Skip to main content

Prognostic value of cell-free DNA in cerebrospinal fluid from lung cancer patients with brain metastases during radiotherapy

Abstract

Background

During the last decades, radiotherapy (RT) for non-small cell lung cancer (NSCLC) with brain metastases (BM) has been developed. However, the lack of predictive biomarkers for therapeutic responses has limited the precision treatment in NSCLC-BM.

Patients and methods

In order to find the predictive biomarkers for RT, we investigated the influence of RT on the cell-free DNA (cfDNA) from cerebrospinal fluid (CSF) and the frequency of T cell subsets of NSCLC patients with BM. A total of 19 patients diagnosed as NSCLC with BM were enrolled. The CSF from 19 patients and matched plasma samples from 11 patients were collected before RT, during RT, and after RT. The cfDNA from CSF and plasma were extracted, and the cerebrospinal fluid tumor mutation burden (cTMB) was calculated after through next-generation sequencing. The frequency of T cell subsets in peripheral blood was using flow cytometry.

Results

The detection rate of cfDNA was higher in CSF compared to plasma in the matched samples. The mutation abundance of cfDNA in CSF was decreased after RT. However, no significant difference was observed in cTMB before and after RT. Although the median intracranial progression-free survival (iPFS) has not yet been reached in patients with decreased or undetectable cTMB, there was a trend that these patients possessed longer iPFS compared to those with stable or increased cTMB (HR 0.28, 95% CI 0.07ā€“1.18, Pā€‰=ā€‰0.067). The proportion of CD4+T cells in peripheral blood was decreased after RT. Conclusion: Our study indicates that cTMB can serve as a prognostic biomarker in NSCLC patients with BMs.

Introduction

Brain metastases (BM) are common and lethal for advanced non-small cell lung carcinoma (NSCLC) [1]. About 10% of BM occurs in newly diagnosed NSCLC patients, while 25ā€“40% of them occur after curative treatment [2]. Although therapeutics, including targeted therapy, immunotherapy, and radiotherapy (RT), were rapidly developed in the past decade, the clinical outcomes and quality of life of NSCLC with BM are relatively poor. There is a lack of evidence-based management of NSCLC patients with BMs. Recently, with intensive studies on cancer driver genes and the emergence of novel targeted therapies, it has been proved that some targeted drugs can pass through the bloodā€“brain barrier, which subsequently prevents or control brain metastases for a while [3,4,5,6]. However, the treatment efficacy is limited to asymptomatic or stable BM [7], and some patients will develop central nervous system progression eventually [8,9,10].

Radiotherapy (RT) is the standard treatment for BMs of NSCLC. For patients with driver gene mutation, a combination of targeted therapy with RT might shed light on those patients. However, the conclusions of previously published retrospective clinical studies are inconsistent [11,12,13]. Currently, the evaluation of treatment efficacy is mainly based on radiographic images, which can neither reflect the treatment efficacy immediately nor the influence of RT on molecular clones of BM, failing to instruct clinicians to design personalized treatment plans. Therefore, biomarkers are urgently needed to predict the efficacy of combination therapy.

For patients without driver gene mutation or those resistant to multiple targeted therapies, immune checkpoint inhibitors (ICIs) provide another promising treatment approach by reinvigorating the immune cells [14]. It has been confirmed that the combination of ICIs and RT can mutually enhance the efficacy in treating BM from NSCLC [15]. However, it is still unclear whether RT can induce and stimulate the immune system of NSCLC patients with BM.

Recently, it has been reported that cell-free DNA (cfDNA) from cerebrospinal fluid (CSF) is more sensitive to the detection of mutated genes than that in plasma, which thus can reflect the treatment response in time [16,17,18,19]. In addition, tumor mutation burden (TMB) can be calculated accurately based on large next-generation sequencing (NGS) panel covering a genome areaā€‰>ā€‰1Ā Mb [20]. In the present study, we explored the dynamic changes of cfDNA in CSF and the frequency of T cell subsets in peripheral blood during RT from NSCLC-BM patients. We figured out for the first time that the change of cerebrospinal fluid tumor mutation burden (cTMB) before and after radiotherapy could predict intracranial progression-free survival (iPFS) in NSCLC patients with BMs.

Methods

Patients and sample collection

Nineteen patients were included in the study. All patients were diagnosed as NSCLC with BMs between March 2018 and January 2020 from Southern Medical University or Guangdong Sanjiu Brain Hospital, and the last follow-up time was April 19th, 2020. All patients received chest computed tomography (CT) or positron emission tomography/computed tomography (PET/CT), gadolinium-enhanced magnetic resonance imaging (MRI) of the brain, and pathological examination or CSF cytologic examination or mutation detection via NGS of cfDNA in CSF to confirm the diagnosis of NSCLC with brain or meningeal metastases. Approximately 10Ā mL CSF was collected by lumbar puncture in each patient before RT (Before RT), in the middle (During RT), and at the end of RT (After RT), and an additional 8Ā mL of peripheral blood was collected at the same time from 8 patients. Plasma was extracted within 2Ā h of blood collection, and all samples were shipped to the central testing laboratory within 48Ā h (Additional file 1: Figure S1). All patients provided signed informed consent, and the study protocol was approved by the research ethics committee of each hospital.

Targeted NGS and data processing

Circulating cfDNA from plasma was extracted using the QIAamp Circulating Nucleic Acid kit (Qiagen). Sequencing libraries were prepared using the KAPA Hyper Prep Kit (KAPA Biosystems) according to the manufacturer's instructions. Customized xGen lockdown probes (Integrated DNA Technologies) targeting 520 or 474 cancer-relevant genes were used for hybridization enrichment. Then, the capture reaction was performed with Dynabeads M-270 (Life Technologies) and xGen Lockdown hybridization and wash kit (Integrated DNA Technologies), following the manufacturers' instructions. Captured libraries were on-beads PCR amplified with Illumina p5 (5ā€² AAT GAT ACG GCG ACC ACC GA 3ā€²) and p7 primers (5ā€² CAA GCA GAA GAC GGC ATA CGA GAT 3ā€²) in KAPA HiFi HotStart ReadyMix (KAPA Biosystems), followed by purification using Agencourt AMPure XP beads. Libraries were quantified by qPCR using the KAPA Library Quantification kit (KAPA Biosystems). Library fragment size was determined by Bioanalyzer 2100 (Agilent Technologies). According to the manufacturer's instruction, the target-enriched library was then sequenced on the HiSeq4000 NGS platform (Illumina). The mean coverage depth was 143Ɨ for the whole blood control samples and 4000Ɨ for cfDNA samples.

Sequence alignment and processing

Base calling was performed on bcl2fastq v2.16.0.10 (Illumina, Inc.) to generate sequence reads in FASTQ format (Illumina 1.8ā€‰+ā€‰encoding). Quality control (QC) was performed with Trimmomatic. High-quality reads were mapped to the human genome (hg19, GRCh37 Genome Reference Consortium Human Reference 37) using the BWA aligner 0.7.12 with the BWA-MEM algorithm and default parameters to create SAM files. Picard 1.119 was used to convert SAM files to compressed BAM files, sorted according to chromosome coordinates. The Genome Analysis Toolkit (GATK, version 3.4ā€“0) was used to locally realign the BAMs files at intervals with indel mismatches and recalibrate base quality scores of reads in BAM files.

Single nucleotide variants (SNV)/short insertions/deletions (InDels)/Copy number variations (CNVs) detections

SNVs and InDels were identified by VarScan2 2.3.9 with minimum variant allele frequency threshold set at 0.01 and P-value threshold for calling variants set at 0.05 to generate Variant Call Format (VCF) files. All SNVs/InDels were annotated with ANNOVAR, and each SNV/InDel was manually checked on the Integrative Genomics Viewer (IGV). CNVs were detected using in-house-developed software.

Statistical analysis

Comparisons of TMB and subset of T cells before RT, during RT, and after RT were done using one-way "repeated measures" ANOVA analysis with a Bonferroni post-hoc test by GraphPad prism 8.0. Comparisons of mutation allele frequency between groups were done using unpaired T-test by GraphPad prism 8.0. Kaplanā€“Meier curves were compared using the log-rank test for survival analyses, and the Cox proportional hazards model calculated hazard ratios (HR). A two-sided P value of less than 0.05 was considered significant for all tests unless indicated otherwise. The rest statistical analyses was done in R (v.3.3.2).

Results

Patient characteristics

Between March 2018 and January 2020, 19 patients were screened for enrolment at Southern Medical University or Guangdong Sanjiu Brain Hospital. All patients were diagnosed with NSCLC with pathological examination, except for D04, who was diagnosed by detecting epidermal growth factor receptor (EGFR) 19del in CSF cfDNA. Baseline demographics and clinical characteristics are listed in Table 1 and Additional file 2: Table S1. We detected cfDNA in all 19 patients. In addition, five patients (D06, D07, S01, S03, S08) were diagnosed with leptomeningeal metastasis by CSF cytologic examination. Leptomeningeal metastasis was also found in 3 patients (S02, S05, S07) by neurological symptoms and gadolinium-enhanced MRI. Notably, four patients (D01, D04, D07, S11) did not receive any concurrent system therapy during RT.

Table 1 Clinicopathological feature of NSCLC patients with brain metastases and changes of CSF mutation status before and after brain radiotherapy

Influence of RT on cfDNA in CSF from NSCLC-BM patients

We compared cfDNA levels in the CSF before, during, and after radiotherapy. The amount of CSF cfDNA noticeable increased in D07 after RT, while the amount of CSF cfDNA decreased in D06 and S01, and no apparent change in the amount of CSF cfDNA occurred after RT in other patients (Fig.Ā 1). And the detection rate of CSF cfDNA during RT and at the end of RT was slightly lower than before RT in the cohort. As expected, the detection rate of cfDNA was higher in CSF compared to plasma in the validation cohort (Fig.Ā 2A). The composition of variants types was similar before and after RT in most patients except D06, with many copy number variants occurring at the end of RT (Additional file 1: Figure S2). A statistical increase in CSF mutation abundance was detected after RT compared to that before RT (Fig.Ā 2B).

Fig. 1
figure 1

The amount of CSF cfDNA. A CSF cfDNA of patients D01-D08 collected at 0Ā day (baseline), 7th day and 10th day (during RT therapy), 20th day (after RT therapy) since the RT therapy began. B CSF cfDNA of patients S01ā€“S11 collected at 0Ā day (baseline), 10th day (during RT therapy), 20th day (after RT) since the RT therapy began. RT radiation therapy

Fig. 2
figure 2

A The cfDNA detection rate of CSF and plasma. RT radiation therapy, CSF cerebrospinal fluid. B Genomic mutations abundance from the CSF. C1 before RT, C2 during RT, and C3 after RT. RT radiation therapy, CSF cerebrospinal fluid

The trend is that mutation abundance in CSF increased in the beginning and then decreased subsequently during RT was observed in 13 patients (D01, D03, D04, D07, D08, S01, S02, S03, S05, S06, S07, S10, S11) while the numbers of mutations were stable or decreased in those patients (Fig.Ā 3). Conversely, CSF mutations abundance and number of mutations obviously increased after RT occurred in D06, S04, and S08. Novel mutations were detected in 7 patients (D03, D06, D07, S03, S04, S06, S08) at the end of RT. And novel mutations detected in D03, S03, and S04 can also be detected in the paired tumor tissue or plasma samples. Additionally, no mutations were detected in the CSF before, during, or after RT in D02, D05, and S09 patients.

Fig. 3
figure 3

Genomic alterations in the CSF of training cohort at different timepoint. INDEL insertion-deletion, CNV copy number variation, CSF Cerebrospinal Fluid, RT radiation therapy

Compared to patients with BM who progressed after systematic treatment (2/5) or newly diagnosed patients with BM (2/6), rare, uncommon, or resistant mutations reported in previous studies were prone to be detected in patients with BM newly diagnosed after systematic treatment (6/7). And the number and abundance of those mutations decreased during RT.

No significant difference was observed in CSF TMB (cTMB) before and after RT (Fig.Ā 4A). Although the median iPFS of patients with decreased or undetectable cTMB has not been reached yet, there was a trend that these patients possessed longer iPFS compared to those with stable or increased cTMB (median: 4.43Ā months, range: 0.23ā€“8.63Ā months) (HR 0.28, 95% CI 0.07ā€“1.18, Pā€‰=ā€‰0.067) (Fig.Ā 4B).

Fig. 4
figure 4

A Cerebrospinal fluid TMB change of cohort before RT, during RT, and after RT. No significant difference was observed. CSF cerebrospinal fluid, TMB tumor mutation burden, RT radiation therapy, ns: no significance. B Kaplanā€“Meier curves of intracranial progression free survival (iPFS) comparing cerebrospinal fluid TMB decrease/undetectable and TMB increase/no change. C Plasma TMB change before RT, during RT, and after RT. No significant difference was observed. TMB tumor mutation burden, RT radiation therapy, ns no significance

Risk factors of intracranial progression-free survival (iPFS)

To figure out independent risk factors predicting iPFS of patients undergoing RT for BMs with or without EGFR-TKIs, we further conducted subgroup analysis based on the diagnostic type of brain metastasis, leptomeninges involvement, performance status (PS) score, Graded Prognostic Assessment (GPA) score and detection of novel mutations at the end of RT (Additional file 1: Figure S3). However, no significant independent risk factors were found, probably due to the limited sample size and short follow-up time. However, in this study, the median iPFS of patients with BMs progression after initial EGFR-TKIs with or without chemotherapy (6.47Ā months) tends to be the shortest compared to patients with newly diagnosed BMs (not yet reached) and patients with newly diagnosed BMs after systematic treatment (with or without TKI) (9.13Ā months). Probable explanations for this phenomenon include those patients with BMs progression after initial EGFR-TKIs with or without chemotherapy substantially possessing dismal survival outcomes, or non-salvage RT could benefit patients with BMs. The exact reasons are worth further exploration.

Influence of RT for BMs on blood immune indexes

Concordant with previous results, there was no significant difference in plasma TMB before and after RT (Fig.Ā 4C). In addition, analyzing the change of T cell subsets in peripheral blood, we observed that the proportion of CD4+ naĆÆve T cells significantly decreased after RT. Although, the change in the proportion of total T cells, natural killer (NK) cells and B cells is not statistically significant, there is an increasing trend of the proportion of total T cells and B cells during radiotherapy. The regulatory T (Treg) cells, a kind of as suppressor T cells, has no significant changes. We did not find significant changes in the proportion of other T cell subsets, including memory T cells (CD4+CD45RO+), cytotoxic T cells (CD3+CD8+), helper T cells (CD3+CD4+), CD8+CD28+ T cells, and CD8+CD28āˆ’ T cells (Additional file 1: Figure S4).

Discussion

BM is a challenge for most clinicians, and whole brain radiotherapy (WBRT) is the acknowledged standard approach with proven efficacy for those patients. Although some studies on targeted therapies posed a challenge to RT in patients with driver gene mutation BM, a head-to-head clinical trial comparing efficacy between targeted therapy alone and combinations of targeted therapy with RT is still lacking. Thus, combining targeted therapy with RT for BMs is still a controversial question drawing intense clinical attention. Besides radiographic images, evaluation of the efficacy of RT alone or in combination therapy for BM from NSCLC remains limited. With the development of NGS, it provides a novel approach for evaluation of therapeutic efficacy by analysis of CSF.

A study presented at the American society of clinical oncology (ASCO) annual meeting 2020 reported that the median time from the initiation of RT (chest: 2.5ā€“4.0Ā Gy/F; brain: 6ā€“9Ā Gy/F) to the peak of the amount of ctDNA was two days (IQR:1ā€“3Ā days) [21]. In plasma, ctDNA accounts for a small fraction of cfDNA because most cfDNA is derived from non-tumor cells, especially blood cells [22]. Compared with plasma cfDNA, most CSF cfDNA with high sensitivity is derived from tumor lysis. During and after radiotherapy, tumor cell destruction leads to the release of intracellular content including cfDNA. In the present study, the amount of CSF cfDNA significantly increased in only 5.3% (1/19) of patients. At the same time, 10.5% (2/19) of patients significantly reduced. There is no dynamic change in CSF cfDNA found during RT (10ā€“14Ā days after initiation) and/or at the end of RT (26ā€“28Ā days after initiation) compared to that before RT. Consistent rate of novel mutation detected in CSF cfDNA reported in previous studies, novel mutations were detected in CSF cfDNA after treatment in 21% (4/19) of patients in the present study. Notably, novel mutations detected in 3 patients (D03, S03, S04) CSF cfDNA at the end of RT, which were absent in CSF cfDNA before treatment, were also detected in corresponding lung tumor or plasma samples. It is suggested that radiation-induced tumor lysis might increase the sensitivity of the detection of CSF cfDNA.

Itā€™s reported that the mechanism of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) includes EGFR T790M mutation, MET or ERBB2 amplification, and a shortened time to progression of patients undergoing EGFR-TKIs was correlated with mutations in ERBB2, MET, and TP53 [23]. Besides, the co-occurrence of deletion of CDKN2A and mutations of EGFR generally indicated poor treatment response to EGFR-TKIs in lung cancer [24, 25]. Subclones resistant to EGFR-TKIs might originate from various genetic alterations, including TP53 mutation, CDKN2A deletion, and RB1 deletion in tumor stem cells [25, 26]. Kenji Sugio reported that loss of PTEN might contribute to the acquired resistance of EGFR-TKIs in patients with uncommon EGFR G719X mutation [27]. MDM2 amplification was found to induce the primary resistance of EGFR-TKIs and predict poor prognosis in NSCLC patients [28]. Itā€™s reported that CDC73, SMAD4, RB1, and PIK3CA might also take an essential part in primary resistance to EGFR-TKIs [29]. Additionally, Keunchil Park found that genetic alterations of TP53, RB1, PTEN, and MDM2 were also independently associated with worse PFS in patients taking third-generation TKI after initial EGFR-TKI failure [30]. Our study previously reported that mutations related to acquired or primary resistance to EGFR-TKIs were detected in 52.63% (10/19) of patients. Except for S08, the abundance or amount of EGFR-TKIs resistance-related mutations was decreased variously during RT in patients harboring these mutations. Notably, 70% (7/10) of these patients possessed longer iPFS (>ā€‰110Ā days), which indicates that RT alone or a combination of EGFR-TKIs with RT might effectively reduce or eliminate EGFR-TKIs resistant clones in BMs from NSCLC.

Considering the large gene panel (>ā€‰300 genes) used in the present study, we also calculated the CSF tumor mutation burden (cTMB) and explored its clinical value in patients with BMs. Notably, the median iPFS of patients with decreased or undetectable cTMB was longer than those without. However, this finding needs to be validated in multicenter, large cohorts. Moreover, whether cTMB can serve as a prognostic biomarker just like blood TMB remains largely unknown, and further research is needed. In addition, whether RT for BMs would stimulate the response of the immune system was also explored preliminarily in our study. Based on analyses of lymphocyte subsets in blood and plasma TMB, we found that the influence imposed on the immune system by RT for BMs was indeterminate.

Conclusion

RT was associated with treatment benefits in advanced NSCLC patients with brain metastases. cfDNA in cerebrospinal fluid is more sensitive to the detection of mutated genes than that in plasma, which could be a more valuable predictive marker of radiotherapy efficacy. Although there was no significant difference in plasma TMB before and after RT, the proportion of CD4ā€‰+ā€‰naĆÆve T cells significantly decreased after RT. Taken together, these results indicate that radiation-induced tumor lysis might increase the sensitivity of the detection of CSF cfDNA. Besides, RT alone or a combination of EGFR-TKIs with RT might effectively reduce or eliminate EGFR-TKIs resistant clones in BMs from NSCLC. Evaluating a real-world population of NSCLC patients with BM will prove the predictive value of cfDNA in cerebrospinal fluid definitively in the future.

Availability of data and materials

Datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Schuette W. Treatment of brain metastases from lung cancer: chemotherapy. Lung Cancer. 2004;45(Suppl 2):S253-257.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  2. Dempke WC, Edvardsen K, Lu S, et al. Brain metastases in NSCLC - are TKIs changing the treatment strategy? Anticancer Res. 2015;35:5797ā€“806.

    CASĀ  PubMedĀ  Google ScholarĀ 

  3. Jeffrey V, Brower H, Ian R. Erlotinib for the treatment of brain metastases in non-small cell lung cancer. Expert Opin Pharmacother. 2016;17(7):1013ā€“21.

    ArticleĀ  Google ScholarĀ 

  4. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med. 2017;377:829ā€“38.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Soria JC, Tan DSW, Chiari R, et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet. 2017;389:917ā€“29.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  6. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 2018;378:113ā€“25.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Protopapa M, Kouloulias V, Nikoloudi S, et al. From whole-brain radiotherapy to immunotherapy: a multidisciplinary approach for patients with brain metastases from NSCLC. J Oncol. 2019;2019:3267409.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  8. Cho BC, Chewaskulyong B, Lee KH, et al. Osimertinib versus standard of care EGFR TKI as first-line treatment in patients with EGFRm advanced NSCLC: FLAURA Asian subset. J Thorac Oncol. 2019;14:99ā€“106.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  9. Su PL, Wu YL, Chang WY, et al. Preventing and treating brain metastases with three first-line EGFR-tyrosine kinase inhibitors in patients with EGFR mutation-positive advanced non-small cell lung cancer. Ther Adv Med Oncol. 2018;10:1758835918797589.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  10. Yang JJ, Zhou C, Huang Y, et al. Icotinib versus whole-brain irradiation in patients with EGFR-mutant non-small-cell lung cancer and multiple brain metastases (BRAIN): a multicentre, phase 3, open-label, parallel, randomised controlled trial. Lancet Respir Med. 2017;5:707ā€“16.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Ponce S, Bruna J, Juan O, et al. Multidisciplinary expert opinion on the treatment consensus for patients with EGFR mutated NSCLC with brain metastases. Crit Rev Oncol Hematol. 2019;138:190ā€“206.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  12. Jiang T, Chunxia Su, Li X, Zhao C, Zhou F, Ren S, Zhou C, Zhang J. EGFR TKIs plus WBRT demonstrated no survival benefit other than that of TKIs alone in patients with NSCLC and EGFR mutation and brain metastases. J Thorac Oncol. 2016;11(10):1718ā€“28.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  13. Magnuson WJ, Lester-Coll NH, Wu AJ, Yang TJ, Lockney NA, Gerber NK, Beal K, Amini A, Patil T, Kavanagh BD, Camidge DR, Braunstein SE, Boreta LC, Balasubramanian SK, Ahluwalia MS, Rana NG, Attia A, Gettinger SN, Contessa JN, Yu JB, Chiang VL. Management of brain metastases in tyrosine kinase inhibitor-naĆÆve epidermal growth factor receptor-mutant non-small-cell lung cancer: a retrospective multi-institutional analysis. J Clin Oncol. 2017;35(10):1070ā€“7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Reck M, Mok TSK, Nishio M, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med. 2019;7:387ā€“401.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Li W, Yu H. Separating or combining immune checkpoint inhibitors (ICIs) and radiotherapy in the treatment of NSCLC brain metastases. J Cancer Res Clin Oncol. 2020;146:137ā€“52.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  16. De Mattos-Arruda L, Mayor R, Ng CKY, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Zheng MM, Li YS, Jiang BY, et al. Clinical utility of cerebrospinal fluid cell-free DNA as liquid biopsy for leptomeningeal metastases in ALK-rearranged NSCLC. J Thorac Oncol. 2019;14:924ā€“32.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Li YS, Jiang BY, Yang JJ, 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;29:945ā€“52.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  19. Xing L, Pan Y, Shi Y, et al. Biomarkers of osimertinib response in patients with refractory, EGFR-T790M-positive non-small cell lung cancer and central nervous system metastases: the APOLLO study. Clin Cancer Res. 2020;26:6168ā€“75.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  20. Fancello L, Gandini S, Pelicci PG, Mazzarella L. Tumor mutational burden quantification from targeted gene panels: major advancements and challenges. J Immunother Cancer. 2019;7:183.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  21. Breadner DA, Vincent MD, Correa RJM, et al. Exploitation of treatment induced tumor lysis to enhance sensitivity of ctDNA analysis: a first-in-human pilot study. Cancer Res. 2020;38:3530ā€“3530.

    Google ScholarĀ 

  22. Mouliere F, Chandrananda D, Piskorz AM, et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med. 2018;10(466):eaat4921.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  23. Yu HA, Suzawa K, Jordan E, et al. Concurrent alterations in EGFR-mutant lung cancers associated with resistance to EGFR kinase inhibitors and characterization of MTOR as a mediator of resistance. Clin Cancer Res. 2018;24:3108ā€“18.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  24. Chang SC, Lai YC, Chang CY, et al. Concomitant genetic alterations are associated with worse clinical outcome in EGFR mutant NSCLC patients treated with tyrosine kinase inhibitors. Transl Oncol. 2019;12:1425ā€“31.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  25. Jiang J, Gu Y, Liu J, et al. Coexistence of p16/CDKN2A homozygous deletions and activating EGFR mutations in lung adenocarcinoma patients signifies a poor response to EGFR-TKIs. Lung Cancer. 2016;102:101ā€“7.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  26. Nahar R, Zhai W, Zhang T, et al. Elucidating the genomic architecture of Asian EGFR-mutant lung adenocarcinoma through multi-region exome sequencing. Nat Commun. 2018;9:216.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  27. Osoegawa A, Hashimoto T, Takumi Y, et al. Acquired resistance to an epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) in an uncommon G719S EGFR mutation. Invest New Drugs. 2018;36:999ā€“1005.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  28. Sun D, Zhu Y, Zhu J, et al. Primary resistance to first-generation EGFR-TKIs induced by MDM2 amplification in NSCLC. Mol Med. 2020;26:66.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  29. Jin Y, Shi X, Zhao J, et al. Mechanisms of primary resistance to EGFR targeted therapy in advanced lung adenocarcinomas. Lung Cancer. 2018;124:110ā€“6.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  30. Kim Y, Lee B, Shim JH, et al. Concurrent genetic alterations predict the progression to target therapy in EGFR-mutated advanced NSCLC. J Thorac Oncol. 2019;14:193ā€“202.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (81572279, 81972853), and Clinical Research Startup Program of Southern Medical University by High-level University Construction Funding of Guangdong Provincial Department of Education (LC2019ZD009).

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: ZXX. Administrative support: ZXX. Provision of study materials or patients: ZXX & CLB. Collection and assembly of data: QSM & HYY. Data analysis and interpretation: DXT & WLJ. Manuscript writing: All authors. Final approval of manuscript: All authors. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Xiaoxia Zhu.

Ethics declarations

Ethics approval and consent to participate

The present study was approved by the research ethics committee of affiliated hospitals of Southern Medical University or Guangdong Sanjiu Brain Hospital.

Competing interests

All authors have completed the ICMJE uniform disclosure form. The authors have no conflicts of interest to declare.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. Supplementary figures.

Additional file 2

. Supplementary tables.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qiao, S., Hao, Y., Cai, L. et al. Prognostic value of cell-free DNA in cerebrospinal fluid from lung cancer patients with brain metastases during radiotherapy. Radiat Oncol 18, 50 (2023). https://doi.org/10.1186/s13014-023-02239-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-023-02239-y

Keywords