Li-Fraumeni syndrome (LFS) is a rare autosomal dominant tumor predisposition syndrome caused by heterozygous germline mutation or deletion of the TP53 tumor suppressor gene on chromosome 17p13. The prevalence of deleterious TP53 germline mutations in humans is estimated to range from 1 in 5,000 to 20,000 [6]. These TP53 germline alterations can either be inherited across generations or arise de novo, of which de novo acquisition is estimated to occur in 20% of affected patients [6]. LFS results in an increased risk of many different cancer types, including breast carcinoma, adrenocortical carcinoma, lymphoblastic leukemia, osteosarcoma, and brain tumors [6]. The three brain tumor types most commonly associated with LFS are choroid plexus carcinoma, medulloblastoma, and glioma [9]. Approximately one third of choroid plexus carcinomas are known to arise in the setting of LFS [11], whereas LFS only accounts for a small fraction of childhood medulloblastomas. The majority of medulloblastomas arising in the setting of LFS occur in the second decade of life and are most commonly large cell/anaplastic tumors belonging to the SHH-activated and TP53-mutant molecular subtype [12]. However, the histologic, molecular, and clinical features of gliomas arising in the setting of LFS have not been well characterized to date.
Here we performed comprehensive genomic characterization and studied the clinicopathologic features of 14 gliomas arising in the setting of LFS (Table 1 and Supplementary Table 1). We identified that gliomas arising in the setting of LFS are diffuse astrocytic neoplasms that can be segregated into two molecular subgroups based on IDH mutation status that are associated with divergent clinicopathologic features and patient outcomes as described below (Fig. 1).
Table 1.
Clinicopathologic features of the 13 patients with gliomas arising in the setting of Li-Fraumeni syndrome.
Patient | Sex | Age | Personal cancer hx | Family cancer hx | Tumor location | Histology | Clinical outcome | Follow-up |
---|---|---|---|---|---|---|---|---|
LF-1 | F | 28 | None | None reported | Cerebral hemisphere | Diffuse astrocytoma | Alive with progressive disease | 24 months |
LF-2A* | F | 23 | Breast DCIS | None reported | Cerebral hemisphere | Diffuse astrocytoma | Died of disease | 79 months |
LF-2B* | 23 | Cerebral hemisphere | Diffuse astrocytoma | Died of unrelated tumor | 79 months | |||
LF-3 | F | 22 | None | Unknown | Cerebral hemisphere | Diffuse astrocytoma | Alive, no evidence of progression | 25 months |
LF-4 | M | 20 | None | Unknown | Cerebral hemisphere | Diffuse astrocytoma | Alive, no evidence of progression | 27 months |
LF-5 | F | 24 | None | Unknown | Cerebral hemisphere | Diffuse astrocytoma | Alive, no evidence of progression | 23 months |
LF-6 | F | 10 | Osteosarcom a, multiple gliomas | Choroid plexus carcinoma (sister) | Cerebral hemisphere | Diffuse astrocytoma | Alive, no evidence of progression | 6 months |
LF-7 | M | 4 | None | None reported | Thalami | Glioblastoma | Died of disease | 2 months |
LF-8 | M | 6 | None | None reported | Cerebral hemisphere | Glioblastoma | Died of disease | 12 months |
LF-9 | M | 18 | Medulloblast oma | None reported | Cerebellum | Anaplastic astrocytoma | Alive, no evidence of progression | 15 months |
LF-10 | M | 18 | None | Adrenal cortical carcinoma (sister) | Cerebral hemisphere | Anaplastic astrocytoma | Alive with progressive disease | 13 months |
LF-11 | F | 4 | None | Phyllodes tumor (mother); osteosarcoma (uncle); rhabdomyosarcoma (aunt); astrocytoma, colon carcinoma, liposarcoma (grandfather) | Cerebral hemisphere | Diffuse astrocytoma | Alive, no evidence of progression | 7 months |
LF-12 | M | 11 | Osteosarcoma | Brain cancer (aunt); ovarian cancer (grandmother); rhabdomyosarcoma (uncle) | Cerebral hemisphere | Anaplastic astrocytoma | Alive, no evidence of progression | 8 months |
LF-13 | M | 6 | None | None reported | Cerebral hemisphere | Glioblastoma | Alive with progressive disease | 4 months |
Patient LF-2 had two spatially distinct gliomas that were synchronously resected and independently evaluated.
Fig. 1.
Clinicopathologic features of gliomas arising in the setting of Li-Fraumeni syndrome (LFS). a Imaging and histology from patient LF-1 (28-year-old woman) showing an expansile, nonenhancing, T2/FLAIR-hyperintense mass centered in the left temporal lobe with histologic features of a diffuse astrocytoma that was IDH-mutant. b Imaging and histology from patient LF-7 (4-year-old boy) showing an expansile and enhancing mass involving the thalami and left lateral ventricle with histologic features of a glioblastoma that was IDH-wildtype. c Oncoprint table of the clinicopathologic and molecular features of the 14 gliomas arising in the setting of LFS. d,e Kaplan-Meier analysis of progression-free survival (d) and disease-specific survival (e) stratified by IDH mutation status for the 14 gliomas arising in the setting of LFS. p value calculated by Log-rank (Mantel-Cox) test.
The IDH-mutant subgroup consisted of seven tumors from six patients, five female and one male, with median age at glioma diagnosis of 23 years (range 10–28). These seven tumors were uniformly located in the cerebral hemispheres and were all IDH-mutant diffuse astrocytomas lacking necrosis, microvascular proliferation, and significant mitotic activity. All seven tumors harbored IDH1 mutations, three of which were p.R132H and four of which were the less common variant p.R132C (Supplementary Table 2). Six tumors harbored biallelic TP53 inactivation due to a germline missense mutation accompanied by somatic loss of heterozygosity (four cases) or a somatic nonsense or missense mutation (two cases; Supplementary Table 3). The seventh tumor (patient LF-1) arose in the setting of constitutional mosaicism for a damaging TP53 missense mutation that was acquired during post-zygotic development, with the glioma arising from a cell affected by the mosaicism that acquired a second somatic nonsense mutation inactivating the remaining TP53 allele (Fig. 1a). Additionally, there were inactivating ATRX mutations in six of the seven tumors, with an intact/wildtype ATRX gene in the youngest patient (LF-7). This absence of ATRX loss in patient LF-7 (10 years of age at time of glioma diagnosis) is similar to what has been previously reported in sporadic IDH-mutant astrocytomas in teenagers which lack the ATRX inactivation that is typical of their IDH-mutant astrocytoma counterparts in adults [3, 7]. Very few additional pathogenic alterations were identified beyond IDH1, TP53, and ATRX, with only one tumor harboring additional CIC nonsense and ZBTB20 missense mutations (Fig. 1c). There were few, if any, chromosomal copy number changes in these seven tumors (Supplementary Table 4). The median progression-free survival for these patients with IDH-mutant astrocytomas arising in the setting of LFS was 57 months (Fig. 1d and e).
The IDH-wildtype subgroup consisted of seven tumors from seven patients, six male and one female, with median age at glioma diagnosis of 6 years (range 4–18). These seven tumors were located in the cerebral hemispheres (five), thalami (one), and cerebellum (one). Histologic diagnoses were glioblastoma (three), anaplastic astrocytoma (three), and diffuse astrocytoma (one). All seven tumors lacked hotspot mutations involving IDH1, IDH2, H3F3A, H3F3B, HIST1H3B, and HIST1H3C. Instead, five of the tumors harbored somatic biallelic inactivation of the NF1 tumor suppressor gene and one harbored multiple EGFR activating missense mutations (p.L861Q and p.R252P) in the absence of EGFR gene amplification (Supplementary Table 2). Additionally, two tumors harbored focal high-level amplification of the MYCN oncogene, and individual tumors also harbored homozygous deletion of CDKN2A/B, focal high-level amplification of CDK6 or IGF1R, an activating missense mutation in PTPN11, and inactivating mutations in PBRM1 or PTPRD (Fig. 1c). All seven tumors harbored biallelic TP53 inactivation due to a germline missense mutation (five cases) or gene deletion (two cases) accompanied by somatic loss of heterozygosity in all cases (Supplementary Table 3). All of the six histologically high-grade astrocytomas demonstrated numerous chromosomal copy number aberrations, whereas the one IDH-wildtype low-grade diffuse astrocytoma from patient LF-12 that was resected after identification on surveillance imaging lacked any chromosomal copy number changes (Supplementary Table 4). The median progression-free survival for these patients with IDH-wildtype astrocytomas arising in the setting of LFS was 5 months (Fig. 1d and e).
Together, these findings indicate that gliomas arising in the setting of Li-Fraumeni syndrome are diffuse astrocytic neoplasms that can be stratified into two molecular subgroups based on IDH status. Based on this cohort, IDH-mutant astrocytomas arising in the setting of LFS most often occur in females in the second and third decades of life, arise supratentorially in the cerebral hemispheres, have low-grade histologic features, harbor co-occurring IDH and ATRX mutations with a paucity of other pathogenic drivers, have near-diploid genomes, and are associated with favorable prognosis. In contrast, IDH-wildtype astrocytomas arising in the setting of LFS most often occur in males in the first and second decades of life, arise throughout the neuroaxis, have high-grade histologic features, harbor frequent NF1 mutations/deletions along with a plethora of other pathogenic drivers including MYCN amplification, have aneuploid genomes, and are associated with unfavorable prognosis compared to their IDH-mutant counterparts. These IDH-wildtype astrocytomas appear to be heterogenous in their anatomic location and genetic drivers (in contrast to the homogeneity of their IDH-mutant counterparts), and therefore may likely represent a biologically diverse group of tumors.
Interestingly, the stepwise genetic progression that occurs in sporadic IDH-mutant astrocytomas starts with initial acquisition of IDH1 mutation, followed by TP53 mutation and then ATRX inactivation [1, 4]. This is in contrast to IDH-mutant astrocytomas arising in the setting of LFS where a germline TP53 mutation is the initiating genetic driver, followed by somatic inactivation of the remaining TP53 allele that occurs prior to acquisition of IDH1 mutation and then ATRX inactivation (inferred based on variant allele frequencies, see Supplementary Table 2). Notably, our cohort confirms a high frequency of variant IDH1 mutations (most commonly p.R132C) that has been previously identified in LFS-associated gliomas [13]. As has recently been appreciated for gliomas arising in the setting of neurofibromatosis type 1 [2], we speculate that gliomas arising in the setting of LFS may represent distinct glioma subtypes that should be distinguished from their sporadic counterparts for more informative diagnostic, prognostic, and therapeutic classification. We suggest the terminology “IDH-mutant [or IDH-wildtype] astrocytoma arising in the setting of Li-Fraumeni syndrome” along with a description of the histologic features (necrosis, microvascular proliferation, mitotic activity) and associated genetic alterations as the best integrated diagnostic framework for these syndromic gliomas pending further investigation.
Methods
The study cohort consisted of nine patients who underwent surgical resection of a glioma that had been prospectively clinically evaluated on the UCSF500 Cancer Panel, one patient who underwent surgical resection of two anatomically and genetically distinct gliomas that were evaluated by exome sequencing on a research basis that has been previously reported (patient 1 from reference 4), and three patients (TCGA-S9-A6U1, TCGA-DB-A64S, and TCGA-VM-A8CH) from the diffuse lower-grade glioma cohort of The Cancer Genome Atlas Research Network with IDH-mutant astrocytomas found to harbor known pathogenic germline TP53 mutations [1]. All of the thirteen patients included in this cohort were genetically confirmed to have Li-Fraumeni syndrome by identification of a known inactivating/pathogenic TP53 mutation or deletion in a constitutional DNA sample via paired tumor-normal sequencing or via germline sequencing analysis at a commercial source. Methodology related to tumor genomic profiling via the UCSF500 Cancer Panel has been previously reported [5, 8, 10].
Supplementary Material
Acknowledgements
We thank the staff of the UCSF Clinical Cancer Genomics Laboratory for assistance with genetic profiling. This study was supported in part by the Sandler Foundation through the UCSF Glioblastoma Precision Medicine Program. D.A.S. is supported by the NIH Director’s Early Independence Award (DP5 OD021403, National Institutes of Health).
Footnotes
Data availability
Mutation and copy number data are available in the electronic supplementary material. Sequencing data files are available from the authors upon request.
Ethical approval
This study was approved by the Committee on Human Research of the University of California, San Francisco, with a waiver of patient consent.
Conflict of interest
The authors declare that they have no competing interests related to this report.
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version
References
- 1.Cancer Genome Atlas Research Network (2015) Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med 372:2481–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.D’Angelo F, Ceccarelli M, Tala et al. (2019) The molecular landscape of glioma in patients with Neurofibromatosis 1. Nat Med 25:176–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ferris SP, Goode B, Joseph NM et al. (2016) IDH1 mutation can be present in diffuse astrocytomas and giant cell glioblastomas of young children under 10 years of age. Acta Neuropathol 132:153–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hayes J, Yu Y, Jalbert LE (2018) Genomic analysis of the origins and evolution of multicentric diffuse lower-grade gliomas. Neuro Oncol 20:632–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kline CN, Joseph NM, Grenert JP et al. (2017) Targeted next-generation sequencing of pediatric neuro-oncology patients improves diagnosis, identifies pathogenic germline mutations, and directs targeted therapy. Neuro Oncol 19:699–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kratz CP, Achatz MI, Brugieres L et al. (2017) Cancer screening recommendations for individuals with Li-Fraumeni syndrome. Clin Cancer Res 23:e38–e45. [DOI] [PubMed] [Google Scholar]
- 7.Lee J, Putnam AR, Chesier SH et al. (2018) Oligodendrogliomas, IDH-mutant and 1p/19q-codeleted,arising during teenage years often lack TERT promoter mutation that is typical of their adult counterparts. Acta Neuropathol Commun 6:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lopez GY, Van Ziffle J, Onodera C et al. (2019). The genetic landscape of gliomas arising after therapeutic radiation. Acta Neuropathol 137:139–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Orr BA, Clay MR, Pinto EM, Kesserwan C (2019). An update on the central nervous system manifestations of Li-Fraumeni syndrome. Acta Neuropathol [Epub Aug 30] doi: 10.1007/s00401-019-02055-3. [DOI] [PubMed] [Google Scholar]
- 10.Pekmezci M, Villanueva-Meyer JE, Goode B et al. (2018) The genetic landscape of ganglioglioma. Acta Neuropathol Commun 6:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tabori U, Shlien A, Baskin B et al. (2010) TP53 alterations determine clinical subgroups and survival of patients with choroid plexus tumors. J Clin Oncol 28:1995–2001. [DOI] [PubMed] [Google Scholar]
- 12.Waszak SM, Northcott PA, Buchhalter I et al. (2018) Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort. Lancet Oncol 19:785–798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Watanabe T, Vital A, Nobusawa S, Kleihues P, Ohgaki H (2009) Selective acquisition of IDH1 R132Cmutations in astrocytomas associated with Li-Fraumeni syndrome. Acta Neuropathol 117:653–656. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.