Network


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

Hotspot


Dive into the research topics where Michael D. Altman is active.

Publication


Featured researches published by Michael D. Altman.


Molecular Endocrinology | 2009

Molecular mechanism of action of pharmacoperone rescue of misrouted GPCR mutants: the GnRH receptor.

Jo Ann Janovick; Akshay Patny; Ralph T. Mosley; Mark T. Goulet; Michael D. Altman; Thomas S. Rush; Anda Cornea; P. Michael Conn

The human GnRH receptor (hGnRHR), a G protein-coupled receptor, is a useful model for studying pharmacological chaperones (pharmacoperones), drugs that rescue misfolded and misrouted protein mutants and restore them to function. This technique forms the basis of a therapeutic approach of rescuing mutants associated with human disease and restoring them to function. The present study relies on computational modeling, followed by site-directed mutagenesis, assessment of ligand binding, effector activation, and confocal microscopy. Our results show that two different chemical classes of pharmacoperones act to stabilize hGnRHR mutants by bridging residues D(98) and K(121). This ligand-mediated bridge serves as a surrogate for a naturally occurring and highly conserved salt bridge (E(90)-K(121)) that stabilizes the relation between transmembranes 2 and 3, which is required for passage of the receptor through the cellular quality control system and to the plasma membrane. Our model was used to reveal important pharmacophoric features, and then identify a novel chemical ligand, which was able to rescue a D(98) mutant of the hGnRHR that could not be rescued as effectively by previously known pharmacoperones.


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.


Bioorganic & Medicinal Chemistry Letters | 2010

The discovery of tricyclic pyridone JAK2 inhibitors. Part 1: hit to lead.

Tony Siu; Ekaterina Kozina; Joon Jung; Craig Rosenstein; Anjili Mathur; Michael D. Altman; Grace Chan; Lin Xu; Eric Bachman; Jan-Rung Mo; Melaney Bouthillette; Thomas S. Rush; Christopher J. Dinsmore; C. Gary Marshall; Jonathan R. Young

This paper describes the discovery and design of a novel class of JAK2 inhibitors. Furthermore, we detail the optimization of a screening hit using ligand binding efficiency and log D. These efforts led to the identification of compound 41, which demonstrates in vivo activity in our study.


Journal of Medicinal Chemistry | 2015

Overcoming mutagenicity and ion channel activity: optimization of selective spleen tyrosine kinase inhibitors.

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.


Bioorganic & Medicinal Chemistry Letters | 2015

Identification of N-(1H-pyrazol-4-yl)carboxamide inhibitors of interleukin-1 receptor associated kinase 4: Bicyclic core modifications

Jongwon Lim; Michael D. Altman; James A. Baker; Jason Brubaker; Hongmin Chen; Yiping Chen; Melanie A. Kleinschek; Chaomin Li; Duan Liu; John Maclean; Erin F. Mulrooney; Larissa Rakhilina; Graham F. Smith; Ruojing Yang

IRAK4 plays a critical role in the IL-1R and TLR signalling, and selective inhibition of the kinase activity of the protein represents an attractive target for the treatment of inflammatory diseases. A series of permeable N-(1H-pyrazol-4-yl)carboxamides was developed by introducing lipophilic bicyclic cores in place of the polar pyrazolopyrimidine core of 5-amino-N-(1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamides. Replacement of the pyrazolo[1,5-a]pyrimidine core with the pyrrolo[2,1-f][1,2,4]triazine, the pyrrolo[1,2-b]pyridazine, and thieno[2,3-b]pyrazine cores guided by cLogD led to the identification of highly permeable IRAK4 inhibitors with excellent potency and kinase selectivity.


Bioorganic & Medicinal Chemistry Letters | 2014

Thiophene carboxamide inhibitors of JAK2 as potential treatments for myleoproliferative neoplasms

Andrew M. Haidle; Anna A. Zabierek; Kaleen Konrad Childers; Craig Rosenstein; Anjili Mathur; Michael D. Altman; Grace Chan; Lin Xu; Eric Bachman; Jan-Rung Mo; Melaney Bouthillette; Thomas S. Rush; Paul Tempest; C. Gary Marshall; Jonathan R. Young

A series of carboxamide-substituted thiophenes demonstrating inhibition of JAK2 is described. Development of this chemical series began with the bioisosteric replacement of a urea substituent by a pyridyl ring. Issues of chemical and metabolic stability were solved using the results of both in vitro and in vivo studies, ultimately delivering compounds such as 24 and 25 that performed well in an acute PK/PD model measuring p-STAT5 inhibition.


Bioorganic & Medicinal Chemistry Letters | 2017

Identification of quinazoline based inhibitors of IRAK4 for the treatment of inflammation.

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.


Journal of Biomolecular Screening | 2016

Development of a High-Throughput Gene Expression Screen for Modulators of RAS-MAPK Signaling in a Mutant RAS Cellular Context

Bryan Severyn; Thi Nguyen; Michael D. Altman; Lixia Li; Kumiko Nagashima; George Naumov; Sriram Sathyanarayanan; Erica Cook; Erick J. Morris; Marc Ferrer; Bill Arthur; Yair Benita; James Watters; Andrey Loboda; Jeff Hermes; D. Gary Gilliland; Michelle A. Cleary; Pamela M. Carroll; Peter Strack; Matt Tudor; Jannik N. Andersen

The RAS-MAPK pathway controls many cellular programs, including cell proliferation, differentiation, and apoptosis. In colorectal cancers, recurrent mutations in this pathway often lead to increased cell signaling that may contribute to the development of neoplasms, thereby making this pathway attractive for therapeutic intervention. To this end, we developed a 26-member gene signature of RAS-MAPK pathway activity utilizing the Affymetrix QuantiGene Plex 2.0 reagent system and performed both primary and confirmatory gene expression–based high-throughput screens (GE-HTSs) using KRAS mutant colon cancer cells (SW837) and leveraging a highly annotated chemical library. The screen achieved a hit rate of 1.4% and was able to enrich for hit compounds that target RAS-MAPK pathway members such as MEK and EGFR. Sensitivity and selectivity performance measurements were 0.84 and 1.00, respectively, indicating high true-positive and true-negative rates. Active compounds from the primary screen were confirmed in a dose–response GE-HTS assay, a GE-HTS assay using 14 additional cancer cell lines, and an in vitro colony formation assay. Altogether, our data suggest that this GE-HTS assay will be useful for larger unbiased chemical screens to identify novel compounds and mechanisms that may modulate the RAS-MAPK pathway.


Bioorganic & Medicinal Chemistry Letters | 2017

Structure guided design of a series of selective pyrrolopyrimidinone MARK inhibitors

Jason D. Katz; Andrew M. Haidle; Kaleen Konrad Childers; Anna A. Zabierek; James P. Jewell; Yongquan Hou; Michael D. Altman; Alexander A. Szewczak; Dapeng Chen; Andreas Harsch; Mansuo L. Hayashi; Lee Warren; Michael Hutton; Hugh Nuthall; Hua-Poo Su; Sanjeev Munshi; Matt G. Stanton; Ian W. Davies; Ben Munoz; Alan B. Northrup

The initial structure activity relationships around an isoindoline uHTS hit will be described. Information gleaned from ligand co-crystal structures allowed for rapid refinements in both MARK potency and kinase selectivity. These efforts allowed for the identification of a compound with properties suitable for use as an in vitro tool compound for validation studies on MARK as a viable target for Alzheimers disease.


Bioorganic & Medicinal Chemistry Letters | 2017

MARK inhibitors: Declaring a No-Go decision on a chemical series based on extensive DMPK experimentation

Andrew M. Haidle; Kaleen Konrad Childers; Anna A. Zabierek; Jason D. Katz; James P. Jewell; Yongquan Hou; Michael D. Altman; Alexander A. Szewczak; Dapeng Chen; Andreas Harsch; Mansuo L. Hayashi; Lee Warren; Michael Hutton; Hugh Nuthall; Matt G. Stanton; Ian W. Davies; Ben Munoz; Alan B. Northrup

Attempts to optimize pharmacokinetic properties in a promising series of pyrrolopyrimidinone MARK inhibitors for the treatment of Alzheimers disease are described. A focus on physical properties and ligand efficiency while prosecuting this series afforded key tool compounds that revealed a large discrepancy in the rat in vitro-in vivo DMPK (Drug Metabolism/Pharmacokinetics) correlation. These differences prompted an in vivo rat disposition study employing a radiolabeled representative of the series, and the results from this experiment justified the termination of any further optimization efforts.

Researchain Logo
Decentralizing Knowledge