Harmonization of postmortem donations for pediatric brain tumors and molecular characterization of diffuse midline gliomas

Madhuri Kambhampati, Eshini Panditharatna, Sridevi Yadavilli, Karim Saoud, Sulgi Lee, Augustine Eze, M I Almira-Suarez, Lauren Hancock, Erin R Bonner, Jamila Gittens, Mojca Stampar, Krutika Gaonkar, Adam C Resnick, Cassie Kline, Cheng-Ying Ho, Angela J Waanders, Maria-Magdalena Georgescu, Naomi E Rance, Yong Kim, Courtney Johnson, Brian R Rood, Lindsay B Kilburn, Eugene I Hwang, Sabine Mueller, Roger J Packer, Miriam Bornhorst, Javad Nazarian, Madhuri Kambhampati, Eshini Panditharatna, Sridevi Yadavilli, Karim Saoud, Sulgi Lee, Augustine Eze, M I Almira-Suarez, Lauren Hancock, Erin R Bonner, Jamila Gittens, Mojca Stampar, Krutika Gaonkar, Adam C Resnick, Cassie Kline, Cheng-Ying Ho, Angela J Waanders, Maria-Magdalena Georgescu, Naomi E Rance, Yong Kim, Courtney Johnson, Brian R Rood, Lindsay B Kilburn, Eugene I Hwang, Sabine Mueller, Roger J Packer, Miriam Bornhorst, Javad Nazarian

Abstract

Children diagnosed with brain tumors have the lowest overall survival of all pediatric cancers. Recent molecular studies have resulted in the discovery of recurrent driver mutations in many pediatric brain tumors. However, despite these molecular advances, the clinical outcomes of high grade tumors, including H3K27M diffuse midline glioma (H3K27M DMG), remain poor. To address the paucity of tissue for biological studies, we have established a comprehensive protocol for the coordination and processing of donated specimens at postmortem. Since 2010, 60 postmortem pediatric brain tumor donations from 26 institutions were coordinated and collected. Patient derived xenograft models and cell cultures were successfully created (76% and 44% of attempts respectively), irrespective of postmortem processing time. Histological analysis of mid-sagittal whole brain sections revealed evidence of treatment response, immune cell infiltration and the migratory path of infiltrating H3K27M DMG cells into other midline structures and cerebral lobes. Sequencing of primary and disseminated tumors confirmed the presence of oncogenic driver mutations and their obligate partners. Our findings highlight the importance of postmortem tissue donations as an invaluable resource to accelerate research, potentially leading to improved outcomes for children with aggressive brain tumors.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Coordination characteristics of postmortem donation program at CNH. (a) Flow chart of personnel and steps involved for coordination. (b) Patient demographics, types of brain tumors collected, logistics, and generation of postmortem preclinical models. Flow chart graphics were made using Adobe Illustrator.
Figure 2
Figure 2
Specimens collected, comprehensive whole brain processing, and utilization of specimens for preclinical models. (a) Detailed figure of processing in a patient with DMG using different platforms and utilization of tumor and healthy regions for frozen, FFPE, preclinical model generation (cell culture or xenograft) and DMSO preservation of whole brain. (b) RNA integrity was not affected by postmortem tissue collection time in DMG tumors. (c) Tissue collected for DMSO preservation representing multiple neuroanatomical locations for future generation of preclinical models. Graphics were made using Adobe Illustrator.
Figure 3
Figure 3
Characterization and fidelity of DMG preclinical models derived from postmortem tissue. (a) Schematic for generation of preclinical models (xenografts or primary cells) from fresh or DMSO cryopreserved tissue. (b) Oncoplot showing preclinical models generated and genomic aberrations of primary tumor obtained from WGS. (c) Characteristics of primary tumor and a matched primary cell line and xenograft model derived from postmortem tissue harboring H3.3K27M. Scale Bar = 100 µm. Schematic graphics were created using Adobe Illustrator.
Figure 4
Figure 4
Identification of patterns of tumor dissemination in H3K27M DMG. (a) Mid-sagittal histological characterization for identification of tumor dissemination (shown by asterisk*) as assessed by a neuropathologist. (b) H&E images of specific locations confirming presence of tumor. (c) Mutational profile identified in primary tumor and other neuroanatomical locations show affected oncogenic pathways (not all locations mentioned in the oncoplot have tumors as per histology or mutational profile). Scale Bar = 100 µm Final figure was combined on Adobe Illustrator.

References

    1. Altekruse SF, K.C., Krapcho M, Neyman N, Aminou R, Waldron W, Ruhl J, Howlader N, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Cronin K, Chen HS, Feuer EJ, Stinchcomb DG, Edwards BK (eds). SEER Cancer Statistics Review: 1975–2007. (2010).
    1. States, C.B.T.R.o.t.U. Statistical Report: Primary brain tumors in the United States, 2004–2007 (2011).
    1. Ostrom QT, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol. 2019;21:v1–v100.
    1. Neftel C, et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell. 2019;178:835–849.
    1. Sturm D, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22:425–437.
    1. Brennan CW, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462–477.
    1. Mistry M, et al. BRAF mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma. J. Clin. Oncol. 2015;33:1015–1022.
    1. Wu G, et al. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat. Genet. 2014;46:444–450.
    1. Schwartzentruber J, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–231.
    1. Mackay A, et al. Integrated molecular meta-analysis of 1,000 pediatric high-grade and diffuse intrinsic pontine glioma. Cancer Cell. 2017;32:520–537.
    1. Albert CM, Davis JL, Federman N, Casanova M, Laetsch TW. TRK fusion cancers in children: a clinical review and recommendations for screening. J. Clin. Oncol. 2019;37:513–524.
    1. Clarke M, et al. Infant high grade gliomas comprise multiple subgroups characterized by novel targetable gene fusions and favorable outcomes. Cancer Discov. 2020 doi: 10.1158/-19-1030.
    1. Hoffman LM, et al. Clinical, radiologic, pathologic, and molecular characteristics of long-term survivors of diffuse intrinsic pontine glioma (DIPG): a collaborative report from the international and European society for pediatric oncology DIPG registries. J. Clin. Oncol. 2018;36:1963–1972.
    1. Filbin MG, et al. Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell RNA-seq. Science. 2018;360:331–335.
    1. Nagaraja S, et al. Transcriptional dependencies in diffuse intrinsic pontine glioma. Cancer Cell. 2017;31:635–652.
    1. Saratsis AM, et al. Comparative multidimensional molecular analyses of pediatric diffuse intrinsic pontine glioma reveals distinct molecular subtypes. Acta Neuropathol. 2014;127:881–895.
    1. Nikbakht H, et al. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma. Nat. Commun. 2016;7:11185.
    1. Paugh BS, et al. Genome-wide analyses identify recurrent amplifications of receptor tyrosine kinases and cell-cycle regulatory genes in diffuse intrinsic pontine glioma. J. Clin. Oncol. 2011;29:3999–4006.
    1. Chan KM, et al. The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression. Genes Dev. 2013;27:985–990.
    1. Castel D, et al. Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta Neuropathol. 2015;130:815–827.
    1. van Zanten SEMV, et al. External validation of the diffuse intrinsic pontine glioma survival prediction model: a collaborative report from the International DIPG Registry and the SIOPE DIPG Registry. J. Neurooncol. 2017;134:231–240.
    1. Baugh J, et al. The international diffuse intrinsic pontine glioma registry: an infrastructure to accelerate collaborative research for an orphan disease. J. Neurooncol. 2017;132:323–331.
    1. Kambhampati M, et al. A standardized autopsy procurement allows for the comprehensive study of DIPG biology. Oncotarget. 2015;6:12740.
    1. Wu G, et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 2012;44:251–253.
    1. Louis DN, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803–820.
    1. Nikbakht H, et al. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma. Nat. Commun. 2016;7:1–8.
    1. Vinci M, et al. Functional diversity and cooperativity between subclonal populations of pediatric glioblastoma and diffuse intrinsic pontine glioma cells. Nat. Med. 2018;24:1204–1215.
    1. Hoffman LM, et al. Spatial genomic heterogeneity in diffuse intrinsic pontine and midline high-grade glioma: implications for diagnostic biopsy and targeted therapeutics. Acta Neuropathol. Commun. 2016;4:1.
    1. Fontebasso AM, et al. Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma. Nat. Genet. 2014;46:462–466.
    1. Buczkowicz P, et al. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 2014;46:451–456.
    1. Taylor KR, et al. Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma. Nat. Genet. 2014;46:457–461.
    1. Pratt D, et al. Diffuse intrinsic pontine glioma-like tumor with EZHIP expression and molecular features of PFA ependymoma. Acta Neuropathol. Commun. 2020;8:37.
    1. Mueller S, et al. A pilot precision medicine trial for children with diffuse intrinsic pontine glioma-PNOC003: a report from the Pacific Pediatric Neuro-Oncology Consortium. Int. J. Cancer. 2019;145:1889–1901.
    1. Bugiani M, et al. Deceptive morphologic and epigenetic heterogeneity in diffuse intrinsic pontine glioma. Oncotarget. 2017;8:60447–60452.
    1. Grasso CS, et al. Functionally defined therapeutic targets in diffuse intrinsic pontine glioma. Nat. Med. 2015;21:555–559.
    1. Tsoli M, et al. Correction to: International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma. J. Neurooncol. 2019;141:265.
    1. Lin GL, Monje M. A protocol for rapid post-mortem cell culture of diffuse intrinsic pontine glioma (DIPG) J. Vis. Exp. 2017;121:e55360.
    1. Broniscer A, et al. Prospective collection of tissue samples at autopsy in children with diffuse intrinsic pontine glioma. Cancer. 2010;116:4632–4637.
    1. Caretti V, et al. Implementation of a multi-institutional diffuse intrinsic pontine glioma autopsy protocol and characterization of a primary cell culture. Neuropathol. Appl. Neurobiol. 2012;39:426–436.
    1. Caretti V, et al. Subventricular spread of diffuse intrinsic pontine glioma. Acta Neuropathol. 2014;128:605–607.
    1. Lin DC, et al. The genomic landscape of nasopharyngeal carcinoma. Nat. Genet. 2014;46:866–871.
    1. El-Ayadi M, et al. Concurrent IDH1 and SMARCB1 mutations in pediatric medulloblastoma: a case report. Front. Neurol. 2018;9:398.
    1. Zhang H, Xiang B, Chen H, Chen X, Cai T. A novel deletion mutation in KMT2A identified in a child with ID/DD and blood eosinophilia. BMC Med. Genet. 2019;20:38.
    1. Tsoli M, et al. International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma. J. Neurooncol. 2019;141:253–263.
    1. Saratsis AM, et al. Comparative multidimensional molecular analyses of pediatric diffuse intrinsic pontine glioma reveals distinct molecular subtypes. Acta Neuropathol. 2013;127:881–895.
    1. Buczkowicz P, Hawkins C. Pathology, molecular genetics and epigenetics of diffuse intrinsic pontine glioma. Front. Oncol. 2015;5:147.
    1. Ballester LY, et al. Morphologic characteristics and immunohistochemical profile of diffuse intrinsic pontine gliomas. Am. J. Surg. Pathol. 2013;37:1357–1364.
    1. Panditharatna E, et al. Clinically relevant and minimally invasive tumor surveillance of pediatric diffuse midline gliomas using patient-derived liquid biopsy. Clin. Cancer Res. 2018;24:5850–5859.

Source: PubMed

3
Sottoscrivi