1
|
Portilho AI, Hermes Monteiro da Costa H, Grando Guereschi M, Prudencio CR, De Gaspari E. Hybrid response to SARS-CoV-2 and Neisseria meningitidis C after an OMV-adjuvanted immunization in mice and their offspring. Hum Vaccin Immunother 2024; 20:2346963. [PMID: 38745461 PMCID: PMC11789737 DOI: 10.1080/21645515.2024.2346963] [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: 12/15/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, and meningococcal disease, caused by Neisseria meningitidis, are relevant infectious diseases, preventable through vaccination. Outer membrane vesicles (OMVs), released from Gram-negative bacteria, such as N. meningitidis, present adjuvant characteristics and may confer protection against meningococcal disease. Here, we evaluated in mice the humoral and cellular immune response to different doses of receptor binding domain (RBD) of SARS-CoV-2 adjuvanted by N. meningitidis C:2a:P1.5 OMVs and aluminum hydroxide, as a combined preparation for these pathogens. The immunization induced IgG antibodies of high avidity for RBD and OMVs, besides IgG that recognized the Omicron BA.2 variant of SARS-CoV-2 with intermediary avidity. Cellular immunity showed IFN-γ and IL-4 secretion in response to RBD and OMV stimuli, demonstrating immunologic memory and a mixed Th1/Th2 response. Offspring presented transferred IgG of similar levels and avidity as their mothers. Humoral immunity did not point to the superiority of any RBD dose, but the group immunized with a lower antigenic dose (0.5 μg) had the better cellular response. Overall, OMVs enhanced RBD immunogenicity and conferred an immune response directed to N. meningitidis too.
Collapse
MESH Headings
- Animals
- Mice
- Immunoglobulin G/blood
- Neisseria meningitidis/immunology
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Adjuvants, Immunologic/administration & dosage
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Immunity, Cellular
- Immunity, Humoral
- Mice, Inbred BALB C
- Meningococcal Infections/prevention & control
- Meningococcal Infections/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Vaccine/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Immunization/methods
- Antibody Affinity
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Meningococcal Vaccines/immunology
- Meningococcal Vaccines/administration & dosage
- Immunologic Memory
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Hernan Hermes Monteiro da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | | | - Carlos Roberto Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Sharif E, Nezafat N, Mohit E. Recombinant ClearColi™-derived outer membrane vesicles as an effective carrier for development of neoepitope-based vaccine candidate against colon carcinoma. Int Immunopharmacol 2024; 143:113283. [PMID: 39418728 DOI: 10.1016/j.intimp.2024.113283] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is the third most common cancer worldwide, with high clonal heterogeneity due to somatic mutations. Poly neoepitope vaccines can inhibit the tumor's escape from the immune system. However, they have rapid clearance and low immunogenicity. Bacteria-derived recombinant outer membrane vesicles (OMVs) have gained increased attention as ideal cancer vaccine candidates due to their unique adjuvant properties and ability to carry antigens. Herein, the benefits of OMV-based and polyneoepitope-based vaccines were combined to obtain a functional individualized cancer vaccine. METHODS OMVs and rOMVs displaying CT26 polytopes were isolated from ClearColi™ and recombinant ClearColi™ containing pET-22b (ClyA-CT26 polytope) by the AS (70 %) + UDF method. BALB/c mice were immunized with OMVs (40 µg) and rOMVs (20 and 40 µg) and subcutaneously challenged with CT26 cells. Then, IgG1 and IgG2a antibodies specific for CT26 M90 and CT26 polytope, the stimulated IFN-γ, TNF-α, and IL-10 cytokines and the stimulated CTL responses by measuring granzyme B were evaluated. To investigate whether pooled sera and pooled splenocytes are indicators of individual responses, pooled and individual methods for determining the elicited immunity were compared. Additionally, the ability of OMVs and rOMVs (20 and 40 µg) to prevent tumor growth against the CT26 challenge was investigated. RESULTS Immunization with rOMVs displaying CT26 polytopes induced a higher titer of CT26 polytope- and CT26 M90 peptide-specific IgG2a than IgG1 antibodies in a dose-dependent manner, thus directing immunity to Th1. The antibody responses determined by pooled sera can be used as indicators of individual responses. In addition, both OMVs and rOMVs displaying CT26 polytopes could induce tumor-suppressing cytokines (IFN-γ and TNF-α). The ability of rOMVs displaying CT26 polytopes to induce these cytokines was higher than OMVs in a dose-dependent way. The results of the granzyme B assay were also in agreement with the cytokine assay. The survival of mice after the CT26 challenge was 100 % in the OMVs and rOMVs groups, and inhibition of tumor growth was significantly higher by rOMVs (40 µg) compared to OMVs. CONCLUSION The bioengineered OMVs displaying CT26 neoepitopes have the potential for the development of personalized tumor vaccines. Our results can provide new insights for developing rOMV-based vaccines displaying polytopes against diseases containing highly variable antigens.
Collapse
Affiliation(s)
- Elham Sharif
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Sharif E, Nezafat N, Ahmadi FM, Mohit E. In Silico Design of CT26 Polytope and its Surface Display by ClearColi™-Derived Outer Membrane Vesicles as a Cancer Vaccine Candidate Against Colon Carcinoma. Appl Biochem Biotechnol 2024; 196:8820-8847. [PMID: 38958886 DOI: 10.1007/s12010-024-04971-x] [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] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Simultaneous targeting of several mutations can be useful in colorectal cancer (CRC) due to its heterogeneity and presence of somatic mutations. As CT26 mutations and expression profiles resemble those of human CRC, we focused on designing a polyepitope vaccine based on CT26 neoepitopes. Due to its low immunogenicity, outer membrane vesicles (rOMV) as an antigen delivery system and adjuvant was applied. Herein, based on previous experimental and our in silico studies four CT26 neoepitopes with the ability to bind MHC-I and MHC-II, TCR, and induce IFN-α production were selected. To increase their immunogenicity, the gp70 and PADRE epitopes were added. The order of the neoepitopes was determined through 3D structure analysis using ProSA, Verify 3D, ERRAT, and Ramachandran servers. The stable peptide-protein docking between the selected epitopes and MHC alleles strengthen our prediction. The CT26 polytope vaccine sequence was fused to the C-terminal of cytolysin A (ClyA) anchor protein and rOMVs were isolated from endotoxin-free ClearColi™ strain. The results of the C-ImmSim server showed that the ClyA-CT26 polytope vaccine could induce T and B cells immunity.The ClyA-CT26 polytope was characterized as a soluble, stable, immunogen, and non-allergen vaccine and optimized for expression in ClearColi™ 24 h after induction with 1 mM IPTG at 25 °C. Western blot analysis confirmed the expression of ClyA-CT26 polytope by ClearColi™ and also on ClearColi™-derived rOMVs. In conclusion, we found that ClearColi™-derived rOMVs with CT26 polytope can deliver CRC neoantigens and induce antitumor immunity, but in vivo immunological studies are needed to confirm vaccine efficacy.
Collapse
Affiliation(s)
- Elham Sharif
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, Tehran, 1991953381, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, Tehran, 1991953381, Iran.
| |
Collapse
|
4
|
Xiang S, Khan A, Yao Q, Wang D. Recent advances in bacterial outer membrane vesicles: Effects on the immune system, mechanisms and their usage for tumor treatment. J Pharm Anal 2024; 14:101049. [PMID: 39840399 PMCID: PMC11750273 DOI: 10.1016/j.jpha.2024.101049] [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: 03/30/2024] [Revised: 06/27/2024] [Accepted: 07/19/2024] [Indexed: 01/23/2025] Open
Abstract
Tumor treatment remains a significant medical challenge, with many traditional therapies causing notable side effects. Recent research has led to the development of immunotherapy, which offers numerous advantages. Bacteria inherently possess motility, allowing them to preferentially colonize tumors and modulate the tumor immune microenvironment, thus influencing the efficacy of immunotherapy. Bacterial outer membrane vesicles (OMVs) secreted by gram-negative bacteria are nanoscale lipid bilayer structures rich in bacterial antigens, pathogen-associated molecular patterns (PAMPs), various proteins, and vesicle structures. These features allow OMVs to stimulate immune system activation, generate immune responses, and serve as efficient drug delivery vehicles. This dual capability enhances the effectiveness of immunotherapy combined with chemotherapy or phototherapy, thereby improving anticancer drug efficacy. Current research has concentrated on engineering OMVs to enhance production yield, minimize cytotoxicity, and improve the safety and efficacy of treatments. Consequently, OMVs hold great promise for applications in tumor immunotherapy, tumor vaccine development, and drug delivery. This article provides an overview of the structural composition and immune mechanisms of OMVs, details various OMVs modification strategies, and reviews the progress in using OMVs for tumor treatment and their anti-tumor mechanisms. Additionally, it discusses the challenges faced in translating OMV-based anti-tumor therapies into clinical practice, aiming to provide a comprehensive understanding of OMVs' potential for in-depth research and clinical application.
Collapse
Affiliation(s)
- Shuo Xiang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
- College of Advanced Materials Engineering, Jiaxing Nanhu University, 572 Yuexiu Road, Jiaxing, Zhejiang, 314001, China
| | - Arshad Khan
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Qiufang Yao
- College of Advanced Materials Engineering, Jiaxing Nanhu University, 572 Yuexiu Road, Jiaxing, Zhejiang, 314001, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| |
Collapse
|
5
|
Huynh DT, Nolfi E, Guleed S, Medfai L, Wolf N, Uijen RF, de Jonge MI, van Ulsen P, Dietrich J, Luirink J, Sijts AJAM, Jong WSP. Intradermal administration of novel particulate Chlamydia trachomatis vaccine candidates drives protective immune responses. Biomed Pharmacother 2024; 180:117563. [PMID: 39405914 DOI: 10.1016/j.biopha.2024.117563] [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: 06/25/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
Chlamydia trachomatis causes the most prevalent bacterial sexually transmitted infection worldwide. Its complex lifecycle and the lack of appropriate antigen delivery vehicles make it difficult to develop an effective C. trachomatis vaccine. Recently, bacterial protein bodies (PBs) have emerged as promising bioparticles for vaccine antigen delivery. By developing a PB-tag for translational fusion, we were able to induce the aggregation of recombinant antigens expressed in Escherichia coli into PBs. Here, we investigated the immunogenicity and efficacy of PBs containing either the C. trachomatis MOMP-derived CTH522-SP or HtrA antigen in mice. Intradermal administration of c-di-AMP-adjuvanted PB-CTH522-SP and PB-HtrA vaccines, produced in an LPS-detoxified E. coli strain, induced antigen-specific cellular immunity, as measured by significant release of IFN-γ and IL17a in draining cervical lymph node and splenic cell cultures. Moreover, significant induction of HtrA-specific IFN-γ expressing CD4+ and CD8+ T cells was detected in the spleens. While immunization with the two PB vaccines led to prominent levels of specific antibodies in both serum and vaginal compartments, only antiserum against PB-CTH522-SP exhibited C. trachomatis-specific neutralization activity. Importantly, intradermal immunization with PB-CTH522-SP significantly reduced bacterial counts following C. trachomatis genital challenge. These data highlight the potential of the PB-based platform for the development of C. trachomatis vaccines.
Collapse
Affiliation(s)
- Dung T Huynh
- Abera Bioscience AB, Uppsala, Sweden; Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Emanuele Nolfi
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Safia Guleed
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Lobna Medfai
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | - Rienke F Uijen
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter van Ulsen
- Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Joen Luirink
- Abera Bioscience AB, Uppsala, Sweden; Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Alice J A M Sijts
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | | |
Collapse
|
6
|
Ho MY, Liu S, Xing B. Bacteria extracellular vesicle as nanopharmaceuticals for versatile biomedical potential. NANO CONVERGENCE 2024; 11:28. [PMID: 38990415 PMCID: PMC11239649 DOI: 10.1186/s40580-024-00434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024]
Abstract
Bacteria extracellular vesicles (BEVs), characterized as the lipid bilayer membrane-surrounded nanoparticles filled with molecular cargo from parent cells, play fundamental roles in the bacteria growth and pathogenesis, as well as facilitating essential interaction between bacteria and host systems. Notably, benefiting from their unique biological functions, BEVs hold great promise as novel nanopharmaceuticals for diverse biomedical potential, attracting significant interest from both industry and academia. Typically, BEVs are evaluated as promising drug delivery platforms, on account of their intrinsic cell-targeting capability, ease of versatile cargo engineering, and capability to penetrate physiological barriers. Moreover, attributing to considerable intrinsic immunogenicity, BEVs are able to interact with the host immune system to boost immunotherapy as the novel nanovaccine against a wide range of diseases. Towards these significant directions, in this review, we elucidate the nature of BEVs and their role in activating host immune response for a better understanding of BEV-based nanopharmaceuticals' development. Additionally, we also systematically summarize recent advances in BEVs for achieving the target delivery of genetic material, therapeutic agents, and functional materials. Furthermore, vaccination strategies using BEVs are carefully covered, illustrating their flexible therapeutic potential in combating bacterial infections, viral infections, and cancer. Finally, the current hurdles and further outlook of these BEV-based nanopharmaceuticals will also be provided.
Collapse
Affiliation(s)
- Ming Yao Ho
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Bengang Xing
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore.
| |
Collapse
|
7
|
Kannan N, Choi A, Rivera De Jesus MA, Wei PM, Sahler JM, Curley SM, August A, DeLisa MP, Whittaker GR, Putnam D. Intranasal Vaccination with Recombinant TLR2-Active Outer Membrane Vesicles Containing Sequential M2e Epitopes Protects against Lethal Influenza a Challenge. Vaccines (Basel) 2024; 12:724. [PMID: 39066362 PMCID: PMC11281606 DOI: 10.3390/vaccines12070724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza is a highly contagious respiratory disease, resulting in an estimated 3 to 5 million cases of severe illness annually. While most influenza vaccines are administered parenterally via injection, one shortcoming is that they do not generate a strong immune response at the site of infection, which can become important in a pandemic. Intranasal vaccines can generate both local and systemic protective immune responses, can reduce costs, and enhance ease of administration. Previous studies showed that parenterally administered outer membrane vesicles (OMVs) that carry sequences of the M2e protein (OMV-M2e) protect against influenza A/PR8 challenge in mice and ferrets. In the current study, we measured the effectiveness of the intranasal route of the OMV-M2e vaccine against the influenza A/PR8 strain in mice. We observed high anti-M2e IgG and IgA titers post-challenge in mice vaccinated intranasally with OMV-M2e. In addition, we observed a Th1/Tc1 bias in the vaccinated mice, and an increased Th17/Tc17 response, both of which correlated with survival to A/PR8 challenge and significantly lower lung viral titers. We conclude that the intranasal-route administration of the OMV-M2e vaccine is a promising approach toward generating protection against influenza A as it leads to an increased proinflammatory immune response correlating with survival to viral challenge.
Collapse
Affiliation(s)
- Nisha Kannan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Mariela A. Rivera De Jesus
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Peter Male Wei
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Julie Marie Sahler
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Stephanie Marie Curley
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Matthew P. DeLisa
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
8
|
Mertes V, Saragliadis A, Mascherin E, Tysvær EB, Roos N, Linke D, Winther-Larsen HC. Recombinant expression of Yersinia ruckeri outer membrane proteins in Escherichia coli extracellular vesicles. Protein Expr Purif 2024; 215:106409. [PMID: 38040272 DOI: 10.1016/j.pep.2023.106409] [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: 08/25/2023] [Revised: 11/11/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023]
Abstract
The secretion of extracellular vesicles (EVs) is a common process in Gram-negative bacteria and can be exploited for biotechnological applications. EVs pose a self-adjuvanting, non-replicative vaccine platform, where membrane and antigens are presented to the host immune system in a non-infectious fashion. The secreted quantity of EVs varies between Gram-negative bacterial species and is comparatively high in the model bacterium E. coli. The outer membrane proteins OmpA and OmpF of the fish pathogen Y. ruckeri have been proposed as vaccine candidates to prevent enteric redmouth disease in aquaculture. In this work, Y.ruckeri OmpA or OmpF were expressed in E. coli and recombinant EVs were isolated. To avoid competition between endogenous E. coli OmpA or OmpF, Y. ruckeri OmpA and OmpF were expressed in E. coli strains lacking ompA, ompF, and in a quadruple knockout strain where the four major outer membrane protein genes ompA, ompC, ompF and lamB were removed. Y.ruckeri OmpA and OmpF were successfully expressed in EVs derived from the E. coli mutants as verified by SDS-PAGE, heat modifiability and proteomic analysis using mass-spectrometry. Transmission electron microscopy revealed the presence of EVs in all E. coli strains, and increased EV concentrations were detected when expressing Y. ruckeri OmpA or OmpF in recombinant EVs compared to empty vector controls as verified by nanoparticle tracking analysis. These results show that E. coli can be utilized as a vector for production of EVs expressing outer membrane antigens from Y. ruckeri.
Collapse
Affiliation(s)
- Verena Mertes
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Athanasios Saragliadis
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, Norway
| | - Elisa Mascherin
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Italy
| | - Ellen-Beate Tysvær
- Section for Physiology and Cell Biology, Department of Biosciences, University of Oslo, Norway
| | - Norbert Roos
- Section for Physiology and Cell Biology, Department of Biosciences, University of Oslo, Norway
| | - Dirk Linke
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, Norway
| | - Hanne C Winther-Larsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway.
| |
Collapse
|
9
|
Effah CY, Ding X, Drokow EK, Li X, Tong R, Sun T. Bacteria-derived extracellular vesicles: endogenous roles, therapeutic potentials and their biomimetics for the treatment and prevention of sepsis. Front Immunol 2024; 15:1296061. [PMID: 38420121 PMCID: PMC10899385 DOI: 10.3389/fimmu.2024.1296061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Sepsis is one of the medical conditions with a high mortality rate and lacks specific treatment despite several years of extensive research. Bacterial extracellular vesicles (bEVs) are emerging as a focal target in the pathophysiology and treatment of sepsis. Extracellular vesicles (EVs) derived from pathogenic microorganisms carry pathogenic factors such as carbohydrates, proteins, lipids, nucleic acids, and virulence factors and are regarded as "long-range weapons" to trigger an inflammatory response. In particular, the small size of bEVs can cross the blood-brain and placental barriers that are difficult for pathogens to cross, deliver pathogenic agents to host cells, activate the host immune system, and possibly accelerate the bacterial infection process and subsequent sepsis. Over the years, research into host-derived EVs has increased, leading to breakthroughs in cancer and sepsis treatments. However, related approaches to the role and use of bacterial-derived EVs are still rare in the treatment of sepsis. Herein, this review looked at the dual nature of bEVs in sepsis by highlighting their inherent functions and emphasizing their therapeutic characteristics and potential. Various biomimetics of bEVs for the treatment and prevention of sepsis have also been reviewed. Finally, the latest progress and various obstacles in the clinical application of bEVs have been highlighted.
Collapse
Affiliation(s)
- Clement Yaw Effah
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Xianfei Ding
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Emmanuel Kwateng Drokow
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Biostatistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiang Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Ran Tong
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Tongwen Sun
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| |
Collapse
|
10
|
Weyant KB, Oloyede A, DeLisa MP. On-Demand Vaccine Production via Dock-and-Display of Biotinylated Antigens on Bacterial Extracellular Vesicles. Methods Mol Biol 2024; 2843:195-216. [PMID: 39141302 DOI: 10.1007/978-1-0716-4055-5_13] [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] [Indexed: 08/15/2024]
Abstract
Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising vaccine technology for developing immunity against diverse pathogens. However, antigen display on OMVs can be challenging to control and highly variable due to bottlenecks in protein expression and localization to the bacterial host cell's outer membrane, especially for bulky and complex antigens. Here, we describe methods related to a universal vaccine technology called AvidVax (avidin-based vaccine antigen crosslinking) for rapid and simplified assembly of antigens on the exterior of OMVs during vaccine development. The AvidVax platform involves remodeling the OMV surface with multiple copies of a synthetic antigen-binding protein (SNAP), which is an engineered fusion protein comprised of an outer membrane scaffold protein linked to a biotin-binding protein. The resulting SNAPs enable efficient decoration of OMVs with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, nucleic acids, and short peptides. We detail the key steps in the AvidVax vaccine production pipeline including preparation and isolation of SNAP-OMVs, biotinylation and enrichment of vaccine antigens, and formulation and characterization of antigen-loaded SNAP-OMVs.
Collapse
Affiliation(s)
| | - Ayomide Oloyede
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Hall, Ithaca, NY, USA
| | - Matthew P DeLisa
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Hall, Ithaca, NY, USA.
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
11
|
Xie J, Li Q, Nie S. Bacterial extracellular vesicles: An emerging postbiotic. Trends Food Sci Technol 2024; 143:104275. [DOI: 10.1016/j.tifs.2023.104275] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Yang YS, Chen HJ, Chen XC, Tang HJ, Chang FJ, Huang YL, Pan YL, Kesavan DK, Chen HY, Shang HS, Kuo SC, Chen TL, Chiang MH. Elizabethkingia anophelis outer membrane vesicles as a novel vaccine candidate against infection: insights into immune response and potential for passive immunity. mSphere 2023; 8:e0040023. [PMID: 38014949 PMCID: PMC10732079 DOI: 10.1128/msphere.00400-23] [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: 07/17/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Elizabethkingia anophelis, a Gram-negative pathogen, causes infections such as bacteraemia, pneumonia, and neonatal meningitis. The pathogen resists most antimicrobial classes, making novel approaches urgently needed. In natural settings, Gram-negative bacteria secrete outer membrane vesicles (OMVs) that carry important molecules in the bacterial life cycle. These OMVs are enriched with proteins involved in virulence, survival, and carbohydrate metabolism, making them a promising source for vaccine development against the pathogen. This study investigated the efficacy of imipenem-induced OMVs (iOMVs) as a vaccine candidate against E. anophelis infection in a mouse pneumonia model. Mice immunized with iOMVs were completely protected during lethal-dose challenges. Passive immunization with hyperimmune sera and splenocytes conferred protection against lethal pneumonia. Further investigation is needed to understand the mechanisms underlying the protective effects of iOMV-induced passive immunity, such as the action on specific antibody subclasses or T cell subsets.
Collapse
Grants
- 109-2320-B-016-002-MY2, 110-2320-B-016-014, 111-2320-B-016-015, 112-2314-B-016-023, 112-2314-B-016-039, 112-2314-B-016-024-MY2 Ministry of Science and Technology, Taiwan (MOST)
- TSGH-E-111244, TSGH-E-112253 Tri-Service General Hospital (TSGH)
- CMNDMC11108, CMNDMC11206 Chi Mei Medical Center
- MND-MAB-110-049, MND-MAB-D-111072, MND-MAB-D-112115, MND-MAB-D-113078 MOD | Medical Affairs Bureau (MAB)
Collapse
Affiliation(s)
- Ya-Sung Yang
- Department of Internal Medicine, Division of Infectious Diseases and Tropical Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jui Chen
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Xiao-Chun Chen
- Department and Graduate institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Fang-Ju Chang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Ling Huang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Dinesh Kumar Kesavan
- School of Material Science, Nanyang Technological University, Singapore, Singapore
| | - Huan-Yuan Chen
- Inflammation Core Facility, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Sheng Shang
- Department of Pathology, Division of Clinical Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
13
|
Mi J, He T, Hu X, Wang Z, Wang T, Qi X, Li K, Gao L, Liu C, Zhang Y, Wang S, Qiu Y, Liu Z, Song J, Wang X, Gao Y, Cui H. Enterococcus faecium C171: Modulating the Immune Response to Acute Lethal Viral Challenge. Int J Antimicrob Agents 2023; 62:106969. [PMID: 37758064 DOI: 10.1016/j.ijantimicag.2023.106969] [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: 01/31/2023] [Revised: 08/08/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Commensal bacteria modulate acute immune responses to infection in hosts. In this study, Enterococcus faecium C171 was screened and isolated. This strain has similar basic characteristics to the reference probiotic, including strong anti-inflammatory and anti-infective effects. E. faecium C171 inhibits the production of pro-Caspase-1 and significantly reduces the production of interleukin-1β (IL-1β) in vitro. These reactions were confirmed using the Transwell system. Live E. faecium C171 mainly exerted an inhibitory effect on acute inflammation, whereas the anti-infective and immune-activating effects were primarily mediated by the E. faecium C171-produced bacterial extracellular vesicles (Efm-C171-BEVs). Furthermore, in the specific pathogen-free (SPF) chicken model, oral administration of E. faecium C171 increased the relative abundance of beneficial microbiota (Enterococcus and Lactobacillus), particularly Enterococcus, the most important functional bacteria of the gut microbiota. E. faecium C171 significantly inhibited the acute inflammatory response induced by a highly virulent infectious disease, and reduced mortality in SPF chickens by 75%. In addition, E. faecium C171 induced high levels of CD3+, CD4-, and CD8- immunoregulatory cells and CD8+ killer T cells, and significantly improved the proliferative activity of T cells in peripheral blood mononuclear cells, and the secretion of interferon-γ. These findings indicate that E. faecium C171 and Efm-C171-BEVs are promising candidates for adjuvant treatment of acute inflammatory diseases and acute viral infections.
Collapse
Affiliation(s)
- Jielan Mi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Tana He
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xinyun Hu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Zhihao Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Tingting Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xiaole Qi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Kai Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Li Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Changjun Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yanping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Suyan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yu Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Zengqi Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Jie Song
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xiaomei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yulong Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Hongyu Cui
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
14
|
Zong R, Ruan H, Liu C, Fan S, Li J. Bacteria and Bacterial Components as Natural Bio-Nanocarriers for Drug and Gene Delivery Systems in Cancer Therapy. Pharmaceutics 2023; 15:2490. [PMID: 37896250 PMCID: PMC10610331 DOI: 10.3390/pharmaceutics15102490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteria and bacterial components possess multifunctional properties, making them attractive natural bio-nanocarriers for cancer diagnosis and targeted treatment. The inherent tropic and motile nature of bacteria allows them to grow and colonize in hypoxic tumor microenvironments more readily than conventional therapeutic agents and other nanomedicines. However, concerns over biosafety, limited antitumor efficiency, and unclear tumor-targeting mechanisms have restricted the clinical translation and application of natural bio-nanocarriers based on bacteria and bacterial components. Fortunately, bacterial therapies combined with engineering strategies and nanotechnology may be able to reverse a number of challenges for bacterial/bacterial component-based cancer biotherapies. Meanwhile, the combined strategies tend to enhance the versatility of bionanoplasmic nanoplatforms to improve biosafety and inhibit tumorigenesis and metastasis. This review summarizes the advantages and challenges of bacteria and bacterial components in cancer therapy, outlines combinatorial strategies for nanocarriers and bacterial/bacterial components, and discusses their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Shaohua Fan
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
15
|
Lamontagne F, Arpin D, Côté‐Cyr M, Khatri V, St‐Louis P, Gauthier L, Archambault D, Bourgault S. Engineered Curli Nanofilaments as a Self-Adjuvanted Antigen Delivery Platform. Adv Healthc Mater 2023; 12:e2300224. [PMID: 37031161 PMCID: PMC11468023 DOI: 10.1002/adhm.202300224] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/22/2023] [Indexed: 04/10/2023]
Abstract
Proteinaceous nanoparticles constitute efficient antigen delivery systems in vaccine formulations due to their size and repetitive nature that mimic most invading pathogens and promote immune activation. Nonetheless, the coadministration of an adjuvant with subunit nanovaccines is usually required to induce a robust, long-lasting, and protective immune response. Herein, the protein Curli-specific gene A (CsgA), which is known to self-assemble into nanofilaments contributing to bacterial biofilm, is exploited to engineer an intrinsically immunostimulatory antigen delivery platform. Three repeats of the M2e antigenic sequence from the influenza A virus matrix 2 protein are merged to the N-terminal domain of engineered CsgA proteins. These chimeric 3M2e-CsgA spontaneously self-assemble into antigen-displaying cross-β-sheet nanofilaments that activate the heterodimeric toll-like receptors 2 and 1. The resulting nanofilaments are avidly internalized by antigen-presenting cells and stimulate the maturation of dendritic cells. Without the need of any additional adjuvants, both assemblies show robust humoral and cellular immune responses, which translate into complete protection against a lethal experimental infection with the H1N1 influenza virus. Notably, these CsgA-based nanovaccines induce neither overt systemic inflammation, nor reactogenicity, upon mice inoculation. These results highlight the potential of engineered CsgA nanostructures as self-adjuvanted, safe, and versatile antigen delivery systems to fight infectious diseases.
Collapse
Affiliation(s)
- Félix Lamontagne
- Department of ChemistryUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
- Quebec Network for Research on Protein FunctionEngineering and Applications (PROTEO)QuebecH3C 3P8Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Dominic Arpin
- Department of ChemistryUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
- Quebec Network for Research on Protein FunctionEngineering and Applications (PROTEO)QuebecH3C 3P8Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Mélanie Côté‐Cyr
- Department of ChemistryUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
- Quebec Network for Research on Protein FunctionEngineering and Applications (PROTEO)QuebecH3C 3P8Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Vinay Khatri
- Department of ChemistryUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
- Quebec Network for Research on Protein FunctionEngineering and Applications (PROTEO)QuebecH3C 3P8Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
| | - Philippe St‐Louis
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Laurie Gauthier
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Denis Archambault
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
- Department of Biological SciencesUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
| | - Steve Bourgault
- Department of ChemistryUniversité du Québec à MontréalC.P.8888, Succursale Centre‐VilleMontrealH3C 3P8Canada
- Quebec Network for Research on Protein FunctionEngineering and Applications (PROTEO)QuebecH3C 3P8Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA)Saint‐HyacintheJ2S 2M2Canada
- The Center of Excellence in Research on Orphan Diseases – Fondation Courtois (CERMO‐FC)MontrealH3C 3P8Canada
| |
Collapse
|
16
|
Park KS, Svennerholm K, Crescitelli R, Lässer C, Gribonika I, Andersson M, Boström J, Alalam H, Harandi AM, Farewell A, Lötvall J. Detoxified synthetic bacterial membrane vesicles as a vaccine platform against bacteria and SARS-CoV-2. J Nanobiotechnology 2023; 21:156. [PMID: 37208676 DOI: 10.1186/s12951-023-01928-w] [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: 01/27/2023] [Accepted: 05/13/2023] [Indexed: 05/21/2023] Open
Abstract
The development of vaccines based on outer membrane vesicles (OMV) that naturally bud off from bacteria is an evolving field in infectious diseases. However, the inherent inflammatory nature of OMV limits their use as human vaccines. This study employed an engineered vesicle technology to develop synthetic bacterial vesicles (SyBV) that activate the immune system without the severe immunotoxicity of OMV. SyBV were generated from bacterial membranes through treatment with detergent and ionic stress. SyBV induced less inflammatory responses in macrophages and in mice compared to natural OMV. Immunization with SyBV or OMV induced comparable antigen-specific adaptive immunity. Specifically, immunization with Pseudomonas aeruginosa-derived SyBV protected mice against bacterial challenge, and this was accompanied by significant reduction in lung cell infiltration and inflammatory cytokines. Further, immunization with Escherichia coli-derived SyBV protected mice against E. coli sepsis, comparable to OMV-immunized group. The protective activity of SyBV was driven by the stimulation of B-cell and T-cell immunity. Also, SyBV were engineered to display the SARS-CoV-2 S1 protein on their surface, and these vesicles induced specific S1 protein antibody and T-cell responses. Collectively, these results demonstrate that SyBV may be a safe and efficient vaccine platform for the prevention of bacterial and viral infections.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Kristina Svennerholm
- Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rossella Crescitelli
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mickael Andersson
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Boström
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Alalam
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- BC Children's Hospital Research Institute, Vaccine Evaluation Center, University of British Columbia, Columbia, Canada
| | - Anne Farewell
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
17
|
Krishnan N, Peng FX, Mohapatra A, Fang RH, Zhang L. Genetically engineered cellular nanoparticles for biomedical applications. Biomaterials 2023; 296:122065. [PMID: 36841215 PMCID: PMC10542936 DOI: 10.1016/j.biomaterials.2023.122065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
In recent years, nanoparticles derived from cellular membranes have been increasingly explored for the prevention and treatment of human disease. With their flexible design and ability to interface effectively with the surrounding environment, these biomimetic nanoparticles can outperform their traditional synthetic counterparts. As their popularity has increased, researchers have developed novel ways to modify the nanoparticle surface to introduce new or enhanced capabilities. Moving beyond naturally occurring materials derived from wild-type cells, genetic manipulation has proven to be a robust and flexible method by which nanoformulations with augmented functionalities can be generated. In this review, an overview of genetic engineering approaches to express novel surface proteins is provided, followed by a discussion on the various biomedical applications of genetically modified cellular nanoparticles.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Fei-Xing Peng
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Animesh Mohapatra
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
18
|
Markthaler D, Ghosh R. Computational prediction of extracellular loops of the Por39 outer membrane porin of Rhodospirillum rubrum suitable for epitope surface display. Comput Struct Biotechnol J 2023; 21:2483-2494. [PMID: 37077176 PMCID: PMC10106341 DOI: 10.1016/j.csbj.2023.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Outer membrane porins from Gram-negative bacteria are established vehicles for the production of vaccines. Typically, one or more of the extracellular loops of a porin are replaced by a peptide encoding a foreign epitope, and recombinant porin is then used as a vaccine. However, many host strains are potentially pathogenic, and also produce toxic lipopolysaccharide (LPS), both of which are undesirable for safety reasons. In contrast, the outer membrane porins from photosynthetic, purple bacteria have no known human pathology and produce only weakly toxic LPS. The purple bacterium Rhodospirillum rubrum is well-suited for large-scale biotechnology, and expresses a major porin, Por39, which is a candidate for a vaccine platform. Unfortunately, the atomic structure of Por39 could not be determined so far, and Por39 shows only a weak homology to other porins of known structure, making the assignment of external loops difficult. Here, we construct a knowledge-based model of Por39 using secondary structure constraints from both the low sequence homology to the 2POR porin from Rhodobacter capsulatus, for which the X-ray structure is known, as well as those obtained using secondary structure prediction packages. The secondary structure predictions were used to constrain a three-dimensional model created using the I-TASSER package. The modelling procedure was validated by predicting the structure of 2POR using the same strategy, but excluding the 2POR X-ray structure from the I-TASSER database. The final Por39 model allows three external loops to be defined precisely, and could also be used to obtain an initial model for the closely related Por41 using molecular modelling. These structures provide a good starting point for the insertion of epitopes with vaccine potential.
Collapse
|
19
|
Pordanjani PM, Bolhassani A, Milani A, Pouriayevali MH. Extracellular vesicles in vaccine development and therapeutic approaches for viral diseases. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
20
|
A modular vaccine platform enabled by decoration of bacterial outer membrane vesicles with biotinylated antigens. Nat Commun 2023; 14:464. [PMID: 36709333 PMCID: PMC9883832 DOI: 10.1038/s41467-023-36101-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/15/2023] [Indexed: 01/29/2023] Open
Abstract
Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising technology for the creation of non-infectious, nanoparticle vaccines against diverse pathogens. However, antigen display on OMVs can be difficult to control and highly variable due to bottlenecks in protein expression and localization to the outer membrane of the host cell, especially for bulky and/or complex antigens. Here, we describe a universal approach for avidin-based vaccine antigen crosslinking (AvidVax) whereby biotinylated antigens are linked to the exterior of OMVs whose surfaces are remodeled with multiple copies of a synthetic antigen-binding protein (SNAP) comprised of an outer membrane scaffold protein fused to a biotin-binding protein. We show that SNAP-OMVs can be readily decorated with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, and short peptides. When the resulting OMV formulations are injected in mice, strong antigen-specific antibody responses are observed that depend on the physical coupling between the antigen and SNAP-OMV delivery vehicle. Overall, these results demonstrate AvidVax as a modular platform that enables rapid and simplified assembly of antigen-studded OMVs for application as vaccines against pathogenic threats.
Collapse
|
21
|
Wo J, Lv ZY, Sun JN, Tang H, Qi N, Ye BC. Engineering probiotic-derived outer membrane vesicles as functional vaccine carriers to enhance immunity against SARS-CoV-2. iScience 2022; 26:105772. [PMID: 36510593 PMCID: PMC9729586 DOI: 10.1016/j.isci.2022.105772] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Because of the continued emergence of SARS-CoV-2 variants, there has been considerable interest in how to display multivalent antigens efficiently. Bacterial outer membrane vesicles (OMVs) can serve as an attractive vaccine delivery system because of their self-adjuvant properties and the ability to be decorated with antigens. Here we set up a bivalent antigen display platform based on engineered OMVs using mCherry and GFP and demonstrated that two different antigens of SARS-CoV-2 could be presented simultaneously in the lumen and on the surface of OMVs. Comparing immunogenicity, ClyA-NG06 fusion and the receptor-binding domain (RBD) of the spike protein in the OMV lumen elicited a stronger humoral response in mice than OMVs presenting either the ClyA-NG06 fusion or RBD alone. Taken together, we provided an efficient approach to display SARS-CoV-2 antigens in the lumen and on the surface of the same OMV and highlighted the potential of OMVs as general multi-antigen carriers.
Collapse
Affiliation(s)
- Jing Wo
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Zhao-Yong Lv
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Jia-Nan Sun
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Hao Tang
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Nan Qi
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Bang-Ce Ye
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China,Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China,Corresponding author
| |
Collapse
|
22
|
Piccioli D, Alfini R, Monaci V, Arato V, Carducci M, Aruta MG, Rossi O, Necchi F, Anemona A, Bartolini E, Micoli F. Antigen presentation by Follicular Dendritic cells to cognate B cells is pivotal for Generalised Modules for Membrane Antigens (GMMA) immunogenicity. Vaccine 2022; 40:6305-6314. [PMID: 36137901 DOI: 10.1016/j.vaccine.2022.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 01/27/2023]
Abstract
GMMA has been proposed as a potent technology platform for the design of safe, effective and affordable vaccines. As GMMA are vesicles blebbing out of the outer membrane of Gram-negative bacteria, they contain lipopolysaccharides, lipoproteins and peptidoglycans that stimulate immune cells via Toll-like Receptors 4 (TLR4) or TLR2. Being basically nanoparticles, GMMA can be efficiently captured by Follicular Dendritic Cells (FDC) for antigen presentation to cognate B cells. GMMA have shown to be highly immunogenic in preclinical and clinical studies and the engagement of TLR4 and TLR2 or antigen presentation by FDC may have a prominent role in GMMA immunogenicity, which is well worth investigating. By using GMMA derived from Shigella sonnei and Salmonella Typhimurium, we show for the first time that the antigen presentation by FDC to cognate B cells plays a major role in the induction of an effective humoral immune response upon immunization with GMMA by using both models. The engagement of TLR4 is critical to elicit an optimal antibody production, but its effect on antibody functionality is dependent on GMMA type and is dispensable when immunizing with Alum adjuvant, whereas TLR2 does not have any role for GMMA immunogenicity. Our findings represent a substantial advancement of the knowledge on GMMA mode of action and shed a light on novel perspectives for the design of safer and more effective GMMA-based vaccines. ONE SENTENCE SUMMARY: The study demonstrated that the antigen presentation by FDC to cognate B cells plays a major role for GMMA immunogenicity.
Collapse
Affiliation(s)
| | - Renzo Alfini
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | - Vanessa Arato
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | | | - Omar Rossi
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | | |
Collapse
|
23
|
Spindler MP, Siu S, Mogno I, Li Z, Yang C, Mehandru S, Britton GJ, Faith JJ. Human gut microbiota stimulate defined innate immune responses that vary from phylum to strain. Cell Host Microbe 2022; 30:1481-1498.e5. [PMID: 36099923 PMCID: PMC9588646 DOI: 10.1016/j.chom.2022.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/10/2022] [Accepted: 08/15/2022] [Indexed: 11/03/2022]
Abstract
The potential of commensal bacteria to modulate host immunity remains largely uncharacterized, largely due to the vast number of strains that comprise the human gut microbiota. We have developed a screening platform to measure the innate immune responses of myeloid cells to 277 bacterial strains isolated from the gut microbiota of healthy individuals and those with inflammatory bowel diseases. The innate immune responses to gut-derived bacteria are as strong as those toward pathogenic bacteria, and they vary from phylum to strain. Myeloid cells differentially rely upon innate receptors TLR2 or TLR4 to sense taxa, with differential sensing of Bacteroidetes and Proteobacteria that predict in vivo functions. These innate immune responses can be modeled using combinations of up to 8 Toll-like receptor (TLR) agonists. Furthermore, the immunogenicity of strains is stable over time and following fecal microbiota transplantation into new human recipients. Collectively, this high-throughput approach provides an insight into how commensal microorganisms shape innate immune phenotypes.
Collapse
Affiliation(s)
- Matthew P Spindler
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sophia Siu
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilaria Mogno
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhihua Li
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chao Yang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Dr. Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Graham J Britton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
24
|
Mat Rani NNI, Alzubaidi ZM, Butt AM, Mohammad Faizal NDF, Sekar M, Azhari H, Mohd Amin MCI. Outer membrane vesicles as biomimetic vaccine carriers against infections and cancers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1784. [PMID: 35194964 DOI: 10.1002/wnan.1784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
In the last decade, nanoparticle-based therapeutic modalities have emerged as promising treatment options for cancer and infectious diseases. To improve prognosis, chemotherapeutic and antimicrobial drugs must be delivered selectively to the target sites. Researchers have increasingly focused their efforts on improving drug delivery, with a particular emphasis on cancer and infectious diseases. When drugs are administered systemically, they become diluted and can diffuse to all tissues but only until the immune system intervenes and quickly removes them from circulation. To enhance and prolong the systemic circulation of drugs, nanocarriers have been explored and used; however, nanocarriers have a major drawback in that they can trigger immune responses. Numerous nanocarriers for optimal drug delivery have been developed using innovative and effective biointerface technologies. Autologous cell-derived drug carriers, such as outer membrane vesicles (OMVs), have demonstrated improved bioavailability and reduced toxicity. Thus, this study investigates the use of biomimetic OMVs as biomimetic vaccine carriers against infections and cancers to improve our understanding in the field of nanotechnology. In addition, discussion on the advantages, disadvantages, and future prospects of OMVs will also be explored. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Nur Najihah Izzati Mat Rani
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, Malaysia
| | - Zahraa M Alzubaidi
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Adeel Masood Butt
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Nur Dini Fatini Mohammad Faizal
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, Malaysia
| | - Hanisah Azhari
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Long Q, Zheng P, Zheng X, Li W, Hua L, Yang Z, Huang W, Ma Y. Engineered bacterial membrane vesicles are promising carriers for vaccine design and tumor immunotherapy. Adv Drug Deliv Rev 2022; 186:114321. [PMID: 35533789 DOI: 10.1016/j.addr.2022.114321] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Bacterial membrane vesicles (BMVs) have emerged as novel and promising platforms for the development of vaccines and immunotherapeutic strategies against infectious and noninfectious diseases. The rich microbe-associated molecular patterns (MAMPs) and nanoscale membrane vesicle structure of BMVs make them highly immunogenic. In addition, BMVs can be endowed with more functions via genetic and chemical modifications. This article reviews the immunological characteristics and effects of BMVs, techniques for BMV production and modification, and the applications of BMVs as vaccines or vaccine carriers. In summary, given their versatile characteristics and immunomodulatory properties, BMVs can be used for clinical vaccine or immunotherapy applications.
Collapse
|
26
|
Yang Z, Hua L, Yang M, Li W, Ren Z, Zheng X, Chen H, Long Q, Bai H, Huang W, Ma Y. Polymerized porin as a novel delivery platform for coronavirus vaccine. J Nanobiotechnology 2022; 20:260. [PMID: 35672856 PMCID: PMC9171476 DOI: 10.1186/s12951-022-01469-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/20/2022] [Indexed: 11/10/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), seriously threatens human life and health. The correct folding and polymerization of the receptor-binding domain (RBD) protein of coronavirus in Escherichia coli may reduce the cost of SARS-CoV-2 vaccines. In this study, we constructed this nanopore by using the principle of ClyA porin polymerization triggered by the cell membrane. We used surfactants to "pick" the ClyA-RBD nanopore from the bacterial outer membrane. More importantly, the polymerized RBD displayed on the ClyA-RBD polymerized porin (RBD-PP) already displays some correct spatial conformational epitopes that can induce neutralizing antibodies. The nanostructures of RBD-PP can target lymph nodes and promote antigen uptake and processing by dendritic cells, thereby effectively eliciting the production of anti-SARS-CoV-2 neutralizing antibodies, systemic cellular immune responses, and memory T cells. We applied this PP-based vaccine platform to fabricate an RBD-based subunit vaccine against SARS-CoV-2, which will provide a foundation for the development of inexpensive coronavirus vaccines. The development of a novel vaccine delivery system is an important part of innovative drug research. This novel PP-based vaccine platform is likely to have additional applications, including other viral vaccines, bacterial vaccines, tumor vaccines, drug delivery, and disease diagnosis.
Collapse
Affiliation(s)
- Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan University, Kunming, 650091, China
| | - Mengli Yang
- National Kunming High-Level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Zhaoling Ren
- The Second Affiliated Hospital of Kunming Medical University, Kunming, 650033, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan University, Kunming, 650091, China
| | - Haoqian Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan Minzu University, Kunming, 650504, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China.
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China.
| |
Collapse
|
27
|
Krishnan N, Kubiatowicz LJ, Holay M, Zhou J, Fang RH, Zhang L. Bacterial membrane vesicles for vaccine applications. Adv Drug Deliv Rev 2022; 185:114294. [PMID: 35436569 DOI: 10.1016/j.addr.2022.114294] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/13/2022] [Accepted: 04/10/2022] [Indexed: 12/11/2022]
Abstract
Vaccines have been highly successful in the management of many diseases. However, there are still numerous illnesses, both infectious and noncommunicable, for which there are no clinically approved vaccine formulations. While there are unique difficulties that must be overcome in the case of each specific disease, there are also a number of common challenges that have to be addressed for effective vaccine development. In recent years, bacterial membrane vesicles (BMVs) have received increased attention as a potent and versatile vaccine platform. BMVs are inherently immunostimulatory and are able to activate both innate and adaptive immune responses. Additionally, BMVs can be readily taken up and processed by immune cells due to their nanoscale size. Finally, BMVs can be modified in a variety of ways, including by genetic engineering, cargo loading, and nanoparticle coating, in order to create multifunctional platforms that can be leveraged against different diseases. Here, an overview of the interactions between BMVs and immune cells is provided, followed by discussion on the applications of BMV vaccine nanotechnology against bacterial infections, viral infections, and cancers.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Luke J Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
28
|
The tremendous biomedical potential of bacterial extracellular vesicles. Trends Biotechnol 2022; 40:1173-1194. [DOI: 10.1016/j.tibtech.2022.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022]
|
29
|
Liu S, Wu X, Chandra S, Lyon C, Ning B, jiang L, Fan J, Hu TY. Extracellular vesicles: Emerging tools as therapeutic agent carriers. Acta Pharm Sin B 2022; 12:3822-3842. [PMID: 36213541 PMCID: PMC9532556 DOI: 10.1016/j.apsb.2022.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are secreted by both eukaryotes and prokaryotes, and are present in all biological fluids of vertebrates, where they transfer DNA, RNA, proteins, lipids, and metabolites from donor to recipient cells in cell-to-cell communication. Some EV components can also indicate the type and biological status of their parent cells and serve as diagnostic targets for liquid biopsy. EVs can also natively carry or be modified to contain therapeutic agents (e.g., nucleic acids, proteins, polysaccharides, and small molecules) by physical, chemical, or bioengineering strategies. Due to their excellent biocompatibility and stability, EVs are ideal nanocarriers for bioactive ingredients to induce signal transduction, immunoregulation, or other therapeutic effects, which can be targeted to specific cell types. Herein, we review EV classification, intercellular communication, isolation, and characterization strategies as they apply to EV therapeutics. This review focuses on recent advances in EV applications as therapeutic carriers from in vitro research towards in vivo animal models and early clinical applications, using representative examples in the fields of cancer chemotherapeutic drug, cancer vaccine, infectious disease vaccines, regenerative medicine and gene therapy. Finally, we discuss current challenges for EV therapeutics and their future development.
Collapse
|
30
|
Curley SM, Putnam D. Biological Nanoparticles in Vaccine Development. Front Bioeng Biotechnol 2022; 10:867119. [PMID: 35402394 PMCID: PMC8984165 DOI: 10.3389/fbioe.2022.867119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Vaccines represent one of the most successful public health initiatives worldwide. However, despite the vast number of highly effective vaccines, some infectious diseases still do not have vaccines available. New technologies are needed to fully realize the potential of vaccine development for both emerging infectious diseases and diseases for which there are currently no vaccines available. As can be seen by the success of the COVID-19 mRNA vaccines, nanoscale platforms are promising delivery vectors for effective and safe vaccines. Synthetic nanoscale platforms, including liposomes and inorganic nanoparticles and microparticles, have many advantages in the vaccine market, but often require multiple doses and addition of artificial adjuvants, such as aluminum hydroxide. Biologically derived nanoparticles, on the other hand, contain native pathogen-associated molecular patterns (PAMPs), which can reduce the need for artificial adjuvants. Biological nanoparticles can be engineered to have many additional useful properties, including biodegradability, biocompatibility, and are often able to self-assemble, thereby allowing simple scale-up from benchtop to large-scale manufacturing. This review summarizes the state of the art in biologically derived nanoparticles and their capabilities as novel vaccine platforms.
Collapse
Affiliation(s)
- Stephanie M. Curley
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| |
Collapse
|
31
|
Thomas SC, Kim JW, Pauletti GM, Hassett DJ, Kotagiri N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front Bioeng Biotechnol 2022; 9:808614. [PMID: 35096795 PMCID: PMC8790084 DOI: 10.3389/fbioe.2021.808614] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are natural cell-derived nanovesicles of endocytic origin that enable cellular crosstalk by transferring encapsulated molecular cargos across biological barriers, thereby holding significantly complex implications in the etiology and progression of diverse disease states. Consequently, the development of exosomes-based nano-theranostic strategies has received immense consideration for advancing therapeutic interventions and disease prognosis. Their favorable biopharmaceutical properties make exosomes a unique nanoparticulate carrier for pharmaceutical drug delivery. This review provides an update on the contemporary strategies utilizing exosomes for theranostic applications in nanomedicine. In addition, we provide a synopsis of exosomal features and insights into strategic modifications that control in vivo biodistribution. We further discuss their opportunities, merits and pitfalls for cell/tissue targeted drug delivery in personalized nanotherapy.
Collapse
Affiliation(s)
- Shindu C Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Giovanni M Pauletti
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, United States
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
32
|
ClearColi as a platform for untagged pneumococcal surface protein A production: cultivation strategy, bioreactor culture, and purification. Appl Microbiol Biotechnol 2022; 106:1011-1029. [PMID: 35024919 PMCID: PMC8755982 DOI: 10.1007/s00253-022-11758-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 11/27/2022]
Abstract
Abstract
Several studies have searched for new antigens to produce pneumococcal vaccines that are more effective and could provide broader coverage, given the great number of serotypes causing pneumococcal diseases. One of the promising subunit vaccine candidates is untagged recombinant pneumococcal surface protein A (PspA4Pro), obtainable in high quantities using recombinant Escherichia coli as a microbial factory. However, lipopolysaccharides (LPS) present in E. coli cell extracts must be removed, in order to obtain the target protein at the required purity, which makes the downstream process more complex and expensive. Endotoxin-free E. coli strains, which synthesize a nontoxic mutant LPS, may offer a cost-effective alternative way to produce recombinant proteins for application as therapeutics. This paper presents an investigation of PspA4Pro production employing the endotoxin-free recombinant strain ClearColi® BL21(DE3) with different media (defined, auto-induction, and other complex media), temperatures (27, 32, and 37 °C), and inducers. In comparison to conventional E. coli cells in a defined medium, ClearColi presented similar PspA4Pro yields, with lower productivities. Complex medium formulations supplemented with salts favored PspA4Pro yields, titers, and ClearColi growth rates. Induction with isopropyl-β-d-thiogalactopyranoside (0.5 mM) and lactose (2.5 g/L) together in a defined medium at 32 °C, which appeared to be a promising cultivation strategy, was reproduced in 5 L bioreactor culture, leading to a yield of 146.0 mg PspA4Pro/g dry cell weight. After purification, the cell extract generated from ClearColi led to 98% purity PspA4Pro, which maintained secondary structure and biological function. ClearColi is a potential host for industrial recombinant protein production. Key points • ClearColi can produce as much PspA4Pro as conventional E. coli BL21(DE3) cells. • 10.5 g PspA4Pro produced in ClearColi bioreactor culture using a defined medium. • Functional PspA4Pro (98% of purity) was obtained in ClearColi bioreactor culture.Graphical abstract ![]() Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-11758-9.
Collapse
|
33
|
Xue K, Wang L, Liu J. Bacterial outer membrane vesicles and their functionalization as vehicles for bioimaging, diagnosis and therapy. MATERIALS ADVANCES 2022; 3:7185-7197. [DOI: 10.1039/d2ma00420h] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
In this review, we summarize the bioactivities of bacterial outer membrane vesicles, including biogenesis, immunogenicity, and interactions, followed by a discussion on their functionalization as nanocarriers for bioimaging, diagnosis, and therapy.
Collapse
Affiliation(s)
- Kaikai Xue
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
34
|
Shkair L, Garanina EE, Martynova EV, Kolesnikova AI, Arkhipova SS, Titova AA, Rizvanov AA, Khaiboullina SF. Immunogenic Properties of MVs Containing Structural Hantaviral Proteins: An Original Study. Pharmaceutics 2022; 14:pharmaceutics14010093. [PMID: 35056989 PMCID: PMC8779827 DOI: 10.3390/pharmaceutics14010093] [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: 11/10/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/01/2023] Open
Abstract
Hemorrhagic fever with renal syndrome (HFRS) is an emerging infectious disease that remains a global public health threat. The highest incidence rate is among zoonotic disease cases in Russia. Most cases of HFRS are reported in the Volga region of Russia, which commonly identifies the Puumala virus (PUUV) as a pathogen. HFRS management is especially challenging due to the lack of specific treatments and vaccines. This study aims to develop new approaches for HFRS prevention. Our goal is to test the efficacy of microvesicles (MVs) as PUUV nucleocapsid (N) and glycoproteins (Gn/Gc) delivery vehicles. Our findings show that MVs could deliver the PUUV N and Gn/Gc proteins in vitro. We have also demonstrated that MVs loaded with PUUV proteins could elicit a specific humoral and cellular immune response in vivo. These data suggest that an MV-based vaccine could control HFRS.
Collapse
|
35
|
Universal influenza vaccine technologies and recombinant virosome production. METHODS IN MICROBIOLOGY 2022. [DOI: 10.1016/bs.mim.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
36
|
Huang Y, Nieh MP, Chen W, Lei Y. Outer membrane vesicles (OMVs) enabled bio-applications: A critical review. Biotechnol Bioeng 2021; 119:34-47. [PMID: 34698385 DOI: 10.1002/bit.27965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 07/28/2021] [Accepted: 10/10/2021] [Indexed: 11/07/2022]
Abstract
Outer membrane vesicles (OMVs) are nanoscale spherical vesicles released from Gram-negative bacteria. The lipid bilayer membrane structure of OMVs consists of similar components as bacterial membrane and thus has attracted more and more attention in exploiting OMVs' bio-applications. Although the endotoxic lipopolysaccharide on natural OMVs may impose potential limits on their clinical applications, genetic modification can reduce their endotoxicity and decorate OMVs with multiple functional proteins. These genetically engineered OMVs have been employed in various fields including vaccination, drug delivery, cancer therapy, bioimaging, biosensing, and enzyme carrier. This review will first briefly introduce the background of OMVs followed by recent advances in functionalization and various applications of engineered OMVs with an emphasis on the working principles and their performance, and then discuss about the future trends of OMVs in biomedical applications.
Collapse
Affiliation(s)
- Yikun Huang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Mu-Ping Nieh
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Yu Lei
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
37
|
Claridge B, Lozano J, Poh QH, Greening DW. Development of Extracellular Vesicle Therapeutics: Challenges, Considerations, and Opportunities. Front Cell Dev Biol 2021; 9:734720. [PMID: 34616741 PMCID: PMC8488228 DOI: 10.3389/fcell.2021.734720] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) hold great promise as therapeutic modalities due to their endogenous characteristics, however, further bioengineering refinement is required to address clinical and commercial limitations. Clinical applications of EV-based therapeutics are being trialed in immunomodulation, tissue regeneration and recovery, and as delivery vectors for combination therapies. Native/biological EVs possess diverse endogenous properties that offer stability and facilitate crossing of biological barriers for delivery of molecular cargo to cells, acting as a form of intercellular communication to regulate function and phenotype. Moreover, EVs are important components of paracrine signaling in stem/progenitor cell-based therapies, are employed as standalone therapies, and can be used as a drug delivery system. Despite remarkable utility of native/biological EVs, they can be improved using bio/engineering approaches to further therapeutic potential. EVs can be engineered to harbor specific pharmaceutical content, enhance their stability, and modify surface epitopes for improved tropism and targeting to cells and tissues in vivo. Limitations currently challenging the full realization of their therapeutic utility include scalability and standardization of generation, molecular characterization for design and regulation, therapeutic potency assessment, and targeted delivery. The fields' utilization of advanced technologies (imaging, quantitative analyses, multi-omics, labeling/live-cell reporters), and utility of biocompatible natural sources for producing EVs (plants, bacteria, milk) will play an important role in overcoming these limitations. Advancements in EV engineering methodologies and design will facilitate the development of EV-based therapeutics, revolutionizing the current pharmaceutical landscape.
Collapse
Affiliation(s)
- Bethany Claridge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - David W. Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
38
|
Zaman M, Huber VC, Heiden DL, DeHaan KN, Chandra S, Erickson D, Ozberk V, Pandey M, Bailly B, Martin G, Langshaw EL, Zaid A, von Itzstein M, Good MF. Combinatorial liposomal peptide vaccine induces IgA and confers protection against influenza virus and bacterial super-infection. Clin Transl Immunology 2021; 10:e1337. [PMID: 34527244 PMCID: PMC8432089 DOI: 10.1002/cti2.1337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Objectives The upper respiratory tract is the major entry site for Streptococcus pyogenes and influenza virus. Vaccine strategies that activate mucosal immunity could significantly reduce morbidity and mortality because of these pathogens. The severity of influenza is significantly greater if a streptococcal infection occurs during the viraemic period and generally viral infections complicated by a subsequent bacterial infection are known as super-infections. We describe an innovative vaccine strategy against influenza virus:S. pyogenes super-infection. Moreover, we provide the first description of a liposomal multi-pathogen-based platform that enables the incorporation of both viral and bacterial antigens into a vaccine and constitutes a transformative development. Methods Specifically, we have explored a vaccination strategy with biocompatible liposomes that express conserved streptococcal and influenza A virus B-cell epitopes on their surface and contain encapsulated diphtheria toxoid as a source of T-cell help. The vaccine is adjuvanted by inclusion of the synthetic analogue of monophosphoryl lipid A, 3D-PHAD. Results We observe that this vaccine construct induces an Immunoglobulin A (IgA) response in both mice and ferrets. Vaccination reduces viral load in ferrets from influenza challenge and protects mice from both pathogens. Notably, vaccination significantly reduces both mortality and morbidity associated with a super-infection. Conclusion The vaccine design is modular and could be adapted to include B-cell epitopes from other mucosal pathogens where an IgA response is required for protection.
Collapse
Affiliation(s)
- Mehfuz Zaman
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Victor C Huber
- Division of Basic Biomedical SciencesSanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Dustin L Heiden
- Division of Basic Biomedical SciencesSanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Katerina N DeHaan
- Division of Basic Biomedical SciencesSanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Sanyogita Chandra
- Division of Basic Biomedical SciencesSanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Demi Erickson
- Division of Basic Biomedical SciencesSanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Victoria Ozberk
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Manisha Pandey
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Benjamin Bailly
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Gael Martin
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Emma L Langshaw
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| | - Ali Zaid
- The Emerging Viruses, Inflammation and Therapeutics GroupMenzies Health Institute QueenslandGriffith UniversityGold CoastQLDAustralia
- School of Medical SciencesGriffith UniversityGold CoastQLDAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastQLDAustralia
| | | | - Michael F Good
- Institute for GlycomicsGriffith UniversityGold CoastQLDAustralia
| |
Collapse
|
39
|
Low endotoxin E. coli strain-derived plasmids reduce rAAV vector-mediated immune responses both in vitro and in vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:293-303. [PMID: 34485612 PMCID: PMC8403685 DOI: 10.1016/j.omtm.2021.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/10/2021] [Indexed: 11/24/2022]
Abstract
The major challenge of recombinant adeno-associated virus (rAAV) vectors is host immunological barriers. Compared to the neutralizing antibody and the cytotoxic T lymphocyte response, the host immune responses induced by unsatisfactory rAAV manufacturing were largely ignored previously. rAAV vector production usually requires large amounts of plasmid DNAs. The DNA are commonly isolated from the DH5α bacterial strain, which contains lipopolysaccharide (LPS) contamination. LPS, also named endotoxin, in plasmid DNA is intractable, and residual endotoxin in the subsequent rAAV vectors may result in substantial host immune response. Recently, a ClearColi K12 bacterial strain is commercially available, with genetically modified LPS that does not trigger endotoxic response in mammalian cells. Here, we produced rAAV-DJ vectors by plasmids yielded from either DH5α or ClearColi K12 bacterial strains. Our data indicated that the ClearColi K12 strain had satisfactory protection for the rAAV inverted terminal repeat (ITR) sequence. As expected, the ClearColi K12-derived rAAV-DJ vectors had lower endotoxin levels. The physical and biological equivalency of the purified viral stocks were confirmed by electron micrographs, Coomassie blue staining, and transduction assays. Most importantly, the ClearColi K12-derived rAAV-DJ vectors triggered reduced nuclear factor-kappa B (NF-κB) signaling pathway both in cell cultures in vitro and in C57BL/6 mice retinas in vivo. We believe that the use of the ClearColi K12 bacterial strain could eliminate the LPS in the purified vector stock at the source. Our data indicate its promising use in future clinical development.
Collapse
|
40
|
Gaspar EB, Prudencio CR, De Gaspari E. Experimental studies using OMV in a new platform of SARS-CoV-2 vaccines. Hum Vaccin Immunother 2021; 17:2965-2968. [PMID: 33950776 PMCID: PMC8108191 DOI: 10.1080/21645515.2021.1920272] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/15/2021] [Indexed: 12/26/2022] Open
Abstract
Although COVID-19 vaccines have recently been approved for emergency use, search for new vaccines are still urgent, since the access of the countries, especially the poorest, to the vaccines, has shown to be slower than the necessary to rapidly control the pandemic. We proposed a novel platform for vaccine using recombinant receptor binding domain (rRBD) from Sars-Cov-2 spike protein and Neisseria meningitidis outer membrane vesicles (OMVs). The antigen preparation produced a humoral and cellular immune response. Taken together our findings suggest a good immunostimulatory patter in response to immunization with rRBD plus N. meningitidis OMV.
Collapse
Affiliation(s)
- Emanuelle B. Gaspar
- Animal Health Department, Embrapa Southern Region Animal Husbandry, Bagé, Brazil
| | | | | |
Collapse
|
41
|
Jung AL, Schmeck B, Wiegand M, Bedenbender K, Benedikter BJ. The clinical role of host and bacterial-derived extracellular vesicles in pneumonia. Adv Drug Deliv Rev 2021; 176:113811. [PMID: 34022269 DOI: 10.1016/j.addr.2021.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022]
Abstract
Pneumonia is among the leading causes of morbidity and mortality worldwide. Due to constant evolution of respiratory bacteria and viruses, development of drug resistance and emerging pathogens, it constitutes a considerable health care threat. To enable development of novel strategies to control pneumonia, a better understanding of the complex mechanisms of interaction between host cells and infecting pathogens is vital. Here, we review the roles of host cell and bacterial-derived extracellular vesicles (EVs) in these interactions. We discuss clinical and experimental as well as pathogen-overarching and pathogen-specific evidence for common viral and bacterial elicitors of community- and hospital-acquired pneumonia. Finally, we highlight the potential of EVs for improved management of pneumonia patients and discuss the translational steps to be taken before they can be safely exploited as novel vaccines, biomarkers, or therapeutics in clinical practice.
Collapse
|
42
|
Mittal N, Sengupta N, Malladi SK, Reddy P, Bhat M, Rajmani RS, Sedeyn K, Saelens X, Dutta S, Varadarajan R. Protective Efficacy of Recombinant Influenza Hemagglutinin Ectodomain Fusions. Viruses 2021; 13:v13091710. [PMID: 34578291 PMCID: PMC8473191 DOI: 10.3390/v13091710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
In current seasonal influenza vaccines, neutralizing antibody titers directed against the hemagglutinin surface protein are the primary correlate of protection. These vaccines are, therefore, quantitated in terms of their hemagglutinin content. Adding other influenza surface proteins, such as neuraminidase and M2e, to current quadrivalent influenza vaccines would likely enhance vaccine efficacy. However, this would come with increased manufacturing complexity and cost. To address this issue, as a proof of principle, we have designed genetic fusions of hemagglutinin ectodomains from H3 and H1 influenza A subtypes. These recombinant H1-H3 hemagglutinin ectodomain fusions could be transiently expressed at high yield in mammalian cell culture using Expi293F suspension cells. Fusions were trimeric, and as stable in solution as their individual trimeric counterparts. Furthermore, the H1-H3 fusion constructs were antigenically intact based on their reactivity with a set of conformation-specific monoclonal antibodies. H1-H3 hemagglutinin ectodomain fusion immunogens, when formulated with the MF59 equivalent adjuvant squalene-in-water emulsion (SWE), induced H1 and H3-specific humoral immune responses equivalent to those induced with an equimolar mixture of individually expressed H1 and H3 ectodomains. Mice immunized with these ectodomain fusions were protected against challenge with heterologous H1N1 (Bel/09) and H3N2 (X-31) mouse-adapted viruses with higher neutralizing antibody titers against the H1N1 virus. Use of such ectodomain-fused immunogens would reduce the number of components in a vaccine formulation and allow for the inclusion of other protective antigens to increase influenza vaccine efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Cross Protection/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Vaccine Efficacy
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Nidhi Mittal
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
| | - Nayanika Sengupta
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
| | - Sameer Kumar Malladi
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
| | - Poorvi Reddy
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru 560012, India; (P.R.); (M.B.)
| | - Madhuraj Bhat
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru 560012, India; (P.R.); (M.B.)
| | - Raju S. Rajmani
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
| | - Koen Sedeyn
- VIB-UGent Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium; (K.S.); (X.S.)
- Department of Biochemistry and Microbiology, Ghent University, 9052 Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium; (K.S.); (X.S.)
- Department of Biochemistry and Microbiology, Ghent University, 9052 Ghent, Belgium
| | - Somnath Dutta
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
| | - Raghavan Varadarajan
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru 560012, India; (N.M.); (N.S.); (S.K.M.); (R.S.R.); (S.D.)
- Correspondence: ; Tel.: +91-80-22932612; Fax: +91-80-23600535
| |
Collapse
|
43
|
Mancini F, Micoli F, Necchi F, Pizza M, Berlanda Scorza F, Rossi O. GMMA-Based Vaccines: The Known and The Unknown. Front Immunol 2021; 12:715393. [PMID: 34413858 PMCID: PMC8368434 DOI: 10.3389/fimmu.2021.715393] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/19/2021] [Indexed: 11/20/2022] Open
Abstract
Generalized Modules for Membrane Antigens (GMMA) are outer membrane vesicles derived from Gram-negative bacteria engineered to provide an over-vesiculating phenotype, which represent an attractive platform for the design of affordable vaccines. GMMA can be further genetically manipulated to modulate the risk of systemic reactogenicity and to act as delivery system for heterologous polysaccharide or protein antigens. GMMA are able to induce strong immunogenicity and protection in animal challenge models, and to be well-tolerated and immunogenic in clinical studies. The high immunogenicity could be ascribed to their particulate size, to their ability to present to the immune system multiple antigens in a natural conformation which mimics the bacterial environment, as well as to their intrinsic self-adjuvanticity. However, GMMA mechanism of action and the role in adjuvanticity are still unclear and need further investigation. In this review, we discuss progresses in the development of the GMMA vaccine platform, highlighting successful applications and identifying knowledge gaps and potential challenges.
Collapse
Affiliation(s)
- Francesca Mancini
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Micoli
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Necchi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Mariagrazia Pizza
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | - Omar Rossi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| |
Collapse
|
44
|
Yang Z, Hua L, Yang M, Liu SQ, Shen J, Li W, Long Q, Bai H, Yang X, Ren Z, Zheng X, Sun W, Ye C, Li D, Zheng P, He J, Chen Y, Huang W, Peng X, Ma Y. RBD-Modified Bacterial Vesicles Elicited Potential Protective Immunity against SARS-CoV-2. NANO LETTERS 2021; 21:5920-5930. [PMID: 34279108 PMCID: PMC8315139 DOI: 10.1021/acs.nanolett.1c00680] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/02/2021] [Indexed: 05/13/2023]
Abstract
The disease caused by SARS-CoV-2 infection threatens human health. In this study, we used high-pressure homogenization technology not only to efficiently drive the bacterial membrane to produce artificial vesicles but also to force the fusion protein ClyA-receptor binding domain (RBD) to pass through gaps in the bacterial membrane to increase the contact between ClyA-RBD and the membrane. Therefore, the load of ClyA-RBD on the membrane is substantially increased. Using this technology, we constructed a "ring-like" bacterial biomimetic vesicle (BBV) loaded with polymerized RBD (RBD-BBV). RBD-BBVs injected subcutaneously can accumulate in lymph nodes, promote antigen uptake and processing, and elicit SARS-CoV-2-specific humoral and cellular immune responses in mice. In conclusion, we evaluated the potential of this novel bacterial vesicle as a vaccine delivery system and provided a new idea for the development of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Zhongqian Yang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Liangqun Hua
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Yunnan
University, Kunming, China
| | - Mengli Yang
- National
Kunming High-level Biosafety Primate Research Center, Institute of
Medical Biology, Chinese Academy of Medical
Sciences and Peking Union Medical College, Kunming, China
| | - Shu-Qun Liu
- Yunnan
University, Kunming, China
- State
Key Laboratory for Conservation and Utilization of Bio-Resources in
Yunnan & School of Life Sciences, Yunnan
University, Kunming, China
| | - Jianxin Shen
- Yunnan
University, Kunming, China
- State
Key Laboratory for Conservation and Utilization of Bio-Resources in
Yunnan & School of Life Sciences, Yunnan
University, Kunming, China
| | - Weiran Li
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Qiong Long
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Hongmei Bai
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Xu Yang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Zhaoling Ren
- The
Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming
Medical University, Kunming, China
| | - Xiao Zheng
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Yunnan
University, Kunming, China
| | - Wenjia Sun
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Chao Ye
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Duo Li
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Department
of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming, China
| | - Peng Zheng
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Jinrong He
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Kunming
Medical University, Kunming, China
| | - Yongjun Chen
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Weiwei Huang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Xiaozhong Peng
- National
Kunming High-level Biosafety Primate Research Center, Institute of
Medical Biology, Chinese Academy of Medical
Sciences and Peking Union Medical College, Kunming, China
- State
Key Laboratory of Medical Molecular Biology, Department of Molecular
Biology and Biochemistry, Institute of Basic Medical Sciences, Medical
Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking
Union Medical College, Beijing, China
| | - Yanbing Ma
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| |
Collapse
|
45
|
Animal Models Utilized for the Development of Influenza Virus Vaccines. Vaccines (Basel) 2021; 9:vaccines9070787. [PMID: 34358203 PMCID: PMC8310120 DOI: 10.3390/vaccines9070787] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/25/2022] Open
Abstract
Animal models have been an important tool for the development of influenza virus vaccines since the 1940s. Over the past 80 years, influenza virus vaccines have evolved into more complex formulations, including trivalent and quadrivalent inactivated vaccines, live-attenuated vaccines, and subunit vaccines. However, annual effectiveness data shows that current vaccines have varying levels of protection that range between 40–60% and must be reformulated every few years to combat antigenic drift. To address these issues, novel influenza virus vaccines are currently in development. These vaccines rely heavily on animal models to determine efficacy and immunogenicity. In this review, we describe seasonal and novel influenza virus vaccines and highlight important animal models used to develop them.
Collapse
|
46
|
Zaruba M, Roschitz L, Sami H, Ogris M, Gerner W, Metzner C. Surface Modification of E. coli Outer Membrane Vesicles with Glycosylphosphatidylinositol-Anchored Proteins: Generating Pro/Eukaryote Chimera Constructs. MEMBRANES 2021; 11:membranes11060428. [PMID: 34199851 PMCID: PMC8228533 DOI: 10.3390/membranes11060428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 11/19/2022]
Abstract
Extracellular vesicles produced by different types of cells have recently attracted great attention, not only for their role in physiology and pathology, but also because of the emerging applications in gene therapy, vaccine production and diagnostics. Less well known than their eukaryotic counterpart, also bacteria produce extracellular vesicles, in the case of the Gram-negative E. coli the main species is termed outer membrane vesicles (OMVs). In this study, we show for the first time the functional surface modification of E. coli OMVs with glycosylphosphatidylinositol (GPI)-anchored protein, exploiting a process variably described as molecular painting or protein engineering in eukaryotic membranes, whereby the lipid part of the GPI anchor inserts in cell membranes. By transferring the process to bacterial vesicles, we can generate a hybrid of perfectly eukaryotic proteins (in terms of folding and post-translational modifications) on a prokaryotic platform. We could demonstrate that two different GPI proteins can be displayed on the same OMV. In addition to fluorescent marker proteins, cytokines, growth factors and antigens canb be potentially transferred, generating a versatile modular platform for a novel vaccine strategy.
Collapse
Affiliation(s)
- Marianne Zaruba
- Institute of Virology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (M.Z.); (L.R.)
| | - Lena Roschitz
- Institute of Virology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (M.Z.); (L.R.)
| | - Haider Sami
- Laboratory of Macromolecular Cancer Therapeutics MMCT, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (H.S.); (M.O.)
| | - Manfred Ogris
- Laboratory of Macromolecular Cancer Therapeutics MMCT, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (H.S.); (M.O.)
| | - Wilhelm Gerner
- Institute of Immunology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Christoph Metzner
- Institute of Virology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (M.Z.); (L.R.)
- Correspondence:
| |
Collapse
|
47
|
Mehanny M, Lehr CM, Fuhrmann G. Extracellular vesicles as antigen carriers for novel vaccination avenues. Adv Drug Deliv Rev 2021; 173:164-180. [PMID: 33775707 DOI: 10.1016/j.addr.2021.03.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/01/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Antigen delivery has always been a challenge in scientific practice of vaccine formulation. Yet, mammalian extracellular vesicles (EVs) or bacterial membrane vesicles (MVs) provide an innovative avenue for safe and effective delivery of antigenic material. They include intrinsically loaded antigens from EV-secreting cells or extrinsically loaded antigens onto pre-formed vesicles. Interestingly, many studies shed light on potential novel anti-cancer vaccination immunotherapy for therapeutic applications from mammalian cell host-derived EVs, as well as conventional vaccination for prophylactic applications using bacterial cell-derived MVs against infectious diseases. Here, we discuss the rationale, status quo and potential for both vaccine applications using EVs.
Collapse
|
48
|
Targeting Antigens for Universal Influenza Vaccine Development. Viruses 2021; 13:v13060973. [PMID: 34073996 PMCID: PMC8225176 DOI: 10.3390/v13060973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Traditional influenza vaccines generate strain-specific antibodies which cannot provide protection against divergent influenza virus strains. Further, due to frequent antigenic shifts and drift of influenza viruses, annual reformulation and revaccination are required in order to match circulating strains. Thus, the development of a universal influenza vaccine (UIV) is critical for long-term protection against all seasonal influenza virus strains, as well as to provide protection against a potential pandemic virus. One of the most important strategies in the development of UIVs is the selection of optimal targeting antigens to generate broadly cross-reactive neutralizing antibodies or cross-reactive T cell responses against divergent influenza virus strains. However, each type of target antigen for UIVs has advantages and limitations for the generation of sufficient immune responses against divergent influenza viruses. Herein, we review current strategies and perspectives regarding the use of antigens, including hemagglutinin, neuraminidase, matrix proteins, and internal proteins, for universal influenza vaccine development.
Collapse
|
49
|
Antonelli G, Cappelli L, Cinelli P, Cuffaro R, Manca B, Nicchi S, Tondi S, Vezzani G, Viviani V, Delany I, Scarselli M, Schiavetti F. Strategies to Tackle Antimicrobial Resistance: The Example of Escherichia coli and Pseudomonas aeruginosa. Int J Mol Sci 2021; 22:4943. [PMID: 34066555 PMCID: PMC8125385 DOI: 10.3390/ijms22094943] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Traditional antimicrobial treatments consist of drugs which target different essential functions in pathogens. Nevertheless, bacteria continue to evolve new mechanisms to evade this drug-mediated killing with surprising speed on the deployment of each new drug and antibiotic worldwide, a phenomenon called antimicrobial resistance (AMR). Nowadays, AMR represents a critical health threat, for which new medical interventions are urgently needed. By 2050, it is estimated that the leading cause of death will be through untreatable AMR pathogens. Although antibiotics remain a first-line treatment, non-antibiotic therapies such as prophylactic vaccines and therapeutic monoclonal antibodies (mAbs) are increasingly interesting alternatives to limit the spread of such antibiotic resistant microorganisms. For the discovery of new vaccines and mAbs, the search for effective antigens that are able to raise protective immune responses is a challenging undertaking. In this context, outer membrane vesicles (OMV) represent a promising approach, as they recapitulate the complete antigen repertoire that occurs on the surface of Gram-negative bacteria. In this review, we present Escherichia coli and Pseudomonas aeruginosa as specific examples of key AMR threats caused by Gram-negative bacteria and we discuss the current status of mAbs and vaccine approaches under development as well as how knowledge on OMV could benefit antigen discovery strategies.
Collapse
Affiliation(s)
- Giada Antonelli
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Luigia Cappelli
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Paolo Cinelli
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Rossella Cuffaro
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Benedetta Manca
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Sonia Nicchi
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Serena Tondi
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Giacomo Vezzani
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Viola Viviani
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Isabel Delany
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
| | - Maria Scarselli
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
| | - Francesca Schiavetti
- GSK Vaccines, 53100 Siena, Italy; (G.A.); (L.C.); (P.C.); (R.C.); (B.M.); (S.N.); (S.T.); (G.V.); (V.V.); (I.D.)
| |
Collapse
|
50
|
Gan Y, Li C, Peng X, Wu S, Li Y, Tan JPK, Yang YY, Yuan P, Ding X. Fight bacteria with bacteria: Bacterial membrane vesicles as vaccines and delivery nanocarriers against bacterial infections. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 35:102398. [PMID: 33901646 DOI: 10.1016/j.nano.2021.102398] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022]
Abstract
Bacterial membrane vesicles (MVs) are particles secreted by bacteria with diameter of 20-400 nm. The pathogen-associated molecular patterns (PAMPs) present on the surface of MVs are capable of activating human immune system, leading to non-specific immune response and specific immune response. Due to the immunostimulatory properties and proteoliposome nanostructures, MVs have been increasingly explored as vaccines or delivery systems for the prevention and treatment of bacterial infections. Herein, the recent progresses of MVs for antibacterial applications are reviewed to provide an overview of MVs vaccines and MVs-related delivery systems. In addition, the safety issues of bacterial MVs are discussed to demonstrate their potential for clinical translation. In the end of this review, the challenges of bacterial MVs as vaccines and delivery systems for clinical applications are highlighted with the purpose of predicting future research directions in this field.
Collapse
Affiliation(s)
- Yingying Gan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xinran Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Shuang Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Jeremy P K Tan
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|