Cargando…

Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma

BACKGROUND: Long-term prognosis of WHO grade II, isocitrate dehydrogenase (IDH)-mutated low-grade glioma (LGG) is poor due to high risks of recurrence and malignant transformation into high-grade glioma. Immunotherapy strategies are attractive given the relatively intact immune system of patients wi...

Descripción completa

Detalles Bibliográficos
Autores principales: Chuntova, Pavlina, Yamamichi, Akane, Chen, Tiffany, Narayanaswamy, Rohini, Ronseaux, Sebastien, Hudson, Christine, Tron, Adriana E, Hyer, Marc L, Montoya, Megan, Mende, Abigail L, Nejo, Takahide, Downey, Kira M, Diebold, David, Lu, Min, Nicolay, Brandon, Okada, Hideho
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9174833/
https://www.ncbi.nlm.nih.gov/pubmed/35606087
http://dx.doi.org/10.1136/jitc-2022-004644
_version_ 1784722324586496000
author Chuntova, Pavlina
Yamamichi, Akane
Chen, Tiffany
Narayanaswamy, Rohini
Ronseaux, Sebastien
Hudson, Christine
Tron, Adriana E
Hyer, Marc L
Montoya, Megan
Mende, Abigail L
Nejo, Takahide
Downey, Kira M
Diebold, David
Lu, Min
Nicolay, Brandon
Okada, Hideho
author_facet Chuntova, Pavlina
Yamamichi, Akane
Chen, Tiffany
Narayanaswamy, Rohini
Ronseaux, Sebastien
Hudson, Christine
Tron, Adriana E
Hyer, Marc L
Montoya, Megan
Mende, Abigail L
Nejo, Takahide
Downey, Kira M
Diebold, David
Lu, Min
Nicolay, Brandon
Okada, Hideho
author_sort Chuntova, Pavlina
collection PubMed
description BACKGROUND: Long-term prognosis of WHO grade II, isocitrate dehydrogenase (IDH)-mutated low-grade glioma (LGG) is poor due to high risks of recurrence and malignant transformation into high-grade glioma. Immunotherapy strategies are attractive given the relatively intact immune system of patients with LGG and the slow tumor growth rate. However, accumulation of the oncometabolite D-2-hydroxyglutarate (D-2HG) in IDH-mutated gliomas leads to suppression of inflammatory pathways in the tumor microenvironment, thereby contributing to the ‘cold’ tumor phenotype. Inhibiting D-2HG production presents an opportunity to generate a robust antitumor response following tumor antigen vaccination and immune checkpoint blockade. METHODS: An IDH1(R132H) glioma model was created in syngeneic HLA-A2/HLA-DR1-transgenic mice, allowing us to evaluate the vaccination with the human leukocyte antigens (HLA)-DR1-restricted, IDH1(R132H) mutation-derived neoepitope. The effects of an orally available inhibitor of mutant IDH1 and IDH2, AG-881, were evaluated as monotherapy and in combination with the IDH1(R132H) peptide vaccination or anti-PD-1 immune checkpoint blockade. RESULTS: The HLA-A2/HLA-DR1-syngeneic IDH1(R132H) cell line expressed the IDH1 mutant protein and formed D-2HG producing orthotopic gliomas in vivo. Treatment of tumor-bearing mice with AG-881 resulted in a reduction of D-2HG levels in IDH1(R132H) glioma cells (10 fold) and tumor-associated myeloid cells, which demonstrated high levels of intracellular D-2HG in the IDH1(R132H) gliomas. AG-881 monotherapy suppressed the progression of IDH1(R132H) gliomas in a CD4(+) and CD8(+) cell-dependent manner, enhanced proinflammatory IFN [Formula: see text]-related gene expression, and increased the number of CD4(+) tumor-infiltrating T-cells. Prophylactic vaccination with the HLA-DR1-restricted IDH1(R132H) peptide or tumor-associated HLA-A2-restricted peptides did not enhance survival of tumor-bearing animals; however, vaccination with both HLA-A2-IDH1(R132H) and DR1-IDH1(R132H) peptides in combination with the IDH inhibitor significantly prolonged survival. Finally, tumor-bearing mice treated with both AG-881 and a PD-1 blocking antibody demonstrated improved survival when compared with either treatment alone. CONCLUSION: The development of effective IDH1(R132H)-targeting vaccine may be enhanced by integration with HLA class I-restricted cytotoxic T cell epitopes and AG-881. Our HLA-A2/HLA-DR1-syngeneic IDH1(R132H) glioma model should allow us to evaluate key translational questions related to the development of novel strategies for patients with IDH-mutant glioma.
format Online
Article
Text
id pubmed-9174833
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-91748332022-06-16 Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma Chuntova, Pavlina Yamamichi, Akane Chen, Tiffany Narayanaswamy, Rohini Ronseaux, Sebastien Hudson, Christine Tron, Adriana E Hyer, Marc L Montoya, Megan Mende, Abigail L Nejo, Takahide Downey, Kira M Diebold, David Lu, Min Nicolay, Brandon Okada, Hideho J Immunother Cancer Clinical/Translational Cancer Immunotherapy BACKGROUND: Long-term prognosis of WHO grade II, isocitrate dehydrogenase (IDH)-mutated low-grade glioma (LGG) is poor due to high risks of recurrence and malignant transformation into high-grade glioma. Immunotherapy strategies are attractive given the relatively intact immune system of patients with LGG and the slow tumor growth rate. However, accumulation of the oncometabolite D-2-hydroxyglutarate (D-2HG) in IDH-mutated gliomas leads to suppression of inflammatory pathways in the tumor microenvironment, thereby contributing to the ‘cold’ tumor phenotype. Inhibiting D-2HG production presents an opportunity to generate a robust antitumor response following tumor antigen vaccination and immune checkpoint blockade. METHODS: An IDH1(R132H) glioma model was created in syngeneic HLA-A2/HLA-DR1-transgenic mice, allowing us to evaluate the vaccination with the human leukocyte antigens (HLA)-DR1-restricted, IDH1(R132H) mutation-derived neoepitope. The effects of an orally available inhibitor of mutant IDH1 and IDH2, AG-881, were evaluated as monotherapy and in combination with the IDH1(R132H) peptide vaccination or anti-PD-1 immune checkpoint blockade. RESULTS: The HLA-A2/HLA-DR1-syngeneic IDH1(R132H) cell line expressed the IDH1 mutant protein and formed D-2HG producing orthotopic gliomas in vivo. Treatment of tumor-bearing mice with AG-881 resulted in a reduction of D-2HG levels in IDH1(R132H) glioma cells (10 fold) and tumor-associated myeloid cells, which demonstrated high levels of intracellular D-2HG in the IDH1(R132H) gliomas. AG-881 monotherapy suppressed the progression of IDH1(R132H) gliomas in a CD4(+) and CD8(+) cell-dependent manner, enhanced proinflammatory IFN [Formula: see text]-related gene expression, and increased the number of CD4(+) tumor-infiltrating T-cells. Prophylactic vaccination with the HLA-DR1-restricted IDH1(R132H) peptide or tumor-associated HLA-A2-restricted peptides did not enhance survival of tumor-bearing animals; however, vaccination with both HLA-A2-IDH1(R132H) and DR1-IDH1(R132H) peptides in combination with the IDH inhibitor significantly prolonged survival. Finally, tumor-bearing mice treated with both AG-881 and a PD-1 blocking antibody demonstrated improved survival when compared with either treatment alone. CONCLUSION: The development of effective IDH1(R132H)-targeting vaccine may be enhanced by integration with HLA class I-restricted cytotoxic T cell epitopes and AG-881. Our HLA-A2/HLA-DR1-syngeneic IDH1(R132H) glioma model should allow us to evaluate key translational questions related to the development of novel strategies for patients with IDH-mutant glioma. BMJ Publishing Group 2022-05-23 /pmc/articles/PMC9174833/ /pubmed/35606087 http://dx.doi.org/10.1136/jitc-2022-004644 Text en © Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Clinical/Translational Cancer Immunotherapy
Chuntova, Pavlina
Yamamichi, Akane
Chen, Tiffany
Narayanaswamy, Rohini
Ronseaux, Sebastien
Hudson, Christine
Tron, Adriana E
Hyer, Marc L
Montoya, Megan
Mende, Abigail L
Nejo, Takahide
Downey, Kira M
Diebold, David
Lu, Min
Nicolay, Brandon
Okada, Hideho
Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title_full Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title_fullStr Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title_full_unstemmed Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title_short Inhibition of D-2HG leads to upregulation of a proinflammatory gene signature in a novel HLA-A2/HLA-DR1 transgenic mouse model of IDH1R132H-expressing glioma
title_sort inhibition of d-2hg leads to upregulation of a proinflammatory gene signature in a novel hla-a2/hla-dr1 transgenic mouse model of idh1r132h-expressing glioma
topic Clinical/Translational Cancer Immunotherapy
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9174833/
https://www.ncbi.nlm.nih.gov/pubmed/35606087
http://dx.doi.org/10.1136/jitc-2022-004644
work_keys_str_mv AT chuntovapavlina inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT yamamichiakane inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT chentiffany inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT narayanaswamyrohini inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT ronseauxsebastien inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT hudsonchristine inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT tronadrianae inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT hyermarcl inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT montoyamegan inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT mendeabigaill inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT nejotakahide inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT downeykiram inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT diebolddavid inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT lumin inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT nicolaybrandon inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma
AT okadahideho inhibitionofd2hgleadstoupregulationofaproinflammatorygenesignatureinanovelhlaa2hladr1transgenicmousemodelofidh1r132hexpressingglioma