Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Exp Immunol ; 182(3): 289-301, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26332605

ABSTRACT

VRC-HIVMAB060-00-AB (VRC01) is a broadly neutralizing HIV-1 monoclonal antibody (mAb) isolated from the B cells of an HIV-infected patient. It is directed against the HIV-1 CD4 binding site and is capable of potently neutralizing the majority of diverse HIV-1 strains. This Phase I dose-escalation study in healthy adults was conducted at the National Institutes of Health (NIH) Clinical Center (Bethesda, MD, USA). Primary objectives were the safety, tolerability and pharmacokinetics (PK) of VRC01 intravenous (i.v.) infusion at 5, 20 or 40 mg/kg, given either once (20 mg/kg) or twice 28 days apart (all doses), and of subcutaneous (s.c.) delivery at 5 mg/kg compared to s.c. placebo given twice, 28 days apart. Cumulatively, 28 subjects received 43 VRC01 and nine received placebo administrations. There were no serious adverse events or dose-limiting toxicities. Mean 28-day serum trough concentrations after the first infusion were 35 and 57 µg/ml for groups infused with 20 mg/kg (n = 8) and 40 mg/kg (n = 5) doses, respectively. Mean 28-day trough concentrations after the second infusion were 56 and 89 µg/ml for the same two doses. Over the 5-40 mg/kg i.v. dose range (n = 18), the clearance was 0.016 l/h and terminal half-life was 15 days. After infusion VRC01 retained expected neutralizing activity in serum, and anti-VRC01 antibody responses were not detected. The human monoclonal antibody (mAb) VRC01 was well tolerated when delivered i.v. or s.c. The mAb demonstrated expected half-life and pharmacokinetics for a human immunoglobulin G. The safety and PK results support and inform VRC01 dosing schedules for planning HIV-1 prevention efficacy studies.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , HIV Antibodies , HIV Infections , HIV-1 , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Broadly Neutralizing Antibodies , Dose-Response Relationship, Drug , Female , HIV Antibodies/administration & dosage , HIV Antibodies/adverse effects , HIV Infections/blood , HIV Infections/drug therapy , Half-Life , Humans , Male , Middle Aged
2.
Clin Vaccine Immunol ; 19(11): 1792-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956656

ABSTRACT

Avian influenza virus causes outbreaks in domestic and wild birds around the world, and sporadic human infections have been reported. A DNA vaccine encoding hemagglutinin (HA) protein from the A/Indonesia/5/05 (H5N1) strain was initially tested in two randomized phase I clinical studies. Vaccine Research Center study 304 (VRC 304) was a double-blinded study with 45 subjects randomized to placebo, 1 mg of vaccine, or 4 mg of vaccine treatment groups (n = 15/group) by intramuscular (i.m.) Biojector injection. VRC 305 was an open-label study to evaluate route, with 44 subjects randomized to intradermal (i.d.) injections of 0.5 mg by needle/syringe or by Biojector or 1 mg delivered as two 0.5-mg Biojector injections in the same deltoid or as 0.5 mg in each deltoid (n = 11/group). Injections were administered at weeks 0, 4, and 8 in both studies. Antibody responses to H5 were assessed by hemagglutination inhibition (HAI) assay, enzyme-linked immunosorbent assay (ELISA), and neutralization assay, and the H5 T cell responses were assessed by enzyme-linked immunospot and intracellular cytokine staining assays. There were no vaccine-related serious adverse events, and the vaccine was well tolerated in all groups. At 1 mg, i.d. vaccination compared to i.m. vaccination induced a greater frequency and magnitude of response by ELISA, but there were no significant differences in the frequency or magnitude of response between the i.d. and i.m. routes in the HAI or neutralization assays. T cell responses were more common in subjects who received the 1- or 4-mg dose i.m. These studies demonstrated that the DNA vaccine encoding H5 is safe and immunogenic and served to define the proper dose and route for further studies. The i.d. injection route did not offer a significant advantage over the i.m. route, and no difference was detected by delivery to one site versus splitting the dose between two sites for i.d. vaccine administration. The 4-mg dose (i.m) was further investigated in prime-boost regimens.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Adult , Antibodies, Viral/blood , Cytokines/metabolism , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza Vaccines/genetics , Injections, Intradermal , Injections, Intramuscular , Male , Middle Aged , Neutralization Tests , Placebos/administration & dosage , T-Lymphocytes/immunology , Vaccines, DNA/genetics , Young Adult
3.
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
4.
J Virol Methods ; 147(2): 197-205, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18079007

ABSTRACT

This report describes a fast, reproducible, inexpensive and convenient assay system for virus titration in the 96-well format. The micromethod substantially increases assay throughput and improves the data reproducibility. A highly simplified variant of virus quantification is based on immunohistochemical detection of virus amplification foci obtained without use of agarose or semisolid overlays. It can be incorporated into several types of routine virological assays successfully replacing the laborious and time-consuming conventional methods based on plaque formation under semisolid overlays. The method does not depend on the development of CPE and can be accommodated to assay viruses with substantial differences in growth properties. The use of enhanced immunohistochemical detection enabled a five- to six-fold reduction of the total assay time. The micromethod was specifically developed to take advantage of multichannel pipettor use to simplify handling of a large number of samples. The method performs well with an inexpensive low-power binocular, thus offering a routine assay system usable outside of specialized laboratory setting, such as for testing of clinical or field samples. When used in focus reduction-neutralization tests (FRNT), the method accommodates very small volumes of immune serum, which is often a decisive factor in experiments involving small rodent models.


Subject(s)
Immunoenzyme Techniques/methods , Virology/methods , Viruses/isolation & purification , Animals , Cell Line , Reproducibility of Results , Sensitivity and Specificity , Viral Plaque Assay
5.
Clin Cancer Res ; 7(10): 3012-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595689

ABSTRACT

A melanoma vaccine composed of HLA-A2-restricted peptide YLEPGPVTA (gp100(280)), with or without a modified T-helper epitope from tetanus toxoid AQYIKANSKFIGITEL, has been evaluated in a Phase I trial to assess safety and immunological response. The vaccines were administered s.c. in either of two adjuvants, Montanide ISA-51 or QS-21, to 22 patients with high-risk resected melanoma (stage IIB-IV). Local and systemic toxicities were mild and transient. We detected CTL responses to the gp100(280) peptide in peripheral blood in 14% of patients. Helper T-cell responses to the tetanus helper peptide were detected in 79% of patients and had a Th1 cytokine profile. One patient with a CTL response to gp100 had a recurrence in a lymph node 2 years later; her nodes contained CD8+ cells reactive to gp100(280) (0.24%), which proliferated in response to peptide. The overall survival of patients is 75% (95% confidence interval, 57-94%) at 4.7 years follow-up, which compares favorably with expected survival. Four of 14 patients who completed at least six vaccines subsequently developed metastases, all of which were solitary and surgically resectable. They remain alive and clinically free of disease at last follow-up. Data from this trial demonstrate immunogenicity of the gp100(280) peptide and suggest that immune responses may persist long-term in some patients. The frequency and magnitude of the CTL response may be improved with more aggressive vaccination regimens. Although this Phase I study was not intended to evaluate clinical benefit, the excellent survival of patients on this protocol suggests the possibility of a benefit that should be assessed in future studies.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Epitopes, T-Lymphocyte/administration & dosage , Melanoma/prevention & control , Membrane Glycoproteins/administration & dosage , Neoplasm Proteins/administration & dosage , Tetanus Toxoid/immunology , Adjuvants, Immunologic/adverse effects , Amino Acid Sequence , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cell Division/drug effects , Cell Line , Cytokines/drug effects , Cytokines/metabolism , Epitopes, T-Lymphocyte/adverse effects , Epitopes, T-Lymphocyte/immunology , Female , Follow-Up Studies , HLA-A2 Antigen/immunology , Headache/chemically induced , Humans , Hypersensitivity, Delayed/immunology , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Melanoma/immunology , Membrane Glycoproteins/adverse effects , Membrane Glycoproteins/immunology , Middle Aged , Molecular Sequence Data , Neoplasm Proteins/adverse effects , Neoplasm Proteins/immunology , Neoplasm Staging , Pain/chemically induced , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Peptides , Saponins/administration & dosage , Saponins/adverse effects , Skin/drug effects , Skin/immunology , Skin Diseases/chemically induced , Skin Tests , Survival Analysis , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/cytology , Th1 Cells/drug effects
6.
Int J Cancer ; 92(5): 703-11, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11340576

ABSTRACT

Many peptide epitopes for cytotoxic T lymphocytes (CTLs) have been identified from melanocytic differentiation proteins. Vaccine trials with these peptides have been limited mostly to those associated with HLA-A2, and immune responses have been detected inconsistently. Cases of clinical regression have been observed after peptide vaccination in some trials, but melanoma regressions have not correlated well with T-cell responses measured in peripheral blood lymphocytes (PBLs). We vaccinated stage IV melanoma patients with a mixture of gp100 and tyrosinase peptides restricted by HLA-A1 (DAEKSDICTDEY), HLA-A2 (YLEPGPVTA and YMDGTMSQV) and HLA-A3 (ALLAVGATK) in an emulsion with GM-CSF and Montanide ISA-51 adjuvant. CTL responses were assessed in PBLs and in a lymph node draining a vaccine site (sentinel immunized node, SIN). We found CTL responses to vaccinating peptides in the SIN in 5/5 patients (100%). Equivalent assays detected peptide-reactive CTLs in PBLs of 2 of these 5 patients (40%). CTLs expanded from the SIN lysed melanoma cells naturally expressing tyrosinase or gp100. We demonstrated immunogenicity for peptides restricted by HLA-A1 and -A3 and for 1 HLA-A2 restricted peptide, YMDGTMSQV. Immune monitoring of clinical trials by evaluation of PBLs alone may under-estimate immunogenicity; evaluation of SIN provides a new and sensitive approach for defining responses to tumor vaccines and correlating these responses with clinical outcomes. This combination of an immunogenic vaccine strategy with a sensitive analysis of CTL responses demonstrates the potential for inducing and detecting anti-tumor immune responses in the majority of melanoma patients.


Subject(s)
Cancer Vaccines/immunology , HLA-A Antigens/immunology , Lymph Nodes/immunology , Melanoma/therapy , Membrane Glycoproteins/immunology , Monophenol Monooxygenase/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Adult , Amino Acid Sequence , Antigens, Neoplasm , Humans , Melanoma/immunology , Melanoma-Specific Antigens , Middle Aged , Molecular Sequence Data , T-Lymphocytes, Cytotoxic/immunology , gp100 Melanoma Antigen
7.
Clin Cancer Res ; 7(3 Suppl): 909s-916s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300491

ABSTRACT

The long-term survival of some patients with metastatic melanoma may be attributable in part to cellular immune responses to melanoma antigens. However, little is known about the level of CTL reactivity in vivo that is required for immunological control of tumor progression. In the present report, T-cell responses were evaluated with lymphocytes obtained from tumor-involved nodes and peripheral blood of a long-term melanoma survivor. Using an ELISPOT assay, naturally occurring functional T cells, which recognize the peptide ALLAVGATK (gp100(17-25)) plus two other HLA-A3 restricted peptides, were detected in a tumor-involved lymph node. The ALLAVGATK-reactive T cells were also evaluated by MHC-tetramers staining and were found to be CD8+ CD45RO+ L-selectin(-) CD11a+, suggesting that they are antigen experienced and have a memory phenotype. Unstimulated peripheral blood lymphocytes from the same patient demonstrated no detectable T-cell responses; however, a single stimulation with ALLAVGATK peptide in vitro resulted in a dramatic expansion of peptide-reactive CTLs. This patient, with evidence of tumor-reactive CTLs targeted to several tumor antigens in a tumor-involved lymph node and with evidence of a circulating memory T-cell response, has remained disease-free for 6 years, despite prior bulky nodal metastasis. In contrast, three HLA-A3+ patients with rapidly progressive metastatic melanoma had no detectable T-cell response in tumor-involved nodes or peripheral blood lymphocytes, even after peptide stimulation ex vivo. The presented data are consistent with a systemic polyvalent immune response against tumor in this long-term survivor. These data provide an estimate of the level of CTL response that may be associated with protection from tumor recurrence.


Subject(s)
Antigens, Neoplasm , Melanoma/immunology , CD11 Antigens/biosynthesis , CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Chromatography, High Pressure Liquid , Disease-Free Survival , HLA-A3 Antigen/metabolism , Humans , Immunohistochemistry , Immunologic Memory , Immunophenotyping , Interferon-gamma/biosynthesis , L-Selectin/biosynthesis , Leukocyte Common Antigens/biosynthesis , Lymphatic Metastasis , Lymphocytes/metabolism , Male , Melanoma/metabolism , Middle Aged , Peptides/chemistry , Phenotype , Time Factors
8.
Int J Cancer ; 83(3): 326-34, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10495424

ABSTRACT

Peptide epitopes for tumor-reactive cytotoxic T-lymphocytes (CTL) have been identified on human cancers and are being used in tumor vaccine trials. However, the pharmacokinetics and pharmacodynamics of such peptides have been inadequately studied. It is predicted that immunogenic tumor peptides would have short half-lives in vivo. The goal of the present work was to evaluate the stability of the immunogenic peptide MART-1(27-35) in fresh normal human plasma (NHP) and to identify modifications that convey protection against enzymatic destruction without loss of immunogenicity. We evaluated the stability of the MART-1(27-35) peptide (AAGIGILTV) and modified forms of that peptide for stability and immune recognition in an in vitro model. The peptides were incubated in plasma for varied time intervals and evaluated for their ability to reconstitute the epitope for MART-1(27-35)-reactive CTL. Loss of CTL reactivity signaled loss of immunoreactive peptide. When 1 microM MART-1(27-35) peptide was incubated in plasma prior to pulsing on target cells, CTL reactivity was lost within 3 hr, and the calculated half-life of this peptide was 22 sec. This degradation was mediated by peptidases. The stability of MART-1(27-35) was markedly prolonged by C-terminal amidation and/or N-terminal acetylation (peptide capping), or by polyethylene-glycol modification (PEGylation) of the C-terminus. These modified peptides were recognized by CTL. The MART-1(27-35) peptide is very unstable in plasma. It is probable that it and other immunogenic peptides will be similarly unstable in vivo. Immunogenicity of these peptides might be enhanced by creating modifications that enhance stability.


Subject(s)
Cancer Vaccines/immunology , Endopeptidases/physiology , Melanoma/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Antigens, Neoplasm , Cell Line , Epitopes, T-Lymphocyte , Humans , MART-1 Antigen , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology
9.
J Immunol ; 158(8): 3635-9, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9103425

ABSTRACT

We previously showed that bacterial DNA contains immunostimulatory motifs consisting of unmethylated CpG dinucleotides flanked by two 5' purines and two 3' pyrimidines. These motifs rapidly trigger an innate immune response, characterized by the production of IL-6, IL-12, and IFN-gamma. Since DNA vaccines are constructed from plasmids of bacterial DNA, we examined whether CpG motifs present in these plasmids contributed to the immunogenicity of DNA vaccines. In vitro experiments showed that DNA plasmids induced production of the same cytokines stimulated by bacterial DNA, an effect eliminated by DNase treatment. In vivo experiments showed that the immunogenicity of a DNA vaccine was significantly reduced by methylating its CpG motifs and was significantly increased by coadministering exogenous CpG-containing DNA. These findings support the conclusion that CpG motifs in the plasmid backbone of DNA vaccines play an important role in the induction of Ag-specific immunity.


Subject(s)
CpG Islands/immunology , DNA, Bacterial/immunology , Vaccines, DNA/immunology , Plasmids/immunology
10.
Springer Semin Immunopathol ; 19(2): 245-56, 1997.
Article in English | MEDLINE | ID: mdl-9406350

ABSTRACT

DNA technology has been harnessed to produce a variety of plasmid-based vaccines designed to prevent viral, bacterial and parasitic infections. The rapid adoption and implementation of this novel vaccine strategy carries with it important safety and efficacy concerns. This review will focus on whether DNA vaccines (1) are likely to induce systemic or organ-specific autoimmune disease, (2) have the potential to induce tolerance rather than immunity, and (3) are as effective in individuals with depressed immune function as they are in healthy adults.


Subject(s)
Vaccines, DNA/standards , Aging/immunology , Animals , Animals, Newborn , Antibodies, Antinuclear/biosynthesis , Autoimmune Diseases/etiology , B-Lymphocytes/immunology , Cytokines/metabolism , Disease Susceptibility , Evaluation Studies as Topic , Humans , Immunity, Cellular , Immunoglobulin G/biosynthesis , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Safety , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
11.
J Clin Invest ; 98(12): 2700-5, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8981914

ABSTRACT

Plasmid DNA vaccines capable of preventing viral, bacterial, and parasitic infections are currently under development. Our labs have shown that a plasmid DNA vaccine encoding the circumsporozoite protein of the malaria parasite elicits protective immunity against live sporozoite challenge in adult BALB/c mice. We now find that the same DNA vaccine induces tolerance rather than immunity when administered to 2-5 d-old mice. Neonatally tolerized animals were unable to mount antibody, cytokine or cytotoxic responses when rechallenged with DNA vaccine in vitro or in vivo. Tolerance was specific for immunogenic epitopes expressed by the vaccine-encoded, endogenously produced antigen. Mice challenged with exogenous circumsporozoite protein produced antibodies against a different set of epitopes, and were not tolerized. These findings demonstrate important differences in the nature and specificity of the immune response elicited by DNA vaccines versus conventional protein immunogens.


Subject(s)
DNA/immunology , Immune Tolerance/immunology , Malaria Vaccines/immunology , Plasmodium yoelii/immunology , Age Factors , Amino Acid Sequence , Animals , Cell Count , Cells, Cultured , Cloning, Molecular , Epitopes/chemistry , Epitopes/immunology , Female , Immunization , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/immunology , Plasmids/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
12.
J Virol ; 70(4): 2669-73, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8642705

ABSTRACT

Previous studies have shown that the glycoprotein cytoplasmic domains of human immunodeficiency virus type 2 (HIV-2) or simian immunodeficiency virus of macaques modulate biological activities of the viral glycoprotein complex, including syncytium formation, exterior glycoprotein conformation, and glycoprotein incorporation into budding virus particles. We have now utilized a recombinant expression system to study interactions of full-length or truncated HIV-2 glycoproteins with coexpressed HIV-2 Gag proteins which self-assemble and bud as virus-like particles. Interestingly, budding of HIV-2 virus-like particles from cells was enhanced 5- to 24-fold when Gag was coexpressed with the full-length HIV-2 glycoprotein, compared with Gag expressed either alone or with a truncated HIV-2 glycoprotein. The results obtained in this model system indicate that an additional effect of the lengthy cytoplasmic domain of the glycoprotein of HIV-2 is enhancement of particle budding. We speculate that the cytoplasmic domain of the viral glycoprotein of HIV-2 enhances budding by (i) potentiation of Gag structure or function or (ii) membrane modulation.


Subject(s)
Glycoproteins/physiology , HIV-2/physiology , Viral Proteins/physiology , Virus Replication , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers , Gene Products, gag/genetics , Gene Products, gag/physiology , Glycoproteins/genetics , HIV-2/genetics , Humans , Molecular Sequence Data , Viral Proteins/genetics
13.
Virology ; 214(1): 50-8, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-8525638

ABSTRACT

The requirements for SIV particle assembly and envelope incorporation were investigated using a baculovirus expression system. The Pr56gag precursor protein expressed under control of the polyhedrin promoter (pPolh) produced high levels of immature retrovirus-like particles (VLP) upon expression in Sf9 insect cells. To determine the optimal conditions for envelope protein (Env) incorporation into VLP, two recombinant baculoviruses expressing the SIV envelope protein under control of a very late pPolh or a hybrid late/very late capsid/polyhedrin (Pcap/polh) promoter and a recombinant expressing a truncated form of the SIV envelope protein (Envt) under the hybrid Pcap/polh promoter were compared. We have observed that utilization of the earlier hybrid promoter resulted in higher levels of Env expression on the cell surface and its incorporation into budding virus particles. We have also found that the Envt protein is transported to the cell surface of insect cells and incorporated into VLP more efficiently than full-length Env. In addition, we examined the effect of coexpression of the protease furin, which has been implicated in the proteolytic cleavage of the Env precursor gp160 in mammalian cells. Coexpression of furin in insect cells resulted in more efficient proteolytic cleavage into gp120 and gp41, and the cleaved proteins were incorporated into VLP.


Subject(s)
Membrane Glycoproteins , Simian Immunodeficiency Virus/physiology , Viral Envelope Proteins/metabolism , Virion/physiology , Virus Assembly , Animals , Base Sequence , Cell Line , DNA Primers , Furin , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genetic Vectors , HIV Envelope Protein gp120/metabolism , Molecular Sequence Data , Nucleopolyhedroviruses/genetics , Protein Precursors/metabolism , Simian Immunodeficiency Virus/ultrastructure , Spodoptera , Subtilisins/metabolism , Viral Envelope Proteins/genetics
14.
Science ; 260(5112): 1323-7, 1993 May 28.
Article in English | MEDLINE | ID: mdl-8493576

ABSTRACT

Although protection in animal models against intravenous challenges with simian immunodeficiency virus (SIV) has been reported, no previous vaccines have protected against a heterosexual route of infection. In this study, five of six macaques were protected against vaginal challenge when immunized with formalin-treated SIV in biodegradable microspheres by the intramuscular plus oral or plus intratracheal route. Oral immunization alone did not protect. After a second vaginal challenge, three of four intramuscularly primed and mucosally boosted macaques remained protected. The data suggest that protection against human immunodeficiency virus vaginal transmission could be provided by microsphere-based booster vaccines when used to immunize women who are systemically primed.


Subject(s)
Antibodies, Viral/analysis , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vagina/immunology , Viral Vaccines , Administration, Oral , Animals , Antibodies, Viral/biosynthesis , Female , Immunization, Secondary , Injections, Intramuscular , Macaca mulatta , Mice , Microspheres , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Trachea , Vaccination , Vagina/microbiology , Viral Vaccines/administration & dosage
15.
J Virol ; 66(6): 3971-5, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1583738

ABSTRACT

To investigate the glycoprotein determinants of viral cytopathology, we constructed chimeric env genes between a noncytopathic strain of human immunodeficiency virus type 2 (HIV-2), designated HIV-2/ST, and a highly fusogenic and cytopathic variant derived from this virus. Expression of the resulting chimeric glycoproteins indicated that efficient syncytium formation in the human T-cell line Sup T1 mapped to the C-terminal region of the transmembrane (TM) glycoprotein subunit. In this region, the wild-type and cytopathic ST glycoproteins differed by only four amino acids and by the presence of a premature termination codon in the cytopathic variant. Subsequent site-directed mutagenesis indicated that the cytoplasmic domain truncation was responsible for the enhanced fusion activity. This modification, however, increased the fusion activity of the glycoprotein only in Sup T1 cells (in which the ST variant arose) but not in Molt 4 clone 8 or peripheral blood mononuclear cells. These observations indicate that the length of the cytoplasmic domain of the HIV-2 glycoprotein modulates the fusion activity of the exterior glycoprotein complex in a cell-specific manner. Such adaptability appears to permit the emergence of fusogenic variants during HIV-2 passage in vitro and may also regulate viral growth or cytopathic effects in selected cell types during natural infection in vivo.


Subject(s)
Cell Fusion/drug effects , Gene Products, env/pharmacology , HIV Infections/pathology , HIV-2/pathogenicity , T-Lymphocytes/pathology , Amino Acid Sequence , Base Sequence , Cell Line , DNA Mutational Analysis , Gene Products, env/biosynthesis , Gene Products, env/genetics , Genetic Vectors/genetics , Giant Cells/microbiology , Giant Cells/pathology , Humans , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship , T-Lymphocytes/microbiology , Vaccinia virus/genetics
16.
Virology ; 187(1): 233-41, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1736526

ABSTRACT

Utilizing a recombinant vaccinia expression system, we investigated the biological properties and CD4 receptor interactions of the envelope glycoproteins of a noncytopathic human immunodeficiency virus type 2 strain, termed HIV-2/ST, and a highly cytopathic variant derived from it. The efficiency and host cell range of syncytium formation by the recombinant glycoproteins of both viruses were highly restricted compared to those of prototypic strains of HIV (HIV-2/ROD or HIV-1/IIIB). However, the glycoprotein of cytopathic but not wild-type ST generated numerous large syncytia in the human T-cell line Sup T1 from which it was derived. A single cell line (Molt 4 clone 8) was permissive to fusion by both wild-type and cytopathic ST envelopes, but only the glycoprotein of cytopathic ST could be inhibited with a soluble form of the viral receptor CD4 (sCD4). While these results indicated major differences in the envelope glycoprotein-CD4 receptor interactions of wild-type versus cytopathic ST, direct and competition binding assays utilizing soluble external glycoprotein (SU) and sCD4 surprisingly revealed equivalent low binding affinity for both viruses. From these experiments we conclude that relevant biological properties (e.g., CD4 binding, cytopathic potential, and sCD4 neutralization) of HIV viruses which differ in their pathogenic potential are reflected in the sCD4 interactions of the assembled native envelope complex (as on cell or virion surfaces) but not the soluble SU glycoprotein.


Subject(s)
CD4 Antigens/metabolism , HIV Envelope Protein gp120/metabolism , HIV-2/metabolism , Cell Fusion/physiology , Cell Line , Cells, Cultured , Cytopathogenic Effect, Viral , Giant Cells , Immunoblotting , Kinetics , Macromolecular Substances , Radioligand Assay , Recombinant Proteins/metabolism , Solubility , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...