Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Article in English | MEDLINE | ID: mdl-32015031

ABSTRACT

Human cytomegalovirus (HCMV) can cause significant disease in immunocompromised patients, and treatment options are limited by toxicities. CSJ148 is a combination of two anti-HCMV human monoclonal antibodies (LJP538 and LJP539) that bind to and inhibit the functions of viral HCMV glycoprotein B (gB) and the pentameric complex, consisting of glycoproteins gH, gL, UL128, UL130, and UL131. In this phase 2, randomized, placebo-controlled trial, we evaluated the safety and efficacy of CSJ148 for prophylaxis of HCMV in patients undergoing allogeneic hematopoietic stem cell transplantation. As would be expected in the study population, all the patients (100%) reported at least one treatment-emergent adverse event. There were 22 deaths during this study, and over 80% of the patients receiving placebo or CSJ148 developed at least one adverse event of grade 3 or higher severity. No subject who received antibody developed a hypersensitivity- or infusion-related reaction. CSJ148-treated patients showed trends toward decreased viral load, shorter median duration of preemptive therapy, and fewer courses of preemptive therapy. However, the estimated probability that CSJ148 decreases the need for preemptive therapy compared to placebo was 69%, with a risk ratio of 0.89 and a 90% credible interval of 0.61 to 1.31. The primary efficacy endpoint was therefore not met, indicating that CSJ148 did not prevent clinically significant HCMV reactivation in recipients of allogeneic hematopoietic cell transplants. (This study has been registered at ClinicalTrials.gov under identifier NCT02268526 and at EudraCT under number 2017-002047-15.).


Subject(s)
Antibodies, Viral/pharmacology , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Administration, Intravenous , Adult , Aged , Antibodies, Viral/administration & dosage , Antibodies, Viral/adverse effects , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cytomegalovirus Infections/etiology , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/mortality , Humans , Male , Middle Aged , Placebos , Treatment Outcome , Viral Load , Young Adult
2.
Invest New Drugs ; 33(6): 1187-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26403509

ABSTRACT

PURPOSE: LY2090314 (LY) is a glycogen synthase kinase 3 inhibitor with preclinical efficacy in xenograft models when combined with platinum regimens. A first-in-human phase 1 dose-escalation study evaluated the combination of LY with pemetrexed/carboplatin. PATIENTS AND METHODS: Forty-one patients with advanced solid tumors received single-dose LY monotherapy lead-in and 37 patients received LY (10-120 mg) plus pemetrexed/carboplatin (500 mg/m(2) and 5-6 AUC, respectively) across 8 dose levels every 21 days. Primary objective was maximum tolerated dose (MTD) determination; secondary endpoints included safety, antitumor activity, pharmacokinetics, and beta-catenin pharmacodynamics. RESULTS: MTD of LY with pemetrexed/carboplatin was 40 mg. Eleven dose-limiting toxicities (DLTs) occurred in ten patients. DLTs during LY monotherapy occurred at ≥ 40 mg: grade 2 visual disturbance (n = 1) and grade 3/4 peri-infusional thoracic pain during or shortly post infusion (n = 4; chest, upper abdominal, and back pain). Ranitidine was added after de-escalation to 80 mg LY to minimize peri-infusional thoracic pain. Following LY with pemetrexed/carboplatin therapy, DLTs included grade 3/4 thrombocytopenia (n = 4) and grade 4 neutropenia (n = 1). Best overall response by RECIST included 5 confirmed partial responses (non-small cell lung cancer [n = 3], mesothelioma, and breast cancer) and 19 patients having stable disease. Systemic LY exposure was approximately linear over dose range studied. Transient upregulation of beta-catenin measured in peripheral blood mononuclear cells (PBMCs) occurred at 40 mg LY. CONCLUSIONS: The initial safety profile of LY2090314 was established. MTD LY dose with pemetrexed/carboplatin is 40 mg IV every 3 weeks plus ranitidine. Efficacy of LY plus pemetrexed/carboplatin requires confirmation in randomized trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carboplatin/administration & dosage , Glycogen Synthase Kinase 3/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/administration & dosage , Maleimides/administration & dosage , Pemetrexed/administration & dosage , Administration, Intravenous , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carboplatin/pharmacokinetics , Cohort Studies , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Glycogen Synthase Kinase 3/metabolism , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Male , Maleimides/pharmacokinetics , Middle Aged , Neoplasms/drug therapy , Neoplasms/enzymology , Pemetrexed/pharmacokinetics
3.
Biomed Res Int ; 2015: 420867, 2015.
Article in English | MEDLINE | ID: mdl-25815314

ABSTRACT

Dengue virus infection is a common tropical disease which often occurs without being detected. These asymptomatic cases provide information in relation to the manifestation of immunological aspects. In this study, we developed an ELISA method to compare neutralizing effects of dengue prM and E antibodies between dengue patients and their asymptomatic household members. Recombinant D2 premembrane (prM) was constructed, cloned, and tested for antigenicity. The recombinant protein was purified and tested with controls by using an indirect ELISA method. Positive dengue serum samples with their asymptomatic pair were then carried out onto the developed ELISA. In addition, commercially available recombinant envelope (E) protein was used to develop an ELISA which was tested with the same set of serum samples in the prM ELISA. Asymptomatic individuals showed preexisting heterotypic neutralizing antibodies. The recombinant prM was antigenically reactive in the developed ELISA. Dengue patients had higher prM and E antibodies compared to their household members. Our study highlights the neutralizing antibodies levels with respect to dengue prM and E between dengue patients and asymptomatic individuals.


Subject(s)
Antibodies, Viral/immunology , Dengue/immunology , Dengue/virology , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Base Sequence , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Family Characteristics , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/immunology , Restriction Mapping , Sequence Analysis, DNA
4.
PLoS One ; 9(4): e92240, 2014.
Article in English | MEDLINE | ID: mdl-24727912

ABSTRACT

OBJECTIVES: Dengue represents one of the most serious life-threatening vector-borne infectious diseases that afflicts approximately 50 million people across the globe annually. Whilst symptomatic infections are frequently reported, asymptomatic dengue remains largely unnoticed. Therefore, we sought to investigate the immune correlates conferring protection to individuals that remain clinically asymptomatic. METHODS: We determined the levels of neutralizing antibodies (nAbs) and gene expression profiles of host immune factors in individuals with asymptomatic infections, and whose cognate household members showed symptoms consistent to clinical dengue infection. RESULTS: We observed broad down-regulation of host defense response (innate, adaptive and matrix metalloprotease) genes in asymptomatic individuals as against symptomatic patients, with selective up-regulation of distinct genes that have been associated with protection. Selected down-regulated genes include: TNF α (TNF), IL8, C1S, factor B (CFB), IL2, IL3, IL4, IL5, IL8, IL9, IL10 and IL13, CD80, CD28, and IL18, MMP8, MMP10, MMP12, MMP15, MMP16, and MMP24. Selected up-regulated genes include: RANTES (CCL5), MIP-1α (CCL3L1/CCL3L3), MIP-1ß (CCL4L1), TGFß (TGFB), and TIMP1. CONCLUSION: Our findings highlight the potential association of certain host genes conferring protection against clinical dengue. These data are valuable to better explore the mysteries behind the hitherto poorly understood immunopathogenesis of subclinical dengue infection.


Subject(s)
Dengue Virus/pathogenicity , Dengue/virology , Antibodies, Neutralizing/metabolism , Cytokines/metabolism , Down-Regulation , Female , Humans , Male , Matrix Metalloproteinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
5.
J Womens Health (Larchmt) ; 19(8): 1459-65, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20626269

ABSTRACT

BACKGROUND: Raloxifene use in postmenopausal women with osteoporosis increases the risk of venous thromboembolic events (VTE) 2-fold compared with placebo. Platelet activation is involved in the pathophysiology of arterial thromboses more than venous thromboses, but aspirin may reduce VTE risk associated with estrogen use. This analysis examines the effects of concomitant antiplatelet therapy on VTE risk in raloxifene-treated women. METHODS: In the Raloxifene Use for the Heart (RUTH) trial, 10,101 postmenopausal women from 177 sites in 26 countries at increased risk of coronary heart disease (CHD) (primary prevention cohort) or with CHD (secondary prevention cohort) were randomized to placebo or raloxifene 60 mg/day and followed for a median 5.6 years. Reports of clinical symptoms of VTE were assessed. Concomitant use of antiplatelet agents (aspirin, clopidogrel, ticlopidine, dipyridamole) was allowed. Cox proportional hazard models, with use of warfarin, presence of fracture, and hospitalization as covariates, were used to estimate hazard ratios (HR) with 95% confidence intervals (CI). RESULTS: Overall, raloxifene use was associated with an increased VTE risk (HR 1.44, 95% CI 1.06-1.95) vs. placebo. Most women (72%) reported using aspirin, and 14.2% reported using nonaspirin antiplatelet agents during the study period. Users of antiplatelet agents were older, more likely to have CHD, and more likely to be hyperlipidemic. They had a higher VTE risk than nonusers. No difference in VTE risk was observed in women who used raloxifene alone vs. those who used raloxifene with antiplatelet agents during the study. The increase in VTE risk with raloxifene compared with placebo was not different between women who used antiplatelet agents at baseline (HR 1.44, 95% CI 0.98, 2.10) and those who did not use antiplatelet agents (HR 1.37, 95% CI 0.83, 2.27) (interaction p = 0.88). Similar conclusions were noted for aspirin and nonaspirin antiplatelet use. CONCLUSIONS: In RUTH, postmenopausal women treated with raloxifene had an increased risk of VTE compared with placebo. Concomitant use of aspirin or nonaspirin antiplatelet agents along with raloxifene did not change VTE risk.


Subject(s)
Platelet Aggregation Inhibitors/therapeutic use , Raloxifene Hydrochloride/adverse effects , Selective Estrogen Receptor Modulators/adverse effects , Venous Thromboembolism/chemically induced , Aspirin/therapeutic use , Cohort Studies , Coronary Disease/prevention & control , Double-Blind Method , Female , Humans , Middle Aged , Placebos , Postmenopause , Proportional Hazards Models , Raloxifene Hydrochloride/therapeutic use , Risk Factors , Selective Estrogen Receptor Modulators/therapeutic use , Treatment Outcome , Venous Thromboembolism/epidemiology , Venous Thromboembolism/prevention & control
6.
Pharm Stat ; 8(1): 5-11, 2009.
Article in English | MEDLINE | ID: mdl-18381588

ABSTRACT

It is challenging to estimate the statistical power when a complicated testing strategy is used to adjust for the type-I error for multiple comparisons in a clinical trial. In this paper, we use the Bonferroni Inequality to estimate the lower bound of the statistical power assuming that test statistics are approximately normally distributed and the correlation structure among test statistics is unknown or only partially known. The method was applied to the design of a clinical study for sample size and statistical power estimation.


Subject(s)
Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Data Interpretation, Statistical , Humans , Models, Statistical , Research Design , Sample Size
7.
Assay Drug Dev Technol ; 5(5): 663-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17939753

ABSTRACT

The Kolmogorov-Smirnov (K-S) test is a statistical method often used for comparing two distributions. In high-throughput screening (HTS) studies, such distributions usually arise from the phenotype of independent cell populations. However, the K-S test has been criticized for being overly sensitive in applications, and it often detects a statistically significant difference that is not biologically meaningful. One major reason is that there is a common phenomenon in HTS studies that systematic drifting exists among the distributions due to reasons such as instrument variation, plate edge effect, accidental difference in sample handling, etc. In particular, in high-content cellular imaging experiments, the location shift could be dramatic since some compounds themselves are fluorescent. This oversensitivity of the K-S test is particularly overpowered in cellular assays where the sample sizes are very big (usually several thousands). In this paper, a modified K-S test is proposed to deal with the nonspecific location-shift problem in HTS studies. Specifically, we propose that the distributions are "normalized" by density curve alignment before the K-S test is conducted. In applications to simulation data and real experimental data, the results show that the proposed method has improved specificity.


Subject(s)
DNA/analysis , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/statistics & numerical data , Algorithms , Antineoplastic Agents/pharmacology , Computer Simulation , Data Interpretation, Statistical , HeLa Cells , Humans , Models, Statistical , Nocodazole/pharmacology , Phenotype , Spectrometry, Fluorescence
8.
J Clin Pharmacol ; 45(9): 1032-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16100297

ABSTRACT

This open-label study investigated the effect of exenatide coadministration on the steady-state plasma pharmacokinetics of digoxin. A total of 21 healthy male subjects received digoxin (day 1, 0.5 mg twice daily; days 2-12, 0.25 mg once daily) and exenatide (days 8-12, 10 microg twice daily). Digoxin plasma and urine concentrations were measured on days 7 and 12. Exenatide coadministration did not change the overall 24-hour steady-state digoxin exposure (AUCtau,ss) and Cmin,ss but caused a 17% decrease in mean plasma digoxin Cmax,ss (1.40 to 1.16 ng/mL) and an increase in digoxin tmax,ss (median increase, 2.5 hours). Although the decrease in digoxin Cmax,ss was statistically significant, peak concentrations were within the therapeutic concentration range in all subjects. Digoxin urinary pharmacokinetic parameters were not altered. Gastrointestinal symptoms, the most common adverse effects of exenatide, decreased over time. Exenatide administration does not cause any changes in digoxin steady-state pharmacokinetics that would be expected to have clinical sequelae; thus, dosage adjustment does not appear warranted, based on pharmacokinetic considerations.


Subject(s)
Anti-Arrhythmia Agents/pharmacokinetics , Cardiotonic Agents/pharmacokinetics , Digoxin/pharmacokinetics , Peptides/pharmacology , Venoms/pharmacology , Adult , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/blood , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/blood , Digoxin/administration & dosage , Digoxin/blood , Drug Interactions , Exenatide , Humans , Male , Nausea/chemically induced , Peptides/administration & dosage , Peptides/adverse effects , Venoms/administration & dosage , Venoms/adverse effects
9.
Genomics ; 84(4): 767-77, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15475255

ABSTRACT

Affymetrix's high-density oligonucleotide arrays offer an exciting technology in biomedical research. With more and more statistical involvement in every step of the process, there has been a constant effort to make sure that the expression data are appropriately extracted in the first place. According to Affymetrix GeneChip technology, each gene is represented by 11-20 oligo probe pairs; the challenge is how to extract one meaningful number, expression, from the 11-20 pairs of numbers. More specifically, there is first a need to differentiate the components of specific binding, nonspecific binding, and optical background noise in both PM and MM probes, and then an expression measure that is proportional to the true abundance of transcripts is to be derived. A new method, SUM, which sums up PM and MM values and then follows a process similar to that of RMA, is considered. The performance of SUM is investigated and compared to the three most popular methods, MAS5, dChip, and RMA. The assessments are based on a well-controlled experiment dataset that is publicly available. The results show that in several respects the performance of SUM is comparable to that of RMA and dChip, and all three of these methods show some advantages over MAS5. There is some evidence showing that SUM has higher differential sensitivity than other methods in certain situations.


Subject(s)
Algorithms , DNA Probes/analysis , DNA Probes/genetics , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Animals , Data Interpretation, Statistical , Humans
10.
Am J Pharmacogenomics ; 4(2): 129-39, 2004.
Article in English | MEDLINE | ID: mdl-15059035

ABSTRACT

INTRODUCTION: The hybridization intensities derived from microarray experiments, for example Affymetrix's MAS5 signals, are very often transformed in one way or another before statistical models are fitted. The motivation for performing transformation is usually to satisfy the model assumptions such as normality and homogeneity in variance. Generally speaking, two types of strategies are often applied to microarray data depending on the analysis need: correlation analysis where all the gene intensities on the array are considered simultaneously, and gene-by-gene ANOVA where each gene is analyzed individually. AIM: We investigate the distributional properties of the Affymetrix GeneChip signal data under the two scenarios, focusing on the impact of analyzing the data at an inappropriate scale. METHODS: The Box-Cox type of transformation is first investigated for the strategy of pooling genes. The commonly used log-transformation is particularly applied for comparison purposes. For the scenario where analysis is on a gene-by-gene basis, the model assumptions such as normality are explored. The impact of using a wrong scale is illustrated by log-transformation and quartic-root transformation. RESULTS: When all the genes on the array are considered together, the dependent relationship between the expression and its variation level can be satisfactorily removed by Box-Cox transformation. When genes are analyzed individually, the distributional properties of the intensities are shown to be gene dependent. Derivation and simulation show that some loss of power is incurred when a wrong scale is used, but due to the robustness of the t-test, the loss is acceptable when the fold-change is not very large.


Subject(s)
Data Interpretation, Statistical , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Algorithms , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Databases, Genetic , Fluorescent Dyes , Gene Expression , Protein Folding , RNA/biosynthesis , RNA/genetics , Rats , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...