Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
J Virol ; 87(18): 10163-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843636

ABSTRACT

Experimental vaccine antigens based upon the HIV-1 envelope glycoproteins (Env) have failed to induce neutralizing antibodies (NAbs) against the majority of circulating viral strains as a result of antibody evasion mechanisms, including amino acid variability and conformational instability. A potential vaccine design strategy is to stabilize Env, thereby focusing antibody responses on constitutively exposed, conserved surfaces, such as the CD4 binding site (CD4bs). Here, we show that a largely trimeric form of soluble Env can be stably cross-linked with glutaraldehyde (GLA) without global modification of antigenicity. Cross-linking largely conserved binding of all potent broadly neutralizing antibodies (bNAbs) tested, including CD4bs-specific VRC01 and HJ16, but reduced binding of several non- or weakly neutralizing antibodies and soluble CD4 (sCD4). Adjuvanted administration of cross-linked or unmodified gp140 to rabbits generated indistinguishable total gp140-specific serum IgG binding titers. However, sera from animals receiving cross-linked gp140 showed significantly increased CD4bs-specific antibody binding compared to animals receiving unmodified gp140. Moreover, peptide mapping of sera from animals receiving cross-linked gp140 revealed increased binding to gp120 C1 and V1V2 regions. Finally, neutralization titers were significantly elevated in sera from animals receiving cross-linked gp140 rather than unmodified gp140. We conclude that cross-linking favors antigen stability, imparts antigenic modifications that selectively refocus antibody specificity and improves induction of NAbs, and might be a useful strategy for future vaccine design.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , HIV Antibodies/blood , HIV Antigens/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/chemistry , AIDS Vaccines/genetics , Adjuvants, Immunologic/administration & dosage , Animals , Cross-Linking Reagents/metabolism , HIV Antigens/chemistry , HIV Antigens/metabolism , Rabbits , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/metabolism
2.
NPJ Vaccines ; 7(1): 152, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36433972

ABSTRACT

The HIV-1 envelope glycoprotein (Env) trimer is the key target for vaccines aimed at inducing neutralizing antibodies (NAbs) against HIV-1. The clinical candidate immunogen ConM SOSIP.v7 is a stabilized native-like HIV-1 Env trimer based on an artificial consensus sequence of all HIV-1 isolates in group M. In preclinical studies ConM SOSIP.v7 trimers induced strong autologous NAb responses in non-human primates (NHPs). To fine-map these responses, we isolated monoclonal antibodies (mAbs) from six cynomolgus macaques that were immunized three times with ConM SOSIP.v7 protein and boosted twice with the closely related ConSOSL.UFO.664 immunogen. A total of 40 ConM and/or ConS-specific mAbs were isolated, of which 18 were retrieved after the three ConM SOSIP.v7 immunizations and 22 after the two immunizations with ConSOSL.UFO.664. 22 mAbs (55%) neutralized the ConM and/or ConS virus. Cross-neutralization of ConS virus by approximately one-third of the mAbs was seen prior to ConSOSL.UFO.664 immunization, albeit with modest potency. Neutralizing antibodies predominantly targeted the V1 and V2 regions of the immunogens, with an apparent extension towards the V3 region. Thus, the V1V2V3 region is immunodominant in the potent NAb response elicited by two consensus sequence native-like HIV-1 Env immunogens. Immunization with these soluble consensus Env proteins also elicited non-neutralizing mAbs targeting the trimer base. These results inform the use and improvement of consensus-based trimer immunogens in combinatorial vaccine strategies.

3.
Nat Med ; 5(5): 526-34, 1999 May.
Article in English | MEDLINE | ID: mdl-10229229

ABSTRACT

Eight different protocols were compared for their ability to raise protection against immunodeficiency virus challenges in rhesus macaques. The most promising containment of challenge infections was achieved by intradermal DNA priming followed by recombinant fowl pox virus booster immunizations. This containment did not require neutralizing antibody and was active for a series of challenges ending with a highly virulent virus with a primary isolate envelope heterologous to the immunizing strain.


Subject(s)
Lentivirus Infections/immunology , Lentivirus Infections/prevention & control , Vaccination , Vaccines, DNA/therapeutic use , Viral Vaccines/therapeutic use , Animals , Antibodies, Viral/blood , Fowlpox virus/genetics , Injections, Intradermal , Macaca , Neutralization Tests , RNA, Viral/blood , T-Lymphocytes, Cytotoxic
4.
Res Sq ; 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33619476

ABSTRACT

One year into the Coronavirus Disease 2019 (COVID-19) pandemic caused by Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), effective treatments are still needed 1-3 . Monoclonal antibodies, given alone or as part of a therapeutic cocktail, have shown promising results in patients, raising the hope that they could play an important role in preventing clinical deterioration in severely ill or in exposed, high risk individuals 4-6 . Here, we evaluated the prophylactic and therapeutic effect of COVA1-18 in vivo , a neutralizing antibody isolated from a convalescent patient 7 and highly potent against the B.1.1.7. isolate 8,9 . In both prophylactic and therapeutic settings, SARS-CoV-2 remained undetectable in the lungs of COVA1-18 treated hACE2 mice. Therapeutic treatment also caused a dramatic reduction in viral loads in the lungs of Syrian hamsters. When administered at 10 mg kg - 1 one day prior to a high dose SARS-CoV-2 challenge in cynomolgus macaques, COVA1-18 had a very strong antiviral activity in the upper respiratory compartments with an estimated reduction in viral infectivity of more than 95%, and prevented lymphopenia and extensive lung lesions. Modelling and experimental findings demonstrate that COVA1-18 has a strong antiviral activity in three different preclinical models and could be a valuable candidate for further clinical evaluation.

5.
Science ; 283(5403): 857-60, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9933172

ABSTRACT

Clinical evidence suggests that cellular immunity is involved in controlling human immunodeficiency virus-1 (HIV-1) replication. An animal model of acquired immune deficiency syndrome (AIDS), the simian immunodeficiency virus (SIV)-infected rhesus monkey, was used to show that virus replication is not controlled in monkeys depleted of CD8+ lymphocytes during primary SIV infection. Eliminating CD8+ lymphocytes from monkeys during chronic SIV infection resulted in a rapid and marked increase in viremia that was again suppressed coincident with the reappearance of SIV-specific CD8+ T cells. These results confirm the importance of cell-mediated immunity in controlling HIV-1 infection and support the exploration of vaccination approaches for preventing infection that will elicit these immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/blood , Disease Progression , Gene Products, gag/blood , Humans , Lymphocyte Count , Lymphocyte Depletion , Macaca mulatta , Neutralization Tests , RNA, Viral/blood , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Viral Load , Viremia/immunology , Viremia/virology , Virus Replication
6.
Science ; 292(5514): 69-74, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11393868

ABSTRACT

Heterologous prime/boost regimens have the potential for raising high levels of immune responses. Here we report that DNA priming followed by a recombinant modified vaccinia Ankara (rMVA) booster controlled a highly pathogenic immunodeficiency virus challenge in a rhesus macaque model. Both the DNA and rMVA components of the vaccine expressed multiple immunodeficiency virus proteins. Two DNA inoculations at 0 and 8 weeks and a single rMVA booster at 24 weeks effectively controlled an intrarectal challenge administered 7 months after the booster. These findings provide hope that a relatively simple multiprotein DNA/MVA vaccine can help to control the acquired immune deficiency syndrome epidemic.


Subject(s)
AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Germinal Center/immunology , HIV Antibodies/blood , HIV Antibodies/immunology , HIV-1/genetics , HIV-1/immunology , HIV-1/physiology , Immunity, Mucosal , Immunization, Secondary , Immunologic Memory , Interferon-gamma/biosynthesis , Lymph Nodes/immunology , Macaca mulatta , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Viral Load
7.
Science ; 290(5491): 486-92, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11039923

ABSTRACT

With accumulating evidence indicating the importance of cytotoxic T lymphocytes (CTLs) in containing human immunodeficiency virus-1 (HIV-1) replication in infected individuals, strategies are being pursued to elicit virus-specific CTLs with prototype HIV-1 vaccines. Here, we report the protective efficacy of vaccine-elicited immune responses against a pathogenic SHIV-89.6P challenge in rhesus monkeys. Immune responses were elicited by DNA vaccines expressing SIVmac239 Gag and HIV-1 89.6P Env, augmented by the administration of the purified fusion protein IL-2/Ig, consisting of interleukin-2 (IL-2) and the Fc portion of immunoglobulin G (IgG), or a plasmid encoding IL-2/Ig. After SHIV-89.6P infection, sham-vaccinated monkeys developed weak CTL responses, rapid loss of CD4+ T cells, no virus-specific CD4+ T cell responses, high setpoint viral loads, significant clinical disease progression, and death in half of the animals by day 140 after challenge. In contrast, all monkeys that received the DNA vaccines augmented with IL-2/Ig were infected, but demonstrated potent secondary CTL responses, stable CD4+ T cell counts, preserved virus-specific CD4+ T cell responses, low to undetectable setpoint viral loads, and no evidence of clinical disease or mortality by day 140 after challenge.


Subject(s)
AIDS Vaccines/therapeutic use , Acquired Immunodeficiency Syndrome/prevention & control , HIV Infections/therapy , HIV-1 , Interleukin-2/therapeutic use , Vaccines, DNA/therapeutic use , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Disease Progression , HIV Antibodies/blood , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , HIV-1/physiology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Lymphocyte Activation , Macaca mulatta , Neutralization Tests , Recombinant Fusion Proteins/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Viral Load , Viremia , Virus Replication
8.
Sci Rep ; 9(1): 20005, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882800

ABSTRACT

Multiple approaches utilizing viral and DNA vectors have shown promise in the development of an effective vaccine against HIV. In this study, an alternative replication-defective flavivirus vector, RepliVax (RV), was evaluated for the delivery of HIV-1 immunogens. Recombinant RV-HIV viruses were engineered to stably express clade C virus Gag and Env (gp120TM) proteins and propagated in Vero helper cells. RV-based vectors enabled efficient expression and correct maturation of Gag and gp120TM proteins, were apathogenic in a sensitive suckling mouse neurovirulence test, and were similar in immunogenicity to recombinant poxvirus NYVAC-HIV vectors in homologous or heterologous prime-boost combinations in mice. In a pilot NHP study, immunogenicity of RV-HIV viruses used as a prime or boost for DNA or NYVAC candidates was compared to a DNA prime/NYVAC boost benchmark scheme when administered together with adjuvanted gp120 protein. Similar neutralizing antibody titers, binding IgG titers measured against a broad panel of Env and Gag antigens, and ADCC responses were observed in the groups throughout the course of the study, and T cell responses were elicited. The entire data demonstrate that RV vectors have the potential as novel HIV-1 vaccine components for use in combination with other promising candidates to develop new effective vaccination strategies.


Subject(s)
AIDS Vaccines/immunology , Defective Viruses/genetics , Flavivirus/genetics , Genetic Vectors , HIV-1/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Cross Reactions , Female , HIV Infections/virology , HIV-1/pathogenicity , Macaca mulatta , Mice , Mice, Inbred BALB C , Vero Cells , Virulence
9.
J Clin Invest ; 92(2): 840-7, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8349820

ABSTRACT

Sera from 11 volunteers immunized with a recombinant HIV-1 gp160-expressing vaccinia virus (HIVAC-1e; Oncogen/Bristol-Myers Squibb, Seattle, WA) and boosted with baculovirus-derived rgp160 (VaxSyn; MicroGeneSys, Inc., Meriden, CT) were evaluated for functional serum antibodies and their epitopes. Sera obtained prior to boosting had undetectable HIV-1-specific IgG and neutralizing activity, and did not block HIV-1 from binding or fusing to CD4+ MT-2 cells. 14 d after boosting, sera from each volunteer contained HIV-1-specific IgG titers of 1:40 to 1:1,280. Five of these sera also contained neutralizing antibodies, where most or all neutralizing activity was blocked by a synthetic peptide corresponding to amino acids 307-330 of the V3 loop of gp120, indicating that neutralizing antibodies were mostly V3 loop-specific. All sera obtained after boosting contained HIV-1 binding/fusion-inhibition antibodies, and a significant portion of their activity was blocked by the V3 loop peptide, a result consistent with the presence of antibodies against the region of the V3 loop that participates in fusion. Three sera with V3 loop-specific neutralizing and fusion-inhibition antibodies were studied further. In competitive antibody binding experiments, antibodies reactive with the conformation-dependent, CD4 binding site of gp120 were undetectable in each serum. When evaluated in combination with a monoclonal antibody to the CD4 binding site of gp120, two sera demonstrated synergism in neutralizing assays, and all three sera demonstrated synergism in binding/fusion-inhibition assays, further indicating that the functional antibodies were primarily V3 loop-specific. The synergism also suggests that a vaccine that elicits strong serum antibody responses to both regions of gp120 may improve the potential for inducing protective immunity.


Subject(s)
AIDS Vaccines/pharmacology , Antibodies, Monoclonal/metabolism , Antigens, CD/metabolism , CD4 Antigens/metabolism , Gene Products, env/pharmacology , HIV Antibodies/blood , HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Immunoglobulin G/blood , Protein Precursors/pharmacology , Vaccines, Synthetic/pharmacology , AIDS Vaccines/immunology , Adult , Binding Sites , Binding, Competitive , Cell Line , Gene Products, env/immunology , HIV Antibodies/pharmacology , HIV Envelope Protein gp160 , HIV-1/immunology , Humans , Immunoglobulin G/pharmacology , Membrane Fusion/drug effects , Membrane Fusion/immunology , Neutralization Tests , Protein Precursors/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/immunology
10.
AIDS Res Hum Retroviruses ; 22(7): 678-83, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16831092

ABSTRACT

A vaccine consisting of DNA priming followed by recombinant modified vaccinia Ankara (rMVA) boosting has achieved long-term control of a pathogenic challenge with a chimera of simian and human immunodeficiency viruses (SHIV-89.6P) in rhesus macaques. Based on these results, clade B HIV-1 DNA and rMVA immunogens have been developed for trials in humans. We conducted a first-time in humans phase I safety trial using the pGA2/JS2 (JS2) HIV-1 DNA priming vector expressing Gag, Pol, Env, Tat, Rev, and Vpu. Thirty HIV-uninfected adults were vaccinated with 0.3 or 3 mg of JS2 DNA, or a saline placebo, by intramuscular injection at months 0 and 2. Both doses of DNA were safe and well-tolerated with no differences between the control, 0.3 mg, or 3 mg groups (n = 6, 12, and 12, respectively) through 12 months of postvaccination follow- up. A chromium-release assay using fresh peripheral blood mononuclear cells (PBMCs) and a validated IFN-gamma ELISpot assay with frozen PBMCs failed to detect CD4(+) or CD8(+) HIV-1-specific T cell responses. HIV-specific neutralizing antibodies were also not detected. The vaccine is being further developed as a priming vector for a combined DNA plus rMVA prime/boost HIV vaccination regimen.


Subject(s)
AIDS Vaccines/adverse effects , HIV-1/immunology , Plasmids/adverse effects , Vaccines, DNA/adverse effects , Vaccinia virus/immunology , AIDS Vaccines/immunology , Adult , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/adverse effects , Genetic Vectors/immunology , HIV Antibodies/metabolism , Humans , Interferon-gamma/metabolism , Male , Plasmids/immunology , Statistics, Nonparametric , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Vaccinia virus/genetics
11.
Mucosal Immunol ; 9(1): 13-23, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25943275

ABSTRACT

There have been encouraging results for the development of an effective HIV vaccine. However, many questions remain regarding the quality of immune responses and the role of mucosal antibodies. We addressed some of these issues by using a simian immunodeficiency virus (SIV) DNA vaccine adjuvanted with plasmid-expressed mucosal chemokines combined with an intravaginal SIV challenge in rhesus macaque (RhM) model. We previously reported on the ability of CCR9 and CCR10 ligand (L) adjuvants to enhance mucosal and systemic IgA and IgG responses in small animals. In this study, RhMs were intramuscularly immunized five times with either DNA or DNA plus chemokine adjuvant delivered by electroporation followed by challenge with SIVsmE660. Sixty-eight percent of all vaccinated animals (P<0.01) remained either uninfected or had aborted infection compared with only 14% in the vaccine naĆÆve group. The highest protection was observed in the CCR10L chemokines group, where six of nine animals had aborted infection and two remained uninfected, leading to 89% protection (P<0.001). The induction of mucosal SIV-specific antibodies and neutralization titers correlated with trends in protection. These results indicate the need to further investigate the contribution of chemokine adjuvants to modulate immune responses and the role of mucosal antibodies in SIV/HIV protection.


Subject(s)
AIDS Vaccines/administration & dosage , Antibodies, Viral/biosynthesis , Chemokines/immunology , Immunity, Mucosal/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, DNA/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Chemokines/administration & dosage , Chemokines/genetics , Female , Immunity, Cellular/drug effects , Ligands , Macaca mulatta , Plasmids/chemistry , Plasmids/immunology , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, CCR10/genetics , Receptors, CCR10/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vagina/drug effects , Vagina/immunology , Vagina/virology
12.
Mucosal Immunol ; 9(2): 414-27, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26242599

ABSTRACT

Despite the risk of transmitting HIV-1, mothers in resource-poor areas are encouraged to breastfeed their infants because of beneficial immunologic and nutritional factors in milk. Interestingly, in the absence of antiretroviral prophylaxis, the overwhelming majority of HIV-1-exposed, breastfeeding infants are naturally protected from infection. To understand the role of HIV-1 envelope (Env)-specific antibodies in breast milk in natural protection against infant virus transmission, we produced 19 HIV-1 Env-specific monoclonal antibodies (mAbs) isolated from colostrum B cells of HIV-1-infected mothers and investigated their specificity, evolution, and anti-HIV-1 functions. Despite the previously reported genetic compartmentalization and gp120-specific bias of colostrum HIV Env-specific B cells, the colostrum Env-specific mAbs described here demonstrated a broad range of gp120 epitope specificities and functions, including inhibition of epithelial cell binding and dendritic cell-mediated virus transfer, neutralization, and antibody-dependent cellular cytotoxicity. We also identified divergent patterns of colostrum Env-specific B-cell lineage evolution with respect to crossreactivity to gastrointestinal commensal bacteria, indicating that commensal bacterial antigens play a role in shaping the local breast milk immunoglobulin G (IgG) repertoire. Maternal vaccine strategies to specifically target this breast milk B-cell population may be necessary to achieve safe breastfeeding for all HIV-1-exposed infants.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , B-Lymphocytes/immunology , Colostrum/immunology , HIV Antibodies/chemistry , HIV Envelope Protein gp120/antagonists & inhibitors , Immunoglobulin G/chemistry , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/isolation & purification , Antibody Affinity , Antibody Specificity , B-Lymphocytes/pathology , B-Lymphocytes/virology , Breast Feeding , Colostrum/cytology , Colostrum/virology , Cross Reactions , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Disease Resistance/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Gastrointestinal Microbiome/immunology , HIV Antibodies/biosynthesis , HIV Antibodies/isolation & purification , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/isolation & purification , Infant , Infectious Disease Transmission, Vertical/prevention & control , Milk, Human/chemistry , Milk, Human/immunology , Milk, Human/virology , Pregnancy , Symbiosis/immunology
13.
AIDS ; 5(5): 513-7, 1991 May.
Article in English | MEDLINE | ID: mdl-1863402

ABSTRACT

Neutralizing and complement-mediated infection-enhancing antibodies to HIV-1 were measured in sera or plasma from 54 HIV-1-positive individuals at various stages of disease, and from an additional 36 HIV-1-positive individuals for whom no clinical data were available. Antibodies were measured in microtiter infection assays utilizing MT-2 cells and the IIIB strain of HIV-1. The frequency of detection of both types of antibodies was identical, being 77 out of 90 cases (86%). Neutralizing and infection-enhancing antibodies were not always found together, and in four cases both were undetectable. No correlation was found between titers of either type of antibody and stage of disease. Furthermore, titers of infection-enhancing antibodies at early stages of disease did not predict rate of disease progression.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Complement System Proteins/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Cell Line , HIV Antibodies/blood , Humans , Neutralization Tests
14.
AIDS ; 12(18): 2407-15, 1998 Dec 24.
Article in English | MEDLINE | ID: mdl-9875578

ABSTRACT

OBJECTIVE: To determine the ability of live attenuated canarypox virus expressing HIV antigens to induce CD8+ cytotoxic T-cell responses and to prime for neutralizing antibody responses to boosting with purified recombinant gp120 subunit vaccine. DESIGN: A prospective, double-blind, randomized, immunogenicity and safety study was conducted in healthy adults at low risk for acquiring HIV infection and who were seronegative for HIV. METHODS: CD8+ cytotoxic T-cells directed against Env or Gag expressing target cells were measured after live recombinant canarypox-HIV-1 vaccine priming (vaccine given at days 0, 7, 14 and 21). Neutralizing antibodies were measured after subunit boosting (vaccine given at days 28 and 84). RESULTS: CD8+ CTL were induced in 64% of volunteers by the live recombinant canarypox-HIV-1 vaccine. All volunteers who received two doses of subunit vaccine after live recombinant canarypox priming developed neutralizing antibodies directed against laboratory strains of HIV-1 and seven out of eight volunteers tested developed neutralizing antibodies to the primary isolate, BZ167, but to none of eight other primary isolates. Unprimed controls had low or absent neutralizing antibodies after two doses of subunit vaccine. CONCLUSIONS: The live canarypox vector was safe, stimulated cytotoxic T-cells and primed for a vigorous neutralizing antibody response upon boosting with subunit gp120 vaccine. This vaccine combination should be evaluated further for inducing protection against HIV infection.


Subject(s)
AIDS Vaccines/immunology , Avipoxvirus/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/adverse effects , Adolescent , Adult , CD8-Positive T-Lymphocytes , Double-Blind Method , HIV Antibodies/blood , Humans , Immunization Schedule , Middle Aged , Neutralization Tests , Peptide Fragments/immunology , Prospective Studies , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic
15.
Article in English | MEDLINE | ID: mdl-2465404

ABSTRACT

Antibody-dependent enhancement (ADE) of human immunodeficiency virus type 1 (HIV-1) infection in vitro has been described recently and was shown to occur by two mechanisms: either participation of the alternative pathway of complement or to involve an Fc receptor-mediated, complement-independent mechanism. Complement-mediated ADE results in an accelerated cytopathic effect in target cells that can abrogate the protective properties of neutralizing antibodies. This study characterizes the surface antigens of MT-2 cells using flow cytometric analysis and shows that these cells express high levels of both CD4 and complement receptor type 2 (CR2) while several CD4+ cell lines that do not demonstrate complement-mediated ADE lack high levels of complement receptors. Further, utilizing MT-2 cell cultures, it is demonstrated that complement-mediated ADE of HIV-1 infection is conferred by the sera from more than 80% of HIV-1 antibody-positive individuals (N = 85). Complement-mediated ADE of HIV-1 infection causes an acceleration of several parameters indicative of HIV-1 infection in vitro including increased HIV-1 antigen synthesis as detected by indirect immunofluorescence, RNA accumulation as measured by a solution hybridization protocol, reverse transcriptase release, and progeny virus production.


Subject(s)
Cytopathogenic Effect, Viral , HIV Antibodies/immunology , HIV-1/physiology , RNA, Viral/biosynthesis , Receptors, Complement/physiology , Cell Line , Flow Cytometry , HIV Antigens/immunology , HIV-1/immunology , HIV-1/metabolism , Humans , RNA Probes , RNA-Directed DNA Polymerase/biosynthesis , Receptors, Complement/analysis , Virus Replication
16.
Immunol Res ; 22(2-3): 263-9, 2000.
Article in English | MEDLINE | ID: mdl-11339361

ABSTRACT

With the AIDS epidemic continuing to spread throughout the world, development of a safe, practical, and effective HIV vaccine is a national priority. HIV vaccine research efforts are currently targeted towards design of HIV immunogens that induce both cellular and humoral immunity. This brief review summarizes ongoing work at the Duke University School of Medicine on HIV vaccine development.


Subject(s)
AIDS Vaccines , Academic Medical Centers , HIV Infections/prevention & control , HIV-1/immunology , Adjuvants, Immunologic , Amino Acid Sequence , Animals , Clinical Trials as Topic , Cytokines/immunology , Epitopes/genetics , HIV Envelope Protein gp120/genetics , HIV-1/genetics , HLA Antigens/immunology , Humans , Molecular Sequence Data , North Carolina , Peptide Fragments/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology
17.
Immunol Lett ; 79(1-2): 57-61, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11595290

ABSTRACT

Accumulating evidence has demonstrated the importance of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes in controlling HIV-1 replication. We have elicited immune responses in rhesus monkeys utilizing DNA vaccines augmented by the administration of IL-2/Ig, a fusion protein consisting of interleukin-2 and the Fc portion of IgG2. These vaccine-elicited immune responses did not prevent infection following a high-dose intravenous challenge with SHIV(89.6P) but did control viremia to nearly undetectable levels and prevented immunodeficiency and clinical disease. In contrast, control monkeys developed high levels of viremia and exhibited a rapid loss of CD4(+) T cells, significant clinical disease progression, and death in half of the animals by day 140 following challenge. Vaccine approaches that elicit immune responses capable of reducing plasma viral loads, but not capable of inducing sterilizing immunity, may still provide substantial clinical benefits.


Subject(s)
AIDS Vaccines/pharmacology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Macaca mulatta/immunology , SAIDS Vaccines/pharmacology , Vaccines, DNA/pharmacology , Animals , CD4 Lymphocyte Count , HIV Antibodies/biosynthesis , HIV Infections/immunology , Immunoglobulin Fc Fragments/administration & dosage , Interleukin-2/administration & dosage , Neutralization Tests , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viremia/immunology , Viremia/prevention & control
18.
Immunol Lett ; 66(1-3): 53-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10203034

ABSTRACT

Highly polymorphic HLA class I molecules may influence rates of disease progression of HIV-infected individuals. Recent evidence suggests that individuals who mount vigorous CTL responses to multiple HIV-1 epitopes have reduced viral loads, and survive longer than individuals that make a less robust or less diverse CTL response. It has been difficult, however, to define associations between particular HLA class I alleles and rates of disease progression. This may be due, in part, to the uncontrolled variables associated with naturally acquired HIV infections. Studies using MHC-defined, non-human primates infected with well characterized viral stocks should help to clarify this relationship. To explore the possibility that MHC class I polymorphism can influence disease progression, we infected four Mamu-DRB-identical individuals from a family of MHC-defined rhesus macaques intravenously with 40 TCID50SIVmac239. Two of these macaques developed severe wasting and were euthanized within 80 days of infection, while the other two survived for more than 400 days without showing any symptoms of disease. Since all four of these macaques were Mamu-DRB-identical, we were able to exclude the MHC class II DRB loci as determinant of disease progression. Interestingly, both of the slow progressors made CTL responses to the same three SIV CTL epitopes, which were restricted by two molecules (Mamu-B*03 and B*04) encoded by their common maternal haplotype. The two rapid progressors did not share this haplotype with the slow progressors, and we were unable to detect CTL responses in these two siblings. These observations implicate products of the Mamu-B*03 and B*04 alleles in resistance to disease progression in this family of SIV-infected macaques, and provide additional evidence that certain MHC class I-restricted CTL responses may play a significant role in delaying the onset of AIDS.


Subject(s)
Histocompatibility Antigens Class I/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Alleles , Amino Acid Sequence , Animals , Cell Line , Disease Progression , Epitopes, T-Lymphocyte/immunology , Gene Products, env/immunology , Gene Products, nef/immunology , Macaca mulatta , Molecular Sequence Data , T-Lymphocytes, Cytotoxic/immunology
19.
Immunol Lett ; 66(1-3): 183-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10203053

ABSTRACT

To assess DNA immunization as a strategy for protecting against HIV infection in humans, we utilized SIVmne infection of Macaca fascicularis as a vaccine challenge model with moderate pathogenic potential. We compared the efficacy of DNA immunization alone and in combination with subunit protein boosts. All of the structural and regulatory genes of SIVmne clone 8 were cloned into mammalian expression vectors under the control of the CMV IE-1 promoter. Eight M. fascicularis were immunized twice with 3 mg of plasmid DNA divided between two sites; intramuscular and intradermal. Four primed macaques received a further two DNA immunizations at weeks 16-36, while the second group of four were boosted with 250 microg recombinant gp160 plus 250 microg recombinant Gag-Pol particles formulated in MF-59 adjuvant. Half of the controls received four immunizations of vector DNA; half received two vector DNA and two adjuvant immunizations. As expected, humoral immune responses were stronger in the macaques receiving subunit boosts, but responses were sustained in both groups. Significant neutralizing antibody titers to SIVmne were detected in one of the subunit-boosted animals and in none of the DNA-only animals prior to challenge. T-cell proliferative responses to gp160 and to Gag were detected in all immunized animals after three immunizations, and these responses increased after four immunizations. Cytokine profiles in PHA-stimulated PBMC taken on the day of challenge showed trends toward Thl responses in 2/4 macaques in the DNA vaccinated group and in 1/4 of the DNA plus subunit vaccinated macaques; Th2 responses in 3/4 DNA plus subunit-immunized macaques; and Th0 responses in 4/4 controls. In bulk CTL culture, SIV specific lysis was low or undetectable, even after four immunizations. However, stable SIV Gag-Pol- and env-specific T-cell clones (CD3+ CD8+) were isolated after only two DNA immunizations, and Gag-Pol- and Nef-specific CTL lines were isolated on the day of challenge. All animals were challenged at week 38 with SIVmne uncloned stock by the intrarectal route. Based on antibody anamnestic responses (western, ELISA, and neutralizing antibodies) and virus detection methods (co-culture of PBMC and LNMC, nested set PCR- of DNA from PBMC and LNMC, and plasma QC-PCR), there were major differences between the groups in the challenge outcome. Surprisingly, sustained low virus loads were observed only in the DNA group, suggesting that four immunizations with DNA only elicited more effective immune responses than two DNA primes combined with two protein boosts. Multigenic DNA vaccines such as these, bearing all structural and regulatory genes, show significant promise and may be a safe alternative to live-attenuated vaccines.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Animals , COS Cells , Macaca fascicularis , Viral Load
20.
AIDS Res Hum Retroviruses ; 3(3): 265-82, 1987.
Article in English | MEDLINE | ID: mdl-2829950

ABSTRACT

The Human Immunodeficiency Virus (HIV), the causative agent of AIDS, is thought to bind to T4+ (CD4+) target cells through the heavily glycosylated gp120 envelope glycoprotein. Plant lectins bind glycoproteins through noncovalent interaction with specific hexose residues; therefore, lectins were evaluated for their ability to inactivate HIV in vitro. The mannose-specific lectins concanavalin-A and succinyl concanavalin-A completely inactivated HIV while lentil lectin, wheat germ agglutinin, and phytohemagglutinin-P substantially inactivated HIV. BS-II, Vicia villosa (hairy vetch), and Ptilota plumosa (red marine algae) failed to alter the infectibility of HIV. Neither simple stearic hindrance, viral aggregation, nor lectin-cell interactions served to explain this phenomenon. Glycoprotein glycosylation was evaluated by differential lectin binding as well as molecular weight changes in gp120 when virus was produced in the presence of swainsonine, a glycosylation inhibitor. Lentil lectin bound gp120 better than concanavalin-A, suggesting the majority of glycosylation sites are fucosylated. The apparent molecular weight of gp120 was reduced by swainsonine, although HIV infectivity and concanavalin-A inactivation were retained. Thus, at least some N-glycosylation sites are complex-type glycoproteins but regions external to the (GlcNAc)2(Man)3 "core" pentasaccharide region are not required for HIV infectivity. It appears that the site or sites involved are nonfucosylated, high mannose and/or biantennary, nonsialylated, N-glycosylated regions of gp120 or gp41. Alternatively, they may be in close approximation to such carbohydrate regions.


Subject(s)
HIV/pathogenicity , Lectins/pharmacology , Mannose/physiology , Alkaloids/pharmacology , Binding Sites , Glycosylation , HIV/drug effects , HIV Envelope Protein gp120 , HIV Envelope Protein gp41 , Retroviridae Proteins/metabolism , Swainsonine , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL