Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Pathol ; 46(4): 408-420, 2018 06.
Article in English | MEDLINE | ID: mdl-29768985

ABSTRACT

Tanezumab, an anti-nerve growth factor (NGF) antibody, is in development for management of chronic pain. During clinical trials of anti-NGF antibodies, some patients reported unexpected adverse events requiring total joint replacements, resulting in a partial clinical hold on all NGF inhibitors. Three nonclinical toxicology studies were conducted to evaluate the effects of tanezumab or the murine precursor muMab911 on selected bone and joint endpoints and biomarkers in cynomolgus monkeys, Sprague-Dawley rats, and C57BL/6 mice. Joint and bone endpoints included histology, immunohistochemistry, microcomputed tomography (mCT) imaging, and serum biomarkers of bone physiology. Responses of bone endpoints to tanezumab were evaluated in monkeys at 4 to 30 mg/kg/week for 26 weeks and in rats at 0.2 to 10 mg/kg twice weekly for 28 days. The effects of muMab911 at 10 mg/kg/week for 12 weeks on selected bone endpoints were determined in mice. Tanezumab and muMab911 had no adverse effects on any bone or joint parameter. There were no test article-related effects on bone or joint histology, immunohistochemistry, or structure. Reversible, higher osteocalcin concentrations occurred only in the rat study. No deleterious effects were observed in joints or bones in monkeys, rats, or mice administered high doses of tanezumab or muMab911.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Bone and Bones/drug effects , Joints/drug effects , Nerve Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/toxicity , Macaca fascicularis , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Tomography, X-Ray Computed
2.
Ann Rheum Dis ; 76(1): 295-302, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27381034

ABSTRACT

OBJECTIVE: To investigate whether the effects of nerve growth factor (NGF) inhibition with tanezumab on rats with medial meniscal tear (MMT) effectively model rapidly progressive osteoarthritis (RPOA) observed in clinical trials. METHODS: Male Lewis rats underwent MMT surgery and were treated weekly with tanezumab (0.1, 1 or 10 mg/kg), isotype control or vehicle for 7, 14 or 28 days. Gait deficiency was measured to assess weight-bearing on the operated limb. Joint damage was assessed via histopathology. A second arm, delayed onset of treatment (starting 3-8 weeks after MMT surgery) was used to control for analgesia early in the disease process. A third arm, mid-tibial amputation, evaluated the dependency of the model on weight-bearing. RESULTS: Gait deficiency in untreated rats was present 3-7 days after MMT surgery, with a return to normal weight-bearing by days 14-28. Prophylactic treatment with tanezumab prevented gait deficiency and resulted in more severe cartilage damage. When onset of treatment with tanezumab was delayed to 3-8 weeks after MMT surgery, there was no increase in cartilage damage. Mid-tibial amputation completely prevented cartilage damage in untreated MMT rats. CONCLUSIONS: These data suggest that analgesia due to NGF inhibition during the acute injury phase is responsible for increased voluntary weight-bearing and subsequent cartilage damage in the rat MMT model. This model failed to replicate the hypotrophic bone response observed in tanezumab-treated patients with RPOA.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cartilage, Articular/injuries , Nerve Growth Factor/antagonists & inhibitors , Tibial Meniscus Injuries/drug therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/toxicity , Arthritis, Experimental/chemically induced , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Gait , Male , Menisci, Tibial/diagnostic imaging , Menisci, Tibial/pathology , Radiography , Rats, Inbred Lew , Tibial Meniscus Injuries/diagnostic imaging , Tibial Meniscus Injuries/pathology , Tibial Meniscus Injuries/physiopathology , Weight-Bearing , X-Ray Microtomography
3.
Neuroreport ; 25(11): 829-832, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-24848615

ABSTRACT

Serum ß-nerve growth factor (NGF) concentrations were determined in pregnant female, nonpregnant female, and male cynomolgus monkeys using a highly selective and sensitive immunoaffinity liquid chromatography-tandem mass spectrometry assay. NGF was significantly higher in pregnant monkeys than in nonpregnant female and male monkeys. NGF increased over pregnancy (mean NGF±SD: 541±448, 1590±520, and 3560±1430 pg/ml during the first, second, and third trimesters, respectively). These data will aid in further understanding the role of NGF during pregnancy.

4.
Toxicol Pathol ; 42(7): 1069-81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24604381

ABSTRACT

Comparative nonclinical studies were conducted with the proposed biosimilar PF-05280586 and rituximab-EU (MabThera®). In side-by-side analyses, peptide maps and complement-dependent cytotoxicity assay results were similar. Sexually-mature cynomolgus monkeys were administered PF-05280586 or rituximab-EU as a single dose of 0, 2, 10, or 20 mg/kg on day 1 and observed for 92 days (single-dose study) or as 5 weekly injections of 0 or 20 mg/kg and necropsied on day 30, the day after the 5th dose, or on day 121 (repeat-dose study). The pharmacokinetic and pharmacodynamic profiles for both molecules were similar. Marked depletion of peripheral blood B cells 4 days after dosing was followed by near or complete repletion (single-dose study) or partial repletion (repeat-dose study). In the single-dose study, anti-drug antibodies (ADA) were detected by day 29 in all animals administered PF-05280586 or rituximab-EU and persisted through day 85, the last day tested. In the repeat-dose study, ADA were detected on day 121 in 50% of animals administered PF-05280586 or rituximab-EU. Both molecules were well tolerated at all doses. In all endpoints evaluated, PF-05280586 exhibited similarity to rituximab-EU.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Animals , Antigens, CD20/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Biosimilar Pharmaceuticals/administration & dosage , Biosimilar Pharmaceuticals/pharmacology , Dose-Response Relationship, Drug , Endpoint Determination , Female , Macaca fascicularis , Male , Reproducibility of Results , Rituximab
5.
Bioorg Med Chem Lett ; 19(9): 2409-12, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19346128

ABSTRACT

The synthesis and SAR of tolylamines with 5-HT(6) receptor antagonist activity is presented. The amine, core aromatic, peripheral aromatic, and ether linker moieties of HTS hit 1 were modulated and the effect on potency at 5-HT(6) examined. Tolylpiperidine ether 9h was found to possess desirable pharmacokinetic (PK) properties, and was also shown to enhance cognition in the rat novel object recognition paradigm.


Subject(s)
Amines/chemistry , Receptors, Serotonin/chemistry , Serotonin Receptor Agonists/chemical synthesis , Animals , Chemistry, Organic/methods , Chemistry, Pharmaceutical/methods , Drug Design , Ethers/chemistry , Inhibitory Concentration 50 , Kinetics , Models, Chemical , Rats , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/pharmacology , Structure-Activity Relationship
6.
Drug Metab Dispos ; 35(8): 1315-24, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17470524

ABSTRACT

Gemfibrozil coadministration generally results in plasma statin area under the curve (AUC) increases, ranging from moderate (2- to 3-fold) with simvastatin, lovastatin, and pravastatin to most significant with cerivastatin (5.6-fold). Inhibition of statin glucuronidation has been postulated as a potential mechanism of interaction (Drug Metab Dispos 30:1280-1287, 2002). This study was conducted to determine the in vitro inhibitory potential of fibrates toward atorvastatin glucuronidation. [(3)H]Atorvastatin, atorvastatin, and atorvastatin lactone were incubated with human liver microsomes or human recombinant UDP-glucuronosyltransferases (UGTs) and characterized using liquid chromatography (LC)/tandem mass spectrometry and LC/UV/beta-radioactivity monitor/mass spectrometry. [(3)H]Atorvastatin yields a minor ether glucuronide (G1) and a major acyl glucuronide (G2) with subsequent pH-dependent lactonization of G2 to yield atorvastatin lactone. Atorvastatin lactonization best fit substrate inhibition kinetics (K(m) = 12 microM, V(max) = 74 pmol/min/mg, K(i) = 75 microM). Atorvastatin lactone yields a single ether glucuronide (G3). G3 formation best fit Michaelis-Menten kinetics (K(m) = 2.6 microM, V(max) = 10.6 pmol/min/mg). Six UGT enzymes contribute to atorvastatin glucuronidation with G2 and G3 formation catalyzed by UGTs 1A1, 1A3, 1A4, 1A8, and 2B7, whereas G1 formation was catalyzed by UGTs 1A3, 1A4, and 1A9. Gemfibrozil, fenofibrate, and fenofibric acid inhibited atorvastatin lactonization with IC(50) values of 346, 320, and 291 microM, respectively. Based on unbound fibrate concentrations at the inlet to the liver, these data predict a small increase in atorvastatin AUC (approximately 1.2-fold) after gemfibrozil coadministration and no interaction with fenofibrate. This result is consistent with recent clinical reports indicating minimal atorvastatin AUC increases ( approximately 1.2- to 1.4-fold) with gemfibrozil.


Subject(s)
Fenofibrate/analogs & derivatives , Fenofibrate/pharmacology , Gemfibrozil/pharmacology , Glucuronic Acid/metabolism , Heptanoic Acids/metabolism , Pyrroles/metabolism , Anticholesteremic Agents/metabolism , Anticholesteremic Agents/pharmacology , Area Under Curve , Atorvastatin , Catalysis/drug effects , Drug Interactions , Glucuronides/analysis , Glucuronides/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Heptanoic Acids/chemistry , Heptanoic Acids/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kinetics , Lactones/analysis , Lactones/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Biological , Molecular Structure , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Recombinant Proteins/metabolism , Tandem Mass Spectrometry , Uridine Diphosphate Glucuronic Acid/metabolism
7.
Drug Metab Dispos ; 33(9): 1349-54, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15980101

ABSTRACT

The predominant metabolic pathway of gemcabene in humans is glucuronidation. The principal human UDP-glucuronosyltransferases (UGTs) involved in the glucuronidation of gemcabene were determined in this study. Glucuronidation of gemcabene was catalyzed by recombinant UGT1A3, recombinant UGT2B7, and recombinant UGT2B17, as well as by human liver microsomes (HLM). Gemcabene glucuronidation in recombinant UGTs and HLM followed non-Michaelis-Menten kinetics consistent with homotropic activation, but pharmacokinetics in humans were linear over the dose range tested (total plasma C(max), 0.06-0.88 mM). Gemcabene showed similar affinity (S(50)) for recombinant UGTs (0.92-1.45 mM) and HLM (1.37 mM). S-Flurbiprofen was identified as a more selective inhibitor of recombinant UGT2B7-catalyzed gemcabene glucuronidation (>23-fold lower IC(50)) when compared with recombinant UGT1A3- or recombinant UGT2B17-catalyzed gemcabene glucuronidation. The IC(50) for S-flurbiprofen inhibition of gemcabene glucuronidation was similar in HLM (60.6 microM) compared with recombinant UGT2B7 (27.4 microM), consistent with a major role for UGT2B7 in gemcabene glucuronidation in HLM. In addition, 5,6,7,3',4',5'-hexamethoxyflavone inhibited recombinant UGT1A3 and recombinant UGT2B17-catalyzed gemcabene glucuronidation (with 4-fold greater potency for recombinant UGT1A3) but did not inhibit gemcabene glucuronidation in HLM, suggesting that UGT1A3 and UGT2B17 do not contribute significantly to gemcabene glucuronidation. Reaction rates for gemcabene glucuronidation from a human liver bank correlated well (r(2)=0.722, P<0.0001; n=24) with rates of glucuronidation of the UGT2B7 probe substrate 3'-azido-3'-deoxythymidine. In conclusion, using the three independent experimental approaches typically used for cytochrome P450 reaction phenotyping, UGT2B7 is the major enzyme contributing to gemcabene glucuronidation in human liver microsomes.


Subject(s)
Caproates/metabolism , Glucuronosyltransferase/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Humans , Microsomes, Liver/enzymology , Minor Histocompatibility Antigens , Phenotype , Recombinant Proteins/metabolism , Zidovudine/analogs & derivatives , Zidovudine/metabolism
8.
Curr Drug Metab ; 4(6): 527-34, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14683480

ABSTRACT

For the pharmaceutical industry, one of the challenges in evaluating the risk of future compound attrition at the discovery stage is the successful prediction of the major routes of clearance in humans. For compounds cleared by metabolism, such information will help to avoid the development of compounds that will exhibit large interpatient differences in pharmacokinetics via 1). routes of metabolism catalyzed by functionally polymorphic enzymes and/or 2). clinically significant metabolic drug-drug interactions, in the later stages of development. The degree of intersubject variability that is acceptable for a drug candidate is uncertain in the discovery stage where knowledge of other important factors is limited or unavailable (i.e. therapeutic index, pharmacodynamic variability, etc). Reaction phenotyping is the semi-quantitative in vitro estimation of the relative contributions of specific drug-metabolizing enzymes to the metabolism of a test compound. However, reaction phenotyping in the discovery stage of drug development is complicated by the absence of radiolabelled parent compound or metabolite bioanalytical standards relative to later stages of development. In this commentary, some of the approaches, based on published data, which can be taken to overcome these challenges are discussed. In addition, knowledge of the molecular structure (i.e. specific chemical substituents), physicochemical properties, and routes of clearance in animals can all help in making a successful prediction for the routes of clearance in humans. In combination, the objective of these studies should be to reduce to a minimum the risk of finding significant inter-patient differences in pharmacokinetics at a later stage in development due to significant metabolism by polymorphic enzymes or drug-drug interactions. Consequently, this data should be used to avoid costly late stage attrition.


Subject(s)
Drug Design , Phenotype , Cytochrome P-450 Enzyme System/metabolism , Drug Industry , Humans , Pharmaceutical Preparations/metabolism
9.
Drug Metab Dispos ; 31(12): 1526-30, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625349

ABSTRACT

Recent research on CYP3A5 in vitro and in humans has provided discordant information on whether CYP3A5 plays a significant role in the metabolism of CYP3A substrates in vivo. For example, six separate studies have reported CYP3A5 to contribute between 2 and 60% of the total hepatic CYP3A. Suggested explanations for the reported differences in hepatic CYP3A5 levels are evaluated in this article. Furthermore, a sensitivity analysis of the contribution of CYP3A5 (in addition to CYP3A4) to the metabolism of a "midazolam"-type substrate based on recently published in vitro and clinical data is compared with the results of two in vivo studies that investigated the influence of CYP3A5 genotype on midazolam pharmacokinetics. The sensitivity analysis predicts an approximately 3-fold lower AUCoral for midazolam for those expressing the highest hepatic and intestinal levels of CYP3A5 (e.g., possessing CYP3A5*1 alleles) compared with those individuals who express insignificant amounts of CYP3A5, assuming CYP3A4 levels are the same in both groups and that CYP3A5 levels do not exceed those of CYP3A4 in CYP3A5*1 homozygotes. In contrast, the two in vivo studies show no statistically significant influence of CYP3A5 genotype on midazolam pharmacokinetics. The discordance between the prediction and the results from the two in vivo studies is discussed.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Pharmaceutical Preparations/metabolism , Animals , Cyclosporine/pharmacokinetics , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Humans , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Midazolam/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...