Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters











Publication year range
1.
J Pharmacol Exp Ther ; 358(3): 537-47, 2016 09.
Article in English | MEDLINE | ID: mdl-27317802

ABSTRACT

Developing therapeutics for neurodegenerative diseases (NDs) prevalent in the aging population remains a daunting challenge. With the growing understanding that many NDs progress by conformational self-templating of specific proteins, the prototypical prion diseases offer a platform for ND drug discovery. We evaluated high-throughput screening hits with the aryl amide scaffold and explored the structure-activity relationships around three series differing in their N-aryl core: benzoxazole, benzothiazole, and cyano. Potent anti-prion compounds were advanced to pharmacokinetic studies, and the resulting brain-penetrant leads from each series, together with a related N-aryl piperazine lead, were escalated to long-term dosing and efficacy studies. Compounds from each of the four series doubled the survival of mice infected with a mouse-passaged prion strain. Treatment with aryl amides altered prion strain properties, as evidenced by the distinct patterns of neuropathological deposition of prion protein and associated astrocytic gliosis in the brain; however, none of the aryl amide compounds resulted in drug-resistant prion strains, in contrast to previous studies on compounds with the 2-aminothiazole (2-AMT) scaffold. As seen with 2-AMTs and other effective anti-prion compounds reported to date, the novel aryl amides reported here were ineffective in prolonging the survival of transgenic mice infected with human prions. Most encouraging is our discovery that aryl amides show that the development of drug resistance is not an inevitable consequence of efficacious anti-prion therapeutics.


Subject(s)
Amides/chemistry , Amides/pharmacology , Drug Discovery , Prion Diseases/drug therapy , Amides/metabolism , Amides/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Creutzfeldt-Jakob Syndrome/drug therapy , Female , Mice , Prion Diseases/metabolism , Structure-Activity Relationship , Survival Analysis
2.
J Pharmacol Exp Ther ; 355(1): 2-12, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26224882

ABSTRACT

Because no drug exists that halts or even slows any neurodegenerative disease, developing effective therapeutics for any prion disorder is urgent. We recently reported two compounds (IND24 and IND81) with the 2-aminothiazole (2-AMT) chemical scaffold that almost doubled the incubation times in scrapie prion-infected, wild-type (wt) FVB mice when given in a liquid diet. Remarkably, oral prophylactic treatment with IND24 beginning 14 days prior to intracerebral prion inoculation extended survival from ∼120 days to over 450 days. In addition to IND24, we evaluated the pharmacokinetics and efficacy of five additional 2-AMTs; one was not followed further because its brain penetration was poor. Of the remaining four new 2-AMTs, IND114338 doubled and IND125 tripled the incubation times of RML-inoculated wt and Tg4053 mice overexpressing wt mouse prion protein (PrP), respectively. Neuropathological examination of the brains from untreated controls showed a widespread deposition of self-propagating, ß-sheet-rich "scrapie" isoform (PrP(Sc)) prions accompanied by a profound astrocytic gliosis. In contrast, mice treated with 2-AMTs had lower levels of PrP(Sc) and associated astrocytic gliosis, with each compound resulting in a distinct pattern of deposition. Notably, IND125 prevented both PrP(Sc) accumulation and astrocytic gliosis in the cerebrum. Progressive central nervous system dysfunction in the IND125-treated mice was presumably due to the PrP(Sc) that accumulated in their brainstems. Disappointingly, none of the four new 2-AMTs prolonged the lives of mice expressing a chimeric human/mouse PrP transgene inoculated with Creutzfeldt-Jakob disease prions.


Subject(s)
Brain/drug effects , Brain/pathology , PrPSc Proteins/metabolism , Thiazoles/chemistry , Thiazoles/pharmacology , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Female , Humans , Mice , PrPSc Proteins/genetics , Scrapie/pathology , Species Specificity , Survival Analysis , Survival Rate , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use , Transgenes/genetics , Treatment Outcome
3.
Bioorg Med Chem ; 22(6): 1960-72, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24530226

ABSTRACT

PURPOSE: Previous studies showed that lowering PrP(C) concomitantly reduced PrP(Sc) in the brains of mice inoculated with prions. We aimed to develop assays that measure PrP(C) on the surface of human T98G glioblastoma and IMR32 neuroblastoma cells. Using these assays, we sought to identify chemical hits, confirmed hits, and scaffolds that potently lowered PrP(C) levels in human brains cells, without lethality, and that could achieve drug concentrations in the brain after oral or intraperitoneal dosing in mice. METHODS: We utilized HTS ELISA assays to identify small molecules that lower PrP(C) levels by ≥30% on the cell surface of human glioblastoma (T98G) and neuroblastoma (IMR32) cells. RESULTS: From 44,578 diverse chemical compounds tested, 138 hits were identified by single point confirmation (SPC) representing 7 chemical scaffolds in T98G cells, and 114 SPC hits representing 6 scaffolds found in IMR32 cells. When the confirmed SPC hits were combined with structurally related analogs, >300 compounds (representing 6 distinct chemical scaffolds) were tested for dose-response (EC50) in both cell lines, only studies in T98G cells identified compounds that reduced PrP(C) without killing the cells. EC50 values from 32 hits ranged from 65 nM to 4.1 µM. Twenty-eight were evaluated in vivo in pharmacokinetic studies after a single 10 mg/kg oral or intraperitoneal dose in mice. Our results showed brain concentrations as high as 16.2 µM, but only after intraperitoneal dosing. CONCLUSIONS: Our studies identified leads for future studies to determine which compounds might lower PrP(C) levels in rodent brain, and provide the basis of a therapeutic for fatal disorders caused by PrP prions.


Subject(s)
Prions/analysis , Small Molecule Libraries/pharmacology , Animals , Brain , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fluorescence , High-Throughput Screening Assays , Humans , Mice , Microscopy, Confocal , Molecular Structure , Protein Isoforms , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Properties , Tissue Distribution , Tumor Cells, Cultured
4.
Bioorg Med Chem ; 21(24): 7999-8012, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24183589

ABSTRACT

During prion diseases, a normally benign, host protein, denoted PrP(C), undergoes alternative folding into the aberrant isoform, PrP(Sc). We used ELISA to identify and confirm hits in order to develop leads that reduce PrP(Sc) in prion-infected dividing and stationary-phase mouse neuroblastoma (ScN2a-cl3) cells. We tested 52,830 diverse small molecules in dividing cells and 49,430 in stationary-phase cells. This led to 3100 HTS and 970 single point confirmed (SPC) hits in dividing cells, 331 HTS and 55 confirmed SPC hits in stationary-phase cells as well as 36 confirmed SPC hits active in both. Fourteen chemical leads were identified from confirmed SPC hits in dividing cells and three in stationary-phase cells. From more than 682 compounds tested in concentration-effect relationships in dividing cells to determine potency (EC50), 102 had EC50 values between 1 and 10 µM and 50 had EC50 values of <1 µM; none affected cell viability. We observed an excellent correlation between EC50 values determined by ELISA and Western immunoblotting for 28 representative compounds in dividing cells (R(2)=0.75; p <0.0001). Of the 55 confirmed SPC hits in stationary-phase cells, 23 were piperazine, indole, or urea leads. The EC50 values of one indole in stationary-phase and dividing ScN2a-cl3 cells were 7.5 and 1.6 µM, respectively. Unexpectedly, the number of hits in stationary-phase cells was ~10% of that in dividing cells. The explanation for this difference remains to be determined.


Subject(s)
PrPSc Proteins/metabolism , Small Molecule Libraries/pharmacology , Animals , Blotting, Western , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Mice , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship
5.
ACS Med Chem Lett ; 4(7): 647-650, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23977416

ABSTRACT

The prion diseases caused by PrPSc, an alternatively folded form of the cellular prion protein (PrPC), are rapidly progressive, fatal, and untreatable neurodegenerative syndromes. We employed HTS ELISA assays to identify compounds that lower the level of PrPSc in prion-infected mouse neuroblastoma (ScN2a-cl3) cells and identified a series of arylamides. SAR studies indicated that small amides with one aromatic, or heteroaromatic ring, on each side of the amide bond are of modest potency. Of note, benzamide (7), with an EC50 of 2200 nM, was one of only a few arylamide hits with a piperazine group on its aniline moiety. The basic piperazine nitrogen can be protonated at physiologic pH, improving solubility, and therefore we wanted to exploit this feature in our search for a drug candidate. An SAR campaign resulted in several key analogs, including a set with biaryl groups introduced on the carbonyl side for improved potency. Several of these biaryl analogs have submicromolar potency, with the most potent analog 17 having an EC50 = 22 nM. More importantly, 17 and several biarylamides (20, 24, 26, 27) were able to traverse the BBB and displayed excellent drug levels in the brains of mice following oral dosing. These biarylamides may represent good starting points for further lead optimization for the identification of potential drug candidates for the treatment of prion diseases.

6.
J Pharmacol Exp Ther ; 347(2): 325-38, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23965382

ABSTRACT

The only small-molecule compound demonstrated to substantially extend survival in prion-infected mice is a biaryl hydrazone termed "Compd B" (4-pyridinecarboxaldehyde,2-[4-(5-oxazolyl)phenyl]hydrazone). However, the hydrazone moiety of Compd B results in toxic metabolites, making it a poor candidate for further drug development. We developed a pharmacophore model based on diverse antiprion compounds identified by high-throughput screening; based on this model, we generated biaryl amide analogs of Compd B. Medicinal chemistry optimization led to multiple compounds with increased potency, increased brain concentrations, and greater metabolic stability, indicating that they could be promising candidates for antiprion therapy. Replacing the pyridyl ring of Compd B with a phenyl group containing an electron-donating substituent increased potency, while adding an aryl group to the oxazole moiety increased metabolic stability. To test the efficacy of Compd B, we applied bioluminescence imaging (BLI), which was previously shown to detect prion disease onset in live mice earlier than clinical signs. In our studies, Compd B showed good efficacy in two lines of transgenic mice infected with the mouse-adapted Rocky Mountain Laboratory (RML) strain of prions, but not in transgenic mice infected with human prions. The BLI system successfully predicted the efficacies in all cases long before extension in survival could be observed. Our studies suggest that this BLI system has good potential to be applied in future antiprion drug efficacy studies.


Subject(s)
Amides/chemistry , Amides/therapeutic use , Hydrazones/chemistry , Hydrazones/therapeutic use , PrPSc Proteins/pathogenicity , Prion Diseases/drug therapy , Amides/chemical synthesis , Amides/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Discovery , Hydrazones/chemical synthesis , Hydrazones/pharmacokinetics , Infectious Disease Incubation Period , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Models, Molecular , Molecular Structure , PrPSc Proteins/genetics , Structure-Activity Relationship
7.
ACS Med Chem Lett ; 4(4): 397-401, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23847718

ABSTRACT

Prion diseases are a group of fatal neurodegenerative disorders that include Creutzfeldt-Jakob disease (CJD) and kuru in humans, BSE in cattle, and scrapie in sheep. Such illnesses are caused by the conversion and accumulation of a misfolded pathogenic isoform (termed PrPSc) of a normally benign, host cellular protein, denoted PrPC. We employed high-throughput screening (HTS) ELISAs to evaluate compounds for their ability to reduce the level of PrPSc in Rocky Mountain Laboratory (RML) prion-infected mouse neuroblastoma cells (ScN2a-cl3). Arylpiperazines were among the active compounds identified but the initial hits suffered from low potency and poor drug-likeness. The best of those hits, such as 1, 7, 13, and 19, displayed moderate antiprion activity with EC50 values in the micromolar range. Key analogs were designed and synthesized based on the SAR, with analogs 41, 44, 46, and 47 found to have sub-micromolar potency. Analogs 41 and 44 were able to penetrate the blood-brain barrier (BBB) and achieved excellent drug concentrations in the brains of mice after oral dosing. These compounds represent good starting points for further lead optimization in our pursuit of potential drug candidates for the treatment of prion diseases.

8.
ChemMedChem ; 8(5): 847-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23509039

ABSTRACT

Recently, we described the aminothiazole lead (4-biphenyl-4-ylthiazol-2-yl)-(6-methylpyridin-2-yl)-amine (1), which exhibits many desirable properties, including excellent stability in liver microsomes, oral bioavailability of ∼40 %, and high exposure in the brains of mice. Despite its good pharmacokinetic properties, compound 1 exhibited only modest potency in mouse neuroblastoma cells overexpressing the disease-causing prion protein PrP(Sc) . Accordingly, we sought to identify analogues of 1 with improved antiprion potency in ScN2a-cl3 cells while retaining similar or superior properties. Herein we report the discovery of improved lead compounds such as (6-methylpyridin-2-yl)-[4-(4-pyridin-3-yl-phenyl)thiazol-2-yl]amine and cyclopropanecarboxylic acid (4-biphenylthiazol-2-yl)amide, which exhibit brain exposure/EC50 ratios at least tenfold greater than that of compound 1.


Subject(s)
Prion Diseases/drug therapy , Thiazoles/pharmacology , Administration, Oral , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Models, Molecular , Molecular Structure , Pregnancy Proteins/biosynthesis , Prion Diseases/metabolism , Quantum Theory , Structure-Activity Relationship , Thiazoles/administration & dosage , Thiazoles/chemistry , Thiazoles/therapeutic use
9.
Pharm Res ; 30(4): 932-50, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23417511

ABSTRACT

PURPOSE: To discover drugs lowering PrP(Sc) in prion-infected cultured neuronal cells that achieve high concentrations in brain to test in mouse models of prion disease and then treat people with these fatal diseases. METHODS: We tested 2-AMT analogs for EC50 and PK after a 40 mg/kg single dose and 40-210 mg/kg/day doses for 3 days. We calculated plasma and brain AUC, ratio of AUC/EC50 after dosing. We reasoned that compounds with high AUC/EC50 ratios should be good candidates going forward. RESULTS: We evaluated 27 2-AMTs in single-dose and 10 in 3-day PK studies, of which IND24 and IND81 were selected for testing in mouse models of prion disease. They had high concentrations in brain after oral dosing. Absolute bioavailability ranged from 27-40%. AUC/EC50 ratios after 3 days were >100 (total) and 48-113 (unbound). Stability in liver microsomes ranged from 30->60 min. Ring hydroxylated metabolites were observed in microsomes. Neither was a substrate for the MDR1 transporter. CONCLUSIONS: IND24 and IND81 are active in vitro and show high AUC/EC50 ratios (total and unbound) in plasma and brain. These will be evaluated in mouse models of prion disease.


Subject(s)
PrPSc Proteins/antagonists & inhibitors , Prion Diseases/drug therapy , Thiazoles/metabolism , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Area Under Curve , Biological Availability , Brain/metabolism , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Humans , Mice , Microsomes, Liver/metabolism , PrPSc Proteins/metabolism , Protein Isoforms/metabolism , Solubility , Thiazoles/chemistry , Thiazoles/pharmacology
10.
Eur Neuropsychopharmacol ; 22(7): 492-500, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22209365

ABSTRACT

While a number of behavioural studies have been conducted to investigate the acute effects of amphetamines on tasks of attention and information processing, there is currently a scarcity of research concerning their electrophysiological effects in healthy adults. It is also unclear as to whether amphetamines exert effects on stimulus evaluation or response selection. In two studies, independent groups of twenty healthy illicit stimulant users aged between 21 and 32 years were administered 0.42 mg/kg d-amphetamine versus placebo, and 0.42 mg/kg d-methamphetamine versus placebo respectively, and completed an auditory oddball task on two separate testing days. A 62-channel EEG was recorded during the completion of the task, and the effects of amphetamines on N200 and P300 ERP components were analysed. d-amphetamine significantly decreased reaction time, improved accuracy, and reduced the latency of the P300 component relative to placebo, while having no effect on the N200 component. d-methamphetamine had no effect on reaction time, accuracy or the P300 component, but reduced the amplitude of the N200 component, relative to placebo. It was concluded that there is tentative support to suggest that d-amphetamine at a dose of 0.42 mg/kg may enhance speed of information processing while d-methamphetamine at a dose of 0.42 mg/kg may reflect changes to stimulus evaluation.


Subject(s)
Brain Waves/drug effects , Central Nervous System Stimulants/pharmacology , Dextroamphetamine/pharmacology , Evoked Potentials/drug effects , Methamphetamine/pharmacology , Acoustic Stimulation , Adult , Analysis of Variance , Dose-Response Relationship, Drug , Electroencephalography , Female , Humans , Male , Neuropsychological Tests , Reaction Time/drug effects , Young Adult
11.
Eur J Med Chem ; 46(9): 4125-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21726921

ABSTRACT

A series of highly potent indole-3-glyoxylamide based antiprion agents was previously characterized, focusing on optimization of structure-activity relationship (SAR) at positions 1-3 of the indole system. New libraries interrogating the SAR at indole C-4 to C-7 now demonstrate that introducing electron-withdrawing substituents at C-6 may improve biological activity by up to an order of magnitude, and additionally confer higher metabolic stability. For the present screening libraries, both the degree of potency and trends in SAR were consistent across two cell line models of prion disease, and the large majority of compounds showed no evidence of toxic effects in zebrafish. The foregoing observations thus make the indole-3-glyoxylamides an attractive lead series for continuing development as potential therapeutic agents against prion disease.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Microsomes/drug effects , Prions/drug effects , Animals , Cell Line , Drug Discovery , Indoles/adverse effects , Structure-Activity Relationship , Zebrafish
12.
Clin Neurophysiol ; 122(11): 2203-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21570341

ABSTRACT

OBJECTIVE: This study examined sensory and cognitive processing in adolescents, young adults and older adults, when exposed to 2nd (2G) and 3rd (3G) generation mobile phone signals. METHODS: Tests employed were the auditory 3-stimulus oddball and the N-back. Forty-one 13-15 year olds, forty-two 19-40 year olds and twenty 55-70 year olds were tested using a double-blind cross-over design, where each participant received Sham, 2G and 3G exposures, separated by at least 4 days. RESULTS: 3-Stimulus oddball task: Behavioural: accuracy and reaction time of responses to targets were not affected by exposure. Electrophysiological: augmented N1 was found in the 2G condition (independent of age group). N-back task: Behavioural: the combined groups performed less accurately during the 3G exposure (compared to Sham), with post hoc tests finding this effect separately in the adolescents only. Electrophysiological: delayed ERD/ERS responses of the alpha power were found in both 3G and 2G conditions (compared to Sham; independent of age group). CONCLUSION: Employing tasks tailored to each individual's ability level, this study provides support for an effect of acute 2G and 3G exposure on human cognitive function. SIGNIFICANCE: The subtlety of mobile phone effect on cognition in our study suggests that it is important to account for individual differences in future mobile phone research.


Subject(s)
Aging/physiology , Brain/radiation effects , Cell Phone/standards , Cognition/radiation effects , Electromagnetic Fields/adverse effects , Adolescent , Adult , Aged , Brain/growth & development , Cognition/physiology , Cognition Disorders/diagnosis , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Computer Simulation , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Models, Neurological , Young Adult
13.
J Med Chem ; 54(4): 1010-21, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21247166

ABSTRACT

2-Aminothiazoles are a new class of small molecules with antiprion activity in prion-infected neuroblastoma cell lines (J. Virol. 2010, 84, 3408). We report here structure-activity studies undertaken to improve the potency and physiochemical properties of 2-aminothiazoles, with a particular emphasis on achieving and sustaining high drug concentrations in the brain. The results of this effort include the generation of informative structure-activity relationships (SAR) and the identification of lead compounds that are orally absorbed and achieve high brain concentrations in animals. The new aminothiazole analogue (5-methylpyridin-2-yl)-[4-(3-phenylisoxazol-5-yl)-thiazol-2-yl]-amine (27), for example, exhibited an EC(50) of 0.94 µM in prion-infected neuroblastoma cells (ScN2a-cl3) and reached a concentration of ∼25 µM in the brains of mice following three days of oral administration in a rodent liquid diet. The studies described herein suggest 2-aminothiazoles as promising new leads in the search for effective therapeutics for prion diseases.


Subject(s)
Prion Diseases/drug therapy , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Animals , Biological Assay , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Magnetic Resonance Spectroscopy , Mice , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Thiazoles/chemistry
14.
Sci Transl Med ; 2(30): 30cm16, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20445199

ABSTRACT

Academic labs have been responsible for virtually all of the basic science discoveries that translate into the discovery and development of innovative new medicines. There is a growing concern that large pharmaceutical and biotechnology companies are not able to sustain research pipelines that bring new compounds into drug development that translate into innovative new medicines, especially in areas with high unmet medical need. To address the needs of patients, caregivers, and society, academic labs have played and can continue to play an important role at one or more stages in the development of innovative medicines, both directly and through collaborations with researchers in pharmaceutical and biotechnology companies. Collaboration is in the best interests of patients and society if it accelerates the translation of basic science discoveries to new medicines that address unmet medical needs.


Subject(s)
Biomedical Research/trends , Drug Discovery/trends , Technology, Pharmaceutical/methods , Academic Medical Centers , Drug Industry/organization & administration , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pharmaceutical Preparations/chemical synthesis , Universities
15.
Neurosci Biobehav Rev ; 34(3): 387-407, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19715722

ABSTRACT

Modulating central serotonergic function by acute tryptophan depletion (ATD) has provided the fundamental insights into which cognitive functions are influenced by serotonin. It may be expected that serotonergic stimulation by tryptophan (Trp) loading could evoke beneficial behavioural changes that mirror those of ATD. The current review examines the evidence for such effects, notably those on cognition, mood and sleep. Reports vary considerably across different cognitive domains, study designs, and populations. It is hypothesised that the effects of Trp loading on performance may be dependent on the initial state of the serotonergic system of the subject. Memory improvements following Trp loading have generally been shown in clinical and sub-clinical populations where initial serotonergic disturbances are known. Similarly, Trp loading appears to be most effective for improving mood in vulnerable subjects, and improves sleep in adults with some sleep disturbances. Research has consistently shown Trp loading impairs psychomotor and reaction time performance, however, this is likely to be attributed to its mild sedative effects.


Subject(s)
Affect/physiology , Cognition/physiology , Sleep/physiology , Tryptophan/metabolism , Animals , Humans
16.
Ann Intern Med ; 150(7): 493-5, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19258550

ABSTRACT

The coverage, cost, and quality problems of the U.S. health care system are evident. Sustainable health care reform must go beyond financing expanded access to care to substantially changing the organization and delivery of care. The FRESH-Thinking Project (www.fresh-thinking.org) held a series of workshops during which physicians, health policy experts, health insurance executives, business leaders, hospital administrators, economists, and others who represent diverse perspectives came together. This group agreed that the following 8 recommendations are fundamental to successful reform: 1. Replace the current fee-for-service payment system with a payment system that encourages and rewards innovation in the efficient delivery of quality care. The new payment system should invest in the development of outcome measures to guide payment. 2. Establish a securely funded, independent agency to sponsor and evaluate research on the comparative effectiveness of drugs, devices, and other medical interventions. 3. Simplify and rationalize federal and state laws and regulations to facilitate organizational innovation, support care coordination, and streamline financial and administrative functions. 4. Develop a health information technology infrastructure with national standards of interoperability to promote data exchange. 5. Create a national health database with the participation of all payers, delivery systems, and others who own health care data. Agree on methods to make de-identified information from this database on clinical interventions, patient outcomes, and costs available to researchers. 6. Identify revenue sources, including a cap on the tax exclusion of employer-based health insurance, to subsidize health care coverage with the goal of insuring all Americans. 7. Create state or regional insurance exchanges to pool risk, so that Americans without access to employer-based or other group insurance could obtain a standard benefits package through these exchanges. Employers should also be allowed to participate in these exchanges for their employees' coverage. 8. Create a health coverage board with broad stakeholder representation to determine and periodically update the affordable standard benefit package available through state or regional insurance exchanges.


Subject(s)
Health Care Reform/organization & administration , Universal Health Insurance/organization & administration , Government Regulation , Health Care Reform/economics , Humans , Insurance, Health, Reimbursement/economics , Total Quality Management/economics , United States , Universal Health Insurance/economics
17.
Psychopharmacology (Berl) ; 179(3): 536-43, 2005 May.
Article in English | MEDLINE | ID: mdl-15614573

ABSTRACT

RATIONALE: The number of road fatalities related to the presence of amphetamines in drivers has been relatively constant over the past 10 years. However, there remains uncertainty as to the extent that these drugs induce driving impairment, and whether any such impairments translate to an increase in road fatalities. OBJECTIVES: To examine the acute effects of 0.42 mg/kg dexamphetamine on simulated driving performance, and to establish which, if any, simulated driving abilities become impaired following dexamphetamine administration. METHODS: A repeated-measures, counter-balanced, double-blind, placebo-controlled design was employed. Twenty healthy volunteers completed two treatment conditions-0.42 mg/kg dexamphetamine and placebo. Performance was assessed using a driving simulator task. Blood and saliva samples were obtained prior to the driving tasks and immediately after task completion (120 min and 170 min post-drug administration, respectively). RESULTS: Mean dexamphetamine blood concentrations were 83 ng/ml and 98 ng/ml at 120 min and 170 min, respectively. Results indicated a decrease in overall simulated driving ability following dexamphetamine administration during the day-time but not the night-time scenario tasks. Contributing to this performance reduction, "incorrect signalling", "failing to stop at a red traffic light" and "slow reaction times" were the behaviours most strongly affected by dexamphetamine. CONCLUSIONS: The decrease in simulated driving ability observed during the day-time driving tasks are consistent with the perceptual narrowing or tunnel vision that is associated with dexamphetamine consumption.


Subject(s)
Automobile Driving , Computer Simulation , Dextroamphetamine/pharmacology , Psychomotor Performance/drug effects , Adult , Double-Blind Method , Female , Humans , Male , Psychomotor Performance/physiology
18.
AAPS PharmSci ; 2(4): E33, 2000.
Article in English | MEDLINE | ID: mdl-11741249

ABSTRACT

The emerging application of pharmacogenomics in the clinical trial setting requires careful comparison with more traditional phenotyping methodologies, particularly in the drug metabolism area where phenotyping is used extensively. The research objectives of this study were 1) to assess the utility of cytochrome P450 2D6 (CYP2D6) genotyping as an alternative to traditional phenotyping as a predictor of poor metabolizer status; 2) to identify issues for consideration when implementing CYP2D6 genotyping in clinical trials; and 3) to outline the advantages and disadvantages of CYP2D6 genotyping compared with phenotyping. DNA samples obtained from 558 previously phenotyped individuals were blindly genotyped at the CYP2D6 locus, and the genotype-phenotype correlation was then determined. The CYP2D6 genotyping methodology successfully predicted all but 1 of the 46 poor metabolizer subjects, and it was determined that this 1 individual had a novel (presumably inactive) mutation within the coding region. In addition, we identified 2 subjects with CYP2D6 genotypes indicative of poor metabolizers who had extensive metabolizer phenotypes as determined by dextromethorphan/dextrorphan ratios. This finding suggests that traditional phenotyping methods do not always offer 100% specificity. Our results suggest that CYP2D6 genotyping is a valid alternative to traditional phenotyping in a clinical trial setting, and in some cases may be better. We also discuss some of the issues and considerations related to the use of genotyping in clinical trials and medical practice.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Debrisoquin/metabolism , Dextromethorphan/metabolism , Genotype , Humans , Phenotype , Polymerase Chain Reaction , Polymorphism, Genetic
19.
J Pharmacol Exp Ther ; 283(1): 46-58, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9336307

ABSTRACT

We describe a comprehensive retrospective analysis in which the abilities of several methods by which human pharmacokinetic parameters are predicted from preclinical pharmacokinetic data and/or in vitro metabolism data were assessed. The prediction methods examined included both methods from the scientific literature as well as some described in this report for the first time. Four methods were examined for their ability to predict human volume of distribution. Three were highly predictive, yielding, on average, predictions that were within 60% to 90% of actual values. Twelve methods were assessed for their utility in predicting clearance. The most successful allometric scaling method yielded clearance predictions that were, on average, within 80% of actual values. The best methods in which in vitro metabolism data from human liver microsomes were scaled to in vivo clearance values yielded predicted clearance values that were, on average, within 70% to 80% of actual values. Human t1/2 was predicted by combining predictions of human volume of distribution and clearance. The best t1/2 prediction methods successfully assigned compounds to appropriate dosing regimen categories (e.g., once daily, twice daily and so forth) 70% to 80% of the time. In addition, correlations between human t1/2 and t1/2 values from preclinical species were also generally successful (72-87%) when used to predict human dosing regimens. In summary, this retrospective analysis has identified several approaches by which human pharmacokinetic data can be predicted from preclinical data. Such approaches should find utility in the drug discovery and development processes in the identification and selection of compounds that will possess appropriate pharmacokinetic characteristics in humans for progression to clinical trials.


Subject(s)
Pharmacokinetics , Animals , Biological Availability , Half-Life , Humans , Metabolic Clearance Rate , Retrospective Studies
20.
J Pharmacol Exp Ther ; 272(2): 953-62, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7853213

ABSTRACT

A novel method is described for quantitative whole-body autoradioluminography using [14C]-radioactive standards prepared from rat red blood cells. MicroComputer Imaging Device model 2 (MCID) and ImageQuant (IQ) imaging systems were evaluated for imaging performance and autoradioluminography quantitation. Weighted linear regression analysis resulted in linearity over five orders of magnitude with a lower limit of quantitation of 2.7 nCi/g. Using IQ, 16 days were necessary for image analysis and data processing of 30 whole-body cryosections and 1080 standards. MCID reduced the image and data processing of the same cryosections and standards to only 4 days. Embedding a series of radioactive standards with each specimen in the same carboxymethyl cellulose block provided an effective method of assessing intrasection and intersection variations in thickness of whole-body cryosections. These results demonstrated that autoradioluminography provided a sensitive, accurate, precise and reproducible method for the quantitative measurement of the tissue distribution of [14C]-radiolabeled xenobiotics in whole-body cryosections. Evaluating the biodistribution of [14C]-xenobiotics by autoradioluminography, not only provides pharmacokinetic data required for predicting the potential tissue deposition of an absorbed dose of radioactivity in man, but also allows for visual and quantitative evaluation of radioactivity in small anatomical structures that otherwise could not be detected or measured by conventional tissue combustion technology.


Subject(s)
Cryoultramicrotomy , Xenobiotics/pharmacokinetics , Animals , Autoradiography , Carbon Radioisotopes , Image Processing, Computer-Assisted , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL