Mason Alan Pairish
Pfizer
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Mason Alan Pairish.
Journal of Medicinal Chemistry | 2011
J. Jean Cui; Michelle Bich Tran-Dube; Hong Shen; Mitchell David Nambu; Pei-Pei Kung; Mason Alan Pairish; Lei Jia; Jerry Meng; Lee Andrew Funk; Iriny Botrous; Michele McTigue; Neil Grodsky; Kevin Ryan; Ellen Padrique; Gordon Alton; Sergei Timofeevski; Shinji Yamazaki; Qiuhua Li; Helen Y. Zou; James G. Christensen; Barbara Mroczkowski; Steve Bender; Robert Steven Kania; Martin Paul Edwards
Because of the critical roles of aberrant signaling in cancer, both c-MET and ALK receptor tyrosine kinases are attractive oncology targets for therapeutic intervention. The cocrystal structure of 3 (PHA-665752), bound to c-MET kinase domain, revealed a novel ATP site environment, which served as the target to guide parallel, multiattribute drug design. A novel 2-amino-5-aryl-3-benzyloxypyridine series was created to more effectively make the key interactions achieved with 3. In the novel series, the 2-aminopyridine core allowed a 3-benzyloxy group to reach into the same pocket as the 2,6-dichlorophenyl group of 3 via a more direct vector and thus with a better ligand efficiency (LE). Further optimization of the lead series generated the clinical candidate crizotinib (PF-02341066), which demonstrated potent in vitro and in vivo c-MET kinase and ALK inhibition, effective tumor growth inhibition, and good pharmaceutical properties.
Journal of Medicinal Chemistry | 2011
Angelica Linton; Ping Kang; Martha Ornelas; Susan Kephart; Qiyue Hu; Mason Alan Pairish; Ying Jiang; Chuangxing Guo
N-{trans-3-[(5-Cyano-6-methylpyridin-2-yl)oxy]-2,2,4,4-tetramethylcyclobutyl}imidazo[1,2-a]pyrimidine-3-carboxamide (1) was recently identified as a full antagonist of the androgen receptor, demonstrating excellent in vivo tumor growth inhibition in castration-resistant prostate cancer (CRPC). However, the imidazo[1,2-a]pyrimidine moiety is rapidly metabolized by aldehyde oxidase (AO). The present paper describes a number of medicinal chemistry strategies taken to avoid the AO-mediated oxidation of this particular system. Guided by an AO protein structure-based model, our investigation revealed the most probable site of AO oxidation and the observation that altering the heterocycle or blocking the reactive site are two of the more effective strategies for reducing AO metabolism. These strategies may be useful for other drug discovery programs.
Bioorganic & Medicinal Chemistry Letters | 2013
Chuangxing Guo; Angelica Linton; Mehran Jalaie; Susan Kephart; Martha Ornelas; Mason Alan Pairish; Samantha Greasley; Paul G. Richardson; Karen Maegley; Michael J. Hickey; John Li; Xin Wu; Xiaodong Ji; Zhi Xie
The M2 isoform of pyruvate kinase is an emerging target for antitumor therapy. In this letter, we describe the discovery of 2-((1H-benzo[d]imidazol-1-yl)methyl)-4H-pyrido[1,2-a]pyrimidin-4-ones as potent and selective PKM2 activators which were found to have a novel binding mode. The original lead identified from high throughput screening was optimized into an efficient series via computer-aided structure-based drug design. Both a representative compound from this series and an activator described in the literature were used as molecular tools to probe the biological effects of PKM2 activation on cancer cells. Our results suggested that PKM2 activation alone is not sufficient to alter cancer cell metabolism.
Journal of Medicinal Chemistry | 2017
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
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.
Bioorganic & Medicinal Chemistry Letters | 2012
Chuangxing Guo; Susan Kephart; Martha Ornelas; Javier Gonzalez; Angelica Linton; Mason Alan Pairish; Asako Nagata; Samantha Greasley; Jeff Elleraas; Natilie Hosea; Jon Engebretsen; Andrea Fanjul
High throughput cell-based screening led to the identification of 3-aryloxy lactams as potent androgen receptor (AR) antagonists. Refinement of these leads to improve the ADME profile and remove residual agonism led to the discovery of 12, a potent full antagonist with greater oral bioavailability. Improvements in the ADME profile were realized by designing more ligand-efficient molecules with reduced molecular weights and lower lipophilicities.
Bioorganic & Medicinal Chemistry Letters | 2012
Cynthia Louise Palmer; Mason Alan Pairish; Susan Kephart; Djamal Bouzida; Jingrong Cui; Judith Gail Deal; Liming Dong; Danlin Gu; Angelica Linton; Indrawan McAlpine; Shinji Yamazaki; Evan Smith; Annette John-Baptiste; Shubha Bagrodia; Robert Steven Kania; Chuangxing Guo
(S)-1-((4-(3-(6-Amino-5-methoxypyridin-3-yl)-1-isopropyl-1H-pyrazol-4-yl)pyrimidin-2-yl)amino)propan-2-ol, 1, was recently identified as a potent inhibitor of the oncogenic kinase bRAF. Compounds containing 3-methoxy-2-aminopyridine, as in 1, comprised a promising lead series because of their high ligand efficiency and excellent ADME profile. However, following metabolic oxidation, compounds in this series also demonstrated two significant safety risks: mutagenic potential and time-dependent drug-drug interaction (TDI). Metabolite identification studies revealed formation of a reactive metabolite. We hypothesized that minimizing or blocking the formation of such a metabolite would mitigate the safety liabilities. Our investigation demonstrated that structural modifications which either reduced the electron density of the 3-methoxy-2-aminopyridine ring or blocked the reactive site following metabolic oxidation were successful in reducing TDI and AMES mutagenicity.
Cancer Research | 2010
Henry Cheng; Shubha Bagrodia; Simon Bailey; Dilip Bhumalkar; Klaus Ruprecht Dress; Martin Paul Edwards; Michael R. Gehring; Lisa Guo; Jacqui Elizabeth Hoffman; Qiyue Hu; Xiaojun Huang; Catherine Johnson; Ted O. Johnson; Robert Steven Kania; Daniel R. Knighton; Phuong T. Le; Haitao Li; Samuel Li; Kevin K.-C. Liu; Zhengyu Liu; Matthew A. Marx; Mitch Nambu; Sacha Ninkovic; Dawn Nowlin; Mason Alan Pairish; Andrew Pannifer; Michael Bruno Plewe; Caroline Rodgers; Graham Smith; Shaoxian Sun
Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC 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. In an effort to discover compounds that inhibit PI3Kα, a high throughput screen was carried out, and 4-methyl-pyrido-pyrimidine (MPP) derivatives were identified as potent and selective inhibitors of PI3Kα. For example, PF-00271897, 8-cyclopentyl-6-[3-(hydroxymethyl)phenyl]-4-methyl-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one demonstrated PI3Kα Ki of 2.2 nM. Multiple crystal structures of inhibitors bound to PI3K gamma were determined to inform design and optimization of the ADMET properties of this lead series. Crystallographic studies with PI3K gamma protein indicated that the aminopyrimidine moiety forms two hydrogen bonds to the kinase backbone, and the aromatic moiety at the 6 position binds in a hydrophobic pocket. The X-ray structure suggested that the 4-methyl group on the MPP core structure conferred the excellent overall kinase selectivity to the series. The structure and SAR suggested optimization could come from keeping N-R group at 2 position very small and maintaining aromatic moiety at 6 position for hydrophobic interaction. Introduction of polar groups to the 8N side chains that are located in the ribose binding pocket increased both metabolic stability and solubility. Based on the overall properties, PF-04691502, 2-amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxypyridin-3-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one, was selected as a clinical candidate. PF-04691502 demonstrated Kis of 1.2-2.2 nM against PI3K α, β, γ and δ isoforms, and Ki of 9.1 nM against recombinant mTOR. PF-04691502 inhibited AKT phosphorylation at S473 in BT20 breast cancer line with IC50 of 12 nM. PF-04691502 is highly selective for inhibition of PI3K family kinases as shown by lack of activity against a panel of >75 protein kinases, including the class III PI3K hVps34. In the in vivo rat PK studies, PF-04691502 demonstrated the following properties: Clearance = 5.2 ml/min/kg, Vdss = 1.4 L/kg, T1/2 = 3.1 h, F% = 63%. The design, synthesis, in vitro potency SAR, selectivity, ADMET of the MPP derivatives will be discussed. The crystal structure of PF-04691502 in PI3K gamma will also be presented. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5779.
Archive | 2004
Jingjong Jean Cui; Joanne Johnson; Stephen A Kolodziej; Pei-Pei Kung; Xiaoyuan Sharon Li; Jason Qishen Lin; Jerry Jialun Meng; Mitchell David Nambu; Christopher G Nelson; Mason Alan Pairish; Hong Shen; Jennifer Zhang; Michelle Bich Tran-Dube; Allison Walter; Fang-Jie Zhang; Cathleen Elizabeth Hanau; Dilip Bhumralkar; Iriny Botrous; Ji Yu Chu; Lee Andrew Funk; Jr. G. Davis Harris; Lei Jia
Archive | 2005
Jingrong Jean Pfizer Global R D Cui; Lee Andrew Funk; Lei Pfizer Global R D Jia; Pei-Pei Pfizer Global R D Kung; Jerry Jialun Pfizer Global R D Meng; Mitchell David Nambu; Mason Alan Pairish; Hong Pfizer Global R D Shen; Michelle Bich Tran-Dube