Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
ACS Med Chem Lett ; 13(5): 848-854, 2022 May 12.
Article in English | MEDLINE | ID: mdl-35586426

ABSTRACT

Lysine-specific demethylase 1 (LSD1/KDM1A) is a promising therapeutic target for the treatment of cancers. Several derivatives of tranylcypromine (trans-2-phenylcyclopropylamine) have been developed as LSD1 inhibitors. One such derivative is S2157; however, this compound has a high hERG channel inhibitory activity and a low microsomal stability, making it unsuitable as a drug candidate. Here, using an in silico hERG inhibition prediction model, we designed, synthesized, and evaluated a novel series of S2157 derivatives characterized by modifications of the benzyloxy and piperazine groups. Among the synthesized derivatives, a compound possessing 2-fluoropyridine and 2,8-diaza-spiro[4.5]decane groups (compound 10) showed the most desirable activities, and its eutomer, S1427, was isolated by the optical resolution of 10. In addition to potent LSD1 inhibitory activity, S1427 exhibited desirable hERG channel inhibition and microsomal stability profiles.

2.
J Virol ; 96(5): e0168621, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34985994

ABSTRACT

Hepatitis B virus (HBV) infects 240 million people worldwide. Current therapy profoundly suppresses HBV replication but requires long-term maintenance therapy. Therefore, there is still a medical need for an efficient HBV cure. HBV enters host cells by binding via the preS1 domain of the viral L protein to the Na+/taurocholate cotransporting polypeptide (NTCP). Thus, NTCP should be a key target for the development of anti-HBV therapeutics. Indeed, myrcludex B, a synthetic form of the myristoylated preS1 peptide, effectively reduces HBV/hepatitis D virus (HDV) infection and has been approved as Hepcludex in Europe for the treatment of patients with chronic HDV infection. We established a monoclonal antibody (MAb), N6HB426-20, that recognizes the extracellular domain of human NTCP and blocks HBV entry in vitro into human liver cells but has much less of an inhibitory effect on bile acid uptake. In vivo, administration of the N6HB426-20 MAb prevented HBV viremia for an extended period of time after HBV inoculation in a mouse model system without strongly inhibiting bile acid absorption. Among the extracellular loops (ECLs) of NTCP, regions of amino acids (aa) 84 to 87 in ECL1 and aa 157 to 165 near ECL2 of transmembrane domain 5 are critically important for HBV/HDV infection. Epitope mapping and the three-dimensional (3D) model of the NTCP structure suggested that the N6HB426-20 MAb may recognize aa 276/277 at the tip of ECL4 and interfere with binding of HBV to the region from aa 84 to 87. In summary, we identified an in vivo neutralizing NTCP-targeting antibody capable of preventing HBV infection. Further improvements in efficacy of this drug will pave the way for its clinical applications. IMPORTANCE A number of entry inhibitors are being developed to enhance the treatment of HBV patients with oral nucleoside/nucleotide analogues (NA). To amplify the effectiveness of NA therapy, several efforts have been made to develop therapeutic MAbs with neutralizing activity against HBs antigens. However, the neutralizing effect of these MAbs may be muted by a large excess of HBsAg-positive noninfectious particles in the blood of infected patients. The advantage of NTCP-targeted HBV entry inhibitors is that they remain effective regardless of viral genotype, viral mutations, and the presence of subviral particles. Although N6HB426-20 requires a higher dose than myrcludex to obtain equivalent suppression of HBV in a model mouse system, it maintained the inhibitory effect for a long time postadministration in proportion to the half-life of an IgG MAb. We believe that further improvements will make this antibody a promising treatment option for patients with chronic hepatitis B.


Subject(s)
Hepatitis B virus , Hepatitis B , Organic Anion Transporters, Sodium-Dependent , Symporters , Virus Internalization , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Hep G2 Cells , Hepatitis B/drug therapy , Hepatitis B/prevention & control , Hepatitis B/virology , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatocytes , Humans , Mice , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Viral Proteins/metabolism , Virus Internalization/drug effects
3.
PLoS One ; 16(2): e0243855, 2021.
Article in English | MEDLINE | ID: mdl-33539347

ABSTRACT

Chagas disease is caused by infection with the protozoan parasite Trypanosoma cruzi (T. cruzi). It was originally a Latin American endemic health problem, but now is expanding worldwide as a result of increasing migration. The currently available drugs for Chagas disease, benznidazole and nifurtimox, provoke severe adverse effects, and thus the development of new drugs is urgently required. Ubiquinone (UQ) is essential for respiratory chain and redox balance in trypanosomatid protozoans, therefore we aimed to provide evidence that inhibitors of the UQ biosynthesis have trypanocidal activities. In this study, inhibitors of the human COQ7, a key enzyme of the UQ synthesis, were tested for their trypanocidal activities because they were expected to cross-react and inhibit trypanosomal COQ7 due to their genetic homology. We show the trypanocidal activity of a newly found human COQ7 inhibitor, an oxazinoquinoline derivative. The structurally similar compounds were selected from the commercially available compounds by 2D and 3D ligand-based similarity searches. Among 38 compounds selected, 12 compounds with the oxazinoquinoline structure inhibited significantly the growth of epimastigotes of T. cruzi. The most effective 3 compounds also showed the significant antitrypanosomal activity against the mammalian stage of T. cruzi at lower concentrations than benznidazole, a commonly used drug today. We found that epimastigotes treated with the inhibitor contained reduced levels of UQ9. Further, the growth of epimastigotes treated with the inhibitors was partially rescued by UQ10 supplementation to the culture medium. These results suggest that the antitrypanosomal mechanism of the oxazinoquinoline derivatives results from inhibition of the trypanosomal UQ synthesis leading to a shortage of the UQ pool. Our data indicate that the UQ synthesis pathway of T. cruzi is a promising drug target for Chagas disease.


Subject(s)
Antiprotozoal Agents/pharmacology , Chagas Disease/drug therapy , Chagas Disease/metabolism , Ubiquinone/metabolism , Animals , Cell Line , Cell Line, Tumor , Chagas Disease/parasitology , Drug Delivery Systems/methods , HeLa Cells , Humans , Mammals/metabolism , Nitroimidazoles/pharmacology , Signal Transduction , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects
4.
Biochem Biophys Res Commun ; 537: 85-92, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33387887

ABSTRACT

Transactive response DNA-binding protein of 43 kDa (TDP-43) abnormally forms aggregates in certain subtypes of frontotemporal lobar degeneration (FTLD) and in amyotrophic lateral sclerosis (ALS). The pathological forms of TDP-43 have reported to be associated with poly(ADP-ribose) (PAR), which regulates the properties of these aggregates. A recent study has indicated that tankyrase, a member of the PAR polymerase (PARP) family, regulates pathological TDP-43 formation under conditions of stress, and tankyrase inhibitors suppress TDP-43 aggregate formation and cytotoxicity. Since we reported the development of tankyrase inhibitors that are more specific than conventional inhibitors, in this study, we examined their effects on the formation of TDP-43 aggregates in cultured cells. Time-lapse imaging showed that TDP-43 aggregates appeared in the nucleus within 30 min of treatment with sodium arsenite. Several tankyrase inhibitors suppressed the formation of aggregates and decreased the levels of the tankyrase protein. Immunohistochemical studies demonstrated that tankyrase was localized to neuronal cytoplasmic inclusions in the spinal cords of patients with ALS. Moreover, the tankyrase protein levels were significantly higher in the brains of patients with FTLD than in the brains of control subjects. These findings suggest that the inhibition of tankyrase activity protects against TDP-43 toxicity. Tankyrase inhibitors may be a potential treatment to suppress the progression of TDP-43 proteinopathies.


Subject(s)
DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Protein Aggregates , Tankyrases/antagonists & inhibitors , Arsenites/toxicity , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , HEK293 Cells , Humans , Poly Adenosine Diphosphate Ribose/toxicity , Protein Aggregates/drug effects , TDP-43 Proteinopathies/pathology , Tankyrases/metabolism
5.
Nat Cancer ; 2(3): 340-356, 2021 03.
Article in English | MEDLINE | ID: mdl-35121960

ABSTRACT

Aggressive therapy-resistant and refractory acute myeloid leukemia (AML) has an extremely poor outcome. By analyzing a large number of genetically complex and diverse, primary high-risk poor-outcome human AML samples, we identified specific pathways of therapeutic vulnerability. Through drug screens followed by extensive in vivo validation and genomic analyses, we found inhibition of cytosolic and mitochondrial anti-apoptotic proteins XIAP, BCL2 and MCL1, and a key regulator of mitosis, AURKB, as a vulnerability hub based on patient-specific genetic aberrations and transcriptional signatures. Combinatorial therapeutic inhibition of XIAP with an additional patient-specific vulnerability eliminated established AML in vivo in patient-derived xenografts (PDXs) bearing diverse genetic aberrations, with no signs of recurrence during off-treatment follow-up. By integrating genomic profiling and drug-sensitivity testing, this work provides a platform for a precision-medicine approach for treating aggressive AML with high unmet need.


Subject(s)
Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-bcl-2 , Apoptosis/genetics , Apoptosis Regulatory Proteins/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-bcl-2/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics
6.
J Med Chem ; 63(8): 4183-4204, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32202790

ABSTRACT

Tankyrases (TNKS/TNKS2) belong to the poly(ADP-ribose) polymerase family. Inhibition of their enzymatic activities attenuates the Wnt/ß-catenin signaling, which plays an important role in cancer pathogenesis. We previously reported the discovery of RK-287107, a spiroindoline-based, highly selective, potent tankyrase inhibitor. Herein we describe the optimization process of RK-287107 leading to RK-582, which exhibits a markedly improved robust tumor growth inhibition in a COLO-320DM mouse xenograft model when orally administered. In addition to the dose-dependent elevation and attenuation of the levels of biomarkers AXIN2 and ß-catenin, respectively, results of the TCF reporter and cell proliferation studies on COLO-320DM are discussed.


Subject(s)
Colonic Neoplasms/drug therapy , Drug Design , Drug Discovery/methods , Enzyme Inhibitors/administration & dosage , Tankyrases/antagonists & inhibitors , Administration, Oral , Animals , Cell Line, Tumor , Colonic Neoplasms/enzymology , Enzyme Inhibitors/chemistry , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Protein Structure, Tertiary , Rats , Tankyrases/chemistry , Tankyrases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays/methods
7.
Bioorg Med Chem ; 28(1): 115182, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31753803

ABSTRACT

Given that the associated clinical manifestations of ubiquinone (UQ, or coenzyme Q) deficiency diseases are highly heterogeneous and complicated, effective new research tools for UQ homeostasis studies are awaited. We set out to develop human COQ7 inhibitors that interfere with UQ synthesis. Systematic structure-activity relationship development starting from a screening hit compound led to the identification of highly potent COQ7 inhibitors that did not disturb physiological cell growth of human normal culture cells. These new COQ7 inhibitors may serve as useful tools for studying the balance between UQ supplementation pathways: de novo UQ synthesis and extracellular UQ uptake.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondrial Proteins/antagonists & inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Mitochondrial Proteins/metabolism , Mixed Function Oxygenases/metabolism , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
8.
J Med Chem ; 62(7): 3407-3427, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30883102

ABSTRACT

The canonical WNT pathway plays an important role in cancer pathogenesis. Inhibition of poly(ADP-ribose) polymerase catalytic activity of the tankyrases (TNKS/TNKS2) has been reported to reduce the Wnt/ß-catenin signal by preventing poly ADP-ribosylation-dependent degradation of AXIN, a negative regulator of Wnt/ß-catenin signaling. With the goal of investigating the effects of tankyrase and Wnt pathway inhibition on tumor growth, we set out to find small-molecule inhibitors of TNKS/TNKS2 with suitable drug-like properties. Starting from 1a, a high-throughput screening hit, the spiroindoline derivative 40c (RK-287107) was discovered as a potent TNKS/TNKS2 inhibitor with >7000-fold selectivity against the PARP1 enzyme, which inhibits WNT-responsive TCF reporter activity and proliferation of human colorectal cancer cell line COLO-320DM. RK-287107 also demonstrated dose-dependent tumor growth inhibition in a mouse xenograft model. These observations suggest that RK-287107 is a promising lead compound for the development of novel tankyrase inhibitors as anticancer agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/chemistry , Indoles/pharmacology , Spiro Compounds/pharmacology , Tankyrases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , High-Throughput Screening Assays , Humans , Mice , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Spiro Compounds/chemistry , Xenograft Model Antitumor Assays
9.
Cancer Sci ; 109(12): 4003-4014, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30238564

ABSTRACT

Aberrant activation of Wnt/ß-catenin signaling causes tumorigenesis and promotes the proliferation of colorectal cancer cells. Porcupine inhibitors, which block secretion of Wnt ligands, may have only limited clinical impact for the treatment of colorectal cancer, because most colorectal cancer is caused by loss-of-function mutations of the tumor suppressor adenomatous polyposis coli (APC) downstream of Wnt ligands. Tankyrase poly(ADP-ribosyl)ates (PARylates) Axin, a negative regulator of ß-catenin. This post-translational modification causes ubiquitin-dependent degradation of Axin, resulting in ß-catenin accumulation. Tankyrase inhibitors downregulate ß-catenin and suppress the growth of APC-mutated colorectal cancer cells. Herein, we report a novel tankyrase-specific inhibitor RK-287107, which inhibits tankyrase-1 and -2 four- and eight-fold more potently, respectively, than G007-LK, a tankyrase inhibitor that has been previously reported as effective in mouse xenograft models. RK-287107 causes Axin2 accumulation and downregulates ß-catenin, T-cell factor/lymphoid enhancer factor reporter activity and the target gene expression in colorectal cancer cells harboring the shortly truncated APC mutations. Consistently, RK-287107 inhibits the growth of APC-mutated (ß-catenin-dependent) colorectal cancer COLO-320DM and SW403 cells but not the APC-wild (ß-catenin-independent) colorectal cancer RKO cells. Intraperitoneal or oral administration of RK-287107 suppresses COLO-320DM tumor growth in NOD-SCID mice. Rates of tumor growth inhibition showed good correlation with the behavior of pharmacodynamic biomarkers, such as Axin2 accumulation and MYC downregulation. These observations indicate that RK-287107 exerts a proof-of-concept antitumor effect, and thus may have potential for tankyrase-directed molecular cancer therapy.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/administration & dosage , Tankyrases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HCT116 Cells , Humans , Mice , Mutation , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Sci Transl Med ; 9(413)2017 Oct 25.
Article in English | MEDLINE | ID: mdl-29070697

ABSTRACT

Numerous variant alleles are associated with human acute myeloid leukemia (AML). However, the same variants are also found in individuals with no hematological disease, making their functional relevance obscure. Through NOD.Cg-PrkdcscidIl2rgtmlWjl/Sz (NSG) xenotransplantation, we functionally identified preleukemic and leukemic stem cell populations present in FMS-like tyrosine kinase 3 internal tandem duplication-positive (FLT3-ITD)+ AML patient samples. By single-cell DNA sequencing, we identified clonal structures and linked mutations with in vivo fates, distinguishing mutations permissive of nonmalignant multilineage hematopoiesis from leukemogenic mutations. Although multiple somatic mutations coexisted at the single-cell level, inhibition of the mutation strongly associated with preleukemic to leukemic stem cell transition eliminated AML in vivo. Moreover, concurrent inhibition of BCL-2 (B cell lymphoma 2) uncovered a critical dependence of resistant AML cells on antiapoptotic pathways. Co-inhibition of pathways critical for oncogenesis and survival may be an effective strategy that overcomes genetic diversity in human malignancies. This approach incorporating single-cell genomics with the NSG patient-derived xenograft model may serve as a broadly applicable resource for precision target identification and drug discovery.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Signal Transduction/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Clone Cells , Female , Genomics , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Analysis, DNA , Single-Cell Analysis , Xenograft Model Antitumor Assays
11.
Sci Transl Med ; 5(181): 181ra52, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23596204

ABSTRACT

Leukemia stem cells (LSCs) that survive conventional chemotherapy are thought to contribute to disease relapse, leading to poor long-term outcomes for patients with acute myeloid leukemia (AML). We previously identified a Src-family kinase (SFK) member, hematopoietic cell kinase (HCK), as a molecular target that is highly differentially expressed in human primary LSCs compared with human normal hematopoietic stem cells (HSCs). We performed a large-scale chemical library screen that integrated a high-throughput enzyme inhibition assay, in silico binding prediction, and crystal structure determination and found a candidate HCK inhibitor, RK-20449, a pyrrolo-pyrimidine derivative with an enzymatic IC50 (half maximal inhibitory concentration) in the subnanomolar range. A crystal structure revealed that RK-20449 bound the activation pocket of HCK. In vivo administration of RK-20449 to nonobese diabetic (NOD)/severe combined immunodeficient (SCID)/IL2rg(null) mice engrafted with highly aggressive therapy-resistant AML significantly reduced human LSC and non-stem AML burden. By eliminating chemotherapy-resistant LSCs, RK-20449 may help to prevent relapse and lead to improved patient outcomes in AML.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Adult , Aged , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Female , Hematopoiesis/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Middle Aged , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Proto-Oncogene Proteins c-hck/chemistry , Proto-Oncogene Proteins c-hck/metabolism , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , RNA, Small Interfering/metabolism , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured , Young Adult
12.
Bioorg Med Chem ; 17(19): 6971-82, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19720539

ABSTRACT

A series of trans-3-oxospiro[(aza)isobenzofuran-1(3H),1'-cyclohexane]-4'-carboxamide derivatives were synthesized to identify potent NPY Y5 receptor antagonists. Of the compounds, 21j showed high Y5 binding affinity, metabolic stability and brain and cerebrospinal fluid (CSF) penetration, and low susceptibility to P-glycoprotein transporters. Oral administration of 21j significantly inhibited the Y5 agonist-induced food intake in rats with a minimum effective dose of 1mg/kg. This compound was selected for proof-of-concept studies in human clinical trials.


Subject(s)
Amides/chemical synthesis , Benzofurans/chemical synthesis , Receptors, Neuropeptide Y/antagonists & inhibitors , Spiro Compounds/chemical synthesis , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Amides/pharmacology , Animals , Benzofurans/pharmacology , Brain/metabolism , Cerebrospinal Fluid/metabolism , Drug Stability , Eating/drug effects , Rats , Spiro Compounds/pharmacology
13.
Bioorg Med Chem Lett ; 19(18): 5436-9, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19679469

ABSTRACT

A series of trans-3-oxospiro[(aza)isobenzofuran-1(3H),1'-cyclohexane]-4'-carboxamide derivatives were synthesized and profiled for NPY Y5 binding affinity, brain and CSF penetrability in rats, and susceptibility to human and mouse P-glycoprotein transporters in order to develop a PET ligand. Compound 12b exhibited an acceptable profile for a PET ligand, and [(11)C]12b was successfully utilized in clinical settings as a Y5 PET ligand.


Subject(s)
Brain/diagnostic imaging , Positron-Emission Tomography/methods , Radioligand Assay/methods , Receptors, Neuropeptide Y/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Brain/metabolism , Cell Line , Cerebrospinal Fluid/diagnostic imaging , Humans , Ligands , Mice , Plasma/diagnostic imaging , Protein Binding , Rats , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 19(18): 5339-45, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19683441

ABSTRACT

Optimization of the lead 2a led to the identification of a novel diarylketoxime class of melanin-concentrating hormone 1 receptor (MCH-1R) antagonists. Our focus was directed toward improvement of hERG activity and metabolic stability. The representative derivative 4b showed potent and dose-dependent body weight reduction in diet-induced obese (DIO) C57BL/6J mice after oral administration. The synthesis and structure-activity relationships of the novel diarylketoxime MCH-1R antagonists are described.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Ether-A-Go-Go Potassium Channels/metabolism , Obesity/drug therapy , Oximes/chemistry , Oximes/therapeutic use , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Body Weight/drug effects , Humans , Mice , Mice, Inbred C57BL , Oximes/pharmacokinetics , Oximes/pharmacology , Protein Binding , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
15.
Bioorg Med Chem ; 17(16): 6106-22, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19616955

ABSTRACT

A series of 2-pyridone-containing imidazoline derivatives was synthesized and evaluated as neuropeptide Y Y5 receptor antagonists. Optimization of the 2-pyridone structure on the 2-position of the imidazoline ring led to identification of 1-(difluoromethyl)-5-[(4S,5S)-4-(4-fluorophenyl)-4-(6-fluoropyridin-3-yl)-5-methyl-4,5-dihydro-1H-imidazol-2-yl]pyridin-2(1H)-one (7m). Compound 7m displayed statistically significant inhibition of food intake in an agonist-induced food intake model in SD rats and no adverse cardiovascular effects in anesthetized dogs. In addition, markedly higher brain penetrability and a lower plasma Occ90 value were observed in P-gp-deficient mdr1a (-/-) mice compared to mdr1a (+/+) mice after oral administration of 7m.


Subject(s)
Anti-Obesity Agents/chemistry , Imidazolines/chemistry , Pyridones/chemistry , Receptors, Neuropeptide Y/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Dogs , Drug Discovery , Humans , Imidazolines/chemical synthesis , Imidazolines/pharmacokinetics , Mice , Pyridones/chemical synthesis , Pyridones/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/metabolism , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 19(16): 4589-93, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19615899

ABSTRACT

A series of imidazo[1,2-a]pyridine derivatives was identified and evaluated for MCH1R binding and antagonistic activity. Introduction of a methyl substituent at the 3-position of imidazo[1,2-a]pyridine provided compounds with a significant improvement in MCH1R affinity. Representative compounds in this series exhibited good potency and brain exposure in rats.


Subject(s)
Anti-Obesity Agents/chemistry , Pyridines/chemistry , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacology , CHO Cells , Cricetinae , Cricetulus , Drug Discovery , Humans , Pyridines/chemical synthesis , Pyridines/pharmacology , Rats , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
17.
Eur J Pharmacol ; 615(1-3): 113-7, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19482021

ABSTRACT

Neuropeptide Y plays a key role in the physiological control of energy homeostasis. Five neuropeptide Y receptor subtypes have been cloned, and multiple neuropeptide Y receptor subtypes are thought to mediate neuropeptide Y activity. However, interactions among neuropeptide Y receptor subtypes have not been elucidated to date. Herein, we examined the interaction between neuropeptide Y(1) and Y(5) receptors in feeding regulation by employing selective neuropeptide Y(1) and Y(5) receptor antagonists in C57BL/6 and neuropeptide Y(1) receptor knockout mice fed a high-fat diet. A single-dose of a neuropeptide Y(1) receptor antagonist (10-30 mg/kg) suppressed spontaneous food intake and reduced body weight in high-fat diet-fed C57BL/6 mice, while treatment with a neuropeptide Y(5) receptor antagonist did not significantly reduce food intake or body weight. Coadministration of a neuropeptide Y(1) receptor antagonist with a neuropeptide Y(5) receptor antagonist further suppressed food intake and reduced body weight. Next, we evaluated the chronic efficacy of a neuropeptide Y(5) receptor antagonist in high-fat diet-fed neuropeptide Y(1) receptor knockout mice in order to mimic chronic combination treatment with neuropeptide Y(1) and Y(5) receptor antagonists. The neuropeptide Y(5) receptor antagonist produced greater body weight reductions in high-fat diet-fed neuropeptide Y(1) receptor knockout mice than in wild-type C57BL/6 mice. These findings confirm an interaction between neuropeptide Y(1) and Y(5) receptors in the regulation of energy homeostasis, as blockade of both the neuropeptide Y(1) and Y(5) receptors produced a greater anti-obesity effect than blocking either receptor alone.


Subject(s)
Anti-Obesity Agents/pharmacology , Eating/drug effects , Energy Metabolism/physiology , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Body Weight/drug effects , Dietary Fats/administration & dosage , Drug Synergism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Obesity/drug therapy , Obesity/metabolism , Pyridines/pharmacology , Receptors, Neuropeptide Y/genetics , Spiro Compounds/pharmacology , Thiazoles/pharmacology
18.
Bioorg Med Chem ; 17(14): 5015-26, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19525116

ABSTRACT

Spiroindoline urea derivatives, designed to act as NPY Y5 receptor antagonists, were synthesized and their structure-activity relationships were investigated. Of these derivatives, compound 3a showed good Y5 binding affinity with favorable pharmacokinetic properties. Compound 3a significantly inhibited bPP Y5 agonist-induced food intake in rats, and suppressed body weight gain in DIO mice.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Biological Availability , Body Weight/drug effects , Eating/drug effects , Indoles/chemical synthesis , Indoles/pharmacokinetics , Mice , Mice, Inbred C57BL , Molecular Structure , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/agonists , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 19(11): 3072-7, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19403308

ABSTRACT

Optimization of high-throughput screening hit 1a led to the identification of a novel spiro-piperidine class of melanin-concentrating hormone 1 receptor (MCH-1R) antagonists. Compound 3c was identified as a highly potent and selective MCH-1R antagonist, which has an IC(50) value of 0.09 nM at hMCH-1R. The synthesis and structure-activity relationships of the novel spiro-piperidine MCH-1R antagonists are described.


Subject(s)
Piperidines/chemistry , Receptors, Somatostatin/antagonists & inhibitors , Spiro Compounds/chemistry , Cell Line , Drug Discovery , Humans , Piperidines/chemical synthesis , Piperidines/pharmacology , Receptors, Somatostatin/metabolism , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacology , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 19(13): 3568-72, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19457661

ABSTRACT

A series of 2-aminobenzimidazole-based MCH1R antagonists was identified by core replacement of the aminoquinoline lead 1. Subsequent modification of the 2- and 5-positions led to improvement in potency and intrinsic clearance. Compound 25 exhibited good plasma and brain exposure, and attenuated MCH induced food intake at 30mg/kg PO in rats.


Subject(s)
Anti-Obesity Agents/chemistry , Benzimidazoles/chemistry , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacokinetics , Eating/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...