Network


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

Hotspot


Dive into the research topics where Dianlin Xie is active.

Publication


Featured researches published by Dianlin Xie.


Cancer Research | 2006

The Structure of Dasatinib (BMS-354825) Bound to Activated ABL Kinase Domain Elucidates Its Inhibitory Activity against Imatinib-Resistant ABL Mutants

John S. Tokarski; John A. Newitt; Chieh Ying J. Chang; Janet D. Cheng; Michael Wittekind; Susan E. Kiefer; Kevin Kish; Francis Y. Lee; Robert Borzillerri; Louis J. Lombardo; Dianlin Xie; Yaqun Zhang; Herbert E. Klei

Chronic myeloid leukemia (CML) is caused by the constitutively activated tyrosine kinase breakpoint cluster (BCR)-ABL. Current frontline therapy for CML is imatinib, an inhibitor of BCR-ABL. Although imatinib has a high rate of clinical success in early phase CML, treatment resistance is problematic, particularly in later stages of the disease, and is frequently mediated by mutations in BCR-ABL. Dasatinib (BMS-354825) is a multitargeted tyrosine kinase inhibitor that targets oncogenic pathways and is a more potent inhibitor than imatinib against wild-type BCR-ABL. It has also shown preclinical activity against all but one of the imatinib-resistant BCR-ABL mutants tested to date. Analysis of the crystal structure of dasatinib-bound ABL kinase suggests that the increased binding affinity of dasatinib over imatinib is at least partially due to its ability to recognize multiple states of BCR-ABL. The structure also provides an explanation for the activity of dasatinib against imatinib-resistant BCR-ABL mutants.


Journal of Medicinal Chemistry | 2008

Discovery of Pyrrolopyridine-Pyridone Based Inhibitors of Met Kinase : Synthesis, X-ray Crystallographic Analysis, and Biological Activities

Kyoung S. Kim; Liping Zhang; Robert J. Schmidt; Zhen-Wei Cai; Donna D. Wei; David K. Williams; Louis J. Lombardo; George L. Trainor; Dianlin Xie; Yaquan Zhang; Yongmi An; John S. Sack; John S. Tokarski; Celia D'Arienzo; Amrita Kamath; Punit Marathe; Yueping Zhang; Jonathan Lippy; Robert Jeyaseelan; Barri Wautlet; Benjamin Henley; Johnni Gullo-Brown; Veeraswamy Manne; John T. Hunt; Joseph Fargnoli; Robert M. Borzilleri

Conformationally constrained 2-pyridone analogue 2 is a potent Met kinase inhibitor with an IC50 value of 1.8 nM. Further SAR of the 2-pyridone based inhibitors of Met kinase led to potent 4-pyridone and pyridine N-oxide inhibitors such as 3 and 4. The X-ray crystallographic data of the inhibitor 2 bound to the ATP binding site of Met kinase protein provided insight into the binding modes of these inhibitors, and the SAR of this series of analogues was rationalized. Many of these analogues showed potent antiproliferative activities against the Met dependent GTL-16 gastric carcinoma cell line. Compound 2 also inhibited Flt-3 and VEGFR-2 kinases with IC50 values of 4 and 27 nM, respectively. It possesses a favorable pharmacokinetic profile in mice and demonstrates significant in vivo antitumor activity in the GTL-16 human gastric carcinoma xenograft model.


Biochemical Journal | 2011

Structural Basis for Carm1 Inhibition by Indole and Pyrazole Inhibitors

John S. Sack; Sandrine Thieffine; Tiziano Bandiera; Marina Fasolini; Gerald J. Duke; Lata Jayaraman; Kevin Kish; Herbert E. Klei; Ashok V. Purandare; Pamela Rosettani; Sonia Troiani; Dianlin Xie; Jay Aaron Bertrand

CARM1 (co-activator-associated arginine methyltransferase 1) is a PRMT (protein arginine N-methyltransferase) family member that catalyses the transfer of methyl groups from SAM (S-adenosylmethionine) to the side chain of specific arginine residues of substrate proteins. This post-translational modification of proteins regulates a variety of transcriptional events and other cellular processes. Moreover, CARM1 is a potential oncological target due to its multiple roles in transcription activation by nuclear hormone receptors and other transcription factors such as p53. Here, we present crystal structures of the CARM1 catalytic domain in complex with cofactors [SAH (S-adenosyl-L-homocysteine) or SNF (sinefungin)] and indole or pyazole inhibitors. Analysis of the structures reveals that the inhibitors bind in the arginine-binding cavity and the surrounding pocket that exists at the interface between the N- and C-terminal domains. In addition, we show using ITC (isothermal titration calorimetry) that the inhibitors bind to the CARM1 catalytic domain only in the presence of the cofactor SAH. Furthermore, sequence differences for select residues that interact with the inhibitors may be responsible for the CARM1 selectivity against PRMT1 and PRMT3. Together, the structural and biophysical information should aid in the design of both potent and specific inhibitors of CARM1.


Protein Science | 2008

Involvement of DPP‐IV catalytic residues in enzyme–saxagliptin complex formation

William Metzler; Joseph Yanchunas; Carolyn A. Weigelt; Kevin Kish; Herbert E. Klei; Dianlin Xie; Yaqun Zhang; Martin J. Corbett; James Tamura; Bin He; Lawrence G. Hamann; Mark S. Kirby; Jovita Marcinkeviciene

The inhibition of DPP‐IV by saxagliptin has been proposed to occur through formation of a covalent but reversible complex. To evaluate further the mechanism of inhibition, we determined the X‐ray crystal structure of the DPP‐IV:saxagliptin complex. This structure reveals covalent attachment between S630 and the inhibitor nitrile carbon (C–O distance <1.3 Å). To investigate whether this serine addition is assisted by the catalytic His‐Asp dyad, we generated two mutants of DPP‐IV, S630A and H740Q, and assayed them for ability to bind inhibitor. DPP‐IVH740Q bound saxagliptin with an ∼1000‐fold reduction in affinity relative to DPP‐IVWT, while DPP‐IVS630A showed no evidence for binding inhibitor. An analog of saxagliptin lacking the nitrile group showed unchanged binding properties to the both mutant proteins, highlighting the essential role S630 and H740 play in covalent bond formation between S630 and saxagliptin. Further supporting mechanism‐based inhibition by saxagliptin, NMR spectra of enzyme–saxagliptin complexes revealed the presence of three downfield resonances with low fractionation factors characteristic of short and strong hydrogen bonds (SSHB). Comparison of the NMR spectra of various wild‐type and mutant DPP‐IV:ligand complexes enabled assignment of a resonance at ∼14 ppm to H740. Two additional DPP‐IV mutants, Y547F and Y547Q, generated to probe potential stabilization of the enzyme–inhibitor complex by this residue, did not show any differences in inhibitor binding either by ITC or NMR. Together with the previously published enzymatic data, the structural and binding data presented here strongly support a histidine‐assisted covalent bond formation between S630 hydroxyl oxygen and the nitrile group of saxagliptin.


Journal of Medicinal Chemistry | 2014

Discovery and Preclinical Characterization of the Cyclopropylindolobenzazepine BMS-791325, A Potent Allosteric Inhibitor of the Hepatitis C Virus NS5B Polymerase.

Robert G. Gentles; Min Ding; John A. Bender; Carl P. Bergstrom; Katharine A. Grant-Young; Piyasena Hewawasam; Thomas William Hudyma; Scott Martin; Andrew Nickel; Alicia Regueiro-Ren; Yong Tu; Zhong Yang; Kap-Sun Yeung; Xiaofan Zheng; Sam T. Chao; Jung-Hui Sun; Brett R. Beno; Daniel M. Camac; Mian Gao; Paul E. Morin; Steven Sheriff; Jeff Tredup; John Wan; Mark R. Witmer; Dianlin Xie; Umesh Hanumegowda; Jay O. Knipe; Kathy Mosure; Kenneth S. Santone; Dawn D. Parker

Described herein are structure-activity relationship studies that resulted in the optimization of the activity of members of a class of cyclopropyl-fused indolobenzazepine HCV NS5B polymerase inhibitors. Subsequent iterations of analogue design and syntheses successfully addressed off-target activities, most notably human pregnane X receptor (hPXR) transactivation, and led to significant improvements in the physicochemical properties of lead compounds. Those analogues exhibiting improved solubility and membrane permeability were shown to have notably enhanced pharmacokinetic profiles. Additionally, a series of alkyl bridged piperazine carboxamides was identified as being of particular interest, and from which the compound BMS-791325 (2) was found to have distinguishing antiviral, safety, and pharmacokinetic properties that resulted in its selection for clinical evaluation.


Journal of Medicinal Chemistry | 2010

Discovery of 6-(Aminomethyl)-5-(2,4-dichlorophenyl)-7-methylimidazo[1,2-a]pyrimidine-2-carboxamides as Potent, Selective Dipeptidyl Peptidase-4 (DPP4) Inhibitors.

Wei Meng; Robert Paul Brigance; Hannguang J. Chao; Aberra Fura; Thomas Harrity; Jovita Marcinkeviciene; Stephen P. O'connor; James Tamura; Dianlin Xie; Yaqun Zhang; Herbert E. Klei; Kevin Kish; Carolyn Weigelt; Huji Turdi; Aiying Wang; Robert Zahler; Mark S. Kirby; Lawrence G. Hamann

Continued structure-activity relationship (SAR) exploration within our previously disclosed azolopyrimidine containing dipeptidyl peptidase-4 (DPP4) inhibitors led us to focus on an imidazolopyrimidine series in particular. Further study revealed that by replacing the aryl substitution on the imidazole ring with a more polar carboxylic ester or amide, these compounds displayed not only increased DPP4 binding activity but also significantly reduced human ether-a-go-go related gene (hERG) and sodium channel inhibitory activities. Additional incremental adjustment of polarity led to permeable molecules which exhibited favorable pharmacokinetic (PK) profiles in preclinical animal species. The active site binding mode of these compounds was determined by X-ray crystallography as exemplified by amide 24c. A subsequent lead molecule from this series, (+)-6-(aminomethyl)-5-(2,4-dichlorophenyl)-N-(1-ethyl-1H-pyrazol-5-yl)-7-methylimidazo[1,2-a]pyrimidine-2-carboxamide (24s), emerged as a potent, selective DPP4 inhibitor that displayed excellent PK profiles and in vivo efficacy in ob/ob mice.


Journal of Biological Chemistry | 2015

Tyrosine Kinase 2-mediated Signal Transduction in T Lymphocytes Is Blocked by Pharmacological Stabilization of Its Pseudokinase Domain

John S. Tokarski; Adriana Zupa-Fernandez; Jeffrey Tredup; Kristen Pike; Chiehying Chang; Dianlin Xie; Lihong Cheng; Donna L. Pedicord; Jodi K. Muckelbauer; Stephen R. Johnson; Sophie Wu; Suzanne C. Edavettal; Yang Hong; Mark R. Witmer; Lisa Elkin; Yuval Blat; William J. Pitts; David S. Weinstein; James R. Burke

Background: Interleukin-23 mediates pathobiology in many autoimmune disorders. Results: A chemogenomics approach identified small molecule agents that block receptor-mediated activation or tyrosine kinase 2 (Tyk2) and downstream signaling. Compounds stabilize the pseudokinase domain of Tyk2. Conclusion: Small molecule ligands of the Tyk2 pseudokinase domain stabilize an autoinhibitory interaction with the catalytic domain. Significance: This work enables the discovery of selective therapeutics targeting Tyk2-dependent pathways critical in autoimmunity. Inhibition of signal transduction downstream of the IL-23 receptor represents an intriguing approach to the treatment of autoimmunity. Using a chemogenomics approach marrying kinome-wide inhibitory profiles of a compound library with the cellular activity against an IL-23-stimulated transcriptional response in T lymphocytes, a class of inhibitors was identified that bind to and stabilize the pseudokinase domain of the Janus kinase tyrosine kinase 2 (Tyk2), resulting in blockade of receptor-mediated activation of the adjacent catalytic domain. These Tyk2 pseudokinase domain stabilizers were also shown to inhibit Tyk2-dependent signaling through the Type I interferon receptor but not Tyk2-independent signaling and transcriptional cellular assays, including stimulation through the receptors for IL-2 (JAK1- and JAK3-dependent) and thrombopoietin (JAK2-dependent), demonstrating the high functional selectivity of this approach. A crystal structure of the pseudokinase domain liganded with a representative example showed the compound bound to a site analogous to the ATP-binding site in catalytic kinases with features consistent with high ligand selectivity. The results support a model where the pseudokinase domain regulates activation of the catalytic domain by forming receptor-regulated inhibitory interactions. Tyk2 pseudokinase stabilizers, therefore, represent a novel approach to the design of potent and selective agents for the treatment of autoimmunity.


Chemical Biology & Drug Design | 2011

X-Ray Crystal Structure of Bone Marrow Kinase in the X Chromosome: A Tec Family Kinase

Jodi K. Muckelbauer; John S. Sack; Nazia Ahmed; James R. Burke; ChiehYing Y. Chang; Mian Gao; Joseph A. Tino; Dianlin Xie; Andrew J. Tebben

Bone marrow kinase in the X chromosome, a member of the Tec family of tyrosine kinases, plays a role in both monocyte/macrophage trafficking as well as cytokine secretion. Although the structures of Tec family kinases Bruton’s tyrosine kinase and IL‐2‐inducible T‐cell kinase are known, the crystal structures of other Tec family kinases have remained elusive. We report the X‐ray crystal structures of bone marrow kinase in the X chromosome in complex with dasatinib at 2.4 Å resolution and PP2 at 1.9 Å resolution. The bone marrow kinase in the X chromosome structures reveal a typical kinase protein fold; with well‐ordered protein conformation that includes an open/extended activation loop and a stabilized DFG‐motif rendering the kinase in an inactive conformation. Dasatinib and PP2 bind to bone marrow kinase in the X chromosome in the ATP binding pocket and display similar binding modes to that observed in other Tec and Src protein kinases. The bone marrow kinase in the X chromosome structures identify conformational elements of the DFG‐motif that could potentially be utilized to design potent and/or selective bone marrow kinase in the X chromosome inhibitors.


Acta Crystallographica Section D-biological Crystallography | 2008

Structural basis for the high-affinity binding of pyrrolotriazine inhibitors of p38 MAP kinase

John S. Sack; Kevin Kish; Matthew E. Pokross; Dianlin Xie; Gerald J. Duke; Jeffrey Tredup; Susan E. Kiefer; John A. Newitt

The crystal structure of unphosphorylated p38alpha MAP kinase complexed with a representative pyrrolotriazine-based inhibitor led to the elucidation of the high-affinity binding mode of this class of compounds at the ATP-binding site. The ligand binds in an extended conformation, with one end interacting with the adenine-pocket hinge region, including a hydrogen bond from the carboxyl O atom of Met109. The other end of the ligand interacts with the hydrophobic pocket of the binding site and with the backbone N atom of Asp168 in the DFG activation loop. Addition of an extended benzylmorpholine group forces the DFG loop to flip out of position and allows the ligand to make additional interactions with the protein.


Acta Crystallographica Section D-biological Crystallography | 2016

Crystal structures of apo and inhibitor-bound TGFβR2 kinase domain: insights into TGFβR isoform selectivity

Andrew J. Tebben; Maxim Ruzanov; Mian Gao; Dianlin Xie; Susan E. Kiefer; Chunhong Yan; John A. Newitt; Liping Zhang; Kyoung S. Kim; Hao Lu; Lisa M. Kopcho; Steven Sheriff

The cytokine TGF-β modulates a number of cellular activities and plays a critical role in development, hemostasis and physiology, as well as in diseases including cancer and fibrosis. TGF-β signals through two transmembrane serine/threonine kinase receptors: TGFβR1 and TGFβR2. Multiple structures of the TGFβR1 kinase domain are known, but the structure of TGFβR2 remains unreported. Wild-type TGFβR2 kinase domain was refractory to crystallization, leading to the design of two mutated constructs: firstly, a TGFβR1 chimeric protein with seven ATP-site residues mutated to their counterparts in TGFβR2, and secondly, a reduction of surface entropy through mutation of six charged residues on the surface of the TGFβR2 kinase domain to alanines. These yielded apo and inhibitor-bound crystals that diffracted to high resolution (<2 Å). Comparison of these structures with those of TGFβR1 reveal shared ligand contacts as well as differences in the ATP-binding sites, suggesting strategies for the design of pan and selective TGFβR inhibitors.

Collaboration


Dive into the Dianlin Xie'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

Mian Gao

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge