Network


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

Hotspot


Dive into the research topics where Meizhong Jin is active.

Publication


Featured researches published by Meizhong Jin.


Organic and Biomolecular Chemistry | 2007

A highly effective one-pot synthesis of quinolines from o-nitroarylcarbaldehydes.

An-Hu Li; Eilaf Ahmed; Xin Chen; Matthew Cox; Andrew P. Crew; Hanqing Dong; Meizhong Jin; Lifu Ma; Bijoy Panicker; Kam W. Siu; Arno G. Steinig; Kathryn M. Stolz; Paula A. R. Tavares; Brian Volk; Qinghua Weng; Doug Werner; Mark J. Mulvihill

A highly effective one-pot Friedländer quinoline synthesis using inexpensive reagents has been developed. o-Nitroarylcarbaldehydes were reduced to o-aminoarylcarbaldehydes with iron in the presence of catalytic HCl (aq.) and subsequently condensed in situ with aldehydes or ketones to form mono- or di-substituted quinolines in high yields (66-100%).


Bioorganic & Medicinal Chemistry Letters | 2011

Imidazo[1,5-a]pyrazines: orally efficacious inhibitors of mTORC1 and mTORC2.

Andrew P. Crew; Shripad V. Bhagwat; Hanqing Dong; Mark Bittner; Anna Chan; Xin Chen; Heather Coate; Andrew Cooke; Prafulla C. Gokhale; Ayako Honda; Meizhong Jin; Jennifer Kahler; Christine Mantis; Mark J. Mulvihill; Paula A. Tavares-Greco; Brian Volk; Jing Wang; Douglas S. Werner; Lee D. Arnold; Jonathan A. Pachter; Robert Wild; Neil W. Gibson

The discovery and optimization of a series of imidazo[1,5-a]pyrazine inhibitors of mTOR is described. HTS hits were optimized for potency, selectivity and metabolic stability to provide the orally bioavailable proof of concept compound 4c that demonstrated target inhibition in vivo and concomitant inhibition of tumor growth in an MDA-MB-231 xenograft model.


Oncology Reviews | 2013

Modulation of insulin-like growth factor-1 receptor and its signaling network for the treatment of cancer: current status and future perspectives

Meizhong Jin; Elizabeth Buck; Mark J. Mulvihill

Based on over three decades of pre-clinical data, insulin-like growth factor-1 receptor (IGF-1R) signaling has gained recognition as a promoter of tumorogenesis, driving cell survival and proliferation in multiple human cancers. As a result, IGF-1R has been pursued as a target for cancer treatment. Early pioneering efforts targeting IGF-1R focused on highly selective monoclonal antibodies, with multiple agents advancing to clinical trials. However, despite some initial promising results, recent clinical disclosures have been less encouraging. Moreover, recent studies have revealed that IGF-1R participates in a dynamic and complex signaling network, interacting with additional targets and pathways thereof through various crosstalk and compensatory signaling mechanisms. Such mechanisms of bypass signaling help to shed some light on the decreased effectiveness of selective IGF-1R targeted therapies (e.g. monoclonal antibodies) and suggest that targeting multiple nodes within this signaling network might be necessary to produce a more effective therapeutic response. Additionally, such findings have led to the development of small molecule IGF-1R inhibitors which also co-inhibit additional targets such as insulin receptor and epidermal growth factor receptor. Such findings have helped to guide the design rationale of numerous drug combinations that are currently being evaluated in clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2013

Novel 6-aminofuro[3,2-c]pyridines as potent, orally efficacious inhibitors of cMET and RON kinases

Arno G. Steinig; An-Hu Li; Jing Wang; Xin Chen; Hanqing Dong; Caterina Ferraro; Meizhong Jin; Mridula Kadalbajoo; Andrew Kleinberg; Kathryn M. Stolz; Paula A. Tavares-Greco; Ti Wang; Mark Albertella; Yue Peng; Linda Crew; Jennifer Kahler; Julie Kan; Ryan Schulz; Andy Cooke; Mark Bittner; Roy Turton; Maryland Franklin; Prafulla C. Gokhale; Darla Landfair; Christine Mantis; Jen Workman; Robert Wild; Jonathan A. Pachter; David M. Epstein; Mark J. Mulvihill

A series of novel 6-aminofuro[3,2-c]pyridines as kinase inhibitors is described, most notably, OSI-296 (6). We discuss our exploration of structure-activity relationships and optimization leading to OSI-296 and disclose its pharmacological activity against cMET and RON in cellular assays. OSI-296 is a potent and selective inhibitor of cMET and RON kinases that shows in vivo efficacy in tumor xenografts models upon oral dosing and is well tolerated.


Bioorganic & Medicinal Chemistry Letters | 2011

Potent and selective cyclohexyl-derived imidazopyrazine insulin-like growth factor 1 receptor inhibitors with in vivo efficacy

Meizhong Jin; Andrew Kleinberg; Andy Cooke; Prafulla C. Gokhale; Kenneth Foreman; Hanqing Dong; Kam W. Siu; Mark Bittner; Kristen Michelle Mulvihill; Yan Yao; Darla Landfair; Matthew O’Connor; Gilda Mak; Jonathan A. Pachter; Robert Wild; Maryland Rosenfeld-Franklin; Qun-Sheng Ji; Mark J. Mulvihill

Preclinical and emerging clinical evidence suggests that inhibiting insulin-like growth factor 1 receptor (IGF-1R) signaling may offer a promising therapeutic strategy for the treatment of several types of cancer. This Letter describes the medicinal chemistry effort towards a series of 8-amino-imidazo[1,5-a]pyrazine derived inhibitors of IGF-1R which features a substituted quinoline moiety at the C1 position and a cyclohexyl linking moiety at the C3 position. Lead optimization efforts which included the optimization of structure-activity relationships and drug metabolism and pharmacokinetic properties led to the identification of compound 9m, a potent, selective and orally bioavailable inhibitor of IGF-1R with in vivo efficacy in an IGF-driven mouse xenograft model.


ACS Medicinal Chemistry Letters | 2013

Discovery of novel insulin-like growth factor-1 receptor inhibitors with unique time-dependent binding kinetics.

Meizhong Jin; Brenda A. Petronella; Andy Cooke; Mridula Kadalbajoo; Kam W. Siu; Andrew Kleinberg; Earl May; Prafulla C. Gokhale; Ryan Schulz; Jennifer Kahler; Mark Bittner; Kenneth Foreman; Jonathan A. Pachter; Robert Wild; David M. Epstein; Mark J. Mulvihill

This letter describes a series of small molecule inhibitors of IGF-1R with unique time-dependent binding kinetics and slow off-rates. Structure-activity and structure-kinetic relationships were elucidated and guided further optimizations within the series, culminating in compound 2. With an IGF-1R dissociative half-life (t 1/2) of >100 h, compound 2 demonstrated significant and extended PD effects in conjunction with tumor growth inhibition in xenograft models at a remarkably low and intermittent dose, which correlated with the observed in vitro slow off-rate properties.


Future Medicinal Chemistry | 2012

Small-molecule ATP-competitive dual IGF-1R and insulin receptor inhibitors: structural insights, chemical diversity and molecular evolution

Meizhong Jin; Jing Wang; Elizabeth Buck; Mark J. Mulvihill

IGF-1R has been recognized as a major target in cancer drug discovery due to its strong implications in various stages of tumorigenesis based on accumulated preclinical data. Recent research on compensatory crosstalk between IGF-1R and insulin receptor (IR) signaling pathways suggests that targeting both IGF-1R and IR should result in a more therapeutically beneficial response, than targeting IGF-1R alone (e.g., IGF-1R-specific antibodies). These findings provided biological rationale and opened the door to the discovery of a variety of small-molecule dual IGF-1R and IR inhibitors. In this review we summarize the recent developments in this field, with a focus on binding modes and binding interactions of these inhibitors with IGF-1R and/or IR. Selectivity of these inhibitors has been discussed in this context as well. This is an important area to be discussed since one of the major challenges in kinase inhibitor drug discovery is to build an optimal selectivity profile based on biological rationale.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of potent, selective and orally bioavailable imidazo[1,5-a]pyrazine derived ACK1 inhibitors

Meizhong Jin; Jing Wang; Andrew Kleinberg; Mridula Kadalbajoo; Kam W. Siu; Andrew Cooke; Mark Bittner; Yan Yao; April Thelemann; Qun-Sheng Ji; Shripad V. Bhagwat; Kristen Michelle Mulvihill; Josef A. Rechka; Jonathan A. Pachter; Andrew P. Crew; David M. Epstein; Mark J. Mulvihill

This Letter describes the medicinal chemistry effort towards a series of novel imidazo[1,5-a]pyrazine derived inhibitors of ACK1. Virtual screening led to the discovery of the initial hit, and subsequent exploration of structure-activity relationships and optimization of drug metabolism and pharmacokinetic properties led to the identification of potent, selective and orally bioavailable ACK1 inhibitors.


ACS Medicinal Chemistry Letters | 2010

Discovery of an Orally Efficacious Imidazo[5,1-f][1,2,4]triazine Dual Inhibitor of IGF-1R and IR.

Meizhong Jin; Prafulla C. Gokhale; Andy Cooke; Kenneth Foreman; Elizabeth Buck; Earl May; Lixin Feng; Mark Bittner; Mridula Kadalbajoo; Darla Landfair; Kam W. Siu; Kathryn M. Stolz; Douglas S. Werner; Radoslaw Laufer; An-Hu Li; Hanqing Dong; Arno G. Steinig; Andrew Kleinberg; Yan Yao; Jonathan A. Pachter; Robert Wild; Mark J. Mulvihill

This report describes the investigation of a series of 5,7-disubstituted imidazo[5,1-f][1,2,4]triazine inhibitors of insulin-like growth factor-1 receptor (IGF-1R) and insulin receptor (IR). Structure-activity relationship exploration and optimization leading to the identification, characterization, and pharmacological activity of compound 9b, a potent, selective, well-tolerated, and orally bioavailable dual inhibitor of IGF-1R and IR with in vivo efficacy in tumor xenograft models, is discussed.


Molecular Cancer Therapeutics | 2015

Abstract PR08: ARV-330: An androgen receptor PROTAC degrader for prostate cancer

James D. Winkler; Meizhong Jin; Andy P. Crew; AnnMarie K. Rossi; Ryan R. Willard; Hanqing Dong; Kam W. Siu; Jing Wang; Deborah A. Gordon; Xin Chen; Caterina Ferraro; Craig M. Crews; Kevin Coleman; Taavi K. Neklesa

Patients with prostate cancer who progress on therapy often have enhanced Androgen Receptor (AR) signaling due to several mechanisms: increased androgen production, increased AR expression, and/or specific AR mutations that render current therapies ineffective. A novel approach to block AR signaling is to specifically target AR for degradation. To do this, we have created AR PROTACs (PROtein-TArgeting Chimeras), bi-functional molecules that have an AR binding moiety on one end and an E3 ligase-recruiting element on the other end, which leads to AR ubiquitination and degradation. We have applied this technology to determine whether it could address mechanisms of resistance to current therapy in prostate cancer models. Our lead AR PROTAC, ARV-330, degrades AR in LNCaP and VCaP cells with 50% degradation concentrations (DC50s) 80% after sc injection. Treatment of mice with ARV-330, at doses ranging from 0.3 to 10 mg/kg, resulted in reduction of AR protein levels and prostate involution in normal mice and, in mice implanted with VCaP tumors, reduction in plasma PSA and blockade of tumor growth. In summary, the AR PROTAC ARV-330 removes AR from prostate cancer cells in a potent manner and produces therapeutic effects as a result. This cellular efficacy has translated into biomarker activity and efficacy in animal models, and ARV-330 is now in preclinical development. Thus, targeted degradation of AR may provide a novel mechanism for providing efficacious therapy for patients with prostate cancer for whom current therapies have failed. Citation Format: James D. Winkler, Meizhong Jin, Andy P. Crew, AnnMarie K. Rossi, Ryan R. Willard, Hanqing Dong, Kam Siu, Jing Wang, Deborah A. Gordon, Xin Chen, Caterina Ferraro, Craig M. Crews, Kevin Coleman, Taavi K. Neklesa. ARV-330: An androgen receptor PROTAC degrader for prostate cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr PR08.

Collaboration


Dive into the Meizhong Jin'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