Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Immunity ; 28(5): 710-22, 2008 May.
Article in English | MEDLINE | ID: mdl-18468462

ABSTRACT

To explore the human T cell response to acute viral infection, we performed a longitudinal analysis of CD8(+) T cells responding to the live yellow fever virus and smallpox vaccines--two highly successful human vaccines. Our results show that both vaccines generated a brisk primary effector CD8(+) T cell response of substantial magnitude that could be readily quantitated with a simple set of four phenotypic markers. Secondly, the vaccine-induced T cell response was highly specific with minimal bystander effects. Thirdly, virus-specific CD8(+) T cells passed through an obligate effector phase, contracted more than 90% and gradually differentiated into long-lived memory cells. Finally, these memory cells were highly functional and underwent a memory differentiation program distinct from that described for human CD8(+) T cells specific for persistent viruses. These results provide a benchmark for CD8(+) T cell responses induced by two of the most effective vaccines ever developed.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Smallpox Vaccine/immunology , T-Lymphocyte Subsets/immunology , Yellow Fever Vaccine/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Lymphocyte Activation , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccination , Vaccinia virus/immunology , Yellow Fever Vaccine/metabolism
2.
Cancer Immunol Immunother ; 62(3): 529-39, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23069871

ABSTRACT

There is a complex interplay between the immune system and a developing tumor that is manifest in the way that the balance of T cell subsets in the local tumor environment reflects clinical outcome. Tumor infiltration by CD8(+) T cells and regulatory T cells (Treg) is associated with improved and reduced survival, respectively, in many cancer types. However, little is known of the prognostic value of immunological parameters measured in peripheral blood. In this study, peripheral CD8(+) T cells and Treg from 43 patients with malignant mesothelioma or advanced non-small-cell lung cancer scheduled to commence palliative chemotherapy were assessed by flow cytometry and evaluated for association with patient survival. Patients had a higher proportion of peripheral Treg, proliferating CD8(+) T cells and CD8(+) T cells with an activated effector phenotype compared with age-matched healthy controls. Higher proportions of Treg and proliferating CD8(+) T cells were both associated with poor survival in univariate analyses (hazard ratio [HR] 3.81, 95 % CI 1.69-8.57; p < 0.01 and HR 2.86, 95 % CI 1.26-6.50; p < 0.05, respectively). CD8(+) T cell proliferation was independently predictive of reduced survival in multivariate analysis (HR 2.58, 95 % CI 1.01-6.61; p < 0.05). These findings suggest that peripheral CD8(+) T cell proliferation can be a useful prognostic marker in patients with thoracic malignancies planned for palliative chemotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Mesothelioma/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Cell Proliferation , Female , Humans , Lung Neoplasms/mortality , Lymphocyte Activation , Male , Mesothelioma/mortality , Middle Aged , Prognosis , T-Lymphocytes, Regulatory/immunology
3.
Front Immunol ; 13: 902840, 2022.
Article in English | MEDLINE | ID: mdl-36311717

ABSTRACT

In the wake of the A/California/7/2009 H1N1 influenza pandemic vaccination campaigns in 2009-2010, an increased incidence of the chronic sleep-wake disorder narcolepsy was detected in children and adolescents in several European countries. Over the last decade, in-depth epidemiological and immunological studies have been conducted to investigate this association, which have advanced our understanding of the events underpinning the observed risk. Narcolepsy with cataplexy (defined as type-1 narcolepsy, NT1) is characterized by an irreversible and chronic deficiency of hypocretin peptides in the hypothalamus. The multifactorial etiology is thought to include genetic predisposition, head trauma, environmental triggers, and/or infections (including influenza virus infections), and an increased risk was observed following administration of the A/California/7/2009 H1N1 vaccine Pandemrix (GSK). An autoimmune origin of NT1 is broadly assumed. This is based on its strong association with a predisposing allele (the human leucocyte antigen DQB1*0602) carried by the large majority of NT1 patients, and on links with other immune-related genetic markers affecting the risk of NT1. Presently, hypotheses on the underlying potential immunological mechanisms center on molecular mimicry between hypocretin and peptides within the A/California/7/2009 H1N1 virus antigen. This molecular mimicry may instigate a cross-reactive autoimmune response targeting hypocretin-producing neurons. Local CD4+ T-cell responses recognizing peptides from hypocretin are thought to play a central role in the response. In this model, cross-reactive DQB1*0602-restricted T cells from the periphery would be activated to cross the blood-brain barrier by rare, and possibly pathogen-instigated, inflammatory processes in the brain. Current hypotheses suggest that activation and expansion of cross-reactive T-cells by H1N1/09 influenza infection could have been amplified following the administration of the adjuvanted vaccine, giving rise to a "two-hit" hypothesis. The collective in silico, in vitro, and preclinical in vivo data from recent and ongoing research have progressively refined the hypothetical model of sequential immunological events, and filled multiple knowledge gaps. Though no definitive conclusions can be drawn, the mechanistical model plausibly explains the increased risk of NT1 observed following the 2009-2010 H1N1 pandemic and subsequent vaccination campaign, as outlined in this review.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Narcolepsy , Child , Adolescent , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Influenza, Human/complications , Orexins , Narcolepsy/etiology , Narcolepsy/genetics , Peptides
4.
Curr Opin Immunol ; 20(5): 545-57, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18573339

ABSTRACT

Tumors can acquire mutations or hijack regulatory pathways of the host immune system to render them resistant to immune attack. Standard first line therapies such as chemotherapy and radiation were not thought to provoke natural immunity to cancer, but recent findings demonstrating that dying tumor cells present and release key signals to stimulate or evade neighboring leukocytes are challenging that view. Killing tumor cells in a manner that provides danger signals and tumor antigens in the right context promotes the engagement of innate and adaptive immunity; however, this response alone will not be effective against established cancer. Coincidently driving the immune response with specific monoclonal antibodies and other immunomodulators that activate and mature dendritic cells and co-stimulate T cells and other lymphocytes is one approach. Additionally releasing immune checkpoints and inhibiting tumor-derived molecules that prevent effective tumor immunity is another. Combined these approaches have enormous potential to improve the current outcomes from conventional cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/immunology , Dendritic Cells/immunology , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Combined Modality Therapy , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Receptors, Pattern Recognition/immunology , T-Lymphocytes, Cytotoxic/metabolism
5.
J Immunol ; 183(12): 7898-908, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20007574

ABSTRACT

Tumors have evolved multiple mechanisms to evade immune destruction. One of these is expression of T cell inhibitory ligands such as programmed death-ligand 1 (PD-L1; B7-H1). In this study, we show that PD-L1 is highly expressed on mesothelioma tumor cells and within the tumor stroma. However, PD-L1 blockade only marginally affected tumor growth and was associated with the emergence of activated programmed death-1(+) ICOS(+) CD4 T cells in tumor-draining lymph nodes, whereas few activated CD8 T cells were present. Full activation of antitumor CD8 T cells, characterized as programmed death-1(+) ICOS(+) Ki-67(+) and displaying CTL activity, was only observed when CD4 T cells were depleted, suggesting that a population of suppressive CD4 T cells exists. ICOS(+) foxp3(+) regulatory T cells were found to be regulated through PD-L1, identifying one potentially suppressive CD4 T cell population. Thus, PD-L1 blockade activates antitumor CD8 T cell most potently in the absence of CD4 T cells. These findings have implications for the development of PD-L1-based therapies.


Subject(s)
Antigens, Surface/physiology , Apoptosis Regulatory Proteins/physiology , B7-1 Antigen/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Membrane Glycoproteins/physiology , Mesothelioma/immunology , Peptides/physiology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , B7-H1 Antigen , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Contraindications , Female , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Membrane Glycoproteins/antagonists & inhibitors , Mesothelioma/pathology , Mesothelioma/therapy , Mice , Mice, Inbred BALB C , Mice, Transgenic , Peptides/antagonists & inhibitors , Programmed Cell Death 1 Receptor , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocytes, Regulatory/metabolism
6.
J Immunol ; 182(9): 5217-24, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380767

ABSTRACT

Topical application of tumors with the TLR7 agonist imiquimod is an effective adjunct treatment for a range of primary dermatological cancers. However, for therapy to be effective against a broad range of solid tumor types, it must promote a strong systemic antitumor response that targets metastases in addition to primary tumor. We therefore investigated the potential of locally delivered imiquimod to stimulate an effective systemic antitumor response in a murine model of malignant mesothelioma (AB1-HA) with primary and distal tumors (dual tumor). Persistent delivery of imiquimod into primary tumor significantly retarded tumor growth in all treated mice compared with vehicle control. This local antitumor immune response required both CD8 T cells and NK cells, but not CD4 T cells, and was reliant on type I IFN induction. In vivo CTL studies and Ly6A/E staining of lymphocytes suggested that local imiquimod treatment had indeed induced a systemic, Ag-specific CD8 response. However, notably this response was not sufficient to retard the growth of an untreated distal tumor. Because local imiquimod treatment did not induce significant CD4 T cell responses, we investigated the efficacy of combining imiquimod with agonistic CD40 Ab (as a surrogate for CD4 T cell help). Combination of locally delivered imiquimod with systemic anti-CD40 immunotherapy not only significantly enhanced the local antitumor response, with 30% complete resolution, but it was also effective at significantly retarding growth of distal tumor. These results demonstrate that antitumor responses induced by locally delivered TLR7 agonists can be harnessed systemically for treating distal tumor.


Subject(s)
Aminoquinolines/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD40 Antigens/immunology , Membrane Glycoproteins/agonists , Mesothelioma/immunology , Mesothelioma/therapy , Toll-Like Receptor 7/agonists , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Aminoquinolines/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Therapy, Combination , Female , Imiquimod , Interferon Type I/administration & dosage , Interferon-gamma/administration & dosage , Killer Cells, Natural/immunology , Ligands , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Mesothelioma/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mice, Transgenic , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/therapeutic use
7.
J Gen Virol ; 91(Pt 10): 2497-506, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20554798

ABSTRACT

The severity of respiratory syncytial virus (RSV) infections appears to differ with age in both humans and bovines. A primary RSV infection in naïve infants and in young calves runs a more severe course when they are 1-6 months old than in their first month of life. The relative lack of clinical signs in the first month of age may be due to high levels of maternally derived neutralizing antibodies or low exposure to infectious virus. This study examined whether age-dependent differences in the pathogenesis of bovine RSV (bRSV) between neonatal and young calves may be due to differences in age-dependent immunocompetence. To study the effect of age and immune parameters on bRSV disease in neonatal and young calves, neonatal (1-day-old) calves without maternally derived antibodies were infected experimentally with bRSV and the severity of disease and immune responses were evaluated in comparison with disease in similar 6-week-old infected calves. Neonatal calves had more extensive virus replication and lung consolidation, but lower pro-inflammatory [in particular tumour necrosis factor alpha (TNF-α)] responses, specific humoral immune responses, lung neutrophilic infiltration and clinical signs of disease than 6-week-old calves. The lack of correlation between virus replication and clinical signs suggests an important role of pro-inflammatory cytokines, in particular TNF-α, in the disease. The capacity to produce pro-inflammatory TNF-α appeared to increase with age, and may explain the age-dependent differences in RSV pathogenesis.


Subject(s)
Cattle Diseases/immunology , Cattle Diseases/pathology , Immunocompetence , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Age Factors , Animals , Animals, Newborn , Antibodies, Viral/immunology , Cattle , Cytokines/immunology , Cytokines/metabolism , Lung/immunology , Lung/pathology , Neutrophils/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Severity of Illness Index
8.
J Virol ; 83(9): 4386-94, 2009 May.
Article in English | MEDLINE | ID: mdl-19211743

ABSTRACT

During some persistent viral infections, virus-specific T-cell responses wane due to the antigen-specific deletion or functional inactivation (i.e., exhaustion) of responding CD8 T cells. T-cell exhaustion often correlates with high viral load and is associated with the expression of the inhibitory receptor PD-1. In other infections, functional T cells are observed despite high levels of pathogen persistence. The reasons for these different T-cell fates during chronic viral infections are not clear. Here, we tracked the fate of virus-specific CD8 T cells in lymphocytic choriomeningitis virus (LCMV)-infected mice during viral clearance, the persistence of wild-type virus, or the selection and persistence of a viral variant that abrogates the presentation of a single epitope. Viral clearance results in PD-1(lo) functional virus-specific CD8 T cells, while the persistence of wild-type LCMV results in high PD-1 levels and T-cell exhaustion. However, following the emergence of a GP35V-->A variant virus that abrogates the presentation of the GP33 epitope, GP33-specific CD8 T cells remained functional, continued to show low levels of PD-1, and reexpressed CD127, a marker of memory T-cell differentiation. In the same animals and under identical environmental conditions, CD8 T cells recognizing nonmutated viral epitopes became physically deleted or were PD-1(hi) and nonfunctional. Thus, the upregulation of PD-1 and the functional inactivation of virus-specific T cells during chronic viral infection is dependent upon continued epitope recognition. These data suggest that optimal strategies for vaccination should induce high-magnitude broadly specific T-cell responses that prevent cytotoxic T-lymphocyte escape and highlight the need to evaluate the function of vaccine-induced T cells in the context of antigens presented during virus persistence.


Subject(s)
Antigens, Differentiation/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Base Sequence , Chronic Disease , Female , Glycoproteins/genetics , Glycoproteins/metabolism , Interleukin-7 Receptor alpha Subunit/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/isolation & purification , Mice , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/metabolism , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Titrimetry , Viral Proteins/genetics , Viral Proteins/metabolism
9.
Sci Transl Med ; 12(569)2020 11 11.
Article in English | MEDLINE | ID: mdl-33177181

ABSTRACT

The current routine use of adjuvants in human vaccines provides a strong incentive to increase our understanding of how adjuvants differ in their ability to stimulate innate immunity and consequently enhance vaccine immunogenicity. Here, we evaluated gene expression profiles in cells from whole blood elicited in naive subjects receiving the hepatitis B surface antigen formulated with different adjuvants. We identified a core innate gene signature emerging 1 day after the second vaccination and that was shared by the recipients of vaccines formulated with adjuvant systems AS01B, AS01E, or AS03. This core signature associated with the magnitude of the hepatitis B surface-specific antibody response and was characterized by positive regulation of genes associated with interferon-related responses or the innate cell compartment and by negative regulation of natural killer cell-associated genes. Analysis at the individual subject level revealed that the higher immunogenicity of AS01B-adjuvanted vaccine was linked to its ability to induce this signature in most vaccinees even after the first vaccination. Therefore, our data suggest that adjuvanticity is not strictly defined by the nature of the receptors or signaling pathways it activates but by the ability of the adjuvant to consistently induce a core inflammatory signature across individuals.


Subject(s)
Hepatitis B Vaccines , Influenza Vaccines , Adjuvants, Immunologic , Antibodies, Viral , Hepatitis B Surface Antigens/genetics , Humans , Immunogenicity, Vaccine , Vaccination
10.
Cancer Immunol Immunother ; 58(8): 1219-28, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19052741

ABSTRACT

Tumor cell death potentially engages with the immune system. However, the efficacy of anti-tumor chemotherapy may be limited by tumor-driven immunosuppression, e.g., through CD25+ regulatory T cells. We addressed this question in a mouse model of mesothelioma by depleting or reconstituting CD25+ regulatory T cells in combination with two different chemotherapeutic drugs. We found that the efficacy of cyclophosphamide to eradicate established tumors, which has been linked to regulatory T cell depletion, was negated by adoptive transfer of CD25+ regulatory T cells. Analysis of post-chemotherapy regulatory T cell populations revealed that cyclophosphamide depleted cycling (Ki-67(hi)) T cells, including foxp3+ regulatory CD4+ T cells. Ki-67(hi) CD4+ T cells expressed increased levels of two markers, TNFR2 and ICOS, that have been associated with a maximally suppressive phenotype according to recently published studies. This suggest that cyclophosphamide depletes a population of maximally suppressive regulatory T cells, which may explain its superior anti-tumor efficacy in our model. Our data suggest that regulatory T cell depletion could be used to improve the efficacy of anti-cancer chemotherapy regimens. Indeed, we observed that the drug gemcitabine, which does not deplete cycling regulatory T cells, eradicates established tumors in mice only when CD25+ CD4+ T cells are concurrently depleted. Cyclophosphamide could be used to achieve regulatory T cell depletion in combination with chemotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion , Mesothelioma/drug therapy , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Inducible T-Cell Co-Stimulator Protein , Kaplan-Meier Estimate , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Mesothelioma/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes, Regulatory/immunology , Gemcitabine
11.
Hum Vaccin Immunother ; 15(12): 2980-2992, 2019.
Article in English | MEDLINE | ID: mdl-31149870

ABSTRACT

The human commensal Staphylococcus aureus (SA) is a leading cause of skin/soft tissue and surgical-site infections, and bacteremia. Functional antibodies and T-cell-mediated immunity, particularly Th1/Th17 responses, are thought to mediate protection. Vaccine development may be hindered by modulation of vaccine-induced T cells by pathogen-activated immunoregulatory responses, e.g., via IL-10.We screened SA proteins for CD4+ T-cell-activating and IL-10/IL-17-inducing capacities using healthy donor-derived PBMCs. Responses were characterized (Th1/Th17/Th22/immunosuppressive IL-10-producing cells) using intracellular cytokine staining and flow cytometry. Phenotypic plasticity of Th1/Th17 cells was evaluated under pro- or anti-inflammatory conditions using modulatory cytokines. The impact of vaccination on SA-specific memory responses was assessed using samples from a clinical trial evaluating AS03-adjuvanted and non-adjuvanted multicomponent (CPS5/CPS8/α-toxin/ClfA) vaccines (NCT01160172).The donors exhibited SA-specific memory T-cell responses, indicative of pre-existing immunity to SA. We identified effective activators of Th1 responses (EbhA/IsaA/SdrE/MntC/Aaa/α-toxin), and Th17 and Th1/Th17 responses (EbhA/IsaA/SdrE and, to a lesser extent, α-toxin), but not of Th22 responses or IL-10 production. MRPII, IsdA, and ClfA were inefficient CD4+ T-cell activators in our assays. IL-10, likely produced by innate immune cells, influenced mainly Th1 cells by suppressing IFN-γ production. The memory CD4+ T-cells observed after long-term stimulation with α-toxin and ClfA indicated that vaccination with these proteins had induced expansion of pre-existing Th1 but not Th17 responses, without apparent adjuvant effect, confirming the trial data. The Th1/Th17-driving proteins (EbhA/IsaA/SdrE) shared low IL-10-promoting abilities and restricted phenotypic plasticity under pro- and anti-inflammatory conditions.Given the complex immunopathology and multiple virulence factors, identification of Th1/Th17-driving antigens, adjuvants and administration routes, and delineation of the role of memory responses, may advance vaccine development.


Subject(s)
Bacterial Proteins/immunology , Cell Plasticity/immunology , Immunologic Memory , Staphylococcal Infections/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Female , Healthy Volunteers , Humans , Immunity, Cellular , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Male , Middle Aged , Phenotype , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Vaccination
12.
Cancer Res ; 66(2): 601-4, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16423984

ABSTRACT

This review shows how tumor antigen cross-presentation is affected by the major therapeutic modalities including chemotherapy, radiotherapy, and surgery. We argue that this process could affect the way that a tumor works as its own cellular vaccine, and that it is differentially modulated by the choice of treatment.


Subject(s)
Antigen Presentation , Cancer Vaccines/immunology , Cross-Priming/immunology , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Combined Modality Therapy , Humans , Neoplasms/immunology , Neoplasms/therapy
13.
Hum Vaccin Immunother ; 14(1): 17-27, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28934066

ABSTRACT

The candidate malaria vaccine RTS,S has demonstrated 45.7% efficacy over 18 months against all clinical disease in a phase-III field study of African children. RTS,S targets the circumsporozoite protein (CSP), which is expressed on the Plasmodium sporozoite during the pre-erythrocyte stage of its life-cycle; the stage between mosquito bite and liver infection. Early in the development of RTS,S, it was recognized that CSP-specific cell-mediated immunity (CMI) was required to complement CSP-specific antibody-mediated immunity. In reviewing RTS,S clinical studies, associations between protection and various types of CMI (CSP-specific CD4+ T cells and INF-γ ELISPOTs) have been identified, but not consistently. It is plausible that certain CD4+ T cells support antibody responses or co-operate with other immune-cell types to potentially elicit protection. However, the identities of vaccine correlates of protection, implicating either CSP-specific antibodies or T cells remain elusive, suggesting that RTS,S clinical trials may benefit from additional immunogenicity analyses that can be informed by the results of controlled human malaria infection studies.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/therapy , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Antigens, Protozoan/immunology , Clinical Trials as Topic , Humans , Immunity, Cellular/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Treatment Outcome
14.
Front Immunol ; 9: 564, 2018.
Article in English | MEDLINE | ID: mdl-29632533

ABSTRACT

Systems biology has the potential to identify gene signatures associated with vaccine immunogenicity and protective efficacy. The main objective of this study was to identify optimal postvaccination time points for evaluating peripheral blood RNA expression profiles in relation to vaccine immunogenicity and potential efficacy in recipients of the candidate tuberculosis vaccine M72/AS01. In this phase II open-label study (NCT01669096; https://clinicaltrials.gov/), healthy Bacillus Calmette-Guérin-primed, HIV-negative adults were administered two doses (30 days apart) of M72/AS01. Twenty subjects completed the study and 18 subjects received two doses. Blood samples were collected pre-dose 1, pre-dose 2, and 1, 7, 10, 14, 17, and 30 days post-dose 2. RNA expression in whole blood (WB) and peripheral blood mononuclear cells (PBMCs) was quantified using microarray technology. Serum interferon-gamma responses and M72-specific CD4+ T cell responses to vaccination, and the observed safety profile were similar to previous trials. Two different approaches were utilized to analyze the RNA expression data. First, a kinetic analysis of RNA expression changes using blood transcription modules revealed early (1 day post-dose 2) activation of several pathways related to innate immune activation, both in WB and PBMC. Second, using a previously identified gene signature as a classifier, optimal postvaccination time points were identified. Since M72/AS01 efficacy remains to be established, a PBMC-derived gene signature associated with the protective efficacy of a similarly adjuvanted candidate malaria vaccine was used as a proxy for this purpose. This approach was based on the assumption that the AS01 adjuvant used in both studies could induce shared innate immune pathways. Subjects were classified as gene signature positive (GS+) or gene signature negative (GS-). Assignments of subjects to GS+ or GS- groups were confirmed by significant differences in RNA expression of the gene signature genes in PBMCs at 14 days post-dose 2 relative to prevaccination and in WB samples at 7, 10, 14, and 17 days post-dose 2 relative to prevaccination. Hence, in comparison with a prevaccination, 7, 10, 14, and 17 days postvaccination appeared to be suitable time points for identifying potentially clinically relevant transcriptome responses to M72/AS01 in WB samples.


Subject(s)
BCG Vaccine/administration & dosage , Lipid A/analogs & derivatives , RNA, Messenger/immunology , Saponins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Drug Combinations , Female , Gene Expression Profiling , Humans , Interferon-gamma/blood , Interferon-gamma/immunology , Kinetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipid A/administration & dosage , Male , Middle Aged , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , RNA, Messenger/blood , RNA, Messenger/genetics , Recombinant Proteins/immunology , Vaccination , Young Adult
15.
Cancer Res ; 65(17): 7580-4, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140921

ABSTRACT

Resection alone is rarely curative for advanced tumors, but the outcome generally improves with adjuvant therapy. We have previously shown that a combination of traditional chemotherapy (gemcitabine) and immunotherapy (anti-CD40/FGK-45) without surgery is synergistic and can lead to long-term cure when applied to small tumors. Such cured animals have immunologic memory and are protected from rechallenge. Here we investigate the effectiveness of combination chemotherapy and immunotherapy after partial or complete surgical debulking of large tumors. We found that complete resection followed by combination chemotherapy/immunotherapy led to a high rate of cure (>80%) but failed to induce a long-term, tumor-specific memory. Partial debulking followed by combination therapy elicited the same proportion of cured animals but in contrast to complete resection, a memory response was invoked. We postulate that chemotherapy induced apoptosis of the residual tumor cells following incomplete resection is absolutely required for the induction of long-term immunologic memory.


Subject(s)
Antibodies/pharmacology , Antimetabolites, Antineoplastic/pharmacology , CD40 Antigens/immunology , Deoxycytidine/analogs & derivatives , Mesothelioma/immunology , Mesothelioma/therapy , Animals , Cell Line, Tumor , Chemotherapy, Adjuvant , Deoxycytidine/pharmacology , Immunization, Passive/methods , Immunologic Memory , Mesothelioma/drug therapy , Mesothelioma/surgery , Mice , Mice, Inbred BALB C , Gemcitabine
16.
Clin Vaccine Immunol ; 24(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-28446441

ABSTRACT

We investigated the role of AS03A (here AS03), an α-tocopherol oil-in-water emulsion-based adjuvant system, on the long-term persistence of humoral and cell-mediated immune responses to A(H1N1)pdm09 influenza vaccines. In two studies, a total of 261 healthy adults (≤60 years old) were randomized to receive two doses of AS03-adjuvanted vaccine containing 3.75 µg of hemagglutinin (HA) or nonadjuvanted vaccine containing 15 µg of hemagglutinin (in study A) or 3.75 µg of hemagglutinin (in study B) 21 days apart. Hemagglutination inhibition (HI) antibody, memory B-cell, and CD4+/CD8+ T-cell responses were characterized up to 1 year following dose 1. We also assessed the effects of age and seasonal influenza vaccination history. AS03-adjuvanted (3.75 µg HA) vaccine and nonadjuvanted vaccine at 15 µg but not at 3.75 µg HA elicited HI antibody responses persisting at levels that continued to meet European licensure criteria through month 12. At month 12, the geometric mean titer for AS03-adjuvanted vaccine was similar to that for nonadjuvanted (15-µg) vaccine in study A (1:86 and 1:88, respectively) and higher than that for nonadjuvanted (3.75-µg) vaccine in study B (1:77 and 1:35, respectively). A(H1N1)pdm09-specific CD4+ T-cell and B-cell responses were stronger in AS03-adjuvanted groups and persisted only in these groups for 12 months at levels exceeding prevaccination frequencies. Advancing age and a seasonal vaccination history tended to reduce HI antibody and memory B-cell responses and, albeit less consistently, CD4+ T-cell responses. Thus, AS03 seemed to enhance the persistence of humoral and cell-mediated responses to A(H1N1)pdm09 vaccine, allowing for antigen sparing and mitigating potential negative effects of age and previous seasonal vaccination. (These studies have been registered at ClinicalTrials.gov under registration no. NCT00968539 and NCT00989287.).


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunity, Humoral , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Polysorbates/administration & dosage , Squalene/administration & dosage , alpha-Tocopherol/administration & dosage , Adolescent , Adult , Age Factors , Antibodies, Viral/blood , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Combinations , Female , Follow-Up Studies , Healthy Volunteers , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity, Cellular , Influenza Vaccines/administration & dosage , Male , Middle Aged , Time Factors , Young Adult
17.
Front Immunol ; 8: 557, 2017.
Article in English | MEDLINE | ID: mdl-28588574

ABSTRACT

The RTS,S candidate malaria vaccine can protect against controlled human malaria infection (CHMI), but how protection is achieved remains unclear. Here, we have analyzed longitudinal peripheral blood transcriptome and immunogenicity data from a clinical efficacy trial in which healthy adults received three RTS,S doses 4 weeks apart followed by CHMI 2 weeks later. Multiway partial least squares discriminant analysis (N-PLS-DA) of transcriptome data identified 110 genes that could be used in predictive models of protection. Among the 110 genes, 42 had known immune-related functions, including 29 that were related to the NF-κB-signaling pathway and 14 to the IFN-γ-signaling pathway. Post-dose 3 serum IFN-γ concentrations were also correlated with protection; and N-PLS-DA of IFN-γ-signaling pathway transcriptome data selected almost all (44/45) of the representative genes for predictive models of protection. Hence, the identification of the NF-κB and IFN-γ pathways provides further insight into how vaccine-mediated protection may be achieved.

19.
J Immunol Methods ; 285(1): 41-54, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871533

ABSTRACT

We describe a three-color flow cytometry assay for the detection of virus-specific CD4+ and CD8+ T cells in the cat. The assay is based upon detection of intracellular TNFalpha using the cross-reactive mAb 6401.1111, raised against the human cytokine. Allophycocyanin-conjugated mAb 6401.1111 specifically stained feline TNFalpha-producing murine cells and also Staphylococcus aureus Enterotoxin B-stimulated feline T cells, thus providing formal evidence for cross-reactivity. By using the anti-TNFalpha mAb in combination with PE- and FITC-conjugated mAbs against feline CD4 and CD8, respectively, antiviral CD4+ and CD8+ T cells could be identified in the peripheral blood and in the spleens of feline infectious peritonitis virus-infected cats. Moreover, feline calicivirus (FCV)-specific CD4+ T cells were detected in the spleens of FCV-vaccinated cats. As antigen-presenting cells (APCs), we used immortalized autologous fibroblast cell lines, PBMC or splenocytes. A straightforward protocol, in which splenocyte preparations served both as APCs and effector cells, consistently yielded best results. The assay will permit further studies of the cellular immune responses in cats during natural and experimental viral infections. It will contribute to vaccine development against feline viruses by facilitating the identification of T cell antigens and epitopes, and by allowing the quantitative detection of virus-specific T cells after vaccination. Furthermore, the assay will add to the value of those systems in which viral infections of the cat serve as models for human disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cats/immunology , Cell Separation/methods , Flow Cytometry/methods , Animals , Antibodies, Monoclonal , Antigen-Presenting Cells/immunology , Base Sequence , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Calicivirus, Feline/immunology , Cell Line , Coronavirus, Feline/immunology , DNA, Complementary/genetics , Fluorescent Dyes , Humans , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Viral Vaccines/immunology
20.
Sci Transl Med ; 6(246): 246ps8, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25101885

ABSTRACT

The study of influenza vaccines has revealed potential interactions between preexisting immunological memory and antigenic context and/or adjuvantation. In the face of antigenic diversity, the process of generating B cell adaptability is driven by cross-reactive CD4 memory cells, such as T follicular helper cells from previous infections or vaccinations. Although such "helped" B cells are capable of adapting to variant antigens, lack of CD4 help could lead to a suboptimal antibody response. Collectively, this indicates an interplay between CD4 T cells, adjuvant, and B cell adaptability.


Subject(s)
Adaptation, Physiological/immunology , B-Lymphocytes/immunology , Influenza, Human/immunology , Adjuvants, Immunologic , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/immunology , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL