126 results on '"Felsberg, J"'
Search Results
2. Does positive MGMT methylation outbalance the limitation of subtotal resection in glioblastoma IDH-wildtype patients?
- Author
-
Müller, M, Staub-Bartelt, F, Ehrmann, J, Hänggi, D, Sabel, M, Felsberg, J, Rapp, M, Müller, M, Staub-Bartelt, F, Ehrmann, J, Hänggi, D, Sabel, M, Felsberg, J, and Rapp, M
- Published
- 2022
3. Establishment of cell culture models from intracerebral carcinoma metastases for personalised preclinical evaluation of novel therapeutic approaches
- Author
-
Jeising, S, Steinmann, J, Felsberg, J, Sohn, D, Kaulich, K, Reifenberger, G, Hänggi, D, Rapp, M, Sabel, M, Nickel, AC, Jeising, S, Steinmann, J, Felsberg, J, Sohn, D, Kaulich, K, Reifenberger, G, Hänggi, D, Rapp, M, Sabel, M, and Nickel, AC
- Published
- 2022
4. Impact of 18F-FET PET imaging to differentiate tumour progression vs therapy induced changes following immunotherapy with DC vaccination in glioblastoma patients
- Author
-
Rapp, M, Datsi, A, Felsberg, J, Quente, C, Galldiks, N, Langen, KJ, Sorg, RV, Sabel, M, Rapp, M, Datsi, A, Felsberg, J, Quente, C, Galldiks, N, Langen, KJ, Sorg, RV, and Sabel, M
- Published
- 2022
5. 18F-FET PET imaging following immunotherapy with DC vaccination in glioblastoma patients
- Author
-
Rapp, M., Stamtsis-Datsi, A., Felsberg, J., Galldiks, N., Langen, K.-J., Dorf, R., and Sabel, M.
- Published
- 2021
- Full Text
- View/download PDF
6. Tumour-infiltrating effector T-cells and false-positive identification of tumour progression by MRI and 18FET-PET after dendritic cell vaccination in newly-diagnosed glioblastoma patients
- Author
-
Rapp, M, Datsi, A, Felsberg, J, Galldiks, N, Kamp, MA, Sorg, RV, and Sabel, M
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Objective: In the phase II GlioVax trial, patients with newly diagnosed glioblastoma are treated with dendritic cell vaccination as add-on to standard radiochemotherapy after fluorescence-guided surgery. Currently, it is not clear, whether dendritic cell vaccination results in effector T cell infiltration[for full text, please go to the a.m. URL], 71. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC), 9. Joint Meeting mit der Japanischen Gesellschaft für Neurochirurgie
- Published
- 2020
- Full Text
- View/download PDF
7. Integrative genomics approach provides insight into molecular characteristics of pediatric glioblastoma
- Author
-
Sturm, D., Remke, M., Rizhova, M., Witt, H., Jacob, K., Milde, T., Witt, O., Frühwald, Michael C., Van Meter, T. E., Kramm, C., Reifenberger, G., Felsberg, J., Deimling, Andreas von, Hartmann, C., Kulozik, A., Korshunov, A., Jabado, N., Lichter, P., and Pfister, S. M.
- Published
- 2020
8. Autologous tumour-lysate loaded dendritic cell vaccination alters the phenotype of tumour-infiltrating lymphocytes in newly diagnosed glioblastoma patients
- Author
-
Datsi, A, Rapp, M, Felsberg, J, Galldiks, N, Kamp, MA, Hänggi, D, Sorg, RV, Sabel, M, Datsi, A, Rapp, M, Felsberg, J, Galldiks, N, Kamp, MA, Hänggi, D, Sorg, RV, and Sabel, M
- Published
- 2021
9. 18F-FET PET imaging following immunotherapy with DC vaccination in glioblastoma patients
- Author
-
Rapp, M, Datsi, A, Felsberg, J, Galldiks, N, Langen, KJ, Kamp, MA, Sorg, RV, Sabel, M, Rapp, M, Datsi, A, Felsberg, J, Galldiks, N, Langen, KJ, Kamp, MA, Sorg, RV, and Sabel, M
- Published
- 2021
10. OC-0322: 4-miRNA signature and MGMT promoter methylation improve risk stratification in glioblastoma.
- Author
-
Unger, K., primary, Fleischmann, D.F., additional, Ruf, V., additional, Felsberg, J., additional, Piehlmaier, D., additional, Samaga, D., additional, Heß, J., additional, Mittelbronn, M., additional, Lauber, K., additional, Budach, W., additional, Sabel, M., additional, Rödel, C., additional, Reifenberger, G., additional, Herms, J., additional, Tonn, J., additional, Zitzelsberger, H., additional, Belka, C., additional, and Niyazi, M., additional
- Published
- 2020
- Full Text
- View/download PDF
11. 360O Telomerase reverse transcriptase (TERT) promoter mutation and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation-mediated sensitivity to temozolomide in IDH-wildtype glioblastoma: Is there a link?
- Author
-
Weller, M., primary, Gramatzki, D., additional, Felsberg, J., additional, Hentschel, B., additional, Wolter, M., additional, Schackert, G., additional, Westphal, M., additional, Regli, L., additional, Thon, N., additional, Tatagiba, M., additional, Wick, W., additional, Schlegel, U., additional, Krex, D., additional, Roth, P., additional, Rushing, E., additional, Pietsch, T., additional, von Deimling, A., additional, Sabel, M., additional, Loeffler, M., additional, and Reifenberger, G., additional
- Published
- 2020
- Full Text
- View/download PDF
12. Prediction of survival in patients with IDH-wildtype astrocytic gliomas using dynamic O-(2-[ 18 F]-fluoroethyl)-L-tyrosine PET
- Author
-
Bauer, E. K, additional, Stoffels, G, additional, Blau, T, additional, Reifenberger, G, additional, Felsberg, J, additional, Werner, JM, additional, Lohmann, P, additional, Rosen, J, additional, Ceccon, G, additional, Tscherpel, C, additional, Rapp, M, additional, Sabel, M, additional, Filss, CP, additional, Shah, NJ, additional, Neumaier, B, additional, Fink, GR, additional, Langen, KJ, additional, and Galldiks, N, additional
- Published
- 2020
- Full Text
- View/download PDF
13. Entwicklung, Implementierung und Evaluation eines interdisziplinären Inverted Classrooms über Hirntumoren: Bericht über Entwicklungsprozess
- Author
-
Malzkorn, B., Felsberg, J., Kamp, M., Hofer, M., and Rotthoff, T.
- Subjects
ddc: 610 ,ddc:610 ,610 Medical sciences ,Medicine - Abstract
Problemstellung/Ziele: In einem durch Vorlesungen geprägten und von Studierenden kritisierten Studienblock sollten interdisziplinäre, multimodale, individualisierte Lernkonzepte entwickelt werden, in denen Studierende theoretische Kenntnisse im Eigenstudium erwerben und die Präsenzzeit[zum vollständigen Text gelangen Sie über die oben angegebene URL], Jahrestagung der Gesellschaft für Medizinische Ausbildung (GMA)
- Published
- 2019
14. Decoding the microstructural correlate of in-vivo brain atrophy patterns in corticobasal syndrome
- Author
-
Dinkelbach, L., Südmeyer, M., Hartmann, CJ, Roeber, S., Arzberger, T., Felsberg, J., Ferrea, S., Moldovan, AS., Schnitzler, A., and Caspers, Svenja
- Published
- 2019
15. OS2.2 Chemotherapy for spinal gliomas in adults
- Author
-
Gramatzki, D, primary, Felsberg, J, additional, Bähr, O, additional, Hentschel, B, additional, Westphal, M, additional, Schackert, G, additional, Tonn, J C, additional, Herrlinger, U, additional, Löffler, M, additional, Pietsch, T, additional, Steinbach, J, additional, Reifenberger, G, additional, Roth, P, additional, and Weller, M, additional
- Published
- 2019
- Full Text
- View/download PDF
16. P14.108 Bevacizumab versus alkylating chemotherapy in recurrent glioblastoma
- Author
-
Seystahl, K, primary, Hentschel, B, additional, Loew, S, additional, Gramatzki, D, additional, Felsberg, J, additional, Herrlinger, U, additional, Westphal, M, additional, Schackert, G, additional, Thon, N, additional, Schlegel, U, additional, Tatagiba, M, additional, Pietsch, T, additional, Reifenberger, G, additional, Löffler, M, additional, Wick, W, additional, and Weller, M, additional
- Published
- 2019
- Full Text
- View/download PDF
17. P14.18 Prognostic value of serial dynamic O-(2-[18F]-fluoroethyl)-L-tyrosine PET in patients with non-resectable malignant glioma undergoing chemoradiation
- Author
-
Rosen, J, primary, Stoffels, G, additional, Lohmann, P, additional, Bauer, E K, additional, Werner, J, additional, Rapp, M, additional, Felsberg, J, additional, Fink, G R, additional, Langen, K, additional, and Galldiks, N, additional
- Published
- 2019
- Full Text
- View/download PDF
18. Catch me if you can! Novel immune escape mechanisms in melanoma metastases of the brain
- Author
-
Herrera-Rios, D., Schmitt, V., Herz, J., Hinze, D., Piercianek, D., Kato, M., Felsberg, J., Singer, B., Sure, U., Reifenberger, G., Dirk Schadendorf, Hoelzel, M., and Helfrich, I.
- Subjects
Medizin - Published
- 2018
19. Entwicklung, Implementierung und Evaluation eines interdisziplinären Inverted Classrooms über Hirntumoren [Bericht über Entwicklungsprozess]
- Author
-
Malzkorn, B, Felsberg, J, Kamp, M, Hofer, M, Rotthoff, T, Malzkorn, B, Felsberg, J, Kamp, M, Hofer, M, and Rotthoff, T
- Published
- 2018
20. Gliomatosis cerebri: molecular pathology and clinical course
- Author
-
Herrlinger, U, Felsberg, J, Küker, W, Bornemann, A, Plasswilm, L, Knobbe, CB, Strik, H, Wick, W, Meyermann, R, Dichgans, J, Bamberg, M, Reifenberger, G, and Weller, M
- Subjects
neoplasms - Abstract
Gliomatosis cerebri is a rare, diffusely growing neuroepithelial tumor characterized by extensive brain infiltration involving more than two cerebral lobes. Among 13 patients with gliomatosis cerebri (median age, 46 years), biopsies showed features of diffuse astrocytoma (n = 4), oligoastrocytoma (n = 1), anaplastic astrocytoma (n = 5), anaplastic oligoastrocytoma (n = 1), or glioblastoma (n = 2). Molecular genetic investigation showed TP53 mutations in three of seven tumors and both PTEN mutation and epidermal growth factor receptor overexpression in one tumor. Amplification of CDK4 or MDM2 or homozygous deletion of CDKN2A was not detected. Three of 10 patients receiving radiotherapy showed a partial response (one patient) or had stable disease (two patients) lasting for more than 1 year. Four of six patients treated with procarbazine, carmustine, vincristine chemotherapy demonstrated partial remission (one patient), minor response (two patients), or stable disease (one patient). Median survival time from diagnosis was 14 months (range, 4-91+ months). Infratentorial involvement was associated with shorter survival. We conclude that (1) the molecular genetic alterations in gliomatosis cerebri resemble those in diffuse astrocytomas; (2) the prognosis of gliomatosis cerebri is variable but for at least 50% of patients as poor as for glioblastoma; and (3) some patients respond to radiotherapy and/or procarbazine, carmustine, vincristine chemotherapy.
- Published
- 2016
- Full Text
- View/download PDF
21. P03.21 Casp9 germline mutation in a family with multiple brain tumors
- Author
-
Ronellenfitsch, M. W., primary, Oh, J., additional, Satomi, K., additional, Sumi, K., additional, Harter, P. N., additional, Steinbach, J. P., additional, Felsberg, J., additional, Capper, D., additional, Voegele, C., additional, Schittenhelm, J., additional, Mittelbronn, M., additional, and Ohgaki, H., additional
- Published
- 2017
- Full Text
- View/download PDF
22. New Brain Tumor Entities Emerge from Molecular Classification of CNS-PNETs
- Author
-
Sturm, D., Orr, B.A., Toprak, U.H., Hovestadt, V., Jones, D.T., Capper, D., Sill, M., Buchhalter, I., Northcott, P.A., Leis, I., Ryzhova, M., Koelsche, C., Pfaff, E., Allen, S.J., Balasubramanian, G., Worst, B.C., Pajtler, K.W., Brabetz, S., Johann, P.D., Sahm, F., Reimand, J., Mackay, A., Carvalho, D.M., Remke, M., Phillips, J.J., Perry, A., Cowdrey, C., Drissi, R., Fouladi, M., Giangaspero, F., Lastowska, M., Grajkowska, W., Scheurlen, W., Pietsch, T., Hagel, C., Gojo, J., Lotsch, D., Berger, W., Slavc, I., Haberler, C., Jouvet, A., Holm, S., Hofer, S., Prinz, M., Keohane, C., Fried, I., Mawrin, C., Scheie, D., Mobley, B.C., Schniederjan, M.J., Santi, M., Buccoliero, A.M., Dahiya, S., Kramm, C.M., Bueren, A.O. von, Hoff, K. von, Rutkowski, S., Herold-Mende, C., Fruhwald, M.C., Milde, T., Hasselblatt, M., Wesseling, P., Rossler, J., Schuller, U., Ebinger, M., Schittenhelm, J., Frank, S., Grobholz, R., Vajtai, I., Hans, V., Schneppenheim, R., Zitterbart, K., Collins, V.P., Aronica, E., Varlet, P., Puget, S., Dufour, C., Grill, J., Figarella-Branger, D., Wolter, M., Schuhmann, M.U., Shalaby, T., Grotzer, M., Meter, T. van, Monoranu, C.M., Felsberg, J., Reifenberger, G., Snuderl, M., Forrester, L.A., Koster, J., Versteeg, R., Volckmann, R., Sluis, P. van, Wolf, S., Mikkelsen, T., Gajjar, A., Aldape, K., Moore, A.S., Taylor, M.D., Jones, C., et al., Sturm, D., Orr, B.A., Toprak, U.H., Hovestadt, V., Jones, D.T., Capper, D., Sill, M., Buchhalter, I., Northcott, P.A., Leis, I., Ryzhova, M., Koelsche, C., Pfaff, E., Allen, S.J., Balasubramanian, G., Worst, B.C., Pajtler, K.W., Brabetz, S., Johann, P.D., Sahm, F., Reimand, J., Mackay, A., Carvalho, D.M., Remke, M., Phillips, J.J., Perry, A., Cowdrey, C., Drissi, R., Fouladi, M., Giangaspero, F., Lastowska, M., Grajkowska, W., Scheurlen, W., Pietsch, T., Hagel, C., Gojo, J., Lotsch, D., Berger, W., Slavc, I., Haberler, C., Jouvet, A., Holm, S., Hofer, S., Prinz, M., Keohane, C., Fried, I., Mawrin, C., Scheie, D., Mobley, B.C., Schniederjan, M.J., Santi, M., Buccoliero, A.M., Dahiya, S., Kramm, C.M., Bueren, A.O. von, Hoff, K. von, Rutkowski, S., Herold-Mende, C., Fruhwald, M.C., Milde, T., Hasselblatt, M., Wesseling, P., Rossler, J., Schuller, U., Ebinger, M., Schittenhelm, J., Frank, S., Grobholz, R., Vajtai, I., Hans, V., Schneppenheim, R., Zitterbart, K., Collins, V.P., Aronica, E., Varlet, P., Puget, S., Dufour, C., Grill, J., Figarella-Branger, D., Wolter, M., Schuhmann, M.U., Shalaby, T., Grotzer, M., Meter, T. van, Monoranu, C.M., Felsberg, J., Reifenberger, G., Snuderl, M., Forrester, L.A., Koster, J., Versteeg, R., Volckmann, R., Sluis, P. van, Wolf, S., Mikkelsen, T., Gajjar, A., Aldape, K., Moore, A.S., Taylor, M.D., Jones, C., and et al.
- Abstract
Item does not contain fulltext, Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors.
- Published
- 2016
23. Combined alterations in MAPK pathway genes, CDKN2A/B and ATRX characterize anaplastic pilocytic astrocytoma
- Author
-
Kratz, A, Sahm, F, Schrimpf, D, Jones, DT, Reuss, D, Koelsche, C, Huang, K, Wefers, AK, Hovestadt, V, Gramatzki, D, Felsberg, J, Koch, A, Thomale, UW, Reifenberger, G, Becker, A, Hans, V, Prinz, M, Staszewski, O, Acker, T, Dohmen-Scheufler, H, Hartmann, C, Mueller, W, Tuffaha, MSA, Paulus, W, Heß, K, Brokinkel, B, Schittenhelm, J, Monoranu, CM, Kessler, AF, Loehr, M, Buslei, R, Deckert, M, Mawrin, C, Kohlhof, P, Hewer, E, Olar, A, Rodriguez, F, Giannini, C, NageswaraRao, AA, Weller, M, Pohl, U, Brandner, S, Pfister, SM, von Deimling, A, Capper, D, Kratz, A, Sahm, F, Schrimpf, D, Jones, DT, Reuss, D, Koelsche, C, Huang, K, Wefers, AK, Hovestadt, V, Gramatzki, D, Felsberg, J, Koch, A, Thomale, UW, Reifenberger, G, Becker, A, Hans, V, Prinz, M, Staszewski, O, Acker, T, Dohmen-Scheufler, H, Hartmann, C, Mueller, W, Tuffaha, MSA, Paulus, W, Heß, K, Brokinkel, B, Schittenhelm, J, Monoranu, CM, Kessler, AF, Loehr, M, Buslei, R, Deckert, M, Mawrin, C, Kohlhof, P, Hewer, E, Olar, A, Rodriguez, F, Giannini, C, NageswaraRao, AA, Weller, M, Pohl, U, Brandner, S, Pfister, SM, von Deimling, A, and Capper, D
- Published
- 2016
24. Paediatric Diffuse Glioma in Constitutional Mismatch Repair Deficiency Syndrome (CMMRD) - Report of Two Affected Sisters
- Author
-
Hans, VH, Leis, A, Angrés, M, Felsberg, J, Erdlenbruch, B, Muleris, M, Knappe, UJ, Hans, VH, Leis, A, Angrés, M, Felsberg, J, Erdlenbruch, B, Muleris, M, and Knappe, UJ
- Published
- 2016
25. Chemotherapy for intracranial ependymoma in adults
- Author
-
Gramatzki, Dorothee, Roth, P, Felsberg, J, Hofer, S, Rushing, E J, Hentschel, B, Westphal, M, Krex, D, Simon, M, Schnell, O, Wick, W, Reifenberger, G, Weller, M, Gramatzki, Dorothee, Roth, P, Felsberg, J, Hofer, S, Rushing, E J, Hentschel, B, Westphal, M, Krex, D, Simon, M, Schnell, O, Wick, W, Reifenberger, G, and Weller, M
- Abstract
BACKGROUND Ependymal tumors in adults are rare, accounting for less than 4 % of primary tumors of the central nervous system in this age group. The low prevalence of intracranial ependymoma in adults limits the ability to perform clinical trials. Therefore, treatment decisions are based on small, mostly retrospective studies and the role of chemotherapy has remained unclear. METHODS We performed a retrospective study on 17 adult patients diagnosed with intracranial World Health Organisation grade II or III ependymoma, who were treated with chemotherapy at any time during the disease course. Benefit from chemotherapy was estimated by applying Macdonald criteria. Progression-free (PFS) and overall survival (OS) were calculated from start of chemotherapy, using the Kaplan-Meier method. RESULTS Eleven patients had supratentorial and 6 infratentorial tumors. Ten patients were treated with temozolomide (TMZ), 3 with procarbazine/lomustine/vincristine (PCV), 3 with platinum-based chemotherapy and 1 patient received epirubicin/ifosfamide. Response rates were as follows: TMZ 8/10 stable disease; PCV 3/3 stable disease; platinum-based chemotherapy 1/3 partial response; epirubicin/ifosfamide 1/1 complete response. PFS rates at 6, 12 and 24 months were 52.9, 35.3 and 23.5 %. OS rates at 6, 12 and 24 months were 82.4, 82.4 and 70.1 %. There was no indication for a favourable prognostic role of O (6) -methylguanyl-DNA-methyltransferase (MGMT) promoter methylation which was detected in 3/12 investigated tumors. CONCLUSIONS Survival outcomes in response to chemotherapy in adult intracranial ependymoma patients vary substantially, but individual patients may respond to any kind of chemotherapy. There were too few patients to compare survival data between chemotherapeutic subgroups.
- Published
- 2016
26. High throughput screening of novel histone deacetylase inhibitors for epigenetic therapy of primary brain tumors
- Author
-
Marquardt, V, primary, Hansen, FK, additional, Felsberg, J, additional, Reifenberger, G, additional, Borkhardt, A, additional, Kurz, T, additional, and Remke, M, additional
- Published
- 2016
- Full Text
- View/download PDF
27. P08.48 Combined alterations in MAPK pathway genes, CDKN2A/B and ATRX characterize anaplastic pilocytic astrocytoma
- Author
-
Kratz, A., primary, Sahm, F., additional, Schrimpf, D., additional, Jones, D. T., additional, Reuss, D. E., additional, Schittenhelm, J., additional, Gramatzki, D., additional, Felsberg, J., additional, von Deimling, A., additional, and Capper, D., additional
- Published
- 2016
- Full Text
- View/download PDF
28. Epigenetische Inaktivierung des Tumor-Suppressor Kandidatengens PROX1 beim sporadischen Mammakarzinom
- Author
-
Versmold, B, Waha, A, Felsberg, J, Pietsch, T, Mallmann, P, and Schmutzler, RK
- Published
- 2024
- Full Text
- View/download PDF
29. Epigenetic silencing of the candidate tumor suppressor gene PROX1 in sporadic breast cancer
- Author
-
Versmold, B, Waha, A, Felsberg, J, Köhler, J, Ehrentraut, D, Pietsch, T, and Schmutzler, RK
- Published
- 2024
- Full Text
- View/download PDF
30. Prediction of survival in patients with IDH-wildtype astrocytic gliomas using dynamic O-(2-[18 F]-fluoroethyl)-L-tyrosine PET.
- Author
-
Bauer, E. K, Stoffels, G, Blau, T, Reifenberger, G, Felsberg, J, Werner, JM, Lohmann, P, Rosen, J, Ceccon, G, Tscherpel, C, Rapp, M, Sabel, M, Filss, CP, Shah, NJ, Neumaier, B, Fink, GR, Langen, KJ, and Galldiks, N
- Published
- 2020
- Full Text
- View/download PDF
31. P14.18 Prognostic value of serial dynamic O-(2-[18F]-fluoroethyl)-L-tyrosine PET in patients with non-resectable malignant glioma undergoing chemoradiation.
- Author
-
Rosen, J, Stoffels, G, Lohmann, P, Bauer, E K, Werner, J, Rapp, M, Felsberg, J, Fink, G R, Langen, K, and Galldiks, N
- Published
- 2019
- Full Text
- View/download PDF
32. P01.013 Long-term analyses of the NOA-08 randomized phase III trial of temozolomide versus radiotherapy for elderly patients with malignant astrocytomas.
- Author
-
Wick, A, Kessler, T, Platten, M, Meisner, C, Bamberg, M, Herrlinger, U, Felsberg, J, Weyerbrock, A, Seidel, C, and Steinbach, J P
- Published
- 2018
- Full Text
- View/download PDF
33. Sellar region atypical teratoid/rhabdoid tumors (ATRT) in adults display DNA methylation profiles of the ATRT-MYC subgroup
- Author
-
Rabea Wagener, Caterina Giannini, Jack M Raisanen, Michael C. Frühwald, Kazunori Arita, Florian Oyen, Gerald F. Reis, Guido Reifenberger, Stefan M. Pfister, Rolf Buslei, Martin Hasselblatt, Sumihito Nobusawa, Reiner Siebert, Reinhard Schneppenheim, Jörg Felsberg, David Capper, Pascal Johann, Werner Paulus, Arie Perry, Susanne Bens, Marcel Kool, Abbas Agaimy, Johann P.D., Bens S., Oyen F., Wagener R., Giannini C., Perry A., Raisanen J.M., Reis G.F., Nobusawa S., Arita K., Felsberg J., Reifenberger G., Agaimy A., Buslei R., Capper D., Pfister S.M., Schneppenheim R., Siebert R., Fruhwald M.C., Paulus W., Kool M., and Hasselblatt M.
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Biology ,Compound heterozygosity ,pituitary ,Pathology and Forensic Medicine ,Epigenesis, Genetic ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,medicine ,Biomarkers, Tumor ,SMARCB1/INI1 ,Humans ,Point Mutation ,Age Factor ,Genetic Predisposition to Disease ,Pituitary Neoplasms ,Epigenetics ,Pituitary Neoplasm ,Allele ,SMARCB1 ,Rhabdoid Tumor ,Aged ,medicine.diagnostic_test ,Point mutation ,Age Factors ,Teratoma ,atypical teratoid/rhabdoid tumor ,SMARCB1 Protein ,DNA Methylation ,Middle Aged ,medicine.disease ,Phenotype ,030220 oncology & carcinogenesis ,DNA methylation ,Atypical teratoid rhabdoid tumor ,outcome ,DNA methylation profiling ,Surgery ,Female ,Anatomy ,030217 neurology & neurosurgery ,Gene Deletion ,Fluorescence in situ hybridization ,Human - Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is a highly malignant brain tumor predominantly encountered in infants. Mutations of the SMARCB1 gene are the characteristic genetic lesion. A small group of ATRT stands out clinically, because these tumors are located in the sellar region of adults. To investigate if sellar region ATRT in adults represents a molecular distinct entity, we characterized molecular alterations in 7 sellar region ATRTs in adults as compared with 150 pediatric ATRTs and 47 pituitary adenomas using SMARCB1 sequencing, multiplex ligation-dependent probe amplification and fluorescence in situ hybridization as well as DNA methylation profiling. The median age of the 6 female and 1 male patients was 56 years. On histopathologic examination, all tumors were malignant rhabdoid tumors showing loss of SMARCB1/INI1 protein expression. Two cases displayed compound heterozygous SMARCB1 point mutations, 3 cases showed heterozygous SMARCB1 deletions with point mutations of the other allele and 1 case a homozygous SMARCB1 deletion; in 1 case, underlying SMARCB1 alterations could not be identified. On unsupervised hierarchical cluster analysis of DNA methylation profiles, sellar region ATRTs did not form a distinct group, but clustered with ATRT-MYC, 1 of 3 recently described molecular subgroups of ATRT. On analysis of DNA methylation array intensity data, only 1 sellar region ATRT showed characteristic features of pediatric ATRT-MYC, that is, major copy number losses affecting the SMARCB1 region. In conclusion, these results suggest that sellar region ATRTs in adults form a clinically distinct entity with a different mutational spectrum, but epigenetic similarities with pediatric ATRTs of the ATRT-MYC subgroup.
- Published
- 2020
34. Anaplastic astrocytoma with piloid features, a novel molecular class of IDH wildtype glioma with recurrent MAPK pathway, CDKN2A/B and ATRX alterations
- Author
-
Camelia M. Monoranu, Andreas von Deimling, Albert J. Becker, Joerg Felsberg, Jens Schittenhelm, Martina Deckert, Marco Prinz, Rolf Buslei, Till Acker, Katharina Heß, Ute Pohl, Volker Hovestadt, Wolf Mueller, Patricia Kohlhof, Dorothee Gramatzki, Muin S. A. Tuffaha, Amulya NageswaraRao, Andrey Korshunov, Benjamin Brokinkel, Daniel Schrimpf, David T.W. Jones, Annekathrin Reinhardt, Ulrich W. Thomale, Werner Paulus, Ekkehard Hewer, Christian Koelsche, Christian Mawrin, Damian Stichel, Ori Staszewski, Wolfgang Wick, David E. Reuss, Almuth F. Kessler, Caterina Giannini, Annika K. Wefers, Michael Platten, Martin Sill, Daniel Hänggi, Kristin Huang, Christian Hartmann, Adriana Olar, David Capper, Volkmar Hans, Andreas Unterberg, Zane Jaunmuktane, Sebastian Brandner, Nuno Miguel Nunes, Christel Herold-Mende, Felix Sahm, Uri Tabori, Guido Reifenberger, Arend Koch, Mario Loehr, Michael Weller, Hildegard Dohmen, Stefan M. Pfister, Fausto J. Rodriguez, Reinhardt A., Stichel D., Schrimpf D., Sahm F., Korshunov A., Reuss D.E., Koelsche C., Huang K., Wefers A.K., Hovestadt V., Sill M., Gramatzki D., Felsberg J., Reifenberger G., Koch A., Thomale U.-W., Becker A., Hans V.H., Prinz M., Staszewski O., Acker T., Dohmen H., Hartmann C., Mueller W., Tuffaha M.S.A., Paulus W., Hess K., Brokinkel B., Schittenhelm J., Monoranu C.-M., Kessler A.F., Loehr M., Buslei R., Deckert M., Mawrin C., Kohlhof P., Hewer E., Olar A., Rodriguez F.J., Giannini C., NageswaraRao A.A., Tabori U., Nunes N.M., Weller M., Pohl U., Jaunmuktane Z., Brandner S., Unterberg A., Hanggi D., Platten M., Pfister S.M., Wick W., Herold-Mende C., Jones D.T.W., von Deimling A., and Capper D.
- Subjects
0301 basic medicine ,Male ,Medizin ,Kaplan-Meier Estimate ,Mitogen-Activated Protein Kinase Kinase ,Histones ,0302 clinical medicine ,CDKN2A ,Retrospective Studie ,Age Factor ,610 Medicine & health ,Child ,DNA Modification Methylases ,Aged, 80 and over ,Pilocytic astrocytoma ,Brain Neoplasms ,DNA Repair Enzyme ,Age Factors ,CDKN2A/B ,Middle Aged ,Molecular characterization ,Isocitrate Dehydrogenase ,Histone ,ATRX ,Child, Preschool ,DNA methylation ,Female ,MGMT ,Human ,Signal Transduction ,Adult ,X-linked Nuclear Protein ,IDH1 ,Adolescent ,Panel sequencing ,Biology ,Astrocytoma ,Pathology and Forensic Medicine ,BRAF ,DNA copy number alteration ,Brain Neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Glioma ,DNA Modification Methylase ,medicine ,Humans ,Gene ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Retrospective Studies ,Pilocytic astrocytoma with anaplasia ,Aged ,Mitogen-Activated Protein Kinase Kinases ,Tumor Suppressor Protein ,Methylation profile based classification ,Tumor Suppressor Proteins ,Infant ,DNA Methylation ,medicine.disease ,Anaplastic pilocytic astrocytoma ,030104 developmental biology ,DNA Repair Enzymes ,FGFR1 ,NF1 ,Mutation ,Cancer research ,570 Life sciences ,biology ,Neurology (clinical) ,030217 neurology & neurosurgery ,Anaplastic astrocytoma - Abstract
Tumors with histological features of pilocytic astrocytoma (PA), but with increased mitotic activity and additional high-grade features (particularly microvascular proliferation and palisading necrosis) have often been designated anaplastic pilocytic astrocytomas. The status of these tumors as a separate entity has not yet been conclusively demonstrated and molecular features have only been partially characterized. We performed DNA methylation profiling of 102 histologically defined anaplastic pilocytic astrocytomas. T-distributed stochastic neighbor-embedding(t-SNE) and hierarchical clustering analysis of these 102 cases against 158 reference cases from 12 glioma reference classes revealed that a subset of 83 of these tumors share a common DNA methylation profile that is distinct from the reference classes. These 83 tumors were thus denominated DNA methylation class anaplastic astrocytoma with piloid features (MC AAP). The 19 remaining tumors were distributed amongst the reference classes, with additional testing confirming the molecular diagnosis in most cases. Median age of patients with MC AAP was 41.5years. The most frequent localization was the posterior fossa (74%). Deletions of CDKN2A/B (66/83, 80%), MAPK pathway gene alterations (49/65, 75%, most frequently affecting NF1, followed by BRAF and FGFR1) and mutations of ATRX or loss of ATRX expression (33/74, 45%) were the most common molecular alterations. All tumors were IDH1/2 wildtype. The MGMT promoter was methylated in 38/83 tumors (45%). Outcome analysis confirmed an unfavorable clinical course in comparison to PA, but better than IDH wildtype glioblastoma. In conclusion, we show that a subset of histologically defined anaplastic pilocytic astrocytomas forms a separate DNA methylation cluster, harbors recurrent alterations in MAPK pathway genes in combination with alterations of CDKN2A/B and ATRX, affects patients who are on average older than those diagnosed with PA and has an intermediate clinical outcome.
- Published
- 2018
- Full Text
- View/download PDF
35. IDH-mutant astrocytomas with primitive neuronal component have a distinct methylation profile and a higher risk of leptomeningeal spread.
- Author
-
Hinz F, Friedel D, Korshunov A, Ippen FM, Bogumil H, Banan R, Brandner S, Hasselblatt M, Boldt HB, Dirse V, Dohmen H, Aronica E, Brodhun M, Broekman MLD, Capper D, Cherkezov A, Deng MY, van Dis V, Felsberg J, Frank S, French PJ, Gerlach R, Göbel K, Goold E, Hench J, Kantelhardt S, Kohlhof-Meinecke P, Krieg S, Mawrin C, Morrison G, Mühlebner A, Ozduman K, Pfister SM, Poliani PL, Prinz M, Reifenberger G, Riemenschneider MJ, Sankowski R, Schrimpf D, Sill M, Snuderl M, Verdijk RM, Voisin MR, Wesseling P, Wick W, Reuss DE, von Deimling A, Sahm F, Maas SLN, and Suwala AK
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, Meningeal Neoplasms genetics, Meningeal Neoplasms pathology, Young Adult, Astrocytoma genetics, Astrocytoma pathology, Isocitrate Dehydrogenase genetics, Brain Neoplasms genetics, Brain Neoplasms pathology, Mutation, DNA Methylation
- Abstract
IDH-mutant astrocytomas are diffuse gliomas that are defined by characteristic mutations in IDH1 or IDH2 and do not have complete 1p/19q co-deletion. The established grading criteria include histological features of brisk mitotic activity (grade 3) and necrosis and/or microvascular proliferation (grade 4). In addition, homozygous deletion of the CDKN2A/B locus has recently been implemented as a molecular marker for grade 4 IDH-mutant astrocytomas. Here, we describe a subgroup of high-grade IDH-mutant astrocytomas characterised by a primitive neuronal component based on histology and a distinct DNA methylation profile (n = 51, ASTRO PNC). Misinterpretation as carcinoma metastasis was common, since GFAP expression was absent in the primitive neuronal component, whereas TTF-1 expression was detected in 15/19 cases (79%) based on immunohistochemistry. Apart from mutations in IDH1, TP53, and ATRX, we observed enrichment for alterations in RB1 (n = 19/51, 37%) and MYCN (n = 14/51, 27%). Homozygous CDKN2A/B deletion (n = 1/51, 2%) and CDK4 amplification (n = 3/51, 6%) were relatively rare events. Clinical (n = 31 patients) and survival data (n = 23 patients) indicate a clinical behaviour similar to other CNS WHO grade 4 IDH-mutant astrocytomas, however with an increased risk for leptomeningeal (n = 7) and extra-axial (n = 2) spread. Taken together, ASTRO PNC is defined by a distinct molecular and histological appearance that can mimic metastatic disease and typically follows an aggressive clinical course., Competing Interests: Declarations. Conflict of interest: MS is scientific advisor and shareholder of Heidelberg Epignostix and Halo Dx, and a scientific advisor of Arima Genomics, and InnoSIGN, and received research funding from Lilly USA, and Illumina USA. DC, SMP, DS, AvD and FS are shareholders of Heidelberg Epignostix. Ethical approval: Collection and analysis of corresponding tissue samples and clinical data was performed in accordance with local ethics regulations (local ethics vote S-318/2022) and in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
36. The clinical and molecular landscape of diffuse hemispheric glioma, H3 G34-mutant.
- Author
-
Le Rhun E, Bink A, Felsberg J, Gramatzki D, Brandner S, Benhamida JK, Wick A, Tonn JC, Mohme M, Tabatabai G, Capper D, Snuderl M, Razis E, Ronellenfitsch MW, Neidert N, Ng HK, Pohl U, Bale T, Quach S, Rieger D, Schüller U, Onken J, Drüschler K, Maurage CA, Regli L, Healy E, Graham M, Hortobagyi T, Paine S, Bridges L, Lausova T, Medici V, Sievers P, Schrimpf D, Wick W, Sahm F, Reifenberger G, von Deimling A, and Weller M
- Abstract
Background: Diffuse hemispheric glioma, histone 3 (H3) G34-mutant, has been newly defined in the 2021 WHO classification of central nervous system tumors. Here we sought to define the prognostic roles of clinical, neuroimaging, pathological, and molecular features of these tumors., Methods: We retrospectively assembled a cohort of 114 patients (median age 22 years) with diffuse hemispheric glioma, H3 G34-mutant, CNS WHO grade 4 and profiled the imaging, histological and molecular landscape of their tumors., Results: Compared with glioblastoma, H3 G34-mutant diffuse hemispheric gliomas exhibited less avid contrast enhancement, necrosis and edema on MRI. Comprehensive analyses of mutational and DNA copy number profiles revealed recurrent mutations in TP53 and ATRX, homozygous deletions of CDKN2A/B, and amplifications of PDGFRA, EGFR, CCND2, and MYCN. MGMT promoter methylation was detected in 79 tumors (75%); 11 tumors (13%) showed DNA copy number profiles suggestive of circumscribed deletions on 10q26.3 involving the MGMT locus. Median survival was 21.5 months. Female sex, gross total resection, and MGMT promoter methylation were positive prognostic factors on univariate analysis. Among radiological, pathological and molecular features, absence of pial invasion, and presence of microvascular proliferation and CDK6 amplification were positive prognostic factors on univariate analyses., Conclusions: This study refines the clinical and molecular landscape of H3 G34-mutant diffuse hemispheric gliomas. Dedicated trials for this novel tumor type are urgently needed., (© The Author(s) 2025. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2025
- Full Text
- View/download PDF
37. A clinically compatible in vitro drug-screening platform identifies therapeutic vulnerabilities in primary cultures of brain metastases.
- Author
-
Jeising S, Nickel AC, Trübel J, Felsberg J, Picard D, Leprivier G, Wolter M, Huynh MK, Olivera MB, Kaulich K, Häberle L, Esposito I, Klau GW, Steinmann J, Beez T, Rapp M, Sabel M, Dietrich S, Remke M, Cornelius JF, Reifenberger G, and Qin N
- Subjects
- Humans, Tumor Cells, Cultured, Drug Screening Assays, Antitumor methods, High-Throughput Nucleotide Sequencing, Female, Male, Middle Aged, High-Throughput Screening Assays methods, Brain Neoplasms drug therapy, Brain Neoplasms secondary, Brain Neoplasms pathology, Brain Neoplasms genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Purpose: Brain metastases represent the most common intracranial tumors in adults and are associated with a poor prognosis. We used a personalized in vitro drug screening approach to characterize individual therapeutic vulnerabilities in brain metastases., Methods: Short-term cultures of cancer cells isolated from brain metastasis patients were molecularly characterized using next-generation sequencing and functionally evaluated using high-throughput in vitro drug screening to characterize pharmacological treatment sensitivities., Results: Next-generation sequencing identified matched genetic alterations in brain metastasis tissue samples and corresponding short-term cultures, suggesting that short-term cultures of brain metastases are suitable models for recapitulating the genetic profile of brain metastases that may determine their sensitivity to anti-cancer drugs. Employing a high-throughput in vitro drug screening platform, we successfully screened the cultures of five brain metastases for response to 267 anticancer compounds and related drug response to genetic data. Among others, we found that targeted treatment with JAK3, HER2, or FGFR3 inhibitors showed anti-cancer effects in individual brain metastasis cultures., Conclusion: Our preclinical study provides a proof-of-concept for combining molecular profiling with in vitro drug screening for predictive evaluation of therapeutic vulnerabilities in brain metastasis patients. This approach could advance the use of patient-derived cancer cells in clinical practice and might eventually facilitate decision-making for personalized drug treatment., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
38. Corrigendum to "Zotiraciclib (TG02) for newly diagnosed glioblastoma in the elderly or for recurrent glioblastoma: The EORTC 1608 STEAM trial" [Eur J Cancer 198 (2024) 113475].
- Author
-
Le Rhun E, Gorlia T, Felsberg J, Jongen J, Maurage CA, Ducray F, Gramatzki D, Hau P, Chinot OL, Preusser M, Cartalat S, Roth P, van den Bent M, Furtner J, Collienne M, Reifenberger G, and Weller M
- Published
- 2024
- Full Text
- View/download PDF
39. Zotiraciclib (TG02) for newly diagnosed glioblastoma in the elderly or for recurrent glioblastoma: The EORTC 1608 STEAM trial.
- Author
-
Le Rhun E, Gorlia T, Felsberg J, Jongen J, Maurage CA, Ducray F, Gramatzki D, Hau P, Chinot OL, Preusser M, Cartalat S, Roth P, van den Bent M, Furtner J, Collienne M, Reifenberger G, and Weller M
- Subjects
- Humans, Aged, Temozolomide therapeutic use, Dacarbazine therapeutic use, Myeloid Cell Leukemia Sequence 1 Protein therapeutic use, Neoplasm Recurrence, Local drug therapy, Enzyme Inhibitors, Antineoplastic Agents, Alkylating therapeutic use, Glioblastoma drug therapy, Glioblastoma radiotherapy, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms radiotherapy, Heterocyclic Compounds, 4 or More Rings
- Abstract
Background: Zotiraciclib (TG02) is an oral multi-cyclin dependent kinase (CDK) inhibitor thought to inhibit tumor growth via CDK-9-dependent depletion of survival proteins such as c-MYC and MCL-1 which are frequently overexpressed in glioblastoma., Methods: EORTC 1608 (NCT03224104) (STEAM) had a three parallel group (A,B,C) phase Ib, open-label, non-randomized, multicenter design in IDH wild-type newly diagnosed glioblastoma or anaplastic astrocytoma. Groups A and B explored the maximum tolerated dose (MTD) of TG02 in elderly patients, in combination with hypofractionated radiotherapy alone (group A) or temozolomide alone (group B), according to O
6 -methylguanine DNA methyltransferase promoter methylation status determined centrally. Group C explored single agent activity of TG02 at first relapse after temozolomide chemoradiotherapy with a primary endpoint of progression-free survival at 6 months (PFS-6). Tumor expression of CDK-9, c-MYC and MCL-1 was determined by immunohistochemistry., Results: The MTD was 150 mg twice weekly in combination with radiotherapy alone (group A) or temozolomide alone (group B). Two dose-limiting toxicities were observed at 150 mg: one in group A (grade 3 seizure), one in group B (multiple grade 1 events). Main toxicities included neutropenia, gastrointestinal disorders and hepatotoxicity. PFS-6 in group C was 6.7%. CDK-9, c-MYC and MCL-1 were confirmed to be expressed and their expression was moderately cross-correlated. High protein levels of MCL-1 were associated with inferior survival., Conclusions: TG02 exhibits overlapping toxicity with alkylating agents and low single agent clinical activity in recurrent glioblastoma. The role of CDK-9 and its down-stream effectors as prognostic factors and therapeutic targets in glioblastoma warrants further study., Competing Interests: Declaration of Competing Interest ELR has received a grant research from Bristol Meyer Squibb and honoraria for lectures or advisory board from Bayer, Janssen, Leo Pharma, Pierre Fabre, Servier and Seattle Genetics. JFu received honoraria for lectures and consultation from the following for-profit companies: Novartis, Seagen. FD received honoraria for advisory board and lectures from Novocure and Servier. PH has an advisory role at BMS, Glaxo Smith Kline, MSD, Novocure, is in the speakers bureau of Lilly, medac, Novocure, Seagen and has received travel grants from Lilly, medac, Novocure, Seagen. OLC received research support from Novocure and honoraria from BMS. MP has received research grants from Pfizer and Roche and honoraria for advisory boards from Bayer. PR has received honoraria for lectures or advisory board participation from Alexion, Bristol-Myers Squibb, Boehringer Ingelheim, Debiopharm, Merck Sharp and Dohme, Midatech Pharma, Novocure, QED, and Roche and research support from Merck Sharp and Dohme and Novocure. MvdB received honoraria for advisory boards from Genenta, Boehringer, Astra Zeneca, Chimerix, Roche, Fore Biotherapeutics and Servier. MW has received research grants from Quercis and Versameb, and honoraria for lectures or advisory board participation or consulting from Bayer, Curevac, Medac, Merck (EMD), Novartis, Novocure, Orbus, Philogen, Roche and Sandoz. TG, JFe, JJ, CAM, DG, SC, MC and GR declare no conflict of interest., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
40. Lymph node and tumor-associated PD-L1 + macrophages antagonize dendritic cell vaccines by suppressing CD8 + T cells.
- Author
-
Sprooten J, Vanmeerbeek I, Datsi A, Govaerts J, Naulaerts S, Laureano RS, Borràs DM, Calvet A, Malviya V, Kuballa M, Felsberg J, Sabel MC, Rapp M, Knobbe-Thomsen C, Liu P, Zhao L, Kepp O, Boon L, Tejpar S, Borst J, Kroemer G, Schlenner S, De Vleeschouwer S, Sorg RV, and Garg AD
- Subjects
- Humans, Animals, Mice, CD8-Positive T-Lymphocytes, B7-H1 Antigen, Macrophages, Dendritic Cells, Lymph Nodes metabolism, Glioblastoma, Vaccines metabolism
- Abstract
Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) response
HIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+ /CD8+ T cells, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+ ) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted tumors., Competing Interests: Declaration of interests A.D.G. received honoraria/funding from Boehringer Ingelheim, Miltenyi Biotec, Novigenix, SOTIO, and IsoPlexis., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
41. Improved prognostic stratification of patients with isocitrate dehydrogenase-mutant astrocytoma.
- Author
-
Weller M, Felsberg J, Hentschel B, Gramatzki D, Kubon N, Wolter M, Reusche M, Roth P, Krex D, Herrlinger U, Westphal M, Tonn JC, Regli L, Maurage CA, von Deimling A, Pietsch T, Le Rhun E, and Reifenberger G
- Subjects
- Humans, Cohort Studies, Homozygote, Prognosis, Retrospective Studies, Sequence Deletion, Astrocytoma genetics, Astrocytoma therapy, Isocitrate Dehydrogenase genetics
- Abstract
Prognostic factors and standards of care for astrocytoma, isocitrate dehydrogenase (IDH)-mutant, CNS WHO grade 4, remain poorly defined. Here we sought to explore disease characteristics, prognostic markers, and outcome in patients with this newly defined tumor type. We determined molecular biomarkers and assembled clinical and outcome data in patients with IDH-mutant astrocytomas confirmed by central pathology review. Patients were identified in the German Glioma Network cohort study; additional cohorts of patients with CNS WHO grade 4 tumors were identified retrospectively at two sites. In total, 258 patients with IDH-mutant astrocytomas (114 CNS WHO grade 2, 73 CNS WHO grade 3, 71 CNS WHO grade 4) were studied. The median age at diagnosis was similar for all grades. Karnofsky performance status at diagnosis inversely correlated with CNS WHO grade (p < 0.001). Despite more intensive treatment upfront with higher grade, CNS WHO grade was strongly prognostic: median overall survival was not reached for grade 2 (median follow-up 10.4 years), 8.1 years (95% CI 5.4-10.8) for grade 3, and 4.7 years (95% CI 3.4-6.0) for grade 4. Among patients with CNS WHO grade 4 astrocytoma, median overall survival was 5.5 years (95% CI 4.3-6.7) without (n = 58) versus 1.8 years (95% CI 0-4.1) with (n = 12) homozygous CDKN2A deletion. Lower levels of global DNA methylation as detected by LINE-1 methylation analysis were strongly associated with CNS WHO grade 4 (p < 0.001) and poor outcome. MGMT promoter methylation status was not prognostic for overall survival. Histomolecular stratification based on CNS WHO grade, LINE-1 methylation level, and CDKN2A status revealed four subgroups of patients with significantly different outcomes. In conclusion, CNS WHO grade, global DNA methylation status, and CDKN2A homozygous deletion are prognostic in patients with IDH-mutant astrocytoma. Combination of these parameters allows for improved prediction of outcome. These data aid in designing upcoming trials using IDH inhibitors., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Spatial immune profiling of glioblastoma identifies an inflammatory, perivascular phenotype associated with longer survival.
- Author
-
Wirsching HG, Felsberg J, Prummer M, Moisoiu V, Lourman R, Hertler C, Antonios M, Cimino PJ, Roth P, Gorlia T, Prins RM, Cloughesy T, Wen PY, Holland EC, Reifenberger G, and Weller M
- Subjects
- Humans, Phenotype, Glioblastoma genetics, Brain Neoplasms genetics
- Published
- 2023
- Full Text
- View/download PDF
43. Integrative multi-omics reveals two biologically distinct groups of pilocytic astrocytoma.
- Author
-
Picard D, Felsberg J, Langini M, Stachura P, Qin N, Macas J, Reiss Y, Bartl J, Selt F, Sigaud R, Meyer FD, Stefanski A, Stühler K, Roque L, Roque R, Pandyra AA, Brozou T, Knobbe-Thomsen C, Plate KH, Roesch A, Milde T, Reifenberger G, Leprivier G, Faria CC, and Remke M
- Subjects
- Child, Humans, Multiomics, Proteomics, Action Potentials, Astrocytoma genetics, Brain Neoplasms genetics
- Abstract
Pilocytic astrocytoma (PA), the most common pediatric brain tumor, is driven by aberrant mitogen-activated protein kinase signaling most commonly caused by BRAF gene fusions or activating mutations. While 5-year overall survival rates exceed 95%, tumor recurrence or progression constitutes a major clinical challenge in incompletely resected tumors. Here, we used similarity network fusion (SNF) analysis in an integrative multi-omics approach employing RNA transcriptomic and mass spectrometry-based proteomic profiling to molecularly characterize PA tissue samples from 62 patients. Thereby, we uncovered that PAs segregated into two molecularly distinct groups, namely, Group 1 and Group 2, which were validated in three non-overlapping cohorts. Patients with Group 1 tumors were significantly younger and showed worse progression-free survival compared to patients with group 2 tumors. Ingenuity pathways analysis (IPA) and gene set enrichment analysis (GSEA) revealed that Group 1 tumors were enriched for immune response pathways, such as interferon signaling, while Group 2 tumors showed enrichment for action potential and neurotransmitter signaling pathways. Analysis of immune cell-related gene signatures showed an enrichment of infiltrating T Cells in Group 1 versus Group 2 tumors. Taken together, integrative multi-omics of PA identified biologically distinct and prognostically relevant tumor groups that may improve risk stratification of this single pathway driven tumor type., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
44. Long-term neurocognitive function and quality of life after multimodal therapy in adult glioma patients: a prospective long-term follow-up.
- Author
-
Pertz M, Schlömer S, Seidel C, Hentschel B, Löffler M, Schackert G, Krex D, Juratli T, Tonn JC, Schnell O, Vatter H, Simon M, Westphal M, Martens T, Sabel M, Bendszus M, Dörner N, Wick A, Fliessbach K, Hoppe C, Klingner M, Felsberg J, Reifenberger G, Gramatzki D, Weller M, and Schlegel U
- Subjects
- Humans, Adult, Follow-Up Studies, Quality of Life, Prospective Studies, Combined Modality Therapy, Brain Neoplasms complications, Brain Neoplasms radiotherapy, Glioma complications, Glioma radiotherapy
- Abstract
Purpose: Multimodal therapies have significantly improved prognosis in glioma. However, in particular radiotherapy may induce long-term neurotoxicity compromising patients' neurocognition and quality of life. The present prospective multicenter study aimed to evaluate associations of multimodal treatment with neurocognition with a particular focus on hippocampal irradiation., Methods: Seventy-one glioma patients (WHO grade 1-4) were serially evaluated with neurocognitive testing and quality of life questionnaires. Prior to (baseline) and following further treatment (median 7.1 years [range 4.6-11.0] after baseline) a standardized computerized neurocognitive test battery (NeuroCog FX) was applied to gauge psychomotor speed and inhibition, verbal short-term memory, working memory, verbal and non-verbal memory as well as verbal fluency. Mean ipsilateral hippocampal radiation dose was determined in a subgroup of 27 patients who received radiotherapy according to radiotherapy plans to evaluate its association with neurocognition., Results: Between baseline and follow-up mean performance in none of the cognitive domains significantly declined in any treatment modality (radiotherapy, chemotherapy, combined radio-chemotherapy, watchful-waiting), except for selective attention in patients receiving chemotherapy alone. Apart from one subtest (inhibition), mean ipsilateral hippocampal radiation dose > 50 Gy (Dmean) as compared to < 10 Gy showed no associations with long-term cognitive functioning. However, patients with Dmean < 10 Gy showed stable or improved performance in all cognitive domains, while patients with > 50 Gy numerically deteriorated in 4/8 domains., Conclusions: Multimodal glioma therapy seems to affect neurocognition less than generally assumed. Even patients with unilateral hippocampal irradiation with > 50 Gy showed no profound cognitive decline in this series., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
45. Long-term survival with IDH wildtype glioblastoma: first results from the ETERNITY Brain Tumor Funders' Collaborative Consortium (EORTC 1419).
- Author
-
Hertler C, Felsberg J, Gramatzki D, Le Rhun E, Clarke J, Soffietti R, Wick W, Chinot O, Ducray F, Roth P, McDonald K, Hau P, Hottinger AF, Reijneveld J, Schnell O, Marosi C, Glantz M, Darlix A, Lombardi G, Krex D, Glas M, Reardon DA, van den Bent M, Lefranc F, Herrlinger U, Razis E, Carpentier AF, Phillips S, Rudà R, Wick A, Tabouret E, Meyronet D, Maurage CA, Rushing E, Rapkins R, Bumes E, Hegi M, Weyerbrock A, Aregawi D, Gonzalez-Gomez C, Pellerino A, Klein M, Preusser M, Bendszus M, Golfinopoulos V, von Deimling A, Gorlia T, Wen PY, Reifenberger G, and Weller M
- Subjects
- Humans, Female, Young Adult, Adult, Middle Aged, Aged, Male, Isocitrate Dehydrogenase genetics, DNA Methylation, Neoplasm Recurrence, Local genetics, Prognosis, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Retrospective Studies, Glioblastoma genetics, Glioblastoma therapy, Glioblastoma pathology, Brain Neoplasms genetics, Brain Neoplasms therapy, Brain Neoplasms diagnosis
- Abstract
Background: Median survival with glioblastoma remains in the range of 12 months on population levels. Only few patients survive for more than 5 years. Patient and disease features associated with long-term survival remain poorly defined., Methods: European Organization for Research and Treatment of Cancer (EORTC) 1419 (ETERNITY) is a registry study supported by the Brain Tumor Funders Collaborative in the US and the EORTC Brain Tumor Group. Patients with glioblastoma surviving at least 5 years from diagnosis were identified at 24 sites in Europe, US, and Australia. In patients with isocitrate dehydrogenase (IDH) wildtype tumours, prognostic factors were analysed using the Kaplan-Meier method and the Cox proportional hazards model. A population-based reference cohort was obtained from the Cantonal cancer registry Zurich., Results: At the database lock of July 2020, 280 patients with histologically centrally confirmed glioblastoma (189 IDH wildtype, 80 IDH mutant, 11 incompletely characterised) had been registered. In the IDH wildtype population, median age was 56 years (range 24-78 years), 96 patients (50.8%) were female, 139 patients (74.3%) had tumours with O
6 -methylguanine DNA methyltransferase (MGMT) promoter methylation. Median overall survival was 9.9 years (95% confidence interval [95% CI] 7.9-11.9). Patients without recurrence experienced longer median survival (not reached) than patients with one or more recurrences (8.92 years) (p < 0.001) and had a high rate (48.8%) of MGMT promoter-unmethylated tumours., Conclusions: Freedom from progression is a powerful predictor of overall survival in long-term survivors with glioblastoma. Patients without relapse often have MGMT promoter-unmethylated glioblastoma and may represent a distinct subtype of glioblastoma., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: C.H. has received a research grant for protected time by the Filling the Gap foundation and honoraria for lecture from Vifor; E.L.R. has received honoraria for lectures or advisory board from Bayer, Janssen, Leo Pharma, Pierre Fabre, Seattle Genetics; J.C. has received research funding from Servier and Merck and consultant for Servier; R.S. reports consultation for Bayer, Astra Zeneca; W.W. reports consultation for Apogenix, Astra Zeneca, Bayer, Enterome, Medac, MSD and Roche/Genentech with honoraria paid to the Medical Faculty at the University of Heidelberg; F.D. has received honoraria for lectures or advisory board participation from Novocure; P.R. has received honoraria for lectures or advisory board participation from Bristol-Myers Squibb, Boehringer Ingelheim, Debiopharm, Merck Sharp and Dohme, Midatech, Novocure, QED, and Roche and research support from Merck Sharp and Dohme and Novocure; P.H. has received honoraria for lectures or advisory board participation or consulting from Bayer, Lilly, medac, Novartis, Novocure and Seagen; A.H. has received honoraria for lectures, consultation or advisory board participation from Novocure and Novartis; G.L. has received funding for a consulting or advisory role from Bayer, AbbVie, Orbus Therapeutics, BrainFarm, Health4U, Novartis, Braun and Janssen, and funding for travel from Roche, Bayer, and Ipsen; D.K. has received honoraria for lectures, consultation or advisory board participation from Novocure and BrainLab; M.G. reports honoraria from Roche, Novartis, UCB, AbbVie, Daiichi Sankyo, Novocure, Seagen, Bayer, Janssen-Cilag, Medac, Merck, Kyowa Kirin, travel support from Novocure and Medac, research grant from Novocure; D.R. reports support from Agenus, Agios; AnHeart Therapeutics, Avita Biomedical, Inc., Blue Rock Therapeutics, Bristol Myers Squibb, Boston Biomedica, CureVac AG, Del Mar Pharma, DNAtrix, Enterome, Hoffman-LaRoche, Ltd, Imvax, Janssen, Kiyatec, Medicenna Therapeutics, Neuvogen, Novartis, Novocure, Pyramid Bio, Sumitomo Dainippon Pharma. Vivacitas Oncology, Inc, Y-mabs Therapeutics; M.v.d.B has received honoraria for consultancy from Genenta, Servier, Astra Zeneca, Boehringer-Ingelheim, Carthera, Nerviano, Chimerix, Roche, Fore Biotherapeutics, Menarini-Stemline, Incyte and Sumitomo Pharma Oncology; F.L. received research funding from the Fonds Erasme; U.H. reports honoraria for lectures and and/or advisory board participation from Medac, Janssen, Bayer; E.R. reports travels grants BMS, Pfizer, MSD, Sanofi, Roche, Karyo labs Honorarium MSD, Servier; AF.C. received honoraria for lectures and/or advisory board participation from Gilead, Novartis; R.Ru. has received honoraria for lectures or consultation or advisory board from UCB, Novocure, Bayer, Genenta; M.P. has received honoraria for lectures, consultation or advisory board participation from the following for-profit companies: Bayer, Bristol-Myers Squibb, Novartis, Gerson Lehrman Group (GLG), CMC Contrast, GlaxoSmithKline, Mundipharma, Roche, BMJ Journals, MedMedia, Astra Zeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dome, Tocagen, Adastra, Gan & Lee Pharmaceuticals; P.W. has received research support from Astra Zeneca/Medimmune, Beigene, Celgene, Chimerix, Eli Lily, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Puma, Servier, Vascular Biogenics, VBI Vaccines and honoraria for advisory board participation or serving on data safety monitoring board from Astra Zeneca, Bayer, Black Diamond, Celularity, Chimerix, Day One Bio, Genenta, Novartis, Prelude Therapeutics, Sapience, Servier, Sagimet, Vascular Biogenics, VBI Vaccines; MW has received research grants from Quercis and Versameb, and honoraria for lectures or advisory board participation or consulting from Bayer, Medac, Merck (EMD), Novartis, Orbus, and Philogen. All remaining authors declare that they have no conflict of interest., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
46. Update on quality assurance in neuropathology: Summary of the round robin trials on TERT promoter mutation, H3-3A mutation, 1p/19q codeletion, and KIAA1549::BRAF fusion testing in Germany in 2020 and 2021.
- Author
-
Pohl S, Dimitrova L, Grassow-Narlik M, Jöhrens K, Acker T, Dohmen H, Herms J, Dorostkar M, Hartmann C, Hasselblatt M, Neumann M, Reifenberger G, Felsberg J, Schüller U, Zoubaa S, Lorenz J, Rothhammer-Hampl T, Mauch-Mücke K, and Riemenschneider MJ
- Subjects
- Child, Humans, Brain Neoplasms diagnosis, Brain Neoplasms genetics, Germany, Membrane Proteins genetics, Oligodendroglioma diagnosis, Oligodendroglioma genetics, Proto-Oncogene Proteins B-raf genetics, Biomarkers, Tumor genetics, Chromosome Deletion, Genetic Testing, Histones genetics, Mutation, Oncogene Proteins, Fusion genetics, Promoter Regions, Genetic genetics, Telomerase genetics
- Abstract
We previously reported on the first neuropathological round robin trials operated together with Quality in Pathology (QuIP) GmbH in 2018 and 2019 in Germany, i.e., the trials on IDH mutational testing and MGMT promoter methylation analysis [1]. For 2020 and 2021, the spectrum of round robin trials has been expanded to cover the most commonly used assays in neuropathological institutions. In addition to IDH mutation and MGMT promoter methylation testing, there is a long tradition for 1p/19q codeletion testing relevant in the context of the diagnosis of oligodendroglioma. With the 5
th edition of the World Health Organization (WHO) classification of the central nervous system tumors, additional molecular markers came into focus: TERT promoter mutation is often assessed as a molecular diagnostic criterion for IDH-wildtype glioblastoma. Moreover, several molecular diagnostic markers have been introduced for pediatric brain tumors. Here, trials on KIAA1549::BRAF fusions (common in pilocytic astrocytomas) and H3-3A mutations (in diffuse midline gliomas, H3-K27-altered and diffuse hemispheric gliomas, H3-G34-mutant) were most desired by the neuropathological community. In this update, we report on these novel round robin trials. In summary, success rates in all four trials ranged from 75 to 96%, arguing for an overall high quality level in the field of molecular neuropathological diagnostics.- Published
- 2023
- Full Text
- View/download PDF
47. Quantitative proteomic landscapes of primary and recurrent glioblastoma reveal a protumorigeneic role for FBXO2-dependent glioma-microenvironment interactions.
- Author
-
Buehler M, Yi X, Ge W, Blattmann P, Rushing E, Reifenberger G, Felsberg J, Yeh C, Corn JE, Regli L, Zhang J, Cloos A, Ravi VM, Wiestler B, Heiland DH, Aebersold R, Weller M, Guo T, and Weiss T
- Subjects
- Humans, Animals, Mice, Proteomics, Mice, Knockout, Brain pathology, Proteins, Tumor Microenvironment, Neoplasm Proteins, Nerve Tissue Proteins, Cell Cycle Proteins, Glioblastoma pathology, Glioma pathology, Brain Neoplasms pathology, F-Box Proteins genetics
- Abstract
Background: Recent efforts have described the evolution of glioblastoma from initial diagnosis to post-treatment recurrence on a genomic and transcriptomic level. However, the evolution of the proteomic landscape is largely unknown., Methods: Sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) was used to characterize the quantitative proteomes of two independent cohorts of paired newly diagnosed and recurrent glioblastomas. Recurrence-associated proteins were validated using immunohistochemistry and further studied in human glioma cell lines, orthotopic xenograft models, and human organotypic brain slice cultures. External spatial transcriptomic, single-cell, and bulk RNA sequencing data were analyzed to gain mechanistic insights., Results: Although overall proteomic changes were heterogeneous across patients, we identified BCAS1, INF2, and FBXO2 as consistently upregulated proteins at recurrence and validated these using immunohistochemistry. Knockout of FBXO2 in human glioma cells conferred a strong survival benefit in orthotopic xenograft mouse models and reduced invasive growth in organotypic brain slice cultures. In glioblastoma patient samples, FBXO2 expression was enriched in the tumor infiltration zone and FBXO2-positive cancer cells were associated with synaptic signaling processes., Conclusions: These findings demonstrate a potential role of FBXO2-dependent glioma-microenvironment interactions to promote tumor growth. Furthermore, the published datasets provide a valuable resource for further studies., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
48. A Sequential Targeting Strategy Interrupts AKT-Driven Subclone-Mediated Progression in Glioblastoma.
- Author
-
Kebir S, Ullrich V, Berger P, Dobersalske C, Langer S, Rauschenbach L, Trageser D, Till A, Lorbeer FK, Wieland A, Wilhelm-Buchstab T, Ahmad A, Fröhlich H, Cima I, Prasad S, Matschke J, Jendrossek V, Remke M, Grüner BM, Roesch A, Siveke JT, Herold-Mende C, Blau T, Keyvani K, van Landeghem FKH, Pietsch T, Felsberg J, Reifenberger G, Weller M, Sure U, Brüstle O, Simon M, Glas M, and Scheffler B
- Subjects
- Humans, Proto-Oncogene Proteins c-akt, Drug Resistance, Neoplasm genetics, Temozolomide, Cell Line, Tumor, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Antineoplastic Agents, Alkylating therapeutic use, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma pathology, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms pathology
- Abstract
Purpose: Therapy resistance and fatal disease progression in glioblastoma are thought to result from the dynamics of intra-tumor heterogeneity. This study aimed at identifying and molecularly targeting tumor cells that can survive, adapt, and subclonally expand under primary therapy., Experimental Design: To identify candidate markers and to experimentally access dynamics of subclonal progression in glioblastoma, we established a discovery cohort of paired vital cell samples obtained before and after primary therapy. We further used two independent validation cohorts of paired clinical tissues to test our findings. Follow-up preclinical treatment strategies were evaluated in patient-derived xenografts., Results: We describe, in clinical samples, an archetype of rare ALDH1A1+ tumor cells that enrich and acquire AKT-mediated drug resistance in response to standard-of-care temozolomide (TMZ). Importantly, we observe that drug resistance of ALDH1A1+ cells is not intrinsic, but rather an adaptive mechanism emerging exclusively after TMZ treatment. In patient cells and xenograft models of disease, we recapitulate the enrichment of ALDH1A1+ cells under the influence of TMZ. We demonstrate that their subclonal progression is AKT-driven and can be interfered with by well-timed sequential rather than simultaneous antitumor combination strategy., Conclusions: Drug-resistant ALDH1A1+/pAKT+ subclones accumulate in patient tissues upon adaptation to TMZ therapy. These subclones may therefore represent a dynamic target in glioblastoma. Our study proposes the combination of TMZ and AKT inhibitors in a sequential treatment schedule as a rationale for future clinical investigation., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
49. Tacedinaline (CI-994), a class I HDAC inhibitor, targets intrinsic tumor growth and leptomeningeal dissemination in MYC-driven medulloblastoma while making them susceptible to anti-CD47-induced macrophage phagocytosis via NF-kB-TGM2 driven tumor inflammation.
- Author
-
Marquardt V, Theruvath J, Pauck D, Picard D, Qin N, Blümel L, Maue M, Bartl J, Ahmadov U, Langini M, Meyer FD, Cole A, Cruz-Cruz J, Graef CM, Wölfl M, Milde T, Witt O, Erdreich-Epstein A, Leprivier G, Kahlert U, Stefanski A, Stühler K, Keir ST, Bigner DD, Hauer J, Beez T, Knobbe-Thomsen CB, Fischer U, Felsberg J, Hansen FK, Vibhakar R, Venkatraman S, Cheshier SH, Reifenberger G, Borkhardt A, Kurz T, Remke M, and Mitra S
- Subjects
- Humans, Mice, Animals, NF-kappa B metabolism, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Protein Glutamine gamma Glutamyltransferase 2, Quality of Life, Phagocytosis, Macrophages, Inflammation metabolism, Medulloblastoma drug therapy, Glioblastoma, Cerebellar Neoplasms
- Abstract
Background: While major advances have been made in improving the quality of life and survival of children with most forms of medulloblastoma (MB), those with MYC-driven tumors (Grp3-MB) still suffer significant morbidity and mortality. There is an urgent need to explore multimodal therapeutic regimens which are effective and safe for children. Large-scale studies have revealed abnormal cancer epigenomes caused by mutations and structural alterations of chromatin modifiers, aberrant DNA methylation, and histone modification signatures. Therefore, targeting epigenetic modifiers for cancer treatment has gained increasing interest, and inhibitors for various epigenetic modulators have been intensively studied in clinical trials. Here, we report a cross-entity, epigenetic drug screen to evaluate therapeutic vulnerabilities in MYC amplified MB, which sensitizes them to macrophage-mediated phagocytosis by targeting the CD47-signal regulatory protein α (SIRPα) innate checkpoint pathway., Methods: We performed a primary screen including 78 epigenetic inhibitors and a secondary screen including 20 histone deacetylase inhibitors (HDACi) to compare response profiles in atypical teratoid/rhabdoid tumor (AT/RT, n=11), MB (n=14), and glioblastoma (n=14). This unbiased approach revealed the preferential activity of HDACi in MYC-driven MB. Importantly, the class I selective HDACi, CI-994, showed significant cell viability reduction mediated by induction of apoptosis in MYC-driven MB, with little-to-no activity in non-MYC-driven MB, AT/RT, and glioblastoma in vitro. We tested the combinatorial effect of targeting class I HDACs and the CD47-SIRPa phagocytosis checkpoint pathway using in vitro phagocytosis assays and in vivo orthotopic xenograft models., Results: CI-994 displayed antitumoral effects at the primary site and the metastatic compartment in two orthotopic mouse models of MYC-driven MB. Furthermore, RNA sequencing revealed nuclear factor-kB (NF-κB) pathway induction as a response to CI-994 treatment, followed by transglutaminase 2 (TGM2) expression, which enhanced inflammatory cytokine secretion. We further show interferon-γ release and cell surface expression of engulfment ('eat-me') signals (such as calreticulin). Finally, combining CI-994 treatment with an anti-CD47 mAb targeting the CD47-SIRPα phagocytosis checkpoint enhanced in vitro phagocytosis and survival in tumor-bearing mice., Conclusion: Together, these findings suggest a dynamic relationship between MYC amplification and innate immune suppression in MYC amplified MB and support further investigation of phagocytosis modulation as a strategy to enhance cancer immunotherapy responses., Competing Interests: Competing interests: SHC and SSM hold a Patent entitled ‘Treatment of pediatric brain tumors with targeting of CD47 pathway’. Other authors hold no potential conflict of interest., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
50. Anaplastic ganglioglioma-A diagnosis comprising several distinct tumour types.
- Author
-
Reinhardt A, Pfister K, Schrimpf D, Stichel D, Sahm F, Reuss DE, Capper D, Wefers AK, Ebrahimi A, Sill M, Felsberg J, Reifenberger G, Becker A, Prinz M, Staszewski O, Hartmann C, Schittenhelm J, Gramatzki D, Weller M, Olar A, Rushing EJ, Bergmann M, Farrell MA, Blümcke I, Coras R, Beckervordersandforth J, Kim SH, Rogerio F, Dimova PS, Niehusmann P, Unterberg A, Platten M, Pfister SM, Wick W, Herold-Mende C, and von Deimling A
- Subjects
- Child, Humans, Retrospective Studies, Isocitrate Dehydrogenase, Ganglioglioma pathology, Glioma pathology, Astrocytoma pathology, Brain Neoplasms genetics, Central Nervous System Neoplasms pathology
- Abstract
Aims: Anaplastic ganglioglioma is a rare tumour, and diagnosis has been based on histological criteria. The 5th edition of the World Health Organization Classification of Tumours of the Central Nervous System (CNS WHO) does not list anaplastic ganglioglioma as a distinct diagnosis due to lack of molecular data in previous publications. We retrospectively compiled a cohort of 54 histologically diagnosed anaplastic gangliogliomas to explore whether the molecular profiles of these tumours represent a separate type or resolve into other entities., Methods: Samples were subjected to histological review, desoxyribonucleic acid (DNA) methylation profiling and next-generation sequencing. Morphological and molecular data were summarised to an integrated diagnosis., Results: The majority of tumours designated as anaplastic gangliogliomas resolved into other CNS WHO diagnoses, most commonly pleomorphic xanthoastrocytoma (16/54), glioblastoma, isocitrate dehydrogenase protein (IDH) wild type and diffuse paediatric-type high-grade glioma, H3 wild type and IDH wild type (11 and 2/54), followed by low-grade glial or glioneuronal tumours including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumour and diffuse leptomeningeal glioneuronal tumour (5/54), IDH mutant astrocytoma (4/54) and others (6/54). A subset of tumours (10/54) was not assignable to a CNS WHO diagnosis, and common molecular profiles pointing to a separate entity were not evident., Conclusions: In summary, we show that tumours histologically diagnosed as anaplastic ganglioglioma comprise a wide spectrum of CNS WHO tumour types with different prognostic and therapeutic implications. We therefore suggest assigning this designation with caution and recommend comprehensive molecular workup., (© 2022 The Authors. Neuropathology and Applied Neurobiology published by John Wiley & Sons Ltd on behalf of British Neuropathological Society.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.