Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 15(4): 424-431, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38628790

ABSTRACT

As we celebrate International Women's Day 2024 with the theme "Inspire Inclusion", the women of the ACS Medicinal Chemistry Division (MEDI) want to foster a sense of belonging, relevance, and empowerment by sharing uplifting stories of what inspired them to become medicinal chemists. In this editorial, we are featuring female medicinal chemistry scientists to provide role models, encouragement, and inspiration to others. We asked women medicinal chemists to contribute a brief paragraph about what inspired them to become medicinal chemists or what inspires them today as medicinal chemists. The responses and contributions highlight their passions and motivations, such as their love of the sciences and their drive to improve human health by contributing to basic research and creating lifesaving drugs.

2.
J Med Chem ; 67(6): 4251-4258, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38456628

ABSTRACT

As we celebrate International Women's Day 2024 with the theme "Inspire Inclusion", the women of the ACS Medicinal Chemistry Division (MEDI) want to foster a sense of belonging, relevance, and empowerment by sharing uplifting stories of what inspired them to become medicinal chemists. In this editorial, we are featuring female medicinal chemistry scientists to provide role models, encouragement, and inspiration to others. We asked women medicinal chemists to contribute a brief paragraph about what inspired them to become medicinal chemists or what inspires them today as medicinal chemists. The responses and contributions highlight their passions and motivations, such as their love of the sciences and their drive to improve human health by contributing to basic research and creating lifesaving drugs.


Subject(s)
Chemistry, Pharmaceutical , Power, Psychological , Humans , Female
3.
EMBO J ; 43(5): 666-694, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38279026

ABSTRACT

The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.


Subject(s)
Anaphase-Promoting Complex-Cyclosome , Neoplasms , Humans , Anaphase-Promoting Complex-Cyclosome/genetics , Dyneins , Kinesins/genetics , Kinetochores , Mitosis , Neoplasms/genetics
4.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38151625

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Subject(s)
Kinesins , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Kinesins/genetics , Kinesins/metabolism , Mitosis/genetics , Cell Line , M Phase Cell Cycle Checkpoints
5.
J Med Chem ; 65(6): 4972-4990, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35286090

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer that results from errors in chromosome segregation during mitosis. Targeting of CIN-associated vulnerabilities is an emerging therapeutic strategy in drug development. KIF18A, a mitotic kinesin, has been shown to play a role in maintaining bipolar spindle integrity and promotes viability of CIN cancer cells. To explore the potential of KIF18A, a series of inhibitors was identified. Optimization of an initial hit led to the discovery of analogues that could be used as chemical probes to interrogate the role of KIF18A inhibition. Compounds 23 and 24 caused significant mitotic arrest in vivo, which was sustained for 24 h. This would be followed by cell death either in mitosis or in the subsequent interphase. Furthermore, photoaffinity labeling experiments reveal that this series of inhibitors binds at the interface of KIF18A and tubulin. This study represents the first disclosure of KIF18A inhibitors with in vivo activity.


Subject(s)
Kinesins , Neoplasms , Cell Death , Humans , Mitosis , Neoplasms/drug therapy , Neoplasms/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
6.
J Med Chem ; 63(1): 52-65, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31820981

ABSTRACT

KRASG12C has emerged as a promising target in the treatment of solid tumors. Covalent inhibitors targeting the mutant cysteine-12 residue have been shown to disrupt signaling by this long-"undruggable" target; however clinically viable inhibitors have yet to be identified. Here, we report efforts to exploit a cryptic pocket (H95/Y96/Q99) we identified in KRASG12C to identify inhibitors suitable for clinical development. Structure-based design efforts leading to the identification of a novel quinazolinone scaffold are described, along with optimization efforts that overcame a configurational stability issue arising from restricted rotation about an axially chiral biaryl bond. Biopharmaceutical optimization of the resulting leads culminated in the identification of AMG 510, a highly potent, selective, and well-tolerated KRASG12C inhibitor currently in phase I clinical trials (NCT03600883).


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Piperazines/therapeutic use , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Pyrimidinones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Dogs , Drug Discovery , Humans , Isomerism , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Mice, Nude , Mutation , Piperazines/chemistry , Piperazines/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/pharmacokinetics , Rats , Structure-Activity Relationship
7.
J Med Chem ; 62(3): 1523-1540, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30624936

ABSTRACT

Pim kinases are a family of constitutively active serine/threonine kinases that are partially redundant and regulate multiple pathways important for cell growth and survival. In human disease, high expression of the three Pim isoforms has been implicated in the progression of hematopoietic and solid tumor cancers, which suggests that Pim kinase inhibitors could provide patients with therapeutic benefit. Herein, we describe the structure-guided optimization of a series of quinazolinone-pyrrolodihydropyrrolone analogs leading to the identification of potent pan-Pim inhibitor 28 with improved potency, solubility, and drug-like properties. Compound 28 demonstrated on-target Pim activity in an in vivo pharmacodynamic assay with significant inhibition of BAD phosphorylation in KMS-12-BM multiple myeloma tumors for 16 h postdose. In a 2-week mouse xenograft model, daily dosing of compound 28 resulted in 33% tumor regression at 100 mg/kg.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyrroles/therapeutic use , Quinazolinones/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Female , Humans , Mice, SCID , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/pharmacokinetics , Quinazolinones/chemical synthesis , Quinazolinones/pharmacokinetics , Structure-Activity Relationship , Swine , Xenograft Model Antitumor Assays
8.
J Med Chem ; 59(13): 6407-30, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27285051

ABSTRACT

The high expression of proviral insertion site of Moloney murine leukemia virus kinases (Pim-1, -2, and -3) in cancers, particularly the hematopoietic malignancies, is believed to play a role in promoting cell survival and proliferation while suppressing apoptosis. The three isoforms of Pim protein appear largely redundant in their oncogenic functions. Thus, a pan-Pim kinase inhibitor is highly desirable. However, cell active pan-Pim inhibitors have proven difficult to develop because Pim-2 has a low Km for ATP and therefore requires a very potent inhibitor to effectively block the kinase activity at cellular ATP concentrations. Herein, we report a series of quinazolinone-pyrrolopyrrolones as potent and selective pan-Pim inhibitors. In particular, compound 17 is orally efficacious in a mouse xenograft model (KMS-12 BM) of multiple myeloma, with 93% tumor growth inhibition at 50 mg/kg QD upon oral dosing.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyrroles/pharmacology , Quinazolinones/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Pyrroles/administration & dosage , Pyrroles/chemistry , Quinazolinones/administration & dosage , Quinazolinones/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem Lett ; 25(19): 4136-42, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26298499

ABSTRACT

Based on lead compound 1, which was discovered from a high-throughput screen, a series of PI3Kα/mTOR inhibitors were evaluated that contained an imidazo[1,2-a]pyridine as a core replacement for the benzimidazole contained in 1. By exploring various ring systems that occupy the affinity pocket, two fragments containing a methoxypyridine were identified that gave <100 nM potency toward PI3Kα in enzyme and cellular assays with moderate stability in rat and human liver microsomes. With the two methoxypyridine groups selected to occupy the affinity pocket, analogs were prepared with various fragments intended to occupy the ribose pocket of PI3Kα and mTOR. From these analogs, tertiary alcohol 18 was chosen for in vivo pharmacodynamic evaluation based on its potency in the PI3Kα cellular assay, microsomal stability, and in vivo pharmacokinetic properties. In a mouse liver pharmacodynamic assay, compound 18 showed 56% inhibition of HFG-induced AKT (Ser473) phosphorylation at a 30 mg/kg dose.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemical synthesis , Rats , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
10.
J Med Chem ; 58(11): 4462-82, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25914941

ABSTRACT

The glucokinase-glucokinase regulatory protein (GK-GKRP) complex plays an important role in controlling glucose homeostasis in the liver. We have recently disclosed a series of arylpiperazines as in vitro and in vivo disruptors of the GK-GKRP complex with efficacy in rodent models of type 2 diabetes mellitus (T2DM). Herein, we describe a new class of aryl sulfones as disruptors of the GK-GKRP complex, where the central piperazine scaffold has been replaced by an aromatic group. Conformational analysis and exploration of the structure-activity relationships of this new class of compounds led to the identification of potent GK-GKRP disruptors. Further optimization of this novel series delivered thiazole sulfone 93, which was able to disrupt the GK-GKRP interaction in vitro and in vivo and, by doing so, increases cytoplasmic levels of unbound GK.


Subject(s)
Aminopyridines/pharmacology , Carrier Proteins/antagonists & inhibitors , Glucokinase/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Liver/drug effects , Small Molecule Libraries/pharmacology , Sulfones/chemistry , Aminopyridines/chemistry , Animals , Carrier Proteins/metabolism , Crystallography, X-Ray , Glucokinase/metabolism , Glucose/metabolism , Hypoglycemic Agents/chemistry , Liver/cytology , Liver/metabolism , Models, Molecular , Molecular Conformation , Molecular Structure , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Sulfones/pharmacology
11.
J Med Chem ; 57(7): 2989-3004, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24597733

ABSTRACT

Transient receptor potential melastatin 8 (TRPM8) is a nonselective cation channel expressed in a subpopulation of sensory neurons in the peripheral nervous system. TRPM8 is the predominant mammalian cold temperature thermosensor and is activated by cold temperatures ranging from 8 to 25 °C and cooling compounds such as menthol or icilin. TRPM8 antagonists are being pursued as potential therapeutics for treatment of pain and bladder disorders. This manuscript outlines new developments in the SAR of a lead series of 1,2,3,4-tetrahydroisoquinoline derivatives with emphasis on strategies to improve pharmacokinetic properties and potency. Selected compounds were profiled in two TRPM8 target-specific in vivo coverage models in rats (the icilin-induced wet dog shake model and the cold pressor test). Compound 45 demonstrated robust efficacy in both pharmacodynamic models with ED90 values <3 mg/kg.


Subject(s)
Behavior, Animal/drug effects , Microsomes, Liver/drug effects , Motor Activity/drug effects , TRPM Cation Channels/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacokinetics , Animals , Circular Dichroism , Cold Temperature , Dogs , Humans , Male , Microsomes, Liver/metabolism , Pyrimidinones/pharmacology , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stereoisomerism , TRPM Cation Channels/metabolism , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacology , Tissue Distribution
12.
J Med Chem ; 57(2): 325-38, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24405213

ABSTRACT

In the previous report , we described the discovery and optimization of novel small molecule disruptors of the GK-GKRP interaction culminating in the identification of 1 (AMG-1694). Although this analogue possessed excellent in vitro potency and was a useful tool compound in initial proof-of-concept experiments, high metabolic turnover limited its advancement. Guided by a combination of metabolite identification and structure-based design, we have successfully discovered a potent and metabolically stable GK-GKRP disruptor (27, AMG-3969). When administered to db/db mice, this compound demonstrated a robust pharmacodynamic response (GK translocation) as well as statistically significant dose-dependent reductions in fed blood glucose levels.


Subject(s)
Carrier Proteins/metabolism , Glucokinase/metabolism , Hypoglycemic Agents/chemistry , Piperazines/chemical synthesis , Sulfonamides/chemical synthesis , Alkynes/chemical synthesis , Alkynes/pharmacokinetics , Alkynes/pharmacology , Animals , Blood Glucose/metabolism , Carrier Proteins/chemistry , Glucokinase/chemistry , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Mice , Microsomes, Liver/metabolism , Models, Molecular , Morpholines/chemical synthesis , Morpholines/pharmacokinetics , Morpholines/pharmacology , Piperazines/pharmacokinetics , Piperazines/pharmacology , Protein Binding , Protein Transport , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
13.
J Med Chem ; 55(17): 7796-816, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22897589

ABSTRACT

The phosphoinositide 3-kinase family catalyzes the phosphorylation of phosphatidylinositol-4,5-diphosphate to phosphatidylinositol-3,4,5-triphosphate, a secondary messenger which plays a critical role in important cellular functions such as metabolism, cell growth, and cell survival. Our efforts to identify potent, efficacious, and orally available phosphatidylinositol 3-kinase (PI3K) inhibitors as potential cancer therapeutics have resulted in the discovery of 4-(2-((6-methoxypyridin-3-yl)amino)-5-((4-(methylsulfonyl)piperazin-1-yl)methyl)pyridin-3-yl)-6-methyl-1,3,5-triazin-2-amine (1). In this paper, we describe the optimization of compound 1, which led to the design and synthesis of pyridyltriazine 31, a potent pan inhibitor of class I PI3Ks with a superior pharmacokinetic profile. Compound 31 was shown to potently block the targeted PI3K pathway in a mouse liver pharmacodynamic model and inhibit tumor growth in a U87 malignant glioma glioblastoma xenograft model. On the basis of its excellent in vivo efficacy and pharmacokinetic profile, compound 31 was selected for further evaluation as a clinical candidate and was designated AMG 511.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Triazines/pharmacology , Crystallography, X-Ray , Models, Molecular , Protein Kinase Inhibitors/chemistry
14.
J Med Chem ; 55(11): 5188-219, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22548365

ABSTRACT

A highly selective series of inhibitors of the class I phosphatidylinositol 3-kinases (PI3Ks) has been designed and synthesized. Starting from the dual PI3K/mTOR inhibitor 5, a structure-based approach was used to improve potency and selectivity, resulting in the identification of 54 as a potent inhibitor of the class I PI3Ks with excellent selectivity over mTOR, related phosphatidylinositol kinases, and a broad panel of protein kinases. Compound 54 demonstrated a robust PD-PK relationship inhibiting the PI3K/Akt pathway in vivo in a mouse model, and it potently inhibited tumor growth in a U-87 MG xenograft model with an activated PI3K/Akt pathway.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Piperazines/chemical synthesis , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , Triazines/chemical synthesis , Animals , Biological Availability , Class I Phosphatidylinositol 3-Kinases/physiology , Crystallography, X-Ray , Drug Design , Female , Humans , Indazoles/chemical synthesis , Indazoles/pharmacokinetics , Indazoles/pharmacology , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Piperazines/pharmacokinetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Purines/chemical synthesis , Purines/pharmacokinetics , Purines/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Signal Transduction , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Sulfones/chemical synthesis , Sulfones/pharmacokinetics , Sulfones/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazines/pharmacokinetics , Triazines/pharmacology , Xenograft Model Antitumor Assays
15.
J Med Chem ; 55(4): 1593-611, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22329507

ABSTRACT

The transient receptor potential melastatin type 8 (TRPM8) is a nonselective cation channel primarily expressed in a subpopulation of sensory neurons that can be activated by a wide range of stimuli, including menthol, icilin, and cold temperatures (<25 °C). Antagonism of TRPM8 is currently under investigation as a new approach for the treatment of pain. As a result of our screening efforts, we identified tetrahydrothienopyridine 4 as an inhibitor of icilin-induced calcium influx in CHO cells expressing recombinant rat TRPM8. Exploration of the structure-activity relationships of 4 led to the identification of a potent and orally bioavailable TRPM8 antagonist, tetrahydroisoquinoline 87. Compound 87 demonstrated target coverage in vivo after oral administration in a rat pharmacodynamic model measuring the prevention of icilin-induced wet-dog shakes (WDS).


Subject(s)
Analgesics/chemical synthesis , Piperidines/chemical synthesis , TRPM Cation Channels/antagonists & inhibitors , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Piperidines/chemistry , Piperidines/pharmacology , Pyrimidinones/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
16.
J Neurosci ; 32(6): 2086-99, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323721

ABSTRACT

We studied N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride (M8-B), a selective and potent antagonist of the transient receptor potential melastatin-8 (TRPM8) channel. In vitro, M8-B blocked cold-induced and TRPM8-agonist-induced activation of rat, human, and murine TRPM8 channels, including those on primary sensory neurons. In vivo, M8-B decreased deep body temperature (T(b)) in Trpm8(+/+) mice and rats, but not in Trpm8(-/-) mice, thus suggesting an on-target action. Intravenous administration of M8-B was more effective in decreasing T(b) in rats than intrathecal or intracerebroventricular administration, indicating a peripheral action. M8-B attenuated cold-induced c-Fos expression in the lateral parabrachial nucleus, thus indicating a site of action within the cutaneous cooling neural pathway to thermoeffectors, presumably on sensory neurons. A low intravenous dose of M8-B did not affect T(b) at either a constantly high or a constantly low ambient temperature (T(a)), but the same dose readily decreased T(b) if rats were kept at a high T(a) during the M8-B infusion and transferred to a low T(a) immediately thereafter. These data suggest that both a successful delivery of M8-B to the skin (high cutaneous perfusion) and the activation of cutaneous TRPM8 channels (by cold) are required for the hypothermic action of M8-B. At tail-skin temperatures <23°C, the magnitude of the M8-B-induced decrease in T(b) was inversely related to skin temperature, thus suggesting that M8-B blocks thermal (cold) activation of TRPM8. M8-B affected all thermoeffectors studied (thermopreferendum, tail-skin vasoconstriction, and brown fat thermogenesis), thus suggesting that TRPM8 is a universal cold receptor in the thermoregulation system.


Subject(s)
Body Temperature/physiology , Cold Temperature , Ganglia, Spinal/physiology , Shivering/physiology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/deficiency , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Behavior, Animal/drug effects , Body Temperature/drug effects , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Ganglia, Spinal/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pharmaceutical Preparations/administration & dosage , Rats , Rats, Wistar , Shivering/drug effects , Thiophenes/pharmacology
17.
J Med Chem ; 54(14): 5174-84, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21714526

ABSTRACT

N-(6-(6-Chloro-5-(4-fluorophenylsulfonamido)pyridin-3-yl)benzo[d]thiazol-2-yl)acetamide (1) is a potent and efficacious inhibitor of PI3Kα and mTOR in vitro and in vivo. However, in hepatocyte and in vivo metabolism studies, 1 was found to undergo deacetylation on the 2-amino substituent of the benzothiazole. As an approach to reduce or eliminate this metabolic deacetylation, a variety of 6,5-heterocyclic analogues were examined as an alternative to the benzothiazole ring. Imidazopyridazine 10 was found to have similar in vitro potency and in vivo efficacy relative to 1, while only minimal amounts of the corresponding deacetylated metabolite of 10 were observed in hepatocytes.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Female , Hepatocytes/metabolism , Imidazoles/chemical synthesis , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Oxazoles/chemical synthesis , Oxazoles/chemistry , Oxazoles/pharmacology , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacology , Transplantation, Heterologous
18.
J Med Chem ; 54(13): 4735-51, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21612232

ABSTRACT

The phosphoinositide 3-kinase (PI3K) family catalyzes the ATP-dependent phosphorylation of the 3'-hydroxyl group of phosphatidylinositols and plays an important role in cell growth and survival. There is abundant evidence demonstrating that PI3K signaling is dysregulated in many human cancers, suggesting that therapeutics targeting the PI3K pathway may have utility for the treatment of cancer. Our efforts to identify potent, efficacious, and orally available PI3K/mammalian target of rapamycin (mTOR) dual inhibitors resulted in the discovery of a series of substituted quinolines and quinoxalines derivatives. In this report, we describe the structure-activity relationships, selectivity, and pharmacokinetic data of this series and illustrate the in vivo pharmacodynamic and efficacy data for a representative compound.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Quinolines/chemical synthesis , Quinoxalines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Biological Availability , Crystallography, X-Ray , Humans , In Vitro Techniques , Liver/blood supply , Liver/metabolism , Male , Mice , Models, Molecular , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Quinolines/pharmacokinetics , Quinolines/pharmacology , Quinoxalines/pharmacokinetics , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , TOR Serine-Threonine Kinases/chemistry , Xenograft Model Antitumor Assays
19.
Bioorg Med Chem Lett ; 18(18): 5118-22, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18722118

ABSTRACT

Clinical candidate AMG 517 (1) is a potent antagonist toward multiple modes of activation of TRPV1; however, it suffers from poor solubility. Analogs with various substituents at the R region of 3 were prepared to improve the solubility while maintaining the potent TRPV1 activity of 1. Compounds were identified that maintained potency, had good pharmacokinetic properties, and improved solubility relative to 1.


Subject(s)
Benzothiazoles/pharmacology , Pyrimidines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Animals , Benzothiazoles/pharmacokinetics , Combinatorial Chemistry Techniques , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
20.
J Pharmacol Exp Ther ; 326(1): 218-29, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18420600

ABSTRACT

Antagonists of the vanilloid receptor TRPV1 (transient receptor potential vanilloid type 1) have been reported to produce antihyperalgesic effects in animal models of pain. These antagonists, however, also caused concomitant hyperthermia in rodents, dogs, monkeys, and humans. Antagonist-induced hyperthermia was not observed in TRPV1 knockout mice, suggesting that the hyperthermic effect is exclusively mediated through TRPV1. Since antagonist-induced hyperthermia is considered a hurdle for developing TRPV1 antagonists as therapeutics, we investigated the possibility of eliminating hyperthermia while maintaining antihyperalgesia. Here, we report four potent and selective TRPV1 modulators with unique in vitro pharmacology profiles (profiles A through D) and their respective effects on body temperature. We found that profile C modulator, (R,E)-N-(2-hydroxy-2,3-dihydro-1H-inden-4-yl)-3-(2-(piperidin-1-yl)-4-(trifluoromethyl)phenyl)acrylamide (AMG8562), blocks capsaicin activation of TRPV1, does not affect heat activation of TRPV1, potentiates pH 5 activation of TRPV1 in vitro, and does not cause hyperthermia in vivo in rats. We further profiled AMG8562 in an on-target (agonist) challenge model, rodent pain models, and tested for its side effects. We show that AMG8562 significantly blocks capsaicin-induced flinching behavior, produces statistically significant efficacy in complete Freund's adjuvant- and skin incision-induced thermal hyperalgesia, and acetic acid-induced writhing models, with no profound effects on locomotor activity. Based on the data shown here, we conclude that it is feasible to modulate TRPV1 in a manner that does not cause hyperthermia while maintaining efficacy in rodent pain models.


Subject(s)
Acrylamides/chemistry , Acrylamides/pharmacology , Analgesics/pharmacology , Fever , Hyperalgesia/drug therapy , Piperidines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/physiology , Acrylamides/pharmacokinetics , Animals , Body Temperature/drug effects , Body Temperature/physiology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fever/chemically induced , Fever/physiopathology , Hyperalgesia/physiopathology , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Piperidines/chemistry , Piperidines/pharmacokinetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...