Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
ACS Med Chem Lett ; 10(1): 50-55, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30655946

ABSTRACT

The further optimization of ER-α degradation efficacy of a series of ER modulators by refining side-chain substitution led to efficacious selective estrogen receptor degraders (SERDs). A fluoromethyl azetidine group was found to be preferred and resulted in the identification of bis-phenol chromene 17ha. In a tamoxifen-resistant breast cancer xenograft model, 17ha (ER-α degradation efficacy = 97%) demonstrated tumor regression, together with robust reduction of intratumoral ER-α levels. However, despite superior oral exposure, 5a (ER-α degradation efficacy = 91%) had inferior activity. This result suggests that optimizing ER-α degradation efficacy leads to compounds with robust effects in a model of tamoxifen-resistant breast cancer. Compound 17ha (GDC-0927) was evaluated in clinical trials in women with metastatic estrogen receptor-positive breast cancer.

3.
Bioorg Med Chem Lett ; 29(3): 367-372, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30587451
4.
J Med Chem ; 61(17): 7917-7928, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30086626

ABSTRACT

About 75% of breast cancers are estrogen receptor alpha (ER-α) positive, and women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, but resistance often emerges. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and shows some activity in patients who have progressed on antihormonal agents. However, fulvestrant must be administered by intramuscular injections that limit its efficacy. We describe the optimization of ER-α degradation efficacy of a chromene series of ER modulators resulting in highly potent and efficacious SERDs such as 14n. When examined in a xenograft model of tamoxifen-resistant breast cancer, 14n (ER-α degradation efficacy = 91%) demonstrated robust activity, while, despite superior oral exposure, 15g (ER-α degradation efficacy = 82%) was essentially inactive. This result suggests that optimizing ER-α degradation efficacy in the MCF-7 cell line leads to compounds with robust effects in models of tamoxifen-resistant breast cancer derived from an MCF-7 background.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzopyrans/chemistry , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Selective Estrogen Receptor Modulators/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice , Rats , Selective Estrogen Receptor Modulators/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Elife ; 52016 07 13.
Article in English | MEDLINE | ID: mdl-27410477

ABSTRACT

ER-targeted therapeutics provide valuable treatment options for patients with ER+ breast cancer, however, current relapse and mortality rates emphasize the need for improved therapeutic strategies. The recent discovery of prevalent ESR1 mutations in relapsed tumors underscores a sustained reliance of advanced tumors on ERα signaling, and provides a strong rationale for continued targeting of ERα. Here we describe GDC-0810, a novel, non-steroidal, orally bioavailable selective ER downregulator (SERD), which was identified by prospectively optimizing ERα degradation, antagonism and pharmacokinetic properties. GDC-0810 induces a distinct ERα conformation, relative to that induced by currently approved therapeutics, suggesting a unique mechanism of action. GDC-0810 has robust in vitro and in vivo activity against a variety of human breast cancer cell lines and patient derived xenografts, including a tamoxifen-resistant model and those that harbor ERα mutations. GDC-0810 is currently being evaluated in Phase II clinical studies in women with ER+ breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Cinnamates/administration & dosage , Indazoles/administration & dosage , Receptors, Estrogen/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Mice , Prospective Studies , Rats , Treatment Outcome
6.
Bioorg Med Chem Lett ; 25(22): 5163-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26463130

ABSTRACT

Selective estrogen receptor degraders (SERDs) have shown promise for the treatment of ER+ breast cancer. Disclosed herein is the continued optimization of our indazole series of SERDs. Exploration of ER degradation and antagonism in vitro followed by in vivo antagonism and oral exposure culminated in the discovery of indazoles 47 and 56, which induce tumor regression in a tamoxifen-resistant breast cancer xenograft.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/therapeutic use , Indazoles/therapeutic use , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cinnamates/therapeutic use , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/metabolism , Female , Indazoles/chemistry , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
J Med Chem ; 58(12): 4888-904, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25879485

ABSTRACT

Approximately 80% of breast cancers are estrogen receptor alpha (ER-α) positive, and although women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, resistance often emerges. Although a variety of resistance mechanism may be at play in this state, there is evidence that in many cases the ER still plays a central role, including mutations in the ER leading to constitutively active receptor. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and is active in patients who have progressed on antihormonal agents. However, fulvestrant suffers from poor pharmaceutical properties and must be administered by intramuscular injections that limit the total amount of drug that can be administered and hence lead to the potential for incomplete receptor blockade. We describe the identification and characterization of a series of small-molecule, orally bioavailable SERDs which are potent antagonists and degraders of ER-α and in which the ER-α degrading properties were prospectively optimized. The lead compound 11l (GDC-0810 or ARN-810) demonstrates robust activity in models of tamoxifen-sensitive and tamoxifen-resistant breast cancer, and is currently in clinical trials in women with locally advanced or metastatic estrogen receptor-positive breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/metabolism , Proteolysis/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Small Molecule Libraries/therapeutic use , Tamoxifen/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Dogs , Drug Discovery , Drug Resistance, Neoplasm/drug effects , Female , Heterografts , Humans , Mice , Rats , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics
8.
Cancer Res ; 72(6): 1494-503, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22266222

ABSTRACT

Continued reliance on the androgen receptor (AR) is now understood as a core mechanism in castration-resistant prostate cancer (CRPC), the most advanced form of this disease. While established and novel AR pathway-targeting agents display clinical efficacy in metastatic CRPC, dose-limiting side effects remain problematic for all current agents. In this study, we report the discovery and development of ARN-509, a competitive AR inhibitor that is fully antagonistic to AR overexpression, a common and important feature of CRPC. ARN-509 was optimized for inhibition of AR transcriptional activity and prostate cancer cell proliferation, pharmacokinetics, and in vivo efficacy. In contrast to bicalutamide, ARN-509 lacked significant agonist activity in preclinical models of CRPC. Moreover, ARN-509 lacked inducing activity for AR nuclear localization or DNA binding. In a clinically valid murine xenograft model of human CRPC, ARN-509 showed greater efficacy than MDV3100. Maximal therapeutic response in this model was achieved at 30 mg/kg/d of ARN-509, whereas the same response required 100 mg/kg/d of MDV3100 and higher steady-state plasma concentrations. Thus, ARN-509 exhibits characteristics predicting a higher therapeutic index with a greater potential to reach maximally efficacious doses in man than current AR antagonists. Our findings offer preclinical proof of principle for ARN-509 as a promising therapeutic in both castration-sensitive and castration-resistant forms of prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Prostatic Neoplasms/drug therapy , Thiohydantoins/therapeutic use , Androgen Antagonists/pharmacokinetics , Anilides/pharmacokinetics , Anilides/therapeutic use , Animals , Antineoplastic Agents, Hormonal/blood , Antineoplastic Agents, Hormonal/pharmacokinetics , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Nitriles/pharmacokinetics , Nitriles/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/blood , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/therapeutic use , Rats , Receptors, Androgen/drug effects , Thiohydantoins/blood , Thiohydantoins/chemical synthesis , Thiohydantoins/pharmacokinetics , Tosyl Compounds/pharmacokinetics , Tosyl Compounds/therapeutic use , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem Lett ; 22(2): 1237-41, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22182498

ABSTRACT

The overproduction of nitric oxide during the biological response to inflammation by the nitric oxide synthase (NOS) enzymes have been implicated in the pathology of many diseases. By removal of the amide core from uHTS-derived quinolone 4, a new series highly potent heteroaromatic-aminomethyl quinolone iNOS inhibitors 8 were identified. SAR studies led to identification of piperazine 22 and pyrimidine 32, both of which reduced plasma nitrates following oral dosing in a mouse lipopolysaccharide challenge assay.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Quinolones/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Ligands , Models, Molecular , Molecular Structure , Nitric Oxide Synthase Type II/metabolism , Quinolones/chemical synthesis , Quinolones/chemistry , Stereoisomerism , Structure-Activity Relationship
10.
J Med Chem ; 53(21): 7739-55, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20931971

ABSTRACT

Three isoforms of nitric oxide synthase (NOS), dimeric enzymes that catalyze the formation of nitric oxide (NO) from arginine, have been identified. Inappropriate or excessive NO produced by iNOS and/or nNOS is associated with inflammatory and neuropathic pain. Previously, we described the identification of a series of amide-quinolinone iNOS dimerization inhibitors that although potent, suffered from high clearance and limited exposure in vivo. By conformationally restricting the amide of this progenitor series, we describe the identification of a novel series of benzimidazole-quinolinone dual iNOS/nNOS inhibitors with low clearance and sustained exposure in vivo. Compounds were triaged utilizing an LPS challenge assay coupled with mouse and rhesus pharmacokinetics and led to the identification of 4,7-imidazopyrazine 42 as the lead compound. 42 (KD7332) (J. Med. Chem. 2009, 52, 3047 - 3062) was confirmed as an iNOS dimerization inhibitor and was efficacious in the mouse formalin model of nociception and Chung model of neuropathic pain, without showing tolerance after repeat dosing. Further 42 did not affect motor coordination up to doses of 1000 mg/kg, demonstrating a wide therapeutic margin.


Subject(s)
Analgesics/chemical synthesis , Fluoroquinolones/chemical synthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pain/drug therapy , Pyrazines/chemical synthesis , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacology , Animals , Cell Line , Drug Tolerance , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacology , Humans , In Vitro Techniques , Mice , Microsomes, Liver/metabolism , Pain/etiology , Pain Measurement , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/etiology , Protein Multimerization , Pyrazines/chemistry , Pyrazines/pharmacology , Rotarod Performance Test , Structure-Activity Relationship
11.
Mol Pharmacol ; 76(1): 153-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19364813

ABSTRACT

Nitric-oxide synthases (NOS) generate nitric oxide (NO) through the oxidation of l-arginine. Inappropriate or excessive production of NO by NOS is associated with the pathophysiology of various disease states. Efforts to treat these disorders by developing arginine mimetic, substrate-competitive NOS inhibitors as drugs have met with little success. Small-molecule-mediated inhibition of NOS dimerization represents an intriguing alternative to substrate-competitive inhibition. An ultra-high-throughput cell-based screen of 880,000 small molecules identified a novel quinolinone with inducible NOS (iNOS) inhibitory activity. Exploratory chemistry based on this initial screening hit resulted in the synthesis of KLYP956, which inhibits iNOS at low nanomolar concentrations. The iNOS inhibitory potency of KLYP956 is insensitive to changes in concentrations of the substrate arginine, or the cofactor tetrahydrobiopterin. Mechanistic analysis suggests that KLYP956 binds the oxygenase domain in the vicinity of the active site heme and inhibits iNOS and neuronal NOS (nNOS) by preventing the formation of enzymatically active dimers. Oral administration of KLYP956 [N-(3-chlorophenyl)-N-((8-fluoro-2-oxo-1,2-dihydroquinolin-4-yl)methyl)-4-methylthiazole-5-carboxamide] inhibits iNOS activity in a murine model of endotoxemia and blocks pain behaviors in a formalin model of nociception. KLYP956 thus represents the first nonimidazole-based inhibitor of iNOS and nNOS dimerization and provides a novel pharmaceutical alternative to previously described substrate competitive inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Fluoroquinolones/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Thiazoles/pharmacology , Administration, Oral , Animals , Cells, Cultured , Dimerization , Humans , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type II/chemistry , Pain/drug therapy , Species Specificity
12.
J Med Chem ; 52(9): 3047-62, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19374401

ABSTRACT

There are three isoforms of dimeric nitric oxide synthases (NOS) that convert arginine to citrulline and nitric oxide. Inducible NOS is implicated in numerous inflammatory diseases and, more recently, in neuropathic pain states. The majority of existing NOS inhibitors are either based on the structure of arginine or are substrate competitive. We describe the identification from an ultra high-throughput screen of a novel series of quinolinone small molecule, nonarginine iNOS dimerization inhibitors. SAR studies on the screening hit, coupled with an in vivo lipopolysaccharide (LPS) challenge assay measuring plasma nitrates and drug levels, rapidly led to the identification of compounds 12 and 42--potent inhibitors of the human and mouse iNOS enzyme that were highly selective over endothelial NOS (eNOS). Following oral dosing, compounds 12 and 42 gave a statistical reduction in pain behaviors in the mouse formalin model, while 12 also statistically reduced neuropathic pain behaviors in the chronic constriction injury (Bennett) model.


Subject(s)
Drug Discovery , Fluoroquinolones/administration & dosage , Fluoroquinolones/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pain/drug therapy , Protein Multimerization/drug effects , Pyrazines/administration & dosage , Pyrazines/pharmacology , Quinolones/administration & dosage , Quinolones/pharmacology , Administration, Oral , Animals , Cell Line , Constriction, Pathologic/chemically induced , Constriction, Pathologic/drug therapy , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Fluoroquinolones/chemistry , Fluoroquinolones/therapeutic use , Formaldehyde/toxicity , Humans , Inhibitory Concentration 50 , Lipopolysaccharides/toxicity , Mice , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , Protein Structure, Quaternary , Pyrazines/chemistry , Pyrazines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Structure-Activity Relationship , Substrate Specificity
13.
Bioorg Med Chem Lett ; 18(23): 6093-6, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18954983

ABSTRACT

We report the identification of KD5170, a potent mercaptoketone-based Class I and II-histone deacetylase inhibitor that demonstrates broad spectrum cytotoxic activity against a range of human tumor-derived cell lines. KD5170 exhibits robust and sustained histone H3 hyperacetylation in HCT-116 xenograft tumors following single oral or i.v. dose and inhibition of tumor growth following chronic dosing.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Prodrugs/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Molecular Structure , Prodrugs/chemistry , Pyridines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 18(24): 6482-5, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18954984

ABSTRACT

In an effort to discover novel non-hydroxamic acid histone deacetylase (HDAC) inhibitors, a novel alpha-mercaptoketone was identified in a high-throughput screen. Lead optimization of the screening hit, led to a number of potent HDAC inhibitors. In particular, alpha-mercaptoketone 19y (KD5150) exhibited nanomolar in vitro activity and inhibition of tumor growth in vivo.


Subject(s)
Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors , Ketones/chemistry , Antineoplastic Agents/therapeutic use , Chelating Agents/pharmacology , Chemistry, Pharmaceutical , Drug Design , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Models, Chemical , Neoplasms/drug therapy , Prodrugs/chemistry , Structure-Activity Relationship , Zinc/chemistry
15.
Mol Cancer Ther ; 7(5): 1054-65, 2008 May.
Article in English | MEDLINE | ID: mdl-18483295

ABSTRACT

Histone deacetylase (HDAC) inhibitors have garnered significant attention as cancer drugs. These therapeutic agents have recently been clinically validated with the market approval of vorinostat (SAHA, Zolinza) for treatment of cutaneous T-cell lymphoma. Like vorinostat, most of the small-molecule HDAC inhibitors in clinical development are hydroxamic acids, whose inhibitory activity stems from their ability to coordinate the catalytic Zn2+ in the active site of HDACs. We sought to identify novel, nonhydroxamate-based HDAC inhibitors with potentially distinct pharmaceutical properties via an ultra-high throughput small molecule biochemical screen against the HDAC activity in a HeLa cell nuclear extract. An alpha-mercaptoketone series was identified and chemically optimized. The lead compound, KD5170, exhibits HDAC inhibitory activity with an IC50 of 0.045 micromol/L in the screening biochemical assay and an EC50 of 0.025 micromol/L in HeLa cell-based assays that monitor histone H3 acetylation. KD5170 also exhibits broad spectrum classes I and II HDAC inhibition in assays using purified recombinant human isoforms. KD5170 shows significant antiproliferative activity against a variety of human tumor cell lines, including the NCI-60 panel. Significant tumor growth inhibition was observed after p.o. dosing in human HCT-116 (colorectal cancer), NCI-H460 (non-small cell lung carcinoma), and PC-3 (prostate cancer) s.c. xenografts in nude mice. In addition, a significant increase in antitumor activity and time to end-point occurred when KD5170 was combined with docetaxel in xenografts of the PC-3 prostate cancer cell line. The biological and pharmaceutical profile of KD5170 supports its continued preclinical and clinical development as a broad spectrum anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pyridines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays
16.
J Pharmacol Exp Ther ; 326(1): 240-51, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18430863

ABSTRACT

Glutamate is a major neurotransmitter in the central nervous system, and abnormal glutamate neurotransmission has been implicated in many neurological disorders, including schizophrenia, Alzheimer's disease, Parkinson's disease, addiction, anxiety, depression, epilepsy, and pain. Metabotropic glutamate receptors (mGluRs) activate intracellular signaling cascades in a G protein-dependent manner, which offer the opportunity for developing drugs that regulate glutamate neurotransmission in a functionally selective manner. In the present study, we further characterize the human mGluR2 (hmGluR2) potentiator binding site by showing that the substitution of the three amino acids found to be required for hmGluR2 potentiation, specifically Ser(688), Gly(689), and Asn(735), with the homologous hmGluR3 amino acids, inactivates the positive allosteric modulator activity of several structurally unique mGluR2 potentiators. Based on the characterization of the hmGluR2 potentiator binding site, we developed a novel scintillation proximity assay that was able to discriminate between compounds that were hmGluR2-specific potentiators, and those that were active on both hmGluR2 and hmGluR3. In addition, we substituted Ser(688), Gly(689), and Asn(735) into hmGluR3 and created an active hmGluR2 allosteric modulation site on the hmGluR3 receptor.


Subject(s)
Allosteric Site/genetics , Amino Acids/metabolism , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Allosteric Regulation/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acids/chemistry , Amino Acids/genetics , Animals , Cell Line , Cells, Cultured , Humans , Male , Molecular Sequence Data , Point Mutation , Protein Binding/genetics , Rats , Rats, Sprague-Dawley
17.
Inorg Chem ; 46(24): 10354-64, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17975915

ABSTRACT

The structural and photophysical properties of Ru(II)-polypyridyl complexes with five- and six-membered chelate rings were studied for two bis-tridentate and two tris-bidentate complexes. The photophysical effect of introducing a six-membered chelate ring is most pronounced for the tridentate complex, leading to a room-temperature excited-state lifetime of 810 ns, a substantial increase from 180 ns for the five-membered chelate ring model complex. Contrasting this, the effect is the opposite in tris-bidentate complexes, in which the lifetime decreases from 430 ns to around 1 ns in going from a five-membered to six-membered chelate ring. All of the complexes were studied spectroscopically at both 80 K and ambient temperatures, and the temperature dependence of the excited-state lifetime was investigated for both of the bis-tridentate complexes. The main reason for the long excited-state lifetime in the six-membered chelate ring bis-tridentate complex was found to be a strong retardation of the activated decay via metal-centered states, largely due to an increased ligand field splitting due to the complex having a more-octahedral geometry.

18.
Bioorg Med Chem Lett ; 15(19): 4354-8, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16046122

ABSTRACT

We have identified and synthesized a series of biphenyl-carboxylic acid indanones as allosteric potentiators of the metabotropic glutamate receptor 2. Structure-activity relationship studies directed toward improving the potency and the brain to plasma ratio of the initial lead led to the discovery of 5 and 23 (EC50=111 and 5 nM, respectively).


Subject(s)
Biphenyl Compounds/chemical synthesis , Indans/chemical synthesis , Receptors, Metabotropic Glutamate/agonists , Allosteric Regulation , Animals , Biphenyl Compounds/metabolism , Biphenyl Compounds/pharmacokinetics , Brain Chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Indans/metabolism , Indans/pharmacokinetics , Rats , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/drug therapy , Structure-Activity Relationship , Tissue Distribution
20.
Bioorg Med Chem Lett ; 15(18): 4068-72, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16005222

ABSTRACT

Metabotropic glutamate receptor 2 (mGluR2) has been implicated in a variety of CNS disorders, including schizophrenia. Disclosed herein is the development of a new series of allosteric potentiators of mGluR2. Structure-activity relationship studies in conjunction with pharmacokinetic data led to the discovery of indole 5, which is active in an animal model for schizophrenia.


Subject(s)
Acetophenones/pharmacology , Disease Models, Animal , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Acetophenones/chemistry , Acetophenones/pharmacokinetics , Allosteric Regulation/drug effects , Animals , Brain/drug effects , Brain/metabolism , Cross-Linking Reagents/chemistry , Humans , Ketamine/pharmacology , Molecular Structure , Rats , Schizophrenia/chemically induced , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...