Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 11(492)2019 05 15.
Article in English | MEDLINE | ID: mdl-31092692

ABSTRACT

Western, Eastern, and Venezuelan equine encephalitis viruses (WEEV, EEEV, and VEEV, respectively) are important mosquito-borne agents that pose public health and bioterrorism threats. Despite considerable advances in understanding alphavirus replication, there are currently no available effective vaccines or antiviral treatments against these highly lethal pathogens. To develop a potential countermeasure for viral encephalitis, we generated a trivalent, or three-component, EEV vaccine composed of virus-like particles (VLPs). Monovalent VLPs elicited neutralizing antibody responses and protected mice and nonhuman primates (NHPs) against homologous challenges, but they were not cross-protective. In contrast, NHPs immunized with trivalent VLPs were completely protected against aerosol challenge by each of these three EEVs. Passive transfer of IgG from immunized NHPs protected mice against aerosolized EEV challenge, demonstrating that the mechanism of protection was humoral. Because they are replication incompetent, these trivalent VLPs represent a potentially safe and effective vaccine that can protect against diverse encephalitis viruses.


Subject(s)
Encephalitis Viruses/immunology , Encephalitis, Arbovirus/immunology , Encephalitis, Arbovirus/prevention & control , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Neutralizing/immunology , Encephalitis, Arbovirus/pathology , Encephalitis, Arbovirus/virology , Immunization , Immunoglobulin G/immunology , Macaca fascicularis , Mice, Inbred BALB C , Vaccines, Virus-Like Particle/ultrastructure
2.
PLoS Pathog ; 12(3): e1005486, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26943673

ABSTRACT

CD8 T cells are involved in pathogen clearance and infection-induced pathology in respiratory syncytial virus (RSV) infection. Studying bulk responses masks the contribution of individual CD8 T cell subsets to protective immunity and immunopathology. In particular, the roles of subdominant responses that are potentially beneficial to the host are rarely appreciated when the focus is on magnitude instead of quality of response. Here, by evaluating CD8 T cell responses in CB6F1 hybrid mice, in which multiple epitopes are recognized, we found that a numerically subdominant CD8 T cell response against DbM187 epitope of the virus matrix protein expressed high avidity TCR and enhanced signaling pathways associated with CD8 T cell effector functions. Each DbM187 T effector cell lysed more infected targets on a per cell basis than the numerically dominant KdM282 T cells, and controlled virus replication more efficiently with less pulmonary inflammation and illness than the previously well-characterized KdM282 T cell response. Our data suggest that the clinical outcome of viral infections is determined by the integrated functional properties of a variety of responding CD8 T cells, and that the highest magnitude response may not necessarily be the best in terms of benefit to the host. Understanding how to induce highly efficient and functional T cells would inform strategies for designing vaccines intended to provide T cell-mediated immunity.


Subject(s)
Immunity, Cellular , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Female , Mice , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology , Viral Load , Virus Replication
3.
Nat Commun ; 6: 8447, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26485194

ABSTRACT

The treatment of AIDS with combination antiretroviral therapy (cART) remains lifelong largely because the virus persists in latent reservoirs. Elimination of latently infected cells could therefore reduce treatment duration and facilitate immune reconstitution. Here we report an approach to reduce the viral reservoir by activating dormant viral gene expression and directing T lymphocytes to lyse previously latent, HIV-1-infected cells. An immunomodulatory protein was created that combines the specificity of a HIV-1 broadly neutralizing antibody with that of an antibody to the CD3 component of the T-cell receptor. CD3 engagement by the protein can stimulate T-cell activation that induces proviral gene expression in latently infected T cells. It further stimulates CD8 T-cell effector function and redirects T cells to lyse these previously latent-infected cells through recognition of newly expressed Env. This immunomodulatory protein could potentially help to eliminate latently infected cells and deplete the viral reservoir in HIV-1-infected individuals.


Subject(s)
Antibodies, Neutralizing/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Gene Expression Regulation, Viral/drug effects , HIV Infections/immunology , HIV-1/drug effects , Immunologic Factors/pharmacology , Lymphocyte Activation/drug effects , Animals , Base Sequence , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytokines/drug effects , Cytokines/immunology , HEK293 Cells , HIV Infections/drug therapy , HIV-1/immunology , Humans , Lymphocyte Activation/immunology , Macaca mulatta , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell , Simian Acquired Immunodeficiency Syndrome/immunology , env Gene Products, Human Immunodeficiency Virus/immunology
4.
Cell ; 162(5): 1090-100, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26279189

ABSTRACT

Epstein-Barr virus (EBV) represents a major global health problem. Though it is associated with infectious mononucleosis and ∼200,000 cancers annually worldwide, a vaccine is not available. The major target of immunity is EBV glycoprotein 350/220 (gp350) that mediates attachment to B cells through complement receptor 2 (CR2/CD21). Here, we created self-assembling nanoparticles that displayed different domains of gp350 in a symmetric array. By focusing presentation of the CR2-binding domain on nanoparticles, potent neutralizing antibodies were elicited in mice and non-human primates. The structurally designed nanoparticle vaccine increased neutralization 10- to 100-fold compared to soluble gp350 by targeting a functionally conserved site of vulnerability, improving vaccine-induced protection in a mouse model. This rational approach to EBV vaccine design elicited potent neutralizing antibody responses by arrayed presentation of a conserved viral entry domain, a strategy that can be applied to other viruses.


Subject(s)
Herpesvirus Vaccines/chemistry , Herpesvirus Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Crystallography, X-Ray , Drug Design , Female , Herpesvirus 4, Human , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/isolation & purification , Macaca fascicularis , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
5.
Nat Med ; 21(9): 1065-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26301691

ABSTRACT

The antibody response to influenza is primarily focused on the head region of the hemagglutinin (HA) glycoprotein, which in turn undergoes antigenic drift, thus necessitating annual updates of influenza vaccines. In contrast, the immunogenically subdominant stem region of HA is highly conserved and recognized by antibodies capable of binding multiple HA subtypes. Here we report the structure-based development of an H1 HA stem-only immunogen that confers heterosubtypic protection in mice and ferrets. Six iterative cycles of structure-based design (Gen1-Gen6) yielded successive H1 HA stabilized-stem (HA-SS) immunogens that lack the immunodominant head domain. Antigenic characterization, determination of two HA-SS crystal structures in complex with stem-specific monoclonal antibodies and cryo-electron microscopy analysis of HA-SS on ferritin nanoparticles (H1-SS-np) confirmed the preservation of key structural elements. Vaccination of mice and ferrets with H1-SS-np elicited broadly cross-reactive antibodies that completely protected mice and partially protected ferrets against lethal heterosubtypic H5N1 influenza virus challenge despite the absence of detectable H5N1 neutralizing activity in vitro. Passive transfer of immunoglobulin from H1-SS-np-immunized mice to naive mice conferred protection against H5N1 challenge, indicating that vaccine-elicited HA stem-specific antibodies can protect against diverse group 1 influenza strains.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Animals , Antibodies, Viral/blood , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Mice , Mice, Inbred BALB C , Nanoparticles , Vaccination
6.
Nat Commun ; 6: 8020, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26282376

ABSTRACT

Mucosal damage to the gastrointestinal (GI) tract with resulting microbial translocation is hypothesized to significantly contribute to the heightened and persistent chronic inflammation and immune activation characteristic to HIV infection. Here we employ a non-human primate model of chemically induced colitis in SIV-uninfected rhesus macaques that we developed using dextran sulfate sodium (DSS), to directly test this hypothesis. DSS treatment results in GI barrier damage with associated microbial translocation, inflammation and immune activation. The progression and severity of colitis are longitudinally monitored by a magnetic resonance imaging approach. DSS treatment of SIV-infected African green monkeys, a natural host species for SIV that does not manifest GI tract damage or chronic immune activation during infection, results in colitis with elevated levels of plasma SIV RNA, sCD14, LPS, CRP and mucosal CD4+ T-cell loss. Together these results support the hypothesis that GI tract damage leading to local and systemic microbial translocation, and associated immune activation, are important determinants of AIDS pathogenesis.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus , Animals , Dextran Sulfate/toxicity , Female , Gastrointestinal Tract/pathology , Macaca mulatta , Male
7.
Nat Commun ; 6: 7712, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26218507

ABSTRACT

The emergence of Middle East respiratory syndrome coronavirus (MERS-CoV) as a cause of severe respiratory disease highlights the need for effective approaches to CoV vaccine development. Efforts focused solely on the receptor-binding domain (RBD) of the viral Spike (S) glycoprotein may not optimize neutralizing antibody (NAb) responses. Here we show that immunogens based on full-length S DNA and S1 subunit protein elicit robust serum-neutralizing activity against several MERS-CoV strains in mice and non-human primates. Serological analysis and isolation of murine monoclonal antibodies revealed that immunization elicits NAbs to RBD and, non-RBD portions of S1 and S2 subunit. Multiple neutralization mechanisms were demonstrated by solving the atomic structure of a NAb-RBD complex, through sequencing of neutralization escape viruses and by constructing MERS-CoV S variants for serological assays. Immunization of rhesus macaques confers protection against MERS-CoV-induced radiographic pneumonia, as assessed using computerized tomography, supporting this strategy as a promising approach for MERS-CoV vaccine development.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Coronavirus Infections/prevention & control , DNA, Viral/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Monoclonal/immunology , Female , HEK293 Cells , Humans , Immunoglobulin G/immunology , Macaca mulatta , Male , Mice , Mice, Inbred BALB C
8.
J Virol ; 89(16): 8334-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041300

ABSTRACT

Broadly neutralizing antibodies (bnAbs) can prevent lentiviral infection in nonhuman primates and may slow the spread of human immunodeficiency virus type 1 (HIV-1). Although protection by passive transfer of human bnAbs has been demonstrated in monkeys, durable expression is essential for its broader use in humans. Gene-based expression of bnAbs provides a potential solution to this problem, although immune responses to the viral vector or to the antibody may limit its durability and efficacy. Here, we delivered an adeno-associated viral vector encoding a simianized form of a CD4bs bnAb, VRC07, and evaluated its immunogenicity and protective efficacy. The expressed antibody circulated in macaques for 16 weeks at levels up to 66 g/ml, although immune suppression with cyclosporine (CsA) was needed to sustain expression. Gene-delivered simian VRC07 protected against simian-human immunodeficiency virus (SHIV) infection in monkeys 5.5 weeks after treatment. Gene transfer of an anti-HIV antibody can therefore protect against infection by viruses that cause AIDS in primates when the host immune responses are controlled.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/prevention & control , HIV-1/immunology , Immunoglobulin G/genetics , Models, Molecular , Amino Acid Sequence , Animals , Antibodies, Neutralizing/genetics , Cyclosporine/pharmacology , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , HIV Antibodies/genetics , Humans , Immunosuppressive Agents/pharmacology , Macaca mulatta , Molecular Sequence Data , Neutralization Tests , Real-Time Polymerase Chain Reaction
9.
J Virol ; 89(11): 5895-903, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25787288

ABSTRACT

UNLABELLED: Pathogen-specific neutralizing antibodies protect against many viral infections and can potentially prevent human immunodeficiency virus (HIV) transmission in humans. However, neutralizing antibodies have so far only been shown to protect nonhuman primates (NHP) against lentiviral infection when given shortly before challenge. Thus, the clinical utility and feasibility of passive antibody transfer to confer long-term protection against HIV-1 are still debated. Here, we investigate the potential of a broadly neutralizing HIV-1 antibody to provide long-term protection in a NHP model of HIV-1 infection. A human antibody was simianized to avoid immune rejection and used to sustain therapeutic levels for ∼5 months. Two months after the final antibody administration, animals were completely protected against viral challenge. These findings demonstrate the feasibility and potential of long-term passive antibody for protection against HIV-1 in humans and provide a model to test antibody therapies for other diseases in NHP. IMPORTANCE: Antibodies against HIV are potential drugs that may be able to prevent HIV infection in humans. However, the long-term protective capacity of antibodies against HIV has not been assessed. Here, we repetitively administered a macaque version of a human anti-HIV antibody to monkeys, after which the antibody persisted in the blood for >5 months. Moreover, the antibody could be sustained at protective levels for 108 days, conferring protection 52 days after the last dose in a monkey model of HIV infection. Thus, passive antibody transfer can provide durable protection against infection by viruses that cause AIDS in primates.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Immunization, Passive , Macaca , Simian Acquired Immunodeficiency Syndrome/virology , Treatment Outcome
10.
J Virol ; 88(21): 12669-82, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142607

ABSTRACT

UNLABELLED: Over the past 5 years, a new generation of highly potent and broadly neutralizing HIV-1 antibodies has been identified. These antibodies can protect against lentiviral infection in nonhuman primates (NHPs), suggesting that passive antibody transfer would prevent HIV-1 transmission in humans. To increase the protective efficacy of such monoclonal antibodies, we employed next-generation sequencing, computational bioinformatics, and structure-guided design to enhance the neutralization potency and breadth of VRC01, an antibody that targets the CD4 binding site of the HIV-1 envelope. One variant, VRC07-523, was 5- to 8-fold more potent than VRC01, neutralized 96% of viruses tested, and displayed minimal autoreactivity. To compare its protective efficacy to that of VRC01 in vivo, we performed a series of simian-human immunodeficiency virus (SHIV) challenge experiments in nonhuman primates and calculated the doses of VRC07-523 and VRC01 that provide 50% protection (EC50). VRC07-523 prevented infection in NHPs at a 5-fold lower concentration than VRC01. These results suggest that increased neutralization potency in vitro correlates with improved protection against infection in vivo, documenting the improved functional efficacy of VRC07-523 and its potential clinical relevance for protecting against HIV-1 infection in humans. IMPORTANCE: In the absence of an effective HIV-1 vaccine, alternative strategies are needed to block HIV-1 transmission. Direct administration of HIV-1-neutralizing antibodies may be able to prevent HIV-1 infections in humans. This approach could be especially useful in individuals at high risk for contracting HIV-1 and could be used together with antiretroviral drugs to prevent infection. To optimize the chance of success, such antibodies can be modified to improve their potency, breadth, and in vivo half-life. Here, knowledge of the structure of a potent neutralizing antibody, VRC01, that targets the CD4-binding site of the HIV-1 envelope protein was used to engineer a next-generation antibody with 5- to 8-fold increased potency in vitro. When administered to nonhuman primates, this antibody conferred protection at a 5-fold lower concentration than the original antibody. Our studies demonstrate an important correlation between in vitro assays used to evaluate the therapeutic potential of antibodies and their in vivo effectiveness.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV-1/immunology , Immunization, Passive/methods , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , HIV Antibodies/administration & dosage , HIV Antibodies/genetics , HIV-1/genetics , Macaca mulatta , Male , Molecular Sequence Data , Sequence Analysis, DNA
11.
Nature ; 514(7524): 642-5, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25119033

ABSTRACT

To protect against human immunodeficiency virus (HIV-1) infection, broadly neutralizing antibodies (bnAbs) must be active at the portals of viral entry in the gastrointestinal or cervicovaginal tracts. The localization and persistence of antibodies at these sites is influenced by the neonatal Fc receptor (FcRn), whose role in protecting against infection in vivo has not been defined. Here, we show that a bnAb with enhanced FcRn binding has increased gut mucosal tissue localization, which improves protection against lentiviral infection in non-human primates. A bnAb directed to the CD4-binding site of the HIV-1 envelope (Env) protein (denoted VRC01) was modified by site-directed mutagenesis to increase its binding affinity for FcRn. This enhanced FcRn-binding mutant bnAb, denoted VRC01-LS, displayed increased transcytosis across human FcRn-expressing cellular monolayers in vitro while retaining FcγRIIIa binding and function, including antibody-dependent cell-mediated cytotoxicity (ADCC) activity, at levels similar to VRC01 (the wild type). VRC01-LS had a threefold longer serum half-life than VRC01 in non-human primates and persisted in the rectal mucosa even when it was no longer detectable in the serum. Notably, VRC01-LS mediated protection superior to that afforded by VRC01 against intrarectal infection with simian-human immunodeficiency virus (SHIV). These findings suggest that modification of FcRn binding provides a mechanism not only to increase serum half-life but also to enhance mucosal localization that confers immune protection. Mutations that enhance FcRn function could therefore increase the potency and durability of passive immunization strategies to prevent HIV-1 infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Histocompatibility Antigens Class I/immunology , Receptors, Fc/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Administration, Rectal , Animals , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/genetics , Antibodies, Viral/analysis , Antibodies, Viral/blood , Antibodies, Viral/genetics , Antibody Affinity/genetics , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites/genetics , CD4 Antigens/metabolism , Female , HIV/chemistry , HIV/immunology , HIV Antibodies/analysis , HIV Antibodies/blood , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/immunology , Half-Life , Immunity, Mucosal/immunology , Immunization, Passive , Intestinal Mucosa/immunology , Macaca mulatta , Male , Mice , Mutagenesis, Site-Directed , Receptors, IgG/immunology , Receptors, IgG/metabolism , Rectum/immunology , Simian Immunodeficiency Virus/immunology , Transcytosis
12.
J Immunol ; 193(4): 1799-811, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25024382

ABSTRACT

Development of a vaccine against pulmonary tuberculosis may require immunization strategies that induce a high frequency of Ag-specific CD4 and CD8 T cells in the lung. The nonhuman primate model is essential for testing such approaches because it has predictive value for how vaccines elicit responses in humans. In this study, we used an aerosol vaccination strategy to administer AERAS-402, a replication-defective recombinant adenovirus (rAd) type 35 expressing Mycobacterium tuberculosis Ags Ag85A, Ag85B, and TB10.4, in bacillus Calmette-Guérin (BCG)-primed or unprimed rhesus macaques. Immunization with BCG generated low purified protein derivative-specific CD4 T cell responses in blood and bronchoalveolar lavage. In contrast, aerosolized AERAS-402 alone or following BCG induced potent and stable Ag85A/b-specific CD4 and CD8 effector T cells in bronchoalveolar lavage that largely produced IFN-γ, as well as TNF and IL-2. Such responses induced by BCG, AERAS-402, or both failed to confer overall protection following challenge with 275 CFUs M. tuberculosis Erdman, although vaccine-induced responses associated with reduced pathology were observed in some animals. Anamnestic T cell responses to Ag85A/b were not detected in blood of immunized animals after challenge. Overall, our data suggest that a high M. tuberculosis challenge dose may be a critical factor in limiting vaccine efficacy in this model. However, the ability of aerosol rAd immunization to generate potent cellular immunity in the lung suggests that using different or more immunogens, alternative rAd serotypes with enhanced immunogenicity, and a physiological challenge dose may achieve protection against M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Vaccines, Synthetic/immunology , Acyltransferases/immunology , Administration, Inhalation , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lung/immunology , Lung/microbiology , Macaca mulatta , Male , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/virology , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccines, DNA , Vaccines, Synthetic/administration & dosage
13.
Sci Transl Med ; 6(243): 243ra88, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24990883

ABSTRACT

HIV-1 infection depends on effective viral entry mediated by the interaction of its envelope (Env) glycoprotein with specific cell surface receptors. Protective antiviral antibodies generated by passive or active immunization must prevent these interactions. Because the HIV-1 Env is highly variable, attention has also focused on blocking the HIV-1 primary cell receptor CD4. We therefore analyzed the in vivo protective efficacy of three potent neutralizing monoclonal antibodies (mAbs) to HIV-1 Env compared to an antibody against the CD4 receptor. Protection was assessed after mucosal challenge of rhesus macaques with simian/HIV (SHIV). Despite its comparable or greater neutralization potency in vitro, the anti-CD4 antibody did not provide effective protection in vivo, whereas the HIV-1-specific mAbs VRC01, 10E8, and PG9, targeting the CD4 binding site, membrane-proximal, and V1V2 glycan Env regions, respectively, conferred complete protection, albeit at different relative potencies. These findings demonstrate the protective efficacy of broadly neutralizing antibodies directed to the HIV-1 Env and suggest that targeting the HIV-1 Env is preferable to the cell surface receptor CD4 for the prevention of HIV-1 transmission.


Subject(s)
Antibodies, Neutralizing/therapeutic use , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/immunology , CD4 Antigens/immunology , Female , HIV Antibodies/immunology , HIV Antibodies/therapeutic use , Macaca mulatta , Male
14.
Nature ; 505(7484): 502-8, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24352234

ABSTRACT

A major challenge for the development of a highly effective AIDS vaccine is the identification of mechanisms of protective immunity. To address this question, we used a nonhuman primate challenge model with simian immunodeficiency virus (SIV). We show that antibodies to the SIV envelope are necessary and sufficient to prevent infection. Moreover, sequencing of viruses from breakthrough infections revealed selective pressure against neutralization-sensitive viruses; we identified a two-amino-acid signature that alters antigenicity and confers neutralization resistance. A similar signature confers resistance of human immunodeficiency virus (HIV)-1 to neutralization by monoclonal antibodies against variable regions 1 and 2 (V1V2), suggesting that SIV and HIV share a fundamental mechanism of immune escape from vaccine-elicited or naturally elicited antibodies. These analyses provide insight into the limited efficacy seen in HIV vaccine trials.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Disease Susceptibility/immunology , Female , Founder Effect , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/chemistry , Humans , Immune Evasion/immunology , Macaca mulatta , Male , Molecular Sequence Data , Phylogeny , Risk , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , env Gene Products, Human Immunodeficiency Virus/immunology
15.
Vaccine ; 31(49): 5879-88, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24099872

ABSTRACT

Nontyphoidal Salmonella (NTS) serovars are a common cause of acute food-borne gastroenteritis worldwide and can cause invasive systemic disease in young infants, the elderly, and immunocompromised hosts, accompanied by high case fatality. Vaccination against invasive NTS disease is warranted where the disease incidence and mortality are high and multidrug resistance is prevalent, as in sub-Saharan Africa. Live-attenuated vaccines that mimic natural infection constitute one strategy to elicit protection. However, they must particularly be shown to be adequately attenuated for consideration of immunocompromised subjects. Accordingly, we examined the safety and tolerability of an oral live attenuated Salmonella typhimurium vaccine candidate, CVD 1921, in an established chronic simian immunodeficiency virus (SIV)-infected rhesus macaque model. We evaluated clinical parameters, histopathology, and measured differences in mucosal permeability to wild-type and vaccine strains. Compared to the wild-type S. typhimurium strain I77 in both SIV-infected and SIV-uninfected nonhuman primate hosts, this live-attenuated vaccine shows reduced shedding and systemic spread, exhibits limited pathological disease manifestations in the digestive tract, and induces low levels of cellular infiltration in tissues. Furthermore, wild-type S. typhimurium induces increased intestinal epithelial damage and permeability, with infiltration of neutrophils and macrophages in both SIV-infected and SIV-uninfected nonhuman primates compared to the vaccine strain. Based on shedding, systemic spread, and histopathology, the live-attenuated S. typhimurium strain CVD 1921 appears to be safe and well-tolerated in the nonhuman primate model, including chronically SIV-infected rhesus macaques.


Subject(s)
Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Simian Acquired Immunodeficiency Syndrome/microbiology , Animals , Bacterial Shedding , Coinfection , Disease Models, Animal , Feces/microbiology , Foodborne Diseases/microbiology , Foodborne Diseases/prevention & control , Immunity, Humoral , Immunocompromised Host , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors/blood , Lipopolysaccharides/blood , Macaca mulatta , Salmonella Infections, Animal/complications , Salmonella Infections, Animal/immunology , Vaccines, Attenuated/immunology
16.
J Virol ; 87(17): 9836-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824823

ABSTRACT

Programmed Death 1 (PD-1) expression by human/simian immunodeficiency virus (HIV/SIV)-specific CD8 T cells has been associated with defective cytokine production and reduced in vitro proliferation capacity. However, the cellular mechanisms that sustain PD-1(high) virus-specific CD8 T cell responses during chronic infection are unknown. Here, we show that the PD-1(high) phenotype is associated with accelerated in vivo CD8 T cell turnover in SIV-infected rhesus macaques, especially within the SIV-specific CD8 T cell pool. Mathematical modeling of 5-bromo-2' deoxyuridine (BrdU) labeling dynamics demonstrated a significantly increased generation rate of PD-1(high) compared to PD-1(low) CD8 T cells in all memory compartments. Simultaneous analysis of Ki67 and BrdU kinetics revealed a complex in vivo turnover profile whereby only a small fraction of PD-1(high) cells, but virtually all PD-1(low) cells, returned to rest after activation. Similar kinetics operated in both chronic and acute SIV infection. Our data suggest that the persistence of PD-1(high) SIV-specific CD8 T cells in chronic infection is maintained in vivo by a mechanism involving high production coupled with a high disappearance rate.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Programmed Cell Death 1 Receptor/metabolism , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Acute Disease , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Chronic Disease , Cytokines/biosynthesis , Immunologic Memory , Lymphocyte Activation , Macaca mulatta , Resting Phase, Cell Cycle , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology
17.
Nature ; 499(7456): 102-6, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23698367

ABSTRACT

Influenza viruses pose a significant threat to the public and are a burden on global health systems. Each year, influenza vaccines must be rapidly produced to match circulating viruses, a process constrained by dated technology and vulnerable to unexpected strains emerging from humans and animal reservoirs. Here we use knowledge of protein structure to design self-assembling nanoparticles that elicit broader and more potent immunity than traditional influenza vaccines. The viral haemagglutinin was genetically fused to ferritin, a protein that naturally forms nanoparticles composed of 24 identical polypeptides. Haemagglutinin was inserted at the interface of adjacent subunits so that it spontaneously assembled and generated eight trimeric viral spikes on its surface. Immunization with this influenza nanoparticle vaccine elicited haemagglutination inhibition antibody titres more than tenfold higher than those from the licensed inactivated vaccine. Furthermore, it elicited neutralizing antibodies to two highly conserved vulnerable haemagglutinin structures that are targets of universal vaccines: the stem and the receptor binding site on the head. Antibodies elicited by a 1999 haemagglutinin-nanoparticle vaccine neutralized H1N1 viruses from 1934 to 2007 and protected ferrets from an unmatched 2007 H1N1 virus challenge. This structure-based, self-assembling synthetic nanoparticle vaccine improves the potency and breadth of influenza virus immunity, and it provides a foundation for building broader vaccine protection against emerging influenza viruses and other pathogens.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Nanoparticles/chemistry , Animals , Binding Sites , Cross Reactions/immunology , Female , Ferrets/immunology , Ferrets/virology , Ferritins/chemistry , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/classification , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Vaccines, Inactivated/immunology
18.
J Clin Invest ; 122(9): 3281-94, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22922258

ABSTRACT

CD4 T follicular helper (TFH) cells interact with and stimulate the generation of antigen-specific B cells. TFH cell interaction with B cells correlates with production of SIV-specific immunoglobulins. However, the fate of TFH cells and their participation in SIV-induced antibody production is not well understood. We investigated the phenotype, function, location, and molecular signature of TFH cells in rhesus macaques. Similar to their human counterparts, TFH cells in rhesus macaques represented a heterogeneous population with respect to cytokine function. In a highly differentiated subpopulation of TFH cells, characterized by CD150lo expression, production of Th1 cytokines was compromised while IL-4 production was augmented, and cells exhibited decreased survival, cycling, and trafficking capacity. TFH cells exhibited a distinct gene profile that was markedly altered by SIV infection. TFH cells were infected by SIV; yet, in some animals, these cells actually accumulated during chronic SIV infection. Generalized immune activation and increased IL-6 production helped drive TFH differentiation during SIV infection. Accumulation of TFH cells was associated with increased frequency of activated germinal center B cells and SIV-specific antibodies. Therefore, chronic SIV does not disturb the ability of TFH cells to help B cell maturation and production of SIV-specific immunoglobulins.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4 Antigens/metabolism , Cell Death , Cell Proliferation , Cytokines/metabolism , Gene Expression , Germinal Center/pathology , Germinal Center/virology , Host-Pathogen Interactions , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymph Nodes/virology , Macaca mulatta , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/physiology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
19.
J Virol ; 86(13): 7052-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532675

ABSTRACT

A major goal of AIDS vaccine development is to design vaccination strategies that can elicit broad and potent protective antibodies. The initial viral targets of neutralizing antibodies (NAbs) early after human or simian immunodeficiency virus (HIV/SIV) infection are not known. The identification of early NAb epitopes that induce protective immunity or retard the progression of disease is important for AIDS vaccine development. The aim of this study was to determine the Env residues targeted by early SIV NAbs and to assess the influence of prior vaccination on neutralizing antibody kinetics and specificity during early infection. We previously described stereotypic env sequence variations in SIVmac251-infected rhesus monkeys that resulted in viral escape from NAbs. Here, we defined the early viral targets of neutralization and determined whether the ability of serum antibody from infected monkeys to neutralize SIV was altered in the setting of prior vaccination. To localize the viral determinants recognized by early NAbs, a panel of mutant pseudoviruses was assessed in a TZM-bl reporter gene neutralization assay to define the precise changes that eliminate recognition by SIV Env-specific NAbs in 16 rhesus monkeys. Changing R420 to G or R424 to Q in V4 of Env resulted in the loss of recognition by NAbs in vaccinated monkeys. In contrast, mutations in the V1 region of Env did not alter the NAb profile. These findings indicate that early NAbs are directed toward SIVmac251 Env V4 but not the V1 region, and that this env vaccination regimen did not alter the kinetics or the breadth of NAbs during early infection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Epitopes, B-Lymphocyte/immunology , Gene Products, env/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Epitopes, B-Lymphocyte/genetics , Gene Products, env/genetics , Macaca mulatta , Male , Mutant Proteins/genetics , Mutant Proteins/immunology , Neutralization Tests , Simian Immunodeficiency Virus/genetics
20.
Virology ; 428(2): 121-7, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22521913

ABSTRACT

To create an HIV-1 vaccine that generates sufficient breadth of immune recognition to protect against the genetically diverse forms of the circulating virus, we have been exploring vaccines based on consensus and mosaic protein designs. Increasing the valency of a mosaic immunogen cocktail increases epitope coverage but with diminishing returns, as increasingly rare epitopes are incorporated into the mosaic proteins. In this study we compared the immunogenicity of 2-valent and 3-valent HIV-1 envelope mosaic immunogens in rhesus monkeys. Immunizations with the 3-valent mosaic immunogens resulted in a modest increase in the breadth of vaccine-elicited T lymphocyte responses compared to the 2-valent mosaic immunogens. However, the 3-valent mosaic immunogens elicited significantly higher neutralizing responses to Tier 1 viruses than the 2-valent mosaic immunogens. These findings underscore the potential utility of polyvalent mosaic immunogens for eliciting both cellular and humoral immune responses to HIV-1.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV-1/immunology , Immunity, Cellular , Immunity, Humoral , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , Animals , Disease Models, Animal , Epitopes/administration & dosage , Epitopes/genetics , Epitopes/immunology , HIV Infections/virology , HIV-1/genetics , Macaca mulatta , env Gene Products, Human Immunodeficiency Virus/administration & dosage , env Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...