Network


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

Hotspot


Dive into the research topics where Simon Paul Planken is active.

Publication


Featured researches published by Simon Paul Planken.


Molecular Cancer Therapeutics | 2012

Effective Targeting of Hedgehog Signaling in a Medulloblastoma Model with PF-5274857, a Potent and Selective Smoothened Antagonist That Penetrates the Blood–Brain Barrier

Allison Rohner; Mary E. Spilker; Justine L. Lam; Bernadette Pascual; Darian Bartkowski; Qing John Li; Amy H. Yang; Greg Stevens; Meirong Xu; Peter A. Wells; Simon Paul Planken; Sajiv K. Nair; Shaoxian Sun

Inhibition of the Smoothened (Smo) represents a promising therapeutic strategy for treating malignant tumors that are dependent on the Hedgehog (Hh) signaling pathway. PF-5274857 is a novel Smo antagonist that specifically binds to Smo with a Ki of 4.6 ± 1.1 nmol/L and completely blocks the transcriptional activity of the downstream gene Gli1 with an IC50 of 2.7 ± 1.4 nmol/L in cells. This Smo antagonist showed robust antitumor activity in a mouse model of medulloblastoma with an in vivo IC50 of 8.9 ± 2.6 nmol/L. The downregulation of Gli1 is closely linked to the tumor growth inhibition in patched+/− medulloblastoma mice. Mathematical analysis of the relationship between the drugs pharmacokinetics and Gli1 pharmacodynamics in patched+/− medulloblastoma tumor models yielded similar tumor and skin Gli1 IC50 values, suggesting that skin can be used as a surrogate tissue for the measurement of tumor Gli1 levels. In addition, PF-5274857 was found to effectively penetrate the blood–brain barrier and inhibit Smo activity in the brain of primary medulloblastoma mice, resulting in improved animal survival rates. The brain permeability of PF-5274857 was also confirmed and quantified in nontumor-bearing preclinical species with an intact blood–brain barrier. PF-5274857 was orally available and metabolically stable in vivo. These findings suggest that PF-5274857 is a potentially attractive clinical candidate for the treatment of tumor types including brain tumors and brain metastasis driven by an activated Hh pathway. Mol Cancer Ther; 11(1); 57–65. ©2011 AACR.


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.


Chemical Biology & Drug Design | 2006

The discovery of small molecule inhibitors of neutral endopeptidase. Structure-activity studies on functionalized glutaramides.

Graham Nigel Maw; Alan Stobie; Simon Paul Planken; David C. Pryde; Vivienne Sanderson; Michelle Y. Platts; Martin Corless; Peter Stacey; Christopher Peter Wayman; Piet H. van der Graaf; Christopher Kohl; Sara Coggon; Kevin Beaumont

A series of small molecule glutaramides were synthesized and evaluated for potency against canine and human neutral endopeptidase using target criteria of molecular weight <400 and log P between 2 and 4.5 to maximize the likelihood of achieving good oral absorption. The structure‐activity relationship (SAR) investigations described in this paper led to the identification of an ethyl 1,3,4‐thiadiazole glutaramide which demonstrated good neutral endopeptidase potency, selectivity and excellent oral absorption in the rat.


Cancer Research | 2015

Abstract 2594: Characterization of a novel irreversible third generation EGFR TKI that targets T790M-mediated resistant EGFR-mutant NSCLC while sparing wild type EGFR

Mike Zientek; Sangita M. Baxi; Henry Cheng; Valeria R. Fantin; Jun Li Feng; Allison M. Given; Zelanna Goldberg; Jie Guo; Michelle Hemkens; John Charles Kath; Jennifer Lafontaine; Gary Li; Pramod P. Mehta; Brion W. Murray; Sajiv K. Nair; Simon Paul Planken; Chad Ray; Yuli Wang; Manli Shi; Anand Sistla; Tod Smeal; Greg Stevens; Wei Tan; Paolo Vicini; Marlena Walls; Liu Yang; Min-Jean Yin; Scott Weinrich

Activating mutations in EGFR confer constitutive activity providing the oncogenic drive in EGFR-mutant NSCLC. First and 2nd generation EGFR tyrosine kinase inhibitors (TKIs) are effective drugs in this setting, but are constrained by dose-limiting toxicities attributed to inhibition of wild type (WT) EGFR and by drug resistance caused, in the majority of cases, via a T790M secondary mutation in EGFR. We report the pharmacology of a novel irreversible 3rd generation EGFR TKI active against EGFR with activating and T790M mutations, but sparing WT EGFR. Our novel 3rd generation EGFR TKI was studied in a variety of in vitro and in vivo models to determine its inhibitory potencies on different EGFR variants, pharmacokinetics (PK), antitumor efficacy, exposure-response relationships, mechanism of action, and predicted human efficacious dose. In enzyme and cell assays, our compound is a highly potent inhibitor of EGFR double mutants (L858R/T790M and Del/T790M) and EGFR activating mutants (L858R and Del), but a weak inhibitor of WT EGFR (26-fold margin over mutant target potencies). Effects on downstream signaling and function indicate the underlying mechanism of the compound is direct inhibition of EGFR, with subsequent inhibition of downstream signaling that results in apoptosis and viable cell decline. In xenograft mouse models, the compound demonstrates tumor growth inhibition and regression at well-tolerated doses in models driven by EGFR double mutants and EGFR activating mutants. The antitumor efficacy is dose-dependent and strongly correlates with inhibition of EGFR phosphorylation and EGFR-mediated downstream signaling, and induction of apoptosis. Plasma concentrations assumed to be sufficient for efficacy (Ceff) were defined using a mathematical model incorporating the plasma levels of the compound, the associated inhibitory effects on EGFR phosphorylation, and the antitumor efficacy in the double and activating mutant xenograft models. Ceff was in agreement across several models and was used with in vitro human PK properties to calculate required human dose. While our compound possesses a similar profile as other recently disclosed 3rd generation EGFR TKIs, this molecule is distinguished by better potency on the activating mutants and by the widest potency margin on WT EGFR. Given that the target potencies and WT margins of 3rd generation EGFR TKIs have been sufficient for tolerated clinical efficacy in preliminary results, it can be inferred that our compound will have similar promise in the clinic. These results support our compound as a novel EGFR TKI with an inhibitory profile and favorable drug-like properties that suggest utility for treating patients with NSCLC with EGFR activating and resistance mutations. Citation Format: Mike Zientek, Sangita Baxi, Henry Cheng, Valeria Fantin, Jun Li Feng, Allison Given, Zelanna Goldberg, Jie Guo, Michelle Hemkens, John Kath, Jennifer Lafontaine, Gary Li, Pramod Mehta, Brion Murray, Sajiv Nair, Simon Planken, Chad Ray, Yuli Wang, Manli Shi, Anand Sistla, Tod Smeal, Greg Stevens, Wei Tan, Paolo Vicini, Marlena Walls, Liu Yang, Min-Jean Yin, Scott L. Weinrich. Characterization of a novel irreversible third generation EGFR TKI that targets T790M-mediated resistant EGFR-mutant NSCLC while sparing wild type EGFR. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2594. doi:10.1158/1538-7445.AM2015-2594


Archive | 2007

Substituted 2-amino-fused heterocyclic compounds

Liming Huang; Song Liu; Elizabeth A. Lunney; Simon Paul Planken


Journal of Medicinal Chemistry | 2016

Discovery of 1-{(3R,4R)-3-[({5-Chloro-2-[(1-methyl-1H-pyrazol-4-yl)amino]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}oxy)methyl]-4-methoxypyrrolidin-1-yl}prop-2-en-1-one (PF-06459988), a Potent, WT Sparing, Irreversible Inhibitor of T790M-Containing EGFR Mutants.

Hengmiao Cheng; Sajiv K. Nair; Brion W. Murray; Chau Almaden; Simon Bailey; Sangita Baxi; Doug Behenna; Sujin Cho-Schultz; Deepak Dalvie; Dac M. Dinh; Martin Paul Edwards; Jun Li Feng; Rose Ann Ferre; Ketan S. Gajiwala; Michelle D. Hemkens; Amy Jackson-Fisher; Mehran Jalaie; Ted O. Johnson; Robert Steven Kania; Susan Kephart; Jennifer Lafontaine; Beth Lunney; Kevin K.-C. Liu; Zhengyu Liu; Jean Matthews; Asako Nagata; Sherry Niessen; Martha Ornelas; Suvi T. M. Orr; Mason Alan Pairish


Journal of Medicinal Chemistry | 2006

Novel selective inhibitors of neutral endopeptidase for the treatment of female sexual arousal disorder. Synthesis and activity of functionalized glutaramides

David C. Pryde; Graham Nigel Maw; Simon Paul Planken; Michelle Y. Platts; Vivienne Sanderson; Martin Corless; Alan Stobie; Christopher Gordon Barber; Rachel Jane Russell; Laura Foster; Laura Barker; Christopher Peter Wayman; Piet H. van der Graaf; Peter Stacey; Debbie Morren; Christopher Kohl; Kevin Beaumont; Sara Coggon; Michael S. Tute


Archive | 2012

Pyrrolopyrimidine and purine derivatives

Hengmiao Cheng; Theodore Otto Johnson; John Charles Kath; Kevin K.-C. Liu; Elizabeth A. Lunney; Asako Nagata; Sajiv K. Nair; Simon Paul Planken; Scott C. Sutton


Archive | 2009

PYRAZOLE DERIVATIVES AS CYTOCHROME P450 INHIBITORS

Simon Paul Planken; Scott C. Sutton; Rongliang Chen


Journal of Medicinal Chemistry | 2007

Cyclization of the Acyl Glucuronide Metabolite of a Neutral Endopeptidase Inhibitor to an Electrophilic Glutarimide : Synthesis, Reactivity, and Mechanistic Analysis

Xiaoli Meng; James L. Maggs; David C. Pryde; Simon Paul Planken; Rosalind E. Jenkins; Torren Peakman; Kevin Beaumont; Christopher Kohl; B. Kevin Park; Andrew V. Stachulski

Collaboration


Dive into the Simon Paul Planken's collaboration.

Researchain Logo
Decentralizing Knowledge