Network


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

Hotspot


Dive into the research topics where Mi Young Cha is active.

Publication


Featured researches published by Mi Young Cha.


International Journal of Cancer | 2012

Antitumor activity of HM781-36B, a highly effective pan-HER inhibitor in erlotinib-resistant NSCLC and other EGFR-dependent cancer models†

Mi Young Cha; Kwang-Ok Lee; Mi-Ra Kim; Ji Yeon Song; Kyu Hang Lee; Jongmin Park; Yun Jung Chae; Young Hoon Kim; Kwee Hyun Suh; Gwan Sun Lee; Seung Bum Park; Maeng Sup Kim

The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases has been implicated in a variety of cancers. In particular, activating mutations such as the L858R point mutation in exon 21 and the small in‐frame deletions in exon 19 of the EGFR tyrosine kinase domain are correlated with sensitivity to EGFR tyrosine kinase inhibitors in non‐small cell lung cancer (NSCLC) patients. Clinical treatment of patients is limited by the development of drug resistance resulting mainly from a gatekeeper mutation (T790M). In this study, we evaluated the therapeutic potential of a novel, irreversible pan‐HER inhibitor, HM781‐36B. The results from this study show that HM781‐36B is a potent inhibitor of EGFR in vitro, including the EGFR‐acquired resistance mutation (T790M), as well as HER‐2 and HER‐4, compared with other EGFR tyrosine kinases inhibitors (erlotinib, lapatinib and BIBW2992). HM781‐36B treatment of EGFR DelE746_A750‐harboring erlotinib‐sensitive HCC827 and EGFR L858R/T790M‐harboring erlotinib‐resistant NCI‐H1975 NSCLC cells results in the inhibition of EGFR phosphorylation and the subsequent deactivation of downstream signaling proteins. Additionally, HM781‐36B shows an excellent efficacy in a variety of EGFR‐ and HER‐2‐dependent tumor xenograft models, including erlotinib‐sensitive HCC827 NSCLC cells, erlotinib‐resistant NCI‐H1975 NSCLC cells, HER‐2 overexpressing Calu‐3 NSCLC cells, NCI‐N87 gastric cancer cells, SK‐Ov3 ovarian cancer cells and EGFR‐overexpressing A431 epidermoid carcinoma cancer cells. On the basis of these preclinical results, HM781‐36B is the most potent pan‐HER inhibitor, which will be advantageous for the treatment of patients with NSCLC including clinical limitation caused by acquired mutation (EGFR T790M), breast cancer and gastric cancer.


Journal of Medicinal Chemistry | 2009

Discovery of A Novel Her-1/Her-2 Dual Tyrosine Kinase Inhibitor for the Treatment of Her-1 Selective Inhibitor-Resistant Non-small Cell Lung Cancer

Mi Young Cha; Kwang-Ok Lee; Jong Woo Kim; Chang Gon Lee; Ji Yeon Song; Young Hoon Kim; Gwan Sun Lee; Seung Bum Park; Maeng Sup Kim

A novel series of (S)-1-acryloyl-N-[4-(arylamino)-7-(alkoxy)quinazolin-6-yl]pyrrolidine-2-carboxamides were synthesized and evaluated as Her-1/Her-2 dual inhibitors. In contrast to the Her-1 selective inhibitors, our novel compounds are irreversible inhibitors of Her-1 and Her-2 tyrosine kinases with the potential to overcome clinically relevant, mutation-induced drug resistance. The selected compounds (19c, 19d) showed excellent EGFR inhibition activity even toward the T790M mutation of Her-1 tyrosine kinase with excellent selectivity. The excellent pharmacokinetic profiles of these compounds in rats and their robust in vivo efficacy in an A431 xenograft model clearly demonstrate that they merit further investigation as novel therapeutic agents for EGFR-targeting treatment of solid tumors, especially Her-1 selective inhibitor-resistant non-small cell lung cancer.


Journal of Medicinal Chemistry | 2012

Synthesis and biological evaluation of pyrimidine-based dual inhibitors of human epidermal growth factor receptor 1 (HER-1) and HER-2 tyrosine kinases.

Mi Young Cha; Kwang-Ok Lee; SeokJong Kang; Young Hee Jung; Ji Yeon Song; Kyung Jin Choi; Joo Yun Byun; Hanjae Lee; Gwan Sun Lee; Seung Bum Park; Maeng Sup Kim

A novel series of N(4)-(3-chlorophenyl)-5-(oxazol-2-yl)pyrimidine-4,6-diamines were synthesized and evaluated as dual inhibitors of HER-1/HER-2 tyrosine kinases. In contrast to the currently approved HER-2-targeted agent (lapatinib, 1), our irreversible HER-1/HER-2 inhibitors have the potential to overcome the clinically relevant and mutation-induced drug resistance. The selected compound (19a) showed excellent inhibitory activity toward HER-1/HER-2 tyrosine kinases with selectivity over 20 other kinases and inhibited the proliferation of both cancer cell types: lapatinib-sensitive cell lines (SK-Br3, MDA-MB-175, and N87) and lapatinib-resistant cell lines (MDA-MB-453, H1781, and H1975). The excellent pharmacokinetic profiles of 19a in mice and rats led us to further investigation of a novel therapeutic agent for HER-2-targeting treatment of solid tumors, especially HER-2-positive breast/gastric cancer and HER-2-mutated lung cancer.


Cancer Research | 2014

Abstract LB-100: Discovery of HM61713 as an orally available and mutant EGFR selective inhibitor

Kwang-Ok Lee; Mi Young Cha; Mi-Ra Kim; Ji Yeon Song; Jaeho Lee; Young Hoon Kim; Young-Mi Lee; Kwee Hyun Suh; Jeewoong Son

Introduction: Activating mutations of EGFR are well known as oncogenic driver mutations in lung adenocarcinoma. Currently, EGFR TKIs including Gefitinib and Erlotinib are used as the first line therapy in NSCLC patients harboring EGFR activating mutations. However, drug resistance caused by T790M mutation limits the efficacy of these 1st generation EGFR TKIs. Currently, some of the next generation EGFR TKIs are under investigation for the treatment of lung cancer patients having T790M mutation. In our current presentation, to obtain HM61713, an EGFR mutant selective inhibitor, as a clinical candidate and the evaluation of HM61713 for mutant EGFR cancer model will be introduced. Method: Novel analogues were designed and synthesized to find active compounds for the T790M mutation as well as EGFR activating mutations with good selectivity over wild- type EGFR. Finally, HM61713 was selected as a clinical candidate through multi-optimization processes including both in vitro and in vivo pharmacologcal studies. Results: HM61713 was designed as an irreversible kinase inhibitor having a Michael acceptor, which covalently binds to a cysteine residue near the kinase domain of mutant the EGFR. In a cell wash out test, HM61713 inhibited phospho-EGFR for a long duration with a half-life of over 24 hours. From in vitro study, HM61713 showed potent activities for H1975 (L858-T790M) and HCC827 (exon 19 del.) with GI50 values of 9.2 nM and 10 nM, respectively. Instead, it showed low potency for H358 (wild type EGFR NSCLC) with GI50 of 2,225 nM. In xenograft studies using H1975 and HCC827, HM61713 resulted in good efficacy without showing any side effects. Conclusion: HM61713 showed excellent in vitro and in vivo activities for H1975 harboring L858R-T790M mutation as well as HCC827 having exon 19 deletion mutation with selectivity over wild-type EGFR. Currently, HM61713 is undergoing phase I study (NCT01588145) for NSCLC patients after the failure of 1st generation EGFR TKIs in Korea. Citation Format: Kwang-Ok Lee, Mi Young Cha, Mira Kim, Ji Yeon Song, Jae-Ho Lee, Young Hoon Kim, Young-Mi Lee, Kwee Hyun Suh, Jeewoong Son. Discovery of HM61713 as an orally available and mutant EGFR selective inhibitor. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-100. doi:10.1158/1538-7445.AM2014-LB-100


Angewandte Chemie | 2014

Phenotypic Screening to Identify Small-Molecule Enhancers for Glucose Uptake: Target Identification and Rational Optimization of Their Efficacy†

Minseob Koh; Jongmin Park; Ja Young Koo; Donghyun Lim; Mi Young Cha; Ala Jo; Jang Hyun Choi; Seung Bum Park

Small-molecule glucose uptake enhancers targeted to myotubes and adipocytes were developed through a phenotypic screening linked with target identification and rational optimization. The target protein of glucose-uptake enhancers was identified as a nuclear receptor PPARγ (peroxisome proliferator-activated receptor gamma). Subsequent optimization of initial hits generated lead compounds with high potency for PPARγ transactivation and cellular glucose uptake. Finally, we confirmed that the chirality of optimized ligands differentiates their PPARγ transcriptional activity, binding affinity, and inhibitory activity toward Cdk5 (cyclin-dependent kinase 5)-mediated phosphorylation of PPARγ at Ser273. Using phenotype-based lead discovery along with early-stage target identification, this study has identified a new small-molecule enhancer of glucose uptake that targets PPARγ.


Bioconjugate Chemistry | 2011

Pharmacophore-based strategy for the development of general and specific scFv biosensors for abused antibiotics.

Mi Young Cha; Hyang Yeon Lee; Yeonjin Ko; Hyunbo Shim; Seung Bum Park

We developed fluorescent biosensor systems that are either general or selective to fluoroquinolone antibiotics by using a single-chain variable-fragment (scFv) as a recognition element. The selectivity of these biosensors to fluoroquinolone antibiotics was rationally tuned through the structural modification on the pharmacophore of fluoroquinolone antibiotics and the subsequent selection of scFv receptor modules against these antibiotics-based antigens using phage display. The resulting A2 and F9 scFvs bound to their representative antigen with a moderate affinity (K(D) in micromolar range as determined by surface plasmon resonance). A2 is a specific binder for enrofloxacin and did not cross-react with other fluoroquinolone antibiotics including structurally similar ciprofloxacin, while F9 is a general fluoroquinolone binder that likely bound to the antigen at the common pyridone-carboxylic acid pharmacophore. These scFv-based receptors were successfully applied to the development of one-step fluorescent biosensor which can detect fluoroquinolone antibiotics at concentrations below the level suggested in animal drug application guidelines. The strategy described in this report can be applied to developing convenient field biosensors that can qualitatively detect overused/misused antibiotics in the livestock drinking water.


Cancer Research | 2011

Abstract LB-389: Antitumor activity of HM781–36B, a highly effective pan-HER inhibitor in erlotinib-resistant NSCLC and other EGFR-dependent cancer models

Mi Young Cha; Kwang-Ok Lee; Young Hun Kim; Kwee Hyun Suh; Seung Bum Park; Maeng Sup Kim; Gwan Sun Lee

The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases has been implicated in a variety of cancers. In particular, activating mutations such as the L858R point mutation in exon 21 and the small in-frame deletions in exon 19 of the EGFR tyrosine kinase domain are correlated with sensitivity to EGFR tyrosine kinase inhibitors in non-small cell lung cancer (NSCLC) patients. But clinical treatment is limited by the development of drug resistance resulting mainly from an acquired mutation (T790M). In this study, we evaluated the therapeutic potential of a novel, irreversible pan-HER inhibitor, HM781–36B. We demonstrated that HM781–36B is a potent EGFR inhibitor including the EGFR -acquired resistance mutation (IC 50 4.2 nM in EGFR T790M and IC 50 2.2 nM in EGFR L858R/T790M ), as well as EGFR, HER-2 and HER-4 (IC 50 3.2 nM, 5.3 nM and 23.5 nM, respectively), compared to other EGFR tyrosine kinases inhibitors (erlotinib, lapatinib and BIBW-2992), with kinase selectivity confirmed by 77 kinases panel assay except for Blk, Bmx and Btk. Strong growth inhibitory activity by the treatment of HM781–36B was confirmed with the range of IC 50 0.6–6nM in NSCLC (HCC827, H358, H1975, Calu-3, and H1781), breast cancer (SK-Br3, BT474, MDA-175, and MDA-453), and gastric cancer (N87 and SNU-216) cell lines. HM781–36B showed 400–4,000 fold low growth inhibitory effect against normal cell lines (Hs-27 and Balb/c 3T3) relative to cancer cell lines. HM781–36B treatment results in the inhibition of EGFR phosphorylation and the subsequent deactivation of downstream signaling proteins in EGFR DelE746_A750-harboring erlotinib-sensitive HCC827 and EGFR L858R/T790M-harboring erlotinib-resistant NCI-H1975 NSCLC cells. HM781–36B interacts directly with its target enzyme ( EGFR WT , EGFR T790M , HER-2), which was confirmed through prolongation study of phosphorylation inhibitory effect in A431 and SK-Br3 cells and direct fluorescence-based SDS-PAGE analysis by the incubation of Cy3-labeled HM781–36B probe. This irreversible occupation at the kinase domain leads to excellent in vivo efficacy of HM781–36B (1mg/kg – 5mg/kg) toward various xenograft models with EGFR-dependent cancers cells, including erlotinib-sensitive HCC827 NSCLC cells, erlotinib-resistant H1975 NSCLC cells, HER-2 overexpressing Calu-3 NSCLC cells, N87 gastric cancer cells, and SK-Ov3 ovarian cancer cells, and EGFR-overexpressing A431 epidermoid carcinoma cancer cells. On the basis of these preclinical results, HM781–36B was selected as a candidate for clinical development and a phase I study of HM781–36B is currently undergoing in South Korea. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-389. doi:10.1158/1538-7445.AM2011-LB-389


Archive | 2005

Quinazoline derivatives for inhibiting cancer cell growth and method for the preparation thereof

Young Jin Ham; Ji Hyeon Gong; Mi Young Cha; Jong-Woo Kim; Maeng Sup Kim; Eun Young Kim; Ji Yeon Song; Chang In Kim; Se Young Kim; Gwan Sun Lee


Archive | 2011

NOVEL FUSED PYRIMIDINE DERIVATIVES FOR INHIBITION OF TYROSINE KINASE ACTIVITY

Mi Young Cha; Seok Jong Kang; Mi Ra Kim; Ju Yeon Lee; Ji Young Jeon; Myoung Gi Jo; Eun Joo Kwak; Kwang Ok Lee; Tae Hee Ha; Kwee Hyun Suh; Maeng Sup Kim


Archive | 2008

Novel amide derivative for inhibiting the growth of cancer cells

Kwang-Ok Lee; Mi Young Cha; Mi Ra Kim; Young Hee Jung; Chang Gon Lee; Se Young Kim; Keukchan Bang; Bum Woo Park; Bo Im Choi; Yun Jung Chae; Mi Young Ko; Han Kyong Kim; Young-Gil Ahn; Maeng Sup Kim; Gwan Sun Lee

Collaboration


Dive into the Mi Young Cha's collaboration.

Top Co-Authors

Avatar

Maeng Sup Kim

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Gwan Sun Lee

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Kwee Hyun Suh

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Young Jin Ham

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Seung Bum Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji Yeon Song

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Se Young Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kwang-Ok Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Young Gil Ahn

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Young Hee Jung

Korea Institute of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge