Network


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

Hotspot


Dive into the research topics where Hengmiao Cheng is active.

Publication


Featured researches published by Hengmiao Cheng.


Molecular Cancer Therapeutics | 2011

PF-04691502, a potent and selective oral inhibitor of PI3K and mTOR kinases with antitumor activity

Jing Yuan; Pramod P. Mehta; Min-Jean Yin; Shaoxian Sun; Aihua Zou; Jeffrey H. Chen; Kristina Rafidi; Zheng Feng; Jeffrey Nickel; Jon Engebretsen; Jill Hallin; Alessandra Blasina; Eric Zhang; Leslie Nguyen; Minghao Sun; Peter K. Vogt; Aileen McHarg; Hengmiao Cheng; James G. Christensen; Julie L.C. Kan; Shubha Bagrodia

Deregulation of the phosphoinositide 3-kinase (PI3K) signaling pathway such as by PTEN loss or PIK3CA mutation occurs frequently in human cancer and contributes to resistance to antitumor therapies. Inhibition of key signaling proteins in the pathway therefore represents a valuable targeting strategy for diverse cancers. PF-04691502 is an ATP-competitive PI3K/mTOR dual inhibitor, which potently inhibited recombinant class I PI3K and mTOR in biochemical assays and suppressed transformation of avian fibroblasts mediated by wild-type PI3K γ, δ, or mutant PI3Kα. In PIK3CA-mutant and PTEN-deleted cancer cell lines, PF-04691502 reduced phosphorylation of AKT T308 and AKT S473 (IC50 of 7.5–47 nmol/L and 3.8–20 nmol/L, respectively) and inhibited cell proliferation (IC50 of 179–313 nmol/L). PF-04691502 inhibited mTORC1 activity in cells as measured by PI3K-independent nutrient stimulated assay, with an IC50 of 32 nmol/L and inhibited the activation of PI3K and mTOR downstream effectors including AKT, FKHRL1, PRAS40, p70S6K, 4EBP1, and S6RP. Short-term exposure to PF-04691502 predominantly inhibited PI3K, whereas mTOR inhibition persisted for 24 to 48 hours. PF-04691502 induced cell cycle G1 arrest, concomitant with upregulation of p27 Kip1 and reduction of Rb. Antitumor activity was observed in U87 (PTEN null), SKOV3 (PIK3CA mutation), and gefitinib- and erlotinib-resistant non–small cell lung carcinoma xenografts. In summary, PF-04691502 is a potent dual PI3K/mTOR inhibitor with broad antitumor activity. PF-04691502 has entered phase I clinical trials. Mol Cancer Ther; 10(11); 2189–99. ©2011 AACR.


MedChemComm | 2010

Discovery of the highly potent PI3K/ mTOR dual inhibitor PF-04691502 through structure based drug design

Hengmiao Cheng; Shubha Bagrodia; Simon Bailey; Martin Paul Edwards; Jacqui Elizabeth Hoffman; Qiyue Hu; Robert Steven Kania; Daniel R. Knighton; Matthew A. Marx; Sacha Ninkovic; Shaoxian Sun; Eric Zhang

The phosphatidylinositol 3-kinase (PI3K) signaling pathway plays crucial roles in cell growth, proliferation and survival. Genomic aberrations in the PI3K pathway, such as mutational activation of PI3Kα or loss of function of tumor suppressor PTEN, have been closely linked to the development and progression of a wide range of cancers. Hence, inhibition of the key targets in the pathway, e.g. PI3K, AKT, mTOR, offers great potential for the treatment of cancer. Lead optimization through integration of structure based drug design (SBDD) and physical properties-based optimization (PPBO) led to the discovery of 2-amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxypyridin-3-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one (PF-04691502, 1) that demonstrated potent in vitro inhibitory activity against both PI3K and mTOR, excellent kinase selectivity, good ADMET, and robust in vivo efficacy in a mouse xenograft tumor growth model. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.


ACS Medicinal Chemistry Letters | 2013

Discovery of the Highly Potent PI3K/mTOR Dual Inhibitor PF-04979064 through Structure-Based Drug Design.

Hengmiao Cheng; Chunze Li; Simon Bailey; Sangita M. Baxi; Lance Goulet; Lisa Guo; Jacqui Elizabeth Hoffman; Ying Jiang; Theodore Otto Johnson; Ted W. Johnson; Daniel R. Knighton; John Li; Kevin Liu; Zhengyu Liu; Matthew A. Marx; Marlena Walls; Peter A. Wells; Min-Jean Yin; JinJiang Zhu; Michael Zientek

PI3K, AKT, and mTOR are key kinases from PI3K signaling pathway being extensively pursued to treat a variety of cancers in oncology. To search for a structurally differentiated back-up candidate to PF-04691502, which is currently in phase I/II clinical trials for treating solid tumors, a lead optimization effort was carried out with a tricyclic imidazo[1,5]naphthyridine series. Integration of structure-based drug design and physical properties-based optimization yielded a potent and selective PI3K/mTOR dual kinase inhibitor PF-04979064. This manuscript discusses the lead optimization for the tricyclic series, which both improved the in vitro potency and addressed a number of ADMET issues including high metabolic clearance mediated by both P450 and aldehyde oxidase (AO), poor permeability, and poor solubility. An empirical scaling tool was developed to predict human clearance from in vitro human liver S9 assay data for tricyclic derivatives that were AO substrates.


Bioorganic & Medicinal Chemistry Letters | 2010

The development and SAR of pyrrolidine carboxamide 11β-HSD1 inhibitors

Hengmiao Cheng; Jacqui Elizabeth Hoffman; Phuong T. Le; Sajiv K. Nair; Stephan James Cripps; Jean Matthews; Christopher Ronald Smith; Michele Yang; Stan Kupchinsky; Klaus Ruprecht Dress; Martin Paul Edwards; Bridget Mccarthy Cole; Evan Walters; Christine Loh; Jacques Ermolieff; Andrea Fanjul; Ganesh B. Bhat; Jocelyn Herrera; Tom Pauly; Natilie Hosea; Genevieve Paderes; Paul A. Rejto

The design and development of a series of highly selective pyrrolidine carboxamide 11beta-HSD1 inhibitors are described. These compounds including PF-877423 demonstrated potent in vitro activity against both human and mouse 11beta-HSD1 enzymes. In an in vivo assay, PF-877423 inhibited the conversion of cortisone to cortisol. Structure guided optimization effort yielded potent and stable 11beta-HSD1 selective inhibitor 42.


Bioorganic & Medicinal Chemistry Letters | 2016

Recent progress on third generation covalent EGFR inhibitors

Hengmiao Cheng; Sajiv K. Nair; Brion W. Murray

First generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (gefitinib and erlotinib) demonstrate excellent clinical efficacy for NSCLC patients carrying EGFR oncogenic mutations (L858R, del exon 19 deletions between amino acids 746 and 750). Invariable, drug resistance occurs with around 60% of it driven by the EGFR-T790M gatekeeper mutation. To counter the T790M-dependent resistance, third generation covalent EGFR inhibitors have been developed with high potency toward T790M containing mutants and selectivity over WT EGFR. This review provides an overview of the third generation drugs currently in clinical trials and also encompasses novel methodologies developed to discover third generation covalent EGFR drugs.


Bioorganic & Medicinal Chemistry Letters | 2010

4-methylpteridinones as orally active and selective PI3K/mTOR dual inhibitors.

Kevin K.-C. Liu; Shubha Bagrodia; Simon Bailey; Hengmiao Cheng; Hui Chen; Lisa Gao; Samantha Greasley; Jacqui Elizabeth Hoffman; Qiyue Hu; Ted O. Johnson; Dan Knighton; Zhengyu Liu; Matthew A. Marx; Mitchell David Nambu; Sacha Ninkovic; Bernadette Pascual; Kristina Rafidi; Caroline Rodgers; Graham L. Smith; Shaoxian Sun; Haitao Wang; Anle Yang; Jing Yuan; Aihua Zou

Pteridinones were designed based on a non-selective kinase template. Because of the uniqueness of the PI3K and mTOR binding pockets, a methyl group was introduced to C-4 position of the peteridinone core to give compounds with excellent selectivity for PI3K and mTOR. This series of compounds were further optimized to improve their potency against PI3Kα and mTOR. Finally, orally active compounds with improved solubility and robust in vivo efficacy in tumor growth inhibition were identified as well.


Bioorganic & Medicinal Chemistry Letters | 2011

Quinazolines with intra-molecular hydrogen bonding scaffold (iMHBS) as PI3K/mTOR dual inhibitors.

Kevin K.-C. Liu; Xiaojun Huang; Shubha Bagrodia; Jeffrey H. Chen; Samantha Greasley; Hengmiao Cheng; Shaoxian Sun; Dan Knighton; Caroline Rodgers; Kristina Rafidi; Aihua Zou; Jiezhan Xiao; Shengyong Yan

Intra-molecular hydrogen bonding was introduced to the quinazoline motif to form a pseudo ring (intra-molecular H-bond scaffold, iMHBS) to mimic our previous published core structures, pyrido[2.3-D]pyrimidin-7-one and pteridinone, as PI3K/mTOR dual inhibitors. This design results in potent PI3K/mTOR dual inhibitors and the purposed intra-molecular hydrogen bonding structure is well supported by co-crystal structure in PI3Kγ enzyme. In addition, a novel synthetic route was developed for these analogs.


Journal of Medicinal Chemistry | 2017

Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR.

Simon Paul Planken; Douglas Carl Behenna; Sajiv Krishnan Nair; Theodore Otto Johnson; Asako Nagata; Chau Almaden; Simon Bailey; T. Eric Ballard; Louise Bernier; Hengmiao Cheng; Sujin Cho-Schultz; Deepak Dalvie; Judith Gail Deal; Dac M. Dinh; Martin Paul Edwards; Rose Ann Ferre; Ketan S. Gajiwala; Michelle Hemkens; Robert Steven Kania; John Charles Kath; Jean Matthews; Brion W. Murray; Sherry Niessen; Suvi T. M. Orr; Mason Alan Pairish; Neal W. Sach; Hong Shen; Manli Shi; James Solowiej; Khanh Tran

Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approximately 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed.1 Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clinical candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compound 21 is currently being evaluated in phase-I clinical trials of mutant EGFR driven NSCLC.


Bioorganic & Medicinal Chemistry Letters | 2013

Structure-based design, SAR analysis and antitumor activity of PI3K/mTOR dual inhibitors from 4-methylpyridopyrimidinone series.

Hengmiao Cheng; Jacqui Elizabeth Hoffman; Phuong T. Le; Mason Alan Pairish; Robert Steven Kania; William Farrell; Shubha Bagrodia; Jing Yuan; Shaoxian Sun; Eric Zhang; Cathy Xiang; Deepak Dalvie; Sadayappan V. Rahavendran

PI3K, AKT and mTOR, key kinases from a frequently dysregulated PI3K signaling pathway, have been extensively pursued to treat a variety of cancers in oncology. Clinical trials of PF-04691502, a highly potent and selective ATP competitive kinase inhibitor of class 1 PI3Ks and mTOR, from 4-methylpyridopyrimidinone series, led to the discovery of a metabolite with a terminal carboxylic acid, PF-06465603. This paper discusses structure-based drug design, SAR and antitumor activity of the MPP derivatives with a terminal alcohol, a carboxylic acid or a carboxyl amide.


Current Cancer Drug Targets | 2013

Targeting the mTOR Pathway in Tumor Malignancy

Hengmiao Cheng; Marlena Walls; Sangita M. Baxi; Min-Jean Yin

The mammalian target of rapamycin (mTOR) plays a critical role in the regulation of cell growth, proliferation,and metabolism by integrating growth factor stimulation and energy/nutrient input through a complex signaling network.The mTOR kinase is a part of two structurally and functionally distinct multiple protein complexes, mTORC1 and mTORC2. The mammalian target of rapamycin complex 1 (mTORC1) is rapamycin-sensitive and mediates temporal control of cell growth by regulating several cellular processes, such as translation, transcription, and nutrient transport while the mammalian target of rapamycin complex 2 (mTORC2) is in sensitive to rapamycin and is involved in spatial control of cell growth via cytoskeleton regulation. Here we discuss the mechanism of mTOR regulation in tumor malignancy through a variety of signaling pathways and the potential of mTOR inhibitors for the treatment of cancer.

Collaboration


Dive into the Hengmiao Cheng's collaboration.

Researchain Logo
Decentralizing Knowledge