Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor

Abstract Background Anti-programmed cell death 1 (PD-1) antibody is an immune checkpoint inhibitor, and anti-PD-1 therapy improves the anti-tumor functions of T cells and affects tumor microenvironment. We previously reported that anti-PD-1 treatment affected tumor glycolysis by using 2-deoxy-2-[18F...

Full description

Bibliographic Details
Main Authors: Mayu Tomita, Motofumi Suzuki, Yusuke Kono, Kohei Nakajima, Takuma Matsuda, Yuji Kuge, Mikako Ogawa
Format: Article
Language:English
Published: SpringerOpen 2020-03-01
Series:EJNMMI Research
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13550-020-0608-4
id doaj-430db2dc891b40f0911dc39209ebe171
record_format Article
spelling doaj-430db2dc891b40f0911dc39209ebe1712020-11-25T03:32:11ZengSpringerOpenEJNMMI Research2191-219X2020-03-0110111010.1186/s13550-020-0608-4Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumorMayu Tomita0Motofumi Suzuki1Yusuke Kono2Kohei Nakajima3Takuma Matsuda4Yuji Kuge5Mikako Ogawa6Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityLaboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityLaboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityLaboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityLaboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityCentral Institute of Isotope Science, Hokkaido UniversityLaboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido UniversityAbstract Background Anti-programmed cell death 1 (PD-1) antibody is an immune checkpoint inhibitor, and anti-PD-1 therapy improves the anti-tumor functions of T cells and affects tumor microenvironment. We previously reported that anti-PD-1 treatment affected tumor glycolysis by using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET). That study showed that anti-PD-1 therapy in a mouse B16F10 melanoma model increased glucose metabolism in cancer cells at the point where anti-PD-1 therapy did not cause a significant inhibition of tumor growth. However, the B16F10 melanoma model is poorly immunogenic, so it is not clear how anti-PD-1 treatment affects glucose metabolism in highly immunogenic cancer models. In this study, we used a cyclic dinucleotide GMP-AMP (cGAMP)-injected B16F10 melanoma model to investigate the effect of anti-PD-1 therapy on [18F]FDG uptake in a highly immune activated tumor in mice. Results To compare the cGAMP-injected B16F10 model with the B16F10 model, experiments were performed as described in our previous manuscript. [18F]FDG-PET was measured before treatment and 7 days after the start of treatment. In this study, [18F]FDG uptake in tumors in the cGAMP/anti-PD-1 combination group was lower than that in the anti-PD-1 treatment group tumors on day 7, as shown by PET and ex vivo validation. Flow-cytometry was performed to assess immune cell populations and glucose metabolism. Anti-PD-1 and/or cGAMP treatment increased the infiltration level of immune cells into tumors. The cGAMP/anti-PD-1 combination group had significantly lower levels of GLUT1high cells/hexokinase IIhigh cells in CD45− cancer cells compared with tumors in the anti-PD-1 treated group. These results suggested that if immune responses in tumors are higher than a certain level, glucose uptake in cancer cells is reduced depending on that level. Such a change of glucose uptake might be caused by the difference in infiltration or activation level of immune cells between the anti-PD-1 treated group and the cGAMP/anti-PD-1 combination group. Conclusions [18F]FDG uptake in cancer cells after anti-PD-1 treatment might be affected by the tumor immune microenvironment including immune cell infiltration, composition, and activation status.http://link.springer.com/article/10.1186/s13550-020-0608-4PD-1Immune checkpoint inhibitor[18F]FDGcGAMP
collection DOAJ
language English
format Article
sources DOAJ
author Mayu Tomita
Motofumi Suzuki
Yusuke Kono
Kohei Nakajima
Takuma Matsuda
Yuji Kuge
Mikako Ogawa
spellingShingle Mayu Tomita
Motofumi Suzuki
Yusuke Kono
Kohei Nakajima
Takuma Matsuda
Yuji Kuge
Mikako Ogawa
Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
EJNMMI Research
PD-1
Immune checkpoint inhibitor
[18F]FDG
cGAMP
author_facet Mayu Tomita
Motofumi Suzuki
Yusuke Kono
Kohei Nakajima
Takuma Matsuda
Yuji Kuge
Mikako Ogawa
author_sort Mayu Tomita
title Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
title_short Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
title_full Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
title_fullStr Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
title_full_unstemmed Influence on [18F]FDG uptake by cancer cells after anti-PD-1 therapy in an enforced-immune activated mouse tumor
title_sort influence on [18f]fdg uptake by cancer cells after anti-pd-1 therapy in an enforced-immune activated mouse tumor
publisher SpringerOpen
series EJNMMI Research
issn 2191-219X
publishDate 2020-03-01
description Abstract Background Anti-programmed cell death 1 (PD-1) antibody is an immune checkpoint inhibitor, and anti-PD-1 therapy improves the anti-tumor functions of T cells and affects tumor microenvironment. We previously reported that anti-PD-1 treatment affected tumor glycolysis by using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET). That study showed that anti-PD-1 therapy in a mouse B16F10 melanoma model increased glucose metabolism in cancer cells at the point where anti-PD-1 therapy did not cause a significant inhibition of tumor growth. However, the B16F10 melanoma model is poorly immunogenic, so it is not clear how anti-PD-1 treatment affects glucose metabolism in highly immunogenic cancer models. In this study, we used a cyclic dinucleotide GMP-AMP (cGAMP)-injected B16F10 melanoma model to investigate the effect of anti-PD-1 therapy on [18F]FDG uptake in a highly immune activated tumor in mice. Results To compare the cGAMP-injected B16F10 model with the B16F10 model, experiments were performed as described in our previous manuscript. [18F]FDG-PET was measured before treatment and 7 days after the start of treatment. In this study, [18F]FDG uptake in tumors in the cGAMP/anti-PD-1 combination group was lower than that in the anti-PD-1 treatment group tumors on day 7, as shown by PET and ex vivo validation. Flow-cytometry was performed to assess immune cell populations and glucose metabolism. Anti-PD-1 and/or cGAMP treatment increased the infiltration level of immune cells into tumors. The cGAMP/anti-PD-1 combination group had significantly lower levels of GLUT1high cells/hexokinase IIhigh cells in CD45− cancer cells compared with tumors in the anti-PD-1 treated group. These results suggested that if immune responses in tumors are higher than a certain level, glucose uptake in cancer cells is reduced depending on that level. Such a change of glucose uptake might be caused by the difference in infiltration or activation level of immune cells between the anti-PD-1 treated group and the cGAMP/anti-PD-1 combination group. Conclusions [18F]FDG uptake in cancer cells after anti-PD-1 treatment might be affected by the tumor immune microenvironment including immune cell infiltration, composition, and activation status.
topic PD-1
Immune checkpoint inhibitor
[18F]FDG
cGAMP
url http://link.springer.com/article/10.1186/s13550-020-0608-4
work_keys_str_mv AT mayutomita influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT motofumisuzuki influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT yusukekono influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT koheinakajima influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT takumamatsuda influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT yujikuge influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
AT mikakoogawa influenceon18ffdguptakebycancercellsafterantipd1therapyinanenforcedimmuneactivatedmousetumor
_version_ 1724569105891065856