Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 11(1)2023 01.
Article in English | MEDLINE | ID: mdl-36593065

ABSTRACT

BACKGROUND: Agonistic anti-CD40 monoclonal antibodies (mAbs) have emerged as promising immunotherapeutic compounds with impressive antitumor effects in mouse models. However, preclinical and clinical studies faced dose-limiting toxicities mediated by necroinflammatory liver disease. An effective prophylactic treatment for liver immune-related adverse events that does not suppress specific antitumor immunity remains to be found. METHODS: We used different mouse models and time-resolved single-cell RNA-sequencing to characterize the pathogenesis of anti-CD40 mAb induced liver toxicity. Subsequently, we developed an antibody-based treatment protocol to selectively target red blood cells (RBCs) for erythrophagocytosis in the liver, inducing an anti-inflammatory liver macrophage reprogramming. RESULTS: We discovered that CD40 signaling in Clec4f+ Kupffer cells is the non-redundant trigger of anti-CD40 mAb-induced liver toxicity. Taking advantage of the highly specific functionality of liver macrophages to clear antibody-tagged RBCs from the blood, we hypothesized that controlled erythrophagocytosis and the linked anti-inflammatory signaling by the endogenous metabolite heme could be exploited to reprogram liver macrophages selectively. Repeated low-dose administration of a recombinant murine Ter119 antibody directed RBCs for selective phagocytosis in the liver and skewed the phenotype of liver macrophages into a Hmoxhigh/Marcohigh/MHCIIlow anti-inflammatory phenotype. This unique mode of action prevented necroinflammatory liver disease following high-dose administration of anti-CD40 mAbs. In contrast, extrahepatic inflammation, antigen-specific immunity, and antitumor activity remained unaffected in Ter119 treated animals. CONCLUSIONS: Our study offers a targeted approach to uncouple CD40-augmented antitumor immunity in peripheral tissues from harmful inflammatoxicity in the liver.


Subject(s)
Antineoplastic Agents , Neoplasms , Mice , Animals , Kupffer Cells/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Liver
2.
Sci Transl Med ; 11(506)2019 08 21.
Article in English | MEDLINE | ID: mdl-31434758

ABSTRACT

Treatment of autoimmune and inflammatory diseases typically involves immune suppression. In an opposite strategy, we show that administration of the highly inflammatory erythrocyte-specific antibody Ter119 into mice remodels the monocyte cellular landscape, leading to resolution of inflammatory disease. Ter119 with intact Fc function was unexpectedly therapeutic in the K/BxN serum transfer model of arthritis. Similarly, it rapidly reversed clinical disease progression in collagen antibody-induced arthritis (CAIA) and collagen-induced arthritis and completely corrected CAIA-induced increase in monocyte Fcγ receptor II/III expression. Ter119 dose-dependently induced plasma chemokines CCL2, CCL5, CXCL9, CXCL10, and CCL11 with corresponding alterations in monocyte percentages in the blood and liver within 24 hours. Ter119 attenuated chemokine production from the synovial fluid and prevented the accumulation of inflammatory cells and complement components in the synovium. Ter119 could also accelerate the resolution of hypothermia and pulmonary edema in an acute lung injury model. We conclude that this inflammatory anti-erythrocyte antibody simultaneously triggers a highly efficient anti-inflammatory effect with broad therapeutic potential.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Erythrocytes/immunology , Inflammation/drug therapy , Acute Lung Injury/blood , Acute Lung Injury/complications , Anemia/blood , Anemia/complications , Animals , Arthritis/blood , Arthritis/complications , Arthritis, Experimental/blood , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Blood Transfusion , Cell Movement , Chemokines/metabolism , Disease Models, Animal , Disease Progression , Glycosylation , Immunoglobulin G/metabolism , Inflammation/blood , Inflammation/complications , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, SCID , Monocytes/metabolism , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/complications , Purpura, Thrombocytopenic, Idiopathic/pathology , Receptors, IgG/metabolism
3.
Respir Res ; 20(1): 99, 2019 May 22.
Article in English | MEDLINE | ID: mdl-31118031

ABSTRACT

BACKGROUND: Recurrent and persistent infections are known to affect airways of patients with Primary Immunodeficiency despite appropriate replacement immunoglobulin serum levels. Interestingly, patients with Chronic Obstructive Pulmonary Disease or with non-CF bronchiectasis also show similar susceptibility to such infections. This may be due to the limited availability of immunoglobulins from the systemic circulation in the conductive airways, resulting in local immunodeficiency. Topical application of nebulized plasma-derived immunoglobulins may represent a means to address this deficiency. In this study, we assessed the feasibility of nebulizing plasma-derived immunoglobulins and delivering them into the airways of rats and non-human primates. METHODS: Distinct human plasma-derived immunoglobulin isotype preparations were nebulized with an investigational eFlow® nebulizer and analyzed in vitro or deposited into animals. Biochemical and immunohistological analysis of nebulized immunoglobulins were then performed. Lastly, efficacy of topically applied human plasma-derived immunoglobulins was assessed in an acute Streptococcus pneumoniae respiratory infection in mice. RESULTS: Characteristics of the resulting aerosols were comparable between preparations, even when using solutions with elevated viscosity. Neither the structural integrity nor the biological function of nebulized immunoglobulins were compromised by the nebulization process. In animal studies, immunoglobulins levels were assessed in plasma, broncho-alveolar lavages (BAL) and on lung sections of rats and non-human primates in samples collected up to 72 h following application. Nebulized immunoglobulins were detectable over 48 h in the BAL samples and up to 72 h on lung sections. Immunoglobulins recovered from BAL fluid up to 24 h after inhalation remained structurally and functionally intact. Importantly, topical application of human plasma-derived immunoglobulin G into the airways of mice offered significant protection against acute pneumococcal pneumonia. CONCLUSION: Taken together our data demonstrate the feasibility of topically applying plasma-derived immunoglobulins into the lungs using a nebulized liquid formulation. Moreover, topically administered human plasma-derived immunoglobulins prevented acute respiratory infection.


Subject(s)
Immunoglobulin A/administration & dosage , Immunoglobulin G/administration & dosage , Immunoglobulin M/administration & dosage , Lung/drug effects , Nebulizers and Vaporizers/trends , Administration, Topical , Animals , Dose-Response Relationship, Drug , Humans , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Lung/metabolism , Macaca fascicularis , Mice, Inbred C57BL , Mice, Transgenic , Primates , Rats , Rats, Sprague-Dawley , Species Specificity
4.
Mucosal Immunol ; 12(4): 1013-1024, 2019 07.
Article in English | MEDLINE | ID: mdl-31105268

ABSTRACT

Recurrent and persistent airway infections remain prevalent in patients with primary immunodeficiency (PID), despite restoration of serum immunoglobulin levels by intravenous or subcutaneous plasma-derived IgG. We investigated the effectiveness of different human Ig isotype preparations to protect mice against influenza when delivered directly to the respiratory mucosa. Four polyvalent Ig preparations from pooled plasma were compared: IgG, monomeric IgA (mIgA), polymeric IgA-containing IgM (IgAM) and IgAM associated with the secretory component (SIgAM). To evaluate these preparations, a transgenic mouse expressing human FcαRI/CD89 within the myeloid lineage was created. CD89 was expressed on all myeloid cells in the lung and blood except eosinophils, reflecting human CD89 expression. Intranasal administration of IgA-containing preparations was less effective than IgG in reducing pulmonary viral titres after infection of mice with A/California/7/09 (Cal7) or the antigenically distant A/Puerto Rico/8/34 (PR8) viruses. However, IgA reduced weight loss and inflammatory mediator expression. Both IgG and IgA protected mice from a lethal dose of PR8 virus and for mIgA, this effect was partially CD89 dependent. Our data support the beneficial effect of topically applied Ig purified from pooled human plasma for controlling circulating and non-circulating influenza virus infections. This may be important for reducing morbidity in PID patients.


Subject(s)
Antigens, CD/genetics , Gene Expression , Immunoglobulin Isotypes/immunology , Receptors, Fc/genetics , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antigens, CD/immunology , Cytokines/biosynthesis , Disease Models, Animal , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Isotypes/administration & dosage , Immunophenotyping , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neutralization Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Protein Binding/immunology , Receptors, Fc/immunology
5.
J Immunol ; 200(8): 2542-2553, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29531170

ABSTRACT

Activation of Fc receptors and complement by immune complexes is a common important pathogenic trigger in many autoimmune diseases and so blockade of these innate immune pathways may be an attractive target for treatment of immune complex-mediated pathomechanisms. High-dose IVIG is used to treat autoimmune and inflammatory diseases, and several studies demonstrate that the therapeutic effects of IVIG can be recapitulated with the Fc portion. Further, recent data indicate that recombinant multimerized Fc molecules exhibit potent anti-inflammatory properties. In this study, we investigated the biochemical and biological properties of an rFc hexamer (termed Fc-µTP-L309C) generated by fusion of the IgM µ-tailpiece to the C terminus of human IgG1 Fc. Fc-µTP-L309C bound FcγRs with high avidity and inhibited FcγR-mediated effector functions (Ab-dependent cell-mediated cytotoxicity, phagocytosis, respiratory burst) in vitro. In addition, Fc-µTP-L309C prevented full activation of the classical complement pathway by blocking C2 cleavage, avoiding generation of inflammatory downstream products (C5a or sC5b-9). In vivo, Fc-µTP-L309C suppressed inflammatory arthritis in mice when given therapeutically at approximately a 10-fold lower dose than IVIG, which was associated with reduced inflammatory cytokine production and complement activation. Likewise, administration of Fc-µTP-L309C restored platelet counts in a mouse model of immune thrombocytopenia. Our data demonstrate a potent anti-inflammatory effect of Fc-µTP-L309C in vitro and in vivo, likely mediated by blockade of FcγRs and its unique inhibition of complement activation.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Autoimmune Diseases/immunology , Complement System Proteins/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , Complement Activation/immunology , Humans , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Phagocytosis/immunology , Receptors, IgG/immunology
6.
Stem Cell Reports ; 7(3): 571-582, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27499199

ABSTRACT

Hematopoiesis is a multistage process involving the differentiation of stem and progenitor cells into distinct mature cell lineages. Here we present Haemopedia, an atlas of murine gene-expression data containing 54 hematopoietic cell types, covering all the mature lineages in hematopoiesis. We include rare cell populations such as eosinophils, mast cells, basophils, and megakaryocytes, and a broad collection of progenitor and stem cells. We show that lineage branching and maturation during hematopoiesis can be reconstructed using the expression patterns of small sets of genes. We also have identified genes with enriched expression in each of the mature blood cell lineages, many of which show conserved lineage-enriched expression in human hematopoiesis. We have created an online web portal called Haemosphere to make analyses of Haemopedia and other blood cell transcriptional datasets easier. This resource provides simple tools to interrogate gene-expression-based relationships between hematopoietic cell types and genes of interest.


Subject(s)
Blood Cells/cytology , Blood Cells/metabolism , Computational Biology , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling , Humans , Mice , Web Browser
7.
Vaccine ; 32(30): 3861-8, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24928062

ABSTRACT

In Australia, during the 2010 Southern Hemisphere (SH) influenza season, there was an unexpected increase in post-marketing adverse event reports of febrile seizures (FS) in children under 5 years of age shortly after vaccination with the CSL trivalent influenza vaccine (CSL 2010 SH TIV) compared to previous CSL TIVs and other licensed 2010 SH TIVs. The present study describes the outcomes of a series of in vitro experiments directed at elucidating the root cause. The scientific investigations found that a subset of paediatric donors displayed elevated cytokine/chemokine responses to the CSL 2010 SH TIV but not to previous CSL TIVs nor other 2010 SH TIVs. The induction of elevated cytokines/chemokines in paediatric whole blood correlated with elevated NF-κB activation in a HEK293 cell reporter assay. The data indicate that the introduction of the B/Brisbane/60/2008 strain within the CSL manufacturing process (such as occurred in the preceding 2009/10 NH season) appears to have raised the pyrogenic potential of the CSL 2009/10 NH TIV but that this was insufficient to elicit FS in children <5 years. The 2010 SH season coincided with the first introduction of the H1N1 A/California/07/2009 in combination with the B/Brisbane/60/2008 strain. Our data demonstrates that the introduction of the H1N1 A/California/07/2009 (and to a much lesser degree, H3N2 A/Wisconsin/15/2009) in combination with B/Brisbane/60/2008 (as expressed through the CSL method of manufacture) combined and likely compounded the bioactivity of the CSL 2010 SH TIV. This was associated with stronger immune responses, which in a proportion of children <5 years were associated with FS. The assays and systems developed during these investigations should greatly assist in determining the bioactivity of new influenza strains, and thus aid with the manufacture of CSL TIVs indicated for use in the paediatric population.


Subject(s)
Chemokines/immunology , Cytokines/immunology , Influenza Vaccines/adverse effects , Influenza, Human/prevention & control , Seizures, Febrile/chemically induced , Australia/epidemiology , Child , Child, Preschool , HEK293 Cells , Humans , Infant , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza B virus , Influenza Vaccines/immunology , NF-kappa B/metabolism , Product Surveillance, Postmarketing
8.
Vaccine ; 32(30): 3869-76, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24681272

ABSTRACT

In Australia, during the 2010 Southern Hemisphere (SH) influenza season, there was an unexpected increase in post-marketing adverse event reports of febrile seizures (FS) in children under 5 years of age shortly after vaccination with the CSL 2010 SH trivalent influenza vaccine (CSL 2010 SH TIV) compared to previous CSL TIVs and other licensed 2010 SH TIVs. In an accompanying study, we described the contribution to these adverse events of the 2010 SH influenza strains as expressed in the CSL 2010 SH TIV using in vitro cytokine/chemokine secretion from whole blood cells and induction of NF-κB activation in HEK293 reporter cells. The aim of the present study was to identify the root cause components that elicited the elevated cytokine/chemokine and NF-κB signature. Our studies demonstrated that the pyrogenic signal was associated with a heat-labile, viral-derived component(s) in the CSL 2010 SH TIV. Further, it was found that viral lipid-mediated delivery of short, fragmented viral RNA was the key trigger for the increased cytokine/chemokine secretion and NF-κB activation. It is likely that the FS reported in children <5 years were due to a combination of the new influenza strains included in the 2010 SH TIV and the CSL standard method of manufacture preserving strain-specific viral components of the new influenza strains (particularly B/Brisbane/60/2008 and to a lesser extent H1N1 A/California/07/2009). These combined to heighten immune activation of innate immune cells, which in a small proportion of children <5 years of age is associated with the occurrence of FS. The data also demonstrates that CSL TIVs formulated with increased levels of splitting agent (TDOC) for the B/Brisbane/60/2008 strain can attenuate the pro-inflammatory signals in vitro, identifying a potential path forward for generating a CSL TIV indicated for use in children <5 years.


Subject(s)
Influenza Vaccines/adverse effects , Lipids/administration & dosage , RNA, Viral/administration & dosage , Seizures, Febrile/chemically induced , Australia/epidemiology , Chemokines/immunology , Child, Preschool , Cytokines/immunology , Drug Carriers/administration & dosage , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype , Influenza B virus , Influenza, Human/prevention & control , NF-kappa B/metabolism , Product Surveillance, Postmarketing
9.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24610009

ABSTRACT

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Subject(s)
Cholesterol/immunology , Immunity/immunology , Inflammasomes/immunology , Interleukin-18/immunology , Phospholipids/immunology , Saponins/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Blotting, Western , Cell Survival/drug effects , Cell Survival/immunology , Cholesterol/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Combinations , Humans , Immunity/drug effects , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lysosomes/drug effects , Lysosomes/immunology , Lysosomes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phospholipids/pharmacology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Saponins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Expert Rev Vaccines ; 12(7): 733-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23885819

ABSTRACT

It is thought that the development of vaccines for the treatment of infectious diseases and cancer is likely to be achieved in the coming decades. This is partially due to a better understanding of the regulatory networks connecting innate with adaptive immune responses. The innate immune response is triggered by the recognition of conserved pathogen-associated molecular patterns by germ line-coded pattern recognition receptors. Several families of pattern recognition receptors have been characterized, including Toll-like receptors and nucleotide-binding domain receptors. The identification of their ligands has driven the development of novel adjuvants many of which have been tested in vaccine clinical trials. Here, the authors review recent preclinical data and clinical trial results supporting the view that combinations of adjuvants are the way forward in vaccine design. Multiadjuvanted vaccines can stimulate the broad and robust protective immune responses required to fight chronic infectious diseases and cancer.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Vaccination/methods , Vaccines/immunology , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Vaccines/administration & dosage
11.
Cancer Immunol Immunother ; 62(2): 321-35, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22923192

ABSTRACT

Inhibitor of apoptosis proteins (IAPs) are critical in regulating apoptosis resistance in cancer. Antagonists of IAPs, such as LCL161, are in clinical development and show promise as anti-cancer agents for solid and hematological cancers, with preliminary data suggesting they may act as immunomodulators. IAP antagonists hypersensitize tumor cells to TNF-α-mediated apoptosis, an effect that may work in synergy with that of cancer vaccines. This study aimed to further investigate the immunomodulatory properties of LCL161 on human immune subsets. T lymphocytes treated with LCL161 demonstrated significantly enhanced cytokine secretion upon activation, with little effect on CD4 and CD8 T-cell survival or proliferation. LCL161 treatment of peripheral blood mononuclear cells significantly enhanced priming of naïve T cells with synthetic peptides in vitro. Myeloid dendritic cells underwent phenotypic maturation upon IAP antagonism and demonstrated a reduced capacity to cross-present a tumor antigen-based vaccine. These effects are potentially mediated through an observed activation of the canonical and non-canonical NF-κB pathways, following IAP antagonism with a resulting upregulation of anti-apoptotic molecules. In conclusion, this study demonstrated the immunomodulatory properties of antagonists at physiologically relevant concentrations and indicates their combination with immunotherapy requires further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , T-Lymphocytes/drug effects , Thiazoles/pharmacology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/pharmacology , Apoptosis/drug effects , Cancer Vaccines/pharmacology , Cells, Cultured , Combined Modality Therapy , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Immunotherapy , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , NF-kappa B/metabolism , Peptides/pharmacology , T-Lymphocytes/immunology , Up-Regulation/drug effects
12.
Vaccine ; 30(51): 7400-6, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23063831

ABSTRACT

During the 2010 Southern Hemisphere (SH) influenza season, there was an unexpected increase in the number of febrile reactions reported in the paediatric population in Australia shortly after vaccination with the CSL 2010 trivalent influenza vaccine (TIV) compared to previous seasons. A series of scientific investigations were initiated to identify the root cause of these adverse events, including in vitro cytokine/chemokine assays following stimulation of adult and paediatric whole blood, as well as mammalian cell lines and primary cells, profiling of molecular signatures using microarrays, and in vivo studies in rabbits, ferrets, new born rats and rhesus non-human primates (NHPs). Various TIVs (approved commercial vaccines as well as re-engineered TIVs) and their individual monovalent pool harvest (MPH) components were examined in these assays and in animal models. Although the scientific investigations are ongoing, the current working hypothesis is that the increase in febrile adverse events reported in Australia after vaccination with the CSL 2010 SH TIV may be due to a combination of both the introduction of three entirely new strains in the CSL 2010 SH TIV, and differences in the manufacturing processes used to manufacture CSL TIVs compared to other licensed TIVs on the market. Identification of the causal component(s) may result in the identification of surrogate assays that can assist in the formulation of TIVs to minimise the future incidence of febrile reactions in the paediatric population.


Subject(s)
Fever/chemically induced , Fever/etiology , Influenza Vaccines/adverse effects , Influenza, Human/prevention & control , Adolescent , Adult , Animals , Australia , Cells, Cultured , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Influenza Vaccines/administration & dosage , Male
13.
J Med Microbiol ; 61(Pt 7): 935-943, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22442293

ABSTRACT

The ISCOMATRIX adjuvant has antigen delivery and presentation properties as well as immunomodulatory capabilities, which combine to provide enhanced and accelerated immune responses. The responses are broad, including a range of subclasses of antibodies as well as CD4(+) and CD8(+) T-cells. A range of ISCOMATRIX vaccines (ISCOMATRIX adjuvant combined with antigen) have now been tested in clinical trials and have been shown to be generally safe and well tolerated as well as immunogenic, generating both antibody (Ab) and T-cell responses. The mechanisms by which ISCOMATRIX adjuvant facilitates its immune effects are the scope of significant study and indicate that ISCOMATRIX adjuvant (i) rapidly traffics antigen into the cytosol of multiple dendritic cell subsets, (ii) induces the induction of an array of cytokines and chemokines and (iii) links the innate and adaptive immune responses in vivo in a Toll-like-receptor-independent but MyD88-dependent manner. These data highlight the clinical utility of ISCOMATRIX adjuvant in the development of prophylactic and therapeutic vaccines for infectious disease.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholesterol/administration & dosage , Communicable Diseases/therapy , Immunotherapy/methods , Phospholipids/administration & dosage , Saponins/administration & dosage , Vaccination/methods , Vaccines/administration & dosage , Vaccines/immunology , Communicable Disease Control , Drug Combinations , Humans
14.
Immunol Cell Biol ; 90(5): 540-52, 2012 May.
Article in English | MEDLINE | ID: mdl-21894173

ABSTRACT

Generating a cytotoxic CD8(+) T-cell response that can eradicate malignant cells is the primary objective of cancer vaccine strategies. In this study we have characterized the innate and adaptive immune response to the ISCOMATRIX adjuvant, and the ability of vaccine antigens formulated with this adjuvant to promote antitumor immunity. ISCOMATRIX adjuvant led to a rapid innate immune cell response at the injection site, followed by the activation of natural killer and dendritic cells (DC) in regional draining lymph nodes. Strikingly, major histocompatibility complex (MHC) class I cross-presentation by CD8α(+) and CD8α(-) DCs was enhanced by up to 100-fold when antigen was formulated with ISCOMATRIX adjuvant. These coordinated features enabled efficient CD8(+) T-cell cross-priming, which exhibited prophylactic and therapeutic tumoricidal activity. The therapeutic efficacy of an ISCOMATRIX vaccine was further improved when co-administered with an anti-CD40 agonist antibody, suggesting that ISCOMATRIX-based vaccines may combine favorably with other immune modifiers in clinical development to treat cancer. Finally, we identified a requirement for the myeloid differentiation primary response gene 88 (MyD88) adapter protein for both innate and adaptive immune responses to ISCOMATRIX vaccines in vivo. Taken together, our findings support the utility of the ISCOMATRIX adjuvant for use in the development of novel vaccines, particularly those requiring strong CD8(+) T-cell immune responses, such as therapeutic cancer vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cholesterol/immunology , Phospholipids/immunology , Saponins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Neoplasm/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/administration & dosage , Cholesterol/administration & dosage , Cross-Priming/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Combinations , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Ovalbumin/immunology , Phospholipids/administration & dosage , Receptor Cross-Talk/drug effects , Saponins/administration & dosage , Signal Transduction/drug effects
15.
J Immunol ; 187(1): 55-63, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21613613

ABSTRACT

Cancer vaccines aim to induce CTL responses against tumors. Challenges for vaccine design are targeting Ag to dendritic cells (DCs) in vivo, facilitating cross-presentation, and conditioning the microenvironment for Th1 type immune responses. In this study, we report that ISCOM vaccines, which consist of ISCOMATRIX adjuvant and protein Ag, meet these challenges. Subcutaneous injection of an ISCOM vaccine in mice led to a substantial influx and activation of innate and adaptive immune effector cells in vaccine site-draining lymph nodes (VDLNs) as well as IFN-γ production by NK and NKT cells. Moreover, an ISCOM vaccine containing the model Ag OVA (OVA/ISCOM vaccine) was efficiently taken up by CD8α(+) DCs in VDLNs and induced their maturation and IL-12 production. Adoptive transfer of transgenic OT-I T cells revealed highly efficient cross-presentation of the OVA/ISCOM vaccine in vivo, whereas cross-presentation of soluble OVA was poor even at a 100-fold higher concentration. Cross-presenting activity was restricted to CD8α(+) DCs in VDLNs, whereas Langerin(+) DCs and CD8α(-) DCs were dispensable. Remarkably, compared with other adjuvant systems, the OVA/ISCOM vaccine induced a high frequency of OVA-specific CTLs capable of tumor cell killing in different tumor models. Thus, ISCOM vaccines combine potent immune activation with Ag delivery to CD8α(+) DCs in vivo for efficient induction of CTL responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cholesterol/administration & dosage , Cross-Priming/immunology , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Phospholipids/administration & dosage , Saponins/administration & dosage , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cells, Cultured , Cholesterol/immunology , Dendritic Cells/metabolism , Drug Combinations , Female , Gene Knock-In Techniques , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Phospholipids/immunology , Quillaja/immunology , Saponins/immunology
16.
Blood ; 114(27): 5522-31, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19837977

ABSTRACT

Host responses controlling blood-stage malaria include both innate and acquired immune effector mechanisms. During Plasmodium chabaudi infection in mice, a population of CD11b(high)Ly6C(+) monocytes are generated in bone marrow, most of which depend on the chemokine receptor CCR2 for migration from bone marrow to the spleen. In the absence of this receptor mice harbor higher parasitemias. Most importantly, splenic CD11b(high)Ly6C(+) cells from P chabaudi-infected wild-type mice significantly reduce acute-stage parasitemia in CCR2(-/-) mice. The CD11b(high)Ly6C(+) cells in this malaria infection display effector functions such as production of inducible nitric oxide synthase and reactive oxygen intermediates, and phagocytose P chabaudi parasites in vitro, and in a proportion of the cells, in vivo in the spleen, suggesting possible mechanisms of parasite killing. In contrast to monocyte-derived dendritic cells, CD11b(high)Ly6C(+) cells isolated from malaria-infected mice express low levels of major histocompatibility complex II and have limited ability to present the P chabaudi antigen, merozoite surface protein-1, to specific T-cell receptor transgenic CD4 T cells and fail to activate these T cells. We propose that these monocytes, which are rapidly produced in the bone marrow as part of the early defense mechanism against invading pathogens, are important for controlling blood-stage malaria parasites.


Subject(s)
Cell Movement/physiology , Monocytes/parasitology , Plasmodium chabaudi/physiology , Spleen/parasitology , Animals , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/parasitology , Antigen-Presenting Cells/pathology , Antigens, Ly/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/pathology , Flow Cytometry , Host-Parasite Interactions , Malaria/blood , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Nitric Oxide Synthase Type II/metabolism , Parasitemia/metabolism , Phagocytosis/physiology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/parasitology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
17.
Mol Biochem Parasitol ; 156(1): 1-11, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17692398

ABSTRACT

The Plasmodium interspersed repeat (pir) genes represent the largest multigene family in Plasmodium genomes, and the only one shared between the human pathogen, P. vivax, the simian malaria species P. knowlesi and the rodent malaria species P.y. yoelii, P. berghei and P.c. chabaudi. PIR have been shown to be expressed on the surface of red blood cells and are thought to play a role in antigenic variation. Here we have used a range of bioinformatic and experimental approaches to investigate the existence of gene subsets within P.y. yoelii pir. We have identified five groups of yir genes which could be further distinguished by chromosomal location and different alternative splicing events. Two of the groups were not highly represented among the transcribed pirs in blood stage parasites. Together these data suggest that different pir genes may be active at different stages of the life cycle of P. yoelii and may have different functions. Analysis of the 5' UTR identified a unique highly conserved yir/bir/cir specific promoter motif, which could serve as a general recognition element for yir transcription. However, its presence in front of all yirs makes it unlikely to play a role in regulating differential expression.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Interspersed Repetitive Sequences/genetics , Multigene Family , Plasmodium yoelii/metabolism , Protozoan Proteins/genetics , Transcription, Genetic , Animals , Base Sequence , Computational Biology , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phylogeny , Plasmodium yoelii/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Protozoan/metabolism
18.
Mol Microbiol ; 58(3): 636-47, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16238615

ABSTRACT

Variant antigens, encoded by multigene families, and expressed at the surface of erythrocytes infected with the human malaria parasite Plasmodium falciparum and the simian parasite Plasmodium knowlesi, are important in evasion of host immunity. The vir multigene family, encoding a very large number of variant antigens, has been identified in the human parasite Plasmodium vivax and homologues (yir) of this family exist in the rodent parasite Plasmodium yoelii. These genes are part of a superfamily (pir) which are found in Plasmodium species infecting rodents, monkeys and humans (P. yoelii, P. berghei, P. chabaudi, P. knowlesi and P. vivax). Here, we show that YIR proteins are expressed on the surface of erythrocytes infected with late-stage asexual parasites, and that host immunity modulates transcription of yir genes. The surface location and expression pattern of YIR is consistent with a role in antigenic variation. This provides a unique opportunity to study the regulation and expression of the pir superfamily, and its role in both protective immunity and antigenic variation, in an easily accessible animal model system.


Subject(s)
Antigenic Variation , Antigens, Protozoan/immunology , Gene Expression Regulation , Immune System/physiology , Malaria/immunology , Plasmodium , Transcription, Genetic , Animals , Antigens, Protozoan/classification , Antigens, Protozoan/genetics , Disease Models, Animal , Erythrocytes/cytology , Erythrocytes/metabolism , Erythrocytes/microbiology , Female , Host-Parasite Interactions , Humans , Malaria/microbiology , Male , Mice , Mice, Inbred BALB C , Multigene Family , Phylogeny , Plasmodium/genetics , Plasmodium/immunology , Polymorphism, Restriction Fragment Length , Protozoan Proteins/classification , Protozoan Proteins/genetics , Protozoan Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...