Cerebrospinal Fluid Cell-Free Tumour DNA As A Liquid Biopsy For Primary Brain Tumours and Central Nervous System Metastases
Cerebrospinal Fluid Cell-Free Tumour DNA As A Liquid Biopsy For Primary Brain Tumours and Central Nervous System Metastases
Cerebrospinal Fluid Cell-Free Tumour DNA As A Liquid Biopsy For Primary Brain Tumours and Central Nervous System Metastases
doi:10.1093/annonc/mdy544
Published online 21 December 2018
REVIEW
*Correspondence to: Prof. Joan Seoane, Vall d’Hebron Institute of Oncology, C/Natzaret, 115-117, 08035 Barcelona, Spain.
Tel: þ34-93-254-34-50; E-mail: [email protected]
Challenges in obtaining tissue specimens from patients with brain tumours limit the diagnosis and molecular characterisation
and impair the development of better therapeutic approaches. The analysis of cell-free tumour DNA in plasma (considered a
liquid biopsy) has facilitated the characterisation of extra-cranial tumours. However, cell-free tumour DNA in plasma is limited in
quantity and may not reliably capture the landscape of genomic alterations of brain tumours. Here, we review recent work
assessing the relevance of cell-free tumour DNA from cerebrospinal fluid in the characterisation of brain cancer. We focus on the
advances in the use of the cerebrospinal fluid as a source of cell-free tumour DNA to facilitate diagnosis, reveal actionable
genomic alterations, monitor responses to therapy, and capture tumour heterogeneity in patients with primary brain tumours
and brain and leptomeningeal metastases. Profiling cerebrospinal fluid cell-free tumour DNA provides the opportunity to
precisely acquire and monitor genomic information in real time and guide precision therapies.
Key words: cerebrospinal fluid, circulating cell-free tumour DNA, glioblastoma, brain metastasis,
liquid biopsy, brain cancer
C The Author(s) 2018. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
V
All rights reserved. For permissions, please email: [email protected].
Review Annals of Oncology
This implies that an intraoperative histological diagnosis may aiding in monitoring response to therapy, and allowing deconvo-
be required possibly delaying the surgical procedure. Repeat lution of tumour heterogeneity in patients with CNS cancer
surgical interventions may be needed to differentiate tumour (Figure 1).
pseudoprogression induced by treatment from true relapse.
The challenges in obtaining tumour tissue have led physicians
to rely on primary archival tumour specimens. Thus, in some
cases, therapies for brain cancer are selected based on the mo- Primary brain tumours
lecular characteristics of the primary tumour that can differ
from the current tumour manifestation [3, 4]. Diagnostic considerations
Plasma cell-free circulating tumour DNA (ctDNA) has been Primary brain tumours encompass a large variety of lesions
used as a ‘liquid biopsy’ in the context of tumour genomic char- with diverse natural course, response to treatment, and prog-
acterisation [5–12]. ctDNA is the fraction of the total cell-free nosis. The histological grade and molecular genetic make-up
DNA that is derived from tumour cells and can be defined by the determine prognosis, with median overall survivals ranging
presence of genomic alterations. ctDNA detected in plasma has from <1 year (e.g. in glioblastoma of the elderly) to long-term
shown promise in characterising tumours and allowing patients survival including cures (e.g. pilocytic astrocytoma and other
and their cancers to be monitored over time. Analyses of muta- rare circumscribed lesions). The clinical hallmark of glioblast-
tions in plasma ctDNA have demonstrated high concordance oma is aggressive growth, local invasiveness, and inexorable re-
with genomic alterations in the tumour [10]. currence [30–32]. In recent years, the development of novel
However, in the context of primary brain tumours and brain sequencing technologies and DNA methylation profiling
metastasis, plasma ctDNA has been shown to be in low abun- coupled to bioinformatics tools has yielded an unparalleled,
dance and present in a limited number of patients [8, 13–16]. comprehensive view of the genome and epigenome of brain
Importantly, the cerebrospinal fluid (CSF) is in intimate contact tumours [33–36].
with brain malignancies and has been recently proved to contain The 2016 update of the WHO classification incorporated well
ctDNA. The CSF space involves the intracerebral ventricles, sub- established molecular parameters into the classification of brain
arachnoid spaces of the spine and brain (cisterns and sulci), and tumours, specifically gliomas. The analysis of the CSF ctDNA of
the central spinal cord canal. The CSF is renewed three to five a cohort of diffuse gliomas indicated that they could be sub-
times daily and is produced by the choroid plexus. The CSF circu- typed by analysing the IDH1 and IDH2, ATRX, TP53, TERT,
lates in a craniocaudal direction from ventricles to spinal sub- H3F3A and HIST1H3B mutational status, facilitating the classi-
arachnoid space from where it is removed via craniocaudal fication of diffuse gliomas and providing prognostic informa-
lymphatic routes and the venous system [17]. The CSF space is tion [28]. Moreover, the presence of mutations in the TERT
separated from the vascular system by the blood–CSF barrier, promoter found in CSF ctDNA correlated with outcome [37].
while the blood–brain barrier is located between the brain paren- In the case of diffuse midline gliomas, the detection of H3F3A
chyma and the vascular system [18]. and HIST1H3B mutations in the CSF could confirm diagnosis
CSF has been explored as a source of ctDNA for precisely char- [28]. This is of major relevance since the anatomical location of
acterising brain cancers. Studies reported before the era of high- this type of tumours increases the risk of obtaining surgical
throughput sequencing showed that some molecular alterations specimens.
or gene mutations can be detected in the DNA present in the CSF CSF ctDNA was detected in a large proportion of patients
of patients with brain tumours [19–24]. Importantly, massively with brain primary tumours (Table 1). However, CSF ctDNA is
parallel sequencing methods have recently been used to analyse not found in all brain tumours. For example, in some low grade
cell-free tumour DNA from CSF to comprehensively characterise gliomas, CSF ctDNA was not detected or was not informative
somatic alterations including gene mutations and copy number [28]. Technological advances may improve sequencing sensitiv-
alterations [15, 25–29] (Table 1). ity in the future, thus reducing the number of non-informative
DNA was isolated from CSF (ranging from 0.75 to 10 ml) usu- cases.
ally obtained from a lumbar puncture and DNA sequencing (i.e.
droplet digital PCR, targeted sequencing, whole-exome sequenc-
ing, or shallow whole-genome sequencing) allowed the identifi-
Therapeutic considerations
cation of ctDNA. Notably, CSF ctDNA enabled the identification ctDNA diagnostic applications with potential therapeutic impli-
of genomic alterations in patients with systemic metastatic bur- cations remain limited for adult patients with primary brain
den including brain metastasis, or disease restricted to the brain tumours. The most relevant biomarker for glioblastoma in terms
(primary tumours and brain metastasis) [15, 25–27]. Higher of choice of therapy remains promoter methylation of the
grade brain tumours were more likely to exhibit detectable CSF MGMT gene [38]. Efforts at the detection of MGMT promoter
ctDNA than lower grade ones [26] and, in some cases, the dis- methylation in CSF of glioma patients showed higher sensitivity
tance of the tumour to CSF spaces could determine the amount than in plasma [39]. Future applications with therapeutic impact
of CSF ctDNA [27]. are likely to include the monitoring of EGFRvIII and amplified
Here, we focus on the studies related to ctDNA obtained from epidermal growth factor receptor (EGFR) in patients undergoing
CSF. Nowadays, the CSF liquid biopsy is increasingly allowing EGFR-targeted therapy [40]. The evaluation of ctDNA during the
molecular diagnoses, providing information on prognosis, facili- follow-up of patients and especially at recurrence can confirm the
tating the identification of new actionable genomic alterations, molecular status and may help to deliver precision therapies.
Patient population Sequencing technologies Method for CSF collection/ Tumour-derived Main findings Comparison Extra-cranial Refs
amount collected DNA found in with plasma disease
Four glioblastoma, 2 medulloblas- Targeted capture massively parallel Lumbar puncture, cerebral shunts, 58–60 CSF ctDNA is enriched in brain 冑 冑 [15]
toma, and 17 brain metastases sequencing, digital droplet PCR autopsy/ tumours and produces better results
(from breast and NSCLC) than plasma ctDNA.
Thirty-five primary brain and spinal
Targeted capture massively parallel Cerebral shunts (during surgical pro- 57–88 All adjacent tumours to the CSF reser- – – [26]
cord tumours sequencing, whole-exome sequenc- cedure)/average of 4.8 ml voir had CSF ctDNA detectable
ing in four cases (range¼0.75–10 ml)
Twelve primary brain tumours and Targeted capture massively parallel Lumbar puncture and one sample 50–63 Drug-resistance mutations in patients – 冑 [27]
41 brain metastases sequencing Ommaya reservoir/5 ml of CSF whose CNS disease progresses during
kinase inhibitor therapy is identified in
CSF ctDNA.
A vestibular schwannoma, a men- Targeted amplicon sequencing and Lumbar puncture, cerebral shunts 85 Tumour mutations were detectable in 冑 – [25]
ingioma, five brain metastases and digital PCR (during surgical procedure)/1–10 ml the CSF ctDNA of patients with differ-
three leptomeningeal metastases of CSF ent types of brain tumours
Twenty primary diffuse glioma Targeted amplicon sequencing and Lumbar puncture before surgery; two 85 A sequencing platform to simultan- – – [28]
tumours digital PCR samples cisterna magna, one sample eously test seven genes IDH1, IDH2,
cerebral shunt/2 ml of CSF TP53, ATRX, TERT, H3F3A, HIST1H3B in
the CSF ctDNA allowing the subclassifi-
cation of diffuse glioma
Thirty-eight TERT mutant glioblast- Sequenced uni-directionally on an Directly after opening the dura (durot- 78–98 CSF ctDNA identifies TERT promoter 冑 [37]
oma (34 primary and 4 recurrent Ion Torrent PGM omy), through dissection of the con- mutations
glioblastoma) NGS system, digital droplet PCR vexity subarachnoid space/2–4 ml of
CSF
Thirteen glioma tumours Untargeted, low-coverage WGS Lumbar puncture/10 ml of CSF 39 Combining analyses of SCNAs with – – [29]
(<0.4) to detect SCNAs DNA fragmentation allows detection
of ctDNA in CSF using sWGS data at
low cost
CNS, central nervous system; GBM, glioblastoma; LM, leptomeningeal metastasis; NSCLC, non-small cell lung cancer; SCNAs, somatic copy number alterations; s-WGS, shallow whole-genome sequencing.
doi:10.1093/annonc/mdy544 | 213
Review
Review Annals of Oncology
Molecular diagnosis
monitoring actionable mutations and therapy resistance using
prognosis CSF ctDNA appears to be an application of CSF-based liquid
biopsies that could be close to clinical practice.
First- (erlotinib and gefitinib) and second-generation (afati-
nib) EGFR tyrosine kinase inhibitors (TKI) have shown activity
against brain metastasis from non-small-cell lung cancers
Brain
GBM metastasis (NSCLC) that harbour EGFR mutations [56–58]. A number of
third-generation EGFR-TKI that also target mutant EGFR
Monitor Actionable T790M, which confers therapeutic resistance, are in various
tumor genomic
CSF alterations
phases of clinical investigation to target brain metastases (osimer-
burden
tinib, rociletinib, ASP-8273, HM-61713). In anaplastic lymph-
oma kinase (ALK) gene-rearranged (ALK)-NSCLC, second-
generation ALK inhibitors with increased potency such as alecti-
ctDNA
nib and ceritinib have apparently superior CNS penetration com-
pared with crizotinib and share significant therapeutic potential
[53–55]. Breast cancer studies have focussed primarily on tar-
geted therapies [e.g. lapatinib, pertuzumab, ado-trastuzumab
Evolving heterogeneity
emtansine (T-DM1)] used for HER2-positive cancers [51, 52, 59,
(tracking diversity and evolution)
60]. In patients with melanoma and brain metastases, substantial
clinical activity has been observed with BRAF and MEK inhibi-
Figure 1. Potential use of CSF ctDNA as a liquid biopsy for primary tors, e.g. dabrafenib plus trametinib [49, 61], resulting in an
brain tumours and brain metastasis. intracranial response rate of nearly 60% [61]. Ongoing clinical
trials exploit the cytotoxic T-lymphocyte-associated antigen 4
Brain metastases and programmed death 1 pathways as target for immune check-
point inhibitor therapy [50, 62]. Actionable genomic alterations
Diagnostic considerations with potential therapeutic implications have been identified in
the CSF ctDNA [15, 25–27], including EGFR, ALK, HER2,
Brain metastases from solid tumours are more frequent than pri- BRAF-targetable kinases, and others associated with DNA integ-
mary brain tumours. They may occur in 20%–40% of advanced rity such as BRCA1 and BRCA2 [63].
stage cancers, particularly in lung cancer, breast cancer and melan- Analysis of CSF ctDNA has also shown gene mutations associ-
oma [41–43]. Recent reports on the branched evolution of cancer ated with therapy resistance [15, 27, 64]. Drug-resistance muta-
at different sites including metastasis to the brain have reinforced tions in patients whose CNS disease progressed during TKI
the need of sequential molecular profiling across the disease trajec- therapy (EGFR, ALK, HER2, or BRAF) were identified in CSF
tory [4, 44]. Brain metastases exhibit different genomic alterations ctDNA in one-third of cases [27]. This included a NRAS G12R
than the primary extra-cranial tumours [4] indicating that the mutation in the CSF of a BRAF V600E-mutant (and NRAS-
brain lesion-specific genomic alterations should be identified to se- negative) melanoma; a PIK3CA H1047R mutation in the CSF of a
lect the optimal therapeutic approach [4, 45]. CSF ctDNA and not HER2-amplified breast cancer patient, potentially associated
plasma ctDNA can be a good surrogate marker in such situations with trastuzumab resistance; and an EGFR T790M mutation in
since ctDNA from brain lesions is enriched in the CSF. the CSF of a patient with EGFR-mutant NSCLC who did not re-
Trunk mutations, present in all cancer cells, as well as private gen- spond to a second-generation EGFR-TKI [27]. ESR1 mutations
omic alterations, present in just a subpopulation of cells or in specif- can confer resistance to aromatase inhibitor therapy in advanced
ic metastatic lesions, can be identified in the CSF [15, 27]. This estrogen receptor-positive breast cancers, but not to fulvestrant
allows opportunities for deconvolving tumour heterogeneity. CSF [65, 66]. A clinical trial is evaluating ESR1 mutations in plasma
and plasma ctDNA were compared in a series of samples that ctDNA to predict the efficacy of a change of the hormone therapy
included multiregional metastatic sites from postmortem speci- (aromatase inhibitor changed to fulvestrant) (ClinicalTrials.gov
mens of patients with disseminated breast cancers including brain Identifier: NCT03079011). Translation of this type of clinical trial
metastases [15]. For example, mutations found in the CSF ctDNA design to the setting of patients with brain metastasis is envi-
allowed to discern the origin of leptomeningeal and brain metasta- sioned using liquid biopsies. In the context of multiple metastases
sis implants separately in a patient with Li Fraumeni syndrome and and discordant clinico-radiological findings, analysing a single-
two concurrent tumours, a metastatic breast cancer and esthesio- lesion biopsy is inadequate in guiding the selection of targeted
neuroblastoma [15]. CSF ctDNA analysis captured trunk mutations therapy [67]. Parallel analyses of serial CSF and plasma ctDNA
and, importantly, private mutations to the brain and to the menin- samples may be warranted.
geal deposits. These observations highlight the potential applica- Although the most common initial clinical presentation of
tions of CSF ctDNA to complement diagnosis of brain metastasis. metastatic HER2-positive breast cancer is with extra-cranial
metastases, CNS progression occurs in a substantial proportion
of patients during the course of the disease [68]. It has also been
Therapeutic considerations shown that extensive extra-CNS disease control, with HER2 tar-
Several targeted therapeutic agents have demonstrated clinical ac- geting, might drive high incidence of CNS progression [69, 70].
tivity against established brain metastases [46–55] and This situation remains a major challenge where genomic analysis