Network


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

Hotspot


Dive into the research topics where Sujal V. Deshmukh is active.

Publication


Featured researches published by Sujal V. Deshmukh.


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 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.


Journal of Medicinal Chemistry | 2013

Discovery of 1-[3-(1-Methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5]cyclohepta[1,2-b]pyridin-7-yl]-N-(pyridin-2-ylmethyl)methanesulfonamide (MK-8033): A Specific c-Met/Ron Dual Kinase Inhibitor with Preferential Affinity for the Activated State of c-Met

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 Pharmacological and Toxicological Methods | 2011

Direct determination of the ratio of unbound fraction in plasma to unbound fraction in microsomal system (fup/fumic) for refined prediction of phase I mediated metabolic hepatic clearance

Sujal V. Deshmukh; Andreas Harsch

At the drug discovery stage, in vivo metabolic hepatic clearance (CL(hep)) is commonly predicted using in vitro parent compound disappearance data generated in liver microsomes or hepatocytes. Correction for the unbound fraction of a compound in the in vitro system and in plasma/serum is known to be critical for the accuracy of metabolic clearance predictions. Discrete generation of these required experimental parameters can be laborious. Herein, we describe a straightforward and direct approach to obtain the ratio of unbound fraction in plasma (fu(p)) to unbound fraction in the microsomal system (fu(mic)) of a small molecule compound using equilibrium dialysis. Experimental conditions were optimized with respect to incubation time, temperature, and plate shaking speed. Results obtained from this system were validated for a set of test compounds by comparison to individually measured fu(p) and fu(mic) data using ultracentrifugation. The correlation for fu(p)/fu(mic) between the two methods for a set of 23 data points was very good with R(2) of 0.94, slope of 1.05 and an intercept of 0.007. The impact of microsomal binding on predicted CL(hep) was illustrated for a tightly bound compound using a series of incubations with increasing concentration of monkey liver microsomal protein. Alteration of this experimental parameter profoundly affected calculated CL(hep) using the well-stirred model. Significant differences were observed in the prediction when the model was corrected for fu(p) only; in contrast, the model corrected for plasma protein and microsomal protein binding predicted clearance values independent of the microsomal protein concentration.


Bioorganic & Medicinal Chemistry Letters | 2015

Potent benzoazepinone γ-secretase modulators with improved bioavailability.

Joey L. Methot; Christian Fischer; Chaomin Li; Alexey Rivkin; Sean P. Ahearn; William Colby Brown; Sam Kattar; Elizabeth Helen Kelley; Dawn M. Mampreian; Adam J. Schell; Andrew Rosenau; Hua Zhou; Richard G. Ball; Sujal V. Deshmukh; Valentina V. Jeliazkova-Mecheva; Damaris Diaz; Lily Y. Moy; Candia M. Kenific; Chris Moxham; Sanjiv Shah; Hugh Nuthall; Alexander A. Szewczak; Armetta D. Hill; Bethany Hughes; Nadya Smotrov; Benito Munoz; Thomas A. Miller; Mark S. Shearman

The triazolyl amide γ-secretase modulators are potent alternatives to the cinnamyl amides that have entered the clinic for the treatment of Alzheimers disease. Herein we build on the lead benzoazepinones described in our prior communication with imidazomethoxyarene moiety alternatives that offer opportunities to fine tune physical properties as well as address hERG binding and PK. Both half-life and bioavailability were significantly improved, especially in dog, with robust brain Aβ42 lowering maintained in both transgenic mouse and rat.


Bioorganic & Medicinal Chemistry Letters | 2012

Lead optimization of 4,4-biaryl piperidine amides as γ-secretase inhibitors.

Joshua Close; Richard Heidebrecht; John Hendrix; Chaomin Li; Ben Munoz; Laura Surdi; Solomon Kattar; Paul Tempest; Paul Moses; Xiaoliu Geng; Bethany Hughes; Nadya Smotrov; Chris Moxham; Jennifer Chapnick; Ilona Kariv; George Nikov; Julie Elizabeth Burke; Sujal V. Deshmukh; Valentina V. Jeliazkova-Mecheva; John Kevin Leach; Damaris Diaz; Lin Xu; Ziping Yang; Gloria Y. Kwei; Lily Y. Moy; Sanjiv Shah; Flobert Tanga; Candia Kenefic; Dan Savage; Mark S. Shearman

Alzheimers disease is a major unmet medical need with pathology characterized by extracellular proteinaceous plaques comprised primarily of β-amyloid. γ-Secretase is a critical enzyme in the cellular pathway responsible for the formation of a range of β-amyloid peptides; one of which, Aβ42, is believed to be responsible for the neuropathological features of the disease. Herein, we report 4,4 disubstituted piperidine γ-secretase inhibitors that were optimized for in vitro cellular potency and pharmacokinetic properties in vivo. Key agents were further characterized for their ability to lower cerebral Aβ42 production in an APP-YAC mouse model. This structural series generally suffered from sub-optimal pharmacokinetics but hypothesis driven lead optimization enabled the discovery of γ-secretase inhibitors capable of lowering cerebral Aβ42 production in mice.


Journal of Pharmacology and Experimental Therapeutics | 2017

Evaluation of JAK3 biology in autoimmune disease using a highly selective, irreversible JAK3 inhibitor

Fiona Elwood; David J. Witter; Jennifer Piesvaux; Brian Kraybill; Nathan Bays; Carla Alpert; Peter Goldenblatt; Yujie Qu; Irena Ivanovska; Hyun-Hee Lee; Chi-Sung Chiu; Hao Tang; Mark E. Scott; Sujal V. Deshmukh; Mark Zielstorff; Alan Byford; Kalyan Chakravarthy; Lauren Dorosh; Alexey Rivkin; Joel A. Klappenbach; Bo-Sheng Pan; Ilona Kariv; Christopher J. Dinsmore; Deborah Slipetz; Peter J. Dandliker

Reversible janus associated kinase (JAK) inhibitors such as tofacitinib and decernotinib block cytokine signaling and are efficacious in treating autoimmune diseases. However, therapeutic doses are limited due to inhibition of other JAK/signal transducer and activator of transcription pathways associated with hematopoiesis, lipid biogenesis, infection, and immune responses. A selective JAK3 inhibitor may have a better therapeutic index; however, until recently, no compounds have been described that maintain JAK3 selectivity in cells, as well as against the kinome, with good physicochemical properties to test the JAK3 hypothesis in vivo. To quantify the biochemical basis for JAK isozyme selectivity, we determined that the apparent Km value for each JAK isozyme ranged from 31.8 to 2.9 μM for JAK1 and JAK3, respectively. To confirm compound activity in cells, we developed a novel enzyme complementation assay that read activity of single JAK isozymes in a cellular context. Reversible JAK3 inhibitors cannot achieve sufficient selectivity against other isozymes in the cellular context due to inherent differences in enzyme ATP Km values. Therefore, we developed irreversible JAK3 compounds that are potent and highly selective in vitro in cells and against the kinome. Compound 2, a potent inhibitor of JAK3 (0.15 nM) was 4300-fold selective for JAK3 over JAK1 in enzyme assays, 67-fold [interleukin (IL)-2 versus IL-6] or 140-fold [IL-2 versus erythropoietin or granulocyte-macrophage colony-stimulating factor (GMCSF)] selective in cellular reporter assays and >35-fold selective in human peripheral blood mononuclear cell assays (IL-7 versus IL-6 or GMCSF). In vivo, selective JAK3 inhibition was sufficient to block the development of inflammation in a rat model of rheumatoid arthritis, while sparing hematopoiesis.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of novel triazolobenzazepinones as γ-secretase modulators with central Aβ42 lowering in rodents and rhesus monkeys.

Christian Fischer; Susan L. Zultanski; Hua Zhou; Joey L. Methot; Sanjiv Shah; Ikuo Hayashi; Bethany Hughes; Christopher M. Moxham; Nathan Bays; Nadya Smotrov; Armetta D. Hill; Bo-Sheng Pan; Zhenhua Wu; Lily Y. Moy; Flobert Tanga; Candia M. Kenific; Jonathan C. Cruz; Deborah Walker; Melanie Bouthillette; George Nikov; Sujal V. Deshmukh; Valentina V. Jeliazkova-Mecheva; Damaris Diaz; Maria S. Michener; Jacquelynn J. Cook; Benito Munoz; Mark S. Shearman

Synthesis and SAR studies of novel triazolobenzazepinones as gamma secretase modulators (GSMs) are presented in this communication. Starting from our azepinone leads, optimization studies toward improving central lowering of Aβ42 led to the discovery of novel benzo-fused azepinones. Several benzazepinones were profiled in vivo and found to lower brain Aβ42 levels in Sprague Dawley rats and transgenic APP-YAC mice in a dose-dependent manner after a single oral dose. Compound 34 was further progressed into a pilot study in our cisterna-magna-ported rhesus monkey model, where we observed robust lowering of CSF Aβ42 levels.


Journal of Medicinal Chemistry | 2017

The Discovery of 3-((4-Chloro-3-methoxyphenyl)amino)-1-((3R,4S)-4-cyanotetrahydro-2H-pyran-3-yl)-1H-pyrazole-4-carboxamide, a Highly Ligand Efficient and Efficacious Janus Kinase 1 Selective Inhibitor with Favorable Pharmacokinetic Properties

Tony Siu; Jason Brubaker; Peter Fuller; Luis Torres; Hongbo Zeng; Joshua Close; Dawn M. Mampreian; Feng Shi; Duan Liu; Xavier Fradera; Kevin Johnson; Nathan Bays; Elma Kadic; Fang He; Peter Goldenblatt; Lynsey Shaffer; Sangita B. Patel; Charles A. Lesburg; Carla Alpert; Lauren Dorosh; Sujal V. Deshmukh; Hongshi Yu; Joel A. Klappenbach; Fiona Elwood; Christopher J. Dinsmore; Rafael Fernández; Lily Y. Moy; Jonathan R. Young

The discovery of a potent selective low dose Janus kinase 1 (JAK1) inhibitor suitable for clinical evaluation is described. As part of an overall goal to minimize dose, we pursued a medicinal chemistry strategy focused on optimization of key parameters that influence dose size, including lowering human Clint and increasing intrinsic potency, bioavailability, and solubility. To impact these multiple parameters simultaneously, we used lipophilic ligand efficiency as a key metric to track changes in the physicochemical properties of our analogs, which led to improvements in overall compound quality. In parallel, structural information guided advancements in JAK1 selectivity by informing on new vector space, which enabled the discovery of a unique key amino acid difference between JAK1 (Glu966) and JAK2 (Asp939). This difference was exploited to consistently produce analogs with the best balance of JAK1 selectivity, efficacy, and projected human dose, ultimately culminating in the discovery of compound 28.

Collaboration


Dive into the Sujal V. Deshmukh's collaboration.

Top Co-Authors

Avatar

Mahmoud S. Ahmed

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Tatiana Nanovskaya

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Bo-Sheng Pan

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary D.V. Hankins

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge