Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Vaccine ; 33(25): 2944-54, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25936724

ABSTRACT

BACKGROUND: Several novel tuberculosis vaccines are currently in clinical trials, including AERAS-402, an adenovector encoding a fusion protein of Mycobacterium tuberculosis antigens 85A, 85B, and TB10.4. A multicentred trial of AERAS-402 safety and immunogenicity in healthy infants was conducted in three countries in sub-Saharan Africa, using an adaptive design. METHODS: In a double-blind, randomised, placebo-controlled, dose-finding trial, we enrolled BCG-vaccinated, HIV-uninfected infants aged 16-26 weeks. Infants in the safety/dose-finding phase received two doses of AERAS-402 across three dose levels, or placebo, intramuscularly on days 0 and 28. Infants in the expanded safety phase received three doses of the highest dose level, with the 3rd dose at day 280. Follow up for safety and immunogenicity was for up to two years. RESULTS: We enrolled 206 infants (52 placebo and 154 AERAS-402 recipients) into the dose-finding phase and 281 (141 placebo and 140 AERAS-402 recipients) into the expanded safety phase. Safety data were acceptable across all dose levels. No vaccine-related deaths were recorded. A single serious adverse event of tachypnoea was deemed related to study vaccine. Antibodies directed largely against Ag85A and Ag85B were detected. Low magnitude CD4+ and CD8+ polyfunctional T cell responses were observed at all dose levels. The addition of a third dose of AERAS-402 at the highest dose level did not increase frequency or magnitude of antibody or CD8+ T cell responses. CONCLUSIONS: AERAS-402 has an acceptable safety profile in infants and was well tolerated at all dose levels. Response rate was lower than previously seen in BCG vaccinated adults, and frequency and magnitude of antigen-specific T cells were not increased by a third dose of vaccine.


Subject(s)
Tuberculosis Vaccines/administration & dosage , Acyltransferases/immunology , Adult , Africa South of the Sahara , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Healthy Volunteers , Humans , Immunity, Humoral , Infant , Interferon-gamma/immunology , Male , Tuberculosis/prevention & control , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Vaccination , Vaccines, DNA
2.
Vaccine ; 29(2): 304-13, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21034824

ABSTRACT

Ebola virus causes irregular outbreaks of severe hemorrhagic fever in equatorial Africa. Case mortality remains high; there is no effective treatment and outbreaks are sporadic and unpredictable. Studies of Ebola virus vaccine platforms in non-human primates have established that the induction of protective immunity is possible and safety and human immunogenicity has been demonstrated in a previous Phase I clinical trial of a 1st generation Ebola DNA vaccine. We now report the safety and immunogenicity of a recombinant adenovirus serotype 5 (rAd5) vaccine encoding the envelope glycoprotein (GP) from the Zaire and Sudan Ebola virus species, in a randomized, placebo-controlled, double-blinded, dose escalation, Phase I human study. Thirty-one healthy adults received vaccine at 2×10(9) (n=12), or 2×10(10) (n=11) viral particles or placebo (n=8) as an intramuscular injection. Antibody responses were assessed by ELISA and neutralizing assays; and T cell responses were assessed by ELISpot and intracellular cytokine staining assays. This recombinant Ebola virus vaccine was safe and subjects developed antigen specific humoral and cellular immune responses.


Subject(s)
Adenoviruses, Human/genetics , Ebola Vaccines/immunology , Genetic Vectors , Hemorrhagic Fever, Ebola/prevention & control , Viral Envelope Proteins/immunology , Adolescent , Adult , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Double-Blind Method , Ebola Vaccines/adverse effects , Ebola Vaccines/genetics , Ebolavirus/genetics , Ebolavirus/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Male , Middle Aged , Neutralization Tests , Placebos/administration & dosage , T-Lymphocytes/immunology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Envelope Proteins/genetics , Young Adult
3.
Vaccine ; 25(12): 2228-37, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17250936

ABSTRACT

The presence of replication-competent adenovirus (RCA) is a safety concern for biologics based on recombinant adenoviruses and RCA testing is therefore mandatory for release of clinical material. RCA, which arises from homologous recombination between Ad5 vectors and HEK-293 cells, can be eliminated by the use of PER.C6 cells in combination with a matched vector. However, little is known on RCA formation with vectors based on adenovirus serotypes other than Ad5 and reliable RCA assays to test them are generally lacking. Here we report on the development and qualification of a sensitive RCA assay for Ad35, a promising alternative to Ad5 vectors. The assay is able to detect 1 RCA in 3x10(10) vector particles with 95% confidence, thus meeting current FDA requirements, and can discriminate between RCA and other rare CPE-causing entities, including helper dependent E1 positive particles (HDEP). Using this assay, the first batches of Ad35 vectors produced in PER.C6 cells were analysed and found to be free of RCA and HDEP. Based on the statistical model used, we anticipate that our approach to RCA assay development can be broadly applicable to other adenoviral vectors.


Subject(s)
Adenoviridae/genetics , Adenovirus E1 Proteins/genetics , Biological Assay/methods , Gene Deletion , Virus Replication , Adenoviridae/growth & development , Cell Line , Cell Line, Tumor , Genetic Vectors/genetics , Humans , Polymerase Chain Reaction , Reproducibility of Results
4.
Infect Immun ; 74(1): 313-20, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16368986

ABSTRACT

Given the promise of recombinant adenovirus type 5 (rAd5) as a malaria vaccine carrier in preclinical models, we evaluated the potency of rAd35 coding for Plasmodium yoelii circumsporozoite protein (rAd35PyCS). We chose rAd35 since a survey with serum samples from African subjects demonstrated that human Ad35 has a much lower seroprevalence of 20% and a much lower geometric mean neutralizing antibody titer (GMT) of 48 compared to Ad5 (seroprevalence, 85%; GMT, 1,261) in countries with a high malaria incidence. We also demonstrated that immunization with rAd35PyCS induced a dose-dependent and potent, CS-specific CD8(+) cellular and humoral immune response and conferred significant inhibition (92 to 94%) of liver infection upon high-dose sporozoite challenge. Furthermore, we showed that in mice carrying neutralizing antibody activity against Ad5, mimicking a human situation, CS-specific T- and B-cell responses were significantly dampened after rAd5PyCS vaccination, resulting in loss of inhibition of liver infection upon sporozoite challenge. In contrast, rAd35 vaccine was as potent in naive mice as in Ad5-preimmunized mice. Finally, we showed that heterologous rAd35-rAd5 prime-boost regimens were more potent than rAd35-rAd35 because of induction of anti-Ad35 antibodies after rAd35 priming. The latter data provide a further rationale for developing rAd prime-boost regimens but indicate that priming and boosting Ad vectors must be immunologically distinct and also should be distinct from Ad5. Collectively, the data presented warrant further development of rAd35-based vaccines against human malaria.


Subject(s)
Adenoviruses, Human/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium yoelii/immunology , Adenoviruses, Human/genetics , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Female , Genetic Vectors/immunology , Humans , Immunization, Secondary , Liver/immunology , Liver/parasitology , Liver Diseases, Parasitic/immunology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Plasmodium yoelii/genetics , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
J Virol Methods ; 110(1): 67-71, 2003 Jun 09.
Article in English | MEDLINE | ID: mdl-12757922

ABSTRACT

Reliable assays for accurate titration of influenza virus in infectious samples are pivotal to both influenza research and vaccine development. A titration assay adopted commonly for this purpose is the plaque assay on Madin-Darby canine kidney (MDCK) cells, despite it being time and labour consuming. A novel assay is described for titration of influenza viruses based on the detection of intracellular viral nucleoprotein (NP) by fluorescence-activated cell sorting (FACS). By using a panel of viruses of different type, subtype and origin, it is demonstrated that there is a mathematical correlation between titres measured by immunotitration and by classical plaque assay on MDCK cells. Moreover, the availability of NP antibodies specific for type A or type B influenza virus ensures the specificity of the assay. Based on speed, accuracy and specificity, it is concluded that the FACS-based immunotitration of influenza virus represents a valid and efficient alternative to the classical plaque assay.


Subject(s)
Flow Cytometry/methods , Influenza A virus/isolation & purification , Influenza B virus/isolation & purification , Nucleoproteins/metabolism , Viral Core Proteins/metabolism , Animals , Cell Line , Dogs , Humans , Influenza A virus/metabolism , Influenza B virus/metabolism , Nucleocapsid Proteins , Time Factors , Viral Plaque Assay
6.
Virology ; 288(2): 236-46, 2001 Sep 30.
Article in English | MEDLINE | ID: mdl-11601895

ABSTRACT

Gene therapy aims to complement or, ideally, correct defective genes. The broad clinical application of this emerging technology requires the development of safe high-capacity gene delivery vehicles that combine efficient transduction of dividing as well as quiescent cells with sustained transgene expression. Here we present a new hybrid vector system that unites favorable attributes of adenoassociated virus (AAV) and adenovirus (Ad) vectors in a single particle. This was achieved by inclusion of Ad packaging elements in different sized recombinant AAV genomes. In the presence of AAV replicative functions and a recombinant helper Ad, AAV/Ad hybrid particles were generated via encapsidation of AAV-dependent replicative intermediates into Ad capsids. In stringent in vitro models based on transduction of proliferating cells we show that AAV/Ad hybrid vectors are superior to Ad vectors in establishing prolonged transgene expression and can be used to deliver DNA fragments of at least 27 kb.


Subject(s)
Adenoviruses, Human/genetics , Capsid/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Virus Assembly/physiology , Virus Replication/physiology , Adenoviruses, Human/physiology , Cell Line, Transformed , Dependovirus/physiology , Gene Expression , Genetic Vectors/physiology , Genome, Viral , Humans , Recombination, Genetic , Transgenes , Virion , Virus Assembly/genetics , Virus Replication/genetics
7.
Vaccine ; 19(17-19): 2716-21, 2001 Mar 21.
Article in English | MEDLINE | ID: mdl-11257414

ABSTRACT

Influenza viruses for vaccine production are currently grown on embryonated eggs. This manufacturing system conveys many major drawbacks such as inflexibility, cumbersome down stream processing, inability of some strains to replicate on eggs to high enough yields, and selection of receptor-binding variants with reduced antigenicity. These limitations emphasize the need for a cell line-based production system that could replace eggs in the production of influenza virus vaccines in a pandemic proof fashion. Here we present the efficient propagation of influenza A and B viruses on the fully characterized and standardized human cell line PER.C6.


Subject(s)
Influenza Vaccines/isolation & purification , Virus Cultivation/methods , Animals , Antigens, Viral/isolation & purification , Bioreactors , Cell Line , Chickens , Edetic Acid , Hemagglutinins, Viral/isolation & purification , Humans , Influenza Vaccines/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae/physiology , Trypsin , Virus Replication
8.
Haematologica ; 79(6): 540-1, 1994.
Article in English | MEDLINE | ID: mdl-7896213

ABSTRACT

Seventy months after diagnosis, minimal residual disease is undetectable in a patient with Philadelphia chromosome-positive chronic myelogenous leukemia (CML) in long-lasting continuous cytogenetic conversion (CCC), achieved through alpha 2a-interferon (IFN-alpha) therapy. Fluctuating molecular remission, evaluated with the two-stage reverse transcriptase-polymerase chain reaction (RT-PCR) with nested primers, has persisted for two years at the maximum tolerable dose of IFN alpha (1.5 x 10(6) IU per day).


Subject(s)
Immunologic Factors/therapeutic use , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Pregnancy Complications, Neoplastic/therapy , Adult , Bone Marrow/pathology , Combined Modality Therapy , DNA, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl/genetics , Humans , Hydroxyurea/therapeutic use , Interferon alpha-2 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Polymerase Chain Reaction , Pregnancy , Recombinant Proteins , Remission Induction
SELECTION OF CITATIONS
SEARCH DETAIL
...