Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Pharm Sci ; 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39343099

ABSTRACT

Over recent years, confidence has been gained that predictive stability modeling approaches using statistical tools, prior knowledge and industry experience enable, in many instances, a robust and reliable shelf-life/expiry or retest period prediction for medicinal products. These science and risk-based approaches can compensate for not having a complete real-time stability data set to be included in regulatory applications at the time of initial submission and, thereby, accelerate the availability of new medicines. Examples of predictive stability modeling include accelerated stability assessment procedure (ASAP), advanced kinetic modeling (AKM), and novel modeling approaches that involve the use of Bayesian statistics and Artificial Intelligence (AI) applications such as Machine Learning (ML), with applicability to both synthetic and biological molecules. For biologics, product-specific and platform prior knowledge could be used to overcome model limitations known for non-quantitative stability indicating attributes. A successful ongoing verification approach by comparing the predicted data with real-time stability data would be an appropriate risk management approach which is intended to address regulatory concerns, and further build confidence in the robustness of these predictive modelling approaches with regulatory agencies. Global regulatory acceptance of stability modeling could allow patients to receive potential life-saving medications faster without compromising quality, safety or efficacy.

2.
Gastroenterology ; 142(7): 1516-25.e3, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22430395

ABSTRACT

BACKGROUND & AIMS: CD4+ T-regulatory (Treg) cells suppress immune responses and control self-tolerance and immunity to pathogens, cancer, and alloantigens. Most pathogens activate Treg cells to minimize immune-mediated tissue damage and prevent clearance, which promotes chronic infections. However, hepatitis A virus (HAV) temporarily inhibits Treg-cell functions. We investigated whether the interaction of HAV with its cellular receptor 1 (HAVCR1), a T-cell co-stimulatory molecule, inhibits the function of Treg cells to control HAV infection. METHODS: We studied the effects of HAV interaction with HAVCR1 on human T cells using binding, signal transduction, apoptosis, activation, suppression, cytokine production, and confocal microscopy analyses. Cytokines were analyzed in sera from 14 patients with HAV infection using bead arrays. RESULTS: Human Treg cells constitutively express HAVCR1. Binding of HAV to HAVCR1 blocked phosphorylation of Akt, prevented activation of the T-cell receptor, and inhibited function of Treg cells. At the peak viremia, patients with acute HAV infection had no Treg-cell suppression function, produced low levels of transforming growth factor-ß , which limited leukocyte recruitment and survival, and produced high levels of interleukin-22, which prevented liver damage. CONCLUSIONS: Interaction between HAV and its receptor HAVCR1 inhibits Treg-cell function, resulting in an immune imbalance that allows viral expansion with limited hepatocellular damage during early stages of infection-a characteristic of HAV pathogenesis. The mechanism by which HAV is cleared in the absence of Treg-cell function could be used as a model to develop anticancer therapies, modulate autoimmune and allergic responses, and prevent transplant rejection.


Subject(s)
Hepatitis A virus/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , T-Lymphocytes, Regulatory/immunology , Virus Attachment , Cell Line , Hepatitis A/immunology , Hepatitis A/metabolism , Hepatitis A Virus Cellular Receptor 1 , Humans , Interleukins/biosynthesis , Proto-Oncogene Proteins c-akt , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Transforming Growth Factor beta1/blood , Interleukin-22
3.
Bioorg Med Chem ; 21(20): 6224-32, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23755885

ABSTRACT

The bioactivity of a CpG-containing phosphorothioate DNA oligonucleotide with thermolytic 2-(N-formyl-N-methylamino)ethyl (fma) thiophosphate groups in mice led us to investigate the parameters affecting the internalization of these thermosensitive DNA prodrugs in various cell lines. Flow cytometry and confocal microscopy analyses indicate that 5'-fluoresceinated fma-phosphorothioate DNA sequences are poorly internalized in Vero, HeLa and GC-2 cells. However, when four fma-thiophosphate groups of a 15-nucleotide long oligothymidylate prodrug are replaced with 3-(N,N-dimethylamino)prop-1-yl thiophosphate functions, internalization of the positively charged prodrug, under physiological conditions, increased fourfold in HeLa and 40-fold in Vero or GC-2 cells. No cytotoxic effects are observed in Vero cells even at an extracellular prodrug concentration of 50 µM over a period of 72 h. Confocal microscopy studies show that internalization of the positively charged oligothymidylate prodrug in Vero cells is time-dependent with early trafficking of the DNA sequence through endosomal vesicles and, eventually, to the nucleus of the cells. Thus, the incorporation of four 3-(N,N-dimethylamino)prop-1-yl thiophosphate groups into thermosentive fma-phosphorothioate DNA prodrugs is an attractive strategy for efficient cellular internalization of these nucleic acid-based drugs for potential therapeutic indications.


Subject(s)
DNA/chemistry , DNA/pharmacokinetics , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Chlorocebus aethiops , Fluoresceins/chemistry , HeLa Cells , Humans , Lipids/chemistry , Lipids/pharmacokinetics , Mice , Microscopy, Confocal , Thionucleotides/chemistry , Thionucleotides/pharmacokinetics , Vero Cells
4.
Cell Mol Immunol ; 14(1): 90-107, 2017 01.
Article in English | MEDLINE | ID: mdl-27569560

ABSTRACT

Neonates are at increased risk of viral encephalopathies that can result in neurological dysfunction, seizures, permanent disability and even death. The neurological damage results from the combined effect of the virus and the immune response it elicits, thus finding tools to facilitate viral clearance from central nervous system (CNS) while minimizing neuron damage remains a critical challenge. Neonatal mice inoculated intraperitoneally with Tacaribe virus (TCRV) develop seizures, hindlimb paralysis and death within 15 days of inoculation. TCRV localizes to the CNS within days of challenge, primarily infecting astrocytes in the cerebellum and brain stem. We show that infection leads to inflammation, T cell and monocyte infiltration into the cerebellar parenchyma, apoptosis of astrocytes, neuronal degeneration and loss of Purkinje cells. Infiltrating antigen-specific T cells fail to clear the virus but drive the disease, as T-cell-deficient CD3ɛ KO mice survive TCRV infection with minimal inflammation or clinical manifestations despite no difference in CNS viral loads in comparison with T-cell sufficient mice. CD8+ T cells drive the pathology, which even in the absence of CD4+ T-cell help, infiltrate the parenchyma and mediate the apoptotic loss of cerebellar astrocytes, neurodegeneration and loss of Purkinje cells resulting in loss of balance, paralysis and death. CD4+ T cells are also pathogenic inducing gliosis and inflammation in the cerebellum and cerebrum that are associated with wasting and death several weeks after CD4+ T-cell transfer. These data demonstrate distinct pathogenic effects of CD4+ and CD8+ T cells and identify them as possible therapeutic targets.


Subject(s)
Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/physiology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Meningoencephalitis/immunology , Meningoencephalitis/virology , Animals , Animals, Newborn , Apoptosis , Arenaviridae Infections/pathology , Astrocytes/pathology , Central Nervous System/pathology , Central Nervous System/virology , Gliosis/pathology , Meningoencephalitis/pathology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Nerve Degeneration/pathology , Neurons/pathology , Purkinje Cells/pathology , T-Lymphocytes/immunology
5.
J Leukoc Biol ; 91(1): 147-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22058422

ABSTRACT

Human I-IFNs include IFN-ß and 13 independently regulated subtypes of IFN-α (I-IFNs). TLR7 and -9 induce I-IFNs, but it is unknown whether their subtype repertoire is similar. This study used new PCR arrays that selectively amplify individual I-IFN subtype genes of human and nonhuman primates to characterize the TLR7- and -9-mediated IFN response in vitro and in vivo. We show that in human PBMCs, TLR7 agonists induce a rapid burst of I-IFN transcripts, consisting primarily of IFN-α1/13, -α2, and -α14. In contrast, TLR9 agonists, regardless of the type used (CpG C-, B-, or D-ODN), prompted slower but sustained expression of IFN-α1/13, -α2, -α7, -α8, -α10, -α14, -α16, and -α21. These qualitative differences were translated downstream as differences in the pattern of IFN-inducible genes. In macaque PBMCs, imiquimod produced a short burst of IFN mRNA, dominated by IFN-α8, whereas C- or D-ODN induced a greater than tenfold increase in transcripts for all I-IFN subtypes by 12 h of culture. Differences were more evident in vivo, where TLR7 and -9 agonists induced significantly different levels of I-IFN transcripts in skin. Although the rates of gene transcription differed significantly for individual TLR9 agonists, their IFN-α subtype signature was almost identical, indicating that the type of receptor dictates the quality of the I-IFN response in vitro and in vivo. These results may underlie the differential therapeutic effects of TLR7 and -9 agonists and should inform future clinical studies.


Subject(s)
Aminoquinolines/pharmacology , Gene Expression Regulation/drug effects , Interferon Type I/biosynthesis , Toll-Like Receptor 7/agonists , Toll-Like Receptor 9/agonists , Animals , Gene Expression Regulation/immunology , Humans , Imiquimod , Interferon Type I/genetics , Interferon Type I/physiology , Macaca mulatta , Oligonucleotide Array Sequence Analysis/methods , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics
6.
Biomaterials ; 29(36): 4808-14, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801571

ABSTRACT

Heparin and low molecular heparins are extensively used in the treatment of a wide range of diseases in addition to their classic anticoagulant activity and can be found coating medical devices such as catheters, stents and filters. Early in 2008, a sharp increase in heparin-associated severe adverse events, including over 80 deaths, was linked to the presence of a contaminant identified as hypersulfated chondroitin sulfate (OS-CS). OS-CS is one of several oversulfated glycosaminoglycans (GAGs) of different origins that can potentially cause similar clinical problems underscoring the need to develop robust screening methods for contaminants in existing and future lots of heparin. This study demonstrates that oversulfated GAGs block the activity of Taq polymerase used for real time PCR. Based on this finding we developed a simple, rapid, sensitive and high throughput screening method to detect and quantify oversulfated chondroitin sulfate (OS-CS) and other potential oversulfated contaminants in commercial lots of heparin. This method requires less than 100 miliUnits (mU) of heparin as starting material, therefore avoiding the need to lyophilize and concentrate samples, and has a limit of detection of <1 ng for all oversulfated GAGs tested.


Subject(s)
Chondroitin Sulfates/analysis , Drug Contamination , Heparin/analysis , Polymerase Chain Reaction/methods , Taq Polymerase/antagonists & inhibitors , Cell Line, Tumor , Humans , RNA, Ribosomal, 18S/genetics
7.
Immunity ; 26(3): 299-310, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363299

ABSTRACT

The T cell immunoglobulin mucin (TIM) receptors are involved in the regulation of immune responses, autoimmunity, and allergy. Structures of the N-terminal ligand binding domain of the murine mTIM-1 and mTIM-2 receptors revealed an immunoglobulin (Ig) fold, with four Cys residues bridging a distinctive CC' loop to the GFC beta-sheet. The structures showed two ligand-recognition modes in the TIM family. The mTIM-1 structure identified a homophilic TIM-TIM adhesion interaction, whereas the mTIM-2 domain formed a dimer that prevented homophilic binding. Biochemical, mutational, and cell adhesion analyses confirmed the divergent ligand-binding modes revealed by the structures. Structural features characteristic of mTIM-1 appear conserved in human TIM-1, which also mediated homophilic interactions. The extracellular mucin domain enhanced binding through the Ig domain, modulating TIM receptor functions. These results explain the divergent immune functions described for the murine receptors and the role of TIM-1 as a cell adhesion receptor in renal regeneration and cancer.


Subject(s)
Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Dimerization , Hepatitis A Virus Cellular Receptor 1 , Humans , Immunity , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Receptors, Immunologic/chemistry
8.
J Virol ; 81(7): 3437-46, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17229699

ABSTRACT

The hepatitis A virus cellular receptor 1 (HAVCR1/TIM1), a member of the T-cell immunoglobulin mucin (TIM) family, is an important atopy susceptibility gene in humans. The exact natural function of HAVCR1/TIM1 and the inverse association between HAV infection and prevention of atopy are not well understood. To identify natural ligands of human HAVCR1/TIM1, we used an expression cloning strategy based on the binding of dog cells transfected with a human lymph node cDNA library to a HAVCR1/TIM1 Fc fusion protein. The transfected cells that bound to the human HAVCR1/TIM1 Fc contained cDNA of human immunoglobulin alpha 1 heavy (Igalpha1) and lambda light (Iglambda) chain and secreted human IgA1lambda antibody that bound to the cell surface. Cotransfection of the isolated Igalpha1 and Iglambda cDNAs to naïve dog cells resulted in the secretion of IgA1lambda that bound to HAVCR1/TIM1 Fc but not to a poliovirus receptor Fc fusion protein in a capture enzyme-linked immunosorbent assay. The interaction of HAVCR1/TIM1 with IgA was inhibited by monoclonal antibodies (MAbs) against Igalpha1 and Iglambda, excess IgA1lambda, or anti-HAVCR1/TIM1 MAb. IgA did not inhibit HAV infection of African green monkey cells, suggesting that the IgA and the virus binding sites are in different epitopes on HAVCR1/TIM1. IgA enhanced significantly the neutralization of HAV by HAVCR1/TIM1 Fc. Our results indicate that IgA1lambda is a specific ligand of HAVCR1/TIM1 and that their association has a synergistic effect in virus-receptor interactions.


Subject(s)
Hepatitis A virus/immunology , Immunoglobulin A/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Receptors, Virus/immunology , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Gene Expression , Hepatitis A Virus Cellular Receptor 1 , Humans , Ligands , Membrane Glycoproteins/genetics , Protein Binding , Receptors, Virus/genetics
9.
Vaccine ; 23(6): 840-5, 2004 Dec 21.
Article in English | MEDLINE | ID: mdl-15542209

ABSTRACT

In the present report, we characterized the immune response and the protection conferred by recombinant baculoviruses or infected insect cells expressing the fusions gp64-P1 and gp64-site A FMDV antigens. Mice, vaccinated intraperitoneally with gp64-P1 immunogens, showed a low-antibody response and a variable degree of protection. However, when mice received recombinant baculoviruses or infected insect cells expressing the fusion protein gp64-site A, high-ELISA and seroneutralizing titers (SNT) against FMDV were elicited. All mice immunized with Sf9 cells expressing FMDV site A developed a protective immune response against challenge with virulent FMDV, indicating that the baculovirus display of foreign epitopes is a promising approach to biosynthetic vaccines.


Subject(s)
Aphthovirus/immunology , Cysteine Endopeptidases/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , 3C Viral Proteases , Animals , Aphthovirus/genetics , Baculoviridae/genetics , Cells, Cultured , Cysteine Endopeptidases/genetics , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spodoptera/cytology , Viral Fusion Proteins/genetics , Viral Proteins/genetics
10.
J Virol ; 77(2): 1219-26, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502839

ABSTRACT

In this work we analyze the antigenic properties and the stability in cell culture of virus mutants recovered upon challenge of peptide-vaccinated cattle with foot-and-mouth disease virus (FMDV) C3 Arg85. Previously, we showed that a significant proportion of 29 lesions analyzed (41%) contained viruses with single amino acid replacements (R141G, L144P, or L147P) within a major antigenic site located at the G-H loop of VP1, known to participate also in interactions with integrin receptors. Here we document that no replacements at this site were found in viruses from 12 lesions developed in six control animals upon challenge with FMDV C3 Arg85. Sera from unprotected, vaccinated animals exhibited poor neutralization titers against mutants recovered from them. Sequence analyses of the viruses recovered upon 10 serial passages in BHK-21 and FBK-2 cells in the presence of preimmune (nonneutralizing) sera revealed that mutants reverted to the parental sequence, suggesting an effect of the amino acid replacements in the interaction of the viruses with cells. Parallel passages in the presence of subneutralizing concentrations of immune homologous sera resulted in the maintenance of mutations R141G and L147P, while mutation L144P reverted to the C3 Arg85 sequence. Reactivity with a panel of FMDV type C-specific monoclonal antibodies indicated that mutant viruses showed altered antigenicity. These results suggest that the selective pressure exerted by host humoral immune response can play a role in both the selection and stability of antigenic FMDV variants and that such variants can manifest alterations in cell tropism.


Subject(s)
Antigens, Viral/immunology , Evolution, Molecular , Foot-and-Mouth Disease Virus/immunology , Tropism , Animals , Antibodies, Monoclonal/immunology , Antigens, Viral/genetics , Base Sequence , Cattle , Cell Line , Cricetinae , DNA Primers , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/growth & development , Serial Passage
SELECTION OF CITATIONS
SEARCH DETAIL