41 results on '"Sergei Timofeevski"'
Search Results
2. Mitotic Checkpoint Kinase Mps1 Has a Role in Normal Physiology which Impacts Clinical Utility.
- Author
-
Ricardo Martinez, Alessandra Blasina, Jill F Hallin, Wenyue Hu, Isha Rymer, Jeffery Fan, Robert L Hoffman, Sean Murphy, Matthew Marx, Gina Yanochko, Dusko Trajkovic, Dac Dinh, Sergei Timofeevski, Zhou Zhu, Peiquing Sun, Patrick B Lappin, and Brion W Murray
- Subjects
Medicine ,Science - Abstract
Cell cycle checkpoint intervention is an effective therapeutic strategy for cancer when applied to patients predisposed to respond and the treatment is well-tolerated. A critical cell cycle process that could be targeted is the mitotic checkpoint (spindle assembly checkpoint) which governs the metaphase-to-anaphase transition and insures proper chromosomal segregation. The mitotic checkpoint kinase Mps1 was selected to explore whether enhancement in genomic instability is a viable therapeutic strategy. The basal-a subset of triple-negative breast cancer was chosen as a model system because it has a higher incidence of chromosomal instability and Mps1 expression is up-regulated. Depletion of Mps1 reduces tumor cell viability relative to normal cells. Highly selective, extremely potent Mps1 kinase inhibitors were created to investigate the roles of Mps1 catalytic activity in tumor cells and normal physiology (PF-7006, PF-3837; Ki
- Published
- 2015
- Full Text
- View/download PDF
3. Design and Synthesis of Functionally Active 5-Amino-6-Aryl Pyrrolopyrimidine Inhibitors of Hematopoietic Progenitor Kinase 1
- Author
-
Rebecca A. Gallego, Louise Bernier, Hui Chen, Sujin Cho-Schultz, Loanne Chung, Michael Collins, Matthew Del Bel, Jeff Elleraas, Cinthia Costa Jones, Ciaran N. Cronin, Martin Edwards, Xu Fang, Timothy Fisher, Mingying He, Jacqui Hoffman, Ruiduan Huo, Mehran Jalaie, Eric Johnson, Ted W. Johnson, Robert S. Kania, Manfred Kraus, Jennifer Lafontaine, Phuong Le, Tongnan Liu, Michael Maestre, Jean Matthews, Michele McTigue, Nichol Miller, Qiming Mu, Xulong Qin, Shijian Ren, Paul Richardson, Allison Rohner, Neal Sach, Li Shao, Graham Smith, Ruirui Su, Bin Sun, Sergei Timofeevski, Phuong Tran, Shuiwang Wang, Wei Wang, Ru Zhou, Jinjiang Zhu, and Sajiv K. Nair
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Tables from Sequential ALK Inhibitors Can Select for Lorlatinib-Resistant Compound ALK Mutations in ALK-Positive Lung Cancer
- Author
-
Alice T. Shaw, Aaron N. Hata, Ted W. Johnson, Jeffrey A. Engelman, Mari Mino-Kenudson, Jochen K. Lennerz, Cyril H. Benes, A. John Iafrate, Rebecca S. Heist, Daria Timonina, Krystina E. Kattermann, Amanda K. Riley, Lorin A. Ferris, Justin F. Gainor, Harper Hubbeling, Sergei Timofeevski, Ibiayi Dagogo-Jack, Kylie Prutisto-Chang, Leila Dardaei, Adam Langenbucher, Luc Friboulet, Benjamin J. Burke, Michael S. Lawrence, Jessica J. Lin, and Satoshi Yoda
- Abstract
suppl tables
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Methods from Sequential ALK Inhibitors Can Select for Lorlatinib-Resistant Compound ALK Mutations in ALK-Positive Lung Cancer
- Author
-
Alice T. Shaw, Aaron N. Hata, Ted W. Johnson, Jeffrey A. Engelman, Mari Mino-Kenudson, Jochen K. Lennerz, Cyril H. Benes, A. John Iafrate, Rebecca S. Heist, Daria Timonina, Krystina E. Kattermann, Amanda K. Riley, Lorin A. Ferris, Justin F. Gainor, Harper Hubbeling, Sergei Timofeevski, Ibiayi Dagogo-Jack, Kylie Prutisto-Chang, Leila Dardaei, Adam Langenbucher, Luc Friboulet, Benjamin J. Burke, Michael S. Lawrence, Jessica J. Lin, and Satoshi Yoda
- Abstract
Supplementary Methods include Enzyme Kinetic Assays and Computational Methods.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figures from Sequential ALK Inhibitors Can Select for Lorlatinib-Resistant Compound ALK Mutations in ALK-Positive Lung Cancer
- Author
-
Alice T. Shaw, Aaron N. Hata, Ted W. Johnson, Jeffrey A. Engelman, Mari Mino-Kenudson, Jochen K. Lennerz, Cyril H. Benes, A. John Iafrate, Rebecca S. Heist, Daria Timonina, Krystina E. Kattermann, Amanda K. Riley, Lorin A. Ferris, Justin F. Gainor, Harper Hubbeling, Sergei Timofeevski, Ibiayi Dagogo-Jack, Kylie Prutisto-Chang, Leila Dardaei, Adam Langenbucher, Luc Friboulet, Benjamin J. Burke, Michael S. Lawrence, Jessica J. Lin, and Satoshi Yoda
- Abstract
Supplementary Figures include Figure S1-S9.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Tables 1-2, Figures 1-2 from Sensitivity of Selected Human Tumor Models to PF-04217903, a Novel Selective c-Met Kinase Inhibitor
- Author
-
James G. Christensen, Tod Smeal, Steven L. Bender, Gerrit Los, Michele McTigue, Jingrong Jean Cui, Sergei Timofeevski, Max Parker, Shinji Yamazaki, Lars D. Engstrom, Ming Qiu, Kristina Burgess, Maria E. Arango, Joseph H. Lee, Qiuhua Li, and Helen Y. Zou
- Abstract
PDF file - 44K
- Published
- 2023
- Full Text
- View/download PDF
8. 1367 A highly-selective HPK1 inhibitor enhances T cell receptor signaling and T cell activation potential, increasing antigen recognition and efficacy of PD-1 therapy
- Author
-
Andrew Nager, David Schaer, Rebecca Gallego, Eleanore Hendrickson, Sergei Timofeevski, Manqing Li, Allison Rohner, Ruth Seelige, Samuel Stoner, Christopher Dillon, Paulina Cuenca, Cinthia Jones, Sarah Firdaus, Robert Amezquita, Clifford Restaino, Derek Bartlett, and Timothy Fisher
- Published
- 2022
- Full Text
- View/download PDF
9. Characterization of Specific N-α-Acetyltransferase 50 (Naa50) Inhibitors Identified Using a DNA Encoded Library
- Author
-
Paul G. Richardson, Xiaoyun Meng, Karen A. Maegley, A.E. Stewart, Pei-Pei Kung, Jose L Montano, Ya-Li Deng, Jinqiao Wan, Michael R. Gehring, Jordan L. Meier, Brigitte S Naughton, Stephan Grant, Dou Dengfeng, Chakrapani Subramanyam, Anthony R. Harris, Samantha Elizabeth Greasley, Barry A. Morgan, Wen Yan, Xuemin Cheng, Prakash B. Palde, William K Sonnenburg, Junli Feng, Chen Qiuxia, Thomas A Paul, Gary M. Gallego, Sylvie K. Sakata, Sergei Timofeevski, Patrick Bingham, Benjamin J. Burke, and Alex Shaginian
- Subjects
chemistry.chemical_classification ,010405 organic chemistry ,Chemistry ,Stereochemistry ,Organic Chemistry ,Target engagement ,Substrate (chemistry) ,01 natural sciences ,Biochemistry ,Cocrystal ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Enzyme ,Mechanism of action ,Acetyltransferase ,Drug Discovery ,medicine ,A-DNA ,medicine.symptom ,Selectivity - Abstract
[Image: see text] Two novel compounds were identified as Naa50 binders/inhibitors using DNA-encoded technology screening. Biophysical and biochemical data as well as cocrystal structures were obtained for both compounds (3a and 4a) to understand their mechanism of action. These data were also used to rationalize the binding affinity differences observed between the two compounds and a MLGP peptide-containing substrate. Cellular target engagement experiments further confirm the Naa50 binding of 4a and demonstrate its selectivity toward related enzymes (Naa10 and Naa60). Additional analogs of inhibitor 4a were also evaluated to study the binding mode observed in the cocrystal structures.
- Published
- 2020
- Full Text
- View/download PDF
10. 737 Inhibition of P21-activated kinase 4 (PAK4) reverts immune exclusion and restores anti-tumor immunity in the tumor microenvironment
- Author
-
Jonathan R. Heyen, Brandy Chavez, Sergei Timofeevski, Murali Gururajan, Keith A. Ching, Szu-Yu Tang, Eleanore Hendrickson, Rui Eugene Yuanjin, Shawn D. Doran, Stephanie T. Shi, Gina Chu, Christopher P. Dillon, Jennifer Kinong, Yu 'Jerry' Zhou, Jon Oyer, Andrew R. Nager, Vinayak Rayannavar, Rajarshi Bhadra, Johnni Gullo-Brown, and Indrawan James Mcalpine
- Subjects
Pharmacology ,Cancer Research ,Tumor microenvironment ,Cell growth ,medicine.medical_treatment ,Immunology ,Wnt signaling pathway ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,chemistry.chemical_compound ,Immune system ,Cytokine ,Oncology ,chemistry ,Cancer cell ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,CXCL10 ,Growth inhibition ,RC254-282 - Abstract
BackgroundP21-activated kinase 4 (PAK4) is a serine/threonine protein kinase that is mostly expressed in tumor and stroma cells. PAK4 activates tumor WNT/β-catenin pathway and regulates cellular morphology, motility, EMT, cell proliferation and survival. Recent studies also showed that PAK4 can actively exclude T cells from tumors, suggesting that therapeutic inhibition of PAK4 can increase T cell infiltration in tumor microenvironment and overcome resistance to checkpoint inhibitor immunotherapy.1MethodsWe generated PAK4 knockout (KO) clones in human and mouse tumor cells to validate its biology in vitro and in vivo. We also performed pharmacological evaluation of PAK4 inhibition using Pfizer compounds (referred to as 'PAK4i compounds' below) for their potential tumor-intrinsic and immune-regulatory roles.ResultsNanostring, qPCR and RNASeq analysis showed that PAK4 depletion led to increase of cytokine expression in tumor, including conventional dendritic cell (cDC)- recruiting chemokine CCL4, and type I IFN / ISG pathway genes that are associated with MHC upregulation such as CXCL10. In addition, PAK4 KO sensitizes B16F10 tumors to anti-PD-1 treatment and increases infiltration of cDC and T cells in the tumor microenvironment.We also showed that small molecule PAK4i compounds induced more potent cancer cell growth inhibition over treated normal PBMCs. PAK4i compounds also increased immune-activating and decreased immune exclusion genes in B16F10 cells and tumor explants in vitro. Although PAK4 target engagement is demonstrated by CETSA assay, the compound potency on modulating PAK4 downstream Wnt/ β-catenin pathway is low, suggesting that the aforementioned phenotypic changes induced by PAK4i compounds may be partially attributed to other off-target effects.ConclusionsCollectively, our data suggests that genetic depletion or pharmacological inhibition of PAK4 may induce immune-activating cytokine production in tumor cells, revert immune cell exclusion in tumor microenvironment, and synergize with checkpoint blockade therapies. However, further optimization on these PAK4i compounds is needed to improve its specificity on modulating PAK4 enzyme activities.ReferenceAbril-Rodriguez G, Torrejon DY, Liu W, Zaretsky JM, Nowicki TS, Tsoi J, Puig-Saus C, Baselga-Carretero I, Medina E, Quist MJ, Garcia AJ, Senapedis W, Baloglu E, Kalbasi A, Cheung-Lau G, Berent-Maoz B, Comin-Anduix B, Hu-Lieskovan S, CWang CY, Grasso CS & Ribas A. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat Cancer 2020;1:46–58.Ethics ApprovalAll animal studies were conducted in accordance with protocols approved by the Institutional Animal Care and Use Committee of Pfizer. Approved protocol # LAJ-2019-01347
- Published
- 2021
11. Abstract 330: Engineering electrophile-sensitive kinase mutants to accelerate oncology target validation
- Author
-
Romelia Salomon-Ferrer, Lawrence G. Lum, Todd VanArsdale, Nathan V. Lee, Andrew C. Wang, Aihua Zou, Koleen Eisele, Eric C. Greenwald, Catherine M. Ambler, Ana Flores-Bojorquez, Bryan Li, Carl Davis, Sergei Timofeevski, Martha A. Ornelas, Zhenxiong Wang, Penney L. Khamphavong, Samantha Elizabeth Greasley, Indrawan James Mcalpine, Michael F. Maestre, Jian Li, Ben Bolaños, Neil B. Grodsky, Yun Huang, Stephanie Scales, Ted William Johnson, Roksolana Melnychuk, Wade Diehl, Jon Oyer, Jacob DeForest, and Sherry Niessen
- Subjects
Cancer Research ,Oncology ,Biochemistry ,Chemistry ,Kinase ,Mutant ,Electrophile - Abstract
Nomination of new oncology targets has been greatly aided by advances in genetic screening and profiling, but developing potent, selective small molecule inhibitors against these targets remains a resource intensive pursuit. To significantly de-risk this process we applied a chemical biology strategy to model pharmacological inhibition of the MASTL kinase and showed that selective enzymatic inhibition accurately mimicked targeted genetic perturbation. Specifically, we engineered an electrophile-sensitive version of MASTL through a single amino acid substitution, Asp117Cys, within the ATP-binding site hinge region. Only 11 human kinases contain a cysteine handle in this particular hinge position (H10), which drastically limits the scope of off-targets prone to any H10 Cys-selective covalent inhibitors but also demonstrates that an H10 Cys is not incompatible with kinase activity. Furthermore, endogenous H10 Cys kinases include several targets of successful covalent inhibitor development campaigns (e.g. EGFR, JAK3 and BTK). This broad availability of optimized compounds allowed us to quickly screen and identify potent inhibitors of the MASTL Asp117Cys recombinant mutant. MASTL overexpression is observed across a broad spectrum of solid tumors, so to directly examine how its inhibition would impact cancer cell growth, Asp117Cys was created in the endogenous MASTL gene through CRISPR-directed gene editing. Pancreatic cancer cells homozygous for MASTL Asp117Cys showed clear dose-dependent growth inhibition when treated with a T790M mutant-specific EGFR covalent inhibitor (PF-06459988). Moreover, inhibitor treatment induced a visible phenotype of large multi-nucleated cells that phenocopies genetic perturbation of MASTL and aligns with MASTL's functional role regulating mitotic division. These inhibitor induced effects were not observed in unmodified parental cells and affinity purification experiments utilizing an alkyne probe of PF-06459988 confirmed binding specifically in engineered Asp117Cys mutant cells. When these Asp117Cys mutant cells are propagated as mouse xenografts, their in vivo tumor growth rate is indistinguishable from parental, but PF-06459988 treatment selectively induces tumor growth inhibition of mutant cells, thus demonstrating that selective enzymatic inhibition is sufficient to drive anti-tumor effects. Herein our application of an electrophile-sensitive mutant demonstrates how existing, optimized covalent small molecule inhibitors can be repurposed as chemical probes against engineered kinase domains. This engineered system provides a valuable orthogonal benchmark in advance of a drug discovery campaign, reveals target-dependent biology, and is likely to translate to additional kinase targets due to structural conservation within this enzymatic domain. Citation Format: Jon A. Oyer, Ted W. Johnson, Andrew C. Wang, Michael F. Maestre, Ana Flores-Bojorquez, Roksolana Melnychuk, Sergei Timofeevski, Sherry Niessen, Zhenxiong Wang, Jian Li, Wade C. Diehl, Koleen J. Eisele, Nathan V. Lee, Aihua Zou, Carl Davis, Eric C. Greenwald, Jacob DeForest, Martha Ornelas, Bryan Li, Stephanie Scales, Penney L. Khamphavong, Catherine M. Ambler, Yun Huang, Romelia Salomon-Ferrer, Samantha E. Greasley, Ben Bolanos, Neil Grodsky, Lawrence Lum, Todd L. VanArsdale, Indrawan J. McAlpine. Engineering electrophile-sensitive kinase mutants to accelerate oncology target validation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 330.
- Published
- 2021
- Full Text
- View/download PDF
12. Multiple conformational states of the HPK1 kinase domain in complex with sunitinib reveal the structural changes accompanying HPK1 trans-regulation
- Author
-
Sergei Timofeevski, Sansana Sawasdikosol, Timothy Scott Fisher, Steven J. Burakoff, Robert Steven Kania, Ted William Johnson, Gallego Rebecca Anne, Ciarán N. Cronin, Eric Johnson, Michele McTigue, and Michael F. Maestre
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,T-Lymphocytes ,Serine threonine protein kinase ,Molecular Dynamics Simulation ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Biochemistry ,03 medical and health sciences ,Adenosine Triphosphate ,medicine ,Sunitinib ,Humans ,Phosphorylation ,Molecular Biology ,Binding Sites ,030102 biochemistry & molecular biology ,Kinase ,Chemistry ,Cell Biology ,Recombinant Proteins ,Cell biology ,Protein Structure, Tertiary ,030104 developmental biology ,Protein kinase domain ,Accelerated Communications ,Mutagenesis, Site-Directed ,Interleukin-2 ,Signal transduction ,Tyrosine kinase ,Dimerization ,medicine.drug - Abstract
Hematopoietic progenitor kinase 1 (HPK1 or MAP4K1) is a Ser/Thr kinase that operates via the c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) signaling pathways to dampen the T-cell response and antitumor immunity. Accordingly, selective HPK1 inhibition is considered a means to enhance antitumor immunity. Sunitinib, a multi-receptor tyrosine kinase (RTK) inhibitor approved for the management of gastrointestinal stromal tumors (GISTs), renal cell carcinoma (RCC), and pancreatic cancer, has been reported to inhibit HPK1 in vitro. In this report, we describe the crystal structures of the native HPK1 kinase domain in both nonphosphorylated and doubly phosphorylated states, in addition to a double phosphomimetic mutant (T165E,S171E), each complexed with sunitinib at 2.17–3.00-Å resolutions. The native nonphosphorylated cocrystal structure revealed an inactive dimer in which the activation loop of each monomer partially occupies the ATP- and substrate-binding sites of the partner monomer. In contrast, the structure of the protein with a doubly phosphorylated activation loop exhibited an active kinase conformation with a greatly reduced monomer–monomer interface. Conversely, the phosphomimetic mutant cocrystal structure disclosed an alternative arrangement in which the activation loops are in an extended domain-swapped configuration. These structural results indicate that HPK1 is a highly dynamic kinase that undergoes trans-regulation via dimer formation and extensive intramolecular and intermolecular remodeling of the activation segment.
- Published
- 2019
13. Reviving B-Factors: Activating ALK Mutations Increase Protein Dynamics of the Unphosphorylated Kinase
- Author
-
Dan Gehlhaar, Alexei Brooun, Mehran Jalaie, Michele McTigue, Sergei Timofeevski, Ben Bolaños, Gallego Rebecca Anne, and Ted William Johnson
- Subjects
0301 basic medicine ,Mutation ,biology ,Chemistry ,Kinase ,Organic Chemistry ,Mutant ,Wild type ,medicine.disease_cause ,Biochemistry ,Receptor tyrosine kinase ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Protein kinase domain ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,Drug Discovery ,biology.protein ,medicine ,Anaplastic lymphoma kinase ,Phosphorylation - Abstract
[Image: see text] Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that can become oncogenic by activating mutations or overexpression. Full kinetic characterization of both phosphorylated and nonphosphorylated wildtype and mutant ALK kinase domain was done. Our structure-based drug design programs directed at ALK allowed us to interrogate whether X-ray crystallography data could be used to support the hypothesis that activation of ALK by mutation occurs due to increased protein dynamics. Crystallographic B-factors were converted to normalized B-factors, which allowed analysis of wildtype ALK, ALK-C1156Y, and ALK-L1196M. This data suggests that mobility of the P-loop, αC-helix, and activation loop (A-loop) may be important in catalytic activity increases, with or without phosphorylation. Both molecular dynamics simulations and hydrogen–deuterium exchange experimental data corroborated the normalized B-factors data.
- Published
- 2018
14. PF-06463922, an ALK/ROS1 Inhibitor, Overcomes Resistance to First and Second Generation ALK Inhibitors in Preclinical Models
- Author
-
Wenyue Hu, Rosa L. Frias, Hovhannes J. Gukasyan, Alice T. Shaw, Valeria Fantin, Ryohei Katayama, Nathan V. Lee, Ruth W. Tang, Timothy Affolter, Eugene Lifshits, Ted William Johnson, Divya Bezwada, David P. Kodack, Lars D. Engstrom, Hieu Lam, Sidra Mahmood, Tod Smeal, Luc Friboulet, Rakesh K. Jain, Hui Wang, Melissa West, Dac M. Dinh, Bhushankumar Patel, Qiuhua Li, Konstantinos Tsaparikos, Helen Y. Zou, Justine L. Lam, Sergei Timofeevski, Shinji Yamazaki, Patrick B. Lappin, Justin F. Gainor, Shibing Deng, and Jinwei Wang
- Subjects
Alectinib ,Cancer Research ,Mutation ,Brigatinib ,Ceritinib ,business.industry ,Cell Biology ,Drug resistance ,Pharmacology ,medicine.disease_cause ,Lorlatinib ,3. Good health ,Oncology ,hemic and lymphatic diseases ,medicine ,ROS1 ,Anaplastic lymphoma kinase ,business ,medicine.drug - Abstract
SummaryWe report the preclinical evaluation of PF-06463922, a potent and brain-penetrant ALK/ROS1 inhibitor. Compared with other clinically available ALK inhibitors, PF-06463922 displayed superior potency against all known clinically acquired ALK mutations, including the highly resistant G1202R mutant. Furthermore, PF-06463922 treatment led to regression of EML4-ALK-driven brain metastases, leading to prolonged mouse survival, in a superior manner. Finally, PF-06463922 demonstrated high selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 will be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors because of secondary ALK kinase domain mutations and/or brain metastases.
- Published
- 2015
- Full Text
- View/download PDF
15. PF-06463922 is a potent and selective next-generation ROS1/ALK inhibitor capable of blocking crizotinib-resistant ROS1 mutations
- Author
-
Melissa West, Victoria A. Appleman, Valeria Fantin, Michele McTigue, Sergei Timofeevski, Matthew D. Falk, Alexei Brooun, Scott Rp McDonnell, Tod Smeal, Al Charest, Wenyue Hu, Wei Liu, Ya-Li Deng, Katy A. Wong, Timothy C. Nichols, Justine L. Lam, Ping Jiang, Helen Y. Zou, Qiuhua Li, Lars D. Engstrom, Timothy Affolter, Ted William Johnson, and Patrick B. Lappin
- Subjects
Models, Molecular ,Alectinib ,Lactams ,Carcinogenesis ,Pyridines ,medicine.drug_class ,Lactams, Macrocyclic ,Aminopyridines ,Biology ,Crystallography, X-Ray ,Mice ,Crizotinib ,Proto-Oncogene Proteins ,ROS1 ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Cell Proliferation ,Multidisciplinary ,Ceritinib ,Kinase ,Point mutation ,Glioma ,Protein-Tyrosine Kinases ,Biological Sciences ,Lorlatinib ,ALK inhibitor ,Disease Models, Animal ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Pyrazoles ,Signal Transduction ,medicine.drug - Abstract
Oncogenic c-ros oncogene1 (ROS1) fusion kinases have been identified in a variety of human cancers and are attractive targets for cancer therapy. The MET/ALK/ROS1 inhibitor crizotinib (Xalkori, PF-02341066) has demonstrated promising clinical activity in ROS1 fusion-positive non-small cell lung cancer. However, emerging clinical evidence has shown that patients can develop resistance by acquiring secondary point mutations in ROS1 kinase. In this study we characterized the ROS1 activity of PF-06463922, a novel, orally available, CNS-penetrant, ATP-competitive small-molecule inhibitor of ALK/ROS1. In vitro, PF-06463922 exhibited subnanomolar cellular potency against oncogenic ROS1 fusions and inhibited the crizotinib-refractory ROS1(G2032R) mutation and the ROS1(G2026M) gatekeeper mutation. Compared with crizotinib and the second-generation ALK/ROS1 inhibitors ceritinib and alectinib, PF-06463922 showed significantly improved inhibitory activity against ROS1 kinase. A crystal structure of the PF-06463922-ROS1 kinase complex revealed favorable interactions contributing to the high-affinity binding. In vivo, PF-06463922 showed marked antitumor activity in tumor models expressing FIG-ROS1, CD74-ROS1, and the CD74-ROS1(G2032R) mutation. Furthermore, PF-06463922 demonstrated antitumor activity in a genetically engineered mouse model of FIG-ROS1 glioblastoma. Taken together, our results indicate that PF-06463922 has potential for treating ROS1 fusion-positive cancers, including those requiring agents with CNS-penetrating properties, as well as for overcoming crizotinib resistance driven by ROS1 mutation.
- Published
- 2015
- Full Text
- View/download PDF
16. Sequential ALK Inhibitors Can Select for Lorlatinib-Resistant Compound ALK Mutations in ALK-Positive Lung Cancer
- Author
-
Alice T. Shaw, Luc Friboulet, Daria Timonina, Benjamin J. Burke, Krystina E. Kattermann, Justin F. Gainor, Jeffrey A. Engelman, Amanda K. Riley, A. John Iafrate, Jochen K. Lennerz, Adam Langenbucher, Sergei Timofeevski, Aaron N. Hata, Leila Dardaei, Cyril H. Benes, Lorin A. Ferris, Jessica J. Lin, Kylie Prutisto-Chang, Ted William Johnson, Harper Hubbeling, Rebecca S. Heist, Ibiayi Dagogo-Jack, Mari Mino-Kenudson, Satoshi Yoda, and Michael S. Lawrence
- Subjects
0301 basic medicine ,Mutation ,medicine.drug_class ,Mutagenesis (molecular biology technique) ,Cancer ,Drug resistance ,Biology ,medicine.disease ,medicine.disease_cause ,Lorlatinib ,Article ,3. Good health ,ALK inhibitor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Lung cancer ,Exome sequencing - Abstract
The cornerstone of treatment for advanced ALK-positive lung cancer is sequential therapy with increasingly potent and selective ALK inhibitors. The third-generation ALK inhibitor lorlatinib has demonstrated clinical activity in patients who failed previous ALK inhibitors. To define the spectrum of ALK mutations that confer lorlatinib resistance, we performed accelerated mutagenesis screening of Ba/F3 cells expressing EML4–ALK. Under comparable conditions, N-ethyl-N-nitrosourea (ENU) mutagenesis generated numerous crizotinib-resistant but no lorlatinib-resistant clones harboring single ALK mutations. In similar screens with EML4–ALK containing single ALK resistance mutations, numerous lorlatinib-resistant clones emerged harboring compound ALK mutations. To determine the clinical relevance of these mutations, we analyzed repeat biopsies from lorlatinib-resistant patients. Seven of 20 samples (35%) harbored compound ALK mutations, including two identified in the ENU screen. Whole-exome sequencing in three cases confirmed the stepwise accumulation of ALK mutations during sequential treatment. These results suggest that sequential ALK inhibitors can foster the emergence of compound ALK mutations, identification of which is critical to informing drug design and developing effective therapeutic strategies. Significance: Treatment with sequential first-, second-, and third-generation ALK inhibitors can select for compound ALK mutations that confer high-level resistance to ALK-targeted therapies. A more efficacious long-term strategy may be up-front treatment with a third-generation ALK inhibitor to prevent the emergence of on-target resistance. Cancer Discov; 8(6); 714–29. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 663
- Published
- 2018
- Full Text
- View/download PDF
17. Engineering of an isolated p110α subunit of PI3Kα permits crystallization and provides a platform for structure-based drug design
- Author
-
Matthew D. Falk, Simon Bergqvist, Ya-Li Deng, Ping Chen, Sergei Timofeevski, Alexei Brooun, and Wei Liu
- Subjects
Structural similarity ,Stereochemistry ,Chemistry ,Protein subunit ,Lipid kinase activity ,Isothermal titration calorimetry ,Binding site ,P110α ,Molecular Biology ,Biochemistry ,Small molecule ,Binding domain - Abstract
PI3Kα remains an attractive target for the development of anticancer targeted therapy. A number of p110α crystal structures in complex with the nSH2-iSH2 fragment of p85 regulatory subunit have been reported, including a few small molecule co-crystal structures, but the utilization of this crystal form is limited by low diffraction resolution and a crystal packing artifact that partially blocks the ATP binding site. Taking advantage of recent data on the functional characterization of the lipid binding properties of p110α, we designed a set of novel constructs allowing production of isolated stable p110α subunit missing the Adapter Binding Domain and lacking or featuring a modified C-terminal lipid binding motif. While this protein is not catalytically competent to phosphorylate its substrate PIP2, it retains ligand binding properties as indicated by direct binding studies with a pan-PI3Kα inhibitor. Additionally, we determined apo and PF-04691502 bound crystal structures of the p110α (105-1048) subunit at 2.65 and 2.85 A, respectively. Comparison of isolated p110α(105-1048) with the p110α/p85 complex reveals a high degree of structural similarity, which validates suitability of this catalytically inactive p110α for iterative SBDD. Importantly, this crystal form of p110α readily accommodates the binding of noncovalent inhibitor by means of a fully accessible ATP site. The strategy presented here can be also applied to structural studies of other members of PI3KIA family.
- Published
- 2014
- Full Text
- View/download PDF
18. Discovery of (10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-ros Oncogene 1 (ROS1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant Mutations
- Author
-
Sergei Timofeevski, Lars D. Engstrom, Mingying He, Michael R. Collins, Martin Paul Edwards, Phuong Le, Graham L. Smith, John Charles Kath, Helen Y. Zou, Deal Judith G, Ya-Li Deng, Robert Steven Kania, Benjamin J. Burke, Cynthia Louise Palmer, Huichun Zhu, A.E. Stewart, Hieu Lam, Dac M. Dinh, Simon Bailey, Jacqui Elizabeth Hoffman, Neal W. Sach, Robert Louis Hoffman, Qinhua Huang, J. Jean Cui, Alexei Brooun, Wei Liu, Laura Lingardo, Tod Smeal, Ted William Johnson, Michele McTigue, Justine L. Lam, Jinjiang Zhu, and Paul F. Richardson
- Subjects
Models, Molecular ,Lactams ,Brigatinib ,Lactams, Macrocyclic ,Aminopyridines ,Antineoplastic Agents ,Crystallography, X-Ray ,Mice ,Structure-Activity Relationship ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Drug Discovery ,medicine ,ROS1 ,Animals ,Humans ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,ADME ,Crizotinib ,Chemistry ,Drug discovery ,Brain ,Receptor Protein-Tyrosine Kinases ,Stereoisomerism ,Protein-Tyrosine Kinases ,Lorlatinib ,Rats ,Drug Resistance, Neoplasm ,Lipophilic efficiency ,Mutation ,Microsomes, Liver ,NIH 3T3 Cells ,Cancer research ,Pyrazoles ,Molecular Medicine ,medicine.drug - Abstract
Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-positive non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addition, some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and physical-property-based optimization, highly potent macrocyclic ALK inhibitors were prepared with good absorption, distribution, metabolism, and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clinically reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.
- Published
- 2014
- Full Text
- View/download PDF
19. Design of Potent and Selective Inhibitors to Overcome Clinical Anaplastic Lymphoma Kinase Mutations Resistant to Crizotinib
- Author
-
Tod Smeal, Wei Liu, Huichun Zhu, Simon Bailey, Michael R. Collins, A.E. Stewart, Jacqui Elizabeth Hoffman, Lars D. Engstrom, Graham L. Smith, J. Jean Cui, Ya-Li Deng, Benjamin J. Burke, Laura Lingardo, Phuong Le, Andrew Simon Cook, Konstantinos Tsaparikos, Hieu Lam, Jinjiang Zhu, Ted William Johnson, Neal W. Sach, Dac M. Dinh, Robert Louis Hoffman, Mingying He, Hui Wang, Alexei Brooun, Dack Kevin Neil, Qinhua Huang, Sergei Timofeevski, Deal Judith G, Justine L. Lam, Qiuhua Li, Hong Shen, Melissa West Lu, Michele McTigue, Paul F. Richardson, Kevin D. Bunker, Robert Steven Kania, Martin Paul Edwards, Helen Y. Zou, Cynthia Louise Palmer, and Patrick S. Johnson
- Subjects
Drug ,Pyridines ,media_common.quotation_subject ,Drug resistance ,Pharmacology ,Crizotinib ,hemic and lymphatic diseases ,Drug Discovery ,medicine ,Humans ,Point Mutation ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,Protein Kinase Inhibitors ,media_common ,Chemistry ,Point mutation ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Protein kinase domain ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Pyrazoles ,Molecular Medicine ,Progressive disease ,medicine.drug - Abstract
Crizotinib (1), an anaplastic lymphoma kinase (ALK) receptor tyrosine kinase inhibitor approved by the U.S. Food and Drug Administration in 2011, is efficacious in ALK and ROS positive patients. Under pressure of crizotinib treatment, point mutations arise in the kinase domain of ALK, resulting in resistance and progressive disease. The successful application of both structure-based and lipophilic-efficiency-focused drug design resulted in aminopyridine 8e, which was potent across a broad panel of engineered ALK mutant cell lines and showed suitable preclinical pharmacokinetics and robust tumor growth inhibition in a crizotinib-resistant cell line (H3122-L1196M).
- Published
- 2014
- Full Text
- View/download PDF
20. The Axl kinase domain in complex with a macrocyclic inhibitor offers first structural insights into an active TAM receptor kinase
- Author
-
Sergei Timofeevski, Junli Feng, Neil B. Grodsky, Meirong Xu, Ben Bolaños, Brion W. Murray, Ted William Johnson, RoseAnn Ferre, Al E. Stewart, and Ketan S. Gajiwala
- Subjects
0301 basic medicine ,Models, Molecular ,Macrocyclic Compounds ,Protein domain ,Receptor Protein-Tyrosine Kinases ,Ligands ,Biochemistry ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Proto-Oncogene Proteins ,Enzyme Stability ,Humans ,Amino Acid Sequence ,Phosphorylation ,Receptor ,Protein kinase A ,Molecular Biology ,Protein Kinase Inhibitors ,Binding Sites ,biology ,Chemistry ,Kinase ,Cell Membrane ,Cell Biology ,Axl Receptor Tyrosine Kinase ,Cell biology ,030104 developmental biology ,Protein kinase domain ,030220 oncology & carcinogenesis ,Drug Design ,Protein Structure and Folding ,biology.protein ,TYRO3 ,Protein Binding - Abstract
The receptor tyrosine kinase family consisting of Tyro3, Axl, and Mer (TAM) is one of the most recently identified receptor tyrosine kinase families. TAM receptors are up-regulated postnatally and maintained at high levels in adults. They all play an important role in immunity, but Axl has also been implicated in cancer and therefore is a target in the discovery and development of novel therapeutics. However, of the three members of the TAM family, the Axl kinase domain is the only one that has so far eluded structure determination. To this end, using differential scanning fluorimetry and hydrogen-deuterium exchange mass spectrometry, we show here that a lower stability and greater dynamic nature of the Axl kinase domain may account for its poor crystallizability. We present the first structural characterization of the Axl kinase domain in complex with a small-molecule macrocyclic inhibitor. The Axl crystal structure revealed two distinct conformational states of the enzyme, providing a first glimpse of what an active TAM receptor kinase may look like and suggesting a potential role for the juxtamembrane region in enzyme activity. We noted that the ATP/inhibitor-binding sites of the TAM members closely resemble each other, posing a challenge for the design of a selective inhibitor. We propose that the differences in the conformational dynamics among the TAM family members could potentially be exploited to achieve inhibitor selectivity for targeted receptors.
- Published
- 2016
21. Discovery of a Novel Class of Exquisitely Selective Mesenchymal-Epithelial Transition Factor (c-MET) Protein Kinase Inhibitors and Identification of the Clinical Candidate 2-(4-(1-(Quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (PF-04217903) for the Treatment of Cancer
- Author
-
Gilles H. Goetz, Michele McTigue, Max Parker, Ya-Li Deng, Nambu Mitchell David, J. Jean Cui, Shinji Yamazaki, Jia Lei, Neil Grodsky, Shirley A. Aguirre, Jacqui Elizabeth Hoffman, Kevin Ryan, Hong Shen, Phuong Le, Sergei Timofeevski, James G. Christensen, Helen Y. Zou, Cheng Hengmiao, Brion W. Murray, Michelle Tran-Dubé, Mehran Jalaie, Qiuhua Li, and Pairish Mason Alan
- Subjects
biology ,Hydrazide ,medicine.disease_cause ,Receptor tyrosine kinase ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Tumor progression ,Drug Discovery ,biology.protein ,medicine ,Cancer research ,Molecular Medicine ,Mesenchymal–epithelial transition ,Transferase ,Oxindole ,Protein kinase A ,Carcinogenesis - Abstract
The c-MET receptor tyrosine kinase is an attractive oncology target because of its critical role in human oncogenesis and tumor progression. An oxindole hydrazide hit 6 was identified during a c-ME...
- Published
- 2012
- Full Text
- View/download PDF
22. Sensitivity of Selected Human Tumor Models to PF-04217903, a Novel Selective c-Met Kinase Inhibitor
- Author
-
James G. Christensen, Sergei Timofeevski, Michele McTigue, Max Parker, Tod Smeal, Shinji Yamazaki, Gerrit Los, Lars D. Engstrom, Maria E. Arango, Kristina Burgess, Bender Steven Lee, Helen Y. Zou, Jingrong Jean Cui, Ming Qiu, Joseph H. Lee, and Qiuhua Li
- Subjects
Cancer Research ,C-Met ,Mice, Nude ,Biology ,Small hairpin RNA ,Mice ,chemistry.chemical_compound ,Growth factor receptor ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Autocrine signalling ,Protein Kinase Inhibitors ,Cell Proliferation ,Oncogene ,Kinase ,Receptor Protein-Tyrosine Kinases ,Triazoles ,Xenograft Model Antitumor Assays ,Molecular biology ,Oncology ,chemistry ,Pyrazines ,Female ,Hepatocyte growth factor ,Signal Transduction ,medicine.drug - Abstract
The c-Met pathway has been implicated in a variety of human cancers for its critical role in tumor growth, invasion, and metastasis. PF-04217903 is a novel ATP-competitive small-molecule inhibitor of c-Met kinase. PF-04217903 showed more than 1,000-fold selectivity for c-Met compared with more than 150 kinases, making it one of the most selective c-Met inhibitors described to date. PF-04217903 inhibited tumor cell proliferation, survival, migration/invasion in MET-amplified cell lines in vitro, and showed marked antitumor activity in tumor models harboring either MET gene amplification or a hepatocyte growth factor (HGF)/c-Met autocrine loop at well-tolerated dose levels in vivo. Antitumor efficacy of PF-04217903 was dose-dependent and showed a strong correlation with inhibition of c-Met phosphorylation, downstream signaling, and tumor cell proliferation/survival. In human xenograft models that express relatively high levels of c-Met, complete inhibition of c-Met activity by PF-04217903 only led to partial tumor growth inhibition (38%–46%) in vivo. The combination of PF-04217903 with Recepteur d'origine nantais (RON) short hairpin RNA (shRNA) knockdown in the HT29 model that also expresses activated RON kinase–induced tumor cell apoptosis and resulted in enhanced antitumor efficacy (77%) compared with either PF-04217903 (38%) or RON shRNA alone (56%). PF-04217903 also showed potent antiangiogenic properties in vitro and in vivo. Furthermore, PF-04217903 strongly induced phospho-PDGFRβ (platelet-derived growth factor receptor) levels in U87MG xenograft tumors, indicating a possible oncogene switching mechanism in tumor cell signaling as a potential resistance mechanism that might compromise tumor responses to c-Met inhibitors. Collectively, these results show the use of highly selective inhibition of c-Met and provide insight toward targeting tumors exhibiting different mechanisms of c-Met dysregulation. Mol Cancer Ther; 11(4); 1036–47. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
23. Enzymatic Characterization of c-Met Receptor Tyrosine Kinase Oncogenic Mutants and Kinetic Studies with Aminopyridine and Triazolopyrazine Inhibitors
- Author
-
Fannie Chau, Helen Y. Zou, Michele McTigue, Karlicek Shannon Marie, Kevin Ryan, Brion W. Murray, James G. Christensen, Jean Cui, Sergei Timofeevski, Gordon Alton, and Jeff Xianchao Zhu
- Subjects
Protein Conformation ,Mutant ,Aminopyridines ,medicine.disease_cause ,Biochemistry ,Catalysis ,Receptor tyrosine kinase ,medicine ,Humans ,Enzyme kinetics ,Phosphorylation ,Binding site ,Protein Kinase Inhibitors ,chemistry.chemical_classification ,Mutation ,Binding Sites ,biology ,Autophosphorylation ,Proto-Oncogene Proteins c-met ,Kinetics ,Enzyme ,chemistry ,Pyrazines ,biology.protein - Abstract
The c-Met receptor tyrosine kinase (RTK) is a key regulator in cancer, in part, through oncogenic mutations. Eight clinically relevant mutants were characterized by biochemical, biophysical, and cellular methods. The c-Met catalytic domain was highly active in the unphosphorylated state (k(cat) = 1.0 s(-1)) and achieved 160-fold enhanced catalytic efficiency (k(cat)/K(m)) upon activation to 425000 s(-1) M(-1). c-Met mutants had 2-10-fold higher basal enzymatic activity (k(cat)) but achieved maximal activities similar to those of wild-type c-Met, except for Y1235D, which underwent a reduction in maximal activity. Small enhancements of basal activity were shown to have profound effects on the acquisition of full enzymatic activity achieved through accelerating rates of autophosphorylation. Biophysical analysis of c-Met mutants revealed minimal melting temperature differences indicating that the mutations did not alter protein stability. A model of RTK activation is proposed to describe how a RTK response may be matched to a biological context through enzymatic properties. Two c-Met clinical candidates from aminopyridine and triazolopyrazine chemical series (PF-02341066 and PF-04217903) were studied. Biochemically, each series produced molecules that are highly selective against a large panel of kinases, with PF-04217903 (1000-fold selective relative to 208 kinases) being more selective than PF-02341066. Although these prototype inhibitors have similar potencies against wild-type c-Met (K(i) = 6-7 nM), significant differences in potency were observed for clinically relevant mutations evaluated in both biochemical and cellular contexts. In particular, PF-02341066 was 180-fold more active against the Y1230C mutant c-Met than PF-04217903. These highly optimized inhibitors indicate that for kinases susceptible to active site mutations, inhibitor design may need to balance overall kinase selectivity with the ability to inhibit multiple mutant forms of the kinase (penetrance).
- Published
- 2009
- Full Text
- View/download PDF
24. Pharmacological characterization of a small molecule inhibitor of c-Jun kinase
- Author
-
Karen Siegel, Helen Kim Cho, David Looper, Shawn C Black, Xiao-Hong Yu, Scott Rp McDonnell, Dawn Kelly-Sullivan, Sergei Timofeevski, Junming Yie, Celia P. Briscoe, Kathleen M. Ogilvie, Ping Chen, Manli Shi, and James S. Fraser
- Subjects
Blood Glucose ,Lipopolysaccharides ,Male ,medicine.medical_specialty ,Proto-Oncogene Proteins c-jun ,Physiology ,Ratón ,Adipose Tissue, White ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Aminopyridines ,Biology ,Eating ,Mice ,Insulin resistance ,3T3-L1 Cells ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Humans ,Insulin ,Mitogen-Activated Protein Kinase 8 ,Obesity ,Phosphorylation ,Protein Kinase Inhibitors ,U937 cell ,Tumor Necrosis Factor-alpha ,Kinase ,Body Weight ,c-jun ,JNK Mitogen-Activated Protein Kinases ,U937 Cells ,medicine.disease ,Dietary Fats ,Small molecule ,Mice, Inbred C57BL ,Endocrinology ,Insulin Receptor Substrate Proteins ,Cytokines ,Insulin Resistance ,Immediate early gene - Abstract
c-Jun NH2-terminal kinase (JNK) plays an important role in insulin resistance; however, identification of pharmacologically potent and selective small molecule JNK inhibitors has been limited. Compound A has a cell IC50 of 102 nM and is at least 100-fold selective against related kinases and 27-fold selective against glycogen synthase kinase-3β and cyclin-dependent kinase-2. In C57BL/6 mice, compound A reduced LPS-mediated increases in both plasma cytokine levels and phosphorylated c-Jun in adipose tissue. Treatment of mice fed a high-fat diet with compound A for 3 wk resulted in a 13.1 ± 1% decrease in body weight and a 9.3 ± 1.5% decrease in body fat, compared with a 6.6 ± 2.1% increase in body weight and a 6.7 ± 2.1% increase in body fat in vehicle-treated mice. Mice pair fed to those that received compound A exhibited a body weight decrease of 7 ± 1% and a decrease in body fat of 1.6 ± 1.3%, suggesting that reductions in food intake could not account solely for the reductions in adiposity observed. Compound A dosed at 30 mg/kg for 13 days in high-fat fed mice resulted in a significant decrease in phosphorylated c-Jun in adipose tissue accompanied by a decrease in weight and reductions in glucose and triglycerides and increases in insulin sensitivity to levels comparable with those in lean control mice. The ability of compound A to reduce the insulin-stimulated phosphorylation of insulin receptor substrate-1 (IRS-1) von Ser307 and partially reverse the free fatty acid inhibition of glucose uptake in 3T3L1 adipocytes, suggests that enhancement of insulin signaling in addition to weight loss may contribute to the effects of compound A on insulin sensitization in vivo. Pharmacological inhibition of JNK using compound A may therefore offer an effective therapy for type 2 diabetes mediated at least in part via weight reduction.
- Published
- 2008
- Full Text
- View/download PDF
25. Resensitization to Crizotinib by the Lorlatinib ALK Resistance Mutation L1198F
- Author
-
Benjamin J. Burke, Bergqvist S, Ya-Li Deng, Tod Smeal, Long P. Le, Michele McTigue, Leila Dardaei, Alexei Brooun, Leonard P. James, Ted William Johnson, Gad Getz, Justin F. Gainor, Anthony J. Iafrate, J. A. Engelman, Ignaty Leshchiner, Luc Friboulet, Ryohei Katayama, W. Liu, Katherine Schultz, Alice T. Shaw, Jennifer A. Logan, Rosa L. Frias, and Sergei Timofeevski
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Lung Neoplasms ,Brigatinib ,Lactams ,medicine.drug_class ,Pyridines ,Lactams, Macrocyclic ,Aminopyridines ,Drug resistance ,Article ,03 medical and health sciences ,0302 clinical medicine ,Crizotinib ,hemic and lymphatic diseases ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Anaplastic lymphoma kinase ,Humans ,Anaplastic Lymphoma Kinase ,Sulfones ,Lung cancer ,Protein Kinase Inhibitors ,Binding Sites ,Molecular Structure ,business.industry ,Liver Neoplasms ,Receptor Protein-Tyrosine Kinases ,General Medicine ,Middle Aged ,medicine.disease ,Resistance mutation ,Lorlatinib ,ALK inhibitor ,030104 developmental biology ,Endocrinology ,Pyrimidines ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Pyrazoles ,Female ,business ,Liver Failure ,medicine.drug - Abstract
In a patient who had metastatic anaplastic lymphoma kinase (ALK)-rearranged lung cancer, resistance to crizotinib developed because of a mutation in the ALK kinase domain. This mutation is predicted to result in a substitution of cysteine by tyrosine at amino acid residue 1156 (C1156Y). Her tumor did not respond to a second-generation ALK inhibitor, but it did respond to lorlatinib (PF-06463922), a third-generation inhibitor. When her tumor relapsed, sequencing of the resistant tumor revealed an ALK L1198F mutation in addition to the C1156Y mutation. The L1198F substitution confers resistance to lorlatinib through steric interference with drug binding. However, L1198F paradoxically enhances binding to crizotinib, negating the effect of C1156Y and resensitizing resistant cancers to crizotinib. The patient received crizotinib again, and her cancer-related symptoms and liver failure resolved. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT01970865.).
- Published
- 2015
26. Mitotic Checkpoint Kinase Mps1 Has a Role in Normal Physiology which Impacts Clinical Utility
- Author
-
Robert Louis Hoffman, Jill Hallin, Patrick B. Lappin, Jeffery Fan, Gina M. Yanochko, Murphy Sean T, Ricardo Martinez, Isha Rymer, Zhou Zhu, Dac M. Dinh, Brion W. Murray, Matthew A. Marx, Wenyue Hu, Peiquing Sun, Sergei Timofeevski, Dusko Trajkovic, and Alessandra Blasina
- Subjects
Cell cycle checkpoint ,Pyridines ,Physiology ,lcsh:Medicine ,Apoptosis ,Cell Cycle Proteins ,Mice, SCID ,Piperazines ,Histones ,Mice ,0302 clinical medicine ,Intestine, Small ,Phosphorylation ,RNA, Small Interfering ,lcsh:Science ,0303 health sciences ,Multidisciplinary ,biology ,Cell cycle process ,Protein-Tyrosine Kinases ,3. Good health ,Spindle checkpoint ,030220 oncology & carcinogenesis ,Female ,RNA Interference ,G1 phase ,Research Article ,Cell Survival ,Transplantation, Heterologous ,Mitosis ,Bone Marrow Cells ,Breast Neoplasms ,Protein Serine-Threonine Kinases ,Palbociclib ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,Humans ,Protein Kinase Inhibitors ,Cell Proliferation ,030304 developmental biology ,Cyclin-dependent kinase 4 ,lcsh:R ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,G1 Phase Cell Cycle Checkpoints ,Rats ,biology.protein ,lcsh:Q ,Cyclin-dependent kinase 6 - Abstract
Cell cycle checkpoint intervention is an effective therapeutic strategy for cancer when applied to patients predisposed to respond and the treatment is well-tolerated. A critical cell cycle process that could be targeted is the mitotic checkpoint (spindle assembly checkpoint) which governs the metaphase-to-anaphase transition and insures proper chromosomal segregation. The mitotic checkpoint kinase Mps1 was selected to explore whether enhancement in genomic instability is a viable therapeutic strategy. The basal-a subset of triple-negative breast cancer was chosen as a model system because it has a higher incidence of chromosomal instability and Mps1 expression is up-regulated. Depletion of Mps1 reduces tumor cell viability relative to normal cells. Highly selective, extremely potent Mps1 kinase inhibitors were created to investigate the roles of Mps1 catalytic activity in tumor cells and normal physiology (PF-7006, PF-3837; K i
- Published
- 2015
27. Enzyme kinetics and distinct modulation of the protein kinase N family of kinases by lipid activators and small molecule inhibitors
- Author
-
Stephan Grant, Ben Bolaños, Alexei Brooun, Sergei Timofeevski, Anke Klippel, Keziban Unsal-Kacmaz, Matthew D. Falk, and Wei Liu
- Subjects
ROCK, Rho-associated kinase ,kinetic mechanism ,lcsh:Life ,lcsh:QR1-502 ,IP3, D-myo-inositol-1,3,5-triphosphate ,PDK1, phosphoinositide-dependent kinase 1 ,Peptide ,Biochemistry ,lcsh:Microbiology ,Adenosine Triphosphate ,DAG, 1,2-dioctanoyl-sn-glycerol ,MS/MS, tandem MS ,Protein Kinase C ,protein kinase N (PKN) ,chemistry.chemical_classification ,CaM, calmodulin ,Arachidonic Acid ,Kinase ,Effector ,PIP3, phosphatidylinositol-3,4,5-triphosphate ,Small molecule ,Isoenzymes ,Gene isoform ,Cdk1/2, cyclin-dependent kinase 1/2 ,kinase inhibitor ,PIF, PDK1-interacting fragment ,Biophysics ,S6 ,Biology ,Catalysis ,lipid ,PKC, protein kinase C ,Humans ,cancer ,Enzyme kinetics ,Protein Kinase Inhibitors ,Molecular Biology ,Original Paper ,S6K, S6 kinase ,Cell Biology ,AGC kinase ,lcsh:QH501-531 ,Kinetics ,Enzyme ,chemistry ,DTT, dithiothreitol ,PIP2, phosphatidylinositol-4,5-bisphosphate ,Product inhibition ,HEK-293 cells, human embryonic kidney 293 cells ,PKG, protein kinase G ,PKA, protein kinase A ,MAPK, mitogen-activated protein kinase ,PKN, protein kinase N - Abstract
The PKN (protein kinase N) family of Ser/Thr protein kinases regulates a diverse set of cellular functions, such as cell migration and cytoskeletal organization. Inhibition of tumour PKN activity has been explored as an oncology therapeutic approach, with a PKN3-targeted RNAi (RNA interference)-derived therapeutic agent in Phase I clinical trials. To better understand this important family of kinases, we performed detailed enzymatic characterization, determining the kinetic mechanism and lipid sensitivity of each PKN isoform using full-length enzymes and synthetic peptide substrate. Steady-state kinetic analysis revealed that PKN1–3 follows a sequential ordered Bi–Bi kinetic mechanism, where peptide substrate binding is preceded by ATP binding. This kinetic mechanism was confirmed by additional kinetic studies for product inhibition and affinity of small molecule inhibitors. The known lipid effector, arachidonic acid, increased the catalytic efficiency of each isoform, mainly through an increase in kcat for PKN1 and PKN2, and a decrease in peptide KM for PKN3. In addition, a number of PKN inhibitors with various degrees of isoform selectivity, including potent (Ki, We conducted kinetic analysis of the relatively unexplored PKN family and effects of lipids, and identified potent inhibitors with various isoform selectivity. The kinetic mechanism, lipid activators and inhibitors could be useful for understanding PKN biology and developing PKN-targeted therapies.
- Published
- 2014
- Full Text
- View/download PDF
28. Discovery of a novel class of exquisitely selective mesenchymal-epithelial transition factor (c-MET) protein kinase inhibitors and identification of the clinical candidate 2-(4-(1-(quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (PF-04217903) for the treatment of cancer
- Author
-
J Jean, Cui, Michele, McTigue, Mitchell, Nambu, Michelle, Tran-Dubé, Mason, Pairish, Hong, Shen, Lei, Jia, Hengmiao, Cheng, Jacqui, Hoffman, Phuong, Le, Mehran, Jalaie, Gilles H, Goetz, Kevin, Ryan, Neil, Grodsky, Ya-li, Deng, Max, Parker, Sergei, Timofeevski, Brion W, Murray, Shinji, Yamazaki, Shirley, Aguirre, Qiuhua, Li, Helen, Zou, and James, Christensen
- Subjects
Models, Molecular ,Indoles ,Protein Conformation ,Molecular Sequence Data ,Antineoplastic Agents ,Proto-Oncogene Proteins c-met ,Triazoles ,High-Throughput Screening Assays ,Oxindoles ,Substrate Specificity ,Drug Stability ,Cell Line, Tumor ,Pyrazines ,Drug Discovery ,Animals ,Humans ,Amino Acid Sequence ,Protein Kinase Inhibitors - Abstract
The c-MET receptor tyrosine kinase is an attractive oncology target because of its critical role in human oncogenesis and tumor progression. An oxindole hydrazide hit 6 was identified during a c-MET HTS campaign and subsequently demonstrated to have an unusual degree of selectivity against a broad array of other kinases. The cocrystal structure of the related oxindole hydrazide c-MET inhibitor 10 with a nonphosphorylated c-MET kinase domain revealed a unique binding mode associated with the exquisite selectivity profile. The chemically labile oxindole hydrazide scaffold was replaced with a chemically and metabolically stable triazolopyrazine scaffold using structure based drug design. Medicinal chemistry lead optimization produced 2-(4-(1-(quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (2, PF-04217903), an extremely potent and exquisitely selective c-MET inhibitor. 2 demonstrated effective tumor growth inhibition in c-MET dependent tumor models with good oral PK properties and an acceptable safety profile in preclinical studies. 2 progressed to clinical evaluation in a Phase I oncology setting.
- Published
- 2012
29. Structure based drug design of crizotinib (PF-02341066), a potent and selective dual inhibitor of mesenchymal-epithelial transition factor (c-MET) kinase and anaplastic lymphoma kinase (ALK)
- Author
-
Michele McTigue, Robert Steven Kania, Pei-Pei Kung, Nambu Mitchell David, Jerry Meng, Steve Bender, Barbara Mroczkowski, Botrous Iriny, J. Jean Cui, Shinji Yamazaki, Pairish Mason Alan, Hong Shen, Jia Lei, James G. Christensen, Sergei Timofeevski, Gordon Alton, Michelle Tran-Dubé, Ellen Padrique, Qiuhua Li, Funk Lee Andrew, Helen Y. Zou, Martin Paul Edwards, Neil Grodsky, and Kevin Ryan
- Subjects
Models, Molecular ,Epithelial-Mesenchymal Transition ,Indoles ,Pyridines ,Molecular Conformation ,Pharmacology ,Crystallography, X-Ray ,Receptor tyrosine kinase ,Structure-Activity Relationship ,Crizotinib ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Anaplastic lymphoma kinase ,Humans ,Anaplastic Lymphoma Kinase ,Epithelial–mesenchymal transition ,Cell Proliferation ,Ligand efficiency ,biology ,Kinase ,Chemistry ,Receptor Protein-Tyrosine Kinases ,Stereoisomerism ,Proto-Oncogene Proteins c-met ,Protein kinase domain ,Drug Design ,biology.protein ,Cancer research ,Molecular Medicine ,Pyrazoles ,Drug Screening Assays, Antitumor ,medicine.drug - Abstract
Because of the critical roles of aberrant signaling in cancer, both c-MET and ALK receptor tyrosine kinases are attractive oncology targets for therapeutic intervention. The cocrystal structure of 3 (PHA-665752), bound to c-MET kinase domain, revealed a novel ATP site environment, which served as the target to guide parallel, multiattribute drug design. A novel 2-amino-5-aryl-3-benzyloxypyridine series was created to more effectively make the key interactions achieved with 3. In the novel series, the 2-aminopyridine core allowed a 3-benzyloxy group to reach into the same pocket as the 2,6-dichlorophenyl group of 3 via a more direct vector and thus with a better ligand efficiency (LE). Further optimization of the lead series generated the clinical candidate crizotinib (PF-02341066), which demonstrated potent in vitro and in vivo c-MET kinase and ALK inhibition, effective tumor growth inhibition, and good pharmaceutical properties.
- Published
- 2011
30. Identification of small-molecule inhibitors of the JIP-JNK interaction
- Author
-
Sergei Timofeevski, William R. Tschantz, Maura E. Charlton, Jeremy Little, Paul Bauer, Tracy Chen, Robert V. Stanton, Natasha M. Kablaoui, Junli Feng, and Ping Chen
- Subjects
Models, Molecular ,MAP kinase kinase kinase ,Molecular Structure ,Cyclin-dependent kinase 2 ,JNK Mitogen-Activated Protein Kinases ,Cell Biology ,Mitogen-activated protein kinase kinase ,Biology ,Biochemistry ,Protein kinase R ,MAP2K7 ,Cell biology ,Protein Structure, Tertiary ,Structure-Activity Relationship ,biology.protein ,Animals ,Humans ,ASK1 ,Cyclin-dependent kinase 9 ,c-Raf ,Organic Chemicals ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Protein Binding - Abstract
JNK1 (c-Jun N-terminal kinase 1) plays a crucial role in the regulation of obesity-induced insulin resistance and is implicated in the pathology of Type 2 diabetes. Its partner, JIP1 (JNK-interacting protein 1), serves a scaffolding function that facilitates JNK1 activation by MKK4 [MAPK (mitogen-activated protein kinase) kinase 4] and MKK7 (MAPK kinase 7). For example, reduced insulin resistance and JNK activation are observed in JIP1-deficient mice. On the basis of the in vivo efficacy of a cell-permeable JIP peptide, the JIP–JNK interaction appears to be a potential target for JNK inhibition. The goal of the present study was to identify small-molecule inhibitors that disrupt the JIP–JNK interaction to provide an alternative approach for JNK inhibition to ATP-competitive inhibitors. High-throughput screening was performed by utilizing a fluorescence polarization assay that measured the binding of JNK1 to the JIP peptide. Multiple chemical series were identified, revealing two categories of JIP/JNK inhibitors: ‘dual inhibitors’ that are ATP competitive and probably inhibit JIP–JNK binding allosterically, and ‘JIP-site binders’ that block binding through interaction with the JIP site. A series of polychloropyrimidines from the second category was characterized by biochemical methods and explored through medicinal-chemistry efforts. As predicted, these inhibitors also inhibited full-length JIP–JNK binding and were selective against a panel of 34 representative kinases, including ones in the MAPK family. Overall, this work demonstrates that small molecules can inhibit protein–protein interactions in vitro in the MAPK family effectively and provides strategies for similar approaches within other target families.
- Published
- 2009
31. Synthesis and SAR of 4-substituted-2-aminopyrimidines as novel c-Jun N-terminal kinase (JNK) inhibitors
- Author
-
Jennifer Lafontaine, Ping Chen, Quyen-Quyen T. Do, Karen Siegel, Lilian Y. Li, Paul S. Humphries, David M. Wilhite, Tianlun Wang, Charles S. Agree, David Alexander, Sergei Timofeevski, Ranjan Jagath Rajapakse, and Elizabeth A. Lunney
- Subjects
Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,Drug Discovery ,Humans ,Mitogen-Activated Protein Kinase 8 ,Molecular Biology ,Protein Kinase Inhibitors ,chemistry.chemical_classification ,biology ,Chemistry ,Kinase ,Cellular Assay ,Organic Chemistry ,c-jun ,JNK Mitogen-Activated Protein Kinases ,In vitro ,Enzyme ,Pyrimidines ,Enzyme inhibitor ,Mitogen-activated protein kinase ,biology.protein ,Microsomes, Liver ,Molecular Medicine - Abstract
The development of a series of novel 4-substituted-2-aminopyrimidines as inhibitors of c-Jun N-terminal kinases is described. The synthesis, in vitro inhibitory values for JNK1, and the in vitro inhibitory value for a c-Jun cellular assay are discussed. Optimization of microsomal clearance led to the identification of 9c, whose kinase selectivity is reported.
- Published
- 2009
32. Abstract 130: PF-06463922, a novel next generation ALK/ROS1 inhibitor, overcomes resistance to 1st and 2nd generation ALK inhibitors in pre-clinical models
- Author
-
Luc Friboulet, Tod Smeal, Konstantinos Tsaparikos, Jinwei Wang, Rakesh K. Jain, Hovhannes J. Gukasyan, Valeria Fantin, Qiuhua Li, Helen Y. Zou, Nathan V. Lee, Timothy Affolter, Melissa West, Patrick B. Lappin, Lars D. Engstrom, Sergei Timofeevski, Ted William Johnson, Eugene Lifshits, Justine L. Lam, Hui Wang, Dac M. Dinh, Bhushankumar Patel, David P. Kodack, Hieu Lam, Sidra Mahmood, Shibing Deng, Shinji Yamazaki, Divya Bezwada, Alice T. Shaw, Wenyue Hu, and Ruth W. Tang
- Subjects
Gerontology ,Cancer Research ,Kinase ,business.industry ,Cancer ,Drug resistance ,medicine.disease ,Oncology ,Protein kinase domain ,hemic and lymphatic diseases ,medicine ,Cancer research ,ROS1 ,Lung cancer ,business ,DISEASE RELAPSE ,Brain metastasis - Abstract
Overcoming resistance to targeted kinase inhibitors is a major clinical challenge in oncology. For 1st and 2nd generation ALK inhibitors acquired resistance due to ALK kinase domain mutations and/or pharmacological drug resistance are major causes for disease relapse. Here, we report the preclinical evaluation of PF-06463922, a potent and brain penetrant ALK/ROS1 inhibitor with sub to low nanomolar cell potency against ALK fusions and all known clinically-acquired resistant mutations. PF-06463922 exhibited marked cytoreductive activity in tumor xenografts driven by various ALK mutants. Furthermore, PF-06463922 led to significant regression of EML4-ALK driven lung cancer brain metastasis and prolonged mouse survival. Compared to other clinically available ALK inhibitors, PF-06463922 is unique in its superior potency against a broad spectrum of acquired ALK mutations, including the highly resistant G1202R mutant and its robust antitumor activity in the brain. Furthermore, PF-06463922 demonstrated remarkable selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 may be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors due to ALK secondary mutations and/or brain metastases. Citation Format: Luc Friboulet, Helen Zou, David P. Kodack, Lars D. Engstrom, Qiuhua Li, Melissa West, Ruth W. Tang, Hui Wang, Konstantinos Tsaparikos, Jinwei Wang, Sergei Timofeevski, Dac M. Dinh, Hieu Lam, Justine L. Lam, Shinji Yamazaki, Wenyue Hu, Bhushankumar Patel, Divya Bezwada, Sidra Mahmood, Eugene Lifshits, Timothy Affolter, Patrick B. Lappin, Hovhannes Gukasyan, Nathan Lee, Shibing Deng, Rakesh K. Jain, Ted W. Johnson, Alice T. Shaw, Valeria R. Fantin, Tod Smeal. PF-06463922, a novel next generation ALK/ROS1 inhibitor, overcomes resistance to 1st and 2nd generation ALK inhibitors in pre-clinical models. [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 130. doi:10.1158/1538-7445.AM2015-130
- Published
- 2015
- Full Text
- View/download PDF
33. Isoform-selective interaction of cyclooxygenase-2 with indomethacin amides studied by real-time fluorescence, inhibition kinetics, and site-directed mutagenesis
- Author
-
Sergei Timofeevski, Carol A. Rouzer, Jeffery J. Prusakiewicz, and Lawrence J. Marnett
- Subjects
Gene isoform ,Spectrometry, Mass, Electrospray Ionization ,Stereochemistry ,Flurbiprofen ,Indomethacin ,Inhibition kinetics ,Biochemistry ,chemistry.chemical_compound ,Mice ,medicine ,Ethanolamide ,Animals ,Cyclooxygenase Inhibitors ,Site-directed mutagenesis ,Nonsteroidal ,Sheep ,biology ,Cyclooxygenase 2 Inhibitors ,Fluorescence ,Amides ,Isoenzymes ,Kinetics ,Spectrometry, Fluorescence ,chemistry ,Cyclooxygenase 2 ,Prostaglandin-Endoperoxide Synthases ,biology.protein ,Mutagenesis, Site-Directed ,Cyclooxygenase ,medicine.drug - Abstract
Conversion of carboxylate-containing nonsteroidal antiinflammatory drugs, such as indomethacin, to esters or amides provides potent and selective inhibitors of cyclooxygenase-2 (COX-2) [Kalgutkar et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 925-930]. Synthesis of cinnamyl- or coumarinyl-substituted ethanolamide derivatives of indomethacin produced fluorescent probes of inhibitor interaction with COX-2 and COX-1. Binding of either derivative to apoCOX-2 or apoCOX-1 resulted in a rapid, reversible enhancement of fluorescence. Following this rapid phase, a slow additional increase in fluorescence was observed with apoCOX-2 but not with apoCOX-1. The slow, COX-2-specific increase in fluorescence was prevented or reversed by addition of the nonfluorescent COX inhibitor (S)-flurbiprofen. Detailed kinetic studies of the interaction of the coumarinyl derivative with COX-2 showed that the observed changes in fluorescence could be described by two sequential equilibria, the first of which is rapid, reversible, and bimolecular in the forward direction. The second equilibrium is slower, reversible, and unimolecular in both directions. The forward rate constant for the slow equilibrium determined by fluorescence enhancement [(3.1 +/- 0.6) x 10(-3) s(-1)] corresponded closely to the forward rate constant for inhibition of COX-2 activity [(6.8 +/- 2.3) x 10(-3) s(-1)], suggesting that the slow fluorescence enhancement is associated with selective COX-2 inhibition. Site-directed mutagenesis indicated that residues in the carboxylate-binding region of the COX-2 active site (Arg-120, Tyr-355, and Glu-524) are critical for the binding of the indomethacin conjugates that leads to slow fluorescence enhancement and cyclooxygenase inhibition. The indomethacin conjugates described herein represent powerful tools for the investigation of a novel class of selective inhibitors of COX-2.
- Published
- 2002
34. Abstract A277: PF-06463922, a novel ROS1/ALK inhibitor, demonstrates sub-nanomolar potency against oncogenic ROS1 fusions and capable of blocking the resistant ROS1G2032R mutant in preclinical tumor models
- Author
-
Helen Y. Zou, Konstantinos Tsaparikos, Hui Wang, Nathan V. Lee, Justine L. Lam, Qiuhua Li, Ruth W. Tang, Sergei Timofeevski, Tod Smeal, Valeria Fantin, Shinji Yamazaki, Ted William Johnson, Melissa West Lu, Lars R. Engstrom, Hovhannes J. Gukasyan, and Wenyue Hu
- Subjects
Cancer Research ,Crizotinib ,medicine.drug_class ,Kinase ,Cancer ,Pharmacology ,Cell cycle ,Biology ,medicine.disease ,ALK inhibitor ,Fusion gene ,Oncology ,Cancer research ,medicine ,ROS1 ,Adenocarcinoma ,medicine.drug - Abstract
The oncogenic ROS1 gene fusion (Fig-ROS1) was first identified in glioblastoma cells over two decades ago. Recently, ROS1 gene rearrangements were further discovered in a variety of human cancers, including lung adenocarcinoma, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma, providing additional evidence for ROS1 as an attractive cancer target. The 1st generation Met/ALK/ROS1 inhibitor XALKORI ® (crizotinib) has demonstrated promising clinical response in ROS1 fusion positive NSCLC. But similar to what was seen with acquired ALK secondary resistant mutations in XALKORI refractory patients, a ROS1 kinase domain mutant–ROS1G2032R has been identified in a ROS1 positive NSCLC patient who developed resistance to XALKORI. Therefore, there is an urgent need to develop agents that can overcome this type of resistance. PF-06463922 is a novel, orally available, ATP-competitive small molecule inhibitor of ROS1/ALK with exquisite potency against ROS1 kinase. PF-06463922 inhibited the catalytic activity of recombinant ROS1 with a mean Ki of < 0.005 nM, and inhibited ROS1 autophosphorylation at IC50 values ranging from 0.1 nM to 1 nM cross a panel of cell lines harboring oncogenic ROS1 fusion variants including CD74-ROS1, SLC34A2-ROS1 and Fig-ROS1. PF-06463922 also inhibited cell proliferation and induced cell apoptosis at sub- to low-nanomolar concentrations in the HCC78 human NSCLC cells harboring SLC34A2-ROS1 fusions and the BaF3-CD74-ROS1 cells expressing human CD74-ROS1. In the BaF3 cells engineered to express the XALKORI resistant CD74-ROS1G2032R mutant, PF-06463922 demonstrated nanomolar potency against either ROS1G2032R cellular activity or cell proliferation. In vivo, PF-06463922 demonstrated marked cytoreductive antitumor efficacy at low nanomolar concentration in the NIH3T3 xenograft models expressing human CD74-ROS1 and Fig-ROS1. The antitumor efficacy of PF-06463922 was dose dependent and strongly correlated to inhibition in ROS1 phosphorylation and the downstream signaling molecules pSHP1, pSHP2 and pErk1/2, as well as inhibition of the cell cycle protein Cyclin D1 in tumors. To our knowledge, PF-06463922 is the first reported ROS1 inhibitor that is capable of blocking the resistant ROS1G2032R mutant at predicted pharmacologically relevant concentrations. Our data indicate that PF-06463922 has great potential for treating ROS1 fusion positive cancers including those from patients who relapsed from XALKORI therapy due to acquired ROS1G2032Rmutation. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A277. Citation Format: Helen Y. Zou, Lars R. Engstrom, Qiuhua Li, Melissa West Lu, Ruth Wei Tang, Hui Wang, Konstantinos Tsaparikos, Sergei Timofeevski, Justine Lam, Shinji Yamazaki, Wenyue Hu, Hovhannes Gukasyan, Nathan Lee, Ted W. Johnson, Valeria Fantin, Tod Smeal. PF-06463922, a novel ROS1/ALK inhibitor, demonstrates sub-nanomolar potency against oncogenic ROS1 fusions and capable of blocking the resistant ROS1G2032R mutant in preclinical tumor models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A277.
- Published
- 2013
- Full Text
- View/download PDF
35. Abstract C253: PF-06463922, a novel brain-penetrating small molecule inhibitor of ALK/ROS1 with potent activity against a broad spectrum of ALK resistant mutations in preclinical models in vitro and in vivo
- Author
-
Jinwei Wang, Sergei Timofeevski, Hovhannes J. Gukasyan, Helen Y. Zou, Wenyue Hu, Konstantinos Tsaparikos, Tod Smeal, Qiuhua Li, Hieu Lam, Shinji Yamazaki, Dac M. Dinh, Justine L. Lam, Timothy Affolter, Ted William Johnson, Lars R. Engstrom, Melissa West Lu, Nathan V. Lee, Valeria Fantin, Patrick B. Lappin, Ruth W. Tang, Hui Wang, and Jennifer M. Tursi
- Subjects
Cancer Research ,Crizotinib ,medicine.drug_class ,Kinase ,Chemistry ,Cancer ,Pharmacology ,medicine.disease ,ALK inhibitor ,Oncology ,Protein kinase domain ,In vivo ,hemic and lymphatic diseases ,Cancer research ,medicine ,ROS1 ,Anaplastic lymphoma kinase ,medicine.drug - Abstract
Oncogenic fusions of Anaplastic Lymphoma Kinase (ALK) define a subset of human lung adenocarcinomas. The 1st generation ALK inhibitor XALKORI ® (crizotinib) demonstrated impressive clinical benefit in ALK-fusion positive lung cancers and was approved by the FDA for the treatment of ALK-fusion positive NSCLC in 2011. However, as seen with most kinase inhibitors, patients treated with XALKORI eventually developed resistance to therapy. Acquired ALK kinase domain mutations and brain metastases are significant contributors to the relapse after XALKORI therapy. To date, multiple types of ALK kinase domain mutations have been identified in XALKORI refractory patients including ALKG1269A, ALKL1196M, ALKC1156Y, ALKL1152R, ALKF1174L, ALKS1206Y, ALK1151Tins and ALKG1202R, accounting for about 1/3 of patient samples tested. Currently, a number of 2nd generation ALK inhibitors are under development aiming to overcome XALKORI resistant mutations. Even though in preclinical models, some ALK mutants such as ALKG1202R and ALK1151Tins confer high-levels of resistance to almost all of the 2nd generation ALK inhibitors tested. Here we report PF-06463922, a novel ATP competitive small molecule inhibitor of ALK/ROS1, with potent and selective inhibitory activity against all known acquired XALKORI resistant mutations identified in patients. PF-06463922 is also capable of penetrating the blood brain barrier in preclinical animal models. In vitro, PF-06463922 demonstrated potent inhibition in catalytic activities of ALK and 8 different ALK mutant kinases in recombinant enzyme and cell based assays (cell IC50s = 1 to 65 nM). PF-06463922 also showed potent growth inhibitory activity and induced apoptosis in the NSCLC cells harboring either non-mutant ALK or mutant ALK fusions (IC50s = 1 to 30 nM). In vivo, PF-06463922 demonstrated marked cytoreductive activity in mice bearing tumor xenografts that express EML4-ALK, EML4-ALKL1196M, EML4-ALKG1269A, EML4-ALKG1202R or NPM-ALK at low nM free plasma concentrations. These effects were associated with significant inhibition in cellular Ki67 and increased cleaved-caspase3 levels in tumors. In addition, PF-06463922 achieved brain exposure of 20-30% of its plasma levels in mice, and significantly regressed the brain tumors and prolonged survival of mice bearing orthotopic EML4-ALK and EML4-ALKL1196M positive brain tumor implants. The antitumor efficacy of PF-06463922 was dose dependent and strongly correlated with inhibition of ALK phosphorylation and downstream signaling. Our data indicate that PF-06463922 is the most potent ALK inhibitor reported to date (to our knowledge, against both non-mutant or mutant ALK in cell assays), and it demonstrates great potential for treating ALK fusion positive cancers including patients who relapsed from XALKORI therapy due to various ALK kinase domain mutations and/or brain metastases. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C253. Citation Format: Helen Y. Zou, Lars R. Engstrom, Qiuhua Li, Melissa West Lu, Ruth Wei Tang, Hui Wang, Konstantinos Tsaparikos, Jinwei Wang, Sergei Timofeevski, Dac M. Dinh, Hieu Lam, Justine Lam, Shinji Yamazaki, Wenyue Hu, Timothy Affolter, Patrick B. Lappin, Hovhannes Gukasyan, Nathan Lee, Jennifer M. Tursi, Ted W. Johnson, Valeria Fantin, Tod Smeal. PF-06463922, a novel brain-penetrating small molecule inhibitor of ALK/ROS1 with potent activity against a broad spectrum of ALK resistant mutations in preclinical models in vitro and in vivo. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C253.
- Published
- 2013
- Full Text
- View/download PDF
36. Abstract 4456: Biochemical and structural characteristics of crizotinib-resistant ALK mutants
- Author
-
Ya-Li Deng, Sergei Timofeevski, Brion W. Murray, Ben Bolaños, Wei Liu, Simon Bergqvist, Alexei Brooun, Michele McTigue, and Karlicek Shannon Marie
- Subjects
Cancer Research ,Mutation ,Crizotinib ,Kinase ,Chemistry ,Autophosphorylation ,Mutant ,medicine.disease_cause ,Molecular biology ,Oncology ,Protein kinase domain ,Biochemistry ,medicine ,Anaplastic lymphoma kinase ,Binding site ,medicine.drug - Abstract
Anaplastic lymphoma kinase (ALK), when aberrantly regulated through gene translocations or mutations, has been implicated as an oncogenic driver in a variety of cancers. XALKORI (crizotinib) is an FDA-approved therapy for locally advanced or metastatic (ALK)-positive non-small cell lung cancer (NSCLC). Mutations of certain residues within the ALK kinase domain have been associated with crizotinib resistance. To understand the molecular basis of mutational resistance, we have kinetically and structurally characterized several clinical mutations of ALK kinase domain. In the basal-state (nonphosphorylated) proteins, mutations C1156Y, F1174L, L1152R and L1196M resulted in 14 to 52-fold increase in catalytic efficiency of phosphorylation of an activation loop peptide. Accordingly, these mutants were more rapidly autophosphorylated, compared to wild-type enzyme. In addition, F1174L, C1156Y and L1156Y had 2 to 6-fold higher catalytic efficiencies when fully activated by autophosphorylation. Conversely, catalytic efficiencies of nonphosphorylated and phosphorylated G1269A variant decreased by 1.2 and 1.6-fold, respectively. Inhibition of L1152R, L1196M and G1269A by crizotinib was reduced by 3-28-fold, compared to wild-type enzyme, from Ki determinations. From direct binding studies of the L1196M “gatekeeper” mutation, the loss of crizotinib binding occurred 6 and 13-fold for phosphorylated and nonphosphorylated proteins, respectively. By crystallographic studies apoenzyme and crizotinib complexes of ALK kinase domain displayed an inactive kinase conformation that is stabilized by an extended hydrophobic network of residues. F1174 and L1196 are part of this hydrophobic core, and mutations of these residues are predicted to destabilize the inactive ALK conformation. Taken together, these results suggest that most of the resistant mutations (e.g. L1196M, F1174L, C1156Y, L1152R) result in a more dynamic protein that increases substrate turnover. In addition, the reduction of crizotinib binding for the mutations in the vicinity of the inhibitor binding site (L1196M, G1269A) is likely a contributing factor for the resistance. This structural and kinetic analysis of mutational resistance may be useful for design of new inhibitors targeting multiple clinical mutations of ALK. Citation Format: Sergei Timofeevski, Wei Liu, Ya-Li Deng, Alexei Brooun, Simon Bergqvist, Shannon Karlicek, Brion Murray, Ben Bolaños, Michele McTigue. Biochemical and structural characteristics of crizotinib-resistant ALK mutants. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4456. doi:10.1158/1538-7445.AM2013-4456
- Published
- 2013
- Full Text
- View/download PDF
37. Correction to Discovery of a Novel Class of Exquisitely Selective Mesenchymal-Epithelial Transition Factor (c-MET) Protein Kinase Inhibitors and Identification of the Clinical Candidate 2-(4-(1-(Quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (PF-04217903) for the Treatment of Cancer
- Author
-
J. Jean Cui, Michele McTigue, Mitchell Nambu, Michelle Tran-Dubé, Mason Pairish, Hong Shen, Lei Jia, Hengmiao Cheng, Jacqui Hoffman, Phuong Le, Mehran Jalaie, Gilles H. Goetz, Marcel Koenig, Tomas Vojkovsky, Fang-Jie Zhang, Steven Do, Iriny Botrous, Kevin Ryan, Neil Grodsky, Ya-li Deng, Max Parker, Sergei Timofeevski, Brion W. Murray, Shinji Yamazaki, Shirley Aguirre, Qiuhua Li, Helen Zou, and James Christensen
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2012
- Full Text
- View/download PDF
38. Abstract 2327: Structural and kinetic characterization of crizotinib with wild-type and mutant anaplastic lymphoma kinase
- Author
-
Sergei Timofeevski, Michele McTigue, Ya-Li Deng, Wei Liu, Alexei Brooun, Jean Cui, and Tami Marrone
- Subjects
Cancer Research ,Crizotinib ,Chemistry ,Kinase ,Mutant ,Gene rearrangement ,Oncology ,Biochemistry ,Protein kinase domain ,hemic and lymphatic diseases ,Cancer research ,medicine ,Anaplastic lymphoma kinase ,Kinase activity ,Tyrosine kinase ,medicine.drug - Abstract
Dysregulation of Anaplastic Lymphoma Kinase (ALK), primarily through gene translocations, has been shown to be involved in a variety of cancers. Crizotinib, an orally available small molecule inhibitor of the ALK tyrosine kinase has demonstrated marked efficacy in clinical trials of NSCLC patients harboring the EML4-ALK oncogenic gene rearrangement. Mutation of some residues within the ALK kinase domain have been reported to confer acquired or de novo resistance to crizotinib. To understand the binding of crizotinib to ALK and the mechanism of resistance of specific mutations we generated and kinetically characterized wild-type (WT) and mutant ALK kinase domains (KD). Additionally, ALK KD crystal structures were determined of the WT nonphosphorylated apoenzyme and complexes with crizotinib bound to WT and a L1196M gatekeeper mutation. No large protein conformational changes are necessary for crizotinib to bind to unliganded ALK. The interactions which crizotinib makes with ALK are similar to its binding to c-Met with the exception of notable differences in the position of the activation loop between ALK and c-Met. Mutation of the L1196 gatekeeper residue to methionine results in a ∼8-fold increase in catalytic efficiency of phosphorylation of an activation loop peptide and also more rapid enzyme auto-phosphorylation. In addition, inhibition of L1196M ALK by crizotinib was reduced ∼9-fold, compared to wild-type enzyme, from Ki determinations. The crystal structures show that L1196 or M1196 make direct contact with crizotinib. The diminished activity of crizotinib against L1196M ALK is therefore likely due to both higher intrinsic kinase activity and a subtle change in the ALK-crizotinib binding interactions. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2327. doi:10.1158/1538-7445.AM2011-2327
- Published
- 2011
- Full Text
- View/download PDF
39. Abstract LB-390: Antitumor efficacy of crizotinib (PF-02341066), a potent and selective ALK and c-Met RTK inhibitor, in EML4-ALK driven NSCLC tumors in vitro and in vivo
- Author
-
Lars D. Engstrom, Zheng Feng, Steve Bender, Patrick B. Lappin, Justine L. Lam, Joseph K. T. Lee, Sergei Timofeevski, Qiuhua Li, Brittany A. Snider, Helen Y. Zou, Timothy Nichols, Melissa West Lu, Alex Young, Tod Smeal, Shinji Yamazaki, Sonya Zabludoff, Anthony Wong, Hovhannes J. Gukasyan, James R. Christensen, Dac M. Dinh, and Jeffrey Nickel
- Subjects
Cancer Research ,C-Met ,Oncogene ,Crizotinib ,business.industry ,Kinase ,Cell growth ,Pharmacology ,chemistry.chemical_compound ,Oncology ,Mechanism of action ,chemistry ,In vivo ,hemic and lymphatic diseases ,Medicine ,medicine.symptom ,business ,Protein kinase B ,medicine.drug - Abstract
EML4-ALK fusion was recently characterized as an “Addicted Oncogene” in a subset of human lung adenocarcinoma, and it plays an essential role in regulation of the tumor cell survival, growth and metastasis. Crizotinib is a potent and selective ATP competitive small molecule inhibitor of ALK and c-Met. It is currently in clinical trials for advanced non-small cell lung cancers positive for ALK fusion. This report summarizes the pre-clinical pharmacology studies for crizotinib in the EML4-Alk positive tumor models to assess the pharmacodynamic inhibition of EML4-ALK, antitumor efficacy, PKPD relationships and antitumor mechanism of action in vitro and in vivo. Crizotinib potently inhibited the catalytic activity of ALK kinase (Ki = 0.5 nM) and the autophosphorylation of cellular EML4-ALK V1, V2, V3a and V3b with IC50 values ranging from 26–74 nM. Crizotinib also inhibited cell proliferation and induced apoptosis in NCI-H3122 human NSCLC cells harboring EML4-ALK V1 fusion with IC50s of 63 nM and 110 nM respectively. In the EML4-ALK V3a/b positive NCI-H2228 human NSCLC cells, crizotinib completely inhibited ALK phosphorylation (IC50 = 74 nM) but only partial inhibited cell proliferation and survival. This observation is consistent with the notion that only a portion of H2228 cells are EML4-ALK positive by Exon Array analysis. Crizotinib demonstrated marked tumor growth inhibition and regression in H3122 xenograft model at well tolerated dose levels. The antitumor efficacy of crizotinib was dose dependent and demonstrated a strong correlation to pharmacodynamic inhibition of ALK phosphorylation in vivo. PKPD modeling was conducted to understand the relationships between crizotinib plasma concentration to ALK target inhibition (EC50 = 19 nM) and antitumor efficacy (EC50 = 23 nM). Collectively, the results from H3122 model indicated that significant inhibition of EML4-ALK during the entire treatment period was necessary to achieve robust antitumor efficacy. Additional in vivo studies with crizotinib demonstrated dose dependent inhibition of EML4-ALK mediated signal transduction (STAT3, AKT, Erk, PLCγ, c-Myc), tumor cell proliferation (Ki67) and induction of apoptosis (caspase-3). Crizotinib also dose dependently inhibited total EML4-ALK levels in H3122 tumors indicating an additional antitumor mechanism of action by crizotinib in these tumors. Furthermore, a dose dependent increase in phospho-EGFR levels was observed after 4 days of drug treatment in H3122 model, indicating a compensation mechanism of “oncogene switching” in tumor cell signaling, and a potential resistance mechanism that may compromise “patient” responses to crizotinib treatment in these tumors. In conclusion, crizotinib was shown to be a potent inhibitor of EML4-ALK. It demonstrated marked antitumor efficacy in EML4-ALK dependent human NSCLC tumors in vitro and in vivo. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-390. doi:10.1158/1538-7445.AM2011-LB-390
- Published
- 2011
- Full Text
- View/download PDF
40. Abstract B258: Investigating PERK biological pathway using protein/peptide microarrays and SAR with small molecule inhibitors
- Author
-
Andrea Fanjul, David Briere, Jacques Ermolieff, Hubert Otlik, Terri Quenzer, Sergei Timofeevski, and Gordon Alton
- Subjects
endocrine system ,Cancer Research ,Cell signaling ,Oncology ,Biochemistry ,Kinase ,Endoplasmic reticulum ,Autophosphorylation ,Unfolded protein response ,Phosphorylation ,PERK inhibitors ,Biology ,Protein kinase A - Abstract
Accumulation of unfolded proteins in the endoplasmic occurs when the cell is subject to stress caused by various pathological conditions such as hypoxia, viral infection, and glucose depravation. Under such stress the cell will initiate an unfolded protein response (UPR), a protective mechanism that is specifically designed to re-establish homeostasis and normal endoplasmic reticulum (ER) function. This adaptive mechanism inhibits overall protein translation, but enhances the translation of a small number of key stress response proteins that will clear the ER of unfolded proteins and send them to the cytoplasm for degradation. The UPR is initiated by several proteins such as IRE1a, ATF6a and PERK, the later being a key Ser/Thr protein kinase in UPR signaling. In response to inducers of ER stress, BiP (GRP78) dissociates from the luminal ER domain of PERK, resulting in the oligomerization, autophosphorylation, and activation of PERK which in turn phosphorylates eIF-2a on Ser51 and Nrf2 (unknown site of phosphorylation). In order to further identify potential substrates for PERK, this enzyme was tested against large protein and peptide microarrays. A significant number of proteins and peptides in the microarrays were found to be phosphorylated by PERK. After additional selection, several of these substrates were further characterized using a microfluidic mobility-shift assay and submitted to LC/MS analysis to identify the site of phosphorylation by PERK. These newly identified peptide substrates were used to develop a robust biochemical assay for the testing of small molecule inhibitors of PERK activity. A limited SAR was established for a set of compounds against PERK and two other related protein kinases, GCN2 and PKR. One of the most potent and selective PERK inhibitors was found to modulate PERK cellular signaling as evidenced by blocking the phosphorylation of (Ser51)eIF-2a as judged by Elisa and Western blot analysis. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B258.
- Published
- 2009
- Full Text
- View/download PDF
41. Identification of small-molecule inhibitors of the JIP–JNK interaction.
- Author
-
Tracy Chen, Natasha Kablaoui, Jeremy Little, Sergei Timofeevski, William R. Tschantz, Ping Chen, Junli Feng, Maura Charlton, Robert Stanton, and Paul Bauer
- Subjects
ENZYME inhibitors ,PROTEIN-protein interactions ,OBESITY treatment ,INSULIN resistance ,GENETIC regulation ,TYPE 2 diabetes diagnosis ,JNK mitogen-activated protein kinases ,ENZYME activation - Abstract
JNK1 (c-Jun N-terminal kinase 1) plays a crucial role in the regulation of obesity-induced insulin resistance and is implicated in the pathology of Type 2 diabetes. Its partner, JIP1 (JNK-interacting protein 1), serves a scaffolding function that facilitates JNK1 activation by MKK4 [MAPK (mitogen-activated protein kinase) kinase 4] and MKK7 (MAPK kinase 7). For example, reduced insulin resistance and JNK activation are observed in JIP1-deficient mice. On the basis of the in vivo efficacy of a cell-permeable JIP peptide, the JIP–JNK interaction appears to be a potential target for JNK inhibition. The goal of the present study was to identify small-molecule inhibitors that disrupt the JIP–JNK interaction to provide an alternative approach for JNK inhibition to ATP-competitive inhibitors. High-throughput screening was performed by utilizing a fluorescence polarization assay that measured the binding of JNK1 to the JIP peptide. Multiple chemical series were identified, revealing two categories of JIP/JNK inhibitors: ‘dual inhibitors’ that are ATP competitive and probably inhibit JIP–JNK binding allosterically, and ‘JIP-site binders’ that block binding through interaction with the JIP site. A series of polychloropyrimidines from the second category was characterized by biochemical methods and explored through medicinal-chemistry efforts. As predicted, these inhibitors also inhibited full-length JIP–JNK binding and were selective against a panel of 34 representative kinases, including ones in the MAPK family. Overall, this work demonstrates that small molecules can inhibit protein–protein interactions in vitro in the MAPK family effectively and provides strategies for similar approaches within other target families. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.