1
|
Ortiz R, Barajas A, Pons-Grífols A, Trinité B, Tarrés-Freixas F, Rovirosa C, Urrea V, Barreiro A, Gonzalez-Tendero A, Rovira-Rigau M, Cardona M, Ferrer L, Clotet B, Carrillo J, Aguilar-Gurrieri C, Blanco J. Production and Immunogenicity of FeLV Gag-Based VLPs Exposing a Stabilized FeLV Envelope Glycoprotein. Viruses 2024; 16:987. [PMID: 38932278 PMCID: PMC11209239 DOI: 10.3390/v16060987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The envelope glycoprotein (Env) of retroviruses, such as the Feline leukemia virus (FeLV), is the main target of neutralizing humoral response, and therefore, a promising vaccine candidate, despite its reported poor immunogenicity. The incorporation of mutations that stabilize analogous proteins from other viruses in their prefusion conformation (e.g., HIV Env, SARS-CoV-2 S, or RSV F glycoproteins) has improved their capability to induce neutralizing protective immune responses. Therefore, we have stabilized the FeLV Env protein following a strategy based on the incorporation of a disulfide bond and an Ile/Pro mutation (SOSIP) previously used to generate soluble HIV Env trimers. We have characterized this SOSIP-FeLV Env in its soluble form and as a transmembrane protein present at high density on the surface of FeLV Gag-based VLPs. Furthermore, we have tested its immunogenicity in DNA-immunization assays in C57BL/6 mice. Low anti-FeLV Env responses were detected in SOSIP-FeLV soluble protein-immunized animals; however, unexpectedly no responses were detected in the animals immunized with SOSIP-FeLV Gag-based VLPs. In contrast, high humoral response against FeLV Gag was observed in the animals immunized with control Gag VLPs lacking SOSIP-FeLV Env, while this response was significantly impaired when the VLPs incorporated SOSIP-FeLV Env. Our data suggest that FeLV Env can be stabilized as a soluble protein and can be expressed in high-density VLPs. However, when formulated as a DNA vaccine, SOSIP-FeLV Env remains poorly immunogenic, a limitation that must be overcome to develop an effective FeLV vaccine.
Collapse
MESH Headings
- Animals
- Mice
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred C57BL
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Leukemia Virus, Feline/immunology
- Leukemia Virus, Feline/genetics
- Gene Products, gag/immunology
- Gene Products, gag/genetics
- Female
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/administration & dosage
- Humans
- Cats
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Raquel Ortiz
- IrsiCaixa, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Barajas
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Anna Pons-Grífols
- IrsiCaixa, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | | | | | | | | | | | | | | | | | | | - Bonaventura Clotet
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Infectious Diseases Department, Germans Trias I Pujol Hospital, 08916 Badalona, Spain
| | | | | | - Julià Blanco
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Germans Trias I Pujol Research Institute (IGTP), 08916 Badalona, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
| |
Collapse
|
2
|
Mietzner R, Barbey C, Lehr H, Ziegler CE, Peterhoff D, Wagner R, Goepferich A, Breunig M. Prolonged delivery of HIV-1 vaccine nanoparticles from hydrogels. Int J Pharm 2024; 657:124131. [PMID: 38643811 DOI: 10.1016/j.ijpharm.2024.124131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Immunization is a straightforward concept but remains for some pathogens like HIV-1 a challenge. Thus, new approaches towards increasing the efficacy of vaccines are required to turn the tide. There is increasing evidence that antigen exposure over several days to weeks induces a much stronger and more sustained immune response compared to traditional bolus injection, which usually leads to antigen elimination from the body within a couple of days. Therefore, we developed a poly(ethylene) glycol (PEG) hydrogel platform to investigate the principal feasibility of a sustained release of antigens to mimic natural infection kinetics. Eight-and four-armed PEG macromonomers of different MWs (10, 20, and 40 kDa) were end-group functionalized to allow for hydrogel formation via covalent cross-linking. An HIV-1 envelope (Env) antigen in its trimeric (Envtri) or monomeric (Envmono) form was applied. The soluble Env antigen was compared to a formulation of Env attached to silica nanoparticles (Env-SiNPs). The latter are known to have a higher immunogenicity compared to their soluble counterparts. Hydrogels were tunable regarding the rheological behavior allowing for different degradation times and release timeframes of Env-SiNPs over two to up to 50 days. Affinity measurements of the VCR01 antibody which specifically recognizes the CD4 binding site of Env, revealed that neither the integrity nor the functionality of Envmono-SiNPs (Kd = 2.1 ± 0.9 nM) and Envtri-SiNPs (Kd = 1.5 ± 1.3 nM), respectively, were impaired after release from the hydrogel (Kd before release: 2.1 ± 0.1 and 7.8 ± 5.3 nM, respectively). Finally, soluble Env and Env-SiNPs which are two physico-chemically distinct compounds, were co-delivered and shown to be sequentially released from one hydrogel which could be beneficial in terms of heterologous immunization or single dose vaccination. In summary, this study presents a tunable, versatile applicable, and effective delivery platform that could improve vaccination effectiveness also for other infectious diseases than HIV-1.
Collapse
Affiliation(s)
- Raphael Mietzner
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Clara Barbey
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Heike Lehr
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Christian E Ziegler
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany.
| |
Collapse
|
3
|
Barbey C, Wolf H, Wagner R, Pauly D, Breunig M. A shift of paradigm: From avoiding nanoparticular complement activation in the field of nanomedicines to its exploitation in the context of vaccine development. Eur J Pharm Biopharm 2023; 193:119-128. [PMID: 37838145 DOI: 10.1016/j.ejpb.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
The complement system plays a central role in our innate immunity to fight pathogenic microorganisms, foreign and altered cells, or any modified molecule. Consequences of complement activation include cell lysis, release of histamines, and opsonization of foreign structures in preparation for phagocytosis. Because nanoparticles interact with the immune system in various ways and can massively activate the complement system due to their virus-mimetic size and foreign texture, detrimental side effects have been described after administration like pro-inflammatory responses, inflammation, mild to severe anaphylactic crisis and potentially complement activated-related pseudoallergy (CARPA). Therefore, application of nanotherapeutics has sometimes been observed with restraint, and avoiding or even suppressing complement activation has been of utmost priority. In contrast, in the field of vaccine development, particularly protein-based immunogens that are attached to the surface of nanoparticles, may profit from complement activation regarding breadth and potency of immune response. Improved transport to the regional lymph nodes, enhanced antigen uptake and presentation, as well as beneficial effects on immune cells like B-, T- and follicular dendritic cells may be exploited by strategic nanoparticle design aimed to activate the complement system. However, a shift of paradigm regarding complement activation by nanoparticular vaccines can only be achieved if these beneficial effects are accurately elicited and overshooting effects avoided.
Collapse
Affiliation(s)
- Clara Barbey
- Department of Pharmaceutical Technology, University Regensburg, Regensburg, Germany
| | - Hannah Wolf
- Department of Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Diana Pauly
- Department of Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Barbey C, Su J, Billmeier M, Stefan N, Bester R, Carnell G, Temperton N, Heeney J, Protzer U, Breunig M, Wagner R, Peterhoff D. Immunogenicity of a silica nanoparticle-based SARS-CoV-2 vaccine in mice. Eur J Pharm Biopharm 2023; 192:41-55. [PMID: 37774890 DOI: 10.1016/j.ejpb.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Safe and effective vaccines have been regarded early on as critical in combating the COVID-19 pandemic. Among the deployed vaccine platforms, subunit vaccines have a particularly good safety profile but may suffer from a lower immunogenicity compared to mRNA based or viral vector vaccines. In fact, this phenomenon has also been observed for SARS-CoV-2 subunit vaccines comprising the receptor-binding domain (RBD) of the spike (S) protein. Therefore, RBD-based vaccines have to rely on additional measures to enhance the immune response. It is well accepted that displaying antigens on nanoparticles can improve the quantity and quality of vaccine-mediated both humoral and cell-mediated immune responses. Based on this, we hypothesized that SARS-CoV-2 RBD as immunogen would benefit from being presented to the immune system via silica nanoparticles (SiNPs). Herein we describe the preparation, in vitro characterization, antigenicity and in vivo immunogenicity of SiNPs decorated with properly oriented RBD in mice. We found our RBD-SiNP conjugates show narrow, homogeneous particle distribution with optimal size of about 100 nm for efficient transport to and into the lymph node. The colloidal stability and binding of the antigen was stable for at least 4 months at storage- and in vivo-temperatures. The antigenicity of the RBD was maintained upon binding to the SiNP surface, and the receptor-binding motif was readily accessible due to the spatial orientation of the RBD. The particles were efficiently taken up in vitro by antigen-presenting cells. In a mouse immunization study using an mRNA vaccine and spike protein as benchmarks, we found that the SiNP formulation was able to elicit a stronger RBD-specific humoral response compared to the soluble protein. For the adjuvanted RBD-SiNP we found strong S-specific multifunctional CD4+ T cell responses, a balanced T helper response, improved auto- and heterologous virus neutralization capacity, and increased serum avidity, suggesting increased affinity maturation. In summary, our results provide further evidence for the possibility of optimizing the cellular and humoral immune response through antigen presentation on SiNP.
Collapse
Affiliation(s)
- Clara Barbey
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
| | - Martina Billmeier
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Nadine Stefan
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
| | - George Carnell
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham ME4 4BF, United Kingdom
| | - Jonathan Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
5
|
Li F, Feng X, Huang J, Zhang M, Liu W, Wang X, Zhu R, Wang X, Wang P, Yu B, Li W, Qiao ZA, Yu X. Periodic Mesoporous Organosilica as a Nanoadjuvant for Subunit Vaccines Elicits Potent Antigen-Specific Germinal Center Responses by Activating Naive B Cells. ACS NANO 2023; 17:15424-15440. [PMID: 37552584 DOI: 10.1021/acsnano.3c00991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Infection diseases such as AIDS and COVID-19 remain challenging in regard to protective vaccine design, while adjuvants are critical for subunit vaccines to induce strong, broad, and durable immune responses against variable pathogens. Here, we demonstrate that periodic mesoporous organosilica (PMO) acts as a multifunctional nanoadjuvant by adsorbing recombinant protein antigens. It can effectively deliver antigens to lymph nodes (LNs), prolong antigen exposure, and rapidly elicit germinal center (GC) responses by directly activating naive B cells via the C-type lectin receptor signaling pathway. In mice, both the gp120 trimer (HIV-1 antigen) and the receptor-binding domain (SARS-CoV-2 antigen) with the PMO nanoadjuvant elicit potent and durable antibodies that neutralize heterologous virus strains. LN immune cells analysis shows that PMO helps to effectively activate the T-follicular helper cells, GC B cells, and memory B cells and eventually develop broad and durable humoral responses. Moreover, the PMO nanoadjuvant elicits a strong cellular immune response and shapes this immune response by eliciting high levels of effector T helper cell cytokines. This study identifies a promising nanoadjuvant for subunit vaccines against multiple pathogens.
Collapse
Affiliation(s)
- Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyao Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiaxing Huang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Mo Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xupu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Rui Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xun Wang
- State Key Laboratory of Genetic Engineering, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Pengfei Wang
- State Key Laboratory of Genetic Engineering, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wei Li
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Zhen-An Qiao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
6
|
Zhang YN, Paynter J, Antanasijevic A, Allen JD, Eldad M, Lee YZ, Copps J, Newby ML, He L, Chavez D, Frost P, Goodroe A, Dutton J, Lanford R, Chen C, Wilson IA, Crispin M, Ward AB, Zhu J. Single-component multilayered self-assembling protein nanoparticles presenting glycan-trimmed uncleaved prefusion optimized envelope trimmers as HIV-1 vaccine candidates. Nat Commun 2023; 14:1985. [PMID: 37031217 PMCID: PMC10082823 DOI: 10.1038/s41467-023-37742-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Uncleaved prefusion-optimized (UFO) design can stabilize diverse HIV-1 envelope glycoproteins (Envs). Single-component, self-assembling protein nanoparticles (1c-SApNP) can display 8 or 20 native-like Env trimers as vaccine candidates. We characterize the biophysical, structural, and antigenic properties of 1c-SApNPs that present the BG505 UFO trimer with wildtype and modified glycans. For 1c-SApNPs, glycan trimming improves recognition of the CD4 binding site without affecting broadly neutralizing antibodies (bNAbs) to major glycan epitopes. In mice, rabbits, and nonhuman primates, glycan trimming increases the frequency of vaccine responders (FVR) and steers antibody responses away from immunodominant glycan holes and glycan patches. The mechanism of vaccine-induced immunity is examined in mice. Compared with the UFO trimer, the multilayered E2p and I3-01v9 1c-SApNPs show 420 times longer retention in lymph node follicles, 20-32 times greater presentation on follicular dendritic cell dendrites, and up-to-4 times stronger germinal center reactions. These findings can inform future HIV-1 vaccine development.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jennifer Paynter
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Aleksandar Antanasijevic
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Joel D Allen
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Mor Eldad
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Maddy L Newby
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Deborah Chavez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Pat Frost
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Anna Goodroe
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - John Dutton
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Robert Lanford
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Christopher Chen
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Max Crispin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
7
|
Peterhoff D, Thalhauser S, Neckermann P, Barbey C, Straub K, Nazet J, Merkl R, Laengst G, Breunig M, Wagner R. Multivalent display of engineered HIV-1 envelope trimers on silica nanoparticles for targeting and in vitro activation of germline VRC01 B cells. Eur J Pharm Biopharm 2022; 181:88-101. [PMID: 36272655 DOI: 10.1016/j.ejpb.2022.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 12/14/2022]
Abstract
Selective targeting of germline B cells with specifically designed germline-targeting HIV-1 envelope immunogens (GT-Env) is considered a feasible vaccination strategy to elicit broadly neutralizing antibodies (bnAbs). BnAbs are extremely valuable because they neutralize genetically distant viral strains at the same time. To overcome its inherently low affinity to germline B cells, the aim of the study was to present GT-Env via different immobilization strategies densely arrayed on the surface of nanoparticles. We engineered a prefusion-stabilized GT-Env trimer with affinity to VRC01 germline B cells using a bioinformatics-supported design approach. Distinct glycan modifications and amino acid substitutions yielded a GT-Env trimer which bound to the receptor with a KD of 11.5 µM. Silica nanoparticles with 200 nm diameter (SiNPs) were used for the multivalent display of the novel GT-Env with a 15 nm mean centre-to-centre spacing either by site-specific, covalent conjugation or at random, non-specific adsorption. Oriented, covalent GT-Env conjugation revealed better binding of structure dependent bnAbs as compared to non-specifically adsorbed GT-Env. In addition, GT-Env covalently attached activated a B cell line expressing the germline VRC01 receptor at an EC50 value in the nanomolar range (4 nM), while soluble GT-Env required 1,000-fold higher concentrations to induce signalling. The significantly lower GT-Env concentration was likely required due to avidity effects, which were in the picomolar range. Thus, low affinity antigens may particularly benefit from a particulate and multivalent delivery. In future, SiNPs are ideal to be modified in a modular design with various GT-Env variants that target different stages of germline and bnAb precursor B cells.
Collapse
Affiliation(s)
- David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany.
| | - Stefanie Thalhauser
- Department of Pharmaceutical Technology, University of Regensburg, 93040 Regensburg, Germany
| | - Patrick Neckermann
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93040 Regensburg, Germany
| | - Clara Barbey
- Department of Pharmaceutical Technology, University of Regensburg, 93040 Regensburg, Germany
| | - Kristina Straub
- Regensburg Center for Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Julian Nazet
- Regensburg Center for Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Rainer Merkl
- Regensburg Center for Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Gernot Laengst
- Regensburg Center for Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, 93040 Regensburg, Germany.
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
8
|
Lamrayah M, Phelip C, Coiffier C, Lacroix C, Willemin T, Trimaille T, Verrier B. A Polylactide-Based Micellar Adjuvant Improves the Intensity and Quality of Immune Response. Pharmaceutics 2022; 14:pharmaceutics14010107. [PMID: 35057003 PMCID: PMC8778782 DOI: 10.3390/pharmaceutics14010107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/21/2022] Open
Abstract
Micelles from amphiphilic polylactide-block-poly(N-acryloxysuccinimide-co-N-vinylpyrrolidone) (PLA-b-P(NAS-co-NVP)) block copolymers of 105 nm in size were characterized and evaluated in a vaccine context. The micelles were non-toxic in vitro (both in dendritic cells and HeLa cells). In vitro fluorescence experiments combined with in vivo fluorescence tomography imaging, through micelle loading with the DiR near infrared probe, suggested an efficient uptake of the micelles by the immune cells. The antigenic protein p24 of the HIV-1 was successfully coupled on the micelles using the reactive N-succinimidyl ester groups on the micelle corona, as shown by SDS-PAGE analyses. The antigenicity of the coupled antigen was preserved and even improved, as assessed by the immuno-enzymatic (ELISA) test. Then, the performances of the micelles in immunization were investigated and compared to different p24-coated PLA nanoparticles, as well as Alum and MF59 gold standards, following a standardized HIV-1 immunization protocol in mice. The humoral response intensity (IgG titers) was substantially similar between the PLA micelles and all other adjuvants over an extended time range (one year). More interestingly, this immune response induced by PLA micelles was qualitatively higher than the gold standards and PLA nanoparticles analogs, expressed through an increasing avidity index over time (>60% at day 365). Taken together, these results demonstrate the potential of such small-sized micellar systems for vaccine delivery.
Collapse
Affiliation(s)
- Myriam Lamrayah
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
- Correspondence: (M.L.); (T.T.)
| | - Capucine Phelip
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Coiffier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Lacroix
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thibaut Willemin
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thomas Trimaille
- Laboratoire Ingénierie des Matériaux Polymères (IMP), CNRS UMR 5223, Université Lyon 1, Université de Lyon, 69622 Villeurbanne, France
- Correspondence: (M.L.); (T.T.)
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| |
Collapse
|
9
|
Li S, Wang B, Jiang S, Pan Y, Shi Y, Kong W, Shan Y. Surface-Functionalized Silica-Coated Calcium Phosphate Nanoparticles Efficiently Deliver DNA-Based HIV-1 Trimeric Envelope Vaccines against HIV-1. ACS APPLIED MATERIALS & INTERFACES 2021; 13:53630-53645. [PMID: 34735127 DOI: 10.1021/acsami.1c16989] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection remains one of the worst crises in global health. The prevention of HIV-1 infection is a crucial task that needs to be addressed due to the absence of a licensed vaccine against HIV-1. DNA vaccines present a promising alternative approach to combat HIV-1 infection due to their excellent safety profile, lack of severe side effects, and relatively rapid fabrication. Traditional vaccines composed of a monomeric envelope or peptide fragments have been indicated to lack protective efficacy mediated by inducing HIV-1-specific neutralizing antibodies in clinical trials. The immunogenicity and protection against HIV-1 induced by DNA vaccines are limited due to the poor uptake of these vaccines by antigen-presenting cells and their ready degradation by DNases and lysosomes. To address these issues of naked DNA vaccines, we described the feasibility of CpG-functionalized silica-coated calcium phosphate nanoparticles (SCPs) for efficiently delivering DNA-based HIV-1 trimeric envelope vaccines against HIV-1. Vaccines comprising the soluble BG505 SOSIP.664 trimer fused to the GCN4-based isoleucine zipper or bacteriophage T4 fibritin foldon motif with excellent simulation of the native HIV-1 envelope were chosen as trimer-based vaccine platforms. Our results showed that SCP-based DNA immunization could significantly induce both broad humoral immune responses and potent cellular immune responses compared to naked DNA vaccination in vivo. To the best of our knowledge, this study is the first to assess the feasibility of CpG-functionalized SCPs for efficiently delivering DNA vaccines expressing a native-like HIV-1 trimer. These CpG-functionalized SCPs for delivering DNA-based HIV-1 trimeric envelope vaccines may lead to the development of promising vaccine candidates against HIV-1.
Collapse
Affiliation(s)
- Shuang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Bo Wang
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Shun Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yi Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
10
|
Broad and potent bispecific neutralizing antibody gene delivery using adeno-associated viral vectors for passive immunization against HIV-1. J Control Release 2021; 338:633-643. [PMID: 34509584 DOI: 10.1016/j.jconrel.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
Broadly neutralizing antibodies (bNAbs) possess favorable safety, and passive immunization using these can prevent or control human immunodeficiency virus type 1 (HIV-1) infection. However, bNAbs generally used for monotherapy (IC80 > 5 μg/mL) have limited breadth and potency and neutralize only 70-90% of all HIV-1 strains. To address the need for broader coverage of the HIV-1 epidemic and enhance the ability of bNAbs to target HIV-1, we fused the single-chain variable antibody fragment (scFv) of bNAbs (PG9, PGT123, or NIH45-46) with full-length ibalizumab (iMab) in an scFv-monoclonal antibody tandem format to construct bispecific bNAbs (BibNAbs). Additionally, we described the feasibility of BibNAb gene delivery mediated by recombinant adeno-associated virus 8 (rAAV8) for generating long-term expression with a single injection as opposed to short-term passive immunization requiring continuous injections. Our results showed that the expressed BibNAbs targeting two distinct epitopes exhibited neutralizing activity against 20 HIV-1 pseudoviruses in vitro. After injecting a single rAAV8 vector, the expression and neutralizing activity of the BibNAbs in serum were sustained for 24 weeks. To the best of our knowledge, very few studies have been published on BibNAb gene delivery using rAAV8 vectors against HIV-1. BibNAb gene delivery using rAAV8 vectors may be promising for passive immunization against HIV-1 infection.
Collapse
|
11
|
Interests of the Non-Human Primate Models for HIV Cure Research. Vaccines (Basel) 2021; 9:vaccines9090958. [PMID: 34579195 PMCID: PMC8472852 DOI: 10.3390/vaccines9090958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Non-human primate (NHP) models are important for vaccine development and also contribute to HIV cure research. Although none of the animal models are perfect, NHPs enable the exploration of important questions about tissue viral reservoirs and the development of intervention strategies. In this review, we describe recent advances in the use of these models for HIV cure research and highlight the progress that has been made as well as limitations using these models. The main NHP models used are (i) the macaque, in which simian immunodeficiency virus (SIVmac) infection displays similar replication profiles as to HIV in humans, and (ii) the macaque infected by a recombinant virus (SHIV) consisting of SIVmac expressing the HIV envelope gene serving for studies analyzing the impact of anti-HIV Env broadly neutralizing antibodies. Lessons for HIV cure that can be learned from studying the natural host of SIV are also presented here. An overview of the most promising and less well explored HIV cure strategies tested in NHP models will be given.
Collapse
|
12
|
Peterhoff D, Thalhauser S, Sobczak JM, Mohsen MO, Voigt C, Seifert N, Neckermann P, Hauser A, Ding S, Sattentau Q, Bachmann MF, Breunig M, Wagner R. Augmenting the Immune Response against a Stabilized HIV-1 Clade C Envelope Trimer by Silica Nanoparticle Delivery. Vaccines (Basel) 2021; 9:642. [PMID: 34208059 PMCID: PMC8230641 DOI: 10.3390/vaccines9060642] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
The delivery of HIV-1 envelope (Env) trimer-based immunogens on the surface of nanoparticles holds promise to promote immunogenicity with the aim of inducing a potent, durable and broad neutralizing antibody (bnAb) response. Towards that goal, we examined the covalent conjugation of Env to 100 nm and 200 nm silica nanoparticles (SiNPs) to optimize conjugation density and attachment stability. Env was redesigned to enable site-specific cysteine-mediated covalent conjugation while maintaining its structural integrity and antigenicity. Env was anchored to different sized SiNPs with a calculated spacing of 15 nm between adjacent trimers. Both particle sizes exhibited high in vitro stability over a seven-day period. After attachment, 100 nm particles showed better colloidal stability compared to 200 nm particles. Importantly, the antigenic profile of Env was not impaired by surface attachment, indicating that the quaternary structure was maintained. In vitro Env uptake by dendritic cells was significantly enhanced when Env was delivered on the surface of nanoparticles compared to soluble Env. Furthermore, multivalent Env displayed efficiently activated B cells even at Env concentrations in the low nanomolar range. In mice, antibody responses to nanoparticle-coupled Env were stronger compared to the free protein and had equivalent effects at lower doses and without adjuvant.
Collapse
Affiliation(s)
- David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stefanie Thalhauser
- Institute of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Germany; (S.T.); (M.B.)
| | - Jan M. Sobczak
- Department for BioMedical Research, University of Bern, 3010 Bern, Switzerland; (J.M.S.); (M.O.M.); (M.F.B.)
- Department of Immunology RI, University Hospital Bern, 3010 Bern, Switzerland
| | - Mona O. Mohsen
- Department for BioMedical Research, University of Bern, 3010 Bern, Switzerland; (J.M.S.); (M.O.M.); (M.F.B.)
- Department of Immunology RI, University Hospital Bern, 3010 Bern, Switzerland
| | - Christoph Voigt
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
| | - Nicole Seifert
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
| | - Patrick Neckermann
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
| | - Alexandra Hauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
| | - Song Ding
- EuroVacc Foundation, 1002 Lausanne, Switzerland;
| | - Quentin Sattentau
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Martin F. Bachmann
- Department for BioMedical Research, University of Bern, 3010 Bern, Switzerland; (J.M.S.); (M.O.M.); (M.F.B.)
- Department of Immunology RI, University Hospital Bern, 3010 Bern, Switzerland
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Miriam Breunig
- Institute of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Germany; (S.T.); (M.B.)
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (C.V.); (N.S.); (P.N.); (A.H.)
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
13
|
Tortella GR, Rubilar O, Diez MC, Padrão J, Zille A, Pieretti JC, Seabra AB. Advanced Material Against Human (Including Covid-19) and Plant Viruses: Nanoparticles As a Feasible Strategy. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000049. [PMID: 33614127 PMCID: PMC7883180 DOI: 10.1002/gch2.202000049] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/06/2020] [Indexed: 05/03/2023]
Abstract
The SARS-CoV-2 virus outbreak revealed that these nano-pathogens have the ability to rapidly change lives. Undoubtedly, SARS-CoV-2 as well as other viruses can cause important global impacts, affecting public health, as well as, socioeconomic development. But viruses are not only a public health concern, they are also a problem in agriculture. The current treatments are often ineffective, are prone to develop resistance, or cause considerable adverse side effects. The use of nanotechnology has played an important role to combat viral diseases. In this review three main aspects are in focus: first, the potential use of nanoparticles as carriers for drug delivery. Second, its use for treatments of some human viral diseases, and third, its application as antivirals in plants. With these three themes, the aim is to give to readers an overview of the progress in this promising area of biotechnology during the 2017-2020 period, and to provide a glance at how tangible is the effectiveness of nanotechnology against viruses. Future prospects are also discussed. It is hoped that this review can be a contribution to general knowledge for both specialized and non-specialized readers, allowing a better knowledge of this interesting topic.
Collapse
Affiliation(s)
- Gonzalo R. Tortella
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio AmbienteCIBAMA‐BIORENUniversidad de La FronteraTemuco4811230Chile
| | - Olga Rubilar
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio AmbienteCIBAMA‐BIORENUniversidad de La FronteraTemuco4811230Chile
- Chemical Engineering DepartmentUniversidad de La FronteraTemuco4811230Chile
| | - María Cristina Diez
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio AmbienteCIBAMA‐BIORENUniversidad de La FronteraTemuco4811230Chile
- Chemical Engineering DepartmentUniversidad de La FronteraTemuco4811230Chile
| | - Jorge Padrão
- Centre for Textile Science and Technology (2C2T)University of MinhoGuimarães4800‐058Portugal
| | - Andrea Zille
- Centre for Textile Science and Technology (2C2T)University of MinhoGuimarães4800‐058Portugal
| | - Joana C. Pieretti
- Center for Natural and Human SciencesUniversidade Federal d ABC (UFABC)Santo André09210‐580Brazil
| | - Amedea B. Seabra
- Center for Natural and Human SciencesUniversidade Federal d ABC (UFABC)Santo André09210‐580Brazil
| |
Collapse
|
14
|
Wibowo D, Jorritsma SHT, Gonzaga ZJ, Evert B, Chen S, Rehm BHA. Polymeric nanoparticle vaccines to combat emerging and pandemic threats. Biomaterials 2020; 268:120597. [PMID: 33360074 PMCID: PMC7834201 DOI: 10.1016/j.biomaterials.2020.120597] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Subunit vaccines are more advantageous than live attenuated vaccines in terms of safety and scale-up manufacture. However, this often comes as a trade-off to their efficacy. Over the years, polymeric nanoparticles have been developed to improve vaccine potency, by engineering their physicochemical properties to incorporate multiple immunological cues to mimic pathogenic microbes and viruses. This review covers recent advances in polymeric nanostructures developed toward particulate vaccines. It focuses on the impact of microbe mimicry (e.g. size, charge, hydrophobicity, and surface chemistry) on modulation of the nanoparticles’ delivery, trafficking, and targeting antigen-presenting cells to elicit potent humoral and cellular immune responses. This review also provides up-to-date progresses on rational designs of a wide variety of polymeric nanostructures that are loaded with antigens and immunostimulatory molecules, ranging from particles, micelles, nanogels, and polymersomes to advanced core-shell structures where polymeric particles are coated with lipids, cell membranes, or proteins.
Collapse
Affiliation(s)
- David Wibowo
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia.
| | - Sytze H T Jorritsma
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Zennia Jean Gonzaga
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Benjamin Evert
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Shuxiong Chen
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia.
| |
Collapse
|
15
|
Suleiman E, Mayer J, Lehner E, Kohlhauser B, Katholnig A, Batzoni M, Damm D, Temchura V, Wagner A, Überla K, Vorauer-Uhl K. Conjugation of Native-Like HIV-1 Envelope Trimers onto Liposomes Using EDC/Sulfo-NHS Chemistry: Requirements and Limitations. Pharmaceutics 2020; 12:E979. [PMID: 33081278 PMCID: PMC7589475 DOI: 10.3390/pharmaceutics12100979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/29/2022] Open
Abstract
The display of native-like human immunodeficiency virus type 1 envelope (HIV-1 Env) trimers on liposomes has gained wide attention over the last few years. Currently, available methods have enabled the preparation of Env-liposome conjugates of unprecedented quality. However, these protocols require the Env trimer to be tagged and/or to carry a specific functional group. For this reason, we have investigated N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide/N-Hydroxysulfosuccinimide (EDC/Sulfo-NHS) chemistry for its potential to covalently conjugate tag-free, non-functionalized native-like Env trimers onto the surface of carboxyl-functionalized liposomes. The preservation of the liposome's physical integrity and the immunogen's conformation required a fine-tuned two-step approach based on the controlled use of β-mercaptoethanol. The display of Env trimers was strictly limited to activated liposomes of positive charge, i.e., liposomes with a positive zeta potential that carry amine-reactive Sulfo-NHS esters on their surface. In agreement with that, conjugation was found to be highly ionic strength- and pH-dependent. Overall, we have identified electrostatic pre-concentration (i.e., close proximity between negatively charged Env trimers and positively charged liposomes established through electrostatic attraction) to be crucial for conjugation reactions to proceed. The present study highlights the requirements and limitations of potentially scalable EDC/Sulfo-NHS-based approaches and represents a solid basis for further research into the controlled conjugation of tag-free, non-functionalized native-like Env trimers on the surface of liposomes, and other nanoparticles.
Collapse
Affiliation(s)
- Ehsan Suleiman
- Polymun Scientific Immunbiologische Forschung GmbH, 3400 Klosterneuburg, Austria;
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
| | - Julia Mayer
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
| | - Elisabeth Lehner
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
| | - Bianca Kohlhauser
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
- University of Vienna, 1010 Vienna, Austria
| | - Alexandra Katholnig
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
| | - Mirjam Batzoni
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
- FH Campus Wien, University of Applied Sciences, 1100 Vienna, Austria
| | - Dominik Damm
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (D.D.); (V.T.); (K.Ü.)
| | - Vladimir Temchura
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (D.D.); (V.T.); (K.Ü.)
| | - Andreas Wagner
- Polymun Scientific Immunbiologische Forschung GmbH, 3400 Klosterneuburg, Austria;
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (D.D.); (V.T.); (K.Ü.)
| | - Karola Vorauer-Uhl
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria; (J.M.); (E.L.); (B.K.); (A.K.); (M.B.); (K.V.-U.)
| |
Collapse
|
16
|
CD4 + T Cell-Mimicking Nanoparticles Broadly Neutralize HIV-1 and Suppress Viral Replication through Autophagy. mBio 2020; 11:mBio.00903-20. [PMID: 32934078 PMCID: PMC7492730 DOI: 10.1128/mbio.00903-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
HIV-1 is a major global health challenge. The development of an effective vaccine and/or a therapeutic cure is a top priority. The creation of vaccines that focus an antibody response toward a particular epitope of a protein has shown promise, but the genetic diversity of HIV-1 hinders this progress. Here we developed an approach using nanoengineered CD4+ T cell membrane-coated nanoparticles (TNP). Not only do TNP effectively neutralize all strains of HIV-1, but they also selectively bind to infected cells and decrease the release of HIV-1 particles through an autophagy-dependent mechanism with no drug-induced off-target or cytotoxic effects on bystander cells. Therapeutic strategies that provide effective and broad‐spectrum neutralization against HIV-1 infection are highly desirable. Here, we investigate the potential of nanoengineered CD4+ T cell membrane-coated nanoparticles (TNP) to neutralize a broad range of HIV-1 strains. TNP displayed outstanding neutralizing breadth and potency; they neutralized all 125 HIV-1-pseudotyped viruses tested, including global subtypes/recombinant forms, and transmitted/founder viruses, with a geometric mean 80% inhibitory concentration (IC80) of 819 μg ml−1 (range, 72 to 8,570 μg ml−1). TNP also selectively bound to and induced autophagy in HIV-1-infected CD4+ T cells and macrophages, while having no effect on uninfected cells. This TNP-mediated autophagy inhibited viral release and reduced cell-associated HIV-1 in a dose- and phospholipase D1-dependent manner. Genetic or pharmacological inhibition of autophagy ablated this effect. Thus, we can use TNP as therapeutic agents to neutralize cell-free HIV-1 and to target HIV-1 gp120-expressing cells to decrease the HIV-1 reservoir.
Collapse
|
17
|
Antanasijevic A, Ueda G, Brouwer PJM, Copps J, Huang D, Allen JD, Cottrell CA, Yasmeen A, Sewall LM, Bontjer I, Ketas TJ, Turner HL, Berndsen ZT, Montefiori DC, Klasse PJ, Crispin M, Nemazee D, Moore JP, Sanders RW, King NP, Baker D, Ward AB. Structural and functional evaluation of de novo-designed, two-component nanoparticle carriers for HIV Env trimer immunogens. PLoS Pathog 2020; 16:e1008665. [PMID: 32780770 PMCID: PMC7418955 DOI: 10.1371/journal.ppat.1008665] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Two-component, self-assembling nanoparticles represent a versatile platform for multivalent presentation of viral antigens. Computational design of protein nanoparticles with differing sizes and geometries enables combination with antigens of choice to test novel multimerization concepts in immunization strategies where the goal is to improve the induction and maturation of neutralizing antibody lineages. Here, we describe detailed antigenic, structural, and functional characterization of computationally designed tetrahedral, octahedral, and icosahedral nanoparticle immunogens displaying trimeric HIV envelope glycoprotein (Env) ectodomains. Env trimers, based on subtype A (BG505) or consensus group M (ConM) sequences and engineered with SOSIP stabilizing mutations, were fused to an underlying trimeric building block of each nanoparticle. Initial screening yielded one icosahedral and two tetrahedral nanoparticle candidates, capable of presenting twenty or four copies of the Env trimer. A number of analyses, including detailed structural characterization by cryo-EM, demonstrated that the nanoparticle immunogens possessed the intended structural and antigenic properties. When the immunogenicity of ConM-SOSIP trimers presented on a two-component tetrahedral nanoparticle or as soluble proteins were compared in rabbits, the two immunogens elicited similar serum antibody binding titers against the trimer component. Neutralizing antibody titers were slightly elevated in the animals given the nanoparticle immunogen and were initially more focused to the trimer apex. Altogether, our findings indicate that tetrahedral nanoparticles can be successfully applied for presentation of HIV Env trimer immunogens; however, the optimal implementation to different immunization strategies remains to be determined.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| | - George Ueda
- Institute for Protein Design, Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | | | - Jeffrey Copps
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| | - Deli Huang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Joel D. Allen
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Christopher A. Cottrell
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| | - Anila Yasmeen
- Weill Cornell Medicine, Cornell University, New York, New York, United States of America
| | - Leigh M. Sewall
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Ilja Bontjer
- Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Thomas J. Ketas
- Weill Cornell Medicine, Cornell University, New York, New York, United States of America
| | - Hannah L. Turner
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| | - Zachary T. Berndsen
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Per Johan Klasse
- Weill Cornell Medicine, Cornell University, New York, New York, United States of America
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - David Nemazee
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - John P. Moore
- Weill Cornell Medicine, Cornell University, New York, New York, United States of America
| | - Rogier W. Sanders
- Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Neil P. King
- Institute for Protein Design, Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - David Baker
- Institute for Protein Design, Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Andrew B. Ward
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
- International AIDS Vaccine Initiative Neutralizing Antibody Center, the Collaboration for AIDS Vaccine Discovery (CAVD) and Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research, La Jolla, California, United States of America
| |
Collapse
|