2019
DOI: 10.1101/mcs.a003863
|View full text |Cite
|
Sign up to set email alerts
|

Lynch syndrome–associated ultra-hypermutated pediatric glioblastoma mimicking a constitutional mismatch repair deficiency syndrome

Abstract: Pediatric glioblastoma multiforme (GBM) has a poor prognosis as a result of recurrence after treatment of surgery and radiochemotherapy. A small subset of pediatric GBMs presenting with an ultra-high tumor mutational burden (TMB) may be sensitive to immune checkpoint inhibition. Here we report a 16-yr-old male with an ultra-hypermutated GBM. After incomplete surgical resection, molecular analysis of the tumor identified unusually high numbers of mutations and intratumor heterogeneity by a hotspot next-generati… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

1
18
0

Year Published

2020
2020
2024
2024

Publication Types

Select...
7
1
1

Relationship

0
9

Authors

Journals

citations
Cited by 23 publications
(19 citation statements)
references
References 39 publications
1
18
0
Order By: Relevance
“…The proportion of MSI/dMMR brain cancers is very low (2% to 3%) [ 8 , 102 ]. Some studies focused on these tumors (related to somatic MMR mutations, to LS, or to CMMRD (Constitutional MisMatch Repair Deficiency) syndrome, which is caused by biallelic germline MMR mutations) [ 103 , 104 ] and showed that they can acquire a secondarily proofreading defect due to a mutation in the POLE (DNA polymerase epsilon, catalytic subunit) gene, resulting in an ultra-mutant phenotype (>100 mutations/Mb) and a unique mutational signature (“MMR first/POLE second”) [ 103 , 104 , 105 ]. Additionally, these tumors seem to be more frequently of a particular and very rare histotype characterized by the presence of multinucleated giant cells [ 106 ].…”
Section: Tumor Characteristics In Lynch Syndromementioning
confidence: 99%
See 1 more Smart Citation
“…The proportion of MSI/dMMR brain cancers is very low (2% to 3%) [ 8 , 102 ]. Some studies focused on these tumors (related to somatic MMR mutations, to LS, or to CMMRD (Constitutional MisMatch Repair Deficiency) syndrome, which is caused by biallelic germline MMR mutations) [ 103 , 104 ] and showed that they can acquire a secondarily proofreading defect due to a mutation in the POLE (DNA polymerase epsilon, catalytic subunit) gene, resulting in an ultra-mutant phenotype (>100 mutations/Mb) and a unique mutational signature (“MMR first/POLE second”) [ 103 , 104 , 105 ]. Additionally, these tumors seem to be more frequently of a particular and very rare histotype characterized by the presence of multinucleated giant cells [ 106 ].…”
Section: Tumor Characteristics In Lynch Syndromementioning
confidence: 99%
“…The sensitivity of MSI has been shown to be lower in non-CRC tumors, especially in endometrium, brain, and urothelial tumors, as a consequence of fewer unstable markers and a shorter instability length [ 25 , 69 , 117 , 118 , 121 , 122 , 123 , 124 , 125 ]. For example, MSI is particularly difficult to detect in brain tumors, with less than 20% of brain tumors from patients with LS (or CMMRD) exhibiting MSI [ 69 , 105 , 126 , 127 , 128 ]. These observations have led to the recommendation to analyze normal DNA in parallel for non-CRC tumors [ 114 , 122 , 124 , 125 ].…”
Section: Tumor Characteristics In Lynch Syndromementioning
confidence: 99%
“…POLE mutational signature is related to recurrent POLE somatic mutations, which may suggest that DNA replication is associated with errors in proofreading activity of Pol ε (32). DNA mismatch repair mutational signatures are usually microsatellite-unstable, serving as a better indicator of response (33). In this study, AA and POLE mutational signatures were only observed in the UTUC cohorts.…”
Section: Discussionmentioning
confidence: 61%
“…Despite these promising results, a recent study reported that PD-1 blockade did not impact mOS in hypermutated gliomas, consistent with an observed lack of TILs in these cancers ( 68 ). However, another study reported significant clinical and radiological responses of nivolumab in two young siblings with biallelic mismatch repair deficiency ( 66 ), suggesting that ICI therapy might benefit pediatric GBM with high mutational burden [e.g., with MSH6 mutations ( 69 )]. It is plausible that treatments that increase mutational burden might synergize with ICI, as has been shown in other cancers ( 70 ).…”
Section: The Future Of Immunotherapies In Gbmmentioning
confidence: 99%