Network


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

Hotspot


Dive into the research topics where Eun Jin Ju is active.

Publication


Featured researches published by Eun Jin Ju.


Biomaterials | 2012

Use of macrophages to deliver therapeutic and imaging contrast agents to tumors

Jinhyang Choi; Hye-Yeong Kim; Eun Jin Ju; Joohee Jung; Jaesook Park; Hye-Kyung Chung; Jin Seong Lee; Jung Shin Lee; Heon Joo Park; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Drug targeting to tumors with limited toxicity and enhanced efficacy of drug is one of the important goals for cancer treatment pharmaceutics. Monocytes/macrophages are able to migrate to tumor sites across the blood barriers by acting as Trojan horses carrying drug cargoes. Taking this advantage, we have intended to develop an efficient administration system using a biologically active carrier of mouse peritoneal macrophage bearing liposomal doxorubicin (macrophage-LP-Dox). We expect that this system could improve the cancer therapeutic efficacy through deeper penetration into tumor even hypoxic region behind tumor blood vessel. We first confirmed that macrophages containing iron oxides could migrate and infiltrate into tumors effectively by MR imaging. Next, we showed that doxorubicin (Dox) encapsulated with liposomes (LP-Dox) was successfully loaded into macrophages, in which the biological activity of macrophage and cytotoxicity of Dox against tumor cells were well preserved. Delivery of Dox into tumor tissue by systemic administration of macrophage-LP-Dox was verified in both subcutaneous and metastasis xenograft tumor models. Importantly, the effective inhibition of in vivo tumor growth was proved with this system. Our results provide the feasibility of macrophages-LP-drug as an active biocarrier for the enhancement of therapeutic effects in cancer treatment and open new perspectives for the active delivery of drugs.


Journal of Controlled Release | 2015

Multifunctional hollow gold nanoparticles designed for triple combination therapy and CT imaging.

Jaesook Park; Jin Park; Eun Jin Ju; Seok Soon Park; Jinhyang Choi; Jae Hee Lee; Kyoung Jin Lee; Seol Hwa Shin; Eun Jung Ko; Intae Park; Chulhee Kim; Jung Jin Hwang; Jung Shin Lee; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Hollow gold nanoparticles (HGNP) are a novel class of hybrid metal nanoparticles whose unique optical and morphological properties have spawned new applications including more effective cancer therapy. The shell thickness of HGNPs can tune the surface plasmon resonance to the near infrared light, resulting in photothermal ablation of tumors with optimal light penetration in tissue. The hollow cavity within a HGNP is able to accommodate a high payload of chemotherapeutic agents. They have also been used for enhancing radiosensitization in tumors during radiotherapy due to the high X-ray absorption capability of gold particles. However, no report has yet been published that utilize HGNPs for the triple combination therapy and CT imaging. In this study, we synthesized HGNPs which exhibit better response to radiation for therapy and imaging and demonstrated the effects of combined chemotherapy, thermal and radiotherapy. This combination strategy presented delayed tumor growth by 4.3-fold and reduced tumors weight by 6.8-fold compared to control tumors. In addition, we demonstrated the feasibility of HGNP as a CT imaging agent. It is expected that translating these capabilities to human cancer patients could dramatically increase the antitumor effect and potentially overcome resistance to chemotherapeutic agents and radiation.


International Journal of Nanomedicine | 2012

Preclinical evaluation of injectable sirolimus formulated with polymeric nanoparticle for cancer therapy

Ha Na Woo; Hye Kyung Chung; Eun Jin Ju; Joohee Jung; Hye Won Kang; Sa-Won Lee; Min-Hyo Seo; Jin Seong Lee; Jung Shin Lee; Heon Joo Park; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Nanoparticles are useful delivery vehicles for promising drug candidates that face obstacles for clinical applicability. Sirolimus, an inhibitor of mammalian target of rapamycin has gained attention for targeted anticancer therapy, but its clinical application has been limited by its poor solubility. This study was designed to enhance the feasibility of sirolimus for human cancer treatment. Polymeric nanoparticle (PNP)–sirolimus was developed as an injectable formulation and has been characterized by transmission electron microscopy and dynamic light scattering. Pharmacokinetic analysis revealed that PNP–sirolimus has prolonged circulation in the blood. In addition, PNP–sirolimus preserved the in vitro killing effect of free sirolimus against cancer cells, and intravenous administration displayed its potent in vivo anticancer efficacy in xenograft tumor mice. In addition, PNP–sirolimus enhanced the radiotherapeutic efficacy of sirolimus both in vitro and in vivo. Clinical application of PNP–sirolimus is a promising strategy for human cancer treatment.


Journal of Nanomaterials | 2012

Immunocytes as a biocarrier to delivery therapeutic and imaging contrast agents to tumors

Jinhyang Choi; Ha-Na Woo; Eun Jin Ju; Joohee Jung; Hye-Kyung Chung; Jaesook Park; Seok Soon Park; Seol Hwa Shin; Hye Ji Park; Jin Seong Lee; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Radiotherapy for cancer treatment has been used for primary or adjuvant treatment in many types of cancer, and approximately half of all cancer patients are undergoing radiation. However, ionizing radiation exposure induces genetic alterations in cancer cells and results in recruitment of monocytes/macrophages by triggering signals released from these cells. Using this characteristic of monocytes/macrophages, we have attempted to develop a biocarrier loading radiosensitizing anticancer agents that can lead to enhance the therapeutic effect of radiation in cancer treatment. The aim of this study is to demonstrate the proof of this concept. THP-1 labeled with Qdot 800 or iron oxide (IO) effectively migrated into tumors of subcutaneous mouse model and increased recruitment after ionizing radiation. Functionalized liposomes carrying a radiosensitizing anticancer agent, doxorubicin, are successfully loaded in THP-1 (THP-1-LP-Dox) with reduced cytotoxicity, and THP-1-LP-Dox also was observed in tumors after intravenous administration. Here, we report that monocytes/macrophages as a biocarrier can be used as a selective tool for amplification of the therapeutic effects on radiotherapy for human cancer treatment.


Radiotherapy and Oncology | 2014

Ibulocydine sensitizes human cancers to radiotherapy by induction of mitochondria-mediated apoptosis.

Seok Soon Park; Eun Jin Ju; Seol Hwa Shin; Jinhyang Choi; Jaesook Park; Jae Hee Lee; Kyoung Jin Lee; Jin Park; Hye Ji Park; Eun Jung Ko; Jung Jin Hwang; Dong-Hoon Jin; Nayoung Suh; Dong-Hyung Cho; Jung Shin Lee; Si Yeol Song; B. Moon Kim; Seong-Yun Jeong; Eun Kyung Choi

BACKGROUND AND PURPOSE Ibulocydine (IB), a novel prodrug of CDK inhibitor, has been reported to have anti-cancer effect in human hepatoma cells. In order to address its feasibility as a radiosensitizer to improve radiotherapeutic efficacy for human cancers, this study was designed. MATERIAL AND METHODS Human cancer cells of lung and colon were treated with IB and/or radiotherapy (RT). The cellular effects were assessed by CCK-8, clonogenic, flow cytometric, and western blotting assays. In vivo radiotherapeutic efficacy was evaluated using the xenograft mouse model. RESULTS Combined treatment of IB and RT significantly reduced viability and survival fraction of the cells. Apoptotic cell death accompanied with activation of caspases, decrease in Bcl-2/Bax expression, loss of mitochondrial membrane potential (MMP) leading to release of cytochrome c into cytosol was observed. Recovery of Bcl-2 expression level by introducing Bcl-2 expressing plasmid DNA compromised the loss of MMP and apoptosis induced by IB and RT. In vivo therapeutic efficacy of combined treatment was verified in the xenograft mouse model, in which tumor growth was markedly delayed by RT with IB. CONCLUSIONS IB demonstrated the property of sensitizing human cancer cells to RT by induction of mitochondria-mediated apoptosis, suggesting that IB deserves to be applied for chemoradiotherapy.


The International Journal of Biochemistry & Cell Biology | 2017

Ibulocydine sensitizes human hepatocellular carcinoma cells to TRAIL-induced apoptosis via calpain-mediated Bax cleavage

Seok Soon Park; Eunjin Jwa; Seol Hwa Shin; Eun Jin Ju; Intae Park; Jhang Ho Pak; Jung Jin Hwang; Dong-Hyung Cho; B. Moon Kim; Sung-Bae Kim; Jung Shin Lee; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells without affecting the majority of normal human cells. However, hepatocellular carcinoma (HCC) cells often display resistance to TRAIL-induced apoptosis. Ibulocydine (IB) is an isobutyrate ester pro-drug of a novel synthetic Cdk inhibitor that targets Cdk7 and Cdk9. In this study, we show that treatment with subtoxic doses of IB in combination with TRAIL displays potent cytotoxicity in TRAIL-resistant human HCC cells. Combination of IB and TRAIL was found to synergistically induce apoptosis through activation of caspases, which was blocked by a pan-caspase inhibitor (zVAD). Although the expression of Mcl-1 and survivin were reduced by IB plus TRAIL, overexpression of Mcl-1 and survivin did not block the cell death induced by co-treatment. Moreover, overexpression of Bcl-xL did not significantly interfere with the cell death induced by co-treatment of IB and TRAIL. Interestingly, the combination treatment induced cleavage of Bax, which was translocated to mitochondria upon induction of apoptosis. Furthermore, down-regulation of Bax by small interfering RNA effectively reduced the cell death and loss of mitochondrial membrane potential (MMP) caused by co-treatment with IB and TRAIL. Finally, pre-treatment of HCC cells with a calpain inhibitor effectively blocked IB plus TRAIL-induced cleavage of Bax and apoptosis. Collectively, our results demonstrate that IB increases the sensitivity of human HCC cells to TRAIL via mitochondria signaling pathway mediated by calpain-induced cleavage of Bax, suggesting that combined treatment with IB and TRAIL may offer an effective therapeutic strategy for human HCC.


International Journal of Nanomedicine | 2015

Nanoparticulated docetaxel exerts enhanced anticancer efficacy and overcomes existing limitations of traditional drugs.

Jinhyang Choi; Eunjung Ko; Hye-Kyung Chung; Jae Hee Lee; Eun Jin Ju; Hyun Kyung Lim; Intae Park; Kab-Sig Kim; Joo-Hwan Lee; Woo-Chan Son; Jung Shin Lee; Joohee Jung; Seong-Yun Jeong; Si Yeol Song; Eun Kyung Choi

Nanoparticulation of insoluble drugs improves dissolution rate, resulting in increased bioavailability that leads to increased stability, better efficacy, and reduced toxicity of drugs. Docetaxel (DTX), under the trade name Taxotere™, is one of the representative anticancer chemotherapeutic agents of this era. However, this highly lipophilic and insoluble drug has many adverse effects. Our novel and widely applicable nanoparticulation using fat and supercritical fluid (NUFS™) technology enabled successful nanoscale particulation of DTX (Nufs-DTX). Nufs-DTX showed enhanced dissolution rate and increased aqueous stability in water. After confirming the preserved mechanism of action of DTX, which targets microtubules, we showed that Nufs-DTX exhibited similar effects in proliferation and clonogenic assays using A549 cells. Interestingly, we observed that Nufs-DTX had a greater in vivo tumor growth delay effect on an A549 xenograft model than Taxotere™, which was in agreement with the improved drug accumulation in tumors according to the biodistribution result, and was caused by the enhanced permeability and retention (EPR) effect. Although both Nufs-DTX and Taxotere™ showed negative results for our administration dose in the hematologic toxicity test, Nufs-DTX showed much less toxicity than Taxotere™ in edema, paralysis, and paw-withdrawal latency on a hot plate analysis that are regarded as indicators of fluid retention, peripheral neuropathy, and thermal threshold, respectively, for toxicological tests. In summary, compared with Taxotere™, Nufs-DTX, which was generated by our new platform technology using lipid, supercritical fluid, and carbon dioxide (CO2), maintained its biochemical properties as a cytotoxic agent and had better tumor targeting ability, better in vivo therapeutic effect, and less toxicity, thereby overcoming the current hurdles of traditional drugs.


International Journal of Oncology | 2015

A cisplatin‑incorporated liposome that targets the epidermal growth factor receptor enhances radiotherapeutic efficacy without nephrotoxicity

Joohee Jung; Seong Yun Jeong; Seok Soon Park; Seol Hwa Shin; Eun Jin Ju; Jinhyang Choi; Jaesook Park; Jae Hee Lee; Inki Kim; Young Ah Suh; Jung Jin Hwang; Shun'ichi Kuroda; Jung Shin Lee; Si Yeol Song; Eun Kyung Choi

Radiotherapy (RT) is one of the major modalities for non‑small cell lung cancer (NSCLC), but its efficacy is often compromised by cellular resistance caused by various mechanisms including the overexpression of epidermal growth factor receptor (EGFR). Although cis‑diamminedichloroplatinum(Ⅱ) (cisplatin, CDDP) has been well characterized as an effective radiosensitizer, its clinical application is limited by its severe nephrotoxic effects. In our current study, we developed a CDDP‑incorporated liposome (LP) conjugated with EGFR antibodies (EGFR:LP‑CDDP) and evaluated its potential to radiosensitize EGFR‑overexpressing cells without exerting nephrotoxic effects. EGFR:LP‑CDDP showed higher cytotoxicity than non‑targeting liposomal CDDP (LP‑CDDP) in the cells expressing EGFR in vitro. In an A549 cell‑derived xenograft tumor mouse model, increased delays in tumor growth were observed in the mice treated with a combination of EGFR:LP‑CDDP and radiation. Notably, the EGFR:LP‑CDDP‑treated animals showed no differences in body weight loss, survival rates of nephrotoxicity compared with untreated control mice. In contrast, the use of CDDP caused lower body weights and poorer survival outcomes accompanied by a significant level of nephrotoxicity [e.g., decreased kidney weight, increased blood urea nitrogen (BUN) and creatinine, and pathological change]. These findings suggest the feasibility of using EGFR:LP‑CDDP to radiosensitize cells in a targeted manner without inducing nephrotoxic effects. This compound may therefore have clinical potential as part of a tailored chemoradiotherapy strategy.


Translational cancer research | 2018

Docetaxel-polymeric nanoparticle enhances radiotherapeutic efficacy in human pancreatic cancer

Jin Park; Seok Soon Park; Kyoung Jin Lee; Eun Jin Ju; Seol Hwa Shin; Eun Jeong Ko; Sa-Won Lee; Min Hyo Seo; Jung Shin Lee; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Background: Nanoparticle therapeutics is promising platform for cancer treatment. In our previous study, we have developed polymeric nanoparticles (PNP) in which docetaxel was incorporated to reduce the side effects and improve the therapeutic efficacy, and recently finished its phase 1 clinical study in patients with solid tumors. Methods: Radiotherapeutic efficacy of the docetaxel-contained PNP (DTX-PNP) in pancreatic cancer cells was determined by both in vitro and in vivo assay such as clonogenic survival assay with cancer cell lines, western blot for apoptotic cell death and tumor growth inhibition assay using several kinds of xenograft models. The tumors derived from human pancreatic cancer AsPC-1 or BxPC-3 cells were analyzed by immunohistochemistry (IHC) to detect in apoptosis and tubulin polymerization induced by DTX-PNP. The combinational therapeutic effect of DTX-PNP and ionizing radiation (IR) was evaluated in vivo mice models of AsPC-1 or BxPC-3 cell line-derived xenograft models and patient-derived xenograft model, and compared to that of reference drugs. Results: DTX-PNP in combination with IR showed high cytotoxicity to pancreatic cancer cells, and ultimate inhibition of cell proliferation as determined via in vitro assays. In vivo radiotherapeutic efficacy was markedly enhanced by intravenous injection of DTX-PNP comparing to Gemzar, a common chemoradiation therapeutic agent in pancreatic cancer. Conclusions: These results suggested DTX-PNP can hold an invaluable and promising position in treating human pancreatic cancer as a novel and effective radiosensitizing agent.


Amino Acids | 2015

Novel peptides functionally targeting in vivo human lung cancer discovered by in vivo peptide displayed phage screening

Kyoung Jin Lee; Jae Hee Lee; Hye Kyung Chung; Jinhyang Choi; Jaesook Park; Seok Soon Park; Eun Jin Ju; Jin Park; Seol Hwa Shin; Hye Ji Park; Eun Jung Ko; Nayoung Suh; Inki Kim; Jung Jin Hwang; Si Yeol Song; Seong-Yun Jeong; Eun Kyung Choi

Collaboration


Dive into the Eun Jin Ju's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge