Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hyun-Jung Sohn is active.

Publication


Featured researches published by Hyun-Jung Sohn.


Cancer Science | 2008

Co‐administration of carcinoembryonic antigen and HIV TAT fusion protein with CpG‐oligodeoxynucleotide induces potent antitumor immunity

Sun-Je Woo; Chang-Hyun Kim; Mi-Young Park; Hye-Sung Kim; Hyun-Jung Sohn; Jung-Sun Park; Hyung Jin Kim; Seong-Taek Oh; Tai-Gyu Kim

Although dendritic cells (DC) have been well demonstrated as a strong cellular adjuvant for a tumor vaccine, there are several limitations for clinical application. A protein‐based vaccine using a potent adjuvant is an appealing approach for tumor antigen‐specific immunotherapy because of their simplicity, safety, efficacy and capacity for repeated administration. CpG‐oligodeoxynucleotides (ODN) have been used as adjuvants to stimulate innate and adaptive immune responses for cancer treatment. The authors evaluated the adjuvant effects of CpG‐ODN in a vaccine incorporating recombinant fusion protein of the HIV TAT PTD domain and carcinoembryonic antigen (TAT‐CEA). Mice vaccinated with TAT‐CEA and CpG‐ODN (TAT‐CEA + CpG) showed enhanced CEA‐specific immunity, including cytotoxic T‐lymphocytes (CTL) activity and interferon (IFN)‐γ secreting T cells compared with CEA and CpG‐ODN (CEA + CpG) or TAT‐CEA vaccination alone. Vaccination with TAT‐CEA + CpG elicited Th1‐based responses, as indicated by the higher ratio of immunoglobulin (Ig)G2a antibody/IgG1 antibodies specific for CEA. The survival rate was significantly increased after vaccination with TAT‐CEA + CpG in a tumor model using MC38/CEA2. Furthermore, the TAT‐CEA ± CpG vaccine groups showed similar antitumor immunity to the CEA peptide‐pulsed DC (CEA peptide/DC) vaccine groups. These data suggest that coadministration of TAT fusion protein with CpG‐ODN may serve as a potential formulation for enhancing antitumor activity. (Cancer Sci 2008; 99: 1034–1039)


PLOS ONE | 2014

Toll like Receptor 3 & 4 Responses of Human Turbinate Derived Mesenchymal Stem Cells: Stimulation by Double Stranded RNA and Lipopolysaccharide

Se Hwan Hwang; Hye Kyung Cho; Sang Hi Park; WeonSun Lee; Hee Jin Lee; Dong Chang Lee; Jeong Hoon Oh; Sun Hwa Park; Tai-Gyu Kim; Hyun-Jung Sohn; Jun Myung Kang; Sung Won Kim

Background and objectives Multipotent mesenchymal stromal cells (MSCs) represent a promising cell-based therapy for a number of inflammatory or autoimmune diseases. Herein, Toll like receptor (TLR) expression by MSCs and their immune regulatory roles are investigated. In this study, we investigated the influence of TLR on the immune response, proliferation, and differentiation potential of human turbinated MSC (hTMSC) cultures in vitro. Subjects and Methods After isolating hTMSCs from discarded inferior turbinate tissue, FACS analysis was used to assess the expression of TLRs such as TLR2, TLR3, TLR4, and TLR5 in hTMSCs and cell proliferation was assessed using a cell counting kit (CCK)-8. Cytokine and chemokine secretions were analyzed with multiplex immunoassays for IL-1α, IL-1β, IL-4, IL-6, IL-8, IL-10, IL-12, IP-10 (CXCL10), RANTES (CCL5), TNF-a, GM-CSF, and IFN-γ. The differentiation potential of hTMSCs was evaluated in the osteogenic, chondogenic, and adipogeinc media and analyzed by histology and gene expression related to differentiation. Results FACS analysis revealed that TLR3 and TLR4 expression consisted of a relatively high percentage of the surface proteins expressed by hTMSCs. The proliferation of hTMSCs was influenced and significantly increased by the presence of TLR4 agonists. In particular, hTMSCs produced a set of cytokines and chemokines and the expression of IL-6, IL-8, IL-12, IP-10 (CXCL10), RANTES (CCL5), TNF-α, and GM-CSF were up-regulated in response to the TLR4 agonist LPS. The osteogenic and adipogeinc differentiation potential of hTMSCs was not affected by TLR agonists. Conclusions We conclude that TLR4 stimulation affects TLR expression, proliferation, and the immunomodulation potential of hTMSCs. Understanding the mechanism behind TLRs influence on hTMSCs and their immunomodulating properties would be useful for providing a novel target to exploit in the improvement of stem cell-based therapeutic strategies.


Vaccine | 2010

Dendritic cell vaccine in addition to FOLFIRI regimen improve antitumor effects through the inhibition of immunosuppressive cells in murine colorectal cancer model.

Hye-Sung Kim; Hye-Mi Park; Jung-Sun Park; Hyun-Jung Sohn; Sung-Guh Kim; Hyung Jin Kim; Seong-Taek Oh; Tai-Gyu Kim

Although chemotherapy is still one of the best treatments for most cancers, immunotherapies such as dendritic cell (DC) vaccines have emerged as an alternative protocol for destroying residual tumors. In this study, we investigated antitumor effects of the combined therapy using DC vaccine and irinotecan plus infusional 5-fluorouracil and leucovorin (FOLFIRI) which have been clinically used for the treatment of colorectal cancer. A maximum tolerated dose of FOLFIRI was preliminarily determined for MC38/CEA2 colorectal cancer model. Vaccination with DC expressing carcinoembryonic antigen (CEA) enhanced antitumor effect after FOLFIRI treatment. The combined therapy also increased CEA-specific Th1 and cytotoxic T-cell responses. Interestingly, although FOLFIRI treatment rather showed a rebound in the number of myeloid-derived suppressor cells (MDSC) and regulatory T-cells (Treg) after 14 days, additional DC vaccine could inhibit the rebound of these immunosuppressive cells. Furthermore, mice cured by the combined therapy showed antigen-specific T-cell responses and resistance against challenge of MC38/CEA2 compared with mice cured with FOLFIRI. These results demonstrated that DC vaccine in addition to FOLFIRI regimen could improve antitumor effects through the inhibition of immunosuppressive tumor environments in murine colorectal cancer model, and may provide knowledge useful for the design of chemo-immunotherapeutic strategies for the treatment of colorectal carcinoma in clinical trials.


Molecular Therapy | 2015

Long-term Outcome of Extranodal NK/T Cell Lymphoma Patients Treated With Postremission Therapy Using EBV LMP1 and LMP2a-specific CTLs

Seok-Goo Cho; Nayoun Kim; Hyun-Jung Sohn; Suk Kyeong Lee; Sang Taek Oh; Hyun-Il Cho; Hyeon Woo Yim; Seung Eun Jung; Gyeongsin Park; Joo Hyun Oh; Byung-Ock Choi; Sung Won Kim; Soo Whan Kim; Nak Gyun Chung; Jong-Wook Lee; Young Seon Hong; Tai-Gyu Kim

Extranodal NK/T-cell lymphoma (ENKTCL) is associated with latent Epstein-Barr virus (EBV) infection and frequent relapse even after complete response (CR) to intensive chemotherapy and radiotherapy. The expression of EBV proteins in the tumor provides targets for adoptive immunotherapy with antigen-specific cytotoxic T cells (CTL). To evaluate the efficacy and safety of EBV latent membrane protein (LMP)-1 and LMP-2a-specific CTLs (LMP1/2a CTLs) stimulated with LMP1/2a RNA-transferred dendritic cells, we treated 10 ENKTCL patients who showed complete response to induction therapy. Patients who completed and responded to chemotherapy, radiotherapy, and/or high-dose therapy followed by stem cell transplantation (HDT/SCT) were eligible to receive eight doses of 2 × 107 LMP1/2a CTLs/m2. Following infusion, there were no immediate or delayed toxicities. The 4-year overall survival (OS) and progression-free survival (PFS) were 100%, and 90% (95% CI: 71.4 to 100%) respectively with a median follow-up of 55·5 months. Circulating IFN-γ secreting LMP1 and LMP2a-specific T cells within the peripheral blood corresponded with decline in plasma EBV DNA levels in patients. Adoptive transfer of LMP1/2a CTLs in ENKTCL patients is a safe and effective postremission therapeutic approach. Further randomized studies will be needed to define the role of EBV-CTLs in preventing relapse of ENKTCL.


Experimental and Molecular Medicine | 2009

Selective addition of CXCR3(+) CCR4(-) CD4(+) Th1 cells enhances generation of cytotoxic T cells by dendritic cells in vitro.

Sung Hee Yoon; Sun Ok Yun; Jung Yong Park; Hee Yeun Won; Eun-Kyung Kim; Hyun-Jung Sohn; Hyun-Il Cho; Tai-Gyu Kim

Increasing importance is being given to the stimulation of Th1 response in cancer immunotherapy because its presence can shift the direction of adaptive immune responses toward protective immunity. Based on chemokine receptor expression, CXCR3+CCR4-CD4+ T cells as Th1-type cells were investigated its capacity in monocyte-derived dendritic cell (DC) maturation and polarization, and induction of antigen specific cytotoxic T lymphocytes (CTL) in vitro. The levels of IL-4, IL-5 and IL-10 were decreased to the basal level compared with high production of IFN-γ, TNF-α, and IL-2 in CXCR3+CCR4-CD4+ T cells stimulated with anti-CD3 and anti-CD28 antibodies. Co-incubation of activated CD4+ or CXCR3+CCR4-CD4+ T cells with DC (CD4+/DC or CXCR3+CD4+/DC, respectively) particularly up-regulated IL-12 and CD80 expression compared with DC matured with TNF-α and LPS (mDC). Although there was no significant difference between the effects of the CXCR3+CCR4-CD4+ and CD4+ T cells on DC phenotype expression, CXCR3+CD4+/DC in CTL culture were able to expand number of CD8+ T cells and increased frequencies of IFN-γ secreting cells and overall cytolytic activity against tumor antigen WT-1. These results demonstrated that the selective addition of CXCR3+CCR4-CD4+ T cells to CTL cultures could enhance the induction of CTLs by DC in vitro, and implicated on a novel strategy for adoptive T cell therapy.


Vaccine | 2008

Efficient generation of survivin-specific cytotoxic T lymphocytes from healthy persons in vitro: Quantitative and qualitative effects of CD4+ T cells

Eun-Kyung Kim; Hyun-Il Cho; Sung Hee Yoon; Min-Ji Park; Hyun-Jung Sohn; Hyung Jin Kim; Seong-Taek Oh; Tai-Gyu Kim

For the adoptive immunotherapy and the study of cytotoxic T lymphocytes (CTLs) in human, efficient in vitro generation of CTLs is needed. However, it is still difficult to induce T cells specific for naïve antigens in vitro even though dendritic cells (DCs) as potent APCs are used. In this study, we investigated quantitative and qualitative effects of CD4+ T cells during in vitro stimulation of CD8+ T cells from healthy donors using DCs transduced with adenovirus vector expressing human survivin (Adv-survivin). CTLs were not efficiently induced in the absence of CD4+ T cells or in CD25+ depleted CD4+ T cells. When the ratio of CD4+:CD8+ T cells was quantitatively decreased from 2:1 to 1:2, proliferation of CTLs specific for survivin was gradually increased. Because DCs pulsed with HCMV pp65 protein could activate CD4+ T cells to secrete Th1 cytokines, the use of pp65 protein as an adjuvant induced higher numbers and frequencies of CTLs. Furthermore, Th1 conditioning of CD4+ T cells augmented this generation of CTLs. These results suggest that both quantitative and qualitative modulation of CD4+ T cells including the number and Th1 polarization may be required for the efficient induction of CTLs specific for tumor antigens in vitro.


European Journal of Immunology | 2008

Efficient antitumor immunity in a murine colorectal cancer model induced by CEA RNA-electroporated B cells

Mi-Young Park; Hye-Sung Kim; Sun-Je Woo; Chang-Hyun Kim; Jung-Sun Park; Hyun-Jung Sohn; Hyung Jin Kim; Seong-Taek Oh; Tai-Gyu Kim

RNA electroporation as a gene delivery method is more feasible and safer as compared with viral vectors. RNA‐loaded dendritic cells (DC) have been used to induce T cell responses against tumor rejection antigens and B cells can also act as antigen‐presenting cells for cellular vaccines. In this study, we compared B cells and DC, after electroporation with carcinoembryonic antigen (CEA) RNA, for their capacity to generate cytotoxic T lymphocytes and antitumor immunity. Vaccination using these B cells induced levels of IFN‐γ‐secreting T cells and cytotoxic T cells comparable to those induced by DC. Intravenous administration was the optimum route for the B cell vaccine, while subcutaneous administration was the optimum route for the DC vaccine. The B cell vaccine predominantly generated CEA‐specific CD4+ T cells, whereas the DC vaccine generated CD8+ T cells. Moreover, the B cell vaccine induced higher levels of anti‐CEA antibodies than the DC vaccine. A heterogeneous prime‐boost using B cells and DC failed to show any synergistic effects; however, the B cell vaccine did inhibit tumor growth and prolonged survival to a similar extent as the DC vaccine. Such RNA‐electroporated B cells may prove useful as cellular tumor vaccines with potential clinical application.


Cancer Science | 2011

Induction of antitumor immunity using dendritic cells electroporated with Polo‐like kinase 1 (Plk1) mRNA in murine tumor models

Jung-Sun Park; Hyun-Jung Sohn; Gyeong-Sin Park; Yeun-Jun Chung; Tai-Gyu Kim

Polo‐like kinase 1 (Plk1), a serine–threonine kinase, plays a key role in the regulation of the cell cycle. Elevated Plk1 expression in various cancers is correlated with poor prognosis and poor patient survival rates. Several Plk1 inhibitors are currently being developed as potential treatments for cancer. In the present study, we investigated whether dendritic cells (DC) electroporated with mouse Plk1RNA (mPlk1RNA/DC) can induce Plk1‐specific immune responses and exert antitumor effects in various murine tumor models. Overexpression of Plk1 protein was confirmed in several mouse and human tumor cell lines and various cancer tissues. Furthermore, Plk1‐specific CD4+ and CD8+ T cells were induced by vaccination with mPlk1RNA/DC and the cytotoxic activity of the T cells was demonstrated against several Plk1‐expressing tumor cell lines. Vaccination with mPlk1RNA/DC inhibited the growth of MC‐38 and B16F10 tumors in C57BL/6 mice and the growth of CT26 tumors in BALB/c mice. Depletion of CD8+ T cells reversed the inhibition of tumor growth by mPlk1RNA/DC vaccination. Homologous human Plk1RNA‐electroporated DC also inhibited tumor growth in MC‐38 tumor‐bearing mice. In addition, Plk1‐specific cytotoxic T lymphocytes from PBMC of healthy donors could be induced using autologous monocyte‐derived DC electroporated with RNA encoding the whole gene of human Plk1. Taken together, the results of the present study suggest that Plk1 could be a universal tumor antigen recognized by cytotoxic T lymphocytes for cancer immunotherapy. (Cancer Sci 2011; 102: 1448–1454)


Immunology Letters | 2010

Efficient co-transduction of adenoviral vectors encoding carcinoembryonic antigen and survivin into dendritic cells by the CAR-TAT adaptor molecule enhance anti-tumor immunity in a murine colorectal cancer model

Hye-Sung Kim; Chang-Hyun Kim; Mi-Young Park; Jung-Sun Park; Hye-Mi Park; Hyun-Jung Sohn; Hyung Jin Kim; Sung-Guh Kim; Seong-Taek Oh; Tai-Gyu Kim

Because multiple tumor antigens, including carcinoembryonic antigen (CEA) and survivin (SVV), have been frequently observed in human colorectal cancer, we investigated whether the expression of both CEA and SVV by co-transduction of adenovirus vectors into dendritic cells (DCs) could improve anti-tumor immunity in a murine colorectal cancer model. The adaptor fusion protein of Coxsackie and adenovirus receptor and TAT-protein transduction domain (CAR-TAT) enhanced co-transduction of adenovirus vectors encoding CEA (AdCEA) and SVV (AdSVV) into DCs, and increased anti-tumor immunity. DCs expressing both CEA and SVV in the presence of CAR-TAT (DC-AdCEA/AdSVV+CAR-TAT) induced T-cell responses specific for CEA and SVV, and enhanced cytotoxic T-cell activity on MC38/CEA2 cells expressing CEA and SVV compared with DCs expressing either CEA or SVV alone. Particularly, DC-AdCEA/AdSVV+CAR-TAT induced higher number of CEA-specific IFN-gamma secreting T cells compared with DC-AdCEA+CAR-TAT. Vaccination with DC-AdCEA/AdSVV+CAR-TAT also more efficiently inhibited tumor growth compared with DCs expressing either CEA or SVV alone in therapeutic tumor models. These results suggest that efficient co-transduction of multiple adenovirus vectors by CAR-TAT could be used to develop various strategies for therapeutic DC vaccines.


Human Gene Therapy | 2010

Fusion of the Human Cytomegalovirus pp65 Antigen with Both Ubiquitin and Ornithine Decarboxylase Additively Enhances Antigen Presentation to CD8+ T Cells in Human Dendritic Cells

Min-Ji Park; Eun-Kyung Kim; Ji-Young Han; Hyun-Woo Cho; Hyun-Jung Sohn; Su-Yeon Kim; Tai-Gyu Kim

Antigenic molecules are modified for targeting to the proteasome by ubiquitin (Ub) or by a Ub-independent system such as ornithine decarboxylase (ODC) to be presented by MHC class I molecules. In this study, we compared the immunogenicity of human cytomegalovirus pp65 antigen fused with Ub and/or ODC, using RNA electroporation of human dendritic cells. Among the C-terminal mutants of Ub (G76, A76, and V76), Ub(G) showed the best ability to enhance the degradation of a target protein and stimulate T cells. The pp65 antigens fused with either Ub(G) or ODC enhanced the stimulation to CD8(+) T cells, and the effects of Ub(G) and ODC were similar. Furthermore, the fusion of both Ub and ODC additively increased immunogenicity compared with the single-fusion proteins. The fusion of Ub(G) and ODC enhanced primarily the stimulation of CD8(+) rather than CD4(+) T cells and more efficiently induced pp65-specific T cells in vitro. These additive effects of Ub and ODC in antigen processing may provide improved strategies to stimulate CD8(+) T cells for the development of immunotherapies against the variety of viral diseases and cancers.

Collaboration


Dive into the Hyun-Jung Sohn's collaboration.

Top Co-Authors

Avatar

Tai-Gyu Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Hyun-Il Cho

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Mi-Young Park

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Chang-Hyun Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Seok-Goo Cho

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Seong-Taek Oh

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Jung-Sun Park

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Dae-Hee Sohn

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Sung Hee Yoon

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Eun-Kyung Kim

Catholic University of Korea

View shared research outputs
Researchain Logo
Decentralizing Knowledge