Network


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

Hotspot


Dive into the research topics where Anne Y. Saiki is active.

Publication


Featured researches published by Anne Y. Saiki.


Journal of Medicinal Chemistry | 2012

Structure-based design of novel inhibitors of the MDM2-p53 interaction.

Yosup Rew; Daqing Sun; Felix Gonzalez-Lopez de Turiso; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jeffrey Deignan; Brian M. Fox; Darin Gustin; Xin Huang; Min Jiang; Xianyun Jiao; Lixia Jin; Frank Kayser; David J. Kopecky; Yihong Li; Mei-Chu Lo; Alexander M. Long; Klaus Michelsen; Jonathan D. Oliner; Tao Osgood; Mark L. Ragains; Anne Y. Saiki; Steve Schneider; Maria M. Toteva; Peter Yakowec; Xuelei Yan; Qiuping Ye

Structure-based rational design led to the discovery of novel inhibitors of the MDM2-p53 protein-protein interaction. The affinity of these compounds for MDM2 was improved through conformational control of both the piperidinone ring and the appended N-alkyl substituent. Optimization afforded 29 (AM-8553), a potent and selective MDM2 inhibitor with excellent pharmacokinetic properties and in vivo efficacy.


Journal of Medicinal Chemistry | 2014

Discovery of AMG 232, a Potent, Selective, and Orally Bioavailable MDM2–p53 Inhibitor in Clinical Development

Daqing Sun; Zhihong Li; Yosup Rew; Michael W. Gribble; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; Xiaoqi Chen; David Chow; Jeffrey Deignan; Jason Duquette; John Eksterowicz; Benjamin Fisher; Brian M. Fox; Jiasheng Fu; Ana Z. Gonzalez; Felix Gonzalez-Lopez de Turiso; Jonathan B. Houze; Xin Huang; Min Jiang; Lixia Jin; Frank Kayser; Jiwen Liu; Mei-Chu Lo; Alexander M. Long; Brian Lucas; Lawrence R. McGee; Joel McIntosh; Jeff Mihalic

We recently reported the discovery of AM-8553 (1), a potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Continued research investigation of the N-alkyl substituent of this series, focused in particular on a previously underutilized interaction in a shallow cleft on the MDM2 surface, led to the discovery of a one-carbon tethered sulfone which gave rise to substantial improvements in biochemical and cellular potency. Further investigation produced AMG 232 (2), which is currently being evaluated in human clinical trials for the treatment of cancer. Compound 2 is an extremely potent MDM2 inhibitor (SPR KD = 0.045 nM, SJSA-1 EdU IC50 = 9.1 nM), with remarkable pharmacokinetic properties and in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft model (ED50 = 9.1 mg/kg).


Journal of Medicinal Chemistry | 2013

Rational Design and Binding Mode Duality of MDM2–p53 Inhibitors

Felix Gonzalez-Lopez de Turiso; Daqing Sun; Yosup Rew; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Tiffany L. Correll; Xin Huang; Lisa Julian; Frank Kayser; Mei-Chu Lo; Alexander M. Long; Dustin L. McMinn; Jonathan D. Oliner; Tao Osgood; Jay P. Powers; Anne Y. Saiki; Steve Schneider; Paul Shaffer; Shou-Hua Xiao; Peter Yakowec; Xuelei Yan; Qiuping Ye; Dongyin Yu; Xiaoning Zhao; Jing Zhou; Julio C. Medina; Steven H. Olson

Structural analysis of both the MDM2-p53 protein-protein interaction and several small molecules bound to MDM2 led to the design and synthesis of tetrasubstituted morpholinone 10, an MDM2 inhibitor with a biochemical IC50 of 1.0 μM. The cocrystal structure of 10 with MDM2 inspired two independent optimization strategies and resulted in the discovery of morpholinones 16 and 27 possessing distinct binding modes. Both analogues were potent MDM2 inhibitors in biochemical and cellular assays, and morpholinone 27 (IC50 = 0.10 μM) also displayed suitable PK profile for in vivo animal experiments. A pharmacodynamic (PD) experiment in mice implanted with human SJSA-1 tumors showed p21(WAF1) mRNA induction (2.7-fold over vehicle) upon oral dosing of 27 at 300 mg/kg.


Journal of Medicinal Chemistry | 2014

Selective and potent morpholinone inhibitors of the MDM2-p53 protein-protein interaction.

Ana Z. Gonzalez; John Eksterowicz; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jason Duquette; Brian M. Fox; Jiasheng Fu; Xin Huang; Jonathan B. Houze; Lixia Jin; Yihong Li; Zhihong Li; Yun Ling; Mei-Chu Lo; Alexander M. Long; Lawrence R. McGee; Joel McIntosh; Dustin L. McMinn; Jonathan D. Oliner; Tao Osgood; Yosup Rew; Anne Y. Saiki; Paul Shaffer; Sarah Wortman; Peter Yakowec; Xuelei Yan; Qiuping Ye

We previously reported the discovery of AMG 232, a highly potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Our continued search for potent and diverse analogues led to the discovery of novel morpholinone MDM2 inhibitors. This change to a morpholinone core has a significant impact on both potency and metabolic stability compared to the piperidinone series. Within this morpholinone series, AM-8735 emerged as an inhibitor with remarkable biochemical potency (HTRF IC50 = 0.4 nM) and cellular potency (SJSA-1 EdU IC50 = 25 nM), as well as pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 41 mg/kg. Lead optimization toward the discovery of this inhibitor as well as key differences between the morpholinone and the piperidinone series will be described herein.


Journal of Medicinal Chemistry | 2014

Novel Inhibitors of the MDM2-p53 Interaction Featuring Hydrogen Bond Acceptors as Carboxylic Acid Isosteres.

Ana Z. Gonzalez; Zhihong Li; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jason Duquette; John Eksterowicz; Brian M. Fox; Jiasheng Fu; Xin Huang; Jonathan B. Houze; Lixia Jin; Yihong Li; Yun Ling; Mei-Chu Lo; Alexander M. Long; Lawrence R. McGee; Joel McIntosh; Jonathan D. Oliner; Tao Osgood; Yosup Rew; Anne Y. Saiki; Paul Shaffer; Sarah Wortman; Peter Yakowec; Xuelei Yan; Qiuping Ye; Dongyin Yu; Xiaoning Zhao

We previously reported the discovery of potent and selective morpholinone and piperidinone inhibitors of the MDM2-p53 interaction. These inhibitors have in common a carboxylic acid moiety that engages in an electrostatic interaction with MDM2-His96. Our continued search for potent and diverse inhibitors led to the discovery of novel replacements for these acids uncovering new interactions with the MDM2 protein. In particular, using pyridine or thiazole as isosteres of the carboxylic acid moiety resulted in very potent analogues. From these, AM-6761 (4) emerged as a potent inhibitor with remarkable biochemical (HTRF IC50 = 0.1 nM) and cellular potency (SJSA-1 EdU IC50 = 16 nM), as well as favorable pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 11 mg/kg. Optimization efforts toward the discovery of these inhibitors as well as the new interactions observed with the MDM2 protein are described herein.


Bioorganic & Medicinal Chemistry Letters | 2011

Improvement of the synthesis and pharmacokinetic properties of chromenotriazolopyrimidine MDM2-p53 protein-protein inhibitors

Hilary P. Beck; Michael DeGraffenreid; Brian M. Fox; John G. Allen; Yosup Rew; Stephen Schneider; Anne Y. Saiki; Dongyin Yu; Jonathan D. Oliner; Kevin Salyers; Qiuping Ye; Steven H. Olson

Human murine double minute 2 (MDM2) is a negative regulator of p53, which plays an important role in cell cycle and apoptosis. We report several optimizations to the synthesis of the chromenotriazolopyrimidine series of MDM2-p53 protein-protein interaction inhibitors. Additionally, the in vitro and in vivo stability, pharmacokinetic properties and solubility were improved through N-substitution.


Antimicrobial Agents and Chemotherapy | 2003

Novel Antibacterial Class

Peter J. Dandliker; Steve D. Pratt; Angela M. Nilius; Candace Black-Schaefer; Xiaoan Ruan; Danli L. Towne; Richard F. Clark; Erika E. Englund; Rolf Wagner; Moshe Weitzberg; Linda E. Chovan; Robert K. Hickman; Melissa M. Daly; Stephan J. Kakavas; Ping Zhong; Zhensheng Cao; Caroline A. David; Xiaoling Xuei; Claude G. Lerner; Niru B. Soni; Mai Bui; Linus L. Shen; Yingna Cai; Philip J. Merta; Anne Y. Saiki; Bruce A. Beutel

ABSTRACT We report the discovery and characterization of a novel ribosome inhibitor (NRI) class that exhibits selective and broad-spectrum antibacterial activity. Compounds in this class inhibit growth of many gram-positive and gram-negative bacteria, including the common respiratory pathogens Streptococcus pneumoniae, Haemophilus influenzae, Staphylococcus aureus, and Moraxella catarrhalis, and are nontoxic to human cell lines. The first NRI was discovered in a high-throughput screen designed to identify inhibitors of cell-free translation in extracts from S. pneumoniae. The chemical structure of the NRI class is related to antibacterial quinolones, but, interestingly, the differences in structure are sufficient to completely alter the biochemical and intracellular mechanisms of action. Expression array studies and analysis of NRI-resistant mutants confirm this difference in intracellular mechanism and provide evidence that the NRIs inhibit bacterial protein synthesis by inhibiting ribosomes. Furthermore, compounds in the NRI series appear to inhibit bacterial ribosomes by a new mechanism, because NRI-resistant strains are not cross-resistant to other ribosome inhibitors, such as macrolides, chloramphenicol, tetracycline, aminoglycosides, or oxazolidinones. The NRIs are a promising new antibacterial class with activity against all major drug-resistant respiratory pathogens.


Molecular Cancer Therapeutics | 2015

The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents

Jude Canon; Tao Osgood; Steven H. Olson; Anne Y. Saiki; Rebecca Robertson; Dongyin Yu; John Eksterowicz; Qiuping Ye; Lixia Jin; Ada Chen; Jing Zhou; David Cordover; Stephen Kaufman; Richard Kendall; Jonathan D. Oliner; Angela Coxon; Robert Radinsky

p53 is a critical tumor suppressor and is the most frequently inactivated gene in human cancer. Inhibition of the interaction of p53 with its negative regulator MDM2 represents a promising clinical strategy to treat p53 wild-type tumors. AMG 232 is a potential best-in-class inhibitor of the MDM2–p53 interaction and is currently in clinical trials. We characterized the activity of AMG 232 and its effect on p53 signaling in several preclinical tumor models. AMG 232 binds the MDM2 protein with picomolar affinity and robustly induces p53 activity, leading to cell-cycle arrest and inhibition of tumor cell proliferation. AMG 232 treatment inhibited the in vivo growth of several tumor xenografts and led to complete and durable regression of MDM2-amplified SJSA-1 tumors via growth arrest and induction of apoptosis. Therapeutic combination studies of AMG 232 with chemotherapies that induce DNA damage and p53 activity resulted in significantly superior antitumor efficacy and regression, and markedly increased activation of p53 signaling in tumors. These preclinical data support the further evaluation of AMG 232 in clinical trials as both a monotherapy and in combination with standard-of-care cytotoxics. Mol Cancer Ther; 14(3); 649–58. ©2015 AACR.


Chemical Biology & Drug Design | 2007

From bacterial genomes to novel antibacterial agents: discovery, characterization, and antibacterial activity of compounds that bind to HI0065 (YjeE) from Haemophilus influenzae.

Claude G. Lerner; Philip J. Hajduk; Rolf Wagner; Frank L. Wagenaar; Charlotte Woodall; Yu-Gui Gu; Xenia B. Searle; Alan S. Florjancic; Tianyuan Zhang; Richard F. Clark; Curt S. Cooper; Jamey Mack; Liping Yu; Mengli Cai; Steven F. Betz; Linda E. Chovan; J. Owen McCall; Candace Black-Schaefer; Stephan J. Kakavas; Mark E. Schurdak; Kenneth M. Comess; Karl A. Walter; Rohinton Edalji; Sarah A. Dorwin; Richard Smith; Eric J. Hebert; John E. Harlan; Randy E. Metzger; Philip J. Merta; John L. Baranowski

As part of a fully integrated and comprehensive strategy to discover novel antibacterial agents, NMR‐ and mass spectrometry‐based affinity selection screens were performed to identify compounds that bind to protein targets uniquely found in bacteria and encoded by genes essential for microbial viability. A biphenyl acid lead series emerged from an NMR‐based screen with the Haemophilus influenzae protein HI0065, a member of a family of probable ATP‐binding proteins found exclusively in eubacteria. The structure–activity relationships developed around the NMR‐derived biphenyl acid lead were consistent with on‐target antibacterial activity as the Staphylococcus aureus antibacterial activity of the series correlated extremely well with binding affinity to HI0065, while the correlation of binding affinity with B‐cell cytotoxicity was relatively poor. Although further studies are needed to conclusively establish the mode of action of the biphenyl series, these compounds represent novel leads that can serve as the basis for the development of novel antibacterial agents that appear to work via an unprecedented mechanism of action. Overall, these results support the genomics‐driven hypothesis that targeting bacterial essential gene products that are not present in eukaryotic cells can identify novel antibacterial agents.


Journal of Biomolecular Screening | 2004

A Strategy for Discovery of Novel Broad-Spectrum Antibacterials Using a High-Throughput Streptococcus pneumoniae Transcription/Translation Screen

Steven D. Pratt; Caroline A. David; Candace Black-Schaefer; Peter J. Dandliker; Xiaoling Xuei; Usha Warrior; David J. Burns; Ping Zhong; Zhensheng Cao; Anne Y. Saiki; Claude G. Lerner; Linda E. Chovan; Niru B. Soni; Angela M. Nilius; Frank L. Wagenaar; Philip J. Merta; Linda Traphagen; Bruce A. Beutel

The authors report the development of a high-throughput screen for inhibitors of Streptococcus pneumoniae transcription and translation (TT) using a luciferase reporter, and the secondary assays used to determine the biochemical spectrum of activity and bacterial specificity. More than 220,000 compounds were screened in mixtures of 10 compounds per well, with 10,000 picks selected for further study. False-positive hits from inhibition of luciferase activity were an extremely common artifact. After filtering luciferase inhibitors and several known classes of antibiotics, approximately 50 hits remained. These compounds were examined for their ability to inhibit Escherichia coli TT, uncoupled S. pneumoniae translation or transcription, rabbit reticulocyte translation, and in vitro toxicity in human and bacterial cells. One of these compounds had the desired profile of broad-spectrum biochemical activity in bacteria and selectivity versus mammalian biochemical and whole-cell assays. (Journal of Biomolecular Screening 2004:3-11)

Collaboration


Dive into the Anne Y. Saiki's collaboration.

Researchain Logo
Decentralizing Knowledge