English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT

Released

Journal Article

KPC-luciferase-expressing cells elicit an anti-tumor immune response in a mouse model of pancreatic cancer

MPS-Authors
/persons/resource/persons292599

Ferrari,  Daniele P.
Research Group of Translational Molecular Imaging, Max Planck Institute for Multidisciplinary Sciences, Max Planck Society;

/persons/resource/persons182362

Ramos Gomes,  Fernanda
Research Group of Translational Molecular Imaging, Max Planck Institute for Multidisciplinary Sciences, Max Planck Society;

/persons/resource/persons182068

Alves,  Frauke
Research Group of Translational Molecular Imaging, Max Planck Institute for Multidisciplinary Sciences, Max Planck Society;

/persons/resource/persons201809

Markus,  M. Andrea
Research Group of Translational Molecular Imaging, Max Planck Institute for Multidisciplinary Sciences, Max Planck Society;

External Resource
No external resources are shared
Fulltext (restricted access)
There are currently no full texts shared for your IP range.
Fulltext (public)

Publisher Version
(Publisher version), 6MB

Supplementary Material (public)
There is no public supplementary material available
Citation

Ferrari, D. P., Ramos Gomes, F., Alves, F., & Markus, M. A. (2024). KPC-luciferase-expressing cells elicit an anti-tumor immune response in a mouse model of pancreatic cancer. Scientific Reports, 14: 13602. doi:10.1038/s41598-024-64053-0.


Cite as: https://hdl.handle.net/21.11116/0000-000F-797E-D
Abstract
Mouse models for the study of pancreatic ductal adenocarcinoma (PDAC) are well-established and representative of many key features observed in human PDAC. To monitor tumor growth, cancer cells that are implanted in mice are often transfected with reporter genes, such as firefly luciferase (Luc), enabling in vivo optical imaging over time. Since Luc can induce an immune response, we aimed to evaluate whether the expression of Luc could affect the growth of KPC tumors in mice by inducing immunogenicity. Although both cell lines, KPC and Luc transduced KPC (KPC-Luc), had the same proliferation rate, KPC-Luc tumors had significantly smaller sizes or were absent 13 days after orthotopic cell implantation, compared to KPC tumors. This coincided with the loss of bioluminescence signal over the tumor region. Immunophenotyping of blood and spleen from KPC-Luc tumor-bearing mice showed a decreased number of macrophages and CD4+ T cells, and an increased accumulation of natural killer (NK) cells in comparison to KPC tumor mice. Higher infiltration of CD8+ T cells was found in KPC-Luc tumors than in their controls. Moreover, the immune response against Luc peptide was stronger in splenocytes from mice implanted with KPC-Luc cells compared to those isolated from KPC wild-type mice, indicating increased immunogenicity elicited by the presence of Luc in the PDAC tumor cells. These results must be considered when evaluating the efficacy of anti-cancer therapies including immunotherapies in immunocompetent PDAC or other cancer mouse models that use Luc as a reporter for bioluminescence imaging.