Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Ther Methods Clin Dev ; 17: 1088-1096, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32478124

ABSTRACT

Antibody-like molecules were evaluated with potent simian immunodeficiency virus (SIV) neutralizing properties (immunoadhesins) that were delivered by a recombinant adeno-associated virus (rAAV) vector in the SIV-infected rhesus macaque model. When injected intramuscularly into the host, the vector directs in vivo production of the transgenes with antibody-like binding properties that lead to serum neutralizing activity against SIV. To extend the half-life of the immunoadhesins, rhesus cluster of differentiation 4 (CD4) and a single-chain antibody (4L6) were fused with albumin molecules, and these constructs were tested in our model of SIV infection. Antibody-based immunoadhesins provided high serum neutralizing titers against the original SIV strain. CD4-based immunoadhesins provided a wider spectrum of neutralization against different SIV strains in comparison to antibody-based therapeutics and had the potential to protect against high viral challenging doses. Although the albumin-antibody fusion immunoadhesin provided strong and prolonged protection of the immunized animals against SIV challenge, the albumin-CD4 fusion altered the specificity and decreased the overall protection effectiveness of the immunoadhesin in comparison to the antibody-based molecules. Albumin-based immunoadhesins increase in vivo longevity of the immune protection; however, they present challenges likely linked to the induction of anti-immunoadhesin antibodies.

2.
Annu Rev Virol ; 4(1): 491-510, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28645240

ABSTRACT

This review discusses recent progress made in developing a vaccine and novel treatments for human immunodeficiency virus (HIV). It highlights the shortcomings of the RV144 vaccination trial [ALVAC-HIV (vCP1521) and AIDSVAX B/E] and the current standard of care and proposes that engineered expression of broadly neutralizing antibodies (bNAbs) against HIV-1 could overcome these shortcomings. Current developments in three major lines of research on HIV prevention and treatment using bNAbs are reviewed: firstly, the use of sequential immunogens to activate B cells to express bNAbs; secondly, the delivery of novel and extremely potent bNAbs through passive administration; and finally, the use of gene transfer using adeno-associated viral vectors to deliver bNAbs.


Subject(s)
Antibodies, Neutralizing/therapeutic use , HIV Antibodies/biosynthesis , HIV Infections/prevention & control , HIV Infections/therapy , HIV-1/immunology , AIDS Vaccines/immunology , AIDS Vaccines/therapeutic use , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/genetics , B-Lymphocytes/immunology , Clinical Trials as Topic , Genetic Engineering , Genetic Therapy , Genetic Vectors , HIV Antibodies/genetics , HIV Antibodies/therapeutic use , HIV Infections/immunology , HIV Infections/virology , Humans , Immunization, Passive , Mice , env Gene Products, Human Immunodeficiency Virus
3.
Health Aff (Millwood) ; 35(11): 2142-2147, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27834256

ABSTRACT

Nearly one in four deaths globally are due to environmental hazards such as air and water pollution, according to the World Health Organization. However, knowledge about how the environment affects health and health equity outcomes has not been well integrated into decisions that shape the conditions in which people live, work, and play. To address this challenge, US foundations have invested millions of dollars to make it easier to incorporate environmental health information into decisions ranging from family purchases and governmental policy making to business, medical, and other professional practices. This article summarizes grant making aimed at improving environmental conditions to improve health and health equity outcomes. We provide examples of environmental health grants that focus on tools that the public, policy makers, and professionals can use in making decisions. We found that the investment in and attention to environmental factors, including in work addressing social determinants of health, have been insufficient to realize the potential for reducing negative environmental impacts on health and health disparities. We argue that the grant making highlighted here has increased knowledge that could enable more widespread consideration of environmental health in many decisions, with positive effects on health and health equity.


Subject(s)
Environmental Health/methods , Environmental Health/standards , Financing, Organized/economics , Foundations/economics , Policy Making , Public Policy , Health Equity , Humans , Social Determinants of Health
4.
Hum Gene Ther ; 27(1): 32-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650966

ABSTRACT

Gene augmentation therapy as a strategy to treat alpha-1 antitrypsin (AAT) deficiency has reached phase 2 clinical testing in humans. Sustained serum levels of AAT have been observed beyond one year after intramuscular administration of a recombinant adeno-associated virus (rAAV) vector expressing the AAT gene. In this study, sequential muscle biopsies obtained at 3 and 12 months after vector injection were examined for the presence of rAAV vector genomes. Each biopsy sample contained readily detectable vector DNA, the majority of which existed as double-stranded supercoiled and open circular episomes. Episomes persisted through 12 months, although at slightly lower levels than observed at 3 months. There was a clear dose response when comparing the low- and mid-vector-dose groups to the high-dose group. The highest absolute copy numbers were found in a high-dose subject, and serum AAT levels at 12 months confirmed that the high-dose group also had the highest sustained serum AAT levels. Sequence analysis revealed that the vast majority of episomes contained double-D inverted terminal repeats ranging from fully intact to severely deleted. Molecular clones of vector genomes derived directly from the biopsies were transcriptionally active, potentially identifying them as the source of serum AAT in the trial subjects.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Muscle, Skeletal/metabolism , Plasmids/genetics , Base Sequence/genetics , Dependovirus/metabolism , Genetic Vectors/genetics , Genome , Humans , Plasmids/metabolism
5.
Vaccines (Basel) ; 3(2): 429-47, 2015 May 26.
Article in English | MEDLINE | ID: mdl-26343196

ABSTRACT

Vaccination has proven to be an invaluable means of preventing infectious diseases by reducing both incidence of disease and mortality. However, vaccines have not been effectively developed for many diseases including HIV-1, hepatitis C virus (HCV), tuberculosis and malaria, among others. The emergence of new technologies with a growing understanding of host-pathogen interactions and immunity may lead to efficacious vaccines against pathogens, previously thought impossible.

6.
Adv Exp Med Biol ; 848: 149-67, 2015.
Article in English | MEDLINE | ID: mdl-25757620

ABSTRACT

This chapter discusses the emerging field of vector-mediated antibody gene transfer as an alternative vaccine for infectious disease, with a specific focus on HIV. However, this methodology need not be confined to HIV-1; the general strategy of vector-mediated antibody gene transfer can be applied to other difficult vaccine targets like hepatitis C virus, malaria, respiratory syncytial virus, and tuberculosis. This approach is an improvement over classical passive immunization strategies that administer antibody proteins to the host to provide protection from infection. With vector-mediated gene transfer, the antibody gene is delivered to the host, via a recombinant adeno-associated virus (rAAV) vector; this in turn results in long-term endogenous antibody expression from the injected muscle that confers protective immunity. Vector-mediated antibody gene transfer can rapidly move existing, potent broadly cross-neutralizing HIV-1-specific antibodies into the clinic. The gene transfer products demonstrate a potency and breadth identical to the original product. This strategy eliminates the need for immunogen design and interaction with the adaptive immune system to generate protection, a strategy that so far has shown limited promise.


Subject(s)
Antibodies, Neutralizing/genetics , Communicable Diseases/therapy , Gene Transfer Techniques , Genes, Immunoglobulin , Genetic Vectors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , HIV Antibodies/administration & dosage , HIV Antibodies/genetics , HIV Infections/therapy , HIV-1/immunology , Humans , Immunization, Passive/methods , Vaccines, DNA/administration & dosage
7.
Curr Opin HIV AIDS ; 9(3): 250-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24638019

ABSTRACT

PURPOSE OF REVIEW: In the present review, we will discuss the emerging field of vector-mediated antibody gene transfer as an alternative HIV vaccine. This approach is an improvement over classical passive immunization strategies that administer antibodies to the host to provide protection from infection. With vector-mediated gene transfer, the antibody gene is delivered to the host, resulting in long-term endogenous antibody expression from the injected muscle that confers protective immunity. RECENT FINDINGS: Large numbers of very potent and broadly neutralizing HIV antibodies have recently been isolated and characterized. Vector-mediated antibody gene transfer allows one to immediately use these antibodies as a vaccine. Gene transfer studies in both mice and monkeys demonstrate long-term antibody expression in serum from a single injection at concentrations that provide sterilizing immunity. SUMMARY: Vector-mediated antibody gene transfer can rapidly move existing, potent anti-HIV molecules into the clinic. The gene transfer products demonstrate a potency and breadth identical to the original product. This strategy eliminates the need for immunogen design and interaction with the adaptive immune system to generate protection, a strategy that so far has shown little promise.


Subject(s)
AIDS Vaccines/administration & dosage , Antibodies, Neutralizing/administration & dosage , Dependovirus/genetics , Drug Delivery Systems/methods , Gene Transfer Techniques , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Haplorhini , Mice
8.
Nature ; 507(7491): 201-6, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24499818

ABSTRACT

Vaccines prevent infectious disease largely by inducing protective neutralizing antibodies against vulnerable epitopes. Several major pathogens have resisted traditional vaccine development, although vulnerable epitopes targeted by neutralizing antibodies have been identified for several such cases. Hence, new vaccine design methods to induce epitope-specific neutralizing antibodies are needed. Here we show, with a neutralization epitope from respiratory syncytial virus, that computational protein design can generate small, thermally and conformationally stable protein scaffolds that accurately mimic the viral epitope structure and induce potent neutralizing antibodies. These scaffolds represent promising leads for the research and development of a human respiratory syncytial virus vaccine needed to protect infants, young children and the elderly. More generally, the results provide proof of principle for epitope-focused and scaffold-based vaccine design, and encourage the evaluation and further development of these strategies for a variety of other vaccine targets, including antigenically highly variable pathogens such as human immunodeficiency virus and influenza.


Subject(s)
Drug Design , Epitopes/chemistry , Epitopes/immunology , Protein Stability , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Amino Acid Motifs , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Macaca mulatta/immunology , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Neutralization Tests , Protein Conformation , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/immunology
9.
Microbiol Spectr ; 2(4): AID-0016-2014, 2014 Aug.
Article in English | MEDLINE | ID: mdl-26104192

ABSTRACT

This article focuses on a novel vaccine strategy known as vector-mediated antibody gene transfer, with a particular focus on human immunodeficiency virus (HIV). This strategy provides a solution to the problem of current vaccines that fail to generate neutralizing antibodies to prevent HIV-1 infection and AIDS. Antibody gene transfer allows for predetermination of antibody affinity and specificity prior to "immunization" and avoids the need for an active humoral immune response against the HIV envelope protein. This approach uses recombinant adeno-associated viral (rAAV) vectors, which have been shown to transduce muscle with high efficiency and direct the long-term expression of a variety of transgenes, to deliver the gene encoding a broadly neutralizing antibody into the muscle. Following rAAV vector gene delivery, the broadly neutralizing antibodies are endogenously synthesized in myofibers and passively distributed to the circulatory system. This is an improvement over classical passive immunization strategies that administer antibody proteins to the host to provide protection from infection. Vector-mediated gene transfer studies in mice and monkeys with anti-HIV and simian immunodeficiency virus (SIV)-neutralizing antibodies demonstrated long-lasting neutralizing activity in serum with complete protection against intravenous challenge with virulent HIV and SIV. These results indicate that existing potent anti-HIV antibodies can be rapidly moved into the clinic. However, this methodology need not be confined to HIV. The general strategy of vector-mediated antibody gene transfer can be applied to other difficult vaccine targets such as hepatitis C virus, malaria, respiratory syncytial virus, and tuberculosis.


Subject(s)
Adenoviridae/genetics , Gene Expression , Genetic Vectors , HIV Antibodies/immunology , HIV/immunology , Immunization, Passive/methods , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , HIV Antibodies/genetics , Haplorhini , Humans , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transduction, Genetic
10.
Curr Top Microbiol Immunol ; 372: 307-24, 2013.
Article in English | MEDLINE | ID: mdl-24362696

ABSTRACT

A respiratory syncytial virus (RSV) vaccine has remained elusive for decades, largely due to the failure of a formalin-inactivated RSV vaccine in the 1960s that resulted in enhanced disease upon RSV exposure in the immunized individuals. Vaccine development has also been hindered by the incomplete immunity conferred by natural infection allowing for re-infection at any time, and the immature immune system and circulating maternal antibodies present in the neonate, the primary target for a vaccine. This chapter will review the use of gene delivery, both nonviral and viral, as a potential vaccine approach for human RSV. Many of these gene-based vaccines vectors elicit protective immune responses in animal models. None of the RSV gene-based platforms have progressed into clinical trials, mostly due to uncertainty regarding the direct translation of animal model results to humans and the hesitancy to invest in costly clinical trials with the potential for unclear and complicated immune responses. The continued development of RSV vaccine gene-based approaches is warranted because of their inherent flexibility with regard to composition and administration. It is likely that multiple candidate vaccines will reach human testing in the next few years.


Subject(s)
Gene Transfer Techniques , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Vaccines, DNA/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Alphavirus/genetics , Alphavirus/immunology , Animals , Child, Preschool , Dependovirus/genetics , Dependovirus/immunology , Genetic Vectors/immunology , Host Specificity , Humans , Infant , Mice , Poxviridae/genetics , Poxviridae/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Human/pathogenicity , Vaccines, DNA/administration & dosage
11.
Science ; 340(6136): 1232910, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23723240

ABSTRACT

Vaccines are among the greatest successes in the history of public health. However, past strategies for vaccine development are unlikely to succeed in the future against major global diseases such as AIDS, tuberculosis, and malaria. For such diseases, the correlates of protection are poorly defined and the pathogens evade immune detection and/or exhibit extensive genetic variability. Recent advances have heralded in a new era of vaccine discovery. However, translation of these advances into vaccines remains impeded by lack of understanding of key vaccinology principles in humans. We review these advances toward vaccine discovery and suggest that for accelerating successful vaccine development, new human immunology-based clinical research initiatives be implemented with the goal of elucidating and more effectively generating vaccine-induced protective immune responses.


Subject(s)
Communicable Disease Control , Vaccines/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antigens/genetics , Antigens/immunology , Antigens/isolation & purification , Disease Models, Animal , Drug Delivery Systems , Humans , Malaria/prevention & control , Tuberculosis/prevention & control , Vaccines/administration & dosage
12.
J Virol ; 86(1): 246-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013035

ABSTRACT

Successful live attenuated vaccines mimic natural exposure to pathogens without causing disease and have been successful against several viruses. However, safety concerns prevent the development of attenuated human immunodeficiency virus (HIV) as a vaccine candidate. If a safe, replicating virus vaccine could be developed, it might have the potential to offer significant protection against HIV infection and disease. Described here is the development of a novel self-replicating chimeric virus vaccine candidate that is designed to provide natural exposure to a lentivirus-like particle and to incorporate the properties of a live attenuated virus vaccine without the inherent safety issues associated with attenuated lentiviruses. The genome from the alphavirus Venezuelan equine encephalitis virus (VEE) was modified to express SHIV89.6P genes encoding the structural proteins Gag and Env. Expression of Gag and Env from VEE RNA in primate cells led to the assembly of particles that morphologically and functionally resembled lentivirus virions and that incorporated alphavirus RNA. Infection of CD4⁺ cells with chimeric lentivirus-like particles was specific and productive, resulting in RNA replication, expression of Gag and Env, and generation of progeny chimeric particles. Further genome modifications designed to enhance encapsidation of the chimeric virus genome and to express an attenuated simian immunodeficiency virus (SIV) protease for particle maturation improved the ability of chimeric lentivirus-like particles to propagate in cell culture. This study provides proof of concept for the feasibility of creating chimeric virus genomes that express lentivirus structural proteins and assemble into infectious particles for presentation of lentivirus immunogens in their native and functional conformation.


Subject(s)
Chimera/physiology , Encephalitis Virus, Venezuelan Equine/physiology , Genetic Vectors/physiology , Virus Replication , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Cell Line , Chimera/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Gene Expression , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , Genetic Vectors/genetics , HIV Infections/prevention & control , Humans , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Virus Assembly
13.
AIDS Res Hum Retroviruses ; 26(8): 933-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20666584

ABSTRACT

The recombinant vaccine, tgAAC09, based on an adeno-associated virus serotype 2 (AAV2) vector encoding HIV-1 subtype C Gag, protease, and part of reverse transcriptase, induced robust T cell and antibody responses in nonhuman primates. In a previous phase I study in 80 healthy HIV-seronegative European and Indian adults, the vaccine was generally safe, well tolerated, and modestly immunogenic when administered once at doses up to 3 x 10(11) DRP. This phase II double-blind, randomized, placebo-controlled trial tested two administrations and a higher dosage of tgAAC009. Ninety-one healthy HIV-seronegative adults from three African countries were given one of three dosage levels of tgAAC09 (3 x 10(10), 3 x 10(11), or 3 x 10(12) DRP) intramuscularly, either at a 6- or 12-month interval; follow-up was 18 months. Overall, 65% and 57% of vaccine recipients experienced local and systemic signs and symptoms, respectively, most being mild. Frequency and severity were not dose related and were similar to those in placebo recipients. No vaccine-related serious adverse events were reported. Overall, HIV-specific T cell responses were detected by IFN-gamma ELISPOT in 17/69 (25%) vaccine recipients with 38% (10/26) responders in the highest dosage group. The response rate improved significantly with boosting at 6, but not 12 months, in the 3 x 10(11) and 3 x 10(12) dosage groups only. Neutralizing antibody titers to the AAV2 did not alter the frequency of immune responses to HIV. Two doses of tgAAC09 were well tolerated at the dosage levels given. Fewer than half the recipients of the highest vaccine dosage, 3 x 10(12) DRP, had T cell responses to HIV.


Subject(s)
AIDS Vaccines/administration & dosage , HIV Infections/prevention & control , AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Adult , Antibodies, Neutralizing/blood , Antibody Formation , Dependovirus/immunology , Double-Blind Method , Female , Genetic Vectors/immunology , HIV Antibodies/blood , HIV Infections/immunology , HIV-1/immunology , Humans , Immunization Schedule , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Young Adult
15.
Infect Immun ; 77(8): 3380-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19487468

ABSTRACT

Current evidence suggests that protective antigen (PA)-based anthrax vaccines may elicit a narrow neutralizing antibody repertoire, and this may represent a vulnerability with PA-based vaccines. In an effort to identify neutralizing specificities which may complement those prevalent in PA antiserum, we evaluated whether sequences within the 2beta2-2beta3 loop of PA, which are apparent in the crystal structure of heptameric but not monomeric PA, might represent a target for an epitope-specific vaccine for anthrax and, further, whether antibodies to these sequences are induced in rabbits immunized with monomeric PA. We evaluated the immunogenicity in rabbits of a multiple antigenic peptide (MAP) displaying copies of amino acids (aa) 305 to 319 of this region. Overall, four out of six rabbits vaccinated with the MAP peptide in Freund's adjuvant developed high-titer, high-avidity antibody responses which cross-reacted with the immobilized peptide sequence comprising aa 305 to 319 and with PA, as determined by an enzyme-linked immunosorbent assay, and which displayed potent and durable neutralization of lethal toxin (LeTx) in vitro, with peak titers which were 452%, 100%, 67%, and 41% of the peak neutralization titers observed in positive-control rabbits immunized with PA. Importantly, analysis of sera from multiple cohorts of rabbits with high-titer immunity to PA demonstrated a virtual absence of this potent antibody specificity, and work by others suggests that this specificity may be present at only low levels in primate PA antiserum. These results highlight the potential importance of this immunologically cryptic neutralizing epitope from PA as a target for alternative and adjunctive vaccines for anthrax.


Subject(s)
Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Epitopes/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Bacterial/blood , Enzyme-Linked Immunosorbent Assay , Female , Neutralization Tests , Rabbits
16.
Nat Med ; 15(8): 901-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19448633

ABSTRACT

The key to an effective HIV vaccine is development of an immunogen that elicits persisting antibodies with broad neutralizing activity against field strains of the virus. Unfortunately, very little progress has been made in finding or designing such immunogens. Using the simian immunodeficiency virus (SIV) model, we have taken a markedly different approach: delivery to muscle of an adeno-associated virus gene transfer vector expressing antibodies or antibody-like immunoadhesins having predetermined SIV specificity. With this approach, SIV-specific molecules are endogenously synthesized in myofibers and passively distributed to the circulatory system. Using such an approach in monkeys, we have now generated long-lasting neutralizing activity in serum and have observed complete protection against intravenous challenge with virulent SIV. In essence, this strategy bypasses the adaptive immune system and holds considerable promise as a unique approach to an effective HIV vaccine.


Subject(s)
Antibodies, Viral/immunology , Genetic Vectors/immunology , Haplorhini/immunology , SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Viral/metabolism , CD4 Immunoadhesins/immunology , Gene Transfer Techniques/veterinary , Haplorhini/genetics , Immunotherapy/methods , Immunotherapy/veterinary , Models, Biological , Neutralization Tests , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Treatment Outcome
17.
Mol Ther ; 17(2): 373-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19002162

ABSTRACT

Bacillus anthracis represents a formidable bioterrorism and biowarfare threat for which new vaccines are needed with improved safety and efficacy over current options. Toward this end, we created recombinant adeno-associated virus type 1 (rAAV1) vectors containing synthetic genes derived from the protective antigen (PA) or lethal factor (LF) of anthrax lethal toxin (LeTx) and tested them for immunogenicity and induction of toxin-neutralizing antibodies in rabbits. Codon-optimized segments encoding activated PA (PA63), or LF, were synthesized and cloned into optimized rAAV1 vectors containing a human cytomegalovirus (hCMV) promoter and synthetic optimized leader. Serum from rabbits immunized intramuscularly with rAAV1/PA (monovalent), rAAV1/LF (monovalent), rAAV1/PA + rAAV1/LF (bivalent), or rAAV1/enhanced green fluorescent protein (control) exhibited substantial PA- and LF-specific antibody responses at 4 weeks by both western blot (> 1:10,000 dilution) and enzyme-linked immunosorbent assay (ELISA) (mean end-point titer: 32,000-260,000), and contained anthrax LeTx-neutralizing activity in vitro, with peak titers approximating those of a rabbit hyperimmune antisera raised against soluble PA and LF. Compared to the monovalent groups (rAAV1/PA or rAAV1/LF), the bivalent group (rAAV1/PA + rAAV1/LF) exhibited marginally higher ELISA and neutralization activity with dual specificity for both PA and LF. The finding of robust neutralizing antibody responses after a single injection of these rAAV1-based vectors supports their further development as candidate anthrax vaccines.


Subject(s)
Anthrax Vaccines/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Animals , Anthrax Vaccines/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Blotting, Western , Cytomegalovirus/genetics , Enzyme-Linked Immunosorbent Assay , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Promoter Regions, Genetic/genetics , Rabbits
18.
J Virol ; 83(3): 1456-64, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19019948

ABSTRACT

Adeno-associated virus (AAV) replication and biology have been extensively studied using cell culture systems, but there is precious little known about AAV biology in natural hosts. As part of our ongoing interest in the in vivo biology of AAV, we previously described the existence of extrachromosomal proviral AAV genomes in human tissues. In the current work, we describe the molecular structure of infectious DNA clones derived directly from these tissues. Sequence-specific linear rolling-circle amplification was utilized to isolate clones of native circular AAV DNA. Several molecular clones containing unit-length viral genomes directed the production of infectious wild-type AAV upon DNA transfection in the presence of adenovirus help. DNA sequence analysis of the molecular clones revealed the ubiquitous presence of a double-D inverted terminal repeat (ITR) structure, which implied a mechanism by which the virus is able to maintain ITR sequence continuity and persist in the absence of host chromosome integration. These data suggest that the natural life cycle of AAV, unlike that of retroviruses, might not have genome integration as an obligatory component.


Subject(s)
Dependovirus/pathogenicity , Adolescent , Base Sequence , Blotting, Western , Child , Child, Preschool , Cloning, Molecular , DNA Primers , Dependovirus/genetics , Dependovirus/isolation & purification , HeLa Cells , Humans , Polymerase Chain Reaction , Virulence
19.
AIDS Res Hum Retroviruses ; 24(6): 873-80, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18544020

ABSTRACT

A novel prophylactic AIDS vaccine candidate, consisting of single-stranded DNA for HIV-1 subtype C gag, protease, and part of reverse transcriptase genes, enclosed within a recombinant adeno-associated virus serotype-2 protein capsid (tgAAC09) induced T cell responses and antibodies in nonhuman primates. In this randomized, dose escalation phase I trial, HIV-uninfected healthy volunteers (50 in Europe, 30 in India) received a single intramuscular injection of tgAAC09 at 3 x 10(9) DNase resistant particles (DRP) (n = 16), 3 x 10(10) DRP (n = 23), 3 x 10(11) DRP (n = 25), or placebo (n = 16). Twenty-one participants in Europe received a second (boost) dose of 3 x 10(11) DRP tgAAC09 or placebo at least 24 weeks after the first injection. The vaccine was safe and well-tolerated after initial and boost vaccination. Local and systemic reactogenicity was experienced by 13-25% of participants and was not dose related. No vaccine-related serious adverse events were reported. Modest HIV-specific T cell responses were detected in 7/64 vaccinees (40-385 SFC/10(6) PBMC), with 16% (4/25) responders in the highest dose group. All responses were to Gag epitopes. tgAAC09 appears to be safe, well-tolerated, and modestly immunogenic. Further evaluation of higher doses of tgAAC09 and boost injections is ongoing in Africa.


Subject(s)
AIDS Vaccines/administration & dosage , HIV-1/immunology , T-Lymphocytes/immunology , Vaccines, Virosome/administration & dosage , gag Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/adverse effects , Adolescent , Adult , Antibody Formation , Capsid/immunology , DNA, Viral/administration & dosage , Dependovirus/immunology , Double-Blind Method , Female , HIV Infections/prevention & control , Humans , Immunity, Cellular , Immunization, Secondary , Injections, Intramuscular , Interferon-gamma/blood , Male , Middle Aged , Neutralization Tests , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Vaccines, Virosome/adverse effects
20.
J Infect Dis ; 196(7): 1058-67, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17763329

ABSTRACT

UNLABELLED: BACKGROUND. Human immunodeficiency virus (HIV) replication and compartmentalization in the central nervous system, including in cerebrospinal fluid (CSF), are associated with severe neurological disease and may contribute to viral persistence during antiretroviral therapy. To understand the relationships between viral populations in multiple compartments, we performed a systematic longitudinal characterization of viral populations in blood plasma and CSF obtained at short time intervals over the full course of infection in 3 macaques infected with simian immunodeficiency virus (SIVsm strain E660). METHODS: Complex viral genetic populations in blood plasma and CSF were characterized using a heteroduplex tracking assay targeted to the V1/V2 hypervariable region of env. To identify signs of neurological disease, monocyte chemoattractant protein (MCP)-1 levels in CSF and CD68(+) monocyte/macrophage infiltration in brain tissues were quantified. RESULTS: Two patterns of blood/CSF viral dynamics were apparent as infection progressed: concordant blood/CSF viral evolution and discordant blood/CSF viral evolution. Perivascular CD68(+) cells in autopsy brain tissue and elevated CSF MCP-1 levels accompanied blood/CSF viral population discordance but not concordance. CONCLUSIONS: Two distinct patterns of blood/CSF viral population dynamics can be observed in SIV-infected macaques, and the patterns may be associated with different neurological disease outcomes.


Subject(s)
Cerebrospinal Fluid/virology , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Immunodeficiency Virus , AIDS Dementia Complex/immunology , AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/virology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/immunology , Brain/virology , Chemokine CCL2/metabolism , Disease Models, Animal , Evolution, Molecular , Genotype , Heteroduplex Analysis , Humans , Macaca mulatta , Macrophages/immunology , Monocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology , Viral Load , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...