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










Publication year range
1.
Nat Med ; 27(10): 1718-1724, 2021 10.
Article in English | MEDLINE | ID: mdl-34621054

ABSTRACT

Human immunodeficiency virus (HIV)-1-specific broadly neutralizing monoclonal antibodies are currently under development to treat and prevent HIV-1 infection. We performed a single-center, randomized, double-blind, dose-escalation, placebo-controlled trial of a single administration of the HIV-1 V3-glycan-specific antibody PGT121 at 3, 10 and 30 mg kg-1 in HIV-uninfected adults and HIV-infected adults on antiretroviral therapy (ART), as well as a multicenter, open-label trial of one infusion of PGT121 at 30 mg kg-1 in viremic HIV-infected adults not on ART (no. NCT02960581). The primary endpoints were safety and tolerability, pharmacokinetics (PK) and antiviral activity in viremic HIV-infected adults not on ART. The secondary endpoints were changes in anti-PGT121 antibody titers and CD4+ T-cell count, and development of HIV-1 sequence variations associated with PGT121 resistance. Among 48 participants enrolled, no treatment-related serious adverse events, potential immune-mediated diseases or Grade 3 or higher adverse events were reported. The most common reactions among PGT121 recipients were intravenous/injection site tenderness, pain and headache. Absolute and relative CD4+ T-cell counts did not change following PGT121 infusion in HIV-infected participants. Neutralizing anti-drug antibodies were not elicited. PGT121 reduced plasma HIV RNA levels by a median of 1.77 log in viremic participants, with a viral load nadir at a median of 8.5 days. Two individuals with low baseline viral loads experienced ART-free viral suppression for ≥168 days following antibody infusion, and rebound viruses in these individuals demonstrated full or partial PGT121 sensitivity. The trial met the prespecified endpoints. These data suggest that further investigation of the potential of antibody-based therapeutic strategies for long-term suppression of HIV is warranted, including in individuals off ART and with low viral load.


Subject(s)
Antiviral Agents/administration & dosage , Broadly Neutralizing Antibodies/administration & dosage , HIV Infections/drug therapy , HIV-1/drug effects , Adult , Antiretroviral Therapy, Highly Active , Antiviral Agents/immunology , Antiviral Agents/pharmacokinetics , Broadly Neutralizing Antibodies/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Double-Blind Method , Female , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/immunology , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Male , Middle Aged , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Placebos , Viral Load/drug effects , Viral Load/immunology , Young Adult
3.
Nature ; 586(7830): 583-588, 2020 10.
Article in English | MEDLINE | ID: mdl-32731257

ABSTRACT

A safe and effective vaccine for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may be required to end the coronavirus disease 2019 (COVID-19) pandemic1-8. For global deployment and pandemic control, a vaccine that requires only a single immunization would be optimal. Here we show the immunogenicity and protective efficacy of a single dose of adenovirus serotype 26 (Ad26) vector-based vaccines expressing the SARS-CoV-2 spike (S) protein in non-human primates. Fifty-two rhesus macaques (Macaca mulatta) were immunized with Ad26 vectors that encoded S variants or sham control, and then challenged with SARS-CoV-2 by the intranasal and intratracheal routes9,10. The optimal Ad26 vaccine induced robust neutralizing antibody responses and provided complete or near-complete protection in bronchoalveolar lavage and nasal swabs after SARS-CoV-2 challenge. Titres of vaccine-elicited neutralizing antibodies correlated with protective efficacy, suggesting an immune correlate of protection. These data demonstrate robust single-shot vaccine protection against SARS-CoV-2 in non-human primates. The optimal Ad26 vector-based vaccine for SARS-CoV-2, termed Ad26.COV2.S, is currently being evaluated in clinical trials.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Macaca mulatta , Pandemics/prevention & control , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , COVID-19 , COVID-19 Vaccines , Disease Models, Animal , Female , Immunity, Cellular , Immunity, Humoral , Macaca mulatta/immunology , Macaca mulatta/virology , Male , SARS-CoV-2 , Vaccination , Viral Load
4.
Science ; 369(6505): 806-811, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32434945

ABSTRACT

The global coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has made the development of a vaccine a top biomedical priority. In this study, we developed a series of DNA vaccine candidates expressing different forms of the SARS-CoV-2 spike (S) protein and evaluated them in 35 rhesus macaques. Vaccinated animals developed humoral and cellular immune responses, including neutralizing antibody titers at levels comparable to those found in convalescent humans and macaques infected with SARS-CoV-2. After vaccination, all animals were challenged with SARS-CoV-2, and the vaccine encoding the full-length S protein resulted in >3.1 and >3.7 log10 reductions in median viral loads in bronchoalveolar lavage and nasal mucosa, respectively, as compared with viral loads in sham controls. Vaccine-elicited neutralizing antibody titers correlated with protective efficacy, suggesting an immune correlate of protection. These data demonstrate vaccine protection against SARS-CoV-2 in nonhuman primates.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Spike Glycoprotein, Coronavirus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Betacoronavirus/physiology , Bronchoalveolar Lavage Fluid/virology , COVID-19 , COVID-19 Vaccines , Coronavirus Infections/immunology , Coronavirus Infections/virology , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunization, Secondary , Immunogenicity, Vaccine , Immunologic Memory , Macaca mulatta , Male , Mutant Proteins/chemistry , Mutant Proteins/immunology , Nasal Mucosa/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Domains , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Vaccination , Vaccines, DNA/administration & dosage , Viral Load , Viral Vaccines/administration & dosage
5.
Science ; 369(6505): 812-817, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32434946

ABSTRACT

An understanding of protective immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical for vaccine and public health strategies aimed at ending the global coronavirus disease 2019 (COVID-19) pandemic. A key unanswered question is whether infection with SARS-CoV-2 results in protective immunity against reexposure. We developed a rhesus macaque model of SARS-CoV-2 infection and observed that macaques had high viral loads in the upper and lower respiratory tract, humoral and cellular immune responses, and pathologic evidence of viral pneumonia. After the initial viral clearance, animals were rechallenged with SARS-CoV-2 and showed 5 log10 reductions in median viral loads in bronchoalveolar lavage and nasal mucosa compared with after the primary infection. Anamnestic immune responses after rechallenge suggested that protection was mediated by immunologic control. These data show that SARS-CoV-2 infection induced protective immunity against reexposure in nonhuman primates.


Subject(s)
Betacoronavirus , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Betacoronavirus/immunology , Betacoronavirus/physiology , Bronchoalveolar Lavage Fluid/virology , COVID-19 , Coronavirus Infections/pathology , Coronavirus Infections/virology , Disease Models, Animal , Female , Immunity, Cellular , Immunity, Humoral , Immunologic Memory , Lung/immunology , Lung/pathology , Lung/virology , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Macaca mulatta , Male , Nasal Mucosa/virology , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Recurrence , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/immunology , Viral Load , Virus Replication
6.
PLoS One ; 13(11): e0205139, 2018.
Article in English | MEDLINE | ID: mdl-30427829

ABSTRACT

BACKGROUND: Live, attenuated viral vectors that express HIV-1 antigens are being investigated as an approach to generating durable immune responses against HIV-1 in humans. We recently developed a replication-competent, highly attenuated Ad26 vector that expresses mosaic HIV-1 Env (rcAd26.MOS1.HIV-Env, "rcAd26"). Here we present the results of a first-in-human, placebo-controlled clinical trial to test the safety, immunogenicity and mucosal shedding of rcAd26 given orally. METHODS: Healthy adults were randomly assigned to receive a single oral dose of vaccine or placebo at 5:1 ratio in a dosage escalation of 10^8 to 10^11 rcAd26 VP (nominal doses) at University of Rochester Medical Center, Rochester, NY, USA. Participants were isolated and monitored for reactogenicity for 10 days post-vaccination, and adverse events were recorded up to day 112. Rectal and oropharyngeal secretions were evaluated for shedding of the vaccine. Humoral and cellular immune responses were measured. Household contacts were monitored for secondary vaccine transmission. RESULTS: We enrolled 22 participants and 11 household contacts between February 7 and June 24, 2015. 18 participants received one dose of HIV-1 vaccine and 4 participants received placebo. The vaccine caused only mild to moderate adverse events. No vaccine-related SAEs were observed. No infectious rcAd26 viral particles were detected in rectal or oropharyngeal secretions from any participant. Env-specific ELISA and ELISPOT responses were undetectable. No household contacts developed vaccine-induced HIV-1 seropositivity or vaccine-associated illness. CONCLUSIONS: The highly attenuated rcAd26.MOS1.HIV-Env vaccine was well tolerated up to 10^11 VP in healthy, HIV-1-uninfected adults, though the single dose was poorly immunogenic suggesting the replicative capacity of the vector was too attenuated. There was no evidence of shedding of infectious virus or secondary vaccine transmission following the isolation period. These data suggest the use of less attenuated viral vectors in future studies of live, oral HIV-1 vaccines. TRIAL REGISTRATION: ClinicalTrials.gov NCT02366013.


Subject(s)
AIDS Vaccines/therapeutic use , Acquired Immunodeficiency Syndrome/therapy , HIV-1/immunology , Immunity, Cellular/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Adenoviridae/genetics , Adult , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/therapeutic use , Female , Genetic Vectors/therapeutic use , HIV-1/genetics , HIV-1/pathogenicity , Humans , Immunity, Cellular/genetics , Male , Middle Aged , Virus Replication/drug effects , Virus Replication/immunology , Young Adult , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/therapeutic use
7.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29298888

ABSTRACT

Human and chimpanzee adenovirus vectors are being developed to circumvent preexisting antibodies against common adenovirus vectors such as Ad5. However, baseline immunity to these vectors still exists in human populations. Traditional cloning of new adenovirus vaccine vectors is a long and cumbersome process that takes 2 months or more and that requires rare unique restriction enzyme sites. Here we describe a novel, restriction enzyme-independent method for rapid cloning of new adenovirus vaccine vectors that reduces the total cloning procedure to 1 week. We developed 14 novel adenovirus vectors from rhesus monkeys that can be grown to high titers and that are immunogenic in mice. All vectors grouped with the unusual adenovirus species G and show extremely low seroprevalence in humans. Rapid cloning of novel adenovirus vectors is a promising approach for the development of new vector platforms. Rhesus adenovirus vectors may prove useful for clinical development.IMPORTANCE To overcome baseline immunity to human and chimpanzee adenovirus vectors, we developed 14 novel adenovirus vectors from rhesus monkeys. These vectors are immunogenic in mice and show extremely low seroprevalence in humans. Rhesus adenovirus vectors may prove useful for clinical development.


Subject(s)
Adenoviridae , Adenovirus Vaccines , Cloning, Molecular , Genetic Vectors , Immunogenicity, Vaccine/genetics , A549 Cells , Adenoviridae/genetics , Adenoviridae/immunology , Adenovirus Vaccines/genetics , Adenovirus Vaccines/immunology , Animals , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Macaca mulatta , Mice
8.
Nature ; 536(7617): 474-8, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27355570

ABSTRACT

Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.


Subject(s)
Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus Infection/virology , Zika Virus/immunology , Adoptive Transfer , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Specificity , Brazil , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Gene Deletion , Humans , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Mice , Microcephaly/complications , Microcephaly/virology , Vaccines, DNA/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Inactivated/chemistry , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/complications , Zika Virus Infection/immunology
9.
Clin Vaccine Immunol ; 22(11): 1166-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26376928

ABSTRACT

Replication-competent adenovirus (rcAd)-based vaccine vectors may theoretically provide immunological advantages over replication-incompetent Ad vectors, but they also raise additional potential clinical and regulatory issues. We produced replication-competent Ad serotype 26 (rcAd26) vectors by adding the E1 region back into a replication-incompetent Ad26 vector backbone with the E3 or E3/E4 regions deleted. We assessed the effect of vectorization on the replicative capacity of the rcAd26 vaccines. Attenuation occurred in a stepwise fashion, with E3 deletion, E4 deletion, and human immunodeficiency virus type 1 (HIV-1) envelope (Env) gene insertion all contributing to reduced replicative capacity compared to that with the wild-type Ad26 vector. The rcAd26 vector with E3 and E4 deleted and containing the Env transgene exhibited 2.7- to 4.4-log-lower replicative capacity than that of the wild-type Ad26 in vitro. This rcAd26 vector is currently being evaluated in a phase 1 clinical trial. Attenuation as a result of vectorization and transgene insertion has implications for the clinical development of replication-competent vaccine vectors.


Subject(s)
Adenovirus Vaccines/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Genetic Vectors , Virus Replication , Adenovirus Vaccines/immunology , Clinical Trials, Phase I as Topic , Gene Expression , Humans , Mutagenesis, Insertional , Serogroup , env Gene Products, Human Immunodeficiency Virus/genetics
10.
J Virol ; 89(3): 1512-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410856

ABSTRACT

UNLABELLED: Adenovirus vectors are widely used as vaccine candidates for a variety of pathogens, including HIV-1. To date, human and chimpanzee adenoviruses have been explored in detail as vaccine vectors. The phylogeny of human and chimpanzee adenoviruses is overlapping, and preexisting humoral and cellular immunity to both are exhibited in human populations worldwide. More distantly related adenoviruses may therefore offer advantages as vaccine vectors. Here we describe the primary isolation and vectorization of three novel adenoviruses from rhesus monkeys. The seroprevalence of these novel rhesus monkey adenovirus vectors was extremely low in sub-Saharan Africa human populations, and these vectors proved to have immunogenicity comparable to that of human and chimpanzee adenovirus vaccine vectors in mice. These rhesus monkey adenoviruses phylogenetically clustered with the poorly described adenovirus species G and robustly stimulated innate immune responses. These novel adenoviruses represent a new class of candidate vaccine vectors. IMPORTANCE: Although there have been substantial efforts in the development of vaccine vectors from human and chimpanzee adenoviruses, far less is known about rhesus monkey adenoviruses. In this report, we describe the isolation and vectorization of three novel rhesus monkey adenoviruses. These vectors exhibit virologic and immunologic characteristics that make them attractive as potential candidate vaccine vectors for both HIV-1 and other pathogens.


Subject(s)
Adenoviridae/classification , Adenoviridae/isolation & purification , Drug Carriers/isolation & purification , Genetic Vectors/isolation & purification , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae Infections/immunology , Adenoviridae Infections/veterinary , Adenoviridae Infections/virology , Africa South of the Sahara , Animals , Antibodies, Viral/blood , Cluster Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Humans , Macaca mulatta , Mice, Inbred BALB C , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA , Seroepidemiologic Studies , Vaccines, Synthetic/genetics
11.
J Virol ; 88(7): 3756-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24429370

ABSTRACT

UNLABELLED: Foreskin is the principal site of heterosexual HIV-1 infection in men. However, little is known about HIV-1-specific immune responses or inflammation in foreskin. To the best of our knowledge, no previous studies have assessed immune responses to candidate HIV-1 vaccines in foreskin. Using the rhesus monkey model, we show that intramuscular immunization with adenovirus serotype 26 and 35 vectors expressing SIV antigens elicited durable SIV Gag-specific CD4(+) and CD8(+) T cell responses in foreskin that were detectable for more than 1 year following vaccination. Gag-specific CD4(+) and CD8(+) T cells were also detectable in foreskin of SIV- and SHIV-infected animals and were at least comparable in magnitude to those in peripheral blood. However, unlike peripheral blood T cells, the majority of foreskin T cells exhibited transitional memory or effector memory phenotype and expressed higher levels of the activation markers CD69, HLA-DR, and CCR5, although vaccination did not further enhance foreskin CD4(+) T cell activation. These findings suggest that systemic vaccination strategies can elicit potentially important SIV-specific cellular immunity in foreskin. Further characterization of vaccine-elicited immune responses and inflammation in foreskin is warranted. IMPORTANCE: We demonstrate here the induction of SIV-specific cellular immune responses in foreskin by adenovirus serotype 26 and 35 vaccine vectors. Foreskin T cells were more activated than peripheral blood T cells, but foreskin T cells were not further activated by vaccination. These findings suggest that alternative serotype adenovirus vectors induce potentially important immune responses in foreskin.


Subject(s)
Adenoviridae/genetics , Drug Carriers , Foreskin/immunology , Genetic Vectors , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Animals , Immunophenotyping , Injections, Intramuscular , Macaca mulatta , Male , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Time Factors
12.
Cell ; 155(3): 531-9, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243013

ABSTRACT

The global diversity of HIV-1 represents a critical challenge facing HIV-1 vaccine development. HIV-1 mosaic antigens are bioinformatically optimized immunogens designed for improved coverage of HIV-1 diversity. However, the protective efficacy of such global HIV-1 vaccine antigens has not previously been evaluated. Here, we demonstrate the capacity of bivalent HIV-1 mosaic antigens to protect rhesus monkeys against acquisition of infection following heterologous challenges with the difficult-to-neutralize simian-human immunodeficiency virus SHIV-SF162P3. Adenovirus/poxvirus and adenovirus/adenovirus vector-based vaccines expressing HIV-1 mosaic Env, Gag, and Pol afforded a significant reduction in the per-exposure acquisition risk following repetitive, intrarectal SHIV-SF162P3 challenges. Protection against acquisition of infection correlated with vaccine-elicited binding, neutralizing, and functional nonneutralizing antibodies, suggesting that the coordinated activity of multiple antibody functions may contribute to protection against difficult-to-neutralize viruses. These data demonstrate the protective efficacy of HIV-1 mosaic antigens and suggest a potential strategy for the development of a global HIV-1 vaccine. PAPERCLIP:


Subject(s)
AIDS Vaccines/immunology , HIV-1 , Animals , Antibody Formation , Female , HIV Antigens/immunology , Human Immunodeficiency Virus Proteins/immunology , Immunity, Cellular , Macaca mulatta , Male , Molecular Sequence Data , Specific Pathogen-Free Organisms
13.
Cell ; 151(2): 253-66, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063120

ABSTRACT

Pathogenic simian immunodeficiency virus (SIV) infection is associated with enteropathy, which likely contributes to AIDS progression. To identify candidate etiologies for AIDS enteropathy, we used next-generation sequencing to define the enteric virome during SIV infection in nonhuman primates. Pathogenic, but not nonpathogenic, SIV infection was associated with significant expansion of the enteric virome. We identified at least 32 previously undescribed enteric viruses during pathogenic SIV infection and confirmed their presence by using viral culture and PCR testing. We detected unsuspected mucosal adenovirus infection associated with enteritis as well as parvovirus viremia in animals with advanced AIDS, indicating the pathogenic potential of SIV-associated expansion of the enteric virome. No association between pathogenic SIV infection and the family-level taxonomy of enteric bacteria was detected. Thus, enteric viral infections may contribute to AIDS enteropathy and disease progression. These findings underline the importance of metagenomic analysis of the virome for understanding AIDS pathogenesis.


Subject(s)
Caliciviridae/isolation & purification , Intestines/virology , Parvoviridae/isolation & purification , Picornaviridae/isolation & purification , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Caliciviridae/classification , Caliciviridae/genetics , Chlorocebus aethiops , Feces/microbiology , Feces/virology , Intestines/microbiology , Molecular Sequence Data , Parvoviridae/classification , Parvoviridae/genetics , Phylogeny , Picornaviridae/classification , Picornaviridae/genetics , Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/microbiology , Simian Immunodeficiency Virus/pathogenicity
14.
Nature ; 482(7383): 89-93, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22217938

ABSTRACT

Preclinical studies of human immunodeficiency virus type 1 (HIV-1) vaccine candidates have typically shown post-infection virological control, but protection against acquisition of infection has previously only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant simian immunodeficiency virus (SIV) challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus-vector-based vaccines expressing SIV(SME543) Gag, Pol and Env antigens resulted in an 80% or greater reduction in the per-exposure probability of infection against repetitive, intrarectal SIV(MAC251) challenges in rhesus monkeys. Protection against acquisition of infection showed distinct immunological correlates compared with post-infection virological control and required the inclusion of Env in the vaccine regimen. These data demonstrate the proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.


Subject(s)
Antibodies, Neutralizing/immunology , Macaca mulatta/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Female , HIV-1/immunology , Male , Neutralization Tests , Viral Vaccines/immunology
15.
J Virol ; 86(2): 1267-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072746

ABSTRACT

The immunogenicity of adenovirus serotype 5 (Ad5) vectors has been shown to be suppressed by neutralizing antibodies (NAbs) directed primarily against the hexon hypervariable regions (HVRs). We previously reported that replacing all seven HVRs with those from the rare serotype virus Ad48 resulted in a chimeric Ad5HVR48(1-7) vector that largely evaded preexisting Ad5 immunity in mice and rhesus monkeys. In this study, we evaluated the extent to which Ad5-specific NAbs are directed against various HVRs. We constructed partial HVR-chimeric Ad5 vectors with only a subset of HVRs exchanged, and we utilized these vectors in both NAb assays and murine immunogenicity studies with and without baseline Ad5 immunity. Our results demonstrate that Ad5-specific NAbs target multiple HVRs, suggesting that replacing all HVRs is required to optimize evasion of anti-Ad5 immunity. These data have important implications for the development of novel vectors for both vaccines and gene therapy.


Subject(s)
Adenovirus Infections, Human/immunology , Adenoviruses, Human/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid Proteins/immunology , Adenovirus Infections, Human/virology , Adenoviruses, Human/genetics , Animals , Capsid Proteins/genetics , Genetic Therapy/instrumentation , Genetic Vectors/genetics , Genetic Vectors/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests
16.
Vaccine ; 29(32): 5203-9, 2011 Jul 18.
Article in English | MEDLINE | ID: mdl-21619905

ABSTRACT

Recombinant adenovirus serotype 5 (rAd5) vaccine vectors for HIV-1 and other pathogens have been shown to be limited by high titers of Ad5 neutralizing antibodies (NAbs) in the developing world. Alternative serotype rAd vectors have therefore been constructed. Here we report Ad5, Ad26, Ad35, and Ad48 NAb titers in 4381 individuals from North America, South America, sub-Saharan Africa, and Southeast Asia. As expected, Ad5 NAb titers were both frequent and high magnitude in sub-Saharan Africa and Southeast Asia. In contrast, Ad35 NAb titers proved infrequent and low in all regions studied, and Ad48 NAbs were rare in all regions except East Africa. Ad26 NAbs were moderately common in adults in sub-Saharan Africa and Southeast Asia, but Ad26 NAb titers proved markedly lower than Ad5 NAb titers in all regions, and these relatively low Ad26 NAb titers did not detectably suppress the immunogenicity of 4×10(10)vp of a rAd26-Gag/Pol/Env/Nef vaccine in rhesus monkeys. These data inform the clinical development of alternative serotype rAd vaccine vectors in the developing world.


Subject(s)
Adenoviridae Infections/epidemiology , Adenoviridae Infections/immunology , Adenoviridae/immunology , Antibodies, Viral/immunology , Viral Vaccines/immunology , AIDS Vaccines/immunology , Adenoviridae/isolation & purification , Adenoviridae Infections/blood , Adenoviridae Infections/virology , Adolescent , Adult , Animals , Antibodies, Viral/blood , Child , HIV-1/immunology , Humans , Infant , Macaca mulatta/immunology , Vaccines, Synthetic/immunology , Young Adult
17.
Nat Med ; 16(3): 319-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20173752

ABSTRACT

The worldwide diversity of HIV-1 presents an unprecedented challenge for vaccine development. Antigens derived from natural HIV-1 sequences have elicited only a limited breadth of cellular immune responses in nonhuman primate studies and clinical trials to date. Polyvalent 'mosaic' antigens, in contrast, are designed to optimize cellular immunologic coverage of global HIV-1 sequence diversity. Here we show that mosaic HIV-1 Gag, Pol and Env antigens expressed by recombinant, replication-incompetent adenovirus serotype 26 vectors markedly augmented both the breadth and depth without compromising the magnitude of antigen-specific T lymphocyte responses as compared with consensus or natural sequence HIV-1 antigens in rhesus monkeys. Polyvalent mosaic antigens therefore represent a promising strategy to expand cellular immunologic vaccine coverage for genetically diverse pathogens such as HIV-1.


Subject(s)
AIDS Vaccines/immunology , AIDS Vaccines/pharmacology , HIV-1/immunology , Immunity, Cellular , Animals , Antibody Formation/immunology , Antibody Formation/physiology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/physiology , HIV Antigens/immunology , HIV Protease/immunology , Immunity, Cellular/immunology , Immunity, Cellular/physiology , Lymphocyte Activation/immunology , Lymphocyte Activation/physiology , Macaca mulatta/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Vaccines, Synthetic , env Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology
18.
Int J Oncol ; 28(3): 747-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465381

ABSTRACT

In murine erythroleukemia (MEL) A20 cells (grown in 20 ng/ml adriamycin), mutation(s) producing 10-fold adriamycin (doxorubicin) resistance emerged via an unknown mechanism. Exposure of A20 cells to further stepwise increasing concentrations of ADR in combination with MDR modulators (PSC833 and verapamil) aimed to amplify the undetermined A20 mechanism while controlling P-glycoprotein (P-gp) overexpression. The growth of the derived cell lines A30P, A40P and A60P (grown in 30, 40 and 60 ng/ml ADR with PSC833 and verapamil) was initially slow, but eventually reached near WT rates. The new cell lines A30P and A40P were only 1.3- and 1.6-fold more resistant to adriamycin than PC4 A20. Resistance to vincristine was unchanged, but resistance to etoposide (VP-16) was 3.7-fold higher in A40P than A20 (itself 97-fold higher than wild-type). Expression of mdr3 and mrp mRNA tested by RT-PCR showed no increase. Daunorubicin and etoposide accumulation was not different among the cell lines, and no changes were detected in the number of daunorubicin fluorescent lysosomes. In comparison to WT, reduced topoisomerase IIalpha (EC 5.99.1.3) activity (20%) and protein expression (80%) was similar to the parental A20 cells. No mutations in the coding sequence of topoisomerase IIalpha could be located to account for the high etoposide resistance levels. The inhibitor combination of verapamil and PSC833 prevented the emergence of transporter mediated MDR, but not ADR selection of cell lines highly resistant to etoposide.


Subject(s)
Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Etoposide/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cyclosporins/pharmacology , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Daunorubicin/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Verapamil/pharmacology
19.
Proc Natl Acad Sci U S A ; 102(5): 1436-41, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15659548

ABSTRACT

Retroviral DNA integration occurs throughout the genome; however, local "hot spots" exist where a strong preference for certain sites over others are seen, and more global preferences associated with genes have been reported. Previous data from our laboratory suggested that there are fewer integration events into a DNA template when it is undergoing active transcription than when it is not. Because these data were generated by using a stably transfected foreign gene that was only weakly inducible, we have extended this observation by comparing integration events into a highly inducible endogenous gene under both induced and uninduced transcriptional states. To examine the influence of transcription on site selection directly, we analyzed the frequency and distribution of integration of avian retrovirus DNA into the metallothionein gene, before and after its induction to a highly sustained level of expression by addition of ZnSO4. We found a 6-fold reduction in integration events after 100-fold induction of transcription. This result implies that, despite an apparent preference for integration of retroviral DNA into transcribed regions of host DNA, high-level transcription can be inhibitory to the integration process. Several possible models for our observation are as follows. First, when a DNA template is undergoing active transcription, integration might be blocked by the RNA polymerase II complex because of steric hindrance. Alternatively, the integrase complex may require DNA to be in a double-stranded conformation, which would not be the case during active transcription. Last, transcription might lead to remodeling of chromatin into a structure that is less favorable for integration.


Subject(s)
Alpharetrovirus/genetics , DNA, Viral/genetics , Transcription, Genetic/genetics , Virus Integration/genetics , Animals , Base Sequence , Cloning, Molecular , DNA Primers , DNA Replication/drug effects , Molecular Sequence Data , Polymerase Chain Reaction/methods , Quail
20.
J Virol ; 77(12): 6709-19, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12767991

ABSTRACT

Alpharetroviruses provide a useful system for the study of the molecular mechanisms of host range and receptor interaction. These viruses can be divided into subgroups based on diverse receptor usage due to variability within the two host range determining regions, hr1 and hr2, in their envelope glycoprotein SU (gp85). In previous work, our laboratory described selection from a subgroup B avian sarcoma-leukosis virus of an extended-host-range variant (LT/SI) with two adjacent amino acid substitutions in hr1. This virus retains its ability to use the subgroup BD receptor but can also infect QT6/BD cells, which bear a related subgroup E receptor (R. A. Taplitz and J. M. Coffin, J. Virol 71:7814-7819, 1997). Here, we report further analysis of this unusual variant. First, one (L154S) of the two substitutions is sufficient for host range extension, while the other (T155I) does not alter host range. Second, these mutations extend host range to non-avian cell types, including human, dog, cat, mouse, rat, and hamster. Third, interference experiments imply that the mutants interact efficiently with the subgroup BD receptor and possibly the related subgroup E receptor, but they have another means of entry that is not dependent on these interactions. Fourth, binding studies indicate that the mutant SU proteins retain the ability to interact as monomers with subgroup BD and BDE receptors but only bind the subgroup E receptor in the context of an Env trimer. Further, the mutant SU proteins bind well to chicken cells but do not bind any better than wild-type subgroup B to QT6 or human cells, even though the corresponding viruses are capable of infecting these cells.


Subject(s)
Avian Leukosis Virus/pathogenicity , Avian Sarcoma Viruses/pathogenicity , Immunoglobulin G/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Avian Leukosis Virus/genetics , Avian Sarcoma Viruses/genetics , Cats , Cell Line , Cells, Cultured , Chick Embryo , Chickens , Cricetinae , Dogs , Humans , Immunoglobulin G/genetics , Mice , Molecular Sequence Data , Mutation , Rats , Recombination, Genetic , Species Specificity , Transfection , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...