Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 May 1.
Published in final edited form as: Drug Discov Today. 2010 Mar 3;15(9-10):371–383. doi: 10.1016/j.drudis.2010.02.011

Progress in corticotropin-releasing factor-1 antagonist development

Eric P Zorrilla 1, George F Koob 1
PMCID: PMC2864802  NIHMSID: NIHMS184385  PMID: 20206287

Abstract

Corticotropin-releasing factor (CRF) receptor antagonists have been sought since the stress-secreted peptide was isolated in 1981. Although evidence suggests the limited efficacy of CRF1 antagonists as antidepressants, CRF1 antagonists might be novel pharmacotherapies for anxiety and addiction. Progress in understanding the two-domain model of ligand–receptor interactions for CRF family receptors might yield chemically novel CRF1 receptor antagonists, including peptide CRF1 antagonists, antagonists with signal transduction selectivity and nonpeptide CRF1 antagonists that act via the extracellular (rather than transmembrane) domains. Novel ligands that conform to prevalent pharmacophore and exhibit drug-like pharmacokinetic properties have been identified. The therapeutic utility of CRF1 antagonists should soon be clearer: several small molecules are currently in Phase II/III clinical trials for depression, anxiety and irritable bowel syndrome.


Corticotropin-releasing factor (CRF) receptor antagonists have been sought since Vale and colleagues isolated the stress-secreted, adrenocorticotropin-releasing hypothalamic peptide in 1981 [1]. The identification of CRF was followed by the discovery of genes encoding three paralogs of CRF (urocortins 1, 2 and 3; Ucn 1, Ucn 2 and Ucn 3) and two G-protein-coupled receptors (CRF1 and CRF2) that the CRF/Ucn[E1] peptides bind and activate with varying affinities [2,3]. Pharmacological and transgenic studies show that brain and pituitary CRF1 receptors mediate endocrine, behavioral and autonomic responses to stress [4]. Consequently, the pharmaceutical industry has sought to develop blood–brain-barrier-penetrating, selective CRF1 receptor antagonists [5]. Previous reviews by us and others have surveyed the biology of CRF systems [2]; the pharmacophore, physiochemical properties and pharmacokinetics of prototypical non-peptide CRF1 receptor antagonists [69]; and the therapeutic potential of CRF1 antagonists for stress-related indications [6,10,11], including major depression [12], anxiety disorders [13] and irritable bowel syndrome [14]. This article, after briefly overviewing the CRF/Ucn system and preclinical data supporting the therapeutic potential of CRF1 antagonists for anxiety, depression and addictive disorders, reviews advances in CRF1 antagonist development since 2005.

Biology of CRF/Ucn receptor systems

CRF-related peptides interact with two known mammalian CRF receptor subtypes, CRF1 and CRF2, which both belong to the class B1 (‘secretin-like’) subfamily of G-protein-coupled receptors. The CRF1 receptor exists in multiple isoforms (e.g. CRF1a–CRF1h), with the best known and functional isoform the CRF1(a) subtype. The CRF2 receptor has three known functional membrane-associated subtypes in humans – CRF2(a), CRF2(b) and CRF2(c) – and a ligand-sequestering, soluble CRF2(a) isoform discovered in mouse. CRF1 and CRF2 receptors have ~70% sequence identity. CRF has high, preferential affinity for CRF1 vs. CRF2 receptors. Ucn 1 is a high-affinity agonist at both receptors, and the type 2 urocortins (Ucn 2 and Ucn 3) are more selective for membrane CRF2 receptors. The biological actions of CRF, Ucn 1 and Ucn 2 in rodents are also modulated by a CRF-binding protein (CRF-BP), a 37-kDa secreted glycoprotein that binds and putatively immunosequesters CRF and Ucn 1 with equal or greater affinity than CRF receptors. Structural requirements for binding to CRF receptors and the CRF-BP differ. Many (if not most) CRF receptor antagonists do not bind the CRF-BP [3,6].

CRF1 receptors mediate not only the hypothalamic–pituitary–adrenal (HPA) axis neuroendocrine response to stress but also other aspects of stress responses in organisms. The distribution of CRF1 receptors in the brain is highly conserved in stress-responsive brain regions, including the neocortex, central extended amygdala, medial septum, hippocampus, hypothalamus, thalamus, cerebellum, and autonomic midbrain and hindbrain nuclei. This receptor distribution, concordant with that of its natural ligands CRF and Ucn 1, is consistent with the recognized role for extrahypothalamic CRF1 receptors in behavioral and autonomic stress responses.

CRF1 antagonists in animal models of anxiety, depression and addictive disorders

Nonpeptide CRF1 antagonists consistently produce anxiolytic-like effects in animal models [6]. For example, in rodents, the compounds reduced conditioned fear [15,16], shock-induced freezing [17], anxiety-like responses to neonatal isolation [18,19] and defensive burying behavior [20,21]. CRF1 antagonists reduced acoustic startle responding [22,23] and showed efficacy in exploration-based models of anxiety, such as the open field, elevated plus maze, light–dark box and defensive withdrawal tests [18,2427], under stressed, but not non-stressed, testing conditions. CRF1 antagonists only exhibited weak activity in punished drinking and punished crossing conflict models (unlike γ-aminobutyric acid anxiolytics) [18,28] but effectively increased social interaction [28,29]. In rodents, little tolerance to the anxiolytic-like actions of CRF1 antagonists is observed with daily administration for up to 14 days [6]. CRF1 antagonists also blocked pain-related synaptic facilitation and anxiety-like behavior [30,31]. In addition, the compounds produced anxiolytic-like effects in intruder tests using non-human primate models [32,33].

Despite initial positive results, however, data with small-molecule CRF1 antagonists have not consistently shown efficacy in animal models that predict antidepressant activity [5]. Regarding positive findings, subchronic treatment with DMP696 and R121919 reduced forced swim immobility in mice [34], and chronic treatment with SSR125543 increased swimming in Flinder Sensitive Line rats, a putative genetic model of depression [35]. Acute antalarmin treatment similarly reduced forced swim immobility in CRF2-receptor-null mutant mice [36], and antalarmin, SSR125543A, LWH234 and CRA1000 acutely reduced immobility in some, but not all, studies of outbred rats [18,37,38]. R278995 reduced hyperemotionality of olfactory bulbectomized rats [39], a putative model of depression [40]. Chronic treatment with antalarmin or SSR125543A also improved coat appearance and reversed reductions in hippocampal neurogenesis in a mouse model of chronic mild stress [18,41,42].

Regarding negative findings, R121919, CP154,526 and R278995 failed to reduce forced swim immobility in rats [38,39], and antalarmin, CP-154,526, DMP904, R121919 and DMP696 failed to reduce forced swim immobility in mice after acute, subchronic or chronic (16 days) dosing [34,43,44]. Furthermore, antalarmin, CP-154526, DMP904, R121919, DMP696 and R278995 were all inactive in the tail suspension test with acute dosing [34,39,45]. Although acute treatment with CP-154526 was first reported to produce antidepressant-like effects in the learned helplessness paradigm [46], a subsequent study with CP-154526 failed to replicate this finding [47]. DMP904, DMP696 and CRA1000 were also inactive in this model after acute dosing [47,48]. Nonetheless, CP-154526, CRA1000 and R2789995 prevented the acquisition, but not the expression, of learned helplessness [39,47,49]. R278995 did not produce antidepressant-like effects in the rat differential-reinforcement-of-low-rate 72-s model [39]. A potential explanation for these mixed findings is that CRF1 antagonists might only exhibit antidepressant-like activity in ‘dysfunction’ models or models that exhibit a depressive-like endophenotype as a result of the animals’ genetic background or environmental manipulation (e.g. olfactory bulbectomy) and not in healthy, normal animals. Such an interpretation is consistent with findings showing that CRF1 antagonists differentially reduce anxiety-like behavior in models of high anxiety [6] and reduce drug or ethanol intake in models of dependence [5058], rather than in healthy, nondependent animals; however, this interpretation might be difficult to reconcile with the inability of CRF1 antagonists to reproducibly reverse learned helplessness behavior that results from repeated inescapable shock. A better understanding of the preclinical conditions under which CRF1 antagonists exert antidepressant-like effects might have translational implications for identifying patient subgroups or conditions under which CRF1 antagonists are more likely to be clinically useful for depression.

Another major action of CRF1 antagonists has been in the context of the activation of brain stress systems in addiction. As reviewed recently [59], both conceptual and neurobiological advances suggest that CRF1 systems contribute to the withdrawal–negative affect and preoccupation–anticipation (‘craving’) stages of the addiction cycle that fuel compulsive drug taking. Regarding the withdrawal–negative affect stage, dysphoria and increased anxiety are associated with both acute and protracted abstinence from most drugs of abuse. Such negative emotional symptoms, via negative reinforcement, drive high levels of drug taking to prevent or relieve the aversive withdrawal state, which is hypothesized to be mediated by CRF1 activation. Consistent with this hypothesis, both the HPA axis and extrahypothalamic CRF systems are activated during acute withdrawal from all major drugs of abuse in animal models [60], and central infusion of non-peptide CRF antagonists block the anxiogenic-like responses observed during acute withdrawal from drugs of abuse, including cocaine, alcohol, nicotine and cannabinoids [61]. Similarly, systemic administration of blood–brain-barrier-penetrating CRF1 antagonists reduced the anxiogenic-, aversive- and hypohedonic-like effects of withdrawal from opioids [6265], nicotine [56,66], benzodiazepines [63] and alcohol [29,54,55,67,68]. Supporting the motivational significance of withdrawal-associated CRF1 system activation for drug taking, administration of small-molecule CRF1 antagonists also reduced the excessive drug intake associated with dependence on alcohol [5055,69], nicotine [56], cocaine [57] and opioids [58]. Relatedly, CRF1 antagonists might also have therapeutic potential in individuals who self-medicate innate negative emotional states by taking excessive amounts of drugs or alcohol. This claim is supported by the anti-drinking efficacy of small-molecule CRF1 antagonists selectively in rats that show high innate anxiety, such as Marchigian alcohol-preferring rats [70] and isolation-reared Fawn-Hooded rats [71].

Stress is a major recognized precipitant of relapse, and CRF1 antagonists, therefore, are also hypothesized to have therapeutic potential in the ‘craving’ stage of the addiction cycle by preventing stress-induced relapse. Accordingly, non-peptide CRF antagonists centrally block stress-induced reinstatement of drug-seeking behavior in animal models [72]. Similarly, systemic administration of small-molecule CRF1 antagonists reduced footshock stress-induced reinstatement of heroin-, cocaine-, nicotine-or alcohol-seeking behavior in rats [55,66,7375] and footshock stress-induced reactivation of conditioned place preference for opioids and cocaine [76,77]. Thus, models of drug withdrawal, excessive drug taking, innate anxiety with comorbid ethanol intake and stress-induced relapse behavior all support the therapeutic potential of CRF1 antagonists for drug dependence.

Peptide CRF1-selective receptor antagonists

Several N-terminally truncated and substituted analogs of CRF act as subtype nonselective competitive partial agonists or full antagonists at CRF receptors. Examples of these, in chronological order of discovery, include the partial agonist [Met18, Lys23, Glu27,29,40, Ala32,41, Leu33,36,38] r/hCRF9-41 (α-helical CRF9-41) and the full receptor antagonists [D-Phe12, Nle21,38 CaMeLeu37] r/hCRF12-41 (D-Phe CRF12-41), cyclo(30-33)[D-Phe12, Nle21,38, Glu30, Lys33] r/hCRF12-41 (astressin) and cyclo(30-33)[D-Phe12, Nle21, CaMeLeu27, Glu30, Lys33, Nle38, CaMeLeu40]Ac-r/hCRF9-41 (astressin-B). These peptide ligands have approximately the same order of binding and antagonist potency at CRF1 vs. CRF2 receptors and do not cross the blood–brain barrier. Thus, they are subtype nonselective, peripherally acting CRF receptor antagonists.

An emerging development within the last five years is that in the course of seeking minimal fragments of CRF that retain antagonist activity, CRF1-preferring peptide antagonists might have been identified. Yamada and colleagues [78] followed up a Solvay patent application [79] that described a peptide comprising the 12 C-terminal residues of astressin as a potent antagonist of CRF receptors. Through amino acid substitution of Nle38 with a lipophilic cyclohexylalanine residue and Ala31 with an unnatural residue (D-Ala), they identified a metabolically stable, high-affinity (Ki ~3 nM) CRF1 antagonist that potently (0.1 mg/kg, i.v.) reduced adrenocorticotropic hormone secretion in a rat sepsis model. This peptide might be a CRF1-preferring antagonist; the Solvay group concurrently reported that the 12-residue N-terminal truncated astressin derivative from which Yamada and colleagues began their studies retains CRF1 affinity but is inactive at the CRF2(a) receptor.[E2] Thus, lactam-bridge-constrained N-terminally truncated astressin derivatives of 12–15 residue length might be preferential CRF1 receptor peptide antagonists. Such compounds could be useful for the treatment of pathologies associated with peripheral CRF1 hyperactivation [80], perhaps including irritable bowel syndrome, premature labor, postoperative gastric ileus, and Cushingoid aspects of severe alcohol dependence, visceral obesity, melancholic major depression and anorexia nervosa [5].

Progress in the two-domain model of ligand–CRF1 receptor interaction

Progress has also been made in understanding the mode of ligand–CRF1 receptor interaction. Natural peptide agonists of CRF receptors are thought to bind and activate the receptor via a two-site mechanism. The carboxyl end of the agonist first binds the N-terminal first extracellular domain (ECD1) (‘N-domain’), and the amino portion of the ligand successively binds the extracellular face of the seven juxtamembrane regions (‘J-domain’), stabilizing an agonist-bound, ‘active’ receptor state that yields signal transduction [81,82]. Interaction with the J-domain might also be important for receptor internalization because short (e.g. 12-residue) C-terminal peptide antagonists that only bind the N-domain do not trigger endocytosis, unlike longer antagonists that also interact with the J-domain (e.g. astressin) [83].

Recently, a minimal cyclic peptide fragment homologous to the 12 C-terminal residues of astressin was used to understand determinants of ligand binding to the N-domain of the CRF1 receptor recombinantly isolated from the J-domain [84]. NMR spectroscopy showed that two hydrophobic residues (Met38 and Ile41 of CRF sequence) and two amide groups (Asn34 and the C-terminal amide) on one face of the bound peptide’s α helix defined the antagonist’s binding epitope. This epitope could potentially be used as a template to develop novel non-peptide competitive antagonists that target the agonist-binding N-domain of the CRF1 receptor, in contrast to existing small-molecule antagonists that allosterically target the J-domain [82].

Recently, different ligands have been shown to address the J-domain of the receptor differently, conferring not only receptor subtype selectivity but also signal transduction pathway selectivity within a given receptor subtype. For example, the NMR solution structures of astressin B, astressin2-B and Ucn 2 exhibit a large (90°) kink that, after binding to the N-domain, orients the ECD1 with its positively charged face toward the negatively charged extracellular loops 2, 3 and 4 of the receptor. By contrast, astressin, stressin1, hUcn1 and hUcn3 have no or much smaller kinks, such that the ECD1 of the receptor would have to orient at a different angle to enable ligand interaction with the J-domain. Grace and colleagues hypothesized that the different ligand–receptor complex confirmations could lead to engagement of different signal transduction pathways [85]. Supporting this hypothesis, single substitutions of Ucn 1 with bulky amino acids (e.g. benzoyl-phenylalanine or naphthylalanine) in residues 6–15, but not at other positions, eliminated the peptide’s ability to stimulate Gi–protein activation while not altering its activation of Gs–protein pathways. The resulting analogs were competitive receptor antagonists for the Gi–protein pathway and agonists for the Gs pathway [86]. Similarly, the nonpeptide antagonist antalarmin, which binds the J-domain of the receptor, had different antagonist potency against urocortin- vs. sauvagine-induced G-protein activation and also exhibited different modes of antagonist action: competitive for receptor coupling to Gs but noncompetitive for Gi activation. By contrast, the peptide antagonist α-helical CRF9-41, which binds the peptide agonist-binding N-domain, uniformly and competitively antagonized urocortin- or sauvagine-induced activation of both Gi and Gs signaling pathways [87]. These results suggest that antagonism of specific CRF1 signal transduction pathways might be possible via ligands that stabilize or destabilize particular ligand–receptor complex confirmations[E3].

A final recent finding regarding the mode of CRF antagonist action was that nonpeptide ligands can allosterically facilitate or inhibit binding of CRF to G-protein-uncoupled CRF1 receptors while uniformly inhibiting signaling efficacy in the CRF-bound, active, G-protein-coupled state. Positive and negative allosteric modulators of CRF affinity for the uncoupled receptor functioned as intrinsic weak agonists or inverse agonists at uncoupled receptors, respectively. Thus, different conformational states can lead to inhibition of CRF signaling. Nonpeptide ligands can act as functional antagonists by stabilizing an inactive (allosteric inverse agonist) or weakly active (allosteric agonist) receptor state, either of which can shift receptor equilibrium away from the CRF-bound, fully active signaling state [88].

Novel, nonpeptide CRF1-selective receptor antagonists

Pharmacophore and selectivity

Since 2005, many small molecules with high and selective CRF1 (vs. CRF2) affinity have been identified (Table 1). Each series follows the previously reviewed general pharmacophore common to most nonpeptide CRF1 antagonists. Prototypical compounds (Figure 1) share one or two aliphatic top units that occupy a hydrophobic pocket of the receptor, a central mono-, bi- or tricyclic ring core and an orthogonal, conformation-stabilizing 2,4-di- or 2,4,6-tri-substituted aromatic bottom group. Each ring core contains a putative proton-accepting ring nitrogen separated from the pendant aromatic by a one- or, more commonly, two-atom spacer. The core ring is typically methylated on the opposite position adjacent to the bonding nitrogen. The hydrogen bond accepting core nitrogen is hypothesized to interact with the imidazole side chain of histidine-199, a polar amino acid in the third transmembrane domain of the CRF1 receptor that is not shared in the CRF2 receptor or CRF-BP sequences. Nonpeptide antagonists of this pharmacophore also require the rotational flexibility present in methionine residue 276 of the CRF1 (and not CRF2) receptor sequence [89], putatively to permit a hydrophobic interaction of the ring core with the fifth transmembrane domain. Accordingly, mutation of the 199 or 276 CRF1 residues (His-199, Met-276) to their corresponding CRF2 amino acids (Val-199, Ile-276) reduced the binding affinity of the selective CRF1 antagonist NBI 27914 by 40- and 200-fold, respectively [90]. A computational model incorporating both structural interaction features recently yielded good affinity predictions for a series of dihydropyridopyrazinone and dihydropteridinone CRF1 antagonists (r2 = 0.71) [91]. An independent in silico receptor docking model reached a similar conclusion regarding the structural mode of antagonist action for diydropyrrolo[2,3-d]pyrimidines [92]. Thus, nonpeptide antagonists of the prevalent pharmacophore seem to be potent and selective CRF1 antagonists in relation to their interactions with features in the third (His-199) and fifth (Met-276) transmembrane receptor domains.

Table 1.

Selected novel nonpeptide CRF1 antagonists in recent peer-reviewed literature (2005–2009)a

Company Core
cycles
Best
exemplars
CAS
registry
CRF1
affinity
(nM)
pKa cLog P cLog
DpH=7
cLog
DpH=2
MW PSA
(Å)
Oral
bioavailability
(F%)
Cl
plasma
(ml/min·kg)
Vd
(L/kg)
t1/2
(h)
Notes Refs
1-Aryl-4-aminoalkylisoquin olines Neurogen Bi 26k 1012325-20-0 Kd = 8 5.84 7.54 7.52 5.07 448.5 25.4 4.8% 14.5 13 5.4 [100]
2-Arylpyrimidines Neurogen Mono 12b 1067229-15-5 Kd = 9 5.00 380.9 6% 13.6 14 8 [101]
1-Methyl-3-dialkylamino-5-aryltriazoles Neurocrine Mono 7r 848648-99-7 Kd = 7 4.65 7.17 7.17 4.85 445.0 43.2 84% 27.7 7.8 3.3 B/P = 0.20 [102]
Pyrrolo[2,3-b]pyridine-based tricyclics Neurocrine Tri 19g 374798-46-6 Kd = 3.5 9.34 8.37 6.25 5.87 396.0 21.1 24% 70 38 6.3 B/P = 0.27; reduced restraint-induced ACTH (10 mg/kg) [103]
Pyrazolo[3,4-b]pyridine-based tricyclics Neurocrine Tri 22a 877672-44-1 Kd = 2.9 13.41 5.43 3.43 2.43 419.4 31.2 7% 43 44 12 B/P = 0.53; reduced restraint-induced ACTH (10 mg/kg) [103]
Imidazo[4,5-b]pyridin-2-ones Neurocrine Tri 16g 268539-94-2 Kd = 2 10.78 2.45 −0.03 −0.05 394.5 48.9 30% 53 7.2 1.6 B/P = 2.8; reduced i.v. CRF-induced ACTH (10 mg/kg) [94]
Tetrahydrotetraaza acenaphthylenes Neurocrine Tri 12a (NBI35965) 603151-83-3 (free base) Kd = 3 7.91 5.14 4.19 2.64 401.3 29.0 34% 17 17.8 12 B/P = 1.25; reduced restraint or i.v. CRF-induced ACTH (10–20 mg/kg, p.o.); blocked i.c.v. CRF-induced colonic motility [104]
Tetrahydrotetraaza acenaphthylenes Neurocrine Tri 12t (NBI34041) 268545-87-5 Kd = 4 7.91 6.22 5.27 3.72 417.4 29.0 Reduced footshock or i.v. CRF-induced ACTH (3 mg/kg) [104,105]
Hexa- and tetrahydrotetra-azaacenaphthylenes GSK Tri 2ba 862901-68-6 IC50 = 14 6.78 6.82 6.25 4.33 484.5 34.0 33% 10 B/P = 1.3; MED = 30 mg/kg p.o. in rat pup ultrasonic vocalization model [106]
Tetrahydrotetraaza acenaphthylenes GSK Tri 3a 476645-12-2 IC50 = 100 9.86 8.49 6.15 5.99 483.5 21.1 71% 8 2.5 5.3 [107]
Cyclopental[d]pyrimidines GSK Bi 3ac 474655-90-8 IC50 = 174 7.19 6.4 5.99 3.90 390.3 29.0 22% 41 5.4 3.1 B/P = 0.35 [7]
Dihydropyrrolo[2,3-d]pyrimidines GSK Bi 4fi 474657-07-3 IC50 = 48 1.84 3.53 3.53 3.30 414.2 46.8 86% 9 3.3 6 B/P = 2.30; ED50 = 2.0 mg/kg p.o. in rat pup ultrasonic vocalization model [7]
‘Top’ heteroaryl-substituted dihydropyrrolo[2,3-d]pyrimidines GSK Bi 2a 474655-69-1 IC50 = 32 0.79 6.04 6.04 6.01 496.4 88.0 52% 12 3.7 5.1 B/P = 2.30; ED50 = 10.0 mg/kg p.o. in rat pup ultrasonic vocalization model [108]
‘Top’ heteroaryl-substituted dihydropyrrolo[2,3-d]pyridines GSK Bi 27 727992-91-8 IC50 = 35 3.93 3.07 3.07 1.23 439.5 84.3 [92]
‘Top’ heteroaryl-substituted dihydropyrrolo[2,3-d]pyridines GSK Bi 3 (GW876008) 786701-83-5 IC50 = 66 4.81 1.54 1.54 −0.79 402.4 75.5 66% 19 2.4 1.6 B/P = 3.7; reduced i.c.v. CRF-induced gerbil forepaw treading (10 mg/kg), marmoset defensive postures (10 mg/kg), rat pup ultrasonic vocalization (30 mg/kg) [95]
Indanylpyrazines Pfizer Mono 19 675198-68-2 Kd = 11 1.46 3.85 3.85 3.65 436.5 69.2 [109]
2-Aryloxy-4-alkylaminopyridines Pfizer Mono 2 (CP-316311) 175139-41-0 IC50 = 7 5.63 6.20 6.18 3.73 327.5 31.4 3.6% (fasted), 37% (fed) 4.1 1.1 8.3 Reduced i.v. CRF-induced ACTH (10 mg/kg), defensive withdrawal (10 mg/kg), fear-potentiated startle (32 mg/kg, p.o.) [96]
2-Aryloxy-4-alkylaminopyridines Pfizer Mono 3a 175140-00-8 IC50 = 5 7.23 7.33 6.9 4.83 326.5 34.2 22% (fasted), 64% (fed) 17 5.3 8.8 Reduced i.v. CRF-induced ACTH and fear-potentiated startle (10 mg/kg, p.o.) [97]
7-Aryl-6,7-dihydroimidazoimidazole BMS Bi 7b 444321-96-4 Kd = 42 6.66 3.68 3.52 1.18 394.5 41.4 32% 35 5.7 B/P = 0.21; anxiolytic-like activity in mouse canopy model (32 mg/kg) [110]
Imidazo[1,2-a]benzimidazoles BMS Tri 8e 444323-42-6 Kd = 23 7.93 8.34 468.6 25.5 35% 5.1 0.6 4.1 B/P = 0.3 [111]
8-Aryl-1,3a,7,8-tetraaza-cyclopenta[a]indenes BMS Tri 9d 444324-30-5 Kd = 7.8 8.45 7.14 451.6 38.4 4% 5.2 0.5 3 anxiolytic-like activity in mouse canopy model (30 mg/kg) [112]
2-Anilno-3-phenylsulfonyl-6-methylpyridines BMS Mono 7f 777940-17-7 Kd = 52 2.53 6.86 6.86 6.22 394.5 67.4 35% 5.2 25 5.1 [113]
Dihydropyridopyrazinones BMS Bi 2a 795300-31-1 IC50 = 0.8 5.46 3.10 3.09 0.63 399.5 79.8 [91]
Dihydropteridines BMS Bi 2d 937723-70-1 IC50 = 10 4.92 3.69 3.69 1.48 386.5 75.6 [91]
Imidazo[1,2-b]pyradizine Eli Lilly/NIAAA Bi MTIP 910551-43-8 Kd = 0.22 3.96 3.55 3.55 1.64 420.0 83.8 91% 4.9 1.7 3.9 Reduced conflict behavior; reduced ‘hangover’ anxiety-like behavior in elevated plus-maze behavior (10 mg/kg); reduced ethanol self-administration (10 mg/kg); reduced ethanol-seeking behavior in ethanol postdependent or alcohol-preferring rats [54,55]
Pyrazolo[1,5-a]pyrimidine Scripps (Dupont/BMS composition patent) Bi MPZP 202579-76-8 Kd = 4.9 5.32 2.95 2.93 0.50 398.5 61.1 Reduced defensive burying behavior; reduced ethanol, nicotine and cocaine self-administration in dependence models [52,53,56, 57]
8-(Pyrid-3-yl)pyrazolo[1,5-a]-1,3,5-triazines BMS Bi 13-15 Pexacerfont (BMS-562,086) 459856-18-9 Kd = 6.1 2.99 2.94 2.94 1.66 340.4 77.2 40% 17.9 14.9 13.5 Anxiolytic-like activity in elevated plus-maze and defensive withdrawal models (10 mg/kg, p.o.); observed logP = 4.32; solubility of 16 μg/ml in water (pH 7.4) and 16.3 mg/ml in 0.01 N HCl (pH 2.5); good pharmacokinetics in nonhuman primates [98]
8-(4-Methoxyphenyl)pyrazolo [1,5-a]-1,3,5-triazines: BMS Bi 6 in 1st 12-3 in 2nd (BMS-561,388) 202578-88-9 (free base) Kd = 4.7 6.63 2.15 1.99 −0.35 399.5 74.0 51% 20 14.6 9.7 Anxiolytic-like activity in elevated plus-maze and defensive withdrawal models (10 mg/kg, p.o.); observed logP = 4.76; solubility < 1 μg/ml in water (pH 7.4) and 2.5 mg/ml in 0.01 N HCl (pH 2.5); inferior oral pharmacokinetics in chimpanzee compared with Pexacerfont [98,114]
a

Intravenous pharmacokinetic parameters include Cl (clearance from plasma), Vd (volume of distribution at steady state) and t1/2 (plasma half-life). Physiochemical properties were calculated using Advanced Chemistry Development (ACD/Labs) Software v. 8.19 for Solaris.

Abbreviations: ACTH, adrenocorticotropic hormone; B/P, brain/plasma ratio; cLogP, calculated LogP; cLogD, calculated LogD at specified pH; CRF, corticotropin-releasing factor; MW, molecular weight; NIAAA, National Institute on Alcohol Abuse and Alcoholism; pKa, negative log of the acid-dissociation constant; PSA, polar surface area.

Figure 1.

Figure 1

Prototypical CRF1 antagonists.

Previously reviewed compounds that depart significantly[E4] from the pharmacophore include oxo-7H-benzo[e]pyrimidine-4-carboxylic acid derivatives (subtype nonselective CRF receptor antagonists discovered by Alanex), CC 2064460 (a moderately potent arylamidrazone CRF1 antagonist that lacks a central ring core with the customary hydrogen-bond-accepting nitrogen[E5]) and stereospecific N-phenylphenylglycines (which also lack a ring core but were identified through computational screening based on a classic pharmacophore training set [6]).

Lipophilicity and pharmacokinetic properties

As we and others have reviewed previously, clinical development of nonpeptide CRF1 antagonists has been hampered because most leads were undesirably lipophilic, with poor water solubility and pharmacokinetic properties [6,9]. Most early CRF1 antagonists failed Lipinski’s ‘rule of five’ criteria for drug candidates because of excessive lipophilicity (cLogP > 5). This benchmark is relevant because of more than 100 drugs marketed as of 1992 for CNS indications, not one had a logD > 4, which can yield poor physiochemical and pharmacokinetic properties. Rather, most (~85%) had a logD of 0–3. Preclinical studies since 2005, therefore, have sought to identify less hydrophobic, more drug-like CRF1 antagonists with favorable pharmacokinetic properties and fewer anticipated toxicities.

Table 1 shows pharmacological, physiochemical and pharmacokinetic properties of several nonpeptide CRF1 receptor antagonists from recent peer-reviewed literature (2005–2008). Many exemplar antagonists continue to exhibit low blood–brain barrier penetration (e.g. brain/plasma [B/P] ratios < 1), poor oral bioavailability (F% < 20%), high volumes of distribution at steady state (VD > 10 L/kg) and high plasma clearance (Clplasma > 45 ml/min/kg) [93]. However, several compounds exhibited more favorable overall pharmacokinetics. One compound from Neurocrine Biosciences was a potent (Ki = 2 nM), albeit rapidly cleared, imidazo[4,5-b]pyridin-2-one with good blood–brain barrier penetration (B/P = 2.8; compound 16g in Ref. [94]). Several from GlaxoSmithKline were less potent (IC50 = 32–100 nM) substituted tetrahydrotetraazaacenaphthylenes and diydropyrrolo[2,3-d]pyrimidines with excellent oral bioavailability (52%–86%), distribution/clearance balance[E6] and central accumulation (B/P = 2.3–3.7), including the clinical candidate GW876008 (see below) [95]. Two from Pfizer were potent (Ki = 5–7 nM) 2-aryloxy-4-alkylaminopyridines, including the clinical candidate CP-316311 (see below) [96] and a successor with better solubility at stomach pH and food-independent oral bioavailability (compound 3a in Ref. [97]). An Eli Lilly and NIAAA collaboration yielded a highly potent (Ki = 0.22 nM) imidazo[1,2-b]pyradizine, with outstanding oral bioavailability (91%; MTIP[E7]) [55]. Finally, Bristol Myers Squibb advanced two promising substituted pyrazolo[1,5-a]-1,3,5-triazines to clinical trials (BMS-561388 and their current lead candidate BMS-562086 [Pexacerfont], IC50 = 6.1 nM), which showed good pharmacokinetics in rat, dog and non-human primate models, no evidence of gastrointestinal or respiratory toxicity, and mild renal effects at doses approximately one order greater than those needed to substantially occupy brain CRF receptors [98]. Each of these compounds was active in vivo at minimum effective oral doses of 2–10 mg/kg in preclinical behavioral or endocrine animal models that are sensitive to CRF1 signaling (Table 1).

Patent literature

As shown in Table 2, small-molecule CRF1 antagonists disclosed recently in the patent literature (2006–2008) are also variations on the prevalent pharmacophore. Newer compounds have involved different cores, including heterocyclic pyrrolotriazinones (e.g. WO 2008136377), benzimadole derivatives (e.g. WO 2008082003, WO 2008051533 and WO 2006116412) and pyrazolo[4,3-d]pyrimidines (e.g. WO 2006126718), as well as monocyclic indanylaminopyrazinypyridines (e.g. US 2006211710). In each of these variants, the proton-accepting nitrogen is present in the cycle adjacent to, rather than distal from, the ‘down’ aromatic (Table 2), which might allow for new substitution chemistries. Another development has been clear confirmation that the ‘top’ unit does not need to be branched alkyl chains (a feature that contributed to the undesirably high lipophilicity of the pharmacophore) but rather can tolerate polar, cyclic amines and related structures (e.g. WO2007039264, WO2006001501, WO2006001511, WO2006126718 and US2006211710). ‘Down’ units that are less lipophilic than the prototypical di- and tri-substituted phenyl aromatic are also now common antagonist features, including sulfur- and nitrogen-containing substituted rings (e.g. WO2008036579, WO2008036541, WO2006102194 and US2006211710).

Table 2.

Recent clinical trials for CRF1 antagonists

Company Compound Stage Date
initiated
Current
status
Trial description Reference
compound
Clinicaltrials.gov or other identifier
GlaxoSmithKline/Neurocrine GW876008 Phase I Nov 2006 Completed Metabolism in smokers vs. nonsmokers NCT00429728
GW876008 Phase I Dec 2006 Completed Safety study; potential interaction with midazolam NCT00423761
GW876008 Phase I Mar 2007 Completed fMRI of emotional processing in healthy volunteers Lorazepam NCT00424697
GW876008 Phase I Jun 2007 Terminated Effects on pharmacokinetics of oral contraceptives NCT00508911
GSK561579 Phase I Oct 2006 Completed Effects on metyrapone-induced ACTH secretion Alprazolam NCT00426608
GSK561579 Phase I Oct 2007 Completed Variability in absorption study NCT00539136
GSK561579 Phase I Sep 2007 Recruiting April 2009 fMRI of emotional processing in healthy volunteers Lorazepam NCT00513565
GSK561679 and GW876008 Phase I/IIa Nov 2006 Completed fMRI of regional cerebral blood flow in IBS patients NCT00376896
GW876008 Phase I/IIa Aug 2007 Terminated Endocrine responses to meals in IBS patients NCT00511563
GW876008 Phase IIa Dec 2006 Completed Gut blood flow and pain sensitivity in IBS patients NCT00385099
GW876008 Phase II Nov 2006 Completed Multi-site, double-blind, placebo-controlled study in IBS patients NCT00421707
GSK561679 and GW876008 Phase I/IIa Mar 2007 Completed fMRI response to public speaking in social anxiety disorder Alprazolam NCT00555139
GW876008 Phase II Nov 2006 Completed Multi-site, double-blind, placebo-controlled study in social anxiety disorder Paroxetine NCT00397722
GSK561679 Phase II Oct 2008 Recruiting Multi-site, double-blind, placebo-controlled study in major depression NCT00733980
GSK586529 Phase I Completed Single-dose, escalating trial toward anxiety and depression indications www.neurocrine.com 3rd quarter, 2008 release
BMS Pexacerfont (BMS-562,086) Phase II/III Jul 2007 Completed Multi-site, double-blind, placebo-controlled study in generalized anxiety disorder Escitalopram NCT00481325
Pexacerfont (BMS-562,086) Phase I/II Nov 2005 Completed Multi-site, double-blind, placebo-controlled study in major depression Escitalopram NCT00135421
Pexacerfont (BMS-562,086) Phase II Dec 2006 Completed Multi-site, double-blind, placebo-controlled study in IBS patients NCT00399438
BMS-561,388 Phase I 2003–2004 Uncertain Effects on reproductive hormones and menstrual cycle in young healthy women http://myprofile.cos.com/miss_kc02
Pfizer CP-316,311 Phase II Apr 2005 Terminated Mar 2006 (negative results) Multi-site, double-blind, placebo-controlled study in major depression Sertraline NCT00143091
PF-57,2778 Phase I Terminated Safety study toward generalized anxiety disorder indication
Ono ONO-2333Ms Phase II Jun 2007 Terminated Jul 2008 (negative results) Multi-site, double-blind, placebo-controlled study in recurrent major depression NCT00514865
Taisho/Janssen (Johnson&Johnson) TAI-041/JNJ-19567470 Phase I Dec 2004 Uncertain
Sanofi-Aventis SSR 125543 Phase I Uncertain http://en.sanofi-aventis.com/research_innovation/rd_strategy/cns/cns.asp (Feb 2008)
Neurocrine NBI-34101 Phase I
NIH Antalarmin Preclinical Completed pre-Phase I toxicity studies [115]

Clinical development

Several CRF1 antagonists from different pharmaceuticals have entered clinical trials since December 2004. Because an open-label Phase IIa trial showed that escalating doses of R121919 exhibited a good overall safety profile, normalized sleep EEG[E8] and reduced depressive and anxious symptoms in depressed patients [6], clinical anticipation of CRF1 antagonists has been high. However, R121919 development was discontinued because of isolated instances of elevated liver enzymes in a parallel trial. Despite major efforts (Table 3), no subsequent CRF1 antagonist has successfully completed a definitive Phase III trial. By contrast, development of ONO-2333Ms and CP-316311 were discontinued because of negative efficacy results in double-blind, placebo-controlled trials for major depression [99]. Currently, the number of additional CRF1 antagonists that are undergoing or have completed undisclosed efficacy trials (Phase II/III) is at least three for major depression (GSK561679, GW876008 and Pexacerfont) and two for irritable bowel syndrome and social anxiety disorder (GSK561679 and GW876008).[E9] Several other candidates are earlier in the pipeline or their status has not been publicly updated by the pharmaceutical industry (e.g. GSK586529, TAI-041/JNJ19567470, SSR125543, NBI-34101 and antalarmin). Results from these trials should provide definitive conclusions regarding the therapeutic potential of CRF1 antagonists for anxiety, depression and irritable bowel disorder and might pave the way for clinical evaluation in addictive disorders.

Table 3.

Selected recent patents for CRF1 antagonists (2006–2008)

Compounds Exemplar structure[E10] Patent No. Inventor(s) Title Notes
Pyrrolo[2,1-f] [1,2,4]triazin-4(3H)-ones WO 2008136377 Saito, Tetsuji; Obitsu, Tetsuo; Kagamiishi, Yoshifum Preparation of pyrrolo[2,1-f] [1,2,4]triazin-4(3H)-ones as CRF antagonists PCT Int. Appl. (2008), 237pp. CODEN: PIXXD2 WO 2008136377 A1 20081113 CAN 149:556672 AN 2008:1360515
Benzimadole derivatives WO 2008082003 Aso, Kazuyoshi; Mochizuki, Michiyo; Kobayashi, Katsumi Benzimidazole derivatives as CRF receptor antagonists and their preparation, pharmaceutical compositions and use in the treatment of diseases PCT Int. Appl. (2008), 77pp. CODEN: PIXXD2 WO 2008082003 A1 20080710 CAN 149:128827 AN 2008:832827
Benzimadole derivatives WO 2008051533 Aso, Kazuyoshi; Mochizuki, Michiyo; Kojima, Takuto; Kobayashi, Katsumi; Pratt, Scott Alan; Gyorkos, Albert Charles; Corrette, Christopher Peter; Cho, Suk Young Preparation of benzimidazole derivatives as CRF receptor antagonists PCT Int. Appl. (2008), 216pp. CODEN: PIXXD2 WO 2008051533 A2 20080502 CAN 148:517721 AN 2008:530180
Thiazolylpyrazolopyrimidines WO 2008036579 Chen, Zhaogen; Hamdouchi, Chafiq Hamdouchi; Hembre, Erik James; Hipskind, Philip Arthur; Myers, Jason Kenneth; Takakuwa, Takako; Toth, James Lee Thiazolylpyrazolopyrimidines as CRF1 receptor antagonists and their preparation, pharmaceutical compositions and use in the treatment of diseases PCT Int. Appl. (2008), 60pp. CODEN: PIXXD2 WO 2008036579 A1 20080327 CAN 148:379656 AN 2008:381087
Thiophene pyrazolopyrimidines WO 2008036541 Chen, Zhaogen; Hamdouchi, Chafiq Hamdouchi; Hembre, Erik James; Hipskind, Philip Arthur; Jia, Shaojuan; Toth, James Lee Preparation of thiophene pyrazolopyrimidine compounds as corticotropin releasing factor 1 (CRF1) receptor antagonists PCT Int. Appl. (2008), 47pp. CODEN: PIXXD2 WO 2008036541 A1 20080327 CAN 148:379655 AN 2008:380601
Pyrrolo[1,2-b]pyridazines US 2007224636 Fu, Jian-Min Pyrrolo [1,2b]pyridazine compound CRF receptor antagonists and their therapeutic use U.S. Pat. Appl. Publ. (2007), 12pp. CODEN: USXXCO US 2007224636 A1 20070927 CAN 147:378405 AN 2007:1088670
Pyrrolopyrimidines JP 2007169216 Nakazato, Atsuro; Okubo, Taketoshi; Nozawa, Hiroshi; Minda, Tomoko; Kenneth, Ludo E.J. Pharmaceuticals containing pyrrolopyrimidines for prophylactic or therapeutic treatment of corticotropin releasing factor (CRF)-associated diseases Jpn. Kokai Tokkyo Koho (2007), 63pp. CODEN: JKXXAF JP 2007169216 A 20070705 CAN 147:134454 AN 2007:726462
Pyrrolopyridazines WO 2007069671 Nakai, Hisao; Saito, Tetsuji; Kagamiishi, Yoshifumi Preparation of bicyclic heterocyclic compounds as antagonists of corticotropin releasing factor (CRF) PCT Int. Appl. (2007), 60pp. CODEN: PIXXD2 WO 2007069671 A1 20070621 CAN 147:72776 AN 2007:670590
Imidazo[1,2-a]pyridines
Pyrazolo[1,5-a]pyridines
Benzimidazole derivatives
WO 2007069565 Nakai, Hisao; Saito, Tetsuji; Kagamiishi, Yoshifumi Preparation of bicyclic heterocyclic compounds as antagonists of corticotropin releasing factor (CRF) PCT Int. Appl. (2007), 93pp. CODEN: PIXXD2 WO 2007069565 A1 20070621 CAN 147:72759 AN 2007:670422
Pyrazolo[1,5-a]pyridines WO 2007039264 Hossner, Frank Pyrazolo[1,5-alpha]pyrimidinyl derivatives useful as corticotropin - releasing factor (CRF) receptor antagonists PCT Int. Appl. (2007), 37pp. CODEN: PIXXD2 WO 2007039264 A1 20070412 CAN 146:428737 AN 2007:409409
Pyrazolo[4,3-d]pyrimidines WO 2006126718 Kashiwagi, Toshihiko; Takamuro, Iwao; Watanabe, Yumi; Yato, Michihisa Preparation of pyrazolo[4,3-d]pyrimidine derivatives as CRF receptor antagonists PCT Int. Appl. (2006), 94pp. CODEN: PIXXD2 WO 2006126718 A1 20061130 CAN 146:27853 AN 2006:1252488
Benzimadole derivatives WO 2006116412 Gyorkos, Albert; Corrette, Christopher; Cho, Suk; Pratt, Scott; Siedem, Christopher; Aso, Kazuyoshi; Gyoten, Michiyo Preparation of fused heterocyclic compounds like benzimidazoles as CRF receptor antagonists PCT Int. Appl. (2006), 249pp. CODEN: PIXXD2 WO 2006116412 A2 20061102 CAN 145:455015 AN 2006:1147073
Pyrrolopyridines WO 2006108689 Andreotti, Daniele; Bacchi, Sergio; Delpogetto, Monica; Guelfi, Simone; Perboni, Alcide; Ribecai, Arianna; Spada, Simone; Stabile, Paolo; Tampieri, Marsia Process for preparing heterocyclylpyrrolopyridines and their use as antagonists of corticotropin -releasing factor (CRF) receptors PCT Int. Appl. (2006), 42 pp. CODEN: PIXXD2 WO 2006108689 A2 20061019 CAN 145:438607 AN 2006:1093832
Imidazo[1,2-b]pyridazines WO 2006107784 Collins, Elizabeth Aaron; Garcia- Losada, Pablo; Hamdouchi, Chafiq; Hipskind, Philip Arthur; Lu, Jianliang; Takakuwa, Takako Preparation of imidazo[1,2-b]pyridazine compounds as corticotropin releasing factor 1 receptor antagonists for treating psychiatric and neurological diseases PCT Int. Appl. (2006), 171pp. CODEN: PIXXD2 WO 2006107784 A1 20061012 CAN 145:419159 AN 2006:1066310
Imidazo[1,2-b]pyridazines WO 2006102194 Barbosa, Heather Janelle; Collins, Elizabeth Aaron; Hamdouchi, Chafiq; Hembre, Erik James; Hipskind, Philip Arthur; Johnston, Richard Duane; Lu, Jianliang; Rupp, Michael John; Takakuwa, Takako; Thompson, Richard Craig Preparation of imidazo[1,2-b]pyridazine compounds as corticotropin releasing factor 1 receptor antagonists for treating psychiatric and neurol. Diseases PCT Int. Appl. (2006), 205pp. CODEN: PIXXD2 WO 2006102194 A1 20060928 CAN 145:356795 AN 2006:1006160
Indanylaminopyrazinypyridines US 2006211710 Verhoest, Patrick R.; Hoffmann, Robert L. Preparation of indanylaminopyrazinylpyridines as corticotropin releasing factor CRF1 antagonists for treatment of CNS disorders U.S. Pat. Appl. Publ. (2006), 11pp. CODEN: USXXCO US 2006211710 A1 20060921 CAN 145:336081 AN 2006:980075
Pyrazolo[1,5-a]pyrimidines WO 2006044958 Lanier, Marion; Luo, Zhiyong; Moorjani, Manisha; Tellew, John Edward; Williams, John P.; Zhang, Xiaohu Preparation of pyrazolo[1,5-a]pyrimidines as corticotropin - releasing factor (CRF) receptor antagonists PCT Int. Appl. (2006), 117 pp. CODEN: PIXXD2 WO 2006044958 A1 20060427 CAN 144:432830 AN 2006:382981
Pyrazolopyridines WO 2006044821 Lanier, Marion; Moorjani, Manisha; Tellew, John Edward; Williams, John P. Preparation of pyrazolopyridines as corticotropin releasing factor (CRF) receptor antagonists PCT Int. Appl. (2006), 65 pp. CODEN: PIXXD2 WO 2006044821 A1 20060427 CAN 144:412502 AN 2006:382953
Pyrrolopyrimidines
Pyrrolopyridines
WO 2006001511 Nakazato, Atsuro; Okubo, Taketoshi; Nozawa, Dai; Tamita, Tomoko; Kennis, Ludo E.J. Preparation of pyrrolopyrimidine and pyrrolopyridine derivatives substituted with a cyclic amino group as CRF antagonists PCT Int. Appl. (2006), 89 pp. CODEN: PIXXD2 WO 2006001511 A1 20060105 CAN 144:108345 AN 2006:13450
Pyrrolopyrimidines
Pyrrolopyridines
WO 2006001501 Nakazato, Atsuro; Okubo, Taketoshi; Nozawa, Dai; Kennis, Ludo E.J.; De Bruyn, Marcel F.L. Preparation of pyrrolopyrimidine and pyrrolopyridine derivatives substituted with tetrahydropyridine as CRF antagonists PCT Int. Appl. (2006), 42 pp. CODEN: PIXXD2 WO 2006001501 A1 20060105 CAN 144:108344 AN 2006:13293

Acknowledgments

This is publication number 21067 from The Scripps Research Institute. Research was supported by National Institutes of Health grants DK26741 and DK070118 from the National Institute of Diabetes and Digestive and Kidney Diseases. The authors thank Michael Arends and Mellany Santos for their help with manuscript preparation.

Footnotes

Disclosure statement

E.P.Z. and G.F.K. are inventors on a provisional patent filed for CRF1 antagonists (application serial #9709/102,422). G.F.K. currently provides consulting services to Alkermes, Boehringer-Ingelheim, Embera Pharmaceuticals, GlaxoSmithKline and Eli Lilly.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Vale W, et al. Characterization of a 41-residue ovine hypothalamic peptide that stimulates the secretion of corticotropin and beta-endorphin. Science. 1981;213:1394–1397. doi: 10.1126/science.6267699. [DOI] [PubMed] [Google Scholar]
  • 2.Bale TL, Vale WW. CRF and CRF receptors: role in stress responsivity and other behaviors. Annu Rev Pharmacol Toxicol. 2004;44:525–557. doi: 10.1146/annurev.pharmtox.44.101802.121410. [DOI] [PubMed] [Google Scholar]
  • 3.Fekete EM, Zorrilla EP. Physiology, pharmacology, and therapeutic relevance of urocortins in mammals: ancient CRF paralogs. Front Neuroendocrinol. 2007;28:1–27. doi: 10.1016/j.yfrne.2006.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Heinrichs SC, Koob GF. Corticotropin-releasing factor in brain: a role in activation, arousal, and affect regulation. J Pharmacol Exp Ther. 2004;311:427–440. doi: 10.1124/jpet.103.052092. [DOI] [PubMed] [Google Scholar]
  • 5.Zorrilla EP, et al. Anti-CRF. In: Fink H, editor. Encyclopedia of Stress. A–E. 2. Vol. 1. Elsevier; 2007. pp. 206–214. [Google Scholar]
  • 6.Zorrilla EP, Koob GF. The therapeutic potential of CRF1 antagonists for anxiety. Expert Opin Investig Drugs. 2004;13:799–828. doi: 10.1517/13543784.13.7.799. [DOI] [PubMed] [Google Scholar]
  • 7.Arban R, et al. Cyclopenta[d]pyrimidines and dihydropyrrolo[2,3-d]pyrimidines as potent and selective corticotropin-releasing factor 1 receptor antagonists. Chem Med Chem. 2007;2:528–540. doi: 10.1002/cmdc.200600257. [DOI] [PubMed] [Google Scholar]
  • 8.Tellew JE, Luo Z. Small molecule antagonists of the corticotropin releasing factor (CRF) receptor: recent medicinal chemistry developments. Curr Top Med Chem. 2008;8:506–520. doi: 10.2174/156802608783955665. [DOI] [PubMed] [Google Scholar]
  • 9.Chen C. Recent advances in small molecule antagonists of the corticotropin-releasing factor type-1 receptor – focus on pharmacology and pharmacokinetics. Curr Med Chem. 2006;13:1261–1282. doi: 10.2174/092986706776873014. [DOI] [PubMed] [Google Scholar]
  • 10.Stahl SM, Wise DD. The potential role of a corticotropin-releasing factor receptor-1 antagonist in psychiatric disorders. CNS Spectr. 2008;13:467–483. doi: 10.1017/s1092852900016709. [DOI] [PubMed] [Google Scholar]
  • 11.Zoumakis E, et al. Potential uses of corticotropin-releasing hormone antagonists. In: Tsigos C, Chrousos GP, editors. Stress, Obesity, and Metabolic Syndrome: Annals New York Academy of Sciences. Vol. 1083. New York: Academy of Sciences; 2006. pp. 239–251. [DOI] [PubMed] [Google Scholar]
  • 12.Holsboer F, Ising M. Central CRH system in depression and anxiety – evidence from clinical studies with CRH1 receptor antagonists. Eur J Pharmacol. 2008;583:350–357. doi: 10.1016/j.ejphar.2007.12.032. [DOI] [PubMed] [Google Scholar]
  • 13.Ising M, Holsboer F. CRH1 receptor antagonists for the treatment of depression and anxiety. Exp Clin Psychopharmacol. 2007;15:519–528. doi: 10.1037/1064-1297.15.6.519. [DOI] [PubMed] [Google Scholar]
  • 14.Stengel A, Taché Y. Neuroendocrine control of the gut during stress: corticotropin-releasing factor signaling pathways in the spotlight. Annu Rev Physiol. doi: 10.1146/annurev.physiol.010908.163221. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kikusui T, et al. Involvement of corticotropin-releasing factor in the retrieval process of fear-conditioned ultrasonic vocalization in rats. Physiol Behav. 2000;71:323–328. doi: 10.1016/s0031-9384(00)00352-8. [DOI] [PubMed] [Google Scholar]
  • 16.Hikichi T, et al. Suppression of conditioned fear by administration of CRF receptor antagonist CP-154,526. Pharmacopsychiatry. 2000;33:189–193. doi: 10.1055/s-2000-7587. [DOI] [PubMed] [Google Scholar]
  • 17.Weninger SC, et al. Stress-induced behaviors require the corticotropin-releasing hormone (CRH) receptor, but not CRH. Proc Natl Acad Sci U S A. 1999;96:8283–8288. doi: 10.1073/pnas.96.14.8283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Griebel G, et al. 4-(2-Chloro-4-methoxy-5-methylphenyl)-N-[(1S)-2-cyclopropyl-1-(3-fluoro-4-methylphenyl)ethyl]5-methyl-N-(2-propynyl)-1, 3-thiazol-2-amine hydrochloride (SSR125543A), a potent and selective corticotrophin-releasing factor(1) receptor antagonist. II Characterization in rodent models of stress-related disorders. J Pharmacol Exp Ther. 2002;301:333–345. doi: 10.1124/jpet.301.1.333. [DOI] [PubMed] [Google Scholar]
  • 19.Kehne JH, et al. Effects of the CRF1 receptor antagonist, CP 154,526, in the separation-induced vocalization anxiolytic test in rat pups. Neuropharmacology. 2000;39:1357–1367. doi: 10.1016/s0028-3908(00)00043-5. [DOI] [PubMed] [Google Scholar]
  • 20.Heinrichs SC, et al. Brain penetrance, receptor occupancy and antistress in vivo efficacy of a small molecule corticotropin releasing factor type I receptor selective antagonist. Neuropsychopharmacology. 2002;27:194–202. doi: 10.1016/S0893-133X(02)00299-3. [DOI] [PubMed] [Google Scholar]
  • 21.Zorrilla E, et al. CRF1 receptor antagonists for anxiety. Eur Neuropsychopharmacol. 2003;13 (Suppl 4):s130–s131. [Google Scholar]
  • 22.Schulz DW, et al. CP-154,526: a potent and selective nonpeptide antagonist of corticotropin releasing factor receptors. Proc Natl Acad Sci U S A. 1996;93:10477–10482. doi: 10.1073/pnas.93.19.10477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chen YL, et al. Synthesis and oral efficacy of a 4-(butylethylamino)pyrrolo[2,3-d]pyrimidine: a centrally active corticotropin-releasing factor1 receptor antagonist. J Med Chem. 1997;40:1749–1754. doi: 10.1021/jm960861b. [DOI] [PubMed] [Google Scholar]
  • 24.Gilligan PJ, et al. The discovery of 4-(3-pentylamino)-2,7-dimethyl-8-(2-methyl-4-methoxyphenyl)-pyrazolo-[1,5-a]-pyrimidine: a corticotropin-releasing factor (hCRF1) antagonist. Bioorg Med Chem. 2000;8:181–189. doi: 10.1016/s0968-0896(99)00271-0. [DOI] [PubMed] [Google Scholar]
  • 25.Lelas S, et al. Anxiolytic-like effects of the corticotropin-releasing factor1 (CRF1) antagonist DMP904 [4-(3-pentylamino)-2,7-dimethyl-8-(2-methyl-4-methoxyphenyl)-pyrazolo-[1,5-a]-pyrimidine] administered acutely or chronically at doses occupying central CRF1 receptors in rats. J Pharmacol Exp Ther. 2004;309:293–302. doi: 10.1124/jpet.103.058784. [DOI] [PubMed] [Google Scholar]
  • 26.Okuyama S, et al. Receptor binding, behavioral, and electrophysiological profiles of nonpeptide corticotropin-releasing factor subtype 1 receptor antagonists CRA1000 and CRA1001. J Pharmacol Exp Ther. 1999;289:926–935. [PubMed] [Google Scholar]
  • 27.Zorrilla EP, et al. Effects of antalarmin, a CRF type 1 receptor antagonist, on anxiety-like behavior and motor activation in the rat. Brain Res. 2002;952:188–199. doi: 10.1016/s0006-8993(02)03189-x. [DOI] [PubMed] [Google Scholar]
  • 28.Millan MJ, et al. Anxiolytic properties of the selective, non-peptidergic CRF(1) antagonists, CP154,526 and DMP695: a comparison to other classes of anxiolytic agent. Neuropsychopharmacology. 2001;25:585–600. doi: 10.1016/S0893-133X(01)00244-5. [DOI] [PubMed] [Google Scholar]
  • 29.Overstreet DH, et al. Modulation of multiple ethanol withdrawal-induced anxiety-like behavior by CRF and CRF1 receptors. Pharmacol Biochem Behav. 2004;77:405–413. doi: 10.1016/j.pbb.2003.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ji G, et al. Pain-related anxiety-like behavior requires CRF1 receptors in the amygdala. Mol Pain. 2007;3:13. doi: 10.1186/1744-8069-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fu Y, Neugebauer V. Differential mechanisms of CRF1 and CRF2 receptor functions in the amygdala in pain-related synaptic facilitation and behavior. J Neurosci. 2008;28:3861–3876. doi: 10.1523/JNEUROSCI.0227-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.He L, et al. 4-(1,3-Dimethoxyprop-2-ylamino)-2,7-dimethyl-8-(2,4-dichlorophenyl)pyrazolo[1,5-a]-1,3,5-triazine: a potent, orally bioavailable CRF(1) receptor antagonist. J Med Chem. 2000;43:449–456. doi: 10.1021/jm9904351. [DOI] [PubMed] [Google Scholar]
  • 33.Habib KE, et al. Oral administration of a corticotropin-releasing hormone receptor antagonist significantly attenuates behavioral, neuroendocrine, and autonomic responses to stress in primates. Proc Natl Acad Sci U S A. 2000;97:6079–6084. doi: 10.1073/pnas.97.11.6079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nielsen DM, et al. Antidepressant-like activity of corticotropin-releasing factor type-1 receptor antagonists in mice. Eur J Pharmacol. 2004;499:135–146. doi: 10.1016/j.ejphar.2004.07.091. [DOI] [PubMed] [Google Scholar]
  • 35.Overstreet DH, Griebel G. Antidepressant-like effects of CRF1 receptor antagonist SSR125543 in an animal model of depression. Eur J Pharmacol. 2004;497:49–53. doi: 10.1016/j.ejphar.2004.06.035. [DOI] [PubMed] [Google Scholar]
  • 36.Bale TL, Vale WW. Increased depression-like behaviors in corticotropin-releasing factor receptor-2-deficient mice: sexually dichotomous responses. J Neurosci. 2003;23:5295–5301. doi: 10.1523/JNEUROSCI.23-12-05295.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Harro J, et al. The effects of CRA 1000, a non-peptide antagonist of corticotropin-releasing factor receptor type 1, on adaptive behaviour in the rat. Neuropeptides. 2001;35:100–109. doi: 10.1054/npep.2001.0851. [DOI] [PubMed] [Google Scholar]
  • 38.Jutkiewicz EM, et al. The effects of CRF antagonists, antalarmin, CP154,526, LWH234, and R121919, in the forced swim test and on swim-induced increases in adrenocorticotropin in rats. Psychopharmacology (Berl) 2005;180:215–223. doi: 10.1007/s00213-005-2164-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chaki S, et al. Anxiolytic- and antidepressant-like profile of a new CRF1 receptor antagonist, R278995/CRA0450. Eur J Pharmacol. 2004;485:145–158. doi: 10.1016/j.ejphar.2003.11.032. [DOI] [PubMed] [Google Scholar]
  • 40.Song C, Leonard BE. The olfactory bulbectomised rat as a model of depression. Neurosci Biobehav Rev. 2005;29:627–647. doi: 10.1016/j.neubiorev.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 41.Ducottet C, et al. Effects of the selective nonpeptide corticotropin-releasing factor receptor 1 antagonist antalarmin in the chronic mild stress model of depression in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:625–631. doi: 10.1016/S0278-5846(03)00051-4. [DOI] [PubMed] [Google Scholar]
  • 42.Alonso R, et al. Blockade of CRF1 or V1b receptors reverses stress-induced suppression of neurogenesis in a mouse model of depression. Mol Psychiatry. 2004;9:278–286. doi: 10.1038/sj.mp.4001464. [DOI] [PubMed] [Google Scholar]
  • 43.Yamano M, et al. Corticotropin-releasing hormone receptors mediate consensus interferon-alpha YM643-induced depression-like behavior in mice. J Pharmacol Exp Ther. 2000;292:181–187. [PubMed] [Google Scholar]
  • 44.Oshima A, et al. Altered serotonergic neurotransmission but normal hypothalamic-pituitary-adrenocortical axis activity in mice chronically treated with the corticotropin-releasing hormone receptor type 1 antagonist NBI 30775. Neuropsychopharmacology. 2003;28:2148–2159. doi: 10.1038/sj.npp.1300267. [DOI] [PubMed] [Google Scholar]
  • 45.Liu X, et al. Tail-suspension induced hyperthermia: a new measure of stress reactivity. J Psychiatr Res. 2003;37:249–259. doi: 10.1016/s0022-3956(03)00004-9. [DOI] [PubMed] [Google Scholar]
  • 46.Mansbach RS, et al. Antidepressant-like effects of CP-154,526, a selective CRF1 receptor antagonist. Eur J Pharmacol. 1997;323:21–26. doi: 10.1016/s0014-2999(97)00025-3. [DOI] [PubMed] [Google Scholar]
  • 47.Takamori K, et al. Involvement of the hypothalamus-pituitary-adrenal axis in antidepressant activity of corticotropin-releasing factor subtype 1 receptor antagonists in the rat learned helplessness test. Pharmacol Biochem Behav. 2001a;69:445–449. doi: 10.1016/s0091-3057(01)00515-9. [DOI] [PubMed] [Google Scholar]
  • 48.Li YW, et al. The pharmacology of DMP696 and DMP904, non-peptidergic CRF1 receptor antagonists. CNS Drug Rev. 2005;11:21–52. doi: 10.1111/j.1527-3458.2005.tb00034.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Takamori K, et al. Involvement of corticotropin-releasing factor subtype 1 receptor in the acquisition phase of learned helplessness in rats. Life Sci. 2001b;69:1241–1248. doi: 10.1016/s0024-3205(01)01217-6. [DOI] [PubMed] [Google Scholar]
  • 50.Sabino V, et al. Dissociation between opioid and CRF1 antagonist sensitive drinking in Sardinian alcohol-preferring rats. Psychopharmacology (Berl) 2006;189:175–186. doi: 10.1007/s00213-006-0546-5. [DOI] [PubMed] [Google Scholar]
  • 51.Funk CK, et al. Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biol Psychiatry. 2007;61:78–86. doi: 10.1016/j.biopsych.2006.03.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Richardson HN, et al. MPZP: a novel small molecule corticotropin-releasing factor type 1 receptor (CRF1) antagonist. Pharmacol Biochem Behav. 2008;88:497–510. doi: 10.1016/j.pbb.2007.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gilpin NW, et al. Effects of CRF1-receptor and opioid-receptor antagonists on dependence-induced increases in alcohol drinking by alcohol-preferring (P) rats. Alcohol Clin Exp Res. 2008;32:1535–1542. doi: 10.1111/j.1530-0277.2008.00745.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sommer WH, et al. Upregulation of voluntary alcohol intake, behavioral sensitivity to stress, and amygdala crhr1 expression following a history of dependence. Biol Psychiatry. 2008;63:139–145. doi: 10.1016/j.biopsych.2007.01.010. [DOI] [PubMed] [Google Scholar]
  • 55.Gehlert DR, et al. 3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo[1,2-b]pyridazine: a novel brain-penetrant, orally available corticotropin-releasing factor receptor 1 antagonist with efficacy in animal models of alcoholism. J Neurosci. 2007;27:2718–2726. doi: 10.1523/JNEUROSCI.4985-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.George O, et al. CRF-CRF1 system activation mediates withdrawal-induced increases in nicotine self-administration in nicotine-dependent rats. Proc Natl Acad Sci U S A. 2007;104:17198–17203. doi: 10.1073/pnas.0707585104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Specio SE, et al. CRF1 receptor antagonists attenuate escalated cocaine self-administration in rats. Psychopharmacology (Berl) 2008;196:473–482. doi: 10.1007/s00213-007-0983-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Greenwell TN, et al. Corticotropin-releasing factor-1 receptor antagonists decrease heroin self-administration in long-, but not short-access rats. Addict Biol. 2009;14:130–143. doi: 10.1111/j.1369-1600.2008.00142.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Koob GF, Zorrilla EP. Neurobiological mechanisms of addiction: focus on corticotropin-releasing factor. Curr Opin Investig Drugs. (in press) [PMC free article] [PubMed] [Google Scholar]
  • 60.Koob GF, Kreek MJ. Stress, dysregulation of drug reward pathways, and the transition to drug dependence. Am J Psychiatry. 2007;164:1149–1159. doi: 10.1176/appi.ajp.2007.05030503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Koob GF. A role for brain stress systems in addiction. Neuron. 2008;59:11–34. doi: 10.1016/j.neuron.2008.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stinus L, et al. Buprenorphine and a CRF1 antagonist block the acquisition of opiate withdrawal-induced conditioned place aversion in rats. Neuropsychopharmacology. 2005;30:90–98. doi: 10.1038/sj.npp.1300487. [DOI] [PubMed] [Google Scholar]
  • 63.Skelton KH, et al. The CRF1 receptor antagonist R121919 attenuates the neuroendocrine and behavioral effects of precipitated lorazepam withdrawal. Psychopharmacology (Berl) 2007;192:385–396. doi: 10.1007/s00213-007-0713-3. [DOI] [PubMed] [Google Scholar]
  • 64.Lu L, et al. Corticotropin-releasing factor receptor type I mediates stress-induced relapse to opiate dependence in rats. Neuroreport. 2000a;11:2373–2378. doi: 10.1097/00001756-200008030-00008. [DOI] [PubMed] [Google Scholar]
  • 65.Contarino A, Papaleo F. The corticotropin-releasing factor receptor-1 pathway mediates the negative affective states of opiate withdrawal. Proc Natl Acad Sci U S A. 2005;102:18649–18654. doi: 10.1073/pnas.0506999102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bruijnzeel AW, Prado M, Isaac S. Corticotropin-releasing factor-1 receptor activation mediates nicotine withdrawal-induced deficit in brain reward function and stress-induced relapse. Biological Psychiatry. 2009 doi: 10.1016/j.biopsych.2009.01.010. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Knapp DJ, et al. SB242084, flumazenil, and CRA1000 block ethanol withdrawal-induced anxiety in rats. Alcohol. 2004;32:101–111. doi: 10.1016/j.alcohol.2003.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Breese GR, et al. Prior multiple ethanol withdrawals enhance stress-induced anxiety-like behavior: inhibition by CRF1- and benzodiazepine-receptor antagonists and a 5-HT1a-receptor agonist. Neuropsychopharmacology. 2005;30:1662–1669. doi: 10.1038/sj.npp.1300706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chu K, et al. Dependence-induced increases in ethanol self-administration in mice are blocked by the CRF1 receptor antagonist antalarmin and by CRF1 receptor knockout. Pharmacol Biochem Behav. 2007;86:813–821. doi: 10.1016/j.pbb.2007.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hansson AC, et al. Variation at the rat Crhr1 locus and sensitivity to relapse into alcohol seeking induced by environmental stress. Proc Natl Acad Sci U S A. 2006;103:15236–15241. doi: 10.1073/pnas.0604419103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lodge DJ, Lawrence AJ. The CRF1 receptor antagonist antalarmin reduces volitional ethanol consumption in isolation-reared Fawn-Hooded rats. Neuroscience. 2003;117:243–247. doi: 10.1016/s0306-4522(02)00793-5. [DOI] [PubMed] [Google Scholar]
  • 72.Shaham Y, et al. The reinstatement model of drug relapse: history, methodology and major findings. Psychopharmacology (Berl) 2003;168:3–20. doi: 10.1007/s00213-002-1224-x. [DOI] [PubMed] [Google Scholar]
  • 73.Le AD, et al. The role of corticotrophin-releasing factor in stress-induced relapse to alcohol-seeking behavior in rats. Psychopharmacology (Berl) 2000;150:317–324. doi: 10.1007/s002130000411. [DOI] [PubMed] [Google Scholar]
  • 74.Shaham Y, et al. CP-154,526, a selective, non-peptide antagonist of the corticotropin-releasing factor1 receptor attenuates stress-induced relapse to drug seeking in cocaine- and heroin-trained rats. Psychopharmacology (Berl) 1998;137:184–190. doi: 10.1007/s002130050608. [DOI] [PubMed] [Google Scholar]
  • 75.Marinelli PW, et al. The CRF1 receptor antagonist antalarmin attenuates yohimbine-induced increases in operant alcohol self-administration and reinstatement of alcohol seeking in rats. Psychopharmacology (Berl) 2007;195:345–355. doi: 10.1007/s00213-007-0905-x. [DOI] [PubMed] [Google Scholar]
  • 76.Lu L, et al. Differential roles of corticotropin-releasing factor receptor subtypes 1 and 2 in opiate withdrawal and in relapse to opiate dependence. Eur J Neurosci. 2000b;12:4398–4404. [PubMed] [Google Scholar]
  • 77.Lu L, et al. Corticotropin-releasing factor receptor type 1 mediates stress-induced relapse to cocaine-conditioned place preference in rats. Eur J Pharmacol. 2001;415:203–208. doi: 10.1016/s0014-2999(01)00840-8. [DOI] [PubMed] [Google Scholar]
  • 78.Yamada Y, et al. New class of corticotropin-releasing factor (CRF) antagonists: small peptides having high binding affinity for CRF receptor. J Med Chem. 2004;47:1075–1078. doi: 10.1021/jm034180+. [DOI] [PubMed] [Google Scholar]
  • 79.Rijkers DTS, et al. Peptides having corticotropin releasing factor (CRF) antagonist activity derived from astressin for treating stress-related disorders. PCT Int Appl. 2001:24. CODEN: PIXXD2 WO 2001029086 A1 20010426 CAN 134:336435 AN 2001:300761 CAPLUS.
  • 80.Yamada, H. et al. (2003) Preparation of cyclic peptides as corticotropin-releasing factor (CRF) receptor antagonists. (Japan Tobacco, Inc., Japan). Jpn. Kokai Tokkyo Koho (2003), 50 pp. CODEN: JKXXAF JP 2003335798 A 20031128 Patent written in Japanese. Application: JP 2002-143881 20020517. Priority: CAN 140:5306 AN 2003:929565 CAPLUS
  • 81.Hoare SR. Mechanisms of peptide and nonpeptide ligand binding to Class B G-protein-coupled receptors. Drug Discov Today. 2005;10:417–427. doi: 10.1016/S1359-6446(05)03370-2. [DOI] [PubMed] [Google Scholar]
  • 82.Hoare SR, et al. Ligand affinity for amino-terminal and juxtamembrane domains of the corticotropin releasing factor type I receptor: regulation by G-protein and nonpeptide antagonists. Biochemistry. 2004;43:3996–4011. doi: 10.1021/bi036110a. [DOI] [PubMed] [Google Scholar]
  • 83.Perry SJ, et al. Distinct conformations of the corticotropin releasing factor type 1 receptor adopted following agonist and antagonist binding are differentially regulated. J Biol Chem. 2005;280:11560–11568. doi: 10.1074/jbc.M412914200. [DOI] [PubMed] [Google Scholar]
  • 84.Mesleh MF, et al. NMR structural characterization of a minimal peptide antagonist bound to the extracellular domain of the corticotropin-releasing factor1 receptor. J Biol Chem. 2007;282:6338–6346. doi: 10.1074/jbc.M609816200. [DOI] [PubMed] [Google Scholar]
  • 85.Grace CR, et al. Common and divergent structural features of a series of corticotropin releasing factor-related peptides. J Am Chem Soc. 2007;129:16102–16114. doi: 10.1021/ja0760933. [DOI] [PubMed] [Google Scholar]
  • 86.Beyermann M, et al. Achieving signalling selectivity of ligands for the corticotropin-releasing factor type 1 receptor by modifying the agonist’s signalling domain. Br J Pharmacol. 2007;151:851–859. doi: 10.1038/sj.bjp.0707293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Berger H, et al. Evidence that corticotropin-releasing factor receptor type 1 couples to Gs- and Gi-proteins through different conformations of its J-domain. Br J Pharmacol. 2006;149:942–947. doi: 10.1038/sj.bjp.0706926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hoare SR, et al. Allosteric ligands for the corticotropin releasing factor type 1 receptor modulate conformational states involved in receptor activation. Mol Pharmacol. 2008;73:1371–1380. doi: 10.1124/mol.107.042978. [DOI] [PubMed] [Google Scholar]
  • 89.Hoare SR, et al. Single amino acid residue determinants of non-peptide antagonist binding to the corticotropin-releasing factor1 (CRF1) receptor. Biochem Pharmacol. 2006;72:244–255. doi: 10.1016/j.bcp.2006.04.007. [DOI] [PubMed] [Google Scholar]
  • 90.Liaw CW, et al. Localization of agonist- and antagonist-binding domains of human corticotropin-releasing factor receptors. Mol Endocrinol. 1997;11:2048–2053. doi: 10.1210/mend.11.13.0034. [DOI] [PubMed] [Google Scholar]
  • 91.Dzierba CD, et al. Dihydropyridopyrazinones and dihydropteridinones as corticotropin-releasing factor-1 receptor antagonists: structure-activity relationships and computational modeling. J Med Chem. 2007;50:2269–2272. doi: 10.1021/jm0611410. [DOI] [PubMed] [Google Scholar]
  • 92.Di Fabio R, et al. Dihydropyrrole[2,3-d]pyridine derivatives as novel corticotropin-releasing factor-1 antagonists: mapping of the receptor binding pocket by in silico docking studies. J Med Chem. 2008a;51:7273–7286. doi: 10.1021/jm800743q. [DOI] [PubMed] [Google Scholar]
  • 93.Kerns EH, Di L. Drug-like Properties: Concepts, Structure Design and Methods—From ADME to Toxicity Optimization. Elsevier; Amsterdam: 2007. [Google Scholar]
  • 94.Guo Z, et al. Design and synthesis of tricyclic imidazo[4,5-b]pyridin-2-ones as corticotropin-releasing factor-1 antagonists. J Med Chem. 2005;48:5104–5107. doi: 10.1021/jm050384+. [DOI] [PubMed] [Google Scholar]
  • 95.Di Fabio R, et al. Synthesis and pharmacological characterization of novel druglike corticotropin-releasing factor 1 antagonists. J Med Chem. 2008b;51:7370–7379. doi: 10.1021/jm800744m. [DOI] [PubMed] [Google Scholar]
  • 96.Chen YL, et al. Synthesis and SAR of 2-aryloxy-4-alkoxy-pyridines as potent orally active corticotropin-releasing factor 1 receptor antagonists. J Med Chem. 2008a;51:1377–1384. doi: 10.1021/jm070578k. [DOI] [PubMed] [Google Scholar]
  • 97.Chen YL, et al. 2-aryloxy-4-alkylaminopyridines: discovery of novel corticotropin-releasing factor 1 antagonists. J Med Chem. 2008b;51:1385–1392. doi: 10.1021/jm070579c. [DOI] [PubMed] [Google Scholar]
  • 98.Gilligan PJ, et al. Synthesis and structure-activity relationships of 8-(pyrid-3-yl)pyrazolo[1,5-a]-1,3,5-triazines: potent, orally bioavailable corticotropin releasing factor receptor-1 (CRF1) antagonists. J Med Chem. 2009a;52:3084–3092. doi: 10.1021/jm900025h. [DOI] [PubMed] [Google Scholar]
  • 99.Binneman B, et al. A 6-week randomized, placebo-controlled trial of CP-316,311 (a selective CRH1 antagonist) in the treatment of major depression. Am J Psychiatry. 2008;165:617–620. doi: 10.1176/appi.ajp.2008.07071199. [DOI] [PubMed] [Google Scholar]
  • 100.Yoon T, et al. The design, synthesis and structure-activity relationships of 1-aryl-4-aminoalkylisoquinolines: a novel series of CRF-1 receptor antagonists. Bioorg Med Chem Lett. 2008a;18:891–896. doi: 10.1016/j.bmcl.2007.12.050. [DOI] [PubMed] [Google Scholar]
  • 101.Yoon T, et al. 2-Arylpyrimidines: novel CRF-1 receptor antagonists. Bioorg Med Chem Lett. 2008b;18:4486–4490. doi: 10.1016/j.bmcl.2008.07.063. [DOI] [PubMed] [Google Scholar]
  • 102.Lowe RF, et al. Rational design, synthesis, and structure-activity relationships of aryltriazoles as novel corticotropin-releasing factor-1 receptor antagonists. J Med Chem. 2005;48:1540–1549. doi: 10.1021/jm049339c. [DOI] [PubMed] [Google Scholar]
  • 103.Dyck B, et al. Potent, orally active corticotropin-releasing factor receptor-1 antagonists containing a tricyclic pyrrolopyridine or pyrazolopyridine core. J Med Chem. 2005;48:4100–4110. doi: 10.1021/jm050070m. [DOI] [PubMed] [Google Scholar]
  • 104.Gross RS, et al. Design and synthesis of tricyclic corticotropin-releasing factor-1 antagonists. J Med Chem. 2005;48:5780–5793. doi: 10.1021/jm049085v. [DOI] [PubMed] [Google Scholar]
  • 105.Ising M, et al. High-affinity CRF1 receptor antagonist NBI-34041: preclinical and clinical data suggest safety and efficacy in attenuating elevated stress response. Neuropsychopharmacology. 2007;32:1941–1949. doi: 10.1038/sj.npp.1301328. [DOI] [PubMed] [Google Scholar]
  • 106.St Denis Y, et al. Substituted tetraazaacenaphthylenes as potent CRF1 receptor antagonists for the treatment of depression and anxiety. Bioorg Med Chem Lett. 2005;15:3713–3716. doi: 10.1016/j.bmcl.2005.05.040. [DOI] [PubMed] [Google Scholar]
  • 107.Gentile G, et al. Novel substituted tetrahydrotriazaacenaphthylene derivatives as potent CRF1 receptor antagonists. Bioorg Med Chem Lett. 2007;17:5218–5221. doi: 10.1016/j.bmcl.2007.06.077. [DOI] [PubMed] [Google Scholar]
  • 108.Sabbatini FM, et al. Heteroaryl-substituted 4-(1H-pyrazol-1-yl)-5,6-dihydro-1H-pyrrolo[2,3-d]pyrimidine derivatives as potent and selective corticotropin-releasing factor receptor-1 antagonists. Chem Med Chem. 2008;3:226–229. doi: 10.1002/cmdc.200700230. [DOI] [PubMed] [Google Scholar]
  • 109.Corbett JW, et al. Heteroatom-linked indanylpyrazines are corticotropin releasing factor type-1 receptor antagonists. Bioorg Med Chem Lett. 2007;17:6250–6256. doi: 10.1016/j.bmcl.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 110.Han X, et al. Synthesis, structure-activity relationships, and anxiolytic activity of 7-aryl-6,7-dihydroimidazoimidazole corticotropin-releasing factor 1 receptor antagonists. Bioorg Med Chem Lett. 2005a;15:3870–3873. doi: 10.1016/j.bmcl.2005.05.117. [DOI] [PubMed] [Google Scholar]
  • 111.Han X, et al. Synthesis and structure-activity relationship of imidazo[1,2-a]benzimidazoles as corticotropin-releasing factor 1 receptor antagonists. Bioorg Med Chem Lett. 2005b;15:4029–4032. doi: 10.1016/j.bmcl.2005.06.028. [DOI] [PubMed] [Google Scholar]
  • 112.Han X, et al. An orally active corticotropin releasing factor 1 receptor antagonist from 8-aryl-1,3a,7,8-tetraaza-cyclopenta[a]indenes. Bioorg Med Chem Lett. 2007;17:2026–2030. doi: 10.1016/j.bmcl.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • 113.Hartz RA, et al. Synthesis and evaluation of 2-anilino-3-phenylsulfonyl-6-methylpyridines as corticotropin-releasing factor1 receptor ligands. Bioorg Med Chem Lett. 2006;16:934–937. doi: 10.1016/j.bmcl.2005.10.097. [DOI] [PubMed] [Google Scholar]
  • 114.Gilligan PJ, et al. 8-(4-Methoxyphenyl)pyrazolo[1,5-a]-1,3,5-triazines: selective and centrally active corticotropin-releasing factor receptor-1 (CRF1) antagonists. J Med Chem. 2009b;52:3073–3083. doi: 10.1021/jm9000242. [DOI] [PubMed] [Google Scholar]
  • 115.Horn TL, et al. Integration of in vivo and in vitro approaches to characterize the toxicity of Antalarmin, a corticotropin-releasing hormone receptor antagonist. Toxicology. 2008;248:8–17. doi: 10.1016/j.tox.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES