Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
NPJ Vaccines ; 6(1): 23, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33558494

ABSTRACT

Nipah and Hendra viruses are highly pathogenic bat-borne paramyxoviruses recently included in the WHO Blueprint priority diseases list. A fully registered horse anti-Hendra virus subunit vaccine has been in use in Australia since 2012. Based on the same immunogen, the Hendra virus attachment glycoprotein ectodomain, a subunit vaccine formulation for use in people is now in a Phase I clinical trial. We report that a single dose vaccination regimen of this human vaccine formulation protects against otherwise lethal challenges of either Hendra or Nipah virus in a nonhuman primate model. The protection against the Nipah Bangladesh strain begins as soon as 7 days post immunization with low dose of 0.1 mg protein subunit. Our data suggest this human vaccine could be utilized as efficient emergency vaccine to disrupt potential spreading of Nipah disease in an outbreak setting.

2.
Mol Syst Biol ; 15(5): e8747, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31048360

ABSTRACT

Characterizing the antigen-binding and innate immune-recruiting properties of the humoral response offers the chance to obtain deeper insights into mechanisms of protection than revealed by measuring only overall antibody titer. Here, a high-throughput, multiplexed Fab-Fc Array was employed to profile rhesus macaques vaccinated with a gp120-CD4 fusion protein in combination with different genetically encoded adjuvants, and subsequently subjected to multiple heterologous simian immunodeficiency virus (SIV) challenges. Systems analyses modeling protection and adjuvant differences using Fab-Fc Array measurements revealed a set of correlates yielding strong and robust predictive performance, while models based on measurements of response magnitude alone exhibited significantly inferior performance. At the same time, rendering Fab-Fc measurements mathematically independent of titer had relatively little impact on predictive performance. Similar analyses for a distinct SIV vaccine study also showed that Fab-Fc measurements performed significantly better than titer. These results suggest that predictive modeling with measurements of antibody properties can provide detailed correlates with robust predictive power, suggest directions for vaccine improvement, and potentially enable discovery of mechanistic associations.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin Fragments/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Macaca mulatta , Membrane Glycoproteins/immunology , Multivariate Analysis , Viral Envelope Proteins/immunology
3.
Viral Immunol ; 30(3): 178-195, 2017 04.
Article in English | MEDLINE | ID: mdl-28085634

ABSTRACT

Vaccination is a proven intervention against human viral diseases; however, success against Herpes Simplex Virus 2 (HSV-2) remains elusive. Most HSV-2 vaccines tested in humans to date contained just one or two immunogens, such as the virion attachment receptor glycoprotein D (gD) and/or the envelope fusion protein, glycoprotein B (gB). At least three factors may have contributed to the failures of subunit-based HSV-2 vaccines. First, immune responses directed against one or two viral antigens may lack sufficient antigenic breadth for efficacy. Second, the antibody responses elicited by these vaccines may have lacked necessary Fc-mediated effector functions. Third, these subunit vaccines may not have generated necessary protective cellular immune responses. We hypothesized that a polyvalent combination of HSV-2 antigens expressed from a DNA vaccine with an adjuvant that polarizes immune responses toward a T helper 1 (Th1) phenotype would compose a more effective vaccine. We demonstrate that delivery of DNA expressing full-length HSV-2 glycoprotein immunogens by electroporation with the adjuvant interleukin 12 (IL-12) generates substantially greater protection against a high-dose HSV-2 vaginal challenge than a recombinant gD subunit vaccine adjuvanted with alum and monophosphoryl lipid A (MPL). Our results further show that DNA vaccines targeting optimal combinations of surface glycoproteins provide better protection than gD alone and provide similar survival benefits and disease symptom reductions compared with a potent live attenuated HSV-2 0ΔNLS vaccine, but that mice vaccinated with HSV-2 0ΔNLS clear the virus much faster. Together, our data indicate that adjuvanted multivalent DNA vaccines hold promise for an effective HSV-2 vaccine, but that further improvements may be required.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/immunology , Herpesvirus Vaccines/immunology , Interleukin-12/administration & dosage , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Glycoproteins/immunology , Herpesvirus Vaccines/administration & dosage , Membrane Proteins/immunology , Mice , Survival Analysis , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
4.
J Control Release ; 244(Pt A): 98-107, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27847326

ABSTRACT

For nearly a century, aluminum salts have been the most widely used vaccine adjuvant formulation, and have thus established a history of safety and efficacy. Nevertheless, for extremely challenging disease targets such as tuberculosis or HIV, the adjuvant activity of aluminum salts may not be potent enough to achieve protective efficacy. Adsorption of TLR ligands to aluminum salts facilitates enhanced adjuvant activity, such as in the human papilloma virus vaccine Cervarix®. However, some TLR ligands such as TLR7/8 agonist imidazoquinolines do not efficiently adsorb to aluminum salts. The present report describes a formulation approach to solving this challenge by developing a lipid-based nanosuspension of a synthetic TLR7/8 ligand (3M-052) that facilitates adsorption to aluminum oxyhydroxide via the structural properties of the helper lipid employed. In immunized mice, the aluminum oxyhydroxide-adsorbed formulation of 3M-052 enhanced antibody and TH1-type cellular immune responses to vaccine antigens for tuberculosis and HIV.


Subject(s)
Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Aluminum Oxide/chemistry , Imidazoles/chemistry , Nanoparticles/chemistry , Quinolines/chemistry , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , AIDS Vaccines/immunology , Adsorption , Animals , Drug Stability , Humans , Imidazoles/immunology , Immunity, Cellular , Immunity, Humoral , Ligands , Lipids/chemistry , Mice , Mice, Inbred C57BL , Particle Size , Quinolines/immunology , Surface Properties , Tuberculosis Vaccines/immunology
5.
Clin Vaccine Immunol ; 23(7): 618-27, 2016 07.
Article in English | MEDLINE | ID: mdl-27193040

ABSTRACT

A promising concept for human immunodeficiency virus (HIV) vaccines focuses immunity on the highly conserved transition state structures and epitopes that appear when the HIV glycoprotein gp120 binds to its receptor, CD4. We are developing chimeric antigens (full-length single chain, or FLSC) in which gp120 and CD4 sequences are flexibly linked to allow stable intrachain complex formation between the two moieties (A. DeVico et al., Proc Natl Acad Sci U S A 104:17477-17482, 2007, doi:10.1073/pnas.0707399104; T. R. Fouts et al., J Virol 74:11427-11436, 2000, doi:10.1128/JVI.74.24.11427-11436.2000). Proof of concept studies with nonhuman primates show that FLSC elicited heterologous protection against simian-human immunodeficiency virus (SHIV)/simian immunodeficiency virus (SIV) (T. R. Fouts et al., Proc Natl Acad Sci U S A 112:E992-E999, 2016, doi:10.1073/pnas.1423669112), which correlated with antibodies against transition state gp120 epitopes. Nevertheless, advancement of any vaccine that comprises gp120-CD4 complexes must consider whether the CD4 component breaks tolerance and becomes immunogenic in the autologous host. To address this, we performed an immunotoxicology study with cynomolgus macaques vaccinated with either FLSC or a rhesus variant of FLSC containing macaque CD4 sequences (rhFLSC). Enzyme-linked immunosorbent assay (ELISA) binding titers, primary CD3(+) T cell staining, and temporal trends in T cell subset frequencies served to assess whether anti-CD4 autoantibody responses were elicited by vaccination. We find that immunization with multiple high doses of rhFLSC did not elicit detectable antibody titers despite robust responses to rhFLSC. In accordance with these findings, immunized animals had no changes in circulating CD4(+) T cell counts or evidence of autoantibody reactivity with cell surface CD4 on primary naive macaque T cells. Collectively, these studies show that antigens using CD4 sequences to stabilize transition state gp120 structures are unlikely to elicit autoimmune antibody responses, supporting the advancement of gp120-CD4 complex-based antigens, such as FLSC, into clinical testing.


Subject(s)
Autoantibodies/blood , CD4 Antigens/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , Recombinant Proteins/immunology , Animals , CD4 Antigens/genetics , CD4 Lymphocyte Count , Enzyme-Linked Immunosorbent Assay , Female , HIV Envelope Protein gp120/genetics , Macaca fascicularis , Male , Recombinant Proteins/genetics , T-Lymphocyte Subsets/immunology
6.
Structure ; 24(5): 697-709, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27041594

ABSTRACT

Evidence supports a role of antibody-dependent cellular cytotoxicity (ADCC) toward transitional epitopes in the first and second constant (C1-C2) regions of gp120 (A32-like epitopes) in preventing HIV-1 infection and in vaccine-induced protection. Here, we describe the first successful attempt at isolating the inner domain (ID) of gp120 as an independent molecule that encapsulates the A32-like region within a minimal structural unit of the HIV-1 Env. Through structure-based design, we developed ID2, which consists of the ID expressed independently of the outer domain and stabilized in the CD4-bound conformation by an inter-layer disulfide bond. ID2 expresses C1-C2 epitopes in the context of CD4-triggered full-length gp120 but without any known neutralizing epitope present. Thus, ID2 represents a novel probe for the analysis and/or selective induction of antibody responses to the A32 epitope region. We also present the crystal structure of ID2 complexed with mAb A32, which defines its epitope.


Subject(s)
Antigen-Antibody Complex/chemistry , HIV Envelope Protein gp120/chemistry , Molecular Docking Simulation , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Binding Sites, Antibody , CD4 Antigens/chemistry , CD4 Antigens/immunology , Epitopes/chemistry , Epitopes/immunology , HIV Envelope Protein gp120/immunology
7.
AIDS Res Hum Retroviruses ; 32(10-11): 964-971, 2016.
Article in English | MEDLINE | ID: mdl-26950606

ABSTRACT

Eradication of human immunodeficiency virus type 1 (HIV-1) by vaccination with epitopes that produce broadly neutralizing antibodies is the ultimate goal for HIV prevention. However, generating appropriate immune responses has proven difficult. Expression of broadly neutralizing antibodies by vaginal colonizing lactobacilli provides an approach to passively target these antibodies to the mucosa. We tested the feasibility of expressing single-chain and single-domain antibodies (dAbs) in Lactobacillus to be used as a topical microbicide/live biotherapeutic. Lactobacilli provide an excellent platform to express anti-HIV proteins. Broadly neutralizing antibodies have been identified against epitopes on the HIV-1 envelope and have been made into active antibody fragments. We tested single-chain variable fragment m9 and dAb-m36 and its derivative m36.4 as prototype antibodies. We cloned and expressed the antibody fragments m9, m36, and m36.4 in Lactobacillus jensenii-1153 and tested the expression levels and functionality. We made a recombinant L. jensenii 1153-1128 that expresses dAb-m36.4. All antibody fragments m9, m36, and m36.4 were expressed by lactobacilli. However, we noted the smaller m36/m36.4 were expressed to higher levels, ≥3 µg/ml. All L. jensenii-expressed antibody fragments bound to gp120/CD4 complex; Lactobacillus-produced m36.4 inhibited HIV-1BaL in a neutralization assay. Using a TZM-bl assay, we characterized the breadth of neutralization of the m36.4. Delivery of dAbs by Lactobacillus could provide passive transfer of these antibodies to the mucosa and longevity at the site of HIV-1 transmission.


Subject(s)
Antibodies, Neutralizing/immunology , Gene Expression , HIV Antibodies/immunology , Lactobacillus/metabolism , Recombinant Proteins/immunology , Antibodies, Neutralizing/genetics , Disease Transmission, Infectious/prevention & control , Female , HIV Antibodies/genetics , HIV Infections/prevention & control , HIV-1 , Humans , Lactobacillus/genetics , Recombinant Proteins/genetics , Vagina/immunology , Vagina/microbiology
8.
Curr HIV Res ; 14(1): 24-36, 2016.
Article in English | MEDLINE | ID: mdl-26354735

ABSTRACT

BACKGROUND: Antiretroviral (ARV) drugs targeting retroviral enzymes have been extensively employed to treat HIV-1 infection. Drawbacks of this approach include cost, toxicity, and the eventual emergence of resistant strains that threaten prophylactic and/or therapeutic efficacy. Accordingly, efforts to develop next-generation ARV approaches are warranted, particularly if they can offer a higher threshold of resistance. We have previously shown that FLSC, a fusion protein containing gp120(BAL) and the D1 and D2 domains of human CD4, specifically binds CCR5, an important cellular co-receptor, and inhibits the entry of R5 HIV isolates. (FLSC) IgG1, a fusion of FLSC and the hinge-C(H)2-C(H)3 region of human IgG1, has an increased antiviral activity, likely due to the resultant bivalency. METHODS: In this study, we show CCR5 reduction upon (FLSC) IgG1 treatment both by standard flow cytometry and visualized using a novel nanoparticle method. A ß-lactamase virus-cell fusion assay was used to quantify (FLSC) IgG1 inhibition of HIV-1 entry into both cell lines and primary cells. Synergistic anti-viral activities of (FLSC) IgG1 and MVC in primary cells were evaluated by measuring supernatant p24 levels via ELISA and calculated using the MacSynergy™ II program. RESULTS: We previously reported that treatment with the CCR5 small molecule antagonist Maraviroc (MVC) increased the apparent exposure of the (FLSC) IgG1 binding sites on CCR5, leading us to wonder if the two compounds used in combination might synergize in their anti-viral activity. Here we show that this is indeed the case. We demonstrate that fusion protein (FLSC) IgG1, strongly synergizes with the CCR5 antagonist Maraviroc to successfully inhibit both MVC-sensitive and MVC-resistant R5 HIV-1. CONCLUSION: Observed synergy between (FLSC) IgG1 and MVC was high in both, cell lines and primary PBMCs. This has relevance for future in vivo studies. In addition, synergy occurred both with MVC-sensitive viruses and MVC-resistant viruses, partially restoring the inhibitory effect of MVC. These findings suggest that a combinatorial treatment based on these two compounds has potential merit and that future in vivo studies are warranted.


Subject(s)
Anti-HIV Agents/therapeutic use , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Immunoglobulin G/pharmacology , Receptors, CCR5/drug effects , Recombinant Fusion Proteins/pharmacology , Triazoles/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Cell Line , Cyclohexanes/therapeutic use , Drug Synergism , Flow Cytometry , HIV Infections/metabolism , Humans , Immunoglobulin G/genetics , Maraviroc , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Recombinant Fusion Proteins/metabolism , Triazoles/therapeutic use , Virus Internalization/drug effects
9.
Cell Rep ; 11(1): 98-110, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25843712

ABSTRACT

Although oncogene-targeted therapy often elicits profound initial tumor responses in patients, responses are generally incomplete because some tumor cells survive initial therapy as residual disease that enables eventual acquired resistance. The mechanisms underlying tumor cell adaptation and survival during initial therapy are incompletely understood. Here, through the study of EGFR mutant lung adenocarcinoma, we show that NF-κB signaling is rapidly engaged upon initial EGFR inhibitor treatment to promote tumor cell survival and residual disease. EGFR oncogene inhibition induced an EGFR-TRAF2-RIP1-IKK complex that stimulated an NF-κB-mediated transcriptional survival program. The direct NF-κB inhibitor PBS-1086 suppressed this adaptive survival program and increased the magnitude and duration of initial EGFR inhibitor response in multiple NSCLC models, including a patient-derived xenograft. These findings unveil NF-κB activation as a critical adaptive survival mechanism engaged by EGFR oncogene inhibition and provide rationale for EGFR and NF-κB co-inhibition to eliminate residual disease and enhance patient responses.


Subject(s)
Adenocarcinoma/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , NF-kappa B/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclohexanones/administration & dosage , Epoxy Compounds/administration & dosage , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Molecular Targeted Therapy , NF-kappa B/antagonists & inhibitors , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism
10.
Proc Natl Acad Sci U S A ; 112(9): E992-9, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25681373

ABSTRACT

A guiding principle for HIV vaccine design has been that cellular and humoral immunity work together to provide the strongest degree of efficacy. However, three efficacy trials of Ad5-vectored HIV vaccines showed no protection. Transmission was increased in two of the trials, suggesting that this vaccine strategy elicited CD4+ T-cell responses that provide more targets for infection, attenuating protection or increasing transmission. The degree to which this problem extends to other HIV vaccine candidates is not known. Here, we show that a gp120-CD4 chimeric subunit protein vaccine (full-length single chain) elicits heterologous protection against simian-human immunodeficiency virus (SHIV) or simian immunodeficiency virus (SIV) acquisition in three independent rhesus macaque repeated low-dose rectal challenge studies with SHIV162P3 or SIVmac251. Protection against acquisition was observed with multiple formulations and challenges. In each study, protection correlated with antibody-dependent cellular cytotoxicity specific for CD4-induced epitopes, provided that the concurrent antivaccine T-cell responses were minimal. Protection was lost in instances when T-cell responses were high or when the requisite antibody titers had declined. Our studies suggest that balance between a protective antibody response and antigen-specific T-cell activation is the critical element to vaccine-mediated protection against HIV. Achieving and sustaining such a balance, while enhancing antibody durability, is the major challenge for HIV vaccine development, regardless of the immunogen or vaccine formulation.


Subject(s)
AIDS Vaccines/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Antibodies/immunology , HIV Infections/prevention & control , Immunity, Cellular/drug effects , AIDS Vaccines/immunology , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4 Antigens/pharmacology , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/pharmacology , HIV Infections/immunology , HIV Infections/pathology , Humans , Immunity, Humoral , Macaca mulatta , Male , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology
11.
Proc Natl Acad Sci U S A ; 110(1): E69-78, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23237851

ABSTRACT

The HIV-1 envelope glycoprotein (Env) undergoes conformational transitions consequent to CD4 binding and coreceptor engagement during viral entry. The physical steps in this process are becoming defined, but less is known about their significance as targets of antibodies potentially protective against HIV-1 infection. Here we probe the functional significance of transitional epitope exposure by characterizing 41 human mAbs specific for epitopes exposed on trimeric Env after CD4 engagement. These mAbs recognize three epitope clusters: cluster A, the gp120 face occluded by gp41 in trimeric Env; cluster B, a region proximal to the coreceptor-binding site (CoRBS) and involving the V1/V2 domain; and cluster C, the coreceptor-binding site. The mAbs were evaluated functionally by antibody-dependent, cell-mediated cytotoxicity (ADCC) and for neutralization of Tiers 1 and 2 pseudoviruses. All three clusters included mAbs mediating ADCC. However, there was a strong potency bias for cluster A, which harbors at least three potent ADCC epitopes whose cognate mAbs have electropositive paratopes. Cluster A epitopes are functional ADCC targets during viral entry in an assay format using virion-sensitized target cells. In contrast, only cluster C contained epitopes that were recognized by neutralizing mAbs. There was significant diversity in breadth and potency that correlated with epitope fine specificity. In contrast, ADCC potency had no relationship with neutralization potency or breadth for any epitope cluster. Thus, Fc-mediated effector function and neutralization coselect with specificity in anti-Env antibody responses, but the nature of selection is distinct for these two antiviral activities.


Subject(s)
Antibodies, Monoclonal/immunology , CD4 Antigens/metabolism , Epitopes/metabolism , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Models, Molecular , Antibodies, Monoclonal/chemistry , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites/genetics , CD4 Antigens/chemistry , CD4 Antigens/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , HIV Antibodies/chemistry , HIV Envelope Protein gp120/genetics , Humans , Neutralization Tests , Protein Conformation
12.
J Virol ; 85(24): 13097-104, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994452

ABSTRACT

Certain antibodies from HIV-infected humans bind conserved transition state (CD4 induced [CD4i]) domains on the HIV envelope glycoprotein, gp120, and demonstrate extreme dependence on the formation of a gp120-human CD4 receptor complex. The epitopes recognized by these antibodies remain undefined although recent crystallographic studies of the anti-CD4i monoclonal antibody (MAb) 21c suggest that contacts with CD4 as well as gp120 might occur. Here, we explore the possibility of hybrid epitopes that demand the collaboration of both gp120 and CD4 residues to enable antibody reactivity. Analyses with a panel of human anti-CD4i MAbs and gp120-CD4 antigens with specific mutations in predicted binding domains revealed one putative hybrid epitope, defined by the human anti-CD4i MAb 19e. In virological and immunological tests, MAb 19e did not bind native or constrained gp120 except in the presence of CD4. This contrasted with other anti-CD4i MAbs, including MAb 21c, which bound unliganded, full-length gp120 held in a constrained conformation. Conversely, MAb 19e exhibited no specific reactivity with free human CD4. Computational modeling of MAb 19e interactions with gp120-CD4 complexes suggested a distinct binding profile involving antibody heavy chain interactions with CD4 and light chain interactions with gp120. In accordance, targeted mutations in CD4 based on this model specifically reduced MAb 19e interactions with stable gp120-CD4 complexes that retained reactivity with other anti-CD4i MAbs. These data represent a rare instance of an antibody response that is specific to a pathogen-host cell protein interaction and underscore the diversity of immunogenic CD4i epitope structures that exist during natural infection.


Subject(s)
Antibodies, Monoclonal/immunology , CD4 Antigens/immunology , Epitopes/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , Antibody Affinity , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Binding
13.
Clin Vaccine Immunol ; 18(6): 922-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21508173

ABSTRACT

Most DNA-encoded adjuvants enhance immune responses to DNA vaccines in small animals but are less effective in primates. Here, we characterize the adjuvant activity of the catalytic A1 domain of cholera toxin (CTA1) for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) antigens in mice and macaques delivered by GeneGun. The inclusion of CTA1 with SIVmac239 Gag dramatically enhanced anti-Gag antibody responses in mice. The adjuvant effects of CTA1 for the secreted antigen HIV gp120 were much less pronounced than those for Gag, as the responses to gp120 were high in the absence of an adjuvant. CTA1 was a stronger adjuvant for Gag than was granulocyte-macrophage colony-stimulating factor (GM-CSF), and it also displayed a wider dose range than GM-CSF in mice. In macaques, CTA1 modestly enhanced the antibody responses to SIV Gag but potently primed for a recombinant Gag protein boost. The results of this study show that CTA1 is a potent adjuvant for SIV Gag when delivered by GeneGun in mice and that CTA1 provides a potent GeneGun-mediated DNA prime for a heterologous protein boost in macaques.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Antigens, Viral/immunology , Cholera Toxin/administration & dosage , SAIDS Vaccines/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adjuvants, Immunologic/genetics , Animals , Antibodies, Viral/blood , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Biolistics , Cholera Toxin/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Macaca , Mice , Mice, Inbred BALB C , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
14.
Methods Mol Biol ; 525: 517-31, xiv, 2009.
Article in English | MEDLINE | ID: mdl-19252841

ABSTRACT

Antibodies can neutralize HIV-1 with potency and cross-reactivity that varies widely and is related but not correlated to their antigen-binding affinity. Therefore, in addition to measuring binding affinity, an evaluation of the antibody neutralizing activity in tissue cultures is important for development of antibody-based therapeutics, design of candidate vaccine immunogens, and understanding the mechanisms of virus entry, neutralization, and evasion of immune responses. The development of a standardized assay for measurement of the in vitro neutralizing activities of the antibody has remained a challenging goal in the last two decades. There are two types of widely used assays, which vary in details between different laboratories--assays based on cell line/pseudovirus and assays based on infection of peripheral blood mononuclear cells (PBMCs). Here we describe in detail the PBMC-based assay, which is more laborious but in our opinion represents a closer approximation of the in vivo situation. As with all other in vitro assays the results of such measurements are only an indication of the antibody potency in vivo, and animal studies and ultimately clinical trials are needed for the development of such antibodies as potential prophylactics and therapeutics.


Subject(s)
Antibodies, Viral/immunology , HIV-1/immunology , Neutralization Tests/methods , HIV Core Protein p24/analysis , HIV Infections/immunology , HIV-1/isolation & purification , Humans , Immunoglobulin Variable Region/immunology , Inhibitory Concentration 50 , Lymphocyte Activation , Lymphocytes/virology , Titrimetry
15.
Proc Natl Acad Sci U S A ; 106(10): 3952-7, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19225108

ABSTRACT

Long-lived memory B cells (B(Mem)) provide an archive of historic Ab responses. By contrast, circulating Abs typically decline once the immunogen is cleared. Consequently, circulating Abs can underestimate the nature of cognate humoral immunity. On the other hand, the B(Mem) pool should provide a comprehensive picture of Ab specificities that arise over the entire course of infection. To test this hypothesis, we compared circulating Ab and B(Mem) from natural virus suppressors who control HIV-1 without therapy and maintain a relatively intact immune system. We found high frequencies of B(Mem) specific for the conserved neutralizing CD4 induced or CD4 binding site epitopes of gp120, whereas low Ab titers to these determinants were detected in contemporaneous plasma. These data suggest that plasma Ab repertoires can underestimate the breadth of humoral immunity, and analyses of B(Mem) should be included in studies correlating Ab specificity with protective immunity to HIV-1.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV-1/immunology , Immunologic Memory/immunology , CD4 Antigens/immunology , Conserved Sequence , Epitopes/immunology , Female , HIV Infections/blood , Humans , Male , Middle Aged , Neutralization Tests , Tissue Donors , Titrimetry
17.
Expert Opin Biol Ther ; 6(5): 523-31, 2006 May.
Article in English | MEDLINE | ID: mdl-16610981

ABSTRACT

The demand for new treatment options against HIV is becoming increasingly desperate as the side effects and the expansion and spread of drug-resistant virus within the infected population limit the clinical benefits provided by available anti-HIV drugs. Preparations of polyclonal antibodies have a long history of proven clinical utility against some viruses; however, they have enjoyed very limited success against HIV. Recent clinical trials and in vitro experiments suggest that monoclonal antibodies against HIV may have promise clinically. These antibodies and antibody-based reagents target either the viral envelope glycoprotein, the receptor (CD4) or coreceptor (CCR5) molecules, or transition-state structures that appear during viral entry. The challenge is whether an antibody-based therapy can be identified (with or without their small molecule brethren) that presents long-term clinical efficacy, low toxicity and minimal risk of clinical failure from viral resistance.


Subject(s)
HIV Antibodies/therapeutic use , HIV Infections/therapy , HIV-1/immunology , Immunotherapy/trends , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , Clinical Trials as Topic , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/immunology , Humans , Receptors, CCR5/immunology
18.
J Leukoc Biol ; 72(5): 962-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429718

ABSTRACT

Pertussis toxin (PT) and adenylate cyclase toxin (AT) are AB enterotoxins produced by Bordetella pertussis. PT is a powerful mucosal adjuvant whose cellular target and mechanism of action are unknown; however, emerging evidence suggests that dendritic cells (DC) may be a principal adjuvant target of PT. Here, we investigate the mechanism underlying the effects of these toxins on human monocyte-derived DC (MDDC) in vitro. We found that the effects of PT and AT on MDDC, including maturation, are mediated by cyclic adenosine monophosphate (cAMP). In this regard, adenosine 5'-diphosphate-ribosylation-defective derivatives of PT failed to induce maturation of MDDC, whereas dibutyryl-cAMP (d-cAMP) and Forskolin mimic the maturation of MDDC and dominant inhibition of cytokine production induced by these toxins. Also, cAMP-dependent kinase inhibitors blocked the ability of PT, AT, d-cAMP, and Forskolin to activate MDDC. Taken together, these results show that the effects of PT and AT on MDDC are mediated strictly by cAMP.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Adjuvants, Immunologic/pharmacology , Cyclic AMP/physiology , Dendritic Cells/immunology , Pertussis Toxin/pharmacology , Antigen Presentation , Bordetella pertussis/pathogenicity , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , Humans , Interleukin-12/biosynthesis , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Culture Test, Mixed , Monocytes/immunology , Pertussis Toxin/chemistry , Protein Structure, Tertiary , Second Messenger Systems , Tumor Necrosis Factor-alpha/biosynthesis
19.
Infect Immun ; 70(10): 5533-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12228279

ABSTRACT

Cholera toxin (CT) and heat-labile enterotoxin (LT) are powerful mucosal adjuvants whose cellular targets and mechanism of action are unknown. There is emerging evidence that dendritic cells (DC) are one of the principal cell types that mediate the adjuvant effects of these toxins in vivo. Here we investigate the effects of CT and LT on the maturation of human monocyte-derived DC (MDDC) in vitro. We found that an enzymatically active A domain is necessary for both CT and LT to induce the maturation of MDDC and that this activation is strictly cyclic AMP (cAMP) dependent. ADP-ribosylation-defective derivatives of these toxins failed to induce maturation of MDDC, whereas dibutyryl-cyclic-3',5'-AMP and Forskolin mimic the maturation of MDDC induced by CT and LT. In addition, an inhibitor of cAMP-dependent kinases, Rp-8-Br-cAMPs, blocked the ability of CT, LT, and Forskolin to activate MDDC. CT, LT, dibutyryl-cyclic-3',5'-AMP, and Forskolin also dominantly inhibit interleukin 12 and tumor necrosis factor alpha production by MDDC in the presence of saturating concentrations of lipopolysaccharide. Taken together, these results show that the effects of CT and LT on MDDC are mediated by cAMP.


Subject(s)
Bacterial Toxins/pharmacology , Cholera Toxin/pharmacology , Cyclic AMP/metabolism , Cytokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Enterotoxins/pharmacology , Escherichia coli Proteins , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Bacterial Toxins/chemistry , Bucladesine/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cell Communication , Cell Differentiation/drug effects , Cholera Toxin/chemistry , Colforsin/pharmacology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enterotoxins/chemistry , Humans , Immunity, Mucosal , In Vitro Techniques , Interleukin-12/biosynthesis , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Protein Structure, Tertiary , Tumor Necrosis Factor-alpha/biosynthesis
20.
J Hum Virol ; 5(1): 17-23, 2002.
Article in English | MEDLINE | ID: mdl-12352264

ABSTRACT

The antigenic diversity, rapid genetic integration into host cell DNA, and immune evasion tactics of human immunodeficiency virus type 1 (HIV-1) create formidable obstacles to the development of an effective vaccine against it. In spite of this, the advent of conformationally constrained HIV-1 Env and gp120 immunogens has made it feasible to formulate HIV-1 vaccines that induce broadly cross-reactive neutralizing antibodies and afford protection through humoral mechanisms. This paper reviews recent advances made by the authors toward the development of an HIV-1 vaccine that elicits such antibodies in both the mucosal and systemic immune compartments.


Subject(s)
AIDS Vaccines/immunology , Drug Design , HIV Antibodies/biosynthesis , HIV Infections/prevention & control , HIV-1/immunology , Immunity, Mucosal , HIV Antibodies/blood , HIV Antibodies/immunology , Humans , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL