Alan B. Northrup
Merck & Co.
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Alan B. Northrup.
Journal of Biological Chemistry | 2011
Kumiko Nagashima; Stuart D. Shumway; Sriram Sathyanarayanan; Albert H. Chen; Brian M. Dolinski; Youyuan Xu; Heike Keilhack; Thi Lien-Anh Nguyen; Maciej Wiznerowicz; Lixia Li; Bart Lutterbach; An Chi; Cloud P. Paweletz; Timothy M. Allison; Youwei Yan; Sanjeev Munshi; Anke Klippel; Manfred Kraus; Ekaterina V. Bobkova; Sujal V. Deshmukh; Zangwei Xu; Uwe Mueller; Alexander A. Szewczak; Bo-Sheng Pan; Victoria M. Richon; Roy M. Pollock; Peter Blume-Jensen; Alan B. Northrup; Jannik N. Andersen
Phosphoinositide-dependent kinase 1 (PDK1) is a critical activator of multiple prosurvival and oncogenic protein kinases and has garnered considerable interest as an oncology drug target. Despite progress characterizing PDK1 as a therapeutic target, pharmacological support is lacking due to the prevalence of nonspecific inhibitors. Here, we benchmark literature and newly developed inhibitors and conduct parallel genetic and pharmacological queries into PDK1 function in cancer cells. Through kinase selectivity profiling and x-ray crystallographic studies, we identify an exquisitely selective PDK1 inhibitor (compound 7) that uniquely binds to the inactive kinase conformation (DFG-out). In contrast to compounds 1–5, which are classical ATP-competitive kinase inhibitors (DFG-in), compound 7 specifically inhibits cellular PDK1 T-loop phosphorylation (Ser-241), supporting its unique binding mode. Interfering with PDK1 activity has minimal antiproliferative effect on cells growing as plastic-attached monolayer cultures (i.e. standard tissue culture conditions) despite reduced phosphorylation of AKT, RSK, and S6RP. However, selective PDK1 inhibition impairs anchorage-independent growth, invasion, and cancer cell migration. Compound 7 inhibits colony formation in a subset of cancer cell lines (four of 10) and primary xenograft tumor lines (nine of 57). RNAi-mediated knockdown corroborates the PDK1 dependence in cell lines and identifies candidate biomarkers of drug response. In summary, our profiling studies define a uniquely selective and cell-potent PDK1 inhibitor, and the convergence of genetic and pharmacological phenotypes supports a role of PDK1 in tumorigenesis in the context of three-dimensional in vitro culture systems.
Journal of Biological Chemistry | 2010
Ekaterina V. Bobkova; Michael Weber; Zangwei Xu; Yan-Ling Zhang; Joon Jung; Peter Blume-Jensen; Alan B. Northrup; Priya Kunapuli; Jannik N. Andersen; Ilona Kariv
The phosphoinositide 3-kinase/AKT signaling pathway plays a key role in cancer cell growth, survival, and angiogenesis. Phosphoinositide-dependent protein kinase-1 (PDK1) acts at a focal point in this pathway immediately downstream of phosphoinositide 3-kinase and PTEN, where it phosphorylates numerous AGC kinases. The PDK1 kinase domain has at least three ligand-binding sites: the ATP-binding pocket, the peptide substrate-binding site, and a groove in the N-terminal lobe that binds the C-terminal hydrophobic motif of its kinase substrates. Based on the unique PDK1 substrate recognition system, ultrahigh throughput TR-FRET and Alphascreen® screening assays were developed using a biotinylated version of the PDK1-tide substrate containing the activation loop of AKT fused to a pseudo-activated hydrophobic motif peptide. Using full-length PDK1, Km values were determined as 5.6 μm for ATP and 40 nm for the fusion peptide, revealing 50-fold higher affinity compared with the classical AKT(Thr-308)-tide. Kinetic and biophysical studies confirmed the PDK1 catalytic mechanism as a rapid equilibrium random bireactant reaction. Following an ultrahigh throughput screen of a large library, 2,000 compounds were selected from the reconfirmed hits by computational analysis with a focus on novel scaffolds. ATP-competitive hits were deconvoluted by dose-response studies at 1× and 10× Km concentrations of ATP, and specificity of binding was assessed in thermal shift assay. Inhibition studies using fusion PDK1-tide1 substrate versus AKT(Thr-308)-tide and kinase selectivity profiling revealed a novel selective alkaloid scaffold that evidently binds to the PDK1-interacting fragment pocket. Molecular modeling suggests a structural paradigm for the design of inhibitory versus activating allosteric ligands of PDK1.
Cancer Research | 2011
Katharine Ellwood-Yen; Heike Keilhack; Kaiko Kunii; Brian Dolinski; Yamicia Connor; Kun Hu; Kumiko Nagashima; Erin O'Hare; Yusuf Erkul; Alessandra Di Bacco; Diana Gargano; Nirah H. Shomer; Minilik Angagaw; Erica Leccese; Paula Andrade; Melissa S. Hurd; Myung K. Shin; Thomas F. Vogt; Alan B. Northrup; Ekaterina V. Bobkova; Shailaja Kasibhatla; Roderick T. Bronson; Martin L. Scott; Giulio Draetta; Victoria M. Richon; Nancy E. Kohl; Peter Blume-Jensen; Jannik N. Andersen; Manfred Kraus
PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.
American Journal of Respiratory Cell and Molecular Biology | 2013
Lily Y. Moy; Yanlin Jia; Michael Caniga; Gissela Lieber; Malgorzata Gil; Xiomara Fernandez; Erich Sirkowski; Richard Miller; Jessica P. Alexander; Hyun-Hee Lee; John D. Shin; J. Michael Ellis; Hongmin Chen; Alan Wilhelm; Hongshi Yu; Stella H. Vincent; Richard W. Chapman; Nancy Kelly; Emily Hickey; William M. Abraham; Alan B. Northrup; Thomas F. Miller; Hani Houshyar; Michael A. Crackower
Spleen tyrosine kinase (SYK) is a key activator of signaling pathways downstream of multiple surface receptors implicated in asthma. SYK function has been extensively studied in mast cells downstream of the high-affinity IgE receptor, FcεR1. Preclinical studies have demonstrated a role for SYK in models of allergic inflammation, but a role in airway constriction has not been demonstrated. Here, we have used a potent and selective pharmacological inhibitor of SYK to determine the role of SYK in allergen-mediated inflammation and airway constriction in preclinical models. Attenuation of allergic airway responses was evaluated in a rat passive anaphylaxis model and rat and sheep inhaled allergen challenge models, as well as an ex vivo model of allergen-mediated airway constriction in rats and cynomolgus monkeys. Pharmacological inhibition of SYK dose-dependently blocked IgE-mediated tracheal plasma extravasation in rats. In a rat ovalbumin-sensitized airway challenge model, oral dosing with an SYK inhibitor led to a dose-dependent reduction in lung inflammatory cells. Ex vivo analysis of allergen-induced airway constriction in ovalbumin-sensitized brown Norway rats showed a complete attenuation with treatment of a SYK inhibitor, as well as a complete block of allergen-induced serotonin release. Similarly, allergen-mediated airway constriction was attenuated in ex vivo studies from nonhuman primate lungs. Intravenous administration of an SYK inhibitor attenuated both early- and late-phase allergen-induced increases in airway resistance in an Ascaris-sensitive sheep allergen challenge model. These data support a key role for SYK signaling in mediating allergic airway responses.
PLOS ONE | 2011
Cloud P. Paweletz; Jannik N. Andersen; Roy Pollock; Kumiko Nagashima; Mansuo L. Hayashi; Shangshuan U. Yu; Hongbo Guo; Ekaterina V. Bobkova; Zangwei Xu; Alan B. Northrup; Peter Blume-Jensen; Ronald C. Hendrickson; An Chi
Pharmacodynamic (PD) biomarkers are an increasingly valuable tool for decision-making and prioritization of lead compounds during preclinical and clinical studies as they link drug-target inhibition in cells with biological activity. They are of particular importance for novel, first-in-class mechanisms, where the ability of a targeted therapeutic to impact disease outcome is often unknown. By definition, proximal PD biomarkers aim to measure the interaction of a drug with its biological target. For kinase drug discovery, protein substrate phosphorylation sites represent candidate PD biomarkers. However, substrate phosphorylation is often controlled by input from multiple converging pathways complicating assessment of how potently a small molecule drug hits its target based on substrate phoshorylation measurements alone. Here, we report the use of quantitative, differential mass-spectrometry to identify and monitor novel drug-regulated phosphorylation sites on target kinases. Autophosphorylation sites constitute clinically validated biomarkers for select protein tyrosine kinase inhibitors. The present study extends this principle to phosphorylation sites in serine/threonine kinases looking beyond the T-loop autophosphorylation site. Specifically, for the 3′-phosphoinositide-dependent protein kinase 1 (PDK1), two phospho-residues p-PDK1Ser410 and p-PDK1Thr513 are modulated by small-molecule PDK1 inhibitors, and their degree of dephosphorylation correlates with inhibitor potency. We note that classical, ATP-competitive PDK1 inhibitors do not modulate PDK1 T-loop phosphorylation (p-PDK1Ser241), highlighting the value of an unbiased approach to identify drug target-regulated phosphorylation sites as these are complementary to pathway PD biomarkers. Finally, we extend our analysis to another protein Ser/Thr kinase, highlighting a broader utility of our approach for identification of kinase drug-target engagement biomarkers.
Journal of Medicinal Chemistry | 2013
Alan B. Northrup; Matthew H. Katcher; Michael D. Altman; Melissa Chenard; Matthew H. Daniels; Sujal V. Deshmukh; Danielle Falcone; David J. Guerin; Harold Hatch; Chaomin Li; Wei Lu; Bart Lutterbach; Timothy J. Allison; Sangita B. Patel; John F. Reilly; Michael H. Reutershan; Keith Rickert; Craig Rosenstein; Stephen M. Soisson; Alexander A. Szewczak; Deborah Walker; Kevin J. Wilson; Jonathan R. Young; Bo Sheng Pan; Christopher J. Dinsmore
This report documents the first example of a specific inhibitor of protein kinases with preferential binding to the activated kinase conformation: 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one 11r (MK-8033), a dual c-Met/Ron inhibitor under investigation as a treatment for cancer. The design of 11r was based on the desire to reduce time-dependent inhibition of CYP3A4 (TDI) by members of this structural class. A novel two-step protocol for the synthesis of benzylic sulfonamides was developed to access 11r and analogues. We provide a rationale for the observed selectivity based on X-ray crystallographic evidence and discuss selectivity trends with additional examples. Importantly, 11r provides full inhibition of tumor growth in a c-Met amplified (GTL-16) subcutaneous tumor xenograft model and may have an advantage over inactive form kinase inhibitors due to equal potency against a panel of oncogenic activating mutations of c-Met in contrast to c-Met inhibitors without preferential binding to the active kinase conformation.
Journal of Biomolecular Screening | 2009
Zangwei Xu; Kumiko Nagashima; Dongyu Sun; Thomas S. Rush; Alan B. Northrup; Jannik N. Andersen; Ilona Kariv; Ekaterina V. Bobkova
The PI3K/Akt signaling pathway plays a key role in cancer cell growth, survival, and tumor angiogenesis. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) is a Ser/Thr protein kinase, which catalyzes the phosphorylation of a conserved residue in the activation loop of a number of AGC kinases, including proto-oncogenes Akt, p70S6K, and RSK kinases. To find new small-molecule inhibitors of this important regulator kinase, the authors have developed PDK1-specific high-throughput enzymatic assays in time-resolved fluorescence resonance energy transfer (TR-FRET) and AlphaScreen® formats, monitoring phosphorylation of a biotinylated peptide substrate derived from the activation loop of Akt. Development of homogeneous assays enabled screening of a focused kinase library of ~21,500 compounds in 1536-well TR-FRET format in duplicate. Upon validation of hits in an alternative 384-well AlphaScreen® assay, several classes of structurally diverse PDK1 inhibitors, including tetracyclics, tricyclics, azaindoles, indazoles, and indenylpyrazoles, were identified, thus confirming the utility and sensitivity of the developed assays. Further testing in PC3 prostate cancer cells confirmed that representatives of the tetracyclic series showed intracellular modulation of the PDK1 activity, as evident from decreased phosphorylation levels of AKT, RSK, and S6-ribosomal protein.
Journal of Medicinal Chemistry | 2015
J. Michael Ellis; Michael D. Altman; Alan S. Bass; John W. Butcher; Alan Byford; Anthony Donofrio; Sheila M. Galloway; Andrew M. Haidle; James P. Jewell; Nancy Kelly; Erica Leccese; Sandra Lee; Matthew L. Maddess; J. Richard Miller; Lily Y. Moy; Ekundayo Osimboni; Ryan D. Otte; M. Vijay Reddy; Kerrie Spencer; Binyuan Sun; Stella H. Vincent; Gwendolyn J. Ward; Grace H. C. Woo; Chiming Yang; Hani Houshyar; Alan B. Northrup
Development of a series of highly kinome-selective spleen tyrosine kinase (Syk) inhibitors with favorable druglike properties is described. Early leads were discovered through X-ray crystallographic analysis, and a systematic survey of cores within a selected chemical space focused on ligand binding efficiency. Attenuation of hERG ion channel activity inherent within the initial chemotype was guided through modulation of physicochemical properties including log D, PSA, and pKa. PSA proved most effective for prospective compound design. Further profiling of an advanced compound revealed bacterial mutagenicity in the Ames test using TA97a Salmonella strain, and subsequent study demonstrated that this mutagenicity was pervasive throughout the series. Identification of intercalation as a likely mechanism for the mutagenicity-enabled modification of the core scaffold. Implementation of a DNA binding assay as a prescreen and models in DNA allowed resolution of the mutagenicity risk, affording molecules with favorable potency, selectivity, pharmacokinetic, and off-target profiles.
Combinatorial Chemistry & High Throughput Screening | 2010
Dongyu Sun; Joon Jung; Thomas S. Rush; Zangwei Xu; Michael Weber; Ekaterina V. Bobkova; Alan B. Northrup; Ilona Kariv
A dynamic, focused screening strategy that utilized a limited but diversified set of target-specific compounds was explored as an efficient means for the identification of inhibitors of the protein kinase PDK1. Approximately 21,500 compounds, including a 19,000 molecule kinase-focused compound collection (KFCC), were screened at two concentrations to identify initial leads. The KFCC included several empirically-derived, general kinase libraries and molecules chosen by PDK1-specific virtual screens. As was expected, this initial screen mostly identified potent leads with limited novelty. In order to overcome this limitation, the data from the screen were used to drive several rounds of a customized iterative focused screening (IFS) campaign. A machine-learning technique was used to build a predictive model to identify compounds to be screened in subsequent rounds. Molecules deemed not to be novel were removed from the training set for the next round, which allowed this campaign to progressively walk away from the chemical space covered by the KFCC. This resulted in the identification of PDK1 inhibitors which are uniquely different from publicly known chemotypes after just three rounds of screenings. A retrospective analysis of this IFS approach against an ultra-high throughput screen (uHTS) indicated that while uHTS is still the most prolific paradigm for lead identification, this dynamic, focused screening approach was successful in discovering novel scaffolds for a medicinal chemistry effort. Finally, a theoretical optimization suggested the dynamic, focused screening approaches could provide either a complementary or alternative approach to uHTS for the efficient and rapid lead identification.
Bioorganic & Medicinal Chemistry Letters | 2017
Graham F. Smith; Michael D. Altman; Brian M. Andresen; James A. Baker; Jason Brubaker; Hongmin Chen; Yiping Chen; Matthew Lloyd Childers; Anthony Donofrio; Heidi Ferguson; Christian Fischer; Thierry O. Fischmann; Craig R. Gibeau; Alexander Hicks; Sue Jin; Sam Kattar; Melanie A. Kleinschek; Erica Leccese; Charles A. Lesburg; Chaomin Li; Jongwon Lim; Duan Liu; John Maclean; Faruk Mansoor; Lilly Y. Moy; Erin F. Mulrooney; Antoaneta S. Necheva; Larissa Rakhilina; Ruojing Yang; Luis Torres
Interleukin-1 receptor associated kinase 4 (IRAK4) has been implicated in IL-1R and TLR based signaling. Therefore selective inhibition of the kinase activity of this protein represents an attractive target for the treatment of inflammatory diseases. Medicinal chemistry optimization of high throughput screening (HTS) hits with the help of structure based drug design led to the identification of orally-bioavailable quinazoline based IRAK4 inhibitors with excellent pharmacokinetic profile and kinase selectivity. These highly selective IRAK4 compounds show activity in vivo via oral dosing in a TLR7 driven model of inflammation.