Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 18(4): e1010467, 2022 04.
Article in English | MEDLINE | ID: mdl-35452496

ABSTRACT

A key challenge for the development of a cure to HIV-1 infection is the persistent viral reservoir established during early infection. Previous studies using Toll-like receptor 7 (TLR7) agonists and broadly neutralizing antibodies (bNAbs) have shown delay or prevention of viral rebound following antiretroviral therapy (ART) discontinuation in simian-human immunodeficiency virus (SHIV)-infected rhesus macaques. In these prior studies, ART was initiated early during acute infection, which limited the size and diversity of the viral reservoir. Here we evaluated in SHIV-infected rhesus macaques that did not initiate ART until 1 year into chronic infection whether the TLR7 agonist vesatolimod in combination with the bNAb PGT121, formatted either as a human IgG1, an effector enhanced IgG1, or an anti-CD3 bispecific antibody, would delay or prevent viral rebound following ART discontinuation. We found that all 3 antibody formats in combination with vesatolimod were able to prevent viral rebound following ART discontinuation in a subset of animals. These data indicate that a TLR7 agonist combined with antibodies may be a promising strategy to achieve long-term ART-free HIV remission in humans.


Subject(s)
HIV Infections , HIV-1 , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Broadly Neutralizing Antibodies , HIV Antibodies/therapeutic use , Immunoglobulin G , Macaca mulatta , Toll-Like Receptor 7/agonists , Viral Load
3.
Nature ; 503(7475): 224-8, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24172905

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian-human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/physiology , Animals , DNA, Viral/blood , HIV Antibodies/immunology , Macaca mulatta , T-Lymphocytes/immunology , Viremia/therapy
4.
J Infect Dis ; 218(9): 1447-1452, 2018 09 22.
Article in English | MEDLINE | ID: mdl-29878133

ABSTRACT

A 48-year-old woman was infected with a vpr-defective human immunodeficiency virus (HIV)-1 molecular clone. Seroconversion was markedly delayed, and without treatment she had durably suppressed viremia and normal T-cell levels. Neutralizing antibody and CD8+ T-cell immune responses against HIV-1 were unremarkable. Viral sequences confirmed the source but evolved defective nef, suggesting an unknown mechanistic link to vpr. There were subtle qualitative defects in T and B cells. To our knowledge, this is the only case of human infection with a characterized defective HIV-1 molecular clone, which furthermore recapitulated live-attenuated vaccination in macaque models of HIV-1 vaccine research.


Subject(s)
AIDS Vaccines/immunology , Gene Products, vpr/immunology , HIV Infections/immunology , HIV-1/immunology , Vaccines, Attenuated/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cloning, Molecular , Female , Humans , Middle Aged , Vaccination/methods
5.
J Infect Dis ; 218(4): 572-580, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29617879

ABSTRACT

Background: Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children. To date, no vaccine is approved for the broad population of healthy infants. MEDI8897, a potent anti-RSV fusion antibody with extended serum half-life, is currently under clinical investigation as a potential passive RSV vaccine for all infants. As a ribonucleic acid virus, RSV is prone to mutation, and the possibility of viral escape from MEDI8897 neutralization is a potential concern. Methods: We generated RSV monoclonal antibody (mAb)-resistant mutants (MARMs) in vitro and studied the effect of the amino acid substitutions identified on binding and viral neutralization susceptibility to MEDI8897. The impact of resistance-associated mutations on in vitro growth kinetics and the prevalence of these mutations in currently circulating strains of RSV in the United States was assessed. Results: Critical residues identified in MARMs for MEDI8897 neutralization were located in the MEDI8897 binding site defined by crystallographic analysis. Substitutions in these residues affected the binding of mAb to virus, without significant impact on viral replication in vitro. The frequency of natural resistance-associated polymorphisms was low. Conclusions: Results from this study provide insights into the mechanism of MEDI8897 escape and the complexity of monitoring for emergence of resistance.


Subject(s)
Amino Acid Substitution , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunologic Factors/pharmacology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Binding Sites , Biological Products/pharmacology , Crystallography, X-Ray , Drug Resistance, Viral , Gene Frequency , Humans , Immune Evasion , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/immunology , Neutralization Tests , Prevalence , Protein Conformation , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , United States/epidemiology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Virus Attachment/drug effects , Virus Replication/drug effects
6.
J Virol ; 90(13): 6127-6139, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27122574

ABSTRACT

UNLABELLED: Although antibodies to the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein have been studied extensively for their ability to block viral infectivity, little data are currently available on nonneutralizing functions of these antibodies, such as their ability to eliminate virus-infected cells by antibody-dependent cell-mediated cytotoxicity (ADCC). HIV-1 Env-specific antibodies of diverse specificities, including potent broadly neutralizing and nonneutralizing antibodies, were therefore tested for ADCC against cells infected with a lab-adapted HIV-1 isolate (HIV-1NL4-3), a primary HIV-1 isolate (HIV-1JR-FL), and a simian-human immunodeficiency virus (SHIV) adapted for pathogenic infection of rhesus macaques (SHIVAD8-EO). In accordance with the sensitivity of these viruses to neutralization, HIV-1NL4-3-infected cells were considerably more sensitive to ADCC, both in terms of the number of antibodies and magnitude of responses, than cells infected with HIV-1JR-FL or SHIVAD8-EO ADCC activity generally correlated with antibody binding to Env on the surfaces of virus-infected cells and with viral neutralization; however, neutralization was not always predictive of ADCC, as instances of ADCC in the absence of detectable neutralization, and vice versa, were observed. These results reveal incomplete overlap in the specificities of antibodies that mediate these antiviral activities and provide insights into the relationship between ADCC and neutralization important for the development of antibody-based vaccines and therapies for combating HIV-1 infection. IMPORTANCE: This study provides fundamental insights into the relationship between antibody-dependent cell-mediated cytotoxicity (ADCC) and virus neutralization that may help to guide the development of antibody-based vaccines and immunotherapies for the prevention and treatment of HIV-1 infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibody-Dependent Cell Cytotoxicity , HIV Antibodies/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibody Specificity , Binding Sites, Antibody , HEK293 Cells , HIV Infections/prevention & control , HIV Infections/therapy , HIV Infections/virology , HIV-1/isolation & purification , Humans , Macaca mulatta , Neutralization Tests , Simian Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism
7.
Proc Natl Acad Sci U S A ; 111(17): 6425-30, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733916

ABSTRACT

Tetherin is an IFN-inducible transmembrane protein that inhibits the detachment of enveloped viruses from infected cells. HIV-1 overcomes this restriction factor by expressing HIV-1 viral protein U (Vpu), which down-regulates and degrades tetherin. We report that mutations in Vpu that impair tetherin antagonism increase the susceptibility of HIV-infected cells to antibody-dependent cell-mediated cytotoxicity (ADCC), and conversely that RNAi knockdown of tetherin, but not other cellular proteins down-modulated by Vpu, decreases the susceptibility of HIV-infected cells to ADCC. These results reveal that Vpu protects HIV-infected cells from ADCC as a function of its ability to counteract tetherin. By serving as link between innate and adaptive immunity, the antiviral activity of tetherin may be augmented by virus-specific antibodies, and hence much greater than previously appreciated.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Cytoprotection , HIV Infections/immunology , HIV Infections/pathology , Human Immunodeficiency Virus Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Substitution , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/metabolism , CD4 Antigens/metabolism , Cytoprotection/drug effects , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Gene Deletion , Humans , Interferon-alpha/pharmacology , RNA Interference/drug effects , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
8.
J Infect Dis ; 214(4): 612-6, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27357340

ABSTRACT

Humanized mice reconstituted with a human immune system can be mucosally infected with human immunodeficiency virus (HIV), opening up the possibility of studying HIV transmission in a small-animal model. Here we report that passive immunization with the broadly neutralizing antibody b12 protected humanized mice against repetitive intravaginal infection in a dose-dependent manner. In addition, treatment with the antibody PGT126, which is more potent in vitro, was more efficacious in vivo and provided sterilizing protection. Our results demonstrate that humanized mice can be used as a small-animal model to study the efficacy and mechanism of broadly neutralizing antibody protection against HIV acquisition.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Disease Models, Animal , HIV Antibodies/administration & dosage , HIV Infections/prevention & control , Immunization, Passive/methods , Animals , Dose-Response Relationship, Immunologic , Female , Mice , Mice, SCID , Treatment Outcome
9.
Nucleic Acids Res ; 42(4): e28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24270790

ABSTRACT

DNA transposon-based vectors have emerged as gene vehicles with a wide biomedical and therapeutic potential. So far, genomic insertion of such vectors has relied on the co-delivery of genetic material encoding the gene-inserting transposase protein, raising concerns related to persistent expression, insertional mutagenesis and cytotoxicity. This report describes potent DNA transposition achieved by direct delivery of transposase protein. By adapting integrase-deficient lentiviral particles (LPs) as carriers of the hyperactive piggyBac transposase protein (hyPBase), we demonstrate rates of DNA transposition that are comparable with the efficiency of a conventional plasmid-based strategy. Embedded in the Gag polypeptide, hyPBase is robustly incorporated into LPs and liberated from the viral proteins by the viral protease during particle maturation. We demonstrate lentiviral co-delivery of the transposase protein and vector RNA carrying the transposon sequence, allowing robust DNA transposition in a variety of cell types. Importantly, this novel delivery method facilitates a balanced cellular uptake of hyPBase, as shown by confocal microscopy, and allows high-efficiency production of clones harboring a single transposon insertion. Our findings establish engineered LPs as a new tool for transposase delivery. We believe that protein transduction methods will increase applicability and safety of DNA transposon-based vector technologies.


Subject(s)
Gene Products, gag/genetics , Genetic Vectors , Lentivirus/genetics , Transposases/metabolism , Animals , Cell Line , Cells, Cultured , Humans , Protein Precursors/genetics , Proteins/genetics , Proteins/metabolism , Transposases/genetics , Virion/genetics , Virion/metabolism
10.
J Virol ; 88(11): 6031-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24623433

ABSTRACT

UNLABELLED: The type I interferon-inducible factor tetherin retains virus particles on the surfaces of cells infected with vpu-deficient human immunodeficiency virus type 1 (HIV-1). While this mechanism inhibits cell-free viral spread, the immunological implications of tethered virus have not been investigated. We found that surface tetherin expression increased the antibody opsonization of vpu-deficient HIV-infected cells. The absence of Vpu also stimulated NK cell-activating FcγRIIIa signaling and enhanced NK cell degranulation and NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC). The deletion of vpu in HIV-1-infected primary CD4(+) T cells enhanced the levels of antibody binding and Fc receptor signaling mediated by HIV-positive-patient-derived antibodies. The magnitudes of antibody binding and Fc signaling were both highly correlated to the levels of tetherin on the surfaces of infected primary CD4 T cells. The affinity of antibody binding to FcγRIIIa was also found to be critical in mediating efficient Fc activation. These studies implicate Vpu antagonism of tetherin as an ADCC evasion mechanism that prevents antibody-mediated clearance of virally infected cells. IMPORTANCE: The ability of the HIV-1 accessory factor to antagonize tetherin has been considered to primarily function by limiting the spread of virus by preventing the release of cell-free virus. This study supports the hypothesis that a major function of Vpu is to decrease the recognition of infected cells by anti-HIV antibodies at the cell surface, thereby reducing recognition by antibody-dependent clearance by natural killer cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD/immunology , Human Immunodeficiency Virus Proteins/immunology , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Signal Transduction/immunology , Viral Regulatory and Accessory Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , Humans , Jurkat Cells , Lysosomal-Associated Membrane Protein 1/immunology , Opsonin Proteins/immunology , Receptors, IgG/metabolism
11.
PLoS Pathog ; 9(11): e1003776, 2013.
Article in English | MEDLINE | ID: mdl-24278022

ABSTRACT

The mechanisms by which human immunodeficiency virus type 1 (HIV-1) crosses mucosal surfaces to establish infection are unknown. Acidic genital secretions of HIV-1-infected women contain HIV-1 likely coated by antibody. We found that the combination of acidic pH and Env-specific IgG, including that from cervicovaginal and seminal fluids of HIV-1-infected individuals, augmented transcytosis across epithelial cells as much as 20-fold compared with Env-specific IgG at neutral pH or non-specific IgG at either pH. Enhanced transcytosis was observed with clinical HIV-1 isolates, including transmitted/founder strains, and was eliminated in Fc neonatal receptor (FcRn)-knockdown epithelial cells. Non-neutralizing antibodies allowed similar or less transcytosis than neutralizing antibodies. However, the ratio of total:infectious virus was higher for neutralizing antibodies, indicating that they allowed transcytosis while blocking infectivity of transcytosed virus. Immunocytochemistry revealed abundant FcRn expression in columnar epithelia lining the human endocervix and penile urethra. Acidity and Env-specific IgG enhance transcytosis of virus across epithelial cells via FcRn and could facilitate translocation of virus to susceptible target cells following sexual exposure.


Subject(s)
Epithelial Cells/immunology , HIV Antibodies/immunology , HIV-1/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Transcytosis/immunology , Cell Line, Tumor , Cervix Uteri/immunology , Cervix Uteri/pathology , Cervix Uteri/virology , Epithelial Cells/pathology , Female , HIV-1/pathogenicity , Humans , Hydrogen-Ion Concentration , Male , Semen/immunology , Urethra/immunology , Urethra/pathology , Urethra/virology
12.
Methods ; 65(1): 127-32, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23811333

ABSTRACT

The mucosal epithelia together with adaptive immune responses, such as local production and secretion of dimeric and polymeric immunoglobulin A (IgA), are a crucial part of the first line of defense against invading pathogens. IgA is primarily secreted as SIgA and plays multiple roles in mucosal defense. The study of SIgA-mediated protection is an important area of research in mucosal immunity but an easy, fast and reproducible method to generate pathogen-specific SIgA in vitro has not been available. We report here a new method to produce SIgA by co-purification of dimeric IgA, containing J chain, and recombinant human SC expressed in CHO cells. We previously reported the generation, production and characterization of the human recombinant monoclonal antibody IgA2 b12. This antibody, derived from the variable regions of the neutralizing anti-HIV-1 mAb IgG1 b12, blocked viral attachment and uptake by epithelial cells in vitro. We used a cloned CHO cell line that expresses monomeric, dimeric and polymeric species of IgA2 b12 for large-scale production of dIgA2 b12. Subsequently, we generated a CHO cell line to express recombinant human secretory component (rhSC). Here, we combined dIgA2 b12 and CHO-expressed rhSC via column chromatography to produce SIgA2 b12 that remains fully intact upon elution with 0.1M citric acid, pH 3.0. We have performed biochemical analysis of the synthesized SIgA to confirm the species is of the expected size and retains the functional properties previously described for IgA2 b12. We show that SIgA2 b12 binds to the HIV-1 gp120 glycoprotein with similar apparent affinity to that of monomeric and dimeric forms of IgA2 b12 and neutralizes HIV-1 isolates with similar potency. An average yield of 6 mg of SIgA2 b12 was achieved from the combination of 20mg of purified dIgA2 b12 and 2L of rhSC-containing CHO cell supernatant. We conclude that synthesized production of stable SIgA can be generated by co-purification. This process introduces a simplified means of generating a variety of pathogen-specific SIgA antibodies for research and clinical applications.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Immunoglobulin A, Secretory/biosynthesis , Animals , Antibodies, Neutralizing/isolation & purification , CHO Cells , Chromatography, Affinity , Cricetinae , Cricetulus , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Humans , Immunoglobulin A, Secretory/isolation & purification , Protein Binding , Protein Engineering , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification
13.
Proc Natl Acad Sci U S A ; 109(46): 18921-5, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23100539

ABSTRACT

Most animal studies using passive administration of HIV broadly neutralizing monoclonal antibodies (bnMAbs) have associated protection against high-dose mucosal viral challenge with relatively high serum concentrations of antibody. We recently identified several bnMAbs remarkable for their in vitro potency against HIV. Of these bnMAbs, PGT121 is one of the most broad and potent antibodies isolated to date and shows 10- to 100-fold higher neutralizing activity than previously characterized bnMAbs. To evaluate the protective potency of PGT121 in vivo, we performed a protection study in rhesus macaques. Animals were i.v. administered 5 mg/kg, 1 mg/kg, or 0.2 mg/kg PGT121 24 h before being vaginally challenged with a single high dose of chimeric simian-human immunodeficiency virus (SHIV)(SF162P3). Sterilizing immunity was achieved in all animals administered 5 mg/kg and 1 mg/kg and three of five animals administered 0.2 mg/kg PGT121, with corresponding average antibody serum concentrations of 95 µg/mL, 15 µg/mL, and 1.8 µg/mL, respectively. The results suggest that a protective serum concentration for PGT121 is in the single-digit µg/mL for SHIV(SF162P3), showing that PGT121 can mediate sterilizing immunity at serum concentrations that are significantly lower than those observed in previous studies and that may be achievable through vaccination with the development of a suitable immunogen.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , HIV Antibodies/pharmacology , HIV Infections/prevention & control , HIV-1/immunology , Immunization, Passive , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Female , HIV Antibodies/immunology , HIV Infections/immunology , Humans , Macaca mulatta
14.
Proc Natl Acad Sci U S A ; 108(27): 11181-6, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21690411

ABSTRACT

To guide vaccine design, we assessed whether human monoclonal antibodies (MAbs) b12 and b6 against the CD4 binding site (CD4bs) on HIV-1 gp120 and F240 against an immundominant epitope on gp41 could prevent vaginal transmission of simian HIV (SHIV)-162P4 to macaques. The two anti-gp120 MAbs have similar monomeric gp120-binding properties, measured in vitro, but b12 is strongly neutralizing and b6 is not. F240 is nonneutralizing. Applied vaginally at a high dose, the strongly neutralizing MAb b12 provided sterilizing immunity in seven of seven animals, b6 in zero of five animals, and F240 in two of five animals. Compared with control animals, the protection by b12 achieved statistical significance, whereas that caused by F240 did not. For two of three unprotected F240-treated animals there was a trend toward lowered viremia. The potential protective effect of F240 may relate to the relatively strong ability of this antibody to capture infectious virions. Additional passive transfer experiments also indicated that the ability of the administered anti-gp120 MAbs to neutralize the challenge virus was a critical influence on protection. Furthermore, when data from all of the experiments were combined, there was a significant increase in the number of founder viruses establishing infection in animals receiving MAb b6, compared with other nonprotected macaques. Thus, a gp120-binding, weakly neutralizing MAb to the CD4bs was, at best, completely ineffective at protection. A nonneutralizing antibody to gp41 may have a limited capacity to protect, but the results suggest that the central focus of HIV-1 vaccine research should be on the induction of potently neutralizing antibodies.


Subject(s)
Antibodies, Neutralizing/administration & dosage , HIV Antibodies/administration & dosage , HIV-1/immunology , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Female , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/genetics , Humans , Immunization, Passive , Macaca mulatta , Membrane Glycoproteins/immunology , Molecular Sequence Data , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics , Vagina/immunology , Vagina/virology , Viral Envelope Proteins/immunology
15.
AIDS ; 38(4): 607-610, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38416554

ABSTRACT

We studied the relationship between viral diversity and susceptibility to broadly neutralizing antibodies (bNAbs) in longitudinal plasma and peripheral blood mononuclear cells from 89 people with HIV who initiated antiretroviral therapy (ART) during acute and early HIV-1 infection (AEHI). HIV-1 diversity and predicted bNAb susceptibility were comparable across AEHI. Diversity evolution was not observed during ART, suggesting (pro)viruses at initiation or during treatment may identify individuals with susceptible virus for bNAb interventional trials.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , HIV Infections/drug therapy , Broadly Neutralizing Antibodies , Leukocytes, Mononuclear
16.
J Virol ; 86(11): 6189-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22457527

ABSTRACT

Eliciting neutralizing antibodies is thought to be a key activity of a vaccine against human immunodeficiency virus (HIV). However, a number of studies have suggested that in addition to neutralization, interaction of IgG with Fc gamma receptors (FcγR) may play an important role in antibody-mediated protection. We have previously obtained evidence that the protective activity of the broadly neutralizing human IgG1 anti-HIV monoclonal antibody (MAb) b12 in macaques is diminished in the absence of FcγR binding capacity. To investigate antibody-dependent cellular cytotoxicity (ADCC) as a contributor to FcγR-associated protection, we developed a nonfucosylated variant of b12 (NFb12). We showed that, compared to fully fucosylated (referred to as wild-type in the text) b12, NFb12 had higher affinity for human and rhesus macaque FcγRIIIa and was more efficient in inhibiting viral replication and more effective in killing HIV-infected cells in an ADCC assay. Despite these more potent in vitro antiviral activities, NFb12 did not enhance protection in vivo against repeated low-dose vaginal challenge in the simian-human immunodeficiency virus (SHIV)/macaque model compared to wild-type b12. No difference in protection, viral load, or infection susceptibility was observed between animals given NFb12 and those given fully fucosylated b12, indicating that FcγR-mediated activities distinct from FcγRIIIa-mediated ADCC may be important in the observed protection against SHIV challenge.


Subject(s)
Antibodies, Monoclonal/administration & dosage , HIV Antibodies/administration & dosage , HIV-1/immunology , Receptors, IgG/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Cells, Cultured , Female , HIV Antibodies/immunology , HIV Antibodies/isolation & purification , HIV Antibodies/metabolism , Humans , Macaca mulatta , Receptors, IgG/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Viral Load
17.
J Virol ; 85(20): 10572-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849450

ABSTRACT

Passive transfer of neutralizing antibodies is effective in protecting rhesus macaques against simian/human immunodeficiency virus (SHIV) challenge. In addition to neutralization, effector functions of the crystallizable fragment (Fc) of antibodies are involved in antibody-mediated protection against a number of viruses. We recently showed that interaction between the Fc fragment of the broadly neutralizing antibody IgG1 b12 and cellular Fcγ receptors (FcγRs) plays an important role in protection against SHIV infection in rhesus macaques. The specific nature of this Fc-dependent protection is largely unknown. To investigate, we generated a panel of 11 IgG1 b12 antibody variants with selectively diminished or enhanced affinity for the two main activating FcγRs, FcγRIIa and FcγRIIIa. All 11 antibody variants bind gp120 and neutralize virus as effectively as does wild-type b12. Binding studies using monomeric (enzyme-linked immunosorbent assay [ELISA] and surface plasmon resonance [SPR]) and cellularly expressed Fcγ receptors show decreased (up to 5-fold) and increased (up to 90-fold) binding to FcγRIIa and FcγRIIIa with this newly generated panel of antibodies. In addition, there was generally a good correlation between b12 variant affinity for Fcγ receptor and variant function in antibody-dependent cell-mediated virus inhibition (ADCVI), phagocytosis, NK cell activation assays, and antibody-dependent cellular cytotoxicity (ADCC) assays. In future studies, these b12 variants will enable the investigation of the protective role of individual FcγRs in HIV infection.


Subject(s)
HIV Antibodies/immunology , HIV Antibodies/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Receptors, Fc/immunology , Receptors, Fc/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Humans , Killer Cells, Natural/immunology , Macaca mulatta , Phagocytosis , Protein Binding , Recombination, Genetic , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Surface Plasmon Resonance
18.
Mol Ther ; 19(8): 1499-510, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21468003

ABSTRACT

It has been previously shown that integrase-defective HIV-1-based gene vectors can serve, with moderate efficiency, as substrate for DNA transposition by a transiently expressed Sleeping Beauty (SB) transposase. Here, we describe the enhanced gene transfer properties of a HIV-1/SB hybrid vector that allows efficient DNA transposition, facilitated by the hyperactive SB100X transposase, from vector DNA intermediates in primary human cells. Potent transposase-dependent integration of genetic cargo carried by the hybrid HIV-1/SB vector (up to 160-fold above background) is reported in human cell lines as well as in primary human fibroblasts and keratinocytes. The efficiency of transgene integration in context of the newly developed hybrid vector is comparable with that of conventional lentiviral vectors (LVs). Integration profiles of integrating HIV-1-derived vectors and SB transposons mobilized from LVs are investigated by deep sequencing of a large number of integration sites. A significant bias of lentiviral integrations in genes is reported, confirming that biological properties of the viral integration machinery facilitate preferred insertion into actively transcribed genomic regions. In sharp contrast, lentiviral insertions catalyzed by the SB100X transposase are far more random with respect to genes. Based on these properties, HIV-1/SB vectors may become valuable tools for genetic engineering and therapeutic gene transfer.


Subject(s)
Genetic Vectors/genetics , HIV-1/genetics , Integrases/metabolism , Transposases/genetics , Transposases/metabolism , Virus Integration/genetics , Base Sequence , Cell Line , DNA/genetics , DNA/metabolism , DNA Transposable Elements , Fibroblasts , Gene Transfer Techniques , Genome , High-Throughput Nucleotide Sequencing , Humans , Integrases/deficiency , Integrases/genetics , Keratinocytes , Sequence Analysis, DNA , Transgenes
19.
AIDS ; 36(2): 205-214, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34586088

ABSTRACT

OBJECTIVE: Persistence of the viral reservoir is the main barrier to curing HIV. Initiation of ART during acute HIV infection can limit the size and diversity of the reservoir. In depth characterization of the reservoir in individuals who initiate ART during acute infection will be critical for clinical trial design and cure strategies. METHODS: Four cohorts with participants who initiated ART during acute infection or during chronic infection were enrolled in a cross-sectional, noninterventional study. Viral reservoir was evaluated by the Intact Proviral DNA Assay (IPDA), the Total HIV DNA Assay (THDA) and the Quantitative Viral Outgrowth Assay (QVOA). Viral diversity and susceptibility to V3-glycan bNAbs were determined by genotyping of the viral envelope gene. RESULTS: Participants who initiated ART during the acute Fiebig I-IV stages had lower level of total HIV DNA than participants who initiated ART during chronic infection whereas no difference was observed in intact HIV DNA or outgrowth virus. Participants who initiated ART during Fiebig I-IV also had lower viral diversity and appeared to have higher susceptibility to bNAbs than participants initiating ART during chronic infection. CONCLUSION: Individuals initiating ART during Fiebig I-IV had small viral reservoirs, low viral diversity, and high susceptibility to bNAbs, and would be an optimal target population for proof-of-concept HIV cure trials.


Subject(s)
HIV Infections , HIV-1 , Anti-Retroviral Agents/therapeutic use , Broadly Neutralizing Antibodies , Cross-Sectional Studies , HIV-1/genetics , Humans , Viral Load
20.
Transgenic Res ; 20(3): 533-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20803249

ABSTRACT

Modelling of human disease in genetically engineered pigs provides unique possibilities in biomedical research and in studies of disease intervention. Establishment of methodologies that allow efficient gene insertion by non-viral gene carriers is an important step towards development of new disease models. In this report, we present transgenic pigs created by Sleeping Beauty DNA transposition in primary porcine fibroblasts in combination with somatic cell nuclear transfer by handmade cloning. Göttingen minipigs expressing green fluorescent protein are produced by transgenesis with DNA transposon vectors carrying the transgene driven by the human ubiquitin C promoter. These animals carry multiple copies (from 8 to 13) of the transgene and show systemic transgene expression. Transgene-expressing pigs carry both transposase-catalyzed insertions and at least one copy of randomly inserted plasmid DNA. Our findings illustrate critical issues related to DNA transposon-directed transgenesis, including coincidental plasmid insertion and relatively low Sleeping Beauty transposition activity in porcine fibroblasts, but also provide a platform for future development of porcine disease models using the Sleeping Beauty gene insertion technology.


Subject(s)
Animals, Genetically Modified , DNA Transposable Elements/genetics , Gene Transfer Techniques , Mutagenesis, Insertional , Transposases/metabolism , Animals , Base Sequence , Fibroblasts/metabolism , Genetic Engineering/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Molecular Sequence Data , Swine , Swine, Miniature , Transgenes/genetics , Transgenes/physiology , Transposases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL