Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Vaccine ; 2024 May 19.
Article in English | MEDLINE | ID: mdl-38769033

ABSTRACT

The emergence and ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need for rapid vaccine development platforms that can be updated to counteract emerging variants of currently circulating and future emerging coronaviruses. Here we report the development of a "train model" subunit vaccine platform that contains a SARS-CoV-2 Wuhan S1 protein (the "engine") linked to a series of flexible receptor binding domains (RBDs; the "cars") derived from SARS-CoV-2 variants of concern (VOCs). We demonstrate that these linked subunit vaccines when combined with Sepivac SWE™, a squalene in water emulsion (SWE) adjuvant, are immunogenic in Syrian hamsters and subsequently provide protection from infection with SARS-CoV-2 VOCs Omicron (BA.1), Delta, and Beta. Importantly, the bivalent and trivalent vaccine candidates offered protection against some heterologous SARS-CoV-2 VOCs that were not included in the vaccine design, demonstrating the potential for broad protection against a range of different VOCs. Furthermore, these formulated vaccine candidates were stable at 2-8 °C for up to 13 months post-formulation, highlighting their utility in low-resource settings. Indeed, our vaccine platform will enable the development of safe and broadly protective vaccines against emerging betacoronaviruses that pose a significant health risk for humans and agricultural animals.

2.
J Radiol Prot ; 42(1)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35037901

ABSTRACT

The field of radiation countermeasures is growing, however, currently there are no effective and non-toxic compounds which could be administered orally to the individuals post exposure to high doses of ionising radiation. The pigment melanin is ubiquitous through all kingdoms of life and provides selective advantage under radiation stress through its role as a chemical and physical shield, and its capacity to respond and react to exposures. Soluble allomelanin was administered to mice following whole-body exposure to lethal or sublethal doses of gamma radiation to determine its capacity to mitigate the effects of acute radiation syndrome, and its utility as a radiation countermeasure. Allomelanin has shown a trend to improve survival post an 8 Gy sublethal radiation exposure when administered up to 48 h post-irradiation. Furthermore, it improved median and overall survival to a 10 Gy lethal radiation exposure, specifically when administered at 24 h post-irradiation. Histological analysis on the jejunum region of the small intestine of this treatment group indicated that alterations of the mucosal and submucosal architecture, and disruption of the lymphatic system associated with lethal radiation exposure were mitigated when allomelanin was administered at 24 h post-irradiation. Based on this work soluble allomelanin derived from a fungal source could serve as an easily sourced, cost-effective, and viable countermeasure to accidental radiation exposure and merits further investigation.


Subject(s)
Acute Radiation Syndrome , Melanins , Animals , Gamma Rays , Mice , Radiation Dosage , Whole-Body Irradiation/adverse effects
3.
Cancer Med ; 10(3): 1128-1140, 2021 02.
Article in English | MEDLINE | ID: mdl-33347715

ABSTRACT

PURPOSE: Despite the availability of new drugs, many patients with acute myeloid leukemia (AML) do not achieve remission and outcomes remain poor. Venetoclax is a promising new therapy approved for use in combination with a hypomethylating agent or with low-dose cytarabine for the treatment of newly diagnosed older AML patients or those ineligible for intensive chemotherapy. 225 Actinium-lintuzumab (225 Ac-lintuzumab) is a clinical stage radioimmunotherapy targeting CD33 that has shown evidence of single-agent activity in relapsed/refractory AML. Increased expression of MCL-1 is a mediator of resistance to venetoclax in cancer. EXPERIMENTAL DESIGN: Here we investigated the potential for 225 Ac-lintuzumab-directed DNA damage to suppress MCL-1 levels as a possible mechanism of reversing resistance to venetoclax in two preclinical in vivo models of AML. RESULTS: We demonstrated that 225 Ac-lintuzumab in combination with venetoclax induced a synergistic increase in tumor cell killing compared to treatment with either drug alone in venetoclax-resistant AML cell lines through both an induction of double-stranded DNA breaks (DSBs) and depletion of MCL-1 protein levels. Further, this combination led to significant tumor growth control and prolonged survival benefit in venetoclax-resistant in vivo AML models. CONCLUSIONS: There results suggest that the combination of 225 Ac-lintuzumab with venetoclax is a promising therapeutic strategy for the treatment of patients with venetoclax-resistant AML. Clinical trial of this combination therapy (NCT03867682) is currently ongoing.


Subject(s)
Actinium/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/immunology , Sulfonamides/pharmacology , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , Cell Proliferation , Female , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncotarget ; 11(39): 3571-3581, 2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33062193

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies, and adoptive cell therapy (ACT) in general, represent one of the most promising anti-cancer strategies. Conditioning has been shown to improve the immune homeostatic environment to enable successful ACT or CAR-T engraftment and expansion in vivo following infusion, and represents potential point of intervention to decrease serious toxicities following CAR-T treatment. In contrast to relatively non-specific chemotherapy-derived lymphodepletion, targeted lymphodepletion with radioimmunotherapy (RIT) directed to CD45 may be a safer and more effective alternative to target and deplete immune cells. Here we describe the results of preclinical studies with an anti-mouse CD45 antibody 30F11, labeled with two different beta-emitters 131Iodine (131I) and 177Lutetium (177Lu), to investigate the effect of anti-CD45 RIT lymphodepletion on immune cell types and on tumor control in a model of adoptive cell therapy. Treatment of mice with 3.7 MBq 131I-30F11 or 1.48 MBq 177Lu-30F11 safely depleted immune cells such as spleen CD4+ and CD8+ T Cells, B and NK cells as well as Tregs in OT I tumor model while sparing RBC and platelets and enabled E. G7 tumor control. Our results support the application of CD45-targeted RIT lymphodepletion with a non-myeloablative dose of 131I-30F11 or 177Lu-30F11 antibody prior to adoptive cell therapy.

5.
Nucl Med Biol ; 82-83: 80-88, 2020.
Article in English | MEDLINE | ID: mdl-32113033

ABSTRACT

BACKGROUND: cART has significantly improved the life expectancy of people living with HIV (PLWH). However, it fails to eliminate the long-lived reservoir of latent HIV-infected cells. Radioimmunotherapy (RIT) relies on antigen-specific monoclonal antibodies (mAbs) for targeted delivery of lethal doses of ionizing radiation to cells. Previously, we have demonstrated that human mAb 2556 against HIV gp41 conjugated with 213Bismuth radioisotope (t1/2 = 46 min, alpha-emitter) selectively killed HIV-infected cells. 225Actinium (t1/2 = 9.92 d, alpha-emitter) and 177Lutetium (t1/2 = 6.7 d, beta-emitter) are two long-lived clinically proven radioisotopes for cancer treatment which might be more effective in killing infected cells systemically and in CNS. METHODS: In this study we have conjugated 2556 mAb with 213Bi, 225Ac and 177Lu, and compared their ability to kill HIV-infected human peripheral blood mononuclear cells (PBMCs) and monocytes. PBMCs and monocytes from healthy donors were infected with HIVp49.5 and treated in vitro with increasing concentrations of 213Bi (4-20 µCi)-, 225Ac (20-100 nCi)- and 177Lu (4-50 µCi)-2556 mAb. RESULTS: After three days post-treatment of infected PBMCs and monocytes, 213Bi- and 177Lu-conjugated 2556 mAb reduced virus production measured by p24 level in a dose-dependent manner, whereas, 225Ac-2556 showed minimal effect. However, seven days post-treatment all three radioisotopes showed significantly more pronounced reduction of virus replication as compared to control labeled mAb with 225Ac-2556 showing the least non-specific killing. CONCLUSION: These results indicate that RIT holds promise as a novel treatment option for the eradication of HIV-infected cells that merits further study in combination with cART and reactivation drugs.


Subject(s)
Antibodies, Monoclonal/immunology , HIV-1/physiology , Membrane Glycoproteins/immunology , Antibodies, Monoclonal/chemistry , Cell Line , DNA Breaks, Double-Stranded/radiation effects , HIV-1/radiation effects , Humans , Isotope Labeling , Leukocytes, Mononuclear/virology , Monocytes/virology
6.
Antiviral Res ; 171: 104594, 2019 11.
Article in English | MEDLINE | ID: mdl-31470041

ABSTRACT

Respiratory syncytial virus (RSV) is responsible for a large proportion of acute lower respiratory tract infections, specifically in children. Pneumonia virus of mice (PVM) causes similar lung pathology and clinical disease in rodents, and is therefore an appropriate model of RSV infection. Previously, we demonstrated that a single intranasal dose of P-I-P, a novel immunomodulator composed of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene, confers protection in Balb/c mice for up to 3 days from lethal PVM-15 infection. In the present study a dual intranasal treatment with P-I-P was shown to extend the duration of the protection conferred by P-I-P from PVM-15 challenge. Balb/c mice treated twice with P-I-P showed higher survival rates and milder clinical signs when compared to animals that received a single P-I-P dose. While the mice treated with two consecutive doses of P-I-P experienced some weight loss, they all recovered. The dual P-I-P treatment mediated infiltration of several innate immune cells into the BALF and lung, including alveolar macrophages, neutrophils, and γδ T cells. Partial depletion of alveolar macrophages decreased survival rates and exacerbated clinical signs of mice subjected to the P-I-P dual treatment regime followed by PVM-15 challenge. This suggests that the alveolar macrophage is at least partially responsible for the protection elicited by this novel prophylactic treatment strategy.


Subject(s)
Immunity, Innate , Immunologic Factors/pharmacology , Macrophages/drug effects , Macrophages/immunology , Murine pneumonia virus/drug effects , Murine pneumonia virus/immunology , Pneumovirus Infections/immunology , Pneumovirus Infections/virology , Animals , Cell Line , Cytokines/biosynthesis , Cytokines/blood , Female , Host-Pathogen Interactions , Immunologic Factors/administration & dosage , Macrophages/metabolism , Macrophages/virology , Mice , Pneumovirus Infections/drug therapy , Pneumovirus Infections/mortality
7.
Expert Rev Vaccines ; 18(5): 505-521, 2019 05.
Article in English | MEDLINE | ID: mdl-31009255

ABSTRACT

INTRODUCTION: Adjuvants form an integral component in most of the inactivated and subunit vaccine formulations. Careful and proper selection of adjuvants helps in promoting appropriate immune responses against target pathogens at both innate and adaptive levels such that protective immunity can be elicited. Areas covered: Herein, we describe the recent progress in our understanding of the mode of action of adjuvants that are licensed for use in human vaccines or in clinical or pre-clinical stages at both innate and adaptive levels. Different pathogens have distinct characteristics, which require the host to mount an appropriate immune response against them. Adjuvants can be selected to elicit a tailor-made immune response to specific pathogens based on their unique properties. Identification of biomarkers of adjuvanticity for several candidate vaccines using omics-based technologies can unravel the mechanism of action of modern and experimental adjuvants. Expert opinion: Adjuvant technology has been revolutionized over the last two decades. In-depth understanding of the role of adjuvants in activating the innate immune system, combined with systems vaccinology approaches, have led to the development of next-generation, novel adjuvants that can be used in vaccines against challenging pathogens and in specific target populations.


Subject(s)
Adaptive Immunity , Adjuvants, Immunologic/pharmacology , Immunity, Innate , Vaccines, Inactivated/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/administration & dosage , Biomedical Research/trends , Humans , Systems Biology/trends , Vaccines, Inactivated/administration & dosage , Vaccines, Subunit/administration & dosage
8.
J Proteome Res ; 18(3): 1145-1161, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30706717

ABSTRACT

Respiratory syncytial virus (RSV) is a significant cause of mortality and morbidity in infants, the elderly, immunocompromised individuals, and patients with congenital heart diseases. Despite extensive efforts, a vaccine against RSV is still not available. We have previously reported the development of a subunit vaccine (ΔF/TriAdj) composed of a truncated version of the fusion protein (ΔF) and a polymer-based combination adjuvant (TriAdj). We compared inflammatory responses of ΔF/TriAdj-vaccinated and unvaccinated mice following intranasal challenge with RSV. Rapid and early inflammatory responses were observed in lung samples from both groups but modulated in the vaccinated group 7 days after the viral challenge. The underlying mechanism of action of ΔF/TriAdj was further studied through LC-MS-based metabolomic profiling by using 12C- or 13C-dansyl labeling for the amine/phenol submetabolome. RSV infection predominantly affected the amino acid biosynthesis pathways and urea cycle, whereas ΔF/TriAdj modulated the concentrations of almost all of the altered metabolites. Tryptophan metabolites were significantly affected, including indole, l-kynurenine, xanthurenic acid, serotonin, 5-hydroxyindoleacetic acid, and 6-hydroxymelatonin. The results from the present study provide further mechanistic insights into the mode of action of this RSV vaccine candidate and have important implications in the design of metabolic therapeutic interventions.


Subject(s)
Immunization/methods , Metabolomics/methods , Respiratory Syncytial Virus Infections/metabolism , Vaccines, Subunit/metabolism , Adjuvants, Immunologic/metabolism , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Humans , Lung/drug effects , Lung/virology , Mice , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/metabolism , Respiratory Syncytial Virus, Human/pathogenicity , Vaccines, Subunit/genetics
9.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769345

ABSTRACT

VP8, the UL47 gene product in bovine herpesvirus-1 (BoHV-1), is a major tegument protein that is essential for virus replication in vivo The major DNA damage response protein, ataxia telangiectasia mutated (ATM), phosphorylates Nijmegen breakage syndrome (NBS1) and structural maintenance of chromosome-1 (SMC1) proteins during the DNA damage response. VP8 was found to interact with ATM and NBS1 during transfection and BoHV-1 infection. However, VP8 did not interfere with phosphorylation of ATM in transfected or BoHV-1-infected cells. In contrast, VP8 inhibited phosphorylation of both NBS1 and SMC1 in transfected cells, as well as in BoHV-1-infected cells, but not in cells infected with a VP8 deletion mutant (BoHV-1ΔUL47). Inhibition of NBS1 and SMC1 phosphorylation was observed at 4 h postinfection by nuclear VP8. Furthermore, UV light-induced cyclobutane pyrimidine dimer (CPD) repair was reduced in the presence of VP8, and VP8 in fact enhanced etoposide or UV-induced apoptosis. This suggests that VP8 blocks the ATM/NBS1/SMC1 pathway and inhibits DNA repair. VP8 induced apoptosis in VP8-transfected cells through caspase-3 activation. The fact that BoHV-1 is known to induce apoptosis through caspase-3 activation is in agreement with this observation. The role of VP8 was confirmed by the observation that BoHV-1 induced significantly more apoptosis than BoHV-1ΔUL47. These data reveal a potential role of VP8 in the modulation of the DNA damage response pathway and induction of apoptosis during BoHV-1 infection.IMPORTANCE To our knowledge, the effect of BoHV-1 infection on the DNA damage response has not been characterized. Since BoHV-1ΔUL47 was previously shown to be avirulent in vivo, VP8 is critical for the progression of viral infection. We demonstrated that VP8 interacts with DNA damage response proteins and disrupts the ATM-NBS1-SMC1 pathway by inhibiting phosphorylation of DNA repair proteins NBS1 and SMC1. Furthermore, interference of VP8 with DNA repair was correlated with decreased cell viability and increased DNA damage-induced apoptosis. These data show that BoHV-1 VP8 developed a novel strategy to interrupt the ATM signaling pathway and to promote apoptosis. These results further enhance our understanding of the functions of VP8 during BoHV-1 infection and provide an additional explanation for the reduced virulence of BoHV-1ΔUL47.


Subject(s)
Apoptosis , Capsid Proteins/metabolism , DNA Damage , Herpesvirus 1, Bovine/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Capsid Proteins/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cattle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , HEK293 Cells , HeLa Cells , Herpesvirus 1, Bovine/genetics , Humans
10.
Vaccine ; 36(17): 2326-2336, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29559168

ABSTRACT

Respiratory syncytial virus (RSV) causes acute respiratory tract infections in infants, the elderly and immunocompromised individuals. No licensed vaccine is available against RSV. We previously reported that intranasal immunization of rodents and lambs with a RSV vaccine candidate (ΔF/TriAdj) induces protective immunity with a good safety profile. ΔF/TriAdj promoted innate immune responses in respiratory mucosal tissues in vivo, by local chemokine and cytokine production, as well as infiltration and activation of immune cells including macrophages. The macrophage is an important cell type in context of both innate and adaptive immune responses against RSV. Therefore, we characterized the effects of ΔF/TriAdj on a murine macrophage cell line, RAW264.7, and bone marrow-derived macrophages (BMMs). A gene expression study of pattern recognition receptors (PRRs) revealed induction of endosomal and cytosolic receptors in RAW264.7 cells and BMMs by ΔF/TriAdj, but no up-regulation by ΔF in PBS. As a secondary response to the PRR gene expression, induction of several chemokines and pro-inflammatory cytokines, as well as up-regulation of MHC-II and co-stimulatory immune markers, was observed. To further investigate the mechanisms involved in ΔF/TriAdj-mediated secondary responses, we used relevant signal transduction pathway inhibitors. Based on inhibition studies at both transcript and protein levels, JNK, ERK1/2, CaMKII, PI3K and JAK pathways were clearly responsible for ΔF/TriAdj-mediated chemokine and pro-inflammatory cytokine responses, while the p38 and NF-κB pathways appeared to be not or minimally involved. ΔF/TriAdj induced IFN-ß, which may participate in the JAK-STAT pathway to further amplify CXCL-10 production, which was strongly up-regulated. Blocking this pathway by a JAK inhibitor almost completely abrogated CXCL-10 production and caused a significant reduction in the cell surface expression of MHC-II and co-stimulatory immune markers. These data demonstrate that ΔF/TriAdj induces multiple signaling pathways in macrophages.


Subject(s)
Macrophages/immunology , Polymers/chemistry , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Signal Transduction/immunology , Viral Fusion Proteins/immunology , Adjuvants, Immunologic/chemistry , Animals , Biomarkers/metabolism , Cell Line , Chemokines/immunology , Immunity, Innate/immunology , Immunization/methods , Inflammation/immunology , Inflammation/metabolism , Macrophages/virology , Mice , RAW 264.7 Cells , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Vaccines/chemistry , Vaccination/methods , Viral Fusion Proteins/chemistry
11.
Vaccine ; 35(35 Pt A): 4486-4489, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28599794

ABSTRACT

Adjuvants are crucial components of many vaccines. They are used to improve the immunogenicity of vaccines with the aim of conferring long-term protection, to enhance the efficacy of vaccines in newborns, elderly or immunocompromised persons, and to reduce the amount of antigen or the number of doses required to elicit effective immunity. Novel combination adjuvants have been tested in both candidate animals and humans vaccines and have generated encouraging results. Recently, we developed a combination adjuvant platform (TriAdj) comprising of three components, namely a TLR agonist, either polyI:C or CpG oligodeoxynucleotides (ODN), host defense peptide and polyphosphazene. This adjuvant platform is stable and highly effective in a wide range of animal and human vaccines tested in mice, cotton rats, pigs, sheep, and koalas. TriAdj with various vaccines antigens induced effective long-term humoral and cellular immunity. Moreover, the adjuvant platform is suitable for maternal immunization and highly effective in neonates even in the presence of maternal antibodies. This novel vaccine platform, offers excellent opportunity for use in present and future generations of vaccines against multiple infectious agents and targets challenging populations.


Subject(s)
Adjuvants, Immunologic/chemistry , Oligodeoxyribonucleotides/chemistry , Organophosphorus Compounds/chemistry , Polymers/chemistry , Vaccines, Combined/chemistry , Vaccines, Combined/immunology , Animals , Drug Design , Female , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunity, Maternally-Acquired/immunology , Infant , Pregnancy , Vaccines, Combined/chemical synthesis
12.
Vaccine ; 35(27): 3466-3473, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28527686

ABSTRACT

Bovine viral diarrhea virus (BVDV) is one of the most serious pathogens in cattle. Recently, we developed a novel adjuvant platform (TriAdj) that includes a toll-like receptor 3 agonist, poly (I:C); an innate defense regulatory peptide; and water-soluble polymer, poly[di(sodiumcarboxylatoethylphenoxy)]-phosphazene (PCEP). To develop a needle-free intradermal (ID) subunit vaccine, the BVDV type-2 E2 protein was formulated with TriAdj, and immune protection was evaluated in calves against a BVDV-2 strain. Intradermal delivery of E2/TriAdj elicited robust virus neutralizing antibodies and cell-mediated immune responses including CD4+ and CD8+ T-cell responses. The development of CD8+ T-cell responses in vaccinated calves indicates that TriAdj promotes cross-presentation. Upon challenge with virulent BVDV-2, the vaccinated calves showed no weight loss, leukopenia or virus shedding, and almost no temperature increase, in contrast to the control animals, which had severe clinical disease and shed virus for three to six days in nasal fluids and white blood cells. Intradermal vaccination was shown to attract various immune cell populations including dendritic cells, the most important antigen presenting cells. These data demonstrate that ID delivery is suitable as an administration route in cattle and that ID delivered, TriAdj-formulated E2 can protect cattle from BVDV-2.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Diarrhea Virus 1, Bovine Viral/immunology , Immunity, Innate , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Body Weight , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cattle , Fever/prevention & control , Injections, Intradermal , Leukopenia/prevention & control , Viral Vaccines/administration & dosage , Virus Shedding
13.
Antiviral Res ; 135: 108-119, 2016 11.
Article in English | MEDLINE | ID: mdl-27771388

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections.


Subject(s)
Immunity, Innate , Immunologic Factors/administration & dosage , Murine pneumonia virus/immunology , Organophosphorus Compounds/administration & dosage , Pneumovirus Infections/prevention & control , Poly I-C/administration & dosage , Polymers/administration & dosage , Adjuvants, Immunologic , Administration, Intranasal , Animals , Cytokines/immunology , Immunologic Factors/chemistry , Immunologic Factors/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Organophosphorus Compounds/immunology , Pneumovirus Infections/immunology , Poly I-C/immunology , Toll-Like Receptor 3/agonists
14.
Virology ; 499: 288-297, 2016 12.
Article in English | MEDLINE | ID: mdl-27721128

ABSTRACT

Respiratory syncytial virus (RSV) causes serious respiratory illness in infants and elderly. RSV infection induces short-lived immunity, which leaves people prone to re-infection. In contrast, the RSV fusion (F) protein formulated with a novel adjuvant (∆F/TriAdj) elicits long term protective immunity. A comparison of RSV-immunized mice to mice vaccinated with a single dose of ∆F/TriAdj showed no difference in IgG1 and IgG2a production; however, local IgA secreting memory B cell development and B cell IgA production were significantly lower in RSV vaccinated mice than in ∆F/TriAdj-immunized mice. This indicates a potential reason as to why long-term immunity is not induced by RSV infection. The comparison also revealed that germinal center lymphocyte populations were higher in ∆F/TriAdj-vaccinated mice. Furthermore, ∆F/TriAdj induced higher gene expression of activation-induced cytidine deaminase (AID), as well as IL-6, IL-21, TGF-ß cytokines, which are key players in IgA class switch recombination, ultimately leading to a sustained long-term memory response.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin A/immunology , Respiratory Mucosa/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Human/immunology , Vaccines, Subunit/administration & dosage , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , B-Lymphocytes/immunology , Cytokines/immunology , Drug Compounding , Female , Humans , Immunization , Mice, Inbred BALB C , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/genetics , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
15.
Vaccine ; 34(42): 5114-5124, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27591951

ABSTRACT

Respiratory syncytial virus (RSV) causes serious upper and lower respiratory tract infections in newborns and infants. Presently, there is no licensed vaccine against RSV. We previously reported the safety and efficacy of a novel vaccine candidate (ΔF/TriAdj) in rodent and lamb models following intranasal immunization. However, the effects of the vaccine on the innate immune system in the upper and lower respiratory tracts, when delivered intranasally, have not been characterized. In the present study, we found that ΔF/TriAdj triggered transient production of chemokines, cytokines and interferons in the nasal tissues and lungs of BALB/c mice. The types of chemokines produced were consistent with the populations of immune cells recruited, i.e. dendritic cells, macrophages and neutrophils, in the nose-associated lymphoid tissue (NALT), lung and their draining lymph nodes of the ΔF/TriAdj-immunized group. In addition, ΔF/TriAdj stimulated cellular activation with generation of mucosal and systemic antibody responses, and conferred complete protection from viral infection in the lungs upon RSV challenge. The effect of ΔF/TriAdj was short-lived in the nasal tissues and more prolonged in the lungs. In addition, both innate and adaptive immune responses were lower when mice were immunized with ΔF alone. These results suggest that ΔF/TriAdj modulates the innate mucosal environment in both upper and lower respiratory tracts, which contributes to robust adaptive immune responses and long-term protective efficacy of this novel vaccine formulation.


Subject(s)
Adaptive Immunity , Adjuvants, Immunologic , Immunity, Innate , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/chemistry , Viral Fusion Proteins/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Chemokines/biosynthesis , Cytokines/biosynthesis , Interferons/biosynthesis , Mice , Mice, Inbred BALB C , Polymers/chemistry , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Respiratory System/immunology , Respiratory System/virology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/chemistry
16.
Parasite Immunol ; 37(7): 368-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824598

ABSTRACT

Isolation of the soluble exogenous antigens (SEAgs), its immune response study and proteome profiling is an essential prerequisite for understanding the molecular pathogenesis of Leishmania donovani. The immunostimulatory potential of L. donovani SEAgs, purified from culture of L. donovani clinical isolate, was evaluated for their ability to induce cellular responses in treated/cured hamsters. SEAgs induced significant proliferative responses in lymphocytes (SI 5.6 ± 2.3; P < 0.01) isolated from cured hamster. In addition, significant NO production in response to SEAgs was also noticed in macrophages of hamsters, mouse and human cell lines (J774A-1 and THP1). Western blot analyses with antibodies against proteophosphoglycan (PPG; surface-expressed and secreted molecule) of L. donovani revealed that PPG molecules are also present in L. donovani SEAgs. Mass spectrometry (MS)-based proteome analysis of 12 protein bands of SEAgs through MALDI-TOF/TOF endorsed the identification of some Th1-stimulatory immunogenic proteins. These immunogenic proteins may offer increased hope for the discovery of new promising vaccine candidates against visceral leishmaniasis (VL). The overall results suggest that immunostimulatory molecules are present in the SEAgs, which may be further exploited, for developing a subunit vaccine against VL a fatal human disease.


Subject(s)
Antigens, Protozoan/immunology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Macrophages/immunology , Animals , Antibodies/immunology , Antigens, Protozoan/isolation & purification , Cell Line , Cell Proliferation , Cricetinae , Humans , Immunization , Lymphocyte Activation/immunology , Lymphocytes/immunology , Macrophage Activation/immunology , Male , Mice , Proteome/immunology
17.
Vaccine ; 32(50): 6758-64, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25454860

ABSTRACT

Bovine viral diarrhea virus (BVDV) is still one of the most serious pathogens in cattle, meriting the development of improved vaccines. Recently, we developed a new adjuvant consisting of poly[di(sodium carboxylatoethylphenoxy)]-phosphazene (PCEP), either CpG ODN or poly(I:C), and an immune defense regulator (IDR) peptide. As this adjuvant has been shown to mediate the induction of robust, balanced immune responses, it was evaluated in an E2 subunit vaccine against BVDV in lambs and calves. The BVDV type 2 E2 protein was produced at high levels in a mammalian expression system and purified. When formulated with either CpG ODN or poly(I:C), together with IDR and PCEP, the E2 protein elicited high antibody titers and production of IFN-γ secreting cells in lambs. As the immune responses were stronger when poly(I:C) was used, the E2 protein with poly(I:C), IDR and PCEP was subsequently tested in cattle. Robust virus neutralizing antibodies as well as cell-mediated immune responses, including CD8(+) cytotoxic T cell (CTL) responses, were induced. The fact that CTL responses were demonstrated in calves vaccinated with an E2 protein subunit vaccine indicates that this adjuvant formulation promotes cross-presentation. Furthermore, upon challenge with a high dose of virulent BVDV-2, the vaccinated calves showed almost no temperature response, weight loss, leukopenia or virus replication, in contrast to the control animals, which had severe clinical disease. These data suggest that this E2 subunit formulation induces significant protection from BVDV-2 challenge, and thus is a promising BVDV vaccine candidate; in addition, the adjuvant platform has applications in bovine vaccines in general.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cattle Diseases/prevention & control , Diarrhea Virus 2, Bovine Viral/immunology , Pestivirus Infections/veterinary , Sheep Diseases/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cattle , Cattle Diseases/pathology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Pestivirus Infections/pathology , Pestivirus Infections/prevention & control , Sheep , Sheep Diseases/pathology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Vaccines/administration & dosage
18.
Expert Rev Vaccines ; 11(12): 1441-57, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23252388

ABSTRACT

Respiratory syncytial virus (RSV) is a common human pathogen that causes cold-like symptoms in most healthy adults and children. However, RSV often moves into the lower respiratory tract in infants and young children predisposed to respiratory illness, making it the most common cause of pediatric broncheolitis and pneumonia. The development of an appropriate balanced immune response is critical for recovery from RSV, while an unbalanced and/or excessively vigorous response may lead to immunopathogenesis. Different dendritic cell (DC) subsets influence the magnitude and quality of the host response to RSV infection, with myeloid DCs mediating and plasmacytoid DCs modulating immunopathology. Furthermore, stimulation of DCs through Toll-like receptors is essential for induction of protective immunity to RSV. These characteristics have implications for the rational design of a RSV vaccine.


Subject(s)
Adaptive Immunity , Dendritic Cells/immunology , Immunity, Innate , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Clinical Trials as Topic , Humans , Lung/immunology , Lung/pathology , Respiratory Syncytial Virus Infections/immunology , Vaccination , Vaccines, Attenuated/immunology
19.
Virology ; 432(2): 271-6, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22704066

ABSTRACT

Miltefosine (Milt) was originally synthesized as an antineoplastic agent but this phospholipid drug is now clinically used as an antiprotozoal compound. We demonstrate here that Milt reduces replication of HIV-1 in cocultures of human dendritic cells (DCs) and CD4(+) T cells. This phenomenon is due to a rapid secretion of soluble factors by DCs. We present evidence that the Milt-mediated repression in virus production is associated with induction of type-I interferon (IFN) in DCs. The Milt-dependent diminution in HIV-1 production was not totally abrogated by B18R, a vaccinia virus-encoded neutralizing type-I IFN receptor, which suggests the involvement of another yet to be identified soluble factor. Altogether, these results suggest that a therapy with Milt when used to control protozoan infections in individuals also carrying HIV-1 might also help to limit viral load. Additional studies are warranted to estimate the exact therapeutic potential of Milt as an anti-HIV-1 agent.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , HIV-1/drug effects , Interferon Type I/metabolism , Phosphorylcholine/analogs & derivatives , Virus Replication/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Line , Coculture Techniques , HIV-1/physiology , Humans , Phosphorylcholine/pharmacology
20.
PLoS Negl Trop Dis ; 3(5): e441, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19468304

ABSTRACT

BACKGROUND: Visceral leishmaniasis has emerged as an important opportunistic disease among patients infected with HIV-1. Both HIV-1 and the protozoan parasite Leishmania can productively infect cells of the macrophage-dendritic cell lineage. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that Leishmania infantum amastigotes increase HIV-1 production when human primary dendritic cells (DCs) are cocultured together with autologous CD4(+) T cells. Interestingly, the promastigote form of the parasite does not modulate virus replication. Moreover, we report that amastigotes promote virus replication in both cell types. Our results indicate that this process is due to secretion of parasite-induced soluble factors by DCs. Luminex micro-beads array system analyses indicate that Leishmania infantum amastigotes induce a higher secretion of several cytokines (i.e. IL-1alpha, IL-2, IL-6, IL-10 and TNF-alpha) and chemokines (i.e. MIP-1alpha, MIP-1beta and RANTES) in these cells. Studies conducted with pentoxifylline and neutralizing antibodies revealed that the Leishmania-dependent augmentation in HIV-1 replication is due to a higher secretion of IL-6 and TNF-alpha. CONCLUSIONS/SIGNIFICANCE: Altogether these findings suggest that the presence of Leishmania within DC/T-cell conjugates leads to an enhancement of virus production and demonstrate that HIV-1 and Leishmania can establish complex interactions in such a cellular microenvironment.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Dendritic Cells/parasitology , HIV-1/growth & development , Interleukin-6/metabolism , Leishmania infantum/growth & development , Leishmania infantum/immunology , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Dendritic Cells/virology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...