Network


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

Hotspot


Dive into the research topics where Carla A. Donatelli is active.

Publication


Featured researches published by Carla A. Donatelli.


ACS Medicinal Chemistry Letters | 2010

Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin.

Steven David Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Juan I. Luengo; Ken A. Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Melody Diamond; Patricia A. Elkins; Christine M. Gardiner; Eric Garver; Seth Gilbert; Richard R. Gontarek; Jeffrey R. Jackson; Kevin L. Kershner; Lusong Luo; Kaushik Raha; Christian S. Sherk; Chiu-Mei Sung; David Sutton; Peter J. Tummino; Ronald Wegrzyn; Kurt R. Auger

Phosphoinositide 3-kinase α (PI3Kα) is a critical regulator of cell growth and transformation, and its signaling pathway is the most commonly mutated pathway in human cancers. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of PI3K/AKT pathway inhibition. 2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide (GSK2126458, 1) has been identified as a highly potent, orally bioavailable inhibitor of PI3Kα and mTOR with in vivo activity in both pharmacodynamic and tumor growth efficacy models. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.


Journal of Medicinal Chemistry | 2010

Discovery of GSK1070916, a Potent and Selective Inhibitor of Aurora B/C Kinase

Nicholas D. Adams; Jerry L. Adams; Joelle L. Burgess; Amita M. Chaudhari; Robert A. Copeland; Carla A. Donatelli; David H. Drewry; Kelly E. Fisher; Toshihiro Hamajima; Mary Ann Hardwicke; William F. Huffman; Kristin K. Koretke-Brown; Zhihong V. Lai; Octerloney B. McDonald; Hiroko Nakamura; Ken A. Newlander; Catherine A. Oleykowski; Cynthia A. Parrish; Denis R. Patrick; Ramona Plant; Martha A. Sarpong; Kosuke Sasaki; Stanley J. Schmidt; Domingos J. Silva; David Sutton; Jun Tang; Christine Thompson; Peter J. Tummino; Jamin C. Wang; Hong Xiang

The Aurora kinases play critical roles in the regulation of mitosis and are frequently overexpressed or amplified in human tumors. Selective inhibitors may provide a new therapy for the treatment of tumors with Aurora kinase amplification. Herein we describe our lead optimization efforts within a 7-azaindole-based series culminating in the identification of GSK1070916 (17k). Key to the advancement of the series was the introduction of a 2-aryl group containing a basic amine onto the azaindole leading to significantly improved cellular activity. Compound 17k is a potent and selective ATP-competitive inhibitor of Aurora B and C with K(i)* values of 0.38 +/- 0.29 and 1.5 +/- 0.4 nM, respectively, and is >250-fold selective over Aurora A. Biochemical characterization revealed that compound 17k has an extremely slow dissociation half-life from Aurora B (>480 min), distinguishing it from clinical compounds 1 and 2. In vitro treatment of A549 human lung cancer cells with compound 17k results in a potent antiproliferative effect (EC(50) = 7 nM). Intraperitoneal administration of 17k in mice bearing human tumor xenografts leads to inhibition of histone H3 phosphorylation at serine 10 in human colon cancer (Colo205) and tumor regression in human leukemia (HL-60). Compound 17k is being progressed to human clinical trials.


ACS Medicinal Chemistry Letters | 2010

Discovery of the First Potent and Selective Inhibitor of Centromere-Associated Protein E: GSK923295.

Xiangping Qian; Andrew Mcdonald; Han-Jie Zhou; Nicholas D. Adams; Cynthia A. Parrish; Kevin J. Duffy; Duke M. Fitch; Rosanna Tedesco; Luke W. Ashcraft; Bing Yao; Hong Jiang; Jennifer Kuo Chen Huang; Melchor V. Marin; Carrie E. Aroyan; Jianchao Wang; Seyed Ahmed; Joelle L. Burgess; Amita M. Chaudhari; Carla A. Donatelli; Michael G. Darcy; Lance H. Ridgers; Ken A. Newlander; Stanley J. Schmidt; Deping Chai; Mariela Colón; Michael N. Zimmerman; Latesh Lad; Roman Sakowicz; Stephen Schauer; Lisa Belmont

Inhibition of mitotic kinesins represents a novel approach for the discovery of a new generation of anti-mitotic cancer chemotherapeutics. We report here the discovery of the first potent and selective inhibitor of centromere-associated protein E (CENP-E) 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide (GSK923295; 1), starting from a high-throughput screening hit, 3-chloro-4-isopropoxybenzoic acid 2. Compound 1 has demonstrated broad antitumor activity in vivo and is currently in human clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of a new series of Aurora inhibitors through truncation of GSK1070916.

Jesus R. Medina; Seth W. Grant; Jeffrey M. Axten; William Henry Miller; Carla A. Donatelli; Mary Ann Hardwicke; Catherine A. Oleykowski; Qiaoyin Liao; Ramona Plant; Hong Xiang

Novel Aurora inhibitors were identified truncating clinical candidate GSK1070916. Many of these truncated compounds retained potent activity against Aurora B with good antiproliferative activity. Mechanistic studies suggested that these compounds, depending on the substitution pattern, may or may not exert their antiproliferative effects via inhibition of Aurora B. The SAR results from this investigation will be presented with an emphasis on the impact structural changes have on the cellular phenotype.


Molecular Cancer Therapeutics | 2009

Abstract C62: Identification of GSK2126458, a highly potent inhibitor of phosphoinositide 3‐kinase (PI3K) and the mammalian target of rapamycin (mTOR)

Steven D. Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Ken Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Patricia A. Elkins; Melody Diamond; Christine M. Gardiner; Eric Garver; Lusong Luo; Kaushik Raha; Chiu-Mei Sung; Peter J. Tummino; Kurt R. Auger; Dashyant Dhanak

Phosphoinositide 3‐kinase (PI3K) is a critical regulator of cell growth and transformation and its signaling pathway is one of the most commonly mutated pathways in human cancer. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of the PI3K/AKT pathway. GSK1059615, our first PI3K clinical compound, progressed to a dose escalation study in patients with refractory malignancies. Following the discovery of GSK1059615, we sought to identify a second inhibitor with improved potency, selectivity, and pharmacokinetics. Key to our approach to achieving the desired levels of PI3K activity was to pursue structure‐based design utilizing crystallography of the more amenable PI3K as a surrogate protein. Following a chemistry lead optimization effort, the pyridylsulfonamide GSK2126458 was identified as a highly potent, orally bioavailable, pan‐PI3K and mTOR inhibitor (PI3K app Ki = 19 pM; mTORC1 app Ki = 180 pM; mTORC2 app Ki = 300 pM). Consistent with potent PI3K and mTORC2 enzyme inhibition, GSK2126458 decreased cellular levels of phosphorylated AKT (BT474 pAKT IC50 = 180 pM) and inhibited cell proliferation in a large panel of cancer cell lines (e.g. BT474 growth IC50 = 2 nM). GSK2126458 showed good exposure in four pre‐clinical animal species and exhibited in vivo activity in both pharmacodynamic and tumor growth efficacy models. GSK2126458 is being evaluated currently in human clinical trials for the treatment of cancer. The discovery, design, and optimization of GSK2126458 and related analogs will be presented. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):C62.


Journal of Medicinal Chemistry | 2018

Discovery of Pyrrolidine Sulfonamides as Selective and Orally Bioavailable Antagonists of Transient Receptor Potential Vanilloid-4 (TRPV4)

Edward J. Brnardic; Guosen Ye; Carl Brooks; Carla A. Donatelli; Linda S. Barton; Jeff J. McAtee; Robert M. Sanchez; Arthur Y. L. Shu; Karl F. Erhard; Lamont Terrell; Grazyna Graczyk-Millbrandt; Yanan He; Melissa H. Costell; David J. Behm; Theresa J. Roethke; Patrick Stoy; Dennis Alan Holt; Brian G. Lawhorn

A novel series of pyrrolidine sulfonamide transient receptor potential vanilloid-4 (TRPV4) antagonists was developed by modification of a previously reported TRPV4 inhibitor (1). Several core-structure modifications were identified that improved TRPV4 activity by increasing structural rigidity and reducing the entropic energy penalty upon binding to the target protein. The new template was initially discovered as a minor regio-isomeric side product formed during routine structure-activity relationship (SAR) studies, and further optimization resulted in highly potent compounds with a novel pyrrolidine diol core. Further improvements in potency and pharmacokinetic properties were achieved through SAR studies on the sulfonamide substituent to give an optimized lead compound GSK3395879 (52) that demonstrated the ability to inhibit TRPV4-mediated pulmonary edema in an in vivo rat model. GSK3395879 is a tool for studying the biology of TRPV4 and an advanced lead for identifying new heart failure medicines.


Journal of Medicinal Chemistry | 2007

Novel ATP-competitive kinesin spindle protein inhibitors

Cynthia A. Parrish; Nicholas D. Adams; Kurt R. Auger; Joelle L. Burgess; Jeffrey D. Carson; Amita M. Chaudhari; Robert A. Copeland; Melody Diamond; Carla A. Donatelli; Kevin J. Duffy; Leo F. Faucette; Jeffrey T. Finer; William F. Huffman; Erin D. Hugger; Jeffrey R. Jackson; Steven David Knight; Lusong Luo; Michael L. Moore; Ken A. Newlander; Lance H. Ridgers; Roman Sakowicz; Antony N. Shaw; Chiu-Mei M. Sung; David Sutton; Kenneth W. Wood; Shu-Yun Zhang; Michael N. Zimmerman; Dashyant Dhanak


Archive | 2008

Quinoline derivatives as p13 kinase inhibitors

Nicholas D. Adams; Joelle L. Burgess; Michael G. Darcy; Carla A. Donatelli; Steven D. Knight; Kenneth A. Newlander; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt


Archive | 2008

Quinoline derivatives as pi3 kinase inhibitors

Nicholas D. Adams; Joelle L. Burgess; Michael Gerrard Darcy; Carla A. Donatelli; Steven David Knight; Kenneth A. Newlander; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt


Archive | 2017

intensificador de inibidores de zeste homólogo 2

Carla A. Donatelli; Kenneth A. Newlander; Steven David Knight; William Henry Miller

Collaboration


Dive into the Carla A. Donatelli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge