314 555

Cited 39 times in

Enhancement of Tumor-Specific T Cell-Mediated Immunity in Dendritic Cell-Based Vaccines by Mycobacterium tuberculosis Heat Shock Protein X

Authors
 In Duk Jung  ;  Sung Jae Shin  ;  Min-Goo Lee  ;  Tae Heung Kang  ;  Hee Dong Han  ;  Seung Jun Lee  ;  Woo Sik Kim  ;  Hong Min Kim  ;  Won Sun Park  ;  Han Wool Kim  ;  Cheol-Heui Yun  ;  Eun Kyung Lee  ;  T.-C. Wu  ;  Yeong-Min Park 
Citation
 JOURNAL OF IMMUNOLOGY, Vol.193(4) : 1233-1245, 2014 
Journal Title
JOURNAL OF IMMUNOLOGY
ISSN
 0022-1767 
Issue Date
2014
MeSH
Animals ; Antigens, Bacterial/administration & dosage* ; Antigens, Bacterial/immunology* ; Bacterial Proteins/administration & dosage* ; Bacterial Proteins/immunology* ; Cell Line, Transformed ; Cell Line, Tumor ; Dendritic Cells/immunology* ; Dendritic Cells/transplantation ; Humans ; Immunity, Cellular ; Immunotherapy, Adoptive/methods* ; Lung Neoplasms/immunology ; Lung Neoplasms/secondary ; Lung Neoplasms/therapy ; Lymphocyte Activation/immunology ; Male ; Melanoma, Experimental/immunology* ; Melanoma, Experimental/secondary ; Melanoma, Experimental/therapy* ; Mice ; Mice, 129 Strain ; Mice, Inbred C57BL ; Mice, Knockout ; Mice, Transgenic ; Primary Cell Culture ; T-Lymphocytes, Cytotoxic/immunology* ; T-Lymphocytes, Cytotoxic/transplantation ; Up-Regulation/immunology*
Abstract
Despite the potential for stimulation of robust antitumor immunity by dendritic cells (DCs), clinical applications of DC-based immunotherapy are limited by the low potency in generating tumor Ag-specific T cell responses. Therefore, optimal conditions for generating potent immunostimulatory DCs that overcome tolerance and suppression are key factors in DC-based tumor immunotherapy. In this study, we demonstrate that use of the Mycobacterium tuberculosis heat shock protein X (HspX) as an immunoadjuvant in DC-based tumor immunotherapy has significant potential in therapeutics. In particular, the treatment aids the induction of tumor-reactive T cell responses, especially tumor-specific CTLs. The HspX protein induces DC maturation and proinflammatory cytokine production (TNF-α, IL-1β, IL-6, and IFN-β) through TLR4 binding partially mediated by both the MyD88 and the TRIF signaling pathways. We employed two models of tumor progression and metastasis to evaluate HspX-stimulated DCs in vivo. The administration of HspX-stimulated DCs increased the activation of naive T cells, effectively polarizing the CD4(+) and CD8(+) T cells to secrete IFN-γ, as well as enhanced the cytotoxicity of splenocytes against HPV-16 E7 (E7)-expressing TC-1 murine tumor cells in therapeutic experimental animals. Moreover, the metastatic capacity of B16-BL6 melanoma cancer cells toward the lungs was remarkably attenuated in mice that received HspX-stimulated DCs. In conclusion, the high therapeutic response rates with tumor-targeted Th1-type T cell immunity as a result of HspX-stimulated DCs in two models suggest that HspX harnesses the exquisite immunological power and specificity of DCs for the treatment of tumors.
Files in This Item:
T201402596.pdf Download
DOI
10.4049/jimmunol.1400656
Appears in Collections:
1. College of Medicine (의과대학) > Dept. of Internal Medicine (내과학교실) > 1. Journal Papers
Yonsei Authors
Shin, Sung Jae(신성재) ORCID logo https://orcid.org/0000-0003-0854-4582
URI
https://ir.ymlib.yonsei.ac.kr/handle/22282913/99434
사서에게 알리기
  feedback

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

Browse

Links