1
|
Zhang H, Liu Z, Li Y, Tao Z, Shen L, Shang Y, Huang X, Liu Q. Adjuvants for Helicobacter pylori vaccines: Outer membrane vesicles provide an alternative strategy. Virulence 2024:2425773. [PMID: 39501551 DOI: 10.1080/21505594.2024.2425773] [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: 01/29/2024] [Revised: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 11/12/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative, spiral-shaped bacterium that colonizes the human stomach, leading to various gastric diseases. The efficacy of traditional treatments, such as bismuth-based triple and quadruple therapies, has reduced due to increasing antibiotic resistance and drug toxicity. As a result, the development of effective vaccines was proposed to control H. pylori-induced infections; however, one of the primary challenges is the lack of potent adjuvants. Although various adjuvants, both toxic (e.g. cholera toxin and Escherichia coli heat-labile toxin) and non-toxic (e.g. aluminium and propolis), have been tested for vaccine development, no clinically favourable adjuvants have been identified due to high toxicity, weak immunostimulatory effects, inability to elicit specific immune responses, or latent side effects. Outer membrane vesicles (OMVs), mainly secreted by gram-negative bacteria, have emerged as promising candidates for H. pylori vaccine adjuvants due to their potential applications. OMVs enhance mucosal immunity and Th1 and Th17 cell responses, which have been recognized to have protective effects and guarantee safety and efficacy. The development of an effective vaccine against H. pylori infection is ongoing, with clinical trials expected in the future.
Collapse
Affiliation(s)
- Hanchi Zhang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yi Li
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Ziwei Tao
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Shen
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinpan Shang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Humaira, Ahmad I, Shakir HA, Khan M, Franco M, Irfan M. Bacterial Extracellular Vesicles: Potential Therapeutic Applications, Challenges, and Future Prospects. J Basic Microbiol 2024; 64:e2400221. [PMID: 39148315 DOI: 10.1002/jobm.202400221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/14/2024] [Accepted: 07/28/2024] [Indexed: 08/17/2024]
Abstract
Almost all cell types naturally secret extracellular vesicles (EVs) in the extracellular space with variable metabolic cargo facilitating intracellular communication, posing immune-modulation capacity. Thus, "bacterial extracellular vesicles" (BEVs), with their great immunoregulatory, immune response stimulation and disease condition-altering potential, have gained importance in the medical and therapeutic industry. Various subtypes of BEVs were observed and reported in the literature, such as exosomes (30-150 nm), microvesicles (100-1000 nm), apoptotic bodies (1000-5000 nm), and oncosomes (1000-10,000 nm). As biological systems are complex entities, inserting BEVs requires extra high purity. Various techniques for BEV isolation have been employed alone or with other strategies, such as ultracentrifugation, precipitation, size-exclusion chromatography, affinity-based separation, ultrafiltration, and field-flow fractionation. But to date, no BEV isolation method is considered perfect as the lack of standard protocols limits their scale-up. Medical research has focused on BEVs to explore their diverse therapeutic potential. This review particularly focused on the recent advancements in the potential medical application of BEVs, current challenges, and prospects associated with their scale-up.
Collapse
Affiliation(s)
- Humaira
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Hafiz Abdullah Shakir
- Institute of Zoology, Faculty of Life Science, University of the Punjab New Campus, Lahore, Pakistan
| | - Muhammad Khan
- Institute of Zoology, Faculty of Life Science, University of the Punjab New Campus, Lahore, Pakistan
| | - Marcelo Franco
- Department of Exact Science, State University of Santa Cruz, Ilheus, Brazil
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| |
Collapse
|
3
|
Bai Z, Wang X, Liang T, Xu G, Cai J, Xu W, Yang K, Hu L, Pei P. Harnessing Bacterial Membrane Components for Tumor Vaccines: Strategies and Perspectives. Adv Healthc Mater 2024; 13:e2401615. [PMID: 38935934 DOI: 10.1002/adhm.202401615] [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: 05/01/2024] [Revised: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Tumor vaccines stand at the vanguard of tumor immunotherapy, demonstrating significant potential and promise in recent years. While tumor vaccines have achieved breakthroughs in the treatment of cancer, they still encounter numerous challenges, including improving the immunogenicity of vaccines and expanding the scope of vaccine application. As natural immune activators, bacterial components offer inherent advantages in tumor vaccines. Bacterial membrane components, with their safer profile, easy extraction, purification, and engineering, along with their diverse array of immune components, activate the immune system and improve tumor vaccine efficacy. This review systematically summarizes the mechanism of action and therapeutic effects of bacterial membranes and its derivatives (including bacterial membrane vesicles and hybrid membrane biomaterials) in tumor vaccines. Subsequently, the authors delve into the preparation and advantages of tumor vaccines based on bacterial membranes and hybrid membrane biomaterials. Following this, the immune effects of tumor vaccines based on bacterial outer membrane vesicles are elucidated, and their mechanisms are explained. Moreover, their advantages in tumor combination therapy are analyzed. Last, the challenges and trends in this field are discussed. This comprehensive analysis aims to offer a more informed reference and scientific foundation for the design and implementation of bacterial membrane-based tumor vaccines.
Collapse
Affiliation(s)
- Zhenxin Bai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuanyu Wang
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| | - Tianming Liang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Guangyu Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jinzhou Cai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| |
Collapse
|
4
|
Wang Y, Li C, Wu F, Mao J, Zhu J, Xie H, Zhou X, Wen C, Tian J. The negative effects of extracellular vesicles in the immune system. Front Immunol 2024; 15:1410273. [PMID: 39372421 PMCID: PMC11449741 DOI: 10.3389/fimmu.2024.1410273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Immunity is a critical self-defense mechanism of the human body, wherein immune cells and immune molecules play a crucial role. Extracellular vesicles (EVs), derived from immune cells or other cells, play a significant role in tumors, autoimmune diseases and other immune-related disorders by serving as carriers and facilitating intercellular communication through the transfer of cargoes. Numerous studies have revealed that EVs can exacerbate disease development by modulating immune responses. Therefore, this paper focuses on the effects of EVs on the number, activity and function of different types of immune cells and the release of immune molecules (such as cytokines, antigens, antibodies, etc) in various diseases, as well as the roles of EVs associated with different types of immune cells in various diseases. We aim to provide a comprehensive review of the negative effects that EVs play in the immune system to provide more ideas and strategies for the management of clinical immune diseases.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Cuifang Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jidong Tian
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Sadeghi L, Bolhassani A, Mohit E, Baesi K, Aghasadeghi MR, Milani A, Agi E. Engineered ClearColi™-derived outer membrane vesicles as functional carriers for development of HIV-1 therapeutic vaccine candidate. Microb Pathog 2024; 193:106749. [PMID: 38879140 DOI: 10.1016/j.micpath.2024.106749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024]
Abstract
Bacteria-derived outer membrane vesicles (OMVs) can be engineered to incorporate foreign antigens. This study explored the potential of ClearColi™-derived OMVs as a natural adjuvant and a carrier (recombinant OMVs or rOMVs) for development of an innovative therapeutic vaccine candidate harboring HIV-1 Nef and Nef-Tat antigens. Herein, the rOMVs containing CytolysinA (ClyA)-Nef and ClyA-Nef-Tat fusion proteins were isolated from ClearColi™ strain. The presence of Nef and Nef-Tat proteins on their surface (rOMVNef and rOMVNef-Tat) was confirmed by western blotting after proteinase K treatment. Immune responses induced by Nef and Nef-Tat proteins emulsified with Montanide® ISA720 or mixed with OMVs, and also rOMVNef and rOMVNef-Tat were investigated in BALB/c mice. Additionally, the potency of splenocytes exposed to single-cycle replicable (SCR) HIV-1 virions was assessed for the secretion of cytokines in vitro. Our findings showed that the rOMVs as an antigen carrier (rOMVNef and rOMVNef-Tat) induced higher levels of IgG2a, IFN-γ and granzyme B compared to OMVs as an adjuvant (Nef + OMV and Nef-Tat + OMV), and also Montanide® ISA720 (Nef + Montanide and Nef-Tat + Montanide). Moreover, IFN-γ level in splenocytes isolated from mice immunized with rOMVNef-Tat was higher than other regimens after exposure to SCR virions. Generally, ClearColi™-derived rOMVs can serve as potent carriers for developing effective vaccines against HIV-1 infection.
Collapse
Affiliation(s)
- Leila Sadeghi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Kazem Baesi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| |
Collapse
|
6
|
Li J, Liao T, Chua EG, Zhang M, Shen Y, Song X, Marshall BJ, Benghezal M, Tang H, Li H. Helicobacter pylori Outer Membrane Vesicles: Biogenesis, Composition, and Biological Functions. Int J Biol Sci 2024; 20:4029-4043. [PMID: 39113715 PMCID: PMC11302881 DOI: 10.7150/ijbs.94156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/06/2024] [Indexed: 08/10/2024] Open
Abstract
Helicobacter pylori has been recognized not only as a causative agent of a spectrum of gastroduodenal diseases including chronic gastritis, peptic ulcer, mucosa-associated lymphoid tissue lymphoma, and gastric cancer, but also as the culprit in several extra-gastric diseases. However, the association of H. pylori infection with extra-gastric diseases remains elusive, prompting a reevaluation of the role of H. pylori-derived outer membrane vesicles (OMVs). Like other gram-negative bacteria, H. pylori constitutively sheds biologically active OMVs for long-distance delivery of bacterial virulence factors in a concentrated and protected form, averting the need of direct bacterial contact with distant host cells to induce extra-gastric diseases associated with this gastric pathogen. Additionally, H. pylori-derived OMVs contribute to bacterial survival and chronic gastric pathogenesis. Moreover, the immunogenic activity, non-replicable nature, and anti-bacterial adhesion effect of H. pylori OMVs make them a desirable vaccine candidate against infection. The immunogenic potency and safety concerns of the OMV contents are challenges in the development of H. pylori OMV-based vaccines. In this review, we discuss recent advances regarding H. pylori OMVs, focusing on new insights into their biogenesis mechanisms and biological functions.
Collapse
Affiliation(s)
- Jiao Li
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | | | - Eng Guan Chua
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands WA 6009, Australia
| | - Mingming Zhang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yalin Shen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiaona Song
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Barry J. Marshall
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands WA 6009, Australia
| | - Mohammed Benghezal
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hong Li
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
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
|
8
|
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 2024:10.1007/s12010-024-04971-x. [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] [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
|
9
|
Li N, Wu M, Wang L, Tang M, Xin H, Deng K. Efficient Isolation of Outer Membrane Vesicles (OMVs) Secreted by Gram-Negative Bacteria via a Novel Gradient Filtration Method. MEMBRANES 2024; 14:135. [PMID: 38921502 PMCID: PMC11205348 DOI: 10.3390/membranes14060135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
Bacterial extracellular vesicles (bEVs) secreted by Gram-negative bacteria are referred to as outer membrane vesicles (OMVs) because they originate in the outer membrane. OMVs are membrane-coated vesicles 20-250 nm in size. They contain lipopolysaccharide (LPS), peptidoglycan, proteins, lipids, nucleic acids, and other substances derived from their parent bacteria and participate in the transmission of information to host cells. OMVs have broad prospects in terms of potential application in the fields of adjuvants, vaccines, and drug delivery vehicles. Currently, there remains a lack of efficient and convenient methods to isolate OMVs, which greatly limits OMV-related research. In this study, we developed a fast, convenient, and low-cost gradient filtration method to separate OMVs that can achieve industrial-scale production while maintaining the biological activity of the isolated OMVs. We compared the gradient filtration method with traditional ultracentrifugation to isolate OMVs from probiotic Escherichia coli Nissle 1917 (EcN) bacteria. Then, we used RAW264.7 macrophages as an in vitro model to study the influence on the immune function of EcN-derived OMVs obtained through the gradient filtration method. Our results indicated that EcN-derived OMVs were efficiently isolated using our gradient filtration method. The level of OMV enrichment obtained via our gradient filtration method was about twice as efficient as that achieved through traditional ultracentrifugation. The EcN-derived OMVs enriched through the gradient filtration method were successfully taken up by RAW264.7 macrophages and induced them to secrete pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α) and interleukins (ILs) 6 and 1β, as well as anti-inflammatory cytokine IL-10. Furthermore, EcN-derived OMVs induced more anti-inflammatory response (i.e., IL-10) than pro-inflammatory response (i.e., TNF-α, IL-6, and IL-1β). These results were consistent with those reported in the literature. The related literature reported that EcN-derived OMVs obtained through ultracentrifugation could induce stronger anti-inflammatory responses than pro-inflammatory responses in RAW264.7 macrophages. Our simple and novel separation method may therefore have promising prospects in terms of applications involving the study of OMVs.
Collapse
Affiliation(s)
- Ning Li
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (M.W.); (L.W.); (M.T.); (H.X.)
| | | | | | | | | | - Keyu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (M.W.); (L.W.); (M.T.); (H.X.)
| |
Collapse
|
10
|
Li X, Guo Q, Chen Q, Chu Y, Zhang Y, Chen H, Zhao Z, Wang Y, Luo Y, Li C, You H, Song H, Su B, Jalili K, Sun T, Jiang C. Reconciling the Cooperative-Competitive Patterns among Tumor and Immune Cells for Triple-Negative Breast Cancer Treatment Using Multimodule Nanocomplexes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312219. [PMID: 38608672 DOI: 10.1002/adma.202312219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/30/2024] [Indexed: 04/14/2024]
Abstract
Targeting the competitive-cooperative relationships among tumor cells and various immune cells can efficiently reverse the immune-dysfunction microenvironment to boost the immunotherapies for the triple-negative breast cancer treatment. Hence, a bacterial outer membrane vesicle-based nanocomplex is designed for specifically targeting malignant cells and immune cells to reconcile the relationships based on metabolic-immune crosstalk. By uniquely utilizing the property of charge-reversal polymers to realize function separation, the nanocomplexes could synergistically regulate tumor cells and immune cells. This approach could reshape the immunosuppressive competition-cooperation pattern into one that is immune-responsive, showcasing significant potential for inducing tumor remission in TNBC models.
Collapse
Affiliation(s)
- Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Qin Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Qinjun Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Yongchao Chu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Yiwen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Hongyi Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Zhenhao Zhao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Yu Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Yifan Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chufeng Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Haoyu You
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Haolin Song
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Boyu Su
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Kiyumars Jalili
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O.Box 51317-11111, Iran
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
- Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| |
Collapse
|
11
|
Sarnelli G, Del Re A, Palenca I, Franzin SB, Lu J, Seguella L, Zilli A, Pesce M, Rurgo S, Esposito G, Sanseverino W, Esposito G. Intranasal administration of Escherichia coli Nissle expressing the spike protein of SARS-CoV-2 induces long-term immunization and prevents spike protein-mediated lung injury in mice. Biomed Pharmacother 2024; 174:116441. [PMID: 38518597 DOI: 10.1016/j.biopha.2024.116441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
While current anti-Spike protein (SP) vaccines have been pivotal in managing the pandemic, their limitations in delivery, storage, and the inability to provide mucosal immunization (preventing infections) highlight the ongoing necessity for research and innovation. To tackle these constraints, our research group developed a bacterial-based vaccine using a non-pathogenic E. coli Nissle 1917 (EcN) strain genetically modified to express the SARS-CoV-2 spike protein on its surface (EcN-pAIDA1-SP). We intranasally delivered the EcN-pAIDA1-SP in two doses and checked specific IgG/IgA production as well as the key immune mediators involved in the process. Moreover, following the initial and booster vaccine doses, we exposed both immunized and non-immunized mice to intranasal delivery of SARS-CoV-2 SP to assess the effectiveness of EcN-pAIDA1-SP in protecting lung tissue from the inflammation damage. We observed detectable levels of anti-SARS-CoV-2 spike IgG in serum samples and IgA in bronchoalveolar lavage fluid two weeks after the initial treatment, with peak concentrations in the respective samples on the 35th day. Moreover, immunoglobulins displayed a progressively enhanced avidity index, suggesting a selective binding to the spike protein. Finally, the pre-immunized group displayed a decrease in proinflammatory markers (TLR4, NLRP3, ILs) following SP challenge, compared to the non-immunized groups, along with better preservation of tissue morphology. Our probiotic-based technology provides an effective immunobiotic tool to protect individuals against disease and control infection spread.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy; Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Alessandro Del Re
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Jie Lu
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Anatomy and Cell Biology, China Medical University, Shenyang 110122, China.
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy.
| | - Giovanni Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, Naples 80131, Italy.
| | - Walter Sanseverino
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Giuseppe Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
12
|
Sangiorgio G, Nicitra E, Bivona D, Bonomo C, Bonacci P, Santagati M, Musso N, Bongiorno D, Stefani S. Interactions of Gram-Positive Bacterial Membrane Vesicles and Hosts: Updates and Future Directions. Int J Mol Sci 2024; 25:2904. [PMID: 38474151 DOI: 10.3390/ijms25052904] [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: 01/24/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayers derived from cell membranes, released by both eukaryotic cells and bacteria into the extracellular environment. During production, EVs carry proteins, nucleic acids, and various compounds, which are then released. While Gram-positive bacteria were traditionally thought incapable of producing EVs due to their thick peptidoglycan cell walls, recent studies on membrane vesicles (MVs) in Gram-positive bacteria have revealed their significant role in bacterial physiology and disease progression. This review explores the current understanding of MVs in Gram-positive bacteria, including the characterization of their content and functions, as well as their interactions with host and bacterial cells. It offers a fresh perspective to enhance our comprehension of Gram-positive bacterial EVs.
Collapse
Affiliation(s)
- Giuseppe Sangiorgio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Emanuele Nicitra
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Dalida Bivona
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Carmelo Bonomo
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Paolo Bonacci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Dafne Bongiorno
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| |
Collapse
|
13
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
14
|
Yunle K, Tong W, Jiyang L, Guojun W. Advances in Helicobacter pylori vaccine research: From candidate antigens to adjuvants-A review. Helicobacter 2024; 29:e13034. [PMID: 37971157 DOI: 10.1111/hel.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/01/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Helicobacter pylori is a Gram-negative, spiral-shaped bacterium that infects approximately 50% of the world's population and has been strongly associated with chronic gastritis, peptic ulcers, gastric mucosa-associated lymphoma, and gastric cancer. The elimination of H. pylori is currently considered one of the most effective strategies for the treatment of gastric-related diseases, so antibiotic therapy is the most commonly used regimen for the treatment of H. pylori infection. Although this therapy has some positive effects, antibiotic resistance has become another clinically prominent problem. Therefore, the development of a safe and efficient vaccine has become an important measure to prevent H. pylori infection. METHODS PubMed and ClinicalTrials.gov were systematically searched from January 1980 to March 2023 with search terms-H. pylori vaccine, adjuvants, immunization, pathogenesis, and H. pylori eradication in the title and/or abstract of literature. A total of 5182 documents were obtained. Based on the principles of academic reliability, authority, nearly publicated, and excluded the similar documents, finally, 75 documents were selected, organized, and analyzed. RESULTS Most of the candidate antigens used as H. pylori vaccines in these literatures are whole-cell antigens and virulence antigens such as UreB, VacA, CagA, and HspA, and the main types of vaccines for H. pylori are whole bacteria vaccines, vector vaccines, subunit vaccines, nucleic acid vaccines, epitope vaccines, etc. Some vaccines have shown good immune protection in animal trials; however, few vaccines show good in clinical trials. The only H. pylori vaccine passed phase 3 clinical trial is a recombinant subunit vaccine using Urease subunit B (UreB) as the vaccine antigen, and it shows good prophylactic effects. Meanwhile, the adjuvant system for vaccines against this bacterium has been developed considerably. In addition to the traditional mucosal adjuvants such as cholera toxin (CT) and E. coli heat labile enterotoxin (LT), there are also promising safer and more effective mucosal adjuvants. All these advances made safe and effective H. pylori vaccines come into service as early as possible. CONCLUSIONS This review briefly summarized the advances of H. pylori vaccines from two aspects, candidates of antigens and adjuvants, to provide references for the development of vaccine against this bacterium. We also present our prospects of exosomal vaccines in H. pylori vaccine research, in the hope of inspiring future researchers.
Collapse
Affiliation(s)
- Kuang Yunle
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wu Tong
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Liu Jiyang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wu Guojun
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
15
|
Abstract
Extracellular vesicles (EVs) are membrane-bound structures released by cells and have become significant players in immune system functioning, primarily by facilitating cell-to-cell communication. Immune cells like neutrophils and dendritic cells release EVs containing bioactive molecules that modulate chemotaxis, activate immune cells, and induce inflammation. EVs also contribute to antigen presentation, lymphocyte activation, and immune tolerance. Moreover, EVs play pivotal roles in antimicrobial host defense. They deliver microbial antigens to antigen-presenting cells (APCs), triggering immune responses, or act as decoys to neutralize virulence factors and toxins. This review discusses host and microbial EVs' multifaceted roles in innate and adaptive immunity, highlighting their involvement in immune cell development, antigen presentation, and antimicrobial responses.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Skylar S. Wright
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Vijay A. Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| |
Collapse
|
16
|
Xiao Y, Wu M, Xue C, Wang Y. Recent Advances in the Development of Membrane-derived Vesicles for Cancer Immunotherapy. Curr Drug Deliv 2024; 21:403-420. [PMID: 37143265 DOI: 10.2174/1567201820666230504120841] [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: 01/17/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
The surface proteins on cell membranes enable the cells to have different properties, such as high biocompatibility, surface modifiability, and homologous targeting ability. Cell-membrane-derived vesicles have features identical to those of their parental cells, which makes them one of the most promising materials for drug delivery. Recently, as a result of the impressive effects of immunotherapy in cancer treatment, an increasing number of researchers have used cell-membrane-derived vesicles to enhance immune responses. To be more specific, the membrane vesicles derived from immune cells, tumor cells, bacteria, or engineered cells have the antigen presentation capacity and can trigger strong anti-tumor effects of the immune system. In this review, we first indicated a brief description of the vesicles and then introduced the detection technology and drug-loading methods for them. Secondly, we concluded the characteristics and applications of vesicles derived from different sources in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuai Xiao
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
17
|
Magaña G, Harvey C, Taggart CC, Rodgers AM. Bacterial Outer Membrane Vesicles: Role in Pathogenesis and Host-Cell Interactions. Antibiotics (Basel) 2023; 13:32. [PMID: 38247591 PMCID: PMC10812699 DOI: 10.3390/antibiotics13010032] [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: 12/04/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Outer membrane vesicles (OMVs) are small, spherical structures released from the outer membranes of Gram-negative bacteria into the surrounding environment. Investigations into OMVs range from their biogenesis and cargo composition to their ability to transfer virulence factors and modulate host immune responses. This emerging understanding of OMVs has unveiled their pivotal role in the pathogenicity of infectious diseases, shedding light on their interactions with host cells, their contributions to inflammation, their potential involvement in antimicrobial resistance, and their promising use for the development of novel treatments and therapies. Numerous studies have associated the OMVs of pathogenic bacteria with the exacerbation of inflammatory diseases, underlining the significance of understanding the mechanisms associated with these vesicles to find alternatives for combating these conditions. Additionally, OMVs possess the ability to act as decoys, absorbing and neutralizing antibiotics, which significantly diminishes the efficacy of a broad spectrum of antimicrobial agents. Another subtopic of interest is OMVs produced by commensal microbiota. These vesicles are increasingly acknowledged for their mutualistic functions, significantly influencing their host's physiology and immune responses. Consequently, OMVs play a crucial role in maintaining a balanced gut microbiota by fostering symbiotic relationships that significantly contribute to the overall health and well-being of the host. This comprehensive review aims to provide an up-to-date review of OMVs derived from Gram-negative bacteria, summarizing current research findings, and elucidating the multifaceted role of these vesicles in diverse biological contexts.
Collapse
Affiliation(s)
| | | | | | - Aoife M. Rodgers
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK; (G.M.); (C.H.); (C.C.T.)
| |
Collapse
|
18
|
Liu Q, Li B, Lu J, Zhang Y, Shang Y, Li Y, Gong T, Zhang C. Recombinant outer membrane vesicles delivering eukaryotic expression plasmid of cytokines act as enhanced adjuvants against Helicobacter pylori infection in mice. Infect Immun 2023; 91:e0031323. [PMID: 37889003 PMCID: PMC10652931 DOI: 10.1128/iai.00313-23] [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: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 10/28/2023] Open
Abstract
The widespread prevalence of Helicobacter pylori (H. pylori) infection remains a great challenge to human health. The existing vaccines are not ideal for preventing H. pylori infection; thus, exploring highly effective adjuvants may improve the immunoprotective efficacy of H. pylori vaccines. In a previous study, we found that the outer membrane vesicles (OMVs), a type of nanoscale particle spontaneously produced by Gram-negative bacteria, could act as adjuvants to boost the immune responses to vaccine antigens. In this study, we explored the potential application of OMVs as delivery vectors for adjuvant development. We constructed recombinant OMVs containing eukaryotic expression plasmid of cytokines, including interleukin 17A or interferon-γ, and evaluated their function as adjuvants in combination with inactivated whole-cell vaccine (WCV) or UreB as vaccine antigens. Our results showed that recombinant OMVs as adjuvants could induce stronger humoral and mucosal immune responses in mice than wild-type H. pylori OMVs and the cholera toxin (CT) adjuvant. Additionally, the recombinant OMVs significantly promoted Th1/Th2/Th17-type immune responses. Furthermore, the recombinant OMV adjuvant induced more potent clearance of H. pylori than CT and wild-type OMVs. Our findings suggest that the recombinant OMVs coupled with cytokines may become potent adjuvants for the development of novel and effective vaccines against H. pylori infection.
Collapse
Affiliation(s)
- Qiong Liu
- Center for Molecular Diagnosis and Precision Medicine, and The Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Biaoxian Li
- Center for Molecular Diagnosis and Precision Medicine, and The Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Jiahui Lu
- Center for Molecular Diagnosis and Precision Medicine, and The Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Yejia Zhang
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Yinpan Shang
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Yi Li
- The Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Tian Gong
- Center for Molecular Diagnosis and Precision Medicine, and The Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengsheng Zhang
- Center for Molecular Diagnosis and Precision Medicine, and The Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Banerjee S, Halder P, Das S, Maiti S, Bhaumik U, Dutta M, Chowdhury G, Kitahara K, Miyoshi SI, Mukhopadhyay AK, Dutta S, Koley H. Pentavalent outer membrane vesicles immunized mice sera confers passive protection against five prevalent pathotypes of diarrhoeagenic Escherichia coli in neonatal mice. Immunol Lett 2023; 263:33-45. [PMID: 37734682 DOI: 10.1016/j.imlet.2023.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Diarrhoeagenic Escherichia coli (DEC) pathotypes are one of the major causative agents of diarrhoea induced childhood morbidity and mortality in developing countries. Licensed vaccines providing broad spectrum protection against DEC mediated infections are not available. Outer membrane vesicles (OMVs) are microvesicles released by gram-negative bacteria during the growth phase and contain multiple immunogenic proteins. Based on prevalence of infections, we have formulated a pentavalent outer-membrane vesicles (POMVs) based immunogen targeting five main pathotypes of DEC responsible for diarrhoeal diseases. Following isolation, OMVs from five DEC pathotypes were mixed in equal proportions to formulate POMVs and 10 µg of the immunogen was intraperitoneally administered to adult BALB/c mice. Three doses of POMVs induced significant humoral immune response against whole cell lysates (WCLs), outer membrane proteins (OMPs) and lipopolysaccharides (LPS) isolated from DEC pathotypes along with significant induction of cellular immune response in adult mice. Passive transfer of POMVs immunized adult mice sera protected neonatal mice significantly against DEC infections. Overall, this study finds POMVs to be immunogenic in conferring broad-spectrum passive protection to neonatal mice against five main DEC pathotypes. Altogether, these findings suggest that POMVs can be used as a potent vaccine candidate to ameliorate the DEC-mediated health burden.
Collapse
Affiliation(s)
- Soumalya Banerjee
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Sanjib Das
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Suhrid Maiti
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Ushasi Bhaumik
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Moumita Dutta
- Division of Electron Microscopy, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Goutam Chowdhury
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India; Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Kei Kitahara
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan; Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Shin-Ichi Miyoshi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan; Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India.
| |
Collapse
|
20
|
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
|
21
|
Rezaei Adriani R, Mousavi Gargari SL, Bakherad H, Amani J. Anti-EGFR bioengineered bacterial outer membrane vesicles as targeted immunotherapy candidate in triple-negative breast tumor murine model. Sci Rep 2023; 13:16403. [PMID: 37775519 PMCID: PMC10541432 DOI: 10.1038/s41598-023-43762-y] [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: 06/20/2023] [Accepted: 09/28/2023] [Indexed: 10/01/2023] Open
Abstract
Cancer immunotherapy employing checkpoint inhibitors holds great promise across diverse cancers; nonetheless, a substantial proportion of patients (ranging from 55 to 87%) remain unresponsive to this treatment. To amplify therapeutic efficiency, we propose a synergistic therapeutic strategy that entails the deployment of targeted nano-sized particles carrying Toll-like receptor (TLR) agonists to the tumor site. This innovative approach seeks to activate intratumoral antigen-presenting cells using bioengineered outer membrane vesicles (OMVs) derived from gram-negative bacteria. These OMVs possess inherent attributes of surface-exposed immune stimulators and TLR-activating components, rendering them intriguing candidates for investigation. These OMVs were meticulously designed to selectively target cancer cells exhibiting an overexpression of epidermal growth factor receptor (EGFR). To gauge the precision of this targeting, the conducted affinity-based assays aimed at determining the equilibrium dissociation constant of the single-chain variable fragment employed for this purpose. In vitro experiments confirmed the OMVs' proficiency in adhering to EGFR-overexpressed cancer cells. Moreover, the evaluation extended to an in vivo context, where the therapeutic effect of nanovesicles was appraised within the tumor microenvironment of the triple-negative breast cancer mouse model. Notably, both intraperitoneal and intratumoral administrations of nanovesicles exhibited the ability to activate natural killer cells and skew M2 macrophage towards an M1 phenotype. The combined scrutiny of in vitro and in vivo findings underscores the potential efficiency of OMVs as a promising strategy for future anti-tumor endeavors.
Collapse
Affiliation(s)
| | | | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, System Biology, and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Xiao M, Li G, Yang H. Microbe-host interactions: structure and functions of Gram-negative bacterial membrane vesicles. Front Microbiol 2023; 14:1225513. [PMID: 37720140 PMCID: PMC10500606 DOI: 10.3389/fmicb.2023.1225513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Bacteria-host interaction is a common, relevant, and intriguing biological phenomena. The host reacts actively or passively to the bacteria themselves, their products, debris, and so on, through various defense systems containing the immune system, the bacteria communicate with the local or distal tissues of the host via their own surface antigens, secreted products, nucleic acids, etc., resulting in relationships of attack and defense, adaptation, symbiosis, and even collaboration. The significance of bacterial membrane vesicles (MVs) as a powerful vehicle for the crosstalk mechanism between the two is growing. In the recent decade, the emergence of MVs in microbial interactions and a variety of bacterial infections, with multiple adhesions to host tissues, cell invasion and evasion of host defense mechanisms, have brought MVs to the forefront of bacterial pathogenesis research. Whereas MVs are a complex combination of molecules not yet fully understood, research into its effects, targeting and pathogenic components will advance its understanding and utilization. This review will summarize structural, extraction and penetration information on several classes of MVs and emphasize the role of MVs in transport and immune response activation. Finally, the potential of MVs as a therapeutic method will be highlighted, as will future research prospects.
Collapse
Affiliation(s)
- Min Xiao
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, China
- Department of Dental Research, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Guiding Li
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, China
- Department of Dental Research, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, China
- Department of Dental Research, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
23
|
Tognetti F, Biagini M, Denis M, Berti F, Maione D, Stranges D. Evolution of Vaccines Formulation to Tackle the Challenge of Anti-Microbial Resistant Pathogens. Int J Mol Sci 2023; 24:12054. [PMID: 37569427 PMCID: PMC10418901 DOI: 10.3390/ijms241512054] [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: 06/21/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
The increasing diffusion of antimicrobial resistance (AMR) across more and more bacterial species emphasizes the urgency of identifying innovative treatment strategies to counter its diffusion. Pathogen infection prevention is among the most effective strategies to prevent the spread of both disease and AMR. Since their discovery, vaccines have been the strongest prophylactic weapon against infectious diseases, with a multitude of different antigen types and formulative strategies developed over more than a century to protect populations from different pathogens. In this review, we review the main characteristics of vaccine formulations in use and under development against AMR pathogens, focusing on the importance of administering multiple antigens where possible, and the challenges associated with their development and production. The most relevant antigen classes and adjuvant systems are described, highlighting their mechanisms of action and presenting examples of their use in clinical trials against AMR. We also present an overview of the analytical and formulative strategies for multivalent vaccines, in which we discuss the complexities associated with mixing multiple components in a single formulation. This review emphasizes the importance of combining existing knowledge with advanced technologies within a Quality by Design development framework to efficiently develop vaccines against AMR pathogens.
Collapse
Affiliation(s)
- Francesco Tognetti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padua, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Vázquez ME, Mesías AC, Acuña L, Spangler J, Zabala B, Parodi C, Thakur M, Oh E, Walper SA, Brandán CP. Exploring the performance of Escherichia coli outer membrane vesicles as a tool for vaccine development against Chagas disease. Mem Inst Oswaldo Cruz 2023; 118:e220263. [PMID: 37222309 DOI: 10.1590/0074-02760220263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/25/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Vaccine development is a laborious craftwork in which at least two main components must be defined: a highly immunogenic antigen and a suitable delivery method. Hence, the interplay of these elements could elicit the required immune response to cope with the targeted pathogen with a long-lasting protective capacity. OBJECTIVES Here we evaluate the properties of Escherichia coli spherical proteoliposomes - known as outer membrane vesicles (OMVs) - as particles with natural adjuvant capacities and as antigen-carrier structures to assemble an innovative prophylactic vaccine for Chagas disease. METHODS To achieve this, genetic manipulation was carried out on E. coli using an engineered plasmid containing the Tc24 Trypanosoma cruzi antigen. The goal was to induce the release of OMVs displaying the parasite protein on their surface. FINDINGS As a proof of principle, we observed that native OMVs - as well as those carrying the T. cruzi antigen - were able to trigger a slight, but functional humoral response at low immunization doses. Of note, compared to the non-immunized group, native OMVs-vaccinated animals survived the lethal challenge and showed minor parasitemia values, suggesting a possible involvement of innate trained immunity mechanism. MAIN CONCLUSION These results open the range for further research on the design of new carrier strategies focused on innate immunity activation as an additional immunization target and venture to seek for alternative forms in which OMVs could be used for optimizing vaccine development.
Collapse
Affiliation(s)
- María Elisa Vázquez
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| | - Andrea Cecilia Mesías
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| | - Leonardo Acuña
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| | - Joseph Spangler
- US Naval Research Laboratory, Center for Bio/Molecular Science & Engineering, Washington, DC, United States of America
| | - Brenda Zabala
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| | - Cecilia Parodi
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| | - Meghna Thakur
- George Mason University, Fairfax, Virginia, United States of America
| | - Eunkeu Oh
- US Naval Research Laboratory, Optical Science Division, Washington, DC, United States of America
| | - Scott Allan Walper
- US Naval Research Laboratory, Center for Bio/Molecular Science & Engineering, Washington, DC, United States of America
| | - Cecilia Pérez Brandán
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Salta, Instituto de Patología Experimental Dr Miguel Ángel Basombrío, Salta, Argentina
| |
Collapse
|
25
|
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
|
26
|
Kawano K, Kamasaka K, Yokoyama F, Kawamoto J, Ogawa T, Kurihara T, Matsuzaki K. Structural factors governing binding of curvature-sensing peptides to bacterial extracellular vesicles covered with hydrophilic polysaccharide chains. Biophys Chem 2023; 299:107039. [PMID: 37209609 DOI: 10.1016/j.bpc.2023.107039] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 05/22/2023]
Abstract
Extracellular vesicles (EVs) have attracted an attention as important targets in the fields of biology and medical science because they contain physiologically active molecules. Curvature-sensing peptides are currently used as novel tools for marker-independent EV detection techniques. A structure-activity correlation study demonstrated that the α-helicity of the peptides is prominently involved in peptide binding to vesicles. However, whether a flexible structure changing from a random coil to an α-helix upon binding to vesicles or a restricted α-helical structure is an important factor in the detection of biogenic vesicles is still unclear. To address this issue, we compared the binding affinities of stapled and unstapled peptides for bacterial EVs with different surface polysaccharide chains. We found that unstapled peptides showed similar binding affinities for bacterial EVs regardless of surface polysaccharide chains, whereas stapled peptides showed substantially decreased binding affinities for bacterial EVs covered with capsular polysaccharides. This is probably because curvature-sensing peptides must pass through the layer of hydrophilic polysaccharide chains prior to binding to the hydrophobic membrane surface. While stapled peptides with restricted structures cannot easily pass through the layer of polysaccharide chains, unstapled peptides with flexible structures can easily approach the membrane surface. Therefore, we concluded that the structural flexibility of curvature-sensing peptides is a key factor for governing the highly sensitive detection of bacterial EVs.
Collapse
Affiliation(s)
- Kenichi Kawano
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Kouhei Kamasaka
- Institute for Chemical Research, Kyoto University, Gokasho, Uji 611-0011, Japan
| | - Fumiaki Yokoyama
- Institute for Chemical Research, Kyoto University, Gokasho, Uji 611-0011, Japan; Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jun Kawamoto
- Institute for Chemical Research, Kyoto University, Gokasho, Uji 611-0011, Japan
| | - Takuya Ogawa
- Institute for Chemical Research, Kyoto University, Gokasho, Uji 611-0011, Japan
| | - Tatsuo Kurihara
- Institute for Chemical Research, Kyoto University, Gokasho, Uji 611-0011, Japan
| | - Katsumi Matsuzaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
27
|
Rezaei M, Esmaeili F, Reza Asadi Karam M, Ehsani P, Abbasnezhad Farsangi Z, Bouzari S. In silico design and in vivo evaluation of two multi-epitope vaccines containing build-in adjuvant with chitosan nanoparticles against uropathogenic Escherichia coli. Int Immunopharmacol 2023; 117:109999. [PMID: 37012877 DOI: 10.1016/j.intimp.2023.109999] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/20/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Urinary pathogenic Escherichia coli (UPEC) is one of the most important bacterial causes of urinary tract infections (UTIs). Rising antimicrobial resistance and serious clinical challenges such as persistent and recurrent UTIs make it a serious public health concern. Therefore, preventative approaches such as vaccinations are required. METHODS In this study, we selected three conserve and protective antigens (FdeC, Hma and UpaB) and also subunit B of cholera toxin (as build-in adjuvant) to design two multi-epitope vaccines (construct B containing B cell epitopes and construct T containing T epitopes) using different bioinformatics methods. The expression of the recombinant protein was performed using the BL21(DE3)/pET28 expression system and purified through a Ni-NTA column. Vaccine proteins were encapsulated in chitosan nanoparticles (CNP) based on ionic gelation via a microfluidic system. Mice were immunized intranasally with different vaccine formulations. Antibody responses and also cytokine expression (IFN-γ and IL-4) were measured by ELISA and real-time PCR respectively. The effectiveness of immune responses was assessed by bladder challenge. RESULTS Based on the in silico study, construct B and construct T have high confidence value and stable structure in vivo. High yield expression of both constructs was confirmed by SDS-PAGE and western blot assay. Immunization of mice with construct B induced strong Th2 (IgG1 and IL4) responses and construct T shift immune responses to Th1 (IFNγ and IgG2a). Vaccine protein-encapsulated CNP elicited higher levels of antibodies and cell-mediated responses than the vaccine proteins alone. CONCLUSIONS The results of this study suggest that intranasal administration of the construct B has the potential to enhance humoral immunity and construct T has the potential to stimulate cellular immunity. In addition, the combination of CTB as a build-in adjuvant and CNP can be proposed as a potent adjuvant for the development of a novel vaccine against UTI.
Collapse
Affiliation(s)
- Maryam Rezaei
- Department of Molecular Biology, Pasteur institute of Iran, Tehran, Iran
| | - Fariba Esmaeili
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Parastoo Ehsani
- Department of Molecular Biology, Pasteur institute of Iran, Tehran, Iran
| | | | - Saeid Bouzari
- Department of Molecular Biology, Pasteur institute of Iran, Tehran, Iran.
| |
Collapse
|
28
|
Sarnelli G, Del Re A, Pesce M, Lu J, Esposito G, Sanseverino W, Corpetti C, Basili Franzin S, Seguella L, Palenca I, Rurgo S, De Palma FDE, Zilli A, Esposito G. Oral Immunization with Escherichia coli Nissle 1917 Expressing SARS-CoV-2 Spike Protein Induces Mucosal and Systemic Antibody Responses in Mice. Biomolecules 2023; 13:biom13030569. [PMID: 36979504 PMCID: PMC10046078 DOI: 10.3390/biom13030569] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/04/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
As of October 2022, the COVID-19 pandemic continues to pose a major public health conundrum, with increased rates of symptomatic infections in vaccinated individuals. An ideal vaccine candidate for the prevention of outbreaks should be rapidly scalable, easy to administer, and able to elicit a potent mucosal immunity. Towards this aim, we proposed an engineered Escherichia coli (E. coli) Nissle 1917 (EcN) strain with SARS-CoV-2 spike protein (SP)-coding plasmid, which was able to expose SP on its cellular surface by a hybridization with the adhesin involved in diffuse adherence 1 (AIDA1). In this study, we presented the effectiveness of a 16-week intragastrically administered, engineered EcN in producing specific systemic and mucosal immunoglobulins against SARS-CoV-2 SP in mice. We observed a time-dependent increase in anti-SARS-CoV-2 SP IgG antibodies in the sera at week 4, with a titre that more than doubled by week 12 and a stable circulating titre by week 16 (+309% and +325% vs. control; both p < 0.001). A parallel rise in mucosal IgA antibody titre in stools, measured via intestinal and bronchoalveolar lavage fluids of the treated mice, reached a plateau by week 12 and until the end of the immunization protocol (+300, +47, and +150%, at week 16; all p < 0.001 vs. controls). If confirmed in animal models of infection, our data indicated that the engineered EcN may be a potential candidate as an oral vaccine against COVID-19. It is safe, inexpensive, and, most importantly, able to stimulate the production of both systemic and mucosal anti-SARS-CoV-2 spike-protein antibodies.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
| | - Alessandro Del Re
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
| | - Jie Lu
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Anatomy and Cell Biology, China Medical University, Shenyang 110122, China
| | - Giovanni Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, 80131 Naples, Italy
| | - Walter Sanseverino
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
| | - Chiara Corpetti
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
| | - Fatima Domenica Elisa De Palma
- Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, 80131 Naples, Italy
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppe Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
29
|
Characterization and Vaccine Potential of Outer Membrane Vesicles from Photobacterium damselae subsp. piscicida. Int J Mol Sci 2023; 24:ijms24065138. [PMID: 36982212 PMCID: PMC10049053 DOI: 10.3390/ijms24065138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Photobacterium damselae subsp. piscicida (Phdp) is a Gram-negative fish pathogen with worldwide distribution and broad host specificity that causes heavy economic losses in aquaculture. Although Phdp was first identified more than 50 years ago, its pathogenicity mechanisms are not completely understood. In this work, we report that Phdp secretes large amounts of outer membrane vesicles (OMVs) when cultured in vitro and during in vivo infection. These OMVs were morphologically characterized and the most abundant vesicle-associated proteins were identified. We also demonstrate that Phdp OMVs protect Phdp cells from the bactericidal activity of fish antimicrobial peptides, suggesting that secretion of OMVs is part of the strategy used by Phdp to evade host defense mechanisms. Importantly, the vaccination of sea bass (Dicentrarchus labrax) with adjuvant-free crude OMVs induced the production of anti-Phdp antibodies and resulted in partial protection against Phdp infection. These findings reveal new aspects of Phdp biology and may provide a basis for developing new vaccines against this pathogen.
Collapse
|
30
|
Liu R, Sun W, Sun T, Zhang W, Nan Y, Zhang Z, Xiang K, Yang H, Wang F, Ge J. Nano selenium-enriched probiotic Lactobacillus enhances alum adjuvanticity and promotes antigen-specific systemic and mucosal immunity. Front Immunol 2023; 14:1116223. [PMID: 36793732 PMCID: PMC9922588 DOI: 10.3389/fimmu.2023.1116223] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 02/17/2023] Open
Abstract
Nano selenium-enriched probiotics have been identified to improve immune responses, such as alleviating inflammation, antioxidant function, treatment of tumors, anticancer activity, and regulating intestinal flora. However, so far, there is little information on improving the immune effect of the vaccine. Here, we prepared nano selenium-enriched Levilactobacillus brevis 23017 (SeL) and heat-inactivated nano selenium-enriched L. brevis 23017 (HiSeL) and evaluated their immune enhancing functions on the alum-adjuvanted, inactivated Clostridium perfringens type A vaccine in mouse and rabbit models, respectively. We found that SeL enhanced immune responses of the vaccine by inducing a more rapid antibody production, eliciting higher immunoglobulin G (IgG) antibody titers, improving secretory immunoglobulin A (SIgA) antibody level and cellular immune response, and regulating Th1/Th2 immune response, thus helping to induce better protective efficacy after challenge. Moreover, we confirmed that the immunoenhancement effects are related to regulating oxidative stress, cytokine secretion, and selenoprotein expression. Meanwhile, similar effects were observed in HiSeL. In addition, they show enhanced humoral immune response at 1/2 and 1/4 standard vaccine doses, which confirms their prominent immune enhancement effect. Finally, the effect of improving vaccine immune responses was further confirmed in rabbits, which shows that SeL stimulates the production of IgG antibodies, generates α toxin-neutralizing antibodies rapidly, and reduces the pathological damage to intestine tissue. Our study demonstrates that nano selenium-enriched probiotics improve the immune effect of the alum adjuvants vaccine and highlight its potential usage in remedying the disadvantages of alum adjuvants.
Collapse
Affiliation(s)
- Runhang Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Weijiao Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tianzhi Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenzhi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yongchao Nan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zheng Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Kongrui Xiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hongliang Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Fang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, China,*Correspondence: Fang Wang, ; Junwei Ge,
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China,Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin, China,*Correspondence: Fang Wang, ; Junwei Ge,
| |
Collapse
|
31
|
Ge Y, Sun F, Zhao B, Kong F, Li Z, Kong X. Bacteria derived extracellular vesicles in the pathogenesis and treatment of gastrointestinal tumours. Front Oncol 2023; 12:1103446. [PMID: 36776356 PMCID: PMC9910087 DOI: 10.3389/fonc.2022.1103446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles are fundamentally significant in the communication between cells. Outer Membrane Vesicles(OMVs) are a special kind of EVs produced by Gram-negative bacteria, which are minute exosome-like particles budding from the outer membrane, which have been found to play essential roles in diverse bacterial life events, including regulation of microbial interactions, pathogenesis promotion, stress responses and biofilm formation. Recently, and more researches have explored the substantial potentials of EVs as natural functional nanoparticles in the bioengineering applications in infectious diseases, cardiovascular diseases, autoimmune diseases and neurological diseases, such as antibacterial therapy, cancer drugs and immunoadjuvants, with several candidates in clinical trials showing promising efficacy. However, due to the poor understanding of sources, membrane structures and biogenesis mechanisms of EVs, progress in clinical applications still remains timid. In this review, we summarize the latest findings of EVs, especially in gastrointestinal tract tumours, to provide a comprehensive introduction of EVs in tumorigenesis and therapeutics.
Collapse
Affiliation(s)
- Yang Ge
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fengyuan Sun
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Bo Zhao
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fanyang Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Xiangyu Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| |
Collapse
|
32
|
Bio-Inspired Drug Delivery Systems: From Synthetic Polypeptide Vesicles to Outer Membrane Vesicles. Pharmaceutics 2023; 15:pharmaceutics15020368. [PMID: 36839691 PMCID: PMC9965272 DOI: 10.3390/pharmaceutics15020368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Nanomedicine is a broad field that focuses on the development of nanocarriers to deliver specific drugs to targeted sites. A synthetic polypeptide is a kind of biomaterial composed of repeating amino acid units that are linked by peptide bonds. The multiplied amphiphilicity segment of the polypeptide could assemble to form polypeptide vesicles (PVs) under suitable conditions. Different from polypeptide vesicles, outer membrane vesicles (OMVs) are spherical buds of the outer membrane filled with periplasmic content, which commonly originate from Gram-negative bacteria. Owing to their biodegradability and excellent biocompatibility, both PVs and OMVs have been utilized as carriers in delivering drugs. In this review, we discuss the recent drug delivery research based on PVs and OMVs. These related topics are presented: (1) a brief introduction to the production methods for PVs and OMVs; (2) a thorough explanation of PV- and OMV-related applications in drug delivery including the vesicle design and biological assessment; (3) finally, we conclude with a discussion on perspectives and future challenges related to the drug delivery systems of PVs and OMVs.
Collapse
|
33
|
Liu Y, Chen J, Raj K, Baerg L, Nathan N, Philpott DJ, Mahadevan R. A Universal Strategy to Promote Secretion of G+/G- Bacterial Extracellular Vesicles and Its Application in Host Innate Immune Responses. ACS Synth Biol 2023; 12:319-328. [PMID: 36592614 DOI: 10.1021/acssynbio.2c00583] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Both Gram-positive and Gram-negative bacteria release nanosized extracellular vesicles called membrane vesicles (MVs, 20-400 nm), which have great potential in various biomedical applications due to their abilities to deliver effector molecules and induce therapeutic responses. To fully utilize bacterial MVs for therapeutic purposes, regulated and enhanced production of MVs would be highly advantageous. In this study, we developed a universal method to enhance MV yields in both G+/G- bacteria through an autonomous controlled peptidoglycan hydrolase (PGase) expression system. A significant increase (9.37-fold) of MV concentration was observed in engineered E. coli Nissle 1917 compared to the wild-type. With the help of this autonomous system, for the first time we experimentally confirmed horizontal gene transfer and nutrient acquisition in a cocultured bacterial consortium. Furthermore, the engineered probiotic E. coli strains with high yield of MVs showed higher activation of the innate immune responses in human embryonic kidney 293T (HEK293T) and human colorectal carcinoma cells (HCT116), thereby demonstrating the great potential of engineering probiotics in immunology and further living therapeutics in humans.
Collapse
Affiliation(s)
- Yilan Liu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Jinjin Chen
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Kaushik Raj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Lauren Baerg
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Nayanan Nathan
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Radhakrishnan Mahadevan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
34
|
Jalalifar S, Morovati Khamsi H, Hosseini-Fard SR, Karampoor S, Bajelan B, Irajian G, Mirzaei R. Emerging role of microbiota derived outer membrane vesicles to preventive, therapeutic and diagnostic proposes. Infect Agent Cancer 2023; 18:3. [PMID: 36658631 PMCID: PMC9850788 DOI: 10.1186/s13027-023-00480-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
The role of gut microbiota and its products in human health and disease is profoundly investigated. The communication between gut microbiota and the host involves a complicated network of signaling pathways via biologically active molecules generated by intestinal microbiota. Some of these molecules could be assembled within nanoparticles known as outer membrane vesicles (OMVs). Recent studies propose that OMVs play a critical role in shaping immune responses, including homeostasis and acute inflammatory responses. Moreover, these OMVs have an immense capacity to be applied in medical research, such as OMV-based vaccines and drug delivery. This review presents a comprehensive overview of emerging knowledge about biogenesis, the role, and application of these bacterial-derived OMVs, including OMV-based vaccines, OMV adjuvants characteristics, OMV vehicles (in conjugated vaccines), cancer immunotherapy, and drug carriers and delivery systems. Moreover, we also highlight the significance of the potential role of these OMVs in diagnosis and therapy.
Collapse
Affiliation(s)
- Saba Jalalifar
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Morovati Khamsi
- Department of Quality Control, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Bahar Bajelan
- School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Irajian
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
35
|
Kong L, Yang C, Zhang Z. Organism-Generated Biological Vesicles In Situ: An Emerging Drug Delivery Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204178. [PMID: 36424135 PMCID: PMC9839880 DOI: 10.1002/advs.202204178] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Biological vesicles, containing genetic materials and proteins of the original cells, are usually used for local or systemic communications among cells. Currently, studies on biological vesicles as therapeutic strategies or drug delivery carriers mainly focus on exogenously generated biological vesicles. However, the limitations of yield and purity caused by the complex purification process still hinder their clinical transformation. Recently, it has been reported that living organisms, including cells and bacteria, can produce functional/therapeutic biological vesicles within body automatically. Therefore, using organisms to produce endogenous biological vesicles in body as drug/bio-information delivery carriers has become a potential therapeutic strategy. In this review, the current development status and application prospects of in situ organism-produced biological vesicles are introduced. The advantages and effects of this endogenous biological vesicles-based strategy in drug delivery and disease treatments are analyzed. According to the type of endogenous biological vesicles, they are divided into four categories: exosomes, platelet-derived microparticles, apoptotic bodies, and bacteria-released outer membrane vesicles. And finally, the shortcomings of current research and future development are analyzed. This review is believed to open up the application of endogenous biological vesicles in the field of biomedicine and shed light on current research.
Collapse
Affiliation(s)
- Li Kong
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
| | - Conglian Yang
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
| | - Zhiping Zhang
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
- Hubei Engineering Research Center for Novel Drug Delivery SystemHuazhong University of Science and TechnologyWuhan430030P. R. China
- National Engineering Research Center for NanomedicineHuazhong University of Science and TechnologyWuhan430030P. R. China
| |
Collapse
|
36
|
Wang H, Ewetse MP, Ma C, Pu W, Xu B, He P, Wang Y, Zhu J, Chen H. The "Light Knife" for Gastric Cancer: Photodynamic Therapy. Pharmaceutics 2022; 15:pharmaceutics15010101. [PMID: 36678730 PMCID: PMC9860630 DOI: 10.3390/pharmaceutics15010101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Photodynamic therapy (PDT) has been used clinically to treat cancer for more than 40 years. Some solid tumors, including esophageal cancer, lung cancer, head and neck cancer, cholangiocarcinoma, and bladder cancer, have been approved for and managed with PDT in many countries globally. Notably, PDT for gastric cancer (GC) has been reported less and is not currently included in the clinical diagnosis and treatment guidelines. However, PDT is a potential new therapeutic modality used for the management of GC, and its outcomes and realization are more and more encouraging. PDT has a pernicious effect on tumors at the irradiation site and can play a role in rapid tumor shrinkage when GC is combined with cardiac and pyloric obstruction. Furthermore, because of its ability to activate the immune system, it still has a specific effect on systemic metastatic lesions, and the adverse reactions are mild. In this Review, we provide an overview of the current application progress of PDT for GC; systematically elaborate on its principle, mechanism, and the application of a new photosensitizer in GC; and focus on the efficacy of PDT in GC and the prospect of combined use with other therapeutic methods to provide a theoretical basis for clinical application.
Collapse
Affiliation(s)
- Haiyun Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | | | - Chenhui Ma
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Weigao Pu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Bo Xu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Puyi He
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Yunpeng Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Jingyu Zhu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Hao Chen
- Department of Oncology Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumor, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-0931-5190550
| |
Collapse
|
37
|
Lieberman LA. Outer membrane vesicles: A bacterial-derived vaccination system. Front Microbiol 2022; 13:1029146. [PMID: 36620013 PMCID: PMC9811673 DOI: 10.3389/fmicb.2022.1029146] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Outer membrane vesicles (OMVs) are non-living spherical nanostructures that derive from the cell envelope of Gram-negative bacteria. OMVs are important in bacterial pathogenesis, cell-to-cell communication, horizontal gene transfer, quorum sensing, and in maintaining bacterial fitness. These structures can be modified to express antigens of interest using glycoengineering and genetic or chemical modification. The resulting OMVs can be used to immunize individuals against the expressed homo- or heterologous antigens. Additionally, cargo can be loaded into OMVs and they could be used as a drug delivery system. OMVs are inherently immunogenic due to proteins and glycans found on Gram negative bacterial outer membranes. This review focuses on OMV manipulation to increase vesiculation and decrease antigenicity, their utility as vaccines, and novel engineering approaches to extend their application.
Collapse
|
38
|
Kong C, Chen X. Combined Photodynamic and Photothermal Therapy and Immunotherapy for Cancer Treatment: A Review. Int J Nanomedicine 2022; 17:6427-6446. [PMID: 36540374 PMCID: PMC9760263 DOI: 10.2147/ijn.s388996] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
Photoactivation therapy based on photodynamic therapy (PDT) and photothermal therapy (PTT) has been identified as a tumour ablation modality for numerous cancer indications, with photosensitisers and photothermal conversion agents playing important roles in the phototherapy process, especially in recent decades. In addition, the iteration of nanotechnology has strongly promoted the development of phototherapy in tumour treatment. PDT can increase the sensitivity of tumour cells to PTT by interfering with the tumour microenvironment, whereas the heat generated by PTT can increase blood flow, improve oxygen supply and enhance the PDT therapeutic effect. In addition, tumour cell debris generated by phototherapy can serve as tumour-associated antigens, evoking antitumor immune responses. In this review, the research progress of phototherapy, and its research effects in combination with immunotherapy on the treatment of tumours are mainly outlined, and issues that may need continued attention in the future are raised.
Collapse
Affiliation(s)
- Cunqing Kong
- Department of medical imaging center, central hospital affiliated to Shandong first medical university, Jinan, People’s Republic of China
| | - Xingcai Chen
- Department of Human Anatomy and Center for Genomics and Personalized Medicine, Nanning, People’s Republic of China,Correspondence: Xingcai Chen, Email
| |
Collapse
|
39
|
Hu K, Palmieri E, Samnuan K, Ricchetti B, Oldrini D, McKay PF, Wu G, Thorne L, Fooks AR, McElhinney LM, Goharriz H, Golding M, Shattock RJ, Micoli F. Generalized Modules for Membrane Antigens (GMMA), an outer membrane vesicle-based vaccine platform, for efficient viral antigen delivery. J Extracell Vesicles 2022; 11:e12247. [PMID: 36377074 PMCID: PMC9663859 DOI: 10.1002/jev2.12247] [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: 09/03/2021] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
Vaccine platforms enable fast development, testing, and manufacture of more affordable vaccines. Here, we evaluated Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles (OMVs) generated by genetically modified Gram-negative bacteria, as a vaccine platform for viral pathogens. Influenza A virus hemagglutinin (HA), either physically mixed with GMMA (HA+STmGMMA mix), or covalently linked to GMMA surface (HA-STmGMMA conjugate), significantly increased antigen-specific humoral and cellular responses, with HA-STmGMMA conjugate inducing further enhancement than HA+STmGMMA mix. HA-STmGMMA conjugate protected mice from lethal challenge. The versatility for this platform was confirmed by conjugation of rabies glycoprotein (RABVG) onto GMMA through the same method. RABVG+STmGMMA mix and RABVG-STmGMMA conjugate exhibited similar humoral and cellular response patterns and protection efficacy as the HA formulations, indicating relatively consistent responses for different vaccines based on the GMMA platform. Comparing to soluble protein, GMMA was more efficiently taken up in vivo and exhibited a B-cell preferential uptake in the draining lymph nodes (LNs). Together, GMMA enhances immunity against viral antigens, and the platform works well with different antigens while retaining similar immunomodulatory patterns. The findings of our study imply the great potential of GMMA-based vaccine platform also against viral infectious diseases.
Collapse
Affiliation(s)
- Kai Hu
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Karnyart Samnuan
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Guanghui Wu
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Leigh Thorne
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Anthony R Fooks
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Lorraine M McElhinney
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Megan Golding
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
40
|
Suri K, D'Souza A, Huang D, Bhavsar A, Amiji M. Bacterial extracellular vesicle applications in cancer immunotherapy. Bioact Mater 2022; 22:551-566. [PMID: 36382022 PMCID: PMC9637733 DOI: 10.1016/j.bioactmat.2022.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/10/2022] [Accepted: 10/22/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer therapy is undergoing a paradigm shift toward immunotherapy focusing on various approaches to activate the host immune system. As research to identify appropriate immune cells and activate anti-tumor immunity continues to expand, scientists are looking at microbial sources given their inherent ability to elicit an immune response. Bacterial extracellular vesicles (BEVs) are actively studied to control systemic humoral and cellular immune responses instead of using whole microorganisms or other types of extracellular vesicles (EVs). BEVs also provide the opportunity as versatile drug delivery carriers. Unlike mammalian EVs, BEVs have already made it to the clinic with the meningococcal vaccine (Bexsero®). However, there are still many unanswered questions in the use of BEVs, especially for chronic systemically administered immunotherapies. In this review, we address the opportunities and challenges in the use of BEVs for cancer immunotherapy and provide an outlook towards development of BEV products that can ultimately translate to the clinic.
Collapse
Affiliation(s)
- Kanika Suri
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA,Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 20115, USA
| | - Di Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA,Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 20115, USA
| | - Aashray Bhavsar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA,Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115, USA,Corresponding author. Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Kashyap D, Panda M, Baral B, Varshney N, R S, Bhandari V, Parmar HS, Prasad A, Jha HC. Outer Membrane Vesicles: An Emerging Vaccine Platform. Vaccines (Basel) 2022; 10:1578. [PMID: 36298443 PMCID: PMC9610665 DOI: 10.3390/vaccines10101578] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2023] Open
Abstract
Vaccine adjuvants are substances that improve the immune capacity of a recombinant vaccine to a great extent and have been in use since the early 1900s; they are primarily short-lived and initiate antigen activity, mainly an inflammatory response. With the developing technologies and innovation, early options such as alum were modified, yet the inorganic nature of major vaccine adjuvants caused several side effects. Outer membrane vesicles, which respond to the stressed environment, are small nano-sized particles secreted by gram-negative bacteria. The secretory nature of OMV gives us many benefits in terms of infection bioengineering. This article aims to provide a detailed overview of bacteria's outer membrane vesicles (OMV) and their potential usage as adjuvants in making OMV-based vaccines. The OMV adjuvant-based vaccines can be a great benefactor, and there are ongoing trials for formulating OMV adjuvant-based vaccines for SARS-CoV-2. This study emphasizes engineering the OMVs to develop better versions for safety purposes. This article will also provide a gist about the advantages and disadvantages of such vaccines, along with other aspects.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Mrutyunjaya Panda
- Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, India
| | - Budhadev Baral
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Nidhi Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Sajitha R
- Amity Institute of Biotechnology, Amity University Noida, Amity 201313, India
| | - Vasundhra Bhandari
- Department of Biological Science, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | | | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| |
Collapse
|
42
|
Wang S, Guo J, Bai Y, Sun C, Wu Y, Liu Z, Liu X, Wang Y, Wang Z, Zhang Y, Hao H. Bacterial outer membrane vesicles as a candidate tumor vaccine platform. Front Immunol 2022; 13:987419. [PMID: 36159867 PMCID: PMC9505906 DOI: 10.3389/fimmu.2022.987419] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer represents a serious concern for human life and health. Due to drug resistance and the easy metastasis of tumors, there is urgent need to develop new cancer treatment methods beyond the traditional radiotherapy, chemotherapy, and surgery. Bacterial outer membrane vesicles (OMVs) are a type of double-membrane vesicle secreted by Gram-negative bacteria in the process of growth and life, and play extremely important roles in the survival and invasion of those bacteria. In particular, OMVs contain a large number of immunogenic components associated with their parent bacterium, which can be used as vaccines, adjuvants, and vectors to treat diseases, especially in presenting tumor antigens or targeted therapy with small-molecule drugs. Some OMV-based vaccines are already on the market and have demonstrated good therapeutic effect on the corresponding diseases. OMV-based vaccines for cancer are also being studied, and some are already in clinical trials. This paper reviews bacterial outer membrane vesicles, their interaction with host cells, and their applications in tumor vaccines.
Collapse
Affiliation(s)
- Shuming Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Jiayi Guo
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Yang Bai
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Cai Sun
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Yanhao Wu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Zhe Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Xiaofei Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Yanfeng Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Zhigang Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
| | - Yongmin Zhang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, China
| | - Huifang Hao
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, China
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, China
- *Correspondence: Huifang Hao,
| |
Collapse
|
43
|
van der Put RMF, Spies A, Metz B, Some D, Scherrers R, Pieters R, Danial M. Validation of an FFF-MALS Method to Characterize the Production and Functionalization of Outer-Membrane Vesicles for Conjugate Vaccines. Anal Chem 2022; 94:12033-12041. [PMID: 36007249 PMCID: PMC9453738 DOI: 10.1021/acs.analchem.2c01590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With the ongoing development of conjugate vaccines battling infectious diseases, there is a need for novel carriers. Although tetanus toxoid and CRM197 belong to the traditional carrier proteins, outer-membrane vesicles (OMVs) are an excellent alternative: in addition to their size, OMVs have self-adjuvanting properties due to the presence of genetically detoxified lipopolysaccharide (LPS) and are therefore ideal as a vaccine component or antigen carrier. An essential aspect of their development for vaccine products is characterization of OMVs with respect to size and purity. We report on the development of a field-flow fractionation multiangle light-scattering (FFF-MALS) method for such characterization. Here, we introduced NIST-traceable particle-size standards and BSA as a model protein to verify the precision of the size and purity analysis of the OMVs. We executed a validation program according to the principles provided in the ICH Guidelines Q2 (R1) to assess the quality attributes of the results obtained by FFF-MALS analysis. All validation characteristics showed excellent results with coefficients of variation between 0.4 and 7.32%. Estimation of limits of detection for hydrodynamic radius and particle concentration revealed that as little as 1 μg OMV still yielded accurate results. With the validated method, we further characterized a full downstream purification process of our proprietary OMV. This was followed by the evaluation of other purified OMVs from different bacterial origin. Finally, functionalizing OMVs with N-γ-(maleimidobutyryl)oxysuccinimide-ester (GMBS), generating ready-to-conjugate OMVs, did not affect the structural integrity of the OMVs and as such, they could be evaluated with the validated FFF-MALS method.
Collapse
Affiliation(s)
- Robert M F van der Put
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, NL-3508 TB Utrecht, The Netherlands.,Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Arnoud Spies
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Bernard Metz
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Daniel Some
- Wyatt Technology Corp., Santa Barbara, California 93117, United States
| | | | - Roland Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, NL-3508 TB Utrecht, The Netherlands
| | - Maarten Danial
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| |
Collapse
|
44
|
Alfini R, Brunelli B, Bartolini E, Carducci M, Luzzi E, Ferlicca F, Buccato S, Galli B, Lo Surdo P, Scarselli M, Romagnoli G, Cartocci E, Maione D, Savino S, Necchi F, Delany I, Micoli F. Investigating the Role of Antigen Orientation on the Immune Response Elicited by Neisseria meningitidis Factor H Binding Protein on GMMA. Vaccines (Basel) 2022; 10:1182. [PMID: 35893831 PMCID: PMC9331691 DOI: 10.3390/vaccines10081182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
GMMA are outer membrane vesicles (OMVs) released from Gram-negative bacteria genetically modified to enhance OMVs formation that have been shown to be optimal systems to enhance immunogenicity of protein antigens. Here, we selected Neisseria meningitidis factor H binding protein (fHbp) and used the conjugation chemistry as a tool to alter antigen orientation on GMMA. Indeed, fHbp was randomly linked to GMMA or selectively attached via the N-terminus to mimic native presentation of the protein on the bacterial surface. Interestingly, protein and peptide array analyses confirmed that antibodies induced by the selective and the random conjugates showed a pattern very similar to fHbp natively expressed on bacterial surfaces or to the recombinant protein mixed with GMMA, respectively. However, the two conjugates elicited antibodies with similar serum bactericidal activity against meningococcal strains, superior to the protein alone or physically mixed with GMMA. Presentation of fHbp on GMMA strongly enhances the functional immune response elicited by the protein but its orientation on the bacterial surface does not have an impact. This study demonstrates the flexibility of the GMMA platform as a display and delivery system for enhancing antigen immunogenicity and further supports the use of such promising technology for the development of effective vaccines.
Collapse
Affiliation(s)
- Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Brunella Brunelli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Erika Bartolini
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Enrico Luzzi
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Ferlicca
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Scilla Buccato
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Barbara Galli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Paola Lo Surdo
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Maria Scarselli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Giacomo Romagnoli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Elena Cartocci
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Domenico Maione
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Silvana Savino
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Isabel Delany
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| |
Collapse
|
45
|
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: 1.5] [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
|
46
|
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: 41] [Impact Index Per Article: 20.5] [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
|
47
|
Palmieri E, Kis Z, Ozanne J, Di Benedetto R, Ricchetti B, Massai L, Carducci M, Oldrini D, Gasperini G, Aruta MG, Rossi O, Kontoravdi C, Shah N, Mawas F, Micoli F. GMMA as an Alternative Carrier for a Glycoconjugate Vaccine against Group A Streptococcus. Vaccines (Basel) 2022; 10:1034. [PMID: 35891202 PMCID: PMC9324507 DOI: 10.3390/vaccines10071034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Group A Streptococcus (GAS) causes about 500,000 annual deaths globally, and no vaccines are currently available. The Group A Carbohydrate (GAC), conserved across all GAS serotypes, conjugated to an appropriate carrier protein, represents a promising vaccine candidate. Here, we explored the possibility to use Generalized Modules for Membrane Antigens (GMMA) as an alternative carrier system for GAC, exploiting their intrinsic adjuvant properties. Immunogenicity of GAC-GMMA conjugate was evaluated in different animal species in comparison to GAC-CRM197; and the two conjugates were also compared from a techno-economic point of view. GMMA proved to be a good alternative carrier for GAC, resulting in a higher immune response compared to CRM197 in different mice strains, as verified by ELISA and FACS analyses. Differently from CRM197, GMMA induced significant levels of anti-GAC IgG titers in mice also in the absence of Alhydrogel. In rabbits, a difference in the immune response could not be appreciated; however, antibodies from GAC-GMMA-immunized animals showed higher affinity toward purified GAC antigen compared to those elicited by GAC-CRM197. In addition, the GAC-GMMA production process proved to be more cost-effective, making this conjugate particularly attractive for low- and middle-income countries, where this pathogen has a huge burden.
Collapse
Affiliation(s)
- Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Zoltán Kis
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (Z.K.); (C.K.); (N.S.)
- Department of Chemical and Biological Engineering, The University of Sheffield, Mappin Street, Sheffield S1 3JD, UK
| | - James Ozanne
- The National Institute for Biological Standards and Control (NIBSC), South Mimms EN6 3QG, UK; (J.O.); (F.M.)
| | - Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Beatrice Ricchetti
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Luisa Massai
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Maria Grazia Aruta
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| | - Cleo Kontoravdi
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (Z.K.); (C.K.); (N.S.)
| | - Nilay Shah
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (Z.K.); (C.K.); (N.S.)
| | - Fatme Mawas
- The National Institute for Biological Standards and Control (NIBSC), South Mimms EN6 3QG, UK; (J.O.); (F.M.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (R.D.B.); (B.R.); (L.M.); (M.C.); (D.O.); (G.G.); (M.G.A.); (O.R.)
| |
Collapse
|
48
|
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: 41] [Impact Index Per Article: 20.5] [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
|
49
|
Huang W, Meng L, Chen Y, Dong Z, Peng Q. Bacterial outer membrane vesicles as potential biological nanomaterials for antibacterial therapy. Acta Biomater 2022; 140:102-115. [PMID: 34896632 DOI: 10.1016/j.actbio.2021.12.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/05/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023]
Abstract
Antibiotic therapy is one of the most important approaches against bacterial infections. However, the improper use of antibiotics and the emergence of drug resistance have compromised the efficacy of traditional antibiotic therapy. In this regard, it is of great importance and significance to develop more potent antimicrobial therapies, including the development of functionalized antibiotics delivery systems and antibiotics-independent antimicrobial agents. Outer membrane vesicles (OMVs), secreted by Gram-negative bacteria and with similar structure to cell-derived exosomes, are natural functional nanomaterials and known to play important roles in many bacterial life events, such as communication, biofilm formation and pathogenesis. Recently, more and more reports have demonstrated the use of OMVs as either active antibacterial agents or antibiotics delivery carriers, implying the great potentials of OMVs in antibacterial therapy. Herein, we aim to provide a comprehensive understanding of OMV and its antibacterial applications, including its biogenesis, biofunctions, isolation, purification and its potentials in killing bacteria, delivering antibiotics and developing vaccine or immunoadjuvants. In addition, the concerns in clinical use of OMVs and the possible solutions are discussed. STATEMENT OF SIGNIFICANCE: The emergence of antibiotic-resistant bacteria has led to the failure of traditional antibiotic therapy, and thus become a big threat to human beings. In this regard, developing more potent antibacterial approaches is of great importance and significance. Recently, bacterial outer membrane vesicles (OMVs), which are natural functional nanomaterials secreted by Gram-negative bacteria, have been used as active agents, drug carriers and vaccine adjuvant for antibacterial therapy. This review provides a comprehensive understanding of OMVs and summarizes the recent progress of OMVs in antibacterial applications. The concerns of OMVs in clinical use and the possible solutions are also discussed. As such, this review may guide the future works in antibacterial OMVs and appeal to both scientists and clinicians.
Collapse
Affiliation(s)
- Wenlong Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lingxi Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zaiquan Dong
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
50
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|