Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 415
Filter
1.
Sci Rep ; 8(1): 8078, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29799015

ABSTRACT

DNA vaccines require a considerable enhancement of immunogenicity. Here, we optimized a prototype DNA vaccine against drug-resistant HIV-1 based on a weak Th2-immunogen, HIV-1 reverse transcriptase (RT). We designed expression-optimized genes encoding inactivated wild-type and drug-resistant RTs (RT-DNAs) and introduced them into mice by intradermal injections followed by electroporation. RT-DNAs were administered as single or double primes with or without cyclic-di-GMP, or as a prime followed by boost with RT-DNA mixed with a luciferase-encoding plasmid ("surrogate challenge"). Repeated primes improved cellular responses and broadened epitope specificity. Addition of cyclic-di-GMP induced a transient increase in IFN-γ production. The strongest anti-RT immune response was achieved in a prime-boost protocol with electroporation by short 100V pulses done using penetrating electrodes. The RT-specific response, dominated by CD4+ T-cells, targeted epitopes at aa 199-220 and aa 528-543. Drug-resistance mutations disrupted the epitope at aa 205-220, while the CTL epitope at aa 202-210 was not affected. Overall, multiparametric optimization of RT strengthened its Th2- performance. A rapid loss of RT/luciferase-expressing cells in the surrogate challenge experiment revealed a lytic potential of anti-RT response. Such lytic CD4+ response would be beneficial for an HIV vaccine due to its comparative insensitivity to immune escape.


Subject(s)
AIDS Vaccines , Drug Resistance, Viral , HIV Infections/therapy , HIV Reverse Transcriptase/immunology , Th2 Cells/immunology , Vaccination/methods , Vaccines, DNA , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Calibration , Cells, Cultured , Codon , Drug Delivery Systems , Drug Resistance, Viral/genetics , Drug Resistance, Viral/immunology , Epitopes/genetics , Epitopes/immunology , HIV Infections/immunology , HIV Reverse Transcriptase/genetics , HIV-1/genetics , HIV-1/immunology , HeLa Cells , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Immunization, Secondary/methods , Immunization, Secondary/standards , Immunogenicity, Vaccine/genetics , Mice , Mice, Inbred BALB C , Quality Improvement , Th2 Cells/metabolism , Vaccination/standards , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
2.
Hum Vaccin Immunother ; 13(12): 2849-2858, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28696158

ABSTRACT

BACKGROUND: Genetic immunization is expected to induce the expression of antigens in a native form. The encoded peptide epitopes are presented on endogenous MHC molecules, mimicking antigen presentation during a viral infection. We have explored the potential of enfuvirtide (T20), a short HIV peptide with antiviral properties, to enhance immune response to HIV antigens. To generate an expression vector, the T20 sequence was cloned into a conventional plasmid, the novel minicircle construct, and a replicon plasmid. In addition, 3 conventional plasmids that express the envelope of HIV-1 subtypes A, B and C and contain T20 in their gp41 sequences were also tested. RESULTS: All combinations induced HIV-specific antibodies and cellular responses. The addition of T20 as a peptide and as an expression cassette in the 3 DNA vectors enhanced antibody responses. The highest anti-HIV-1 Env titers were obtained by the replicon T20 construct. This demonstrates that besides its known antiviral activity, T20 promotes immune responses. We also confirm that the combination of slightly divergent antigens improves immune responses. CONCLUSIONS: The antiretroviral T20 HIV-1 sequence can be used as an immunogen to elicit binding and neutralizing antibodies against HIV-1. These, or similarly modified gp41 genes/peptides, can be used as priming or boosting components for induction of broadly neutralizing anti-HIV antibodies. Future comparative studies will reveal the optimal mode of T20 administration.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , Cross Reactions , HIV Antibodies/blood , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Peptide Fragments/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Animals , Enfuvirtide , Female , HIV Envelope Protein gp41/genetics , Mice, Inbred BALB C , Peptide Fragments/genetics , Vaccines, DNA/administration & dosage
3.
Heliyon ; 3(6): e00339, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28721397

ABSTRACT

BACKGROUND: In order to develop a more effective prophylactic HIV-1 vaccine it is important optimize the components, improve Envelope glycoprotein immunogenicity as well as to explore prime-boost immunization schedules. It is also valuable to include several HIV-1 subtype antigens representing the world-wide epidemic. METHODS: HIVIS-DNA plasmids which include Env genes of subtypes A, B and C together with Gag subtypes A and B and RTmut/Rev of subtype B were modified as follows: the Envelope sequences were shortened, codon optimized, provided with an FT4 sequence and an immunodominant region mutated. The reverse transcriptase (RT) gene was shortened to contain the most immunogenic N-terminal fragment and fused with an inactivated viral protease vPR gene. HIVISopt-DNA thus contains fewer plasmids but additional PR epitopes compared to the native HIVIS-DNA. DNA components were delivered intradermally to young Balb/c mice once, using a needle-free Biojector® immediately followed by dermal electroporation. Vaccinia-based MVA-CMDR boosts including Env gene E and Gag-RT genes A were delivered intramuscularly by needle, once or twice. RESULTS: Both HIVIS-DNA and HIVISopt-DNA primed humoral and cell mediated responses well. When boosted with heterologous MVA-CMDR (subtypes A and E) virus inhibitory neutralizing antibodies were obtained to HIV-1 subtypes A, B, C and AE. Both plasmid compositions boosted with MVA-CMDR generated HIV-1 specific cellular responses directed against HIV-1 Env, Gag and Pol, as measured by IFNγ ELISpot. It was shown that DNA priming augmented the vector MVA immunological boosting effects, the HIVISopt-DNA with a trend to improved (Env) neutralization, the HIVIS-DNA with a trend to better (Gag) cell mediated immune reponses. CONCLUSIONS: HIVIS-DNA was modified to obtain HIVISopt-DNA that had fewer plasmids, and additional epitopes. Even with one DNA prime followed by two MVA-CMDR boosts, humoral and cell-mediated immune responses were readily induced by priming with either DNA construct composition. Priming by HIV-DNA augmented neutralizing antibody responses revealed by boosting with the vaccinia-based heterologous sequences. Cellular and antibody responses covered selected strains representing HIV-1 subtypes A, B, C and CRF01_AE. We assume this is related to the inclusion of heterologous full genes in the vaccine schedule.

4.
J Virus Erad ; 1(3): 134-139, 2015.
Article in English | MEDLINE | ID: mdl-26893908

ABSTRACT

The EPIICAL (Early-treated Perinatally HIV-infected Individuals: Improving Children's Actual Life with Novel Immunotherapeutic Strategies) project arises from the firm belief that perinatally infected children treated with suppressive antiretroviral therapy (ART) from early infancy represent the optimal population model in which to study novel immunotherapeutic strategies aimed at achieving ART-free remission. This is because HIV-infected infants treated within 2-3 months of life have a much reduced viral reservoir size, and rarely show HIV-specific immunity but preserve normal immune development. The goal of EPIICAL is the establishment of an international collaboration to develop a predictive platform using this model to select promising HIV therapeutic vaccine candidates, leading to prioritisation or deprioritisation of novel immunotherapeutic strategies. To establish this platform, the EPIICAL Consortium aims to: develop predictive models of virological and immunological dynamics associated with response to early ART and to treatment interruption using available data from existing cohorts/studies of early-treated perinatally HIV-infected children; optimise methodologies to better characterise immunological, virological and genomic correlates/profiles associated with viral control; test novel immunotherapeutic strategies using in vivo proof-of-concept (PoC) studies with the aim of inducing virological, immunological and transcriptomic correlates/profiles equivalent to those defined by the predictive model. This approach will strengthen the capacity for discovery, development and initial testing of new therapeutic vaccine strategies through the integrated efforts of leading international scientific groups, with the aim of improving the health of HIV-infected individuals.

5.
Vaccine ; 29(33): 5558-66, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21300092

ABSTRACT

Immunotherapy in patients with HIV-1 infection aims to restore and broaden immunological competence, reduce viral load and thereby permit longer periods without combined antiretroviral treatment (cART). Twelve HIV-1-infected patients on cART were immunized on the skin with DNA plasmids containing genes of several HIV-1 subtypes with or without the addition of hydroxyurea (HU), or with placebo. The mean net gain of HIV-specific CD8+ T cell responses were higher and broader in the HIV DNA vaccine groups compared to non-vaccinated individuals (p<0.05). The vaccine-induced immune responses per se had no direct effect on viral replication. In all patients combined, including placebo, the viral set point after a final structured therapy interruption (STI) was lower than prior to initiation of cART (p=0.003). Nadir CD4 levels appeared to strongly influence the post-STI viral titers. After the sixth immunization or placebo, patients could stay off cART for a median time of 15 months. The study shows that HIV DNA immunization induces broader and higher magnitudes of HIV-specific immune responses compared to structured therapy interruptions alone. Although compromised by small numbers of patients, the study also demonstrates that well-monitored STI may safely function as an immunological read out of HIV vaccine efficacy.


Subject(s)
AIDS Vaccines/administration & dosage , Antigens, Viral/immunology , HIV Infections/prevention & control , HIV Infections/therapy , HIV-1/immunology , Immunotherapy/methods , Vaccines, DNA/administration & dosage , AIDS Vaccines/immunology , Adult , Anti-HIV Agents/administration & dosage , Antigens, Viral/genetics , Antiretroviral Therapy, Highly Active/methods , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Double-Blind Method , HIV Infections/immunology , HIV-1/genetics , Humans , Licensure , Lymphocyte Count , Male , Middle Aged , Placebos/administration & dosage , Plasmids/administration & dosage , Vaccines, DNA/immunology , Viral Load
6.
Vaccine ; 26(40): 5079-82, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18514370

ABSTRACT

As more HIV-1 infected patients receive anti-retroviral drug treatment, the occurrence of drug-resistant variants of the virus is increasing. We have previously shown that mutated HIV peptide sequences represent mutations induced by antiretroviral drugs are equally good and often better immunogens than wild type peptides. The non-toxic B subunit of cholera toxin (CTB) is an active substance in the oral cholera vaccine, and has been shown to bind ganglioside receptors and activate mucosal cells. By fusing mutant epitopes deriving from HIV-1 enzymes with the B subunit of cholera toxin, we aim is to induce cellular responses against virus harboring drug-induced mutations. We successfully created conjugates of HIV peptide sequences fused to rCTB. The immune response against the different peptides was strongly enhanced by the fusion to the toxin. Moreover, immunization with sequence containing drug-induced mutation triggered a cross-reactive immune response against the wild type epitope. Long-term follow-up of immunized animals revealed a persistence of cellular immune response for over 4 months, which could readily be boosted with an additional late immunization. By linking HIV-peptides to the B subunit of cholera toxin it is thus possible to stimulate a strong and long-lasting immune response, significantly better than that evoked by the peptide alone.


Subject(s)
AIDS Vaccines , Cholera Toxin , Epitopes, T-Lymphocyte/immunology , HIV Infections/immunology , HIV Reverse Transcriptase , Recombinant Fusion Proteins , Vaccines, Conjugate , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Cholera Toxin/administration & dosage , Cholera Toxin/genetics , Cholera Toxin/immunology , Cholera Toxin/metabolism , HIV Antibodies/blood , HIV Infections/prevention & control , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunization , Immunologic Memory , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/genetics , Vaccines, Conjugate/immunology
7.
Vaccine ; 26(40): 5182-7, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18479789

ABSTRACT

Individuals infected with different subtypes of HIV-1 (A, B, C, D, CRF01_AE and CRF02_AG) were analyzed for their antigen-specific immune response with respect to their HLA genetics. The p24 Gag protein was selected for analysis, since previous studies of the same cohort of patients had shown that almost 80% of these individuals responded to Gag peptides of subtypes A, B and/or C. A large number of Gag antigen-specific responses were recorded. Both previously recognized as well as new epitopes were identified, assumed to bind HLA classes I and/or II. Fifteen individuals showed class I cellular responses to T cell epitopes irrespective of the infecting virus subtype. For five individuals infected with subtypes A, B, D and CRF02_AG, new T cell epitopes are described. Responses related to the patient's class I alleles are frequent, and several new putative class II responses were found.


Subject(s)
Alleles , Epitopes, T-Lymphocyte , HIV Core Protein p24/immunology , HIV-1/classification , Histocompatibility Antigens Class II , Histocompatibility Antigens Class I , Amino Acid Sequence , CD4 Lymphocyte Count , Cross Reactions , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Products, gag/chemistry , Gene Products, gag/immunology , HIV Core Protein p24/chemistry , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Testing , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology
8.
Vaccine ; 25(32): 5968-77, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17629365

ABSTRACT

The immune response to HIV-1 virus-like particles (VLPs), presenting a clade A Ugandan gp120, has been evaluated in a mouse model by intra-nasal (i.n.) administration by a VLP+VLP homologous or a DNA+VLP heterologous prime-boost immunization protocol, including a HIV-1 DNA gp160/rev plasmid. Furthermore, the effect of the Eurocine lipid-based mucosal L3 adjuvant on the VLP immunogenicity has been assessed as well. The designed heterologous protocol is able to increase the env-specific humoral and cellular immune response, compared to the homologous protocol, which is to some extent increased by the administration of L3-adjuvanted VLP boosting dose. The anti-gag response is statistically increased in both homologous and heterologous protocols, particularly when the VLP boosting dose is adjuvanted. Immune sera from immunized animals exhibit >50% ex vivo neutralizing activity against heterologous A and B-clade viral isolates. An envelope B-cell epitope mapping shows an enhanced response against V3 epitopes all across the C2-V5 region in the heterologous prime-boost immunization strategy. The induction of humoral immunity at mucosal sites, which represents the main port of entry for the HIV-1 infection, is extremely relevant. In this framework, the DNA-VLP heterologous prime-boost protocol appears a promising preventive vaccine approach which can significantly benefit from specific mucosal adjuvants, as the Eurocine L3.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/immunology , HIV-1/classification , HIV-1/immunology , Immunity, Cellular/immunology , Immunity, Mucosal/immunology , Immunization, Secondary , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Administration, Intranasal , Animals , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Female , HIV Antibodies/blood , HIV Envelope Protein gp160/immunology , Immunoglobulin G/blood , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Spleen/cytology , Spleen/immunology , Vaccines, DNA/administration & dosage
9.
Scand J Immunol ; 66(1): 43-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17587345

ABSTRACT

In preparation for a clinical trial in patients diagnosed with colorectal cancer, a vaccination strategy targeting the carcinoembryonic antigen (CEA) was evaluated in mice using a GMP-produced plasmid DNA vaccine, CEA66, encoding a truncated form of the tumour-associated antigen, CEA. The GMP-produced CEA DNA vaccine was also evaluated for toxicity. Repeated intradermal administration of the GMP-produced vaccine using a novel needle-free jet injection device (Biojector) induced robust CD4 and CD8 T-cell responses in mice, and did not result in any vaccine-related toxicity. In a heterologous DNA prime/protein boost setting, cellular immune responses were of higher magnitude in animals primed with CEA66 DNA than in animals receiving repeated doses of recombinant CEA protein. These responses were further enhanced if recombinant murine granulocyte-macrophage colony-stimulating factor was given as an adjuvant prior to vaccination. In contrast to repeated administration of recombinant CEA protein as a single modality vaccine, the heterologous CEA66 DNA prime/rCEA boost vaccination strategy resulted in a qualitatively broader immune response, and supports clinical testing of this vaccination regimen in humans.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/prevention & control , Vaccines, DNA/immunology , Adjuvants, Immunologic/pharmacology , Animals , Blotting, Western , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Colorectal Neoplasms/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Guanosine Monophosphate/immunology , HeLa Cells , Humans , Injections, Jet , Mice , Plasmids , Recombinant Proteins , T-Lymphocytes/immunology , Transfection , Transgenes , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/toxicity
10.
Microbes Infect ; 7(14): 1414-23, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16257558
11.
AIDS Res Hum Retroviruses ; 20(8): 860-70, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15320990

ABSTRACT

The aim of this study was to evaluate the immunological responses induced by DNA plasmids containing HIV regulatory genes administered in combination in HIV-1-infected patients with pretreatment with highly active antiretroviral treatment (HAART). The study is a double-blind, randomized, and placebo-controlled study, including 15 asymptomatic HIV-1-infected patients on stable HAART for at least 6 months and with plasma HIV RNA levels below 50 copies/ml. Ten patients received a combination of rev, tat, and nef intramuscularly (im) at weeks 0, 4, and 16 at increasing doses giving totals of 300 (100 x 3), 900 (300 x 3), and 1800 (600 x 3) micrograms DNA. Five patients received saline in the same amounts im. Antigen-specific cytotoxic T lymphocyte (CTL) levels were preserved or increased and new T lymphocyte proliferative responses were induced in the group immunized with the HIV DNA genes. No increase in antibody levels was noted. Despite a 10-fold higher vaccine dose, patients on HAART did not respond better to vaccination compared to non-HAART patients included in a previous study where the genes were administered separately. Combining the regulatory genes rev, tat, and nef in increasing doses may reduce the anticipated augmentation of HIV-specific T cell proliferative and CTL responses. Viral suppression did not seem to further improve the initial vaccine responses of patients with comparable CD4 levels.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV Infections/therapy , HIV-1 , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Anti-HIV Agents/therapeutic use , Antigens, Viral/biosynthesis , Antiretroviral Therapy, Highly Active , Double-Blind Method , Genes, Viral , HIV-1/genetics , HIV-1/immunology , Humans , Vaccines, DNA/administration & dosage , Viral Load
12.
J Gen Virol ; 85(Pt 8): 2407-2419, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15269383

ABSTRACT

The immunogenicity and protective efficacy of a DNA and recombinant modified vaccinia Ankara (MVA) vaccine administered by two different routes were investigated. DNA expressing HIV-1 IIIB env, gag, RT, rev, tat and nef, and MVA expressing HIV-1 IIIB nef, tat and rev and simian immunodeficiency virus (SIV) macJ5 gag/pol and vaccinia HIV-1 env, were used as immunogens. Four cynomolgus macaques received DNA intramuscularly (i.m.) at month 0 and intrarectally (i.r.) and intra-orally (i.o.) at 2 months, followed by MVA i.m. at 4 months and i.r. and i.o. at 8 months. Another group of four monkeys received the same immunogens but only i.m. Overall, stronger cellular immune responses measured by ELISPOT and T-cell proliferation assay were detected in the group primed i.m. and boosted mucosally. Following homologous intravenous simian-human immunodeficiency virus (SHIV) challenge, one of eight vaccinated animals was completely protected. This monkey, immunized i.m. and i.r.+i.o., exhibited the highest levels of HIV Env, Nef and Tat antibodies, high HIV Tat cytotoxic T-lymphocyte activity and T-lymphocyte proliferative responses to HIV Env. Four weeks post-challenge none of the monkeys immunized i.m. and i.r.+i.o., and only two out of four animals immunized i.m., demonstrated detectable plasma viral RNA levels. In contrast, all eight control animals had demonstrable plasma viral RNA levels 4 weeks post-challenge. Thus, stronger cellular immune responses and reduction of challenge virus burden were demonstrated in animals immunized i.m. as well as mucosally, compared with animals immunized i.m. only. The breadth and magnitude of the induced immune responses correlated with protective efficacy.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Vaccines, DNA/immunology , Vaccinia virus/immunology , Animals , HIV Antibodies/blood , Immunization , Interferon-gamma/biosynthesis , Macaca fascicularis , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/administration & dosage , Viral Load
13.
Vaccine ; 21(19-20): 2263-7, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12744856

ABSTRACT

To improve immune responses induced by DNA immunization, murine polyomavirus major capsid protein (VP1) pseudocapsids were complexed with a DNA plasmid encoding the p37 (p24 and p17) nucleocapsid proteins of the human immunodeficiency virus type 1 (HIV-1). A 10-fold increase in antibody titer was noted in mice given DNA plasmid together with VP1 pseudocapsids in comparison to animals that received DNA plasmid alone. Cell mediated responses to HIV-1 p24 occurred, but were not significantly augmented by delivering the DNA as a VP1 complex. We have consequently for the first time shown a carrier/adjuvant effect of polyomavirus pseudocapsids that strongly increased the humoral immune response in DNA immunization.


Subject(s)
Capsid Proteins/immunology , Polyomavirus Infections/immunology , Polyomavirus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Antibody Formation , Capsid Proteins/genetics , Cloning, Molecular , Mice , Mice, Inbred C57BL
14.
HIV Med ; 4(2): 101-10, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12702130

ABSTRACT

OBJECTIVE: To study the impact of effective highly active antiretroviral therapy (HAART) on the preservation of long-term CD4 memory cells induced by vaccines in HIV-1-infected patients. METHODS: Thirty HIV-1-positive patients on HAART with undetectable viral load were randomized into three groups: 10 received HIV-1 rgp160 vaccine, 10 received tetanus vaccine and 10 patients were not immunized. As controls, 10 HIV-negative volunteers were immunized with tetanus vaccine. The results were compared with an rgp160 vaccine study performed before the era of HAART on patients with comparable CD4 levels. All patients were monitored for 2 years for T-cell proliferative responses, T-cell subset levels, serum IgG and viral load. RESULTS: After 1 year all patients immunized with rgp160 had strong T-cell responses to the rgp160 antigen. After 2 years this response was preserved in the HAART-treated group, but not in the rgp160 immunized non-HAART group, despite comparable CD4 levels. Recall effects of the CD4-specific responses towards other antigens were seen in the rgp160-immunized HAART group. CONCLUSION: Immunization with rpg 160 leads to positive effects on HIV-specific T-cell proliferative responses in patients both with and without HAART. Immune responses after immunization is better preserved in HAART-treated patients who have low viral amounts than in individuals with high viral load and no HAART despite comparable CD4 levels during 2 years' follow-up. Interruption of HAART with return of a high viral load might thus have negative effects on T-cell functions in the long term, even if CD4 levels are kept at clinically acceptable levels.


Subject(s)
AIDS Vaccines/administration & dosage , HIV Infections/immunology , HIV Infections/therapy , HIV-1 , Adult , Antibodies, Viral/blood , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay/methods , Female , Follow-Up Studies , HIV-1/genetics , HIV-1/immunology , Humans , Immunologic Memory , Male , Middle Aged , RNA, Viral/blood , Statistics, Nonparametric , T-Lymphocytes/immunology , Tetanus Toxoid/administration & dosage
15.
Clin Microbiol Infect ; 8(8): 504-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12197872

ABSTRACT

Smallpox is a disease that followed humanity for thousands of years up until 30 years ago. It was possible to eradicate, because an effective live vaccine from crossreacting vaccinia could be developed. Twenty years have passed since vaccinations stopped and very few people are protected against the disease today. Variola today has become an object of discussion due to the possibility that it can be used as a bioweapon. Due to the number of complications that can be expected a general vaccination is probably not possible. Research is ongoing to develop new vaccines. Many countries are improving their capabilities to respond to a renewed threat of a smallpox epidemic.


Subject(s)
Bioterrorism/trends , Cytosine/analogs & derivatives , Organophosphonates , Smallpox , Antiviral Agents/therapeutic use , Biological Warfare/trends , Cidofovir , Cytosine/therapeutic use , Europe/epidemiology , History, 16th Century , History, 18th Century , History, 20th Century , History, Ancient , History, Medieval , Humans , Organophosphorus Compounds/therapeutic use , Smallpox/drug therapy , Smallpox/history , Smallpox/prevention & control , Smallpox/virology , Smallpox Vaccine/immunology , Smallpox Vaccine/supply & distribution , USSR
16.
Neuropharmacology ; 42(5): 731-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11985832

ABSTRACT

The therapeutic effects of ABR-215062, which is a new immunoregulator derived from Linomide, have been evaluated in experimental autoimmune neuritis (EAN), a CD4(+) T cell-mediated animal model of Guillain-Barré syndrome in man. In previous studies, we reported that Linomide suppressed the clinical EAN and myelin antigen-reactive T and B cell responses. Here EAN induced in Lewis rats by inoculation with peripheral nerve myelin P0 protein peptide 180-199 and Freund's complete adjuvant was strongly suppressed by ABR-215062 administered daily subcutaneously from the day of inoculation. ABR-215062 dose-dependently reduced the incidence of EAN, ameliorated clinical signs and inhibited P0 peptide 180-199-specific T cell responses as well as also the decreased inflammation and demyelination in the peripheral nerves. The suppression of clinical EAN was associated with inhibition of the inflammatory cytokines IFN-gamma and TNF-alpha, as well as the enhancement of anti-inflammatory cytokine IL-4 in lymph node cells and periphery nerve tissues, respectively, in a dose-dependent manner. These effects indicate that ABR-215062 may mediate its effects by regulation of Th1/Th2 cytokine balance and suggest that ABR-215062 is potentially a new chemical entity for effective treatment of autoimmune diseases.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cell Movement/drug effects , Hydroxyquinolines/therapeutic use , Neuritis, Autoimmune, Experimental/prevention & control , Peripheral Nerves/immunology , Peripheral Nerves/pathology , Th1 Cells/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Cell Movement/immunology , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Male , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Peripheral Nerves/drug effects , Peripheral Nerves/metabolism , Quinolones , Rats , Rats, Inbred Lew , Th1 Cells/drug effects , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
17.
Vaccine ; 20(3-4): 397-405, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11672902

ABSTRACT

DNA encoding HIV-1 env is a poorly efficient B-cell immunogen and one probable explanation is that the numerous gp120 N-linked glycans gp120 may interfere with B-cell epitope presentation. The N306 glycan in gp120 shields HIV-1 from neutralizing antibodies. A DNA immunogen lacking the N306 glycosylation signal (T308A) was constructed to determine whether this glycan affected the immune response. Mice were immunized intranasally twice with DNA containing either the wild type or the mutant env. Two additional groups were primed with wild type or mutant env and boosted with rgp160 protein, containing the complete set of N-linked glycans. Immunization with DNA alone resulted in priming of B-cell clones but was not sufficient to induce a complete antibody response. Animals primed with the N306 mutant and subsequently boosted with rgp160 protein displayed higher serum IgG-binding titers to gp120 than animals primed with wild type env DNA. The manipulation of the glycosylation sites of the env DNA strongly primes antibody responses (but non-neutralizing) as well as T-cell responses to the wild type strain gp160. However, priming with mutant plasmid did not result in higher neutralization titers to wild type or T308A-mutated virus than did the wild type plasmid. With the N306 mutant DNA we thus immunized a non-neutralization epitope, but obtained strong env-binding IgG after rgp160 boosting.


Subject(s)
AIDS Vaccines/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Vaccines, DNA/immunology , Animals , Female , Glycosylation , HIV Antibodies/blood , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp160/immunology , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Structure-Activity Relationship , T-Lymphocytes/immunology
18.
J Clin Virol ; 23(1-2): 1-15, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11595579

ABSTRACT

BACKGROUND: Identification of human cytomegalovirus (CMV) genome variation is important for understanding mutations associated with drug resistance. OBJECTIVES: To investigate the CMV resistance to foscarnet (PFA) and ganciclovir (GCV) in patients treated with antiviral drugs and to identify the DNA polymerase (UL54) and phosphotransferase (UL97) gene mutations inducing resistance. STUDY DESIGN: Antiviral susceptibility of CMV strains/isolates for PFA and GCV was compared by plaque reduction assay and in situ ELISA. UL54 and UL97 gene mutations were identified by sequencing. Growth phenotype of two CMV recombinants with mutations in UL54 was studied. RESULTS: Six of seven GCV resistant strains had alterations within the UL97. Five of them also had alterations in the UL54 (F412C, L802M or K513E), previously shown to induce GCV resistance. Seven isolates had no or reduced susceptibility to PFA, which had alterations in the UL54 (D588N, E756K, V781I or L802M). By in vitro mutagenesis, it was shown that a mutation at codon D588N of UL54 conferred 9-fold reduced susceptibility to PFA, while a mutation at codon V781I induced 4-fold reduced susceptibility to PFA and GCV. Both recombinants showed the same kinetics of protein expression (IE, E, and L antigen) and virus yields as the CMV Towne strain. CONCLUSIONS: The recombinants containing alterations within the UL54 (D588N and V781I) showed a reduced susceptibility to antiviral drugs but no change in the replication rate compared to the CMV Towne.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , DNA-Directed DNA Polymerase/genetics , Foscarnet/pharmacology , Ganciclovir/pharmacology , Genes, Viral , Phosphotransferases/genetics , Codon , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/virology , Drug Resistance, Multiple, Viral/genetics , Genetic Variation , Humans , Microbial Sensitivity Tests , Virus Replication
19.
Hum Gene Ther ; 12(13): 1623-37, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11535166

ABSTRACT

DNA plasmid immunization has the important advantage over traditional vaccines of making it possible to combine selected genes into one vaccine. The efficacy of a combination of DNA plasmids encoding the nef, rev, and tat HIV-1 regulatory genes in inducing cellular immune responses was analyzed in asymptomatic HIV-1-infected patients. Patients initially selected for having low or no detectable immune responses to Nef, Rev, or Tat antigens developed MHC class I-restricted cytolytic activities as well as enhanced bystander effects. The induction of memory cells against target cells infected with the whole HIV-1 genome was analyzed by using a pseudovirus HIV-1/murine leukemia virus (MuLV), and target cells infected with vaccinia virus carrying the respective gene. The most remarkable change observed after immunization with the gene combination was an increase in cytotoxic T lymphocyte (CTL) precursors to target cells infected with the whole HIV-1 genome. Infection by the pseudotype HIV-1/MuLV virus should result in a multitude of HIV-1 peptides presented on the target cell surface, representative of the in vivo situation. An in vitro assessment of the expression of the single and combined gene products showed that this was consistent with the induction of CTL responses in vivo. No clinical advantage or adverse effects were noted. Therapeutic effects of such immunization may become measurable by structured therapy interruption.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , HIV Antigens/genetics , HIV Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/therapeutic use , CD4 Lymphocyte Count , CpG Islands/genetics , Cytotoxicity, Immunologic , Gene Expression , Gene Products, nef/biosynthesis , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, nef/therapeutic use , Gene Products, rev/biosynthesis , Gene Products, rev/genetics , Gene Products, rev/immunology , Gene Products, rev/therapeutic use , Gene Products, tat/biosynthesis , Gene Products, tat/genetics , Gene Products, tat/immunology , Gene Products, tat/therapeutic use , Genes, Viral/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV Antigens/biosynthesis , HIV Antigens/immunology , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , HeLa Cells , Histocompatibility Antigens Class I/immunology , Humans , Leukemia Virus, Murine/genetics , Lymphocyte Activation , Plasmids/genetics , T-Lymphocytes, Cytotoxic/cytology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use , Vaccinia virus/genetics , nef Gene Products, Human Immunodeficiency Virus , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
20.
Vaccine ; 19(31): 4434-44, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11483269

ABSTRACT

We studied the use of a DNA vaccine expressing the matrix (M) gene of the influenza virus A/PR/8/34. Mice were immunized by painting the DNA vaccine three times on the skin after removal of its keratinocytic layers. Immunization by this method produced M-specific antibodies and cytotoxic T lymphocyte (CTL) response, and acquired resistance against influenza virus challenge. This protection was abrogated by the in vivo injection of anti-CD8 or anti-CD4 monoclonal antibody. We further found that simultaneous topical application (t.a.) of GM-CSF expression plasmid (pGM-CSF) or liposomes plus mannan produced stronger immune response competence and enhanced the protective effect against influenza virus challenge. The present study revealed that administering DNA vaccine by topical application can elicit both humoral and cell-mediated immunity (CMI).


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/therapeutic use , Orthomyxoviridae Infections/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use , Administration, Cutaneous , Animals , Antibodies, Viral/biosynthesis , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Edema/pathology , Male , Mice , Mice, Inbred BALB C , Skin/pathology , Survival Rate , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...