Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mucosal Immunol ; 16(3): 275-286, 2023 06.
Article in English | MEDLINE | ID: mdl-36935091

ABSTRACT

The stimulation of local immunity by vaccination is desirable for controlling virus replication in the respiratory tract. However, the local immune stimulatory effects of adjuvanted vaccines administered through the non-mucosal route are poorly understood. Here, we clarify the mechanisms by which non-mucosal inoculation of adjuvants stimulates the plasmacytoid dendritic cell (pDC)-dependent immunoglobulin (Ig)A response in the lungs. After systemic inoculation with type 1 interferon (IFN)-inducing adjuvants, type 1 IFN promotes CXCL9/10/11 release from alveolar endothelial and epithelial cells and recruits CXCR3-expressing pDCs into the lungs. Because adjuvant-activated pulmonary pDCs highly express major histocompatibility complex II, cluster of differentiation 80, and cluster of differentiation 86, transplantation of such cells into the lungs successfully enhances antigen-specific IgA production by the intranasally sensitized vaccine. In contrast, pDC accumulation in the lungs and subsequent IgA production are impaired in pDC-depleted mice and Ifnar1-/- mice. Notably, the combination of systemic inoculation with type 1 IFN-inducing adjuvants and intranasal antigen sensitization protects mice against influenza virus infection due to the pDC-dependent IgA response and type I IFN response. Our results provide insights into the novel mucosal vaccine strategies using non-mucosal inoculated adjuvants.


Subject(s)
Influenza Vaccines , Interferon Type I , Animals , Mice , Immunoglobulin A , Immunoglobulin G , Immunity, Mucosal , Antibodies, Viral , Adjuvants, Immunologic , Administration, Intranasal , Dendritic Cells , Mice, Inbred BALB C
2.
J Toxicol Sci ; 47(11): 439-451, 2022.
Article in English | MEDLINE | ID: mdl-36328534

ABSTRACT

Aluminum salt adjuvants (Als) have been the most widely used adjuvants in vaccines and known to be effective in intramuscular inoculation. However, in rare cases, some Al containing vaccines caused serious adverse events such as chronic pain at the site of the injection. The Als cause mild tissue damage at the inoculation site, allowing the antigen to be locally retained at the inoculation site and thus potentiate innate immunity. This is required to elicit effectiveness of vaccination. However, there is concern that chronic muscle damage might potentially lead to serious adverse events, such as autoimmune disease and movement disorders. In this study, muscle damage caused by several Al containing vaccines were examined in guinea pigs. Mild and moderate inflammation were observed following Al containing split influenza virus vaccine, formalin-inactivated diphtheria-pertussis-tetanus and Salk polio vaccine. While massive inflammation and muscle damage were observed in Al-containing human papillomavirus vaccine-inoculated animals. However, the severities of damage were not associated with their Al contents. Masson's trichrome staining and immunostaining revealed that injured muscle at the inoculated site recovered within one month of vaccination, whereas inflammatory nodules remained. Flow cytometric analyses of the infiltrating cells revealed that the number of CD45+ lymphocytes and potential granulocytes were increased following vaccination. The number of infiltrated cells seemed to be associated with severity of muscle damages. These observations revealed that Al containing vaccine-induced muscle damage is reparable, and severity of transient muscle damages seemed to be determined by type of antigen or types of Al salts rather than Al content.


Subject(s)
Aluminum , Vaccines , Guinea Pigs , Humans , Animals , Aluminum/toxicity , Vaccines/adverse effects , Adjuvants, Immunologic/toxicity , Inflammation/chemically induced , Muscles
3.
J Immunotoxicol ; 18(1): 163-172, 2021 12.
Article in English | MEDLINE | ID: mdl-34761701

ABSTRACT

The importance of alveolar macrophages has been reported in many toxicology/immunology studies. Alveolar macrophages release interleukin (IL)-1α as a damage-associated molecular pattern (DAMP) when stimulated by fine particles. However, it is unclear whether cell isolation procedures affect ex vivo particle-induced responses in primary mouse alveolar macrophages (mAM). In this study, effects of injection buffer volume used to perform bronchoalveolar lavage fluid (BALF) collection to isolate mAM for use in ex vivo particle-induced responses were assessed. Among the mAM obtained from BALF collected using a 0.55 or 0.75 ml, but not a 1.0 ml buffer injection volume, decreased cell viability and IL-1α release were observed when cells were stimulated ex vivo with silica crystal or aluminum salt. Injected buffer composition did not affect the IL-1α release. On the other hand, IL-6 secretion induced by lipopolysaccharide (LPS) did not differ among mAM obtained from BALF collected using the different volumes. Expression levels of cell surface markers like CD11c, SiglecF, and CD64 did not differ among mAM obtained from BALF collected using the different injection buffer volumes. IL-1α release (and also necroptosis) induced by ex vivoparticle stimulation was suppressed by RIPK3 inhibitor or cytochalasin D co-treatment. Decreases in RIPK3 phosphorylation were noted in mAM obtained in BALF collected using the 1.0 ml injection volume compared with mAM obtained in BALF using 0.55 or 0.75 ml buffer. These observations illustrate that larger volumes of buffer used to collect BALF from mice can affect sensitivity of the isolated mAM to ex vivo particle-induced responses by inhibiting their functions.


Subject(s)
Macrophages, Alveolar , Silicon Dioxide , Animals , Bronchoalveolar Lavage Fluid , Cell Separation , Lipopolysaccharides , Mice
4.
PLoS Pathog ; 17(8): e1009890, 2021 08.
Article in English | MEDLINE | ID: mdl-34460865

ABSTRACT

Aluminum hydroxide salts (alum) have been added to inactivated vaccines as safe and effective adjuvants to increase the effectiveness of vaccination. However, the exact cell types and immunological factors that initiate mucosal immune responses to alum adjuvants are unclear. In this study, the mechanism of action of alum adjuvant in nasal vaccination was investigated. Alum has been shown to act as a powerful and unique adjuvant when added to a nasal influenza split vaccine in mice. Alum is cytotoxic in the alveoli and stimulates the release of damage-associated molecular patterns, such as dsDNA, interleukin (IL)-1α, and IL-33. We found that Ag-specific IgA antibody (Ab) production was markedly reduced in IL-33-deficient mice. However, no decrease was observed in Ag-specific IgA Ab production with DNase I treatment, and no decrease was observed in IL-1α/ß or IL-6 production in IL-33-deficient mice. From the experimental results of primary cultured cells and immunofluorescence staining, although IL-1α was secreted by alveolar macrophage necroptosis, IL-33 release was observed in alveolar epithelial cell necroptosis but not in alveolar macrophages. Alum- or IL-33-dependent Ag uptake enhancement and elevation of OX40L expression were not observed. By stimulating the release of IL-33, alum induced Th2 immunity via IL-5 and IL-13 production in group 2 innate lymphoid cells (ILC2s) and increased MHC class II expression in antigen-presenting cells (APCs) in the lung. Our results suggest that IL-33 secretion by epithelial cell necroptosis initiates APC- and ILC2-mediated T cell activation, which is important for the enhancement of Ag-specific IgA Ab production by alum.


Subject(s)
Aluminum Hydroxide/chemistry , Alveolar Epithelial Cells/immunology , Immunoglobulin A/metabolism , Influenza Vaccines/administration & dosage , Interleukin-33/physiology , Orthomyxoviridae Infections/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/administration & dosage , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Animals , Antibodies, Viral/immunology , Antibody Formation , Female , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoglobulin A/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/chemistry , Nasal Mucosa/metabolism , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Vaccination
5.
Front Immunol ; 11: 2171, 2020.
Article in English | MEDLINE | ID: mdl-33013912

ABSTRACT

The efficacy of vaccine adjuvants depends on their ability to appropriately enhance the immunogenicity of vaccine antigens, which is often insufficient in non-adjuvanted vaccines. Genomic analyses of immune responses elicited by vaccine adjuvants provide information that is critical for the rational design of adjuvant vaccination strategies. In this study, biomarker genes from the genomic analyses of lungs after priming were used to predict the efficacy and toxicity of vaccine adjuvants. Based on the results, it was verified whether the efficacy and toxicity of the tested adjuvants could be predicted based on the biomarker gene profiles after priming. Various commercially available adjuvants were assessed by combining them with the split influenza vaccine and were subsequently administered in mice through nasal inoculation. The expression levels of lung biomarker genes within 24 h after priming were analyzed. Furthermore, we analyzed the antibody titer, cytotoxic T lymphocyte (CTL) induction, IgG1/IgG2a ratio, leukopenic toxicity, and cytotoxicity in mice vaccinated at similar doses. The association between the phenotypes and the changes in the expression levels of biomarker genes were analyzed. The ability of the adjuvants to induce the production of antigen-specific IgA could be assessed based on the levels of Timp1 expression. Furthermore, the expression of this gene partially correlated with the levels of other damage-associated molecular patterns in bronchoalveolar lavage fluid. Additionally, the changes in the expression of proteasome- and transporter-related genes involved in major histocompatibility complex class 1 antigen presentation could be monitored to effectively assess the expansion of CTL by adjuvants. The monitoring of certain genes is necessary for the assessment of leukopenic toxicity and cytotoxicity of the tested adjuvant. These results indicate that the efficacy and toxicity of various adjuvants can be characterized by profiling lung biomarker genes after the first instance of immunization. This approach could make a significant contribution to the development of optimal selection and exploratory screening strategies for novel adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Biomarkers , Immunization/methods , Influenza Vaccines/immunology , Lung/drug effects , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Administration, Intranasal , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Bronchoalveolar Lavage Fluid , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Immunologic , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , T-Lymphocyte Subsets/immunology , Th1-Th2 Balance/drug effects , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
6.
Sci Rep ; 9(1): 16249, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700085

ABSTRACT

The safety evaluation of vaccines is critical to avoid the development of side effects in humans. To increase the sensitivity of detection for toxicity tests, it is important to capture not only pathological changes but also physiological changes. 1H nuclear magnetic resonance (NMR) spectroscopy analysis of biofluids produces profiles that show characteristic responses to changes in physiological status. In this study, mouse urine metabolomics analysis with 1H NMR was performed using different influenza vaccines of varying toxicity to assess the usefulness of 1H NMR in evaluating vaccine toxicity. Two types of influenza vaccines were used as model vaccines: a toxicity reference vaccine (RE) and a hemagglutinin split vaccine. According to the blood biochemical analyses, the plasma alanine transaminase levels were increased in RE-treated mice. Changes in metabolite levels between mice administered different types of influenza vaccines were observed in the 1H NMR spectra of urine, and a tendency toward dosage-dependent responses for some spectra was observed. Hierarchical clustering analyses and principal component analyses showed that the changes in various urine metabolite levels allowed for the classification of different types of vaccines. Among them, two liver-derived metabolites were shown to largely contribute to the formation of the cluster. These results demonstrate the possibility that urine metabolomics analysis could provide information about vaccine-induced toxicity and physiological changes.


Subject(s)
Influenza Vaccines/pharmacology , Metabolomics , Urinalysis , Animals , Blood Chemical Analysis , Body Weight/immunology , Female , Leukocytes/cytology , Mice , Vaccines, Inactivated/pharmacology
7.
Biologicals ; 55: 43-52, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30017557

ABSTRACT

Recently, many vaccine adjuvants have been developed; however, most of the newly developed adjuvants have been dropped out of preclinical and clinical trials owing to their unexpected toxicity. Thus, the development of highly quantitative and comparable screening methods for evaluating adjuvant safety is needed. In a previous study, we identified specific biomarkers for evaluating the safety of an intranasal influenza vaccine with CpG K3 adjuvant by comparing biomarker expression. We hypothesized that these biomarkers might be useful for screening newly developed adjuvant safety. We compared the expression of biomarkers in mouse lungs by the intranasal administration of 4 types of adjuvants: Alum, Pam3CSK4, NanoSiO2, and DMXAA with subvirion influenza vaccine. The control adjuvant alum did not show any significant increase in biomarker expression or preclinical parameters; however, NanoSiO2 and Pam3CSK4 increased the expression of biomarkers, such as Timp1 and Csf1. DMXAA at 300 µg induced the expression of over 80% of biomarkers. Hierarchical clustering analysis showed that 300 µg DMXAA was classified in the toxicity reference whole-particle influenza vaccine cluster. FACS analysis to confirm specific phenotypes that the number of T cells decreased in DMXAA-treated mouse lungs. Thus, our biomarkers are useful for initial adjuvant safety and toxicity screening.


Subject(s)
Adjuvants, Immunologic , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Drug Evaluation, Preclinical , Female , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C
8.
Oncotarget ; 9(40): 25751-25763, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29899819

ABSTRACT

Safety evaluation of a human vaccine is critical for vaccine development and for preventing an unexpected adverse reaction in humans. Nonetheless, to date, very few systems have been described for preclinical studies of human adverse reactions in vivo. Previously, we have identified biomarker genes expressed in the lungs for evaluation of influenza vaccine safety, and their usefulness in rodent models and for adjuvant-containing vaccines has already been reported. Here, our purpose was to develop a novel humanized mouse model retaining human innate-immunity-related cells to assess the safety of influenza vaccines using the previously identified biomarker genes. In the present study, we tested whether the two humanized models, a short-term and long-term reconstitution model of NOD/Shi-scid IL2rγnull mice, are suitable for biomarker gene-based safety evaluation. In the short-term model, human CD14+ cells, plasmacytoid dendritic cells, CD4+ and CD8+ T cells, and B cells were retained in the lungs. Among these cells, human CD14+ cells and plasmacytoid dendritic cells were not detected in the lungs of the long-term model. After the vaccination, the expression levels of human biomarker genes were elevated only in the short-term model when the toxicity reference vaccine was inoculated. This phenomenon was not observed in the long-term model. The levels of human cytokines and chemokines in the lungs increased in response to the toxicity reference vaccine in the short-term mouse model. According to these results, the short-term model provides a better platform for evaluating vaccine safety in terms of human peripheral blood mononuclear cell-mediated initial reactions in vivo.

9.
PLoS One ; 13(2): e0191896, 2018.
Article in English | MEDLINE | ID: mdl-29408882

ABSTRACT

Historically, vaccine safety assessments have been conducted by animal testing (e.g., quality control tests and adjuvant development). However, classical evaluation methods do not provide sufficient information to make treatment decisions. We previously identified biomarker genes as novel safety markers. Here, we developed a practical safety assessment system used to evaluate the intramuscular, intraperitoneal, and nasal inoculation routes to provide robust and comprehensive safety data. Influenza vaccines were used as model vaccines. A toxicity reference vaccine (RE) and poly I:C-adjuvanted hemagglutinin split vaccine were used as toxicity controls, while a non-adjuvanted hemagglutinin split vaccine and AddaVax (squalene-based oil-in-water nano-emulsion with a formulation similar to MF59)-adjuvanted hemagglutinin split vaccine were used as safety controls. Body weight changes, number of white blood cells, and lung biomarker gene expression profiles were determined in mice. In addition, vaccines were inoculated into mice by three different administration routes. Logistic regression analyses were carried out to determine the expression changes of each biomarker. The results showed that the regression equations clearly classified each vaccine according to its toxic potential and inoculation amount by biomarker expression levels. Interestingly, lung biomarker expression was nearly equivalent for the various inoculation routes. The results of the present safety evaluation were confirmed by the approximation rate for the toxicity control. This method may contribute to toxicity evaluation such as quality control tests and adjuvant development.


Subject(s)
Adjuvants, Immunologic/adverse effects , Gene Expression Profiling , Influenza Vaccines/adverse effects , Models, Biological , Animals , Mice , Mice, Inbred BALB C
10.
Biologicals ; 50: 100-108, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28838806

ABSTRACT

The leukopenic toxicity test (LTT) is used to evaluate the safety and lot-to-lot consistency of influenza hemagglutinin split vaccine (HAv) and is included in the Japanese Minimum Requirements for Biological Products. LTT assesses the reduced leukocyte levels in murine peripheral blood after HAv administration. However, they require large numbers of animals, and therefore it would be beneficial to develop a more accurate and sensitive alternative method. In this study, we selected biomarkers of leukocyte reduction from 18 previously identified marker genes that were associated with an abnormal toxicity test (ATT). Among these 18 genes, the expressions of 15 marker genes were strongly associated with leukocyte reduction levels. A stepwise single addition multiple regression analysis was used to further extract the genes responsible for leukocyte reduction, with significant (p < 0.25) regression coefficients. The expression of 7 genes significantly predicted the leukocyte reduction. The prediction accuracy of this approach was approximately >90% (mean) for the direct measurement of leukocyte numbers. These results indicate that the expression of these 18 previously identified genes can provide information for both ATT and LTT.


Subject(s)
Cytotoxicity Tests, Immunologic/methods , Influenza Vaccines/immunology , Leukocytes/immunology , Vaccines, Inactivated/immunology , Animals , Biomarkers/metabolism , Female , Gene Expression Profiling/methods , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/pharmacology , Influenza Vaccines/standards , Leukocyte Count , Leukocytes/drug effects , Leukocytes/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Vaccination , Vaccines, Inactivated/pharmacology , Vaccines, Inactivated/standards
11.
Oncotarget ; 7(32): 51027-51043, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27340921

ABSTRACT

Adult T-cell leukemia (ATL) is a malignant disease caused by human T-lymphotropic virus type 1. In aggressive ATL, the response to chemotherapy is extremely poor. We hypothesized that this poor response is due to the existence of chemotherapy-resistant cells, such as leukemic stem cells. Previously, we successfully identified an ATL stem cell (ATLSC) candidate as the c-kit+/CD38-/CD71- cells in an ATL mouse model using Tax transgenic mice. Here, with a new ATL mouse model using HBZ-transgenic mice, we further discovered that the functional ATLSC candidate, which commonly expresses c-kit, is drug-resistant and has the ability to initiate tumors and reconstitute lymphomatous cells. We characterized the ATLSCs as c-kit+/CD4-/CD8- cells and found that they have a similar gene expression profile as T cell progenitors. Additionally, we found that AP-1 gene family members, including Junb, Jund, and Fosb, were up-regulated in the ATLSC fraction. The results of an in vitro assay showed that ATLSCs cultured with cytokines known to promote stem cell expansion, such as stem cell factor (SCF), showed highly proliferative activity and maintained their stem cell fraction. Inhibition of c-kit-SCF signaling with the neutralizing antibody ACK2 affected ATLSC self-renewal and proliferation. Experiments in Sl/Sld mice, which have a mutation in the membrane-bound c-kit ligand, found that ATL development was completely blocked in these mice. These results clearly suggest that the c-kit-SCF signal plays a key role in ATLSC self-renewal and in ATL initiation and disease progression.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Neoplastic Stem Cells/metabolism , Retroviridae Proteins/genetics , Signal Transduction/physiology , Stem Cell Factor/metabolism , Animals , Disease Models, Animal , Disease Progression , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Mice, Transgenic , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-kit/metabolism
12.
PLoS One ; 10(4): e0124392, 2015.
Article in English | MEDLINE | ID: mdl-25909814

ABSTRACT

We have previously identified 17 biomarker genes which were upregulated by whole virion influenza vaccines, and reported that gene expression profiles of these biomarker genes had a good correlation with conventional animal safety tests checking body weight and leukocyte counts. In this study, we have shown that conventional animal tests showed varied and no dose-dependent results in serially diluted bulk materials of influenza HA vaccines. In contrast, dose dependency was clearly shown in the expression profiles of biomarker genes, demonstrating higher sensitivity of gene expression analysis than the current animal safety tests of influenza vaccines. The introduction of branched DNA based-concurrent expression analysis could simplify the complexity of multiple gene expression approach, and could shorten the test period from 7 days to 3 days. Furthermore, upregulation of 10 genes, Zbp1, Mx2, Irf7, Lgals9, Ifi47, Tapbp, Timp1, Trafd1, Psmb9, and Tap2, was seen upon virosomal-adjuvanted vaccine treatment, indicating that these biomarkers could be useful for the safety control of virosomal-adjuvanted vaccines. In summary, profiling biomarker gene expression could be a useful, rapid, and highly sensitive method of animal safety testing compared with conventional methods, and could be used to evaluate the safety of various types of influenza vaccines, including adjuvanted vaccine.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Animals , Branched DNA Signal Amplification Assay/methods , Gene Expression Profiling/methods , Genetic Markers , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Male , Quality Control , Rats , Rats, Wistar , Safety , Up-Regulation , Vaccines, Virosome/administration & dosage , Vaccines, Virosome/adverse effects , Vaccines, Virosome/pharmacology
13.
PLoS One ; 9(7): e101835, 2014.
Article in English | MEDLINE | ID: mdl-25010690

ABSTRACT

Vaccines are beneficial and universal tools to prevent infectious disease. Thus, safety of vaccines is strictly evaluated in the preclinical phase of trials and every vaccine batch must be tested by the National Control Laboratories according to the guidelines published by each country. Despite many vaccine production platforms and methods, animal testing for safety evaluation is unchanged thus far. We recently developed a systems biological approach to vaccine safety evaluation where identification of specific biomarkers in a rat pre-clinical study evaluated the safety of vaccines for pandemic H5N1 influenza including Irf7, Lgals9, Lgalsbp3, Cxcl11, Timp1, Tap2, Psmb9, Psme1, Tapbp, C2, Csf1, Mx2, Zbp1, Ifrd1, Trafd1, Cxcl9, ß2m, Npc1, Ngfr and Ifi47. The current study evaluated whether these 20 biomarkers could evaluate the safety, batch-to-batch and manufacturer-to-manufacturer consistency of seasonal trivalent influenza vaccine using a multiplex gene detection system. When we evaluated the influenza HA vaccine (HAv) from four different manufactures, the biomarker analysis correlated to findings from conventional animal use tests, such as abnormal toxicity test. In addition, sensitivity of toxicity detection and differences in HAvs were higher and more accurate than with conventional methods. Despite a slight decrease in body weight caused by HAv from manufacturer B that was not statistically significant, our results suggest that HAv from manufacturer B is significantly different than the other HAvs tested with regard to Lgals3bp, Tapbp, Lgals9, Irf7 and C2 gene expression in rat lungs. Using the biomarkers confirmed in this study, we predicted batch-to-batch consistency and safety of influenza vaccines within 2 days compared with the conventional safety test, which takes longer. These biomarkers will facilitate the future development of new influenza vaccines and provide an opportunity to develop in vitro methods of evaluating batch-to-batch consistency and vaccine safety as an alternative to animal testing.


Subject(s)
Influenza Vaccines/adverse effects , Safety , Toxicity Tests/methods , Transcriptome/immunology , Animals , Biomarkers/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Male , Rats , Seasons
14.
Biologicals ; 37(1): 8-17, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18805705

ABSTRACT

Vaccines differ from other pharmaceutical products. The quality and safety of batches are regulated to high standards by national regulatory authorities. Various quality control and safety tests have been developed, including the abnormal toxicity test (ATT), which is described in the World Health Organization (WHO) guidelines and in each country's pharmacopoeia. However, the criteria for abnormal results are not well defined in these guidelines. In addition, the animal grade to be used in ATT, classified on the basis of microbial colonization, was not designated in either guideline. In this study, we report a new and improved method of performing ATT, including statistical, histopathological analysis and hematological findings. It is based on the observation that there are body weight changes characteristic to each vaccine, and such standardized changes can be used as references for evaluating test vaccines. In addition, histopathological data are useful for determining vaccine quality and safety. Combined with histopathological examination, the improved ATT will be of great use for evaluating the consistency, quality and safety of different batches of vaccine. The results of these analyses were similar using either 'clean' or specific pathogen-free guinea pigs.


Subject(s)
Body Weight/physiology , Consumer Product Safety/standards , Product Surveillance, Postmarketing/methods , Toxicity Tests/standards , Vaccines, Attenuated/standards , Animals , Body Weight/immunology , Disease/etiology , Female , Guinea Pigs , Japan , Models, Biological , Product Surveillance, Postmarketing/standards , Quality Control , Random Allocation , Specific Pathogen-Free Organisms , Toxicity Tests/methods , Vaccination/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...