Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Pharmacol Exp Ther ; 362(1): 108-118, 2017 07.
Article in English | MEDLINE | ID: mdl-28465372

ABSTRACT

LY2584702 is an inhibitor of p70 S6 kinase-1 previously developed for the treatment of cancer. In two phase 1 trials in oncology patients, significant reductions of total cholesterol, low-density lipoprotein cholesterol (LDL-C), and triglyceride were observed. In the current study, we sought to understand the potential mechanism of action of this compound in regulating lipid metabolism. In Long Evans diet-induced obese (DIO) rats, oral administration of LY2584702 for 3-4 weeks led to robust reduction of LDL-C up to 60%. An unexpected finding of liver triglyceride (TG) increase implicated a metabolite of LY2584702, 4-aminopyrazolo[3,4-day]pyrimidine (4-APP), in modulation of lipid metabolism in these rats. We showed that low-dose 4-APP, when administered orally for 3-4 weeks to Long Evans DIO rats, produced lipoprotein profile changes that were strikingly similar to LY2584702. Kinetic studies suggested that both LY2584702 and 4-APP had no effect on chylomicron-TG secretion and only exerted a modest effect on hepatic very low-density lipoprotein (VLDL)-TG secretion. In human hepatoma HepG2 cells, 4-APP, but not LY2584702, increased LDL uptake. We hypothesize that generation of the 4-APP metabolite may contribute to the efficacy of LY2584702 in lowering LDL-C in rats and potentially in humans as well. This mechanism of LDL-C lowering may include inhibition of VLDL production and increase in LDL clearance.


Subject(s)
Adenine/analogs & derivatives , Hypolipidemic Agents/pharmacology , Obesity/blood , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/pharmacology , Animals , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Cholesterol, VLDL/biosynthesis , Cholesterol, VLDL/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipoproteins, LDL/metabolism , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Long-Evans , Triglycerides/metabolism
2.
J Clin Pharmacol ; 64(1): 94-102, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37566903

ABSTRACT

Lasmiditan is an in vitro inhibitor of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) efflux transporters. We aimed to confirm predictions from physiologically based pharmacokinetic models of lasmiditan, and assess the safety and tolerability of rosuvastatin and dabigatran co-administered with lasmiditan. In this open-label, post-marketing drug-drug interaction, phase 1 clinical trial, eligible participants were adults aged 21-70 years with a body mass index of 18.5-35.0 kg/m2 . Part 1 (P-gp, 150 mg dabigatran etexilate with 200 mg lasmiditan) and part 2 (BCRP, 10 mg rosuvastatin with 200 mg lasmiditan) employed similar designs: a single dose of probe substrate administered on day -2 with pharmacokinetic evaluation; 1-week washout; lasmiditan administered on days 8 and 9 alone; lasmiditan co-administered with a single dose of probe substrate on day 10, with pharmacokinetic evaluation of probe substrate and lasmiditan. Sixty-six participants were included in part 1 and 30 participants were included in part 2. Following dabigatran co-administration with lasmiditan, versus dabigatran alone, 90% confidence intervals for geometric least-squares (LS) mean ratios of area under the plasma concentration-time curve from time 0 extrapolated to infinity (AUC0-∞ ) and maximum observed drug concentration (Cmax ) were not contained within the non-effect boundaries (0.80 to 1.25). Dabigatran AUC0-∞ increased by 25% and Cmax increased by 22%. The median time of maximum observed drug concentration (tmax ) for dabigatran was 2.0 to 3.0 hours. Following rosuvastatin co-administration with lasmiditan, versus rosuvastatin alone, 90%CIs for geometric LS mean ratios of AUC0-∞ and Cmax were contained within non-effect boundaries (0.80-1.25). Rosuvastatin AUC0-∞ increased by 15% and Cmax increased by 7%. The median tmax for rosuvastatin was 4.0 hours. Results suggest that lasmiditan has a weak effect on P-gp substrates and no clinically relevant effect on BCRP substrates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Breast Neoplasms , Adult , Female , Humans , Area Under Curve , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Cross-Over Studies , Dabigatran , Drug Interactions , Neoplasm Proteins , Rosuvastatin Calcium/pharmacokinetics , Young Adult , Middle Aged , Aged
3.
Eur J Nucl Med Mol Imaging ; 40(2): 245-53, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135321

ABSTRACT

PURPOSE: Two allosteric modulators of the group I metabotropic glutamate receptors (mGluR1 and mGluR5) were evaluated as positron emission tomography (PET) radioligands for mGluR1. METHODS: LY2428703, a full mGluR1 antagonist (IC(50) 8.9 nM) and partial mGluR5 antagonist (IC(50) 118 nM), and LSN2606428, a full mGluR1 and mGluR5 antagonist (IC(50) 35.3 nM and 10.2 nM, respectively) were successfully labeled with (11)C and evaluated as radioligands for mGluR1. The pharmacology of LY2428703 was comprehensively assessed in vitro and in vivo, and its biodistribution was investigated by liquid chromatography-mass spectrometry/mass spectrometry, and by PET imaging in the rat. In contrast, LSN2606428 was only evaluated in vitro; further evaluation was stopped due to its unfavorable pharmacological properties and binding affinity. RESULTS: (11)C-LY2428703 showed promising characteristics, including: (1) high potency for binding to human mGluR1 (IC(50) 8.9 nM) with no significant affinity for other human mGlu receptors (mGluR2 through mGluR8); (2) binding to brain displaceable by administration of an mGluR1 antagonist; (3) only one major radiometabolite in both plasma and brain, with a negligible brain concentration (with 3.5 % of the total radioactivity in cerebellum) and no receptor affinity; (4) a large specific and displaceable signal in the mGluR1-rich cerebellum with no significant in vivo affinity for mGluR5, as shown by PET studies in rats; and (5) lack of substrate behavior for efflux transporters at the blood-brain barrier, as shown by PET studies conducted in wild-type and knockout mice. CONCLUSION: (11)C-LY2428703, a new PET radioligand for mGluR1 quantification, displayed promising characteristics both in vitro and in vivo in rodents.


Subject(s)
Brain/pathology , Carbon Isotopes/pharmacology , Positron-Emission Tomography/methods , Receptors, Metabotropic Glutamate/metabolism , Allosteric Site , Animals , Blood-Brain Barrier , Chromatography, Liquid/methods , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Ligands , Male , Mice , Mice, Knockout , Models, Chemical , Rats , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods
4.
Eur J Clin Pharmacol ; 69(12): 2011-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23955175

ABSTRACT

PURPOSE: To assess the impact of hepatic or renal impairment on the pharmacokinetics (PK) of edivoxetine. METHODS: Two separate multi-center, open-label studies with males and females were conducted. Subjects were categorized according to their hepatic function, determined by the Child-Pugh classification, or renal function, determined by creatinine clearance using the Cockcroft-Gault equation. Subjects received a single dose of 18 mg in the hepatic impairment study or 6 mg in the renal impairment study. Noncompartmental PK parameters were computed from the edivoxetine plasma concentration-time data. RESULTS: In the hepatic study, the geometric least squares mean (GLSM) and 90 % confidence interval (CI) of the ratio [impaired : normal] of area under the concentration versus time curve from time zero to infinity (AUC0-∞; h × ng/mL) was 1.24 (0.93, 1.64) in the mild, 1.60 (1.21, 2.12) in the moderate, and 1.70 (1.28, 2.24) in the severe group. In the renal impairment study, the GLSM (90 % CI) of the ratio [impaired : normal] of AUC0-∞ was 1.13 (0.73, 1.73) in mild, 1.90 (1.28, 2.82) in moderate, 1.55 (0.94, 2.55) in severe, and 1.03 (0.66, 1.59) in ESRD groups. Overall, the GLSM of the ratio [impaired : normal] of Cmax was slightly less than or approximately 1 across the hepatic and renal impairment groups. Across both studies, there were no clinically significant changes in vital signs and laboratory values, the adverse events were mild in severity and mostly related to nervous system and gastrointestinal disorder-related events. CONCLUSIONS: PK changes in subjects with hepatic or renal impairment were of small magnitude and did not appear to impact overall subject tolerability. Daily dosing of edivoxetine in a larger population of impaired subjects, including those with dual impairment, would aid in establishing edivoxetine tolerability and PK in a clinical practice scenario.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Liver Diseases/blood , Morpholines/pharmacokinetics , Phenylethyl Alcohol/analogs & derivatives , Renal Insufficiency/blood , Adrenergic Uptake Inhibitors/blood , Adult , Aged , Female , Humans , Male , Middle Aged , Morpholines/blood , Phenylethyl Alcohol/blood , Phenylethyl Alcohol/pharmacokinetics
5.
Birth Defects Res ; 115(7): 782-796, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36916488

ABSTRACT

BACKGROUND: Olaratumab (Lartruvo™) is a recombinant human IgG1 monoclonal antibody that specifically binds PDGFRα. In order to support use of Lartruvo in pediatric patients, a definitive juvenile animal study in neonatal mice was conducted with a human anti-mouse PDGFRα antibody analog of olaratumab (LSN3338786). METHODS: A pilot study was used to set doses for the definitive juvenile mouse study. In the definitive study, juvenile mice were administered vehicle, 50, 100, or 150 mg/kg LSN3338786 by subcutaneous (SC) injection every 3 days between postnatal days (PND) 1 and 49, for a total of 17 doses. Blood samples were collected on PND 49 for antibody analysis and toxicokinetic evaluation. Tissues were collected on PND 52 for histopathologic examination. RESULTS: Results of the pilot study indicated that dosing neonatal mice starting on PND 1 via SC administration every 3 days was logistically feasible, produced exposures consistent with prior animal studies, and the selected dose levels were well tolerated by juvenile mice. In the definitive juvenile study, there were no LSN3338786-related deaths, clinical findings, and no effects on mean body weights, body weight gains, or food consumption. Additionally, there were no adverse LSN3338786-related hematology findings, and no macroscopic, organ weight, or microscopic findings of note. The highest dose evaluated, 150 mg/kg, was considered the NOAEL for juvenile toxicity. CONCLUSIONS: In conclusion, the juvenile animal studies did not identify any new toxicities or increased sensitivities for the intended pediatric patient population. The use of the surrogate antibody approach in a standard rodent model enabled the de-risking of theoretical concerns for toxicity in pediatric patients due to disruption of the PDGFRα pathway during early human development, such as pulmonary development.


Subject(s)
Antibodies, Monoclonal , Receptor, Platelet-Derived Growth Factor alpha , Animals , Mice , Humans , Child , Pilot Projects , Antibodies, Monoclonal/adverse effects , No-Observed-Adverse-Effect Level
6.
Chem Res Toxicol ; 22(2): 243-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19138063

ABSTRACT

The FDA issued a guidance on the safety testing of metabolites in February 2008, in which they stated that metabolites of concern are those that are detected at levels greater than 10% of the systemic exposure of the parent at steady state. This has presented many challenges in determining the circulating human metabolites at an early stage of development. The intention of this perspective is to address the question of how effective in vitro metabolism and early exploratory clinical data are in predicting the circulating metabolites from both a qualitative and a quantitative perspective. To this end, data were reviewed from 17 molecules in the Lilly portfolio for which there were in vitro data and a radiolabeled study in humans. Twelve example cases are presented in detail to demonstrate trends for when in vitro data adequately predicted in vivo (41%), when in vitro data underpredicted the circulating metabolites (35%), and when in vitro data overpredicted the circulating metabolites (24%). In addition, cases that present special challenges due to very low levels of the circulating parent or long half-lives of the parent and/or metabolites are presented. The trends indicate that the more complex the metabolism, the less likely the in vitro data were to predict the circulating metabolites. The in vitro data were also less predictive for N-glucuronidations and non-P450-mediated cleavage reactions. Although the in vitro data were better at predicting clearance pathways, the data set often failed to predict the quantity of metabolites, which is needed in consideration of whether or not a "disproportionate" metabolite may be circulating in human plasma.


Subject(s)
Drug Evaluation, Preclinical/methods , Pharmaceutical Preparations/metabolism , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Guidelines as Topic , Humans , Mass Spectrometry , Metabolic Networks and Pathways , Models, Animal , Pharmaceutical Preparations/blood , Predictive Value of Tests
7.
Chem Res Toxicol ; 22(2): 348-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19170655

ABSTRACT

The role of metabolism in genotoxicity and carcinogenicity of many chemicals is well established. Accordingly, both in vitro metabolic activation systems and in vivo assays are routinely utilized for genotoxic hazard identification of drug candidates prior to clinical investigations. This should, in most cases provide a high degree of confidence that the genotoxic potential of the parent and associated metabolites have been characterized. However, it is well known that significant differences can exist between human metabolism and that which occurs with in vitro and in vivo genotoxicity tests. This poses challenges when considering the adequacy of hazard identification and cancer risk assessment if a human metabolite of genotoxic concern is identified during the course of drug development. Since such challenges are particularly problematic when recognized in the later stages of drug development, a framework for conducting a carcinogenic risk assessment for human genotoxic metabolites is desirable. Here, we propose a risk assessment method that is dependent upon the availability of quantitative human and rodent ADME (absorption, distribution, metabolism, excretion) data, such that exposures to a metabolite of genotoxic concern can be estimated at the intended human efficacious dose and the maximum dose used in the 2-year rodent bioassay(s). The exposures are then applied to the risk assessment framework, based on known cancer potencies, that allows one to understand the probability of a known or suspect genotoxic metabolite posing a carcinogenic risk in excess of 1 in 100,000. Practical case examples are presented to illustrate the application of the risk assessment method within the context of drug development and to highlight its utility and limitations.


Subject(s)
Mutagenicity Tests/methods , Mutagens/metabolism , Mutagens/toxicity , Animals , Body Burden , Carcinogens/metabolism , Carcinogens/toxicity , Humans , Metabolic Networks and Pathways , Rats , Risk Assessment
8.
Drug Metab Rev ; 40(3): 447-63, 2008.
Article in English | MEDLINE | ID: mdl-18642142

ABSTRACT

A Draft Guidance for Industry on "Safety Testing of Drug Metabolites" was released by FDA in 2005. According to these recommendations, there may be instances when the safety profile of human metabolites may mandate their direct safety testing in animals prior to registration and approval of new molecular entity. In response to this evolving regulatory environment, pragmatic and scientifically driven approaches should be used to assess which (if any) metabolites may require direct safety testing in animals. A specific Lilly case study highlights a strategic approach for evaluation of unique and major human metabolites of a drug in Phase 2 development.


Subject(s)
Clinical Trials as Topic , Drug Evaluation, Preclinical , Drugs, Investigational/pharmacokinetics , Toxicity Tests , Animals , Biotransformation , Clinical Trials as Topic/legislation & jurisprudence , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Approval , Drugs, Investigational/toxicity , Government Regulation , Guidelines as Topic , Humans , Risk Assessment , United States , United States Food and Drug Administration
9.
Birth Defects Res ; 110(18): 1358-1371, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30367709

ABSTRACT

BACKGROUND: Olaratumab (Lartruvo™) is a recombinant human IgG1 monoclonal antibody that specifically binds PDGFRα. The maternal and in utero embryo-fetal toxicity and toxicokinetics of a human anti-mouse PDGFRα antibody (LSN3338786) were investigated in pregnant mice. METHODS: A pilot study was used to set doses for the definitive study. In the definitive study, mice were administered vehicle, 5, 50, or 150 mg/kg LSN3338786 by intravenous injection on gestation days (GD) 6, 9, 12, and 15. Fetal tissues and/or serum samples were collected on GD 10, 12, 15, and 18 to evaluate exposure of antibody. RESULTS: There were no adverse maternal effects at 50 and 150 mg/kg although maternal deaths and adverse clinical signs were observed at 5 mg/kg. LSN3338786 crossed the placenta as early as GD 10 during organogenesis. Elimination half-life of LSN3338786 in dams decreased between GD 6 and 15. On GD 18, fetal serum concentrations of antibody were substantially higher than maternal serum concentrations at all doses. Increased incidences of malformations consisting of open and partially open eye and increased incidences of skeletal variation frontal/parietal additional ossification site occurred in fetuses from mid- and high-dose groups. CONCLUSIONS: The majority of transplacental migration of antibody occurred in concert with rapid maternal serum clearance before parturition. The no-observed effect level for teratogenicity of 5 mg/kg was associated with GD 15 maternal serum concentrations 3-11 times lower than clinical exposure of olaratumab, suggesting that olaratumab may cause fetal harm when administered to pregnant women.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/toxicity , Receptor, Platelet-Derived Growth Factor alpha/immunology , Animals , Antibodies, Monoclonal/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Fetal Development/drug effects , Fetus , Maternal Exposure/adverse effects , Mice , Mice, Inbred Strains , No-Observed-Adverse-Effect Level , Organogenesis , Pilot Projects , Placenta , Pregnancy , Toxicity Tests/methods
10.
Clin Pharmacokinet ; 57(3): 355-365, 2018 03.
Article in English | MEDLINE | ID: mdl-28620891

ABSTRACT

BACKGROUND AND OBJECTIVES: Olaratumab is a recombinant human monoclonal antibody that binds to platelet-derived growth factor receptor-α (PDGFRα). In a randomized phase II study, olaratumab plus doxorubicin met its predefined primary endpoint for progression-free survival and achieved a highly significant improvement in overall survival versus doxorubicin alone in patients with advanced or metastatic soft tissue sarcoma (STS). In this study, we characterize the pharmacokinetics (PKs) of olaratumab in a cancer patient population. METHODS: Olaratumab was tested at 15 or 20 mg/kg in four phase II studies (in patients with nonsmall cell lung cancer, glioblastoma multiforme, STS, and gastrointestinal stromal tumors) as a single agent or in combination with chemotherapy. PK sampling was performed to measure olaratumab serum levels. PK data were analyzed by nonlinear mixed-effect modeling techniques using NONMEM®. RESULTS: The PKs of olaratumab were best described by a two-compartment PK model with linear clearance (CL). Patient body weight was found to have a significant effect on both CL and central volume of distribution (V 1), whereas tumor size significantly affected CL. A small subset of patients developed treatment-emergent anti-drug antibodies (TE-ADAs); however, TE-ADAs did not have any effect on CL or PK time course of olaratumab. There was no difference in the PKs of olaratumab between patients who received olaratumab as a single agent or in combination with chemotherapy. CONCLUSION: The PKs of olaratumab were best described by a model with linear disposition. Patient body weight and tumor size were found to be significant covariates. The PKs of olaratumab were not affected by immunogenicity or chemotherapeutic agents.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Models, Biological , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Body Weight , Disease-Free Survival , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Nonlinear Dynamics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Survival Rate , Time Factors , Young Adult
11.
J Clin Pharmacol ; 58(4): 412-424, 2018 04.
Article in English | MEDLINE | ID: mdl-29178617

ABSTRACT

The safety, pharmacokinetic, and pharmacodynamic effects of LY2584702, a selective inhibitor for p70 S6 serine/threonine protein kinase-1, were evaluated in healthy dyslipidemic volunteers. LY2584702 was tolerated well as a monotherapy and dose-dependently reduced low-density lipoprotein cholesterol and triglycerides by up to 60% and 50%, respectively, without significantly changing high-density lipoprotein cholesterol levels in plasma. LY2584702 also dose-dependently decreased factor V activity. Alanine aminotransferase elevations were noted in 2 subjects when LY2584702 was given with atorvastatin. We suspect that the formation of 4-aminopyrazolo[3,4-d]pyrimidine (4-APP) during metabolism may have contributed to some of the adverse effects of LY2584702, and the contribution of 4-APP to the pharmacology merits further investigation. Although clinical investigation of LY2584702 has been terminated because of hepatotoxicity risk, we suggest that a selective inhibitor of p70 S6 serine/threonine protein kinase-1 with a larger margin of safety and without the possibility of being metabolized to 4-APP may be useful in the treatment of dyslipidemia.


Subject(s)
Cholesterol, LDL/blood , Dyslipidemias/blood , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/metabolism , Adult , Aged , Alanine Transaminase/blood , Cross-Over Studies , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/blood , Pyrazoles/blood , Pyrimidines/blood , Single-Blind Method , Young Adult
12.
J Med Chem ; 61(5): 2041-2051, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29425457

ABSTRACT

Two 2-aminoimidazole-based inhibitors, LY3031207 (1) and LY3023703 (2), of the microsomal prostaglandin E synthase-1 (mPGES-1) enzyme were found to cause drug-induced liver injury (DILI) in humans. We studied imidazole ring substitutions to successfully mitigate reactive metabolite (RM) formation. These studies support the conclusion that RM formation may play a role in the observations of DILI and the consideration of 2-aminoimidazoles as structure alerts, due to the high likelihood of bioactivation to generate RMs.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Imidazoles/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Humans , Imidazoles/adverse effects , Imidazoles/metabolism , Safety-Based Drug Withdrawals , Structure-Activity Relationship
13.
Clin Cancer Res ; 24(4): 847-857, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29191969

ABSTRACT

Purpose: Platelet-derived growth factor receptor α (PDGFRα) is implicated in several adult and pediatric malignancies, where activated signaling in tumor cells and/or cells within the microenvironment drive tumorigenesis and disease progression. Olaratumab (LY3012207/IMC-3G3) is a human mAb that exclusively binds to PDGFRα and recently received accelerated FDA approval and conditional EMA approval for treatment of advanced adult sarcoma patients in combination with doxorubicin. In this study, we investigated olaratumab in preclinical models of pediatric bone and soft tissue tumors.Experimental Design: PDGFRα expression was evaluated by qPCR and Western blot analysis. Olaratumab was investigated in in vitro cell proliferation and invasion assays using pediatric osteosarcoma and rhabdoid tumor cell lines. In vivo activity of olaratumab was assessed in preclinical mouse models of pediatric osteosarcoma and malignant rhabdoid tumor.Results:In vitro olaratumab treatment of osteosarcoma and rhabdoid tumor cell lines reduced proliferation and inhibited invasion driven by individual platelet-derived growth factors (PDGFs) or serum. Furthermore, olaratumab delayed primary tumor growth in mouse models of pediatric osteosarcoma and malignant rhabdoid tumor, and this activity was enhanced by combination with either doxorubicin or cisplatin.Conclusions: Overall, these data indicate that olaratumab, alone and in combination with standard of care, blocks the growth of some preclinical PDGFRα-expressing pediatric bone and soft tissue tumor models. Clin Cancer Res; 24(4); 847-57. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line , Cell Line, Tumor , Child , Disease-Free Survival , Humans , Mice, Nude , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sarcoma/genetics , Sarcoma/metabolism , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
14.
J Med Chem ; 59(10): 4753-68, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27067148

ABSTRACT

Transmembrane AMPA receptor regulatory proteins (TARPs) are a family of scaffolding proteins that regulate AMPA receptor trafficking and function. TARP γ-8 is one member of this family and is highly expressed within the hippocampus relative to the cerebellum. A selective TARP γ-8-dependent AMPA receptor antagonist (TDAA) is an innovative approach to modulate AMPA receptors in specific brain regions to potentially increase the therapeutic index relative to known non-TARP-dependent AMPA antagonists. We describe here, for the first time, the discovery of a noncompetitive AMPA receptor antagonist that is dependent on the presence of TARP γ-8. Three major iteration cycles were employed to improve upon potency, CYP1A2-dependent challenges, and in vivo clearance. An optimized molecule, compound (-)-25 (LY3130481), was fully protective against pentylenetetrazole-induced convulsions in rats without the motor impairment associated with non-TARP-dependent AMPA receptor antagonists. Compound (-)-25 could be utilized to provide proof of concept for antiepileptic efficacy with reduced motor side effects in patients.


Subject(s)
Calcium Channels/metabolism , Drug Discovery , Receptors, AMPA/antagonists & inhibitors , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Molecular Structure , Receptors, AMPA/metabolism
16.
Expert Opin Drug Metab Toxicol ; 8(8): 985-97, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22681256

ABSTRACT

INTRODUCTION: An evolution in bioanalytical methodologies to identify and quantify drug metabolites has led to a wealth of biotransformation information during preclinical and early clinical testing phases. However, this abundance of metabolism data has not clarified how to select the most important circulating human metabolites for safety assessment. Consequently, more stringent regulatory expectations for a comprehensive approach to human metabolism have led pharmaceutical sponsors to employ a variety of novel methods to estimate circulating drug metabolites in humans and animals. AREAS COVERED: This review provides context for 'why' human circulating metabolites must be qualified for safety in animals. A detailed overview is also presented concerning 'where,' 'how' and 'when' to conduct these assessments during drug development. EXPERT OPINION: A human metabolite qualification strategy is now a required element of the drug safety package submitted with a new drug application (NDA). The important question is whether or not this additional information, about metabolite safety, is making human drugs any safer. Currently, this is a debatable issue, especially because stand-alone metabolite testing is fraught with its own challenges. As drug development moves into the twenty-first century, there is a pressing need for more sophisticated methodologies to address human drug and metabolite safety.


Subject(s)
Biotransformation , Drug Evaluation, Preclinical/standards , Pharmacokinetics , Animals , Databases, Factual , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/metabolism , Guidelines as Topic , Humans , Inactivation, Metabolic , Pharmaceutical Preparations/metabolism , United States , United States Food and Drug Administration
18.
Bioanalysis ; 2(7): 1195-210, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21083234

ABSTRACT

BACKGROUND: It has become important for metabolism scientists to identify and quantify prominent circulating human metabolites in order to develop a metabolite safety-qualification package that meets regulatory standards. Often these metabolites cannot be analyzed using traditional bioanalytical methods because a standard is not available. RESULTS: A radiocalibration method is described that can estimate circulating metabolite concentrations in nonradioactive human and animal plasma. The key to this method is application of a pseudo internal standard (PIS) that is present in both radioactive reference and nonradioactive (i.e., unknown) samples. Metabolite exposure in the unknown samples is estimated from measured PIS exposure using a relative molar ratio established between the metabolite and PIS (usually parent drug). CONCLUSION: Two case studies demonstrate that the method can be used to establish human metabolite safety coverage in animal plasma and method validation is demonstrated by comparing estimated metabolite concentrations in human plasma with concentrations obtained directly using a metabolite calibration curve.


Subject(s)
Blood Chemical Analysis/methods , Blood Chemical Analysis/standards , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/metabolism , Animals , Calibration , Dogs , Female , Humans , Male , Pharmaceutical Preparations/administration & dosage , Radioactivity , Rats , Reference Standards , Reproducibility of Results , Tandem Mass Spectrometry
19.
Bioanalysis ; 2(7): 1249-61, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21083238

ABSTRACT

Regulatory guidelines on MIST were initially established in 2005 and finalized in 2008 by the US FDA and this has led to much discussion and debate on how to apply these recommendations in today's resource-constrained pharmaceutical environment. There are four aspects of MIST that impact on the field of bioanalysis: definition of a disproportionate human metabolite, establishment of nonclinical (animal) safety coverage for important human metabolites, degree of rigor in validation of bioanalytical methods to quantify metabolites when synthetic standards are available, and semiquantitation of metabolites when synthetic standards are not available. In this manuscript, each of these points has been addressed from a pharmaceutical industry standpoint, including a perspective on the necessary convergence of the fields of metabolite safety testing and bioanalysis.


Subject(s)
Drug Industry , Drug-Related Side Effects and Adverse Reactions , Pharmaceutical Preparations/metabolism , Toxicity Tests/methods , Animals , Humans , Pharmaceutical Preparations/analysis , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL