Network


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

Hotspot


Dive into the research topics where Ching-Ping Chen is active.

Publication


Featured researches published by Ching-Ping Chen.


Journal of Medicinal Chemistry | 2010

Design and Synthesis of Tetrahydropyridothieno[2,3-d]pyrimidine Scaffold Based Epidermal Growth Factor Receptor (EGFR) Kinase Inhibitors: The Role of Side Chain Chirality and Michael Acceptor Group for Maximal Potency

Chia-Hsien Wu; Mohane Selvaraj Coumar; Chang-Ying Chu; Wen-Hsing Lin; Yi-Rong Chen; Chiung-Tong Chen; Hui-Yi Shiao; Shaik Rafi; Sing-Yi Wang; Hui Hsu; Chun-Hwa Chen; Chun-Yu Chang; Teng-Yuan Chang; Tzu-Wen Lien; Ming-Yu Fang; Kai-Chia Yeh; Ching-Ping Chen; Teng-Kuang Yeh; Su-Huei Hsieh; John T.-A. Hsu; Chun-Chen Liao; Yu-Sheng Chao; Hsing-Pang Hsieh

HTS hit 7 was modified through hybrid design strategy to introduce a chiral side chain followed by introduction of Michael acceptor group to obtain potent EGFR kinase inhibitors 11 and 19. Both 11 and 19 showed over 3 orders of magnitude enhanced HCC827 antiproliferative activity compared to HTS hit 7 and also inhibited gefitinib-resistant double mutant (DM, T790M/L858R) EGFR kinase at nanomolar concentration. Moreover, treatment with 19 shrinked tumor in nude mice xenograft model.


Journal of Medicinal Chemistry | 2010

Synthesis and biological activities of 2-amino-1-arylidenamino imidazoles as orally active anticancer agents.

Wen-Tai Li; Der-Ren Hwang; Jen-Shin Song; Ching-Ping Chen; Jiunn-Jye Chuu; Chih-Bo Hu; Heng-Liang Lin; Chen-Lung Huang; Huang Cy; Huan-Yi Tseng; Chu-Chung Lin; Tung-Wei Chen; Chi-Hung Lin; Hsin-Sheng Wang; Chien-Chang Shen; Chung-Ming Chang; Yu-Sheng Chao; Chiung-Tong Chen

2-Amino-1-arylidenaminoimidazoles, a novel class of orally (po) active microtubule-destabilizing anticancer agents, were synthesized. The compounds were designed from a hit compound identified in a drug discovery platform by using cancer cell-based high throughput screening assay. Selective synthesized compounds exerted cell cytotoxicity against human cancer cells. The underlying mechanisms for the anticancer activity were demonstrated as interacting with the tubulins and inhibiting microtubule assembly, leading to proliferation inhibition and apoptosis induction in the human tumor cells. Furthermore, two compounds showed in vivo anticancer activities in both po and intravenously (iv) administered routes and prolonged the life spans of murine leukemic P388 cells-inoculated mice. These new po active antimitotic anticancer agents are to be further examined in preclinical studies and developed for clinical uses.


Bioorganic & Medicinal Chemistry | 2011

Discovery and evaluation of 3-phenyl-1H-5-pyrazolylamine-based derivatives as potent, selective and efficacious inhibitors of FMS-like tyrosine kinase-3 (FLT3)

Wen-Hsing Lin; Shu-Yi Hsieh; Shih-Chieh Yen; Chiung-Tong Chen; Teng-Kuang Yeh; Tsu Hsu; Cheng-Tai Lu; Ching-Ping Chen; Chun-Wha Chen; Ling-Hui Chou; Yu-Lin Huang; An-Huei Cheng; Yun-I Chang; Ya-Ju Tseng; Kuei-Rong Yen; Yu-Sheng Chao; John T.-A. Hsu; Weir-Torn Jiaang

Preclinical investigations and early clinical trial studies suggest that FLT3 inhibitors offer a viable therapy for acute myeloid leukemia. However, early clinical data for direct FLT3 inhibitors provided only modest results because of the failure to fully inhibit FLT3. We have designed and synthesized a novel class of 3-phenyl-1H-5-pyrazolylamine-derived compounds as FLT3 inhibitors which exhibit potent FLT3 inhibition and high selectivity toward different receptor tyrosine kinases. The structure-activity relationships led to the discovery of two series of FLT3 inhibitors, and some potent compounds within these two series exhibited comparable potency to FLT3 inhibitors sorafenib (3) and ABT-869 (4) in both wt-FLT3 enzyme inhibition and FLT3-ITD inhibition on cell growth (MOLM-13 and MV4;11 cells). In particular, the selected compound 12a exhibited the ability to regress tumors in mouse xenograft models using MOLM-13 and MV4;11 cells.


Pharmacological Research | 2010

Antitumor activities and pharmacokinetics of silatecans DB-67 and DB-91

Teng-Kuang Yeh; Chien-Ming Li; Ching-Ping Chen; Jiuun-Jye Chuu; Chen-Lung Huang; Hsin-Sheng Wang; Chien-Chang Shen; Tien-Yi Lee; Chi-Yen Chang; Chung-Ming Chang; Yu-Sheng Chao; Chin-Tarng Lin; Jang Yang Chang; Chiung-Tong Chen

DB-67 and its lactone homolog DB-91 are derivatives of topoisomerase I inhibitor camptothecin (CPT) with silyl moiety, which may exhibit a slower inactivation process by changed kinetics of protein binding and/or hydrolysis of its lactone ring and result in increased antitumor activity and decreased toxicity. Pharmacokinetic properties and antitumor activities of the two silatecans were studied and compared. The lactone ring of DB-91 is more stable than those of all the other CPT derivatives in mouse plasma. Both silatecans were metabolized faster than CPT in mouse and human liver microsomes. Pharmacokinetic study revealed a plasma elimination half-life (t(1/2)) of 33 and 94min for DB-67 and DB-91, respectively; similar systemic exposure in plasma between DB-67 and DB-91; and similar volume of distribution at the steady state between DB-67 and DB-91, approximately 15-fold smaller than that of CPT. While DB-91 showed limited activities, DB-67 exhibited activities against the growth of in vivo-like histocultured human tumors and s.c. xenografted human tumors in nude mice. In conclusion, DB-67 is more effective, compared to DB-91, against human tumor growth in in vitro, in vivo-like and in vivo systems. Further pre-clinical and clinical investigations of DB-67 are warranted.


PLOS ONE | 2014

Evaluation of the antitumor effects of BPR1J-340, a potent and selective FLT3 inhibitor, alone or in combination with an HDAC inhibitor, vorinostat, in AML cancer.

Wen-Hsing Lin; Teng-Kuang Yeh; Weir-Torn Jiaang; Kuei-Jung Yen; Chun-Hwa Chen; Chin Ting Huang; Shih-Chieh Yen; Shu-Yi Hsieh; Ling-Hui Chou; Ching-Ping Chen; Chun-Hsien Chiu; Li Chun Kao; Yu-Sheng Chao; Chiung-Tong Chen; John T.-A. Hsu

Overexpression or/and activating mutation of FLT3 kinase play a major driving role in the pathogenesis of acute myeloid leukemia (AML). Hence, pharmacologic inhibitors of FLT3 are of therapeutic potential for AML treatment. In this study, BPR1J-340 was identified as a novel potent FLT3 inhibitor by biochemical kinase activity (IC50 approximately 25 nM) and cellular proliferation (GC50 approximately 5 nM) assays. BPR1J-340 inhibited the phosphorylation of FLT3 and STAT5 and triggered apoptosis in FLT3-ITD+ AML cells. The pharmacokinetic parameters of BPR1J-340 in rats were determined. BPR1J-340 also demonstrated pronounced tumor growth inhibition and regression in FLT3-ITD+ AML murine xenograft models. The combination treatment of the HDAC inhibitor vorinostat (SAHA) with BPR1J-340 synergistically induced apoptosis via Mcl-1 down-regulation in MOLM-13 AML cells, indicating that the combination of selective FLT3 kinase inhibitors and HDAC inhibitors could exhibit clinical benefit in AML therapy. Our results suggest that BPR1J-340 may be further developed in the preclinical and clinical studies as therapeutics in AML treatments.


Bioorganic & Medicinal Chemistry | 2013

Discovery of 3-phenyl-1H-5-pyrazolylamine derivatives containing a urea pharmacophore as potent and efficacious inhibitors of FMS-like tyrosine kinase-3 (FLT3)

Wen-Hsing Lin; John T.-A. Hsu; Shu-Yi Hsieh; Chiung-Tong Chen; Jen-Shin Song; Shih-Chieh Yen; Tsu Hsu; Cheng-Tai Lu; Chun-Hwa Chen; Ling-Hui Chou; Yung-Ning Yang; Ching-Hui Chiu; Ching-Ping Chen; Ya-Ju Tseng; Kuei-Jung Yen; Ching-Fang Yeh; Yu-Sheng Chao; Teng-Kuang Yeh; Weir-Torn Jiaang

Preclinical investigations and early clinical trials suggest that FLT3 inhibitors are a viable therapy for acute myeloid leukemia. However, early clinical data have been underwhelming due to incomplete inhibition of FLT3. We have developed 3-phenyl-1H-5-pyrazolylamine as an efficient template for kinase inhibitors. Structure-activity relationships led to the discovery of sulfonamide, carbamate and urea series of FLT3 inhibitors. Previous studies showed that the sulfonamide 4 and carbamate 5 series were potent and selective FLT3 inhibitors with good in vivo efficacy. Herein, we describe the urea series, which we found to be potent inhibitors of FLT3 and VEGFR2. Some inhibited growth of FLT3-mutated MOLM-13 cells more strongly than the FLT3 inhibitors sorafenib (2) and ABT-869 (3). In preliminary in vivo toxicity studies of the four most active compounds, 10f was found to be the least toxic. A further in vivo efficacy study demonstrated that 10f achieved complete tumor regression in a higher proportion of MOLM-13 xenograft mice than 4 and 5 (70% vs 10% and 40%). These results show that compound 10f possesses improved pharmacologic and selectivity profiles and could be more effective than previously disclosed FLT3 inhibitors in the treatment of acute myeloid leukemia.


Investigational New Drugs | 2012

Synthesis and biological evaluation of 2-amino-1-thiazolyl imidazoles as orally active anticancer agents.

Wen-Tai Li; Der-Ren Hwang; Jen-Shin Song; Ching-Ping Chen; Tung-Wei Chen; Chi-Hung Lin; Jiunn-Jye Chuu; Tzu-Wen Lien; Tsu-An Hsu; Chen-Lung Huang; Huan-Yi Tseng; Chu-Chung Lin; Heng-Liang Lin; Chung-Ming Chang; Yu-Sheng Chao; Chiung-Tong Chen

SummaryDesigned from a high throughput screened hit compound, novel 2-amino-1-thiazolyl imidazoles were synthesized and demonstrated cytotoxicity against human cancer cells. 1-(4-Phenylthiazol-2-yl)-4-(thiophen-2-yl)-1H-imidazol-2-amine (compound 2), a 2-amino-1-thiazolyl imidazole, inhibited tubulin polymerization, interacted with the colchicine-binding sites of tubulins, and caused cell cycle arrest at the G2/M phase in human gastric cancer cells. Disruption of the microtubule structure in cancer cells by compound 2 was also observed. Compound 2 concentration-dependently inhibited the proliferation of cancer cells in histocultured human gastric and colorectal tumors. Given orally, compound 2 prolonged the lifespans of leukemia mice intraperitoneally inoculated with the murine P388 leukemic cells. We report 2-amino-1-thiazolyl imidazoles as a novel class of orally active microtubule-destabilizing anticancer agents.


Anti-Cancer Drugs | 2013

BPR0C305, an orally active microtubule-disrupting anticancer agent.

Wen-Tai Li; Teng-Kuang Yeh; Jen-Shin Song; Yung-Ning Yang; Tung-Wei Chen; Chi-Hung Lin; Ching-Ping Chen; Chien-Chang Shen; Chih-Chien Hsieh; Heng-Liang Lin; Yu-Sheng Chao; Chiung-Tong Chen

BPR0C305 is a novel N-substituted indolyl glyoxylamide previously reported with in-vitro cytotoxic activity against a panel of human cancer cells including P-gp-expressing multiple drug-resistant cell sublines. The present study further examined the underlying molecular mechanism of anticancer action and evaluated the in-vivo antitumor activities of BPR0C305. BPR0C305 is a novel synthetic small indole derivative that demonstrates in-vitro activities against human cancer cell growth by inhibiting tubulin polymerization, disrupting cellular microtubule assembly, and causing cell cycle arrest at the G2/M phase. It is also orally active against leukemia and solid tumor growths in mouse models. Findings of these pharmacological and pharmacokinetic studies suggest that BPR0C305 is a promising lead compound for further preclinical developments.


Oncotarget | 2016

Discovery of BPR1K871, a quinazoline based, multi-kinase inhibitor for the treatment of AML and solid tumors: Rational design, synthesis, in vitro and in vivo evaluation

Yung Chang Hsu; Mohane Selvaraj Coumar; Wen-Chieh Wang; Hui-Yi Shiao; Yi-Yu Ke; Wen-Hsing Lin; Ching-Chuan Kuo; Chun-Wei Chang; Fu-Ming Kuo; Pei-Yi Chen; Sing-Yi Wang; An-Siou Li; Chun-Hwa Chen; Po-Chu Kuo; Ching-Ping Chen; Ming-Hsine Wu; Chen-Lung Huang; Kuei-Jung Yen; Yun-I Chang; John T.-A. Hsu; Chiung-Tong Chen; Teng-Kuang Yeh; Jen-Shin Song; Chuan Shih; Hsing-Pang Hsieh

The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents. BPR1K871 showed potent anti-proliferative activities in MOLM-13 and MV4-11 AML cells (EC50 ∼ 5 nM). Moreover, kinase profiling and cell-line profiling revealed BPR1K871 to be a potential multi-kinase inhibitor. Functional studies using western blot and DNA content analysis in MV4-11 and HCT-116 cell lines revealed FLT3 and AURKA/B target modulation inside the cells. In vivo efficacy in AML xenograft models (MOLM-13 and MV4-11), as well as in solid tumor models (COLO205 and Mia-PaCa2), led to the selection of BPR1K871 as a preclinical development candidate for anti-cancer therapy. Further detailed studies could help to investigate the full potential of BPR1K871 as a multi-kinase inhibitor.


Journal of Medicinal Chemistry | 2003

Synthesis and biological evaluation of N-heterocyclic indolyl glyoxylamides as orally active anticancer agents.

Wen-Tai Li; Der-Ren Hwang; Ching-Ping Chen; Chien-Wei Shen; Chen-Long Huang; Tung-Wei Chen; Chi-Hung Lin; Yee-Ling Chang; Ying-Ying Chang; Yue-Kan Lo; Huan-Yi Tseng; Chu-Chung Lin; Jeng-Shin Song; Hua-Chien Chen; Shu-Jen Chen; Se−Hui Wu; Chiung-Tong Chen

Collaboration


Dive into the Ching-Ping Chen's collaboration.

Top Co-Authors

Avatar

Chiung-Tong Chen

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Yu-Sheng Chao

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Teng-Kuang Yeh

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Chen-Lung Huang

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Jen-Shin Song

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Chi-Hung Lin

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

John T.-A. Hsu

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Tung-Wei Chen

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Wen-Hsing Lin

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Wen-Tai Li

National Health Research Institutes

View shared research outputs
Researchain Logo
Decentralizing Knowledge