Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 197(12): 4593-4602, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27849166

RESUMO

FR104 is a monovalent pegylated Fab' Ab, antagonist of CD28, under development for treatment of transplant rejection and autoimmune diseases. In contrast to CD80/86 antagonists (CTLA4-Ig), FR104 selectively blunts CD28 costimulation while sparing CTLA-4 and PD-L1 coinhibitory signals. In the present work, FR104 has been evaluated in a first-in-human study to evaluate the safety, pharmacokinetics, pharmacodynamics, and potency of i.v. administrations in healthy subjects. Sixty-four subjects were randomly assigned to four single ascending dose groups, two double dose groups and four single ascending dose groups challenged with keyhole limpet hemocyanin. Subjects were followed up over a maximum of 113 d. Overall, the pharmacokinetics of FR104 after a single and double infusions was approximately linear at doses ≥0.200 mg/kg. CD28 receptor occupancy by FR104 was saturated at the first sampling time point (0.5 h) at doses above 0.02 mg/kg and returned to 50% in a dose-dependent manner, by day 15 (0.020 mg/kg) to 85 (1.500 mg/kg). FR104 was well tolerated, with no evidence of cytokine-release syndrome and no impact on blood lymphocyte subsets. Inhibition of anti-keyhole limpet hemocyanin Ab response was dose-dependent in FR104 recipients and was already apparent at a dose of 0.02 mg/kg. Abs to FR104 were detected in 22/46 (48%) of FR104 recipients and only 1/46 (2.2%) was detected during drug exposure. In conclusion, selective blockade of CD28 with FR104 was safe and well tolerated at the doses tested. The observed immunosuppressive activity indicated that FR104 has potential to show clinical activity in the treatment of immune-mediated diseases.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doenças Autoimunes/terapia , Rejeição de Enxerto/prevenção & controle , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Transplante de Órgãos , Administração Intravenosa , Adulto , Anticorpos Monoclonais/farmacologia , Doenças Autoimunes/imunologia , Antígenos CD28/antagonistas & inibidores , Antígenos CD28/imunologia , Protocolos Clínicos , Estudos de Coortes , Feminino , Seguimentos , Rejeição de Enxerto/imunologia , Voluntários Saudáveis , Humanos , Imunidade Humoral/efeitos dos fármacos , Imunossupressores , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade
3.
Toxicol Sci ; 200(1): 11-30, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38588579

RESUMO

Drug hypersensitivity reactions (DHRs) are a type of adverse drug reaction that can occur with different classes of drugs and affect multiple organ systems and patient populations. DHRs can be classified as allergic or non-allergic based on the cellular mechanisms involved. Whereas nonallergic reactions rely mainly on the innate immune system, allergic reactions involve the generation of an adaptive immune response. Consequently, drug allergies are DHRs for which an immunological mechanism, with antibody and/or T cell, is demonstrated. Despite decades of research, methods to predict the potential for a new chemical entity to cause DHRs or to correctly attribute DHRs to a specific mechanism and a specific molecule are not well-established. This review will focus on allergic reactions induced by systemically administered low-molecular weight drugs with an emphasis on drug- and patient-specific factors that could influence the development of DHRs. Strategies for predicting and diagnosing DHRs, including potential tools based on the current state of the science, will also be discussed.


Assuntos
Hipersensibilidade a Drogas , Humanos , Hipersensibilidade a Drogas/diagnóstico , Hipersensibilidade a Drogas/imunologia
4.
JCI Insight ; 7(20)2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36278483

RESUMO

BACKGROUNDAntigen-specific regulation of autoimmune disease is a major goal. In seropositive rheumatoid arthritis (RA), T cell help to autoreactive B cells matures the citrullinated (Cit) antigen-specific immune response, generating RA-specific V domain glycosylated anti-Cit protein antibodies (ACPA VDG) before arthritis onset. Low or escalating antigen administration under "sub-immunogenic" conditions favors tolerance. We explored safety, pharmacokinetics, and immunological and clinical effects of s.c. DEN-181, comprising liposomes encapsulating self-peptide collagen II259-273 (CII) and NF-κB inhibitor 1,25-dihydroxycholecalciferol.METHODSA double-blind, placebo-controlled, exploratory, single-ascending-dose, phase I trial assessed the impact of low, medium, and high DEN-181 doses on peripheral blood CII-specific and bystander Cit64vimentin59-71-specific (Cit-Vim-specific) autoreactive T cell responses, cytokines, and ACPA in 17 HLA-DRB1*04:01+ or *01:01+ ACPA+ RA patients on methotrexate.RESULTSDEN-181 was well tolerated. Relative to placebo and normalized to baseline values, Cit-Vim-specific T cells decreased in patients administered medium and high doses of DEN-181. Relative to placebo, percentage of CII-specific programmed cell death 1+ T cells increased within 28 days of DEN-181. Exploratory analysis in DEN-181-treated patients suggested improved RA disease activity was associated with expansion of CII-specific and Cit-Vim-specific T cells; reduction in ACPA VDG, memory B cells, and inflammatory myeloid populations; and enrichment in CCR7+ and naive T cells. Single-cell sequencing identified T cell transcripts associated with tolerogenic TCR signaling and exhaustion after low or medium doses of DEN-181.CONCLUSIONThe safety and immunomodulatory activity of low/medium DEN-181 doses provide rationale to further assess antigen-specific immunomodulatory therapy in ACPA+ RA.TRIAL REGISTRATIONAnzctr.org.au identifier ACTRN12617001482358, updated September 8, 2022.FUNDINGInnovative Medicines Initiative 2 Joint Undertaking (grant agreement 777357), supported by European Union's Horizon 2020 research and innovation programme and European Federation of Pharmaceutical Industries and Associations; Arthritis Queensland; National Health and Medical Research Council (NHMRC) Senior Research Fellowship; and NHMRC grant 2008287.


Assuntos
Artrite Reumatoide , Calcitriol , Humanos , Lipossomos , Metotrexato , NF-kappa B , Receptores CCR7 , Artrite Reumatoide/tratamento farmacológico , Peptídeos , Imunoterapia , Fatores Imunológicos , Citocinas , Colágeno , Receptores de Antígenos de Linfócitos T
5.
Antimicrob Agents Chemother ; 55(11): 5200-4, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896908

RESUMO

Moxidectin, registered worldwide as a veterinary antiparasitic agent, is currently under development for humans for the treatment of onchocerciasis in collaboration with the World Health Organization. The objective of this study was to assess the pharmacokinetics of moxidectin in healthy lactating women, including the excretion into breast milk. Twelve women, ages 23 to 38 years, weighing 54 to 79 kg, all more than 5 months postpartum, were enrolled, following their plan to wean their infants and provision of informed consent. A single 8-mg, open-label dose was administered orally after consumption of a standard breakfast. Complete milk collection was done for approximately 28 days, and plasma samples were collected for 90 days. Moxidectin concentrations were measured by high-performance liquid chromatography (HPLC) with fluorescence detection, with a validated range of 0.08 to 120 ng/ml. Noncompartmental pharmacokinetic methods were used to find the following results: peak concentration in plasma (C(max)), 87 ± 25 ng/ml; time to C(max) (t(max)), 4.18 ± 1.59 h; terminal-phase elimination half-life (t(1/2)), 832 ± 321 h; total area under the concentration-time curve (AUC), 4,046 ± 1,796 ng · h/ml; apparent oral dose clearance (CL/F), 2.35 ± 1.07 l/h; ratio of CL/F to the terminal-phase disposition rate constant, λ(z) (Vλ(z)/F), 2,526 ± 772 liters; percentage of maternal dose excreted in milk, 0.701 ± 0.299%; absolute amount excreted in milk, 0.056 ± 0.024 mg; relative infant dose, 8.73 ± 3.17% of maternal dose assuming complete absorption; clearance in milk (CL(milk)), 0.016 ± 0.009 liter/h. Nine of 12 subjects reported adverse events, all of which were considered treatment emergent but not drug related and were mostly reported during the long outpatient period 8 to 90 days after dose administration. The most frequently reported adverse events were headache and nausea (n = 4), oropharyngeal pain (n = 2), rhinitis, viral pharyngitis, and viral upper respiratory tract infection (n = 2).


Assuntos
Lactação/metabolismo , Leite Humano/metabolismo , Adulto , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Macrolídeos/metabolismo , Macrolídeos/farmacocinética , Adulto Jovem
6.
J Pharm Biomed Anal ; 41(1): 189-95, 2006 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-16321496

RESUMO

PURPOSE: Measurements of cytokine release in whole blood after ex vivo stimulation are useful in drug development. The components contributing to variation within such assays have not been clearly defined. Therefore, we characterized the sources of variability within an ex vivo stimulation assay for TNF-alpha release. METHOD: Fresh whole blood or mononuclear cells from a cell preparation tube were added to silanized, screw-top tubes with a final concentration of 1 microg/mL lipopolysaccharide (LPS). Each tube was purged with 95% air/5%CO2 and incubated 4 or 6 h at 37 degrees C in a metabolic water bath. Plasma TNF-alpha was next measured in supernatants by immunoassay. Total method variability was assessed in 10 normal donors each drawn in the morning and afternoon over 3 days. Four additional samples were pre-treated with dexamethasone to investigate inhibition of TNF-alpha release. RESULTS: Our analysis indicated precise temperature control, the timing and duration of stimulation, and the surface properties of the stimulation vessel most significantly influenced assay performance. A comparison of multiple anticoagulants indicated that careful consideration should be taken in selecting the optimal anticoagulant. The estimated total assay CV for all anticoagulants tested was less than 33.81%. The analytical variability (stimulation and measurement) was less than 25.88% CV. The one exception was mononuclear cells collected in sodium heparin. The total variability estimate incorporated day-to-day, diurnal, inter-donor, tube-to-tube and immunoassay variability. Using our optimized conditions, TNF-alpha release was inhibited by dexamethasone with a mean IC50 of 33.3 +/- 4.6 nM. CONCLUSIONS: We have described an optimal set of conditions for collection, storage and processing of an ex vivo cytokine stimulation assay. These conditions were selected for operational feasibility, minimal imprecision and elimination of potential confounding factors. The end result is a more robust method that can be applied to clinical drug development.


Assuntos
Técnicas de Química Analítica/métodos , Química Farmacêutica/métodos , Citocinas/química , Anticoagulantes/farmacologia , Citocinas/análise , Citocinas/sangue , Dexametasona/farmacologia , Desenho de Fármacos , Humanos , Imunoensaio/métodos , Concentração Inibidora 50 , Lipopolissacarídeos/metabolismo , Temperatura , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo
7.
Clin Ther ; 38(6): 1417-1434.e2, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27112532

RESUMO

PURPOSE: SBI-087 is a Small Modular Immunopharmaceutical Protein™(SMIP™) drug that binds to CD20 and has been reported to deplete B cells in murine/primate studies. The safety, tolerability and pharmacokinetic/pharmacodynamic properties of SBI-087 were evaluated in patients with rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE). METHODS: Single-dose SBI-087 was evaluated in 2 Phase I, open-label, escalating-dose studies in patients with RA or SLE. The studies included 6 IV/4 SC escalating doses (RA) and 1 IV/4 SC escalating doses (SLE). Escalation was determined by tolerability/rate of B-cell depletion. Serum was collected for analyses of pharmacokinetic and pharmacodynamic (CD19(+) B cells) properties and immunogenicity. Patients were followed until B-cell counts were normalized or stabilized. Safety, tolerability was evaluated from adverse events, physical examinations, vital sign measurements, ECG, and clinical laboratory results. FINDINGS: Sixty patients with RA (IV, 28; SC, 32) and 30 patients with SLE (6 per cohort) were enrolled. Mild to moderate infusion reactions occurred in several patients at the top doses in the RA study despite a pretreatment regimen of IV doses. Unanticipated reactions after SC administration of SBI-087 included fever, chills, and malaise, seen on the day of dosing in the lowest-dose cohorts in both studies. These events were abrogated in subsequent cohorts by a pre/postdose treatment regimen consisting of oral corticosteroids, acetaminophen, and an antihistamine. SBI-087 clearance (IV) ranged from 22 to 229 mL/h; volume of distribution at steady state ranged from 5 to 12 L. Apparent clearance (SC) ranged from 44.7 to 105 mL/h; volume of distribution ranged from 14.3 to 32.1 L. Overall, PK properties were similar at equivalent doses between IV/SC administrations in patients with RA/SLE. Mean t½ (IV) ranged from 2.1 to 10.7 days (less at lower doses). SBI-087 concentration and B-cell depletion were generally dose proportional across IV and SC cohorts. However, the extent of B-cell depletion was less, and rate of repletion was faster, in patients with SLE versus RA. In both studies, B-cell repletion to baseline did not occur in the majority of patients by the end of the observation period. Overall, the prevalence and type of adverse events were similar to those seen with other anti-CD20-depleting agents. IMPLICATIONS: In patients with mild RA/SLE, SBI-087 was well tolerated when administered intravenously or subcutaneously with pre- and posttreatment regimens. B-cell depletion is long lasting, and the duration and extent of depletion may be greater in RA compared with SLE. SBI-087 exhibited slow elimination and low distribution in both populations. Clinicaltrials.gov identifiers: NCT00641225 (RA) and NCT00714116 (SLE).


Assuntos
Antígenos CD20/imunologia , Antirreumáticos/administração & dosagem , Artrite Reumatoide/tratamento farmacológico , Linfócitos B/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Proteínas/administração & dosagem , Administração Intravenosa , Adolescente , Adulto , Idoso , Antirreumáticos/efeitos adversos , Antirreumáticos/farmacocinética , Antirreumáticos/farmacologia , Artrite Reumatoide/imunologia , Linfócitos B/patologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Pessoa de Meia-Idade , Proteínas/efeitos adversos , Proteínas/farmacocinética , Proteínas/farmacologia , Adulto Jovem
8.
J Rheumatol ; 43(12): 2094-2100, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27909139

RESUMO

OBJECTIVE: To evaluate subcutaneous SBI-087 to treat rheumatoid arthritis (RA). METHODS: A total of 210 adult patients with active RA were randomized to receive either 200 mg SBI-087 or placebo (Pbo), according to one of these patterns: SBI/Pbo/Pbo (SBI on Day 1), SBI/SBI/Pbo (SBI days 1 and 15), SBI/Pbo/SBI (SBI days 1 and 84), SBI/SBI/SBI (SBI days 1, 15, and 84), or Pbo/Pbo/Pbo (Pbo all 3 days). All patients were seropositive and taking background methotrexate. The primary endpoint was proportion of patients achieving 20% improvement from baseline at Week 16 by American College of Rheumatology criteria (ACR20). Other outcomes included 28-joint Disease Activity Score (DAS28)-C-reactive protein (CRP), physician's and patient's global assessments of disease activity (PGA and PtGA, respectively) and Health Assessment Questionnaire-Disability Index (HAQ-DI). Peripheral CD19+ B cells were measured by high-sensitivity flow cytometer. Statistical significance was set at 2-sided α 0.10 level. RESULTS: The SBI/SBI/SBI group demonstrated significant improvement in ACR20 and DAS28-CRP from Week 8 onward, sustained improvement in CRP levels from Week 12 onward, and significant improvements in PGA and PtGA in weeks 16 through 24, and in HAQ-DI at Week 24. The SBI/Pbo/Pbo and SBI/SBI/Pbo groups did not meet the primary endpoint but demonstrated improvements in several secondary endpoints. All treatment groups exhibited depletion of peripheral CD19+ B cells throughout the study. Overall, 61.5% of patients receiving SBI-087 and 55.0% of patients receiving Pbo reported adverse events. CONCLUSION: SBI-087 effectively depleted peripheral CD20 B cells and was well tolerated. Improvements were consistently observed in the SBI/SBI/SBI group for the majority of efficacy and quality-of-life outcomes.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Linfócitos B/efeitos dos fármacos , Depleção Linfocítica/métodos , Proteínas/uso terapêutico , Adulto , Idoso , Antirreumáticos/administração & dosagem , Antirreumáticos/efeitos adversos , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Injeções Subcutâneas , Masculino , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Proteínas/administração & dosagem , Proteínas/efeitos adversos , Resultado do Tratamento
9.
Hum Immunol ; 65(3): 248-54, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15041164

RESUMO

We determined the association between clinical outcomes after heart transplantation and gene polymorphism in five cytokines (tumor necrosis factor-alpha, transforming growth factor (TGF)-beta, interleukin-10, interleukin-6, and interferon-gamma) reported to influence expression in vitro. Ninety-five patients were studied. Cytokine genotyping was performed by sequence specific priming polymerase chain reaction. Clinical outcomes studied in the first posttransplant year included: (1) documented viral, bacterial, or fungal infection; (2) cytomegalovirus infection; (3) acute cellular rejection (International Society for Heart and Lung Transplantation > or = grade IIIA); (4) time to first rejection episode; and (5) the development of allograft vasculopathy. Patients with the TGF-beta genotype 10 T/T 25 G/G or 10 T/C 25 G/G had a longer time to first rejection (median time to first rejection episode 321 days) than those with the TGF-beta genotype 10C/C 25 G/C or 10 C/C 25 C/C (median time to first rejection 88 days). There was a trend toward a higher frequency of the tumor necrosis factor-alpha genotype -308 G/A or A/A in patients without infection (19/59, 32%) as compared with patients with infection (5/31, 16%). In both cases, these differences failed to reach significance when adjusted for multiple comparisons. No other significant association was found with clinical outcomes and polymorphisms in the five cytokine genes studied in this population.


Assuntos
Transplante de Coração/imunologia , Interleucinas/genética , Polimorfismo Genético/genética , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética , Adulto , Primers do DNA , Feminino , Rejeição de Enxerto/tratamento farmacológico , Humanos , Imunossupressores/farmacologia , Infecções , Interferon gama/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase
10.
J Clin Pharmacol ; 54(1): 70-4, 2014 01.
Artigo em Inglês | MEDLINE | ID: mdl-24155157

RESUMO

The goal of the this study was to re-evaluate tigecycline bone concentrations in subjects undergoing elective orthopedic surgery, using multiple doses and a more robust bone assay than was used in a previous study. Each subject received three intravenous doses of tigecycline (one 100-mg infusion followed by two 50-mg infusions, each administered over 30 minutes). A single bone sample was collected from each subject at one of the following times: 1, 2, 4, 6, 8, or 12 hours after the third dose. Four blood samples were collected from each subject: before the first dose, before and after the third dose, and within 15 minutes of the collection time of the bone sample. Noncompartmental pharmacokinetic analysis serum and bone area under the curve for the given dose interval (AUCτ ) values were 2,402 ng h/mL and 11,465 ng h/g, and maximum concentration (Cmax ) values were 974 ng/mL and 2,262 ng/g, respectively. The bone to serum ratio calculated using the AUCτ values was 4.77, confirming tigecycline penetration into bone.


Assuntos
Antibacterianos/farmacocinética , Osso e Ossos/metabolismo , Minociclina/análogos & derivados , Procedimentos Ortopédicos , Adulto , Antibacterianos/sangue , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Espectrometria de Massas , Minociclina/sangue , Minociclina/farmacocinética , Tigeciclina
11.
Clin Pharmacol Drug Dev ; 3(2): 151-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27128460

RESUMO

In order to evaluate the potential for CYP3A4 induction by moxidectin, midazolam pharmacokinetic (PK) parameters were compared before and after moxidectin administration. Healthy subjects received a single 8 mg dose of moxidectin and 3 single 7.5 mg doses of midazolam 3 days before, and 7 and 89 days after the moxidectin. Blood samples were taken for 24 hours to measure midazolam and metabolites in plasma, and for 89 days to measure moxidectin in plasma after dose administration. Noncompartmental PK analyses were performed for each analyte. Analysis of variance was performed on log-transformed midazolam parameters with treatment day as a fixed effect. Adverse events were recorded and laboratory tests, physical examinations, pulse oximetry monitoring, vital sign measurement, and electrocardiograms performed. Thirty-nine subjects were enrolled in the study; PK data were available for 37 subjects. Moxidectin PK parameters were similar to previous studies. There were no significant changes in PK for midazolam or its metabolites 7 or 89 days after moxidectin administration. Adverse events were generally mild and there were no relevant changes in safety assessments. Thus, 8 mg moxidectin does not induce CYP3A4 activity and other CYP3A4 substrates are unlikely to be affected by moxidectin co-administration.

12.
Clin Pharmacol Drug Dev ; 1(2): 52-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27121219

RESUMO

Availability of a lower dose tablet would add to the dosing flexibility of currently available 1- and 2-mg sirolimus tablets for optimal concentrations and patient compliance. A randomized, 3-period crossover study was conducted in 30 fasting healthy volunteers (29 men, aged 31 ± 8 years, weight 79 ± 12 kg). Subjects were given 5 mg of sirolimus, either as doses of the 0.5-mg nonshellac-core prototype, 0.5-mg shellac-core prototype, or approved 1-mg tablet. Whole blood samples were collected at selected time points for 144 hours after dosing and analyzed using LC/MS/MS assay. Noncompartmental pharmacokinetic analysis was performed, followed by bioequivalence assessment. Twenty-four subjects completed all dosing periods, and no formulation-associated adverse events were reported. Ratios of maximum plasma concentration (Cmax ), area under the concentration-time curve to the last measured concentration (AUCT ), and area under the concentration-time curve from time 0 to infinity (AUC) for the nonshellac-core prototype compared with the 1-mg tablet were within the 80% to 125% range dictated by bioequivalence conventions. Similar results were observed when comparing the ratios of AUCT and AUC for the shellac-core prototype, while 90% confidence interval of the ratio of Cmax values was 105% to 129%. Within the context of clinical equivalence standards established by a phase 3 study comparing liquid to tablet formulations, it was concluded that both prototypes were clinically bioequivalent to the reference formulation.

13.
Am J Trop Med Hyg ; 86(1): 122-125, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22232462

RESUMO

The objective of this study was to assess the effect of a high-fat meal on the pharmacokinetics of moxidectin. Healthy male subjects were randomized to receive single oral 8 mg doses of moxidectin after an overnight fast or high-fat breakfast. In fasted subjects (N = 27), mean [SD] parameters were C(max): 58.9 [12.5] ng/mL; t(max): 3.7 [1.5] h; area under concentration-time curve (AUC): 3,387 [1,328] ng/h/mL; Vλ(z)/F: 2,829 [1,267] L; CL/F: 2.76 [1.28] L/h; and t(1/2): 784 [347] h. Compared with fasted subjects, fed subjects (N = 27) exhibited a 34% increase in C(max), delay in t(max) to 5.3 [2.1] h, 44% increase in AUC, 40% decrease in Vλ(z)/F, and a 35% decrease in CL/F. There was no significant change in t(1/2). The changes are consistent with an increase in moxidectin bioavailability following administration with food. There were no clinically relevant changes in vital signs, laboratory tests, or electrocardiograms.


Assuntos
Antinematódeos/farmacocinética , Gorduras na Dieta/farmacologia , Interações Alimento-Droga , Administração Oral , Adulto , Antinematódeos/administração & dosagem , Área Sob a Curva , Disponibilidade Biológica , Gorduras na Dieta/administração & dosagem , Jejum , Humanos , Macrolídeos/administração & dosagem , Macrolídeos/farmacocinética , Masculino , Pessoa de Meia-Idade , Adulto Jovem
14.
J Immunotoxicol ; 6(1): 1-10, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19519157

RESUMO

The number of anti-inflammatory and immunomodulatory drugs being developed in the pharmaceutical industry has increased considerably in the past decade. This increase in research and development has been paralleled by questions from both regulatory agencies and industry on how best to assess decreased host resistance to infections or adverse immunostimulation caused by immunomodulatory agents such as anti-cytokine antibodies (e.g., the tumor necrosis factor-alpha inhibitors), anti-adhesion molecule antibodies (e.g., anti-alpha-4 integrin inhibitors) and immunostimulatory molecules (e.g., anti-CD28 antibodies). Although several methods have been developed for nonclinical assessment of immunotoxicity, highly publicized adverse events have brought to light significant gaps in the application of nonclinical immunotoxicity testing in assessing potential risk in humans. Confounding this problem is inconsistent application of immunotoxicology methods for risk assessment within the scientific community, limited understanding of appropriate immunotoxicity testing strategy for immunomodulators and inconsistent testing requests by regulatory agencies. To address these concerns, The Immunotoxicology Technical Committee (ITC) of the International Life Science Institute (ILSI) Health and Environmental Sciences Institute (HESI) organized a workshop on Immunomodulators and Clinical Immunotoxicology in May 2007. The Workshop was convened to identify key gaps in nonclinical and clinical immunotoxicity testing of anti-inflammatory and immunomodulatory agents and to begin to develop consistent approaches for immunotoxicity testing and risk assessment. This paper summarizes the outcome of the HESI ITC Immunomodulators and Clinical Immunotoxicology Workshop. Topics not discussed at the Workshop were outside the scope of this report. Although more work is needed to develop consistent approaches for immunotoxicity assessment of immunomodulators, this Workshop provided the foundation for future discussion.


Assuntos
Ensaios Clínicos como Assunto/tendências , Avaliação Pré-Clínica de Medicamentos/tendências , Fatores Imunológicos/efeitos adversos , Testes de Toxicidade/tendências , Animais , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/toxicidade , Ensaios Clínicos como Assunto/normas , Consenso , Coleta de Dados , Aprovação de Drogas/legislação & jurisprudência , Avaliação Pré-Clínica de Medicamentos/normas , Indústria Farmacêutica/normas , Indústria Farmacêutica/tendências , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Fatores Imunológicos/farmacologia , Fatores Imunológicos/toxicidade , Modelos Animais , Medição de Risco/normas , Medição de Risco/tendências , Testes de Toxicidade/normas
15.
J Infect Dis ; 186(7): 1007-12, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12232842

RESUMO

The intensity of malaria transmission is related to the pattern of malarial disease observed in different regions, but populations may also differ in their underlying predispositions to severe malarial anemia or cerebral malaria. In western Kenya, where severe malarial anemia is much more common than cerebral malaria, the distributions of tumor necrosis factor (TNF)-alpha, interleukin (IL)-10, transforming growth factor (TGF)-beta, IL-6, and interferon (IFN)-gamma alleles were examined in a cohort of young men. The cohort displayed a marked bias toward genotypes associated with low expression of IFN-gamma and IL-6, cytokines that, at high levels, have been implicated in malarial anemia and poor malaria outcomes. By contrast, the frequency of the TNF-alpha -238A allele, which has been associated with severe malarial anemia, was found to be similar to the frequency previously reported in comparison populations in Africa and elsewhere. IFN-gamma and IL-6 genotypes may play roles in the development of severe malaria and could contribute to the relative frequency of severe malarial anemia or cerebral malaria in exposed populations.


Assuntos
Anemia/complicações , Interferon gama/genética , Interleucina-6/genética , Malária Falciparum/complicações , Adolescente , Adulto , Alelos , Anemia/genética , Anemia/imunologia , Criança , Estudos de Coortes , Predisposição Genética para Doença , Humanos , Interleucina-10/genética , Quênia , Malária Falciparum/genética , Malária Falciparum/imunologia , Masculino , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA