Network


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

Hotspot


Dive into the research topics where Alexander A. Szewczak is active.

Publication


Featured researches published by Alexander A. Szewczak.


Bioorganic & Medicinal Chemistry Letters | 2008

Exploration of the internal cavity of histone deacetylase (HDAC) with selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Prasun K. Chakravarty; Melissa Chenard; Joshua Close; Jonathan C. Cruz; William K. Dahlberg; Judith C. Fleming; Christopher Hamblett; Julie E. Hamill; Paul Harrington; Andreas Harsch; Richard Heidebrecht; Bethany Hughes; Joon Jung; Candia M. Kenific; Astrid M. Kral; Peter T. Meinke; Richard E. Middleton; Nicole Ozerova; David L. Sloman; Matthew G. Stanton; Alexander A. Szewczak; Sriram Tyagarajan; David J. Witter; J. Paul Secrist; Thomas A. Miller

We report herein the initial exploration of novel selective HDAC1/HDAC2 inhibitors (SHI-1:2). Optimized SHI-1:2 structures exhibit enhanced intrinsic activity against HDAC1 and HDAC2, and are greater than 100-fold selective versus other HDACs, including HDAC3. Based on the SAR of these agents and our current understanding of the HDAC active site, we postulate that the SHI-1:2 extend the existing HDAC inhibitor pharmacophore to include an internal binding domain.


Cancer Research | 2010

MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor

Bo-Sheng Pan; Grace K.Y. Chan; Melissa Chenard; An Chi; Lenora Davis; Sujal V. Deshmukh; Jackson B. Gibbs; Susana Gil; Gaozhen Hang; Harold Hatch; James P. Jewell; Ilona Kariv; Jason D. Katz; Kaiko Kunii; Wei Lu; Bart Lutterbach; Cloud P. Paweletz; Xianlu Qu; John F. Reilly; Alexander A. Szewczak; Qinwen Zeng; Nancy E. Kohl; Christopher J. Dinsmore

The receptor tyrosine kinase c-Met is an attractive target for therapeutic blockade in cancer. Here, we describe MK-2461, a novel ATP-competitive multitargeted inhibitor of activated c-Met. MK-2461 inhibited in vitro phosphorylation of a peptide substrate recognized by wild-type or oncogenic c-Met kinases (N1100Y, Y1230C, Y1230H, Y1235D, and M1250T) with IC(50) values of 0.4 to 2.5 nmol/L. In contrast, MK-2461 was several hundredfold less potent as an inhibitor of c-Met autophosphorylation at the kinase activation loop. In tumor cells, MK-2461 effectively suppressed constitutive or ligand-induced phosphorylation of the juxtamembrane domain and COOH-terminal docking site of c-Met, and its downstream signaling to the phosphoinositide 3-kinase-AKT and Ras-extracellular signal-regulated kinase pathways, without inhibiting autophosphorylation of the c-Met activation loop. BIAcore studies indicated 6-fold tighter binding to c-Met when it was phosphorylated, suggesting that MK-2461 binds preferentially to activated c-Met. MK-2461 displayed significant inhibitory activities against fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor, and other receptor tyrosine kinases. In cell culture, MK-2461 inhibited hepatocyte growth factor/c-Met-dependent mitogenesis, migration, cell scatter, and tubulogenesis. Seven of 10 MK-2461-sensitive tumor cell lines identified from a large panel harbored genomic amplification of MET or FGFR2. In a murine xenograft model of c-Met-dependent gastric cancer, a well-tolerated oral regimen of MK-2461 administered at 100 mg/kg twice daily effectively suppressed c-Met signaling and tumor growth. Similarly, MK-2461 inhibited the growth of tumors formed by s.c. injection of mouse NIH-3T3 cells expressing oncogenic c-Met mutants. Taken together, our findings support further preclinical development of MK-2461 for cancer therapy.


Journal of Medicinal Chemistry | 2011

Discovery of 1-Amino-5H-pyrido[4,3-b]indol-4-carboxamide Inhibitors of Janus Kinase 2 (JAK2) for the Treatment of Myeloproliferative Disorders

Jongwon Lim; Brandon M. Taoka; Ryan D. Otte; Kerrie Spencer; Christopher J. Dinsmore; Michael D. Altman; Grace Chan; Craig Rosenstein; Sujata Sharma; Hua-Poo Su; Alexander A. Szewczak; Lin Xu; Hong Yin; Joan Zugay-Murphy; C. Gary Marshall; Jonathan R. Young

The JAK-STAT pathway mediates signaling by cytokines, which control survival, proliferation, and differentiation of a variety of cells. In recent years, a single point mutation (V617F) in the tyrosine kinase JAK2 was found to be present with a high incidence in myeloproliferative disorders (MPDs). This mutation led to hyperactivation of JAK2, cytokine-independent signaling, and subsequent activation of downstream signaling networks. The genetic, biological, and physiological evidence suggests that JAK2 inhibitors could be effective in treating MPDs. De novo design efforts of new scaffolds identified 1-amino-5H-pyrido[4,3-b]indol-4-carboxamides as a new viable lead series. Subsequent optimization of cell potency, metabolic stability, and off-target activities of the leads led to the discovery of 7-(2-aminopyrimidin-5-yl)-1-{[(1R)-1-cyclopropyl-2,2,2-trifluoroethyl]amino}-5H-pyrido[4,3-b]indole-4-carboxamide (65). Compound 65 is a potent, orally active inhibitor of JAK2 with excellent selectivity, PK profile, and in vivo efficacy in animal models.


Journal of Biological Chemistry | 2011

Genetic and Pharmacological Inhibition of PDK1 in Cancer Cells CHARACTERIZATION OF A SELECTIVE ALLOSTERIC KINASE INHIBITOR

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 Medicinal Chemistry | 2011

Discovery of a 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one (MK-2461) inhibitor of c-Met kinase for the treatment of cancer.

Jason D. Katz; James P. Jewell; David J. Guerin; Jongwon Lim; Christopher J. Dinsmore; Sujal V. Deshmukh; Bo-Sheng Pan; C. Gary Marshall; Wei Lu; Michael D. Altman; William K. Dahlberg; Lenora Davis; Danielle Falcone; Ana E. Gabarda; Gaozhen Hang; Harold Hatch; Rachael Holmes; Kaiko Kunii; Kevin J. Lumb; Bart Lutterbach; Robert J. Mathvink; Naim Nazef; Sangita B. Patel; Xianlu Qu; John Reilly; Keith Rickert; Craig Rosenstein; Stephen M. Soisson; Kerrie Spencer; Alexander A. Szewczak

c-Met is a transmembrane tyrosine kinase that mediates activation of several signaling pathways implicated in aggressive cancer phenotypes. In recent years, research into this area has highlighted c-Met as an attractive cancer drug target, triggering a number of approaches to disrupt aberrant c-Met signaling. Screening efforts identified a unique class of 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one kinase inhibitors, exemplified by 1. Subsequent SAR studies led to the development of 81 (MK-2461), a potent inhibitor of c-Met that was efficacious in preclinical animal models of tumor suppression. In addition, biochemical studies and X-ray analysis have revealed that this unique class of kinase inhibitors binds preferentially to the activated (phosphorylated) form of the kinase. This report details the development of 81 and provides a description of its unique biochemical properties.


Bioorganic & Medicinal Chemistry Letters | 2010

Piperazinyl pyrimidine derivatives as potent γ-secretase modulators

Alexey Rivkin; Sean P. Ahearn; Stephanie M. Chichetti; Yoona R. Kim; Chaomin Li; Andrew Rosenau; Sam Kattar; Joon Jung; Sanjiv Shah; Bethany Hughes; Jamie L. Crispino; Richard E. Middleton; Alexander A. Szewczak; Benito Munoz; Mark S. Shearman

The development of a novel series of piperazinyl pyrimidines as gamma-secretase modulators for potential use in the treatment of Alzheimers disease is disclosed herein. Optimization of a screening hit provided a series of potent gamma-secretase modulators with >180-fold in vitro selectivity over inhibition of Notch cleavage.


Bioorganic & Medicinal Chemistry Letters | 2010

Purine derivatives as potent γ-secretase modulators

Alexey Rivkin; Sean P. Ahearn; Stephanie M. Chichetti; Christopher Hamblett; Yudith Garcia; Michelle Martinez; Jed L. Hubbs; Michael H. Reutershan; Matthew H. Daniels; Phieng Siliphaivanh; Karin M. Otte; Chaomin Li; Andrew Rosenau; Laura Surdi; Joon Jung; Bethany Hughes; Jamie L. Crispino; George Nikov; Richard E. Middleton; Christopher M. Moxham; Alexander A. Szewczak; Sanjiv Shah; Lily Y. Moy; Candia M. Kenific; Flobert Tanga; Jonathan C. Cruz; Paula Andrade; Minilik Angagaw; Nirah H. Shomer; Thomas A. Miller

The development of a novel series of purines as gamma-secretase modulators for potential use in the treatment of Alzheimers disease is disclosed herein. Optimization of a previously disclosed pyrimidine series afforded a series of potent purine-based gamma-secretase modulators with 300- to 2000-fold in vitro selectivity over inhibition of Notch cleavage and that selectively reduces Alphabeta42 in an APP-YAC transgenic mouse model.


Bioorganic & Medicinal Chemistry Letters | 2008

SAR profiles of spirocyclic nicotinamide derived selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Christopher Hamblett; Dawn M. Mampreian; Joon Jung; Andreas Harsch; Alexander A. Szewczak; William K. Dahlberg; Richard E. Middleton; Bethany Hughes; Judith C. Fleming; Hongmei Wang; Astrid M. Kral; Nicole Ozerova; Jonathan C. Cruz; Brian B. Haines; Melissa Chenard; Candia M. Kenific; J. Paul Secrist; Thomas A. Miller

A potent family of spirocyclic nicotinyl aminobenzamide selective HDAC1/HDAC2 inhibitors (SHI-1:2) is profiled. The incorporation of a biaryl zinc-binding motif into a nicotinyl scaffold resulted in enhanced potency and selectivity versus HDAC3, but also imparted hERG activity. It was discovered that increasing polar surface area about the spirocycle attenuates this liability. Compound 12 induced a 4-fold increase in acetylated histone H2B in an HCT-116 xenograft model study with acute exposure, and inhibited tumor growth in a 21-day efficacy study with qd dosing.


Bioorganic & Medicinal Chemistry Letters | 2010

Fluorinated piperidine acetic acids as γ-secretase modulators

Matthew G. Stanton; Jed L. Hubbs; David L. Sloman; Christopher Hamblett; Paula Andrade; Minilik Angagaw; Grace Bi; Regina M. Black; Jamie L. Crispino; Jonathan C. Cruz; Eric Fan; Georgia Farris; Bethany Hughes; Candia M. Kenific; Richard E. Middleton; George Nikov; Peter Sajonz; Sanjiv Shah; Nirah H. Shomer; Alexander A. Szewczak; Flobert Tanga; Matthew T. Tudge; Mark S. Shearman; Benito Munoz

We report herein a novel series of difluoropiperidine acetic acids as modulators of gamma-secretase. Synthesis of 2-aryl-3,3-difluoropiperidine analogs was facilitated by a unique and selective beta-difluorination with Selectfluor. Compounds 1f and 2c were selected for in vivo assessment and demonstrated selective lowering of Abeta42 in a genetically engineered mouse model of APP processing. Moreover, in a 7-day safety study, rats treated orally with compound 1f (250mg/kg per day, AUC(0-24)=2100microMh) did not exhibit Notch-related effects.


Bioorganic & Medicinal Chemistry Letters | 2008

Phenylglycine and phenylalanine derivatives as potent and selective HDAC1 inhibitors (SHI-1)

Kevin J. Wilson; David J. Witter; Jonathan Grimm; Phieng Siliphaivanh; Karin M. Otte; Astrid M. Kral; Judith C. Fleming; Andreas Harsch; Julie E. Hamill; Jonathan C. Cruz; Melissa Chenard; Alexander A. Szewczak; Richard E. Middleton; Bethany Hughes; William K. Dahlberg; J. Paul Secrist; Thomas A. Miller

An HTS screening campaign identified a series of low molecular weight phenols that showed excellent selectivity (>100-fold) for HDAC1/HDAC2 over other Class I and Class II HDACs. Evolution and optimization of this HTS hit series provided HDAC1-selective (SHI-1) compounds with excellent anti-proliferative activity and improved physical properties. Dose-dependent efficacy in a mouse HCT116 xenograft model was demonstrated with a phenylglycine SHI-1 analog.

Researchain Logo
Decentralizing Knowledge