114
Views
0
CrossRef citations to date
0
Altmetric
Review

The early recognition and diagnosis of neoplastic meningitis

ORCID Icon, ORCID Icon, , & ORCID Icon
Pages 105-116 | Received 18 Jul 2023, Accepted 06 Dec 2023, Published online: 25 Dec 2023

References

  • Ozcan G, Singh M, Vredenburgh JJ. Leptomeningeal metastasis from non-small cell lung cancer and Current landscape of treatments. Clin Cancer Res. 2023;29(1):11–29. doi: 10.1158/1078-0432.CCR-22-1585
  • Lamba N, Cagney DN, Catalano PJ, et al. Incidence proportion and prognosis of leptomeningeal disease among patients with breast versus non-breast primaries. Neuro Oncol. 2022 Nov 11;25(5):973–983.
  • Figura NB, Rizk VT, Armaghani AJ, et al. Breast leptomeningeal disease: a review of current practices and updates on management. Breast Cancer Res Treat. 2019;177(2):277–294. doi: 10.1007/s10549-019-05317-6
  • Khaled ML, Tarhini AA, Forsyth PA, et al. Leptomeningeal disease (LMD) in patients with melanoma metastases. Cancers (Basel). 2023;15(6):1884. doi: 10.3390/cancers15061884
  • 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(5):924–932. doi: 10.1016/j.jtho.2019.01.007
  • Li YS, Jiang BY, Yang JJ, et al. Leptomeningeal metastases in patients with NSCLC with EGFR mutations. J Thorac Oncol. 2016;11(11):1962–1969. doi: 10.1016/j.jtho.2016.06.029
  • Abouharb S, Ensor J, Loghin ME, et al. Leptomeningeal disease and breast cancer: the importance of tumor subtype. Breast Cancer Res Treat. 2014;146(3):477–86. doi: 10.1007/s10549-014-3054-z
  • Morikawa A, Jordan L, Rozner R, et al. Characteristics and outcomes of patients with breast cancer with leptomeningeal metastasis. Clin Breast Cancer. 2017;17(1):23–28. doi: 10.1016/j.clbc.2016.07.002
  • Chorti E, Kebir S, Ahmed MS, et al. Leptomeningeal disease from melanoma-poor prognosis despite new therapeutic modalities. Eur J Cancer. 2021;148:395–404. doi: 10.1016/j.ejca.2021.02.016
  • Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41. doi: 10.1038/s41568-019-0205-x
  • Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, et al. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med. 2020;18(1):1. doi: 10.1186/s12967-019-02189-8
  • Mo F, Pellerino A, Soffietti R, et al. Blood-brain barrier in brain tumors: biology and clinical relevance. Int J Mol Sci. 2021 Nov 23;22(23):12654. doi: 10.3390/ijms222312654
  • Keep RF, Jones HC, Hamilton MG, et al. A year in review: brain barriers and brain fluids research in 2022. Fluids Barriers CNS. 2023;20(1):30. doi: 10.1186/s12987-023-00429-0
  • Hu X, Deng Q, Ma L, et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020 Mar;30(3):229–243. doi: 10.1038/s41422-020-0287-8
  • Le Rhun E, Weller M, Brandsma D, et al. EANO executive board and ESMO Guidelines committee. EANO-ESMO clinical practice Guidelines for diagnosis, treatment and follow-up of patients with leptomeningeal metastasis from solid tumours. Ann Oncol. 2017;28(suppl_4):iv84–iv99. doi: 10.1093/annonc/mdx221
  • Le Rhun E, Weller M, van den Bent M, et al. EANO Guidelines committee and ESMO guidelines committee. Electronic address: [email protected]. Leptomeningeal metastasis from solid tumours: EANO-ESMO clinical practice guideline for diagnosis, treatment and follow-up. ESMO Open. 2023 Oct;8(5):101624. doi: 10.1016/j.esmoop.2023.101624
  • Le Rhun E, Devos P, Winklhofer S, et al. Prospective validation of a new imaging scorecard to assess leptomeningeal metastasis: a joint EORTC BTG and RANO effort. Neuro Oncol. 2022 Oct 3;24(10):1726–1735. doi: 10.1093/neuonc/noac043
  • Pellerino A, Brastianos PK, Rudà R, et al. Leptomeningeal metastases from solid tumors: recent advances in diagnosis and molecular approaches. Cancers (Basel). 2021;13(12):2888. doi: 10.3390/cancers13122888
  • Le Rhun E, Devos P, Weller J, et al. Prognostic validation and clinical implications of the EANO ESMO classification of leptomeningeal metastasis from solid tumors. Neuro Oncol. 2021;23(7):1100–1112. doi: 10.1093/neuonc/noaa298
  • Brastianos PK, Carter SL, Santagata S, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 2015;5(11):1164–1177. doi: 10.1158/2159-8290.CD-15-0369
  • Shih DJH, Nayyar N, Bihun I, et al. Genomic characterization of human brain metastases identifies drivers of metastatic lung adenocarcinoma. Nat Genet. 2020;52(4):371–377. doi: 10.1038/s41588-020-0592-7
  • Huang RSP, Harries L, Decker B, et al. Clinicopathologic and genomic landscape of non-small cell lung Cancer brain metastases. Oncology. 2022;27(10):839–848. doi: 10.1093/oncolo/oyac094
  • Lengel HB, Mastrogiacomo B, Connolly JG, et al. Genomic mapping of metastatic organotropism in lung adenocarcinoma. Cancer Cell. 2023 May 8;41(5):970–985.e3. doi: 10.1016/j.ccell.2023.03.018
  • Magbanua MJ, Melisko M, Roy R, et al. Molecular profiling of tumor cells in cerebrospinal fluid and matched primary tumors from metastatic breast cancer patients with leptomeningeal carcinomatosis. Cancer Res. 2013 Dec 1;73(23):7134–7143. doi: 10.1158/0008-5472.CAN-13-2051
  • Li X, Zhang Y, Ding J, et al. Clinical significance of detecting CSF-derived tumor cells in breast cancer patients with leptomeningeal metastasis. Oncotarget. 2017;9(2):2705–2714. doi: 10.18632/oncotarget.23597
  • Cordone I, Masi S, Summa V, et al. Overexpression of syndecan-1, MUC-1, and putative stem cell markers in breast cancer leptomeningeal metastasis: a cerebrospinal fluid flow cytometry study. Breast Cancer Res. 2017;2017(1):46. doi: 10.1186/s13058-017-0827-4
  • Le Rhun E, Guckenberger M, Smits M, et al. EANO executive board and ESMO guidelines committee. Electronic address: [email protected]. EANO-ESMO clinical practice Guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours. Ann Oncol. 2021;32(11):1332–1347. doi: 10.1016/j.annonc.2021.07.016
  • Bander ED, Yuan M, Reiner AS, et al. Cerebrospinal fluid diversion for leptomeningeal metastasis: palliative, procedural and oncologic outcomes. J Neurooncol. 2021 Sep;154(3):301–313. doi: 10.1007/s11060-021-03827-2
  • Boire A, Brandsma D, Brastianos PK, et al. Liquid biopsy in central nervous system metastases: a RANO review and proposals for clinical applications. Neuro Oncol. 2019 May 6;21(5):571–584. doi: 10.1093/neuonc/noz012
  • van Bussel MTJ, Pluim D, Bol M, et al. EpCAM-based assays for epithelial tumor cell detection in cerebrospinal fluid. J Neurooncol. 2018;137(1):1–10. doi: 10.1007/s11060-017-2691-6
  • van Bussel MTJ, Pluim D, Milojkovic Kerklaan B, et al. Circulating epithelial tumor cell analysis in CSF in patients with leptomeningeal metastases. Neurology. 2020 Feb 4;94(5):e521–e528.
  • Campoli MR, Chang CC, Kageshita T, et al. Human high molecular weight-melanoma-associated antigen (HMW-MAA): a melanoma cell surface chondroitin sulfate proteoglycan (MSCP) with biological and clinical significance. Crit Rev Immunol. 2004;24(4):267–96. doi: 10.1615/critrevimmunol.v24.i4.40
  • Le Rhun E, Tu Q, De Carvalho Bittencourt M, et al. Detection and quantification of CSF malignant cells by the CellSearch technology in patients with melanoma leptomeningeal metastasis. Med Oncol. 2013 Jun;30(2):538. doi: 10.1007/s12032-013-0538-3
  • Boral D, Vishnoi M, Liu HN, et al. Molecular characterization of breast cancer CTCs associated with brain metastasis. Nat Commun. 2017;8(1):196. doi: 10.1038/s41467-017-00196-1
  • Riebensahm C, Joosse SA, Mohme M, et al. Clonality of circulating tumor cells in breast cancer brain metastasis patients. Breast Cancer Res. 2019;21(1):101. doi: 10.1186/s13058-019-1184-2
  • Klotz R, Thomas A, Teng T, et al. Circulating Tumor Cells Exhibit Metastatic Tropism and Reveal Brain Metastasis Drivers. Cancer Discov. 2020;10(1):86–103. doi: 10.1158/2159-8290.CD-19-0384
  • Darlix A, Cayrefourcq L, Pouderoux S, et al. Detection of circulating tumor cells in cerebrospinal fluid of patients with suspected breast cancer leptomeningeal metastases: a prospective study. Clin Chem. 2022 Oct 6;68(10):1311–1322. doi: 10.1093/clinchem/hvac127
  • Patel AS, Allen JE, Dicker DT, et al. Identification and enumeration of circulating tumor cells in the cerebrospinal fluid of breast cancer patients with central nervous system metastases. Oncotarget. 2011 Oct;2(10):752–60. doi: 10.18632/oncotarget.336
  • Le Rhun E, Massin F, Tu Q, et al. Development of a new method for identification and quantification in cerebrospinal fluid of malignant cells from breast carcinoma leptomeningeal metastasis. BMC Clin Pathol. 2012;12:21. doi: 10.1186/1472-6890-12-21
  • Subirá D, Serrano C, Castañón S, et al. Role of flow cytometry immunophenotyping in the diagnosis of leptomeningeal carcinomatosis. Neuro Oncol. 2012;14(1):43–52. doi: 10.1093/neuonc/nor172
  • Nayak L, Fleisher M, Gonzalez-Espinoza R, et al. Rare cell capture technology for the diagnosis of leptomeningeal metastasis in solid tumors. Neurology. 2013;80(17):1598–605. discussion 1603 doi: 10.1212/WNL.0b013e31828f183f
  • Lee JS, Melisko ME, Magbanua MJ, et al. Detection of cerebrospinal fluid tumor cells and its clinical relevance in leptomeningeal metastasis of breast cancer. Breast Cancer Res Treat. 2015;154(2):339–349. doi: 10.1007/s10549-015-3610-1
  • Subirá D, Simó M, Illán J, et al. Diagnostic and prognostic significance of flow cytometry immunophenotyping in patients with leptomeningeal carcinomatosis. Clin Exp Metastasis. 2015;32(4):383–91. doi: 10.1007/s10585-015-9716-3
  • Tu Q, Wu X, Le Rhun E, 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(2):352–357. doi: 10.1016/j.lungcan.2015.09.008
  • Acosta M, Pereira J, Arroz M. Screening of carcinoma metastasis by flow cytometry: a study of 238 cases. Cytometry B Clin Cytom. 2016;90(3):289–94. doi: 10.1002/cyto.b.21258
  • Milojkovic Kerklaan B, Pluim D, Bol M, et al. EpCAM-based flow cytometry in cerebrospinal fluid greatly improves diagnostic accuracy of leptomeningeal metastases from epithelial tumors. Neuro Oncol. 2016;18(6):855–862. doi: 10.1093/neuonc/nov273
  • Ma C, Lv Y, Jiang R, et al. Novel method for the detection and quantification of malignant cells in the CSF of patients with leptomeningeal metastasis of lung cancer. Oncol Lett. 2016;11(1):619–623. doi: 10.3892/ol.2015.3971
  • Jiang BY, Li YS, Guo WB, 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 Sep 15;23(18):5480–5488. doi: 10.1158/1078-0432.CCR-17-0047
  • Lin X, Fleisher M, Rosenblum M, et al. Cerebrospinal fluid circulating tumor cells: a novel tool to diagnose leptomeningeal metastases from epithelial tumors. Neuro Oncol. 2017;19(9):1248–1254. doi: 10.1093/neuonc/nox066
  • Nevel KS, DiStefano N, Lin X, et al. A retrospective, quantitative assessment of disease burden in patients with leptomeningeal metastases from non-small-cell lung cancer. Neuro Oncol. 2020 May 15;22(5):675–683. doi: 10.1093/neuonc/noz208
  • Malani R, Fleisher M, Kumthekar P, et al. Cerebrospinal fluid circulating tumor cells as a quantifiable measurement of leptomeningeal metastases in patients with HER2 positive cancer. J Neurooncol. 2020 Jul;148(3):599–606. doi: 10.1007/s11060-020-03555-z
  • Diaz M, Singh P, Kotchetkov IS, et al. Quantitative assessment of circulating tumor cells in cerebrospinal fluid as a clinical tool to predict survival in leptomeningeal metastases. J Neurooncol. 2022;157(1):81–90. doi: 10.1007/s11060-022-03949-1
  • Dirix L, Buys A, Oeyen S, et al. Circulating tumor cell detection: a prospective comparison between CellSearch® and RareCyte® platforms in patients with progressive metastatic breast cancer. Breast Cancer Res Treat. 2022 Jun;193(2):437–444. doi: 10.1007/s10549-022-06585-5
  • Sandri A, Bazhenov N, Orosco H, et al. SDPS-47. Circulating tumor cell analysis from the cerebrospinal fluid informs early diagnosis, treatment and prognosis in leptomeningeal carcinomatosis. Neurooncol Adv. 2023;5(Suppl_3):iii26. doi: 10.1093/noajnl/vdad070.101
  • Wijetunga NA, Boire A, Young RJ, et al. Quantitative cerebrospinal fluid circulating tumor cells are a potential biomarker of response for proton craniospinal irradiation for leptomeningeal metastasis. Neurooncol Adv. 2021 Dec 4;3(1):vdab181. doi: 10.1093/noajnl/vdab181
  • Kumthekar P, Youssef M, Sharma A, et al. The HER2 flip: HER2 amplification of tumor cells in the cerebrospinal fluid (CSF-TCs) of patients with solid tumor leptomeningeal metastasis [abstract]. Neuro Oncol. 2022;23(Supplement_7):vii4–vii5.
  • Gabriel MT, Calleja LR, Chalopin A, et al. Circulating tumor cells: a review of non-EpCAM-Based approaches for cell enrichment and isolation. Clin Chem. 2016 Apr;62(4):571–581. doi: 10.1373/clinchem.2015.249706
  • Kustanovich A, Schwartz R, Peretz T, et al. Life and death of circulating cell-free DNA. Cancer Biol Ther. 2019;20(8):1057–1067. doi: 10.1080/15384047.2019.1598759
  • Tivey A, Church M, Rothwell D, et al. Circulating tumour DNA - looking beyond the blood. Nat Rev Clin Oncol. 2022 Sep;19(9):600–612. doi: 10.1038/s41571-022-00660-y
  • Swinkels DW, de Kok JB, Hanselaar A, et al. Early detection of leptomeningeal metastasis by PCR examination of tumor-derived K-ras DNA in cerebrospinal fluid. Clin Chem. 2000 Jan;46(1):132–133. doi: 10.1093/clinchem/46.1.132
  • Momtaz P, Pentsova E, Abdel-Wahab O, et al. Quantification of tumor-derived cell free DNA(cfDNA) by digital PCR (DigPCR) in cerebrospinal fluid of patients with BRAFV600 mutated malignancies. Oncotarget. 2016 Dec 20;7(51):85430–85436. doi: 10.18632/oncotarget.13397
  • Pentsova EI, Shah RH, Tang J, et al. Evaluating cancer of the central nervous system through next-generation sequencing of cerebrospinal fluid. J Clin Oncol. 2016 Jul 10;34(20):2404–15. doi: 10.1200/JCO.2016.66.6487
  • Marchiò C, Mariani S, Bertero L, et al. Liquoral liquid biopsy in neoplastic meningitis enables molecular diagnosis and mutation tracking: a proof of concept. Neuro Oncol. 2017 Mar 1;19(3):451–453. doi: 10.1093/neuonc/now244
  • Fan Y, Zhu X, Xu Y, et al. Cell-Cycle and DNA-Damage response pathway is involved in leptomeningeal metastasis of non-small cell lung cancer. Clin Cancer Res. 2018 Jan 1;24(1):209–216. doi: 10.1158/1078-0432.CCR-17-1582
  • 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 Apr 1;29(4):945–952. doi: 10.1093/annonc/mdy009
  • Huang R, Xu X, Li D, et al. Digital PCR-Based detection of EGFR mutations in paired plasma and CSF samples of lung adenocarcinoma patients with central nervous system metastases. Target Oncol. 2019 Jun;14(3):343–350. doi: 10.1007/s11523-019-00645-5
  • Ma C, Yang X, Xing W, et al. Detection of circulating tumor DNA from non-small cell lung cancer brain metastasis in cerebrospinal fluid samples. Thorac Cancer. 2020 Mar;11(3):588–593. doi: 10.1111/1759-7714.13300
  • Li H, Xie Y, Lin Y, et al. Different gene mutation spectrum of the paired CSF and plasma samples in lung adenocarcinoma with leptomeningeal metastases: the liquid biopsy based on circulating tumor DNA. Chin J Lung Cancer. 2020;23(8):646–654. doi: 10.3779/j.issn.1009-3419.2020.102.14
  • Zheng MM, Li YS, Tu HY, et al. Genotyping of cerebrospinal fluid associated with osimertinib response and resistance for leptomeningeal metastases in EGFR-Mutated NSCLC. J Thorac Oncol. 2021 Feb;16(2):250–258.
  • Carausu M, Melaabi S, Pierga JY, et al. ESR1 mutation detection and Dynamics in meningeal carcinomatosis in breast cancer. J Breast Cancer. 2019;23(2):218–223. doi: 10.4048/jbc.2020.23.e4
  • Angus L, Deger T, Jager A, et al. Detection of Aneuploidy in Cerebrospinal Fluid from Patients with Breast Cancer Can Improve Diagnosis of Leptomeningeal Metastases. Clin Cancer Res. 2021 May 15;27(10):2798–2806. doi: 10.1158/1078-0432.CCR-20-3954
  • Ballester LY, Glitza OI, Douse DY, et al. Evaluating circulating tumor DNA from the cerebrospinal fluid of patients with melanoma and leptomeningeal disease. J Neuropathol Exp Neurol. 2018 Jul 1;77(7):628–635. doi: 10.1093/jnen/nly046
  • Wu X, Xing P, Shi M, et al. Cerebrospinal fluid cell-free DNA-Based detection of high level of genomic instability is associated with poor prognosis in NSCLC patients with leptomeningeal metastases. Front Oncol. 2022 Apr 28;12:664420.
  • 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 Nov 10;6: 8839. doi: 10.1038/ncomms9839
  • Wang Y, Springer S, Zhang M, et al. Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord. Proc Natl Acad Sci U S A. 2015 Aug 4;112(31):9704–9. doi: 10.1073/pnas.1511694112
  • Pan W, Gu W, Nagpal S, et al. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem. 2015 Mar;61(3):514–22.
  • Adalsteinsson VA, Ha G, Freeman SS, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017 Nov 6;8(1):1324. doi: 10.1038/s41467-017-00965-y
  • Fitzpatrick A, Iravani M, Mills A, et al. Assessing CSF ctDNA to improve diagnostic accuracy and therapeutic monitoring in breast cancer leptomeningeal metastasis. Clin Cancer Res. 2022 Mar 15;28(6):1180–1191. doi: 10.1158/1078-0432.CCR-21-3017
  • White MD, Klein RH, Shaw B, et al. Detection of leptomeningeal disease using cell-free DNA from cerebrospinal fluid. JAMA Netw Open. 2021 Aug 2;4(8):e2120040. doi: 10.1001/jamanetworkopen.2021.20040
  • Chiang CL, Ho HL, Yeh YC, et al. Efficacy of different platforms in detecting EGFR mutations using cerebrospinal fluid cell-free DNA from non-small-cell lung cancer patients with leptomeningeal metastases. Thorac Cancer. 2023 May;14(14):1251–1259. doi: 10.1111/1759-7714.14866
  • Wang H, Wang L, Fang C, et al. Comparison of the diagnostic value of liquid biopsy in leptomeningeal metastases: a systematic review and meta-analysis. Front Oncol. 2022 Dec 19;12:1079796. doi: 10.3389/fonc.2022.1079796
  • Nakasu Y, Deguchi S, Nakasu S, et al. Diagnostic accuracy of cerebrospinal fluid liquid biopsy and MRI for leptomeningeal metastases in solid cancers: a systematic review and meta-analysis. Neurooncol Adv. 2023 Mar 5;5(1):vdad002. doi: 10.1093/noajnl/vdad002.
  • Smalley I, Law V, Wyatt C, et al. Proteomic analysis of CSF from patients with leptomeningeal melanoma metastases identifies signatures associated with disease progression and therapeutic resistance. Clin Cancer Res. 2020 May 1;26(9):2163–2175. doi: 10.1158/1078-0432.CCR-19-2840
  • Glitza Oliva IC, Tawbi HA. “Liquid gold” - the unTapped potential of cerebrospinal fluid analysis? Clin Cancer Res. 2020 May 1;26(9):2083–2084.
  • Schmid D, Warnken U, Latzer P, et al. Diagnostic biomarkers from proteomic characterization of cerebrospinal fluid in patients with brain malignancies. J Neurochem. 2021 Jul;158(2):522–538.
  • Kwon JW, Im JH, Lee KY, et al. Different Metabolomic and Proteomic Profiles of Cerebrospinal Fluid in Ventricular and Lumbar Compartments in Relation to Leptomeningeal Metastases. Metabolites. 2022 Jan 14;12(1):80. doi: 10.3390/metabo12010080
  • Conrad C, Dorzweiler K, Miller MA, et al. Profiling of metalloprotease activities in cerebrospinal fluids of patients with neoplastic meningitis. Fluids Barriers CNS. 2017 Aug 14;14(1):22. doi: 10.1186/s12987-017-0070-5
  • Boire A, Zou Y, Shieh J, et al. Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis. Cell. 2017 Mar 9;168(6):1101–1113.e13. doi: 10.1016/j.cell.2017.02.025
  • Chi Y, Remsik J, Kiseliovas V, et al. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science. 2020 Jul 17;369(6501):276–282. doi: 10.1126/science.aaz2193
  • Wilcox J, Modelevsky LR, Cremers TTS, et al. A phase Ia/Ib study of intrathecal deferoxamine in patients with leptomeningeal metastases. J Clin Oncol. 2022;40(16_suppl):TPS2074–TPS2074. doi: 10.1200/JCO.2022.40.16_suppl.TPS2074
  • Boire A, Brastianos PK, Garzia L, et al. Brain metastasis. Nat Rev Cancer. 2020 Jan;20(1):4–11. doi: 10.1038/s41568-019-0220-y
  • Wessels PH, Boelens MC, Monkhorst K, et al. A review on genetic alterations in CNS metastases related to breast cancer treatment. Is there a role for liquid biopsies in CSF? J Neurooncol. 2023 Mar;162(1):1–13. doi: 10.1007/s11060-023-04261-2
  • Seoane J, De Mattos-Arruda L, Le Rhun E, et al. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann Oncol. 2019 Feb 1;30(2):211–218. doi: 10.1093/annonc/mdy544
  • Morganti S, Parsons HA, Lin NU, et al. Liquid biopsy for brain metastases and leptomeningeal disease in patients with breast cancer. NPJ Breast Cancer. 2023 May 24;9(1):43. doi: 10.1038/s41523-023-00550-1
  • Wijaya JH, Patel UD, Quintero-Consuegra MD, et al. Liquid biopsy in the setting of leptomeningeal metastases: a systematic review and meta-analysis. J Neurooncol. 2023 Nov 29. doi: 10.1007/s11060-023-04519-9

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.