Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Clin Pharmacol Ther ; 115(5): 1007-1014, 2024 May.
Article in English | MEDLINE | ID: mdl-38073049

ABSTRACT

A model-based meta-analysis (MBMA) was conducted to compare the efficacy of bimekizumab with other psoriatic arthritis (PsA) treatment regimens using ≥ 20%/50%/70% improvements in American College of Rheumatology (ACR) criteria (ACR20/50/70) for patients with PsA. Forty-nine trials of 16 drugs were identified in the literature, comprising 21,340 patients. Trial-level covariates, including prior biologic use, concomitant methotrexate use, time since diagnosis, trial completion year, and active comparator were considered for exploratory models. The final model was selected using leave-one-out cross-validation (LOO CV) to assess predictive performance based on prespecified criteria. LOO CV was conducted for 15 trials; the final model demonstrated that 91.5% (952/1,040) of the observed treatment differences, and 96.1% of the observed ACR20/50/70 response rates were within the 95% prediction interval (PI). Median ACR50 response rates (95% PI) at week 16 in biologic-naïve patients were predicted to be 44% (40-49%) for bimekizumab 160 mg, among the highest of all treatments analyzed. Response rates for secukinumab 150 mg and risankizumab 150 mg were 28% (25-32%) and 27% (24-31%), respectively. The MBMA was also used to predict the probability of success (PoS) of potential head-to-head trials using ACR50 response as the end point with varying sample sizes: vs. secukinumab 150 mg, the PoS for bimekizumab 160 mg was 62% (N = 200) and 90% (N = 400). Versus risankizumab 150 mg, the PoS for bimekizumab 160 mg was 68% (N = 200) and 94% (N = 400). In summary, a predictive MBMA described ACR20/50/70 outcomes in PsA, allowing accurate and precise treatment comparisons and robust PoS calculations.


Subject(s)
Antibodies, Monoclonal, Humanized , Antirheumatic Agents , Arthritis, Psoriatic , Biological Products , Rheumatology , Humans , Arthritis, Psoriatic/diagnosis , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/chemically induced , Antirheumatic Agents/therapeutic use , Biological Products/therapeutic use , Treatment Outcome
2.
Clin Pharmacol Ther ; 112(6): 1291-1302, 2022 12.
Article in English | MEDLINE | ID: mdl-36104012

ABSTRACT

Myostatin, a negative regulator of skeletal muscle growth, is a therapeutic target in muscle-wasting diseases. Domagrozumab, a humanized recombinant monoclonal antibody, binds myostatin and inhibits activity. Domagrozumab was investigated in a phase II trial (NCT02310763) as a potential treatment for boys with Duchenne muscular dystrophy (DMD). Pharmacokinetic/pharmacodynamic (PK/PD) modeling is vital in clinical trial design, particularly for determining dosing regimens in pediatric populations. This analysis sought to establish the PK/PD relationship between free domagrozumab and total myostatin concentrations in pediatric patients with DMD using a prior semimechanistic model developed from a phase I study in healthy adult volunteers (NCT01616277) and following inclusion of phase II data. The refined model was developed using a multiple-step approach comprising structural, random effects, and covariate model development; assessment of model adequacy (goodness-of-fit); and predictive performance. Differences in PKs/PDs between healthy adult volunteers and pediatric patients with DMD were quantitatively accounted for and evaluated by predicting myostatin coverage (the percentage of myostatin bound by domagrozumab). The final model parameter estimates and semimechanistic target-mediated drug disposition structure sufficiently described both domagrozumab and myostatin concentrations in pediatric patients with DMD, and most population parameters were comparable with the prior model (in healthy adult volunteers). Predicted myostatin coverage for phase II patients with DMD was consistently > 90%. Baseline serum myostatin was ~ 65% lower than in healthy adult volunteers. This study provides insights into the regulation of myostatin in healthy adults and pediatric patients with DMD. Clinicaltrials.gov identifiers: NCT01616277 and NCT02310763.


Subject(s)
Muscular Dystrophy, Duchenne , Humans , Child , Adult , Male , Muscular Dystrophy, Duchenne/drug therapy , Myostatin/metabolism , Myostatin/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Healthy Volunteers , Muscle, Skeletal/metabolism
3.
Gene Ther ; 29(10-11): 608-615, 2022 11.
Article in English | MEDLINE | ID: mdl-34737451

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, degenerative muscle disorder caused by mutations in the DMD gene, leading to severe reduction or absence of the protein dystrophin. Gene therapy strategies that aim to increase expression of a functional dystrophin protein (mini-dystrophin) are under investigation. The ability to accurately quantify dystrophin/mini-dystrophin is essential in assessing the level of gene transduction. We demonstrated the validation and application of a novel peptide immunoaffinity liquid chromatography-tandem mass spectrometry (IA-LC-MS/MS) assay. Data showed that dystrophin expression in Becker muscular dystrophy and DMD tissues, normalized against the mean of non-dystrophic control tissues (n = 20), was 4-84.5% (mean 32%, n = 20) and 0.4-24.1% (mean 5%, n = 20), respectively. In a DMD rat model, biceps femoris tissue from dystrophin-deficient rats treated with AAV9.hCK.Hopti-Dys3978.spA, an adeno-associated virus vector containing a mini-dystrophin transgene, showed a dose-dependent increase in mini-dystrophin expression at 6 months post-dose, exceeding wildtype dystrophin levels at high doses. Validation data showed that inter- and intra-assay precision were ≤20% (≤25% at the lower limit of quantification [LLOQ]) and inter- and intra-run relative error was within ±20% (±25% at LLOQ). IA-LC-MS/MS accurately quantifies dystrophin/mini-dystrophin in human and preclinical species with sufficient sensitivity for immediate application in preclinical/clinical trials.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Humans , Rats , Animals , Dystrophin/genetics , Dystrophin/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Chromatography, Liquid , Tandem Mass Spectrometry , Muscle, Skeletal/metabolism , Genetic Therapy/methods
4.
Br J Anaesth ; 123(2): e194-e203, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30915991

ABSTRACT

BACKGROUND: This study investigated the analgesic effects of two doses (15 and 65 mg) of PF-06372865, a novel α2/α3/α5 gamma-aminobutyric acid A (GABAA) subunit selective partial positive allosteric modulator (PAM), compared with placebo and pregabalin (300 mg) as a positive control. METHODS: We performed a randomised placebo-controlled crossover study (NCT02238717) in 20 healthy subjects, using a battery of pain tasks (electrical, pressure, heat, cold and inflammatory pain, including a paradigm of conditioned pain modulation). Pharmacodynamic measurements were performed at baseline and up to 10 h after dose. RESULTS: A dose of 15 mg PF-06372865 increased pain tolerance thresholds (PTTs) for pressure pain at a ratio of 1.11 (90% confidence interval [CI]: 1.02, 1.22) compared with placebo. A dose of 65 mg PF-06372865 led to an increase in PTT for the cold pressor at a ratio of 1.17 (90% CI: 1.03, 1.32), and pressure pain task: 1.11 (90% CI: 1.01, 1.21). Pregabalin showed an increase in PTT for pressure pain at a ratio of 1.15 (95% CI: 1.06, 1.26) and cold pressor task: 1.31 (90% CI: 1.16, 1.48). CONCLUSION: We conclude that PF-06372865 has analgesic potential at doses that do not induce significant sedation or other intolerable adverse events limiting its clinical use. In addition, the present study established the potential role for this battery of pain tasks as a tool in the development of analgesics with a novel mechanism of action, for the treatment of various pain states including neuropathic pain and to establish proof-of-concept. CLINICAL TRIALS REGISTRATION: NCT0223871.


Subject(s)
Analgesics/pharmacology , Imidazoles/pharmacology , Pain/drug therapy , Pyridazines/pharmacology , Adult , Analgesics/therapeutic use , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Imidazoles/therapeutic use , Male , Pyridazines/therapeutic use
5.
Eur J Pain ; 23(6): 1129-1140, 2019 07.
Article in English | MEDLINE | ID: mdl-30793411

ABSTRACT

BACKGROUND: Although reproducibility is considered essential for any method used in scientific research, it is investigated only rarely; thus, strikingly little has been published regarding the reproducibility of evoked pain models involving human subjects. Here, we studied the reproducibility of a battery of evoked pain models for demonstrating the analgesic effects of two analgesic compounds. METHODS: A total of 81 healthy subjects participated in four studies involving a battery of evoked pain tests in which mechanical, thermal and electrical stimuli were used to measure pain detection and tolerance thresholds. Pharmacodynamic outcome variables were analysed using a mixed model analysis of variance, and a coefficient of variation was calculated by dividing the standard deviation by the least squares means. RESULTS: A total of 76 subjects completed the studies. After being administered pregabalin, the subjects' pain tolerance thresholds in the cold pressor and pressure stimulation tests were significantly increased compared to the placebo group. Moreover, the heat pain detection threshold in UVB-irradiated skin was significantly increased in subjects who were administered ibuprofen compared to the placebo group. Variation among all evoked pain tests ranged from 2.2% to 30.6%. CONCLUSIONS: Four studies using a similar design showed reproducibility with respect to the included evoked pain models. The relatively high consistency and reproducibility of two analgesics at doses known to be effective in treating clinically relevant pain supports the validity of using this pain test battery to investigate the analgesic activity and determine the active dosage of putative analgesic compounds in early clinical development. SIGNIFICANCE: The consistency and reproducibility of measuring the profile of an analgesic at clinically relevant doses illustrates that this pain test battery is a valid tool for demonstrating the analgesic activity of a test compound and for determining the optimal active dose in early clinical drug development.


Subject(s)
Analgesics/therapeutic use , Pain Measurement/methods , Pain/drug therapy , Adult , Cross-Over Studies , Double-Blind Method , Drug Tolerance , Female , Healthy Volunteers , Humans , Ibuprofen/pharmacology , Male , Middle Aged , Pain Threshold/drug effects , Pregabalin/pharmacology , Reproducibility of Results , Skin/drug effects , Skin/radiation effects
6.
Pharmacol Res Perspect ; 7(1): e00467, 2019 02.
Article in English | MEDLINE | ID: mdl-30784208

ABSTRACT

During a randomized Phase 1 clinical trial the drug candidate, PF-04895162 (ICA-105665), caused transaminase elevations (≥grade 1) in six of eight healthy subjects treated at 300 mg twice daily for 2-weeks (NCT01691274). This was unexpected since studies in rats (<6 months) and cynomolgus monkeys (<9 months) treated up to 100 mg/kg/day did not identify the liver as a target organ. Mechanistic studies showed PF-04895162 had low cytotoxic potential in human hepatocytes, but inhibited liver mitochondrial function and bile salt export protein (BSEP) transport. Clinical relevance of these postulated mechanisms of liver injury was explored in three treated subjects that consented to analysis of residual pharmacokinetic plasma samples. Compared to a nonresponder, two subjects with transaminase elevations displayed higher levels of miRNA122 and total/conjugated bile acid species, whereas one demonstrated impaired postprandial clearance of systemic bile acids. Elevated taurine and glycine conjugated to unconjugated bile acid ratios were observed in two subjects, one before the onset of elevated transaminases. Based on the affinity of conjugated bile acid species for transport by BSEP, the profile of plasma conjugated/unconjugated bile acid species was consistent with inhibition of BSEP. These data collectively suggest that the human liver injury by PF-04895162 was due to alterations in bile acid handling driven by dual BSEP/mitochondrial inhibition, two important risk factors associated with drug-induced liver injury in humans. Alterations in systemic bile acid composition were more important than total bile acids in the manifestation of clinical liver injury and may be a very early biomarker of BSEP inhibition.


Subject(s)
Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Adult , Animals , Biological Transport/drug effects , Chemical and Drug Induced Liver Injury/physiopathology , Double-Blind Method , Hepatocytes/metabolism , Homeostasis , Humans , Macaca fascicularis , Male , Mitochondria, Liver/metabolism , Rats , Risk Factors , Species Specificity , Transaminases/metabolism , Young Adult
7.
J Clin Psychopharmacol ; 39(1): 20-27, 2019.
Article in English | MEDLINE | ID: mdl-30531477

ABSTRACT

BACKGROUND: Generalized anxiety disorder (GAD) is a common psychiatric disorder, but many patients experience only partial relief of symptoms with existing therapies. Benzodiazepines are effective in many cases but are limited by a number of significant adverse effects. PF-06372865 is a subtype-selective gamma-aminobutyric acid A (GABAA)-positive allosteric modulator lacking in functional activity at alpha 1-containing receptors that are believed to mediate many of these adverse effects. METHODS: PF-06372865 was evaluated as an adjunct to current GAD treatment in a double-blind, placebo-controlled, sequential parallel comparison study in patients with GAD who showed an incomplete response to current standard-of-care pharmacotherapy. A total of 90 subjects (of the planned 384) were randomized into the study before the decision to terminate the study. Two doses of PF-06372865 (2.5 mg twice daily and 7.5 mg twice daily) were compared with placebo. RESULTS: Neither dose of PF-06372865 differentiated from placebo on week 4 Hamilton Anxiety Inventory total (primary end point) or on the Sheehan Disability Scale total score (secondary end point). Adverse events including dizziness, headache, and somnolence were observed, and the 7.5 mg dose demonstrated some impairment on the Digit Symbol Substitution test and the Epworth Sleepiness Scale relative to placebo and the 2.5 mg dose. CONCLUSIONS: Factors contributing to the negative results include the limited sample size and failure to explore a broader range of doses.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/drug therapy , Imidazoles/therapeutic use , Pyridazines/therapeutic use , Adolescent , Adult , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/blood , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Imidazoles/adverse effects , Imidazoles/blood , Male , Middle Aged , Outpatients , Pyridazines/adverse effects , Pyridazines/blood , Standard of Care , Treatment Outcome , Withholding Treatment , Young Adult
8.
Front Med (Lausanne) ; 5: 160, 2018.
Article in English | MEDLINE | ID: mdl-29904633

ABSTRACT

Gout is the most common form of inflammatory arthritis and is a multifactorial disease typically characterized by hyperuricemia and monosodium urate crystal deposition predominantly in, but not limited to, the joints and the urinary tract. The prevalence of gout and hyperuricemia has increased in developed countries over the past two decades and research into the area has become progressively more active. We review the current field of knowledge with emphasis on active areas of hyperuricemia research including the underlying physiology, genetics and epidemiology, with a focus on studies which suggest association of hyperuricemia with common comorbidities including cardiovascular disease, renal insufficiency, metabolic syndrome and diabetes. Finally, we discuss current therapies and emerging drug discovery efforts aimed at delivering an optimized clinical treatment strategy.

9.
Pain ; 159(9): 1742-1751, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29787472

ABSTRACT

The effect of PF-06372865, a subtype-selective positive allosteric modulator of the γ-aminobutyric acid type A (GABAA) receptor, on chronic low back pain was investigated in a randomised, placebo- and active-controlled phase 2 clinical trial. The parallel treatment group trial consisted of a 1-week single-blind placebo run in the phase, followed by 4-week double-blind treatment. Patients were randomised to receive either PF-06372865, naproxen, or placebo twice a day for 4 weeks. The primary end point was the numerical rating score of low back pain intensity after 4 weeks of active treatment. Secondary end points included the Roland Morris Disability Questionnaire and the Hopkins Verbal Learning Test-Revised. The trial had predefined decision rules based on the probability that PF-06372865 was better than placebo. The study was stopped at the interim analysis for futility. At this time, a total of 222 patients were randomised and the mean PF-06372865 4-week response on the low back pain intensity was 0.16 units higher (worse) than placebo (90% confidence interval -0.28 to 0.60). There were small, statistically significant reductions in the delayed recall test score with PF-06372865, as measured by Hopkins Verbal Learning Test-Revised. The effects of naproxen were in line with expectations. PF-06372865 was well tolerated. The most common treatment-related adverse events in the PF-06372865 arm were somnolence (5 mild and 4 moderate), dizziness (2 mild and 3 moderate), and nausea (2 mild). Although the reason for the lack of analgesic effect is not completely clear, it may be a result of not achieving sufficient receptor occupancy to drive efficacy.


Subject(s)
Chronic Pain/drug therapy , GABA Modulators/therapeutic use , Low Back Pain/drug therapy , Receptors, GABA-A , Adolescent , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Double-Blind Method , Female , Humans , Male , Middle Aged , Naproxen/therapeutic use , Pain Measurement , Treatment Outcome , Young Adult
10.
Br J Pharmacol ; 175(4): 708-725, 2018 02.
Article in English | MEDLINE | ID: mdl-29214652

ABSTRACT

BACKGROUND AND PURPOSE: Benzodiazepines, non-selective positive allosteric modulators (PAMs) of GABAA receptors, have significant side effects that limit their clinical utility. As many of these side effects are mediated by the α1 subunit, there has been a concerted effort to develop α2/3 subtype-selective PAMs. EXPERIMENTAL APPROACH: In vitro screening assays were used to identify molecules with functional selectivity for receptors containing α2/3 subunits over those containing α1 subunits. In vivo receptor occupancy (RO) was conducted, prior to confirmation of in vivo α2/3 and α1 pharmacology through quantitative EEG (qEEG) beta frequency and zolpidem drug discrimination in rats respectively. PF-06372865 was then progressed to Phase 1 clinical trials. KEY RESULTS: PF-06372865 exhibited functional selectivity for those receptors containing α2/3/5 subunits, with significant positive allosteric modulation (90-140%) but negligible activity (≤20%) at GABAA receptors containing α1 subunits. PF-06372865 exhibited concentration-dependent occupancy of GABAA receptors in preclinical species. There was an occupancy-dependent increase in qEEG beta frequency and no generalization to a GABAA α1 cue in the drug-discrimination assay, clearly demonstrating the lack of modulation at the GABAA receptors containing an α1 subtype. In a Phase 1 single ascending dose study in healthy volunteers, evaluation of the pharmacodynamics of PF-06372865 demonstrated a robust increase in saccadic peak velocity (a marker of α2/3 pharmacology), increases in beta frequency qEEG and a slight saturating increase in body sway. CONCLUSIONS AND IMPLICATIONS: PF-06372865 has a unique clinical pharmacology profile and a highly predictive translational data package from preclinical species to the clinical setting.


Subject(s)
GABA Modulators/pharmacology , Receptors, GABA-A/physiology , Translational Research, Biomedical/methods , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , GABA Modulators/chemistry , HEK293 Cells , Humans , Male , Positron-Emission Tomography/methods , Rats , Rats, Sprague-Dawley
11.
Br J Clin Pharmacol ; 84(2): 301-309, 2018 02.
Article in English | MEDLINE | ID: mdl-29178434

ABSTRACT

AIM: Inhibitors of nerve growth factor (NGF) reduce pain in several chronic pain indications. NGF signals through tyrosine kinase receptors of the tropomyosin-related kinase (Trk) family and the unrelated p75 receptor. PF-06273340 is a small molecule inhibitor of Trks A, B and C that reduces pain in nonclinical models, and the present study aimed to investigate the pharmacodynamics of this first-in-class molecule in humans. METHODS: A randomized, double-blind, single-dose, placebo- and active-controlled five-period crossover study was conducted in healthy human subjects (NCT02260947). Subjects received five treatments: PF-06273340 50 mg, PF-06273340 400 mg, pregabalin 300 mg, ibuprofen 600 mg and placebo. The five primary endpoints were the pain detection threshold for the thermal pain tests and the pain tolerance threshold for the cold pressor, electrical stair and pressure pain tests. The trial had predefined decision rules based on 95% confidence that the PF-06273340 effect was better than that of placebo. RESULTS: Twenty subjects entered the study, with 18 completing all five periods. The high dose of PF-06273340 met the decision rules on the ultraviolet (UV) B skin thermal pain endpoint [least squares (LS) mean vs. placebo: 1.13, 95% confidence interval: 0.64-1.61], but not on the other four primary endpoints. The low dose did not meet the decision criteria for any of the five primary endpoints. Pregabalin (cold pressor and electrical stair tests) and ibuprofen (UVB thermal pain) showed significant analgesic effects on expected endpoints. CONCLUSIONS: The study demonstrated, for the first time, the translation of nonclinical effects into man in an inflammatory pain analgesic pharmacodynamic endpoint using a pan-Trk inhibitor.


Subject(s)
Analgesics/therapeutic use , Hyperalgesia/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Receptor, trkA/antagonists & inhibitors , Receptor, trkB/antagonists & inhibitors , Receptor, trkC/antagonists & inhibitors , Adolescent , Adult , Analgesics/administration & dosage , Analgesics/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Humans , Hyperalgesia/enzymology , Male , Middle Aged , Pain Threshold/drug effects , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Young Adult
12.
Clin Rheumatol ; 35(8): 2045-2051, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27094945

ABSTRACT

The objective of these clinical studies was to assess the safety and urate lowering activity of a novel urate transporter 1 (URAT1)/ xanthine oxidase (XO) inhibitor PF-06743649 in healthy subjects and gout patients. Escalating doses of PF-06743649 or placebo were given to healthy young subjects, healthy elderly subjects and gout patients. Serum uric acid (sUA) and urinary pharmacodynamic markers were assayed, and safety was assessed by collection of adverse events and assessment of safety labs, ECGs and vital signs. Administration of PF-06743649 led to rapid decrease in sUA in all cohorts; in gout patients, a change from baseline of 69 % was observed for the 40 mg dose. Urinary and serum biomarkers were consistent with inhibition of both URAT1 and XO. Although dosing was otherwise well tolerated, two subjects experienced serious adverse events of acute kidney injury. Both subjects exhibited increased serum creatinine and blood urea nitrogen in the first 3 days post first dose and were hospitalised. One subject exhibited oliguria for the first 24 h. Both subjects made a complete recovery with minimal intervention. PF-06743649 was effective at rapidly lowering sUA, but further development was terminated for an identified renal safety risk.


Subject(s)
Acute Kidney Injury/chemically induced , Gout Suppressants/adverse effects , Gout Suppressants/pharmacokinetics , Gout/drug therapy , Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Xanthine Oxidase/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Double-Blind Method , Female , Humans , Hyperuricemia/blood , Male , Middle Aged , United States , Uric Acid/blood , Young Adult
13.
Interface Focus ; 3(2): 20120071, 2013 Apr 06.
Article in English | MEDLINE | ID: mdl-24427523

ABSTRACT

The nerve growth factor (NGF) pathway is of great interest as a potential source of drug targets, for example in the management of certain types of pain. However, selecting targets from this pathway either by intuition or by non-contextual measures is likely to be challenging. An alternative approach is to construct a mathematical model of the system and via sensitivity analysis rank order the targets in the known pathway, with respect to an endpoint such as the diphosphorylated extracellular signal-regulated kinase concentration in the nucleus. Using the published literature, a model was created and, via sensitivity analysis, it was concluded that, after NGF itself, tropomyosin receptor kinase A (TrkA) was one of the most sensitive druggable targets. This initial model was subsequently used to develop a further model incorporating physiological and pharmacological parameters. This allowed the exploration of the characteristics required for a successful hypothetical TrkA inhibitor. Using these systems models, we were able to identify candidates for the optimal drug targets in the known pathway. These conclusions were consistent with clinical and human genetic data. We also found that incorporating appropriate physiological context was essential to drawing accurate conclusions about important parameters such as the drug dose required to give pathway inhibition. Furthermore, the importance of the concentration of key reactants such as TrkA kinase means that appropriate contextual data are required before clear conclusions can be drawn. Such models could be of great utility in selecting optimal targets and in the clinical evaluation of novel drugs.

14.
Lancet Neurol ; 11(2): 131-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22226929

ABSTRACT

BACKGROUND: Monoclonal antibody therapy against α4ß-integrin is efficacious in patients with multiple sclerosis (MS) with some safety concerns. We assessed the safety and efficacy of firategrast, a small oral anti-α4ß-integrin molecule, in patients with relapsing remitting MS. METHODS: We did a multicentre, phase 2, randomised, double-blind, placebo-controlled, dose-ranging study in participants with clinically definite relapsing-remitting MS. A 24-week treatment period was followed by 12 weeks of core follow-up and 40 weeks of extended follow-up. Participants were randomly assigned, via computer-generated block randomisation in a 1:2:2:2 ratio, to receive one of four treatments twice a day: firategrast 150 mg, firategrast 600 mg, or firategrast 900 mg (women) or 1200 mg (men), or placebo. Brain scans were obtained at 4-week intervals to the end of core follow-up. The primary outcome was cumulative number of new gadolinium-enhancing brain lesions during the treatment phase and was analysed using a generalised linear model with an underlying negative binomial distribution, adjusted for sex, baseline number of new gadolinium-enhancing lesions, and country. This study is registered with ClinicalTrials.gov, NCT00395317. FINDINGS: Of 343 individuals enrolled, 49 received firategrast 150 mg, 95 received firategrast 600 mg, 100 received firategrast 900 mg or 1200 mg, and 99 received placebo. A 49% reduction (95% CI 21·2-67·6; p=0·0026) in the cumulative number of new gadolinium-enhancing lesions was seen for the 900 mg or 1200 mg firategrast group (n=92, mean number of lesions 2·69 [SE 1·18]) versus the placebo group (90, 5·31 [1·18]). In the 600 mg group (86, 4·12 [SE 1·19]), a non-significant 22% reduction (95% CI -21·3 to 49·7; p=0·2657) occurred in mean number of new gadolinium-enhanced lesions relative to placebo; for the 150 mg group (47, 9·51 [SE 1·24]), a 79% increase (95% CI 4·1-308·1; p=0·0353) occurred relative to placebo. Firategrast was generally well tolerated at all doses. The frequency of all adverse events was similar across all treatment groups except for an increased rate of urinary tract infections in the high-dose firategrast group. No cases of progressive multifocal leukoencephalopathy or evidence of reactivation of JC virus were identified. INTERPRETATION: This study showed efficacy on imaging endpoints for firategrast at the highest dose tested, and suggests that further investigation of oral short-acting α4ß integrin blockade therapies is warranted. FUNDING: GlaxoSmithKline.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Integrin alpha4beta1/antagonists & inhibitors , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adult , Antibodies, Monoclonal/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Gadolinium , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/pathology , Placebos , Treatment Outcome
15.
Med Biol Eng Comput ; 48(6): 543-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20405230

ABSTRACT

This article presents model predictive controllers (MPCs) and multi-parametric model-based controllers for delivery of anaesthetic agents. The MPC can take into account constraints on drug delivery rates and state of the patient but requires solving an optimization problem at regular time intervals. The multi-parametric controller has all the advantages of the MPC and does not require repetitive solution of optimization problem for its implementation. This is achieved by obtaining the optimal drug delivery rates as a set of explicit functions of the state of the patient. The derivation of the controllers relies on using detailed models of the system. A compartmental model for the delivery of three drugs for anaesthesia is developed. The key feature of this model is that mean arterial pressure, cardiac output and unconsciousness of the patient can be simultaneously regulated. This is achieved by using three drugs: dopamine (DP), sodium nitroprusside (SNP) and isoflurane. A number of dynamic simulation experiments are carried out for the validation of the model. The model is then used for the design of model predictive and multi-parametric controllers, and the performance of the controllers is analyzed.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Drug Delivery Systems/methods , Models, Biological , Anesthetics, Inhalation/pharmacology , Blood Pressure/drug effects , Cardiotonic Agents/pharmacology , Dopamine/pharmacology , Drug Monitoring/methods , Humans , Isoflurane/administration & dosage , Isoflurane/pharmacology , Nitroprusside/pharmacology , Vasodilator Agents/pharmacology
16.
Med Biol Eng Comput ; 47(3): 343-52, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214613

ABSTRACT

For people with type 1 diabetes, automatic controllers aim to maintain the blood glucose concentration within the desired range of 60-120 mg/dL by infusing the appropriate amount of insulin in the presence of meal and exercise disturbances. Blood glucose concentration outside the desired range can be harmful to an individual's health but concentration below 60 mg/dL, a state known as hypoglycemia, is considered to be more harmful than the concentration above 120 mg/dL, a state known as hyperglycemia. In this paper, two techniques to address this issue within a multi-parametric model based control framework are presented. The first technique introduces asymmetry into the objective function to penalize the deviation towards hypoglycemia more than the deviation towards hyperglycemia. The second technique is based upon placing higher priority on satisfaction of constraints on hypoglycemia than on satisfaction of constraints on hyperglycemia. The performance of both the control techniques is analyzed and compared in the presence of disturbances.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Models, Biological , Diabetes Mellitus, Type 1/drug therapy , Drug Administration Schedule , Drug Delivery Systems , Humans , Hyperglycemia/prevention & control , Hypoglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage
17.
IEEE Trans Biomed Eng ; 53(8): 1478-91, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16916082

ABSTRACT

An advanced model-based control technique for regulating the blood glucose for patients with Type 1 diabetes is presented. The optimal insulin delivery rate is obtained off-line as an explicit function of the current blood glucose concentration of the patient by using novel parametric programming algorithms, developed at Imperial College London. The implementation of the optimal insulin delivery rate, therefore, requires simple function evaluation and minimal on-line computations. The proposed framework also addresses the uncertainty in the model due to interpatient and intrapatient variability by identifying the model parameters which ensure that a feasible control law can be obtained. The developments reported in this paper are expected to simplify the insulin delivery mechanism, thereby enhancing the quality of life of the patient.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Drug Therapy, Computer-Assisted/methods , Insulin/administration & dosage , Insulin/blood , Models, Biological , Blood Glucose/analysis , Computer Simulation , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/administration & dosage , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...