Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Pharmacol Exp Ther ; 338(1): 318-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21493753

ABSTRACT

The biological functions of the aryl hydrocarbon receptor (AHR) can be delineated into dioxin response element (DRE)-dependent or -independent activities. Ligands exhibiting either full or partial agonist activity, e.g., 2,3,7,8-tetrachlorodibenzo-p-dioxin and α-naphthoflavone, have been demonstrated to potentiate both DRE-dependent and -independent AHR function. In contrast, the recently identified selective AHR modulators (SAhRMs), e.g., 1-allyl-3-(3,4-dimethoxyphenyl)-7-(trifluoromethyl)-1H-indazole (SGA360), bias AHR toward DRE-independent functionality while displaying antagonism with regard to ligand-induced DRE-dependent transcription. Recent studies have expanded the physiological role of AHR to include modulation of hematopoietic progenitor expansion and immunoregulation. It remains to be established whether such physiological roles are mediated through DRE-dependent or -independent pathways. Here, we present evidence for a third class of AHR ligand, "pure" or complete antagonists with the capacity to suppress both DRE-dependent and -independent AHR functions, which may facilitate dissection of physiological AHR function with regard to DRE or non-DRE-mediated signaling. Competitive ligand binding assays together with in silico modeling identify N-(2-(1H-indol-3-yl)ethyl)-9-isopropyl-2-(5-methylpyridin-3-yl)-9H-purin-6-amine (GNF351) as a high-affinity AHR ligand. DRE-dependent reporter assays, in conjunction with quantitative polymerase chain reaction analysis of AHR targets, reveal GNF351 as a potent AHR antagonist that demonstrates efficacy in the nanomolar range. Furthermore, unlike many currently used AHR antagonists, e.g., α-naphthoflavone, GNF351 is devoid of partial agonist potential. It is noteworthy that in a model of AHR-mediated DRE-independent function, i.e., suppression of cytokine-induced acute-phase gene expression, GNF351 has the capacity to antagonize agonist and SAhRM-mediated suppression of SAA1. Such data indicate that GNF351 is a pure antagonist with the capacity to inhibit both DRE-dependent and -independent activity.


Subject(s)
Allyl Compounds/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , Purines/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Allyl Compounds/metabolism , Animals , Binding Sites/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Indazoles/metabolism , Indoles/chemistry , Indoles/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Purines/chemistry , Purines/metabolism
2.
ACS Med Chem Lett ; 12(12): 1912-1919, 2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34917254

ABSTRACT

The selective inhibition of RET kinase as a treatment for relevant cancer types including lung adenocarcinoma has garnered considerable interest in recent years and prompted a variety of efforts toward the discovery of small-molecule therapeutics. Hits uncovered via the analysis of archival kinase data ultimately led to the identification of a promising pyrrolo[2,3-d]pyrimidine scaffold. The optimization of this pyrrolo[2,3-d]pyrimidine core resulted in compound 1, which demonstrated potent in vitro RET kinase inhibition and robust in vivo efficacy in RET-driven tumor xenografts upon multiday dosing in mice. The administration of 1 was well-tolerated at established efficacious doses (10 and 30 mg/kg, po, qd), and plasma exposure levels indicated a minimal risk of KDR or hERG inhibition in vivo, as evaluated by Miles assay and free plasma concentrations, respectively.

3.
Bioorg Med Chem Lett ; 20(24): 7259-64, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21074436

ABSTRACT

Antagonists of the corticotropin-releasing factor (CRF) neuropeptide may prove effective in treating stress and anxiety related disorders. In an effort to identify antagonists with improved physico-chemical properties a new series of CRF(1) antagonists were designed to substitute the propyl groups at the C7 position of the pyrazolo[1,5-a]pyrimidine core of 1 with heterocycles. Compound (S)-8d was identified as a high affinity ligand with a pK(i) value of 8.2 and a functional CRF(1) antagonist with pIC(50) value of 7.0 in the in vitro CRF ACTH production assay.


Subject(s)
Azabicyclo Compounds/chemistry , Oxadiazoles/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacokinetics , Humans , Microsomes, Liver/metabolism , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Protein Binding , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
ACS Med Chem Lett ; 11(4): 558-565, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292564

ABSTRACT

RET (REarranged during Transfection) kinase gain-of-function aberrancies have been identified as potential oncogenic drivers in lung adenocarcinoma, along with several other cancer types, prompting the discovery and assessment of selective inhibitors. Internal mining and analysis of relevant kinase data informed the decision to investigate a pyrazolo[1,5-a]pyrimidine scaffold, where subsequent optimization led to the identification of compound WF-47-JS03 (1), a potent RET kinase inhibitor with >500-fold selectivity against KDR (Kinase insert Domain Receptor) in cellular assays. In subsequent mouse in vivo studies, compound 1 demonstrated effective brain penetration and was found to induce strong regression of RET-driven tumor xenografts at a well-tolerated dose (10 mg/kg, po, qd). Higher doses of 1, however, were poorly tolerated in mice, similar to other pyrazolo[1,5-a]pyrimidine compounds at or near the efficacious dose, and indicative of the narrow therapeutic windows seen with this scaffold.

5.
J Med Chem ; 63(5): 2013-2027, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31059256

ABSTRACT

Direct pharmacological inhibition of RAS has remained elusive, and efforts to target CRAF have been challenging due to the complex nature of RAF signaling, downstream of activated RAS, and the poor overall kinase selectivity of putative RAF inhibitors. Herein, we describe 15 (LXH254, Aversa, R.; et al. Int. Patent WO2014151616A1, 2014), a selective B/C RAF inhibitor, which was developed by focusing on drug-like properties and selectivity. Our previous tool compound, 3 (RAF709; Nishiguchi, G. A.; et al. J. Med. Chem. 2017, 60, 4969), was potent, selective, efficacious, and well tolerated in preclinical models, but the high human intrinsic clearance precluded further development and prompted further investigation of close analogues. A structure-based approach led to a pyridine series with an alcohol side chain that could interact with the DFG loop and significantly improved cell potency. Further mitigation of human intrinsic clearance and time-dependent inhibition led to the discovery of 15. Due to its excellent properties, it was progressed through toxicology studies and is being tested in phase 1 clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , Mutation/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Discovery/trends , Humans , Molecular Docking Simulation/methods , Molecular Docking Simulation/trends , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays/methods
6.
Bioorg Med Chem Lett ; 18(20): 5420-3, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18815029

ABSTRACT

The discovery and synthesis of a series of (dimethoxyphenoxy)alkylamino acetamides as orexin-2 receptor antagonists from a small-molecule combinatorial library using a high-throughput calcium mobilization functional assay (HEK293-human OX2-R cell line) is described. Active compounds show a good correlation between high-throughput single concentration screening data and measured IC(50)s. Specific examples exhibit IC(50) values of approximately 20 nM using human orexin A as the peptide agonist for the orexin-2 receptor.


Subject(s)
Acetamides/chemical synthesis , Chemistry, Pharmaceutical/methods , Intracellular Signaling Peptides and Proteins/chemistry , Neuropeptides/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Acetamides/chemistry , Calcium/chemistry , Cell Line , Combinatorial Chemistry Techniques , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Mutation , Orexin Receptors , Orexins , Temperature
7.
Bioorg Med Chem Lett ; 18(6): 1778-83, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18329269
9.
Bioorg Med Chem Lett ; 18(20): 5402-5, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18835161

ABSTRACT

In this report, the strategy and outcome of expanding SAR exploration to improve solubility and metabolic stability are discussed. Compound 35 exhibited excellent potency, selectivity over A(1) and improved solubility of >4 mg/mL at pH 8.0. In addition, compound 35 had good metabolic stability with a scaled intrinsic clearance of 3 mL/min/kg (HLM) and demonstrated efficacy in the haloperidol induced catalepsy model.


Subject(s)
Adenosine A2 Receptor Antagonists , Aminopyridines/chemistry , Chemistry, Pharmaceutical/methods , Pyrimidines/chemical synthesis , Drug Design , Haloperidol/chemistry , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Models, Chemical , Parkinson Disease/therapy , Protein Binding , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2A/chemistry , Solubility , Structure-Activity Relationship
10.
Cancer Res ; 78(6): 1537-1548, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29343524

ABSTRACT

Resistance to the RAF inhibitor vemurafenib arises commonly in melanomas driven by the activated BRAF oncogene. Here, we report antitumor properties of RAF709, a novel ATP-competitive kinase inhibitor with high potency and selectivity against RAF kinases. RAF709 exhibited a mode of RAF inhibition distinct from RAF monomer inhibitors such as vemurafenib, showing equal activity against both RAF monomers and dimers. As a result, RAF709 inhibited MAPK signaling activity in tumor models harboring either BRAFV600 alterations or mutant N- and KRAS-driven signaling, with minimal paradoxical activation of wild-type RAF. In cell lines and murine xenograft models, RAF709 demonstrated selective antitumor activity in tumor cells harboring BRAF or RAS mutations compared with cells with wild-type BRAF and RAS genes. RAF709 demonstrated a direct pharmacokinetic/pharmacodynamic relationship in in vivo tumor models harboring KRAS mutation. Furthermore, RAF709 elicited regression of primary human tumor-derived xenograft models with BRAF, NRAS, or KRAS mutations with excellent tolerability. Our results support further development of inhibitors like RAF709, which represents a next-generation RAF inhibitor with unique biochemical and cellular properties that enables antitumor activities in RAS-mutant tumors.Significance: In an effort to develop RAF inhibitors with the appropriate pharmacological properties to treat RAS mutant tumors, RAF709, a compound with potency, selectivity, and in vivo properties, was developed that will allow preclinical therapeutic hypothesis testing, but also provide an excellent probe to further unravel the complexities of RAF kinase signaling. Cancer Res; 78(6); 1537-48. ©2018 AACR.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Proto-Oncogene Proteins B-raf/genetics , raf Kinases/antagonists & inhibitors , ras Proteins/genetics , 2,2'-Dipyridyl/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice, Nude , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Xenograft Model Antitumor Assays , raf Kinases/metabolism
11.
J Med Chem ; 60(12): 4869-4881, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28557458

ABSTRACT

RAS oncogenes have been implicated in >30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , raf Kinases/antagonists & inhibitors , ras Proteins/genetics , 2,2'-Dipyridyl/chemistry , 2,2'-Dipyridyl/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzamides/chemistry , Crystallography, X-Ray , Dogs , Drug Design , Drug Discovery , Drug Stability , Humans , Inhibitory Concentration 50 , Mice , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
12.
J Med Chem ; 48(1): 171-9, 2005 Jan 13.
Article in English | MEDLINE | ID: mdl-15634011

ABSTRACT

In a previous report we demonstrated that merging together key structural elements present in an AT(1) receptor antagonist (1, irbesartan) with key structural elements in a biphenylsulfonamide ET(A) receptor antagonist (2) followed by additional optimization provided compound 3 as a dual-action receptor antagonist (DARA), which potently blocked both AT(1) and ET(A) receptors. Described herein are our efforts directed toward improving both the pharmacokinetic profile as well as the AT(1) and ET(A) receptor potency of 3. Our efforts centered on modifying the 2'-side chain of 3 and examining the isoxazolylsulfonamide moiety in 3. This effort resulted in the discovery of 7 as a highly potent second-generation DARA. Compound 7 also showed substantially improved pharmacokinetic properties compared to 3. In rats, DARA 7 reduced blood pressure elevations caused by intravenous infusion of Ang II or big ET-1 to a greater extent and with longer duration than DARA 3 or AT(1) or ET(A) receptor antagonists alone. Compound 7 clearly demonstrated superiority over irbesartan (an AT(1) receptor antagonist) in the normal SHR model of hypertension in a dose-dependent manner, demonstrating the synergy of AT(1) and ET(A) receptor blockade in a single molecule.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Endothelin A Receptor Antagonists , Isoxazoles/chemistry , Isoxazoles/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Sulfonamides/chemistry , Sulfonamides/pharmacology , Administration, Oral , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Biological Availability , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Dogs , Humans , Hypertension/drug therapy , Irbesartan , Isoxazoles/pharmacokinetics , Macaca fascicularis , Male , Rats , Rats, Inbred SHR , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Tetrazoles/chemistry , Tetrazoles/pharmacology
13.
J Med Chem ; 48(16): 5104-7, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-16078829

ABSTRACT

The synthesis and SAR studies of tricyclic imidazo[4,5-b]pyridin-2-ones as human corticotropin-releasing factor receptor (CRF(1)) antagonists are discussed herein. Compound 16g was identified as a functional antagonist that inhibited CRF-stimulated cyclic adenosine monophosphate production and CRF-induced adrenocorticotrophic hormone release. Pharmacokinetics studies in rats showed that 16g was orally bioavailable, had good brain penetration, and had a moderate half-life. In our effort to identify CRF(1) antagonists with improved pharmacokinetics properties, 16g exhibited a favorably lower volume of distribution.


Subject(s)
Heterocyclic Compounds, 3-Ring/chemical synthesis , Imidazoles/chemical synthesis , Pyridines/chemical synthesis , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Administration, Oral , Adrenocorticotropic Hormone/blood , Animals , Blood-Brain Barrier/metabolism , CHO Cells , Corticotropin-Releasing Hormone/pharmacology , Cricetinae , Cricetulus , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/biosynthesis , Drug Design , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Injections, Intravenous , Male , Pyridines/pharmacokinetics , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
14.
J Med Chem ; 45(18): 3829-35, 2002 Aug 29.
Article in English | MEDLINE | ID: mdl-12190306

ABSTRACT

The ET(A) receptor antagonist (2) (N-(3,4-dimethyl-5-isoxazolyl)-4'-(2-oxazolyl)-[1,1'-biphenyl]-2-sulfonamide, BMS-193884) shares the same biphenyl core as a large number of AT(1) receptor antagonists, including irbesartan (3). Thus, it was hypothesized that merging the structural elements of 2 with those of the biphenyl AT(1) antagonists (e.g., irbesartan) would yield a compound with dual activity for both receptors. This strategy led to the design, synthesis, and discovery of (15) (4'-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(3,4-dimethyl-5-isoxazolyl)-2'-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-[1,1'-biphenyl]-2-sulfonamide, BMS-248360) as a potent and orally active dual antagonist of both AT(1) and ET(A) receptors. Compound 15 represents a new approach to treating hypertension.


Subject(s)
Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Endothelin Receptor Antagonists , Isoxazoles/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , CHO Cells , Cricetinae , Crystallography, X-Ray , Isoxazoles/chemistry , Isoxazoles/pharmacology , Molecular Structure , Radioligand Assay , Rats , Receptor, Angiotensin, Type 1 , Receptor, Endothelin A , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
15.
J Org Chem ; 61(18): 6153-6161, 1996 Sep 06.
Article in English | MEDLINE | ID: mdl-11667449

ABSTRACT

We report our synthesis of the C(26)-C(37) fragment of serine/threonine protein phosphatase PP1 and PP2A inhibitor calyculin C (1). Outlined in this paper are synthetic approaches to the two components based on disconnection at the C(33)-N(3) amide bond. We report the successful synthesis of the C(33)-C(37) aza-sugar derived from D-lyxose which was coupled onto a C(26)-C(32) aminooxazole originating from L-pyroglutamic acid. Elaboration of the resulting amide to a fully deprotected C(26)-C(37) fragment of calyculin C completed our synthesis. This provided an appropriate phosphonium salt for use in a Wittig olefination for joining both halves of the natural product.

16.
Cancer Cell ; 23(5): 594-602, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23680146

ABSTRACT

ATP competitive inhibitors of the BRAF(V600E) oncogene paradoxically activate downstream signaling in cells bearing wild-type BRAF (BRAF(WT)). In this study, we investigate the biochemical mechanism of wild-type RAF (RAF(WT)) activation by multiple catalytic inhibitors using kinetic analysis of purified BRAF(V600E) and RAF(WT) enzymes. We show that activation of RAF(WT) is ATP dependent and directly linked to RAF kinase activity. These data support a mechanism involving inhibitory autophosphorylation of RAF's phosphate-binding loop that, when disrupted either through pharmacologic or genetic alterations, results in activation of RAF and the mitogen-activated protein kinase (MAPK) pathway. This mechanism accounts not only for compound-mediated activation of the MAPK pathway in BRAF(WT) cells but also offers a biochemical mechanism for BRAF oncogenesis.


Subject(s)
MAP Kinase Signaling System/drug effects , raf Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/physiology , Cell Line, Tumor , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/physiology , raf Kinases/genetics , raf Kinases/metabolism
18.
J Med Chem ; 52(3): 709-17, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19140664

ABSTRACT

In the present article, we report on a strategy to improve the physical properties of a series of small molecule human adenosine 2A (hA2A) antagonists. One of the aromatic rings typical of this series of antagonists is replaced with a series of aliphatic groups, with the aim of disrupting crystal packing of the molecule to lower the melting point and in turn to improve the solubility. Herein, we describe the SAR of a new series of water-soluble 2,4,6-trisubstituted pyrimidines where R1 is an aromatic heterocycle, R2 is a short-chain alkyl amide, and the typical R3 aromatic heterocyclic substituent is replaced with an aliphatic amino substituent. This approach significantly enhanced aqueous solubility and lowered the log P of the system to provide molecules without significant hERG or CYP liabilities and robust in vivo efficacy.


Subject(s)
Acetamides/therapeutic use , Adenosine A2 Receptor Antagonists , Pyrimidines/therapeutic use , Acetamides/chemical synthesis , Adenosine A1 Receptor Antagonists , Animals , Behavior, Animal/drug effects , Catalepsy/chemically induced , Catalepsy/drug therapy , Drug Synergism , Haloperidol , Humans , Pyrimidines/chemical synthesis , Rats , Rotation , Solubility , Structure-Activity Relationship
19.
Curr Top Med Chem ; 8(6): 506-20, 2008.
Article in English | MEDLINE | ID: mdl-18397172

ABSTRACT

Antagonists of the corticotropin releasing factor (CRF or CRH) receptor have shown promise for the treatment of anxiety, depression, and irritable bowel syndrome. In the present article, medicinal chemistry developments surrounding small molecule CRF receptor antagonists are reviewed, focusing on publications and patents from mid-2004 through the first quarter of 2006. While the CRF type 2 receptor remains an intractable target, incremental progress has been made in the search for drug-like antagonists of the CRF type 1 receptor. Most recent work has not ventured far from previously-established pharmacophoric topologies. A common theme in recent patent disclosures is the addition of novel polar substituents to known heterocyclic core structures to reduce overall lipophilicity. New disclosures of pharmacokinetic (PK) data for several series of antagonists reveal that achieving appropriate PK remains a challenge for the field. The recent publication of selection patents and patents relating to salt and crystal forms of particular compounds suggests that several second generation compounds are nearing or have entered clinical development.


Subject(s)
Anti-Anxiety Agents/chemistry , Antidepressive Agents/chemistry , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Chemistry, Pharmaceutical , Depression/drug therapy , Irritable Bowel Syndrome/drug therapy , Structure-Activity Relationship
20.
J Med Chem ; 51(22): 7099-110, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18947224

ABSTRACT

4-Acetylamino-2-(3,5-dimethylpyrazol-1-yl)-pyrimidines bearing substituted pyridyl groups as C-6 substituents were prepared as selective adenosine hA2A receptor antagonists for the treatment of Parkinson's disease. The 5-methoxy-3-pyridyl derivative 6g (hA2A Ki 2.3 nM, hA1 Ki 190 nM) was orally active at 3 mg/kg in a rat HIC model but exposure was poor in nonrodent species, presumably due to poor aqueous solubility. Follow-on compound 16a (hA2A Ki 0.83 nM, hA1 Ki 130 nM), bearing a 6-(morpholin-4-yl)-2-pyridyl substituent at C-6, had improved solubility and was orally efficacious (3 mg/kg, HIC) but showed time-dependent cytochrome P450 3A4 inhibition, possibly related to morpholine ring metabolism. Compound 16j (hA2A Ki 0.44 nM, hA1 Ki 80 nM), bearing a 6-(4-methoxypiperidin-1-yl)-2-pyridyl substituent at C-6, was sparingly soluble but had good oral exposure in rodent and nonrodent species, had no cytochrome P450 or human ether-a-go-go related gene channel issues, and was orally efficacious at 1 mg/kg in HIC and at 3 mg/kg for potentiation of l-dopa-induced contralateral rotations in 6-hydroxydopamine-lesioned rats.


Subject(s)
Adenosine A2 Receptor Antagonists , Parkinson Disease/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Catalepsy/chemically induced , Catalepsy/drug therapy , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Haloperidol , Humans , Ligands , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL