Network


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

Hotspot


Dive into the research topics where Erin L. Mullady is active.

Publication


Featured researches published by Erin L. Mullady.


Journal of Medicinal Chemistry | 2011

Discovery and Optimization of a Series of Benzothiazole Phosphoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors

Noel D. D’Angelo; Tae-Seong Kim; Kristin L. Andrews; Shon Booker; Sean Caenepeel; Kui Chen; Derin C. D’Amico; Daniel J. Freeman; Jian Jiang; Longbin Liu; John D. McCarter; Tisha San Miguel; Erin L. Mullady; Michael L. Schrag; Raju Subramanian; Jin Tang; Robert C. Wahl; Ling Wang; Douglas A. Whittington; Tian Wu; Ning Xi; Yang Xu; Peter Yakowec; Kevin Yang; Leeanne Zalameda; Nancy R. Zhang; Paul E. Hughes; Mark H. Norman

Phosphoinositide 3-kinase α (PI3Kα) is a lipid kinase that plays a key regulatory role in several cellular processes. The mutation or amplification of this kinase in humans has been implicated in the growth of multiple tumor types. Consequently, PI3Kα has become a target of intense research for drug discovery. Our studies began with the identification of benzothiazole compound 1 from a high throughput screen. Extensive SAR studies led to the discovery of sulfonamide 45 as an early lead, based on its in vitro cellular potency. Subsequent modifications of the central pyrimidine ring dramatically improved enzyme and cellular potency and led to the identification of chloropyridine 70. Further arylsulfonamide SAR studies optimized in vitro clearance and led to the identification of 82 as a potent dual inhibitor of PI3K and mTOR. This molecule exhibited potent enzyme and cell activity, low clearance, and high oral bioavailability. In addition, compound 82 demonstrated tumor growth inhibition in U-87 MG, A549, and HCT116 tumor xenograft models.


Journal of Medicinal Chemistry | 2011

Structure-activity relationships of phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) dual inhibitors: investigations of various 6,5-heterocycles to improve metabolic stability.

Markian Stec; Kristin L. Andrews; Shon Booker; Sean Caenepeel; Daniel J. Freeman; Jian Jiang; Hongyu Liao; John D. McCarter; Erin L. Mullady; Tisha San Miguel; Raju Subramanian; Nuria A. Tamayo; Ling Wang; Kevin Yang; Leeanne Zalameda; Nancy Zhang; Paul E. Hughes; Mark H. Norman

N-(6-(6-Chloro-5-(4-fluorophenylsulfonamido)pyridin-3-yl)benzo[d]thiazol-2-yl)acetamide (1) is a potent and efficacious inhibitor of PI3Kα and mTOR in vitro and in vivo. However, in hepatocyte and in vivo metabolism studies, 1 was found to undergo deacetylation on the 2-amino substituent of the benzothiazole. As an approach to reduce or eliminate this metabolic deacetylation, a variety of 6,5-heterocyclic analogues were examined as an alternative to the benzothiazole ring. Imidazopyridazine 10 was found to have similar in vitro potency and in vivo efficacy relative to 1, while only minimal amounts of the corresponding deacetylated metabolite of 10 were observed in hepatocytes.


Journal of Medicinal Chemistry | 2013

Discovery of Novel, Induced-Pocket Binding Oxazolidinones as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors

Howard Bregman; Nagasree Chakka; Angel Guzman-Perez; Hakan Gunaydin; Yan Gu; Xin Huang; Virginia Berry; Jingzhou Liu; Yohannes Teffera; Liyue Huang; Bryan Egge; Erin L. Mullady; Steve Schneider; Paul S. Andrews; Ankita Mishra; John Newcomb; Randy Serafino; Craig A. Strathdee; Susan M. Turci; Cindy Wilson; Erin F. DiMauro

Tankyrase (TNKS) is a poly-ADP-ribosylating protein (PARP) whose activity suppresses cellular axin protein levels and elevates β-catenin concentrations, resulting in increased oncogene expression. The inhibition of tankyrase (TNKS1 and 2) may reduce the levels of β-catenin-mediated transcription and inhibit tumorigenesis. Compound 1 is a previously described moderately potent tankyrase inhibitor that suffers from poor pharmacokinetic properties. Herein, we describe the utilization of structure-based design and molecular modeling toward novel, potent, and selective tankyrase inhibitors with improved pharmacokinetic properties (39, 40).


Journal of Medicinal Chemistry | 2012

Structure-based design of a novel series of potent, selective inhibitors of the class I phosphatidylinositol 3-kinases.

Adrian L. Smith; Noel D'angelo; Yunxin Y. Bo; Shon Booker; Victor J. Cee; Brad Herberich; Fang-Tsao Hong; Claire L.M. Jackson; Brian A. Lanman; Longbin Liu; Nobuko Nishimura; Liping H. Pettus; Anthony B. Reed; Seifu Tadesse; Nuria A. Tamayo; Ryan Wurz; Kevin Yang; Kristin L. Andrews; Douglas A. Whittington; John D. McCarter; Tisha San Miguel; Leeanne Zalameda; Jian Jiang; Raju Subramanian; Erin L. Mullady; Sean Caenepeel; Daniel J. Freeman; Ling Wang; Nancy R. Zhang; Tian Wu

A highly selective series of inhibitors of the class I phosphatidylinositol 3-kinases (PI3Ks) has been designed and synthesized. Starting from the dual PI3K/mTOR inhibitor 5, a structure-based approach was used to improve potency and selectivity, resulting in the identification of 54 as a potent inhibitor of the class I PI3Ks with excellent selectivity over mTOR, related phosphatidylinositol kinases, and a broad panel of protein kinases. Compound 54 demonstrated a robust PD-PK relationship inhibiting the PI3K/Akt pathway in vivo in a mouse model, and it potently inhibited tumor growth in a U-87 MG xenograft model with an activated PI3K/Akt pathway.


Journal of Medicinal Chemistry | 2012

Selective Class I Phosphoinositide 3-Kinase Inhibitors: Optimization of a Series of Pyridyltriazines Leading to the Identification of a Clinical Candidate, AMG 511

Mark H. Norman; Kristin L. Andrews; Yunxin Y. Bo; Shon Booker; Sean Caenepeel; Victor J. Cee; Noel D. D’Angelo; Daniel J. Freeman; Bradley J. Herberich; Fang-Tsao Hong; Claire L.M. Jackson; Jian Jiang; Brian A. Lanman; Longbin Liu; John D. McCarter; Erin L. Mullady; Nobuko Nishimura; Liping H. Pettus; Anthony B. Reed; Tisha San Miguel; Adrian L. Smith; Markian Stec; Seifu Tadesse; Andrew Tasker; Divesh Aidasani; Xiaochun Zhu; Raju Subramanian; Nuria A. Tamayo; Ling Wang; Douglas A. Whittington

The phosphoinositide 3-kinase family catalyzes the phosphorylation of phosphatidylinositol-4,5-diphosphate to phosphatidylinositol-3,4,5-triphosphate, a secondary messenger which plays a critical role in important cellular functions such as metabolism, cell growth, and cell survival. Our efforts to identify potent, efficacious, and orally available phosphatidylinositol 3-kinase (PI3K) inhibitors as potential cancer therapeutics have resulted in the discovery of 4-(2-((6-methoxypyridin-3-yl)amino)-5-((4-(methylsulfonyl)piperazin-1-yl)methyl)pyridin-3-yl)-6-methyl-1,3,5-triazin-2-amine (1). In this paper, we describe the optimization of compound 1, which led to the design and synthesis of pyridyltriazine 31, a potent pan inhibitor of class I PI3Ks with a superior pharmacokinetic profile. Compound 31 was shown to potently block the targeted PI3K pathway in a mouse liver pharmacodynamic model and inhibit tumor growth in a U87 malignant glioma glioblastoma xenograft model. On the basis of its excellent in vivo efficacy and pharmacokinetic profile, compound 31 was selected for further evaluation as a clinical candidate and was designated AMG 511.


Journal of Medicinal Chemistry | 2013

Development of Novel Dual Binders as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors

Zihao Hua; Howard Bregman; John L. Buchanan; Nagasree Chakka; Angel Guzman-Perez; Hakan Gunaydin; Xin Huang; Yan Gu; Virginia Berry; Jingzhou Liu; Yohannes Teffera; Liyue Huang; Bryan Egge; Renee Emkey; Erin L. Mullady; Steve Schneider; Paul S. Andrews; Lisa Acquaviva; Jennifer Dovey; Ankita Mishra; John Newcomb; Douglas Saffran; Randy Serafino; Craig A. Strathdee; Susan M. Turci; Mary K. Stanton; Cindy Wilson; Erin F. DiMauro

Tankyrases (TNKS1 and TNKS2) are proteins in the poly ADP-ribose polymerase (PARP) family. They have been shown to directly bind to axin proteins, which negatively regulate the Wnt pathway by promoting β-catenin degradation. Inhibition of tankyrases may offer a novel approach to the treatment of APC-mutant colorectal cancer. Hit compound 8 was identified as an inhibitor of tankyrases through a combination of substructure searching of the Amgen compound collection based on a minimal binding pharmacophore hypothesis and high-throughput screening. Herein we report the structure- and property-based optimization of compound 8 leading to the identification of more potent and selective tankyrase inhibitors 22 and 49 with improved pharmacokinetic properties in rodents, which are well suited as tool compounds for further in vivo validation studies.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of triazine-benzimidazoles as selective inhibitors of mTOR.

Emily A. Peterson; Paul S. Andrews; Xuhai Be; Alessandro Boezio; Tammy L. Bush; Alan C. Cheng; James R. Coats; Adria E. Colletti; Katrina W. Copeland; Michelle DuPont; Russell Graceffa; Barbara Grubinska; Jean-Christophe Harmange; Joseph L. Kim; Erin L. Mullady; Philip R. Olivieri; Laurie B. Schenkel; Mary K. Stanton; Yohannes Teffera; Douglas A. Whittington; Ti Cai; Daniel S. La

mTOR is part of the PI3K/AKT pathway and is a central regulator of cell growth and survival. Since many cancers display mutations linked to the mTOR signaling pathway, mTOR has emerged as an important target for oncology therapy. Herein, we report the discovery of triazine benzimidazole inhibitors that inhibit mTOR kinase activity with up to 200-fold selectivity over the structurally homologous kinase PI3Kα. When tested in a panel of cancer cell lines displaying various mutations, a selective inhibitor from this series inhibited cellular proliferation with a mean IC(50) of 0.41 μM. Lead compound 42 demonstrated up to 83% inhibition of mTOR substrate phosphorylation in a murine pharmacodynamic model.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery and optimization of potent and selective imidazopyridine and imidazopyridazine mTOR inhibitors

Emily A. Peterson; Alessandro Boezio; Paul S. Andrews; Christiane Boezio; Tammy L. Bush; Alan C. Cheng; Deborah Choquette; James R. Coats; Adria E. Colletti; Katrina W. Copeland; Michelle DuPont; Russell Graceffa; Barbara Grubinska; Joseph L. Kim; Richard T. Lewis; Jingzhou Liu; Erin L. Mullady; Michele Potashman; Karina Romero; Paul L. Shaffer; Mary K. Stanton; John Stellwagen; Yohannes Teffera; Shuyan Yi; Ti Cai; Daniel S. La

mTOR is a critical regulator of cellular signaling downstream of multiple growth factors. The mTOR/PI3K/AKT pathway is frequently mutated in human cancers and is thus an important oncology target. Herein we report the evolution of our program to discover ATP-competitive mTOR inhibitors that demonstrate improved pharmacokinetic properties and selectivity compared to our previous leads. Through targeted SAR and structure-guided design, new imidazopyridine and imidazopyridazine scaffolds were identified that demonstrated superior inhibition of mTOR in cellular assays, selectivity over the closely related PIKK family and improved in vivo clearance over our previously reported benzimidazole series.


Bioorganic & Medicinal Chemistry Letters | 2012

Synthesis and structure-activity relationships of dual PI3K/mTOR inhibitors based on a 4-amino-6-methyl-1,3,5-triazine sulfonamide scaffold

Ryan Wurz; Longbin Liu; Kevin Yang; Nobuko Nishimura; Yunxin Bo; Liping H. Pettus; Sean Caenepeel; Daniel J. Freeman; John D. McCarter; Erin L. Mullady; Tisha San Miguel; Ling Wang; Nancy Zhang; Kristin L. Andrews; Douglas A. Whittington; Jian Jiang; Raju Subramanian; Paul E. Hughes; Mark H. Norman

Phosphoinositide 3-kinase (PI3K) is an important target in oncology due to the deregulation of the PI3K/Akt signaling pathway in a wide variety of tumors. A series of 4-amino-6-methyl-1,3,5-triazine sulfonamides were synthesized and evaluated as inhibitors of PI3K. The synthesis, in vitro biological activities, pharmacokinetic and in vivo pharmacodynamic profiling of these compounds are described. The most promising compound from this investigation (compound 3j) was found to be a pan class I PI3K inhibitor with a moderate (>10-fold) selectivity over the mammalian target of rapamycin (mTOR) in the enzyme assay. In a U87 MG cellular assay measuring phosphorylation of Akt, compound 3j displayed low double digit nanomolar IC(50) and exhibited good oral bioavailability in rats (F(oral)=63%). Compound 3j also showed a dose dependent reduction in the phosphorylation of Akt in a U87 tumor pharmacodynamic model with a plasma EC(50)=193 nM (91 ng/mL).


Bioorganic & Medicinal Chemistry Letters | 2015

The imidazo[1,2-a]pyridine ring system as a scaffold for potent dual phosphoinositide-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitors.

Markian Stec; Kristin L. Andrews; Yunxin Bo; Sean Caenepeel; Hongyu Liao; John D. McCarter; Erin L. Mullady; Tisha San Miguel; Raju Subramanian; Nuria A. Tamayo; Douglas A. Whittington; Ling Wang; Tian Wu; Leeanne Zalameda; Nancy Zhang; Paul E. Hughes; Mark H. Norman

Based on lead compound 1, which was discovered from a high-throughput screen, a series of PI3Kα/mTOR inhibitors were evaluated that contained an imidazo[1,2-a]pyridine as a core replacement for the benzimidazole contained in 1. By exploring various ring systems that occupy the affinity pocket, two fragments containing a methoxypyridine were identified that gave <100 nM potency toward PI3Kα in enzyme and cellular assays with moderate stability in rat and human liver microsomes. With the two methoxypyridine groups selected to occupy the affinity pocket, analogs were prepared with various fragments intended to occupy the ribose pocket of PI3Kα and mTOR. From these analogs, tertiary alcohol 18 was chosen for in vivo pharmacodynamic evaluation based on its potency in the PI3Kα cellular assay, microsomal stability, and in vivo pharmacokinetic properties. In a mouse liver pharmacodynamic assay, compound 18 showed 56% inhibition of HFG-induced AKT (Ser473) phosphorylation at a 30 mg/kg dose.

Researchain Logo
Decentralizing Knowledge