65 results on '"Amankulor N"'
Search Results
2. A Phase II Study to Determine the Efficacy of Pre-Operative Stereotactic Radiosurgery Followed by Resection for Brain Metastasis
- Author
-
Iheagwara, U.K., primary, Siddiqui, Z., additional, Holeva, K., additional, Engh, J., additional, Amankulor, N., additional, Burton, S.A., additional, and Clump, D.A., additional
- Published
- 2021
- Full Text
- View/download PDF
3. Outcomes and Toxicity of Single- And Multi-Fraction Spine Radiosurgery for Three or More Vertebral Bodies
- Author
-
Skiba, J.H., primary, Iarrobino, N., additional, Burton, S.A., additional, Flickinger, J.C., additional, Gerszten, P.C., additional, Amankulor, N., additional, and Olson, A.C., additional
- Published
- 2020
- Full Text
- View/download PDF
4. Book Review: Poetic Heritage: Igbo Traditional Verse. Compiled and translated with an introduction by Romanus N. Egudu and Donatus I. Nwoga. Enugu, Nigeria: Nwankwo-Ifejika & Co., 1971.
- Author
-
Amankulor, N. N.
- Abstract
No abstract
- Published
- 1973
5. Recursive Partitioning Analysis of Patients with Spine Metastases Treated with Stereotactic Body Radiotherapy
- Author
-
Rodriguez-Lopez, J.L., primary, Flickinger, J.C., additional, Gerszten, P.C., additional, Amankulor, N., additional, Burton, S.A., additional, and Olson, A.C., additional
- Published
- 2019
- Full Text
- View/download PDF
6. Immuno-Profile of a Phase II Clinical Trial Utilizing Preoperative Stereotactic Radiosurgery Followed by Surgery in Cancer Patient with Metastatic Lesions in the Brain
- Author
-
Iheagwara, U.K., primary, Holeva, K., additional, Bakkenist, C.J., additional, Shogan, J., additional, Engh, J., additional, Amankulor, N., additional, and Clump, D.A., additional
- Published
- 2018
- Full Text
- View/download PDF
7. Long-Term Outcomes After Stereotactic Radiosurgery for Spine Metastases: Radiation Dose Response for Late Toxicity
- Author
-
Ling, D.C., primary, Flickinger, J.C., additional, Burton, S.A., additional, Heron, D.E., additional, Quinn, A., additional, Ghassan, B.K., additional, Engh, J., additional, Gerszten, P.C., additional, Amankulor, N., additional, and Vargo, J.A., additional
- Published
- 2017
- Full Text
- View/download PDF
8. NT-03 * DRAMATIC RESPONSE INDUCED BY VEMURAFENIB IN A BRAF V600E-MUTATED BEVACIZUMAB REFRACTORY GLIOBLASTOMA
- Author
-
Arvanitis, L., primary, Marchioli, C., additional, Flickinger, J., additional, Pritchard, A., additional, Engh, J., additional, Amankulor, N., additional, Lieberman, F., additional, Nikiforova, M., additional, Murdoch, G., additional, and Drappatz, J., additional
- Published
- 2014
- Full Text
- View/download PDF
9. IT-33 * USE OF IFN-ALPHA AND POLY-ICLC AS CHEMOKINE MODULATORS FOR IMMUNOTHERAPY IN GLIOMAS
- Author
-
Smith-Cohn, M., primary, Kohanbash, G., additional, Muthuswamy, R., additional, Ohkuri, T., additional, Kosaka, A., additional, Amankulor, N., additional, Engh, J., additional, Potter, D., additional, Kalinski, P., additional, and Okada, H., additional
- Published
- 2014
- Full Text
- View/download PDF
10. IT-23 * INDUCTION OF ROBUST TYPE-1 CD8+ T-CELL RESPONSES IN WHO GRADE II LOW-GRADE GLIOMA PATIENTS RECEIVING PEPTIDE-BASED VACCINES IN COMBINATION WITH POLY-ICLC
- Author
-
Okada, H., primary, Butterfield, L., additional, Hamilton, R., additional, Ahn, B., additional, Kohanbash, G., additional, Drappatz, J., additional, Engh, J., additional, Amankulor, N., additional, Lively, M., additional, Chan, M., additional, Salazar, A., additional, Shaw, E., additional, Potter, D., additional, and Lieberman, F., additional
- Published
- 2014
- Full Text
- View/download PDF
11. IB-03 * IDH MUTANT GLIOMAS ARE RESISTANT TO NATURAL KILLER CELL-MEDIATED CYTOLYSIS
- Author
-
Brancho, M., primary, Chang, Y., additional, Zhang, X., additional, Okada, H., additional, Li, A., additional, Grandi, P., additional, Lotze, M., additional, and Amankulor, N., additional
- Published
- 2014
- Full Text
- View/download PDF
12. Postoperative Stereotactic Radiosurgery to the Resection Cavity for Large Brain Metastases
- Author
-
Ling, D.C., primary, Vargo, J.A., additional, Wegner, R.E., additional, Flickinger, J.C., additional, Burton, S.A., additional, Engh, J., additional, Amankulor, N., additional, Quinn, A.E., additional, Ozhasoglu, C., additional, and Heron, D.E., additional
- Published
- 2014
- Full Text
- View/download PDF
13. ROBUST INDUCTION OF TYPE-1 CD8+ T-CELL RESPONSES IN WHO GRADE II LOW-GRADE GLIOMA PATIENTS RECEIVING PEPTIDE-BASED VACCINES IN COMBINATION WITH POLY-ICLC
- Author
-
Okada, H., primary, Butterfield, L. H., additional, Hamilton, R. L., additional, Hoji, A., additional, Sakaki, M., additional, Ahn, B. J., additional, Kohanbash, G., additional, Drappatz, J., additional, Engh, J., additional, Amankulor, N., additional, Lively, M. O., additional, Chan, M. D., additional, Salazar, A. M., additional, Potter, D. M., additional, Shaw, E. G., additional, and Lieberman, F. S., additional
- Published
- 2014
- Full Text
- View/download PDF
14. IMMUNOTHERAPY/BIOLOGICAL THERAPIES
- Author
-
Campian, J., primary, Gladstone, D., additional, Ambady, P., additional, Ye, X., additional, King, K., additional, Borrello, I., additional, Petrik, S., additional, Golightly, M., additional, Holdhoff, M., additional, Grossman, S., additional, Bhardwaj, R., additional, Chakravadhanula, M., additional, Ozols, V., additional, Georges, J., additional, Carlson, E., additional, Hampton, C., additional, Decker, W., additional, Chiba, Y., additional, Hashimoto, N., additional, Kagawa, N., additional, Hirayama, R., additional, Tsuboi, A., additional, Oji, Y., additional, Oka, Y., additional, Sugiyama, H., additional, Yoshimine, T., additional, Choi, B., additional, Gedeon, P., additional, Herndon, J., additional, Sanchez-Perez, L., additional, Mitchell, D., additional, Bigner, D., additional, Sampson, J., additional, Choi, Y. A., additional, Pandya, H., additional, Gibo, D. M., additional, Debinski, W., additional, Cloughesy, T. F., additional, Liau, L. M., additional, Chiocca, E. A., additional, Jolly, D. J., additional, Robbins, J. M., additional, Ostertag, D., additional, Ibanez, C. E., additional, Gruber, H. E., additional, Kasahara, N., additional, Vogelbaum, M. A., additional, Kesari, S., additional, Mikkelsen, T., additional, Kalkanis, S., additional, Landolfi, J., additional, Bloomfield, S., additional, Foltz, G., additional, Pertschuk, D., additional, Everson, R., additional, Jin, R., additional, Safaee, M., additional, Lisiero, D., additional, Odesa, S., additional, Liau, L., additional, Prins, R., additional, Gholamin, S., additional, Mitra, S. S., additional, Richard, C. E., additional, Achrol, A., additional, Kahn, S. A., additional, Volkmer, A. K., additional, Volkmer, J. P., additional, Willingham, S., additional, Kong, D., additional, Shin, J. J., additional, Monje-Deisseroth, M., additional, Cho, Y.-J., additional, Weissman, I., additional, Cheshier, S. H., additional, Kanemura, Y., additional, Sumida, M., additional, Yoshioka, E., additional, Yamamoto, A., additional, Kanematsu, D., additional, Takada, A., additional, Nonaka, M., additional, Nakajima, S., additional, Goto, S., additional, Kamigaki, T., additional, Takahara, M., additional, Maekawa, R., additional, Shofuda, T., additional, Moriuchi, S., additional, Yamasaki, M., additional, Kebudi, R., additional, Cakir, F. B., additional, Gorgun, O., additional, Agaoglu, F. Y., additional, Darendeliler, E., additional, Lin, Y., additional, Wang, Y., additional, Qiu, X., additional, Jiang, T., additional, Zhang, G., additional, Wang, J., additional, Okada, H., additional, Butterfield, L., additional, Hamilton, R., additional, Drappatz, J., additional, Engh, J., additional, Amankulor, N., additional, Lively, M., additional, Chan, M., additional, Salazar, A., additional, Potter, D., additional, Shaw, E., additional, Lieberman, F., additional, Choi, Y., additional, Park, J., additional, Phuphanich, S., additional, Wheeler, C., additional, Rudnick, J., additional, Hu, J., additional, Mazer, M., additional, Wang, H., additional, Nuno, M., additional, Guevarra, A., additional, Sanchez, C., additional, Fan, X., additional, Ji, J., additional, Chu, R., additional, Bender, J., additional, Hawkins, E., additional, Black, K., additional, Yu, J., additional, Reap, E., additional, Archer, G., additional, Norberg, P., additional, Schmittling, R., additional, Nair, S., additional, Cui, X., additional, Snyder, D., additional, Chandramohan, V., additional, Kuan, C.-T., additional, Yan, H., additional, Reardon, D., additional, Li, G., additional, Recht, L., additional, Fink, K., additional, Nabors, L., additional, Tran, D., additional, Desjardins, A., additional, Chandramouli, N., additional, Duic, J. P., additional, Groves, M., additional, Clarke, A., additional, Hawthorne, T., additional, Green, J., additional, Yellin, M., additional, Rigakos, G., additional, Spyri, O., additional, Nomikos, P., additional, Stavridi, F., additional, Grossi, I., additional, Theodorakopoulou, I., additional, Assi, A., additional, Kouvatseas, G., additional, Papadopoulou, E., additional, Nasioulas, G., additional, Labropoulos, S., additional, Razis, E., additional, Ravi, A., additional, Tang, D. N., additional, Sharma, P., additional, Sengupta, S., additional, Sampath, P., additional, Soto, H., additional, Erickson, K., additional, Malone, C., additional, Hickey, M., additional, Ha, E., additional, Young, E., additional, Ellingson, B., additional, Kruse, C., additional, Sul, J., additional, Hilf, N., additional, Kutscher, S., additional, Schoor, O., additional, Lindner, J., additional, Reinhardt, C., additional, Kreisl, T., additional, Iwamoto, F., additional, Fine, H., additional, Singh-Jasuja, H., additional, Teijeira, L., additional, Gil-Arnaiz, I., additional, Hernandez-Marin, B., additional, Martinez-Aguillo, M., additional, Sanchez, S. d. l. C., additional, Viudez, A., additional, Hernandez-Garcia, I., additional, Lecumberri, M. J., additional, Grandez, R., additional, de Lascoiti, A. F., additional, Garcia, R. V., additional, Thomas, A., additional, Fisher, J., additional, Baron, U., additional, Olek, S., additional, Rhodes, H., additional, Gui, J., additional, Hampton, T., additional, Tafe, L., additional, Tsongalis, G., additional, Lefferts, J., additional, Wishart, H., additional, Kleen, J., additional, Miller, M., additional, Ernstoff, M., additional, Fadul, C., additional, Vlahovic, G., additional, Peters, K., additional, Ranjan, T., additional, Friedman, A., additional, Friedman, H., additional, Lally-Goss, D., additional, Wainwright, D., additional, Dey, M., additional, Chang, A., additional, Cheng, Y., additional, Han, Y., additional, Lesniak, M., additional, Weller, M., additional, Kaulich, K., additional, Hentschel, B., additional, Felsberg, J., additional, Gramatzki, D., additional, Pietsch, T., additional, Simon, M., additional, Westphal, M., additional, Schackert, G., additional, Tonn, J. C., additional, Loeffler, M., additional, Reifenberger, G., additional, Xu, M., additional, and Patil, C., additional
- Published
- 2013
- Full Text
- View/download PDF
15. NEUROSURGICAL TREATMENTS
- Author
-
Nanney, A. D., primary, Adel, J. G., additional, Smith, T. R., additional, Chandler, J. P., additional, Kimmell, K. T., additional, Walter, K., additional, Zacharia, B. E., additional, Deibert, C., additional, Malone, H. R., additional, Sonabend, A. M., additional, Neugut, A. I., additional, Spencer, B., additional, Bruce, J. N., additional, Wang, Y., additional, Li, S., additional, Zhang, Z., additional, Chen, X., additional, You, G., additional, Yang, P., additional, Yan, W., additional, Bao, Z., additional, Yao, K., additional, Liu, Y., additional, Wang, L., additional, Jiang, T., additional, Farhoud, M. K., additional, Ruge, M. I., additional, Brandes, A. A., additional, Ermani, M., additional, Fioravanti, A., additional, Andreoli, A., additional, Pozzati, E., additional, Bacci, A., additional, Bartolini, S., additional, Poggi, R., additional, Crisi, G., additional, Franceschi, E., additional, Recinos, P. F., additional, Grabowski, M. M., additional, Nowacki, A. S., additional, Thompson, N., additional, Vogelbaum, M. A., additional, Sun, P., additional, Krueger, D., additional, Liu, Z., additional, Kohrman, M., additional, Dagens, A. B., additional, Rachinger, W., additional, Kunz, M., additional, Eigenbrod, S., additional, Lutz, J., additional, Tonn, J.-C., additional, Kreth, F.-W., additional, Duong, H. T., additional, Chaloner, C., additional, Bordo, G., additional, Eisenberg, A., additional, Rosenthal, K., additional, Sim, M.-S., additional, Boasberg, P., additional, Faries, M. B., additional, Hamid, O., additional, Kelly, D. F., additional, Thon, N., additional, Simon, M., additional, Westphal, M., additional, Schackert, G., additional, Nikkhah, G., additional, Hentschel, B., additional, Pietsch, T., additional, Reifenberger, G., additional, Weller, M., additional, Ironside, S., additional, Perry, J., additional, Tsao, M., additional, Mainprize, T., additional, Keith, J., additional, Laperrierre, N., additional, Paszat, L., additional, Sahgal, A., additional, Hoover, J. M., additional, Nwojo, M., additional, Puffer, R., additional, Parney, I. F., additional, Tanaka, S., additional, Nakada, M., additional, Hayashi, Y., additional, Hamada, J.-i., additional, Lee, I. Y., additional, Ekram, T., additional, Jain, R., additional, Scarpace, L., additional, Omodon, M., additional, Rock, J., additional, Rosenblum, M., additional, Kalkanis, S., additional, Amankulor, N. M., additional, Kim, J.-H., additional, Tabar, V., additional, Peck, K. K., additional, Holodny, A. I., additional, Gutin, P. H., additional, Kim, C.-Y., additional, Kim, Y.-H., additional, Kim, T., additional, Kim, I. K., additional, Kim, J. W., additional, Kim, Y. H., additional, Han, J. H., additional, Park, C.-K., additional, Kim, D. G., additional, Jung, H.-W., additional, Nonaka, M., additional, Bamba, Y., additional, Kanemura, Y., additional, and Nakajima, S., additional
- Published
- 2012
- Full Text
- View/download PDF
16. Tumor Models (In Vivo/In Vitro)
- Author
-
Doucette, T. A., primary, Kong, L.-Y., additional, Yang, Y., additional, Wei, J., additional, Wang, J., additional, Fuller, G. N., additional, Heimberger, A. B., additional, Rao, G., additional, Ajewung, N., additional, Kamnasaran, D., additional, Katz, A. M., additional, Amankulor, N., additional, Squatrito, M., additional, Hambardzumyan, D., additional, Holland, E. C., additional, Poschl, J., additional, Lorenz, A., additional, Von Bueren, A., additional, Li, S., additional, Peraud, A., additional, Tonn, J.-C., additional, Herms, J., additional, Xiang, M., additional, Rutkowski, S., additional, Kretzschmar, H., additional, Schuller, U., additional, Studebaker, A., additional, Raffel, C., additional, Aoki, Y., additional, Hashizume, R., additional, Ozawa, T., additional, Gupta, N., additional, James, C. D., additional, Navis, A. C., additional, Hamans, B. C., additional, Claes, A., additional, Heerschap, A., additional, Wesseling, P., additional, Jeuken, J. W., additional, Leenders, W. P., additional, Agudelo, P. A., additional, Williams, S., additional, Nowicki, M. O., additional, Johnson, J., additional, Li, P. K., additional, Chiocca, E. A., additional, Lannutti, J. J., additional, Lawler, S. E., additional, Viapiano, M. S., additional, Bergeron, J., additional, Aliaga, A., additional, Bedell, B., additional, Soderquist, C., additional, Sonabend, A., additional, Lei, L., additional, Crisman, C., additional, Yun, J. P., additional, Sisti, J., additional, Castelli, M., additional, Bruce, J. N., additional, Canoll, P., additional, Kirsch, M., additional, Stelling, A., additional, Salzer, R., additional, Krafft, C., additional, Schackert, G., additional, Steiner, G., additional, Balvers, R. K., additional, van den Hengel, S. K., additional, Wakimoto, H., additional, Hoeben, R. C., additional, Leenstra, S., additional, Dirven, C. M., additional, Lamfers, M. L., additional, Sabha, N. S., additional, Agnihotri, S., additional, Wolf, A., additional, von Deimling, A., additional, Croul, S., additional, Guha, A., additional, Trojahn, U. S., additional, Lenferink, A., additional, O'Connor-McCourt, M., additional, Kanai, R., additional, Curry, W. T., additional, Yip, S., additional, Barnard, Z. R., additional, Mohapatra, G., additional, Stemmer-Rachamimov, A. O., additional, Martuza, R. L., additional, Rabkin, S. D., additional, Binder, Z. A., additional, Salmasi, V., additional, Lim, M., additional, Weingart, J., additional, Brem, H., additional, Olivi, A., additional, Riggins, G. J., additional, Gallia, G. L., additional, Rong, Y., additional, Zhang, Z., additional, Gang, C., additional, Tucker-Burden, C., additional, Van Meir, E., additional, Brat, D. J., additional, Kloezeman, J. J., additional, Kleijn, A., additional, French, P. J., additional, Spoor, J. K., additional, Bazzoli, E., additional, Fomchenko, E. I., additional, Schultz, N., additional, Brennan, C., additional, DeAngelis, L. M., additional, Nimer, S. D., additional, Mohyeldin, A., additional, Hsu, W., additional, Shah, S. R., additional, Adams, H., additional, Shah, P., additional, Katuri, L., additional, Kosztowski, T., additional, Loeb, D. M., additional, Wolinsky, J.-P., additional, Gokaskan, Z. L., additional, Quinones-Hinojosa, A., additional, Daphu, I. K., additional, Immervoll, H., additional, Bjerkvig, R., additional, Thorsen, F., additional, Caretti, V., additional, Idema, S., additional, Zondervan, I., additional, Meijer, D. H., additional, Lagerweij, T., additional, Barazas, M., additional, Vos, W., additional, Hamans, B., additional, van der Stoop, P., additional, Hulleman, E., additional, van der Valk, P., additional, Bugiani, M., additional, Vandertop, W. P., additional, Noske, D., additional, Kaspers, G. J., additional, Molthoff, C., additional, Wurdinger, T., additional, Chow, L. M., additional, Endersby, R., additional, Zhu, X., additional, Rankin, S., additional, Qu, C., additional, Zhang, J., additional, Ellison, D. W., additional, Baker, S. J., additional, Tabar, V., additional, LaFaille, F., additional, and Studer, L., additional
- Published
- 2010
- Full Text
- View/download PDF
17. ChemInform Abstract: N‐Alkoxy Analogues of 3,4,5‐Trihydroxypiperidine.
- Author
-
SUN, L., primary, LI, P., additional, AMANKULOR, N., additional, TANG, W., additional, LANDRY, D. W., additional, and ZHAO, K., additional
- Published
- 1999
- Full Text
- View/download PDF
18. ChemInform Abstract: Syntheses of Isoxazolinyl and Isoxazolidinyl Nucleoside Analogues
- Author
-
PAN, S., primary, AMANKULOR, N. M., additional, and ZHAO, K., additional
- Published
- 1998
- Full Text
- View/download PDF
19. Machine Learning-based Prognostic Subgrouping of Glioblastoma: A Multi-center Study.
- Author
-
Akbari H, Bakas S, Sako C, Fathi Kazerooni A, Villanueva-Meyer J, Garcia JA, Mamourian E, Liu F, Cao Q, Shinohara RT, Baid U, Getka A, Pati S, Singh A, Calabrese E, Chang S, Rudie J, Sotiras A, LaMontagne P, Marcus DS, Milchenko M, Nazeri A, Balana C, Capellades J, Puig J, Badve C, Barnholtz-Sloan JS, Sloan AE, Vadmal V, Waite K, Ak M, Colen RR, Park YW, Ahn SS, Chang JH, Choi YS, Lee SK, Alexander GS, Ali AS, Dicker AP, Flanders AE, Liem S, Lombardo J, Shi W, Shukla G, Griffith B, Poisson LM, Rogers LR, Kotrotsou A, Booth TC, Jain R, Lee M, Mahajan A, Chakravarti A, Palmer JD, DiCostanzo D, Fathallah-Shaykh H, Cepeda S, Santonocito OS, Di Stefano AL, Wiestler B, Melhem ER, Woodworth GF, Tiwari P, Valdes P, Matsumoto Y, Otani Y, Imoto R, Aboian M, Koizumi S, Kurozumi K, Kawakatsu T, Alexander K, Satgunaseelan L, Rulseh AM, Bagley SJ, Bilello M, Binder ZA, Brem S, Desai AS, Lustig RA, Maloney E, Prior T, Amankulor N, Nasrallah MLP, O'Rourke DM, Mohan S, and Davatzikos C
- Abstract
Background: Glioblastoma is the most aggressive adult primary brain cancer, characterized by significant heterogeneity, posing challenges for patient management, treatment planning, and clinical trial stratification., Methods: We developed a highly reproducible, personalized prognostication and clinical subgrouping system using machine learning (ML) on routine clinical data, MRI, and molecular measures from 2,838 demographically diverse patients across 22 institutions and 3 continents. Patients were stratified into favorable, intermediate, and poor prognostic subgroups (I, II, III) using Kaplan-Meier analysis (Cox proportional model and hazard ratios [HR])., Results: The ML model stratified patients into distinct prognostic subgroups with HRs between subgroups I-II and I-III of 1.62 (95%CI: 1.43-1.84, p<0.001) and 3.48 (95%CI: 2.94-4.11, p<0.001), respectively. Analysis of imaging features revealed several tumor properties contributing unique prognostic value, supporting the feasibility of a generalizable prognostic classification system in a diverse cohort., Conclusions: Our ML model demonstrates extensive reproducibility and online accessibility, utilizing routine imaging data rather than complex imaging protocols. This platform offers a unique approach for personalized patient management and clinical trial stratification in glioblastoma., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
20. An ERK5-PFKFB3 axis regulates glycolysis and represents a therapeutic vulnerability in pediatric diffuse midline glioma.
- Author
-
Casillo SM, Gatesman TA, Chilukuri A, Varadharajan S, Johnson BJ, David Premkumar DR, Jane EP, Plute TJ, Koncar RF, Stanton AJ, Biagi-Junior CAO, Barber CS, Halbert ME, Golbourn BJ, Halligan K, Cruz AF, Mansi NM, Cheney A, Mullett SJ, Land CV, Perez JL, Myers MI, Agrawal N, Michel JJ, Chang YF, Vaske OM, MichaelRaj A, Lieberman FS, Felker J, Shiva S, Bertrand KC, Amankulor N, Hadjipanayis CG, Abdullah KG, Zinn PO, Friedlander RM, Abel TJ, Nazarian J, Venneti S, Filbin MG, Gelhaus SL, Mack SC, Pollack IF, and Agnihotri S
- Subjects
- Animals, Child, Humans, Mice, Extracellular Signal-Regulated MAP Kinases, Glycolysis, Phosphofructokinase-2, Phosphoric Monoester Hydrolases, Signal Transduction, Glioma genetics, Histones genetics
- Abstract
Metabolic reprogramming in pediatric diffuse midline glioma is driven by gene expression changes induced by the hallmark histone mutation H3K27M, which results in aberrantly permissive activation of oncogenic signaling pathways. Previous studies of diffuse midline glioma with altered H3K27 (DMG-H3K27a) have shown that the RAS pathway, specifically through its downstream kinase, extracellular-signal-related kinase 5 (ERK5), is critical for tumor growth. Further downstream effectors of ERK5 and their role in DMG-H3K27a metabolic reprogramming have not been explored. We establish that ERK5 is a critical regulator of cell proliferation and glycolysis in DMG-H3K27a. We demonstrate that ERK5 mediates glycolysis through activation of transcription factor MEF2A, which subsequently modulates expression of glycolytic enzyme PFKFB3. We show that in vitro and mouse models of DMG-H3K27a are sensitive to the loss of PFKFB3. Multi-targeted drug therapy against the ERK5-PFKFB3 axis, such as with small-molecule inhibitors, may represent a promising therapeutic approach in patients with pediatric diffuse midline glioma., Competing Interests: Declaration of interests I.F.P. and S.A. have a utility use patent on ERK5 inhibitors for use in pediatric brain tumors., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
21. A novel ARIH1::BRAF fusion in a glioma.
- Author
-
Xu E, Stone SL, Zhong Y, Golenberg N, Qiu L, Abdullaev Z, Aldape K, Bagley L, Halpern CH, Amankulor N, and Nasrallah MP
- Subjects
- Humans, Proto-Oncogene Proteins B-raf genetics, Mutation genetics, Oncogene Proteins, Fusion genetics, Ubiquitin-Protein Ligases, Glioma genetics, Astrocytoma, Brain Neoplasms genetics
- Published
- 2023
- Full Text
- View/download PDF
22. TP53-PTEN-NF1 depletion in human brain organoids produces a glioma phenotype in vitro .
- Author
-
Singh SK, Wang Y, Habib A, Priyadarshini M, Kodavali CV, Chen A, Ma W, Wang J, Hameed NUF, Hu B, Fuller GN, Kulich SM, Amankulor N, Colen RR, Edwards LA, and Zinn PO
- Abstract
Glioblastoma (GBM) is fatal and the study of therapeutic resistance, disease progression, and drug discovery in GBM or glioma stem cells is often hindered by limited resources. This limitation slows down progress in both drug discovery and patient survival. Here we present a genetically engineered human cerebral organoid model with a cancer-like phenotype that could provide a basis for GBM-like models. Specifically, we engineered a doxycycline-inducible vector encoding shRNAs enabling depletion of the TP53, PTEN, and NF1 tumor suppressors in human cerebral organoids. Designated as inducible short hairpin-TP53-PTEN-NF1 (ish-TPN), doxycycline treatment resulted in human cancer-like cerebral organoids that effaced the entire organoid cytoarchitecture, while uninduced ish-TPN cerebral organoids recapitulated the normal cytoarchitecture of the brain. Transcriptomic analysis revealed a proneural GBM subtype. This proof-of-concept study offers a valuable resource for directly investigating the emergence and progression of gliomas within the context of specific genetic alterations in normal cerebral organoids., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2023 Singh, Wang, Habib, Priyadarshini, Kodavali, Chen, Ma, Wang, Hameed, Hu, Fuller, Kulich, Amankulor, Colen, Edwards and Zinn.)
- Published
- 2023
- Full Text
- View/download PDF
23. Super-enhancer hijacking drives ectopic expression of hedgehog pathway ligands in meningiomas.
- Author
-
Youngblood MW, Erson-Omay Z, Li C, Najem H, Coșkun S, Tyrtova E, Montejo JD, Miyagishima DF, Barak T, Nishimura S, Harmancı AS, Clark VE, Duran D, Huttner A, Avşar T, Bayri Y, Schramm J, Boetto J, Peyre M, Riche M, Goldbrunner R, Amankulor N, Louvi A, Bilgüvar K, Pamir MN, Özduman K, Kilic T, Knight JR, Simon M, Horbinski C, Kalamarides M, Timmer M, Heimberger AB, Mishra-Gorur K, Moliterno J, Yasuno K, and Günel M
- Subjects
- Humans, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Ligands, Signal Transduction, Meningioma genetics, Meningeal Neoplasms genetics
- Abstract
Hedgehog signaling mediates embryologic development of the central nervous system and other tissues and is frequently hijacked by neoplasia to facilitate uncontrolled cellular proliferation. Meningiomas, the most common primary brain tumor, exhibit Hedgehog signaling activation in 6.5% of cases, triggered by recurrent mutations in pathway mediators such as SMO. In this study, we find 35.6% of meningiomas that lack previously known drivers acquired various types of somatic structural variations affecting chromosomes 2q35 and 7q36.3. These cases exhibit ectopic expression of Hedgehog ligands, IHH and SHH, respectively, resulting in Hedgehog signaling activation. Recurrent tandem duplications involving IHH permit de novo chromatin interactions between super-enhancers within DIRC3 and a locus containing IHH. Our work expands the landscape of meningioma molecular drivers and demonstrates enhancer hijacking of Hedgehog ligands as a route to activate this pathway in neoplasia., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
24. Dissecting the tumor microenvironment of epigenetically driven gliomas: Opportunities for single-cell and spatial multiomics.
- Author
-
Sussman JH, Xu J, Amankulor N, and Tan K
- Abstract
Malignant gliomas are incurable brain neoplasms with dismal prognoses and near-universal fatality, with minimal therapeutic progress despite billions of dollars invested in research and clinical trials over the last 2 decades. Many glioma studies have utilized disparate histologic and genomic platforms to characterize the stunning genomic, transcriptomic, and immunologic heterogeneity found in gliomas. Single-cell and spatial omics technologies enable unprecedented characterization of heterogeneity in solid malignancies and provide a granular annotation of transcriptional, epigenetic, and microenvironmental states with limited resected tissue. Heterogeneity in gliomas may be defined, at the broadest levels, by tumors ostensibly driven by epigenetic alterations (IDH- and histone-mutant) versus non-epigenetic tumors (IDH-wild type). Epigenetically driven tumors are defined by remarkable transcriptional programs, immunologically distinct microenvironments, and incompletely understood topography (unique cellular neighborhoods and cell-cell interactions). Thus, these tumors are the ideal substrate for single-cell multiomic technologies to disentangle the complex intra-tumoral features, including differentiation trajectories, tumor-immune cell interactions, and chromatin dysregulation. The current review summarizes the applications of single-cell multiomics to existing datasets of epigenetically driven glioma. More importantly, we discuss future capabilities and applications of novel multiomic strategies to answer outstanding questions, enable the development of potent therapeutic strategies, and improve personalized diagnostics and treatment via digital pathology., Competing Interests: None., (© The Author(s) 2023. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
25. PHGDH-mediated endothelial metabolism drives glioblastoma resistance to chimeric antigen receptor T cell immunotherapy.
- Author
-
Zhang D, Li AM, Hu G, Huang M, Yang F, Zhang L, Wellen KE, Xu X, Conn CS, Zou W, Kahn M, Rhoades SD, Weljie AM, Fuchs SY, Amankulor N, Yoshor D, Ye J, Koumenis C, Gong Y, and Fan Y
- Subjects
- Animals, Mice, Humans, Phosphoglycerate Dehydrogenase metabolism, Cell Line, Tumor, Immunotherapy, Adoptive, T-Lymphocytes metabolism, Tumor Microenvironment, Glioblastoma therapy, Glioblastoma metabolism, Receptors, Chimeric Antigen
- Abstract
The efficacy of immunotherapy is limited by the paucity of T cells delivered and infiltrated into the tumors through aberrant tumor vasculature. Here, we report that phosphoglycerate dehydrogenase (PHGDH)-mediated endothelial cell (EC) metabolism fuels the formation of a hypoxic and immune-hostile vascular microenvironment, driving glioblastoma (GBM) resistance to chimeric antigen receptor (CAR)-T cell immunotherapy. Our metabolome and transcriptome analyses of human and mouse GBM tumors identify that PHGDH expression and serine metabolism are preferentially altered in tumor ECs. Tumor microenvironmental cues induce ATF4-mediated PHGDH expression in ECs, triggering a redox-dependent mechanism that regulates endothelial glycolysis and leads to EC overgrowth. Genetic PHGDH ablation in ECs prunes over-sprouting vasculature, abrogates intratumoral hypoxia, and improves T cell infiltration into the tumors. PHGDH inhibition activates anti-tumor T cell immunity and sensitizes GBM to CAR T therapy. Thus, reprogramming endothelial metabolism by targeting PHGDH may offer a unique opportunity to improve T cell-based immunotherapy., Competing Interests: Declaration of interests Y.F. and D.Z. are inventors on a patent application related to PHGDH inhibition for cancer immunotherapy., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Machine Learning Identification of Immunotherapy Targets in Low-Grade Glioma Using RNA Sequencing Expression Data.
- Author
-
Agarwal P, Beale OM, Zhang X, Sandlesh P, Jaman E, and Amankulor N
- Subjects
- Humans, Immunotherapy, Isocitrate Dehydrogenase genetics, Machine Learning, RNA, Sequence Analysis, RNA, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms therapy, Glioma genetics, Glioma metabolism, Glioma therapy
- Abstract
Objective: Immunotherapy has revolutionized cancer treatment in the past decade, but significant hurdles remain. Human studies with immune checkpoint inhibitors targeting programmed cell death protein have demonstrated suboptimal efficacy in the setting of low-grade gliomas (LGGs). Identification of mechanisms leading to inadequate anti-tumor immunity is paramount. The current study evaluates and validates barriers to immunotherapy using a novel machine learning algorithm., Methods: We utilized The Cancer Genome Atlas (TCGA) to generate expression levels of 28 immune genes related to known immunotherapeutic targets or lymphocyte cytolytic activity. We created training and testing groups and 3 machine learning models to determine the genes most highly correlated to cytolytic activity (CYT). The 3 models were run through multiple regression by exhaustive selection, LASSO, and random forest. We validated computational results by comparing expression of pertinent genes in patient-derived glioma samples., Results: Our models demonstrated linearity, a low mean-squared error, and consistent results with respect to the most important variables. Expression of ICOS, IDO1, and CD40 were the most important variables in all models and demonstrated positive correlation with CYT. Other variables included TIGIT and CD137. Genetic analysis from 3 IDH-mutants (IDHm) and 3 IDH-wild type (IDHwt) patient-derived glioma samples validated TCGA data and demonstrated lower levels of CYT in IDHm gliomas compared with IDHwt., Conclusions: This novel methodology has elucidated 3 potential targets for immunotherapy development in LGGs. We also demonstrated a novel method of analyzing data using advanced statistical techniques that can be further used in developing treatments for other diseases as well., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling.
- Author
-
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, and Zinn PO
- Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Beiriger, Habib, Jovanovich, Kodavali, Edwards, Amankulor and Zinn.)
- Published
- 2022
- Full Text
- View/download PDF
28. The dominant TP53 hotspot mutation in IDH -mutant astrocytoma, R273C, has distinctive pathologic features and sex-specific prognostic implications.
- Author
-
Marker DF, Agnihotri S, Amankulor N, Murdoch GH, and Pearce TM
- Abstract
Background: Infiltrative astrocytic tumors with and without isocitrate dehydrogenase (IDH) mutation frequently contain mutations in the TP53 tumor suppressor gene. Disruption of normal p53 protein activity confers neoplastic cells with a number of oncogenic properties and is a common feature of aggressive malignancies. However, the high prevalence of TP53 mutation and its pathogenic role in IDH-mutant (IDHmut) astrocytoma is not well understood., Methods: We performed a retrospective analysis of molecular and clinical data from patients with IDHmut astrocytoma at the University of Pittsburgh Medical Center between 2015 and 2019 as our initial cohort. We validated and expanded our findings using molecular and clinical data from The Cancer Genome Atlas., Results: We show that the TP53 mutational spectrum in IDHmut astrocytomas is dominated by a single hotspot mutation that codes for the R273C amino acid change. This mutation is not enriched in IDH-wildtype astrocytomas. The high prevalence of TP53
R273C mutation is not readily explained by known mutagenic mechanisms, and TP53R273C mutant tumors have lower transcriptional levels of proliferation-related genes compared to IDHmut astrocytomas harboring other forms of mutant p53. Despite lower proliferation, TP53R273C mutant tumors tend to progress more quickly and have a shorter overall survival than those with other TP53 mutations, particularly in male patients., Conclusions: Our findings suggest that compared to other TP53 mutations, IDHmut astrocytomas may select for TP53R273C mutations during tumorigenesis. The genotype, sex, and mutation-specific findings are clinically relevant and should prompt further investigation of TP53R273C ., (© The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)- Published
- 2021
- Full Text
- View/download PDF
29. The Evolving Role of Induced Pluripotent Stem Cells and Cerebral Organoids in Treating and Modeling Neurosurgical Diseases.
- Author
-
Jovanovich N, Habib A, Kodavali C, Edwards L, Amankulor N, and Zinn PO
- Subjects
- Animals, Central Nervous System Diseases surgery, Humans, Models, Biological, Central Nervous System Diseases physiopathology, Cerebral Cortex physiopathology, Induced Pluripotent Stem Cells physiology, Neurosurgical Procedures, Organoids physiopathology
- Abstract
Over the past decade, the use of induced pluripotent stem cells (IPSCs), as both direct therapeutics and building blocks for 3D in vitro models, has exhibited exciting potential in both helping to elucidate pathogenic mechanisms and treating diseases relevant to neurosurgery. Transplantation of IPSCs is being studied in neurological injuries and diseases, such as spinal cord injury and Parkinson's disease, whose clinical manifestations stem from underlying neuronal and/or axonal degeneration. Both animal models and clinical trials have shown that IPSCs have the ability to regenerate damaged neural tissue. Such evidence makes IPSCs a potentially promising therapeutic modality for patients who suffer from these neurological injuries/diseases. In addition, the cerebral organoid, a 3D assembly of IPSC aggregates that develops heterogeneous brain regions, has become the first in vitro model to closely recapitulate the complexity of the brain extracellular matrix, a 3-dimensional network of molecules that structurally and biochemically support neighboring cells. Cerebral organoids have become an exciting prospect for modeling and testing drug susceptibility of brain tumors, such as glioblastoma and metastatic brain cancer. As patient-derived organoid models are becoming more faithful to the brain, they are becoming an increasingly accurate substitute for patient clinical trials; such patient-less trials would protect the patient from potentially ineffective drugs, and speed up trial results and optimize cost. In this review, we aim to describe the role of IPSCs and cerebral organoids in treating and modeling diseases that are relevant to neurosurgery., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. Re-evaluating Biopsy for Recurrent Glioblastoma: A Position Statement by the Christopher Davidson Forum Investigators.
- Author
-
Nduom EK, Gephart MH, Chheda MG, Suva ML, Amankulor N, Battiste JD, Campian JL, Dacey RG, Das S, Fecci PE, Hadjipanayis CG, Hoang KB, Jalali A, Orringer D, Patel AJ, Placantonakis D, Rodriguez A, Yang I, Yu JS, Zipfel GJ, Dunn GP, Leuthardt EC, and Kim AH
- Subjects
- Biopsy, Humans, Mutation, Neoplasm Recurrence, Local diagnosis, Brain Neoplasms diagnosis, Brain Neoplasms genetics, Brain Neoplasms therapy, Glioblastoma diagnosis, Glioblastoma genetics, Glioblastoma therapy
- Abstract
Patients with glioblastoma (GBM) need bold new approaches to their treatment, yet progress has been hindered by a relative inability to dynamically track treatment response, mechanisms of resistance, evolution of targetable mutations, and changes in mutational burden. We are writing on behalf of a multidisciplinary group of academic neuro-oncology professionals who met at the collaborative Christopher Davidson Forum at Washington University in St Louis in the fall of 2019. We propose a dramatic but necessary change to the routine management of patients with GBM to advance the field: to routinely biopsy recurrent GBM at the time of presumed recurrence. Data derived from these samples will identify true recurrence vs treatment effect, avoid treatments with little chance of success, enable clinical trial access, and aid in the scientific advancement of our understanding of GBM., (© Congress of Neurological Surgeons 2021.)
- Published
- 2021
- Full Text
- View/download PDF
31. Associations of meningioma molecular subgroup and tumor recurrence.
- Author
-
Youngblood MW, Miyagishima DF, Jin L, Gupte T, Li C, Duran D, Montejo JD, Zhao A, Sheth A, Tyrtova E, Özduman K, Iacoangeli F, Peyre M, Boetto J, Pease M, Avşar T, Huttner A, Bilguvar K, Kilic T, Pamir MN, Amankulor N, Kalamarides M, Erson-Omay EZ, Günel M, and Moliterno J
- Subjects
- Epigenomics, Genomics, Humans, Kruppel-Like Factor 4, Male, Neoplasm Recurrence, Local genetics, Retrospective Studies, Meningeal Neoplasms genetics, Meningioma genetics
- Abstract
Background: We and others have identified mutually exclusive molecular subgroups of meningiomas; however, the implications of this classification for clinical prognostication remain unclear. Integrated genomic and epigenomic analyses implicate unique oncogenic processes associated with each subgroup, suggesting the potential for divergent clinical courses. The aim of this study was to understand the associated clinical outcomes of each subgroup, as this could optimize treatment for patients., Methods: We analyzed outcome data for 469 meningiomas of known molecular subgroup, including extent of resection, postoperative radiation, surveillance imaging, and time to recurrence, when applicable. Statistical relationships between outcome variables and subgroup were assessed. Features previously associated with recurrence were further investigated after stratification by subgroup. We used Kaplan-Meier analyses to compare progression-free survival, and identified factors significantly associated with recurrence using Cox proportional hazards modeling., Results: Meningioma molecular subgroups exhibited divergent clinical courses at 2 years of follow-up, with several aggressive subgroups (NF2, PI3K, HH, tumor necrosis factor receptor-associated factor 7 [TRAF7]) recurring at an average rate of 22 times higher than others (KLF4, POLR2A, SMARCB1). PI3K-activated tumors recurred earlier than other subgroups but had intermediate long-term outcome. Among low-grade tumors, HH and TRAF7 meningiomas exhibited elevated recurrence compared with other subgroups. Recurrence of NF2 tumors was associated with male sex, high grade, and elevated Ki-67. Multivariate analysis identified molecular subgroup as an independent predictor of recurrence, along with grade and previous recurrence., Conclusion: We describe distinct clinical outcomes and recurrence rates associated with meningioma molecular subgroups. Our findings emphasize the importance of genomic characterization to guide postoperative management decisions for meningiomas., (© The Author(s) 2020. 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
- 2021
- Full Text
- View/download PDF
32. Oncolytic HSV Vectors and Anti-Tumor Immunity.
- Author
-
Glorioso JC, Cohen JB, Goins WF, Hall B, Jackson JW, Kohanbash G, Amankulor N, Kaur B, Caligiuri MA, Chiocca EA, Holland EC, and Quéva C
- Subjects
- Adaptive Immunity drug effects, Adaptive Immunity immunology, Animals, Humans, Immune Checkpoint Inhibitors pharmacology, T-Lymphocytes, Cytotoxic immunology, Immune Checkpoint Inhibitors immunology, Neoplasms immunology, Neoplasms therapy, Oncolytic Viruses immunology, Simplexvirus immunology
- Abstract
The therapeutic promise of oncolytic viruses (OVs) rests on their ability to both selectively kill tumor cells and induce anti-tumor immunity. The potential of tumors to be recognized and eliminated by an effective anti-tumor immune response has been spurred on by the discovery that immune checkpoint inhibition can overcome tumor-specific cytotoxic T cell (CTL) exhaustion and provide durable responses in multiple tumor indications. OV-mediated tumor destruction is now recognized as a powerful means to assist in the development of anti-tumor immunity for two important reasons: (i) OVs, through the elicitation of an anti-viral response and the production of type I interferon, are potent stimulators of inflammation and can be armed with transgenes to further enhance anti-tumor immune responses; and (ii) lytic activity can promote the release of tumor-associated antigens (TAAs) and tumor neoantigens that function as in situ tumor-specific vaccines to elicit adaptive immunity. Oncolytic herpes simplex viruses (oHSVs) are among the most widely studied OVs for the treatment of solid malignancies, and Amgen's oHSV Imlygic® for the treatment of melanoma is the only OV approved in major markets. Here we describe important biological features of HSV that make it an attractive OV, clinical experience with HSV-based vectors, and strategies to increase applicability to cancer treatment.
- Published
- 2021
- Full Text
- View/download PDF
33. A contemporary update on glioblastoma: molecular biology, current management, and a vision towards bio-adaptable personalized care.
- Author
-
Habib A, Pease M, Kodavali CV, Amankulor N, and Zinn PO
- Subjects
- Brain Neoplasms pathology, Combined Modality Therapy, Disease Management, Glioblastoma pathology, Humans, Brain Neoplasms therapy, Glioblastoma therapy, Immunotherapy methods, Molecular Targeted Therapy methods, Precision Medicine
- Abstract
Introduction: Glioblastoma (GBM) is the most fatal brain tumor in adults. Current survival rates of GBM remain below 2 years due to GBM's aggressive cellular migration and genetically driven treatment escape pathways. Despite our rapidly increasing understanding of GBM biology, earlier diagnoses, and refined surgical techniques, only moderate survival benefits have been achieved. Nonetheless, the pressing need for better survival rates has brought forward a multitude of newer therapeutic approaches and opened the door for potential personalization of these modalities in the near future., Methods: We reviewed the published literature discussing the current state of knowledge regarding GBM biology and therapy and summarized the information that may point toward future personalized therapeutic strategies., Results: Several novel modalities such as oncolytic viruses, targeted immune, and molecular therapies, and tumor treating fields have been introduced. To date, there is no single treatment modality for GBM, but rather a wide spectrum of combined modalities that address intratumoral cellular and genetic variabilities. While the current state of GBM research and clinical trial landscape may hold promise, current literature lacks any fruitful progress towards personalized GBM therapy., Conclusion: In this review, we are discussing our recent knowledge of the GBM genetic biologic landscape and the current advances in therapy, as well as providing a blueprint for an envisioned GBM management paradigm that should be personalized and adaptable to accommodate each patient's diverse genetic variations and therapy response/escape patterns.
- Published
- 2021
- Full Text
- View/download PDF
34. Long-term control of leptomeningeal disease after radiation therapy and nivolumab in a metastatic melanoma patient.
- Author
-
Wu RC, Newman W, Patanowitz L, Branstetter BF, Amankulor N, and Tarhini AA
- Subjects
- Adult, Brain Neoplasms genetics, Brain Neoplasms pathology, Fatal Outcome, Humans, Male, Melanoma genetics, Melanoma pathology, Mutation genetics, Programmed Cell Death 1 Receptor antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Remission Induction, Spinal Cord Diseases genetics, Spinal Cord Diseases pathology, Antineoplastic Agents, Immunological therapeutic use, Brain Neoplasms therapy, Melanoma therapy, Meninges pathology, Nivolumab therapeutic use, Radioimmunotherapy methods, Spinal Cord Diseases therapy
- Abstract
Background: Leptomeningeal disease (LMD) from melanoma is rapidly fatal with median overall survival between 6.9 weeks and 3.5 months. It is not known whether immune checkpoint inhibitors have a role in treating LMD. Case presentation: We report a 33-year-old male patient who developed LMD from a BRAF V600E-mutated melanoma brain metastasis, despite prior treatment with surgical resection, radiotherapy and dabrafenib/trametinib. He underwent whole brain radiotherapy with stereotactic radiotherapy to the lumbosacral spine, and was started on nivolumab, which led to prolonged remission lasting 2 years and 3 months, before disease progression and death. Conclusion: This is the first case report to highlight a potential long-term efficacy of radiotherapy and anti-PD-1 immunotherapy, in treating LMD from metastatic melanoma that is resistant to targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
35. Correlations between genomic subgroup and clinical features in a cohort of more than 3000 meningiomas.
- Author
-
Youngblood MW, Duran D, Montejo JD, Li C, Omay SB, Özduman K, Sheth AH, Zhao AY, Tyrtova E, Miyagishima DF, Fomchenko EI, Hong CS, Clark VE, Riche M, Peyre M, Boetto J, Sohrabi S, Koljaka S, Baranoski JF, Knight J, Zhu H, Pamir MN, Avşar T, Kilic T, Schramm J, Timmer M, Goldbrunner R, Gong Y, Bayri Y, Amankulor N, Hamilton RL, Bilguvar K, Tikhonova I, Tomak PR, Huttner A, Simon M, Krischek B, Kalamarides M, Erson-Omay EZ, Moliterno J, and Günel M
- Abstract
Objective: Recent large-cohort sequencing studies have investigated the genomic landscape of meningiomas, identifying somatic coding alterations in NF2, SMARCB1, SMARCE1, TRAF7, KLF4, POLR2A, BAP1, and members of the PI3K and Hedgehog signaling pathways. Initial associations between clinical features and genomic subgroups have been described, including location, grade, and histology. However, further investigation using an expanded collection of samples is needed to confirm previous findings, as well as elucidate relationships not evident in smaller discovery cohorts., Methods: Targeted sequencing of established meningioma driver genes was performed on a multiinstitution cohort of 3016 meningiomas for classification into mutually exclusive subgroups. Relevant clinical information was collected for all available cases and correlated with genomic subgroup. Nominal variables were analyzed using Fisher's exact tests, while ordinal and continuous variables were assessed using Kruskal-Wallis and 1-way ANOVA tests, respectively. Machine-learning approaches were used to predict genomic subgroup based on noninvasive clinical features., Results: Genomic subgroups were strongly associated with tumor locations, including correlation of HH tumors with midline location, and non-NF2 tumors in anterior skull base regions. NF2 meningiomas were significantly enriched in male patients, while KLF4 and POLR2A mutations were associated with female sex. Among histologies, the results confirmed previously identified relationships, and observed enrichment of microcystic features among "mutation unknown" samples. Additionally, KLF4-mutant meningiomas were associated with larger peritumoral brain edema, while SMARCB1 cases exhibited elevated Ki-67 index. Machine-learning methods revealed that observable, noninvasive patient features were largely predictive of each tumor's underlying driver mutation., Conclusions: Using a rigorous and comprehensive approach, this study expands previously described correlations between genomic drivers and clinical features, enhancing our understanding of meningioma pathogenesis, and laying further groundwork for the use of targeted therapies. Importantly, the authors found that noninvasive patient variables exhibited a moderate predictive value of underlying genomic subgroup, which could improve with additional training data. With continued development, this framework may enable selection of appropriate precision medications without the need for invasive sampling procedures.
- Published
- 2019
- Full Text
- View/download PDF
36. GBM-Targeted oHSV Armed with Matrix Metalloproteinase 9 Enhances Anti-tumor Activity and Animal Survival.
- Author
-
Sette P, Amankulor N, Li A, Marzulli M, Leronni D, Zhang M, Goins WF, Kaur B, Bolyard C, Cripe TP, Yu J, Chiocca EA, Glorioso JC, and Grandi P
- Abstract
The use of mutant strains of oncolytic herpes simplex virus (oHSV) in early-phase human clinical trials for the treatment of glioblastoma multiforme (GBM) has proven safe, but limited efficacy suggests that more potent vector designs are required for effective GBM therapy. Inadequate vector performance may derive from poor intratumoral vector replication and limited spread to uninfected cells. Vector replication may be impaired by mutagenesis strategies to achieve vector safety, and intratumoral virus spread may be hampered by vector entrapment in the tumor-specific extracellular matrix (ECM) that in GBM is composed primarily of type IV collagen. In this report, we armed our previously described epidermal growth factor receptor (EGFR)vIII-targeted, neuronal microRNA-sensitive oHSV with a matrix metalloproteinase (MMP9) to improve intratumoral vector distribution. We show that vector-expressed MMP9 enhanced therapeutic efficacy and long-term animal survival in a GBM xenograft model., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
37. Arming oHSV with ULBP3 drives abscopal immunity in lymphocyte-depleted glioblastoma.
- Author
-
Wirsching HG, Zhang H, Szulzewsky F, Arora S, Grandi P, Cimino PJ, Amankulor N, Campbell JS, McFerrin L, Pattwell SS, Ene C, Hicks A, Ball M, Yan J, Zhang J, Kumasaka D, Pierce RH, Weller M, Finer M, Quéva C, Glorioso JC, Houghton AM, and Holland EC
- Subjects
- Animals, Antigen Presentation genetics, Antineoplastic Agents, Immunological pharmacology, Brain pathology, Brain Neoplasms genetics, Brain Neoplasms immunology, Brain Neoplasms mortality, Cell Line, Tumor, Combined Modality Therapy methods, Disease Models, Animal, Female, GPI-Linked Proteins genetics, GPI-Linked Proteins immunology, Gene Expression Regulation, Neoplastic immunology, Glioblastoma genetics, Glioblastoma immunology, Glioblastoma mortality, Humans, Intercellular Signaling Peptides and Proteins genetics, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase immunology, Kaplan-Meier Estimate, Male, Mice, Mice, Transgenic, Oncolytic Viruses genetics, Primary Cell Culture, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Recombinant Proteins genetics, Recombinant Proteins immunology, Simplexvirus genetics, Up-Regulation, Antineoplastic Agents, Immunological therapeutic use, Brain Neoplasms therapy, Glioblastoma therapy, Intercellular Signaling Peptides and Proteins immunology, Oncolytic Virotherapy methods, Oncolytic Viruses immunology, Simplexvirus immunology
- Abstract
Oncolytic viruses induce local tumor destruction and inflammation. Whether virotherapy can also overcome immunosuppression in noninfected tumor areas is under debate. To address this question, we have explored immunologic effects of oncolytic herpes simplex viruses (oHSVs) in a genetically engineered mouse model of isocitrate dehydrogenase (IDH) wild-type glioblastoma, the most common and most malignant primary brain tumor in adults. Our model recapitulates the genomics, the diffuse infiltrative growth pattern, and the extensive macrophage-dominant immunosuppression of human glioblastoma. Infection with an oHSV that was armed with a UL16-binding protein 3 (ULBP3) expression cassette inhibited distant tumor growth in the absence of viral spreading (abscopal effect) and yielded accumulation of activated macrophages and T cells. There was also abscopal synergism of oHSVULBP3 with anti-programmed cell death 1 (anti-PD-1) against distant, uninfected tumor areas; albeit consistent with clinical trials in patients with glioblastoma, monotherapy with anti-PD-1 was ineffective in our model. Arming oHSV with ULBP3 led to upregulation of antigen processing and presentation gene sets in myeloid cells. The cognate ULBP3 receptor NKG2D, however, is not present on myeloid cells, suggesting a noncanonical mechanism of action of ULBP3. Overall, the myeloid-dominant, anti-PD-1-sensitive abscopal effect of oHSVULBP3 warrants further investigation in patients with IDH wild-type glioblastoma.
- Published
- 2019
- Full Text
- View/download PDF
38. Elevated Na/H exchanger 1 (SLC9A1) emerges as a marker for tumorigenesis and prognosis in gliomas.
- Author
-
Guan X, Luo L, Begum G, Kohanbash G, Song Q, Rao A, Amankulor N, Sun B, Sun D, and Jia W
- Subjects
- Animals, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glioma drug therapy, Glioma metabolism, Glioma pathology, Guanidines administration & dosage, Guanidines pharmacology, Humans, Male, Mice, Mice, Inbred C57BL, Neoplasm Grading, Neoplasm Transplantation, Prognosis, Sequence Analysis, RNA, Sodium-Hydrogen Exchanger 1 metabolism, Sulfones administration & dosage, Sulfones pharmacology, Survival Analysis, Brain Neoplasms genetics, Glioma genetics, Sodium-Hydrogen Exchanger 1 genetics, Up-Regulation
- Abstract
Background: Sodium/hydrogen exchanger 1 (NHE1), encoded by the SLC9A1 gene (SoLute Carrier family 9A1) in humans, is the main H
+ efflux mechanism in maintaining alkaline intracellular pH (pHi ) and Warburg effects in glioma. However, to date, there are no clinical studies exploring pharmacological inhibition of NHE1 protein in cancer treatment. In this study, we investigated NHE1 expression in gliomas and its relationship with glioma clinical outcome., Methods: The Chinese Glioma Genome Atlas (CGGA) dataset containing transcriptome sequencing data of 325 glioma samples and the Cancer Genome Atlas (TCGA) with 698 glioma mRNAseq data were analyzed in this study. Mouse SB28 and GL26 intracranial syngeneic glioma models in C57BL/6 J mice were established to investigate NHE1 expression and impact of NHE1 protein inhibition with its inhibitor HOE642 on tumorigenesis and anti-PD1 therapy. Tumor angiogenesis, immunogenicity, and progression were assessed by immunofluorescence staining and flow cytometric profiling., Results: Analysis of SLC9A1 mRNA expression in two data sets, CGGA and TCGA, reveals significantly higher SLC9A1 mRNA levels in higher grade gliomas. The SLC9A1 mRNA expression was especially enriched in isocitrate dehydrogenase (IDH)1/2 wild-type glioblastoma (GBM) and in mesenchymal glioma subtypes. Worsened survival probabilities were correlated with the elevated SLC9A1 mRNA levels in gliomas. The underlying mechanisms include promoting angiogenesis, and extracellular matrix remodeling. Increased SLC9A1 mRNA expression was also associated with tumor-associated macrophage accumulation. NHE1 inhibitor HOE642 reduced glioma volume, invasion, and prolonged overall survival in mouse glioma models. Blockade of NHE1 protein also stimulated immunogenic tumor microenvironment via activating CD8 T-cell accumulation, increasing expression of interferon-gamma (Ifng), and sensitized animals to anti-PD-1 therapy., Conclusion: Our findings strongly suggest that NHE1 protein emerges as a marker for tumorigenesis and prognosis in glioma. Blocking NHE1 protein is a novel strategy for adjuvant anti-cancer therapies.- Published
- 2018
- Full Text
- View/download PDF
39. Core Entrustable Professional Activities in Clinical Pharmacology for Entering Residency: Value of Interprofessional Health-Care Teams in Medication Prescribing and Medication Error Prevention.
- Author
-
Cohen LJ, Donnenberg VS, Wiernik PH, Newman WC, and Amankulor N
- Subjects
- Clinical Competence, Comprehension, Cooperative Behavior, Drug Prescriptions standards, Health Occupations standards, Humans, Interdisciplinary Communication, Internship and Residency, Interprofessional Relations, Physician-Patient Relations, Health Occupations education, Patient Safety standards, Pharmacology, Clinical education
- Abstract
In recent years, health care has been increasingly delivered by interprofessional teams in the inpatient, outpatient, and transition-of-care arenas. For many reasons, effective communication between patient-centered care teams and patients is critically important in order to optimize care, ensure patient safety, and prevent medical and medication misadventures. In rapid-paced, high-stress medical environments, it is especially important to carefully evaluate the causes of all misadventures in a manner that avoids assigning blame and identifies the root causes and, through team activity, leads to development of remedies that reduce the likelihood of future misadventures. Using a series of illustrative cases, this paper seeks to bring attention to these issues and provide insights regarding some tools developed to assist in improving patient safety and effective team communication., (© 2018, The American College of Clinical Pharmacology.)
- Published
- 2018
- Full Text
- View/download PDF
40. The Utility of Early Postoperative Head Computed Tomography in Brain Tumor Surgery: A Retrospective Analysis of 755 Cases.
- Author
-
Alkhalili K, Zenonos G, Tataryn Z, Amankulor N, and Engh J
- Subjects
- Adult, Aged, Brain Neoplasms surgery, Craniotomy, Female, Humans, Male, Middle Aged, Postoperative Period, Retrospective Studies, Tomography, X-Ray Computed, Brain Neoplasms diagnostic imaging
- Abstract
Objective: Scheduled early postoperative computed tomography (EPOCT) after craniotomy for brain tumor resection is standard at many institutions. We analyzed utility of preplanned EPOCT after elective craniotomy for brain tumor resection., Methods: We retrospectively analyzed 755 brain tumor resections for which EPOCT was performed within 4 hours of surgery. Postoperative clinical neurologic examination results were classified into expected (baseline or predicted postoperative examination), changed (from baseline examination), and unreliable (sedated or baseline comatose patient). Scans were analyzed for unexpected and/or worrisome findings (e.g., hemorrhagic or ischemic stroke). In cases of unexpected findings, management changes were correlated to patient's neurologic examination. Demographic information, tumor histology, and tumor location were analyzed to determine risk factors for unexpected findings., Results: Rate of unexpected EPOCT findings was 4.1%. Patients with expected postoperative examinations were at significantly lower risk of abnormal findings (odds ratio [OR] = 0.074, P < 0.001). Patients with intraventricular tumors (OR = 5.7, P = 0.001) were at higher risk compared with patients with metastatic tumors (OR = 0.24, P = 0.06). No unexpected EPOCT findings led to management changes in patients with expected postoperative neurologic examinations. All unexpected EPOCT findings in patients with changed postoperative neurologic examinations led to management changes. Patients with nonreliable neurologic examinations were at significantly higher risk for unexpected findings on EPOCT (OR = 6.33, P < 0.001) and subsequent management changes., Conclusions: Routine EPOCT is not indicated for patients undergoing brain tumor resection if postoperative neurologic examination is unchanged, as imaging is unlikely to result in management changes. EPOCT should be obtained in all patients with worrisome changes in examination or nonreliable examinations, as both groups have high rates of unexpected findings on imaging that lead to management changes., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. Genomic analysis of 220 CTCLs identifies a novel recurrent gain-of-function alteration in RLTPR (p.Q575E).
- Author
-
Park J, Yang J, Wenzel AT, Ramachandran A, Lee WJ, Daniels JC, Kim J, Martinez-Escala E, Amankulor N, Pro B, Guitart J, Mendillo ML, Savas JN, Boggon TJ, and Choi J
- Subjects
- Amino Acid Sequence, Amino Acid Substitution genetics, Base Sequence, Genome, Human, HEK293 Cells, Humans, Jurkat Cells, Microfilament Proteins chemistry, Mutation genetics, NF-kappa B metabolism, Oncogenes, Receptors, Antigen, T-Cell metabolism, Sequence Analysis, DNA, Signal Transduction genetics, rhoA GTP-Binding Protein genetics, Genomics, Lymphoma, T-Cell, Cutaneous genetics, Microfilament Proteins genetics
- Abstract
Cutaneous T-cell lymphoma (CTCL) is an incurable non-Hodgkin lymphoma of the skin-homing T cell. In early-stage disease, lesions are limited to the skin, but in later-stage disease, the tumor cells can escape into the blood, the lymph nodes, and at times the visceral organs. To clarify the genomic basis of CTCL, we performed genomic analysis of 220 CTCLs. Our analyses identify 55 putative driver genes, including 17 genes not previously implicated in CTCL. These novel mutations are predicted to affect chromatin ( BCOR , KDM6A , SMARCB1 , TRRAP ), immune surveillance ( CD58 , RFXAP ), MAPK signaling ( MAP2K1 , NF1 ), NF-κB signaling ( PRKCB , CSNK1A1 ), PI-3-kinase signaling ( PIK3R1 , VAV1 ), RHOA/cytoskeleton remodeling ( ARHGEF3 ), RNA splicing ( U2AF1 ), T-cell receptor signaling ( PTPRN2 , RLTPR ), and T-cell differentiation ( RARA ). Our analyses identify recurrent mutations in 4 genes not previously identified in cancer. These include CK1α (encoded by CSNK1A1 ) (p.S27F; p.S27C), PTPRN2 (p.G526E), RARA (p.G303S), and RLTPR (p.Q575E). Last, we functionally validate CSNK1A1 and RLTPR as putative oncogenes. RLTPR encodes a recently described scaffolding protein in the T-cell receptor signaling pathway. We show that RLTPR (p.Q575E) increases binding of RLTPR to downstream components of the NF-κB signaling pathway, selectively upregulates the NF-κB pathway in activated T cells, and ultimately augments T-cell-receptor-dependent production of interleukin 2 by 34-fold. Collectively, our analysis provides novel insights into CTCL pathogenesis and elucidates the landscape of potentially targetable gene mutations., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
42. IDH mutant gliomas escape natural killer cell immune surveillance by downregulation of NKG2D ligand expression.
- Author
-
Zhang X, Rao A, Sette P, Deibert C, Pomerantz A, Kim WJ, Kohanbash G, Chang Y, Park Y, Engh J, Choi J, Chan T, Okada H, Lotze M, Grandi P, and Amankulor N
- Subjects
- Antineoplastic Agents pharmacology, Azacitidine analogs & derivatives, Azacitidine pharmacology, Brain Neoplasms genetics, Brain Neoplasms immunology, DNA Methylation, Decitabine, Down-Regulation, GPI-Linked Proteins biosynthesis, Glioma genetics, Glioma immunology, Humans, Immunologic Surveillance, Killer Cells, Natural immunology, Mutation, Real-Time Polymerase Chain Reaction, Transcriptome, Tumor Escape drug effects, Brain Neoplasms pathology, Gene Expression Regulation, Neoplastic genetics, Glioma pathology, Intercellular Signaling Peptides and Proteins biosynthesis, Isocitrate Dehydrogenase genetics, Tumor Escape genetics
- Abstract
Background: Diffuse gliomas are poorly immunogenic, fatal brain tumors. The basis for insufficient antitumor immunity in diffuse gliomas is unknown. Gain-of-function mutations in isocitrate dehydrogenases (IDH1 and IDH2) promote diffuse glioma formation through epigenetic reprogramming of a number of genes, including immune-related genes. Here, we identify epigenetic dysregulation of natural killer (NK) cell ligand genes as significant contributors to immune escape in glioma., Methods: We analyzed the database of The Cancer Genome Atlas for immune gene expression patterns in IDH mutant or wild-type gliomas and identified differentially expressed immune genes. NKG2D ligand expression levels and NK cell-mediated lysis were measured in IDH mutant and wild-type patient-derived glioma stem cells and genetically engineered astrocytes. Finally, we assessed the impact of hypomethylating agent 5-aza-2'deoxycytodine (decitabine) as a potential NK cell sensitizing agent in IDH mutant cells., Results: IDH mutant glioma stemlike cell lines exhibited significantly lower expression of NKG2D ligands compared with IDH wild-type cells. Consistent with these findings, IDH mutant glioma cells and astrocytes are resistant to NK cell-mediated lysis. Decitabine increases NKG2D ligand expression and restores NK-mediated lysis of IDH mutant cells in an NKG2D-dependent manner., Conclusions: IDH mutant glioma cells acquire resistance to NK cells through epigenetic silencing of NKG2D ligands ULBP1 and ULBP3. Decitabine-mediated hypomethylation restores ULBP1 and ULBP3 expression in IDH mutant glioma cells and may provide a clinically useful method to sensitize IDH mutant gliomas to NK cell-mediated immune surveillance in patients with IDH mutated diffuse gliomas., (© The Author(s) 2016. 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
- 2016
- Full Text
- View/download PDF
43. Hyalinizing Clear Cell Carcinoma with Biopsy-Proven Spinal Metastasis: Case Report and Review of Literature.
- Author
-
Newman WC, Williams L, Duvvuri U, Clump DA 2nd, and Amankulor N
- Subjects
- Aged, Diagnosis, Differential, Female, Genetic Testing, Humans, Salivary Gland Neoplasms genetics, Sarcoma, Clear Cell genetics, Spinal Neoplasms genetics, Salivary Gland Neoplasms pathology, Sarcoma, Clear Cell pathology, Sarcoma, Clear Cell secondary, Spinal Neoplasms pathology, Spinal Neoplasms secondary
- Abstract
Background: Hyalinizing clear cell carcinoma (HCCC) is a rare epithelial malignant neoplasm typically arising from the minor salivary glands. Although it has been described as a benign neoplasm, there are increasing reports of malignant features and metastases to the lungs; we present the first case of biopsy-proven spinal metastases from HCCC and an overview of the literature., Case Description: This is a single-patient case report in which we used immunohistochemistry and fluorescence in situ hybridization for Ewing sarcoma breakpoint region 1 translocation to confirm the diagnosis of HCCC in a spinal metastasis. The diagnosis of metastatic HCCC was confirmed on the basis of histopathology, immunohistochemistry, and fluorescence in situ hybridization studies., Conclusions: We present the first pathologically confirmed case of a spinal metastasis in HCCC. As increasing of metastatic HCCC arise, a reconsideration of HCCC as a potentially high-grade disease seems increasingly necessary as it may impact the current treatment paradigm., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Ommaya reservoir with ventricular catheter placement for chemotherapy with frameless and pinless electromagnetic surgical neuronavigation.
- Author
-
Weiner GM, Chivukula S, Chen CJ, Ding D, Engh JA, and Amankulor N
- Subjects
- Adult, Aged, Aged, 80 and over, Drug Delivery Systems methods, Electromagnetic Fields, Female, Humans, Hydrocephalus etiology, Male, Middle Aged, Retrospective Studies, Tomography, X-Ray Computed methods, Catheters, Hydrocephalus therapy, Neuronavigation, Neurosurgical Procedures methods
- Abstract
Background: Accuracy in Ommaya reservoir catheter placement is critical to chemotherapy infusion. Most frameless image guidance is light emitting diode (LED) based, requiring a direct line of communication between instrument and tracker, limiting freedom of instrument movement within the surgical field. Electromagnetic neuronavigation may overcome this challenge., Objective: To compare Ommaya reservoir ventricular catheter placement using electromagnetic neuronavigation to LED-based optical navigation, with emphasis on placement accuracy, operative time and complication rate., Methods: Twenty-eight patients who underwent placement of Ommaya reservoirs at our institution between 2010 and 2014 with either electromagnetic (12 patients) or optical neuronavigation (16 patients) were retrospectively reviewed., Results: Half of the patients were male. Their mean age was 56 years (range 28-87 years). Accuracy and precision in catheter tip placement at the target site (foramen of Monro) were both higher (p=0.038 and p=0.043, respectively) with electromagnetic neuronavigation. Unintended placement of the distal catheter contralateral to the target site occurred more frequently with optical navigation, as did superior or inferior positioning by more than 5 mm. Mean operative times were shorter (p=0.027) with electromagnetic neuronavigation (43.2 min) than with optical navigation (51.0 min). There were three complications (10.7%)--one case each of cytotoxic edema, post-operative wound infection, and urinary tract infection. The rate of complication did not differ between groups., Conclusion: In contrast with optical neuronavigation, frameless and pinless electromagnetic image guidance allows the ability to track instrument depth in real-time. It may increase ventricular catheter placement accuracy and precision, and decrease operative times., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
45. Postoperative stereotactic radiosurgery to the resection cavity for large brain metastases: clinical outcomes, predictors of intracranial failure, and implications for optimal patient selection.
- Author
-
Ling DC, Vargo JA, Wegner RE, Flickinger JC, Burton SA, Engh J, Amankulor N, Quinn AE, Ozhasoglu C, and Heron DE
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms mortality, Female, Humans, Male, Middle Aged, Multivariate Analysis, Postoperative Period, Radiosurgery mortality, Retrospective Studies, Brain Neoplasms secondary, Brain Neoplasms surgery, Radiosurgery methods
- Abstract
Background: Postoperative stereotactic radiosurgery for brain metastases potentially offers similar local control rates and fewer long-term neurocognitive sequelae compared to whole brain radiation therapy, although patients remain at risk for distant brain failure (DBF)., Objective: To describe clinical outcomes of adjuvant stereotactic radiosurgery for large brain metastases and identify predictors of intracranial failure and their implications on optimal patient selection criteria., Methods: We performed a retrospective review on 100 large (>3 cm) brain metastases in 99 patients managed by resection followed by postoperative stereotactic radiosurgery to a median dose of 22 Gy (range, 10-28) in 1 to 5 fractions (median, 3). Primary histology was nonsmall cell lung in 40%, breast cancer in 18%, and melanoma in 17%. Forty (40%) patients had uncontrolled systemic disease., Results: With a median follow-up of 12.2 months (range, 0.6-87.4), the 1-year Kaplan-Meier local control was 72%, DBF 64%, and overall survival 55%. Nine patients (9%) developed evidence of radiation injury, and 6 (6%) developed leptomeningeal disease. Uncontrolled systemic disease (P=.03), melanoma histology (P=.04), and increasing number of brain metastases (P<.001) were significant predictors of DBF on Cox multivariate analysis. Patients with <4 metastases, controlled systemic disease, and nonmelanoma primary (n=47) had a 1-year DBF of 48.6% vs 80.1% for all others (P=.01)., Conclusion: Postoperative stereotactic radiosurgery to the resection cavity safely and effectively augments local control of large brain metastases. Patients with <4 metastases and controlled systemic disease have significantly lower rates of DBF and are ideal treatment candidates.
- Published
- 2015
- Full Text
- View/download PDF
46. Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptide-based vaccines in combination with poly-ICLC.
- Author
-
Okada H, Butterfield LH, Hamilton RL, Hoji A, Sakaki M, Ahn BJ, Kohanbash G, Drappatz J, Engh J, Amankulor N, Lively MO, Chan MD, Salazar AM, Shaw EG, Potter DM, and Lieberman FS
- Subjects
- Adult, Antigens, Neoplasm administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines administration & dosage, Carboxymethylcellulose Sodium administration & dosage, Carboxymethylcellulose Sodium analogs & derivatives, Disease-Free Survival, Female, Glioma immunology, Glioma mortality, Glioma pathology, Humans, Male, Middle Aged, Neoplasm Grading, Pilot Projects, Poly I-C administration & dosage, Polylysine administration & dosage, Polylysine analogs & derivatives, Treatment Outcome, Vaccines, Subunit administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Glioma drug therapy
- Abstract
Purpose: WHO grade 2 low-grade gliomas (LGG) with high risk factors for recurrence are mostly lethal despite current treatments. We conducted a phase I study to evaluate the safety and immunogenicity of subcutaneous vaccinations with synthetic peptides for glioma-associated antigen (GAA) epitopes in HLA-A2(+) adults with high-risk LGGs in the following three cohorts: (i) patients without prior progression, chemotherapy, or radiotherapy (RT); (ii) patients without prior progression or chemotherapy but with prior RT; and (iii) recurrent patients., Experimental Design: GAAs were IL13Rα2, EphA2, WT1, and Survivin. Synthetic peptides were emulsified in Montanide-ISA-51 and given every 3 weeks for eight courses with intramuscular injections of poly-ICLC, followed by q12 week booster vaccines., Results: Cohorts 1, 2, and 3 enrolled 12, 1, and 10 patients, respectively. No regimen-limiting toxicity was encountered except for one case with grade 3 fever, fatigue, and mood disturbance (cohort 1). ELISPOT assays demonstrated robust IFNγ responses against at least three of the four GAA epitopes in 10 and 4 cases of cohorts 1 and 3, respectively. Cohort 1 patients demonstrated significantly higher IFNγ responses than cohort 3 patients. Median progression-free survival (PFS) periods since the first vaccine are 17 months in cohort 1 (range, 10-47+) and 12 months in cohort 3 (range, 3-41+). The only patient with large astrocytoma in cohort 2 has been progression-free for more than 67 months since diagnosis., Conclusion: The current regimen is well tolerated and induces robust GAA-specific responses in WHO grade 2 glioma patients. These results warrant further evaluations of this approach. Clin Cancer Res; 21(2); 286-94. ©2014 AACR., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
47. Use of miRNA response sequences to block off-target replication and increase the safety of an unattenuated, glioblastoma-targeted oncolytic HSV.
- Author
-
Mazzacurati L, Marzulli M, Reinhart B, Miyagawa Y, Uchida H, Goins WF, Li A, Kaur B, Caligiuri M, Cripe T, Chiocca EA, Amankulor N, Cohen JB, Glorioso JC, and Grandi P
- Subjects
- Animals, Base Sequence, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Chromosomes, Artificial, Bacterial chemistry, Chromosomes, Artificial, Bacterial metabolism, ErbB Receptors genetics, ErbB Receptors metabolism, Gene Expression Regulation, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, HEK293 Cells, Herpesvirus 1, Human metabolism, Humans, Immediate-Early Proteins antagonists & inhibitors, Immediate-Early Proteins metabolism, Injections, Intraventricular, Mice, Mice, Nude, MicroRNAs metabolism, Molecular Sequence Data, Neuroglia metabolism, Neuroglia pathology, Neurons metabolism, Neurons pathology, Virus Replication, Xenograft Model Antitumor Assays, 3' Untranslated Regions, Brain Neoplasms therapy, Glioblastoma therapy, Herpesvirus 1, Human genetics, Immediate-Early Proteins genetics, MicroRNAs genetics, Oncolytic Virotherapy methods
- Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer for which there is no effective treatment. Oncolytic HSV vectors (oHSVs) are attenuated lytic viruses that have shown promise in the treatment of human GBM models in animals, but their efficacy in early phase patient trials has been limited. Instead of attenuating the virus with mutations in virulence genes, we engineered four copies of the recognition sequence for miR-124 into the 3'UTR of the essential ICP4 gene to protect healthy tissue against lytic virus replication; miR-124 is expressed in neurons but not in glioblastoma cells. Following intracranial inoculation into nude mice, the miR-124-sensitive vector failed to replicate or show overt signs of pathogenesis. To address the concern that this safety feature may reduce oncolytic activity, we inserted the miR-124 response elements into an unattenuated, human receptor (EGFR/EGFRvIII)-specific HSV vector. We found that miR-124 sensitivity did not cause a loss of treatment efficiency in an orthotopic model of primary human GBM in nude mice. These results demonstrate that engineered miR-124 responsiveness can eliminate off-target replication by unattenuated oHSV without compromising oncolytic activity, thereby providing increased safety.
- Published
- 2015
- Full Text
- View/download PDF
48. The SHH/Gli pathway is reactivated in reactive glia and drives proliferation in response to neurodegeneration-induced lesions.
- Author
-
Pitter KL, Tamagno I, Feng X, Ghosal K, Amankulor N, Holland EC, and Hambardzumyan D
- Subjects
- Animals, Astrocytes drug effects, Astrocytes pathology, Cell Proliferation drug effects, Central Nervous System Agents, Disease Models, Animal, Hippocampus pathology, Hippocampus physiopathology, Kainic Acid, Kruppel-Like Transcription Factors genetics, Mice, Mice, Transgenic, Microglia drug effects, Microglia pathology, Neurodegenerative Diseases pathology, Neurogenesis drug effects, Neurogenesis physiology, Neurons pathology, Neurons physiology, Seizures pathology, Seizures physiopathology, Severity of Illness Index, Signal Transduction drug effects, Veratrum Alkaloids pharmacology, Zinc Finger Protein GLI1, Astrocytes physiology, Cell Proliferation physiology, Hedgehog Proteins metabolism, Kruppel-Like Transcription Factors metabolism, Microglia physiology, Neurodegenerative Diseases physiopathology
- Abstract
In response to neurodegeneration, the adult mammalian brain activates a cellular cascade that results in reactive astrogliosis and microgliosis. The mechanism through which astrocytes become reactive and the physiological consequences of their activation in response to neurodegeneration is complex. While the activation and proliferation of astrocytes has been shown to occur during massive neuronal cell death, the functional relationship between these two events has not been clearly elucidated. Here we show that in response to kainic acid- (KA) induced neurodegeneration, the mitogen sonic hedgehog (SHH) is upregulated in reactive astrocytes. SHH activity peaks at 7 days and is accompanied by increased Gli activity and elevated proliferation in several cell types. To determine the functional role of SHH-Gli signaling following KA lesions, we used a pharmacological approach to show that SHH secreted by astrocytes drives the activation and proliferation of astrocytes and microglia. The consequences of SHH-Gli signaling in KA-induced lesions appear to be independent of the severity of neurodegeneration., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
49. GM-CSF promotes the immunosuppressive activity of glioma-infiltrating myeloid cells through interleukin-4 receptor-α.
- Author
-
Kohanbash G, McKaveney K, Sakaki M, Ueda R, Mintz AH, Amankulor N, Fujita M, Ohlfest JR, and Okada H
- Subjects
- Animals, Apoptosis, Arginase genetics, Arginase metabolism, Blotting, Western, Bone Marrow immunology, Bone Marrow metabolism, Bone Marrow pathology, Cell Proliferation, Cells, Cultured, Glioma metabolism, Glioma pathology, Humans, Immune Tolerance, Interleukin-13 genetics, Interleukin-13 metabolism, Lymphocyte Activation, Mice, Mice, Inbred BALB C, Mice, Knockout, Myeloid Cells metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes metabolism, T-Lymphocytes pathology, Tumor Microenvironment, Glioma immunology, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Immunosuppression Therapy, Myeloid Cells immunology, Myeloid Cells pathology, Receptors, Cell Surface physiology, T-Lymphocytes immunology
- Abstract
Malignant gliomas are lethal cancers in the brain and heavily infiltrated by myeloid cells. Interleukin-4 receptor-α (IL-4Rα) mediates the immunosuppressive functions of myeloid cells, and polymorphisms in the IL-4Rα gene are associated with altered glioma risk and prognosis. In this study, we sought to evaluate a hypothesized causal role for IL-4Rα and myeloid suppressor cells in glioma development. In both mouse de novo gliomas and human glioblastoma cases, IL-4Rα was upregulated on glioma-infiltrating myeloid cells but not in the periphery or in normal brain. Mice genetically deficient for IL-4Rα exhibited a slower growth of glioma associated with reduced production in the glioma microenvironment of arginase, a marker of myeloid suppressor cells, which is critical for their T-cell inhibitory function. Supporting this result, investigations using bone marrow-derived myeloid cells showed that IL-4Rα mediates IL-13-induced production of arginase. Furthermore, glioma-derived myeloid cells suppressed T-cell proliferation in an IL-4Rα-dependent manner, consistent with their identification as myeloid-derived suppressor cells (MDSC). Granulocyte macrophage colony-stimulating factor (GM-CSF) plays a central role for the induction of IL-4Rα expression on myeloid cells, and we found that GM-CSF is upregulated in both human and mouse glioma microenvironments compared with normal brain or peripheral blood samples. Together, our findings establish a GM-CSF-induced mechanism of immunosuppression in the glioma microenvironment via upregulation of IL-4Rα on MDSCs.
- Published
- 2013
- Full Text
- View/download PDF
50. A novel cranioplasty technique incorporating cancellous femoral bone and recombinant bone morphogenic protein 2.
- Author
-
Wang F, Hoang D, Medvecky M, Amankulor N, Teng E, and Narayan D
- Subjects
- Adult, Humans, Male, Middle Aged, Prostheses and Implants, Transplantation, Autologous, Treatment Outcome, Bone Diseases, Infectious surgery, Bone Morphogenetic Protein 2 pharmacology, Bone Transplantation methods, Craniotomy methods, Femur transplantation, Plastic Surgery Procedures methods
- Abstract
Large calvarial defects in the setting of chronic infection and limited autologous donor sites present challenging problems for the reconstructive surgeon. We report on a novel osteogenic implant as a potential calvarial reconstruction solution. Two patients with an extensive history of severe traumatic brain injury and multiple cranial operations desired delayed calvarial reconstruction subsequent to removal of infected acrylic and titanium implants. These patients underwent harvesting of cancellous femoral bone and bone marrow, which was placed between 2 layers of recombinant bone morphogenic protein 2 impregnated in acellular collagen sponges and an outer layer of sturdily pliable Vicryl mesh to create a moldable osteogenic implant construct. The construct was molded into the shape of the defect and secured with sutures to the skull surrounding the defect. The osteogenic implant showed evidence of bone formation and maintained coverage over the original calvarial defect at 26 months postoperatively as well as subjective satisfaction by both patients and the senior surgeon. This technically straightforward technique with acceptable functional and aesthetic outcomes may be useful to reconstructing selected calvarial defects.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.