Network


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

Hotspot


Dive into the research topics where Paul E. Morin is active.

Publication


Featured researches published by Paul E. Morin.


Structure | 2012

Structures of adnectin/protein complexes reveal an expanded binding footprint.

Vidhyashankar Ramamurthy; Stanley R. Krystek; Alexander Bush; Anzhi Wei; Stuart Emanuel; Ruchira Das Gupta; Ahsen Janjua; Lin Cheng; Melissa Murdock; Bozena Abramczyk; Daniel Cohen; Zheng Lin; Paul E. Morin; Jonathan Davis; Michael Dabritz; Douglas C. McLaughlin; Katie A. Russo; Ginger Chao; Martin C. Wright; Victoria Jenny; Linda Engle; Eric Furfine; Steven Sheriff

Adnectins are targeted biologics derived from the tenth type III domain of human fibronectin (¹⁰Fn3), a member of the immunoglobulin superfamily. Target-specific binders are selected from libraries generated by diversifying the three ¹⁰Fn3 loops that are analogous to the complementarity determining regions of antibodies. The crystal structures of two Adnectins were determined, each in complex with its therapeutic target, EGFR or IL-23. Both Adnectins bind different epitopes than those bound by known monoclonal antibodies. Molecular modeling suggests that some of these epitopes might not be accessible to antibodies because of the size and concave shape of the antibody combining site. In addition to interactions from the Adnectin diversified loops, residues from the N terminus and/or the β strands interact with the target proteins in both complexes. Alanine-scanning mutagenesis confirmed the calculated binding energies of these β strand interactions, indicating that these nonloop residues can expand the available binding footprint.


Journal of Medicinal Chemistry | 2014

Tetrahydroquinoline derivatives as potent and selective factor XIa inhibitors.

Mimi L. Quan; Pancras C. Wong; Cailan Wang; Francis J. Woerner; Joanne M. Smallheer; Frank A. Barbera; Jeffrey M. Bozarth; Randi L. Brown; Mark R. Harpel; Joseph M. Luettgen; Paul E. Morin; Tara L. Peterson; Vidhyashankar Ramamurthy; Alan R. Rendina; Karen A. Rossi; Carol A. Watson; Anzhi Wei; Ge Zhang; Dietmar A. Seiffert; Ruth R. Wexler

Antithrombotic agents that are inhibitors of factor XIa (FXIa) have the potential to demonstrate robust efficacy with a low bleeding risk profile. Herein, we describe a series of tetrahydroquinoline (THQ) derivatives as FXIa inhibitors. Compound 1 was identified as a potent and selective tool compound for proof of concept studies. It exhibited excellent antithrombotic efficacy in rabbit thrombosis models and did not prolong bleeding times. This demonstrates proof of concept for the FXIa mechanism in animal models with a reversible, small molecule inhibitor.


Journal of Medicinal Chemistry | 2014

Discovery and Preclinical Characterization of the Cyclopropylindolobenzazepine BMS-791325, A Potent Allosteric Inhibitor of the Hepatitis C Virus NS5B Polymerase.

Robert G. Gentles; Min Ding; John A. Bender; Carl P. Bergstrom; Katharine A. Grant-Young; Piyasena Hewawasam; Thomas William Hudyma; Scott Martin; Andrew Nickel; Alicia Regueiro-Ren; Yong Tu; Zhong Yang; Kap-Sun Yeung; Xiaofan Zheng; Sam T. Chao; Jung-Hui Sun; Brett R. Beno; Daniel M. Camac; Mian Gao; Paul E. Morin; Steven Sheriff; Jeff Tredup; John Wan; Mark R. Witmer; Dianlin Xie; Umesh Hanumegowda; Jay O. Knipe; Kathy Mosure; Kenneth S. Santone; Dawn D. Parker

Described herein are structure-activity relationship studies that resulted in the optimization of the activity of members of a class of cyclopropyl-fused indolobenzazepine HCV NS5B polymerase inhibitors. Subsequent iterations of analogue design and syntheses successfully addressed off-target activities, most notably human pregnane X receptor (hPXR) transactivation, and led to significant improvements in the physicochemical properties of lead compounds. Those analogues exhibiting improved solubility and membrane permeability were shown to have notably enhanced pharmacokinetic profiles. Additionally, a series of alkyl bridged piperazine carboxamides was identified as being of particular interest, and from which the compound BMS-791325 (2) was found to have distinguishing antiviral, safety, and pharmacokinetic properties that resulted in its selection for clinical evaluation.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and SAR of indole-and 7-azaindole-1,3-dicarboxamide hydroxyethylamine inhibitors of BACE-1.

Mendi A. Higgins; F. Christopher Zusi; Yunhui Zhang; Michael F. Dee; Michael F. Parker; Jodi K. Muckelbauer; Daniel M. Camac; Paul E. Morin; Vidhyashankar Ramamurthy; Andrew J. Tebben; Kimberley A. Lentz; James E. Grace; Jovita Marcinkeviciene; Lisa M. Kopcho; Catherine R. Burton; Donna M. Barten; Jeremy H. Toyn; Jere E. Meredith; Charles F. Albright; Joanne J. Bronson; John E. Macor; Lorin A. Thompson

Heterocyclic replacement of the isophthalamide phenyl ring in hydroxyethylamine (HEA) BACE-1 inhibitors was explored. A variety of indole-1,3-dicarboxamide HEAs exhibited potent BACE-1 enzyme inhibition, but displayed poor cellular activity. Improvements in cellular activity and aspartic protease selectivity were observed for 7-azaindole-1,3-dicarboxamide HEAs. A methylprolinol-bearing derivative (10n) demonstrated robust reductions in rat plasma Aβ levels, but did not lower rat brain Aβ due to poor central exposure. The same analog exhibited a high efflux ratio in a bidirectional Caco-2 assay and was likely a substrate of the efflux transporter P-glycoprotein. X-ray crystal structures are reported for two indole HEAs in complex with BACE-1.


Bioorganic & Medicinal Chemistry Letters | 2011

Syntheses and initial evaluation of a series of indolo-fused heterocyclic inhibitors of the polymerase enzyme (NS5B) of the hepatitis C virus.

Xiaofan Zheng; Thomas W. Hudyma; Scott W. Martin; Carl P. Bergstrom; Min Ding; Feng He; Jeffrey L. Romine; Michael A. Poss; John F. Kadow; John Wan; Mark R. Witmer; Paul E. Morin; Daniel M. Camac; Steven Sheriff; Brett R. Beno; Karen Rigat; Ying-Kai Wang; Robert A. Fridell; Julie A. Lemm; Dike Qiu; Mengping Liu; Stacey Voss; Lenore Pelosi; Susan B. Roberts; Min Gao; Jay O. Knipe; Robert G. Gentles

Herein, we present initial SAR studies on a series of bridged 2-arylindole-based NS5B inhibitors. The introduction of bridging elements between the indole N1 and the ortho-position of the 2-aryl moiety resulted in conformationally constrained heterocycles that possess multiple additional vectors for further exploration. The binding mode and pharmacokinetic (PK) properties of select examples, including: 13-cyclohexyl-6-oxo-6,7-dihydro-5H-indolo[2,1-d][1,4]benzodiazepine-10-carboxylic acid (7) (IC(50)=0.07 μM, %F=18), are reported.


Bioorganic & Medicinal Chemistry Letters | 2008

Pyridine amides as potent and selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1

Haixia Wang; Zheming Ruan; James J. Li; Ligaya M. Simpkins; Rebecca A. Smirk; Shung C. Wu; Robert Hutchins; David S. Nirschl; Katy Van Kirk; Christopher B. Cooper; James C. Sutton; Zhengping Ma; Rajasree Golla; Ramakrishna Seethala; Mary Ellen K. Salyan; Akbar Nayeem; Stanley R. Krystek; Steven Sheriff; Daniel M. Camac; Paul E. Morin; Brian Carpenter; Jeffrey A. Robl; Robert Zahler; David A. Gordon; Lawrence G. Hamann

Several series of pyridine amides were identified as selective and potent 11beta-HSD1 inhibitors. The most potent inhibitors feature 2,6- or 3,5-disubstitution on the pyridine core. Various linkers (CH(2)SO(2), CH(2)S, CH(2)O, S, O, N, bond) between the distal aryl and central pyridyl groups are tolerated, and lipophilic amide groups are generally favored. On the distal aryl group, a number of substitutions are well tolerated. A crystal structure was obtained for a complex between 11beta-HSD1 and the most potent inhibitor in this series.


Archives of Biochemistry and Biophysics | 2003

Comparative studies of active site–ligand interactions among various recombinant constructs of human β-amyloid precursor protein cleaving enzyme

Lisa M. Kopcho; Jianhong Ma; Jovita Marcinkeviciene; Zhihong Lai; Mark R. Witmer; Janet Cheng; Joseph Yanchunas; Jeffrey Tredup; Martin J. Corbett; Deepa Calambur; Michael Wittekind; Manjula Paruchuri; Dharti Kothari; Grace Lee; Subinay Ganguly; Vidhyashankar Ramamurthy; Paul E. Morin; Daniel M. Camac; Robert W King; Amy L Lasut; O Harold Ross; Milton C Hillman; Barbara Fish; Keqiang Shen; Randine L. Dowling; Young Bun Kim; Nilsa R. Graciani; Dale Collins; Andrew P. Combs; Henry J. George

Amyloid precursor protein (APP) cleaving enzyme (BACE) is the enzyme responsible for beta-site cleavage of APP, leading to the formation of the amyloid-beta peptide that is thought to be pathogenic in Alzheimers disease (AD). Hence, BACE is an attractive pharmacological target, and numerous research groups have begun searching for potent and selective inhibitors of this enzyme as a potential mechanism for therapeutic intervention in AD. The mature enzyme is composed of a globular catalytic domain that is N-linked glycosylated in mammalian cells, a single transmembrane helix that anchors the enzyme to an intracellular membrane, and a short C-terminal domain that extends outside the phospholipid bilayer of the membrane. Here we have compared the substrate and active site-directed inhibitor binding properties of several recombinant constructs of human BACE. The constructs studied here address the importance of catalytic domain glycosylation state, inclusion of domains other than the catalytic domain, and incorporation into a membrane bilayer on the interactions of the enzyme active site with peptidic ligands. We find no significant differences in ligand binding properties among these various constructs. These data demonstrate that the nonglycosylated, soluble catalytic domain of BACE faithfully reflects the ligand binding properties of the full-length mature enzyme in its natural membrane environment. Thus, the use of the nonglycosylated, soluble catalytic domain of BACE is appropriate for studies aimed at understanding the determinants of ligand recognition by the enzyme active site.


Journal of Medicinal Chemistry | 2014

Phenylimidazoles as Potent and Selective Inhibitors of Coagulation Factor XIa with in Vivo Antithrombotic Activity

Jon J. Hangeland; Todd J. Friends; Karen A. Rossi; Joanne M. Smallheer; Cailan Wang; Zhong Sun; James R. Corte; Tianan Fang; Pancras C. Wong; Alan R. Rendina; Frank A. Barbera; Jeffrey M. Bozarth; Joseph M. Luettgen; Carol A. Watson; Ge Zhang; Anzhi Wei; Vidhyashankar Ramamurthy; Paul E. Morin; Gregory S. Bisacchi; Srinath Subramaniam; Piramanayagam Arunachalam; Arvind Mathur; Dietmar A. Seiffert; Ruth R. Wexler; Mimi L. Quan

Novel inhibitors of FXIa containing an (S)-2-phenyl-1-(4-phenyl-1H-imidazol-2-yl)ethanamine core have been optimized to provide compound 16b, a potent, reversible inhibitor of FXIa (Ki = 0.3 nM) having in vivo antithrombotic efficacy in the rabbit AV-shunt thrombosis model (ID50 = 0.6 mg/kg + 1 mg kg(-1) h(-1)). Initial analog selection was informed by molecular modeling using compounds 11a and 11h overlaid onto the X-ray crystal structure of tetrahydroquinoline 3 complexed to FXIa. Further optimization was achieved by specific modifications derived from careful analysis of the X-ray crystal structure of the FXIa/11h complex. Compound 16b was well tolerated and enabled extensive pharmacologic evaluation of the FXIa mechanism up to the ID90 for thrombus inhibition.


Bioorganic & Medicinal Chemistry Letters | 2008

Structure–activity relationships of anthranilamide-based factor Xa inhibitors containing piperidinone and pyridinone P4 moieties

James R. Corte; Tianan Fang; Donald J. P. Pinto; Wei Han; Zilun Hu; Xiangjun Jiang; Yun-Long Li; Jolicia F. Gauuan; Mark Hadden; Darren Orton; Alan R. Rendina; Joseph M. Luettgen; Pancras C. Wong; Kan He; Paul E. Morin; Daniel L. Cheney; Robert M. Knabb; Ruth R. Wexler; Patrick Y.S. Lam

Introduction of the phenyl piperidinone and phenyl pyridinone P4 moieties in the anthranilamide scaffold led to potent, selective, and orally bioavailable inhibitors of factor Xa. Anthranilamide 28 displayed comparable efficacy to apixaban in the rabbit arteriovenous-shunt (AV) thrombosis model.


ACS Medicinal Chemistry Letters | 2014

Optimization of 1,2,4-Triazolopyridines as Inhibitors of Human 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD-1).

Jun Li; Lawrence J. Kennedy; Haixia Wang; James J. Li; Steven J. Walker; Zhenqiu Hong; Stephen P. O’Connor; Akbar Nayeem; Daniel M. Camac; Paul E. Morin; Steven Sheriff; Mengmeng Wang; Timothy W. Harper; Rajasree Golla; Ramakrishna Seethala; Thomas Harrity; Randolph Ponticiello; Nathan Morgan; Joseph R. Taylor; Rachel Zebo; David A. Gordon; Jeffrey A. Robl

Small alkyl groups and spirocyclic-aromatic rings directly attached to the left side and right side of the 1,2,4-triazolopyridines (TZP), respectively, were found to be potent and selective inhibitors of human 11β-hydroxysteroid dehydrogenase-type 1 (11β-HSD-1) enzyme. 3-(1-(4-Chlorophenyl)cyclopropyl)-8-cyclopropyl-[1,2,4]triazolo[4,3-a]pyridine (9f) was identified as a potent inhibitor of the 11β-HSD-1 enzyme with reduced Pregnane-X receptor (PXR) transactivation activity. The binding orientation of this TZP series was revealed by X-ray crystallography structure studies.

Collaboration


Dive into the Paul E. Morin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge