Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.

BACKGROUND: Lung cancer is the most common malignancy in humans and its high fatality means that no effective treatment is available. Developing new therapeutic strategies for lung cancer is urgently needed. Malaria has been reported to stimulate host immune responses, which are believed to be effic...

Full description

Bibliographic Details
Main Authors: Lili Chen, Zhengxiang He, Li Qin, Qinyan Li, Xibao Shi, Siting Zhao, Ling Chen, Nanshan Zhong, Xiaoping Chen
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2011-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3170332?pdf=render
id doaj-7f42ecce629a446580ccc1d69febfbee
record_format Article
spelling doaj-7f42ecce629a446580ccc1d69febfbee2020-11-25T02:27:08ZengPublic Library of Science (PLoS)PLoS ONE1932-62032011-01-0169e2440710.1371/journal.pone.0024407Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.Lili ChenZhengxiang HeLi QinQinyan LiXibao ShiSiting ZhaoLing ChenNanshan ZhongXiaoping ChenBACKGROUND: Lung cancer is the most common malignancy in humans and its high fatality means that no effective treatment is available. Developing new therapeutic strategies for lung cancer is urgently needed. Malaria has been reported to stimulate host immune responses, which are believed to be efficacious for combating some clinical cancers. This study is aimed to provide evidence that malaria parasite infection is therapeutic for lung cancer. METHODOLOGY/PRINCIPAL FINDINGS: Antitumor effect of malaria infection was examined in both subcutaneously and intravenously implanted murine Lewis lung cancer (LLC) model. The results showed that malaria infection inhibited LLC growth and metastasis and prolonged the survival of tumor-bearing mice. Histological analysis of tumors from mice infected with malaria revealed that angiogenesis was inhibited, which correlated with increased terminal deoxynucleotidyl transferase-mediated (TUNEL) staining and decreased Ki-67 expression in tumors. Through natural killer (NK) cell cytotoxicity activity, cytokine assays, enzyme-linked immunospot assay, lymphocyte proliferation, and flow cytometry, we demonstrated that malaria infection provided anti-tumor effects by inducing both a potent anti-tumor innate immune response, including the secretion of IFN-γ and TNF-α and the activation of NK cells as well as adaptive anti-tumor immunity with increasing tumor-specific T-cell proliferation and cytolytic activity of CD8(+) T cells. Notably, tumor-bearing mice infected with the parasite developed long-lasting and effective tumor-specific immunity. Consequently, we found that malaria parasite infection could enhance the immune response of lung cancer DNA vaccine pcDNA3.1-hMUC1 and the combination produced a synergistic antitumor effect. CONCLUSIONS/SIGNIFICANCE: Malaria infection significantly suppresses LLC growth via induction of innate and adaptive antitumor responses in a mouse model. These data suggest that the malaria parasite may provide a novel strategy or therapeutic vaccine vector for anti-lung cancer immune-based therapy.http://europepmc.org/articles/PMC3170332?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Lili Chen
Zhengxiang He
Li Qin
Qinyan Li
Xibao Shi
Siting Zhao
Ling Chen
Nanshan Zhong
Xiaoping Chen
spellingShingle Lili Chen
Zhengxiang He
Li Qin
Qinyan Li
Xibao Shi
Siting Zhao
Ling Chen
Nanshan Zhong
Xiaoping Chen
Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
PLoS ONE
author_facet Lili Chen
Zhengxiang He
Li Qin
Qinyan Li
Xibao Shi
Siting Zhao
Ling Chen
Nanshan Zhong
Xiaoping Chen
author_sort Lili Chen
title Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
title_short Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
title_full Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
title_fullStr Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
title_full_unstemmed Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity.
title_sort antitumor effect of malaria parasite infection in a murine lewis lung cancer model through induction of innate and adaptive immunity.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2011-01-01
description BACKGROUND: Lung cancer is the most common malignancy in humans and its high fatality means that no effective treatment is available. Developing new therapeutic strategies for lung cancer is urgently needed. Malaria has been reported to stimulate host immune responses, which are believed to be efficacious for combating some clinical cancers. This study is aimed to provide evidence that malaria parasite infection is therapeutic for lung cancer. METHODOLOGY/PRINCIPAL FINDINGS: Antitumor effect of malaria infection was examined in both subcutaneously and intravenously implanted murine Lewis lung cancer (LLC) model. The results showed that malaria infection inhibited LLC growth and metastasis and prolonged the survival of tumor-bearing mice. Histological analysis of tumors from mice infected with malaria revealed that angiogenesis was inhibited, which correlated with increased terminal deoxynucleotidyl transferase-mediated (TUNEL) staining and decreased Ki-67 expression in tumors. Through natural killer (NK) cell cytotoxicity activity, cytokine assays, enzyme-linked immunospot assay, lymphocyte proliferation, and flow cytometry, we demonstrated that malaria infection provided anti-tumor effects by inducing both a potent anti-tumor innate immune response, including the secretion of IFN-γ and TNF-α and the activation of NK cells as well as adaptive anti-tumor immunity with increasing tumor-specific T-cell proliferation and cytolytic activity of CD8(+) T cells. Notably, tumor-bearing mice infected with the parasite developed long-lasting and effective tumor-specific immunity. Consequently, we found that malaria parasite infection could enhance the immune response of lung cancer DNA vaccine pcDNA3.1-hMUC1 and the combination produced a synergistic antitumor effect. CONCLUSIONS/SIGNIFICANCE: Malaria infection significantly suppresses LLC growth via induction of innate and adaptive antitumor responses in a mouse model. These data suggest that the malaria parasite may provide a novel strategy or therapeutic vaccine vector for anti-lung cancer immune-based therapy.
url http://europepmc.org/articles/PMC3170332?pdf=render
work_keys_str_mv AT lilichen antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT zhengxianghe antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT liqin antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT qinyanli antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT xibaoshi antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT sitingzhao antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT lingchen antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT nanshanzhong antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
AT xiaopingchen antitumoreffectofmalariaparasiteinfectioninamurinelewislungcancermodelthroughinductionofinnateandadaptiveimmunity
_version_ 1724844118202384384