Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 934638, 2022.
Article in English | MEDLINE | ID: mdl-35837107

ABSTRACT

Background: Glioblastoma (GBM) is the most common primary, malignant brain tumor in adults and has a poor prognosis. The median progression-free survival (mPFS) of newly diagnosed GBM is approximately 6 months. The recurrence rate approaches 100%, and the case-fatality ratio approaches one. Half the patients die within 8 months of recurrence, and 5-year survival is less than 10%. Advances in treatment options are urgently needed. We report on the efficacy and safety of a therapeutic vaccine (SITOIGANAP: Epitopoietic Research Corporation) administered to 21 patients with recurrent GBM (rGBM) under a Right-to-Try/Expanded Access program. SITOIGANAP is composed of both autologous and allogeneic tumor cells and lysates. Methods: Twenty-one patients with rGBM received SITOIGANAP on 28-day cycles in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), cyclophosphamide, bevacizumab, and an anti-programmed cell death protein-1 (anti-PD-1) monoclonal antibody (either nivolumab or pembrolizumab). Results: The mPFS was 9.14 months, and the median overall survival (mOS) was 19.63 months from protocol entry. Currently, 14 patients (67%) are at least 6 months past their first SITOIGANAP cycle; 10 patients (48%) have received at least six cycles and have a mOS of 30.64 months and 1-year survival of 90%. The enrollment and end-of-study CD3+/CD4+ T-lymphocyte counts strongly correlate with OS. Conclusions: The addition of SITOIGANAP/GM-CSF/cyclophosphamide to bevacizumab and an anti-PD-1 monoclonal antibody resulted in a significant survival benefit compared to historic control values in rGBM with minimal toxicity compared to current therapy.

2.
Int J Mol Sci ; 19(9)2018 Aug 27.
Article in English | MEDLINE | ID: mdl-30150597

ABSTRACT

Glioblastoma is the most common form of brain cancer in adults that produces severe damage to the brain leading to a very poor survival prognosis. The standard of care for glioblastoma is usually surgery, as well as radiotherapy followed by systemic temozolomide chemotherapy, resulting in a median survival time of about 12 to 15 months. Despite these therapeutic efforts, the tumor returns in the vast majority of patients. When relapsing, statistics suggest an imminent death dependent on the size of the tumor, the Karnofsky Performance Status, and the tumor localization. Following the standard of care, the administration of Bevacizumab, inhibiting the growth of the tumor vasculature, is an approved medicinal treatment option approved in the United States, but not in the European Union, as well as the recently approved alternating electric fields (AEFs) generator NovoTTF/Optune. However, it is clear that regardless of the current treatment regimens, glioma patients continue to have dismal prognosis and novel treatments are urgently needed. Here, we describe different approaches of recently developed therapeutic glioma brain cancer vaccines, which stimulate the patient's immune system to recognize tumor-associated antigens (TAA) on cancer cells, aiming to instruct the immune system to eventually attack and destroy the brain tumor cells, with minimal bystander damage to normal brain cells. These distinct immunotherapies may target particular glioma TAAs which are molecularly defined, but they may also target broad patient-derived tumor antigen preparations intentionally evoking a very broad polyclonal antitumor immune stimulation.


Subject(s)
Brain Neoplasms/immunology , Cancer Vaccines/immunology , Glioblastoma/immunology , Immunization/methods , Brain/drug effects , Brain/immunology , Brain/pathology , Brain Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Glioblastoma/drug therapy , Humans , Immune System/drug effects , Immune System/immunology , Survival Analysis
3.
Vaccines (Basel) ; 3(1): 148-71, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-26344951

ABSTRACT

The existence of pathogens that escape recognition by specific vaccines, the need to improve existing vaccines and the increased availability of therapeutic (non-infectious disease) vaccines necessitate the rational development of novel vaccine concepts based on the induction of protective cell-mediated immune responses. For naive T-cell activation, several signals resulting from innate and adaptive interactions need to be integrated, and adjuvants may interfere with some or all of these signals. Adjuvants, for example, are used to promote the immunogenicity of antigens in vaccines, by inducing a pro-inflammatory environment that enables the recruitment and promotion of the infiltration of phagocytic cells, particularly antigen-presenting cells (APC), to the injection site. Adjuvants can enhance antigen presentation, induce cytokine expression, activate APC and modulate more downstream adaptive immune reactions (vaccine delivery systems, facilitating immune Signal 1). In addition, adjuvants can act as immunopotentiators (facilitating Signals 2 and 3) exhibiting immune stimulatory effects during antigen presentation by inducing the expression of co-stimulatory molecules on APC. Together, these signals determine the strength of activation of specific T-cells, thereby also influencing the quality of the downstream T helper cytokine profiles and the differentiation of antigen-specific T helper populations (Signal 3). New adjuvants should also target specific (innate) immune cells in order to facilitate proper activation of downstream adaptive immune responses and homing (Signal 4). It is desirable that these adjuvants should be able to exert such responses in the context of mucosal administered vaccines. This review focuses on the understanding of the potential working mechanisms of the most well-known classes of adjuvants to be used effectively in vaccines.

4.
Vaccine ; 33(23): 2690-6, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25865468

ABSTRACT

Glioblastoma multiforme (GBM) patients have a poor prognosis. After tumor recurrence statistics suggest an imminent death within 1-4.5 months. Supportive preclinical data, from a rat model, provided the rational for a prototype clinical vaccine preparation, named Gliovac (or ERC 1671) composed of autologous antigens, derived from the patient's surgically removed tumor tissue, which is administered together with allogeneic antigens from glioma tissue resected from other GBM patients. We now report the first results of the Gliovac treatment for treatment-resistant GBM patients. Nine (9) recurrent GBM patients, after standard of care treatment, including surgery radio- and chemotherapy temozolomide, and for US patients, also bevacizumab (Avastin™), were treated under a compassionate use/hospital exemption protocol. Gliovac was given intradermally, together with human GM-CSF (Leukine(®)), and preceded by a regimen of regulatory T cell-depleting, low-dose cyclophosphamide. Gliovac administration in patients that have failed standard of care therapies showed minimal toxicity and enhanced overall survival (OS). Six-month (26 weeks) survival for the nine Gliovac patients was 100% versus 33% in control group. At week 40, the published overall survival was 10% if recurrent, reoperated patients were not treated. In the Gliovac treated group, the survival at 40 weeks was 77%. Our data suggest that Gliovac has low toxicity and a promising efficacy. A phase II trial has recently been initiated in recurrent, bevacizumab naïve GBM patients (NCT01903330).


Subject(s)
Glioblastoma/therapy , Immunotherapy, Active/methods , Precision Medicine/methods , Vaccination/methods , Adult , Aged , Animals , Female , Humans , Immunotherapy, Active/adverse effects , Male , Mesothelin , Middle Aged , Rats , Recurrence , Treatment Outcome , United States , Vaccination/adverse effects
5.
Curr Protoc Immunol ; 106: 2.18.1-2.18.7, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25081910

ABSTRACT

Vaccine adjuvants are critical components in experimental and licensed vaccines used in human and veterinary medicine. When aiming to evoke an immune response to a purified antigen, the administration of antigen alone is often insufficient, unless the antigen contains microbial structures or has a natural particulate structure. In most cases, the rationale to use an adjuvant is obvious to the experimental immunologist or the professional vaccinologist, who is familiar with the nature of the antigen, and the aim of the vaccine to elicit a specific antibody response and/or a specific type of T cell response. In this unit, we describe protocols to formulate antigens with oil-based emulsions. Such emulsions represent a major prototype adjuvant category that is frequently used in experimental preclinical vaccines, as well as veterinary and human vaccines.


Subject(s)
Adjuvants, Immunologic , Antigens , Oils , Vaccines , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Antigens/chemistry , Antigens/immunology , Antigens/pharmacology , Emulsions , Humans , Oils/chemistry , Oils/pharmacology , Vaccines/chemistry , Vaccines/immunology , Vaccines/pharmacology
6.
Oncoimmunology ; 1(3): 298-305, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22737605

ABSTRACT

The efficacy of immunotherapeutic TLR7/8 activation by resiquimod (R848) was evaluated in vivo, in the CNS-1 rat glioma model syngeneic to Lewis rats. The immune treatment was compared with cytotoxic cyclophosphamide chemotherapy, and as well, was compared with the combination cytotoxic and immunotherapeutic treatments. We found that parenteral treatment with the TLR7/8 agonist, resiquimod, eventually induced complete tumor regression of CNS-1 glioblastoma tumors in Lewis rats. Cyclophosphamide (CY) treatment also resulted in dramatic CNS-1 remission, while the combined treatment showed similar antitumor effects. The resiquimod efficacy appeared not to be associated with direct injury to CNS-1 growth, while CY proved to exert tumoricidal cytotoxicity to the tumor cells. Rats that were cured by treatment with the innate immune response modifier resiquimod proved to be fully immune to secondary CNS-1 tumor rechallenge. They all remained tumor-free and survived. In contrast, rats that controlled CNS-1 tumor growth as a result of CY treatment did not develop immune memory, as demonstrated by their failure to reject a secondary CNS-1 tumor challenge; they showed a concomittant outgrowth of the primary tumor upon secondary tumor exposure. Rechallenge of rats that initially contained tumor growth by combination chemo-immunotherapy also failed to reject secondary tumor challenge, indicating that the cytotoxic effect of the CY likely extended to the endogenous memory immune cells as well as to the tumor. These data demonstrate strong therapeutic antitumor efficacy for the immune response modifier resiquimod leading to immunological memory, and suggest that CY treatment, although effective as chemotherapeutic agent, may be deleterious to maintenance of long-term antitumor immune memory. These data also highlight the importance of the sequence in which a multi-modal therapy is administered.

7.
J Clin Cell Immunol ; Suppl 5: 004, 2012.
Article in English | MEDLINE | ID: mdl-24955288

ABSTRACT

The efficacy of a various immunotherapeutic immunisation strategies for malignant glioma brain cancer was evaluated in the syngeneic CNS-1 Lewis rat glioma model. A prototype glioma cancer vaccine, which was composed of multivalent antigens derived from allogeneic and syngeneic cells and lysates, formed the prototype preparation of antigens. These antigens reflect the autologous antigens derived from the patient's surgically removed tumor tissue, as well as allogeneic antigens form glioma tumor tissue surgically removed from donor patients. This antigen mixture provides a broad spectrum of tumor associated antigens (TAA) and helps to prevent escape of tumor immune surveillance when given as a vaccine. This antigen preparation was administered in a therapeutic setting with distinct single or multiple co-stimulation-favouring immunostimulants and evaluated for inhibition of tumor growth. Our prototype vaccine was able to arrest progression of tumor growth when co-delivered in a specific regimen together with the costimulating multi-TLR agonist, Bacille Calmette Guerin (BCG) and interleukin-2, or with the Toll-Like receptor (TLR) 7/8 activator resiquimod.

8.
Expert Rev Vaccines ; 10(4): 539-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21506650

ABSTRACT

Adjuvants are essential components of most clinically used vaccines. This is because the majority of nonliving vaccines are relatively poor inducers of adaptive immunity unless effective adjuvants are co-administered. Aluminum salts (alum) have been used as adjuvants with great success for almost a century and have been particularly effective at promoting protective humoral immunity. However, alum is not optimally effective for diseases where cell-mediated immunity is required for protection. Furthermore, adjuvants including oil-in-water emulsions have shown improved efficacy for avian influenza protection suggesting that even for diseases where humoral immunity can confer protection, there is scope for developing improved adjuvants. There have been major developments in antigen discovery over the past decade, which has accelerated the vaccine development process for new indications and this demands a new generation of adjuvants that can drive and specifically direct the desired immune responses. A number of systems are under investigation that combine different types of adjuvants into specific formulations with greater activity. Additionally, targeting of vaccines to specific immune cells shows great promise. In the case of cancer and chronic infectious diseases, it may be difficult to develop effective vaccines without blocking immune regulatory pathways, which impede cell-mediated responses. However, increased understanding of immunology and particularly the innate immune system is informing vaccine adjuvant research and consequently driving the development of novel and specifically directed vaccine adjuvant strategies. In this article we address the importance of adjuvants in vaccine development, the known mode of action of specific adjuvants and recent developments in this important field.


Subject(s)
Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Biomedical Research/trends , Communicable Disease Control , Humans , Neoplasms/therapy , Vaccines/immunology
9.
PLoS One ; 6(1): e15986, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21305002

ABSTRACT

BACKGROUND: The innate immune system relies upon a wide range of germ-line encoded receptors including a large number of immunoglobulin superfamily (IgSF) receptors. Different Ig-like immune receptor families have been reported in mammals, birds, amphibians and fish. Most innate immune receptors of the IgSF are type I transmembrane proteins containing one or more extracellular Ig-like domains and their regulation of effector functions is mediated intracellularly by distinct stimulatory or inhibitory pathways. METHODOLOGY/PRINCIPAL FINDINGS: Carp SITR was found in a substracted cDNA repertoire from carp macrophages, enriched for genes up-regulated in response to the protozoan parasite Trypanoplasma borreli. Carp SITR is a type I protein with two extracellular Ig domains in a unique organisation of a N-proximal V/C2 (or I-) type and a C-proximal V-type Ig domain, devoid of a transmembrane domain or any intracytoplasmic signalling motif. The carp SITR C-proximal V-type Ig domain, in particular, has a close sequence similarity and conserved structural characteristics to the mammalian CD300 molecules. By generating an anti-SITR antibody we could show that SITR protein expression was restricted to cells of the myeloid lineage. Carp SITR is abundantly expressed in macrophages and is secreted upon in vitro stimulation with the protozoan parasite T. borreli. Secretion of SITR protein during in vivo T. borreli infection suggests a role for this IgSF receptor in the host response to this protozoan parasite. Overexpression of carp SITR in mouse macrophages and knock-down of SITR protein expression in carp macrophages, using morpholino antisense technology, provided evidence for the involvement of carp SITR in the parasite-induced NO production. CONCLUSION/SIGNIFICANCE: We report the structural and functional characterization of a novel soluble immune-type receptor (SITR) in a teleost fish and propose a role for carp SITR in the NO-mediated response to a protozoan parasite.


Subject(s)
Carps/parasitology , Nitric Oxide/immunology , Receptors, Immunologic/immunology , Animals , Carps/immunology , Fishes , Immunity, Innate , Macrophages/parasitology , Mice , Receptors, Immunologic/chemistry , Solubility , Trypanosoma/immunology
10.
Methods Mol Biol ; 626: 1-14, 2010.
Article in English | MEDLINE | ID: mdl-20099117

ABSTRACT

In recent times vaccine adjuvants, or immunopotentiators, received abundant attention in the media as critical ingredients of current and future vaccines. Indeed, vaccine adjuvants are recognized to make the difference between competing vaccines based on identical antigens. Moreover, it is recognized that vaccines designed for certain indications require a matching combination of selected antigen(s) together with a critical immunopotentiator that selectively drives the required immune pathway with minimal adverse reactions. Recently, the mechanistic actions of some immunopotentiators have become clearer as a result of research focused on innate immunity receptors. These insights enable more rational adjuvant and vaccine design, which, ideally, is based on predictable immunophenotypes following vaccination.This chapter addresses immunopotentiators, classed according to their (presumed) mechanisms of action. They are categorized functionally in two major groups as facilitators of signal 1 and/or signal 2. The mode(s) of action of some well-known adjuvant prototypes is discussed in the context of this classification.


Subject(s)
Adjuvants, Immunologic/pharmacology , Drug Design , Vaccines/immunology , Animals , Antigens/immunology , Humans
11.
Vaccine ; 26(14): 1764-72, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18336964

ABSTRACT

In the present study we investigated whether allogeneic glioma cells can be utilized to evoke prophylactic or therapeutic immune-mediated elimination of syngeneic glioma in two rat strains. Fisher 344 and Sprague-Dawley (SD) rats were injected with two syngeneic glioma cell lines, 9L and C6, respectively, resulting in progressive tumor growth. 9L is syngeneic to the Fisher 344 and allogeneic to the SD rats, while C6 cells are syngeneic to SD rats and allogeneic to Fisher 344 rats. Both rat strains were subcutaneously injected with their respective allogeneic tumor cells, which proved unable to grow progressively. The allogeneic cells were either rejected immediately in SD rats or within 25 days in Fisher rats, after limited tumor outgrowth. Both rat strains were subsequently challenged with their respective syngeneic glioma tumor cells and once more 10 days later with a fivefold higher dose. SD rats, even after reinjection with five times the original dosage of C6 cells, remained tumor free for at least 360 days. Similarly, Fisher rats, after initially rejecting allogeneic tumors, failed to develop syngeneic tumors. To determine anti-tumor immunity against established glioma tumors under more demanding therapeutic conditions, rats were first injected subcutaneously with their respective syngeneic tumor and vaccinated once or repeatedly (at 5-day intervals) with a mixture of the allogeneic or xenogeneic cells, with or without a lysate from the same syngeneic tumor, which served as a therapeutic vaccine preparations. The control group received either no treatment or syngeneic instead of allogeneic cells. In both strains of rats, we demonstrated that the therapeutically vaccinated groups were able to significantly reduce tumor growth, while complete rejection of tumors was noted in the SD rats. Immunization with syngeneic tumor cells alone failed to evoke anti-tumor immunity. We conclude that therapeutic immunization with a combination of allogeneic cells and syngeneic lysates induces rejection of malignant gliomas and offers a protective effect against challenge with syngeneic tumor cells. This immunization approach may prove useful as a post-surgery adjuvant therapy in future cancer treatment protocols, or even as a stand-alone therapeutic tumor vaccination.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/prevention & control , Cancer Vaccines/therapeutic use , Glioma/immunology , Glioma/prevention & control , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/pathology , Immunohistochemistry , Neoplasm Recurrence, Local/prevention & control , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Species Specificity , Transplantation, Isogeneic
12.
Avian Pathol ; 36(5): 361-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17899459

ABSTRACT

The release requirements for vaccine antigens delivered by adjuvants with presumed depot function are poorly understood. Water-in-oil (W/O) emulsions are routinely used in many poultry vaccines. They strongly activate antibody production, and are regarded as a depot from which antigens are slowly released, resulting in prolonged antigen residence. However, from earlier studies we concluded that W/O adjuvant activity is partly based on the immunostimulatory activity of the oil phase. Here we assess the dose and regimen requirements for viral antigen in immunization experiments in chickens. Three-week-old to 4-week-old White Leghorn chickens were repeatedly injected with inactivated infectious bursal disease virus antigen over 48 days. Our aim was to compare the antibody responses in repeatedly injected animals, receiving fractioned doses of antigen, with the responses in animals receiving only one injection of the full dose of antigen formulated in either a W/O emulsion or in saline. We observed that repeated administration of small amounts of antigen results in a gradual increase of specific humoral immune responses during the immunization regimen. Immunization with a higher first dose evoked an early higher antibody response, which, however, reached a similar plateau level at the end of the regimen. When compared with lower first-dose regimens, a slow decline of serum antibody titre 2 weeks after the end of antigen injections indicated that repeated injection of small doses of antigen indeed mimics the efficacy of depot-forming adjuvants. All regimens of fractioned antigen in saline, however, proved less effective, when compared with a single-dose vaccination of the cumulative amount of antigen formulated in a W/O emulsion. From our data we confirm that W/O emulsions are very effective vaccine vehicles for improving antigen-specific humoral responses in chickens, owing to a combination of antigen residence-prolonging activity and direct immune stimulation.


Subject(s)
Chickens/immunology , Infectious bursal disease virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Antibody Formation , Antigens, Viral/immunology , Birnaviridae Infections/prevention & control , Birnaviridae Infections/veterinary , Delayed-Action Preparations , Dose-Response Relationship, Immunologic , Drug Administration Schedule , Emulsions , Female , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Specific Pathogen-Free Organisms
13.
Vaccine ; 24(35-36): 6096-109, 2006 Aug 28.
Article in English | MEDLINE | ID: mdl-16781024

ABSTRACT

In a respiratory-infection-model with the avian influenza A H9N2 virus we studied lung and splenic immune reactions in chickens using a recently developed 5K chicken immuno-microarray. Groups of chickens were either mock-immunized (referred to as non-immune), vaccinated with inactivated viral antigen only (immune) or with viral antigen in a water-in-oil (W/O) immunopotentiator (immune potentiated). Three weeks after vaccination all animals were given a respiratory infection. Immune potentiated birds developed inhibitory antiviral antibodies, showed minimal lung histopathology and no detectable viral sequences, while non-immune animals showed microscopic immunopathology and detectable virus. Immune birds, receiving antigen in saline only, showed minimal microscopic histopathology, and intermediate levels of virus detection. These classical features in the different groups were mirrored by overlapping or specific mRNA gene expression profiles in lungs and spleen using microarray analysis. To our knowledge this is the first study demonstrating pneumonia-associated lung pathology of the low pathogenic avian influenza H9N2 virus. Our data provide insights into the molecular interaction of this virus with its natural host when naive or primed by vaccination.


Subject(s)
Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/therapeutic use , Influenza in Birds/immunology , Lung/immunology , Spleen/immunology , Animals , Antigen Presentation/physiology , Antigens, CD/metabolism , Apoptosis/physiology , B-Lymphocytes/metabolism , Chickens , Complement System Proteins/metabolism , Cytokines/metabolism , Immunity, Innate , Influenza in Birds/pathology , Influenza in Birds/prevention & control , Interferon Type I/metabolism , Lung/pathology , Myeloid Cells/metabolism , Spleen/pathology , Toll-Like Receptors/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Up-Regulation
14.
Vaccine ; 24(26): 5400-5, 2006 Jun 29.
Article in English | MEDLINE | ID: mdl-16675072

ABSTRACT

Oil-based emulsions are well-known immunopotentiators for inactivated, "killed" vaccines. We addressed the relationship between emulsion structure and levels of in vivo antibody formation to inactivated New Castle Disease virus (NDV) and Infectious Bronchitis virus (IBV) as antigens in 3-week-old chickens. The use of a polymeric emulsifier allowed for direct comparison of three types of emulsions, water-in-oil (W/O), oil-in-water (O/W) and W/O-in-water (W/O/W), while maintaining an identical content of components for each vehicle. They were prepared with either non-metabolizable, mineral oil or metabolizable, Miglyol 840. In addition, we assessed the inherent release capacity of each emulsion variant in vitro. Remarkably, we noted that W/O-type emulsions induced the best immune responses, while they released no antigen during 3 weeks. In general, mineral oil vaccines showed superior efficacy compared to Miglyol 840-based vaccines.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/immunology , Newcastle Disease/prevention & control , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral , Chemistry, Pharmaceutical , Chickens , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Diglycerides/immunology , Emulsions , Hexoses , Mineral Oil , Polysorbates , Time Factors , Viral Vaccines/adverse effects
15.
BMC Genomics ; 7: 49, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16533398

ABSTRACT

BACKGROUND: The development of microarray resources for the chicken is an important step in being able to profile gene expression changes occurring in birds in response to different challenges and stimuli. The creation of an immune-related array is highly valuable in determining the host immune response in relation to infection with a wide variety of bacterial and viral diseases. RESULTS: Here we report the development of chicken immune-related cDNA libraries and the subsequent construction of a microarray containing 5190 elements (in duplicate). Clones on the array originate from tissues known to contain high levels of cells related to the immune system, namely Bursa, Peyers patch, thymus and spleen. Represented on the array are genes that are known to cluster with existing chicken ESTs as well as genes that are unique to our libraries. Some of these genes have no known homologies and represent novel genes in the chicken collection. A series of reference genes (ie. genes of known immune function) are also present on the array. Functional annotation data is also provided for as many of the genes on the array as is possible. CONCLUSION: Six new chicken immune cDNA libraries have been created and nearly 10,000 sequences submitted to GenBank [GenBank: AM063043-AM071350; AM071520-AM072286; AM075249-AM075607]. A 5 K immune-related array has been developed from these libraries. Individual clones and arrays are available from the ARK-Genomics resource centre.


Subject(s)
Chickens/genetics , Chickens/immunology , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Animals , B-Lymphocytes/immunology , Base Sequence , Chickens/metabolism , Female , Gene Expression Profiling/standards , Gene Library , Genomics , Immune System/immunology , Male , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/standards , T-Lymphocytes/immunology , Vocabulary, Controlled
16.
Vaccine ; 23(33): 4212-8, 2005 Jul 21.
Article in English | MEDLINE | ID: mdl-15919141

ABSTRACT

Mammalian interleukin-18 (IL-18) is one of the pro-inflammatory cytokines involved in innate immune responses to microbial infection preceding the induction of both cellular and humoral immune responses. We assessed the potential of Escherichia coli-expressed His-tag purified recombinant chicken IL-18 (rHis-ChIL-18) as a potentiator of vaccine-induced immune responses in 3 week-old SPF chickens and compared it with several commonly used traditional immunostimulating adjuvants. We found that rHis-ChIL-18 significantly enhanced antibody responses to Clostridium perfringens alpha-toxoid and Newcastle disease virus (NDV) antigens, comparable to the Al(OH)3-gel, Miglyol and chitosan adjuvant. However, antibody responses were lower than water-in-oil (w/o) emulsions and combinations of rHis-ChIL-18 with Al(OH)3-gel, Miglyol or chitosan. Improved HI-titers for infectious bronchitis virus (IBV) were not noted. We conclude that rHis-ChIL-18, when delivered in the proper context, is a novel and safe immunostimulator in chickens.


Subject(s)
Adjuvants, Immunologic , Chickens/immunology , Immunity/drug effects , Interleukin-18/administration & dosage , Animals , Antibodies, Bacterial/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Interleukin-18/immunology , Recombinant Proteins/immunology , Vaccination
17.
Vet Microbiol ; 105(3-4): 163-7, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15708812

ABSTRACT

Mammals developed an immune system able to functionally polarize into so-called type 1 or type 2 immune pathways, to resolve infections with intracellular and extracellular pathogens, respectively. In the well-studied avian immune system of the chicken, however, no evidence for polarized immunity could be found, as yet. To investigate whether these two major arms of mammalian immunity, regulated by a T helper (Th)1/Th2 cytokine balance, evolved similarly in birds, chickens were exposed to a prevalent intracellular (viral) or extracellular (helminth) infection. By using semi-quantitative RT-PCR analysis we provide evidence that polarization of Th1/Th2 type immunity extends beyond mammalian species, and, therefore, has been evolutionary conserved for more than 300 million years, when the lineages of mammalian and avian vertebrates are assumed to have segregated.


Subject(s)
Ascaridiasis/veterinary , Chickens/immunology , Newcastle Disease/immunology , Poultry Diseases/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Ascaridia/immunology , Ascaridiasis/immunology , Base Sequence , Biological Evolution , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Ileum/immunology , Immunity, Cellular , Newcastle disease virus/immunology , Poultry Diseases/parasitology , Poultry Diseases/virology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Specific Pathogen-Free Organisms , Spleen/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism
18.
Vaccine ; 23(11): 1343-50, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15661382

ABSTRACT

Dendritic cells (DCs) induce different types of immune responses depending on their lineage and activation signals. When exposed to inactivated pseudorabiesvirus (iPRV), plasmacytoid but not myeloid DCs released IFN-alpha and IL-12. Remarkably, both iPRV-pulsed DC types were able to induce primary IFN-gamma producing T cells and IgG isotype switching in vivo. In contrast, tetanus toxoid pulsed DCs did not induce detectable primary immune responses. The efficacy of antiviral T and B cell priming proved dependent on the recipient's genotype. We conclude that either plasmacytoid or myeloid DCs pulsed with inactivated virus suffice to induce primary Th1-polarised immune responses.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Herpesvirus 1, Suid/immunology , Th1 Cells/immunology , Viral Vaccines/immunology , Animals , B-Lymphocytes/immunology , Dendritic Cells/cytology , Immunoglobulin G/analysis , Interferon-alpha/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-5 , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Plasma Cells/immunology , Tetanus Toxoid/immunology , Virion/immunology
19.
Vaccine ; 23(8): 1053-60, 2005 Jan 11.
Article in English | MEDLINE | ID: mdl-15620479

ABSTRACT

Water-in-oil (W/O) emulsions are known as most effective adjuvants to generate high and durable antibody responses to vaccine antigens following a single immunization. However, their structural requirements remain poorly understood. Here we addressed the significance of certain pharmaceutical characteristics including water/oil ratios--ranging from 60/40 to 30/70 (w/w(%))--droplet size and type of oil, i.e. non-metabolizable (mineral oil) versus metabolizable (Miglyol 840). Stability of emulsions was accomplished by the use of a polymeric emulsifier. Distinct W/O emulsions were formulated with inactivated (i) infectious bronchitis virus (iIBV) and Newcastle disease virus (iNDV), and evaluated in immunized chickens for magnitude and duration of in vivo antiviral antibody formation and local reactions. A high mineral oil content proved most effective for antibody response formation. In general, a larger droplet size evoked higher antibody responses for both oil types. Inoculum residues proved lower using biodegradable Miglyol, when compared to mineral oil, for all emulsion variants. Especially water-to-oil ratio and droplet size may provide useful parameters for improving (antiviral) antibody production by W/O emulsions.


Subject(s)
Antibody Formation , Antigen-Antibody Reactions/drug effects , Epitopes/immunology , Oils/chemistry , Vaccines/chemistry , Vaccines/immunology , Water/chemistry , Animals , Antigen-Antibody Reactions/immunology , Chemistry, Pharmaceutical , Chickens , Epitopes/chemistry , Female , Oils/pharmacology , Structure-Activity Relationship , Vaccines/pharmacology , Water/pharmacology
20.
J Gen Virol ; 85(Pt 4): 795-803, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039522

ABSTRACT

Replicating viruses generally induce type 1 immune responses, with high interferon (IFN)-gamma levels and antibodies of the IgG2a isotype. In the present study we demonstrate the intrinsic ability of non-replicating virions to induce comparable immune responses in the notable absence of any adjuvant. Injection of inactivated pseudorabies virus, an alphaherpesvirus, by various routes into mice resulted in the generation of T helper (Th) 1 type immune response. Co-delivery of inactivated pseudorabies herpesvirus (iPRV) with protein redirected IgG1-dominated tetanus toxoid-specific responses towards an IgG1/IgG2a balanced response. Also inactivated preparations of viruses from the paramyxo- (Newcastle disease virus), rhabdo- (rabies virus), corona- (infectious bronchitis virus) and reovirus (avian reovirus) families led to IgG2a antibody responses; however, the genetic background of the host did result in considerable variation. Because disrupted virions also induced type 1 immune responses, we conclude that structural elements of virions inherently contribute to IFN-gamma-dependent isotype switching by inactivated viruses. Strikingly, immunizations in gene-disrupted mice showed that a functional IFN-alpha/beta, IFN-gamma or interleukin (IL)-12 pathway was not required for the generation of a polarized Th1 type immune response initiated by inactivated virus particles. These findings have a bearing on the understanding of immune responsiveness to virus structures and the design of vaccines containing virus components.


Subject(s)
Cytokines/biosynthesis , Th1 Cells/immunology , Viruses/immunology , Animals , Antibodies, Viral/biosynthesis , Herpesvirus 1, Suid/immunology , Immunoglobulin Class Switching , Immunoglobulin G/biosynthesis , Infectious bronchitis virus/immunology , Interferon Type I/deficiency , Interferon Type I/genetics , Interferon-gamma/metabolism , Interleukin-12/deficiency , Interleukin-12/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Newcastle disease virus/immunology , Orthoreovirus, Avian/immunology , Rabies virus/immunology , Vaccines, Inactivated/immunology , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...