[en] Patient-based cancer models are essential tools for studying tumor biology and for the assessment of drug responses in a translational context. We report the establishment a large cohort of unique organoids and patient-derived orthotopic xenografts (PDOX) of various glioma subtypes, including gliomas with mutations in IDH1, and paired longitudinal PDOX from primary and recurrent tumors of the same patient. We show that glioma PDOXs enable long-term propagation of patient tumors and represent clinically relevant patient avatars that retain histopathological, genetic, epigenetic, and transcriptomic features of parental tumors. We find no evidence of mouse-specific clonal evolution in glioma PDOXs. Our cohort captures individual molecular genotypes for precision medicine including mutations in IDH1, ATRX, TP53, MDM2/4, amplification of EGFR, PDGFRA, MET, CDK4/6, MDM2/4, and deletion of CDKN2A/B, PTCH, and PTEN. Matched longitudinal PDOX recapitulate the limited genetic evolution of gliomas observed in patients following treatment. At the histological level, we observe increased vascularization in the rat host as compared to mice. PDOX-derived standardized glioma organoids are amenable to high-throughput drug screens that can be validated in mice. We show clinically relevant responses to temozolomide (TMZ) and to targeted treatments, such as EGFR and CDK4/6 inhibitors in (epi)genetically defined subgroups, according to MGMT promoter and EGFR/CDK status, respectively. Dianhydrogalactitol (VAL-083), a promising bifunctional alkylating agent in the current clinical trial, displayed high therapeutic efficacy, and was able to overcome TMZ resistance in glioblastoma. Our work underscores the clinical relevance of glioma organoids and PDOX models for translational research and personalized treatment studies and represents a unique publicly available resource for precision oncology.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
Golebiewska, Anna; Luxembourg Institute of Health
Hau, Ann-Christin; Luxembourg Institute of Health
Oudin, Anaïs
Stieber, Daniel
Yabo, Yahaya
Baus, Virginie
Barthélémy, Vanessa
Klein, Eliane
Bougnaud, Sébastien
Keunen, Olivier
Wantz, May
Michelucci, Alessandro
Neirinckx, Virginie ; Luxembourg Institute of Health > Department of oncology > NorLux Neuro-Oncology Laboratory
Muller, Arnaud
Kaoma, Tony
Nazarov, Petr
Azuaje, Francisco
De Falco, Alfonso
Flies, Ben
Richart, Lorraine
Poovathingal, Suresh
Arns, Thais
Grzyb, Kamil
Mock, Andreas
Herold-Mende, Christel
Steino, Anne
Brown, Dennis
May, Patrick
Miletic, Hrvoje
Malta, Tathiane
Noushmehr, Houtan
Kwon, Yong-Jun
Jahn, Winnie
Klink, Barbara
Tanner, Georgette
Stead, Lucy
Mittelbronn, Michel
Skupin, Alexander
Hertel, Frank
Bjerkvig, Rolf
Niclou, Simone; Luxembourg Institute of Health > Department of Oncology > NorLux Neuro-oncology laboratory
scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.
Bibliography
Abdul Rahim SA, Dirkse A, Oudin A, Schuster A, Bohler J, Barthelemy V et al (2017) Regulation of hypoxia-induced autophagy in glioblastoma involves ATG9A. Br J Cancer 117:813–825. 10.1038/bjc.2017.263 DOI: 10.1038/bjc.2017.263
Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR et al (2019) Challenges to curing primary brain tumours. Nat Rev Clin Oncol 16:509–520. 10.1038/s41571-019-0177-5 DOI: 10.1038/s41571-019-0177-5
Alonso R, Salavert F, Garcia-Garcia F, Carbonell-Caballero J, Bleda M, Garcia-Alonso L et al (2015) Babelomics 5.0: functional interpretation for new generations of genomic data. Nucleic Acids Res 43:W117–121. 10.1093/nar/gkv384 DOI: 10.1093/nar/gkv384
Balvers RK, Kleijn A, Kloezeman JJ, French PJ, Kremer A, van den Bent MJ et al (2013) Serum-free culture success of glial tumors is related to specific molecular profiles and expression of extracellular matrix-associated gene modules. Neuro Oncol 15:1684–1695. 10.1093/neuonc/not116 DOI: 10.1093/neuonc/not116
Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E et al (2019) Longitudinal molecular trajectories of diffuse glioma in adults. Nature. 10.1038/s41586-019-1775-1
Baskaran S, Mayrhofer M, Kultima HG, Bergstrom T, Elfineh L, Cavelier L et al (2018) Primary glioblastoma cells for precision medicine: a quantitative portrait of genomic (in)stability during the first 30 passages. Neuro Oncol 20:1080–1091. 10.1093/neuonc/noy024 DOI: 10.1093/neuonc/noy024
Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J et al (2017) Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet 49:1567–1575. 10.1038/ng.3967 DOI: 10.1038/ng.3967
Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B (Methodol) 57:289–300
Binder ZA, Thorne AH, Bakas S, Wileyto EP, Bilello M, Akbari H et al (2018) Epidermal growth factor receptor extracellular domain mutations in glioblastoma present opportunities for clinical imaging and therapeutic development. Cancer Cell 34(163–177):e167. 10.1016/j.ccell.2018.06.006 DOI: 10.1016/j.ccell.2018.06.006
Bjerkvig R, Tonnesen A, Laerum OD, Backlund EO (1990) Multicellular tumor spheroids from human gliomas maintained in organ culture. J Neurosurg 72:463–475. 10.3171/jns.1990.72.3.0463 DOI: 10.3171/jns.1990.72.3.0463
Bonavia R, Inda MM, Cavenee WK, Furnari FB (2011) Heterogeneity maintenance in glioblastoma: a social network. Cancer Res 71:4055–4060. 10.1158/0008-5472.CAN-11-0153 DOI: 10.1158/0008-5472.CAN-11-0153
Bougnaud S, Golebiewska A, Oudin A, Keunen O, Harter PN, Mader L et al (2016) Molecular crosstalk between tumour and brain parenchyma instructs histopathological features in glioblastoma. Oncotarget 7:31955–31971. 10.18632/oncotarget.7454 DOI: 10.18632/oncotarget.7454
Brabetz S, Leary SES, Grobner SN, Nakamoto MW, Seker-Cin H, Girard EJ et al (2018) A biobank of patient-derived pediatric brain tumor models. Nat Med 24:1752–1761. 10.1038/s41591-018-0207-3 DOI: 10.1038/s41591-018-0207-3
Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR et al (2013) The somatic genomic landscape of glioblastoma. Cell 155:462–477. 10.1016/j.cell.2013.09.034 DOI: 10.1016/j.cell.2013.09.034
Butler A, Hoffman P, Smibert P, Papalexi E, Satija R (2018) Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol 36:411–420. 10.1038/nbt.4096 DOI: 10.1038/nbt.4096
Byrne AT, Alferez DG, Amant F, Annibali D, Arribas J, Biankin AV et al (2017) Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer 17:254–268. 10.1038/nrc.2016.140 DOI: 10.1038/nrc.2016.140
Callari M, Batra AS, Batra RN, Sammut SJ, Greenwood W, Clifford H et al (2018) Computational approach to discriminate human and mouse sequences in patient-derived tumour xenografts. BMC Genomics 19:19. 10.1186/s12864-017-4414-y DOI: 10.1186/s12864-017-4414-y
Campos B, Wan F, Farhadi M, Ernst A, Zeppernick F, Tagscherer KE et al (2010) Differentiation therapy exerts antitumor effects on stem-like glioma cells. Clin Cancer Res 16:2715–2728 DOI: 10.1158/1078-0432.CCR-09-1800
Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA et al (2013) The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45:1113–1120. 10.1038/ng.2764 DOI: 10.1038/ng.2764
Capper D, Jones DTW, Sill M, Hovestadt V, Schrimpf D, Sturm D et al (2018) DNA methylation-based classification of central nervous system tumours. Nature 555:469–474. 10.1038/nature26000 DOI: 10.1038/nature26000
Carlson BL, Pokorny JL, Schroeder MA, Sarkaria JN (2011) Establishment, maintenance and in vitro and in vivo applications of primary human glioblastoma multiforme (GBM) xenograft models for translational biology studies and drug discovery. Curr Protoc Pharmacol Chapter 14:14–16. 10.1002/0471141755.ph1416s52
Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray BA et al (2016) Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164:550–563. 10.1016/j.cell.2015.12.028 DOI: 10.1016/j.cell.2015.12.028
Chen S, Zhou Y, Chen Y, Gu J (2018) fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34:i884–i890. 10.1093/bioinformatics/bty560 DOI: 10.1093/bioinformatics/bty560
Chen X, Schulz-Trieglaff O, Shaw R, Barnes B, Schlesinger F, Kallberg M et al (2016) Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics 32:1220–1222. 10.1093/bioinformatics/btv710 DOI: 10.1093/bioinformatics/btv710
Cho J, Pastorino S, Zeng Q, Xu X, Johnson W, Vandenberg S et al (2011) Glioblastoma-derived epidermal growth factor receptor carboxyl-terminal deletion mutants are transforming and are sensitive to EGFR-directed therapies. Cancer Res 71:7587–7596. 10.1158/0008-5472.CAN-11-0821 DOI: 10.1158/0008-5472.CAN-11-0821
Christensen K, Aaberg-Jessen C, Andersen C, Goplen D, Bjerkvig R, Kristensen BW (2010) Immunohistochemical expression of stem cell, endothelial cell, and chemosensitivity markers in primary glioma spheroids cultured in serum-containing and serum-free medium. Neurosurgery 66:933–947. 10.1227/01.NEU.0000368393.45935.46 DOI: 10.1227/01.NEU.0000368393.45935.46
Cingolani P, Platts A, le Wang L, Coon M, Nguyen T, Wang L et al (2012) A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 6:80–92. 10.4161/fly.19695 DOI: 10.4161/fly.19695
Claes A, Schuuring J, Boots-Sprenger S, Hendriks-Cornelissen S, Dekkers M, van der Kogel AJ et al (2008) Phenotypic and genotypic characterization of orthotopic human glioma models and its relevance for the study of anti-glioma therapy. Brain Pathol 18:423–433. 10.1111/j.1750-3639.2008.00141.x DOI: 10.1111/j.1750-3639.2008.00141.x
Consortium G (2018) Glioma through the looking GLASS: molecular evolution of diffuse gliomas and the Glioma Longitudinal Analysis Consortium. Neuro Oncol 20:873–884. 10.1093/neuonc/noy020 DOI: 10.1093/neuonc/noy020
Conte N, Mason JC, Halmagyi C, Neuhauser S, Mosaku A, Yordanova G et al (2019) PDX Finder: a portal for patient-derived tumor xenograft model discovery. Nucleic Acids Res 47:D1073–D1079. 10.1093/nar/gky984 DOI: 10.1093/nar/gky984
Darmanis S, Sloan SA, Croote D, Mignardi M, Chernikova S, Samghababi P et al (2017) Single-Cell RNA-seq analysis of infiltrating neoplastic cells at the migrating front of human glioblastoma. Cell reports 21:1399–1410. 10.1016/j.celrep.2017.10.030 DOI: 10.1016/j.celrep.2017.10.030
de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y et al (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol 12:233–242. 10.1093/neuonc/nop027 DOI: 10.1093/neuonc/nop027
de Souza CF, Sabedot TS, Malta TM, Stetson L, Morozova O, Sokolov A et al (2018) A distinct DNA methylation shift in a subset of glioma CpG island methylator phenotypes during tumor recurrence. Cell reports 23:637–651. 10.1016/j.celrep.2018.03.107 DOI: 10.1016/j.celrep.2018.03.107
De Witt Hamer PC, Van Tilborg AA, Eijk PP, Sminia P, Troost D, Van Noorden CJ et al (2008) The genomic profile of human malignant glioma is altered early in primary cell culture and preserved in spheroids. Oncogene 27:2091–2096 DOI: 10.1038/sj.onc.1210850
deCarvalho AC, Kim H, Poisson LM, Winn ME, Mueller C, Cherba D, Koeman J, Seth S et al (2018) Discordant inheritance of chromosomal and extrachromosomal DNA elements contributes to dynamic disease evolution in glioblastoma. Nat Genet 50:708–717. 10.1038/s41588-018-0105-0 DOI: 10.1038/s41588-018-0105-0
Dirkse A, Golebiewska A, Buder T, Nazarov PV, Muller A, Poovathingal S et al (2019) Stem cell-associated heterogeneity in Glioblastoma results from intrinsic tumor plasticity shaped by the microenvironment. Nature Commun 10:1787. 10.1038/s41467-019-09853-z DOI: 10.1038/s41467-019-09853-z
Doh I, Kwon YJ, Ku B, Lee DW (2019) Drug efficacy comparison of 3D forming and preforming sphere models with a micropillar and microwell chip platform. SLAS Disc Adv Life Sci R & D 24:476–483. 10.1177/2472555218821292 DOI: 10.1177/2472555218821292
Draaisma K, Chatzipli A, Taphoorn M, Kerkhof M, Weyerbrock A, Sanson M et al (2019) Molecular Evolution of IDH wild-type glioblastomas treated with standard of care affects survival and design of precision medicine trials: a report from the EORTC 1542 study. J Clin Oncol. 10.1200/JCO.19.00367
Eckhardt S, Csetenyi J, Horvath IP, Kerpel-Fronius S, Szamel I, Institoris L et al (1977) Uptake of labeled dianhydrogalactitol into human gliomas and nervous tissue. Cancer Treat Rep 61:841–847
Eirew P, Steif A, Khattra J, Ha G, Yap D, Farahani H et al (2015) Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 518:422–426. 10.1038/nature13952 DOI: 10.1038/nature13952
Eskilsson E, Rosland GV, Solecki G, Wang Q, Harter PN, Graziani G et al (2018) EGFR heterogeneity and implications for therapeutic intervention in glioblastoma. Neuro Oncol 20:743–752. 10.1093/neuonc/nox191 DOI: 10.1093/neuonc/nox191
Evrard YA, Srivastava A, Randjelovic J, Consortium NP, Doroshow JH, Dean DA et al (2020) Systematic establishment of robustness and standards in patient-derived xenograft experiments and analysis. Cancer Res. 10.1158/0008-5472.CAN-19-3101
Fack F, Tardito S, Hochart G, Oudin A, Zheng L, Fritah S et al (2017) Altered metabolic landscape in IDH-mutant gliomas affects phospholipid, energy, and oxidative stress pathways. EMBO Mol Med 9:1681–1695. 10.15252/emmm.201707729 DOI: 10.15252/emmm.201707729
Francies HE, Barthorpe A, McLaren-Douglas A, Barendt WJ, Garnett MJ (2019) Drug sensitivity assays of human cancer organoid cultures. Methods Mol Biol 1576:339–351. 10.1007/7651_2016_10 DOI: 10.1007/7651_2016_10
Francis JM, Zhang CZ, Maire CL, Jung J, Manzo VE, Adalsteinsson VA et al (2014) EGFR variant heterogeneity in glioblastoma resolved through single-nucleus sequencing. Cancer Discov 4:956–971. 10.1158/2159-8290.CD-13-0879 DOI: 10.1158/2159-8290.CD-13-0879
Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M et al (2015) High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nature Med 21:1318–1325. 10.1038/nm.3954
Golebiewska A, Bougnaud S, Stieber D, Brons NH, Vallar L, Hertel F et al (2013) Side population in human glioblastoma is non-tumorigenic and characterizes brain endothelial cells. Brain J Neurol 136:1462–1475. 10.1093/brain/awt025 DOI: 10.1093/brain/awt025
Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:997–1003. 10.1056/NEJMoa043331 DOI: 10.1056/NEJMoa043331
Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC et al (2016) A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer Res 76:2465–2477. 10.1158/0008-5472.CAN-15-2402 DOI: 10.1158/0008-5472.CAN-15-2402
Hulten A, Kerstell J, Larsson J, Olsson R, Svanborg A (1968) A method of calculating the nursing work load. Lakartidningen 65:1683–1686
Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH et al (2020) A Patient-Derived Glioblastoma Organoid Model And Biobank Recapitulates Inter- And Intra-Tumoral Heterogeneity. Cell 180(188–204):e122. 10.1016/j.cell.2019.11.036 DOI: 10.1016/j.cell.2019.11.036
Joo KM, Kim J, Jin J, Kim M, Seol HJ, Muradov J et al (2013) Patient-specific orthotopic glioblastoma xenograft models recapitulate the histopathology and biology of human glioblastomas in situ. Cell reports 3:260–273. 10.1016/j.celrep.2012.12.013 DOI: 10.1016/j.celrep.2012.12.013
Keunen O, Johansson M, Oudin A, Sanzey M, Abdul Rahim SA, Fack F et al (2011) Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma. Proc Natl Acad Sci USA. 10.1073/pnas.1014480108
Kim H, Zheng S, Amini SS, Virk SM, Mikkelsen T, Brat DJ et al (2015) Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution. Genome Res 25:316–327. 10.1101/gr.180612.114 DOI: 10.1101/gr.180612.114
Klink B, Miletic H, Stieber D, Huszthy PC, Valenzuela JA, Balss J et al (2013) A novel, diffusely infiltrative xenograft model of human anaplastic oligodendroglioma with mutations in FUBP1, CIC, and IDH1. PLoS One 8:e59773. 10.1371/journal.pone.0059773 DOI: 10.1371/journal.pone.0059773
Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH et al (2018) The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med 24:1611–1624. 10.1038/s41591-018-0156-x DOI: 10.1038/s41591-018-0156-x
Korber V, Yang J, Barah P, Wu Y, Stichel D, Gu Z et al (2019) Evolutionary trajectories of IDH(WT) glioblastomas reveal a common path of early tumorigenesis instigated years ahead of initial diagnosis. Cancer Cell 35(692–704):e612. 10.1016/j.ccell.2019.02.007 DOI: 10.1016/j.ccell.2019.02.007
Kriebel J, Herder C, Rathmann W, Wahl S, Kunze S, Molnos S et al (2016) Association between DNA Methylation in whole blood and measures of glucose metabolism: KORA F4 study. Plos One 11:e0152314. 10.1371/journal.pone.0152314 DOI: 10.1371/journal.pone.0152314
Kristensen BW, Priesterbach-Ackley LP, Petersen JK, Wesseling P (2019) Molecular pathology of tumors of the central nervous system. Ann Oncol. 10.1093/annonc/mdz164 DOI: 10.1093/annonc/mdz164
Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S et al (2018) ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res 46:D1062–D1067. 10.1093/nar/gkx1153 DOI: 10.1093/nar/gkx1153
Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM et al (2006) Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9:391–403. 10.1016/j.ccr.2006.03.030 DOI: 10.1016/j.ccr.2006.03.030
Lee JC, Vivanco I, Beroukhim R, Huang JH, Feng WL, DeBiasi RM et al (2006) Epidermal growth factor receptor activation in glioblastoma through novel missense mutations in the extracellular domain. PLoS Med 3:e485. 10.1371/journal.pmed.0030485 DOI: 10.1371/journal.pmed.0030485
Lee JK, Liu Z, Sa JK, Shin S, Wang J, Bordyuh M et al (2018) Pharmacogenomic landscape of patient-derived tumor cells informs precision oncology therapy. Nat Genet 50:1399–1411. 10.1038/s41588-018-0209-6 DOI: 10.1038/s41588-018-0209-6
Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N et al (2009) The Sequence alignment/map format and SAMtools. Bioinformatics 25:2078–2079. 10.1093/bioinformatics/btp352
Liffers K, Lamszus K, Schulte A (2015) EGFR amplification and glioblastoma stem-like cells. Stem Cells Int 2015:427518. 10.1155/2015/427518 DOI: 10.1155/2015/427518
Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK et al (2016) The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820. 10.1007/s00401-016-1545-1 DOI: 10.1007/s00401-016-1545-1
Luchman HA, Chesnelong C, Cairncross JG, Weiss S (2013) Spontaneous loss of heterozygosity leading to homozygous R132H in a patient-derived IDH1 mutant cell line. Neuro Oncol 15:979–980. 10.1093/neuonc/not064 DOI: 10.1093/neuonc/not064
Luchman HA, Stechishin OD, Dang NH, Blough MD, Chesnelong C, Kelly JJ et al (2012) An in vivo patient-derived model of endogenous IDH1-mutant glioma. Neuro Oncol 14:184–191. 10.1093/neuonc/nor207 DOI: 10.1093/neuonc/nor207
Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M et al (2015) Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161:1202–1214. 10.1016/j.cell.2015.05.002 DOI: 10.1016/j.cell.2015.05.002
Mahesparan R, Read TA, Lund-Johansen M, Skaftnesmo KO, Bjerkvig R, Engebraaten O (2003) Expression of extracellular matrix components in a highly infiltrative in vivo glioma model. Acta Neuropathol 105:49–57. 10.1007/s00401-002-0610-0 DOI: 10.1007/s00401-002-0610-0
Meehan TF, Conte N, Goldstein T, Inghirami G, Murakami MA, Brabetz S et al (2017) PDX-MI: minimal information for patient-derived tumor xenograft models. Cancer Res 77:e62–e66. 10.1158/0008-5472.CAN-17-0582 DOI: 10.1158/0008-5472.CAN-17-0582
Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ et al (2005) Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353:2012–2024. 10.1056/NEJMoa051918 DOI: 10.1056/NEJMoa051918
Muller F, Scherer M, Assenov Y, Lutsik P, Walter J, Lengauer T et al (2019) RnBeads 2.0: comprehensive analysis of DNA methylation data. Genome biology 20:55. 10.1186/s13059-019-1664-9
Navis AC, Niclou SP, Fack F, Stieber D, van Lith S, Verrijp K et al (2013) Increased mitochondrial activity in a novel IDH1-R132H mutant human oligodendroglioma xenograft model: in situ detection of 2-HG and alpha-KG. Acta Neuropathol Commun 1:18. 10.1186/2051-5960-1-18 DOI: 10.1186/2051-5960-1-18
Nazarov PV, Wienecke-Baldacchino AK, Zinovyev A, Czerwinska U, Muller A, Nashan D et al (2019) Deconvolution of transcriptomes and miRNomes by independent component analysis provides insights into biological processes and clinical outcomes of melanoma patients. BMC Med Genomics 12:132. 10.1186/s12920-019-0578-4 DOI: 10.1186/s12920-019-0578-4
Neftel C, Laffy J, Filbin MG, Hara T, Shore ME, Rahme GJ et al (2019) An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178(835–849):e821. 10.1016/j.cell.2019.06.024 DOI: 10.1016/j.cell.2019.06.024
Niclou SP, Danzeisen C, Eikesdal HP, Wiig H, Brons NH, Poli AM et al (2008) A novel eGFP-expressing immunodeficient mouse model to study tumor-host interactions. Faseb J 22:3120–3128. 10.1096/fj.08-109611 DOI: 10.1096/fj.08-109611
Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP et al (2010) Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17:510–522. 10.1016/j.ccr.2010.03.017 DOI: 10.1016/j.ccr.2010.03.017
Obad N, Espedal H, Jirik R, Sakariassen PO, Brekke Rygh C, Lund-Johansen M et al (2018) Lack of functional normalisation of tumour vessels following anti-angiogenic therapy in glioblastoma. J Cerebral Blood Flow Metabol 38:1741–1753. 10.1177/0271678X17714656 DOI: 10.1177/0271678X17714656
Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H et al (2014) Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344:1396–1401. 10.1126/science.1254257 DOI: 10.1126/science.1254257
Pires-Afonso Y, Niclou SP, Michelucci A (2020) Revealing and harnessing tumour-associated microglia/macrophage heterogeneity in glioblastoma. Int J Mol Sci. 10.3390/ijms21030689 DOI: 10.3390/ijms21030689
Reis GF, Pekmezci M, Hansen HM, Rice T, Marshall RE, Molinaro AM et al (2015) CDKN2A loss is associated with shortened overall survival in lower-grade (World Health Organization Grades II-III) astrocytomas. J Neuropathol Exp Neurol 74:442–452. 10.1097/NEN.0000000000000188 DOI: 10.1097/NEN.0000000000000188
Roth A, Khattra J, Yap D, Wan A, Laks E, Biele J et al (2014) PyClone: statistical inference of clonal population structure in cancer. Nat Methods 11:396–398. 10.1038/nmeth.2883 DOI: 10.1038/nmeth.2883
Sahm F, Schrimpf D, Jones DT, Meyer J, Kratz A, Reuss D et al (2016) Next-generation sequencing in routine brain tumor diagnostics enables an integrated diagnosis and identifies actionable targets. Acta Neuropathol 131:903–910. 10.1007/s00401-015-1519-8 DOI: 10.1007/s00401-015-1519-8
Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Mahesparan R, Molthoff C et al (2006) Angiogenesis-independent tumor growth mediated by stem-like cancer cells. Proc Natl Acad Sci USA 103:16466–16471. 10.1073/pnas.0607668103 DOI: 10.1073/pnas.0607668103
Seshan VE, A O (2019) DNAcopy: DNA copy number data analysis. R package version 1600
Shen Y, Grisdale CJ, Islam SA, Bose P, Lever J, Zhao EY et al (2019) Comprehensive genomic profiling of glioblastoma tumors, BTICs, and xenografts reveals stability and adaptation to growth environments. Proc Natl Acad Sci USA 116:19098–19108. 10.1073/pnas.1813495116 DOI: 10.1073/pnas.1813495116
Shirahata M, Ono T, Stichel D, Schrimpf D, Reuss DE, Sahm F et al (2018) Novel, improved grading system(s) for IDH-mutant astrocytic gliomas. Acta Neuropathol 136:153–166. 10.1007/s00401-018-1849-4 DOI: 10.1007/s00401-018-1849-4
Smyth G (2005) Limma: linear models for microarray data. In: Springer (ed) Bioinformatics and Computational Biology Solutions using R and Bioconductor. New York, pp 397–420
Sottoriva A, Spiteri I, Piccirillo SG, Touloumis A, Collins VP, Marioni JC et al (2013) Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci USA 110:4009–4014. 10.1073/pnas.1219747110 DOI: 10.1073/pnas.1219747110
Stieber D, Golebiewska A, Evers L, Lenkiewicz E, Brons NH, Nicot N et al (2014) Glioblastomas are composed of genetically divergent clones with distinct tumourigenic potential and variable stem cell-associated phenotypes. Acta Neuropathol 127:203–219. 10.1007/s00401-013-1196-4 DOI: 10.1007/s00401-013-1196-4
Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd et al (2019) Comprehensive integration of single-cell data. Cell 177(1888–1902):e1821. 10.1016/j.cell.2019.05.031 DOI: 10.1016/j.cell.2019.05.031
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996. 10.1056/NEJMoa043330
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA et al (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 102:15545–15550. 10.1073/pnas.0506580102 DOI: 10.1073/pnas.0506580102
Szerlip NJ, Pedraza A, Chakravarty D, Azim M, McGuire J, Fang Y et al (2012) Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci USA 109:3041–3046. 10.1073/pnas.1114033109 DOI: 10.1073/pnas.1114033109
Talasila KM, Soentgerath A, Euskirchen P, Rosland GV, Wang J, Huszthy PC et al (2013) EGFR wild-type amplification and activation promote invasion and development of glioblastoma independent of angiogenesis. Acta Neuropathol 125:683–698. 10.1007/s00401-013-1101-1 DOI: 10.1007/s00401-013-1101-1
Talevich E, Shain AH, Botton T, Bastian BC (2016) CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS Comput Biol 12:e1004873. 10.1371/journal.pcbi.1004873 DOI: 10.1371/journal.pcbi.1004873
Tateishi K, Nakamura T, Juratli TA, Williams EA, Matsushita Y, Miyake S et al (2019) PI3K/AKT/mTOR pathway alterations promote malignant progression and xenograft formation in oligodendroglial tumors. Clin Cancer Res 25:4375–4387. 10.1158/1078-0432.CCR-18-4144 DOI: 10.1158/1078-0432.CCR-18-4144
Teschendorff AE, Marabita F, Lechner M, Bartlett T, Tegner J, Gomez-Cabrero D et al (2013) A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics 29:189–196. 10.1093/bioinformatics/bts680 DOI: 10.1093/bioinformatics/bts680
Tew BY, Legendre C, Schroeder MA, Triche T, Gooden GC, Huang Y et al (2019) Patient-derived xenografts of central nervous system metastasis reveal expansion of aggressive minor clones. Neuro Oncol. 10.1093/neuonc/noz137 DOI: 10.1093/neuonc/noz137
Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH et al (2018) The immune landscape of cancer. Immunity 48:812–830 e814. 10.1016/j.immuni.2018.03.023
Torsvik A, Stieber D, Enger PO, Golebiewska A, Molven A, Svendsen A et al (2014) U-251 revisited: genetic drift and phenotypic consequences of long-term cultures of glioblastoma cells. Cancer Med 3:812–824. 10.1002/cam4.219 DOI: 10.1002/cam4.219
Unruh D, Mirkov S, Wray B, Drumm M, Lamano J, Li YD et al (2019) Methylation-dependent tissue factor suppression contributes to the reduced malignancy of IDH1-mutant gliomas. Clin Cancer Res 25:747–759. 10.1158/1078-0432.CCR-18-1222 DOI: 10.1158/1078-0432.CCR-18-1222
Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A et al (2013) From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protocols Bioinformatics 43:111011–111033. 10.1002/0471250953.bi1110s43
Vaubel RA, Tian S, Remonde D, Schroeder MA, Mladek AC, Kitange GJ et al (2020) Genomic and phenotypic characterization of a broad panel of patient-derived xenografts reflects the diversity of glioblastoma. Clin Cancer Res 26:1094–1104. 10.1158/1078-0432.CCR-19-0909 DOI: 10.1158/1078-0432.CCR-19-0909
Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD et al (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer CELL 17:98–110. 10.1016/j.ccr.2009.12.020
Wakimoto H, Mohapatra G, Kanai R, Curry WT Jr, Yip S, Nitta M et al (2012) Maintenance of primary tumor phenotype and genotype in glioblastoma stem cells. Neuro Oncol 14:132–144. 10.1093/neuonc/nor195 DOI: 10.1093/neuonc/nor195
Wakimoto H, Tanaka S, Curry WT, Loebel F, Zhao D, Tateishi K et al (2014) Targetable signaling pathway mutations are associated with malignant phenotype in IDH-mutant gliomas. Clin Cancer Res 20:2898–2909. 10.1158/1078-0432.CCR-13-3052 DOI: 10.1158/1078-0432.CCR-13-3052
Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S et al (2016) Clonal evolution of glioblastoma under therapy. Nat Genet 48:768–776. 10.1038/ng.3590 DOI: 10.1038/ng.3590
Wang J, Miletic H, Sakariassen PO, Huszthy PC, Jacobsen H, Brekka N et al (2009) A reproducible brain tumour model established from human glioblastoma biopsies. BMC Cancer 9:465. 10.1186/1471-2407-9-465 DOI: 10.1186/1471-2407-9-465
Wang Q, Hu B, Hu X, Kim H, Squatrito M, Scarpace L et al (2017) Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell 32(42–56):e46. 10.1016/j.ccell.2017.06.003 DOI: 10.1016/j.ccell.2017.06.003
Weng Q, Wang J, Wang J, He D, Cheng Z, Zhang F et al (2019) Single-cell transcriptomics uncovers glial progenitor diversity and cell fate determinants during development and gliomagenesis. Cell Stem Cell 24(707–723):e708. 10.1016/j.stem.2019.03.006 DOI: 10.1016/j.stem.2019.03.006
Williams JA (2018) Using PDX for preclinical cancer drug discovery: the evolving field. J Clin Med. 10.3390/jcm7030041 DOI: 10.3390/jcm7030041
Woo XY, Giordano J, Srivastava A, Zhao Z-M, Lloyd MW, de Bruijn R et al (2019) Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts. bioRxiv:861393. 10.1101/861393
Yang RR, Shi ZF, Zhang ZY, Chan AK, Aibaidula A, Wang WW et al (2019) IDH-mutant lower-grade (WHO Grades II/III) astrocytomas can be stratified for risk by CDKN2A, CDK4 and PDGFRA copy number alterations. Brain Pathol. 10.1111/bpa.12801 DOI: 10.1111/bpa.12801
Zhai B, Steino A, Bacha J, Brown D, Daugaard M (2018) Dianhydrogalactitol induces replication-dependent DNA damage in tumor cells preferentially resolved by homologous recombination. Cell Death Dis 9:1016. 10.1038/s41419-018-1069-9 DOI: 10.1038/s41419-018-1069-9
Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O et al (2019) Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nature Commun 10:1523. 10.1038/s41467-019-09234-6 DOI: 10.1038/s41467-019-09234-6
This website uses cookies to improve user experience. Read more
Save & Close
Accept all
Decline all
Show detailsHide details
Cookie declaration
About cookies
Strictly necessary
Performance
Strictly necessary cookies allow core website functionality such as user login and account management. The website cannot be used properly without strictly necessary cookies.
This cookie is used by Cookie-Script.com service to remember visitor cookie consent preferences. It is necessary for Cookie-Script.com cookie banner to work properly.
Performance cookies are used to see how visitors use the website, eg. analytics cookies. Those cookies cannot be used to directly identify a certain visitor.
Used to store the attribution information, the referrer initially used to visit the website
Cookies are small text files that are placed on your computer by websites that you visit. Websites use cookies to help users navigate efficiently and perform certain functions. Cookies that are required for the website to operate properly are allowed to be set without your permission. All other cookies need to be approved before they can be set in the browser.
You can change your consent to cookie usage at any time on our Privacy Policy page.