AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma

Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated...

Full description

Bibliographic Details
Main Authors: M.I. Strecker, K. Wlotzka, F. Strassheimer, B. Roller, G. Ludmirski, S. König, J. Röder, C. Opitz, T. Alekseeva, J. Reul, L. Sevenich, T. Tonn, W.S. Wels, J.P. Steinbach, C.J. Buchholz, M.C. Burger
Format: Article
Language:English
Published: Taylor & Francis Group 2022-12-01
Series:OncoImmunology
Subjects:
Online Access:https://www.tandfonline.com/doi/10.1080/2162402X.2022.2127508
_version_ 1797995666086035456
author M.I. Strecker
K. Wlotzka
F. Strassheimer
B. Roller
G. Ludmirski
S. König
J. Röder
C. Opitz
T. Alekseeva
J. Reul
L. Sevenich
T. Tonn
W.S. Wels
J.P. Steinbach
C.J. Buchholz
M.C. Burger
author_facet M.I. Strecker
K. Wlotzka
F. Strassheimer
B. Roller
G. Ludmirski
S. König
J. Röder
C. Opitz
T. Alekseeva
J. Reul
L. Sevenich
T. Tonn
W.S. Wels
J.P. Steinbach
C.J. Buchholz
M.C. Burger
author_sort M.I. Strecker
collection DOAJ
description Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.
first_indexed 2024-04-11T10:05:19Z
format Article
id doaj.art-94d82fff92f741b8ae2df7293f6ae1d4
institution Directory Open Access Journal
issn 2162-402X
language English
last_indexed 2024-04-11T10:05:19Z
publishDate 2022-12-01
publisher Taylor & Francis Group
record_format Article
series OncoImmunology
spelling doaj.art-94d82fff92f741b8ae2df7293f6ae1d42022-12-22T04:30:15ZengTaylor & Francis GroupOncoImmunology2162-402X2022-12-0111110.1080/2162402X.2022.2127508AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastomaM.I. Strecker0K. Wlotzka1F. Strassheimer2B. Roller3G. Ludmirski4S. König5J. Röder6C. Opitz7T. Alekseeva8J. Reul9L. Sevenich10T. Tonn11W.S. Wels12J.P. Steinbach13C.J. Buchholz14M.C. Burger15Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanyFrankfurt Cancer Institute, Goethe University, Frankfurt, GermanyInstitute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, GermanyFrankfurt Cancer Institute, Goethe University, Frankfurt, GermanyPaul-Ehrlich-Institut, Molecular Biotechnology and Gene Therapy, Langen, GermanyGerman Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, GermanyInstitute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, GermanyGerman Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanyFrankfurt Cancer Institute, Goethe University, Frankfurt, GermanySenckenberg Institute of Neurooncology, Goethe University, Frankfurt, GermanyGlioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.https://www.tandfonline.com/doi/10.1080/2162402X.2022.2127508Immunotherapytumor targetingcheckpoint inhibitionCAR-NK celladeno-associated virusHER2
spellingShingle M.I. Strecker
K. Wlotzka
F. Strassheimer
B. Roller
G. Ludmirski
S. König
J. Röder
C. Opitz
T. Alekseeva
J. Reul
L. Sevenich
T. Tonn
W.S. Wels
J.P. Steinbach
C.J. Buchholz
M.C. Burger
AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
OncoImmunology
Immunotherapy
tumor targeting
checkpoint inhibition
CAR-NK cell
adeno-associated virus
HER2
title AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
title_full AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
title_fullStr AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
title_full_unstemmed AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
title_short AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
title_sort aav mediated gene transfer of a checkpoint inhibitor in combination with her2 targeted car nk cells as experimental therapy for glioblastoma
topic Immunotherapy
tumor targeting
checkpoint inhibition
CAR-NK cell
adeno-associated virus
HER2
url https://www.tandfonline.com/doi/10.1080/2162402X.2022.2127508
work_keys_str_mv AT mistrecker aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT kwlotzka aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT fstrassheimer aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT broller aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT gludmirski aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT skonig aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT jroder aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT copitz aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT talekseeva aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT jreul aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT lsevenich aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT ttonn aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT wswels aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT jpsteinbach aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT cjbuchholz aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma
AT mcburger aavmediatedgenetransferofacheckpointinhibitorincombinationwithher2targetedcarnkcellsasexperimentaltherapyforglioblastoma