Network


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

Hotspot


Dive into the research topics where Min Hee Hong is active.

Publication


Featured researches published by Min Hee Hong.


Scientific Reports | 2016

PD-L1 expression on immune cells, but not on tumor cells, is a favorable prognostic factor for head and neck cancer patients

Hye Ryun Kim; Sang Jun Ha; Min Hee Hong; Su Jin Heo; Yoon Woo Koh; Eun Chang Choi; Eun-Kyung Kim; Kyoung Ho Pyo; Inkyung Jung; Daekwan Seo; Jae Woo Choi; Byoung Chul Cho; Sun Och Yoon

To investigate the expression of programmed death-ligand 1 (PD-L1) and immune checkpoints and their prognostic value for resected head and neck squamous cell cancer (HNSCC). PD-L1 expression on tumor cells (TC) and tumor-infiltrating immune cells (IC), abundance of tumor-infiltrating lymphocytes (TILs), and expression of the immune checkpoints were investigated in 402 HNSCC patients. PD-L1 expression on TC and IC was categorized into four groups according to the percentage of PD-L1-positive cells. PD-L1 positivity was defined as ≥5% of cells based on immunohistochemistry. High PD-L1 expression on IC, but not TC, was an independent favorable prognostic factor for RFS and OS adjusted for age, gender, smoking, stage, and HPV. High frequencies of CD3+ or CD8+ TILs, Foxp3+ Tregs, and PD-1+ TILs were strongly associated with favorable prognosis. PD-L1 was exclusively expressed on either TC or IC. Transcriptome analysis demonstrated that IC3 expressed higher levels of the effector T cell markers than TC3, suggesting that PD-L1 expression is regulated via an adaptive IFNγ-mediated mechanism. High PD-L1 expression on IC, but not TC, and high abundance of PD-1+ T cells and Foxp3+ Tregs are favorable prognostic factors for resected HNSCC. This study highlights the importance of comprehensive assessment of both TC and IC.


Annals of Oncology | 2017

Co-clinical trials demonstrate predictive biomarkers for dovitinib, an FGFR inhibitor, in lung squamous cell carcinoma

H.R. Kim; H. N. Kang; Hyo Sup Shim; Eun Young Kim; J. Kim; Dong Joon Kim; Jin Gu Lee; Chang Young Lee; Min Hee Hong; S.-M. Kim; H. Kim; K.-H. Pyo; M. R. Yun; H. J. Park; J. Y. Han; H. A. Youn; Myung Ju Ahn; Soonmyung Paik; T.-M. Kim; Byoung Chul Cho

BackgroundnWe conducted co-clinical trials in patient-derived xenograft (PDX) models to identify predictive biomarkers for the multikinase inhibitor dovitinib in lung squamous cell carcinoma (LSCC).nnnMethodsnThe PDX01-02 were established from LSCC patients enrolled in the phase II trial of dovitinib (NCT01861197) and PDX03-05 were established from LSCC patients receiving surgery. These five PDX tumors were subjected to in vivo test of dovitinib efficacy, whole exome sequencing and gene expression profiling.nnnResultsnThe PDX tumors recapitulate histopathological properties and maintain genomic characteristics of originating tumors. Concordant with clinical outcomes of the trial enrolled-LSCC patients, dovitinib produced substantial tumor regression in PDX-01 and PDX-05, whereas it resulted in tumor progression in PDX-02. PDX-03 and -04 also displayed poor antitumor efficacy to dovitinib. Mutational and genome-wide copy number profiles revealed no correlation between genomic alterations of FGFR1-3 and sensitivity to dovitinib. Of note, gene expression profiles revealed differentially expressed genes including FGF3 and FGF19 between PDX-01 and 05 and PDX-02-04. Pathway analysis identified two FGFR signaling-related gene sets, FGFR ligand binding/activation and SHC-mediated cascade pathway were substantially up-regulated in PDX-01 and 05, compared with PDX-02-04. The comparison of gene expression profiles between dovitinib-sensitive versus -resistant lung cancer cell lines in the Cancer Cell Line Encyclopedia database also found that transcriptional activation of 18u2009key signaling components in FGFR pathways can predict the sensitivity to dovitinib both in cell lines and PDX tumors. These results highlight FGFR pathway activation as a key molecular determinant for sensitivity to dovitinib.nnnConclusionsnFGFR gene expression signatures are predictors for the response to dovitinib in LSCC.


Oncotarget | 2017

Concordance of programmed death-ligand 1 expression between primary and metastatic non-small cell lung cancer by immunohistochemistry and RNA in situ hybridization

Hye Ryun Kim; Yoon Jin Cha; Min Hee Hong; Manoj Gandhi; Shauna Levinson; Inkyung Jung; Jin Gu Lee; Chang Young Lee; Byoung Chul Cho; Sang Jun Ha; Hyo Sup Shim

We investigated the concordance of programmed death-ligand 1 (PD-L1) expression between primary cancer at initial diagnosis and metastasis at recurrence in resected non-small cell lung cancer (NSCLC). PD-L1 expression was evaluated using the SP142 assay in 37 NSCLC patients with paired primary lung cancer and surgically resected metastases at recurrence. PD-L1 positivity was defined as immunohistochemistry (IHC) and also evaluated by RNA in situ hybridization (RISH). The concordance rate of PD-L1 between primaries and metastases and correlation with clinicopathological factors were analyzed. PD-L1 expression was higher in squamous cell carcinoma, wild-type EGFR, and smokers than in non-squamous carcinoma, mutant EGFR, and never smokers, respectively. PD-L1 positivity was observed in 18.9% of primaries and 21.6% of metastases. IHC demonstrated 78.4% concordance of PD-L1 positivity between primary and metastatic cancers. In 10.8% of cases, PD-L1 positivity was higher in primaries than in metastases, and vice versa in the remaining 10.8%. By PD-L1 RISH, 35.1% of primaries and 27.0% of metastases demonstrated PD-L1 positivity. There was 62.2% concordance in PD-L1 by RISH between the primaries and metastases. Our results thus highlight the clinical importance of replacing metastases with primary archival tissue, particularly when re-biopsy is difficult at recurrence.


Lung Cancer | 2018

Establishment of a platform of non-small-cell lung cancer patient-derived xenografts with clinical and genomic annotation

Han Na Kang; Jae Woo Choi; Hyo Sup Shim; Jinna Kim; Dae Joon Kim; Chang Young Lee; Min Hee Hong; Seong Yong Park; A-Young Park; Eun Joo Shin; Seo Yoon Lee; Kyoung-Ho Pyo; Mi Ran Yun; Hun Mi Choi; S. Lee; Seok-Young Kim; Hanna Lee; Soonmyung Paik; Byoung Chul Cho; Jin Gu Lee; Hye Ryun Kim

BACKGROUNDnPreclinical models that can better predict therapeutic activity in clinical trials are needed in this era of personalized cancer treatment. Herein, we established genomically and clinically annotated patient-derived xenografts (PDXs) from non-small-cell lung cancer (NSCLC) patients and investigated whether these PDXs would faithfully recapitulate patient responses to targeted therapy.nnnMETHODSnPatient-derived tumors were implanted in immunodeficient mice and subsequently expanded via re-implantation. Established PDXs were examined by light microscopy, genomic profiling, and in vivo drug testing, and the successful engraft rate was analyzed with the mutation profile, histology, or acquisition method. Finally, the drug responses of PDXs were compared with the clinical responses of the respective patients.nnnRESULTSnUsing samples from 122 patients, we established 41 NSCLC PDXs [30 adenocarcinoma (AD), 11 squamous cell carcinoma (SQ)], among which the following driver mutation were observed: 13 EGFR-mutant, 4 ALK-rearrangement, 1 ROS1-rearrangement, 1 PIK3CA-mutant, 1 FGFR1-amplification, and 2 KRAS-mutant. We rigorously characterized the relationship of clinical features to engraftment rate and latency rates. The engraft rates were comparable across histologic type. The AD engraft rate tended to be higher for surgically resected tissues relative to biopsies, whereas similar engraft rates was observed for SQ, irrespective of the acquisition method. Notably, EGFR-mutants demonstrated significantly longer latency time than EGFR-WT (86 vs. 37days, Pu2009=u20090.007). The clinical responses were recapitulated by PDXs harboring driver gene alteration (EGFR, ALK, ROS1, or FGFR1) which regressed to their target inhibitors, suggesting that established PDXs comprise a clinically relevant platform.nnnCONCLUSIONnThe establishment of genetically and clinically annotated NSCLC PDXs can yield a robust preclinical tool for biomarker, therapeutic target, and drug discovery.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2018

Efficacy and safety of vinorelbine plus cisplatin chemotherapy for patients with recurrent and/or metastatic salivary gland cancer of the head and neck

Min Hee Hong; Chang Gon Kim; Yoon Woo Koh; Eun Chang Choi; Jinna Kim; Sun Och Yoon; Hye Ryun Kim; Byoung Chul Cho

The purpose of this study was to investigate the efficacy and safety of vinorelbine plus cisplatin chemotherapy in patients with recurrent and/or metastatic salivary gland cancer of the head and neck.


Journal of Thoracic Oncology | 2017

P2.06-003 A Phase Ib Study of the Combination of Afatinib and Ruxolitinib in EGFR Mutant Non-Small Cell Lung Cancer (NSCLC) Progressed on EGFR-TKI: Topic: Phase I Trials

Ji Soo Park; Hye Ryun Kim; Min Hee Hong; Byoung Chul Cho


Journal of Thoracic Oncology | 2018

MA26.09 Lazertinib, a Third Generation EGFR-TKI, in Patients with EGFR-TKI-Resistant NSCLC: Updated Results of a Phase I/II Study

Byoung Chul Cho; J. Han; S. Kim; Kyung-Jong Lee; D-W. Kim; Y. Lee; Gyeong-Won Lee; J. Lee; Eou-Sik Cho; J. Kim; Soo-Hyun Lee; Young Joo Min; Sung Wook Shin; H.R. Kim; Min Hee Hong; J.S. Ahn; Shin Myung Kang; S.B. Jang; So-Jung Choi; Myung-Ju Ahn


Journal of Thoracic Oncology | 2018

P1.01-01 ROS1-Positive Non-Small Cell Lung Cancer: Real-World Data in Korea

B.C. Ahn; Sunhoo Park; Sung Won Lim; H.R. Kim; Min Hee Hong; Byoung Chul Cho; Myung-Ju Ahn


Annals of Oncology | 2018

1929PTumor-infiltrating CD3+ lymphocytes and ICOS+ T-cells predict a favorable survival in resected esophageal squamous cell carcinoma

Min Hee Hong; S-J Shin; Y.S. Choi; Byoung Chul Cho; H.R. Kim


Annals of Oncology | 2018

1429PMolecular landscape of osimertinib resistance revealed by targeted panel sequencing and patient-derived cancer models in non-small cell lung cancer patients

Min Hee Hong; Min Hwan Kim; S-Y Kim; S G Heo; H-N Kang; C-W Park; J C Barrett; D Stetson; J Chmielecki; A Markovets; H.R. Kim; Byoung Chul Cho

Collaboration


Dive into the Min Hee Hong'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