Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 975
Filter
1.
J Virol ; 96(17): e0119122, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000845

ABSTRACT

Epitopes with evidence of HLA-II-associated adaptation induce poorly immunogenic CD4+ T-cell responses in HIV-positive (HIV+) individuals. Many such escaped CD4+ T-cell epitopes are encoded by HIV-1 vaccines being evaluated in clinical trials. Here, we assessed whether this viral adaptation adversely impacts CD4+ T-cell responses following HIV-1 vaccination, thereby representing escaped epitopes. When evaluated in separate peptide pools, vaccine-encoded adapted epitopes (AE) induced CD4+ T-cell responses less frequently than nonadapted epitopes (NAE). We also demonstrated that in a polyvalent vaccine, where both forms of the same epitope were encoded, AE were less immunogenic. NAE-specific CD4+ T cells had increased, albeit low, levels of interferon gamma (IFN-γ) cytokine production. Single-cell transcriptomic analyses showed that NAE-specific CD4+ T cells expressed interferon-related genes, while AE-specific CD4+ T cells resembled a Th2 phenotype. Importantly, the magnitude of NAE-specific CD4+ T-cell responses, but not that of AE-specific responses, was found to positively correlate with Env-specific antibodies in a vaccine efficacy trial. Together, these findings show that HLA-II-associated viral adaptation reduces CD4+ T-cell responses in HIV-1 vaccine recipients and suggest that vaccines encoding a significant number of AE may not provide optimal B-cell help for HIV-specific antibody production. IMPORTANCE Despite decades of research, an effective HIV-1 vaccine remains elusive. Vaccine strategies leading to the generation of broadly neutralizing antibodies are likely needed to provide the best opportunity of generating a protective immune response against HIV-1. Numerous studies have demonstrated that T-cell help is necessary for effective antibody generation. However, immunogen sequences from recent HIV-1 vaccine efficacy trials include CD4+ T-cell epitopes that have evidence of immune escape. Our study shows that these epitopes, termed adapted epitopes, elicit lower frequencies of CD4+ T-cell responses in recipients from multiple HIV-1 vaccine trials. Additionally, the counterparts to these epitopes, termed nonadapted epitopes, elicited CD4+ T-cell responses that correlated with Env-specific antibodies in one efficacy trial. These results suggest that vaccine-encoded adapted epitopes dampen CD4+ T-cell responses, potentially impacting both HIV-specific antibody production and efficacious vaccine efforts.


Subject(s)
AIDS Vaccines , Antibody Formation , CD4-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , HIV Infections , HIV-1 , HLA-D Antigens , Immune Evasion , AIDS Vaccines/immunology , Broadly Neutralizing Antibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Clinical Trials as Topic , Epitopes, T-Lymphocyte/immunology , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , HLA-D Antigens/immunology , Humans
2.
Nat Med ; 27(12): 2234-2245, 2021 12.
Article in English | MEDLINE | ID: mdl-34887575

ABSTRACT

The development of a protective vaccine remains a top priority for the control of the HIV/AIDS pandemic. Here, we show that a messenger RNA (mRNA) vaccine co-expressing membrane-anchored HIV-1 envelope (Env) and simian immunodeficiency virus (SIV) Gag proteins to generate virus-like particles (VLPs) induces antibodies capable of broad neutralization and reduces the risk of infection in rhesus macaques. In mice, immunization with co-formulated env and gag mRNAs was superior to env mRNA alone in inducing neutralizing antibodies. Macaques were primed with a transmitted-founder clade-B env mRNA lacking the N276 glycan, followed by multiple booster immunizations with glycan-repaired autologous and subsequently bivalent heterologous envs (clades A and C). This regimen was highly immunogenic and elicited neutralizing antibodies against the most prevalent (tier-2) HIV-1 strains accompanied by robust anti-Env CD4+ T cell responses. Vaccinated animals had a 79% per-exposure risk reduction upon repeated low-dose mucosal challenges with heterologous tier-2 simian-human immunodeficiency virus (SHIV AD8). Thus, the multiclade env-gag VLP mRNA platform represents a promising approach for the development of an HIV-1 vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , Genes, env , Genes, gag , HIV Antibodies/biosynthesis , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology , Animals , HIV Antibodies/immunology , Immunization, Secondary , Macaca mulatta , Risk Factors , Simian Acquired Immunodeficiency Syndrome/immunology , Vaccines, Synthetic/administration & dosage , mRNA Vaccines/administration & dosage
3.
Front Immunol ; 12: 757811, 2021.
Article in English | MEDLINE | ID: mdl-34745131

ABSTRACT

Induction of broadly neutralizing antibodies (bNAbs) is a major goal for HIV vaccine development. HIV envelope glycoprotein (Env)-specific bNAbs isolated from HIV-infected individuals exhibit substantial somatic hypermutation and correlate with T follicular helper (Tfh) responses. Using the VC10014 DNA-protein co-immunization vaccine platform consisting of gp160 plasmids and gp140 trimeric proteins derived from an HIV-1 infected subject that developed bNAbs, we determined the characteristics of the Env-specific humoral response in vaccinated rhesus macaques in the context of CD4+ T cell depletion. Unexpectedly, both CD4+ depleted and non-depleted animals developed comparable Tier 1 and 2 heterologous HIV-1 neutralizing plasma antibody titers. There was no deficit in protection from SHIV challenge, no diminution of titers of HIV Env-specific cross-clade binding antibodies, antibody dependent cellular phagocytosis, or antibody-dependent complement deposition in the CD4+ depleted animals. These collective results suggest that in the presence of diminished CD4+ T cell help, HIV neutralizing antibodies were still generated, which may have implications for developing effective HIV vaccine strategies.


Subject(s)
AIDS Vaccines , Broadly Neutralizing Antibodies/biosynthesis , HIV Antibodies/biosynthesis , Macaca mulatta/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , Broadly Neutralizing Antibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Female , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp160/immunology , HIV-1/immunology , Immunization, Secondary , Male , Phagocytosis , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vaccine Development , Vaccines, Synthetic , Viral Load , env Gene Products, Human Immunodeficiency Virus/immunology
4.
Cell Rep Med ; 2(6): 100314, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34195680

ABSTRACT

Increasing evidence suggests infants develop unique neutralizing antibody (nAb) responses to HIV compared to adults. Here, we dissected the nAb response of an infant whose virus is in clinical trials as a vaccine immunogen, with a goal of characterizing the broad responses in the infant to this antigen. We isolated 73 nAbs from infant BG505 and identified a large number of clonal families. Twenty-six antibodies neutralized tier 2 viruses-in some cases, viruses from the same clade as BG505, and in others, a different clade, although none showed notable breadth. Several nAbs demonstrated antibody-dependent cellular cytotoxicity activity and targeted the V3 loop. These findings suggest an impressive polyclonal response to HIV infection in infant BG505, adding to the growing evidence that the nAb response to HIV in infants is polyclonal-a desirable vaccine response to a rapidly evolving virus like HIV.


Subject(s)
Antibodies, Neutralizing/biosynthesis , B-Lymphocytes/immunology , HIV Antibodies/biosynthesis , HIV Infections/prevention & control , HIV-1/immunology , Immunoglobulin G/biosynthesis , Adult , Amino Acid Sequence , Antibodies, Neutralizing/classification , Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/virology , Child, Preschool , Clone Cells , Epitopes/chemistry , HIV Antibodies/classification , HIV Infections/immunology , HIV Infections/virology , Humans , Immunoglobulin G/classification , Male
5.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33637649

ABSTRACT

A vaccine which is effective against the HIV virus is considered to be the best solution to the ongoing global HIV/AIDS epidemic. In the past thirty years, numerous attempts to develop an effective vaccine have been made with little or no success, due, in large part, to the high mutability of the virus. More recent studies showed that a vaccine able to elicit broadly neutralizing antibodies (bnAbs), that is, antibodies that can neutralize a high fraction of global virus variants, has promise to protect against HIV. Such a vaccine has been proposed to involve at least three separate stages: First, activate the appropriate precursor B cells; second, shepherd affinity maturation along pathways toward bnAbs; and, third, polish the Ab response to bind with high affinity to diverse HIV envelopes (Env). This final stage may require immunization with a mixture of Envs. In this paper, we set up a framework based on theory and modeling to design optimal panels of antigens to use in such a mixture. The designed antigens are characterized experimentally and are shown to be stable and to be recognized by known HIV antibodies.


Subject(s)
AIDS Vaccines/biosynthesis , Antigens, Viral/chemistry , Broadly Neutralizing Antibodies/biosynthesis , Epitopes/chemistry , HIV Antibodies/biosynthesis , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/chemistry , AIDS Vaccines/genetics , Amino Acid Sequence , Antigens, Viral/genetics , Antigens, Viral/immunology , Binding Sites , Broadly Neutralizing Antibodies/chemistry , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Crystallography, X-Ray , Epitopes/genetics , Epitopes/immunology , HIV Antibodies/chemistry , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/metabolism , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Engineering/methods , Protein Interaction Domains and Motifs
6.
FEBS Lett ; 595(3): 379-388, 2021 02.
Article in English | MEDLINE | ID: mdl-33263189

ABSTRACT

The tobacco variant Nicotiana benthamiana has recently emerged as a versatile host for the manufacturing of protein therapeutics, but the fidelity of many recombinant proteins generated in this system is compromised by inadvertent proteolysis. Previous studies have revealed that the anti-HIV-1 antibodies 2F5 and PG9 as well as the protease inhibitor α1 -antitrypsin (A1AT) are particularly susceptible to N. benthamiana proteases. Here, we identify two subtilisin-like serine proteases (NbSBT1 and NbSBT2) whose combined action is sufficient to account for all major cleavage events observed upon expression of 2F5, PG9 and A1AT in N. benthamiana. We propose that downregulation of NbSBT1 and NbSBT2 activities could constitute a powerful means to optimize the performance of this promising platform for the production of biopharmaceuticals. DATABASES: NbSBT sequence data are available in the DDBJ/EMBL/GenBank databases under the accession numbers MN534996 to MN535005.


Subject(s)
Antibodies, Monoclonal/chemistry , HIV Antibodies/chemistry , Nicotiana/genetics , Plant Proteins/antagonists & inhibitors , Subtilisins/antagonists & inhibitors , alpha 1-Antitrypsin/chemistry , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Gene Expression , HIV Antibodies/biosynthesis , HIV Antibodies/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Phenylmethylsulfonyl Fluoride/pharmacology , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified , Protease Inhibitors/pharmacology , Proteolysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Subtilisins/genetics , Subtilisins/metabolism , Nicotiana/drug effects , Nicotiana/enzymology , alpha 1-Antitrypsin/biosynthesis , alpha 1-Antitrypsin/genetics
7.
Virology ; 550: 99-108, 2020 11.
Article in English | MEDLINE | ID: mdl-32980676

ABSTRACT

The membrane proximal external region (MPER) of HIV-1 gp41 is targeted by several neutralizing antibodies (NAbs) and is of interest for vaccine design. In this study, we identified novel MPER peptide mimotopes and evaluated their reactivity with HIV + plasma antibodies to characterize the diversity of the immune responses to MPER during natural infection. We utilized phage display technology to generate novel mimotopes that fit antigen-binding sites of MPER NAbs 4E10, 2F5 and Z13. Plasma antibodies from 10 HIV + patients were mapped by phage immunoprecipitation, to identify unique patient MPER binding profiles that were distinct from, and overlapping with, those of MPER NAbs. 4E10 mimotope binding profiles correlated with plasma neutralization of HIV-2/HIV-1 MPER chimeric virus, and with overall plasma neutralization breadth and potency. When administered as vaccines, 4E10 mimotopes elicited low titer NAb responses in mice. HIV mimotopes may be useful for detailed analysis of plasma antibody specificity.


Subject(s)
Epitopes/chemistry , HIV Antibodies/biosynthesis , HIV Envelope Protein gp41/chemistry , HIV Infections/prevention & control , HIV-1/immunology , HIV-2/immunology , AIDS Vaccines/administration & dosage , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antibody Specificity , Antigens, Viral/chemistry , Antigens, Viral/immunology , Antigens, Viral/metabolism , Binding, Competitive , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/immunology , Epitopes/metabolism , Female , HIV Envelope Protein gp41/immunology , HIV Envelope Protein gp41/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , HIV-2/drug effects , HIV-2/genetics , Humans , Immunity, Humoral/drug effects , Immunization/methods , Mice , Mice, Inbred BALB C , Peptide Library , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Protein Binding
8.
Viruses ; 12(2)2020 01 31.
Article in English | MEDLINE | ID: mdl-32023860

ABSTRACT

Non-human primates (NHP) are the only animal model suitable to evaluate the protection efficacy of HIV-1 vaccines. It is important to understand how and when neutralizing antibodies (nAbs) with specificities similar to those of human broadly neutralizing antibodies (bnAbs) develop in NHPs. To address these questions, we determined plasma neutralization specificities in two macaques which developed neutralization breadth after long-term simian/human immunodeficiency virus (SHIV) infection and identified neutralization escape mutations by analyzing the env sequences from longitudinal plasma samples. Neutralization activities targeting V2, CD4bs, V3 and gp120-gp41 interface only became detectable in week 350 plasma from macaques G1015R and G1020R using 25710 env mutants. When mapped with CAP45 env mutants, only V2 specificity was detected at week 217 and persisted until week 350 in G1015R. Neutralization escape mutations were found in CD4bs and V2 regions. However, all of them were different from those resistant mutations identified for human bnAbs. These results show that nAbs with specificities similar to human bnAbs are only detectable after long-term SHIV infection and that neutralization escape mutations in macaques are different from those found in HIV-1-infected individuals. These findings can have important implications in the best utilization of the NHP model to evaluate HIV-1 vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , HIV Antibodies/immunology , HIV-1/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Genes, env/genetics , Genes, env/immunology , HIV Antibodies/biosynthesis , HIV Seropositivity , Macaca mulatta/immunology , Mutation , Neutralization Tests , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Time Factors
9.
Front Immunol ; 10: 2793, 2019.
Article in English | MEDLINE | ID: mdl-31867001

ABSTRACT

There is an urgent need for the development of potent vaccination regimens that are able to induce specific T and B cell responses against human immunodeficiency virus type 1 (HIV-1). Here, we describe the generation and characterization of a fusion antigen comprised of the HIV-1 envelope GP120 glycoprotein from clade C (GP120C) fused at its C-terminus, with the modified vaccinia virus (VACV) 14K protein (A27L gene) (termed GP120C14K). The design is directed toward improving the immunogenicity of the GP120C protein through its oligomerization facilitated by the fused VACV 14K protein that results in hexamer-like structures. Two different immunogens were generated: a recombinant GP120C14K fusion protein (purified from a stable CHO-K1 cell line) and a recombinant modified vaccinia virus Ankara (MVA) poxvirus vector expressing the GP120C14K fusion protein (termed MVA-GP120C14K). The GP120C14K fusion protein is recognized by broadly neutralizing antibodies (bNAbs) against HIV-1. In a murine model, a heterologous prime/boost immunization regimen with MVA-GP120C14K prime followed by adjuvanted GP120C14K protein boost generated stronger and polyfunctional HIV-1 Env-specific CD8 T cell responses when compared with the delivery of the monomeric GP120C form. Furthermore, the immunization protocol MVA-GP120C14K/GP120C14K elicited higher HIV-1 Env-specific T follicular helper cells, germinal center B cells and antibody responses than monomeric GP120. In addition, a similar MVA-GP120C14K prime/GP120C14K protein boost regimen performed in rabbits triggered high HIV-1-Env-specific IgG binding antibody titers that were capable of neutralizing HIV-1 pseudoviruses. The extent of HIV-1 neutralization was comparable to that elicited by the current standard GP140 SOSIP trimers from clades B and C when immunized as MVA-SOSIP prime/SOSIP protein boost regimen. Overall, the novel fusion antigen and the corresponding immunization scheme provided in this report can therefore be considered as potential vaccine strategies against HIV-1.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/biosynthesis , CHO Cells , Cricetulus , Female , Humans , Immunization , Mice , Mice, Inbred BALB C , Rabbits , Recombinant Fusion Proteins/chemistry
10.
Sci Rep ; 9(1): 15515, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664100

ABSTRACT

Recombinant Mycobacterium strains such as recombinant BCG (rBCG) have received considerable attention for the HIV-1 vaccine development. Recently, we described a temperature-sensitive Mycobacterium paragordonae (Mpg) strain as a novel live tuberculosis vaccine that is safer and showed an enhanced protective effect against mycobacterial infection compared to BCG. We studied the possibility of developing a vaccine against HIV-1 infection using rMpg strain expressing the p24 antigen (rMpg-p24). We observed that rMpg-p24 can induce an increased p24 expression in infected antigen presenting cells (APCs) compared to rBCG-p24. We also observed that rMpg-p24 can induce enhanced p24 specific immune responses in vaccinated mice as evidenced by increased p24-specific T lymphocyte proliferation, gamma interferon induction, antibody production and cytotoxic T lymphocyte (CTL) responses. Furthermore, an rMpg-p24 prime and plasmid DNA boost showed an increased CTL response and antibody production compared to rBCG or rMpg alone. In summary, our study indicates that a live rMpg-p24 strain induced enhanced immune responses against HIV-1 Gag in vaccinated mice. Thus, rMpg-p24 may have potential as a preventive prime vaccine in a heterologous prime-boost regimen for HIV-1 infection.


Subject(s)
AIDS Vaccines/immunology , Gene Products, gag/immunology , HIV Infections/prevention & control , HIV-1/immunology , Mycobacterium/genetics , Recombination, Genetic , Animals , Cell Proliferation , Cytokines/biosynthesis , Female , HIV Antibodies/biosynthesis , HIV Infections/immunology , HIV-1/genetics , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology
11.
Appl Microbiol Biotechnol ; 103(21-22): 8875-8888, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31641814

ABSTRACT

Monoclonal antibodies (mABs) are of great biopharmaceutical importance for the diagnosis and treatment of diseases. However, their production in mammalian expression hosts usually requires extensive production times and is expensive. Escherichia coli has become a new platform for production of functional small antibody fragment variants. In this study, we have used a rhamnose-inducible expression system that allows precise control of protein expression levels. The system was first evaluated for the cytoplasmic production of super folder green fluorescence protein (sfGFP) in various production platforms and then for the periplasmic production of the anti-HIV single-chain variable antibody fragment (scFv) of PGT135. Anti-HIV broadly neutralizing antibodies, like PGT135, have potential for clinical use to prevent HIV transmission, to promote immune responses and to eradicate infected cells. Different concentrations of L-rhamnose resulted in the controlled production of both sfGFP and scFv PGT135 antibody. In addition, by optimizing the culture conditions, the amount of scFv PGT135 antibody that was expressed soluble or as inclusions bodies could be modulated. The proteins were produced in batch bioreactors, with yields of 4.9 g/L for sfGFP and 0.8 g/L for scFv. The functionality of the purified antibodies was demonstrated by their ability to neutralize a panel of different HIV variants in vitro. We expect that this expression system will prove very useful for the development of a more cost-effective production process for proteins and antibody fragments in microbial cells.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Escherichia coli/metabolism , HIV Antibodies/biosynthesis , HIV Infections/therapy , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Bioreactors/microbiology , Escherichia coli/genetics , Gene Expression/genetics , HIV Antibodies/therapeutic use , HIV-1/immunology , Promoter Regions, Genetic/genetics , Single-Chain Antibodies/immunology
12.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31619555

ABSTRACT

Soluble recombinant native-like (NL) envelope glycoprotein (Env) trimers of various human immunodeficiency virus type 1 (HIV-1) genotypes are being developed as vaccine candidates aimed at the induction of broadly neutralizing antibodies (bNAbs). The prototypic design, designated BG505 SOSIP.664, incorporates an intersubunit disulfide bond (SOS) to covalently link the gp120 and gp41 ectodomain (gp41ECTO) subunits and a point substitution, I559P (IP), to further stabilize the gp41ECTO components. Without the SOS and IP changes, proteolytically cleaved trimers tend to disintegrate into their constituent gp120 and gp41ECTO subunits. We show, however, that NL trimers lacking the SOS and/or IP change can be affinity purified in amounts sufficient for analyses of their antigenicity and thermal stability. In general, these trimer variants have properties highly comparable to those of the fully stabilized SOSIP.664 version. We conclude that the major effect of the SOS and IP changes is to substantially increase trimer stability during and after the expression process, thereby allowing useful amounts to be produced. However, once the trimers have been purified, the SOS and IP changes have only subtle impacts on thermostability and the antigenicity of bNAb and other epitopes.IMPORTANCE Recombinant trimeric proteins based on HIV-1 env genes are being developed for vaccine trials in humans. A feature of these proteins is their mimicry of the envelope glycoprotein structure on virus particles that is targeted by neutralizing antibodies, i.e., antibodies that prevent cells from becoming infected. One vaccine concept under exploration is that recombinant trimers may be able to elicit virus-neutralizing antibodies when delivered as immunogens. A commonly used design is designated SOSIP.664, a term reflecting the sequence changes that are used to stabilize the trimers and allow their production in practically useful amounts. Here, we show that these stabilizing changes act to increase trimer yield during the biosynthesis process within the producer cell but have little impact on the properties of purified trimers.


Subject(s)
AIDS Vaccines/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/genetics , HIV-1/genetics , Recombinant Fusion Proteins/genetics , env Gene Products, Human Immunodeficiency Virus/genetics , AIDS Vaccines/biosynthesis , Animals , Antibodies, Neutralizing/biosynthesis , CHO Cells , Cricetulus , Disulfides/chemistry , Gene Expression , Genotype , HEK293 Cells , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/immunology , HIV-1/classification , HIV-1/immunology , Humans , Point Mutation , Protein Domains , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Temperature , env Gene Products, Human Immunodeficiency Virus/immunology
13.
PLoS Pathog ; 15(9): e1008026, 2019 09.
Article in English | MEDLINE | ID: mdl-31527908

ABSTRACT

The CD4 binding site (CD4bs) of the HIV-1 envelope glycoprotein is susceptible to multiple lineages of broadly neutralizing antibodies (bnAbs) that are attractive to elicit with vaccines. The CH235 lineage (VH1-46) of CD4bs bnAbs is particularly attractive because the most mature members neutralize 90% of circulating strains, do not possess long HCDR3 regions, and do not contain insertions and deletions that may be difficult to induce. We used virus neutralization to measure the interaction of CH235 unmutated common ancestor (CH235 UCA) with functional Env trimers on infectious virions to guide immunogen design for this bnAb lineage. Two Env mutations were identified, one in loop D (N279K) and another in V5 (G458Y), that acted synergistically to render autologous CH505 transmitted/founder virus susceptible to neutralization by CH235 UCA. Man5-enriched N-glycans provided additional synergy for neutralization. CH235 UCA bound with nanomolar affinity to corresponding soluble native-like Env trimers as candidate immunogens. A cryo-EM structure of CH235 UCA bound to Man5-enriched CH505.N279K.G458Y.SOSIP.664 revealed interactions of the antibody light chain complementarity determining region 3 (CDR L3) with the engineered Env loops D and V5. These results demonstrate that virus neutralization can directly inform vaccine design and suggest a germline targeting and reverse engineering strategy to initiate and mature the CH235 bnAb lineage.


Subject(s)
AIDS Vaccines/immunology , Broadly Neutralizing Antibodies/biosynthesis , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , HIV-1/genetics , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/chemistry , AIDS Vaccines/genetics , Amino Acid Substitution , Antibody Affinity , Binding Sites , CD4 Antigens/metabolism , Drug Design , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , HEK293 Cells , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/pathogenicity , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Engineering , Protein Multimerization , Protein Structure, Quaternary , env Gene Products, Human Immunodeficiency Virus/chemistry
14.
Gene Ther ; 26(9): 363-372, 2019 09.
Article in English | MEDLINE | ID: mdl-31300730

ABSTRACT

Self-amplifying RNA (saRNA) is a promising biotherapeutic tool that has been used as a vaccine against both infectious diseases and cancer. saRNA has been shown to induce protein expression for up to 60 days and elicit immune responses with lower dosing than messenger RNA (mRNA). Because saRNA is a large (~9500 nt), negatively charged molecule, it requires a delivery vehicle for efficient cellular uptake and degradation protection. Lipid nanoparticles (LNPs) have been widely used for RNA formulations, where the prevailing paradigm is to encapsulate RNA within the particle, including the first FDA-approved small-interfering siRNA therapy. Here, we compared LNP formulations with cationic and ionizable lipids with saRNA either on the interior or exterior of the particle. We show that LNPs formulated with cationic lipids protect saRNA from RNAse degradation, even when it is adsorbed to the surface. Furthermore, cationic LNPs deliver saRNA equivalently to particles formulated with saRNA encapsulated in an ionizable lipid particle, both in vitro and in vivo. Finally, we show that cationic and ionizable LNP formulations induce equivalent antibodies against HIV-1 Env gp140 as a model antigen. These studies establish formulating saRNA on the surface of cationic LNPs as an alternative to the paradigm of encapsulating RNA.


Subject(s)
Nanoparticles , RNA, Messenger/administration & dosage , Animals , Cations , Fatty Acids, Monounsaturated/chemistry , Female , HEK293 Cells , HIV Antibodies/biosynthesis , Humans , Lipids/administration & dosage , Lipids/chemistry , Luciferases, Firefly/metabolism , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Particle Size , Quaternary Ammonium Compounds/chemistry , RNA Stability , RNA, Messenger/immunology , RNA, Messenger/metabolism , Ribonucleases/metabolism , Transfection , env Gene Products, Human Immunodeficiency Virus/immunology
15.
PLoS One ; 14(5): e0216949, 2019.
Article in English | MEDLINE | ID: mdl-31100082

ABSTRACT

The development of an effective vaccine against HIV infection remains a global priority. Dendritic cell (DC)-based HIV immunotherapeutic vaccine is a promising approach which aims at optimizing the HIV-specific immune response using primed DCs to promote and enhance both the cellular and humoral arms of immunity. Since the Ebola virus envelope glycoprotein (EboGP) has strong DC-targeting ability, we investigated whether EboGP is able to direct HIV particles towards DCs efficiently and promote potent HIV-specific immune responses. Our results indicate that the incorporation of EboGP into non-replicating virus-like particles (VLPs) enhances their ability to target human monocyte-derived dendritic cells (MDDCs) and monocyte-derived macrophages (MDMs). Also, a mucin-like domain deleted EboGP (EboGPΔM) can further enhanced the MDDCs and MDMs-targeting ability. Furthermore, we investigated the effect of EboGP on HIV immunogenicity in mice, and the results revealed a significantly stronger HIV-specific humoral immune response when immunized with EboGP-pseudotyped HIV VLPs compared with those immunized with HIV VLPs. Splenocytes harvested from mice immunized with EboGP-pseudotyped HIV VLPs secreted increased levels of macrophage inflammatory proteins-1α (MIP-1α) and IL-4 upon stimulation with HIV Env and/or Gag peptides compared with those harvested from mice immunized with HIV VLPs. Collectively, this study provides evidence for the first time that the incorporation of EboGP in HIV VLPs can facilitate DC and macrophage targeting and induce more potent immune responses against HIV.


Subject(s)
AIDS Vaccines/immunology , Dendritic Cells/drug effects , HIV Antibodies/biosynthesis , HIV Infections/prevention & control , Macrophages/drug effects , Vaccines, Virus-Like Particle/immunology , Viral Envelope Proteins/genetics , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Ebolavirus/chemistry , Female , Gene Expression , HEK293 Cells , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/immunology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Primary Cell Culture , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Viral Envelope Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
16.
Nat Commun ; 10(1): 948, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30814513

ABSTRACT

An array of carbohydrates masks the HIV-1 surface protein Env, contributing to the evasion of humoral immunity. In most HIV-1 isolates 'glycan holes' occur due to natural sequence variation, potentially revealing the underlying protein surface to the immune system. Here we computationally design epitopes that mimic such surface features (carbohydrate-occluded neutralization epitopes or CONE) of Env through 'epitope transplantation', in which the target region is presented on a carrier protein scaffold with preserved structural properties. Scaffolds displaying the four CONEs are examined for structure and immunogenicity. Crystal structures of two designed proteins reflect the computational models and accurately mimic the native conformations of CONEs. The sera from rabbits immunized with several CONE immunogens display Env binding activity. Our method determines essential structural elements for targets of protective antibodies. The ability to design immunogens with high mimicry to viral proteins also makes possible the exploration of new templates for vaccine development.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Biophysical Phenomena , Carbohydrates/chemistry , Carbohydrates/immunology , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , HIV Antigens/chemistry , HIV Antigens/genetics , HIV Antigens/immunology , Humans , Models, Molecular , Protein Conformation , Protein Engineering , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics
17.
Plant Biotechnol J ; 17(8): 1646-1656, 2019 08.
Article in English | MEDLINE | ID: mdl-30729651

ABSTRACT

The discovery of broadly neutralizing antibodies (bNAbs) has been a major step towards better prophylactic and therapeutic agents against human immunodeficiency virus type 1 (HIV-1). However, effective therapy will likely require a combination of anti-HIV agents to avoid viral evasion. One possible solution to this problem is the creation of bispecific molecules that can concurrently target two vulnerable sites providing synergistic inhibitory effects. Here, we describe the production in plants and anti-HIV activity of a novel bispecific fusion protein consisting of the antigen-binding fragment (Fab) of the CD4 binding site-specific bNAb VRC01 and the antiviral lectin Avaren, which targets the glycan shield of the HIV-1 envelope (VRC01Fab -Avaren). This combination was justified by a preliminary experiment demonstrating the synergistic HIV-1 neutralization activity of VRC01 and Fc-fused Avaren dimer (Avaren-Fc). Using the GENEWARE® tobacco mosaic virus vector, VRC01Fab -Avaren was expressed in Nicotiana benthamiana and purified using a three-step chromatography procedure. Surface plasmon resonance and ELISA demonstrated that both the Avaren and VRC01Fab moieties retain their individual binding specificities. VRC01Fab -Avaren demonstrated enhanced neutralizing activity against representative HIV-1 strains from A, B and C clades, compared to equimolar combinations of VRC01Fab and Avaren. Notably, VRC01Fab -Avaren showed significantly stronger neutralizing effects than the bivalent parent molecules VRC01 IgG and Avaren-Fc, with IC50 values ranging from 48 to 310 pm. These results support the continued development of bispecific anti-HIV proteins based on Avaren and bNAbs, to which plant-based transient overexpression systems will provide an efficient protein engineering and production platform.


Subject(s)
Antibodies, Bispecific/biosynthesis , HIV Antibodies/biosynthesis , HIV-1 , Lectins/biosynthesis , Protein Engineering , Recombinant Fusion Proteins/biosynthesis , Antibodies, Bispecific/pharmacology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/pharmacology , HIV Antibodies/pharmacology , Lectins/pharmacology , Recombinant Fusion Proteins/pharmacology , Nicotiana
18.
Retrovirology ; 15(1): 63, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30208933

ABSTRACT

An effective HIV-1 vaccine probably will need to be able to induce broadly neutralizing HIV-1 antibodies (bNAbs) in order to be efficacious. The many bNAbs that have been isolated from HIV-1 infected patients illustrate that the human immune system is able to elicit this type of antibodies. The elucidation of the structure of the HIV-1 envelope glycoprotein (Env) trimer has further fueled the search for Env immunogens that induce bNAbs, but while native Env trimer mimetics are often capable of inducing strain-specific neutralizing antibodies (NAbs) against the parental virus, they have not yet induced potent bNAb responses. To improve the performance of Env trimer immunogens, researchers have studied the immune responses that Env trimers have induced in animals; they have evaluated how to best use Env trimers in various immunization regimens; and they have engineered increasingly stabilized Env trimer variants. Here, we review the different approaches that have been used to increase the stability of HIV-1 Env trimer immunogens with the aim of improving the induction of NAbs. In particular, we draw parallels between the various approaches to stabilize Env trimers and ones that have been used by nature in extremophile microorganisms in order to survive in extreme environmental conditions.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV-1/chemistry , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Epitopes/immunology , HIV Antibodies/biosynthesis , HIV-1/immunology , Humans , Immunization , Models, Molecular , Protein Multimerization , env Gene Products, Human Immunodeficiency Virus/genetics
19.
Proc Natl Acad Sci U S A ; 115(33): E7854-E7862, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061386

ABSTRACT

The transmission of HIV can be prevented by the application of neutralizing monoclonal antibodies and lectins. Traditional recombinant protein manufacturing platforms lack sufficient capacity and are too expensive for developing countries, which suffer the greatest disease burden. Plants offer an inexpensive and scalable alternative manufacturing platform that can produce multiple components in a single plant, which is important because multiple components are required to avoid the rapid emergence of HIV-1 strains resistant to single microbicides. Furthermore, crude extracts can be used directly for prophylaxis to avoid the massive costs of downstream processing and purification. We investigated whether rice could simultaneously produce three functional HIV-neutralizing proteins (the monoclonal antibody 2G12, and the lectins griffithsin and cyanovirin-N). Preliminary in vitro tests showed that the cocktail of three proteins bound to gp120 and achieved HIV-1 neutralization. Remarkably, when we mixed the components with crude extracts of wild-type rice endosperm, we observed enhanced binding to gp120 in vitro and synergistic neutralization when all three components were present. Extracts of transgenic plants expressing all three proteins also showed enhanced in vitro binding to gp120 and synergistic HIV-1 neutralization. Fractionation of the rice extracts suggested that the enhanced gp120 binding was dependent on rice proteins, primarily the globulin fraction. Therefore, the production of HIV-1 microbicides in rice may not only reduce costs compared to traditional platforms but may also provide functional benefits in terms of microbicidal potency.


Subject(s)
Anti-HIV Agents , Antibodies, Monoclonal , Endosperm , HIV Antibodies , HIV Envelope Protein gp120/antagonists & inhibitors , HIV-1/chemistry , Oryza , Plants, Genetically Modified , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Endosperm/chemistry , Endosperm/genetics , Endosperm/metabolism , HIV Antibodies/biosynthesis , HIV Antibodies/chemistry , HIV Antibodies/genetics , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , Oryza/chemistry , Oryza/genetics , Oryza/metabolism , Plants, Genetically Modified/chemistry , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism
20.
Adv Exp Med Biol ; 1075: 3-30, 2018.
Article in English | MEDLINE | ID: mdl-30030787

ABSTRACT

Despite progress in antiretroviral therapy, pre-exposure prophylaxis, microbicides, and other preventive strategies, a vaccine to prevent HIV-1 infection remains desperately needed. Development of an effective vaccine is challenged by several immunologic features of HIV-1 evidenced by the failure of five of the six HIV-1 candidate vaccine efficacy trials to date. This chapter reviews these efficacy trials with a focus on the Phase 3 RV144 trial in Thailand, the only HIV-1 vaccine efficacy trial to show a moderate protective effect of 31% with respect to placebo administration. Although modest, this protection has allowed for the study of potential immunologic correlates of protection to improve development of future HIV-1 pox-protein and other vaccine strategies. Trials in Thailand and South Africa have built upon the RV144 framework to provide additional immunologic insights which enable current and future efficacy testing of related vaccine candidates.


Subject(s)
AIDS Vaccines , Clinical Trials as Topic , HIV Infections/prevention & control , HIV-1/immunology , Clinical Trials, Phase III as Topic , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , Humans , Immunogenicity, Vaccine , International Cooperation , South Africa , Thailand , Vaccine Potency , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...