Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 95(22): e0038721, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34469243

ABSTRACT

Preexisting immune responses toward adenoviral vectors limit the use of a vector based on particular serotypes and its clinical applicability for gene therapy and/or vaccination. Therefore, there is a significant interest in vectorizing novel adenoviral types that have low seroprevalence in the human population. Here, we describe the discovery and vectorization of a chimeric human adenovirus, which we call HAdV-20-42-42. Full-genome sequencing revealed that this virus is closely related to human serotype 42, except for the penton base, which is derived from serotype 20. The HAdV-20-42-42 vector could be propagated stably to high titers on existing E1-complementing packaging cell lines. Receptor-binding studies revealed that the vector utilized both CAR and CD46 as receptors for cell entry. Furthermore, the HAdV-20-42-42 vector was potent in transducing human and murine cardiovascular cells and tissues, irrespective of the presence of blood coagulation factor X. In vivo characterizations demonstrate that when delivered intravenously (i.v.) in mice, HAdV-20-42-42 mainly targeted the lungs, liver, and spleen and triggered robust inflammatory immune responses. Finally, we demonstrate that potent T-cell responses against vector-delivered antigens could be induced upon intramuscular vaccination in mice. In summary, from the data obtained we conclude that HAdV-20-42-42 provides a valuable addition to the portfolio of adenoviral vectors available to develop efficacious products in the fields of gene therapy and vaccination. IMPORTANCE Adenoviral vectors are under investigation for a broad range of therapeutic indications in diverse fields, such as oncology and gene therapy, as well as for vaccination both for human and veterinary use. A wealth of data shows that preexisting immune responses may limit the use of a vector. Particularly in the current climate of global pandemic, there is a need to expand the toolbox with novel adenoviral vectors for vaccine development. Our data demonstrate that we have successfully vectorized a novel adenovirus type candidate with low seroprevalence. The cell transduction data and antigen-specific immune responses induced in vivo demonstrate that this vector is highly promising for the development of gene therapy and vaccine products.


Subject(s)
Adenoviruses, Human , Genetic Therapy/methods , Genetic Vectors , Vaccine Development/methods , A549 Cells , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Genetic Vectors/genetics , Genetic Vectors/immunology , HEK293 Cells , Humans , Male , Mice , Seroepidemiologic Studies
2.
Clin Vaccine Immunol ; 17(11): 1687-94, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20826614

ABSTRACT

The most advanced malaria vaccine, RTS,S, is comprised of a portion of the Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed with the hepatitis B virus surface antigen, and an adjuvant [corrected].This vaccine confers short-term protection against malaria infection, with an efficacy of about 50%, and induces particularly B-cell and CD4(+) T-cell responses.In the present study, we tested the hypothesis that the Th1 immune response to CS protein,in particular the CD8(+) T-cell response, which is needed for strong and lasting malaria immunity, is boosted to sustainable levels by adenovirus vectors 35 and 26 with a homologous insert (Ad35.CS/Ad26.CS) [corrected]. In this study, we evaluated immune responses induced with vaccination regimens based on an adjuvant-containing, yeast-produced complete CS protein followed by two recombinant low-seroprevalence adenoviruses expressing P. falciparum CS antigen, Ad35.CS (subgroup B) and Ad26.CS (subgroup D). Our results show that (i) the yeast (Hansenula polymorpha)produced, adjuvanted full-length CS protein is highly potent in inducing high CS-specific humoral responses in mice but produces poor T-cell responses, (ii) the Ad35.CS vector boosts the gamma interferon-positive (IFN-γ(+)) CD8(+) T-cell response induced by the CS protein immunization and shifts the immune response toward the Th1 type, and (iii) a three-component heterologous vaccination comprised of a CS protein prime followed by boosts with Ad35.CS and Ad26.CS elicits an even more robust and sustainable IFN-γ(+) CD8(+) T-cell response than one- or two-component regimens. The Ad35.CS/Ad26.CS combination boosted particularly the IFN-γ(+) and tumor necrosis factor alpha-positive (TNF-α(+)) T cells, confirming the shift of the immune response from the Th2 type to the Th1 type. These results support the notion of first immunizations of infants with an adjuvanted CS protein vaccine, followed by a booster Ad35.CS/Ad26.CS vaccine at a later age, to induce lasting protection against malaria for which the Th1 response and immune memory is required.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Th1 Cells/immunology , Animals , Antibodies, Protozoan/blood , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunization, Secondary/methods , Interferon-gamma/metabolism , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Pichia/genetics , Protozoan Proteins/genetics , Vaccination/methods , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
3.
J Virol ; 82(10): 4844-52, 2008 May.
Article in English | MEDLINE | ID: mdl-18337575

ABSTRACT

Recombinant adenovirus serotype 5 (rAd5) vaccine vectors for human immunodeficiency virus type 1 (HIV-1) and other pathogens have been shown to elicit antigen-specific cellular immune responses. Rare serotype rAd vectors have also been constructed to circumvent preexisting anti-Ad5 immunity and to facilitate the development of novel heterologous rAd prime-boost regimens. Here we show that rAd5, rAd26, and rAd48 vectors elicit qualitatively distinct phenotypes of cellular immune responses in rhesus monkeys and can be combined as potent heterologous prime-boost vaccine regimens. While rAd5-Gag induced primarily gamma interferon-positive (IFN-gamma(+)) and IFN-gamma(+)/tumor necrosis factor alpha(+) (TNF-alpha(+)) T-lymphocyte responses, rAd26-Gag and rAd48-Gag induced higher proportions of interleukin-2(+) (IL-2(+)) and polyfunctional IFN-gamma(+)/TNF-alpha(+)/IL-2(+) T-lymphocyte responses. Priming with the rare serotype rAd vectors proved remarkably effective for subsequent boosting with rAd5 vectors. These data demonstrate that the rare serotype rAd vectors elicited T-lymphocyte responses that were phenotypically distinct from those elicited by rAd5 vectors and suggest the functional relevance of polyfunctional CD8(+) and CD4(+) T-lymphocyte responses. Moreover, qualitative differences in cellular immune responses may prove critical in determining the overall potency of heterologous rAd prime-boost regimens.


Subject(s)
Adenoviridae/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , SAIDS Vaccines/immunology , Animals , Gene Products, gag/genetics , Gene Products, gag/immunology , Immunization/methods , Immunization, Secondary/methods , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Macaca mulatta , Tumor Necrosis Factor-alpha/biosynthesis
4.
J Virol ; 81(9): 4654-63, 2007 May.
Article in English | MEDLINE | ID: mdl-17329340

ABSTRACT

Recombinant adenovirus serotype 5 (rAd5) vector-based vaccines are currently being developed for both human immunodeficiency virus type 1 and other pathogens. The potential limitations associated with rAd5 vectors, however, have led to the construction of novel rAd vectors derived from rare Ad serotypes. Several rare serotype rAd vectors have already been described, but a detailed comparison of multiple rAd vectors from subgroups B and D has not previously been reported. Such a comparison is critical for selecting optimal rAd vectors for advancement into clinical trials. Here we describe the construction of three novel rAd vector systems from Ad26, Ad48, and Ad50. We report comparative seroprevalence and immunogenicity studies involving rAd11, rAd35, and rAd50 vectors from subgroup B; rAd26, rAd48, and rAd49 vectors from subgroup D; and rAd5 vectors from subgroup C. All six rAd vectors from subgroups B and D exhibited low seroprevalence in a cohort of 200 individuals from sub-Saharan Africa, and they elicited Gag-specific cellular immune responses in mice both with and without preexisting anti-Ad5 immunity. The rAd vectors from subgroup D were also evaluated using rhesus monkeys and were shown to be immunogenic after a single injection. The rAd26 vectors proved the most immunogenic among the rare serotype rAd vectors studied, although all rare serotype rAd vectors were still less potent than rAd5 vectors in the absence of anti-Ad5 immunity. These studies substantially expand the portfolio of rare serotype rAd vectors that may prove useful as vaccine vectors for the developing world.


Subject(s)
Adenoviridae Infections/epidemiology , Adenoviridae/genetics , Genetic Vectors/genetics , Vaccines, Synthetic/genetics , Viral Vaccines/genetics , Adenoviridae Infections/blood , Africa South of the Sahara/epidemiology , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/immunology , Humans , Macaca mulatta , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutralization Tests , Sequence Analysis, DNA , Seroepidemiologic Studies , Serotyping
5.
J Clin Microbiol ; 44(10): 3781-3, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17021110

ABSTRACT

We assessed neutralizing antibody titers to adenovirus serotype 5 (Ad5) and six rare adenovirus serotypes, serotypes 11, 35, 50, 26, 48, and 49, in pediatric populations in sub-Saharan Africa. We observed a clear age dependence of Ad5-specific neutralizing antibody titers. These data will help to guide the development of Ad vector-based vaccines for human immunodeficiency virus type 1 and other pathogens.


Subject(s)
Adenovirus Infections, Human/immunology , Adenoviruses, Human/immunology , Aging , Antibodies, Viral/blood , Adenovirus Infections, Human/blood , Adenovirus Infections, Human/epidemiology , Adolescent , Africa South of the Sahara/epidemiology , Child , Child, Preschool , Humans , Infant , Infectious Disease Transmission, Vertical , Seroepidemiologic Studies
6.
J Virol ; 80(24): 12009-16, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035318

ABSTRACT

The high prevalence of preexisting immunity to adenovirus serotype 5 (Ad5) in human populations has led to the development of recombinant adenovirus (rAd) vectors derived from rare Ad serotypes as vaccine candidates for human immunodeficiency virus type 1 and other pathogens. Vaccine vectors have been constructed from Ad subgroup B, including rAd11 and rAd35, as well as from Ad subgroup D, including rAd49. However, the optimal combination of vectors for heterologous rAd prime-boost vaccine regimens and the extent of cross-reactive vector-specific neutralizing antibodies (NAbs) remain poorly defined. We have shown previously that the closely related vectors rAd11 and rAd35 elicited low levels of cross-reactive NAbs. Here we show that these cross-reactive NAbs correlated with substantial sequence homology in the hexon hypervariable regions (HVRs) and suppressed the immunogenicity of heterologous rAd prime-boost regimens. In contrast, vectors with lower hexon HVR homology, such as rAd35 and rAd49, did not elicit detectable cross-reactive vector-specific NAbs. Consistent with these findings, rAd35-rAd49 vaccine regimens proved more immunogenic than both rAd35-rAd5 and rAd35-rAd11 regimens in mice with anti-Ad5 immunity. These data suggest that optimal heterologous rAd prime-boost regimens should include two vectors that are both rare in human populations to circumvent preexisting antivector immunity as well as sufficiently immunologically distinct to avoid cross-reactive antivector immunity.


Subject(s)
Adenoviridae/immunology , Cross Reactions/immunology , Genetic Vectors/immunology , Vaccines, Synthetic/immunology , Adenoviridae/genetics , Animals , Antibodies/immunology , Antigens, Viral/genetics , Capsid Proteins/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Neutralization Tests
7.
J Gen Virol ; 87(Pt 10): 2891-2899, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16963747

ABSTRACT

Recombinant adenoviral vectors based on type 5 (rAd5) show great promise as a vaccine carrier. However, neutralizing activity against Ad5 is prevalent and high-titred among human populations, and significantly dampens Ad5-based vaccine modalities. The generation of alternative adenoviral vectors with low seroprevalence thus receives much research attention. Here, it is shown that a member from human adenovirus subgroup D, i.e. Ad49, does not cross-react with Ad5 neutralizing activity, making it a candidate serotype for vector development. Therefore, a plasmid system that allows formation of replication-incompetent adenovirus serotype 49 vaccine vectors (rAd49) was constructed and it was demonstrated that rAd49 can be successfully propagated to high titres on existing Ad5.E1-complementing cell lines such as PER.C6. Using an rAd49 vector carrying the luciferase marker gene, detailed seroprevalence studies were performed, demonstrating that rAd49 has low seroprevalence and neutralizing antibody titres worldwide. Also, we have initiated rAd49 vector receptor usage suggesting that rAd49 utilizes hCD46 as a cellular receptor. Finally, the immunogenicity of the rAd49 vector was assessed and it was shown that an rAd49.SIVGag vaccine induces strong anti-SIVGag CD8+ T-lymphocytes in naïve mice, albeit less than an rAd5.SIVGag vaccine. However, in mice with high anti-Ad5 immunity the rAd5.SIVGag vaccine was severely blunted, whereas the anti-SIVGag response was not significantly suppressed using the rAd49.SIVGag vaccine. These data demonstrate the potential of a replication deficient human group D adenoviral vector for vaccination purposes.


Subject(s)
Adenoviruses, Human/growth & development , Adenoviruses, Human/immunology , Virus Replication , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Antibodies, Viral , Cell Line , Genetic Engineering , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Membrane Cofactor Protein , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Organ Specificity , Vaccines, Synthetic , Viral Vaccines/immunology
8.
Nature ; 441(7090): 239-43, 2006 May 11.
Article in English | MEDLINE | ID: mdl-16625206

ABSTRACT

A common viral immune evasion strategy involves mutating viral surface proteins in order to evade host neutralizing antibodies. Such immune evasion tactics have not previously been intentionally applied to the development of novel viral gene delivery vectors that overcome the critical problem of anti-vector immunity. Recombinant, replication-incompetent adenovirus serotype 5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 and other pathogens have proved highly immunogenic in preclinical studies but will probably be limited by the high prevalence of pre-existing anti-Ad5 immunity in human populations, particularly in the developing world. Here we show that rAd5 vectors can be engineered to circumvent anti-Ad5 immunity. We constructed novel chimaeric rAd5 vectors in which the seven short hypervariable regions (HVRs) on the surface of the Ad5 hexon protein were replaced with the corresponding HVRs from the rare adenovirus serotype Ad48. These HVR-chimaeric rAd5 vectors were produced at high titres and were stable through serial passages in vitro. HVR-chimaeric rAd5 vectors expressing simian immunodeficiency virus Gag proved comparably immunogenic to parental rAd5 vectors in naive mice and rhesus monkeys. In the presence of high levels of pre-existing anti-Ad5 immunity, the immunogenicity of HVR-chimaeric rAd5 vectors was not detectably suppressed, whereas the immunogenicity of parental rAd5 vectors was abrogated. These data demonstrate that functionally relevant Ad5-specific neutralizing antibodies are focused on epitopes located within the hexon HVRs. Moreover, these studies show that recombinant viral vectors can be engineered to circumvent pre-existing anti-vector immunity by removing key neutralizing epitopes on the surface of viral capsid proteins. Such chimaeric viral vectors may have important practical implications for vaccination and gene therapy.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Engineering , Genetic Vectors/genetics , Genetic Vectors/immunology , Adenoviridae/classification , Adenoviridae/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , DNA, Recombinant/genetics , Genetic Therapy , Macaca mulatta/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests , Vaccines
9.
J Virol ; 79(22): 14161-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16254351

ABSTRACT

Preexisting immunity to adenovirus serotype 5 (Ad5) has been shown to suppress the immunogenicity of recombinant Ad5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 (HIV-1) in both preclinical studies and clinical trials. A potential solution to this problem is to utilize rAd vectors derived from rare Ad serotypes, such as Ad35. However, rAd35 vectors have appeared less immunogenic than rAd5 vectors in preclinical studies to date. In this study, we explore the hypothesis that the differences in immunogenicity between rAd5 and rAd35 vectors may be due in part to differences between the fiber proteins of these viruses. We constructed capsid chimeric rAd35 vectors containing the Ad5 fiber knob (rAd35k5) and compared the immunogenicities of rAd5, rAd35k5, and rAd35 vectors expressing simian immunodeficiency virus Gag and HIV-1 Env in mice and rhesus monkeys. In vitro studies demonstrated that rAd35k5 vectors utilized the Ad5 receptor CAR rather than the Ad35 receptor CD46. In vivo studies showed that rAd35k5 vectors were more immunogenic than rAd35 vectors in both mice and rhesus monkeys. These data suggest that the Ad5 fiber knob contributes substantially to the immunogenicity of rAd vectors. Moreover, these studies demonstrate that capsid chimeric rAd vectors can be constructed to combine beneficial immunologic and serologic properties of different Ad serotypes.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Capsid Proteins/genetics , Viral Vaccines , Adenoviridae/classification , Adenoviridae/genetics , Animals , Epitopes/chemistry , Epitopes/immunology , Immunization , Macaca mulatta , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Serotyping , Virus Replication
10.
J Virol ; 79(15): 9694-701, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014931

ABSTRACT

The high prevalence of preexisting immunity to adenovirus serotype 5 (Ad5) in human populations will likely limit the immunogenicity and clinical utility of recombinant Ad5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 and other pathogens. A potential solution to this problem is to utilize rAd vaccine vectors derived from rare Ad serotypes such as Ad35 and Ad11. We have previously reported that rAd35 vectors were immunogenic in the presence of anti-Ad5 immunity, but the immunogenicity of heterologous rAd prime-boost regimens and the extent that cross-reactive anti-vector immunity may limit this approach have not been fully explored. Here we assess the immunogenicity of heterologous vaccine regimens involving rAd5, rAd35, and novel rAd11 vectors expressing simian immunodeficiency virus Gag in mice both with and without anti-Ad5 immunity. Heterologous rAd prime-boost regimens proved significantly more immunogenic than homologous regimens, as expected. Importantly, all regimens that included rAd5 were markedly suppressed by anti-Ad5 immunity. In contrast, rAd35-rAd11 and rAd11-rAd35 regimens elicited high-frequency immune responses both in the presence and in the absence of anti-Ad5 immunity, although we also detected clear cross-reactive Ad35/Ad11-specific humoral and cellular immune responses. Nevertheless, these data suggest the potential utility of heterologous rAd prime-boost vaccine regimens using vectors derived from rare human Ad serotypes.


Subject(s)
Adenoviruses, Human/immunology , Genetic Vectors/immunology , Reassortant Viruses/immunology , Viral Vaccines/immunology , Animals , Antibody Formation , Cross Reactions , Drug Evaluation, Preclinical , Gene Products, gag/genetics , Genetic Therapy , Immunity, Cellular , Immunization, Secondary , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Simian Immunodeficiency Virus/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
11.
J Immunol ; 174(11): 7179-85, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905562

ABSTRACT

The utility of recombinant adenovirus serotype 5 (rAd5) vector-based vaccines for HIV-1 and other pathogens will likely be limited by the high prevalence of pre-existing Ad5-specific neutralizing Abs (NAbs) in human populations. However, the immunodominant targets of Ad5-specific NAbs in humans remain poorly characterized. In this study, we assess the titers and primary determinants of Ad5-specific NAbs in individuals from both the United States and the developing world. Importantly, median Ad5-specific NAb titers were >10-fold higher in sub-Saharan Africa compared with the United States. Moreover, hexon-specific NAb titers were 4- to 10-fold higher than fiber-specific NAb titers in these cohorts by virus neutralization assays using capsid chimeric viruses. We next performed adoptive transfer studies in mice to evaluate the functional capacity of hexon- and fiber-specific NAbs to suppress the immunogenicity of a prototype rAd5-Env vaccine. Hexon-specific NAbs were remarkably efficient at suppressing Env-specific immune responses elicited by the rAd5 vaccine. In contrast, fiber-specific NAbs exerted only minimal suppressive effects on rAd5 vaccine immunogenicity. These data demonstrate that functionally significant Ad5-specific NAbs are directed primarily against the Ad5 hexon protein in both humans and mice. These studies suggest a potential strategy for engineering novel Ad5 vectors to evade dominant Ad5-specific NAbs.


Subject(s)
Adenoviruses, Human/immunology , Antibodies, Viral/physiology , Capsid Proteins/immunology , Genetic Vectors/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Adenoviruses, Human/genetics , Adult , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Dose-Response Relationship, Immunologic , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutralization Tests , Seroepidemiologic Studies , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
12.
J Leukoc Biol ; 77(3): 337-43, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15601667

ABSTRACT

Recently, we described the molecular identification of dendritic cell-specific TrAnsMembrane protein (DC-STAMP), a multimembrane-spanning protein preferentially expressed by human DC (hDC). In this report, we describe the identification and expression profile of the murine homologue of DC-STAMP (mDC-STAMP) as well as the characterization of the DC-STAMP protein. The results demonstrate that mDC-STAMP is over 90% homologous to hDC-STAMP and is also preferentially expressed by DC in vitro and ex vivo. mDC-STAMP expression is enhanced by interleukin-4 and down-regulated upon DC maturation. Analysis of differently tagged DC-STAMP proteins further demonstrates that hDC-STAMP and mDC-STAMP are glycosylated and primarily localize to an intracellular compartment. Applying confocal microscopy and electron microscopy, we demonstrate that hDC-STAMP localizes to the endoplasmic reticulum (ER) in human embryonic kidney 293 cells as well as hDC transduced with an adenovirus encoding hDC-STAMP-green fluorescent protein fusion protein. These data imply that DC-STAMP may exert its effect in the ER.


Subject(s)
Dendritic Cells/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid
13.
Eur J Epidemiol ; 19(6): 513-6, 2004.
Article in English | MEDLINE | ID: mdl-15330122

ABSTRACT

Considerable progress has been made over the past several years in the development of an HIV vaccine. As a result, a growing number of vaccine modalities are being investigated in pre-clinical and phase I/II clinical trials. However, a number of major scientific challenges still remain. It is widely believed that the ideal vaccine should elicit both neutralizing antibodies and cytotoxic T lymphocytes (CTL) against diverse isolates of HIV, but the precise correlates of immunity have not been defined. Recombinant live vector-based vaccines and plasmid DNA vaccines have been shown to induce CTL, either alone or in combination, and these CTL-based vaccines have shown partial protective efficacy in nonhuman primates challenge studies. An immunogen that elicits broadly reactive neutralizing antibodies, however, has yet to be developed.


Subject(s)
AIDS Vaccines , Acquired Immunodeficiency Syndrome/prevention & control , HIV/immunology , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , Humans , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...