Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2015 Jul 20;112(31):9704–9709. doi: 10.1073/pnas.1511694112

Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord

Yuxuan Wang a,1, Simeon Springer a,1, Ming Zhang a,1, K Wyatt McMahon a, Isaac Kinde a, Lisa Dobbyn a, Janine Ptak a, Henry Brem b, Kaisorn Chaichana b, Gary L Gallia b, Ziya L Gokaslan b, Mari L Groves b, George I Jallo b, Michael Lim b, Alessandro Olivi b, Alfredo Quinones-Hinojosa b, Daniele Rigamonti b, Greg J Riggins b, Daniel M Sciubba b, Jon D Weingart b, Jean-Paul Wolinsky b, Xiaobu Ye b, Sueli Mieko Oba-Shinjo c, Suely K N Marie c, Matthias Holdhoff d, Nishant Agrawal a,e, Luis A Diaz Jr a, Nickolas Papadopoulos a, Kenneth W Kinzler a, Bert Vogelstein a,2, Chetan Bettegowda a,b,2
PMCID: PMC4534284  PMID: 26195750

Significance

Outcomes for individuals with central nervous system (CNS) malignancies remain abysmal. A major challenge in managing these patients is the lack of reliable biomarkers to monitor tumor dynamics. Consequently, many patients undergo invasive surgical procedures to determine disease status or experience treatment delays when radiographic testing fails to show disease progression. We show here that primary CNS malignancies shed detectable levels of tumor DNA into the surrounding cerebrospinal fluid (CSF), which could serve as a sensitive and exquisitely specific marker for quantifying tumor burden without invasive biopsies. Therefore, assessment of such tumor-derived DNA in the CSF has the potential to improve the management of patients with primary CNS tumors.

Keywords: CSF-tDNA, CNS tumors, biomarker

Abstract

Cell-free DNA shed by cancer cells has been shown to be a rich source of putative tumor-specific biomarkers. Because cell-free DNA from brain and spinal cord tumors cannot usually be detected in the blood, we studied whether the cerebrospinal fluid (CSF) that bathes the CNS is enriched for tumor DNA, here termed CSF-tDNA. We analyzed 35 primary CNS malignancies and found at least one mutation in each tumor using targeted or genome-wide sequencing. Using these patient-specific mutations as biomarkers, we identified detectable levels of CSF-tDNA in 74% [95% confidence interval (95% CI) = 57–88%] of cases. All medulloblastomas, ependymomas, and high-grade gliomas that abutted a CSF space were detectable (100% of 21 cases; 95% CI = 88–100%), whereas no CSF-tDNA was detected in patients whose tumors were not directly adjacent to a CSF reservoir (P < 0.0001, Fisher’s exact test). These results suggest that CSF-tDNA could be useful for the management of patients with primary tumors of the brain or spinal cord.


Approximately 25,000 individuals each year are diagnosed with a malignant brain or spinal cord tumor in the United States, and more than one-half of these patients will die from their disease (1). Although there are a number of different subtypes of primary CNS cancers, nearly all are treated with maximal safe surgical resection followed by radiation and in some cases, chemotherapy. Given the lack of clinically available biomarkers for CNS malignancies, the conventional method for disease monitoring in these patients is radiographic using either computed tomography or MRI (2). Unfortunately, anatomic changes detected by these imaging modalities are often nonspecific and slow to change, even in the face of progressing or regressing disease. Moreover, it can be difficult to discriminate between treatment effect and cancer growth with imaging alone (3). Patients must, therefore, have additional surgeries for definitive tissue diagnosis or inappropriately wait for radiographic findings to change as their disease progresses. As a result, there is a great need for more sensitive and specific tumor biomarkers in neurooncology.

The recent success of detecting circulating tumor cells in the peripheral blood of glioblastoma patients represents an important step toward this goal, with reported sensitivities between 21% and 39% (46). Circulating tumor DNA (ctDNA) is found in the plasma of patients with most forms of malignancies (711). However, brain tumors, including high-grade gliomas and medulloblastomas, are an exception, with only a minority giving rise to detectable levels of ctDNA, perhaps because of the blood–brain barrier (8).

Other studies have shown that tumor-derived DNA can be found in anatomically relevant fluids, such as urine in bladder cancer patients, sputum in lung cancer patients, stool in patients with colorectal carcinomas, and endocervical fluid in patients with gynecological malignancies (1217). Based on this concept, we wondered whether primary brain and spinal cord tumors might shed appreciable levels of tDNA into the cerebrospinal fluid (CSF) that bathes the CNS (Fig. 1). We coined the term “CSF-tDNA” to describe tumor DNA shed into the CSF. The experiments below were designed to test this hypothesis in an exploratory study of tumors of diverse histology and locations within the CNS.

Fig. 1.

Fig. 1.

Schematic showing the shedding of CSF-tDNA from CNS malignancies. Tumor cells from primary brain and spinal cord tumors shed DNA into the CSF that bathes the CNS. DNA purified from the CSF is analyzed for tumor-specific mutations.

Results

Patient and Tumor Characteristics.

Thirty-five patients with CNS cancers were enrolled in this study. Their ages, sexes, races, and preoperative symptoms are listed in Table S1. In total, 6 patients had medulloblastomas, and 29 patients had gliomas; 7, 9, 2, and 17 of the tumors were classified as World Health Organization (WHO) grades I–IV, respectively. Twenty-nine (83%) of 35 patients provided CSF during the initial surgery, whereas the remaining 6 (17%) did so during a repeat resection. The tumors were distributed throughout the brain and spinal cord, with 14 arising in the posterior fossa (including six medulloblastomas), 8 arising in the supratentorial compartment of the brain, and 13 arising in the spinal cord (Table 1).

Table S1.

Patient demographics

Patient Sex Recurrence Age (y) Race Preoperative symptoms Duration of symptoms (mo) Hydrocephalus Enhancing on CT or MRI Tumor size (cm3)
CGLI 02 Female No 33 W Dysesthesias, urinary retention 33 No No 2 × 1 × 1
CGLI 03 Male No 13 W Incidental Not applicable No Yes 3 × 3.2 × 3.8
CGLI 06 Female No 13 AA Headache, vision loss 2 Yes Yes 4.9 × 5 × 5.6
CGLI 11 Female No 50 W Dysesthesias, numbness 36 No Yes 5.3 × 1 × 1.2
CGLI 12 Male No 5 W Headache, vomiting 1 Yes Yes 4.1 × 4.2 × 3.8
CGLI 13 Male No 27 W Back pain, sciatica 15 No NA NA
CGLI 14 Female No 62 W Back pain 5 No Yes 4.5 × 0.8 × 1.2
CGLI 15 Male No 52 W Neck pain 1 No Yes 2.1 × 0.9 × 1.2
CGLI 20 Female No 14 W Headache, vomiting 3 Yes Yes 3.4 × 3.2 × 2.6
CGLI 22 Female No 18 A Headache, vomiting 3 No Yes 5 × 4.4 × 4.3
CGLI 25 Female No 72 W Back pain, arm pain 3 No Yes 4.3 × 1.2 × 0.7
CGLI 26 Female No 46 W Left leg pain 13 No No 5 × 1.4 × 1.2
CGLI 28 Female No 79 A Left arm weakness 3 No Yes 4.4 × 5.7 × 5.8
CGLI 29 Male Yes 18 W Neck pain 28 No Yes 3.1 × 1.1
CGLI 31 Male No 74 AA Seizure 0.5 No Yes 4.5 × 5.2 × 4.3
CGLI 35 Male No 70 AA Memory loss 2 No Yes 6.1 × 3 × 3.5
CGLI 36 Male No 11 W Back pain 2 No Yes 6.5 × 1.7 × 2.2
CGLI 39 Male No 56 W Paresthesias 3 No Yes 1.7 × 1.1 × 1.6
CGLI 40 Male No 7 W Headache, ataxia 5 No Yes 4.0 × 4.0 × 4.8
CGLI 41 Male Yes 34 W Gait imbalance, incoordination 0.5 No Yes 4.0 × 3.0 × 2.9
CGLI 42 Female No 31 W Back pain 24 No NA NA
CGLI 43 Male Yes 66 W Dysesthesias 12 No Yes 3.8 × 1.4 × 1.4
CGLI 44 Female No 6 A Headache, vomiting 2 Yes Yes 3.4 × 3.8 × 3.4
CGLI 47 Female Yes 71 W Headache 2 No Yes 6.0 × 3.7 × 3.3
CGLI 48 Male No 56 W Blurry vision, mental status change 2 No Yes 6.1 × 7.7 × 4.9
CGLI 50 Female No 69 W Vision 1 No Yes 5.1 × 5.0 × 3.8
CGLI 51 Female No 84 W Asymptomatic Not applicable No Yes 4.0 × 3.3 × 2.9
CGLI 55 Male No 7 ME Headache, difficulty walking 1 No Yes 3.1 × 3.8 × 3.4
CGLI 56 Male No 3 W Headache, difficulty walking 1 Yes Yes 4.1 × 4.2 × 4.0
CGLI 58 Male No 32 ME Numbness, myelopathy 24 No No 5.1 × 1.9 × 1.2
CGLI 60 Female No 9 W Headache 3 No Yes 2.4 × 2.2 × 2.0
CGLI 61 Female No 12 W Headache, nausea, vomiting 1 No Yes 5.5 × 5.3 × 5.2
CGLI 63 Male No 22 ME Headache, vomiting, ataxia 1 No Yes 3.0 × 2.2 × 2.0
CGLI 101 Male Yes 49 W Headache, nausea, vomiting NA Yes Yes 2.6 × 2.0 × 1.6
CGLI 254 Male Yes 7 W Headache, ataxia, vomiting NA Yes Yes 1.7 × 1.2 × 1.2

A, Asian; AA, African American; CT, computed tomography; ME, Middle Eastern; NA, not available; W, white.

Table 1.

Tumor characteristics and the detection of CSF-tDNA

Patient Diagnosis Tumor grade Tumor location Location of CSF sampling Tumor abutting CSF space CSF-tDNA
CGLI 02 Glioma WHO II, diffuse astrocytoma T11 spinal cord Spinal subarachnoid space Yes Not detected
CGLI 03 Anaplastic astrocytoma WHO III, anaplastic astrocytoma Pons Basal cistern Yes Positive
CGLI 06 Pilocytic astrocytoma WHO I, pilocytic astrocytoma Cerebellar vermis Basal cistern Yes Positive
CGLI 11 Spinal ependymoma WHO II, ependymoma C7-T3 spinal cord Spinal subarachnoid space Yes Positive
CGLI 12 Intracranial ependymoma WHO II, ependymoma Fourth ventricle Basal cistern Yes Positive
CGLI 13 Myxopapillary ependymoma WHO I, myxopapillary ependymoma L2-3 spinal cord Spinal subarachnoid space NA Positive
CGLI 14 Intramedullary spinal cord lesion Low-grade neoplasm T7-9 spinal cord Spinal subarachnoid space Yes Positive
CGLI 15 Spinal ependymoma WHO II, ependymoma C3-4 spinal cord Spinal subarachnoid space No Not detected
CGLI 20 Medulloblastoma WHO IV, medulloblastoma Fourth ventricle Basal cistern Yes Positive
CGLI 22 Pilocytic astrocytoma WHO I, pilocytic astrocytoma Cerebellar hemisphere Basal cistern No Not detected
CGLI 25 Myxopapillary ependymoma WHO I, myxopapillary ependymoma L2-3 spinal cord Spinal subarachnoid space Yes Positive
CGLI 26 Intramedullary spinal cord tumor WHO II, infiltrating astrocytoma with oligodendroglial features C3-6 spinal cord Spinal subarachnoid space Yes Positive
CGLI 28 Anaplastic astrocytoma WHO III, anaplastic astrocytoma Right frontal/butterfly Ventricle Yes Positive
CGLI 29 Glioblastoma WHO IV, glioblastoma C4-6 spinal cord Spinal subarachnoid space Yes Positive
CGLI 31 Glioblastoma WHO IV, glioblastoma Right frontal/butterfly Ventricle Yes Positive
CGLI 35 Glioblastoma WHO IV, glioblastoma Right temporal Ventricle Yes Positive
CGLI 36 Spinal cord glioblastoma WHO IV, glioblastoma T10-L1 spinal cord Spinal subarachnoid space Yes Positive
CGLI 39 Intramedullary spinal cord tumor WHO II, low-grade glioma likely ependymoma C2-3 spinal cord Spinal subarachnoid space No Not detected
CGLI 40 Medulloblastoma WHO IV, medulloblastoma Fourth ventricle Basal cistern Yes Positive
CGLI 41 Glioblastoma WHO IV, glioblastoma Cerebellar hemisphere Basal cistern Yes Positive
CGLI 42 Spinal ependymoma WHO II, ependymoma T1-7 spinal cord Spinal subarachnoid space NA Positive
CGLI 43 Low-grade glioma WHO II, low-grade glioma T10 spinal cord Spinal subarachnoid space Yes Not detected
CGLI 44 Pilocytic astrocytoma WHO I, pilocytic astrocytoma Cerebellar vermis Basal cistern No Not detected
CGLI 47 Glioblastoma WHO IV, glioblastoma Right temporal Ventricle Yes Positive
CGLI 48 Glioblastoma WHO IV, glioblastoma Left temporal Ventricle Yes Positive
CGLI 50 Glioblastoma WHO IV, glioblastoma Right temporal Ventricle Yes Positive
CGLI 51 Glioblastoma WHO IV, glioblastoma Right frontal Ventricle Yes Positive
CGLI 55 Brainstem glioblastoma WHO IV, glioblastoma Midbrain Basal cistern Yes Positive
CGLI 56 Medulloblastoma WHO IV, medulloblastoma Fourth ventricle Basal cistern Yes Positive
CGLI 58 Diffuse astrocytoma WHO II, diffuse astrocytoma T2-4 spinal cord Spinal subarachnoid space Yes Not detected
CGLI 60 Medulloblastoma WHO IV, medulloblastoma Fourth ventricle Basal cistern Yes Positive
CGLI 61 Pilocytic astrocytoma WHO I, pilocytic astrocytoma Cerebellar vermis Basal cistern Yes Not detected
CGLI 63 Medulloblastoma WHO IV, medulloblastoma Cerebellum Basal cistern No Not detected
CGLI 101 Glioblastoma WHO IV, glioblastoma Cerebellar vermis Basal cistern Yes Positive
CGLI 254 Medulloblastoma WHO IV, medulloblastoma Fourth ventricle Basal cistern Yes Positive

NA, not available.

Identification of Somatic Mutations.

At least one mutation was identified in each of 35 tumors analyzed using a tiered approach [targeted sequencing followed by whole-exome sequencing (WES)] described in Materials and Methods.

With the targeted sequencing approach, we identified mutations in 13 tumors. The mutations in these samples occurred in TP53 (tumor protein p53; n = 5), IDH1 (isocitrate dehydrogenase 1; n = 2), or the TERT promoter (telomerase reverse transcriptase; n = 6) (Table S2). In the remaining 22 tumors, WES was used to identify at least one mutation per sample (Dataset S1). Genes mutated in these samples included well-known drivers, such as NF2, PIK3R1, PTCH1, and PTEN (18). The fractions of mutant alleles in tumors were generally high, averaging 46% (with an SD of 18%). This finding is consistent with the expected early development of driver gene mutations during tumor evolution and the presence of nonneoplastic cells in all tumors, even macrodissected ones, such as the samples used here. All mutations identified were confirmed to be absent in DNA from matched noncancerous (normal) cells from each patient.

Table S2.

Mutations detected in the CSF or tumor of each patient

Patient Volume of CSF analyzed (mL) DNA in CSF (ng) Sequencing method Mutation Location Gene Transcript Protein cDNA Mutant allele fraction in tumor (%) Mutant allele fraction in CSF (%)
CGLI 02 4 245 WES NM_144631.5 (ZNF513):c.413C > T Exon ZNF513 NM_144631.5 p.P138L c.413C > T 49 Not detected
CGLI 03 7 314 WES NM_181523.2 (PIK3R1):c.1690A > G Exon PIK3R1 NM_181523.2 p.N564D c.1690A > G 39 0.1
CGLI 06 8 148 WES NR_024604.1 (CLCA3P):c.446G > A Exon CLCA3P NR_024604.1 p.R149Q c.446G > A 36 0.2
CGLI 11 5 290 WES NM_133450.3 (ANKS3):c.1738G > A Exon ANKS3 NM_133450.3 p.E580K c.1738G > A 47 0.3
CGLI 12 5 71 WES NM_003531.2 (HIST1H3C):c.83T > A Exon HIST1H3C NM_003531.2 p.K28M c.83T > A 44 0.2
CGLI 13 1.5 6 WES NM_144965.1 (TTC16):c.1016T > A Exon TTC16 NM_144965.1 p.V339E c.1016T > A 48 1.0
CGLI 14 7.5 187 Panel NM_000546.5 (TP53):c.638G > A Exon TP53 NM_000546.5 p.R213Q c.638G > A 59 19.6
CGLI 15 5 482 WES NM_004990.3 (MARS):c.1487C > T Exon MARS NM_004990.3 p.T496I c.1487C > T 13 Not detected
CGLI 20 3 127 WES NM_198229.2 (RGS12):c.2980C > T Exon RGS12 NM_198229.2 p.R994W c.2980C > T 52 52.6
CGLI 22 5 8 WES NM_001080522.2 (CC2D2A):c.1938G > A Exon CC2D2A NM_001080522.2 p.W646X c.1938G > A 54 Not detected
CGLI 25 10 253 WES NM_001795.3 (CDH5):c.1211G > A Exon CDH5 NM_001795.3 p.S404N c.1211G > A 50 0.3
CGLI 26 7 20 Panel NM_005896.2 (IDH1):c.394C > T Exon IDH1 NM_005896.2 p.R132C c.394C > T 43 0.3
CGLI 28 1 2,361 Panel NM_005896.2 (IDH1):c.395G > A Exon IDH1 NM_005896.2 p.R132H c.395G > A 33 33.0
CGLI 29 3.5 206 Panel NM_000546.5 (TP53):c.742C > T Exon TP53 NM_000546.5 p.R248W c.742C > T 69 0.2
CGLI 31 4 1,416 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 37 8.2
CGLI 35 2.5 432 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 46 1.4
CGLI 36 5.5 448 Panel NM_000546.5 (TP53):c.742C > T Exon TP53 NM_000546.5 p.R248W c.742C > T 65 14.3
CGLI 39 8.5 154 WES NM_000268.3 (NF2):c.592C > T Exon NF2 NM_000268.3 p.R198X c.592C > T 80 Not detected
CGLI 40 1.5 1,169 WES NM_000388.3 (CASR):c.2549C > T Exon CASR NM_000388.3 p.A850V c.2549C > T 45 2.8
CGLI 41 4.25 87 Panel NM_000546.5 (TP53):c.396G > C Exon TP53 NM_000546.5 p.K132N c.396G > C 43 1.4
CGLI 42 0.75 20 WES NM_001848.2 (COL6A1):c.1417G > A Exon COL6A1 NM_001848.2 p.G473R c.1417G > A 39 0.5
CGLI 43 4.5 100 WES NM_000268.3 (NF2):c.1009C > T Exon NF2 NM_000268.3 p.Q337* c.1009C > T 61 Not detected
CGLI 44 1.5 64 WES NM_001004439.1 (ITGA11):c.2140C > T Exon ITGA11 NM_001004439.1 p.R714* c.2140C > T 16 Not detected
CGLI 47 6.8 29 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 44 22.1
CGLI 48 5 1,400 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 52 1.0
CGLI 50 5.6 820 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 47 18.8
CGLI 51 0.75 64 Panel NM_198253.2 (TERT):c.1–124C > T Promoter TERT NM_198253.2 NA c.1–124C > T 10 5.9
CGLI 55 9 146 WES NM_000314.4 (PTEN):c.388C > T Exon PTEN NM_000314.4 p.R130* c.388C > T 66 33.2
CGLI 56 6.5 1,155 WES NM_014691.2 (AQR):c.1746delT Exon AQR NM_014691.2 p.P582fs c.1746delT 39 1.8
CGLI 58 9 90 WES NM_052853.3 (ADCK2):c.1057T > A Exon ADCK2 NM_052853.3 p.F353I c.1057T > A 23 Not detected
CGLI 60 8 1,335 WES NM_004687.4 (MTMR4):c.2110C > G Exon MTMR4 NM_004687.4 p.L704V c.2110C > G 40 18.7
CGLI 61 3 99 WES NM_001122679.1 (TENM2):c.799G > A Exon TENM2 NM_001122679.1 p.A267T c.799G > A 21 Not detected
CGLI 63 4 411 WES NM_000264.3 (PTCH1):c.746+1C > A Splice site PTCH1 NM_000264.3 NA c.746+1C > A 71 Not detected
CGLI 101 3.5 14 Panel NM_000546.5 (TP53):c.731G > A Exon TP53 NM_000546.5 p.G244D c.731G > A 36 0.9
CGLI 254 2.5 168 WES NM_021140.2 (KDM6A):c.4153C > T Exon KDM6A NM_021140.2 p.Q1385X c.4153C > T 91 77.3

NA, not available; panel, directed sequencing of codons 130–139 of IDH1; codons 126–155, 144–178, and 250–262 of IDH2; all coding exons of TP53; and the TERT promoter (Materials and Methods).

The presence of one of the mutations detected in each patient’s tumor was then assessed in the CSF of the same patient using a sensitive sequencing-based method. This method reliably detects mutations with allele fractions as low as 0.01% (8, 19). An average of 4.8 mL CSF (SD of 2.6) was collected from 35 patients (Table S2). DNA could be purified from all CSF samples, although the amounts varied considerably (average of 417 ng; SD of 553 ng) (Table S2). Primers were designed to amplify each of 35 mutations as previously described (8, 19). Using this technology, we found that 74% of 35 CSF samples contained detectable levels of tumor DNA. The detectability of tumor DNA present in the CSF was not correlated with demographic characteristics, symptom duration, presence of hydrocephalus, contrast enhancement on imaging, or mutation type (Table S3). The fraction of mutant alleles in the CSF was, as expected, usually lower than the fraction in the primary tumors, and it was also much more variable than in the primary tumors. The average detectable mutant allele fraction in CSF was 12.2% (range = 0.1–77%).

Table S3.

Associations between clinical characteristics and detection of CSF-tDNA

Characteristic assessed All patients (n = 35) CSF undetected (n = 9) CSF detected (n = 26) P value
Age (y) median (range) 32 (3–84) 32 (6–66) 32.5 (3–84) 0.65
Sex, no. (%) 1
 Female 16 (46) 4 (44) 12 (46)
 Male 19 (54) 5 (55) 14 (54)
Recurrent, no. (%) 1
 No 29 (83) 8 (89) 21 (81)
 Yes 6 (17) 1 (11) 5 (19)
Race, no. (%) 0.08
 A 3 (9) 2 (22) 1 (4)
 AA 3 (9) 0 (0) 3 (12)
 ME 3 (9) 2 (22) 1 (4)
 W 26 (74) 5 (56) 21 (81)
Hydrocephalus, no. (%) 0.65
 No 28 (80) 8 (89) 20 (77)
 Yes 7 (20) 1 (11) 6 (23)
Tumor grade, no. (%) 0.01
 1 6 (17) 3 (33) 3 (12)
 2 9 (26) 5 (56) 4 (15)
 3 2 (6) 0 (0) 2 (8)
 4 17 (49) 1 (11) 16 (62)
 NA 1 (3) 0 (0) 1 (4)
Tumor grade, no. (%) 0.004
 1 and 2 15 (43) 8 (89) 7 (27)
 3 and 4 19 (54) 1 (11) 18 (69)
 NA 1 (3) 0 (0) 1 (4)
Enhancing CT or MRI, no. (%) 0.17
 No 3 (9) 2 (22) 1 (4)
 Yes 30 (86) 7 (78) 23 (88)
 NA 2 (6) 0 (0) 2 (8)
Tumor location, no. (%) 0.16
 Infratentorial 14 (40) 4 (44) 10 (38)
 Spinal 13 (37) 5 (56) 8 (31)
 Supratentorial 8 (23) 0 (0) 8 (31)
Abutting CSF space, no. (%) <0.001
 No 5 (14) 5 (56) 0 (0)
 Yes 28 (80) 4 (44) 24 (92)
 NA 2 (6) 0 (0) 2 (8)
Sequencing method, no. (%) 0.01
 Panel 13 (37) 0 (0) 13 (50)
 WES 22 (63) 9 (100) 13 (50)
Location, no. (%) 0.1
 Exon 28 (80) 8 (89) 20 (77)
 Promoter 6 (17) 0 (0) 6 (23)
 Splice site 1 (3) 1 (11) 0 (0)
Location, no. (%) 0.3
 Promoter 6 (17) 0 (0) 6 (23)
 Exon and splice site 29 (83) 9 (100) 20 (77)
Symptom duration
 Median (range) 3 (0.5–36) 3 (1–33) 2.5 (0.5–36) 0.65
Tumor size
 Median (range) 34.8 (2–230) 11.6 (2–152) 37.4 (2.45–230) 0.41
CSF volume (mL)
 Median (range) 5 (0.75–10) 4.5 (1.5–9) 5 (0.75–10) 0.88
Quantity of DNA in CSF (ng)
 Median (range) 168 (6–2,361) 100 (8–482) 196 (6–2,361) 0.03
Mutant allele fraction in tumor
 Median (range) 0.45 (0.1–0.91) 0.49 (0.13–0.80) 0.44 (0.10–0.91) 0.68

A, Asian; AA, African American; CT, computed tomography; ME, Middle Eastern; NA, not available; panel, directed sequencing of codons 130–139 of IDH1; codons 126–155, 144–178, and 250–262 of IDH2; all coding exons of TP53; and the TERT promoter (Materials and Methods); W, white.

Relationship Between Mutations and Clinical Features.

The great variation in mutant allele fraction among the CSF samples suggested that there might be some anatomical or biological factor underlying the differences. The tumors were distributed among the brain and spinal cord (Table 1), and malignancies arising in both organs were detected at similar frequencies (P = 0.16; t test). High-grade (WHO grades III and IV) tumors were more likely to have detectable CSF-tDNA than low-grade lesions (P = 0.004) (Table S3), which was evidenced by the fact that all but one high-grade tumor (18 of 19) was detected. The levels of CSF-tDNA were also higher in high-grade lesions than in low-grade lesions (mutant allele fractions of 16.3 ± 21.2% vs. 2.8 ± 6.8%). Eighteen of 19 (∼95%) high-grade (WHO grade III or IV) tumors had detectable levels of CSF-tDNA. However, tumor size was not a statistically significant factor in predicting CSF-tDNA detectability or level (P = 0.41) (Table S3).

Another important factor associated with CSF-tDNA levels was anatomic location. MRI scans were examined for the presence of contrast enhancement adjacent to a large CSF space (Table 1). Representative examples are provided in Fig. S1. Patients with lesions adjacent to a CSF reservoir in the brain or spinal cord were much more likely to have detectable levels of CSF-tDNA than those with the remaining lesions. Such reservoirs included the cortical surfaces and ventricles as well as the basal and other cisterns. Accordingly, 86% of 28 cases in which tumors were adjacent to a CSF reservoir had detectable levels of CSF-tDNA. These cases included all 13 high-grade gliomas, all 3 ependymomas, and all 5 medulloblastomas that were in contact with the CSF. The four tumors in CSF contact that were not detectable were all low-grade gliomas. Moreover, zero of five patients whose tumors were entirely encapsulated by the brain or spinal cord parenchyma had detectable levels of CSF-tDNA (P < 0.001) (Table S2). On multivariate logistic regression, only the location of tumors with respect to CSF and the tumor grade were statistically significant (Table S4).

Fig. S1.

Fig. S1.

Representative magnetic resonance images. (A) An example of a tumor (red arrow) abutting a CSF space is shown. (B) An example of tumor (red arrow) not in contact with a CSF space is shown. Corresponding T2 images are provided for easier visualization of CSF.

Table S4.

Univariate and multivariate analyses to identify clinical characteristics associated with the detection of CSF-tDNA

Characteristic assessed in univariate or multivariate analysis Odds ratio (95% confidence interval) P value
Univariate logistic regression
 Sex, female vs. male 1.07 (0.23–4.92) 0.93
 Recurrent, yes vs. no 1.91 (0.19–18.93) 0.58
 Race
  A vs. W 0.15 (0.01–1.92) 0.15
  AA vs. W 1.79 (0.05–62.48) 0.75
  ME vs. W 0.15 (0.01–1.92) 0.15
  AA vs. A 11.67 (0.19–736) 0.25
  ME vs. A 1.00 (0.04–27.27) 1
  ME vs. AA 0.09 (0.001–5.40) 0.25
 Hydrocephalus, yes vs. no 2.40 (0.25–23.24) 0.45
 Tumor grade, III and IV vs I and II 20.57 (2.16–196) 0.01
 Enhancing imaging, yes vs. no 6.57 (0.52–83.75) 0.15
 Tumor location
  Spinal vs. infratentorial 0.66 (0.13–3.27) 0.61
  Supratentorial vs. infratentorial 7.29 (0.29–185) 0.23
  Supratentorial vs. spinal 11.0 (0.44–276) 0.14
 Abutting CSF space, yes vs. no 59.9 (2.14–1,669) 0.02
 Sequencing method, panel vs. WES 19.0 (0.90–401) 0.06
 Location, promoter vs. exon and splice site 6.03 (0.25–148) 0.27
 Age 1.01 (0.98–1.04) 0.64
 Symptom duration 0.98 (0.91–1.06) 0.63
 Tumor size 1.01 (0.99–1.02) 0.41
 CSF volume 0.98 (0.72–1.32) 0.88
 Quantity of DNA in CSF 1.00 (1.00–1.01) 0.2
 Mutant allele fraction in tumor 3.55 (0.04–298) 0.58
Multivariate logistic regression
 Tumor grade, III and IV vs I and II 26.51 (1.45–485) 0.03
 CSF space, yes vs. no 90.59 (1.07–7,670) 0.05

A, Asian; AA, African American; ME, Middle Eastern; panel, directed sequencing of codons 130–139 of IDH1; codons 126–155, 144–178, and 250–262 of IDH2; all coding exons of TP53; and the TERT promoter (Materials and Methods); W, white.

Genome-Wide Sequencing of DNA from the CSF.

The results described above were found after identifying at least one mutation in the primary tumor of each patient. In four patients with either brainstem or intramedullary spinal cord tumors, we also tested whether CSF-tDNA could be detected directly in their CSF by WES without prior knowledge of the tumor genotype. These four samples were selected based on the critical and highly sensitive location of the malignancies, making surgery treacherous. We found that two of four cases analyzed had levels of CSF-tDNA that were comparable with the levels identified through single-amplicon sequencing (Safe-SeqS) when the same mutation was assessed (Table 2). Both detectable cases had greater than 10% mutant allele fractions in the CSF as measured by single-amplicon sequencing. In contrast, the two cases in which WES was unable to identify CSF-tDNA had mutant allele fractions <1% as assessed by single-amplicon sequencing. As controls, we also performed WES on matched normal tissues and tumor tissues. The mutations were found in the tumors at a high frequency, but they were absent in normal tissues.

Table 2.

Detection of CSF-tDNA using WES

Patient and sample type Mutation Genomic coordinate Distinct coverage (SafeSeqS) Mutant (%; SafeSeqS) Distinct coverage (WES) Mutant (%; WES)
CGLI 03
 CSF PIK3R1 p.N564D, c.A1690G Chr5:67591097 57,921 0.1 76 0.0
 Normal PIK3R1 p.N564D, c.A1690G Chr5:67591097 284 0.0 61 0.0
 Primary tumor PIK3R1 p.N564D, c.A1690G Chr5:67591097 377 38.7 147 12.9
CGLI 29
 CSF TP53 p.R248W, c.C742T Chr17:7577539 13,964 0.2 64 0.0
 Normal TP53 p.R248W, c.C742T Chr17:7577539 NA 0.0 55 0.0
 Primary tumor TP53 p.R248W, c.C742T Chr17:7577539 6,418 69.0 58 70.7
CGLI 36
 CSF TP53 p.R248W, c.C742T Chr17:7577539 376,434 14.3 74 9.5
 Normal TP53 p.R248W, c.C742T Chr17:7577539 57,818 0.0 75 0.0
 Primary tumor TP53 p.R248W, c.C742T Chr17:7577539 44,981 65.0 25 72.0
CGLI 55
 CSF PTEN p.R130*, c.C388T Chr10:89692904 251,609 33.2 63 42.9
 Primary tumor PTEN p.R130*, c.C388T Chr10:89692904 91,515 65.9 56 66.1

Genomic coordinates refer to the human reference genome hg19 release (Genome Reference Consortium GRCh37, February of 2009). NA, not available.

Discussion

Minimally invasive techniques to monitor disease burden have been a challenge for many diseases of the CNS, including cancer. This challenge is highlighted by the high risks associated with neurosurgical procedures and the widely recognized limitations of current imaging modalities. In cancer patients, there is no reliable way of parsing out treatment effects from tumor recurrence, causing many patients to undergo unnecessary repeat surgeries. For example, in ∼30% of patients with glioblastoma who undergo a repeat resection for presumptive recurrence, pathologic examination of the resected specimen reveals necrosis, scarring, or other treatment-related effects rather than recurrent disease (20). Conversely, while patients are waiting for or recovering from surgery for suspicious lesions, chemotherapy or radiation therapy cannot be administered, providing time for unabated tumor growth. Finally, patients are often kept on ineffective medication regimens until definitive signs of tumor progression appear on imaging. This delay in detection precludes potential opportunities to undergo new targeted therapies that might be effective for their disease (21). The health costs of these missed opportunities will increase with the expected advances in therapeutic modalities.

Given the need for sensitive and specific markers to monitor tumor dynamics, we asked whether tumor-derived DNA could be found in the CSF of patients with primary CNS tumors. This study was stimulated by our inability to consistently detect ctDNA in the plasma of these patients (8) and inspired by previous demonstrations that tumor-derived DNA could be found in fluids located in the proximity of neoplastic lesions. For example, a recent pilot study by Pan et al. (22) suggests that tumor-derived DNA can be detected in the spinal fluid of individuals whose primary tumors have metastasized to the brain. Although lumbar puncture to obtain CSF is not a noninvasive procedure, it qualifies as minimally invasive and is currently routinely performed to follow some brain tumor patients, particularly those with medulloblastomas (23, 24). Unfortunately, the examination of these CSF samples by cytology is usually of limited use, with relatively low sensitivities achieved even using large volumes of CSF (25, 26). Only 1 of 35 patients evaluated in our study had concomitant cytologic studies of CSF, precluding direct comparison. The results of this study suggest that the rates of tumor-derived DNA found in the CSF (74%) closely approximate the levels found in body fluids adjacent to other tumor types. For example, urine in bladder cancer was found to have tumor-derived DNA in 70% of cases, whereas sputum in lung cancer was positive in 79% of cases (27, 28). Although the rate of detection observed in this study was not 100%, its sensitivity was comparable with or superior to other noninvasive tests for malignancies in general. Moreover and as noted below, it was particularly sensitive for tumors that abutted a CSF reservoir or cortical surface. Finally, from a technological standpoint, the average fraction of mutant DNA (12.2%) far exceeded the limit of detection of the sequencing assay used (0.01%). This assay could be performed with any commercially available next generation sequencing instrument at relatively small cost.

Our study revealed a significant association between the location and type of the tumor and the presence of CSF-tDNA. In particular, we were able to detect all 13 WHO grade III or IV gliomas (also known as anaplastic astrocytoma and glioblastoma, respectively), all 5 medulloblastomas, and all 3 ependymomas that abutted a CSF reservoir or cortical surface. It is in these aggressive tumors where the need for a robust biomarker is most desperate. There are also emerging data that some brain tumors, particularly those with genotypes susceptible to targeted therapies, may be able to be treated primarily with medical therapies, thereby obviating the need for surgery if appropriate noninvasive diagnostic tools were available (2932). It is also worth noting that surgical resection nearly always creates an opening extending from the surface to the deep-seated tumor. This passageway typically persists and may enable tDNA from any residual or recurrent tumor to enter the CSF. Even without such surgically induced openings, the vast majority of medulloblastomas and ependymomas arise within or communicate with a ventricular reservoir, making them well-suited for CSF monitoring (2426, 33, 34). Future studies will be required to directly compare CSF-tDNA with CSF cytology. Rather than replacing cytology, we envision that CSF-tDNA will be used in combination with it and other biomarkers under development as well as with radiographic and clinical parameters (3538). This could substantially increase the accuracy of the estimates of tumor burden at various points during the management of patients.

Given the invasive and risky nature of surgical interventions on the brain and spinal cord, it would be useful to be able to identify a neoplastic process without performing surgery. Our results provide a glimpse of the potential for this form of diagnosis in the future. We evaluated four patients: one patient with a tumor in the midbrain, one patient with a tumor in the pons, and two patients with a tumor in the spinal cord. Using WES, we were able to detect CSF-tDNA in two of four cases by comparing the data with those obtained by targeted sequencing with SafeSeqS. The results were consistent with expectation, in that the mutant fractions revealed by genome-wide sequencing were in accord with those identified by targeted sequencing (Table 2). Additional cases will need to be tested to elucidate the potential of this approach in patients in whom biopsies are challenging, but our results show that genome-wide analysis of the DNA from CSF is feasible in at least some cases.

Although the results described above are promising, we caution that this is an exploratory study designed primarily to determine whether it was possible to detect CSF-tDNA in patients with primary CNS tumors. A secondary goal was to document the anatomical and pathologic characteristics of the tumors that shed DNA into the CSF. The most important technical limitation of our study is that CSF samples were obtained at the time of surgery, and they were often from the ventricles rather than from a lumbar puncture. CSF has been shown to quickly circulate throughout the ventricles and spinal reservoirs (39, 40). It is, therefore, very likely that the DNA in the spinal fluid obtained through lumbar puncture will be similar to that of the ventricles, although the fluid obtained from lumbar puncture is farther away from the site of malignancy. An additional consideration is that, in individuals with a bulky mass that obstructs spinal fluid flow or elevates intracranial pressure, a lumbar puncture might be unsafe. However, these patients will almost always require surgical decompression to reduce the mass effect generated by the tumor, and CSF could be safely obtained after opening the dura. The exact method and location of CSF sampling in patients with CNS neoplasms will need to be individualized, and they will be based on a number of factors, including tumor location, ease of CSF sampling, and clinical characteristics. For example, patients may initially undergo CSF sampling from an intracranial space at the time of surgery to determine baseline levels of CSF-tDNA, but lumbar punctures could be used to longitudinally monitor CSF-tDNA levels.

Now that it has been documented that most primary brain tumors release tDNA into the CSF, the stage is set for a longitudinal study of the clinical use of this biomarker. Our results suggest specific guidelines for such a follow-up study. The optimal patients to follow would be those with medulloblastomas, ependymomas, or high-grade gliomas that abut a CSF space, because the CSF-tDNA assay is particularly sensitive in such cases and these tumor types are relatively common. CSF-tDNA should be evaluated intraoperatively to establish a baseline, and a concomitant lumbar puncture should be performed when possible to ensure concordance between the two fluid samples. Subsequent evaluations of CSF obtained through lumbar puncture or an implanted reservoir should be compared with other clinical and laboratory features, with the goal of determining the use of CSF-tDNA to detect minimal residual disease. For example, patients whose mass persists on MRI but CSF-tDNA is undetectable might be spared a second biopsy. Alternatively, patients in whom residual disease is evident on CSF-tDNA analysis but equivocal on imaging analysis might be well-served by additional therapy. In the future, it is likely that most brain tumors will be routinely assessed for mutations in various genes of interest for both prognostic and therapeutic purposes (4143). The availability of such sequencing data should make the approach described here more cost-effective and easier to implement.

Materials and Methods

Patient Samples.

All samples were collected after approval was obtained from the Johns Hopkins Institutional Review Board and informed consent was provided. Whole blood and CSF were collected at the time of surgery before surgical manipulation of the tumor. A WBC pellet was prepared from the blood sample after hypotonic lysis of RBCs by centrifugation at 200 × g. CSF was frozen in its entirety at −80 °C until DNA purification, and the entire volume of CSF (cells plus fluid) was used for DNA purification. The amount of CSF used averaged 4.8 mL (range = 0.75–10 mL). When fresh tumor tissue from surgical specimens was available, it was immediately frozen at −80 °C. When frozen tissue was not available, formalin-fixed, paraffin-embedded tissues were used for DNA purification. In either case (fresh frozen or formalin-fixed, paraffin-embedded), tumors were macrodissected to ensure neoplastic cellularity exceeding 50%. DNA was purified from the white cell pellet, CSF, and tumor using an ALLPrep Kit (Qiagen).

Statistical Analysis.

Clinical characteristics were compared between the CSF samples with and without detectable CSF-tDNA with Fisher’s exact test or t test. Correlation coefficients among outcomes were estimated using Pearson correlation statistics. A logistic regression model was used to estimate the odds of detecting CSF-tDNA under different conditions. All P values are two-sided, and all analyses were conducted using SAS software (version 9.2; SAS Institute).

Tumor Mutational Profiling.

A tiered approach was used to determine a somatic mutation within each tumor. Initially, a PCR-based approach testing for mutations in codons 130–139 of IDH1; codons 126–155, 144–178, and 250–262 of IDH2; all coding exons of TP53; and the TERT promoter was used (4448). If no mutations were present within these genes, paired-end libraries of DNA from the tumors and WBC pellets were prepared and captured (SureSelect; Agilent) as previously described (47). Massively parallel sequencing was carried out on an Illumina HiSeq Instrument at either the Goldman Sequencing Facility at Johns Hopkins Medical Institutions or Personal Genome Diagnostics. Mutations were identified as previously described (47, 4952).

Mutation Detection in CSF.

DNA from tumor, WBCs, and CSF was used to validate the somatic mutations identified by targeted sequencing and determine whether these mutations could be found in the CSF; 3–5 ng tumor and WBC DNA was used for each assay, whereas all DNA from the CSF (for cases with <20 ng CSF DNA available) or 20 ng CSF DNA was used for each assay (Table S2). For this purpose, primers were designed to amplify an ∼100-bp region surrounding each mutation. The two primers had universal sequences at their 5′ ends, allowing a second round of PCR to be performed using a second set of primers containing these sequences (19, 47). The sequences of the primers used to assess each mutation are listed in Table S5. Oligonucleotides used in this study were synthesized by TriLink Biotechnologies. The final PCR products (after two rounds of PCR) were purified with AMPure (Beckman) and sequenced using an Illumina MiSeq Instrument. The data were analyzed with the SafeSeqS Pipeline, allowing mutations occurring as infrequently as 0.01% to be detected and quantified with confidence using the experimental conditions applied (19). In every case, DNA from the normal cells served as a control to ensure that the mutations were not the result of errors generated during the DNA purification, amplification, or sequencing processes. Four paired-end libraries for CSF samples were also generated and exome-captured (Table 2). Preparation of the genomic library was performed using the TruSeq DNA Sample Prep Kit (Illumina) according to the manufacturer’s recommendations. Exomic capture (SureSelect; Agilent) and massively parallel sequencing were carried out as described above.

Table S5.

Primers used for mutation detection

Patient Gene Mutation Location Forward primer sequence Reverse primer sequence
CGLI 02 ZNF513 NM_144631.5 (ZNF513):c.413C > T Exon CACACAGGAAACAGCTATGACCATGCTTCAGGTGGCTCGAGTAGTG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGTGAGGGGCCGTGTTGT
CGLI 03 PIK3R1 NM_181523.2 (PIK3R1):c.1690A > G Exon CACACAGGAAACAGCTATGACCATGTTTCTTTTGCCTGCAGGATT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCTGAATTGTAGCAATCACCAA
CGLI 06 CLCA3P NR_024604.1 (CLCA3P):c.446G > A Exon CACACAGGAAACAGCTATGACCATGTGGACAATGTGGAGATAAAGGAC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCACCAGTGGAAATAAGAACAGA
CGLI 11 ANKS3 NM_133450.3 (ANKS3):c.1738G > A Exon CACACAGGAAACAGCTATGACCATGCTGGTCCTGCCTCACTCG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTCCCTCCCCTAGAAGGTCTG
CGLI 12 HIST1H3C NM_003531.2 (HIST1H3C):c.83T > A Exon CACACAGGAAACAGCTATGACCATGGAAGCAAACAGCTCGCAAGT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGCGGTAGCGATGAGGTTTC
CGLI 13 TTC16 NM_144965.1 (TTC16):c.1016T > A Exon CACACAGGAAACAGCTATGACCATGGCCAGCTGTTGCTGACCTA CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNAGAGTCCTTTCTCCTGCTGCT
CGLI 14 TP53 NM_000546.5 (TP53):c.638G > A Exon CACACAGGAAACAGCTATGACCATGAGACCTCAGGCGGCTCATAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTTGCGTGTGGAGTATTTGGA
CGLI 15 MARS NM_004990.3 (MARS):c.1487C > T Exon CACACAGGAAACAGCTATGACCATGGAGTGGTTGGGGAGGACATT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGTTCCCCATTTGAGGTCTCG
CGLI 20 RGS12 NM_198229.2 (RGS12):c.2980C > T Exon CACACAGGAAACAGCTATGACCATGCTGTCAAGGCGGGCTTCT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGCAGCACGAGGGTCAGG
CGLI 22 CC2D2A NM_001080522.2 (CC2D2A):c.1938G > A Exon CACACAGGAAACAGCTATGACCATGATCGGGCAGTGATAGAGCAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTCATTGGGTGTTACGCTTCC
CGLI 25 CDH5 NM_001795.3 (CDH5):c.1211G > A Exon CACACAGGAAACAGCTATGACCATGGAAGCCTCTGATTGGCACAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNAGCAGGCATAGGAACATCCA
CGLI 26 IDH1 NM_005896.2 (IDH1):c.394C > T Exon CACACAGGAAACAGCTATGACCATGGCTTGTGAGTGGATGGGTAAAACCTATCAT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNATGCAAAATCACATTATTGCCAACATGACT
CGLI 28 IDH1 NM_005896.2 (IDH1):c.395G > A Exon CACACAGGAAACAGCTATGACCATGGCTTGTGAGTGGATGGGTAAAACCTATCAT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNATGCAAAATCACATTATTGCCAACATGACT
CGLI 29 TP53 NM_000546.5 (TP53):c.742C > T Exon CACACAGGAAACAGCTATGACCATGTCCACTACAACTACATGTGTAACAGTTC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTGTGATGATGGTGAGGATGG
CGLI 31 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 35 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 36 TP53 NM_000546.5 (TP53):c.742C > T Exon CACACAGGAAACAGCTATGACCATGTGGCTCTGACTGTACCACCATC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGTGGCAAGTGGCTCCTGA
CGLI 39 NF2 NM_000268.3 (NF2):c.592C > T Exon CACACAGGAAACAGCTATGACCATGCGGAAATGTGGGAGGAGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNAAAGCCCATAAAGGAATGTAAACC
CGLI 40 CASR NM_000388.3 (CASR):c.2549C > T Exon CACACAGGAAACAGCTATGACCATGCCAGCACCTATGGCAAGTTT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCGGGATGGCTTGAAGAGA
CGLI 41 TP53 NM_000546.5 (TP53):c.396G > C Exon CACACAGGAAACAGCTATGACCATGGCCCTGACTTTCAACTCTGTCT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGGGGGTGTGGAATCAACC
CGLI 42 COL6A1 NM_001848.2 (COL6A1):c.1417G > A Exon CACACAGGAAACAGCTATGACCATGGTCCAACGTGCCATATCCAT CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGGTGGGGAGTGGGACTCA
CGLI 43 NF2 NM_000268.3 (NF2):c.1009C > T Exon CACACAGGAAACAGCTATGACCATGCCTTGTGGCACCCTAGGTC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGTTCAGCCTCCTCCCTCATC
CGLI 44 ITGA11 NM_001004439.1 (ITGA11):c.2140C > T Exon CACACAGGAAACAGCTATGACCATGGGAAGTTGATCCGCTCACA CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCATCAGATACAACGCCACCAT
CGLI 47 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 48 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 50 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 51 TERT NM_198253.2 (TERT):c.1–124C > T Promoter CACACAGGAAACAGCTATGACCATGGCGGAAAGGAAGGGGAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNCCCGTCCCGACCCCT
CGLI 55 PTEN NM_000314.4 (PTEN):c.388C > T Exon CACACAGGAAACAGCTATGACCATGGCAATTCACTGTAAAGCTGGAAA CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTGGTCCTTACTTCCCCATAGAA
CGLI 56 AQR NM_014691.2 (AQR):c.1746delT Exon CACACAGGAAACAGCTATGACCATGTCTGACATAAACCAGGCCAAC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNACCAGGTCTTCGTAAGCATGA
CGLI 58 ADCK2 NM_052853.3 (ADCK2):c.1057T > A Exon CACACAGGAAACAGCTATGACCATGCCGAGTCCTGGGAGTTTTG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTACAGCTGAGGGGAGGAGAA
CGLI 60 MTMR4 NM_004687.4 (MTMR4):c.2110C > G Exon CACACAGGAAACAGCTATGACCATGCGAGGCACTGCGGTATTAAG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTGGGTCTTCCTCCTCCTCTG
CGLI 61 TENM2 NM_001122679.1 (TENM2):c.799G > A Exon CACACAGGAAACAGCTATGACCATGACAGCCAGTCGACTCTGAGG CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNATCTGACTCCGCCGATTG
CGLI 63 PTCH1 NM_000264.3 (PTCH1):c.746+1C > A Splice site CACACAGGAAACAGCTATGACCATGAATGGAACAAACAATGATAAGCAA CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNGGAAGGGGCGAAATTACAGT
CGLI 101 TP53 NM_000546.5 (TP53):c.731G > A Exon CACACAGGAAACAGCTATGACCATGTCCACTACAACTACATGTGTAACAGTTC CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNTGTGATGATGGTGAGGATGG
CGLI 254 KDM6A NM_021140.2 (KDM6A):c.4153C > T Exon CACACAGGAAACAGCTATGACCATGACTCACATGTTGATTTGACTTACTAATGTA CGACGTAAAACGACGGCCAGTNNNNNNNNNNNNNNACAGTACAAAATGGAGGACCTGA

Ns in primer sequences represent bases with an equal probability of A, C, T, and G.

Supplementary Material

Supplementary File
pnas.1511694112.sd01.xlsx (170.4KB, xlsx)

Acknowledgments

We thank our patients for their courage and generosity. We also thank N. Silliman, J. Schaefer, C. Blair, K. Judge, and M. Popoli for technical assistance. This work was supported by a Burroughs Wellcome Career Award for Medical Scientists, Alex’s Lemonade Stand/Malia’s Cord Foundation, the Pediatric Brain Tumor Foundation, a Johns Hopkins Clinician Scientist Award, Doris Duke Charitable Foundation Grant 2014107, The Virginia and D. K. Ludwig Fund for Cancer Research, The Banyan Gate Foundation, Swim Across America, The Sol Goldman Sequencing Facility at Johns Hopkins, and NIH Grants CA43460 and NS70024.

Footnotes

The authors declare no conflict of interest.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1511694112/-/DCSupplemental.

References

  • 1.Ostrom QT, et al. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro-oncol. 2013;15(Suppl 2):ii1–ii56. doi: 10.1093/neuonc/not151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kros JM, et al. Circulating glioma biomarkers. Neuro-oncol. 2014 doi: 10.1093/neuonc/nou207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Van Mieghem E, et al. Defining pseudoprogression in glioblastoma multiforme. Eur J Neurol. 2013;20(10):1335–1341. doi: 10.1111/ene.12192. [DOI] [PubMed] [Google Scholar]
  • 4.Müller C, et al. Hematogenous dissemination of glioblastoma multiforme. Sci Transl Med. 2014;6(247):247ra101. doi: 10.1126/scitranslmed.3009095. [DOI] [PubMed] [Google Scholar]
  • 5.Macarthur KM, et al. Detection of brain tumor cells in the peripheral blood by a telomerase promoter-based assay. Cancer Res. 2014;74(8):2152–2159. doi: 10.1158/0008-5472.CAN-13-0813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sullivan JP, et al. Brain tumor cells in circulation are enriched for mesenchymal gene expression. Cancer Discov. 2014;4(11):1299–1309. doi: 10.1158/2159-8290.CD-14-0471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Diehl F, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–990. doi: 10.1038/nm.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bettegowda C, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6(224):224ra24. doi: 10.1126/scitranslmed.3007094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dawson SJ, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368(13):1199–1209. doi: 10.1056/NEJMoa1213261. [DOI] [PubMed] [Google Scholar]
  • 10.Newman AM, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20(5):548–554. doi: 10.1038/nm.3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Martignetti JA, et al. Personalized ovarian cancer disease surveillance and detection of candidate therapeutic drug target in circulating tumor DNA. Neoplasia. 2014;16(1):97–103. doi: 10.1593/neo.131900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sidransky D, et al. Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors. Science. 1992;256(5053):102–105. doi: 10.1126/science.1566048. [DOI] [PubMed] [Google Scholar]
  • 13.Sidransky D, et al. Identification of p53 gene mutations in bladder cancers and urine samples. Science. 1991;252(5006):706–709. doi: 10.1126/science.2024123. [DOI] [PubMed] [Google Scholar]
  • 14.Ralla B, et al. Nucleic acid-based biomarkers in body fluids of patients with urologic malignancies. Crit Rev Clin Lab Sci. 2014;51(4):200–231. doi: 10.3109/10408363.2014.914888. [DOI] [PubMed] [Google Scholar]
  • 15.Hubers AJ, Prinsen CF, Sozzi G, Witte BI, Thunnissen E. Molecular sputum analysis for the diagnosis of lung cancer. Br J Cancer. 2013;109(3):530–537. doi: 10.1038/bjc.2013.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Diehl F, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135(2):489–498. doi: 10.1053/j.gastro.2008.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kinde I, et al. Evaluation of DNA from the Papanicolaou test to detect ovarian and endometrial cancers. Sci Transl Med. 2013;5(167):167ra4. doi: 10.1126/scitranslmed.3004952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Vogelstein B, et al. Cancer genome landscapes. Science. 2013;339(6127):1546–1558. doi: 10.1126/science.1235122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 2011;108(23):9530–9535. doi: 10.1073/pnas.1105422108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Woodworth GF, et al. Histopathological correlates with survival in reoperated glioblastomas. J Neurooncol. 2013;113(3):485–493. doi: 10.1007/s11060-013-1141-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Krueger DA, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. 2010;363(19):1801–1811. doi: 10.1056/NEJMoa1001671. [DOI] [PubMed] [Google Scholar]
  • 22.Pan W, Gu W, Nagpal S, Gephart MH, Quake SR. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem. 2015;61(3):514–522. doi: 10.1373/clinchem.2014.235457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.von Hoff K, Rutkowski S. Medulloblastoma. Curr Treat Options Neurol. 2012;14(4):416–426. doi: 10.1007/s11940-012-0183-8. [DOI] [PubMed] [Google Scholar]
  • 24.Bartlett F, Kortmann R, Saran F. Medulloblastoma. Clin Oncol (R Coll Radiol) 2013;25(1):36–45. doi: 10.1016/j.clon.2012.09.008. [DOI] [PubMed] [Google Scholar]
  • 25.Glass JP, Melamed M, Chernik NL, Posner JB. Malignant cells in cerebrospinal fluid (CSF): The meaning of a positive CSF cytology. Neurology. 1979;29(10):1369–1375. doi: 10.1212/wnl.29.10.1369. [DOI] [PubMed] [Google Scholar]
  • 26.Preusser M, Hainfellner JA. CSF and laboratory analysis (tumor markers) Handb Clin Neurol. 2012;104:143–148. doi: 10.1016/B978-0-444-52138-5.00011-6. [DOI] [PubMed] [Google Scholar]
  • 27.Allory Y, et al. Telomerase reverse transcriptase promoter mutations in bladder cancer: High frequency across stages, detection in urine, and lack of association with outcome. Eur Urol. 2014;65(2):360–366. doi: 10.1016/j.eururo.2013.08.052. [DOI] [PubMed] [Google Scholar]
  • 28.Destro A, et al. K-ras and p16(INK4A)alterations in sputum of NSCLC patients and in heavy asymptomatic chronic smokers. Lung Cancer. 2004;44(1):23–32. doi: 10.1016/j.lungcan.2003.10.002. [DOI] [PubMed] [Google Scholar]
  • 29.Mack SC, et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature. 2014;506(7489):445–450. doi: 10.1038/nature13108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gajjar A, Pfister SM, Taylor MD, Gilbertson RJ. Molecular insights into pediatric brain tumors have the potential to transform therapy. Clin Cancer Res. 2014;20(22):5630–5640. doi: 10.1158/1078-0432.CCR-14-0833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rudin CM, et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med. 2009;361(12):1173–1178. doi: 10.1056/NEJMoa0902903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Thompson MC, et al. Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations. J Clin Oncol. 2006;24(12):1924–1931. doi: 10.1200/JCO.2005.04.4974. [DOI] [PubMed] [Google Scholar]
  • 33.Moreno L, et al. Utility of cerebrospinal fluid cytology in newly diagnosed childhood ependymoma. J Pediatr Hematol Oncol. 2010;32(6):515–518. doi: 10.1097/MPH.0b013e3181d7adf5. [DOI] [PubMed] [Google Scholar]
  • 34.Weston CL, Glantz MJ, Connor JR. Detection of cancer cells in the cerebrospinal fluid: Current methods and future directions. Fluids Barriers CNS. 2011;8(1):14. doi: 10.1186/2045-8118-8-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Khwaja FW, et al. Proteomic identification of biomarkers in the cerebrospinal fluid (CSF) of astrocytoma patients. J Proteome Res. 2007;6(2):559–570. doi: 10.1021/pr060240z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Roy S, et al. Protein biomarker identification in the CSF of patients with CNS lymphoma. J Clin Oncol. 2008;26(1):96–105. doi: 10.1200/JCO.2007.12.1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bougel S, et al. Methylation of the hTERT promoter: A novel cancer biomarker for leptomeningeal metastasis detection in cerebrospinal fluids. Clin Cancer Res. 2013;19(8):2216–2223. doi: 10.1158/1078-0432.CCR-12-1246. [DOI] [PubMed] [Google Scholar]
  • 38.Samuel N, Remke M, Rutka JT, Raught B, Malkin D. Proteomic analyses of CSF aimed at biomarker development for pediatric brain tumors. J Neurooncol. 2014;118(2):225–238. doi: 10.1007/s11060-014-1432-3. [DOI] [PubMed] [Google Scholar]
  • 39.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(1):42–45. doi: 10.1093/neuonc/3.1.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gajjar A, et al. Comparison of lumbar and shunt cerebrospinal fluid specimens for cytologic detection of leptomeningeal disease in pediatric patients with brain tumors. J Clin Oncol. 1999;17(6):1825–1828. doi: 10.1200/JCO.1999.17.6.1825. [DOI] [PubMed] [Google Scholar]
  • 41.Thomas L, Di Stefano AL, Ducray F. Predictive biomarkers in adult gliomas: The present and the future. Curr Opin Oncol. 2013;25(6):689–694. doi: 10.1097/CCO.0000000000000002. [DOI] [PubMed] [Google Scholar]
  • 42.Olar A, Aldape KD. Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol. 2014;232(2):165–177. doi: 10.1002/path.4282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gajjar AJ, Robinson GW. Medulloblastoma-translating discoveries from the bench to the bedside. Nat Rev Clin Oncol. 2014;11(12):714–722. doi: 10.1038/nrclinonc.2014.181. [DOI] [PubMed] [Google Scholar]
  • 44.Horn S, et al. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339(6122):959–961. doi: 10.1126/science.1230062. [DOI] [PubMed] [Google Scholar]
  • 45.Huang FW, et al. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339(6122):957–959. doi: 10.1126/science.1229259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Killela PJ, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci USA. 2013;110(15):6021–6026. doi: 10.1073/pnas.1303607110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bettegowda C, et al. Exomic sequencing of four rare central nervous system tumor types. Oncotarget. 2013;4(4):572–583. doi: 10.18632/oncotarget.964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kinde I, et al. TERT promoter mutations occur early in urothelial neoplasia and are biomarkers of early disease and disease recurrence in urine. Cancer Res. 2013;73(24):7162–7167. doi: 10.1158/0008-5472.CAN-13-2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhang M, et al. Somatic mutations of SUZ12 in malignant peripheral nerve sheath tumors. Nat Genet. 2014;46(11):1170–1172. doi: 10.1038/ng.3116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Agrawal N, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333(6046):1154–1157. doi: 10.1126/science.1206923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Agrawal N, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov. 2012;2(10):899–905. doi: 10.1158/2159-8290.CD-12-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bettegowda C, et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science. 2011;333(6048):1453–1455. doi: 10.1126/science.1210557. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary File
pnas.1511694112.sd01.xlsx (170.4KB, xlsx)

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES