Network


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

Hotspot


Dive into the research topics where Lusong Luo is active.

Publication


Featured researches published by Lusong Luo.


Cancer Research | 2006

Demonstration of a Genetic Therapeutic Index for Tumors Expressing Oncogenic BRAF by the Kinase Inhibitor SB-590885

Alastair J. King; Denis R. Patrick; Roberta S. Batorsky; Maureen L. Ho; Hieu T. Do; Shu Yun Zhang; Rakesh Kumar; David W. Rusnak; Andrew K. Takle; David M. Wilson; Erin D. Hugger; Lifu Wang; Florian A. Karreth; Julie Lougheed; Jae Lee; David Hau Wing Chau; Thomas J. Stout; Earl W. May; Cynthia M. Rominger; Michael D. Schaber; Lusong Luo; Ami S. Lakdawala; Jerry L. Adams; Rooja G. Contractor; Keiran S.M. Smalley; Meenhard Herlyn; Michael M. Morrissey; David A. Tuveson; Pearl S. Huang

Oncogenic BRAF alleles are both necessary and sufficient for cellular transformation, suggesting that chemical inhibition of the activated mutant protein kinase may reverse the tumor phenotype. Here, we report the characterization of SB-590885, a novel triarylimidazole that selectively inhibits Raf kinases with more potency towards B-Raf than c-Raf. Crystallographic analysis revealed that SB-590885 stabilizes the oncogenic B-Raf kinase domain in an active configuration, which is distinct from the previously reported mechanism of action of the multi-kinase inhibitor, BAY43-9006. Malignant cells expressing oncogenic B-Raf show selective inhibition of mitogen-activated protein kinase activation, proliferation, transformation, and tumorigenicity when exposed to SB-590885, whereas other cancer cell lines and normal cells display variable sensitivities or resistance to similar treatment. These studies support the validation of oncogenic B-Raf as a target for cancer therapy and provide the first evidence of a correlation between the expression of oncogenic BRAF alleles and a positive response to a selective B-Raf inhibitor.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Antitumor activity of an allosteric inhibitor of centromere-associated protein-E

Kenneth W. Wood; Latesh Lad; Lusong Luo; Xiangping Qian; Steven D. Knight; Neysa Nevins; Katjuša Brejc; David Sutton; Aidan G. Gilmartin; Penelope Chua; Radhika Desai; Stephen Schauer; Dean E. McNulty; Roland S. Annan; Lisa Belmont; Carlos Garcia; Yan Lee; Melody Diamond; Leo F. Faucette; Michele Giardiniere; Shu-Yun Zhang; Chiu-Mei Sun; Justin D. Vidal; Serge Lichtsteiner; William D. Cornwell; Joel Greshock; Richard Wooster; Jeffrey T. Finer; Robert A. Copeland; Pearl S. Huang

Centromere-associated protein-E (CENP-E) is a kinetochore-associated mitotic kinesin that is thought to function as the key receptor responsible for mitotic checkpoint signal transduction after interaction with spindle microtubules. We have identified GSK923295, an allosteric inhibitor of CENP-E kinesin motor ATPase activity, and mapped the inhibitor binding site to a region similar to that bound by loop-5 inhibitors of the kinesin KSP/Eg5. Unlike these KSP inhibitors, which block release of ADP and destabilize motor-microtubule interaction, GSK923295 inhibited release of inorganic phosphate and stabilized CENP-E motor domain interaction with microtubules. Inhibition of CENP-E motor activity in cultured cells and tumor xenografts caused failure of metaphase chromosome alignment and induced mitotic arrest, indicating that tight binding of CENP-E to microtubules is insufficient to satisfy the mitotic checkpoint. Consistent with genetic studies in mice suggesting that decreased CENP-E function can have a tumor-suppressive effect, inhibition of CENP-E induced tumor cell apoptosis and tumor regression.


Biochemical Journal | 2008

Effects of oncogenic p110α subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase

Jeffrey D. Carson; Glenn S. Van Aller; Ruth Lehr; Robert H. Sinnamon; Robert B. Kirkpatrick; Kurt R. Auger; Dashyant Dhanak; Robert A. Copeland; Richard R. Gontarek; Peter J. Tummino; Lusong Luo

The PIK3CA gene, encoding the p110α catalytic subunit of Class IA PI3Ks (phosphoinositide 3-kinases), is frequently mutated in many human tumours. The three most common tumour-derived alleles of p110α, H1047R, E542K and E545K, were shown to potently activate PI3K signalling in human epithelial cells. In the present study, we examine the biochemical activity of the recombinantly purified PI3K oncogenic mutants. The kinetic characterizations of the wt (wild-type) and the three ‘hot spot’ PI3K mutants show that the mutants all have approx. 2-fold increase in lipid kinase activities. Interestingly, the phosphorylated IRS-1 (insulin receptor substrate-1) protein shows activation of the lipid kinase activity for the wt and H1047R but not E542K and E545K PI3Kα, suggesting that these mutations represent different mechanisms of lipid kinase activation and hence transforming activity in cancer cells.


Biochemical and Biophysical Research Communications | 2011

Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor.

Glenn S. Van Aller; Jeffrey D. Carson; Wei Tang; Hao Peng; Lin Zhao; Robert A. Copeland; Peter J. Tummino; Lusong Luo

The PI3K signaling pathway is activated in a broad spectrum of human cancers, either directly by genetic mutation or indirectly via activation of receptor tyrosine kinases or inactivation of the PTEN tumor suppressor. The key nodes of this pathway have emerged as important therapeutic targets for the treatment of cancer. In this study, we show that (-)-epigallocatechin-3-gallate (EGCG), a major component of green tea, is an ATP-competitive inhibitor of both phosphoinositide-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) with K(i) values of 380 and 320nM respectively. The potency of EGCG against PI3K and mTOR is within physiologically relevant concentrations. In addition, EGCG inhibits cell proliferation and AKT phosphorylation at Ser473 in MDA-MB-231 and A549 cells. Molecular docking studies show that EGCG binds well to the PI3K kinase domain active site, agreeing with the finding that EGCG competes for ATP binding. Our results suggest another important molecular mechanism for the anticancer activities of EGCG.


Biochemistry | 2008

Mechanism of Inhibition of Human KSP by Ispinesib

Latesh Lad; Lusong Luo; Jeffrey D. Carson; Kenneth W. Wood; James J. Hartman; Robert A. Copeland; Roman Sakowicz

KSP, also known as HsEg5, is a kinesin that plays an essential role in the formation of a bipolar mitotic spindle and is required for cell cycle progression through mitosis. Ispinesib is the first potent, highly specific small-molecule inhibitor of KSP tested for the treatment of human disease. This novel anticancer agent causes mitotic arrest and growth inhibition in several human tumor cell lines and is currently being tested in multiple phase II clinical trials. In this study we have used steady-state and pre-steady-state kinetic assays to define the mechanism of KSP inhibition by ispinesib. Our data show that ispinesib alters the ability of KSP to bind to microtubules and inhibits its movement by preventing the release of ADP without preventing the release of the KSP-ADP complex from the microtubule. This type of inhibition is consistent with the physiological effect of ispinesib on cells, which is to prevent KSP-driven mitotic spindle pole separation. A comparison of ispinesib to monastrol, another small-molecule inhibitor of KSP, reveals that both inhibitors share a common mode of inhibition.


Protein Science | 2007

Characterization of an exosite binding inhibitor of matrix metalloproteinase 13

Lata T. Gooljarsingh; Ami S. Lakdawala; Frank T. Coppo; Lusong Luo; Gregg B. Fields; Peter J. Tummino; Richard R. Gontarek

Matrix metalloproteinase 13 (MMP13) is a key enzyme implicated in the degradation of the extracellular matrix in osteoarthritis. Clinical administration of broad spectrum MMP inhibitors such as marimastat has been implicated in severe musculo‐skeletal side effects. Consequently, research has been focused on designing inhibitors that selectively inhibit MMP13, thereby circumventing musculo‐skeletal toxicities. A series of pyrimidine dicarboxamides were recently shown to be highly selective inhibitors of MMP13 with a novel binding mode. We have applied a molecular ruler to this exosite by dual inhibition studies involving a potent dicarboxamide in the presence of two metal chelators of different sizes. A larger hydroxamate mimic overlaps and antagonizes binding of the dicarboxamide to the exosite whereas the much smaller acetohydroxamate synergizes with the dicarboxamide. These studies elucidate the steric requirement for compounds that fit exclusively into the active site, a mandate for generating highly selective MMP13 inhibitors.


Biochemical Journal | 2011

Biochemical characterization of human HIF hydroxylases using HIF protein substrates that contain all three hydroxylation sites

Melissa B. Pappalardi; Dean E. McNulty; John D. Martin; Kelly E. Fisher; Yong Jiang; Matthew C. Burns; Huizhen Zhao; Thau Ho; Sharon Sweitzer; Benjamin Schwartz; Roland S. Annan; Robert A. Copeland; Peter J. Tummino; Lusong Luo

The HIF (hypoxia-inducible factor) plays a central regulatory role in oxygen homoeostasis. HIF proteins are regulated by three Fe(II)- and α-KG (α-ketoglutarate)-dependent prolyl hydroxylase enzymes [PHD (prolyl hydroxylase domain) isoenzymes 1-3 or PHD1, PHD2 and PHD3] and one asparaginyl hydroxylase [FIH (factor inhibiting HIF)]. The prolyl hydroxylases control the abundance of HIF through oxygen-dependent hydroxylation of specific proline residues in HIF proteins, triggering subsequent ubiquitination and proteasomal degradation. FIH inhibits the HIF transcription activation through asparagine hydroxylation. Understanding the precise roles and regulation of these four Fe(II)- and α-KG-dependent hydroxylases is of great importance. In the present paper, we report the biochemical characterization of the first HIF protein substrates that contain the CODDD (C-terminal oxygen-dependent degradation domain), the NODDD (N-terminal oxygen-dependent degradation domain) and the CAD (C-terminal transactivation domain). Using LC-MS/MS (liquid chromatography-tandem MS) detection, we show that all three PHD isoenzymes have a strong preference for hydroxylation of the CODDD proline residue over the NODDD proline residue and the preference is observed for both HIF1α and HIF2α protein substrates. In addition, steady-state kinetic analyses show differential substrate selectivity for HIF and α-KG in reference to the three PHD isoforms and FIH.


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.


Biochemistry | 2008

Biochemical Characterization of Human Prolyl Hydroxylase Domain Protein 2 Variants Associated with Erythrocytosis

Melissa B. Pappalardi; John D. Martin; Yong Jiang; Matthew C. Burns; Huizhen Zhao; Thau Ho; Sharon Sweitzer; Leng Lor; Benjamin J. Schwartz; Kevin J. Duffy; Richard R. Gontarek; Peter J. Tummino; Robert A. Copeland; Lusong Luo

Prolyl hydroxylase domain proteins (PHD isozymes 1-3) regulate levels of the alpha-subunit of the hypoxia inducible factor (HIF) through proline hydroxylation, earmarking HIFalpha for proteosome-mediated degradation. Under hypoxic conditions, HIF stabilization leads to enhanced transcription and regulation of a multitude of processes, including erythropoiesis. Herein, we examine the biochemical characterization of PHD2 variants, Arg371His and Pro317Arg, identified from patients with familial erythrocytosis. The variants display differential effects on catalytic rate and substrate binding, implying that partial inhibition or selective inhibition with regard to HIFalpha isoforms of PHD2 could result in the phenotype displayed by patients with familial erythrocytosis.

Collaboration


Dive into the Lusong Luo's collaboration.

Top Co-Authors

Avatar

Lai Wang

Fudan University Shanghai Medical College

View shared research outputs
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