Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Hum Vaccin Immunother ; 17(2): 554-559, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32750273

ABSTRACT

SeVRSV is a replication-competent Sendai virus (SeV)-based vaccine carrying the respiratory syncytial virus (RSV) fusion protein (F) gene. Unmanipulated, non-recombinant SeV is a murine parainfluenza virus type 1 (PIV-1) and serves as a Jennerian vaccine for human PIV-1 (hPIV-1). SeV protects African green monkeys (AGM) from infection after hPIV-1 challenge. The recombinant SeVRSV additionally targets RSV and protects AGM from lower respiratory infections after RSV challenge. The present study is the first to report on the safety, viral genome detection, and immunogenicity following SeVRSV vaccination of healthy adults. Seventeen and four healthy adults received intranasal SeVRSV and PBS, respectively, followed by six months of safety monitoring. Virus genome (in nasal wash) and vaccine-specific antibodies (in sera) were monitored for two and four weeks, respectively, post-vaccination. The vaccine was well-tolerated with only mild to moderate reactions that were also present in the placebo group. No severe reactions occurred. As expected, due to preexisting immunity toward hPIV-1 and RSV in adults, vaccine genome detection was transient. There were minimal antibody responses to SeV and negligible responses to RSV F. Results encourage further studies of SeVRSV with progression toward a clinical trial in seronegative children. Abbreviations: AE-adverse event; SAE-serious adverse event; SeV-Sendai virus; RSV-respiratory syncytial virus; PIV-1-parainfluenza virus-type 1; hPIV-1-human parainfluenza virus-type 1; F-RSV fusion protein; SeVRSV-recombinant SeV carrying the RSV F gene; Ab-antibody; MSW-medically significant wheezing; NOCMC-new onset chronic medical condition, mITT-modified Intent to Treat; ALRI-acute lower respiratory tract infection.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Adult , Animals , Antibodies, Viral , Chlorocebus aethiops , Humans , Immunogenicity, Vaccine , Parainfluenza Virus 1, Human/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Sendai virus/genetics , Viral Fusion Proteins/genetics
2.
Viral Immunol ; 33(3): 233-236, 2020 04.
Article in English | MEDLINE | ID: mdl-32286171

ABSTRACT

This article is dedicated to Dr. Peter Doherty. While Peter continues to make groundbreaking discoveries in the field of immunology, he also provides outstanding scientific mentorship to his trainees. Here we contemplate our past training with Peter, Peter's teachings of basic immunological principles, and how basic principles may instruct the design of a successful human immunodeficiency virus-type 1 vaccine.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Drug Development , Humans
3.
Obstet Gynecol ; 127(2): 213-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26942345

ABSTRACT

OBJECTIVE: To evaluate the safety and immunogenicity of an investigational trivalent group B streptococcal vaccine in pregnant women and antibody transfer to their newborns. METHODS: The primary outcome of this observer-blind, randomized study was to estimate placental antibody transfer rates at birth. Secondary outcomes included measurement of serotype-specific antibodies at screening, 30 days postvaccination, at delivery, and 91 days postpartum, infant antibody levels at 3 months of age, the potential effect on routine infant diphtheria vaccination at 1 month after the third infant series dose, and safety in mother and infant participants through at least 5 months postpartum. Sample size was based on 60 participants in the vaccine group giving a probability of observing at least one adverse event of 90% if the actual rate of the event was 3.8%. RESULTS: From September 2011 to October 2013, 86 pregnant women were allocated in a 3:2 ratio to receive an investigational group B streptococcal vaccine containing glycoconjugates of serotypes Ia, Ib, and III or placebo. Demographics were similar across groups. Transfer ratios were 66-79% and maternal geometric mean concentrations increased 16-, 23-, and 20-fold by delivery against serotypes Ia, Ib, and III, respectively, Women with no detectable antibodies at inclusion had lower responses than those with detectable antibodies. Three months after birth, infant antibody concentrations were 22-25% of birth levels. Antidiphtheria geometric mean concentrations were similar across groups. In the vaccine and placebo groups, 32 of 51 women (63%) and 26 of 35 women (74%) reported adverse effects, respectively. CONCLUSION: The investigational vaccine was well-tolerated without safety signals in recipients and their infants or interference with routine infant diphtheria vaccination, although further studies on safety and effectiveness are needed. The investigational vaccine was immunogenic for all serotypes, particularly among women with detectable antibody levels at baseline. Antibody transfer to neonates was at similar levels to other maternally administered polysaccharide vaccines. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, www.clinicaltrials.gov, NCT01446289.


Subject(s)
Streptococcal Infections/prevention & control , Streptococcal Vaccines , Streptococcus agalactiae/immunology , Adult , Female , Humans , Immunization , Infant, Newborn , Pregnancy , Young Adult
4.
BMC Pediatr ; 15: 143, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26427955

ABSTRACT

BACKGROUND: Group B Streptococcus (GBS) is a leading cause of serious infection in very young infants. Robust incidence data from many geographic regions, including Latin America and Asia, are however lacking. METHODS: A multicenter, hospital-based observational study was performed in Panama, Dominican Republic, Hong Kong and Bangladesh. All represented urban, tertiary referral hospitals, except Bangladesh. GBS cases (microbiological isolation from normally sterile sites in infants aged 0-89 days) were collected over 12 months. RESULTS: At 2.35 (95% CI: 1.74-3.18) cases per 1000 live births, the incidence of early onset GBS disease (EOD) was highest in the Dominican Republic, compared with 0.76 (95% CI: 0.41-1.39) in Hong Kong and 0.77 (95% CI: 0.44-1.35) in Panama, while no cases were identified in Bangladesh. Over 90% of EOD cases occurred on the first day of life, with case fatality ratios ranging from 6.7% to 40%, varying by center, age of onset and clinical presentation. Overall, 90% of GBS (EOD and late onset disease) was due to serotypes Ia, Ib and III. CONCLUSIONS: The incidence rate of early onset GBS infection reported in Dominican Republic was not dissimilar from that described in the United States prior to screening and intrapartum antibiotic prophylaxis, while the incidence in Hong Kong was higher than previously reported in the Asian region. The failure to identify GBS cases in Bangladesh highlights a need to better understand the contribution of population, healthcare and surveillance practice to variation in reported incidence. Overall, the identified disease burden and serotype distribution support the need for effective prevention methods in these populations, and the need for community based surveillance studies in rural areas where access to healthcare may be challenging.


Subject(s)
Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus agalactiae , Antibiotic Prophylaxis , Bangladesh/epidemiology , Dominican Republic/epidemiology , Female , Hong Kong/epidemiology , Humans , Incidence , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Male , Panama/epidemiology , Pregnancy , Pregnancy Complications, Infectious/prevention & control , Risk Factors , Serogroup , Streptococcal Infections/prevention & control , Streptococcal Infections/transmission , Streptococcus agalactiae/classification , Streptococcus agalactiae/genetics
5.
Clin Vaccine Immunol ; 22(3): 298-303, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552633

ABSTRACT

Human parainfluenza virus type 1 (hPIV-1) is the most common cause of laryngotracheobronchitis (croup), resulting in tens of thousands of hospitalizations each year in the United States alone. No licensed vaccine is yet available. We have developed murine PIV-1 (Sendai virus [SeV]) as a live Jennerian vaccine for hPIV-1. Here, we describe vaccine testing in healthy 3- to 6-year-old hPIV-1-seropositive children in a dose escalation study. One dose of the vaccine (5 × 10(5), 5 × 10(6), or 5 × 10(7) 50% egg infectious doses) was delivered by the intranasal route to each study participant. The vaccine was well tolerated by all the study participants. There was no sign of vaccine virus replication in the airway in any participant. Most children exhibited an increase in antibody binding and neutralizing responses toward hPIV-1 within 4 weeks from the time of vaccination. In several children, antibody responses remained above incoming levels for at least 6 months after vaccination. Data suggest that SeV may provide a benefit to 3- to 6-year-old children, even when vaccine recipients have preexisting cross-reactive antibodies due to previous exposures to hPIV-1. Results encourage the testing of SeV administration in young seronegative children to protect against the serious respiratory tract diseases caused by hPIV-1 infections.


Subject(s)
Antibodies, Viral/blood , Parainfluenza Virus 1, Human/immunology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/immunology , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Child , Child, Preschool , Cross Reactions , Female , Humans , Infant , Male , Mice , Sendai virus/growth & development , United States , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Viral Vaccines/adverse effects , Viral Vaccines/immunology
6.
Int Immunol ; 27(5): 229-36, 2015 May.
Article in English | MEDLINE | ID: mdl-25477211

ABSTRACT

The respiratory syncytial virus (RSV) is responsible for as many as 199000 annual deaths worldwide. Currently, there is no standard treatment for RSV disease and no vaccine. Sendai virus (SeV) is an attractive pediatric vaccine candidate because it elicits robust and long-lasting virus-specific B cell and T cell activities in systemic and mucosal tissues. The virus serves as a gene delivery system as well as a Jennerian vaccine against its close cousin, human parainfluenza virus type 1. Here we describe the testing of a recombinant SeV (SeVRSV-Fs) that expresses an unconstrained, secreted RSV-F protein as a vaccine against RSV in cotton rats. After a single intranasal immunization of cotton rats with SeVRSV-Fs, RSV-specific binding and neutralizing antibodies were generated. These antibodies exhibited cross-reactivity with both RSV A and B isolates. RSV-F-specific IFN-γ-producing T cells were also activated. The SeVRSV-Fs vaccine conferred protection against RSV challenge without enhanced immunopathology. In total, results showed that an SeV recombinant that expresses RSV F in an unconstrained, soluble form can induce humoral and cellular immunity that protects against infection with RSV.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/metabolism , Sendai virus/immunology , T-Lymphocytes/immunology , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/metabolism , Disease Models, Animal , Gene Transfer Techniques , Humans , Immunization , Interferon-gamma/metabolism , Lymphocyte Activation , Rats , Recombinant Fusion Proteins/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/genetics , Sendai virus/genetics , Sigmodontinae , T-Lymphocytes/virology , Viral Fusion Proteins/genetics , Viral Vaccines/genetics
7.
Vaccine ; 32(26): 3264-73, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24721531

ABSTRACT

Respiratory syncytial virus (RSV) is the cause of significant morbidity and mortality among infants, and despite decades of research there remains no licensed vaccine. SeVRSV is a Sendai virus (SeV)-based live intranasal vaccine that expresses the full length RSV fusion (F) gene. SeV is the murine counterpart of human parainfluenza virus type 1. Given that the target population of SeVRSV is young infants, we questioned whether maternal antibodies typical of this age group would inhibit SeVRSV vaccine efficacy. After measuring SeV- and RSV-specific serum neutralizing antibody titers in human infants, we matched these defined titers in cotton rats by the passive transfer of polyclonal or monoclonal antibody products. Animals were then vaccinated with SeVRSV followed by a 3 month rest period to allow passively transferred antibodies to wane. Animals were finally challenged with RSV to measure the de novo vaccine-induced immune responses. Despite the presence of passively-transferred serum neutralizing antibodies at the time of vaccination, SeVRSV induced immune responses that were protective against RSV challenge. The data encourage advancement of SeVRSV as a candidate vaccine for the protection of children from morbidity and mortality caused by RSV.


Subject(s)
Antibodies, Viral/immunology , Immunity, Maternally-Acquired , Immunization, Passive , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Sendai virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Female , Humans , Infant , Infant, Newborn , Neutralization Tests , Respiratory Syncytial Viruses , Sigmodontinae
8.
Viruses ; 2(2): 435-467, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20407589

ABSTRACT

Currently, there are more than 30 million people infected with HIV-1 and thousands more are infected each day. Vaccination is the single most effective mechanism for prevention of viral disease, and after more than 25 years of research, one vaccine has shown somewhat encouraging results in an advanced clinical efficacy trial. A modified intent-to-treat analysis of trial results showed that infection was approximately 30% lower in the vaccine group compared to the placebo group. The vaccine was administered using a heterologous prime-boost regimen in which both target antigens and delivery vehicles were changed during the course of inoculations. Here we examine the complexity of heterologous prime-boost immunizations. We show that the use of different delivery vehicles in prime and boost inoculations can help to avert the inhibitory effects caused by vector-specific immune responses. We also show that the introduction of new antigens into boost inoculations can be advantageous, demonstrating that the effect of `original antigenic sin' is not absolute. Pre-clinical and clinical studies are reviewed, including our own work with a three-vector vaccination regimen using recombinant DNA, virus (Sendai virus or vaccinia virus) and protein. Promising preliminary results suggest that the heterologous prime-boost strategy may possibly provide a foundation for the future prevention of HIV-1 infections in humans.

9.
Blood ; 115(5): 925-35, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19880495

ABSTRACT

T-cell immunotherapy that takes advantage of Epstein-Barr virus (EBV)-stimulated immunity has the potential to fill an important niche in targeted therapy for EBV-related cancers. To address questions of long-term efficacy, safety, and practicality, we studied 114 patients who had received infusions of EBV-specific cytotoxic T lymphocytes (CTLs) at 3 different centers to prevent or treat EBV(+) lymphoproliferative disease (LPD) arising after hematopoietic stem cell transplantation. Toxicity was minimal, consisting mainly of localized swelling at sites of responsive disease. None of the 101 patients who received CTL prophylaxis developed EBV(+) LPD, whereas 11 of 13 patients treated with CTLs for biopsy-proven or probable LPD achieved sustained complete remissions. The gene-marking component of this study enabled us to demonstrate the persistence of functional CTLs for up to 9 years. A preliminary analysis indicated that a patient-specific CTL line can be manufactured, tested, and infused for $6095, a cost that compares favorably with other modalities used in the treatment of LPD. We conclude that the CTL lines described here provide safe and effective prophylaxis or treatment for lymphoproliferative disease in transplantation recipients, and the manufacturing methodology is robust and can be transferred readily from one institution to another without loss of reproducibility.


Subject(s)
Epstein-Barr Virus Infections/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Herpesvirus 4, Human/immunology , Lymphoproliferative Disorders/therapy , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adoptive Transfer/economics , Adoptive Transfer/methods , Adult , Child , Child, Preschool , Epstein-Barr Virus Infections/etiology , Epstein-Barr Virus Infections/virology , Female , Follow-Up Studies , Humans , Immunophenotyping , Infant , Lymphoma/etiology , Lymphoma/mortality , Lymphoma/therapy , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/mortality , Male , Survival Rate , T-Lymphocytes, Cytotoxic/transplantation , Time Factors , Treatment Outcome , Young Adult
10.
Vaccine ; 27(12): 1848-57, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19200447

ABSTRACT

The human parainfluenza viruses (hPIVs) and respiratory syncytial viruses (RSVs) are the leading causes of hospitalizations due to respiratory viral disease in infants and young children, but no vaccines are yet available. Here we describe the use of recombinant Sendai viruses (rSeVs) as candidate vaccine vectors for these respiratory viruses in a cotton rat model. Two new Sendai virus (SeV)-based hPIV-2 vaccine constructs were generated by inserting the fusion (F) gene or the hemagglutinin-neuraminidase (HN) gene from hPIV-2 into the rSeV genome. The inoculation of either vaccine into cotton rats elicited neutralizing antibodies toward both homologous and heterologous hPIV-2 virus isolates. The vaccines elicited robust and durable antibodies toward hPIV-2, and cotton rats immunized with individual or mixed vaccines were fully protected against hPIV-2 infections of the lower respiratory tract. The immune responses toward a single inoculation with rSeV vaccines were long-lasting and cotton rats were protected against viral challenge for as long as 11 months after vaccination. One inoculation with a mixture of the hPIV-2-HN-expressing construct and two additional rSeVs (expressing the F protein of RSV and the HN protein of hPIV-3) resulted in protection against challenge viruses hPIV-1, hPIV-2, hPIV-3, and RSV. Results identify SeV vectors as promising vaccine candidates for four different paramyxoviruses, each responsible for serious respiratory infections in children.


Subject(s)
Parainfluenza Vaccines/therapeutic use , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Sendai virus/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Humans , Neutralization Tests , Sigmodontinae , Vaccines, Synthetic/immunology
11.
Int Rev Immunol ; 28(1): 49-68, 2009.
Article in English | MEDLINE | ID: mdl-19241253

ABSTRACT

The human immune system has evolved to recognize antigenic diversity, a strength that has been harnessed by vaccine developers to combat numerous pathogens (e.g., pneumococcus, influenza virus, rotavirus). In each case, vaccine cocktails were formulated to include antigenic variants of the target. To combat HIV-1 diversity, we assembled a cocktail vaccine comprising dozens of envelopes, delivered as recombinant DNA, vaccinia virus, and protein for testing in a clinical trial. One vaccinee has now completed vaccinations with no serious adverse events. Preliminary analyses demonstrate early proof-of-principle that a multi-envelope vaccine can elicit neutralizing antibody responses toward heterologous HIV-1 in humans.


Subject(s)
AIDS Vaccines/therapeutic use , DNA, Recombinant/immunology , HIV Infections/prevention & control , HIV-1/immunology , Vaccines, DNA/therapeutic use , Vaccinia virus/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/adverse effects , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antigenic Variation/immunology , Clinical Trials as Topic , DNA, Recombinant/genetics , Humans , Mice , Vaccines, DNA/adverse effects , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus/genetics , env Gene Products, Human Immunodeficiency Virus/genetics
12.
Vaccine ; 26(27-28): 3480-8, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18499307

ABSTRACT

The human parainfluenza viruses (hPIVs) and respiratory syncytial virus (RSV) are the leading causes of serious respiratory illness in the human pediatric population. Despite decades of research, there are currently no licensed vaccines for either the hPIV or RSV pathogens. Here we describe the testing of hPIV-3 and RSV candidate vaccines using Sendai virus (SeV, murine PIV-1) as a vector. SeV was selected as the vaccine backbone, because it has been shown to elicit robust and durable immune activities in animal studies, and has already advanced to human safety trials as a xenogenic vaccine for hPIV-1. Two new SeV-based hPIV-3 vaccine candidates were first generated by inserting either the fusion (F) gene or hemagglutinin-neuraminidase (HN) gene from hPIV-3 into SeV. The resultant rSeV-hPIV3-F and rSeV-hPIV3-HN vaccines expressed their inserted hPIV-3 genes upon infection. The inoculation of either vaccine into cotton rats elicited binding and neutralizing antibody activities, as well as interferon-gamma-producing T cells. Vaccination of cotton rats resulted in protection against subsequent challenges with either homologous or heterologous hPIV-3. Furthermore, vaccination of cotton rats with a mixture of rSeV-hPIV3-HN and a previously described recombinant SeV expressing the F protein of RSV resulted in protection against three different challenge viruses: hPIV-3, hPIV-1 and RSV. Results encourage the continued development of the candidate recombinant SeV vaccines to combat serious respiratory infections of children.


Subject(s)
HN Protein/immunology , Parainfluenza Vaccines/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respirovirus Infections/prevention & control , Sendai virus/genetics , Viral Fusion Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Female , Genetic Vectors , HN Protein/genetics , Lung/virology , Molecular Sequence Data , Parainfluenza Vaccines/genetics , Rats , Respiratory Syncytial Virus Vaccines/genetics , Sigmodontinae , T-Lymphocytes/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Fusion Proteins/genetics
13.
AIDS ; 22(1): 149-51, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18090404

ABSTRACT

To combat HIV-1 diversity, we are developing a multienvelope vaccine (comprising DNA, vaccinia virus and protein vectors). Toward this goal, we conducted a phase I clinical trial of EnvPro, a gp140 protein formulated in alum. The vaccine was well tolerated and elicited an immune response in every trial participant.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV-1/immunology , Immunization , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , Adult , Female , HIV Antibodies/blood , HIV Infections/blood , Headache/chemically induced , Humans , Immunization Schedule , Injections, Intramuscular , Male , Recombinant Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/biosynthesis
14.
Front Biosci ; 13: 609-20, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981574

ABSTRACT

A major obstacle to the design of a global HIV-1 vaccine is viral diversity. At present, data suggest that a vaccine comprising a single antigen will fail to generate broadly reactive B-cell and T-cell responses able to confer protection against the diverse isolates of HIV-1. While some B-cell and T-cell epitopes lie within the more conserved regions of HIV-1 proteins, many are localized to variable regions and differ from one virus to the next. Neutralizing B-cell responses may vary toward viruses with different i) antibody contact residues and/or ii) protein conformations while T-cell responses may vary toward viruses with different (i) T-cell receptor contact residues and/or (ii) amino acid sequences pertinent to antigen processing. Here we review previous and current strategies for HIV-1 vaccine development. We focus on studies at St. Jude Children's Research Hospital (SJCRH) dedicated to the development of an HIV-1 vaccine cocktail strategy. The SJCRH multi-vectored, multi-envelope vaccine has now been shown to elicit HIV-1-specific B- and T-cell functions with a diversity and durability that may be required to prevent HIV-1 infections in humans.


Subject(s)
AIDS Vaccines/chemistry , Drug Design , HIV Infections/prevention & control , HIV-1/metabolism , Animals , Chemistry, Pharmaceutical/methods , HIV Antibodies/chemistry , Human Immunodeficiency Virus Proteins/chemistry , Humans , Immune System/virology , Neutralization Tests , Primates
15.
Vaccine ; 25(52): 8782-93, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18037543

ABSTRACT

The respiratory syncytial virus (RSV) is a serious pediatric pathogen for which there is currently no clinically approved vaccine. This report describes the design and testing of a new RSV vaccine construct (rSV-RSV-F), created by the recombination of an RSV F sequence with the murine parainfluenza virus-type 1 (Sendai virus, SV) genome. SV was selected as the vaccine backbone for this study, because it has previously been shown to elicit high-magnitude, durable immune activities in animal studies and has advanced to human safety trials as a xenogenic vaccine for human parainfluenza virus-type 1 (hPIV-1). Cells infected with the recombinant SV expressed RSV F protein, but F was not incorporated into progeny SV virions. When cotton rats were inoculated with the vaccine, high-titer RSV-binding and neutralizing antibodies as well as interferon-gamma-producing T-cells were induced. Most striking was the protection against intra-nasal RSV challenge conferred by the vaccine. The rSV-RSV-F construct was also tested as a mixture with a second SV construct expressing the RSV G protein, but no clear advantage was demonstrated by combining the two vaccines. As a final analysis, the efficacy of the rSV-RSV-F vaccine was tested against an array of RSV isolates. Results showed that neutralizing and protective responses were effective against RSV isolates of both A and B subtypes. Together, experimental results encourage promotion of this recombinant SV construct as a vaccine candidate for the prevention of RSV in humans.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Sendai virus/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon-gamma/biosynthesis , Lung/pathology , Lung/virology , Molecular Sequence Data , Neutralization Tests , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/genetics , Sendai virus/genetics , Sigmodontinae , T-Lymphocytes/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Fusion Proteins/genetics , Viral Plaque Assay , Viral Vaccines/genetics
16.
J Virol ; 81(22): 12535-42, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17652379

ABSTRACT

The importance of antigen-specific CD4(+) helper T cells in virus infections is well recognized, but their possible role as direct mediators of virus clearance is less well characterized. Here we describe a recombinant Sendai virus strategy for probing the effector role(s) of CD4(+) T cells. Mice were vaccinated with DNA and vaccinia virus recombinant vectors encoding a secreted human immunodeficiency virus type 1 (HIV-1) envelope protein and then challenged with a Sendai virus carrying a homologous HIV-1 envelope gene. The primed mice showed (i) prompt homing of numerous envelope-primed CD4(+) T cell populations to the virus-infected lung, (ii) substantial production of gamma interferon, and interleukin-2 (IL-2), IL-4, and IL-5 in that site, and (iii) significantly reduced pulmonary viral load. The challenge experiments were repeated with immunoglobulin(-/-) microMT mice in the presence or absence of CD8(+) and/or CD4(+) T cells. These selectively immunodeficient mice were protected by primed CD4(+) T cells in the absence of antibody or CD8(+) T cells. Together, these results highlight the role of CD4(+) T cells as direct effectors in vivo and, because this protocol gives such a potent response, identify an outstanding experimental model for further dissecting CD4(+) T-cell-mediated immunity in the lung.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Lung/immunology , Sendai virus/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , HIV Envelope Protein gp120/genetics , HIV-1/genetics , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Sendai virus/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination , Vaccinia virus/genetics , Vaccinia virus/immunology
17.
AIDS Res Hum Retroviruses ; 23(3): 471-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17411381

ABSTRACT

HIV-1-specific T cell responses correlate with control of infection and disease, thus encouraging a full understanding of the peptides and antigen-processing mechanisms that govern T cell activation. We have previously demonstrated that CD4(+) T cell epitopes cluster nonrandomly within envelope protein "hotspot" regions. The current study was initiated to determine whether envelope-specific CD8(+) T cells might share epitope "hotspots" with the CD4(+) T cell population. Identification of CD8(+) T cell determinants by ELISPOT assays with peripheral blood mononuclear cells from four HIV-1-infected individuals, in conjunction with a survey of determinants in the Los Alamos database, revealed similarities among "hotspot" positions for CD4(+) and CD8T(+) cells within mice and humans. These results emphasized the important influence that envelope peptide position may have on antigen processing, and the consequent impact such processing may have on HIV-1-specific CD4(+) and CD8(+) T-cell activities.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocyte Subsets , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Humans , Mice , Molecular Sequence Data , Peptide Fragments/immunology , Sequence Alignment , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology
18.
J Pediatr Pharmacol Ther ; 12(2): 68-76, 2007 Apr.
Article in English | MEDLINE | ID: mdl-23055844

ABSTRACT

The St. Jude Children's Research Hospital (St. Jude) HIV-1 vaccine program is based on the observation that multiple antigenically distinct HIV-1 envelope protein structures are capable of mediating HIV-1 infection. A cocktail vaccine comprising representatives of these diverse structures (immunotypes) is therefore considered necessary to elicit lymphocyte populations that prevent HIV-1 infection. This strategy is reminiscent of that used to design a currently licensed and successful 23-valent pneumococcus vaccine. Three recombinant vector systems are used for the delivery of envelope cocktails (DNA, vaccinia virus, and purified protein), and each of these has been tested individually in phase I safety trials. A fourth FDA-approved clinical trial, in which diverse envelopes and vectors are combined in a prime-boost vaccination regimen, has recently begun. This trial will continue to test the hypothesis that a multi-vector, multi-envelope vaccine can elicit diverse B- and T-cell populations that can prevent HIV-1 infections in humans.

19.
Vaccine ; 23(46-47): 5306-20, 2005 Nov 16.
Article in English | MEDLINE | ID: mdl-16095768

ABSTRACT

A central obstacle to the design of a global HIV-1 vaccine is virus diversity. Pathogen diversity is not unique to HIV-1, and has been successfully conquered in other fields by the creation of vaccine cocktails. Here we describe the testing of an HIV-1 envelope cocktail vaccine. Six macaques received the vaccine, delivered by successive immunizations with recombinant DNA, recombinant vaccinia virus and recombinant envelope proteins. Following vaccination, animals developed a diversity of anti-envelope antibody binding and neutralizing activities toward proteins and viruses that were not represented by sequence in the vaccine. T-cells were also elicited, as measured by gamma-interferon production assays with envelope-derived peptide pools. Vaccinated and control animals were then challenged with the heterologous pathogenic SHIV, 89.6P. Vaccinated monkeys experienced significantly lower virus titers and better maintenance of CD4+ T-cells than unvaccinated controls. The B- and T-cell immune responses were far superior post-challenge in the vaccinated group. Four of six vaccinated animals and only one of six control animals survived a 44-week observation period post-challenge. The present report is the first to describe pathogenic SHIV disease control mediated by a heterologous HIV-1 vaccine, devoid of 89.6 or SIV derivatives.


Subject(s)
AIDS Vaccines/therapeutic use , HIV-1/immunology , Immunity, Cellular/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , CD4 Lymphocyte Count , Enzyme-Linked Immunosorbent Assay , Macaca mulatta , Neutralization Tests , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccination , Viral Load
20.
Viral Immunol ; 18(2): 255-66, 2005.
Article in English | MEDLINE | ID: mdl-16035938

ABSTRACT

Respiratory syncytial virus (RSV) is among the most important and serious pediatric respiratory diseases, and yet after more than four decades of research an effective vaccine is still unavailable. This review examines the role of the immune response in reducing disease severity; considers the history of RSV vaccine development; and advocates the potential utility of Sendai virus (a murine paramyxovirus) as a xenogenic vaccine vector for the delivery of RSV antigens. The immunogenicity and protective efficacy of RSV-recombinant Sendai virus vectors constructed using reverse genetics is examined. RSV-recombinant Sendai virus is easy to grow (i.e., achieves extremely high titers in eggs), is easy to administer (intranasal drops), and elicits both B- and T-cell responses leading to protection from RSV challenge in a small-animal model. Unmodified Sendai virus is currently being studied in clinical trials as a vaccine for its closely related human cognate (human parainfluenza virus type 1). Sendai virus may prove an enormously valuable vaccine platform, permitting the delivery of recombinants targeting important pediatric respiratory pathogens, RSV chief among them.


Subject(s)
Recombination, Genetic , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human/immunology , Sendai virus/immunology , Animals , Child , Genetic Vectors , History, 21st Century , Humans , Mice , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/genetics , Sendai virus/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...