Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Mol Ther ; 26(3): 917-931, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29433935

ABSTRACT

Loss of adenosine deaminase activity leads to severe combined immunodeficiency (ADA-SCID); production and function of T, B, and natural killer (NK) cells are impaired. Gene therapy (GT) with an autologous CD34+-enriched cell fraction that contains CD34+ cells transduced with a retroviral vector encoding the human ADA cDNA sequence leads to immune reconstitution in most patients. Here, we report short- and medium-term safety analyses from 18 patients enrolled as part of single-arm, open-label studies or compassionate use programs. Survival was 100% with a median of 6.9 years follow-up (range, 2.3 to 13.4 years). Adverse events were mostly grade 1 or grade 2 and were reported by all 18 patients following GT. Thirty-nine serious adverse events (SAEs) were reported by 15 of 18 patients; no SAEs were considered related to GT. The most common adverse events reported post-GT include upper respiratory tract infection, gastroenteritis, rhinitis, bronchitis, oral candidiasis, cough, neutropenia, diarrhea, and pyrexia. Incidence rates for all of these events were highest during pre-treatment, treatment, and/or 3-month follow-up and then declined over medium-term follow-up. GT did not impact the incidence of neurologic/hearing impairments. No event indicative of leukemic transformation was reported.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Genetic Therapy , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/administration & dosage , Adenosine Deaminase/immunology , Adenosine Deaminase/metabolism , Agammaglobulinemia/immunology , Agammaglobulinemia/metabolism , Autoimmunity , Child , Child, Preschool , Combined Modality Therapy , Enzyme Replacement Therapy , Follow-Up Studies , Gene Expression , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Male , Phenotype , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/metabolism , Transgenes , Treatment Outcome
2.
Blood ; 128(1): 45-54, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27129325

ABSTRACT

Adenosine deaminase (ADA) deficiency is a rare, autosomal-recessive systemic metabolic disease characterized by severe combined immunodeficiency (SCID). The treatment of choice for ADA-deficient SCID (ADA-SCID) is hematopoietic stem cell transplant from an HLA-matched sibling donor, although <25% of patients have such a donor available. Enzyme replacement therapy (ERT) partially and temporarily relieves immunodeficiency. We investigated the medium-term outcome of gene therapy (GT) in 18 patients with ADA-SCID for whom an HLA-identical family donor was not available; most were not responding well to ERT. Patients were treated with an autologous CD34(+)-enriched cell fraction that contained CD34(+) cells transduced with a retroviral vector encoding the human ADA complementary DNA sequence (GSK2696273) as part of single-arm, open-label studies or compassionate use programs. Overall survival was 100% over 2.3 to 13.4 years (median, 6.9 years). Gene-modified cells were stably present in multiple lineages throughout follow up. GT resulted in a sustained reduction in the severe infection rate from 1.17 events per person-year to 0.17 events per person-year (n = 17, patient 1 data not available). Immune reconstitution was demonstrated by normalization of T-cell subsets (CD3(+), CD4(+), and CD8(+)), evidence of thymopoiesis, and sustained T-cell proliferative capacity. B-cell function was evidenced by immunoglobulin production, decreased intravenous immunoglobulin use, and antibody response after vaccination. All 18 patients reported infections as adverse events; infections of respiratory and gastrointestinal tracts were reported most frequently. No events indicative of leukemic transformation were reported. Trial details were registered at www.clinicaltrials.gov as #NCT00598481.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/therapy , Genetic Therapy , Recovery of Function , Retroviridae , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Agammaglobulinemia/mortality , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Male , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/mortality , Survival Rate
3.
J Allergy Clin Immunol ; 133(4): 989-96, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24582316

ABSTRACT

BACKGROUND: Approximately 5% to 10% of asthmatic patients achieve incomplete symptom control on current therapies. The association of IL-13 with asthma pathology and reduced corticosteroid sensitivity suggests a potential benefit of anti-IL-13 therapy in refractory asthma. GSK679586, a humanized mAb, inhibits IL-13 binding to both IL-13 receptor α1 and α2. OBJECTIVES: We sought to evaluate the efficacy and safety of GSK679586 in patients with severe asthma refractory to maximally indicated doses of inhaled corticosteroids. METHODS: Patients who remained symptomatic (Asthma Control Questionnaire score ≥1.5) after uptitration to 1000 µg/d fluticasone propionate or greater were randomized to 3 once-monthly intravenous infusions of 10 mg/kg GSK679586 (n = 99) or placebo (n = 99). RESULTS: Treatment differences in adjusted mean change from baseline over 12 weeks were nonsignificant for Asthma Control Questionnaire symptom scores (the primary end point; GSK679586 = -0.31, placebo = -0.17, P = .058) and FEV1 (GSK679586 = -0.01, placebo = 0.03, P = .276). Similar analyses in patients with increased serum IgE levels, blood eosinophil counts, or both were also negative. Incidence of asthma exacerbations was similar between treatments. Most adverse events were nonserious and unrelated to treatment. Two GSK679586-treated patients had treatment-related serious adverse events (lethargy and supraventricular extrasystoles). CONCLUSIONS: Although well tolerated, GSK679586 did not demonstrate clinically meaningful improvements in asthma control, pulmonary function, or exacerbations in patients with severe asthma. Further studies are needed to determine whether therapies targeting IL-13, the functionally related IL-4 cytokine, or both can provide clinical benefit in patients with severe refractory asthma or a subpopulation of these patients beyond that achievable with high-dose corticosteroids.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Interleukin-13/antagonists & inhibitors , Adult , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacokinetics , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Asthma/physiopathology , Female , Humans , Male , Middle Aged , Severity of Illness Index , Treatment Outcome
4.
Br J Clin Pharmacol ; 75(1): 118-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22616628

ABSTRACT

AIMS: IL-13 is implicated as an important mediator of the pathology of asthma. This first clinical study with GSK679586, a novel humanized anti-IL-13 IgG1 monoclonal antibody, evaluated the safety, pharmacokinetics and pharmacodynamics of escalating single and repeat doses of GSK679586. METHODS: In this randomized, double-blind study, healthy subjects received single intravenous infusions of GSK679586 (0.005, 0.05, 0.5, 2.5, 10 mg kg(-1)) or placebo and mild intermittent asthmatics received two once monthly intravenous infusions of GSK679586 (2.5, 10, 20 mg kg(-1)) or placebo. RESULTS: GSK679586 displayed approximately linear pharmacokinetics (based on AUC and C(max)) with limited accumulation upon repeat administration. In mild intermittent asthmatics, treatment with GSK679586 produced an increase in serum total IL-13 concentrations, indicative of GSK679586-IL-13 complex formation. Additionally, mean levels of exhaled nitric oxide (FeNO), a marker of pulmonary inflammation, were reduced relative to baseline at 2.5, 10 and 20 mg kg(-1) doses of GSK679586 at both 2 weeks (19%, 44% and 52% decreases) and 8 weeks (29%, 55% and 42% decreases) after the second infusion. GSK679586 was well tolerated; the incidence of AEs was comparable across all presumed biologically active doses and there were no treatment-related SAEs. CONCLUSIONS: GSK679586 demonstrated dose-dependent pharmacological activity in the lungs of mild intermittent asthmatics. These findings, together with the favourable safety profile and advantageous PK characteristics of a monoclonal antibody (e.g. a long half-life supporting less frequent dosing), warrant further investigation of GSK679586 in a broader asthma patient population.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Asthma/drug therapy , Interleukin-13/antagonists & inhibitors , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Breath Tests , Double-Blind Method , Humans , Male , Nitric Oxide/metabolism
5.
J Clin Endocrinol Metab ; 93(12): 4810-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18812476

ABSTRACT

CONTEXT: Native glucagon-like peptide-1 increases insulin secretion, decreases glucagon secretion, and reduces appetite but is rapidly inactivated by dipeptidyl peptidase-4. Albiglutide is a novel dipeptidyl peptidase-4-resistant glucagon-like peptide-1 dimer fused to human albumin designed to have sustained efficacy in vivo. OBJECTIVES: The objectives were to investigate pharmacodynamics, pharmacokinetics, safety, and tolerability of albiglutide in type 2 diabetes subjects. METHODS: In a single-blind dose-escalation study, 54 subjects were randomized to receive placebo or 9-, 16-, or 32-mg albiglutide on d 1 and 8. In a complementary study, 46 subjects were randomized to a single dose (16 or 64 mg) of albiglutide to the arm, leg, or abdomen. RESULTS: Significant dose-dependent reductions in 24-h mean weighted glucose [area under the curve((0-24 h))] were observed, with placebo-adjusted least squares means difference values in the 32-mg cohort of -34.8 and -56.4 mg/dl [95% confidence interval (-54.1, -15.5) and (-82.2, -30.5)] for d 2 and 9, respectively. Placebo-adjusted fasting plasma glucose decreased by -26.7 and -50.7 mg/dl [95% confidence interval (-46.3, -7.06) and (-75.4, -26.0)] on d 2 and 9, respectively. Postprandial glucose was also reduced. No hypoglycemic episodes were detected in the albiglutide cohorts. The frequency and severity of the most common adverse events, headache and nausea, were comparable with placebo controls. Albiglutide half-life ranged between 6 and 7 d. The pharmacokinetics or pharmacodynamic of albiglutide was unaffected by injection site. CONCLUSIONS: Albiglutide improved fasting plasma glucose and postprandial glucose with a favorable safety profile in subjects with type 2 diabetes. Albiglutide's long half-life may allow for once-weekly or less frequent dosing.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/physiology , Hypoglycemic Agents/therapeutic use , Adolescent , Adult , Aged , Area Under Curve , Blood Glucose/metabolism , Dose-Response Relationship, Drug , Female , Glucagon-Like Peptide 1/adverse effects , Glucagon-Like Peptide 1/pharmacokinetics , Glucagon-Like Peptide 1/therapeutic use , Glycated Hemoglobin/metabolism , Half-Life , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Injections, Intravenous , Male , Middle Aged , Molecular Mimicry , Young Adult
6.
Plasmid ; 54(1): 57-69, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15907539

ABSTRACT

The Enterococcus faecalis plasmid pAM373 (36.7kb) encodes a mating response to the sex pheromone cAM373 secreted by recipient (plasmid-free) bacteria. Like certain other conjugative enterococcal plasmids, a key regulator of the pheromone response is a negatively acting protein, TraA, which is believed to interact with internalized pheromone to influence expression from a key transcriptional promoter P(0). An earlier report showed that in the case of pAM373 most, but not all, transposon-insertion mutations in traA differed from those in the case of pAD1 and pCF10 in that they did not give rise to the normally characteristic constitutive clumping. We show here that this phenomenon relates to a host effect involving an RpoB-related mutation associated with rifampin resistance. When harboring traA mutants, rifampin-sensitive hosts exhibited constitutive clumping, whereas rifampin-resistant hosts did not-despite the fact that the latter host exhibited a normal pheromone-inducible clumping response when harboring a wild-type plasmid. The data imply that TraA normally remains associated with the transcription complex after induction. In addition the promoter of traA, designated P(a), was shown to be located about 600bp upstream of the translational start site, as clones containing traA required this site to complement traA mutants in trans. Transcription from P(a) also gave rise to a short (130 nt) transcript, mD, expressed at a high level in uninduced cells. An earlier observation suggesting that TraA negatively affected transcriptional readthrough into the 3' end of traA from the t(ac) intrinsic bidirectional terminator between traA and the opposing, adjacent traC was supported by TraA complementation studies. Evidence is also presented suggesting that this regulation at t(ac) also involves an additional, possibly cis-acting, element.


Subject(s)
Bacterial Proteins/metabolism , Enterococcus faecalis/genetics , Escherichia coli Proteins/metabolism , Fimbriae Proteins/metabolism , Pheromones/genetics , Plasmids/physiology , Bacterial Proteins/genetics , Base Sequence , Codon, Terminator , DNA-Directed RNA Polymerases , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/physiology , Escherichia coli Proteins/genetics , Fimbriae Proteins/genetics , Gene Expression Regulation, Bacterial , Molecular Sequence Data , Mutation , Plasmids/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Rifampin/pharmacology
7.
Clin Infect Dis ; 38(12): 1716-23, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15227617

ABSTRACT

Atovaquone-proguanil has been shown to be effective and well tolerated for malaria prophylaxis in residents of countries of endemicity and in nonimmune adult travelers, but data about traveling children are limited. In a randomized, open-label, multicenter prophylaxis trial, 221 nonimmune pediatric travelers (age, 2-17 years) received either atovaquone-proguanil or chloroquine-proguanil. Safety and clinical outcome were evaluated 7, 28, and 60 days after travel. By posttravel day 7, a total of 39 (35%) of 110 atovaquone-proguanil and 41 (37%) of 111 chloroquine-proguanil recipients reported > or =1 adverse event. The data indicate that, over the course of treatment, fewer atovaquone-proguanil recipients had treatment-related adverse events (8% vs. 14%), including gastrointestinal complaints (5% vs. 10%). Two subjects discontinued prophylaxis because of drug-related adverse events; both had received chloroquine-proguanil. Observed compliance with prophylaxis was similar before and during travel, but it was higher for atovaquone-proguanil in the posttravel period. No study participant developed malaria. Atovaquone-proguanil was well tolerated and is an important addition to the limited arsenal of prophylactic agents available to children.


Subject(s)
Antimalarials/therapeutic use , Chloroquine/therapeutic use , Malaria/prevention & control , Naphthoquinones/therapeutic use , Proguanil/therapeutic use , Travel , Adolescent , Antimalarials/administration & dosage , Antimalarials/adverse effects , Atovaquone , Child , Child, Preschool , Chloroquine/administration & dosage , Chloroquine/adverse effects , Drug Therapy, Combination , Humans , Infant , Naphthoquinones/administration & dosage , Naphthoquinones/adverse effects , Patient Compliance , Proguanil/administration & dosage , Proguanil/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL