Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Virol ; 97(10): e0067423, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37830821

ABSTRACT

IMPORTANCE: Vaccines targeting highly conserved proteins can protect broadly against diverse viral strains. When a vaccine is administered to the respiratory tract, protection against disease is especially powerful. However, it is important to establish that this approach is safe. When vaccinated animals later encounter viruses, does reactivation of powerful local immunity, including T cell responses, damage the lungs? This study investigates the safety of mucosal vaccination of the respiratory tract. Non-replicating adenoviral vaccine vectors expressing conserved influenza virus proteins were given intranasally. This vaccine-induced protection persists for at least 15 months. Vaccination did not exacerbate inflammatory responses or tissue damage upon influenza virus infection. Instead, vaccination with nucleoprotein reduced cytokine responses and histopathology, while neutrophil and T cell responses resolved earlier. The results are promising for safe vaccination at the site of infection and thus have implications for the control of influenza and other respiratory viruses.


Subject(s)
Influenza Vaccines , Orthomyxoviridae Infections , Animals , Mice , Antibodies, Viral , Influenza Vaccines/immunology , Lung , Mice, Inbred BALB C , Orthomyxoviridae , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Adenoviridae
2.
J Virol ; 96(12): e0032022, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35638848

ABSTRACT

Vaccination against influenza virus infection can protect the vaccinee and also reduce transmission to contacts. Not all types of vaccines induce sterilizing immunity via neutralizing antibodies; some instead permit low-level, transient infection. There has been concern that infection-permissive influenza vaccines may allow continued spread in the community despite minimizing symptoms in the vaccinee. We have explored that issue for a universal influenza vaccine candidate that protects recipients by inducing T cell responses and nonneutralizing antibodies. Using a mouse model, we have shown previously that an adenoviral vectored vaccine expressing nucleoprotein (NP) and matrix 2 (M2) provides broad protection against diverse strains and subtypes of influenza A viruses and reduces transmission to contacts in an antigen-specific manner. Here, we use this mouse model to further explore the mechanism and features of that reduction in transmission. Passive immunization did not reduce transmission from infected donors to naive contact animals to whom passive serum had been transferred. Vaccination of antibody-deficient mIgTg-JHD-/- mice, which have intact T cell responses and antigen presentation, reduced transmission in an antigen-specific manner, despite the presence of some virus in the lungs and nasal wash, pointing to a role for cellular immunity. Vaccination at ages ranging from 8 to 60 weeks was able to achieve reduction in transmission. Finally, the immune-mediated reduction in transmission persisted for at least a year after a single-dose intranasal vaccination. Thus, this infection-permissive vaccine reduces virus transmission in a long-lasting manner that does not require antibodies. IMPORTANCE Universal influenza virus vaccines targeting antigens conserved among influenza A virus strains can protect from severe disease but do not necessarily prevent infection. Despite allowing low-level infection, intranasal immunization with adenovirus vectors expressing the conserved antigens influenza nucleoprotein (A/NP) and M2 reduces influenza virus transmission from vaccinated to unvaccinated contact mice. Here, we show that antibodies are not required for this transmission reduction, suggesting a role for T cells. We also show that transmission blocking could be achieved in recipients of different ages and remained effective for at least a year following a single-dose vaccination. Such vaccines could have major public health impacts by limiting viral transmission in the community.


Subject(s)
Influenza A virus , Influenza Vaccines , Orthomyxoviridae Infections , Adenoviridae , Animals , Antibodies, Viral , Humans , Immunity, Cellular , Influenza A virus/genetics , Influenza Vaccines/immunology , Influenza, Human , Nucleocapsid Proteins/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/transmission , T-Lymphocytes/immunology , Vaccination , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology
3.
Vaccine ; 39(33): 4628-4640, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34226103

ABSTRACT

Current influenza vaccines rely on inducing antibody responses to the rapidly evolving hemagglutinin (HA) and neuraminidase (NA) proteins, and thus need to be strain-matched. However, predictions of strains that will circulate are imperfect, and manufacturing of new vaccines based on them takes months. As an alternative, universal influenza vaccines target highly conserved antigens. In proof of concept studies of universal vaccine candidates in animal models challenge is generally conducted only a short time after vaccination, but protective immunity lasting far longer is important for the intended public health impact. We address the challenge of providing long-term protection. We demonstrate here broad, powerful, and long-lasting immune protection for a promising universal vaccine candidate. A single intranasal dose of recombinant adenoviruses (rAd) expressing influenza A nucleoprotein (A/NP) and matrix 2 (M2) was used. Extending our previous studies of this type of vaccine, we show that antibody and T-cell responses persist for over a year without boosting, and that protection against challenge persists a year after vaccination and remains broad, covering both group 1 and 2 influenza A viruses. In addition, we extend the work to influenza B. Immunization with influenza B nucleoprotein (B/NP)-rAd also gives immune responses that last a year without boosting and protect against challenge with influenza B viruses of mismatched HA lineages. Despite host immunity to adenoviral antigens, effective readministration is possible a year after primary vaccination, as shown by successful immunization to a transgene product the animals had not seen before. Protection against challenge with divergent and highly pathogenic A/H7N9 virus was weaker but was enhanced by a second dose of vaccine. Thus, this mucosal vaccination to conserved influenza antigens confers very long-lasting immune protection in animals against a broad range of influenza A and B viruses.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Animals , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunity , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Vaccination
4.
J Virol ; 95(9)2021 04 12.
Article in English | MEDLINE | ID: mdl-33627390

ABSTRACT

Current influenza vaccines, live attenuated or inactivated, do not protect against antigenically novel influenza A viruses (IAVs) of pandemic potential, which has driven interest in the development of universal influenza vaccines. Universal influenza vaccine candidates targeting highly conserved antigens of IAV nucleoprotein (NP) are promising as vaccines that induce T cell immunity, but concerns have been raised about the safety of inducing robust CD8 T cell responses in the lungs. Using a mouse model, we systematically evaluated effects of recombinant adenovirus vectors (rAd) expressing IAV NP (A/NP-rAd) or influenza B virus (IBV) NP (B/NP-rAd) on pulmonary inflammation and function after vaccination and following live IAV challenge. After A/NP-rAd or B/NP-rAd vaccination, female mice exhibited robust systemic and pulmonary vaccine-specific B cell and T cell responses and experienced no morbidity (e.g., body mass loss). Both in vivo pulmonary function testing and lung histopathology scoring revealed minimal adverse effects of intranasal rAd vaccination compared with unvaccinated mice. After IAV challenge, A/NP-rAd-vaccinated mice experienced significantly less morbidity, had lower pulmonary virus titers, and developed less pulmonary inflammation than unvaccinated or B/NP-rAd-vaccinated mice. Based on analysis of pulmonary physiology using detailed testing not previously applied to the question of T cell damage, mice protected by vaccination also had better lung function than controls. Results provide evidence that, in this model, adenoviral universal influenza vaccine does not damage pulmonary tissue. In addition, adaptive immunity, in particular, T cell immunity in the lungs, does not cause damage when restimulated but instead mitigates pulmonary damage following IAV infection.IMPORTANCE Respiratory viruses can emerge and spread rapidly before vaccines are available. It would be a tremendous advance to use vaccines that protect against whole categories of viruses, such as universal influenza vaccines, without the need to predict which virus will emerge. The nucleoprotein (NP) of influenza virus provides a target conserved among strains and is a dominant T cell target. In animals, vaccination to NP generates powerful T cell immunity and long-lasting protection against diverse influenza strains. Concerns have been raised, but not evaluated experimentally, that potent local T cell responses might damage the lungs. We analyzed lung function in detail in the setting of such a vaccination. Despite CD8 T cell responses in the lungs, lungs were not damaged and functioned normally after vaccination alone and were protected upon subsequent infection. This precedent provides important support for vaccines based on T cell-mediated protection, currently being considered for both influenza and SARS-CoV-2 vaccines.


Subject(s)
Adenoviridae , Genetic Vectors , Influenza B virus , Influenza Vaccines , Lung , Orthomyxoviridae Infections , Adenoviridae/genetics , Adenoviridae/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Disease Models, Animal , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Immunity, Cellular , Influenza B virus/genetics , Influenza B virus/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
PLoS Pathog ; 16(9): e1008583, 2020 09.
Article in English | MEDLINE | ID: mdl-32970783

ABSTRACT

The prospect of universal influenza vaccines is generating much interest and research at the intersection of immunology, epidemiology, and viral evolution. While the current focus is on developing a vaccine that elicits a broadly cross-reactive immune response in clinical trials, there are important downstream questions about global deployment of a universal influenza vaccine that should be explored to minimize unintended consequences and maximize benefits. Here, we review and synthesize the questions most relevant to predicting the population benefits of universal influenza vaccines and discuss how existing information could be mined to begin to address these questions. We review three research topics where computational modeling could bring valuable evidence: immune imprinting, viral evolution, and transmission. We address the positive and negative consequences of imprinting, in which early childhood exposure to influenza shapes and limits immune responses to future infections via memory of conserved influenza antigens. However, the mechanisms at play, their effectiveness, breadth of protection, and the ability to "reprogram" already imprinted individuals, remains heavily debated. We describe instances of rapid influenza evolution that illustrate the plasticity of the influenza virus in the face of drug pressure and discuss how novel vaccines could introduce new selective pressures on the evolution of the virus. We examine the possible unintended consequences of broadly protective (but infection-permissive) vaccines on the dynamics of epidemic and pandemic influenza, compared to conventional vaccines that have been shown to provide herd immunity benefits. In conclusion, computational modeling offers a valuable tool to anticipate the benefits of ambitious universal influenza vaccine programs, while balancing the risks from endemic influenza strains and unpredictable pandemic viruses. Moving forward, it will be important to mine the vast amount of data generated in clinical studies of universal influenza vaccines to ensure that the benefits and consequences of these vaccine programs have been carefully modeled and explored.


Subject(s)
Antibodies, Viral/immunology , Biomedical Research/trends , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Clinical Trials as Topic , Humans , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/virology
6.
PLoS One ; 14(4): e0215321, 2019.
Article in English | MEDLINE | ID: mdl-30986224

ABSTRACT

Current approaches to influenza control rely on vaccines matched to viruses in circulation. Universal influenza vaccines would offer the advantage of providing broad protection against diverse strains of influenza virus. Candidate universal vaccines are developed using model systems, often testing in naïve animals. Yet the human population is not naïve, having varied immune histories that include exposure to viruses. We studied a candidate universal influenza vaccine (replication deficient adenoviruses expressing the conserved influenza A antigens NP and M2 [A/NP+M2-rAd]) given intranasally, the route previously shown to be most effective. To model recipients exposed to viruses, we used mice given rhinovirus (RV1B), respiratory syncytial virus (RSV-A2), influenza B virus, or influenza A virus before or after universal influenza vaccine. Vaccine performance was assessed by measuring immune responses to NP and M2, and monitoring weight loss and survival following influenza A challenge. Prior influenza A virus infection enhanced the response to the vaccine by priming to conserved influenza A antigens. RSV-A2 or RV1B had no effect on antibody responses to NP and M2 in serum. None of the viruses inhibited the ability of the vaccine to protect against influenza A virus challenge. The study demonstrates that the usefulness of this universal vaccine is not confined to the immunologically naïve and supports possible use in a human population with a varied history of respiratory infections.


Subject(s)
Common Variable Immunodeficiency/immunology , Coxsackievirus Infections/immunology , Enterovirus/immunology , Immunogenicity, Vaccine , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Common Variable Immunodeficiency/virology , Coxsackievirus Infections/pathology , Female , HeLa Cells , Humans , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology
7.
Am J Epidemiol ; 187(12): 2603-2614, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30084906

ABSTRACT

Despite all we have learned since 1918 about influenza virus and immunity, available influenza vaccines remain inadequate to control outbreaks of unexpected strains. Universal vaccines not requiring strain matching would be a major improvement. Their composition would be independent of predicting circulating viruses and thus potentially effective against unexpected drift or pandemic strains. This commentary explores progress with candidate universal vaccines based on various target antigens. Candidates include vaccines based on conserved viral proteins such as nucleoprotein and matrix, on the conserved hemagglutinin (HA) stem, and various combinations. Discussion covers the differing evidence for each candidate vaccine demonstrating protection in animals against influenza viruses of widely divergent HA subtypes and groups; durability of protection; routes of administration, including mucosal, providing local immunity; and reduction of transmission. Human trials of some candidate universal vaccines have been completed or are underway. Interestingly, the HA stem, like nucleoprotein and matrix, induces immunity that permits some virus replication and emergence of escape mutants fit enough to cause disease. Vaccination with multiple target antigens will thus have advantages over use of single antigens. Ultimately, a universal vaccine providing long-term protection against all influenza virus strains might contribute to pandemic control and routine vaccination.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Antibodies, Viral/immunology , Cross Protection/immunology , Drug Administration Routes , Epitopes, T-Lymphocyte/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/administration & dosage , Neuraminidase/immunology , Nucleoproteins/immunology , Viral Matrix Proteins/immunology
8.
Vaccine ; 36(32 Pt B): 4910-4918, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30037481

ABSTRACT

Transmission of influenza virus between susceptible hosts mediates spread of infection in the population and can occur via direct-contact or airborne routes. Mathematical models suggest that vaccines that reduce viral transmission from infected individuals could substantially reduce viral spread in an epidemic or pandemic, even if they do not completely protect against infection. Vaccines targeting conserved nucleoprotein (A/NP) and matrix 2 (M2) antigens of influenza virus do not completely prevent infection upon influenza virus challenge, but reduce viral replication, morbidity, and mortality. Using a mouse model of influenza virus transmission, we have previously shown that immunization with recombinant adenovirus vectors expressing the combination of A/NP and M2 can reduce viral transmission to unimmunized contacts. Here we demonstrate that transmission reduction is more effective when mice are immunized against A/NP and M2 intranasally than via the intramuscular route. We show that immunization against the combination of A/NP and M2 is more effective at reducing transmission than either antigen alone, with a clear hierarchy of effectiveness (A/NP + M2 > A/NP > M2). Transmission reduction is seen to a similar degree under both direct-contact and airborne transmission conditions. Finally, using seroconversion as an indicator of infection, we show that immunizing contact mice against A/NP and M2 prevents a significant fraction (∼50%) from becoming infected under direct-contact conditions. These findings suggest that when strain-matched vaccines are unavailable, conserved antigen vaccines could not only reduce severity of disease in vaccinated individuals but also limit the spread of virus during influenza epidemics or pandemics.


Subject(s)
Antigens, Viral/immunology , Influenza Vaccines/therapeutic use , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Adenoviridae/genetics , Animals , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza Vaccines/immunology , Mice , Vaccination/methods
9.
Vaccine ; 36(7): 1008-1015, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29249542

ABSTRACT

Universal influenza vaccines are designed to protect against diverse strains of influenza virus. Preclinical testing of new vaccine candidates is usually done in naïve animals, despite intended use in the human population with its varied immune history including responses to previous vaccinations. As an approach more relevant to human use, we tested a candidate universal influenza vaccine in mice with a history of conventional vaccination. Female BALB/c mice were given two intramuscular doses of inactivated influenza vaccine (IIV) or diphtheria and tetanus toxoids vaccine (DT), one month apart. Another group was given two intranasal doses of live attenuated influenza virus (LAIV). One month after the second dose, mice were given the universal influenza vaccine: recombinant adenoviruses expressing influenza A nucleoprotein (A/NP) and matrix 2 (M2) (A/NP + M2-rAd). Immune responses to universal vaccine antigens A/NP and M2 were assessed by ELISA and interferon-γ ELISPOT. Protection was tested by challenge with mouse-adapted A/FM/1/47 (H1N1) and monitoring for weight loss and survival. Universal vaccine performance was enhanced, inhibited or unaffected by particular prior vaccinations. Mice given Afluria IIV and LAIV had greater antibody and T-cell response to A/NP than mice without prior vaccination, providing examples of enhanced A/NP + M2-rAd performance. Though Fluvirin IIV partially inhibited, the universal vaccine still provided considerable protection unlike conventional vaccination. Fluzone IIV and DT had no effect on A/NP + M2-rAd performance. Thus our results demonstrate that universal vaccine candidate A/NP + M2-rAd was at least partially effective in mice with diverse prior histories. However, the degree of protection and nature of the immune responses may be affected by a history of conventional vaccination and suggests that performance in humans would be influenced by immune history.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Disease Models, Animal , Female , Immunity, Cellular/immunology , Immunity, Mucosal , Immunization , Influenza Vaccines/classification , Mice , Outcome Assessment, Health Care , Vaccines, Inactivated/immunology , Viral Proteins/immunology
10.
Open Forum Infect Dis ; 4(2): ofx023, 2017.
Article in English | MEDLINE | ID: mdl-28730155

ABSTRACT

BACKGROUND: Antibody and T-cell immunity to conserved influenza virus antigens can protect animals against infection with diverse influenza strains. Although immunity against conserved antigens occurs in humans, whether such responses provide cross-protection in humans and could be harnessed as the basis for universal influenza vaccines is controversial. The 2009 pandemic provided an opportunity to investigate whether pre-existing cross-reactive immunity affected susceptibility to infection. METHODS: In 2009, we banked sera and peripheral blood mononuclear cells (PBMC) from blood donors, then monitored them for pandemic influenza infection (pH1N1) by polymerase chain reaction or seroconversion. Antibodies to hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix 2 (M2), and HA-pseudotypes were measured in sera. T-cell inteferon-γ enzyme-linked immunospot responses were measured in PBMC. RESULTS: There were 13 infections in 117 evaluable donors. Pre-existing T-cell reactivity to pH1N1 was substantial (of 153 donors tested, 146 had >100 spot-forming cells/106 cells). Antibodies reactive with pH1N1 were common: anti-NP (all donors) and anti-M2 (44% of donors). Pseudotype-neutralizing antibodies to H1 were detected, but not to highly conserved HA epitopes. Unexpectedly, donors with symptomatic pH1N1 infection had sharp rises in HA pseudotype-neutralizing antibodies, not only pH1N1 but also against multiple seasonal H1s. In addition, an exploratory study of a T-cell marker (response to NP418-426) identified probable infection missed by standard criteria. CONCLUSIONS: Although the number of infections was inadequate for conclusions about mechanisms of protection, this study documents the wide variety of pre-existing, cross-reactive, humoral and cellular immune responses to pandemic influenza virus antigens in humans. These responses can be compared with results of other studies and explored in universal influenza vaccine studies.

11.
Science ; 354(6313): 706-707, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27846592

Subject(s)
Influenza, Human , Humans
12.
PLoS One ; 11(4): e0153195, 2016.
Article in English | MEDLINE | ID: mdl-27055234

ABSTRACT

Influenza has a major impact on the elderly due to increased susceptibility to infection with age and poor response to current vaccines. We have studied universal influenza vaccine candidates based on influenza A nucleoprotein and matrix 2 (A/NP+M2). Long-lasting protection against influenza virus strains of divergent subtypes is induced, especially with mucosal immunization. Here, we tested universal vaccination in BALB/c mice of different ages. Vaccination used intramuscular DNA priming to A/NP+M2 followed by intranasal (i.n.) boosting with recombinant adenoviruses (rAd) expressing the same antigens, or only A/NP+M2-rAd given i.n. Antigen-specific systemic antibody responses were induced in young, middle-aged, and elderly mice (2, 11-17, and 20 months old, respectively), but decreased with age. Antibody responses in bronchoalveolar lavage (BAL) were detected only in young mice. Antigen-specific T cell responses were seen in young and middle-aged but not elderly mice. A/NP+M2 vaccination by the two regimens above protected against stringent challenge in young and middle-aged mice, but not in elderly mice. However, mice vaccinated with A/NP-rAd or A/M2-rAd during their youth were partially protected against challenge 16 months later when they were elderly. In addition, a regimen of two doses of A/NP+M2-rAd given i.n. one month apart beginning in old age protected elderly mice against stringent challenge. This study highlights the potential benefit of cross-protective vaccines through middle age, and suggests that their performance might be enhanced in elderly individuals who had been exposed to influenza antigens early in life, as most humans have been, or by a two-dose rAd regimen given later in life.


Subject(s)
Antibodies, Viral/blood , Immunity, Mucosal/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/therapeutic use , Orthomyxoviridae Infections/immunology , Adenoviridae/genetics , Age Factors , Animals , Antibodies, Viral/immunology , Cross Protection , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Vaccination
13.
J Virol ; 88(11): 6019-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24623430

ABSTRACT

UNLABELLED: Pandemic influenza is a major public health concern, but conventional strain-matched vaccines are unavailable early in a pandemic. Candidate "universal" vaccines targeting the viral antigens nucleoprotein (NP) and matrix 2 (M2), which are conserved among all influenza A virus strains and subtypes, could be manufactured in advance for use at the onset of a pandemic. These vaccines do not prevent infection but can reduce disease severity, deaths, and virus titers in the respiratory tract. We hypothesized that such immunization may reduce virus transmission from vaccinated, infected animals. To investigate this hypothesis, we studied mouse models for direct-contact and airborne transmission of H1N1 and H3N2 influenza viruses. We established conditions under which virus transmission occurs and showed that transmission efficiency is determined in part at the level of host susceptibility to infection. Our findings indicate that virus transmission between mice has both airborne and direct-contact components. Finally, we demonstrated that immunization with recombinant adenovirus vectors expressing NP and M2 significantly reduced the transmission of virus to cohoused, unimmunized mice in comparison to controls. These findings have broad implications for the impact of conserved-antigen vaccines, not only in protecting the vaccinated individual but also in protecting others by limiting influenza virus transmission and potentially reducing the size of epidemics. IMPORTANCE: Using a mouse model of influenza A virus transmission, we demonstrate that a candidate "universal" influenza vaccine both protects vaccinated animals from lethal infection and reduces the transmission of virus from vaccinated to nonvaccinated mice. This vaccine induces immunity against proteins conserved among all known influenza A virus strains and subtypes, so it could be used early in a pandemic before conventional strain-matched vaccines are available and could potentially reduce the spread of infection in the community.


Subject(s)
Disease Susceptibility/virology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/transmission , Adenoviridae , Animals , Bronchoalveolar Lavage , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Hemagglutination Inhibition Tests , Immunity, Mucosal , Mice , Neutralization Tests , Nucleoproteins/metabolism , Viral Matrix Proteins/metabolism
15.
PLoS One ; 8(3): e55435, 2013.
Article in English | MEDLINE | ID: mdl-23536756

ABSTRACT

Among approximately 1000 adenoviruses from chimpanzees and bonobos studied recently, the Pan Adenovirus type 3 (PanAd3, isolated from a bonobo, Pan paniscus) has one of the best profiles for a vaccine vector, combining potent transgene immunogenicity with minimal pre-existing immunity in the human population. In this study, we inserted into a replication defective PanAd3 a transgene expressing a fusion protein of conserved influenza antigens nucleoprotein (NP) and matrix 1 (M1). We then studied antibody and T cell responses as well as protection from challenge infection in a mouse model. A single intranasal administration of PanAd3-NPM1 vaccine induced strong antibody and T cell responses, and protected against high dose lethal influenza virus challenge. Thus PanAd3 is a promising candidate vector for vaccines, including universal influenza vaccines.


Subject(s)
Adenoviruses, Simian/immunology , Antigens, Viral/immunology , Genetic Vectors/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Adenoviruses, Human/immunology , Adenoviruses, Simian/genetics , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/genetics , Cross Reactions/immunology , Female , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Immunity, Mucosal , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Mice , Molecular Sequence Data , Nucleocapsid Proteins , Nucleophosmin , Pan paniscus , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
16.
Proc Natl Acad Sci U S A ; 109(8): 3173-7, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22323589

ABSTRACT

Large-scale immunization has profoundly impacted control of many infectious diseases such as measles and smallpox because of the ability of vaccination campaigns to maintain long-term herd immunity and, hence, indirect protection of the unvaccinated. In the case of human influenza, such potential benefits of mass vaccination have so far proved elusive. The central difficulty is a considerable viral capacity for immune escape; new pandemic variants, as well as viral escape mutants in seasonal influenza, compromise the buildup of herd immunity from natural infection or deployment of current vaccines. Consequently, most current influenza vaccination programs focus mainly on protection of specific risk groups, rather than mass prophylactic protection. Here, we use epidemiological models to show that emerging vaccine technologies, aimed at broad-spectrum protection, could qualitatively alter this picture. We demonstrate that sustained immunization with such vaccines could--through potentially lowering transmission rates and improving herd immunity--significantly moderate both influenza pandemic and seasonal epidemics. More subtly, phylodynamic models indicate that widespread cross-protective immunization could slow the antigenic evolution of seasonal influenza; these effects have profound implications for a transition to mass vaccination strategies against human influenza, and for the management of antigenically variable viruses in general.


Subject(s)
Cross Protection/immunology , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Influenza, Human/transmission , Antigens, Viral/immunology , Humans , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Models, Immunological , Vaccination , Virus Shedding/immunology
17.
Mol Ther ; 19(11): 1990-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21829176

ABSTRACT

A critical aspect in defining the utility of a vector for gene therapy applications is the cell tropism and biodistribution of the vector. Adeno-associated virus type 12 (AAV12) has several unique biological and immunological properties that could be exploited for gene therapy purposes, including a unique cell surface receptor, transduction of epithelial cells, and limited neutralization by pooled human antibodies. However, little is known about its cell tropism and biodistribution in vivo. In vivo biodistribution studies with AAV12 vectors encoding a cytomegalovirus promoted luciferase transgene indicated preferential transduction of the nasal epithelia which was not observed with AAV2-based vectors. Expression peaked 2 weeks postadministration, before decreasing to a persistent level. The level of neutralizing antibodies (Nab) induced was sevenfold lower for AAV12 than for AAV2, an advantage for use in repeat administration. Furthermore, vectors encoding influenza A nucleoprotein (NP), an antigen which has previously been shown to induce immune protection against challenge, resulted in generation of both anti-A/NP antibodies and lung anti-A/NP T cells. Our findings suggest further evaluation of AAV12 as a vector for gene therapy and as a potential nasal vaccine.


Subject(s)
Dependovirus/genetics , Dependovirus/immunology , Genetic Vectors/administration & dosage , Nasal Mucosa/metabolism , Transduction, Genetic , Transgenes , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , COS Cells , Chlorocebus aethiops , Genes, Reporter/immunology , Genetic Vectors/pharmacokinetics , HEK293 Cells , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Intramuscular , Injections, Intraperitoneal , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Respiratory Mucosa/metabolism , T-Lymphocytes/immunology , Time Factors , Transgenes/immunology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
18.
PLoS One ; 6(7): e21937, 2011.
Article in English | MEDLINE | ID: mdl-21789196

ABSTRACT

BACKGROUND: The rapid spread of the 2009 H1N1 pandemic influenza virus (pH1N1) highlighted problems associated with relying on strain-matched vaccines. A lengthy process of strain identification, manufacture, and testing is required for current strain-matched vaccines and delays vaccine availability. Vaccines inducing immunity to conserved viral proteins could be manufactured and tested in advance and provide cross-protection against novel influenza viruses until strain-matched vaccines became available. Here we test two prototype vaccines for cross-protection against the recent pandemic virus. METHODOLOGY/PRINCIPAL FINDINGS: BALB/c and C57BL/6 mice were intranasally immunized with a single dose of cold-adapted (ca) influenza viruses from 1977 or recombinant adenoviruses (rAd) expressing 1934 nucleoprotein (NP) and consensus matrix 2 (M2) (NP+M2-rAd). Antibodies against the M2 ectodomain (M2e) were seen in NP+M2-rAd immunized BALB/c but not C57BL/6 mice, and cross-reacted with pH1N1 M2e. The ca-immunized mice did not develop antibodies against M2e. Despite sequence differences between vaccine and challenge virus NP and M2e epitopes, extensive cross-reactivity of lung T cells with pH1N1 peptides was detected following immunization. Both ca and NP+M2-rAd immunization protected BALB/c and C57BL/6 mice against challenge with a mouse-adapted pH1N1 virus. CONCLUSION/SIGNIFICANCE: Cross-protective vaccines such as NP+M2-rAd and ca virus are effective against pH1N1 challenge within 3 weeks of immunization. Protection was not dependent on recognition of the highly variable external viral proteins and could be achieved with a single vaccine dose. The rAd vaccine was superior to the ca vaccine by certain measures, justifying continued investigation of this experimental vaccine even though ca vaccine is already available. This study highlights the potential for cross-protective vaccines as a public health option early in an influenza pandemic.


Subject(s)
Adaptation, Physiological/immunology , Adenoviridae/immunology , Cross Protection/immunology , Immunity/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Cold Temperature , Epitopes/chemistry , Epitopes/immunology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pandemics , T-Lymphocytes/immunology , Vaccination , Viral Load/immunology
19.
J Immunol ; 186(4): 2422-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242525

ABSTRACT

Maturation of dendritic cells (DC) to competent APC is essential for the generation of acquired immunity and is a major function of adjuvants. dsRNA, a molecular signature of viral infection, drives DC maturation by activating TLR3, but the size of dsRNA required to activate DC and the expression patterns of TLR3 protein in DC subsets have not been established. In this article, we show that cross-priming CD8α(+) and CD103(+) DC subsets express much greater levels of TLR3 than other DC. In resting DC, TLR3 is located in early endosomes and other intracellular compartments but migrates to LAMP1(+) endosomes on stimulation with a TLR3 ligand. Using homogeneous dsRNA oligonucleotides (ONs) ranging in length from 25 to 540 bp, we observed that a minimum length of ∼90 bp was sufficient to induce CD86, IL-12p40, IFN-ß, TNF-α, and IL-6 expression, and to mature DC into APC that cross-presented exogenous Ags to CD8(+) T cells. TLR3 was essential for activation of DC by dsRNA ONs, and the potency of activation increased with dsRNA length and varied between DC subsets. In vivo, dsRNA ONs, in a size-dependent manner, served as adjuvants for the generation of Ag-specific CTL and for inducing protection against lethal challenge with influenza virus when given with influenza nucleoprotein as an immunogen. These results provide the basis for the development of TLR3-specific adjuvants capable of inducing immune responses tailored for viral pathogens.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cross-Priming/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Oligodeoxyribonucleotides/therapeutic use , Orthomyxoviridae Infections/prevention & control , RNA, Double-Stranded/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 3/immunology , Adaptive Immunity/genetics , Animals , Cells, Cultured , CpG Islands/immunology , Cytotoxicity Tests, Immunologic , Dendritic Cells/metabolism , Dendritic Cells/virology , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/therapeutic use , Female , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/virology , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/therapeutic use
20.
Expert Rev Vaccines ; 9(11): 1325-41, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21087110

ABSTRACT

Antigenic changes in influenza virus occur gradually, owing to mutations (antigenic drift), and abruptly, owing to reassortment among subtypes (antigenic shift). Availability of strain-matched vaccines often lags behind these changes, resulting in a shortfall in public health. In animal models, cross-protection by vaccines based on conserved antigens does not completely prevent infection, but greatly reduces morbidity, mortality, virus replication and, thus, viral shedding and spread. Such immunity is especially effective and long-lasting with mucosal administration. Cross-protective immunity in humans is controversial, but is suggested by some epidemiological findings. 'Universal' vaccines protective against all influenza A viruses might substantially reduce severity of infection and limit spread of disease during outbreaks. These vaccines could be used 'off the shelf' early in an outbreak or pandemic, before strain-matched vaccines are available.


Subject(s)
Cross Protection , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Animals , Disease Models, Animal , Humans , Influenza, Human/pathology , Influenza, Human/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...