Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 161
Filtrer
1.
NPJ Vaccines ; 7(1): 152, 2022 Nov 25.
Article de Anglais | MEDLINE | ID: mdl-36433972

RÉSUMÉ

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.

2.
Res Sq ; 2021 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-33619476

RÉSUMÉ

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.

3.
Sci Rep ; 9(1): 20005, 2019 12 27.
Article de Anglais | MEDLINE | ID: mdl-31882800

RÉSUMÉ

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.


Sujet(s)
Vaccins contre le SIDA/immunologie , Virus défectifs/génétique , Flavivirus/génétique , Vecteurs génétiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Vaccins synthétiques/immunologie , Animaux , Anticorps neutralisants/immunologie , Chlorocebus aethiops , Réactions croisées , Femelle , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/pathogénicité , Macaca mulatta , Souris , Souris de lignée BALB C , Cellules Vero , Virulence
4.
Mucosal Immunol ; 9(2): 414-27, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26242599

RÉSUMÉ

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.


Sujet(s)
Anticorps monoclonaux/composition chimique , Anticorps neutralisants/composition chimique , Lymphocytes B/immunologie , Colostrum/immunologie , Anticorps anti-VIH/composition chimique , Protéine d'enveloppe gp120 du VIH/antagonistes et inhibiteurs , Immunoglobuline G/composition chimique , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/isolement et purification , Anticorps neutralisants/biosynthèse , Anticorps neutralisants/isolement et purification , Affinité des anticorps , Spécificité des anticorps , Lymphocytes B/anatomopathologie , Lymphocytes B/virologie , Allaitement naturel , Colostrum/cytologie , Colostrum/virologie , Réactions croisées , Cellules dendritiques/immunologie , Cellules dendritiques/anatomopathologie , Cellules dendritiques/virologie , Résistance à la maladie/immunologie , Cellules épithéliales/immunologie , Cellules épithéliales/anatomopathologie , Cellules épithéliales/virologie , Femelle , Microbiome gastro-intestinal/immunologie , Anticorps anti-VIH/biosynthèse , Anticorps anti-VIH/isolement et purification , Protéine d'enveloppe gp120 du VIH/immunologie , Infections à VIH/immunologie , Infections à VIH/anatomopathologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Humains , Immunoglobuline G/biosynthèse , Immunoglobuline G/isolement et purification , Nourrisson , Transmission verticale de maladie infectieuse/prévention et contrôle , Lait humain/composition chimique , Lait humain/immunologie , Lait humain/virologie , Grossesse , Symbiose/immunologie
5.
Mucosal Immunol ; 9(1): 13-23, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-25943275

RÉSUMÉ

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 naïve 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.


Sujet(s)
Vaccins contre le SIDA/administration et posologie , Anticorps antiviraux/biosynthèse , Chimiokines/immunologie , Immunité muqueuse/effets des médicaments et des substances chimiques , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Vaccins à ADN/administration et posologie , Vaccins contre le SIDA/génétique , Vaccins contre le SIDA/immunologie , Adjuvants immunologiques/administration et posologie , Animaux , Chimiokines/administration et posologie , Chimiokines/génétique , Femelle , Immunité cellulaire/effets des médicaments et des substances chimiques , Ligands , Macaca mulatta , Plasmides/composition chimique , Plasmides/immunologie , Récepteurs CCR/génétique , Récepteurs CCR/immunologie , Récepteurs CCR10/génétique , Récepteurs CCR10/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/effets des médicaments et des substances chimiques , Virus de l'immunodéficience simienne/immunologie , Vaccination , Vaccins à ADN/génétique , Vaccins à ADN/immunologie , Vagin/effets des médicaments et des substances chimiques , Vagin/immunologie , Vagin/virologie
6.
J Virol ; 87(18): 10163-72, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23843636

RÉSUMÉ

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.


Sujet(s)
Vaccins contre le SIDA/immunologie , Anticorps neutralisants/sang , Anticorps anti-VIH/sang , Antigènes du VIH/immunologie , Produits du gène env du virus de l'immunodéficience humaine/immunologie , Vaccins contre le SIDA/administration et posologie , Vaccins contre le SIDA/composition chimique , Vaccins contre le SIDA/génétique , Adjuvants immunologiques/administration et posologie , Animaux , Réactifs réticulants/métabolisme , Antigènes du VIH/composition chimique , Antigènes du VIH/métabolisme , Lapins , Produits du gène env du virus de l'immunodéficience humaine/composition chimique , Produits du gène env du virus de l'immunodéficience humaine/métabolisme
7.
Mucosal Immunol ; 6(4): 692-703, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23299618

RÉSUMÉ

Prevention of HIV-1 transmission at mucosal surfaces will likely require durable pre-existing mucosal anti-HIV-1 antibodies (Abs). Defining the ontogeny, specificities and potentially protective nature of the initial mucosal virus-specific B-cell response will be critical for understanding how to induce protective Ab responses by vaccination. Genital fluids from patients within the earliest stages of acute HIV-1 infection (Fiebig I-VI) were examined for multiple anti-HIV specificities. Gp41 (but not gp120) Env immunoglobulin (Ig)A Abs were frequently elicited in both plasma and mucosal fluids within the first weeks of transmission. However, shortly after induction, these initial mucosal gp41 Env IgA Abs rapidly declined with a t(½) of ∼2.7 days. B-cell-activating factor belonging to the TNF family (BAFF) was elevated immediately preceding the appearance of gp41 Abs, likely contributing to an initial T-independent Ab response. HIV-1 transmission frequently elicits mucosal HIV-1 envelope-specific IgA responses targeted to gp41 that have a short half-life.


Sujet(s)
Protéine d'enveloppe gp41 du VIH/immunologie , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Immunoglobuline A/immunologie , Spécificité des anticorps/immunologie , Facteur d'activation des lymphocytes B/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Femelle , Infections à VIH/métabolisme , Infections à VIH/transmission , Humains , Immunité muqueuse , Immunoglobuline A/sang , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Activation des lymphocytes/immunologie , Mâle , Facteurs temps
8.
Vaccine ; 28(39): 6483-90, 2010 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-20654666

RÉSUMÉ

The development of an effective AIDS vaccine remains one of the highest priorities in HIV research. The live, attenuated varicella-zoster virus (VZV) Oka vaccine, safe and effective for prevention of chickenpox and zoster, also has potential as a recombinant vaccine against other pathogens, including human immunodeficiency virus (HIV). The simian varicella model, utilizing simian varicella virus (SVV), offers an approach to evaluate recombinant varicella vaccine candidates. Recombinant SVV (rSVV) vaccine viruses expressing simian immunodeficiency virus (SIV) env and gag antigens were constructed. The hypothesis tested was that a live, attenuated rSVV-SIV vaccine will induce immune responses against SIV in the rhesus macaques and provide protection against SIV challenge. The results demonstrated that rSVV-SIV vaccination induced low levels of neutralizing antibodies and cellular immune responses to SIV in immunized rhesus macaques and significantly reduced viral loads following intravenous challenge with pathogenic SIVmac251-CX-1.


Sujet(s)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , Vaccin contre la varicelle/immunologie , Vaccins contre le SIDA simien/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Charge virale , Animaux , Chlorocebus aethiops , Produits du gène env/immunologie , Produits du gène gag/immunologie , Immunité cellulaire , Interféron gamma/immunologie , Macaca mulatta , Mâle , Tests de neutralisation , Virus de l'immunodéficience simienne/immunologie , Lymphocytes T/immunologie , Vaccins synthétiques/immunologie , Cellules Vero
9.
Mucosal Immunol ; 2(6): 536-50, 2009 Nov.
Article de Anglais | MEDLINE | ID: mdl-19741603

RÉSUMÉ

Preventive human immunodeficiency virus (HIV) vaccination may require induction of virus-specific immune responses at mucosal sites to contain viral infection locally after exposure, as most HIV infections occur through mucosal surfaces. We compared the efficacy of an intranasal or intramuscular Simian immunodeficiency virus (SIV)+ interleukin (IL)-2+IL-15 DNA/SIV-MVA (modified vaccinia virus Ankara) vaccination in preventing disease progression in SIVmac251 intrarectally challenged rhesus macaques. SIV-specific rectal IgA responses were more significantly persistent in nasally vaccinated than in intramuscularly vaccinated animals. No significant differences were observed in the magnitude of systemic T-cell responses between the two groups, although the nasal immunization induced more significant anti-SIV T-cell responses in the colorectal mucosa. After challenge, CD4(+) central memory (C(M)) T-cell preservation and significant disease-delay were observed in both vaccination groups. However, nasally vaccinated animals had more significant early preservation of circulating and colorectal CD4(+) C(M) T cells, of circulating CD4(+)/alpha4beta7(+) effector memory (E(M)) T cells, and a longer disease-free interval when compared with the intramuscularly vaccinated or control groups. Regardless of vaccination status, long-term viremia control and preservation of CD4(+) C(M) T cells was detected in animals with significantly higher systemic CD8(+)/tumor necrosis factor (TNF)-alpha(+) and CD8(+)/interferon (IFN)-gamma(+) T-cell responses and higher SIV-specific CD4(+)/IL-2(+) responses in colorectal T cells.


Sujet(s)
Vaccins contre le SIDA simien/administration et posologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Administration par voie nasale , Animaux , Lymphocytes T CD4+ , Lymphocytes T CD8+ , Évolution de la maladie , Immunité muqueuse/immunologie , Injections musculaires , Muqueuse intestinale/immunologie , Muqueuse intestinale/virologie , Sous-populations de lymphocytes , Macaca mulatta
10.
AIDS Res Hum Retroviruses ; 22(7): 678-83, 2006 Jul.
Article de Anglais | MEDLINE | ID: mdl-16831092

RÉSUMÉ

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.


Sujet(s)
Vaccins contre le SIDA/effets indésirables , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Plasmides/effets indésirables , Vaccins à ADN/effets indésirables , Virus de la vaccine/immunologie , Vaccins contre le SIDA/immunologie , Adulte , Méthode en double aveugle , Test ELISA , Femelle , Vecteurs génétiques/effets indésirables , Vecteurs génétiques/immunologie , Anticorps anti-VIH/métabolisme , Humains , Interféron gamma/métabolisme , Mâle , Plasmides/immunologie , Statistique non paramétrique , Lymphocytes T cytotoxiques/immunologie , Vaccins à ADN/immunologie , Virus de la vaccine/génétique
11.
J Virol ; 79(17): 11214-24, 2005 Sep.
Article de Anglais | MEDLINE | ID: mdl-16103173

RÉSUMÉ

Human immunodeficiency virus type 1 (HIV-1) is a difficult target for vaccine development, in part because of its ever-expanding genetic diversity and attendant capacity to escape immunologic recognition. Vaccine efficacy might be improved by maximizing immunogen antigenic similarity to viruses likely to be encountered by vaccinees. To this end, we designed a prototype HIV-1 envelope vaccine using a deduced ancestral state for the env gene. The ancestral state reconstruction method was shown to be >95% accurate by computer simulation and 99.8% accurate when estimating the known inoculum used in an experimental infection study in rhesus macaques. Furthermore, the deduced ancestor gene differed from the set of sequences used to derive the ancestor by an average of 12.3%, while these latter sequences were an average of 17.3% different from each other. A full-length ancestral subtype B HIV-1 env gene was constructed and shown to produce a glycoprotein of 160 kDa that bound and fused with cells expressing the HIV-1 coreceptor CCR5. This Env was also functional in a virus pseudotype assay. When either gp160- or gp140-expressing plasmids and recombinant gp120 were used to immunize rabbits in a DNA prime-protein boost regimen, the artificial gene induced immunoglobulin G antibodies capable of weakly neutralizing heterologous primary HIV-1 strains. The results were similar for rabbits immunized in parallel with a natural isolate, HIV-1 SF162. Further design efforts to better present conserved neutralization determinants are warranted.


Sujet(s)
Vaccins contre le SIDA/immunologie , Anticorps anti-VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Immunisation , Vaccins synthétiques/immunologie , Protéines de l'enveloppe virale/immunologie , Séquence d'acides aminés , Animaux , Produits du gène env/composition chimique , Produits du gène env/génétique , Produits du gène env/immunologie , Protéine d'enveloppe gp120 du VIH/génétique , Protéine d'enveloppe gp120 du VIH/immunologie , Protéine d'enveloppe gp160 du VIH/génétique , Protéine d'enveloppe gp160 du VIH/immunologie , Protéine d'enveloppe gp160 du VIH/métabolisme , Rappel de vaccin , Données de séquences moléculaires , Tests de neutralisation , Phylogenèse , Lapins , Récepteurs CCR5/métabolisme , Protéines recombinantes/immunologie , Solubilité , Protéines de l'enveloppe virale/génétique , Produits du gène env du virus de l'immunodéficience humaine
12.
J Virol ; 78(24): 13819-28, 2004 Dec.
Article de Anglais | MEDLINE | ID: mdl-15564490

RÉSUMÉ

Further advances are required in understanding protection from AIDS by T-cell immunity. We analyzed a set of multigenic simian/human immunodeficiency virus (SHIV) DNA and fowlpox virus priming and boosting vaccines for immunogenicity and protective efficacy in outbred pigtail macaques. The number of vaccinations required, the effect of DNA vaccination alone, and the effect of cytokine (gamma interferon) coexpression by the fowlpox virus boost was also studied. A coordinated induction of high levels of broadly reactive CD4 and CD8 T-cell immune responses was induced by sequential DNA and fowlpox virus vaccination. The immunogenicity of regimens utilizing fowlpox virus coexpressing gamma interferon, a single DNA priming vaccination, or DNA vaccines alone was inferior. Significant control of a virulent SHIV challenge was observed despite a loss of SHIV-specific proliferating T cells. The outcome of challenge with virulent SHIV(mn229) correlated with vaccine immunogenicity except that DNA vaccination alone primed for protection almost as effectively as the DNA/fowlpox virus regimen despite negligible immunogenicity by standard assays. These studies suggest that priming of immunity with DNA and fowlpox virus vaccines could delay AIDS in humans.


Sujet(s)
Vaccins contre le SIDA/immunologie , Virus de la variole de la volaille/immunologie , Infections à VIH/prévention et contrôle , Immunisation , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Vaccins à ADN/immunologie , Vaccins contre le SIDA/administration et posologie , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Humains , Calendrier vaccinal , Rappel de vaccin , Interféron gamma/immunologie , Interféron gamma/métabolisme , Activation des lymphocytes , Macaca nemestrina , Vaccins contre le SIDA simien/administration et posologie , Vaccins contre le SIDA simien/immunologie , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/immunologie , Résultat thérapeutique , Vaccins à ADN/administration et posologie
13.
J Med Primatol ; 31(4-5): 217-27, 2002 Aug.
Article de Anglais | MEDLINE | ID: mdl-12390544

RÉSUMÉ

The ever increasing number of people infected by human immunodeficiency virus (HIV) throughout the world renders the development of effective vaccines an urgent priority. Herein, we report on an attempt to induce and enhance antiviral responses using a deoxyribonucleic acid (DNA) prime/virus-like particles (VLP) protein boost strategy adjuvanted with interleukin (IL)-12/GM-CSF in rhesus macaques challenged with simian immunodeficiency virus (SIV). Thus, groups of monkeys were administered three consecutive doses of pVecB7 a plasmid expressing VLP with or without plasmids expressing IL-12 and GM-CSF at weeks 0, 13 and 26. The VLP boost was administered at week 39 with or without IL-12. All monkeys were challenged intrarectally with SIVsmE660 2 months following the protein boost. All except one immunized monkey became infected. While all immunized monkeys showed a marked reduction of acute viral peaks, reduction of viral load set points was only achieved in groups whose prime-boost immunizations were supplemented with IL-12/GM-CSF (prime) and/or with IL-12 (boost). Control of viremia correlated with lack of disease progression and survival. Detection of virus in rectal washes at 1 year post-challenge was only successful in monkeys whose immunizations did not include cytokine adjuvant, but these loads did not correlate with plasma viral loads. In summary, use of IL-12 and/or GM-CSF was shown to provide significant differences in the outcome of SIV challenge of prime/boost immunized monkeys.


Sujet(s)
Facteur de stimulation des colonies de granulocytes et de macrophages/immunologie , Interleukine-12/immunologie , Macaca mulatta/immunologie , Vaccins contre le SIDA simien/administration et posologie , Vaccins contre le SIDA simien/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne/immunologie , Adjuvants immunologiques , Animaux , Anticorps antiviraux/immunologie , Modèles animaux de maladie humaine , Évolution de la maladie , Calendrier d'administration des médicaments , Association de médicaments , Facteur de stimulation des colonies de granulocytes et de macrophages/administration et posologie , Interleukine-12/administration et posologie , Macaca mulatta/virologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/physiologie , Taux de survie , Facteurs temps , Vaccins à ADN/administration et posologie , Vaccins à ADN/immunologie , Charge virale
14.
J Med Primatol ; 31(3): 109-19, 2002 Jun.
Article de Anglais | MEDLINE | ID: mdl-12190851

RÉSUMÉ

Simian-human immunodeficiency viruses (SHIV) allow the evaluation of antiviral strategies that target the envelope glycoproteins of the human immunodeficiency virus 1 (HIV-1) in macaques. We previously protected neonates from oral challenge with cell-free SHIV-vpu+ by passive immunization with synergistic human neutralizing monoclonal antibodies (mAbs) (Baba et al., Nat Med 6:200-206, 2000). mAbs were administered prenatally to pregnant dams and postnatally to the neonates. Here, we used solely postnatal or postexposure mAb treatment, thus significantly reducing the amount of mAbs necessary. All neonatal monkeys were also protected with these abbreviated mAb regimens. Our results are directly relevant for humans because we used mAbs that target HIV-1 envelope glycoproteins. Thus, the large-scale use of passive immunization with neutralizing mAbs may be feasible in human neonates. The mAbs, being natural human proteins, can be expected to have low toxicity. Passive immunization has promise to prevent intrapartum as well as milk-borne virus transmission from HIV-1-infected women to their infants.


Sujet(s)
Animaux nouveau-nés/immunologie , VIH (Virus de l'Immunodéficience Humaine)/immunologie , Immunisation passive/méthodes , Macaca mulatta/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne/immunologie , Administration par voie orale , Animaux , Anticorps monoclonaux/immunologie , Technique de Western , Anticorps anti-VIH/immunologie , Protéines du virus de l'immunodéficience humaine , Humains , Immunité muqueuse , Syndrome d'immunodéficience acquise du singe/transmission , Facteurs temps , Charge virale , Protéines virales régulatrices ou accessoires/physiologie
15.
J Med Primatol ; 31(1): 40-60, 2002 Feb.
Article de Anglais | MEDLINE | ID: mdl-12076047

RÉSUMÉ

Newborn macaques were vaccinated against a chimeric simian human immunodeficiency (SHIV) virus, SHIV-vpu+, by DNA priming and boosting with homologous HIV-1 gp160. Following SHIV-vpu+ challenge, containment of infection was observed in 4 of 15 animals given DNA priming/protein boost vaccination and in three of four animals given gp160 boosts only. Rechallenge with homologous virus of six animals that contained the first challenge virus resulted in rapid viral clearance or low viral loads. Upon additional rechallenge with heterologous, pathogenic SHIV89.6P, four of these six animals maintained normal CD4+ T-cell counts with no or limited SHIV89.6P infection. Our data suggest that humoral and cellular immune mechanisms may have contributed to the containment of SHIV89.6P; however, viral interference with SHIV-vpu+ could also have played a role. Our results indicate that immunogenicity and efficacy of candidate AIDS vaccines are not affected when vaccination is initiated during infancy as compared with later in life.


Sujet(s)
Vaccins contre le SIDA/immunologie , VIH (Virus de l'Immunodéficience Humaine)/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Animaux nouveau-nés , Numération des lymphocytes CD4 , Chimère , ADN viral , VIH (Virus de l'Immunodéficience Humaine)/pathogénicité , Rappel de vaccin/médecine vétérinaire , Macaca mulatta/virologie , Plasmides , Virus de l'immunodéficience simienne/pathogénicité , Vaccination/médecine vétérinaire
16.
J Virol ; 76(1): 292-302, 2002 Jan.
Article de Anglais | MEDLINE | ID: mdl-11739694

RÉSUMÉ

T-cell-mediated immune effector mechanisms play an important role in the containment of human immunodeficiency virus/simian immunodeficiency virus (HIV/SIV) replication after infection. Both vaccination- and infection-induced T-cell responses are dependent on the host major histocompatibility complex classes I and II (MHC-I and MHC-II) antigens. Here we report that both inherent, host-dependent immune responses to SIVmac251 infection and vaccination-induced immune responses to viral antigens were able to reduce virus replication and/or CD4+ T-cell loss. Both the presence of the MHC-I Mamu-A*01 genotype and vaccination of rhesus macaques with ALVAC-SIV-gag-pol-env (ALVAC-SIV-gpe) contributed to the restriction of SIVmac251 replication during primary infection, preservation of CD4+ T cells, and delayed disease progression following intrarectal challenge exposure of the animals to SIV(mac251 (561)). ALVAC-SIV-gpe immunization induced cytotoxic T-lymphocyte (CTL) responses cumulatively in 67% of the immunized animals. Following viral challenge, a significant secondary virus-specific CD8+ T-cell response was observed in the vaccinated macaques. In the same immunized macaques, a decrease in virus load during primary infection (P = 0.0078) and protection from CD4 loss during both acute and chronic phases of infection (P = 0.0099 and P = 0.03, respectively) were observed. A trend for enhanced survival of the vaccinated macaques was also observed. Neither boosting the ALVAC-SIV-gpe with gp120 immunizations nor administering the vaccine by the combination of mucosal and systemic immunization routes increased significantly the protective effect of the ALVAC-SIV-gpe vaccine. While assessing the role of MHC-I Mamu-A*01 alone in the restriction of viremia following challenge of nonvaccinated animals with other SIV isolates, we observed that the virus load was not significantly lower in Mamu-A*01-positive macaques following intravenous challenge with either SIV(mac251 (561)) or SIV(SME660). However, a significant delay in CD4+ T-cell loss was observed in Mamu-A*01-positive macaques in each group. Of interest, in the case of intravenous or intrarectal challenge with the chimeric SIV/HIV strains SHIV(89.6P) or SHIV(KU2), respectively, MHC-I Mamu-A*01-positive macaques did not significantly restrict primary viremia. The finding of the protective effect of the Mamu-A*01 molecule parallels the protective effect of the B*5701 HLA allele in HIV-1-infected humans and needs to be accounted for in the evaluation of vaccine efficacy against SIV challenge models.


Sujet(s)
Produits du gène env/administration et posologie , Produits du gène gag/administration et posologie , Produits du gène pol/administration et posologie , Antigènes d'histocompatibilité de classe I/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne/immunologie , Vaccination , Vaccins antiviraux/administration et posologie , Animaux , Produits du gène env/immunologie , Produits du gène gag/immunologie , Produits du gène pol/immunologie , Macaca , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de la vaccine , Vaccins antiviraux/immunologie
17.
J Infect Dis ; 184(12): 1603-7, 2001 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-11740737

RÉSUMÉ

According to the principle of original antigenic sin, neutralizing antibodies (NAbs) initially directed against a single virus strain compromise the immune system's ability to subsequently mount adequate responses against antigenically divergent virus strains. In this study, rhesus macaques, after vaccination and breakthrough infection with homologous simian-human immunodeficiency virus (SHIV), developed strong SHIV-IIIB strain-directed NAb responses that were mostly V3 loop specific. After superinfection with heterologous SHIV89.6P, all macaques developed high-titer SHIV89.6P-specific NAbs without significant boosting of SHIV-IIIB-specific NAbs. These results indicate that prior B cell responses against a single immunodeficiency virus strain do not preclude the later development of NAbs against a divergent strain of the same virus.


Sujet(s)
Anticorps antiviraux/sang , Anticorps anti-VIH/sang , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Virus de l'immunodéficience simienne/immunologie , Vaccins contre le SIDA/immunologie , Séquence d'acides aminés , Animaux , Anticorps antiviraux/immunologie , Spécificité des anticorps , Anticorps anti-VIH/immunologie , Antigènes du VIH/immunologie , Protéine d'enveloppe gp120 du VIH/immunologie , Infections à VIH/immunologie , Humains , Immunisation , Mémoire immunologique , Macaca mulatta , Données de séquences moléculaires , Tests de neutralisation , Fragments peptidiques/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie
18.
Proc Natl Acad Sci U S A ; 98(23): 13288-93, 2001 Nov 06.
Article de Anglais | MEDLINE | ID: mdl-11687611

RÉSUMÉ

The risks and benefits of structured treatment interruption (STI) in HIV-1-infected subjects are not fully understood. A pilot study was performed to compare STI with continuous highly active antiretroviral therapy (HAART) in chronic HIV-1-infected subjects with HIV-1 plasma RNA levels (VL) <400 copies per ml and CD4(+) T cells >400 per microl. CD4(+) T cells, VL, HIV-1-specific neutralizing antibodies, and IFN-gamma-producing HIV-1-specific CD8(+) and CD4(+) T cells were measured in all subjects. STIs of 1-month duration separated by 1 month of HAART, before a final 3-month STI, resulted in augmented CD8(+) T cell responses in all eight STI subjects (P = 0.003), maintained while on HAART up to 22 weeks after STI, and augmented neutralization titers to autologous HIV-1 isolate in one of eight subjects. However, significant decline of CD4(+) T cell count from pre-STI level, and VL rebound to pre-HAART baseline, occurred during STI (P = 0.001 and 0.34, respectively). CD4(+) T cell counts were regained on return to HAART. Control subjects (n = 4) maintained VL <400 copies per ml and stable CD4(+) T cell counts, and showed no enhancement of antiviral CD8(+) T cell responses. Despite increases in antiviral immunity, no control of VL was observed. Future studies of STI should proceed with caution.


Sujet(s)
Calendrier d'administration des médicaments , Infections à VIH/traitement médicamenteux , Inhibiteurs de protéase du VIH/administration et posologie , Inhibiteurs de la transcriptase inverse/administration et posologie , Adulte , Thérapie antirétrovirale hautement active , Numération des lymphocytes CD4 , Lymphocytes T CD8+/immunologie , Association de médicaments , Femelle , Anticorps anti-VIH/immunologie , Inhibiteurs de protéase du VIH/usage thérapeutique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/isolement et purification , Humains , Mâle , Adulte d'âge moyen , Tests de neutralisation , Inhibiteurs de la transcriptase inverse/usage thérapeutique , Charge virale
19.
J Virol ; 75(21): 10200-7, 2001 Nov.
Article de Anglais | MEDLINE | ID: mdl-11581388

RÉSUMÉ

Immediate treatment of acute human immunodeficiency virus type 1 (HIV-1) infection has been associated with subsequent control of viremia in a subset of patients after therapy cessation, but the immune responses contributing to control have not been fully defined. Here we examined neutralizing antibodies as a correlate of viremia control following treatment interruption in HIV-1-infected individuals in whom highly active antiretriviral therapy (HAART) was initiated during early seroconversion and who remained on therapy for 1 to 3 years. Immediately following treatment interruption, neutralizing antibodies were undetectable with T-cell-line adapted strains and the autologous primary HIV-1 isolate in seven of nine subjects. Env- and Gag-specific antibodies as measured by enzyme-linked immunosorbent assay were also low or undetectable at this time. Despite this apparent poor maturation of the virus-specific B-cell response during HAART, autologous neutralizing antibodies emerged rapidly and correlated with a spontaneous downregulation in rebound viremia following treatment interruption in three subjects. Control of rebound viremia was seen in other subjects in the absence of detectable neutralizing antibodies. The results indicate that virus-specific B-cell priming occurs despite the early institution of HAART, allowing rapid secondary neutralizing-antibody production following treatment interruption in a subset of individuals. Since early HAART limits viral diversification, we hypothesize that potent neutralizing-antibody responses to autologous virus are able to mature and that in some persons these responses contribute to the control of plasma viremia after treatment cessation.


Sujet(s)
Syndrome d'immunodéficience acquise/traitement médicamenteux , Thérapie antirétrovirale hautement active , Anticorps anti-VIH/sang , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Virémie/traitement médicamenteux , Syndrome d'immunodéficience acquise/immunologie , Maladie aigüe , Humains , Lymphocytes T cytotoxiques/immunologie , Virémie/immunologie
20.
Transfus Clin Biol ; 8(4): 350-8, 2001 Aug.
Article de Anglais | MEDLINE | ID: mdl-11642027

RÉSUMÉ

Neonatal macaques were completely protected against oral challenge with SHIV-vpu+, a simian-human immunodeficiency virus that encodes the envelope gene of a laboratory-adapted HIV strain, by pre- and post-natal treatment with a triple combination of human neutralizing monoclonal antibodies (mAbs). The mAbs were directed either against the CD4 binding site, a glycosylation-dependent gp120 epitope, or against a linear epitope on gp41. This triple combination was highly synergistic in vitro and neutralized primary HIV completely. Subsequently, oral challenge was performed with pathogenic SHIV89.6P, an animal-passaged variant of a chimeric virus that encodes the envelope gene of the primary, dual-tropic HIV89.6. Only post-natal treatment with a similar triple mAb combination was used. One out of 4 mAb-treated infants was completely protected from infection. In the other 3 treated animals, there was a tendency towards lower peak viral RNA loads compared with untreated controls. Two out of 4 mAb-treated infants maintained normal CD4+ T-cell numbers, in contrast to all controls that had steep declines at 2 weeks post-challenge. We conclude that the triple mAb combination significantly protected the neonates, even against mucosal challenge with pathogenic SHIV89.6P. Passively administered synergistic human mAbs may play a role in preventing mother-infant transmission of HIV, both against intrapartum transmission as well as against infection through breast milk. As passive immunization is a tool to assess correlates of immune protection, we conclude that the epitopes recognized by the mAbs in our combinations are important for AIDS vaccine development. Future passive immunization studies may reveal other important conserved epitopes.


Sujet(s)
Vaccins contre le SIDA/administration et posologie , Anticorps monoclonaux/administration et posologie , Anticorps anti-VIH/administration et posologie , Infections à VIH/prévention et contrôle , VIH (Virus de l'Immunodéficience Humaine)/immunologie , Immunisation passive , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne/immunologie , Vaccination , Vaccins contre le SIDA/immunologie , Administration par voie orale , Animaux , Animaux nouveau-nés , Anticorps monoclonaux/immunologie , Numération des lymphocytes CD4 , Césarienne , Accouchement (procédure) , Modèles animaux de maladie humaine , Femelle , Anticorps anti-VIH/immunologie , Protéine d'enveloppe gp120 du VIH/immunologie , Protéine d'enveloppe gp41 du VIH/immunologie , Humains , Immunité acquise d'origine maternelle , Nouveau-né , Transmission verticale de maladie infectieuse/prévention et contrôle , Lactation , Macaca mulatta , Échange foetomaternel , Lait/virologie , Tests de neutralisation , Projets pilotes , Grossesse , Complications infectieuses de la grossesse/virologie , Spécificité d'espèce , Assemblage viral , Excrétion virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...