Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Brain Behav Immun ; 112: 163-172, 2023 08.
Article in English | MEDLINE | ID: mdl-37315700

ABSTRACT

One of the chief advantages of using highly standardised biological models including model organisms is that multiple variables can be precisely controlled so that the variable of interest is more easily studied. However, such an approach often obscures effects in sub-populations resulting from natural population heterogeneity. Efforts to expand our fundamental understanding of multiple sub-populations are in progress. However, such stratified or personalised approaches require fundamental modifications of our usual study designs that should be implemented in Brain, Behavior and Immunity (BBI) research going forward. Here we explore the statistical feasibility of asking multiple questions (including incorporating sex) within the same experimental cohort using statistical simulations of real data. We illustrate and discuss the large explosion in sample numbers necessary to detect effects with appropriate power for every additional question posed using the same data set. This exploration highlights the strong likelihood of type II errors (false negatives) for standard data and type I errors when dealing with complex genomic data, where studies are too under-powered to appropriately test these interactions. We show this power may differ for males and females in high throughput data sets such as RNA sequencing. We offer a rationale for the use of alternative experimental and statistical strategies based on interdisciplinary insights and discuss the real-world implications of increasing the complexities of our experimental designs, and the implications of not attempting to alter our experimental designs going forward.


Subject(s)
Animal Experimentation , Research Design , Male , Animals , Causality
2.
Trials ; 24(1): 202, 2023 Mar 18.
Article in English | MEDLINE | ID: mdl-36934272

ABSTRACT

BACKGROUND: The need for coronavirus 2019 (COVID-19) vaccination in different age groups and populations is a subject of great uncertainty and an ongoing global debate. Critical knowledge gaps regarding COVID-19 vaccination include the duration of protection offered by different priming and booster vaccination regimens in different populations, including homologous or heterologous schedules; how vaccination impacts key elements of the immune system; how this is modified by prior or subsequent exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and future variants; and how immune responses correlate with protection against infection and disease, including antibodies and effector and T cell central memory. METHODS: The Platform Trial In COVID-19 priming and BOOsting (PICOBOO) is a multi-site, multi-arm, Bayesian, adaptive, randomised controlled platform trial. PICOBOO will expeditiously generate and translate high-quality evidence of the immunogenicity, reactogenicity and cross-protection of different COVID-19 priming and booster vaccination strategies against SARS-CoV-2 and its variants/subvariants, specific to the Australian context. While the platform is designed to be vaccine agnostic, participants will be randomised to one of three vaccines at trial commencement, including Pfizer's Comirnaty, Moderna's Spikevax or Novavax's Nuvaxovid COVID-19 vaccine. The protocol structure specifying PICOBOO is modular and hierarchical. Here, we describe the Core Protocol, which outlines the trial processes applicable to all study participants included in the platform trial. DISCUSSION: PICOBOO is the first adaptive platform trial evaluating different COVID-19 priming and booster vaccination strategies in Australia, and one of the few established internationally, that is designed to generate high-quality evidence to inform immunisation practice and policy. The modular, hierarchical protocol structure is intended to standardise outcomes, endpoints, data collection and other study processes for nested substudies included in the trial platform and to minimise duplication. It is anticipated that this flexible trial structure will enable investigators to respond with agility to new research questions as they arise, such as the utility of new vaccines (such as bivalent, or SARS-CoV-2 variant-specific vaccines) as they become available for use. TRIAL REGISTRATION: Australian and New Zealand Clinical Trials Registry ACTRN12622000238774. Registered on 10 February 2022.


Subject(s)
COVID-19 , Vaccines , Humans , SARS-CoV-2 , COVID-19/prevention & control , COVID-19 Vaccines , Bayes Theorem , Australia , Vaccination , Randomized Controlled Trials as Topic
3.
Expert Rev Vaccines ; 18(2): 133-151, 2019 02.
Article in English | MEDLINE | ID: mdl-30601095

ABSTRACT

INTRODUCTION: The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED: This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY: Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.


Subject(s)
Immunization/methods , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Adjuvants, Immunologic/administration & dosage , Global Health , Humans , Immunity, Cellular/immunology , Malaria/epidemiology , Malaria/immunology , Seasons , Time Factors
4.
Cytometry A ; 91(4): 336-350, 2017 04.
Article in English | MEDLINE | ID: mdl-28264143

ABSTRACT

The balance of inflammation and immunosuppression driven by changed ratios in diverse myeloid and T cell subsets, as well as their state of activation and ability to migrate to lymphoid compartments or inflammatory sites, has emerged as a highly active area of study across clinical trials of vaccines and therapies against cancer, trauma, as well as autoimmune and infectious diseases. There is a need for effective protocols which maximally use the possibilities offered by modern flow cytometers to characterize such immune cell changes in peripheral blood using small volumes of human blood. Additionally, longitudinal clinical studies often use cryopreserved samples, which can impact flow cytometric results. To efficiently gauge both the innate and the adaptive immune response, two novel 15-color antibody panels to identify key myeloid and T cell subsets and their functional potential were established. This approach was used to compare cellular immune profiles in fresh whole blood and in matched cryopreserved peripheral blood mononuclear cells (PBMCs). Cocktail I was designed to identify and characterize myeloid cell populations including dendritic cells (DCs), monocytic monocyte-derived suppressor cells (MO-MDSC), and monocytes, determining further core aspects of their state of maturity, T cell stimulatory (or inhibitory) potential, and migration capability. Cocktail II was used for phenotyping diverse T cells subsets, and their key migration and functional regulatory capabilities. The two 15-color antibody panels for the evaluation of both immune-stimulating and immunosuppressive processes presented herein allowed for efficient evaluation of the balance of immune activation versus immunosuppression across key blood cells, with good resolution for all 15 markers stained for in each panel. Gating strategies for the myeloid and T cells are presented to further support specific subset identification. This protocol was shown to be reproducible across donors and useful to study both RBC-lysed whole blood and cryopreserved PBMCs. © 2017 International Society for Advancement of Cytometry.


Subject(s)
Flow Cytometry/methods , Immunophenotyping/methods , Myeloid Cells/cytology , T-Lymphocyte Subsets/cytology , Cryopreservation , Humans , Immunity, Innate , Leukocytes, Mononuclear/cytology
5.
Gene Ther ; 21(2): 212-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24352195

ABSTRACT

Dendritic cells (DC) targeting vaccines require high efficiency for uptake, followed by DC activation and maturation. We used magnetic vectors comprising polyethylenimine (PEI)-coated superparamagnetic iron oxide nanoparticles, with hyaluronic acid (HA) of different molecular weights (<10 and 900 kDa) to reduce cytotoxicity and to facilitate endocytosis of particles into DCs via specific surface receptors. DNA encoding Plasmodium yoelii merozoite surface protein 1-19 and a plasmid encoding yellow fluorescent gene were added to the magnetic complexes with various % charge ratios of HA: PEI. The presence of magnetic fields significantly enhanced DC transfection and maturation. Vectors containing a high-molecular-weight HA with 100% charge ratio of HA: PEI yielded a better transfection efficiency than others. This phenomenon was attributed to their longer molecular chains and higher mucoadhesive properties aiding DNA condensation and stability. Insights gained should improve the design of more effective DNA vaccine delivery systems.


Subject(s)
Ferric Compounds/metabolism , Hyaluronic Acid/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/metabolism , Vaccines, DNA/administration & dosage , Vaccines, DNA/chemistry , Animals , Antigens, Protozoan/genetics , Dendritic Cells , Ferric Compounds/chemistry , Genetic Vectors , Magnetic Phenomena , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/adverse effects , Nanoparticles/chemistry , Plasmodium yoelii/genetics , Transfection
6.
Curr Med Chem ; 20(11): 1437-44, 2013.
Article in English | MEDLINE | ID: mdl-23409714

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of myeloid progenitors that can play a major role in tumour development and chronic inflammation. The importance of the suppressive function ofMDSCs was first suggested by studies involving cancer patients and cancer-bearing mice. In addition, recent studies have demonstrated that MDSCs can also be involved in many other pathological conditions. MDSCs have unique ways of abrogatingan immune response in addition to those utilised by other immune-suppressive cell types, for example via the induction of arginase-1 and consequent upregulation in reactive oxygen species (ROS) production. Due to their heterogeneity,they further can express a variety of lineage markers, which overlap with other myeloid cell types such as Gr1,CD11b, MHCIIlo, Ly6C and Ly6G, making it difficult to identify them by surface phenotype alone. The disparity between mouse and human MDSCs further complicates the identification of these elusive cell populations. In this review, we will summarise the recent updates on the methods for eliciting and studying different MDSC subsets, including newly proposed surface phenotypes, as well as insights into how their function is being characterised in both mice and humans. In addition, exciting new discoveries suggesting their involvement across a number of different pathological settings, such as sepsis, autoimmunity and Leishmaniasis, will be discussed.


Subject(s)
Immune Tolerance , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Neoplasms/immunology , Animals , Humans , Inflammation/immunology , Inflammation/pathology , Myeloid Progenitor Cells/cytology , Neoplasms/pathology
7.
Clin Exp Immunol ; 143(2): 363-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16412062

ABSTRACT

The alveolar macrophage (AM) is believed to be of central importance in the immune response against infection and tumour. We examined patients with lung cancer in order to evaluate the immuno-stimulatory potential of AM in lung cancer. Bronchoalveolar lavage fluid samples were obtained from patients with adenocarcinoma, squamous cell carcinoma, large cell undifferentiated lung carcinoma, small cell carcinoma and control subjects. AM were isolated and phagocytic function, flow cytometry and cytokine analysis were assessed. AM from patients with small and squamous cell carcinoma had impaired uptake in vitro of 40 nm fluorescent polystyrene beads. AM from patients with small, squamous and large cell undifferentiated carcinoma showed impaired uptake of 1000 nm fluorescent polystyrene beads. Secreted levels of TNF-alpha and IL-1 from AM of patients with small, squamous, and large cell undifferentiated carcinoma were decreased compared to controls. Secreted AM IL-6 levels were decreased in small and large cell undifferentiated carcinoma. AM from adenocarcinoma patients showed similar levels of IL-10, IL-6, IL-1 and TNF-alpha compared to controls. Phenotypic analysis demonstrated that patients with small cell carcinoma were the only group that showed a decrease in MHC class II surface expression. Surface expression of ICAM-1 and CD83 was decreased on AM from patients with large, squamous and small cell carcinoma compared to controls but not adenocarcinoma. Mannose receptor levels were only decreased on AM from patients with squamous and small cell carcinoma but not adenocarcinoma and large cell undifferentiated carcinoma. We conclude that there are type-specific alterations in uptake ability, cytokine secretion and phenotype of AM from lung cancer patients, which may result in an inability to stimulate anti-tumour immunity. The observed differences between lung cancer subgroups may explain previously reported inconsistencies in descriptions of AM characteristics in lung cancer.


Subject(s)
Lung Neoplasms/immunology , Macrophages/immunology , Pulmonary Alveoli/immunology , Adenocarcinoma/immunology , Aged , Antigens, CD/analysis , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/immunology , Carcinoma, Large Cell/immunology , Carcinoma, Small Cell/immunology , Carcinoma, Squamous Cell/immunology , Cytokines/immunology , HLA-DR Antigens/analysis , Humans , Immunoglobulins/analysis , Intercellular Adhesion Molecule-1/analysis , Lectins, C-Type/analysis , Mannose Receptor , Mannose-Binding Lectins/analysis , Membrane Glycoproteins/analysis , Microspheres , Middle Aged , Phenotype , Polystyrenes/pharmacokinetics , Receptors, Cell Surface/analysis , CD83 Antigen
8.
Article in English | MEDLINE | ID: mdl-16375694

ABSTRACT

The immune system is a tightly regulated network that is able to maintain a balance of immune homeostasis under normal physiological conditions. Normally, when challenged with foreign antigen, specific appropriate responses are initiated that are aimed at restoring homeostasis. However under particular circumstances, this balance is not maintained and immune responses either under or over react. Cancer is an example of a situation where the immune response can be inefficient or unresponsive, resulting in uncontrolled growth of the cancer cells. Conversely, when the immune response over-reacts, this can result in conditions such as autoimmunity or pathology following infection. Many drug therapies have been developed that aim to alleviate or prevent such immune disorders and restore immune homeostasis. This review highlights recent advances in immunotherapies, with an emphasis on specific examples in the treatment of cancer, autoimmune disease (multiple sclerosis) and viral infection (respiratory syncytial virus).


Subject(s)
Autoimmune Diseases/immunology , Homeostasis/immunology , Immunotherapy , Neoplasms/immunology , Virus Diseases/immunology , Animals , Autoimmune Diseases/therapy , Humans , Immunotherapy/methods , Neoplasms/therapy , Virus Diseases/therapy
9.
J Postgrad Med ; 50(1): 30-4, 2004.
Article in English | MEDLINE | ID: mdl-15047996

ABSTRACT

The interaction between the malaria parasite and the human host involves a number of interactions that result in the parasite evading the human immune system. Since the stages of the malaria lifecycle are complex, this allows the use of various immune evasion strategies by the malaria parasite and has major implications in the development of a vaccine for malaria endemic areas. The present review highlights key host:parasite interactions. Plasmodia puts selection pressure on human gene frequencies, and studies into host genetic factors such as the Duffy blood group and sickle cell anaemia offer insight into the host- parasite relationship. In addition, parasite interactions with the different effector arms of the immune system can result in altered peptide ligand (APL) antagonism which alters the immune response from a pro- to an anti-inflammatory T cell response. Recent insights into the interaction between professional antigen presenting cells, dendritic cells (DCs), and malaria parasites is discussed in detail.


Subject(s)
Host-Parasite Interactions/physiology , Plasmodium/physiology , Protozoan Proteins/immunology , Animals , Antigens, CD , Dendritic Cells/immunology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Malaria Vaccines , Peptide Fragments , Plasmodium/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/parasitology , T-Lymphocytes/immunology
10.
Clin Exp Immunol ; 135(2): 286-93, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14738458

ABSTRACT

Vaccination of malaria-naive humans with recombinant RTS,S/AS02, which includes the C-terminus of the circumsporozoite protein (CS), has been shown to induce strong T cell responses to both the whole protein antigen and to peptides from CS. Here we show that strong T cell responses were also observed in a semi-immune population in The Gambia, West Africa. In a Phase I study, 20 adult male volunteers, lifelong residents in a malaria-endemic region, were given three doses of RTS,S/AS02 at 0, 1 and 6 months. Responses to RTS,S, hepatitis B surface antigen and peptides from CS were tested using lymphocyte proliferation, interferon (IFN)-gamma production in microcultures, and IFN-gamma ex vivo and cultured ELISPOT, before and after vaccination. Cytotoxic responses were tested only after vaccination and none were detected. Before vaccination, the majority of the volunteers (15/20) had detectable responses in at least one of the tests. After vaccination, responses increased in all assays except cytotoxicity. The increase was most marked for proliferation; all donors responded to RTS,S after the third dose and all except one donor responded to at least one peptide after the second or third dose. There was a lack of close association of peptide responses detected by the different assays, although in microcultures IFN-gamma responses were found only when proliferative responses were high, and responses by cultured ELISPOT and proliferation were found together more frequently after vaccination. We have therefore identified several peptide-specific T cell responses induced by RTS,S/AS02 which provides a mechanism to investigate potentially protective immune responses in the field.


Subject(s)
Immunity, Cellular/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Adolescent , Adult , Antigens, Protozoan/immunology , Cells, Cultured , Cohort Studies , Cytotoxicity Tests, Immunologic/methods , Enzyme-Linked Immunosorbent Assay/methods , Gambia , Hepatitis B Surface Antigens/immunology , Histocompatibility Testing/methods , Humans , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Malaria, Falciparum/prevention & control , Male , Middle Aged , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology
11.
Curr Med Chem ; 9(4): 411-20, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11945117

ABSTRACT

The development of a vaccine for cancer has been difficult compared to the effective vaccines of infectious diseases. Most tumor antigens are not entirely foreign and are expressed on normal tissues, thus, making it difficult to induce strong immune responses against self antigens. A peptide mimic, however, may have the potential to generate greater immune responses than those induced to self peptides. In this review we discuss applications of peptide mimics for cancer immunotherapy which may ultimately prove useful in humans.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Molecular Mimicry , Neoplasms/therapy , Peptides/chemistry , Animals , Antibodies, Anti-Idiotypic/immunology , Autoimmune Diseases/immunology , Drug Design , Humans , Immunotherapy, Active , Models, Molecular , Peptide Library , Peptides/immunology
12.
Infect Immun ; 70(3): 1468-74, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11854234

ABSTRACT

T-cell responses directed against the circumsporozoite protein (CS) of Plasmodium falciparum can mediate protection against malaria. We determined the frequency of T cells reactive to different regions of the CS in the blood of donors naturally exposed to P. falciparum by examining T1 (gamma interferon [IFN-gamma] ELISPOT assay), T2 (interleukin 4 [IL-4] ELISPOT assay), and proliferative T-cell responses. The proliferative responses were weak, which confirmed previous observations. The responses to the CS in the IL-4 and IFN-gamma ELISPOT assays were also weak (<40 responding cells per 10(6) cells), much weaker than the response to the purified protein derivative of Mycobacterium tuberculosis in the same donors. Moreover, a response in one assay could not be used to predict a response in either of the other assays, suggesting that although these assays may measure different responding cells, all of the responses are weakly induced by natural exposure. Interestingly, the two different study populations used had significantly different T1 and T2 biases in their responses in the C terminus of the protein, suggesting that the extent of P. falciparum exposure can affect regulation of the immune system.


Subject(s)
Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Epitope Mapping , Female , Gambia , Geography , Humans , Interferon-gamma/analysis , Malaria, Falciparum/epidemiology , Male , Middle Aged , Molecular Sequence Data , Tuberculin/immunology
13.
J Immunol ; 167(8): 4729-37, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591804

ABSTRACT

Natural immunity to malaria is characterized by low level CD4 T cell reactivity detected by either lymphoproliferation or IFN-gamma secretion. Here we show a doubling in the detection rate of responders to the carboxyl terminus of circumsporozoite protein (CS) of Plasmodium falciparum by employing three T cell assays simultaneously: rapid IFN-gamma secretion (ex vivo ELISPOT), IFN-gamma secretion after reactivation of memory T cells and expansion in vitro (cultured ELISPOT), and lymphoproliferation. Remarkably, for no individual peptide did a positive response for one T cell effector function correlate with any other. Thus these CS epitopes elicited unique T cell response patterns in malaria-exposed donors. Novel or important epitope responses may therefore be missed if only one T cell assay is employed. A borderline correlation was found between anti-CS Ab levels and proliferative responses, but no correlation was found with ex vivo or cultured IFN-gamma responses. This suggested that the proliferating population, but not the IFN-gamma-secreting cells, contained cells that provide help for Ab production. The data suggest that natural immunity to malaria is a complex function of T cell subgroups with different effector functions and has important implications for future studies of natural T cell immunity.


Subject(s)
Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Animals , Epitopes , Gambia , Humans , Immunity, Cellular , Immunodominant Epitopes , Immunologic Memory , Interferon-gamma/metabolism , Lymphocyte Activation , Male , Molecular Sequence Data , Peptide Fragments/immunology , T-Lymphocyte Subsets/immunology
14.
Am J Trop Med Hyg ; 64(3-4): 194-203, 2001.
Article in English | MEDLINE | ID: mdl-11442217

ABSTRACT

The merozoite surface protein-1 (MSP1) is the most studied malaria blood-stage vaccine candidate. Lymphokines such as interferon gamma (IFN-gamma) and interleukin 4 (IL-4) may mediate blood-stage specific protection. Here we identify Plasmodiumfalciparum MSP1 T-cell epitopes capable of rapid induction of IFN-gamma and/or IL-4 from peripheral blood mononuclear cells of East and West African donors. Both allelic forms of these novel MSP1 T-cell epitopes were stimulatory. An unusually high numbers of Gambian responders (> 80%) to these epitopes were observed, suggesting that MSPI reactivity may have been underestimated previously in this population. Surprisingly, IFN-gamma responses to allelic T-cell epitopes failed to correlate with differential antigenic exposure in The Gambia compared to Kenya. These results suggest an unexpected level of immunoregulation of IFN-gamma response with variable allelic T-cell reactivity independent of the level of antigenic exposure. Further analysis of the mechanisms determining this response pattern may be required if vaccines are to overcome this allelic reactivity bias in malaria-exposed populations.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Alleles , Amino Acid Sequence , Animals , Case-Control Studies , DNA Primers , Female , Gambia/epidemiology , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Kenya/epidemiology , Malaria, Falciparum/epidemiology , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Prevalence
15.
Curr Opin Immunol ; 12(4): 437-41, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10899022

ABSTRACT

As global malaria mortality increases the urgency for vaccine development, analysis of immune responses in naturally exposed populations is providing clues to the nature of protective immunity. Recently, sophisticated immune evasion strategies adopted by the parasite have been analysed at the molecular level. More immunogenic vaccination strategies have been identified, providing renewed optimism that effective malaria control through vaccination should be feasible.


Subject(s)
Malaria/immunology , Animals , Endemic Diseases , Erythrocytes/immunology , Hematopoietic Stem Cells/immunology , Humans , Malaria/epidemiology , Malaria/prevention & control
16.
Curr Opin Mol Ther ; 2(4): 441-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11249775

ABSTRACT

Vaccination has eradicated smallpox and greatly decreased mortality and morbidity associated with a variety of viral and bacterial infectious diseases. However, conventional methodologies have failed to provide vaccines against many widespread deadly human diseases, among them HIV, malaria and cancer. Recombinant DNA vaccines have shown great promise in animal models in inducing protective immunity. In this review we will discuss their potential safe use in humans following recent advances in their use in animals, including non-human primates.


Subject(s)
Vaccines, DNA/therapeutic use , Animals , Antigen Presentation , Antigens/administration & dosage , Antigens/genetics , Biotechnology , Clinical Trials as Topic , DNA, Recombinant , Humans , Lymphocyte Activation , Primates , Safety , T-Lymphocytes/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
17.
Dev Biol (Basel) ; 104: 171-9, 2000.
Article in English | MEDLINE | ID: mdl-11713817

ABSTRACT

A generic approach to inducing high level CD8+ T cell responses would be of value for prophylactic and therapeutic immunisation against several infectious diseases. However, it has been very difficult to achieve such immune responses using available vaccination strategies. Malaria is one of several diseases against which a new generation of better CD8+ T cell-inducing vaccines might be useful and is unusual in that it allows assessment of vaccine efficacy in small numbers of volunteers in carefully controlled challenge studies. Here we review the identification of a heterologous prime-boost regime using DNA priming and recombinant modified vaccinia Ankara (MVA) boosting that induces high level T cell responses in both mice and non-human primates. Clinical trials to determine whether this prime-boost approach is immunogenic in humans are in progress.


Subject(s)
Malaria Vaccines/administration & dosage , Vaccines, DNA/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/immunology , Clinical Trials, Phase I as Topic , Genetic Vectors , Humans , Immunization, Secondary , Immunoassay , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Primates , Vaccinia virus/genetics , Vaccinia virus/immunology
18.
Immunol Rev ; 170: 29-38, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10566139

ABSTRACT

One of the current challenges in vaccine design is the development of antigen delivery systems or vaccination strategies that induce high protective levels of CD8+ T cells. These cells are crucial for protection against certain tumours and intracellular pathogens such as the liver-stage parasite of malaria. A liver-stage malaria vaccine should therefore include CD8+ T-cell-inducing components. This review provides an overview of prime-boost immunisation strategies that result in protective CD8+ T-cell responses against malaria with an emphasis on work from our laboratory. Possible mechanisms explaining why heterologous prime-boost strategies, in particular boosting with replication-impaired recombinant poxviruses, are so effective are discussed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary/methods , Adjuvants, Immunologic/administration & dosage , Animals , Epitopes/administration & dosage , Genetic Vectors , Humans , Malaria/immunology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Mice , Primates , Vaccines, DNA/administration & dosage
19.
J Infect Dis ; 180(5): 1656-64, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10515829

ABSTRACT

The RTS,S/SBAS2 vaccine confers sterile protection against Plasmodium falciparum sporozoite challenge. The mechanisms underlying this are of great interest, yet little is known about the immune effector mechanisms induced by this vaccine. The immune responses induced by RTS,S/SBAS2 were characterized in 10 malaria-naive volunteers. Several epitopes in the circumsporozoite protein (CSP) were identified as targets of cultured interferon (IFN)-gamma-secreting CD4+ T cells. RTS,S-specific IFN-gamma-secreting effector T cells were induced in 8 subjects; this ex vivo response mapped to a single peptide in Th2R. CSP-specific CD8+ cytotoxic T lymphocytes were not detected. RTS, S-specific IFN-gamma production was universal, whereas interleukin-4 and -5 production was rare. RTS,S-specific lymphoproliferative responses and antibodies to CSP were strongly induced in all volunteers. Responses waned with time but were boostable. Thus, RTS, S/SBAS2 is a potent inducer of Th1-type cellular and humoral immunity. These results highlight possible immune mechanisms of protection and have important implications for vaccine design in general.


Subject(s)
Antibodies, Protozoan/blood , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Th1 Cells/immunology , Vaccines, Synthetic/immunology , Adult , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , Epitopes/chemistry , Hepatitis B Surface Antigens/immunology , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Middle Aged , Molecular Sequence Data , Peptides/immunology , T-Lymphocytes, Regulatory/immunology , Vaccination , Vaccines, Synthetic/administration & dosage
20.
J Biol Chem ; 274(34): 24113-23, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446183

ABSTRACT

Monocyte/macrophages play important roles in regulating tissue growth and angiogenesis through the controlled release of heparin-binding growth factors such as fibroblast growth factor (FGF), vascular endothelial growth factor, and heparin binding epidermal growth factor. The action of these potent growth mediators is known to be regulated by adsorption to heparan sulfate proteoglycans (HSPGs) on the surface and within the extracellular matrix of other neighboring cells, which respectively promote or restrict interactions with their signal-transducing receptors on target cells. Here we report on the nature of HSPGs inducibly expressed on the surface of macrophages that confer these cells with the capacity to regulate endogenous growth factor activity. We reveal that activated human macrophages express only a single major 48-kDa cell surface HSPG, syndecan-2 (fibroglycan) as the result of de novo RNA and protein synthesis. In addition, we demonstrate this macrophage HSPG selectively binds the macrophage-derived growth factors FGF-2, vascular endothelial growth factor and heparin binding EGF and can present FGF-2 in a form that transactivates receptor-bearing BaF32 cells. These results define a novel and unique proteoglycan profile for macrophages and imply a key role for syndecan-2 in the delivery of sequestered growth factors by inflammatory macrophages for productive binding to their appropriate target cells in vivo.


Subject(s)
Fibroblast Growth Factors/physiology , Macrophage Activation , Macrophages/metabolism , Membrane Glycoproteins/physiology , Proteoglycans/physiology , Angiogenesis Inducing Agents/physiology , Cell Line , Heparin/metabolism , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Proteoglycans/analysis , Proteoglycans/genetics , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/physiology , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/physiology , Syndecan-2
SELECTION OF CITATIONS
SEARCH DETAIL
...