Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur J Drug Metab Pharmacokinet ; 49(1): 7-21, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37921950

ABSTRACT

BACKGROUND AND OBJECTIVE: Cenobamate is an antiseizure medication (ASM) approved for treatment of focal epilepsy in adults. The objective of this study was to characterize the distribution, metabolism, and excretion of cenobamate in adult and pre- and postnatal rats, including pregnant and lactating females and nursing pups. METHODS: Distribution, metabolic, and excretion profiles were determined for 14C-labeled and unlabeled cenobamate using liquid scintillation counting, radiochromatography, LCMS, and LCMS/MS after oral or intravenous (IV) administration. RESULTS: Distribution of 14C-cenobamate-related material in adult male rats was widespread throughout the body, with nearly 1:1 tissue-to-plasma ratios observed for most tissues, including brain. Cenobamate administered to pregnant females was also transferred across the placental barrier into amniotic fluid and fetal plasma. Following administration to lactating F0 females, cenobamate was detected in breast milk and in plasma of nursing pups. 14C-cenobamate administered to adult male rats as a single oral dose was extensively metabolized with nine metabolites identified in urine and feces, including a principal dihydrodiol metabolite. Cenobamate was the principal drug-related material in rat plasma. Following a single dose of 14C-cenobamate to male and female rats, radioactivity was excreted equally into urine and feces, with mass balance achieved by 48 h postdose. CONCLUSIONS: Distribution of cenobamate was widespread into many rat tissues, including brain, amniotic fluid, fetal plasma, breast milk, and breastfeeding rat pups. These distribution findings, along with the results of the metabolism and excretion studies, may help inform treatment decisions for patients with epilepsy being treated with cenobamate, including pregnant or nursing mothers.


Subject(s)
Carbamates , Chlorophenols , Lactation , Placenta , Tetrazoles , Humans , Rats , Male , Animals , Pregnancy , Female , Placenta/metabolism , Milk/metabolism , Feces/chemistry
2.
Heliyon ; 9(8): e18920, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37636350

ABSTRACT

Background: Cenobamate is an antiseizure medication used to treat partial-onset (focal) seizures. It is a molecule with one chiral center and a unique dual mechanism of action: enhancement of fast and slow inactivation of sodium channels with preferential inhibition of the persistent current and positive allosteric modulation of GABAA receptor-mediated ion channels. Aims/Methods: Anticonvulsant effects of cenobamate (YKP3089; R-enantiomer), YKP3090 (S-enantiomer), and YKP1983 (racemate) were evaluated in chemically and electrically induced focal and generalized seizure models in rodents. The Genetic Absence Epilepsy Rat from Strasbourg (GAERS) model examined the effect of cenobamate on spike-wave seizures. Motor coordination was assessed with rotarod tests and minimal motor impairment exams. Results: Early in development, cenobamate was found to have activity in focal and generalized seizure models in animals and was selected for continued development. Cenobamate prevented seizures in a dose-dependent manner, prevented seizure spread, and increased seizure threshold without potentiating seizure initiation or the development of tolerance to its anticonvulsant effects. In contrast, YKP3090 and YKP1983 were only effective against generalized tonic-clonic seizures. Cenobamate also protected mice from 6 Hz psychomotor-induced seizures. Cenobamate showed significant dose-dependent reductions in the number and cumulative duration of spike-and-wave discharges in the GAERS model. Discussion: Cenobamate showed efficacy or efficacy signals in all animal models of epilepsy tested with a favorable risk-versus-benefit ratio, supporting its clinical use in the treatment of partial-onset (focal) seizures in adults and warranting further clinical research in generalized seizures and absence seizures.

3.
Clin Transl Sci ; 15(4): 899-911, 2022 04.
Article in English | MEDLINE | ID: mdl-34877801

ABSTRACT

This study was designed to evaluate the effects of cenobamate, an antiseizure medication for focal seizures, on the pharmacokinetics of cytochrome P450 probes (bupropion, CYP2B6; midazolam, CYP3A4/5; warfarin, CYP2C9; and omeprazole, CYP2C19) in healthy subjects. Probes were administered alone on days 1 (bupropion) and 7 (midazolam/warfarin/omeprazole), and with cenobamate 100 mg/day on day 69 (midazolam) and cenobamate 200 mg/day on days 99 (bupropion) and 105 (midazolam/warfarin/omeprazole). No significant interaction was concluded if 90% confidence intervals (CIs) for geometric mean ratios (GMRs) for area under the curve (AUC) and maximum concentration of CYP substrates and/or their metabolites were within the no-effect interval (0.80-1.25). When co-administered with cenobamate 100 mg/day, AUC from time of administration up to the time of the last quantifiable concentration (AUC0-last ) GMR (90% CIs) for midazolam was 0.734 (0.647-0.832). When co-administered with cenobamate 200 mg/day, AUC0-last GMRs (90% CI) for midazolam, bupropion, S-warfarin, and omeprazole were 0.277 (0.238-0.323), 0.615 (0.522-0.724), 1.14 (1.10-1.18), and 2.07 (1.44-2.98), respectively. Co-administration of cenobamate with midazolam and bupropion probes led to values that were outside and below the no effect boundary, indicating that cenobamate induces the CYP3A4/5 and CYP2B6 enzymes. Co-administration of cenobamate led to omeprazole values which were outside and above the no-effect boundary, but with high variability, suggesting that cenobamate may moderately inhibit CYP2C19 activity. No effect on CYP2C9 was observed with the cenobamate and warfarin combination. Co-administration of cenobamate with these probes drugs was well-tolerated. In this study, 200 mg/day cenobamate moderately induced CYP3A4/5 (dose-dependently; 100 mg/day was a weak inducer), was a weak inducer of CYP2B6, moderately inhibited CYP2C19, and had a negligible effect on CYP2C9.


Subject(s)
Cytochrome P-450 CYP3A , Midazolam , Bupropion/pharmacokinetics , Carbamates , Chlorophenols , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Healthy Volunteers , Humans , Midazolam/pharmacokinetics , Omeprazole/pharmacokinetics , Pharmaceutical Preparations , Tetrazoles , Warfarin/pharmacokinetics
4.
Eur J Pharmacol ; 879: 173117, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32325146

ABSTRACT

Cenobamate is a novel antiepileptic drug under investigation for use in patients with focal (partial-onset) seizures. To understand its potential molecular mechanism of action, the effects of cenobamate on GABAA-mediated currents and GABAA receptors in rodent hippocampal neurons were examined. Cenobamate potentiated GABA-induced currents (IGABA) in acutely isolated CA3 pyramidal cells in a concentration-dependent manner (EC50, 164 µM), which was not affected by flumazenil, a benzodiazepine receptor antagonist. Cenobamate enhanced tonic GABAA currents (Itonic), which is defined as a holding current shift by the GABAA receptor antagonist bicuculline (EC50, 36.63 µM). At therapeutically relevant concentrations, cenobamate induced minimal changes in the frequency, amplitudes, and decay time of spontaneous inhibitory postsynaptic currents in the CA1 neurons. Flumazenil failed to affect cenobamate-potentiated Itonic and Iphasic in CA1 neurons. Cenobamate showed positive allosteric modulation of GABA-induced IGABA mediated by GABAA receptors. This effect was similar for all tested hGABAA receptors containing six different alpha subunits (α1ß2γ2 or α2-6ß3γ2), with EC50 values ranging from 42 to 194 µM. Cenobamate did not displace the binding of flunitrazepam, a benzodiazepine derivative, or flumazenil to GABAA receptors. The results showed that cenobamate, a novel antiepileptic drug, acts as a positive allosteric modulator of high-affinity GABAA receptors, activated by GABA at a site independent of the benzodiazepine binding site and efficiently enhances Itonic inhibition in hippocampal neurons, which could be an underlying molecular mechanism stabilizing neural circuits of the epileptic hippocampus.


Subject(s)
Anticonvulsants/pharmacology , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Carbamates/pharmacology , Chlorophenols/pharmacology , Neurons/drug effects , Receptors, GABA-A/physiology , Tetrazoles/pharmacology , Animals , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Female , HEK293 Cells , Humans , Male , Neurons/physiology , Rats, Sprague-Dawley , Rats, Wistar
5.
Eur J Drug Metab Pharmacokinet ; 45(4): 513-522, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32301064

ABSTRACT

BACKGROUND AND OBJECTIVE: Cenobamate is an antiepileptic drug for the treatment of partial-onset seizures. The current study was designed to assess the mass balance and the metabolic profiling of cenobamate in humans. METHODS: Absorption, metabolism, and excretion of cenobamate were investigated in healthy male subjects after a single oral dose of 400 mg of cenobamate containing 50 µCi of [14C]-cenobamate as capsule formulation. RESULTS: Cenobamate was rapidly (median time to maximum plasma concentration of 1.25 h) and extensively (≥ 88% of dose) absorbed. The mean cenobamate plasma concentration-time profile revealed a multiphasic elimination profile whereas the mean plasma/blood concentration-time curve for total radioactivity did not appear to be multiphasic, suggesting that elimination mechanisms for cenobamate and its metabolites may be different. Blood/plasma ratios observed for the area under the concentration-time curve (AUC) and peak concentration (both ~ 0.60) suggest a limited penetration of cenobamate and metabolites into red blood cells (RBCs). Eight cenobamate metabolites were identified across plasma, urine, and feces. Cenobamate was the main plasma radioactive component and M1 was the only metabolite detected in plasma (> 98% and < 2% total radioactivity AUC, respectively). All detected metabolites were found in urine, with M1 as the major radioactive component (mean cumulative recovery 37.7% of dose); unchanged cenobamate accounted for 6%. Metabolites comprised ~ 88% of the dose recovered in urine, indicating extensive metabolism by the kidneys and/or metabolites formed in the liver were rapidly eliminated from the bloodstream. However, cenobamate metabolites appear to be formed slowly. Minor amounts of cenobamate (0.48%) and five metabolites (≤ 1.75% each; M1, M3, M6, M7, M11) were recovered in feces. CONCLUSION: This study indicates that cenobamate is primarily eliminated in urine as metabolites. Cenobamate is the major circulating component in plasma after oral administration and has a limited penetration into RBCs.


Subject(s)
Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Chlorophenols/administration & dosage , Chlorophenols/pharmacokinetics , Renal Elimination , Tetrazoles/administration & dosage , Tetrazoles/pharmacokinetics , Administration, Oral , Adult , Anticonvulsants/blood , Biotransformation , Carbamates/blood , Chlorophenols/blood , Gastrointestinal Absorption , Healthy Volunteers , Humans , Intestinal Elimination , Male , Metabolomics , Middle Aged , New Jersey , Tetrazoles/blood , Young Adult
6.
Br J Clin Pharmacol ; 82(4): 1022-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27277189

ABSTRACT

AIMS: This study aimed to evaluate the impact of esomeprazole on the pharmacokinetics of sonidegib. METHODS: This Phase I study evaluated the impact of the proton pump inhibitor (PPI) esomeprazole on the oral absorption and pharmacokinetics (PKs) of a single dose of sonidegib under fasted conditions. A total of 42 healthy subjects were enrolled to receive either sonidegib alone (200 mg single dose) or sonidegib in combination with esomeprazole (40 mg pre-treatment 5 days and combination were given on day 6). Primary PK parameters assessed in the study were area under the concentration-time curve (AUC) from 0-14 days and 0-7 days and maximum observed plasma concentration (Cmax ). RESULTS: The plasma exposure (AUC0-14d, AUC0-7d and Cmax ) of a single 200 mg oral dose of sonidegib was decreased by 32-38% when sonidegib was co-administered with esomeprazole compared with sonidegib alone, with no apparent change in elimination slope and tmax . Baseline gastric pH was similar between the two arms. CONCLUSIONS: These results suggest a modest reduction in the extent of sonidegib absorption by esomeprazole. There was no obvious metabolic drug-drug interaction between the two agents. Both sonidegib and esomeprazole were well tolerated in the study population.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Esomeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Pyridines/pharmacokinetics , Adolescent , Adult , Biphenyl Compounds/adverse effects , Biphenyl Compounds/blood , Drug Interactions , Esomeprazole/adverse effects , Female , Healthy Volunteers , Humans , Male , Middle Aged , Proton Pump Inhibitors/adverse effects , Pyridines/adverse effects , Pyridines/blood , Young Adult
7.
Xenobiotica ; 46(8): 667-76, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26586447

ABSTRACT

1. Idasanutlin (RG7388) is a potent p53-MDM2 antagonist currently in clinical development for treatment of cancer. The purpose of the present studies was to investigate the cause of marked decrease in plasma exposure after repeated oral administration of RG7388 in monkeys and whether the autoinduction observed in monkeys is relevant to humans. 2. In monkey liver and intestinal microsomes collected after repeated oral administration of RG7388 to monkeys, significantly increased activities of homologue CYP3A8 were observed (ex vivo). Investigation using a physiologically based pharmacokinetic (PBPK) model suggested that the loss of exposure was primarily due to induction of metabolism in the gut of monkeys. 3. Studies in monkey and human primary hepatocytes showed that CYP3A induction by RG7388 only occurred in monkey hepatocytes but not in human hepatocytes, which suggests the observed CYP3A induction is monkey specific. 4. The human PK data obtained from the first cohorts confirmed the lack of relevant induction as predicted by the human hepatocytes and the PBPK modelling based on no induction in humans.


Subject(s)
Antineoplastic Agents/pharmacology , Macaca fascicularis/physiology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrrolidines/pharmacology , para-Aminobenzoates/pharmacology , Animals , Antineoplastic Agents/metabolism , Humans , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrrolidines/metabolism , para-Aminobenzoates/metabolism
8.
PLoS One ; 10(11): e0142845, 2015.
Article in English | MEDLINE | ID: mdl-26571496

ABSTRACT

PURPOSE: The paradigm shift in cancer treatment from cytotoxic drugs to tumor targeted therapies poses new challenges, including optimization of dose and schedule based on a biologically effective dose, rather than the historical maximum tolerated dose. Optimal dosing is currently determined using concentrations of tyrosine kinase inhibitors in plasma as a surrogate for tumor concentrations. To examine this plasma-tumor relationship, we explored the association between lapatinib levels in tumor and plasma in mice and humans, and those effects on phosphorylation of human epidermal growth factor receptors (HER) in human tumors. EXPERIMENTAL DESIGN: Mice bearing BT474 HER2+ human breast cancer xenografts were dosed once or twice daily (BID) with lapatinib. Drug concentrations were measured in blood, tumor, liver, and kidney. In a randomized phase I clinical trial, 28 treatment-naïve female patients with early stage HER2+ breast cancer received lapatinib 1000 or 1500 mg once daily (QD) or 500 mg BID before evaluating steady-state lapatinib levels in plasma and tumor. RESULTS: In mice, lapatinib levels were 4-fold higher in tumor than blood with a 4-fold longer half-life. Tumor concentrations exceeded the in vitro IC90 (~ 900 nM or 500 ng/mL) for inhibition of HER2 phosphorylation throughout the 12-hour dosing interval. In patients, tumor levels were 6- and 10-fold higher with QD and BID dosing, respectively, compared to plasma trough levels. The relationship between tumor and plasma concentration was complex, indicating multiple determinants. HER receptor phosphorylation varied depending upon lapatinib tumor concentrations, suggestive of changes in the repertoire of HER homo- and heterodimers. CONCLUSION: Plasma lapatinib concentrations underestimated tumor drug levels, suggesting that optimal dosing should be focused on the site of action to avoid to inappropriate dose escalation. Larger clinical trials are required to determine optimal dose and schedule to achieve tumor concentrations that maximally inhibit HER receptors. CLINICAL TRIAL REGISTRATION: NCT00359190.


Subject(s)
Antineoplastic Agents/blood , ErbB Receptors/metabolism , Quinazolines/blood , Administration, Oral , Adult , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Area Under Curve , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Administration Schedule , Drug Dosage Calculations , ErbB Receptors/antagonists & inhibitors , Female , Half-Life , Humans , Immunohistochemistry , Lapatinib , Mice , Mice, SCID , Phosphorylation/drug effects , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , ROC Curve , Tandem Mass Spectrometry , Transplantation, Heterologous
9.
Bioorg Med Chem ; 22(15): 4001-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24997575

ABSTRACT

The field of small-molecule inhibitors of protein-protein interactions is rapidly advancing and the specific area of inhibitors of the p53/MDM2 interaction is a prime example. Several groups have published on this topic and multiple compounds are in various stages of clinical development. Building on the strength of the discovery of RG7112, a Nutlin imidazoline-based compound, and RG7388, a pyrrolidine-based compound, we have developed additional scaffolds that provide opportunities for future development. Here, we report the discovery and optimization of a highly potent and selective series of spiroindolinone small-molecule MDM2 inhibitors, culminating in RO8994.


Subject(s)
Indoles/chemistry , Indolizidines/chemistry , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Spiro Compounds/chemistry , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Imidazolines/chemistry , Indoles/therapeutic use , Indoles/toxicity , Indolizidines/therapeutic use , Indolizidines/toxicity , Molecular Dynamics Simulation , Neoplasms/drug therapy , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrrolidines/chemistry , Spiro Compounds/therapeutic use , Spiro Compounds/toxicity , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , para-Aminobenzoates/chemistry
10.
ACS Med Chem Lett ; 5(2): 124-7, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24900784

ABSTRACT

The development of small-molecule MDM2 inhibitors to restore dysfunctional p53 activities represents a novel approach for cancer treatment. In a previous communication, the efforts leading to the identification of a non-imidazoline MDM2 inhibitor, RG7388, was disclosed and revealed the desirable in vitro and in vivo pharmacological properties that this class of pyrrolidine-based inhibitors possesses. Given this richness and the critical need for a wide variety of chemical structures to ensure success in the clinic, research was expanded to evaluate additional derivatives. Here we report two new potent, selective, and orally active p53-MDM2 antagonists, RO5353 and RO2468, as follow-ups with promising potential for clinical development.

11.
Clin Cancer Res ; 20(14): 3742-52, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24812409

ABSTRACT

PURPOSE: Antitumor clinical activity has been demonstrated for the MDM2 antagonist RG7112, but patient tolerability for the necessary daily dosing was poor. Here, utilizing RG7388, a second-generation nutlin with superior selectivity and potency, we determine the feasibility of intermittent dosing to guide the selection of initial phase I scheduling regimens. EXPERIMENTAL DESIGN: A pharmacokinetic-pharmacodynamic (PKPD) model was developed on the basis of preclinical data to determine alternative dosing schedule requirements for optimal RG7388-induced antitumor activity. This PKPD model was used to investigate the pharmacokinetics of RG7388 linked to the time-course of the antitumor effect in an osteosarcoma xenograft model in mice. These data were used to prospectively predict intermittent and continuous dosing regimens, resulting in tumor stasis in the same model system. RESULTS: RG7388-induced apoptosis was delayed relative to drug exposure with continuous treatment not required. In initial efficacy testing, daily dosing at 30 mg/kg and twice a week dosing at 50 mg/kg of RG7388 were statistically equivalent in our tumor model. In addition, weekly dosing of 50 mg/kg was equivalent to 10 mg/kg given daily. The implementation of modeling and simulation on these data suggested several possible intermittent clinical dosing schedules. Further preclinical analyses confirmed these schedules as viable options. CONCLUSION: Besides chronic administration, antitumor activity can be achieved with intermittent schedules of RG7388, as predicted through modeling and simulation. These alternative regimens may potentially ameliorate tolerability issues seen with chronic administration of RG7112, while providing clinical benefit. Thus, both weekly (qw) and daily for five days (5 d on/23 off, qd) schedules were selected for RG7388 clinical testing.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bone Neoplasms/drug therapy , Imidazolines/pharmacokinetics , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Drug Administration Schedule , Female , Humans , Imidazolines/therapeutic use , Mice, Nude , Osteosarcoma/drug therapy , Pyrrolidines/pharmacology , Xenograft Model Antitumor Assays , para-Aminobenzoates/pharmacology
12.
Exp Hematol ; 42(2): 137-45.e5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24309210

ABSTRACT

The tumor suppressor p53 is thought to play a role in megakaryocyte (MK) development. To assess the influence of the p53 regulatory pathway further, we studied the effect of RG7112, a small molecule MDM2 antagonist that activates p53 by preventing its interaction with MDM2, on normal megakaryocytopoiesis and platelet production. This drug has been previously been evaluated in clinical trials of cancer patients where thrombocytopenia was one of the major dose-limiting toxicities. In this study, we demonstrated that administration of RG7112 in vivo in rats and monkeys results in thrombocytopenia. In addition, we identified two distinct mechanisms by which RG7112-mediated activation of p53 affected human megakaryocytopoiesis and platelet production in vitro. RG7112 promoted apoptosis of MK progenitor cells, resulting in a reduction of their numbers and RG7112 affected mature MK by blocking DNA synthesis during endomitosis and impairing platelet production. Together, the disruption of these events provides an explanation for RG7112-induced thrombocytopenia and insight into the role of the p53-MDM2 auto-regulatory loop in normal megakaryocytopoiesis.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazolines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Thrombopoiesis/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Female , Humans , Male , Mice , Platelet Count
13.
Bioorg Med Chem Lett ; 23(24): 6610-5, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24239188

ABSTRACT

DYRK1B is a kinase over-expressed in certain cancer cells (including colon, ovarian, pancreatic, etc.). Recent publications have demonstrated inhibition of DYRK1B could be an attractive target for cancer therapy. From a data-mining effort, the team has discovered analogues of pyrido[2,3-d]pyrimidines as potent enantio-selective inhibitors of DYRK1B. Cells treated with a tool compound from this series showed the same cellular effects as down regulation of DYRK1B with siRNA. Such effects are consistent with the proposed mechanism of action. Progress of the SAR study is presented.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Half-Life , Humans , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacokinetics , Rats , Structure-Activity Relationship , Dyrk Kinases
14.
J Med Chem ; 56(14): 5979-83, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23808545

ABSTRACT

Restoration of p53 activity by inhibition of the p53-MDM2 interaction has been considered an attractive approach for cancer treatment. However, the hydrophobic protein-protein interaction surface represents a significant challenge for the development of small-molecule inhibitors with desirable pharmacological profiles. RG7112 was the first small-molecule p53-MDM2 inhibitor in clinical development. Here, we report the discovery and characterization of a second generation clinical MDM2 inhibitor, RG7388, with superior potency and selectivity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrrolidines/chemical synthesis , Tumor Suppressor Protein p53/antagonists & inhibitors , para-Aminobenzoates/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Humans , Male , Mice , Mice, Inbred C57BL , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , para-Aminobenzoates/pharmacology , para-Aminobenzoates/therapeutic use
15.
ACS Med Chem Lett ; 4(5): 466-9, 2013 May 09.
Article in English | MEDLINE | ID: mdl-24900694

ABSTRACT

The p53 tumor suppressor is a potent transcription factor that plays a key role in the regulation of cellular responses to stress. It is controlled by its negative regulator MDM2, which binds directly to p53 and inhibits its transcriptional activity. MDM2 also targets p53 for degradation by the proteasome. Many tumors produce high levels of MDM2, thereby impairing p53 function. Restoration of p53 activity by inhibiting the p53-MDM2 interaction may represent a novel approach to cancer treatment. RG7112 (2g) is the first clinical small-molecule MDM2 inhibitor designed to occupy the p53-binding pocket of MDM2. In cancer cells expressing wild-type p53, RG7112 stabilizes p53 and activates the p53 pathway, leading to cell cycle arrest, apoptosis, and inhibition or regression of human tumor xenografts.

16.
Cancer Res ; 69(19): 7672-80, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19773430

ABSTRACT

Notch signaling is an area of great interest in oncology. RO4929097 is a potent and selective inhibitor of gamma-secretase, producing inhibitory activity of Notch signaling in tumor cells. The RO4929097 IC50 in cell-free and cellular assays is in the low nanomolar range with >100-fold selectivity with respect to 75 other proteins of various types (receptors, ion channels, and enzymes). RO4929097 inhibits Notch processing in tumor cells as measured by the reduction of intracellular Notch expression by Western blot. This leads to reduced expression of the Notch transcriptional target gene Hes1. RO4929097 does not block tumor cell proliferation or induce apoptosis but instead produces a less transformed, flattened, slower-growing phenotype. RO4929097 is active following oral dosing. Antitumor activity was shown in 7 of 8 xenografts tested on an intermittent or daily schedule in the absence of body weight loss or Notch-related toxicities. Importantly, efficacy is maintained after dosing is terminated. Angiogenesis reverse transcription-PCR array data show reduced expression of several key angiogenic genes. In addition, comparative microarray analysis suggests tumor cell differentiation as an additional mode of action. These preclinical results support evaluation of RO4929097 in clinical studies using an intermittent dosing schedule. A multicenter phase I dose escalation study in oncology is under way.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Benzazepines/pharmacology , Neoplasms/drug therapy , Protease Inhibitors/pharmacology , Receptors, Notch/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Hair Follicle/drug effects , Hair Follicle/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Neoplasms/enzymology , Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
17.
J Rural Health ; 19 Suppl: 314-20, 2003.
Article in English | MEDLINE | ID: mdl-14526514

ABSTRACT

CONTEXT: Many state, federal, and foundation resources have been invested in improving the recruitment of primary care providers to rural communities. The Southern Rural Access Program of the Robert Wood Johnson Foundation (RWJF) has provided varying levels of support to several southern states to assist with retention of those providers. PURPOSE: This study describes the strategies that 6 states used to develop and implement practice management technical assistance services for rural health care providers. METHODS: Practice managers in each of the 6 states were surveyed regarding how their service was structured, what types of entities were eligible, and the nature of the technical assistance offered. Information regarding what types of entities used the service, characteristics of the practices, and the number of practices served was also collected. FINDINGS: The survey results showed that almost half (46%) of all practices assisted were private stand-alone physician practices, with overall practice assessments being the practice management service rendered most often. Although the type of organisational home for the technical assistance services varied by state, overall states employed an average of 1.67 full-time equivalent practice managers (0.81 full-time equivalent supported by RWJF) and received an average of $136,055 per state from the RWJF for the 2-year period beginning April 2002 for practice management support. CONCLUSIONS: Overall, the study found that the type of organizational home did not appear to affect the type of technical assistance services offered. However, the type of orgnizational home did appear to affect what types of providers used the service, with trade associations assisting their members or constituents at least half the time.


Subject(s)
Community Health Planning/organization & administration , Medically Underserved Area , Practice Management, Medical/organization & administration , Program Development , Rural Health Services/organization & administration , Financing, Organized , Foundations , Health Planning Technical Assistance , Health Services Research , Humans , Organizational Case Studies , Primary Health Care , Southeastern United States , State Government
SELECTION OF CITATIONS
SEARCH DETAIL
...