Network


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

Hotspot


Dive into the research topics where Joel A. Cassel is active.

Publication


Featured researches published by Joel A. Cassel.


Journal of Medicinal Chemistry | 2008

Potent, Orally Bioavailable Delta Opioid Receptor Agonists for the Treatment of Pain : Discovery of N,N-Diethyl-4-(5-hydroxyspiro-[chromene-2,4'-piperidine]-4-yl)benzamide (ADL5859)

Bertrand Le Bourdonnec; Rolf T. Windh; Christopher W. Ajello; Lara K. Leister; Minghua Gu; Guo-Hua Chu; Paul A. Tuthill; William M. Barker; Michael Koblish; Daniel D. Wiant; Thomas M. Graczyk; Serge Belanger; Joel A. Cassel; Marina S. Feschenko; Bernice L. Brogdon; Steven A. Smith; David D. Christ; Michael J. Derelanko; Steve Kutz; Patrick J. Little; Robert N. DeHaven; Diane L. DeHaven-Hudkins; Roland E. Dolle

Selective delta opioid receptor agonists are promising potential therapeutic agents for the treatment of various types of pain conditions. A spirocyclic derivative was identified as a promising hit through screening. Subsequent lead optimization identified compound 20 (ADL5859) as a potent, selective, and orally bioavailable delta agonist. Compound 20 was selected as a clinical candidate for the treatment of pain.


Journal of Medicinal Chemistry | 2009

Spirocyclic Delta Opioid Receptor Agonists for the Treatment of Pain: Discovery of N,N-Diethyl-3-hydroxy-4-(spiro[chromene-2,4'-piperidine]-4-yl) Benzamide (ADL5747)

Bertrand Le Bourdonnec; Rolf T. Windh; Lara K. Leister; Q. Jean Zhou; Christopher W. Ajello; Minghua Gu; Guo-Hua Chu; Paul A. Tuthill; William M. Barker; Michael Koblish; Daniel D. Wiant; Thomas M. Graczyk; Serge Belanger; Joel A. Cassel; Marina S. Feschenko; Bernice L. Brogdon; Steven A. Smith; Michael J. Derelanko; Steve Kutz; Patrick J. Little; Robert N. DeHaven; Diane L. DeHaven-Hudkins; Roland E. Dolle

Selective, nonpeptidic delta opioid receptor agonists have been the subject of great interest as potential novel analgesic agents. The discoveries of BW373U86 (1) and SNC80 (2) contributed to the rapid expansion of research in this field. However, poor drug-like properties and low therapeutic indices have prevented clinical evaluation of these agents. Doses of 1 and 2 similar to those required for analgesic activity produce convulsions in rodents and nonhuman primates. Recently, we described a novel series of potent, selective, and orally bioavailable delta opioid receptor agonists. The lead derivative, ADL5859 (4), is currently in phase II proof-of-concept studies for the management of pain. Further structure activity relationship exploration has led to the discovery of ADL5747 (36), which is approximately 50-fold more potent than 4 in an animal model of inflammatory pain. On the basis of its favorable efficacy, safety, and pharmacokinetic profile, 36 was selected as a clinical candidate for the treatment of pain.


FEBS Letters | 1998

Endomorphins fully activate a cloned human mu opioid receptor.

Jianhua Gong; Judith A. Strong; Shengwen Zhang; Xia Yue; Robert N. DeHaven; Jeffrey D. Daubert; Joel A. Cassel; Guangling Yu; Erik Mansson; Lei Yu

Endomorphins were recently identified as endogenous ligands with high selectivity for mu opioid receptors. We have characterized the ability of endomorphins to bind to and functionally activate the cloned human mu opioid receptor. Both endomorphin‐1 and endomorphin‐2 exhibited binding selectivity for the mu opioid receptor over the delta and kappa opioid receptors. Both agonists inhibited forskolin‐stimulated increase of cAMP in a dose‐dependent fashion. When the mu opioid receptor was coexpressed in Xenopus oocytes with G protein‐activated K+ channels, application of either endomorphin activated an inward K+ current. This activation was dose‐dependent and blocked by naloxone. Both endomorphins acted as full agonists with efficacy similar to that of [d‐Ala2,N‐Me‐Phe4,Gly‐ol5]enkephalin (DAMGO). These data indicate that endomorphins act as full agonists at the human mu opioid receptor, capable of stimulating the receptor to inhibit the cAMP/adenylyl cyclase pathway and activate G‐protein‐activated inwardly rectifying potassium (GIRK) channels.


Life Sciences | 2002

Antipruritic and antihyperalgesic actions of loperamide and analogs

Diane L. DeHaven-Hudkins; Alan Cowan; Luz Cortes Burgos; Jeffrey D. Daubert; Joel A. Cassel; Robert N. DeHaven; George B. Kehner; Virendra Kumar

Loperamide and three of its analogs were evaluated for their ability to inhibit binding to cloned human opioid receptor subtypes and to produce antipruritus and antinociception following local s.c. administration to rodents. All four compounds were fully efficacious agonists with affinities of 2 to 4 nM for the cloned human mu opioid receptor. Local s.c. injection of loperamide, ADL 01-0001 or ADL 01-0002 at the same site as the introduction of the pruritogenic compound 48/80 resulted in antipruritic activity in a mouse model of itch. Similarly, i.paw or i.pl. administration of compounds ADL 01-0001, ADL 01-0002 and ADL 01-0003 to inflamed paws caused potent antinociception, inhibiting late phase formalin-induced flinching, Freunds adjuvant-induced mechanical hyperalgesia and tape stripping-induced mechanical hyperalgesia. Loperamide and its analogs were efficacious in animal models of itch and inflammatory pain, and may have potential therapeutic utility as antipruritic and antihyperalgesic agents.


Bioorganic & Medicinal Chemistry Letters | 2000

Arylacetamides as peripherally restricted kappa opioid receptor agonists.

Virendra Kumar; Michael Anthony Marella; Luz Cortesburgos; An-Chih Chang; Joel A. Cassel; Jeffrey D. Daubert; Robert N. DeHaven; Diane L. DeHaven-Hudkins; Susan L. Gottshall; Erik Mansson; Alan L. Maycock

Analogues of the kappa (kappa) opioid receptor agonist, ICI 199441, were prepared. Ki values for these analogues at the cloned human kappa opioid receptor ranged from 0.058 to 25 nM. Trifluoromethylaryl derivatives were potent analgesics when administered subcutaneously in the rat and were more peripherally restricted than the parent compound, ICI 199441.


Bioorganic & Medicinal Chemistry Letters | 2009

CB2 selective sulfamoyl benzamides: optimization of the amide functionality.

Allan J. Goodman; Christopher W. Ajello; Karin Worm; Bertrand Le Bourdonnec; Markku A. Savolainen; Heather O’Hare; Joel A. Cassel; Gabriel J. Stabley; Robert N. DeHaven; Christopher J. LaBuda; Michael Koblish; Patrick J. Little; Bernice L. Brogdon; Steven A. Smith; Roland E. Dolle

Previous research within our laboratories identified sulfamoyl benzamides as novel cannabinoid receptor ligands. Optimization of the amide linkage led to the reverse amide 40. The compound exhibited robust antiallodynic activity in a rodent pain model when administered intraperitoneally. Efficacy after oral administration was observed only when ABT, a cytochrome P450 suicide inhibitor, was coadministered.


Bioorganic & Medicinal Chemistry Letters | 2008

Sulfamoyl benzamides as novel CB2 cannabinoid receptor ligands.

Karin Worm; Q. Jean Zhou; Christopher T. Saeui; Rosalyn C. Green; Joel A. Cassel; Gabriel J. Stabley; Robert N. DeHaven; Nathalie Conway-James; Christopher J. LaBuda; Michael Koblish; Patrick J. Little; Roland E. Dolle

Sulfamoyl benzamides were identified as a novel series of cannabinoid receptor ligands. Starting from a screening hit 8 that had modest affinity for the cannabinoid CB(2) receptor, a parallel synthesis approach and initial SAR are described, leading to compound 27 with 120-fold functional selectivity for the CB(2) receptor. This compound produced robust antiallodynic activity in rodent models of postoperative pain and neuropathic pain without traditional cannabinergic side effects.


Journal of Medicinal Chemistry | 2003

Structure-activity relationships of dynorphin a analogues modified in the address sequence.

Georg Schlechtingen; Robert N. DeHaven; Jeffrey D. Daubert; Joel A. Cassel; Nga N. Chung; Peter W. Schiller; Joseph P. Taulane; Murray Goodman

The peptide [Pro3]Dyn A(1-11)-NH2 2 exhibits high affinity (K(i) = 2.4 nM) and over 2000-fold selectivity for the opioid receptor. Stepwise removal of the C-terminal residues from this ligand demonstrated that its positively charged Arg residues, particularly Arg6 and Arg7, were crucial for binding to the kappa receptor. Analogues shorter than seven amino acids lacked significant affinity for opioid receptors. Comparison with a series of truncated analogues of Dyn A showed that the relative losses in binding potency differed only slightly between the two series. The neutral residues Ile8 and Pro10 could be removed without significant loss in affinity for the kappa receptor. Their replacement, in the Pro3 analogue, with additional Arg residues led to analogues with improved kappa affinity (e.g., [Pro3,Arg8]Dyn A(1-11)-NH2 20: K(i)(kappa) = 0.44 nM). This type of modification did not compromise the high kappa selectivity of the Pro3 analogues. These findings support the view that a negatively charged domain in the putative second extracellular loop of the kappa receptor selectively recognizes residues 6-11 of dynorphin through electrostatic interactions. As with parent compound 2, analogue 20 and related compounds displayed kappa antagonist properties.


Bioorganic & Medicinal Chemistry Letters | 2009

Novel pyridine derivatives as potent and selective CB2 cannabinoid receptor agonists.

Guo-Hua Chu; Christopher T. Saeui; Karin Worm; Damian G. Weaver; Allan J. Goodman; Robert L. Broadrup; Joel A. Cassel; Robert N. DeHaven; Christopher J. LaBuda; Michael Koblish; Bernice L. Brogdon; Steve Smith; Bertrand Le Bourdonnec; Roland E. Dolle

Replacement of the phenyl ring in our previous (morpholinomethyl)aniline carboxamide cannabinoid receptor ligands with a pyridine ring led to the discovery of a novel chemical series of CB2 ligands. Compound 3, that is, 2,2-dimethyl-N-(5-methyl-4-(morpholinomethyl)pyridin-2-yl)butanamide was identified as a potent and selective CB2 agonist exhibiting in vivo efficacy after oral administration in a rat model of neuropathic pain.


Bioorganic & Medicinal Chemistry Letters | 2010

Novel sulfamoyl benzamides as selective CB2 agonists with improved in vitro metabolic stability

Ian Sellitto; Bertrand Le Bourdonnec; Karin Worm; Allan J. Goodman; Markku A. Savolainen; Guo-Hua Chu; Christopher W. Ajello; Christopher T. Saeui; Lara K. Leister; Joel A. Cassel; Robert N. DeHaven; Christopher J. LaBuda; Michael Koblish; Patrick J. Little; Bernice L. Brogdon; Steven A. Smith; Roland E. Dolle

A lead optimization campaign in our previously reported sulfamoyl benzamide class of CB(2) agonists was conducted to improve the in vitro metabolic stability profile in this series while retaining high potency and selectivity for the CB(2) receptor. From this study, compound 14, N-(3,4-dimethyl-5-(morpholinosulfonyl)phenyl)-2,2-dimethylbutanamide, was identified as a potent and selective CB(2) agonist exhibiting moderate in vitro metabolic stability and oral bioavailability. Compound 14 demonstrated in vivo efficacy in a rat model of post-surgical pain.

Collaboration


Dive into the Joel A. Cassel'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

Guo-Hua Chu

Cubist Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge