Clinical trial links oncolytic immunoactivation to survival in glioblastoma

Ling, Alexander L. and Solomon, Isaac H. and Landivar, Ana Montalvo and Nakashima, Hiroshi and Woods, Jared K. and Santos, Andres and Masud, Nafisa and Fell, Geoffrey and Mo, Xiaokui and Yilmaz, Ayse S. and Grant, James and Zhang, Abigail and Bernstock, Joshua D. and Torio, Erickson and Ito, Hirotaka and Liu, Junfeng and Shono, Naoyuki and Nowicki, Michal O. and Triggs, Daniel and Halloran, Patrick and Piranlioglu, Raziye and Soni, Himanshu and Stopa, Brittany and Bi, Wenya Linda and Peruzzi, Pierpaolo and Chen, Ethan and Malinowski, Seth W. and Prabhu, Michael C. and Zeng, Yu and Carlisle, Anne and Rodig, Scott J. and Wen, Patrick Y. and Lee, Eudocia Quant and Nayak, Lakshmi and Chukwueke, Ugonma and Gonzalez Castro, L. Nicolas and Dumont, Sydney D. and Batchelor, Tracy and Kittelberger, Kara and Tikhonova, Ekaterina and Miheecheva, Natalia and Tabakov, Dmitry and Shin, Nara and Gorbacheva, Alisa and Shumskiy, Artemy and Frenkel, Felix and Aguilar-Cordova, Estuardo and Aguilar, Laura K. and Krisky, David and Wechuck, James and Manzanera, Andrea and Matheny, Chris and Tak, Paul P. and Barone, Francesca and Kovarsky, Daniel and Tirosh, Itay and Suvà, Mario L. and Wucherpfennig, Kai W. and Ligon, Keith and Reardon, David A. and Chiocca, E. Antonio (2023) Clinical trial links oncolytic immunoactivation to survival in glioblastoma. Nature, 623 (7985). pp. 157-166. ISSN 0028-0836

[thumbnail of s41586-023-06623-2.pdf] Text
s41586-023-06623-2.pdf - Published Version

Download (42MB)

Abstract

Immunotherapy failures can result from the highly suppressive tumour microenvironment that characterizes aggressive forms of cancer such as recurrent glioblastoma (rGBM)1,2. Here we report the results of a first-in-human phase I trial in 41 patients with rGBM who were injected with CAN-3110—an oncolytic herpes virus (oHSV)3. In contrast to other clinical oHSVs, CAN-3110 retains the viral neurovirulence ICP34.5 gene transcribed by a nestin promoter; nestin is overexpressed in GBM and other invasive tumours, but not in the adult brain or healthy differentiated tissue4. These modifications confer CAN-3110 with preferential tumour replication. No dose-limiting toxicities were encountered. Positive HSV1 serology was significantly associated with both improved survival and clearance of CAN-3110 from injected tumours. Survival after treatment, particularly in individuals seropositive for HSV1, was significantly associated with (1) changes in tumour/PBMC T cell counts and clonal diversity, (2) peripheral expansion/contraction of specific T cell clonotypes; and (3) tumour transcriptomic signatures of immune activation. These results provide human validation that intralesional oHSV treatment enhances anticancer immune responses even in immunosuppressive tumour microenvironments, particularly in individuals with cognate serology to the injected virus. This provides a biological rationale for use of this oncolytic modality in cancers that are otherwise unresponsive to immunotherapy (ClinicalTrials.gov: NCT03152318).

Item Type: Article
Subjects: Apsci Archives > Multidisciplinary
Depositing User: Unnamed user with email support@apsciarchives.com
Date Deposited: 10 Nov 2023 05:59
Last Modified: 10 Nov 2023 05:59
URI: http://eprints.go2submission.com/id/eprint/2184

Actions (login required)

View Item
View Item