Network


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

Hotspot


Dive into the research topics where Yeonju Lee is active.

Publication


Featured researches published by Yeonju Lee.


Scientific Reports | 2015

Microfluidic cytometric analysis of cancer cell transportability and invasiveness

Zongbin Liu; Yeonju Lee; Joon Hee Jang; Ying Li; Xin Han; Kenji Yokoi; Mauro Ferrari; Ledu Zhou; Lidong Qin

The extensive phenotypic and functional heterogeneity of cancer cells plays an important role in tumor progression and therapeutic resistance. Characterizing this heterogeneity and identifying invasive phenotype may provide possibility to improve chemotherapy treatment. By mimicking cancer cell perfusion through circulatory system in metastasis, we develop a unique microfluidic cytometry (MC) platform to separate cancer cells at high throughput, and further derive a physical parameter ‘transportability’ to characterize the ability to pass through micro-constrictions. The transportability is determined by cell stiffness and cell-surface frictional property, and can be used to probe tumor heterogeneity, discriminate more invasive phenotypes and correlate with biomarker expressions in breast cancer cells. Decreased cell stiffness and cell-surface frictional force leads to an increase in transportability and may be a feature of invasive cancer cells by promoting cell perfusion through narrow spaces in circulatory system. The MC-Chip provides a promising microfluidic platform for studying cell mechanics and transportability could be used as a novel marker for probing tumor heterogeneity and determining invasive phenotypes.


Journal of Visualized Experiments | 2015

Porous silicon microparticles for delivery of siRNA therapeutics

Jianliang Shen; Xiaoyan Wu; Yeonju Lee; Joy Wolfram; Zhizhou Yang; Zong-Wan Mao; Mauro Ferrari; Haifa Shen

Small interfering RNA (siRNA) can be used to suppress gene expression, thereby providing a new avenue for the treatment of various diseases. However, the successful implementation of siRNA therapy requires the use of delivery platforms that can overcome the major challenges of siRNA delivery, such as enzymatic degradation, low intracellular uptake and lysosomal entrapment. Here, a protocol for the preparation and use of a biocompatible and effective siRNA delivery system is presented. This platform consists of polyethylenimine (PEI) and arginine (Arg)-grafted porous silicon microparticles, which can be loaded with siRNA by performing a simple mixing step. The silicon particles are gradually degraded over time, thereby triggering the formation of Arg-PEI/siRNA nanoparticles. This delivery vehicle provides a means for protecting and internalizing siRNA, without causing cytotoxicity. The major steps of polycation functionalization, particle characterization, and siRNA loading are outlined in detail. In addition, the procedures for determining particle uptake, cytotoxicity, and transfection efficacy are also described.


Clinical Cancer Research | 2016

Transforming Growth Factor-β Limits Secretion of Lumican by Activated Stellate Cells within Primary Pancreatic Adenocarcinoma Tumors.

Ya'an Kang; David Roife; Yeonju Lee; Hailong Lv; Rei Suzuki; Jianhua Ling; Mayrim V. Rios Perez; Xinqun Li; Bingbing Dai; Michael Pratt; Mark J. Truty; Deyali Chatterjee; Huamin Wang; Ryan M. Thomas; Yu Wang; Eugene J. Koay; Paul J. Chiao; Matthew H. Katz; Jason B. Fleming

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is lethal cancer whose primary tumor is characterized by dense composition of cancer cells, stromal cells, and extracellular matrix (ECM) composed largely of collagen. Within the PDAC tumor microenvironment, activated pancreatic stellate cells (PSC) are the dominant stromal cell type and responsible for collagen deposition. Lumican is a secreted proteoglycan that regulates collagen fibril assembly. We have previously identified that the presence of lumican in the ECM surrounding PDAC cells is associated with improved patient outcome after multimodal therapy and surgical removal of localized PDAC. Experimental Design: Lumican expression in PDAC from 27 patients was determined by IHC and quantitatively analyzed for colocalization with PSCs. In vitro studies examined the molecular mechanisms of lumican transcription and secretion from PSCs (HPSCs and HPaSteC), and cell adhesion and migration assays examined the effect of lumican on PSCs in a collagen-rich environment. Results: Here we identify PSCs as a significant source of extracellular lumican production through quantitative IHC analysis. We demonstrate that the cytokine, TGF-β, negatively regulates lumican gene transcription within HPSCs through its canonical signaling pathway and binding of SMAD4 to novel SBEs identified within the promoter region. In addition, we found that the ability of HPSCs to produce and secrete extracellular lumican significantly enhances HPSCs adhesion and mobility on collagen. Conclusions: Our results demonstrate that activated pancreatic stellate cells within PDAC secrete lumican under the negative control of TGF-β; once secreted, the extracellular lumican enhances stellate cell adhesion and mobility in a collagen-rich environment. Clin Cancer Res; 22(19); 4934–46. ©2016 AACR.


Oncogene | 2017

Prolonged exposure to extracellular lumican restrains pancreatic adenocarcinoma growth

Xinqun Li; Y Kang; David Roife; Yeonju Lee; Michael Pratt; Mayrim V. Rios Perez; Bingbing Dai; E J Koay; J B Fleming

We previously demonstrated that pancreatic stellate cells within pancreatic ductal adenocarcinoma (PDAC) stroma secrete lumican and its presence is associated with prolonged survival of patients with localized PDAC. Here, we observed that extracellular lumican decreases PDAC tumour cell growth in xenograft and syngeneic orthotopic animal models, and induces growth inhibition of low-passage human PDAC cells in a species-specific manner. PDAC cells grown in variant culture conditions and exposed to extracellular lumican display typical characterizations of cancer cell in a quiescent state, such as growth inhibition, apoptosis, G0/G1 arrest and chemoresistance. Importantly, extracellular lumican is associated with diminished ERK1/2 phosphorylation and increased p38 phosphorylation within PDAC cells. We further demonstrated that extracellular lumican physically binds with EGFR to trigger EGFR internalization and downregulation of EGFR and its downstream signal molecule ERK. Lumican enhances casitas B-lineage lymphoma expression, which stabilized the TGFβ Type II receptor sensitizing PDAC cells to TGFβ-mediated activation of p38 and SMAD signals. These provide a mechanism for the shift in signalling and phenotypic changes we observed after prolonged exposure to lumican. Together, our findings demonstrate that stromal lumican restrains PDAC cell growth through mediating cell entry into a quiescent state.


Advances in radiation oncology | 2017

Dose escalation with an IMRT technique in 15 to 28 fractions is better tolerated than standard doses of 3DCRT for LAPC

L.E. Colbert; Shalini Moningi; Awalpreet S. Chadha; Ahmed M. Amer; Yeonju Lee; Robert A. Wolff; Gauri R. Varadhachary; Jason B. Fleming; Matthew H. Katz; Prajnan Das; Sunil Krishnan; Eugene J. Koay; Peter J. Park; Christopher H. Crane; Cullen M. Taniguchi

Purpose To review acute and late toxicities after chemoradiation for locally advanced pancreatic ductal adenocarcinoma in patients who were treated with escalated dose radiation (EDR). Methods and materials Maximum Common Terminology Criteria for Adverse Events Version 4.0 acute toxicities (AT) during radiation and within 60 days after radiation were recorded for both acute gastrointestinal toxicity and overall toxicity (OT). Late toxicities were also recorded. EDR was generally delivered with daily image guidance and breath-hold techniques using intensity modulated radiation therapy (IMRT) planning. These were compared with patients who received standard dose radiation (SDR) delivered as 50.4 Gy in 28 fractions using 3-dimensional chemoradiation therapy planning. Results A total of 59 of 154 patients (39%) received EDR with biologically equivalent doses >70 Gy. The most frequent schedules were 63 Gy in 28 fractions (19 of 154 patients), 67.5 Gy in 15 fractions (10 of 154 patients), and 70 Gy in 28 fractions (15 of 154 patients). No grade 4 or grade 5 OT or late toxicities were reported. Rates of grade 3 acute gastrointestinal toxicity were significantly lower in patients who received EDR compared with SDR (1% vs 14%; P < .001). Similarly, rates of grade 3 OT were also lower for EDR compared with SDR (4% vs 16%; P = .004). The proportion of patients who experienced no AT was higher in the EDR group than the SDR group (36% vs 15%; P = .001). For EDR patients treated with IMRT, a lower risk of AT was associated with a later treatment year (P = .007), nonpancreatic head tumor location (P = .01), breath-hold (P = .002), 4-dimensional computed tomography (P = .003), computed tomography on rails (P = .002), and lower stomach V40 (P = .03). With a median time of 12 months (range, 1-79 months) from the start of radiation therapy to the last known follow-up in the EDR group, 51 of 59 patients (86%) had no late toxicity. Six of 59 EDR patients (10%) had either strictures or gastrointestinal bleeding that required intervention. No significant predictors of late toxicity were identified. Conclusion Overall acute and late toxicity rates were low with EDR using an IMRT technique with image guidance and respiratory gating.


Cell Death & Differentiation | 2018

Hypoxia-induced autophagy of stellate cells inhibits expression and secretion of lumican into microenvironment of pancreatic ductal adenocarcinoma

Xinqun Li; Yeonju Lee; Ya’an Kang; Bingbing Dai; Mayrim V. Rios Perez; Michael Pratt; Eugene J. Koay; Michael Kim; Rolf A. Brekken; Jason B. Fleming

Lumican is secreted by pancreatic stellate cells and inhibits cancer progression. Extracellular lumican inhibits cancer cell replication and restrains growth of early-stage pancreatic adenocarcinoma (PDAC) such that patients with tumors containing stromal lumican experience a three-fold longer survival after treatment. In the present study, patient tumor tissues, ex-vivo cultures of patient-derived xenografts (PDX), PDAC stellate and tumor cells were used to investigate whether hypoxia (1% O2) within the tumor microenvironment influences stromal lumican expression and secretion. We observed that hypoxia significantly reduced lumican expression and secretion from pancreatic stellate cells, but not cancer cells. Although hypoxia enhanced lactate dehydrogenase A (LDHA) expression and lactate secretion from all cells, neither hypoxia-induced nor exogenous lactate influenced lumican expression. Autophagy was induced by hypoxia in ex vivo cultures of PDX and pancreatic stellate cells, but not cancer cells cultured in 2D. Autophagic flux inhibitors, bafilomycin A1, chloroquine diphosphate salt, and ammonium chloride prevented hypoxia-mediated reduction in lumican expression in stellate cells. Furthermore, inhibition of AMP-regulated protein kinase (AMPK) phosphorylation or hypoxia-inducible factor (HIF)-1α expression within hypoxic stellate cells restored lumican expression levels. Hypoxia did not affect lumican mRNA expression, indicating that hypoxia-induced reduction of lumican occurs post-transcriptionally; in addition, AMPK inhibition prevented hypoxia-reduced phosphorylation of the mTOR/p70S6K/4EBP signaling pathway, a key contributor to protein synthesis. Taken together, these findings demonstrate that hypoxia reduces stromal lumican in PDAC through autophagy-mediated degradation and reduction in protein synthesis within pancreatic cancer stellate cells.


Cancer | 2018

Imaging-based biomarkers: Changes in the tumor interface of pancreatic ductal adenocarcinoma on computed tomography scans indicate response to cytotoxic therapy

Ahmed M. Amer; Mohamed Zaid; Baishali Chaudhury; Dalia Elganainy; Yeonju Lee; Christopher Wilke; Jordan M. Cloyd; Huamin Wang; Anirban Maitra; Robert A. Wolff; Gauri R. Varadhachary; Michael J. Overman; J. E. Lee; Jason B. Fleming; Ching Wei Tzeng; Matthew H. Katz; Emma B. Holliday; Sunil Krishnan; Bruce D. Minsky; Joseph M. Herman; Cullen M. Taniguchi; Prajnan Das; Christopher H. Crane; Ott Le; Priya Bhosale; Eric P. Tamm; Eugene J. Koay

The assessment of pancreatic ductal adenocarcinoma (PDAC) response to therapy remains challenging. The objective of this study was to investigate whether changes in the tumor/parenchyma interface are associated with response.


Cancer Letters | 2017

Gemcitabine enhances the transport of nanovector-albumin-bound paclitaxel in gemcitabine-resistant pancreatic ductal adenocarcinoma

Carlotta Borsoi; Fransisca Leonard; Yeonju Lee; Mohamed Zaid; Dalia Elganainy; Jenolyn F. Alexander; Megumi Kai; Yan Ting Liu; Ya'an Kang; Xuewu Liu; Eugene J. Koay; Mauro Ferrari; Biana Godin; Kenji Yokoi


Clinical Cancer Research | 2018

A Visually Apparent and Quantifiable CT Imaging Feature Identifies Biophysical Subtypes of Pancreatic Ductal Adenocarcinoma

Eugene J. Koay; Yeonju Lee; Vittorio Cristini; John Lowengrub; Ya'an Kang; F. Anthony San Lucas; Brian P. Hobbs; Rong Ye; Dalia Elganainy; Muayad Almahariq; Ahmed M. Amer; Deyali Chatterjee; Huaming Yan; Peter C. Park; Mayrim V. Rios Perez; Dali Li; Naveen Garg; Kim Anna Reiss; Shun Yu; Anil K. Chauhan; Mohamed Zaid; Newsha Nikzad; Robert A. Wolff; Milind Javle; Gauri R. Varadhachary; Rachna T. Shroff; Prajnan Das; Jeffrey E. Lee; Mauro Ferrari; Anirban Maitra


Journal of Clinical Oncology | 2017

Tumor interface stability on CT imaging is an early marker of response to cytotoxic therapy in pancreatic ductal adenocarcinoma.

Christopher Wilke; Ahmed M. Amer; Dalia Elganainy; Priya Bhosale; Ott Le; Yeonju Lee; Jordan M. Cloyd; Huamin Wang; Joseph M. Herman; Cullen M. Taniguchi; Andrew D. Rhim; Jason B. Fleming; Anirban Maitra; Robert A. Wolff; Gauri R. Varadhachary; Jeffrey E. Lee; Matthew H. Katz; Eric P. Tamm; Eugene Jon Koay

Collaboration


Dive into the Yeonju Lee's collaboration.

Top Co-Authors

Avatar

Jason B. Fleming

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eugene J. Koay

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gauri R. Varadhachary

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew H. Katz

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ahmed M. Amer

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cullen M. Taniguchi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Prajnan Das

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Robert A. Wolff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher H. Crane

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dalia Elganainy

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge