpoint and were alive at 3 months 
    Secondary outcome: toxicity profile. 6/18 patients developed grade 3 or higher AE 
SRS and immunotherapy (nivolumab or pembrolizumab) vs. postoperative SRS alone nivolumab or pembrolizumab Minniti et al. (119)
∼retrospective study (2021) 
56/129 patients with NSCLC resected brain metastasis 15 months LM development after treatments 12-month LM development rates were 22% in SRS alone and 6% in the combined treatment group (P = 0.007) 
     SRS with immunotherapy decreased the LM development rate among patients with NSCLC who underwent surgery for brain metastasis 
Pembrolizumab Naidoo et al. (116)
∼ phase 2 clinical trial (2021) 
3/16 patients with lung cancer with LM Closed early due to poor accrual Central nervous system (CNS) response after four cycles 3 patients achieved CR but developed grade 3 side effect, thus therapy stopped 
     2/3 patients reported to have stable disease 
Pembrolizumab Brastianos et al. (115)
∼ phase II clinical trial (2020) 
20 patients with LM secondary solid tumor (2/20 with lung cancer) Median follow-up: 6.3 months (2.2–12.5 months) Primary endpoint: OS at 3 months 60% of patients reached primary endpoint and were alive at 3 months 
    Secondary endpoint: toxicity profile, response rate (RR), time to progression 40% patients developed grade 3 or higher AE 
Nivolumab Bover et al. (151)
∼ case report (2020) 
4 years 4-year OS Symptomatic and radiologic improvement (in interim analysis resolution of metabolic activity in PET imaging) reported 
Pembrolizumab or nivolumab Hendriks et al. (11319 13 months Median OS of 3.7 months (0.9–6.6 months) N/A 
 ∼ retrospective cohort study (2019)   6 months PFS was 21%  
Pembrolizumab Brastianos et al. (1522 patients with lung cancer (18 total) 6 months 3-month OS rate of 44% among all LM group, not specified as NSCLC N/A 
 ∼prospective study (2018)     
Nivolumab Dudnik et al. (153)
∼ case series (2016) 
28 weeks One patient had partial response; another patient had complete response. 7 months OS for only one patient reported. Both patients were asymptomatic at baseline. Reported radiologic response in 1 patient. 
      
Immunotherapy treatment usedPublicationNumber of patients observedDuration of follow-upOutcomes measuredSymptomatic or radiographic improvement
Pembrolizumab or combined ipilimumab and nivolumab Prakadan et al. (120)
  • -combined analysis of two clinical trials (NCT02886585 and NCT02939300)

 
19 patients with LM secondary to any histology (n = 10 patients with PD-1 inhibitor treatment, n = 9 patients with PD-1 and CTLA-4 inhibitor treatment) NA CD8 cells in CSF post-ICI treatment In both trials, there were overall higher levels of IFN- signaling and cytotoxicity in CD8+ T cells posttreatment 
    IFNγ response and antigen presentation Active immune response in CSF after intravenous ICI was correlated with OS 
Nivolumab (n = 21), pembrolizumab (n = 9), or atezolizumab (n = 2) Zheng et al. (118)
∼metanalysis 
32 patients with LM secondary to NSCLC who received ICI therapy NA In single agent ICI group:
PFS: 2 months
OS: 4 months 
62.5% of patients had neurologic symptom improvement 
  18.8% patients had PD-L1 expression >80%   ECOG-PS score was associated with longer PFS (P = 0.04) 
  37.5% patients had druggable mutations (EGFR/ALK/BRAF/MET/ERBB2)   No difference seen in OS between monotherapy with ICI vs. combined therapy 
    In combined group:  
    PFS: 3 months OS: 5.4 months  
Combined therapy with ipilimumab and nivolumab Brastianos et al. (117)
∼phase II clinical trial 
18 patients with LM secondary to solid tumors 8 months (median follow-up for patients who were alive) Primary end point: 3-month OS 8/18 patients reached the primary endpoint and were alive at 3 months 
    Secondary outcome: toxicity profile. 6/18 patients developed grade 3 or higher AE 
SRS and immunotherapy (nivolumab or pembrolizumab) vs. postoperative SRS alone nivolumab or pembrolizumab Minniti et al. (119)
∼retrospective study (2021) 
56/129 patients with NSCLC resected brain metastasis 15 months LM development after treatments 12-month LM development rates were 22% in SRS alone and 6% in the combined treatment group (P = 0.007) 
     SRS with immunotherapy decreased the LM development rate among patients with NSCLC who underwent surgery for brain metastasis 
Pembrolizumab Naidoo et al. (116)
∼ phase 2 clinical trial (2021) 
3/16 patients with lung cancer with LM Closed early due to poor accrual Central nervous system (CNS) response after four cycles 3 patients achieved CR but developed grade 3 side effect, thus therapy stopped 
     2/3 patients reported to have stable disease 
Pembrolizumab Brastianos et al. (115)
∼ phase II clinical trial (2020) 
20 patients with LM secondary solid tumor (2/20 with lung cancer) Median follow-up: 6.3 months (2.2–12.5 months) Primary endpoint: OS at 3 months 60% of patients reached primary endpoint and were alive at 3 months 
    Secondary endpoint: toxicity profile, response rate (RR), time to progression 40% patients developed grade 3 or higher AE 
Nivolumab Bover et al. (151)
∼ case report (2020) 
4 years 4-year OS Symptomatic and radiologic improvement (in interim analysis resolution of metabolic activity in PET imaging) reported 
Pembrolizumab or nivolumab Hendriks et al. (11319 13 months Median OS of 3.7 months (0.9–6.6 months) N/A 
 ∼ retrospective cohort study (2019)   6 months PFS was 21%  
Pembrolizumab Brastianos et al. (1522 patients with lung cancer (18 total) 6 months 3-month OS rate of 44% among all LM group, not specified as NSCLC N/A 
 ∼prospective study (2018)     
Nivolumab Dudnik et al. (153)
∼ case series (2016) 
28 weeks One patient had partial response; another patient had complete response. 7 months OS for only one patient reported. Both patients were asymptomatic at baseline. Reported radiologic response in 1 patient. 
      

Abbreviations: CTLA-4, cytotoxic T-lymphocyte–associated antigen 4; LM, leptomeningeal disease; NSCLC, non–small cell lung cancer; N/A, not applicable; OS, overall survival; PD-1, program death 1.

Supportive measurements

Supportive care is essential for LM management. Pain management, steroids, treatment of seizures, ventricular peritoneal (VP) shunt placement for relief of symptomatic hydrocephalus, and elevated ICP are all supportive therapies in patients with LM. In LM, there are no robust data to support the use of antiepileptics as seizure prophylaxis. There is also no agreement on systemic steroid treatment in LM other than at the lowest dose for the shortest time feasible (36, 41, 122). Peritoneal carcinomatosis reported as very rare complication of VP shunt placement in literature, although incidence remains unknown. A retrospective study of patients with LM from solid tumors (123) showed that patients who underwent LP or VP shunt placement reported a 50% improvement in symptoms, with 34% of total symptom relief after the procedure. Shunt-related complications included infection, shunt malfunction, and need for shunt revision. Another study involving 31 patients with hydrocephalus secondary to LM from lung adenocarcinoma who subsequently underwent palliative shunt placement reported symptom alleviation in 90.3% of patients, with a median survival of 7.7 months following LM diagnosis (124). Another retrospective study involving 190 patients with LM secondary to solid tumors reported that 83% of patients had symptomatic relief, and complications included infection (5%), shunt repair/externalization/removal (8%), and symptomatic subdural hygroma/hematoma (6.3%) whereas no abdominal seeding was seen (125). In day-to-day practice, the benefits of VP shunting in patients with increased intracranial pressure outweigh the theoretical risk of abdominal seeding (125).

Future directions

As discussed above, the landscape of treatments for LM from NSCLC is quickly changing with the new advancement in diagnostic tools and treatment options. There are several prospective clinical studies currently underway to investigate new diagnostic procedures, monitoring measures, and therapy options for LM from NSCLC. Ongoing clinical trials of LM are presented in Table 4.

Table 4.

Ongoing clinical trials on LM from NSCLC and selected clinical trials on LM from solid tumors including NSCLC (from https://clinicaltrials.gov, last update April 15, 2022).

Name of the studyDescriptionProtocol numberStatusNo. of patientsTreatmentPrimary outcomes to be measuredEstimated study completion date
Efficacy and Safety of Recombinant Human Endostatin in Non–Small Cell Lung Cancer with Leptomeningeal Metastasis Phase IV: Single group assignment NCT04356118 Not yet enrolling 30 Recombinant human endostatin 7.5 mg/m2/day once a day for 2 weeks and 1 week off, start the next cycle, up to four cycles +intrathecal MTX + targeted therapy (EGFR TKIs or ALK inhibitors) Leptomeningeal metastasis OS June 2023 
      NPFS  
      The incidence of AEs  
Efficacy and Safety of Durvalumab in Non–Small Cell Lung Cancer with Leptomeningeal Metastasis Phase IV: Single group assignment NCT04356222 Not yet enrolling 30 Durvalumab 10 mg/kg every 2 weeks plus intrathecal MTX OS June 2023 
      NPFS  
      The incidence of AEs  
Osimertinib with Bevacizumab for LM from EGFR-mutation non–small cell lung cancer Phase II: Single group assignment NCT04425681 Recruiting 20 Osimertinib 80 mg daily + bevacizumab 7.5 mg/kg every 3 weeks LM PFS June 2021 
      ORR  
Clinical efficacy and safety of EGFR-TKI combined with nimotuzumab in the treatment of leptomeningeal metastases from lung cancer Phase II NCT04833205 Recruiting 30 Nimotuzumab 200 mg for 8 weeks + third generation EGFR-TKI PFS April 2023 
Study of AZD9291 in NSCLC patients harboring T790M mutation who failed EGFR TKI and with brain and/or LMS Phase II: Single group assignment NCT03257124 Active, not recruiting 80 AZD9291 (osimertinib) 160 mg daily ORR in CNS -brain metastasis cohort December 2021 
      OS - Leptomeningeal with or without brain metastasis cohort  
A dose exploration study of almonertinib for EGFRm NSCLC Patients with Brain/LM (ARTISTRY) Single group assignment NCT04778800 Recruiting 60 Almonertinib
  • Arm 1: 110 mg daily

  • Arm 2: 160 mg daily

  • Arm 3: 220 mg daily

 
Intracranial progression-free survival (iPFS) February 2024 
Study of osimertinib in patients with a lung cancer with brain or leptomeningeal metastases with EGFR mutation (ORBITAL) Phase II: Single group assignment NCT04233021 Recruiting 113 Osimertinib 80 mg/day ORR July 2022 
      Objective response rate at 6 months using EANO-ESMO criteria (cohort 1) and RECIST1.1 criteria (cohorts 2, 3, 4)  
Study of TY-9591 in Patients with a lung cancer with brain or leptomeningeal metastases with EGFR mutation Phase II: Single group assignment NCT05146219 Recruiting 60 TY-9591 tablets 160 mg/day iORR December 2024 
      eORR  
Efficacy and safety of 80 mg osimertinib in patients with NSCLC (BLOSSOM) Phase II: Single group assignment NCT04563871 Enrolling by invitation 80 Osimertinib 80 mg/day OS February 2024 
177Lu-DTPA-omburtamab radioimmunotherapy for LM from solid tumors (breast, NSCLC, malignant melanoma)a Phase I/II: Single group assignment NCT04315246 Recruiting 63 177Lu-DTPA-omburtamab Incidence of AEs and SAEs December 2024 
     Intracerebroventricular administration of 177Lu-DTPA-omburtamab for up to five cycles   
Proton craniospinal radiation therapy vs. partial photon radiotherapy for LM from solid tumorsa Phase II: Randomized parallel assignment NCT04343573 Active, not recruiting 102 Arm 1: standard of care (involved field photon RT including WBRT and/or focal spine RT followed by standard of care systemic treatments per physician choice) NPFS March 2022 
     Arm 2: proton CSI followed by standard of care systemic treatments per physician choice   
GDC-0084 with radiotherapy for people with PIK3CA-mutated solid tumor brain metastases or leptomeningeal metastasesa Phase I: Single group assignment NCT04192981 Recruiting 36 WBRT (30 Gy/10 fr) plus GDC-0084 in 3+3 dose-escalation cohort: 45, 60, 75 mg daily, with a potential dose de-escalation cohort to 30 mg MTD October 2021 
Avelumab with radiotherapy in patients with leptomeningeal diseasea Phase Ib: Single group assignment NCT03719768 Recruiting 23 Avelumab 800 mg iv every 2 weeks plus WBRT (30 Gr/10 fr) Safety and dose limiting toxicity March 2025 
Pembrolizumab and lenvatinib in leptomeningeal metastasesa Phase II: Single group assignment NCT04729348 Recruiting 19 Pembrolizumab + lenvatinib daily every 3 weeks Proportion of participants alive at 6 months December 2022 
Name of the studyDescriptionProtocol numberStatusNo. of patientsTreatmentPrimary outcomes to be measuredEstimated study completion date
Efficacy and Safety of Recombinant Human Endostatin in Non–Small Cell Lung Cancer with Leptomeningeal Metastasis Phase IV: Single group assignment NCT04356118 Not yet enrolling 30 Recombinant human endostatin 7.5 mg/m2/day once a day for 2 weeks and 1 week off, start the next cycle, up to four cycles +intrathecal MTX + targeted therapy (EGFR TKIs or ALK inhibitors) Leptomeningeal metastasis OS June 2023 
      NPFS  
      The incidence of AEs  
Efficacy and Safety of Durvalumab in Non–Small Cell Lung Cancer with Leptomeningeal Metastasis Phase IV: Single group assignment NCT04356222 Not yet enrolling 30 Durvalumab 10 mg/kg every 2 weeks plus intrathecal MTX OS June 2023 
      NPFS  
      The incidence of AEs  
Osimertinib with Bevacizumab for LM from EGFR-mutation non–small cell lung cancer Phase II: Single group assignment NCT04425681 Recruiting 20 Osimertinib 80 mg daily + bevacizumab 7.5 mg/kg every 3 weeks LM PFS June 2021 
      ORR  
Clinical efficacy and safety of EGFR-TKI combined with nimotuzumab in the treatment of leptomeningeal metastases from lung cancer Phase II NCT04833205 Recruiting 30 Nimotuzumab 200 mg for 8 weeks + third generation EGFR-TKI PFS April 2023 
Study of AZD9291 in NSCLC patients harboring T790M mutation who failed EGFR TKI and with brain and/or LMS Phase II: Single group assignment NCT03257124 Active, not recruiting 80 AZD9291 (osimertinib) 160 mg daily ORR in CNS -brain metastasis cohort December 2021 
      OS - Leptomeningeal with or without brain metastasis cohort  
A dose exploration study of almonertinib for EGFRm NSCLC Patients with Brain/LM (ARTISTRY) Single group assignment NCT04778800 Recruiting 60 Almonertinib
  • Arm 1: 110 mg daily

  • Arm 2: 160 mg daily

  • Arm 3: 220 mg daily

 
Intracranial progression-free survival (iPFS) February 2024 
Study of osimertinib in patients with a lung cancer with brain or leptomeningeal metastases with EGFR mutation (ORBITAL) Phase II: Single group assignment NCT04233021 Recruiting 113 Osimertinib 80 mg/day ORR July 2022 
      Objective response rate at 6 months using EANO-ESMO criteria (cohort 1) and RECIST1.1 criteria (cohorts 2, 3, 4)  
Study of TY-9591 in Patients with a lung cancer with brain or leptomeningeal metastases with EGFR mutation Phase II: Single group assignment NCT05146219 Recruiting 60 TY-9591 tablets 160 mg/day iORR December 2024 
      eORR  
Efficacy and safety of 80 mg osimertinib in patients with NSCLC (BLOSSOM) Phase II: Single group assignment NCT04563871 Enrolling by invitation 80 Osimertinib 80 mg/day OS February 2024 
177Lu-DTPA-omburtamab radioimmunotherapy for LM from solid tumors (breast, NSCLC, malignant melanoma)a Phase I/II: Single group assignment NCT04315246 Recruiting 63 177Lu-DTPA-omburtamab Incidence of AEs and SAEs December 2024 
     Intracerebroventricular administration of 177Lu-DTPA-omburtamab for up to five cycles   
Proton craniospinal radiation therapy vs. partial photon radiotherapy for LM from solid tumorsa Phase II: Randomized parallel assignment NCT04343573 Active, not recruiting 102 Arm 1: standard of care (involved field photon RT including WBRT and/or focal spine RT followed by standard of care systemic treatments per physician choice) NPFS March 2022 
     Arm 2: proton CSI followed by standard of care systemic treatments per physician choice   
GDC-0084 with radiotherapy for people with PIK3CA-mutated solid tumor brain metastases or leptomeningeal metastasesa Phase I: Single group assignment NCT04192981 Recruiting 36 WBRT (30 Gy/10 fr) plus GDC-0084 in 3+3 dose-escalation cohort: 45, 60, 75 mg daily, with a potential dose de-escalation cohort to 30 mg MTD October 2021 
Avelumab with radiotherapy in patients with leptomeningeal diseasea Phase Ib: Single group assignment NCT03719768 Recruiting 23 Avelumab 800 mg iv every 2 weeks plus WBRT (30 Gr/10 fr) Safety and dose limiting toxicity March 2025 
Pembrolizumab and lenvatinib in leptomeningeal metastasesa Phase II: Single group assignment NCT04729348 Recruiting 19 Pembrolizumab + lenvatinib daily every 3 weeks Proportion of participants alive at 6 months December 2022 

Abbreviations: AE, adverse events; CSI, craniospinal irradiation; eORR, extracranial overall response rate; iORR, intracranial overall response rate; LM: leptomeningeal metastases; MTX, methotrexate; NSCLC, non–small cell lung cancer; NPFS, neurologic PFS; ORR, objective response rate; SAE, serious adverse events; WBRT, whole-brain radiotherapy.

aStudies on LM secondary to solid tumors including lung cancer, but not exclusively lung cancer.

LM remains a fatal complication of advanced cancer, and its incidence is increasing, given the success of advanced cancer therapies. Drug penetration is limited to the leptomeningeal region, given the blood–brain barrier. Initial diagnostic workup should include CSF cytology, and neuraxial imaging with MRI. Given emerging novel diagnostics such as CSF liquid biopsy and the promise for early diagnosis and can offer valuable information to guide the therapy. The treatment of LM secondary to NSCLC needs to tailor according to an individual patient and requires multimodal treatment. The development of novel therapeutic agents with better CNS penetration, especially with. Patients with actionable mutations are candidates for targeted therapy. The third-generation EGFR inhibitor high-dose osimertinib (160 mg daily) recommended LM from EGFR-mutated NSCLC and has shown a survival advantage compared with standard therapies, such as RT, chemotherapy, and first- and second-generation TKIs. Patients with ALK-rearrangement should be treated with ALK inhibitors. Radiotherapy offers palliative relief in symptomatic patients with bulky disease. Although there is no consensus, WBRT did not show an OS benefit. Ongoing trials are investigating the role of WBRT in LM. Systemic or IT chemotherapy can be options for patients with no actionable mutation with systemic metastasis or progressive disease. There are several studies that showed promising results with a combined or single-agent immunotherapy. Clinical trials in LM secondary to NSCLC focus on monitoring clinical and radiologic responses, expanding data on other targetable mutations, and investigating response and resistance indicators are urgently needed.

No author disclosures were reported.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1.
Clarke
JL
,
Perez
HR
,
Jacks
LM
,
Panageas
KS
,
Deangelis
LM
.
Leptomeningeal metastases in the MRI era
.
Neurology
2010
;
74
:
1449
54
.
2.
Wasserstrom
WR
,
Glass
JP
,
Posner
JB
.
Diagnosis and treatment of leptomeningeal metastases from solid tumors: experience with 90 patients
.
Cancer
1982
;
49
:
759
72
.
3.
Kaplan
JG
,
DeSouza
TG
,
Farkash
A
,
Shafran
B
,
Pack
D
,
Rehman
F
, et al
.
Leptomeningeal metastases: comparison of clinical features and laboratory data of solid tumors, lymphomas and leukemias
.
J Neurooncol
1990
;
9
:
225
9
.
4.
Chamberlain
MC
.
Combined-modality treatment of leptomeningeal gliomatosis
.
Neurosurgery
2003
;
52
:
324
29
.
5.
Beauchesne
P
.
Intrathecal chemotherapy for treatment of leptomeningeal dissemination of metastatic tumours
.
Lancet Oncol
2010
;
11
:
871
9
.
6.
Zheng
MM
,
Li
YS
,
Jiang
BY
,
Tu
HY
,
Tang
WF
,
Yang
JJ
, et al
.
Clinical utility of cerebrospinal fluid cell-free DNA as liquid biopsy for leptomeningeal metastases in ALK-rearranged NSCLC
.
J Thorac Oncol
2019
;
14
:
924
32
.
7.
Li
YS
,
Jiang
BY
,
Yang
JJ
,
Tu
HY
,
Zhou
Q
,
Guo
WB
, et al
.
Leptomeningeal metastases in patients with NSCLC with EGFR mutations
.
J Thorac Oncol
2016
;
11
:
1962
9
.
8.
Lee
Y
,
Han
JY
,
Kim
HT
,
Yun
T
,
Lee
GK
,
Kim
HY
, et al
.
Impact of EGFR tyrosine kinase inhibitors versus chemotherapy on the development of leptomeningeal metastasis in never smokers with advanced adenocarcinoma of the lung
.
J Neurooncol
2013
;
115
:
95
101
.
9.
Liao
BC
,
Lee
JH
,
Lin
CC
,
Chen
YF
,
Chang
CH
,
Ho
CC
, et al
.
Epidermal growth factor receptor tyrosine kinase inhibitors for non-small-cell lung cancer patients with leptomeningeal carcinomatosis
.
J Thorac Oncol
2015
;
10
:
1754
61
.
10.
Umemura
S
,
Tsubouchi
K
,
Yoshioka
H
,
Hotta
K
,
Takigawa
N
,
Fujiwara
K
, et al
.
Clinical outcome in patients with leptomeningeal metastasis from non-small cell lung cancer: Okayama Lung Cancer Study Group
.
Lung Cancer
2012
;
77
:
134
9
.
11.
Riess
JW
,
Nagpal
S
,
Iv
M
,
Zeineh
M
,
Gubens
MA
,
Ramchandran
K
, et al
.
Prolonged survival of patients with non-small-cell lung cancer with leptomeningeal carcinomatosis in the modern treatment era
.
Clin Lung Cancer
2014
;
15
:
202
6
.
12.
Le Rhun
E
,
Devos
P
,
Boulanger
T
,
Smits
M
,
Brandsma
D
,
Ruda
R
, et al
.
The RANO Leptomeningeal Metastasis Group proposal to assess response to treatment: lack of feasibility and clinical utility and a revised proposal
.
Neuro Oncol
2019
;
21
:
648
58
.
13.
Lee
SJ
,
Lee
JI
,
Nam
DH
,
Ahn
YC
,
Han
JH
,
Sun
JM
, et al
.
Leptomeningeal carcinomatosis in non-small-cell lung cancer patients: impact on survival and correlated prognostic factors
.
J Thorac Oncol
2013
;
8
:
185
91
.
14.
Yin
K
,
Li
YS
,
Zheng
MM
,
Jiang
BY
,
Li
WF
,
Yang
JJ
, et al
.
A molecular graded prognostic assessment (molGPA) model specific for estimating survival in lung cancer patients with leptomeningeal metastases
.
Lung Cancer
2019
;
131
:
134
8
.
15.
Chi
Y
,
Remsik
J
,
Kiseliovas
V
,
Derderian
C
,
Sener
U
,
Alghader
M
, et al
.
Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis
.
Science
2020
;
369
:
276
82
.
16.
Boire
A
,
Zou
Y
,
Shieh
J
,
Macalinao
DG
,
Pentsova
E
,
Massagué
J
.
Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis
.
Cell
2017
;
168
:
1101
13
.
17.
Norris
LK
,
Grossman
SA
,
Olivi
A
.
Neoplastic meningitis following surgical resection of isolated cerebellar metastasis: a potentially preventable complication
.
J Neurooncol
1997
;
32
:
215
23
.
18.
DeAngelis
LM
,
Mandell
LR
,
Thaler
HT
,
Kimmel
DW
,
Galicich
JH
,
Fuks
Z
, et al
.
The role of postoperative radiotherapy after resection of single brain metastases
.
Neurosurgery
1989
;
24
:
798
805
.
19.
Johnson
MD
,
Avkshtol
V
,
Baschnagel
AM
,
Meyer
K
,
Ye
H
,
Grills
IS
, et al
.
Surgical resection of brain metastases and the risk of leptomeningeal recurrence in patients treated with stereotactic radiosurgery
.
Int J Radiat Oncol Biol Phys
2016
;
94
:
537
43
.
20.
Clarke
JL
.
Leptomeningeal metastasis from systemic cancer
.
Continuum (N Y)
2012
;
18
:
328
42
.
21.
Demopoulos
A
.
Clinical features and diagnosis of leptomeningeal metastases from solid tumors
.
In: UpToDate, Post TW (Ed), UpToDate
,
Waltham, MA
Available from
: https://www.uptodate.com/contents/clinical-features-and-diagnosis-of-leptomeningeal-disease-from-solid-tumors.
22.
Chamberlain
M
,
Soffietti
R
,
Raizer
J
,
Ruda
R
,
Brandsma
D
,
Boogerd
W
, et al
.
Leptomeningeal metastasis: a Response Assessment in Neuro-Oncology critical review of endpoints and response criteria of published randomized clinical trials
.
Neuro Oncol
2014
;
16
:
1176
85
.
23.
Le Rhun
E
,
Weller
M
,
Brandsma
D
,
Van den Bent
M
,
de Azambuja
E
,
Henriksson
R
, 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
:
iv84
99
.
24.
Le Rhun
E
,
Devos
P
,
Weller
J
,
Seystahl
K
,
Mo
F
,
Compter
A
, et al
.
Prognostic validation and clinical implications of the EANO ESMO classification of leptomeningeal metastasis from solid tumors
.
Neuro Oncol
2021
;
23
:
1100
12
.
25.
Glantz
MJ
,
Cole
BF
,
Glantz
LK
,
Cobb
J
,
Mills
P
,
Lekos
A
, et al
.
Cerebrospinal fluid cytology in patients with cancer: minimizing false-negative results
.
Cancer
1998
;
82
:
733
9
.
26.
Chamberlain
MC
,
Kormanik
PA
,
Glantz
MJ
.
A comparison between ventricular and lumbar cerebrospinal fluid cytology in adult patients with leptomeningeal metastases
.
Neuro Oncol
2001
;
3
:
42
5
.
27.
Straathof
CS
,
de Bruin
HG
,
Dippel
DW
,
Vecht
CJ
.
The diagnostic accuracy of magnetic resonance imaging and cerebrospinal fluid cytology in leptomeningeal metastasis
.
J Neurol
1999
;
246
:
810
4
.
28.
Ruff
RL
,
Dougherty
JH
Jr
.
Complications of lumbar puncture followed by anticoagulation
.
Stroke
1981
;
12
:
879
81
.
29.
Rinehardt
H
,
Kassem
M
,
Morgan
E
,
Palettas
M
,
Stephens
JA
,
Suresh
A
, et al
.
Assessment of leptomeningeal carcinomatosis diagnosis, management and outcomes in patients with solid tumors over a decade of experience
.
Eur J Breast Health
2021
;
17
:
371
7
.
30.
Hoon
DS
,
Kuo
CT
,
Wascher
RA
,
Fournier
P
,
Wang
HJ
,
O'Day
SJ
.
Molecular detection of metastatic melanoma cells in cerebrospinal fluid in melanoma patients
.
J Invest Dermatol
2001
;
117
:
375
8
.
31.
Malkin
MG
,
Posner
JB
.
Cerebrospinal fluid tumor markers for the diagnosis and management of leptomeningeal metastases
.
Eur J Cancer Clin Oncol
1987
;
23
:
1
4
.
32.
Nakagawa
H
,
Kubo
S
,
Murasawa
A
,
Nakajima
S
,
Nakajima
Y
,
Izumoto
S
, et al
.
Measurements of CSF biochemical tumor markers in patients with meningeal carcinomatosis and brain tumors
.
J Neurooncol
1992
;
12
:
111
20
.
33.
Bernstein
WB
,
Kemp
JD
,
Kim
GS
,
Johnson
VV
.
Diagnosing leptomeningeal carcinomatosis with negative CSF cytology in advanced prostate cancer
.
J Clin Oncol
2008
;
26
:
3281
4
.
34.
Kosmas
C
,
Tsavaris
NB
,
Soukouli
G
,
Gouveris
P
,
Tsakonas
G
,
Katselis
J
, et al
.
Changes of cerebrospinal fluid tumor marker levels may predict response to treatment and survival of carcinomatous meningitis in patients with advanced breast cancer
.
Med Oncol
2005
;
22
:
123
8
.
35.
Jiang
BY
,
Li
YS
,
Guo
WB
,
Zhang
XC
,
Chen
ZH
,
Su
J
, et al
.
Detection of driver and resistance mutations in leptomeningeal metastases of NSCLC by next-generation sequencing of cerebrospinal fluid circulating tumor cells
.
Clin Cancer Res
2017
;
23
:
5480
8
.
36.
Nayak
L
,
Fleisher
M
,
Gonzalez-Espinoza
R
,
Lin
O
,
Panageas
K
,
Reiner
A
, et al
.
Rare cell capture technology for the diagnosis of leptomeningeal metastasis in solid tumors
.
Neurology
2013
;
80
:
1598
605
.
37.
Tu
Q
,
Wu
X
,
Le Rhun
E
,
Blonski
M
,
Wittwer
B
,
Taillandier
L
, et al
.
CellSearch technology applied to the detection and quantification of tumor cells in CSF of patients with lung cancer leptomeningeal metastasis
.
Lung Cancer
2015
;
90
:
352
7
.
38.
Patel
AS
,
Allen
JE
,
Dicker
DT
,
Peters
KL
,
Sheehan
JM
,
Glantz
MJ
, et al
.
Identification and enumeration of circulating tumor cells in the cerebrospinal fluid of breast cancer patients with central nervous system metastases
.
Oncotarget
2011
;
2
:
752
60
.
39.
Nevel
KS
,
DiStefano
N
,
Lin
X
,
Skakodub
A
,
Ogilvie
SQ
,
Reiner
AS
, et al
.
A retrospective, quantitative assessment of disease burden in patients with leptomeningeal metastases from non-small-cell lung cancer
.
Neuro Oncol
2020
;
22
:
675
83
.
40.
Pellerino
A
,
Brastianos
PK
,
Ruda
R
,
Soffietti
R
.
Leptomeningeal metastases from solid tumors: recent advances in diagnosis and molecular approaches
.
Cancers
2021
;
13
:
2888
.
41.
Lin
X
,
Fleisher
M
,
Rosenblum
M
,
Lin
O
,
Boire
A
,
Briggs
S
, et al
.
Cerebrospinal fluid circulating tumor cells: a novel tool to diagnose leptomeningeal metastases from epithelial tumors
.
Neuro Oncol
2017
;
19
:
1248
54
.
42.
van Bussel
MTJ
,
Pluim
D
,
Milojkovic Kerklaan
B
,
Bol
M
,
Sikorska
K
,
Linders
DTC
, et al
.
Circulating epithelial tumor cell analysis in CSF in patients with leptomeningeal metastases
.
Neurology
2020
;
94
:
e521
e8
.
43.
De Mattos-Arruda
L
,
Mayor
R
,
Ng
CKY
,
Weigelt
B
,
Martinez-Ricarte
F
,
Torrejon
D
, et al
.
Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma
.
Nat Commun
2015
;
6
:
8839
.
44.
Li
YS
,
Jiang
BY
,
Yang
JJ
,
Zhang
XC
,
Zhang
Z
,
Ye
JY
, et al
.
Unique genetic profiles from cerebrospinal fluid cell-free DNA in leptomeningeal metastases of EGFR-mutant non-small-cell lung cancer: a new medium of liquid biopsy
.
Ann Oncol
2018
;
29
:
945
52
.
45.
Li
Y-S
,
Zheng
M-M
,
Jiang
B-Y
,
Tu
H-Y
,
Yang
J-J
,
Zhang
X-C
, et al
.
Association of cerebrospinal fluid tumor DNA genotyping with survival among patients with lung adenocarcinoma and central nervous system metastases
.
JAMA Netw Open
2020
;
3
:
e209077
.
46.
Zheng
MM
,
Li
YS
,
Tu
HY
,
Jiang
BY
,
Yang
JJ
,
Zhou
Q
, et al
.
Genotyping of cerebrospinal fluid associated with osimertinib response and resistance for leptomeningeal metastases in EGFR-mutated NSCLC
.
J Thorac Oncol
2021
;
16
:
250
8
.
47.
Chamberlain
MC
,
Sandy
AD
,
Press
GA
.
Leptomeningeal metastasis: a comparison of gadolinium-enhanced MR and contrast-enhanced CT of the brain
.
Neurology
1990
;
40
:
435
8
.
48.
Collie
DA
,
Brush
JP
,
Lammie
GA
,
Grant
R
,
Kunkler
I
,
Leonard
R
, et al
.
Imaging features of leptomeningeal metastases
.
Clin Radiol
1999
;
54
:
765
71
.
49.
Singh
SK
,
Leeds
NE
,
Ginsberg
LE
.
MR imaging of leptomeningeal metastases: comparison of three sequences
.
AJNR Am J Neuroradiol
2002
;
23
:
817
21
.
50.
Mittl
RL
Jr
,
Yousem
DM
.
Frequency of unexplained meningeal enhancement in the brain after lumbar puncture
.
AJNR Am J Neuroradiol
1994
;
15
:
633
8
.
51.
Remon
J
,
Le Rhun
E
,
Besse
B
.
Leptomeningeal carcinomatosis in non-small cell lung cancer patients: a continuing challenge in the personalized treatment era
.
Cancer Treat Rev
2017
;
53
:
128
37
.
52.
Ko
Y
,
Gwak
HS
,
Park
EY
,
Joo
J
,
Lee
YJ
,
Lee
SH
, et al
.
Association of MRI findings with clinical characteristics and prognosis in patients with leptomeningeal carcinomatosis from non-small cell lung cancer
.
J Neurooncol
2019
;
143
:
553
62
.
53.
Sze
G
,
Soletsky
S
,
Bronen
R
,
Krol
G
.
MR imaging of the cranial meninges with emphasis on contrast enhancement and meningeal carcinomatosis
.
AJR Am J Roentgenol
1989
;
153
:
1039
49
.
54.
Glantz
MJ
,
Hall
WA
,
Cole
BF
,
Chozick
BS
,
Shannon
CM
,
Wahlberg
L
, et al
.
Diagnosis, management, and survival of patients with leptomeningeal cancer based on cerebrospinal fluid-flow status
.
Cancer
1995
;
75
:
2919
31
.
55.
Chamberlain
MC
,
Kormanik
PA
.
Prognostic significance of 111indium-DTPA CSF flow studies in leptomeningeal metastases
.
Neurology
1996
;
46
:
1674
7
.
56.
Mason
WP
,
Yeh
SD
,
DeAngelis
LM
.
111Indium-diethylenetriamine pentaacetic acid cerebrospinal fluid flow studies predict distribution of intrathecally administered chemotherapy and outcome in patients with leptomeningeal metastases
.
Neurology
1998
;
50
:
438
44
.
57.
2 NCCNCNSCV
.
2020 April 2022
. <www.nccn.org/professionals/physician_gls/pdf/cns.pdf>.
April
2022
.
58.
Cheng
H
,
Perez-Soler
R
.
Leptomeningeal metastases in non-small-cell lung cancer
.
Lancet Oncol
2018
;
19
:
e43
55
.
59.
Pellerino
A
,
Internò
V
,
Muscolino
E
,
Mo
F
,
Bruno
F
,
Pronello
E
, et al
.
Leptomeningeal metastases from non-small cell lung cancer: state of the art and recent advances
.
J Cancer Metastasis Treat
2020
;
6
:
41
.
60.
Kim
HJ
,
Im
SA
,
Keam
B
,
Kim
YJ
,
Han
SW
,
Kim
TM
, et al
.
Clinical outcome of central nervous system metastases from breast cancer: differences in survival depending on systemic treatment
.
J Neurooncol
2012
;
106
:
303
13
.
61.
Alexander
M
,
Lin
E
,
Cheng
H
.
Leptomeningeal metastases in non-small cell lung cancer: optimal systemic management in NSCLC with and without driver mutations
.
Curr Treat Options Oncol
2020
;
21
:
72
.
62.
Yan
W
,
Liu
Y
,
Li
J
,
Han
A
,
Kong
L
,
Yu
J
, et al
.
Whole brain radiation therapy does not improve the overall survival of EGFR-mutant NSCLC patients with leptomeningeal metastasis
.
Radiat Oncol
2019
;
14
:
168
.
63.
Buszek
SM
,
Chung
C
.
Radiotherapy in leptomeningeal disease: a systematic review of randomized and non-randomized trials
.
Front Oncol
2019
;
9
:
1224
.
64.
Nguyen
TK
,
Nguyen
EK
,
Soliman
H
.
An overview of leptomeningeal disease
.
Ann Palliat Med
2020
;
10
:
909
22
.
65.
Ozdemir
Y
,
Yildirim
BA
,
Topkan
E
.
Whole brain radiotherapy in management of non-small-cell lung carcinoma associated leptomeningeal carcinomatosis: evaluation of prognostic factors
.
J Neurooncol
2016
;
129
:
329
35
.
66.
El Shafie
RA
,
Bohm
K
,
Weber
D
,
Lang
K
,
Schlaich
F
,
Adeberg
S
, et al
.
Outcome and prognostic factors following palliative craniospinal irradiation for leptomeningeal carcinomatosis
.
Cancer Manag Res
2019
;
11
:
789
801
.
67.
Yang
TJ
,
Wijetunga
NA
,
Yamada
J
,
Wolden
S
,
Mehallow
M
,
Goldman
DA
, et al
.
Clinical trial of proton craniospinal irradiation for leptomeningeal metastases
.
Neuro Oncol
2021
;
23
:
134
43
.
68.
Ariyasu
R
,
Horiike
A
,
Koyama
J
,
Saiki
M
,
Sonoda
T
,
Kawashima
Y
, et al
.
Efficacy of bevacizumab and erlotinib combination for leptomeningeal carcinomatosis after failure of erlotinib
.
Anticancer Drugs
2017
;
28
:
565
7
.
69.
Wu
YL
,
Zhou
L
,
Lu
Y
.
Intrathecal chemotherapy as a treatment for leptomeningeal metastasis of non-small cell lung cancer: a pooled analysis
.
Oncol Lett
2016
;
12
:
1301
14
.
70.
Pan
Z
,
Yang
G
,
Cui
J
,
Li
W
,
Li
Y
,
Gao
P
, et al
.
A pilot phase 1 study of intrathecal pemetrexed for refractory leptomeningeal metastases from non-small-cell lung cancer
.
Front Oncol
2019
;
9
:
838
.
71.
Fan
C
,
Zhao
Q
,
Li
L
,
Shen
W
,
Du
Y
,
Teng
C
, et al
.
Efficacy and safety of intrathecal pemetrexed combined with dexamethasone for treating tyrosine kinase inhibitor-failed leptomeningeal metastases from EGFR-mutant NSCLC-a prospective, open-label, single-arm phase 1/2 clinical trial (Unique Identifier: ChiCTR1800016615)
.
J Thorac Oncol
2021
;
16
:
1359
68
.
72.
Zheng
MM
,
Li
YS
,
Sun
H
,
Chen
HJ
,
Wu
YL
.
Intrathecal pemetrexed: another potential treatment modality for tyrosine kinase inhibitor-failed leptomeningeal metastases?
J Thorac Oncol
2021
;
16
:
e82
e4
.
73.
Pan
Z
,
Yang
G
,
He
H
,
Cui
J
,
Li
W
,
Yuan
T
, et al
.
Intrathecal pemetrexed combined with involved-field radiotherapy as a first-line intra-CSF therapy for leptomeningeal metastases from solid tumors: a phase I/II study
.
Ther Adv Med Oncol
2020
;
12
:
1758835920937953
.
74.
Zairi
F
,
Le Rhun
E
,
Bertrand
N
,
Boulanger
T
,
Taillibert
S
,
Aboukais
R
, et al
.
Complications related to the use of an intraventricular access device for the treatment of leptomeningeal metastases from solid tumor: a single centre experience in 112 patients
.
J Neurooncol
2015
;
124
:
317
23
.
75.
Montes de Oca Delgado
M
,
Cacho Diaz
B
,
Santos Zambrano
J
,
Guerrero Juarez
V
,
Lopez Martinez
MS
,
Castro Martinez
E
, et al
.
The comparative treatment of intraventricular chemotherapy by ommaya reservoir vs. lumbar puncture in patients with leptomeningeal carcinomatosis
.
Front Oncol
2018
;
8
:
509
.
76.
Glantz
MJ
,
Van Horn
A
,
Fisher
R
,
Chamberlain
MC
.
Route of intracerebrospinal fluid chemotherapy administration and efficacy of therapy in neoplastic meningitis
.
Cancer
2010
;
116
:
1947
52
.
77.
Gaughan
EM
,
Costa
DB
.
Genotype-driven therapies for non-small cell lung cancer: focus on EGFR, KRAS and ALK gene abnormalities
.
Ther Adv Med Oncol
2011
;
3
:
113
25
.
78.
Kuiper
JL
,
Hendriks
LE
,
van der Wekken
AJ
,
de Langen
AJ
,
Bahce
I
,
Thunnissen
E
, et al
.
Treatment and survival of patients with EGFR-mutated non-small cell lung cancer and leptomeningeal metastasis: a retrospective cohort analysis
.
Lung Cancer
2015
;
89
:
255
61
.
79.
Jackman
DM
,
Holmes
AJ
,
Lindeman
N
,
Wen
PY
,
Kesari
S
,
Borras
AM
, 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
:
4517
20
.
80.
Kawamura
T
,
Hata
A
,
Takeshita
J
,
Fujita
S
,
Hayashi
M
,
Tomii
K
, et al
.
High-dose erlotinib for refractory leptomeningeal metastases after failure of standard-dose EGFR-TKIs
.
Cancer Chemother Pharmacol
2015
;
75
:
1261
6
.
81.
So
T
,
Inoue
M
,
Chikaishi
Y
,
Nose
N
,
Sugio
K
,
Yasumoto
K
.
Gefitinib and a ventriculo-peritoneal shunt to manage carcinomatous meningitis from non-small-cell lung cancer: report of two cases
.
Surg Today
2009
;
39
:
598
602
.
82.
Yi
HG
,
Kim
HJ
,
Kim
YJ
,
Han
SW
,
Oh
DY
,
Lee
SH
, 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
:
80
4
.
83.
Kashima
J
,
Okuma
Y
,
Miwa
M
,
Hosomi
Y
.
Retrospective analysis of survival in patients with leptomeningeal carcinomatosis from lung adenocarcinoma treated with erlotinib and gefitinib
.
Jpn J Clin Oncol
2017
;
47
:
357
62
.
84.
Nanjo
S
,
Arai
S
,
Wang
W
,
Takeuchi
S
,
Yamada
T
,
Hata
A
, et al
.
MET copy number gain is associated with gefitinib resistance in leptomeningeal carcinomatosis of EGFR-mutant lung cancer
.
Mol Cancer Ther
2017
;
16
:
506
15
.
85.
Tamiya
A
,
Tamiya
M
,
Nishihara
T
,
Shiroyama
T
,
Nakao
K
,
Tsuji
T
, 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
:
4177
82
.
86.
Hochmair
M
.
Medical treatment options for patients with epidermal growth factor receptor mutation-positive non-small cell lung cancer suffering from brain metastases and/or leptomeningeal disease
.
Target Oncol
2018
;
13
:
269
85
.
87.
Liu
J
,
Jin
B
,
Su
H
,
Qu
X
,
Liu
Y
.
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
:
702
.
88.
Yang
JCH
,
Kim
SW
,
Kim
DW
,
Lee
JS
,
Cho
BC
,
Ahn
JS
, et al
.
Osimertinib in patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer and leptomeningeal metastases: The BLOOM Study
.
J Clin Oncol
2020
;
38
:
538
47
.
89.
Ahn
MJ
,
Chiu
CH
,
Cheng
Y
,
Han
JY
,
Goldberg
SB
,
Greystoke
A
, 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
:
637
48
.
90.
Zheng
MM
,
Li
YS
,
Sun
H
,
Wu
YL
.
Osimertinib leads the way toward improving outcomes of EGFR-mutant NSCLC with leptomeningeal metastases
.
J Thorac Oncol
2021
;
16
:
e12
e4
.
91.
Zeng
Q
,
Wang
J
,
Cheng
Z
,
Chen
K
,
Johnström
P
,
Varnäs
K
, 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
:
8200
15
.
92.
Yang
Z
,
Guo
Q
,
Wang
Y
,
Chen
K
,
Zhang
L
,
Cheng
Z
, et al
.
AZD3759, a BBB-penetrating EGFR inhibitor for the treatment of EGFR mutant NSCLC with CNS metastases
.
Sci Transl Med
2016
;
8
:
368ra172
.
93.
Ahn
MJ
,
Kim
DW
,
Cho
BC
,
Kim
SW
,
Lee
JS
,
Ahn
JS
, et al
.
Activity and safety of AZD3759 in EGFR-mutant non-small-cell lung cancer with CNS metastases (BLOOM): a phase 1, open-label, dose-escalation and dose-expansion study
.
Lancet Respir Med
2017
;
5
:
891
902
.
94.
Shaw
AT
,
Kim
DW
,
Nakagawa
K
,
Seto
T
,
Crino
L
,
Ahn
MJ
, et al
.
Crizotinib versus chemotherapy in advanced ALK-positive lung cancer
.
N Engl J Med
2013
;
368
:
2385
94
.
95.
Costa
DB
,
Kobayashi
S
,
Pandya
SS
,
Yeo
WL
,
Shen
Z
,
Tan
W
, et al
.
CSF concentration of the anaplastic lymphoma kinase inhibitor crizotinib
.
J Clin Oncol
2011
;
29
:
e443
5
.
96.
Okimoto
T
,
Tsubata
Y
,
Hotta
T
,
Hamaguchi
M
,
Nakao
M
,
Hamaguchi
S-I
, et al
.
A low crizotinib concentration in the cerebrospinal fluid causes ineffective treatment of anaplastic lymphoma kinase-positive non-small cell lung cancer with carcinomatous meningitis
.
Intern Med
2019
;
58
:
703
5
.
97.
Metro
G
,
Lunardi
G
,
Floridi
P
,
Pascali
JP
,
Marcomigni
L
,
Chiari
R
, et al
.
CSF concentration of crizotinib in two ALK-positive non-small-cell lung cancer patients with CNS metastases deriving clinical benefit from treatment
.
J Thorac Oncol
2015
;
10
:
e26
e7
.
98.
Mok
T
,
Camidge
DR
,
Gadgeel
SM
,
Rosell
R
,
Dziadziuszko
R
,
Kim
DW
, et al
.
Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study
.
Ann Oncol
2020
;
31
:
1056
64
.
99.
Ou
SH
,
Sommers
KR
,
Azada
MC
,
Garon
EB
.
Alectinib induces a durable (>15 months) complete response in an ALK-positive non-small cell lung cancer patient who progressed on crizotinib with diffuse leptomeningeal carcinomatosis
.
Oncologist
2015
;
20
:
224
6
.
100.
Gainor
JF
,
Ou
SH
,
Logan
J
,
Borges
LF
,
Shaw
AT
.
The central nervous system as a sanctuary site in ALK-positive non-small-cell lung cancer
.
J Thorac Oncol
2013
;
8
:
1570
3
.
101.
Huber
RM
,
Hansen
KH
,
Paz-Ares Rodriguez
L
,
West
HL
,
Reckamp
KL
,
Leighl
NB
, et al
.
Brigatinib in crizotinib-refractory ALK+ NSCLC: 2-year follow-up on systemic and intracranial outcomes in the phase 2 ALTA trial
.
J Thorac Oncol
2020
;
15
:
404
15
.
102.
Li
Z
,
Li
P
,
Yan
B
,
Gao
Q
,
Jiang
X
,
Zhan
Z
, et al
.
Sequential ALK inhibitor treatment benefits patient with leptomeningeal metastasis harboring non-EML4-ALK rearrangements detected from cerebrospinal fluid: a case report
.
Thorac Cancer
2020
;
11
:
176
80
.
103.
Gaye
E
,
Geier
M
,
Bore
P
,
Guilloique
M
,
Lucia
F
,
Quere
G
, et al
.
Intra-cranial efficacy of brigatinib in an ALK-positive non-small cell lung cancer patient presenting leptomeningeal carcinomatosis
.
Lung Cancer
2019
;
133
:
1
3
.
104.
Zou
Z
,
Xing
P
,
Hao
X
,
Wang
Y
,
Song
X
,
Shan
L
, et al
.
Intracranial efficacy of alectinib in ALK-positive NSCLC patients with CNS metastases-a multicenter retrospective study
.
BMC Med
2022
;
20
:
12
.
105.
Camidge
DR
,
Kim
DW
,
Tiseo
M
,
Langer
CJ
,
Ahn
MJ
,
Shaw
AT
, et al
.
Exploratory analysis of brigatinib activity in patients with anaplastic lymphoma kinase-positive non-small-cell lung cancer and brain metastases in two clinical trials
.
J Clin Oncol
2018
;
36
:
2693
701
.
106.
Bauer
TM
,
Shaw
AT
,
Johnson
ML
,
Navarro
A
,
Gainor
JF
,
Thurm
H
, et al
.
Brain penetration of lorlatinib: cumulative incidences of CNS and non-CNS progression with lorlatinib in patients with previously treated ALK-positive non-small-cell lung cancer
.
Target Oncol
2020
;
15
:
55
65
.
107.
Shaw
AT
,
Bauer
TM
,
de Marinis
F
,
Felip
E
,
Goto
Y
,
Liu
G
, et al
.
First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer
.
N Engl J Med
2020
;
383
:
2018
29
.
108.
Dagogo-Jack
I
,
Oxnard
GR
,
Evangelist
M
,
Digumarthy
SR
,
Lin
JJ
,
Gainor
JF
, et al
.
Phase II study of lorlatinib in patients with anaplastic lymphoma kinase-positive lung cancer and CNS-specific relapse
.
JCO Precis Oncol
2022
;
6
:
e2100522
.
109.
Sharma
SV
,
Haber
DA
,
Settleman
J
.
Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents
.
Nat Rev Cancer
2010
;
10
:
241
53
.
110.
Guo
R
,
Schreyer
M
,
Chang
JC
,
Rothenberg
SM
,
Henry
D
,
Cotzia
P
, et al
.
Response to selective RET inhibition with LOXO-292 in a patient with RET fusion-positive lung cancer with leptomeningeal metastases
.
JCO Precis Oncol
2019
;
3
:
PO.19.00021
.
111.
Kim
DW
,
Barcena
E
,
Mehta
UN
,
Rohlfs
ML
,
Kumar
AJ
,
Penas-Prado
M
, et al
.
Prolonged survival of a patient with metastatic leptomeningeal melanoma treated with BRAF inhibition-based therapy: a case report
.
BMC Cancer
2015
;
15
:
400
.
112.
McLoughlin
EM
,
Fadul
CE
,
Patel
SH
,
Hall
RD
,
Gentzler
RD
.
Clinical and radiographic response of leptomeningeal and brain metastases to encorafenib and binimetinib in a patient with BRAF V600E-mutated lung adenocarcinoma
.
J Thorac Oncol
2019
;
14
:
e269
e71
.
113.
Hendriks
LEL
,
Bootsma
G
,
Mourlanette
J
,
Henon
C
,
Mezquita
L
,
Ferrara
R
, et al
.
Survival of patients with non-small cell lung cancer having leptomeningeal metastases treated with immune checkpoint inhibitors
.
Eur J Cancer
2019
;
116
:
182
9
.
114.
Gion
M
,
Remon
J
,
Caramella
C
,
Soria
JC
,
Besse
B
.
Symptomatic leptomeningeal metastasis improvement with nivolumab in advanced non-small cell lung cancer patient
.
Lung Cancer
2017
;
108
:
72
4
.
115.
Brastianos
PK
,
Lee
EQ
,
Cohen
JV
,
Tolaney
SM
,
Lin
NU
,
Wang
N
, et al
.
Single-arm, open-label phase 2 trial of pembrolizumab in patients with leptomeningeal carcinomatosis
.
Nat Med
2020
;
26
:
1280
4
.
116.
Naidoo
J
,
Schreck
KC
,
Fu
W
,
Hu
C
,
Carvajal-Gonzalez
A
,
Connolly
RM
, et al
.
Pembrolizumab for patients with leptomeningeal metastasis from solid tumors: efficacy, safety, and cerebrospinal fluid biomarkers
.
J Immunother Cancer
2021
;
9
:
e002473
.
117.
Brastianos
PK
,
Strickland
MR
,
Lee
EQ
,
Wang
N
,
Cohen
JV
,
Chukwueke
U
, et al
.
Phase II study of ipilimumab and nivolumab in leptomeningeal carcinomatosis
.
Nat Commun
2021
;
12
:
5954
.
118.
Zheng
MM
,
Tu
HY
,
Yang
JJ
,
Zhang
XC
,
Zhou
Q
,
Xu
CR
, et al
.
Clinical outcomes of non-small cell lung cancer patients with leptomeningeal metastases after immune checkpoint inhibitor treatments
.
Eur J Cancer
2021
;
150
:
23
30
.
119.
Minniti
G
,
Lanzetta
G
,
Capone
L
,
Giraffa
M
,
Russo
I
,
Cicone
F
, et al
.
Leptomeningeal disease and brain control after postoperative stereotactic radiosurgery with or without immunotherapy for resected brain metastases
.
J Immunother Cancer
2021
;
9
:
e003730
.
120.
Prakadan
SM
,
Alvarez-Breckenridge
CA
,
Markson
SC
,
Kim
AE
,
Klein
RH
,
Nayyar
N
, et al
.
Genomic and transcriptomic correlates of immunotherapy response within the tumor microenvironment of leptomeningeal metastases
.
Nat Commun
2021
;
12
:
5955
.
121.
Rubio-Perez
C
,
Planas-Rigol
E
,
Trincado
JL
,
Bonfill-Teixidor
E
,
Arias
A
,
Marchese
D
, et al
.
Immune cell profiling of the cerebrospinal fluid enables the characterization of the brain metastasis microenvironment
.
Nat Commun
2021
;
12
:
1503
.
122.
Le Rhun
E
,
Preusser
M
,
van den Bent
M
,
Andratschke
N
,
Weller
M
.
How we treat patients with leptomeningeal metastases
.
ESMO Open
2019
;
4
:
e000507
.
123.
Kim
HS
,
Park
JB
,
Gwak
HS
,
Kwon
JW
,
Shin
SH
,
Yoo
H
.
Clinical outcome of cerebrospinal fluid shunts in patients with leptomeningeal carcinomatosis
.
World J Surg Oncol
2019
;
17
:
59
.
124.
Mitsuya
K
,
Nakasu
Y
,
Hayashi
N
,
Deguchi
S
,
Takahashi
T
,
Murakami
H
, et al
.
Palliative cerebrospinal fluid shunting for leptomeningeal metastasis-related hydrocephalus in patients with lung adenocarcinoma: a single-center retrospective study
.
PLoS One
2019
;
14
:
e0210074
.
125.
Bander
ED
,
Yuan
M
,
Reiner
AS
,
Garton
ALA
,
Panageas
KS
,
Brennan
CW
, et al
.
Cerebrospinal fluid diversion for leptomeningeal metastasis: palliative, procedural and oncologic outcomes
.
J Neurooncol
2021
;
154
:
301
13
.
126.
Yi
Y
,
Cai
J
,
Xu
P
,
Xiong
L
,
Lu
Z
,
Zeng
Z
, et al
.
Potential benefit of osismertinib plus bevacizumab in leptomeningeal metastasis with EGFR mutant non-small-cell lung cancer
.
J Transl Med
2022
;
20
:
122
.
127.
Zhang
H
,
Wang
Y
,
Wu
H
,
Zhou
S
,
Li
S
,
Meng
X
, et al
.
Olaparib combined with dacomitinib in osimertinib-resistant brain and leptomeningeal metastases from non-small cell lung cancer: a case report and systematic review
.
Front Oncol
2022
;
12
:
877279
.
128.
Zhang
M
,
Ma
W
,
Liu
H
,
Jiang
Y
,
Qin
L
,
Li
W
, et al
.
Osimertinib improves overall survival in patients with leptomeningeal metastases associated with EGFR-mutated non-small-cell lung cancer regardless of cerebrospinal fluid T790M mutational status
.
Evid Based Complement Alternat Med
2021
;
2021
:
6968194
.
129.
Mizusaki
S
,
Otsubo
K
,
Ninomiya
T
,
Arimura
H
,
Tsuchiya-Kawano
Y
,
Inoue
K
.
Remarkable response to dacomitinib in a patient with leptomeningeal carcinomatosis due to EGFR-mutant non-small cell lung cancer
.
Thorac Cancer
2021
;
12
:
114
6
.
130.
Xu
H
,
Zhou
L
,
Lu
Y
,
Su
X
,
Cheng
P
,
Li
D
, et al
.
Dual targeting of the epidermal growth factor receptor using combination of nimotuzumab and erlotinib in advanced non-small-cell lung cancer with leptomeningeal metastases: a report of three cases
.
Onco Targets Ther
2020
;
13
:
647
56
.
131.
Lee
J
,
Choi
Y
,
Han
J
,
Park
S
,
Jung
HA
,
Su
JM
, et al
.
Osimertinib improves overall survival in patients with EGFR-mutated NSCLC with leptomeningeal metastases regardless of T790M mutational status
.
J Thorac Oncol
2020
;
15
:
1758
66
.
132.
Park
S
,
Lee
M-H
,
Seong
M
,
Kim
S
,
Kang
J-H
,
Cho
B
, 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
:
1397
404
.
133.
Sakaguchi
M
,
Maebayashi
T
,
Aizawa
T
,
Ishibashi
N
,
Saito
T
.
Successful treatment of nonsmall cell lung cancer patients with leptomeningeal metastases using whole brain radiotherapy and tyrosine kinase inhibitors
.
J Cancer Res Ther
2020
;
16
:
930
2
.
134.
Saboundji
K
,
Auliac
JB
,
Pérol
M
,
François
G
,
Janicot
H
,
Marcq
M
, et al
.
Efficacy of osimertinib in EGFR-mutated non-small cell lung cancer with leptomeningeal metastases pretreated with EGFR-tyrosine kinase inhibitors
.
Target Oncol
2018
;
13
:
501
7
.
135.
Cho
BC
,
Ahn
M-J
,
Lee
J-S
,
Kim
D-W
,
Kim
S-W
,
John
T
, et al
.
Phase I study (BLOOM) of AZD3759, a BBB penetrable EGFR inhibitor, in EGFRm NSCLC patients with leptomeningeal metastasis (LM) who progressed after other anti-cancer therapy
.
American Society of Clinical Oncology
;
2017
.
136.
Nanjo
S
,
Hata
A
,
Okuda
C
,
Kaji
R
,
Okada
H
,
Tamura
D
, et al
.
Standard-dose osimertinib for refractory leptomeningeal metastases in T790M-positive EGFR-mutant non-small cell lung cancer
.
Br J Cancer
2018
;
118
:
32
7
.
137.
Gong
L
,
Xiong
M
,
Huang
Z
,
Miao
L
,
Fan
Y
.
Icotinib might be effective for the treatment of leptomeningeal carcinomatosis in non-small cell lung cancer with sensitive EGFR mutations
.
Lung Cancer
2015
;
89
:
268
73
.
138.
Jackman
DM
,
Cioffredi
LA
,
Jacobs
L
,
Sharmeen
F
,
Morse
LK
,
Lucca
J
, et al
.
A phase I trial of high dose gefitinib for patients with leptomeningeal metastases from non-small cell lung cancer
.
Oncotarget
2015
;
6
:
4527
.
139.
Yang
H
,
Yang
X
,
Zhang
Y
,
Liu
X
,
Deng
Q
,
Zhao
M
, et al
.
Erlotinib in combination with pemetrexed/cisplatin for leptomeningeal metastases and cerebrospinal fluid drug concentrations in lung adenocarcinoma patients after gefitinib faliure
.
Target Oncol
2015
;
10
:
135
40
.
140.
Lee
E
,
Keam
B
,
Kim
DW
,
Kim
TM
,
Lee
SH
,
Chung
DH
, et al
.
Erlotinib versus gefitinib for control of leptomeningeal carcinomatosis in non-small-cell lung cancer
.
J Thorac Oncol
2013
;
8
:
1069
74
.
141.
Grommes
C
,
Oxnard
GR
,
Kris
MG
,
Miller
VA
,
Pao
W
,
Holodny
AI
, et al
.
“Pulsatile” high-dose weekly erlotinib for CNS metastases from EGFR mutant non-small cell lung cancer
.
Neuro Oncol
2011
;
13
:
1364
9
.
142.
Chow
LQM
,
Barlesi
F
,
Bertino
EM
,
van den Bent
MJ
,
Wakelee
HA
,
Wen
PY
, et al
.
ASCEND-7: efficacy and safety of ceritinib treatment in patients with ALK-positive non-small cell lung cancer metastatic to the brain and/or leptomeninges
.
Clin Cancer Res
2022
;
28
:
2506
16
.
143.
Frost
N
,
Christopoulos
P
,
Kauffmann-Guerrero
D
,
Stratmann
J
,
Riedel
R
,
Schaefer
M
, et al
.
Lorlatinib in pretreated ALK- or ROS1-positive lung cancer and impact of TP53 co-mutations: results from the German early access program
.
Ther Adv Med Oncol
2021
;
13
:
1758835920980558
.
144.
Pellerino
A
,
Buffoni
L
,
Ruda
R
,
Soffietti
R
.
Complete response of spinal metastases from non-small cell lung cancer with ALK inhibitors
.
Neurology
2019
;
93
:
217
9
.
145.
Gainor
JF
,
Chi
AS
,
Logan
J
,
Hu
R
,
Oh
KS
,
Brastianos
PK
, et al
.
Alectinib dose escalation reinduces central nervous system responses in patients with anaplastic lymphoma kinase-positive non-small cell lung cancer relapsing on standard dose alectinib
.
J Thorac Oncol
2016
;
11
:
256
60
.
146.
Metro
G
,
Lunardi
G
,
Bennati
C
,
Chiarini
P
,
Sperduti
I
,
Ricciuti
B
, et al
.
Alectinib's activity against CNS metastases from ALK-positive non-small cell lung cancer: a single institution case series
.
J Neurooncol
2016
;
129
:
355
61
.
147.
Gainor
JF
,
Sherman
CA
,
Willoughby
K
,
Logan
J
,
Kennedy
E
,
Brastianos
PK
, et al
.
Alectinib salvages CNS relapses in ALK-positive lung cancer patients previously treated with crizotinib and ceritinib
.
J Thorac Oncol
2015
;
10
:
232
6
.
148.
Dudnik
E
,
Siegal
T
,
Zach
L
,
Allen
AM
,
Flex
D
,
Yust-Katz
S
, et al
.
Durable brain response with pulse-dose crizotinib and ceritinib in ALK-positive non-small cell lung cancer compared with brain radiotherapy
.
J Clin Neurosci
2016
;
26
:
46
9
.
149.
Arrondeau
J
,
Ammari
S
,
Besse
B
,
Soria
JC
.
LDK378 compassionate use for treating carcinomatous meningitis in an ALK translocated non-small-cell lung cancer
.
J Thorac Oncol
2014
;
9
:
e62
3
.
150.
Ahn
HK
,
Han
B
,
Lee
SJ
,
Lim
T
,
Sun
JM
,
Ahn
JS
, et al
.
ALK inhibitor crizotinib combined with intrathecal methotrexate treatment for non-small cell lung cancer with leptomeningeal carcinomatosis
.
Lung Cancer
2012
;
76
:
253
4
.
151.
Bover
M
,
Yarza
R
,
Docampo
LI
.
Four-year lasting sustained complete response after nivolumab in a patient with non–small-cell lung cancer and confirmed leptomeningeal carcinomatosis: changing the paradigm
.
Clin Lung Cancer
2020
;
21
:
e1
e5
.
152.
Brastianos
PK
,
Prakadan
S
,
Alvarez-Breckenridge
C
,
Lee
EQ
,
Tolaney
SM
,
Nayak
L
, et al
.
Phase II study of pembrolizumab in leptomeningeal carcinomatosis
.
American Society of Clinical Oncology
;
2018
.
153.
Dudnik
E
,
Yust-Katz
S
,
Nechushtan
H
,
Goldstein
DA
,
Zer
A
,
Flex
D
, et al
.
Intracranial response to nivolumab in NSCLC patients with untreated or progressing CNS metastases
.
Lung Cancer
2016
;
98
:
114
7
.