Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Proc Natl Acad Sci U S A ; 117(31): 18649-18660, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690687

ABSTRACT

Starting at birth, the immune system of newborns and children encounters and is influenced by environmental challenges. It is still not completely understood how γδ T cells emerge and adapt during early life. Studying the composition of T cell receptors (TCRs) using next-generation sequencing (NGS) in neonates, infants, and children can provide valuable insights into the adaptation of T cell subsets. To investigate how neonatal γδ T cell repertoires are shaped by microbial exposure after birth, we monitored the γ-chain (TRG) and δ-chain (TRD) repertoires of peripheral blood T cells in newborns, infants, and young children from Europe and sub-Saharan Africa. We identified a set of TRG and TRD sequences that were shared by all children from Europe and Africa. These were primarily public clones, characterized by simple rearrangements of Vγ9 and Vδ2 chains with low junctional diversity and usage of non-TRDJ1 gene segments, reminiscent of early ontogenetic subsets of γδ T cells. Further profiling revealed that these innate, public Vγ9Vδ2+ T cells underwent an immediate TCR-driven polyclonal proliferation within the first 4 wk of life. In contrast, γδ T cells using Vδ1+ and Vδ3+TRD rearrangements did not significantly expand after birth. However, different environmental cues may lead to the observed increase of Vδ1+ and Vδ3+TRD sequences in the majority of African children. In summary, we show how dynamic γδ TCR repertoires develop directly after birth and present important differences among γδ T cell subsets.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/immunology , Africa South of the Sahara , Bacteria/immunology , Child , Child, Preschool , Europe , Gene Rearrangement, T-Lymphocyte/genetics , Gene Rearrangement, T-Lymphocyte/immunology , Humans , Infant , Infant, Newborn , Longitudinal Studies , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology
2.
Proc Natl Acad Sci U S A ; 116(39): 19753-19759, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31506353

ABSTRACT

Hypoxia is a ubiquitous feature of cancers, encouraging glycolytic metabolism, proliferation, and resistance to therapy. Nonetheless, hypoxia is a poorly defined term with confounding features described in the literature. Redox biology provides an important link between the external cellular microenvironment and the cell's response to changing oxygen pressures. In this paper, we demonstrate a correlation between intracellular redox potential (measured using optical nanosensors) and the concentrations of microRNAs (miRNAs) involved in the cell's response to changes in oxygen pressure. The correlations were established using surprisal analysis (an approach derived from thermodynamics and information theory). We found that measured redox potential changes reflect changes in the free energy computed by surprisal analysis of miRNAs. Furthermore, surprisal analysis identified groups of miRNAs, functionally related to changes in proliferation and metastatic potential that played the most significant role in the cell's response to changing oxygen pressure.


Subject(s)
Cell Hypoxia/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Cytoplasm/metabolism , Humans , Hypoxia/metabolism , MCF-7 Cells/metabolism , Oxidation-Reduction , Reactive Oxygen Species , Thermodynamics , Tumor Microenvironment/genetics
3.
N Engl J Med ; 372(18): 1689-99, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25923549

ABSTRACT

BACKGROUND: The EGFR T790M mutation is the most common mechanism of drug resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in patients who have lung cancer with an EGFR mutation (EGFR-mutated lung cancer). In preclinical models, the EGFR inhibitor AZD9291 has been shown to be effective against both EGFR tyrosine kinase inhibitor-sensitizing and T790M resistance mutations. METHODS: We administered AZD9291 at doses of 20 to 240 mg once daily in patients with advanced lung cancer who had radiologically documented disease progression after previous treatment with EGFR tyrosine kinase inhibitors. The study included dose-escalation cohorts and dose-expansion cohorts. In the expansion cohorts, prestudy tumor biopsies were required for central determination of EGFR T790M status. Patients were assessed for safety, pharmacokinetics, and efficacy. RESULTS: A total of 253 patients were treated. Among 31 patients enrolled in the dose-escalation cohorts, no dose-limiting toxic effects occurred at the doses evaluated. An additional 222 patients were treated in five expansion cohorts. The most common all-cause adverse events were diarrhea, rash, nausea, and decreased appetite. The overall objective tumor response rate was 51% (95% confidence interval [CI], 45 to 58). Among 127 patients with centrally confirmed EGFR T790M who could be evaluated for response, the response rate was 61% (95% CI, 52 to 70). In contrast, among 61 patients without centrally detectable EGFR T790M who could be evaluated for response, the response rate was 21% (95% CI, 12 to 34). The median progression-free survival was 9.6 months (95% CI, 8.3 to not reached) in EGFR T790M-positive patients and 2.8 months (95% CI, 2.1 to 4.3) in EGFR T790M-negative patients. CONCLUSIONS: AZD9291 was highly active in patients with lung cancer with the EGFR T790M mutation who had had disease progression during prior therapy with EGFR tyrosine kinase inhibitors. (Funded by AstraZeneca; ClinicalTrials.gov number, NCT01802632.).


Subject(s)
Acrylamides/administration & dosage , Aniline Compounds/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Acrylamides/adverse effects , Acrylamides/pharmacokinetics , Adult , Aged , Aged, 80 and over , Aniline Compounds/adverse effects , Aniline Compounds/pharmacokinetics , Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Disease-Free Survival , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , Female , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics
4.
Br J Clin Pharmacol ; 84(12): 2877-2888, 2018 12.
Article in English | MEDLINE | ID: mdl-30171779

ABSTRACT

AIM: We report on two Phase 1, open-label, single-arm studies assessing the effect of osimertinib on simvastatin (CYP3A substrate) and rosuvastatin (breast cancer resistance protein substrate [BCRP] substrate) exposure in patients with advanced epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer who have progressed after treatment with an EGFR tyrosine kinase inhibitor, to determine, upon coadministration, whether osimertinib could affect the exposure of these agents. METHODS: Fifty-two patients in the CYP3A study (pharmacokinetic [PK] analysis, n = 49), and 44 patients in the BCRP study were dosed (PK analysis, n = 44). In the CYP3A study, patients received single doses of simvastatin 40 mg on Days 1 and 31, and osimertinib 80 mg once daily on Days 3-32. In the BCRP study, single doses of rosuvastatin 20 mg were given on Days 1 and 32, and osimertinib 80 mg once daily on Days 4-34. RESULTS: Geometric least squares mean (GLSM) ratios (90% confidence intervals) of simvastatin plus osimertinib for area under the plasma concentration-time curves from zero to infinity (AUC) were 91% (77-108): entirely contained within the predefined no relevant effect limits, and Cmax of 77% (63, 94) which was not contained within the limits. GLSM ratios of rosuvastatin plus osimertinib for AUC were 135% (115-157) and Cmax were 172 (146, 203): outside the no relevant effect limits. CONCLUSIONS: Osimertinib is unlikely to have any clinically relevant interaction with CYP3A substrates and has a weak inhibitory effect on BCRP. No new safety concerns were identified in either study.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Rosuvastatin Calcium/pharmacokinetics , Simvastatin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/physiology , Acrylamides/administration & dosage , Acrylamides/adverse effects , Adult , Aged , Aged, 80 and over , Aniline Compounds/administration & dosage , Aniline Compounds/adverse effects , Area Under Curve , Cytochrome P-450 CYP3A/physiology , Female , Humans , Hydroxycholesterols/blood , Male , Middle Aged , Neoplasm Proteins/physiology
5.
Br J Clin Pharmacol ; 84(6): 1156-1169, 2018 06.
Article in English | MEDLINE | ID: mdl-29381826

ABSTRACT

AIMS: We investigated the effects of a strong CYP3A4 inhibitor (itraconazole) or inducer (rifampicin) on the pharmacokinetics of the epidermal growth factor receptor-tyrosine kinase inhibitor osimertinib, in patients with advanced non-small cell lung cancer in two Phase I, open-label, two-part clinical studies. Part one of both studies is reported. METHODS: In the itraconazole study (NCT02157883), patients received single-dose osimertinib 80 mg on Days 1 and 10 and itraconazole (200 mg twice daily) on Days 6-18 orally. In the rifampicin study (NCT02197247), patients received osimertinib 80 mg once daily on Days 1-77 and rifampicin 600 mg once daily on Days 29-49. RESULTS: In the itraconazole study (n = 36), the geometric least squares mean (GMLSM) ratios (osimertinib plus itraconazole/osimertinib alone) for Cmax and AUC were 80% (90% CI 73, 87) and 124% (90% CI 115, 135), respectively, below the predefined no-effect upper limit of 200%. In the rifampicin study (n = 40), the GMLSM ratios (osimertinib plus rifampicin/osimertinib alone) for Css,max and AUCτ were 27% (90% CI 24, 30) and 22% (90% CI 20, 24), respectively, below the predefined no-effect lower limit of 50%. The induction effect of rifampicin was apparent within 7 days of initiation; osimertinib Css,max and AUCτ values returned to pre-rifampicin levels within 3 weeks of rifampicin discontinuation. No new osimertinib safety findings were observed. CONCLUSIONS: Osimertinib can be co-administered with CYP3A4 inhibitors, but strong CYP3A inducers should be avoided if possible.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Itraconazole/administration & dosage , Lung Neoplasms/drug therapy , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Rifampin/administration & dosage , Acrylamides , Administration, Oral , Adult , Aged , Aged, 80 and over , Aniline Compounds , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Cytochrome P-450 CYP3A Inducers/adverse effects , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drug Administration Schedule , Drug Interactions , Female , Humans , Itraconazole/adverse effects , Lung Neoplasms/pathology , Male , Middle Aged , Models, Biological , Piperazines/administration & dosage , Piperazines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Rifampin/adverse effects , Risk Assessment , Risk Factors , Treatment Outcome
6.
Clin Infect Dis ; 63(9): 1213-1226, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27436422

ABSTRACT

Vaccines can have nontargeted heterologous effects that manifest as increased protection against nonvaccine infections, as described for measles vaccine (MV), or increased susceptibility to infections and death, as described following diphtheria-tetanus-whole cell pertussis (DTP) vaccination. The mechanisms are unknown, and high-quality immunological studies are lacking. This study was designed to investigate the heterologous effects of MV and DTP in 302 Gambian infants. The results support a sex-differential immunosuppressive effect of DTP on innate proinflammatory responses and T-cell immunity. Males but not females receiving MV had enhanced proinflammatory innate responses. The results point to modified signaling via Toll-like receptor 4 (TLR4) as a possible mechanism for the effects on innate immunity. When both vaccines were administered together, purified protein derivative responses were enhanced in females but downregulated in males. Collectively, these data indicate immunological effects that could account for heterologous effects of MV and DTP, to take forward into prospective trials.


Subject(s)
Diphtheria-Tetanus-Pertussis Vaccine/immunology , Measles Vaccine/immunology , Sex Characteristics , Antibodies, Viral/blood , Cohort Studies , Cytokines/blood , Female , Gambia , Genome, Human , Humans , Immunity, Innate , Immunoglobulin G/blood , Immunosuppression Therapy , Infant , Longitudinal Studies , Male , RNA , T-Lymphocytes/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Transcriptome
7.
Drug Metab Dispos ; 44(8): 1201-12, 2016 08.
Article in English | MEDLINE | ID: mdl-27226351

ABSTRACT

Preclinical and clinical studies were conducted to determine the metabolism and pharmacokinetics of osimertinib and key metabolites AZ5104 and AZ7550. Osimertinib was designed to covalently bind to epidermal growth factor receptors, allowing it to achieve nanomolar cellular potency (Finlay et al., 2014). Covalent binding was observed in incubations of radiolabeled osimertinib with human and rat hepatocytes, human and rat plasma, and human serum albumin. Osimertinib, AZ5104, and AZ7550 were predominantly metabolized by CYP3A. Seven metabolites were detected in human hepatocytes, also observed in rat or dog hepatocytes at similar or higher levels. After oral administration of radiolabeled osimertinib to rats, drug-related material was widely distributed, with the highest radioactivity concentrations measured at 6 hours postdose in most tissues; radioactivity was detectable in 42% of tissues 60 days postdose. Concentrations of [(14)C]-radioactivity in blood were lower than in most tissues. After the administration of a single oral dose of 20 mg of radiolabeled osimertinib to healthy male volunteers, ∼19% of the dose was recovered by 3 days postdose. At 84 days postdose, mean total radioactivity recovery was 14.2% and 67.8% of the dose in urine and feces. The most abundant metabolite identified in feces was AZ5104 (∼6% of dose). Osimertinib accounted for ∼1% of total radioactivity in the plasma of non-small cell lung cancer patients after 22 days of 80-mg osimertinib once-daily treatment; the most abundant circulatory metabolites were AZ7550 and AZ5104 (<10% of total osimertinib-related material). Osimertinib is extensively distributed and metabolized in humans and is eliminated primarily via the fecal route.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Acrylamides , Adult , Aged , Aniline Compounds , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Binding Sites , Biotransformation , Carcinoma, Non-Small-Cell Lung/blood , Cysteine , Cytochrome P-450 CYP3A/metabolism , Dogs , Drug Administration Schedule , ErbB Receptors/chemistry , Feces/chemistry , Female , Hepatocytes/enzymology , Humans , Lung Neoplasms/blood , Male , Mice , Middle Aged , Piperazines/administration & dosage , Piperazines/blood , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Rats, Wistar , Serum Albumin, Human/metabolism , Tissue Distribution
8.
Proc Natl Acad Sci U S A ; 110(8): 3053-8, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23391734

ABSTRACT

Severe infectious disease in children may be a manifestation of primary immunodeficiency. These genetic disorders represent important experiments of nature with the capacity to elucidate nonredundant mechanisms of human immunity. We hypothesized that a primary defect of innate antiviral immunity was responsible for unusually severe viral illness in two siblings; the proband developed disseminated vaccine strain measles following routine immunization, whereas an infant brother died after a 2-d febrile illness from an unknown viral infection. Patient fibroblasts were indeed abnormally permissive for viral replication in vitro, associated with profound failure of type I IFN signaling and absence of STAT2 protein. Sequencing of genomic DNA and RNA revealed a homozygous mutation in intron 4 of STAT2 that prevented correct splicing in patient cells. Subsequently, other family members were identified with the same genetic lesion. Despite documented infection by known viral pathogens, some of which have been more severe than normal, surviving STAT2-deficient individuals have remained generally healthy, with no obvious defects in their adaptive immunity or developmental abnormalities. These findings imply that type I IFN signaling [through interferon-stimulated gene factor 3 (ISGF3)] is surprisingly not essential for host defense against the majority of common childhood viral infections.


Subject(s)
Genetic Predisposition to Disease , STAT2 Transcription Factor/genetics , Virus Diseases/genetics , Base Sequence , Cells, Cultured , Child, Preschool , DNA Primers , Female , Humans , Interferon Type I/metabolism , Oligonucleotide Array Sequence Analysis , Pedigree , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Virus Diseases/metabolism
9.
Nucleic Acids Res ; 41(17): 8107-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23832230

ABSTRACT

Interferons (IFN) play a pivotal role in innate immunity, orchestrating a cell-intrinsic anti-pathogenic state and stimulating adaptive immune responses. The complex interplay between the primary response to IFNs and its modulation by positive and negative feedback loops is incompletely understood. Here, we implement the combination of high-resolution gene-expression profiling of nascent RNA with translational inhibition of secondary feedback by cycloheximide. Unexpectedly, this approach revealed a prominent role of negative feedback mechanisms during the immediate (≤60 min) IFNα response. In contrast, a more complex picture involving both negative and positive feedback loops was observed on IFNγ treatment. IFNγ-induced repression of genes associated with regulation of gene expression, cellular development, apoptosis and cell growth resulted from cycloheximide-resistant primary IFNγ signalling. In silico promoter analysis revealed significant overrepresentation of SP1/SP3-binding sites and/or GC-rich stretches. Although signal transducer and activator of transcription 1 (STAT1)-binding sites were not overrepresented, repression was lost in absence of STAT1. Interestingly, basal expression of the majority of these IFNγ-repressed genes was dependent on STAT1 in IFN-naïve fibroblasts. Finally, IFNγ-mediated repression was also found to be evident in primary murine macrophages. IFN-repressed genes include negative regulators of innate and stress response, and their decrease may thus aid the establishment of a signalling perceptive milieu.


Subject(s)
Gene Expression Regulation , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Promoter Regions, Genetic , Transcription, Genetic , Animals , Cells, Cultured , Computer Simulation , Cycloheximide/pharmacology , Feedback, Physiological , Gene Expression Profiling , Gene Expression Regulation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , NIH 3T3 Cells , Protein Synthesis Inhibitors/pharmacology , Response Elements , STAT1 Transcription Factor/physiology , Thiouridine , Transcription, Genetic/drug effects
10.
PLoS Pathog ; 8(2): e1002517, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22319449

ABSTRACT

The early host response to viral infections involves transient activation of pattern recognition receptors leading to an induction of inflammatory cytokines such as interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNFα). Subsequent activation of cytokine receptors in an autocrine and paracrine manner results in an inflammatory cascade. The precise mechanisms by which viruses avert an inflammatory cascade are incompletely understood. Nuclear factor (NF)-κB is a central regulator of the inflammatory signaling cascade that is controlled by inhibitor of NF-κB (IκB) proteins and the IκB kinase (IKK) complex. In this study we show that murine cytomegalovirus inhibits the inflammatory cascade by blocking Toll-like receptor (TLR) and IL-1 receptor-dependent NF-κB activation. Inhibition occurs through an interaction of the viral M45 protein with the NF-κB essential modulator (NEMO), the regulatory subunit of the IKK complex. M45 induces proteasome-independent degradation of NEMO by targeting NEMO to autophagosomes for subsequent degradation in lysosomes. We propose that the selective and irreversible degradation of a central regulatory protein by autophagy represents a new viral strategy to dampen the inflammatory response.


Subject(s)
I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muromegalovirus/metabolism , NF-kappa B/antagonists & inhibitors , Phagosomes/immunology , Animals , Autophagy , Inflammation , Interleukin-1beta/metabolism , Mice , NF-kappa B/metabolism , NIH 3T3 Cells , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS Biol ; 9(3): e1000598, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21408089

ABSTRACT

Little is known about the protective role of inflammatory processes in modulating lipid metabolism in infection. Here we report an intimate link between the innate immune response to infection and regulation of the sterol metabolic network characterized by down-regulation of sterol biosynthesis by an interferon regulatory loop mechanism. In time-series experiments profiling genome-wide lipid-associated gene expression of macrophages, we show a selective and coordinated negative regulation of the complete sterol pathway upon viral infection or cytokine treatment with IFNγ or ß but not TNF, IL1ß, or IL6. Quantitative analysis at the protein level of selected sterol metabolic enzymes upon infection shows a similar level of suppression. Experimental testing of sterol metabolite levels using lipidomic-based measurements shows a reduction in metabolic output. On the basis of pharmacologic and RNAi inhibition of the sterol pathway we show augmented protection against viral infection, and in combination with metabolite rescue experiments, we identify the requirement of the mevalonate-isoprenoid branch of the sterol metabolic network in the protective response upon statin or IFNß treatment. Conditioned media experiments from infected cells support an involvement of secreted type 1 interferon(s) to be sufficient for reducing the sterol pathway upon infection. Moreover, we show that infection of primary macrophages containing a genetic knockout of the major type I interferon, IFNß, leads to only a partial suppression of the sterol pathway, while genetic knockout of the receptor for all type I interferon family members, ifnar1, or associated signaling component, tyk2, completely abolishes the reduction of the sterol biosynthetic activity upon infection. Levels of the proteolytically cleaved nuclear forms of SREBP2, a key transcriptional regulator of sterol biosynthesis, are reduced upon infection and IFNß treatment at both the protein and de novo transcription level. The reduction in srebf2 gene transcription upon infection and IFN treatment is also found to be strictly dependent on ifnar1. Altogether these results show that type 1 IFN signaling is both necessary and sufficient for reducing the sterol metabolic network activity upon infection, thereby linking the regulation of the sterol pathway with interferon anti-viral defense responses. These findings bring a new link between sterol metabolism and interferon antiviral response and support the idea of using host metabolic modifiers of innate immunity as a potential antiviral strategy.


Subject(s)
Down-Regulation , Herpesviridae Infections/immunology , Interferon-beta/physiology , Interferon-gamma/physiology , Muromegalovirus/immunology , Sterols/biosynthesis , Animals , Antiviral Agents/pharmacology , Cholesterol/metabolism , Herpesviridae Infections/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/pharmacology , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , RNA Interference , Signal Transduction , Simvastatin/pharmacology , Sterol Regulatory Element Binding Protein 2/physiology
12.
Curr Opin Infect Dis ; 26(3): 213-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23449137

ABSTRACT

PURPOSE OF REVIEW: Sepsis is a serious complication in preterm and term infants, yet our understanding of how neonates respond to infection remains poorly defined. RECENT FINDINGS: We describe our current clinical, cellular and molecular understanding of the neonatal host systemic response to infection. We find that host resilience essentially relies on innate immune mechanisms despite there being a complete repertoire of cellular components of the adaptive immune arm. The functional interplay between metabolism, immunity and microbiome further suggests that neonatal vulnerability to infection is not simply due to immaturity of the immune system but how immune homeostasis is regulated. Further research is required for exploring regulatory homeostatic mechanisms between innate and adaptive responses and microbiome colonization at birth, but which can impart an adverse trajectory to infection. SUMMARY: The vulnerability and resilience against infection in neonates, including extreme preterm infants, still remains poorly understood. We advance the view that greater consideration should be given to understanding the set point in the regulation of homeostatic control of innate and adaptive immunity and its interplay with metabolism and the newly acquired microbiome.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Sepsis/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular/physiology , Infant, Newborn , Infant, Premature/immunology , Metagenome/immunology , T-Lymphocytes/immunology
13.
Proc Natl Acad Sci U S A ; 107(31): 13830-5, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20643939

ABSTRACT

Although the functional parameters of microRNAs (miRNAs) have been explored in some depth, the roles of these molecules in viral infections remain elusive. Here we report a general method for global analysis of miRNA function that compares the significance of both overexpressing and inhibiting each mouse miRNA on the growth properties of different viruses. Our comparative analysis of representatives of all three herpesvirus subfamilies identified host miRNAs with broad anti- and proviral properties which extend to a single-stranded RNA virus. Specifically, we demonstrate the broad antiviral capacity of miR-199a-3p and illustrate that this individual host-encoded miRNA regulates multiple pathways required and/or activated by viruses, including PI3K/AKT and ERK/MAPK signaling, oxidative stress signaling, and prostaglandin synthesis. Global miRNA expression analysis further demonstrated that the miR-199a/miR-214 cluster is down-regulated in both murine and human cytomegalovirus infection and manifests similar antiviral properties in mouse and human cells. Overall, we report a general strategy for examining the contributions of individual host miRNAs in viral infection and provide evidence that these molecules confer broad inhibitory potential against multiple viruses.


Subject(s)
Antiviral Agents/analysis , Genome-Wide Association Study/methods , Herpesviridae/drug effects , MicroRNAs/analysis , Animals , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Gene Expression Regulation/drug effects , Humans , Mice , MicroRNAs/pharmacology , NIH 3T3 Cells , Signal Transduction/drug effects
14.
J Virol ; 85(19): 10286-99, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775459

ABSTRACT

Activated macrophages play a central role in controlling inflammatory responses to infection and are tightly regulated to rapidly mount responses to infectious challenge. Type I interferon (alpha/beta interferon [IFN-α/ß]) and type II interferon (IFN-γ) play a crucial role in activating macrophages and subsequently restricting viral infections. Both types of IFNs signal through related but distinct signaling pathways, inducing a vast number of interferon-stimulated genes that are overlapping but distinguishable. The exact mechanism by which IFNs, particularly IFN-γ, inhibit DNA viruses such as cytomegalovirus (CMV) is still not fully understood. Here, we investigate the antiviral state developed in macrophages upon reversible inhibition of murine CMV by IFN-γ. On the basis of molecular profiling of the reversible inhibition, we identify a significant contribution of a restricted type I IFN subnetwork linked with IFN-γ activation. Genetic knockout of the type I-signaling pathway, in the context of IFN-γ stimulation, revealed an essential requirement for a primed type I-signaling process in developing a full refractory state in macrophages. A minimal transient induction of IFN-ß upon macrophage activation with IFN-γ is also detectable. In dose and kinetic viral replication inhibition experiments with IFN-γ, the establishment of an antiviral effect is demonstrated to occur within the first hours of infection. We show that the inhibitory mechanisms at these very early times involve a blockade of the viral major immediate-early promoter activity. Altogether our results show that a primed type I IFN subnetwork contributes to an immediate-early antiviral state induced by type II IFN activation of macrophages, with a potential further amplification loop contributed by transient induction of IFN-ß.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Macrophages/immunology , Macrophages/virology , Muromegalovirus/growth & development , Muromegalovirus/immunology , Animals , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , Time Factors
15.
Mol Pharm ; 9(11): 3034-45, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23009633

ABSTRACT

The aim of this study was to investigate the mechanisms that might explain the observed route-dependent stereoselective pharmacokinetics (PK) of R/S-verapamil (R/S-VER) following oral and intravenous (iv) administration, by using a novel pig-specific physiologically based pharmacokinetic (PBPK) model suitable for investigations of first-pass extraction in the gut (E(G)) and the liver (E(H)). The PBPK model consisted of eight tissue compartments and was designed to simultaneously model the plasma concentration-time (PCT) profiles from three sampling sites after intrajejunal (ij) or iv administration of VER. The PBPK model successfully described the observed PCT profiles and E(H) over time for R- and S-VER. Extensive tissue binding to gut mucosa, liver, and lungs was an important determinant of the observed PK data. The stereoselective PK of VER was explained by a combination of several processes, including enantioselective plasma protein binding, blood-to-plasma partition, and gut mucosa and liver tissue distribution. The absence of stereoselectivity after iv dosing indicates that the first-pass tissue binding effect is an important factor in determining the steroselective PK of R/S-VER after oral administration. Additionally a combination of extensive liver tissue binding and a metabolite inhibition mechanism explained the time-dependent E(H) for both R- and S-VER. An in vitro-in vivo correlation of absorption needs to consider these processes because tissue binding may confound analysis of a drug's biopharmaceutical properties when using classical deconvolution or convolution techniques. In conclusion, a combination of PK data from multiple plasma sampling sites and a PBPK modeling approach provided a mechanistic understanding of processes involved in the intestinal absorption and first-pass extraction of R- and S-VER.


Subject(s)
Antifungal Agents/metabolism , Intestinal Mucosa/metabolism , Ketoconazole/metabolism , Liver/metabolism , Models, Biological , Vasodilator Agents/metabolism , Verapamil/metabolism , Administration, Oral , Animals , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacokinetics , Drug Combinations , Drug Interactions , Intestines/drug effects , Ketoconazole/administration & dosage , Ketoconazole/pharmacokinetics , Liver/drug effects , Protein Binding , Stereoisomerism , Swine , Tissue Distribution , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacokinetics , Verapamil/administration & dosage , Verapamil/pharmacokinetics
16.
Xenobiotica ; 42(9): 917-28, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22559211

ABSTRACT

In this study an advanced multisampling site pig model, with simultaneous venous blood sampling pre- and post liver, was applied to quantify the role of the intestine in relation to the liver in first-pass glucuronidation of raloxifene in vivo. The pharmacokinetic of raloxifene (a BCS/BDDCS class II compound) in humans is characterized by extensive metabolism (>90%) and the major metabolite is the 4'-ß-glucuronide (R-4-G). Following intra-jejunal (i.j.) single dose administration in pigs raloxifene was metabolized in the gut (E(G)) during first-pass to more than 70% and a high concentration (AUC(0-6 h) ratio R-4-G/raloxifene >100) of R-4-G was reached in the portal vein. The hepatic extraction (E(H)) of raloxifene was ~50% and as in humans the bioavailability become low (~7%) in pigs. Interestingly the E(H) of raloxifene and R-4-G was time-dependent after i.j. administration. It is clear that the gut was the dominating organ in first-pass extraction of raloxifene in vivo in pigs. The quantification in this study support earlier human data and emphasize that intestinal glucuronidation should be considered early in the pharmaceutical development.


Subject(s)
Glucuronides/metabolism , Intestinal Mucosa/metabolism , Raloxifene Hydrochloride/metabolism , Raloxifene Hydrochloride/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Glucuronides/chemistry , Molecular Structure , Raloxifene Hydrochloride/administration & dosage , Raloxifene Hydrochloride/chemistry , Sus scrofa
17.
Eur J Pharm Sci ; 179: 106296, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36184958

ABSTRACT

Long acting injectables (LAI) products are a popular intervention for treating a number of chronic conditions, with their long drug release reducing the administration frequency and thus improving patient adherence. The extended release, however, can provide a major challenge to bioequivalence (BE) testing since the long absorption half-life results in a long washout period, meaning that a traditional BE study can be many months or years in length. The unique PK profile for LAI products also means that it is critical to have appropriate metrics to summarise the plasma concentration profile. In this work, we use paliperidone as a case study to demonstrate how Population PK modelling can be utilised to explore sensitivity of summary metrics to different products. We also determine a range of products that are bioequivalent after both multiple dosing and single dosing. Finally, we show how the modelling can be used in a (virtual) PK study as an alternative approach to determining bioequivalence. This work demonstrates the potential for Population PK modelling in bioequivalence assessment, opening doors to more streamlined product development.


Subject(s)
Paliperidone Palmitate , Humans , Therapeutic Equivalency , Drug Liberation
18.
Pharmaceutics ; 14(5)2022 May 07.
Article in English | MEDLINE | ID: mdl-35631595

ABSTRACT

A webinar series that was organised by the Academy of Pharmaceutical Sciences Biopharmaceutics focus group in 2021 focused on the challenges of developing clinically relevant dissolution specifications (CRDSs) for oral drug products. Industrial scientists, together with regulatory and academic scientists, came together through a series of six webinars, to discuss progress in the field, emerging trends, and areas for continued collaboration and harmonisation. Each webinar also hosted a Q&A session where participants could discuss the shared topic and information. Although it was clear from the presentations and Q&A sessions that we continue to make progress in the field of CRDSs and the utility/success of PBBM, there is also a need to continue the momentum and dialogue between the industry and regulators. Five key areas were identified which require further discussion and harmonisation.

19.
Biomicrofluidics ; 16(2): 024108, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35464137

ABSTRACT

Drug-induced liver injury (DILI) results in over 100 000 hospital attendances per year in the UK alone and is a leading cause for the post-marketing withdrawal of new drugs, leading to significant financial losses. MicroRNA-122 (miR-122) has been proposed as a sensitive DILI marker although no commercial applications are available yet. Extracellular blood microRNAs (miRNAs) are promising clinical biomarkers but their measurement at point of care remains time-consuming, technically challenging, and expensive. For circulating miRNA to have an impact on healthcare, a key challenge to overcome is the development of rapid and reliable low-cost sample preparation. There is an acknowledged issue with miRNA stability in the presence of hemolysis and platelet activation, and no solution has been demonstrated for fast and robust extraction at the site of blood draw. Here, we report a novel microfluidic platform for the extraction of circulating miR-122 from blood enabled by a vertical approach and gravity-based bubble mixing. The performance of this disposable cartridge was verified by standard quantitative polymerase chain reaction analysis on extracted miR-122. The cartridge performed equivalently or better than standard bench extraction kits. The extraction cartridge was combined with electrochemical impedance spectroscopy to detect miR-122 as an initial proof-of-concept toward an application in point-of-care detection. This platform enables the standardization of sample preparation and the detection of miRNAs at the point of blood draw and in resource limited settings and could aid the introduction of miRNA-based assays into routine clinical practice.

20.
PeerJ ; 9: e10681, 2021.
Article in English | MEDLINE | ID: mdl-33569251

ABSTRACT

PURPOSE: To assess whether a model-based analysis increased statistical power over an analysis of final day volumes and provide insights into more efficient patient derived xenograft (PDX) study designs. METHODS: Tumour xenograft time-series data was extracted from a public PDX drug treatment database. For all 2-arm studies the percent tumour growth inhibition (TGI) at day 14, 21 and 28 was calculated. Treatment effect was analysed using an un-paired, two-tailed t-test (empirical) and a model-based analysis, likelihood ratio-test (LRT). In addition, a simulation study was performed to assess the difference in power between the two data-analysis approaches for PDX or standard cell-line derived xenografts (CDX). RESULTS: The model-based analysis had greater statistical power than the empirical approach within the PDX data-set. The model-based approach was able to detect TGI values as low as 25% whereas the empirical approach required at least 50% TGI. The simulation study confirmed the findings and highlighted that CDX studies require fewer animals than PDX studies which show the equivalent level of TGI. CONCLUSIONS: The study conducted adds to the growing literature which has shown that a model-based analysis of xenograft data improves statistical power over the common empirical approach. The analysis conducted showed that a model-based approach, based on the first mathematical model of tumour growth, was able to detect smaller size of effect compared to the empirical approach which is common of such studies. A model-based analysis should allow studies to reduce animal use and experiment length providing effective insights into compound anti-tumour activity.

SELECTION OF CITATIONS
SEARCH DETAIL