Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Immunol Immunother ; 68(7): 1211-1222, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31069460

ABSTRACT

Human tumor cells express antigens that serve as targets for the host cellular immune system. This phase 1 dose-escalating study was conducted to assess safety and tolerability of G305, a recombinant NY-ESO-1 protein vaccine mixed with glucopyranosyl lipid A (GLA), a synthetic TLR4 agonist adjuvant, in a stable emulsion (SE). Twelve patients with solid tumors expressing NY-ESO-1 were treated using a 3 + 3 design. The NY-ESO-1 dose was fixed at 250 µg, while GLA-SE was increased from 2 to 10 µg. Safety, immunogenicity, and clinical responses were assessed prior to, during, and at the end of therapy. G305 was safe and immunogenic at all doses. All related AEs were Grade 1 or 2, with injection site soreness as the most commonly reported event (100%). Overall, 75% of patients developed antibody response to NY-ESO-1, including six patients with increased antibody titer ( ≥ 4-fold rise) and three patients with seroconversion from negative (titer < 100) to positive (titer ≥ 100). CD4 T-cell responses were observed in 44.4% of patients; 33.3% were new responses and 1 was boosted ( ≥ 2-fold rise). Following treatment, 8 of 12 patients had stable disease for 3 months or more; at the end of 1 year, three patients had stable disease and nine patients were alive. G305 is a potent immunotherapeutic agent that can stimulate NY-ESO-1-specific antibody and T-cell responses. The vaccine was safe at all doses of GLA-SE (2-10 µg) and showed potential clinical benefit in this population of patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Glucosides/administration & dosage , Lipid A/administration & dosage , Membrane Proteins/administration & dosage , Neoplasms/therapy , Adjuvants, Immunologic/adverse effects , Adult , Aged , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Female , Glucosides/adverse effects , Glucosides/immunology , Humans , Immunogenicity, Vaccine , Injections, Intramuscular , Lipid A/adverse effects , Lipid A/immunology , Male , Membrane Proteins/adverse effects , Membrane Proteins/immunology , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Young Adult
2.
Mol Ther ; 25(2): 494-503, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153096

ABSTRACT

To be effective against HIV type 1 (HIV-1), vaccine-induced T cells must selectively target epitopes, which are functionally conserved (present in the majority of currently circulating and reactivated HIV-1 strains) and, at the same time, beneficial (responses to which are associated with better clinical status and control of HIV-1 replication), and rapidly reach protective frequencies upon exposure to the virus. Heterologous prime-boost regimens using virally vectored vaccines are currently the most promising vaccine strategies; nevertheless, induction of robust long-term memory remains challenging. To this end, lentiviral vectors induce high frequencies of memory cells due to their low-inflammatory nature, while typically inducing only low anti-vector immune responses. Here, we describe construction of novel candidate vaccines ZVex.tHIVconsv1 and ZVex.tHIVconsv2, which are based on an integration-deficient lentiviral vector platform with preferential transduction of human dendritic cells and express a bivalent mosaic of conserved-region T cell immunogens with a high global HIV-1 match. Each of the two mosaic vaccines was individually immunogenic. When administered together in heterologous prime-boost regimens with chimpanzee adenovirus and/or poxvirus modified vaccinia virus Ankara (MVA) vaccines to BALB/c and outbred CD1-Swiss mice, they induced a median frequency of over 6,000 T cells/106 splenocytes, which were plurifunctional, broadly specific, and cross-reactive. These results support further development of this vaccine concept.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Vectors/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Lentivirus/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Conserved Sequence , Disease Models, Animal , Epitopes/genetics , Epitopes/immunology , Female , Gene Order , HIV Infections/virology , Humans , Immunity, Cellular , Mice , Peptides/genetics , Peptides/immunology
3.
PLoS Pathog ; 11(11): e1005276, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26587982

ABSTRACT

Arenaviruses such as Lassa virus (LASV) can cause severe hemorrhagic fever in humans. As a major impediment to vaccine development, delayed and weak neutralizing antibody (nAb) responses represent a unifying characteristic of both natural infection and all vaccine candidates tested to date. To investigate the mechanisms underlying arenavirus nAb evasion we engineered several arenavirus envelope-chimeric viruses and glycan-deficient variants thereof. We performed neutralization tests with sera from experimentally infected mice and from LASV-convalescent human patients. NAb response kinetics in mice correlated inversely with the N-linked glycan density in the arenavirus envelope protein's globular head. Additionally and most intriguingly, infection with fully glycosylated viruses elicited antibodies, which neutralized predominantly their glycan-deficient variants, both in mice and humans. Binding studies with monoclonal antibodies indicated that envelope glycans reduced nAb on-rate, occupancy and thereby counteracted virus neutralization. In infected mice, the envelope glycan shield promoted protracted viral infection by preventing its timely elimination by the ensuing antibody response. Thus, arenavirus envelope glycosylation impairs the protective efficacy rather than the induction of nAbs, and thereby prevents efficient antibody-mediated virus control. This immune evasion mechanism imposes limitations on antibody-based vaccination and convalescent serum therapy.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Arenavirus/immunology , Hemorrhagic Fevers, Viral/immunology , Polysaccharides/immunology , Animals , HIV Antibodies/immunology , HIV-1/immunology , Humans , Mice, Inbred C57BL , Molecular Sequence Data
4.
J Infect Dis ; 214(suppl 3): S342-S354, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27456709

ABSTRACT

The 2013-2016 West African Ebola virus (EBOV) disease outbreak was the largest filovirus outbreak to date. Over 28 000 suspected, probable, or confirmed cases have been reported, with a 53% case-fatality rate. The magnitude and international impact of this EBOV outbreak has highlighted the urgent need for a safe and efficient EBOV vaccine. To this end, we demonstrate the immunogenicity and protective efficacy of FILORAB1, a recombinant, bivalent, inactivated rabies virus-based EBOV vaccine, in rhesus and cynomolgus monkeys. Our results demonstrate that the use of the synthetic Toll-like receptor 4 agonist glucopyranosyl lipid A in stable emulsion (GLA-SE) as an adjuvant increased the efficacy of FILORAB1 to 100% protection against lethal EBOV challenge, with no to mild clinical signs of disease. Furthermore, all vaccinated subjects developed protective anti-rabies virus antibody titers. Taken together, these results support further development of FILORAB1/GLA-SE as an effective preexposure EBOV vaccine.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Glucosides/immunology , Hemorrhagic Fever, Ebola/prevention & control , Lipid A/immunology , Rabies virus/immunology , Rabies/prevention & control , Adjuvants, Immunologic , Animals , Antibodies, Viral/immunology , Emulsions , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Macaca fascicularis , Macaca mulatta , Male , Rabies/immunology , Rabies/virology , Rabies Vaccines/immunology , Toll-Like Receptor 4/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology
5.
J Virol ; 89(3): 1781-93, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410871

ABSTRACT

UNLABELLED: Varicella-zoster virus (VZV) is a human neurotropic alphaherpesvirus and the etiological agent of varicella (chickenpox) and herpes zoster (HZ, shingles). Previously, inoculation of monkeys via the subcutaneous, intratracheal, intravenous, or oral-nasal-conjunctival routes did not recapitulate all the hallmarks of VZV infection, including varicella, immunity, latency, and reactivation. Intrabronchial inoculation of rhesus macaques (RMs) with simian varicella virus (SVV), a homolog of VZV, recapitulates virologic and immunologic hallmarks of VZV infection in humans. Given that VZV is acquired primarily via the respiratory route, we investigated whether intrabronchial inoculation of RMs with VZV would result in a robust model. Despite the lack of varicella and viral replication in either the lungs or whole blood, all four RMs generated an immune response characterized by the generation of VZV-specific antibodies and T cells. Two of 4 VZV-inoculated RMs were challenged with SVV to determine cross-protection. VZV-immune RMs displayed no varicella rash and had lower SVV viral loads and earlier and stronger humoral and cellular immune responses than controls. In contrast to the results for SVV DNA, no VZV DNA was detected in sensory ganglia at necropsy. In summary, following an abortive VZV infection, RMs developed an adaptive immune response that conferred partial protection against SVV challenge. These data suggest that a replication-incompetent VZV vaccine that does not establish latency may provide sufficient protection against VZV disease and that VZV vaccination of RMs followed by SVV challenge provides a model to evaluate new vaccines and therapeutics against VZV. IMPORTANCE: Although VZV vaccine strain Oka is attenuated, it can cause mild varicella, establish latency, and in rare cases, reactivate to cause herpes zoster (HZ). Moreover, studies suggest that the HZ vaccine (Zostavax) only confers short-lived immunity. The development of more efficacious vaccines would be facilitated by a robust animal model of VZV infection. The data presented in this report show that intrabronchial inoculation of rhesus macaques (RMs) with VZV resulted in an abortive VZV infection. Nevertheless, all animals generated a humoral and cellular immune response that conferred partial cross-protection against simian varicella virus (SVV) challenge. Additionally, VZV DNA was not detected in the sensory ganglia, suggesting that viremia might be required for the establishment of latency. Therefore, VZV vaccination of RMs followed by SVV challenge is a model that will support the development of vaccines that boost protective T cell responses against VZV.


Subject(s)
Chickenpox/veterinary , Cross Protection , Herpesvirus 3, Human/immunology , Primate Diseases/prevention & control , Animals , Antibodies, Viral/blood , Chickenpox/immunology , Chickenpox/pathology , Chickenpox/prevention & control , DNA, Viral/genetics , DNA, Viral/isolation & purification , Ganglia/virology , Macaca mulatta , Male , Primate Diseases/immunology , T-Lymphocytes/immunology
7.
Opt Express ; 22 Suppl 3: A589-600, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24922367

ABSTRACT

We report on the fabrication and simulation of a green OLED with an Internal Light Extraction (ILE) layer. The optical behavior of these devices is simulated using both Rigorous Coupled Wave Analysis (RCWA) and Finite Difference Time-Domain (FDTD) methods. Results obtained using these two different techniques show excellent agreement and predict the experimental results with good precision. By verifying the validity of both simulation methods on the internal light extraction structure we pave the way to optimization of ILE layers using either of these methods.

8.
J Clin Microbiol ; 51(9): 2970-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824772

ABSTRACT

Pathogenic bacteria produce several virulence factors that help them establish infection in permissive hosts. Bacterial toxins are a major class of virulence factors and hence are attractive therapeutic targets for vaccine development. Here, we describe the development of a rapid, sensitive, and high-throughput assay that can be used as a versatile platform to measure the activities of bacterial toxins. We have exploited the ability of these toxins to cause cell death via apoptosis of sensitive cultured cell lines as a readout for measuring toxin activity. Caspases (cysteine-aspartic proteases) are induced early in the apoptotic pathway, and so we used their induction to measure the activities of Clostridium difficile toxins A (TcdA) and B (TcdB) and binary toxin (CDTa-CDTb), Corynebacterium diphtheriae toxin (DT), and Pseudomonas aeruginosa exotoxin A (PEA). Caspase induction in the cell lines, upon exposure to toxins, was optimized by toxin concentration and intoxication time, and the specificity of caspase activity was established using a genetically mutated toxin and a pan-caspase inhibitor. In addition, we demonstrate the utility of the caspase assay for measuring toxin potency, as well as neutralizing antibody (NAb) activity against C. difficile toxins. Furthermore, the caspase assay showed excellent correlation with the filamentous actin (F-actin) polymerization assay for measuring TcdA and TcdB neutralization titers upon vaccination of hamsters. These results demonstrate that the detection of caspase induction due to toxin exposure using a chemiluminescence readout can support potency and clinical immunogenicity testing for bacterial toxin vaccine candidates in development.


Subject(s)
Bacterial Toxins/analysis , Caspases/metabolism , Cytological Techniques/methods , Animals , Apoptosis , Bacterial Toxins/toxicity , Chlorocebus aethiops , HeLa Cells , High-Throughput Screening Assays/methods , Humans , Luminescent Measurements/methods , Vero Cells
9.
J Virol ; 86(24): 13434-44, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015722

ABSTRACT

The hemagglutinin protein (HA) on the surface of influenza virus is essential for viral entry into the host cells. The HA1 subunit of HA is also the primary target for neutralizing antibodies. The HA2 subunit is less exposed on the virion surface and more conserved than HA1. We have previously designed an HA2-based immunogen derived from the sequence of the H3N2 A/HK/68 virus. In the present study, we report the design of an HA2-based immunogen from the H1N1 subtype (PR/8/34). This immunogen (H1HA0HA6) and its circular permutant (H1HA6) were well folded and provided complete protection against homologous viral challenge. Antisera of immunized mice showed cross-reactivity with HA proteins of different strains and subtypes. Although no neutralization was observable in a conventional neutralization assay, sera of immunized guinea pigs competed with a broadly neutralizing antibody, CR6261, for binding to recombinant Viet/04 HA protein, suggesting that CR6261-like antibodies were elicited by the immunogens. Stem domain immunogens from a seasonal H1N1 strain (A/NC/20/99) and a recent pandemic strain (A/Cal/07/09) provided cross-protection against A/PR/8/34 viral challenge. HA2-containing stem domain immunogens therefore have the potential to provide subtype-specific protection.


Subject(s)
Escherichia coli/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Amino Acid Sequence , Animals , Circular Dichroism , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Female , Guinea Pigs , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Immune Sera , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Spectrometry, Fluorescence , Surface Plasmon Resonance
10.
J Virol ; 86(7): 3819-27, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278233

ABSTRACT

We have discovered the first indigenous African hantavirus, Sangassou virus (SANGV). The virus was isolated from an African wood mouse (Hylomyscus simus), trapped in a forest habitat in Guinea, West Africa. Here, we report on the characterization of the genetic and functional properties of the virus. The complete genome of SANGV was determined and showed typical hantavirus organization. The small (S), medium (M), and large (L) genome segments containing genes encoding nucleocapsid protein, two envelope glycoproteins, and viral polymerase were found to be 1,746, 3,650, and 6,531 nucleotides long, respectively. The exact 5' and 3' termini for all three segments of the SANGV genome were determined and were predicted to form the panhandle structures typical of bunyaviruses. Phylogenetic analyses of all three segment sequences confirmed SANGV as a Murinae-associated hantavirus most closely related to the European Dobrava-Belgrade virus. We showed, however, that SANGV uses ß(1) integrin rather than ß(3) integrin and decay-accelerating factor (DAF)/CD55 as an entry receptor. In addition, we demonstrated a strong induction of type III lambda interferon (IFN-λ) expression in type I IFN-deficient Vero E6 cells by SANGV. These properties are unique within Murinae-associated hantaviruses and make the virus useful in comparative studies focusing on hantavirus pathogenesis.


Subject(s)
Genetic Variation , Hantavirus Infections/veterinary , Murinae/virology , Orthohantavirus/genetics , Orthohantavirus/isolation & purification , Rodent Diseases/virology , Africa , Animals , Base Sequence , Cell Line , Genome, Viral , Orthohantavirus/classification , Hantavirus Infections/virology , Humans , Mice , Molecular Sequence Data , Phylogeny
11.
Proc Natl Acad Sci U S A ; 107(31): 13701-6, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20615991

ABSTRACT

Influenza HA is the primary target of neutralizing antibodies during infection, and its sequence undergoes genetic drift and shift in response to immune pressure. The receptor binding HA1 subunit of HA shows much higher sequence variability relative to the metastable, fusion-active HA2 subunit, presumably because neutralizing antibodies are primarily targeted against the former in natural infection. We have designed an HA2-based immunogen using a protein minimization approach that incorporates designed mutations to destabilize the low pH conformation of HA2. The resulting construct (HA6) was expressed in Escherichia coli and refolded from inclusion bodies. Biophysical studies and mutational analysis of the protein indicate that it is folded into the desired neutral pH conformation competent to bind the broadly neutralizing HA2 directed monoclonal 12D1, not the low pH conformation observed in previous studies. HA6 was highly immunogenic in mice and the mice were protected against lethal challenge by the homologous A/HK/68 mouse-adapted virus. An HA6-like construct from another H3 strain (A/Phil/2/82) also protected mice against A/HK/68 challenge. Regions included in HA6 are highly conserved within a subtype and are fairly well conserved within a clade. Targeting the highly conserved HA2 subunit with a bacterially produced immunogen is a vaccine strategy that may aid in pandemic preparedness.


Subject(s)
Escherichia coli/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Binding Sites , Circular Dichroism , Escherichia coli/genetics , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Models, Molecular , Mutation , Protein Structure, Quaternary , Protein Structure, Tertiary
12.
Sci Rep ; 13(1): 4648, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36944687

ABSTRACT

SARS-CoV-2 continues to circulate in the human population necessitating regular booster immunization for its long-term control. Ideally, vaccines should ideally not only protect against symptomatic disease, but also prevent transmission via asymptomatic shedding and cover existing and future variants of the virus. This may ultimately only be possible through induction of potent and long-lasting immune responses in the nasopharyngeal tract, the initial entry site of SARS-CoV-2. To this end, we have designed a vaccine based on recombinantly expressed receptor binding domain (RBD) of SARS-CoV-2, fused to the C-terminus of C. perfringens enterotoxin, which is known to target Claudin-4, a matrix molecule highly expressed on mucosal microfold (M) cells of the nasal and bronchial-associated lymphoid tissues. To further enhance immune responses, the vaccine was adjuvanted with a novel toll-like receptor 3/RIG-I agonist (Riboxxim™), consisting of synthetic short double stranded RNA. Intranasal prime-boost immunization of mice induced robust mucosal and systemic anti-SARS-CoV-2 neutralizing antibody responses against SARS-CoV-2 strains Wuhan-Hu-1, and several variants (B.1.351/beta, B.1.1.7/alpha, B.1.617.2/delta), as well as systemic T-cell responses. A combination vaccine with M-cell targeted recombinant HA1 from an H1N1 G4 influenza strain also induced mucosal and systemic antibodies against influenza. Taken together, the data show that development of an intranasal SARS-CoV-2 vaccine based on recombinant RBD adjuvanted with a TLR3 agonist is feasible, also as a combination vaccine against influenza.


Subject(s)
COVID-19 Vaccines , COVID-19 , Influenza, Human , Animals , Humans , Mice , Adjuvants, Immunologic , Adjuvants, Pharmaceutic , Antibodies, Neutralizing , Antibodies, Viral , Clostridium perfringens , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Gastric Mucosa , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , M Cells , SARS-CoV-2 , Toll-Like Receptor 3
13.
JAMA Oncol ; 9(12): 1660-1668, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37824131

ABSTRACT

Importance: Metastatic soft tissue sarcomas (STSs) have limited systemic therapy options, and immunomodulation has not yet meaningfully improved outcomes. Intratumoral (IT) injection of the toll-like receptor 4 (TLR4) agonist glycopyranosyl lipid A in stable-emulsion formulation (GLA-SE) has been studied as immunotherapy in other contexts. Objective: To evaluate the safety, efficacy, and immunomodulatory effects of IT GLA-SE with concurrent radiotherapy in patients with metastatic STS with injectable lesions. Design, Setting, and Participants: This phase 1 nonrandomized controlled trial of patients with STS was performed at a single academic sarcoma specialty center from November 17, 2014, to March 16, 2016. Data analysis was performed from August 2016 to September 2022. Interventions: Two doses of IT GLA-SE (5 µg and 10 µg for 8 weekly doses) were tested for safety in combination with concurrent radiotherapy of the injected lesion. Main Outcomes and Measures: Primary end points were safety and tolerability. Secondary and exploratory end points included local response rates as well as measurement of antitumor immunity with immunohistochemistry and T-cell receptor (TCR) sequencing of tumor-infiltrating and circulating lymphocytes. Results: Twelve patients (median [range] age, 65 [34-78] years; 8 [67%] female) were treated across the 2 dose cohorts. Intratumoral GLA-SE was well tolerated, with only 1 patient (8%) experiencing a grade 2 adverse event. All patients achieved local control of the injected lesion after 8 doses, with 1 patient having complete regression (mean regression, -25%; range, -100% to 4%). In patients with durable local response, there were detectable increases in tumor-infiltrating lymphocytes. In 1 patient (target lesion -39% at 259 days of follow-up), TCR sequencing revealed expansion of preexisting and de novo clonotypes, with convergence of numerous rearrangements coding for the same binding sequence (suggestive of clonal convergence to antitumor targets). Single-cell sequencing identified these same expanded TCR clones in peripheral blood after treatment; these T cells had markedly enhanced Tbet expression, suggesting TH1 phenotype. Conclusions and Relevance: In this nonrandomized controlled trial, IT GLA-SE with concurrent radiotherapy was well tolerated and provided more durable local control than radiotherapy alone. Patients with durable local response demonstrated enhanced IT T-cell clonal expansion, with matched expansion of these clonotypes in the circulation. Additional studies evaluating synergism of IT GLA-SE and radiotherapy with systemic immune modulation are warranted. Trial Registration: ClinicalTrials.gov Identifier: NCT02180698.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Humans , Female , Aged , Male , Toll-Like Receptor 4/agonists , T-Lymphocytes , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/radiotherapy , Sarcoma/drug therapy , Sarcoma/radiotherapy , Receptors, Antigen, T-Cell
14.
J Infect Dis ; 204 Suppl 3: S785-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987751

ABSTRACT

A needlestick injury occurred during an animal experiment in the biosafety level 4 laboratory in Hamburg, Germany, in March 2009. The syringe contained Zaire ebolavirus (ZEBOV) mixed with Freund's adjuvant. Neither an approved treatment nor a postexposure prophylaxis (PEP) exists for Ebola hemorrhagic fever. Following a risk-benefit assessment, it was recommended the exposed person take an experimental vaccine that had shown PEP efficacy in ZEBOV-infected nonhuman primates (NHPs) [12]. The vaccine, which had not been used previously in humans, was a live-attenuated recombinant vesicular stomatitis virus (recVSV) expressing the glycoprotein of ZEBOV. A single dose of 5 × 10(7) plaque-forming units was injected 48 hours after the accident. The vaccinee developed fever 12 hours later and recVSV viremia was detectable by polymerase chain reaction (PCR) for 2 days. Otherwise, the person remained healthy, and ZEBOV RNA, except for the glycoprotein gene expressed in the vaccine, was never detected in serum and peripheral blood mononuclear cells during the 3-week observation period.


Subject(s)
Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola/prevention & control , Laboratory Infection/prevention & control , Needlestick Injuries , Post-Exposure Prophylaxis/methods , Animals , Containment of Biohazards , Ebola Vaccines/administration & dosage , Ebola Vaccines/standards , Germany , Humans , Mice , Needlestick Injuries/virology , Occupational Exposure , RNA, Viral/blood , Research Personnel , Vaccines, Attenuated , Vaccines, DNA/immunology , Vesiculovirus/genetics , Viremia
15.
J Clin Microbiol ; 49(3): 1157-61, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21191050

ABSTRACT

Recent Lassa virus strains from Nigeria were completely or partially sequenced. Phylogenetic analysis revealed the predominance of lineage II and III strains, the existence of a previously undescribed (sub)lineage in Nigeria, and the directional spread of virus in the southern part of the country. The Bayesian analysis also provided estimates for divergence times within the Lassa virus clade.


Subject(s)
Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/classification , Lassa virus/isolation & purification , Cluster Analysis , Humans , Lassa virus/genetics , Molecular Epidemiology , Molecular Sequence Data , Nigeria/epidemiology , Phylogeny , Polymorphism, Genetic , RNA, Viral/genetics , Sequence Analysis, DNA
16.
J Infect Dis ; 201(7): 1031-4, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20187741

ABSTRACT

We recently discovered a novel hantavirus, Sangassou virus, in Guinea, West Africa. Using enzyme-linked immunosorbent assays followed by confirmatory and serotyping assays, we retrospectively detected hantavirus antibodies in 3 (4.4%) of 68 patients with fever of unknown origin in Sangassou village, Forest Guinea. A population-based survey in Forest Guinea (n = 649) found the prevalence of hantavirus antibodies to be 1.2%. Specific neutralizing antibodies against Sangassou virus were demonstrated in serum samples from 2 patients and in 2 serum samples of the serosurvey. Our data allow us to conclude that hantavirus infections may be a significant unrecognized medical problem in at least this part of Africa.


Subject(s)
Antibodies, Viral/blood , Hantavirus Infections/blood , Orthohantavirus/isolation & purification , Antibodies, Neutralizing/blood , Child , Enzyme-Linked Immunosorbent Assay , Female , Guinea/epidemiology , Orthohantavirus/immunology , Hantavirus Infections/epidemiology , Hantavirus Infections/immunology , Hantavirus Infections/transmission , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Retrospective Studies , Seroepidemiologic Studies
17.
PLoS One ; 16(12): e0259301, 2021.
Article in English | MEDLINE | ID: mdl-34855754

ABSTRACT

Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.


Subject(s)
Glucosides/pharmacology , Immunologic Memory/drug effects , Interleukin-12/genetics , Lipid A/pharmacology , Neoplasms, Experimental/immunology , Toll-Like Receptor 4/agonists , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunologic Memory/genetics , Immunotherapy/methods , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-12/immunology , Lentivirus/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
18.
Nanomaterials (Basel) ; 11(2)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572813

ABSTRACT

Bio-inspired surfaces with superamphiphobic properties are well known as effective candidates for antifouling technology. However, the limitation of large-area mastering, patterning and pattern collapsing upon physical contact are the bottleneck for practical utilization in marine and medical applications. In this study, a roll-to-plate nanoimprint lithography (R2P NIL) process using Morphotonics' automated Portis NIL600 tool was used to replicate high aspect ratio (5.0) micro-structures via reusable intermediate flexible stamps that were fabricated from silicon master molds. Two types of Morphotonics' in-house UV-curable resins were used to replicate a micro-pillar (PIL) and circular rings with eight stripe supporters (C-RESS) micro-structure onto polycarbonate (PC) and polyethylene terephthalate (PET) foil substrates. The pattern quality and surface wettability was compared to a conventional polydimethylsiloxane (PDMS) soft lithography process. It was found that the heights of the R2P NIL replicated PIL and C-RESS patterns deviated less than 6% and 5% from the pattern design, respectively. Moreover, the surface wettability of the imprinted PIL and C-RESS patterns was found to be superhydro- and oleophobic and hydro- and oleophobic, respectively, with good robustness for the C-RESS micro-structure. Therefore, the R2P NIL process is expected to be a promising method to fabricate robust C-RESS micro-structures for large-scale anti-biofouling application.

19.
J Virol ; 83(7): 3228-37, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19153226

ABSTRACT

The cell entry and humoral immune response of the human pathogen Lassa virus (LV), a biosafety level 4 (BSL4) Old World arenavirus, are not well characterized. LV pseudoparticles (LVpp) are a surrogate model system that has been used to decipher factors and routes involved in LV cell entry under BSL2 conditions. Here, we describe LVpp, which are highly infectious, with titers approaching those obtained with pseudoparticles displaying G protein of vesicular stomatitis virus and their the use for the characterization of LV cell entry and neutralization. Upon cell attachment, LVpp utilize endocytic vesicles for cell entry as described for many pH-dependent viruses. However, the fusion of the LV glycoproteins is activated at unusually low pH values, with optimal fusion occurring between pH 4.5 and 3, a pH range at which fusion characteristics of viral glycoproteins have so far remained largely unexplored. Consistent with a shifted pH optimum for fusion activation, we found wild-type LV and LVpp to display a remarkable resistance to exposure to low pH. Finally, LVpp allow the fast and quantifiable detection of neutralizing antibodies in human and animal sera and will thus facilitate the study of the humoral immune response in LV infections.


Subject(s)
Lassa virus/immunology , Lassa virus/physiology , Virosomes/immunology , Virus Internalization , Animals , Antibodies, Viral/blood , Cell Line , Endocytosis , Endosomes/virology , Humans , Hydrogen-Ion Concentration , Neutralization Tests
20.
Front Oncol ; 10: 1438, 2020.
Article in English | MEDLINE | ID: mdl-32974162

ABSTRACT

Intratumoral (IT) injections of Glucopyranosyl lipid A (G100), a synthetic toll-like receptor 4 (TLR4) agonist formulated in a stable emulsion, resulted in T-cell inflammation of the tumor microenvironment (TME) and complete cure of 60% of mice with large established A20 lymphomas. Strong abscopal effects on un-injected lesions were observed in a bilateral tumor model and surviving mice resisted a secondary tumor challenge. Depletion of CD8 T-cells, but not CD4 or NK cells, abrogated the anti-tumor effect. Unexpectedly, TLR4 knock-out rendered A20 tumors completely non-responsive to G100. In vitro studies showed that GLA has direct effect on A20 cells, but not on A20 cells deficient for TLR4. As shown by genotyping and phenotyping analysis, G100 strongly activated antigen presentation functions in A20 cells in vitro and in vivo and induced their apoptosis in a dose dependent manner. Similarly, the TLR4 positive human mantle cell lymphoma line Mino showed in vitro activation with G100 that was blocked with an anti-TLR4 antibody. In the A20 model, direct activation of B-lymphoma cells with G100 is sufficient to induce protective CD8 T-cell responses and TLR4 expressing human B-cell lymphomas may be amenable to this therapy as well.

SELECTION OF CITATIONS
SEARCH DETAIL