1
|
Feng R, Du M, Chen Z. Application of bacteria-mediated gene therapy in tumor treatment. Expert Opin Drug Deliv 2025:1-14. [PMID: 40325972 DOI: 10.1080/17425247.2025.2502638] [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: 01/06/2025] [Revised: 03/25/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION Gene therapy refers to the use of vectors to introduce target genes into target cells to exert a therapeutic effect on tumors. As a new type of tumor therapy, gene therapy has the advantage of precision and specificity. Excellent delivery vehicles have a major impact on the efficiency, precision and safety of gene therapy. Unlike traditional vectors, bacteria based on prokaryotes have the advantages of good targeting, large load, and simplicity. In addition, different types of bacteria also have characteristics that can be used in various scenarios. AREAS COVERED In this review, we searched the gene therapy-related literature in PubMed, mainly in the last five years, and compared the characteristics of different gene vectors, focusing on the bacterial gene therapy and aiming to explore excellent bacterial gene therapy programs. EXPERT OPINION Compared with traditional tumor gene therapy vectors, bacteria have many advantages, such as good targeting, large carrying capacity, and simple production. Meanwhile, the combination of artificial intelligence technology, bacterial imaging probe technology and suicide genes will be expected to control the bacterial therapy process, improve the safety of treatment, and promote the translational application of bacterial gene therapy.
Collapse
Affiliation(s)
- Renjie Feng
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Institute for Future Sciences, University of South China, Changsha, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Institute for Future Sciences, University of South China, Changsha, Hunan, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Institute for Future Sciences, University of South China, Changsha, Hunan, China
- Department of Medical Imaging, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
2
|
Senevirathne A, Lloren KKS, Aganja RP, Kwon J, Lee JH. Transforming bacterial pathogens into wonder tools in cancer immunotherapy. Mol Ther 2025; 33:866-882. [PMID: 39825565 PMCID: PMC11897747 DOI: 10.1016/j.ymthe.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/02/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
Cancer immunotherapy has revolutionized cancer treatment due to its precise, target-specific approach compared with conventional therapies. However, treating solid tumors remains challenging as these tumors are inherently immunosuppressive, and their tumor microenvironment (TME) often limits therapeutic efficacy. Interestingly, certain bacterial species offer a promising alternative by exhibiting an innate ability to target and proliferate within tumor environments. Bacterial structural and functional components can activate innate and adaptive immune responses, creating tumor-suppressive conditions that reduce tumor mass. Additionally, bacteria can deliver effector molecules directly into tumor cells, inducing apoptotic and necrotic cell death. Despite their potential, the use of bacteria in cancer immunotherapy poses risks due to possible toxicities and unpredictable in vivo behavior. Advances in genetic engineering have addressed these concerns by enabling the development of attenuated bacterial strains with enhanced anticancer properties for safer medical applications. This review highlights the role of bacteria in TME modulation, recent strategies to bioengineer bacterial pathogens as therapeutic tools, and the synergistic effects of combining bacteria with other immunotherapies. It also discusses the challenges and prospects of translating this innovative approach into clinical practice, offering a comprehensive overview of bacteria-based cancer immunotherapy's potential to reshape the future of cancer treatment.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do 54596, Republic of Korea
| | - Khristine Kaith S Lloren
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do 54596, Republic of Korea
| | - Ram Prasad Aganja
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do 54596, Republic of Korea
| | - Jun Kwon
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do 54596, Republic of Korea.
| |
Collapse
|
3
|
Yang J, Chen R, Sun M, Yuan R, Xiao YF, Sun Y, Zhou G, Wen Y, Wang Y, Wu R, Zhao Q, Du S, Cao S, Huang X. Development and immunogenicity evaluation of attenuated Salmonella typhimurium delivering porcine Deltacoronavirus S1 gene. Int J Biol Macromol 2025; 288:138615. [PMID: 39674474 DOI: 10.1016/j.ijbiomac.2024.138615] [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: 11/13/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is a swine enteropathogenic coronavirus that causes severe diarrhea in piglets, the development of novel vaccines is of great value in the prevention and control of PDCoV. Here, we selected attenuated Salmonella typhimurium SL7207 to deliver pVAX1-S1, resulting in the oral vaccine strain, SL7207 (pVAX1-S1). In immunized mice, SL7207 (pVAX1-S1) induced PDCoV-specific humoral IgG, IgA, neutralizing antibodies, mucosal sIgA, up-regulation of CD8+ T cells, and increased levels of Th1 cytokines (IFN-γ and IL-2). In piglets, SL7207 (pVAX1-S1) induced high levels of PDCoV-specific humoral IgG and neutralizing antibodies but no detectable IgA, and only low levels of mucosal sIgA. SL7207 (pVAX1-S1) also promoted T cell differentiation into CD4+ and CD8+ T cells, and increased the expression level of IFN-γ and IL-4 in peripheral blood. Challenge experiments in piglets showed SL7207 (pVAX1-S1) alleviated diarrhea, decreased fecal virus load and intestinal lesions compared with control groups. In conclusion, this study systematically evaluated the immunogenicity and feasibility of attenuated Salmonella typhimurium delivering PDCoV S1 gene, which will provide helpful reference information for further exploration of novel PDCoV oral vaccine.
Collapse
Affiliation(s)
- Junpeng Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Mengke Sun
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rong Yuan
- Chengdu Livestork and Poultry Genetic Resources Protection Center, China
| | - Ying Feng Xiao
- Chengdu Livestork and Poultry Genetic Resources Protection Center, China
| | - Ying Sun
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Guiping Zhou
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu 611130, China; International joint Research Center for Animal Disease Prevention and control of Sichuan Province, Chengdu 611130, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu 611130, China; International joint Research Center for Animal Disease Prevention and control of Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
4
|
Yang Y, Yuan H, Zhang Y, Luan J, Wang H. Progress in African Swine Fever Vector Vaccine Development. Int J Mol Sci 2025; 26:921. [PMID: 39940691 PMCID: PMC11816837 DOI: 10.3390/ijms26030921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
African swine fever (ASF) is a highly lethal, infectious, hemorrhagic fever disease, characterized by an acute mortality rate approaching 100%. It is highly contagious, and results in significant losses to the global hog industry as it spreads. Despite incremental progress in research on the African swine fever virus (ASFV), a safe and effective commercial vaccine has yet to be developed. Vector vaccines, a promising type of vaccine, offer unique advantages, and are a primary focus in ASFV vaccine research. This paper focuses on the characteristics of viral, bacterial, and yeast vector vaccines; elucidates the immunological mechanisms associated with antigens; lists the types of antigens that have significant potential; discusses the feasibility of using exogenously expressed cytokines to enhance the protective power of vector vaccines; and, finally, discusses the types of vectors that are commonly used and the latest advances in this field.
Collapse
Affiliation(s)
| | | | | | | | - Hailong Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, Shandong University, Qingdao 266237, China; yangyue-@mail.sdu.edu.cn (Y.Y.); (H.Y.); (Y.Z.); (J.L.)
| |
Collapse
|
5
|
Park JY, Senevirathne A, Lee JH. Development of a candidate vaccine against severe fever with thrombocytopenia syndrome virus using Gn/Gc glycoprotein via multiple expression vectors delivered by attenuated Salmonella confers effective protection in hDC-SIGN transduced mice. Vaccine 2025; 43:126524. [PMID: 39547019 DOI: 10.1016/j.vaccine.2024.126524] [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: 09/02/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
In this study, we developed two plasmid constructs, pJHL270 and pJHL305, for the dual expression of Severe Fever with Thrombocytopenia Syndrome Virus (SFTSV) Gn and Gc glycoproteins in both prokaryotic and eukaryotic systems. The constructs feature a prokaryotic expression controlled by the Ptrc promoter and a eukaryotic expression driven by the cytomegalovirus promoter and Semliki Forest Virus RNA-dependent RNA polymerase. The Gn/Gc antigenic epitope was derived from consensus sequences of 12 SFTSV M segments collected in South Korea and designed for optimal antigen expression. The full antigen was expressed eukaryotically for post-translational modifications, while the epitope construct was expressed prokaryotically. These constructs were electroporated into an attenuated Salmonella Typhimurium strain (JOL2500) for plasmid delivery, resulting in JOL3042 and JOL3045. Successful expression was confirmed via qRT-PCR and western blot analysis. Mice immunized with JOL3042 showed antibody responses as early as two weeks, while JOL3045 elicited responses at six weeks, skewed toward a Th1 response initially, later balancing with Th2. Flow cytometry revealed significant CD3+CD4+ and CD3+CD8+ T-cell responses. Both constructs generated neutralizing antibodies, and a challenge study indicated significant reductions in viral loads in the serum, liver, and spleen of vaccinated mice, demonstrating the effectiveness of the Salmonella-mediated delivery system against SFTSV infection. The outcome of the current study may pave the way to develop a safer and more effective Salmonella-mediated vaccine against lethal SFTSV infection in vulnerable populations.
Collapse
MESH Headings
- Animals
- Phlebovirus/immunology
- Phlebovirus/genetics
- Mice
- Salmonella typhimurium/immunology
- Salmonella typhimurium/genetics
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Severe Fever with Thrombocytopenia Syndrome/prevention & control
- Severe Fever with Thrombocytopenia Syndrome/immunology
- Genetic Vectors/immunology
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/genetics
- Mice, Inbred BALB C
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/genetics
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Plasmids/genetics
- Plasmids/immunology
- Glycoproteins/immunology
- Glycoproteins/genetics
- Receptors, Cell Surface
Collapse
Affiliation(s)
- Ji-Young Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea.
| |
Collapse
|
6
|
Aganja RP, Kim IS, Tae HJ, Lee JH. Expression and delivery of HA1-M2e antigen using an innovative attenuated Salmonella-mediated delivery system confers promising protection against H9N2 avian influenza challenge. Poult Sci 2025; 104:104602. [PMID: 39631285 PMCID: PMC11665344 DOI: 10.1016/j.psj.2024.104602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/28/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
This study explores a dual expression vector system for delivering prokaryotic and eukaryotic antigens to improve conventional vaccination strategies. To enhance immune protection against H9N2 avian influenza virus (AIV), which threatens poultry and humans, we used the previously constructed pJHL270 and pJHL305 plasmids with the Ptrc and CMV promoters to stimulate MHC class II and I responses through exogenous and endogenous antigenic presentation. Salmonella Gallinarum (SG), a delivery vector, was engineered to have defective lipopolysaccharide structures through lon, pagL, and rfaL deletion. It demonstrated a safety profile with lower induction of inflammatory cytokines than the wild-type strain. Bioinformatics tools predicted that the HA1 and M2e sequences, which were designed as consensus sequences of South Korean strains (2000-2021), would have high antigenicity and favorable structures. In vitro expression of the vaccine constructs was validated by western blotting. Birds immunized with attenuated SG harboring pJHL270 (JOL3025) or pJHL305 (JOL3027) containing HA-M2e showed significant increases in serum IgY and mucosal IgA antibodies, indicating strong humoral and mucosal immune responses, comparable with inactivated commercial vaccine. Post-immunization, we found a substantial rise in the hemagglutination inhibition titer, suggesting effective prevention of viral attachment and robust cell-mediated immunity, with a 1.96-fold and 2.80-fold increase in CD4+ and CD8+ T cells, respectively, for JOL3025 and a 1.75-fold and 2.49-fold increase for JOL3027. Furthermore, MHC class I and II expression increased 1.35-fold and 1.63-fold, for JOL3025, and 1.61-fold and 1.68-fold, respectively, for JOL3027. The IL-4 and IFN-γ levels were elevated, indicating a balanced Th-1 and Th-2 response. Post-challenge, birds immunized with vaccine candidates or the commercial vaccine exhibited minimal to no clinical signs, reduced lesions, lower lung viral titers, and negligible impacts on egg production compared to controls. In conclusion, both plasmids successfully delivered HA1-M2e immunogens through the engineered SG strains, eliciting strong humoral, mucosal, and cell-mediated immune responses and co-stimulating MHC class I and II antigen presentation pathways to provide effective protection against H9N2 AIV with minimal adverse effects.
Collapse
Affiliation(s)
- Ram Prasad Aganja
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, Iksan 54596, Republic of Korea; College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Campus, Iksan 54596, Republic of Korea
| | - In-Shik Kim
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Campus, Iksan 54596, Republic of Korea
| | - Hyun-Jin Tae
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Campus, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, Iksan 54596, Republic of Korea; College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Campus, Iksan 54596, Republic of Korea.
| |
Collapse
|
7
|
Lloren KKS, Senevirathne A, Lee JH. Advancing vaccine technology through the manipulation of pathogenic and commensal bacteria. Mater Today Bio 2024; 29:101349. [PMID: 39850273 PMCID: PMC11754135 DOI: 10.1016/j.mtbio.2024.101349] [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: 08/15/2024] [Revised: 10/30/2024] [Accepted: 11/15/2024] [Indexed: 01/25/2025] Open
Abstract
Advancements in vaccine technology are increasingly focused on leveraging the unique properties of both pathogenic and commensal bacteria. This revolutionary approach harnesses the diverse immune modulatory mechanisms and bacterial biology inherent in different bacterial species enhancing vaccine efficacy and safety. Pathogenic bacteria, known for their ability to induce robust immune responses, are being studied for their potential to be engineered into safe, attenuated vectors that can target specific diseases with high precision. Concurrently, commensal bacteria, which coexist harmlessly with their hosts and contribute to immune system regulation, are also being explored as novel delivery systems and in microbiome-based therapy. These bacteria can modulate immune responses, offering a promising avenue for developing effective and personalized vaccines. Integrating the distinctive characteristics of pathogenic and commensal bacteria with advanced bacterial engineering techniques paves the way for innovative vaccine and therapeutic platforms that could address a wide range of infectious diseases and potentially non-infectious conditions. This holistic approach signifies a paradigm shift in vaccine development and immunotherapy, emphasizing the intricate interplay between the bacteria and the immune systems to achieve optimal immunological outcomes.
Collapse
Affiliation(s)
- Khristine Kaith S. Lloren
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| |
Collapse
|
8
|
Lloren KKS, Sivasankar C, Lee JH. Comparative immunogenic and immunoprotective activities of PCV2d Cap and Rep antigens delivered by an efficient eukaryotic expression system engineered into a Salmonella vaccine vector. Vet Microbiol 2024; 295:110151. [PMID: 38870752 DOI: 10.1016/j.vetmic.2024.110151] [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: 03/07/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Porcine circovirus type 2 (PCV2) stands as a predominant etiological agent in porcine circovirus-associated diseases. To manage the spread of the disease, it is necessary to develop a next-generation vaccine expressing PCV2 antigens that target the prevailing genotype such as PCV2d. A bacterial-mediated vaccine delivery by live-attenuated Salmonella has attracted interest for its low-cost production and highly effective vaccine delivery. Thus, in this study, we utilized the advantages of the Salmonella-mediated vaccine delivery by cloning PCV2d cap and rep into a eukaryotic expression plasmid pJHL204 and electroporation into an engineered live-attenuated Salmonella Typhimurium JOL2500 (Δlon, ΔcpxR, ΔsifA, Δasd). The eukaryotic antigen expression by JOL2995 (p204:cap) and JOL2996 (p204:rep) was confirmed in vitro and in vivo which showed efficient antigen delivery. Furthermore, vaccination of mice model with the vaccine candidates elicited humoral and cell-mediated immune responses as depicted by high levels of PCV2-specific antibodies, CD4+ and CD8+ T cells, and neutralizing antibodies, especially by JOL2995 (p204:cap) which correlated with the significant decrease in the viral load in PCV2d-challenged mice. Interestingly, JOL2996 (p204:rep) may not have elicited high levels of neutralizing antibodies and protective efficacy, but it elicited considerably higher cell-mediated immune responses. This study demonstrated Salmonella-mediated vaccine delivery system coupled with the eukaryotic expression vector can efficiently deliver and express the target PCV2d antigens for strong induction of immune response and protective efficacy in mice model, further supporting the potential application of the Salmonella-mediated vaccine delivery system as an effective novel approach in vaccine strategies for PCV2d.
Collapse
Affiliation(s)
- Khristine Kaith S Lloren
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea.
| |
Collapse
|
9
|
Sivasankar C, Hewawaduge C, Muthuramalingam P, Lee JH. Tumor-targeted delivery of lnc antisense RNA against RCAS1 by live-attenuated tryptophan-auxotrophic Salmonella inhibited 4T1 breast tumors and metastasis in mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102053. [PMID: 37941832 PMCID: PMC10628790 DOI: 10.1016/j.omtn.2023.102053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Emerging chemo- and radiotherapy resistance exacerbated the cancer risk and necessitated novel treatment strategies. Although RNA therapeutics against pro-oncogenic genes are highly effective, tumor-specific delivery remains a barrier to the implementation of this valuable tool. In this study, we report a tryptophan-auxotrophic Salmonella typhimurium strain as an onco-therapeutic delivery system with tumor-targeting ability using 4T1 mice breast-cancer model. The receptor-binding cancer antigen expressed on SiSo cell (RCAS1) is a cancer-specific protein that induces the apoptosis of peripheral lymphocytes and confers tumor immune evasion. We designed a long non-coding antisense-RNA against RCAS1 (asRCAS1) and delivered by Salmonella using a non-antibiotic, auxotrophic-selective, eukaryotic expression plasmid, pJHL204. After in vivo tumor-to-tumor passaging, the JOL2888 (ΔtrpA, ΔtrpE, Δasd + asRCAS1) strain exhibited high sustainability in tumors, but did not last in healthy organs, thereby demonstrating tumor specificity and safety. RCAS1 inhibition in the tumor was confirmed by western blotting and qPCR. In mice, JOL2888 treatment reduced tumor-associated macrophages, improved the T cell population, elicited cell-mediated immunity, and suppressed cancer-promoting genes. Consequently, the JOL2888 treatment significantly decreased the tumor volume by 80%, decreased splenomegaly by 30%, and completely arrested lung metastasis. These findings highlight the intrinsic tumor-targeting ability of tryptophan-auxotrophic Salmonella for delivering onco-therapeutic macromolecules.
Collapse
Affiliation(s)
- Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | | | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| |
Collapse
|
10
|
Hewawaduge C, Kwon J, Sivasankar C, Park JY, Senevirathne A, Lee JH. Salmonella delivers H9N2 influenza virus antigens via a prokaryotic and eukaryotic dual-expression vector and elicits bivalent protection against avian influenza and fowl typhoid. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105058. [PMID: 37714394 DOI: 10.1016/j.dci.2023.105058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
The H9N2 avian influenza virus significantly affects the health of poultry and humans. We identified a prokaryotic and eukaryotic dual-expression vector system, pJHL270, that can provide simultaneous MHC class I and II stimulation of the host immune system, and we designed vaccine antigens by selecting the consensus HA1 sequence and M2e antigens from H9N2 virus circulating in South Korea from 2000 to 2021. The genes were cloned into the pJHL270 vector, and the cloned plasmid was delivered by a live-attenuated Salmonella Gallinarum (SG) strain. The immunity and protective efficacy of the SG-based H9N2 vaccine construct, JOL2922, against avian influenza and fowl typhoid (FT) were evaluated. The Ptrc and CMV promoters conferred antigen expression in prokaryotic and eukaryotic cells to induce balanced Th-1/Th-2 immunity. Chickens immunized with JOL2922 yielded high antigen-specific humoral and mucosal immune responses. qRT-PCR revealed that the strain generated polyfunctional IFN-γ and IL-4 secretion in immunized chickens. Furthermore, a FACS analysis showed increased CD3CD4+ and CD3CD8+ T-cell subpopulations following immunization. Peripheral Blood Mononuclear Cells (PBMCs) harvested from the immunized chickens significantly increased MHC class I and II expression, 3.5-fold and 2.5-fold increases, respectively. Serum collected from the immunized groups had an evident hemagglutinin inhibition titer of ≥6 log2. Immunization reduced the lung viral titer by 3.8-fold within 5 days post-infection. The strain also generated SG-specific humoral and cellular immune responses. The immunized birds all survived a virulent SG wild-type challenge. In addition, the bacterial burden was reduced by 2.7-fold and 2.1-fold in spleen and liver tissue, respectively, collected from immunized chickens. Our data indicate that an attenuated SG strain successfully delivered the dual-expression vector system and co-stimulated MHC class I and II antigen presentation pathways via exogenous and endogenous antigen presentation, thereby triggering a balanced Th-1/Th-2-based immune response and conferring effective protection against avian influenza and FT.
Collapse
Affiliation(s)
- Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Jun Kwon
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Ji-Young Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea.
| |
Collapse
|
11
|
Lloren KKS, Lee JH. Live-Attenuated Salmonella-Based Oral Vaccine Candidates Expressing PCV2d Cap and Rep by Novel Expression Plasmids as a Vaccination Strategy for Mucosal and Systemic Immune Responses against PCV2d. Vaccines (Basel) 2023; 11:1777. [PMID: 38140182 PMCID: PMC10748173 DOI: 10.3390/vaccines11121777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Oral vaccines are highly envisaged for veterinary applications due to their convenience and ability to induce protective mucosal immunity as the first line of defense. The present investigation harnessed live-attenuated Salmonella Typhimurium to orally deliver novel expression vector systems containing the Cap and Rep genes from porcine circovirus type 2 (PCV2), a significant swine pathogen. The antigen expression by the vaccine candidates JOL2885 and JOL2886, comprising eukaryotic pJHL204 and pro-eukaryotic expression pJHL270 plasmids, respectively, was confirmed by Western blot and IFA. We evaluated their immunogenicity and protective efficacy through oral vaccination in a mouse model. This approach elicited both mucosal and systemic immunity against PCV2d. Oral administration of the candidates induced PCV2-specific sIgA, serum IgG antibodies, and neutralizing antibodies, resulting in reduced viral loads in the livers and lungs of PCV2d-challenged mice. T-lymphocyte proliferation and flow-cytometry assays confirmed enhanced cellular immune responses after oral inoculation. The synchronized elicitation of both Th1 and Th2 responses was also confirmed by enhanced expression of TNF-α, IFN-γ, IL-4, MHC-I, and MHC-II. Our findings highlight the effectiveness and safety of the constructs with an engineered-attenuated S. Typhimurium, suggesting its potential application as an oral PCV2 vaccine candidate.
Collapse
Affiliation(s)
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea;
| |
Collapse
|
12
|
Park JY, Hewawaduge C, Sivasankar C, Lloren KKS, Oh B, So MY, Lee JH. An mRNA-Based Multiple Antigenic Gene Expression System Delivered by Engineered Salmonella for Severe Fever with Thrombocytopenia Syndrome and Assessment of Its Immunogenicity and Protection Using a Human DC-SIGN-Transduced Mouse Model. Pharmaceutics 2023; 15:pharmaceutics15051339. [PMID: 37242581 DOI: 10.3390/pharmaceutics15051339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Currently, there are no commercial vaccines or therapeutics against severe fever with thrombocytopenia syndrome (SFTS) virus. This study explored an engineered Salmonella as a vaccine carrier to deliver a eukaryotic self-mRNA replicating vector, pJHL204. This vector expresses multiple SFTS virus antigenic genes for the nucleocapsid protein (NP), glycoprotein precursor (Gn/Gc), and nonstructural protein (NS) to induce host immune responses. The engineered constructs were designed and validated through 3D structure modeling. Western blot and qRT-PCR analyses of transformed HEK293T cells confirmed the delivery and expression of the vaccine antigens. Significantly, mice immunized with these constructs demonstrated a cell-mediated and humoral response as balanced Th1/Th2 immunity. The JOL2424 and JOL2425 delivering NP and Gn/Gc generated strong immunoglobulin IgG and IgM antibodies and high neutralizing titers. To further examine the immunogenicity and protection, we utilized a human DC-SIGN receptor transduced mouse model for SFTS virus infection by an adeno-associated viral vector system. Among the SFTSV antigen constructs, the construct with full-length NP and Gn/Gc and the construct with NP and selected Gn/Gc epitopes induced robust cellular and humoral immune responses. These were followed by adequate protection based on viral titer reduction and reduced histopathological lesions in the spleen and liver. In conclusion, these data indicate that recombinant attenuated Salmonella JOL2424 and JOL2425 delivering NP and Gn/Gc antigens of SFTSV are promising vaccine candidates that induce strong humoral and cellular immune responses and protection against SFTSV. Moreover, the data proved that the hDC-SIGN transduced mice as a worthy tool for immunogenicity study for SFTSV.
Collapse
Affiliation(s)
- Ji-Young Park
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Chandran Sivasankar
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Khristine Kaith S Lloren
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Byungkwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Mi Young So
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| |
Collapse
|
13
|
Bhilare KD, Jawalagatti V, Alam MJ, Chen B, Kim B, Lee JH, Kim JH. Immune response following safer administration of recombinant Salmonella Typhimurium harboring ASFV antigens in pigs. Vet Immunol Immunopathol 2023; 259:110596. [PMID: 37119725 DOI: 10.1016/j.vetimm.2023.110596] [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: 02/02/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/01/2023]
Abstract
African swine fever virus (ASFV) is a contagious epizootic pathogen adversely affecting porcine industry in Asian and European countries. Till date, 8 serotypes and 24 genotypes of the virus have been reported. Few live attenuated virus vaccine studies have reported to provide complete protection against ASFV infection but biohazard concern still remain. Recombinant subunit antigens are capable of providing cellular and humoral immunity in porcine, but not a single vaccine has hit the market yet. In the present study, we attempted to use recombinant Salmonella Typhimurium JOL912 strain harboring ASFV antigens (rSal-ASFV) to investigate its immunostimulant effect in porcine. Post intramuscular administration, we observed significant increment in the levels of helper T cells, cytotoxic T cells, natural killer (NK) cells, and immunoglobulin (i.e. IgG, IgA, and IgM) levels in rSal-ASFV treated groups. Further RT-PCR analysis indicated the increased expression of MHC-I, MHC-II, CD80/86, NK cell receptors (NKp30, NKp44, and NKp46) and cytokines while ELIspot analysis revealed significant production of IFN-γ in rSal-ASFV treated groups. Taken together, we are able to demonstrate that rSal-ASFV could elicit a non-specific cellular as well as humoral immune response. However, additional antigen specific immunity data is needed to evaluate its efficacy. Intramuscular administration of rSal-ASFV was found to be safe and immunostimulant in nature without any side-effects and may serve as an excellent option for in-vivo antigen delivery in pigs.
Collapse
Affiliation(s)
- Kiran D Bhilare
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - Vijay Jawalagatti
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - Md Jahangir Alam
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - Baicheng Chen
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - John-Hwa Lee
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan-city Pincode-54596, Jeollabuk-Do, Republic of Korea.
| |
Collapse
|
14
|
Salmonella-mediated oral delivery of multiple-target vaccine constructs with conserved and variable regions of SARS-CoV-2 protect against the Delta and Omicron variants in hamster. Microbes Infect 2023; 25:105101. [PMID: 36657635 PMCID: PMC9841750 DOI: 10.1016/j.micinf.2023.105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/07/2022] [Accepted: 01/10/2023] [Indexed: 01/17/2023]
Abstract
Since the emergence of the pandemic COVID19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the development of vaccines has been the prime strategy to control the disease transmission. Most of the developed vaccines target the spike protein, however, the emerging variants have alterations, particularly at the same region which may pose resistance to neutralizing antibodies. In this study, we explored the variable and conserved regions of SARS-CoV-2 as a potential inclusion in a multiple-target vaccine with the exploitation of Salmonella-based vector for oral mRNA vaccine against Delta and Omicron variants. Increased IgG and IgA levels imply the induction of humoral response and the CD4+, CD8+ and IFN-γ+ sub-population level exhibits cell-mediated immune responses. The degree of CD44+ cells indicates the induction of memory cells corresponding to long-term immune responses. Furthermore, we assessed the protective efficacy of the vaccines against the Delta and Omicron variants in the hamster model. The vaccine constructs induced neutralizing antibodies and protected the viral-challenged hamsters with significant decrease in lung viral load and reduced histopathological lesions. These results reinforce the use of the conserved and variable regions as potential antigen targets of SARS-CoV-2 as well as the exploitation of bacteria-mediated delivery for oral mRNA vaccine development.
Collapse
|
15
|
Aganja RP, Sivasankar C, Hewawaduge C, Lee JH. Safety assessment of compliant, highly invasive, lipid A-altered, O-antigen-defected Salmonella strains as prospective vaccine delivery systems. Vet Res 2022; 53:76. [PMID: 36183131 PMCID: PMC9526937 DOI: 10.1186/s13567-022-01096-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
In the present study, two prospective Salmonella delivery strains, JOL2782 and JOL2837, were developed by gene deletions of lon and cpxR, which are related to cellular adhesion and intracellular survival. Additionally, sifA deletion was introduced for JOL2782, which confers immune susceptibility and improves antigen delivery. Similarly, the rfaL deletion and lpxE substitution for pagL were accomplished in JOL2837 to reduce virulence and endotoxicity. Thus, enhanced adhesion and invasion and reduced intracellular survival were attained. Furthermore, aspartic acid auxotrophic (asd) was deleted to impose Darwinian selection on retention of the foreign antigen-expressing plasmid. Both delivery strains induced sufficient cytokine expression, but the level was significantly lower than that of the wild-type strain; the lowest cytokine expression was induced by the JOL2837 strain, indicating reduced endotoxicity. In parallel, IgG production was significantly enhanced by both delivery strains. Thus, the innate and adaptive immunogenicity of the strains was ensured. The environmental safety of these strains was ascertained through faecal dissemination assays. The nonpathogenicity of these strains to the host was confirmed by body weight monitoring, survival assays, and morphological and histological assessments of the vital organs. The in vitro assay in murine and human cell lines and in vivo safety assessments in mice suggest that these novel strains possess safety, invasiveness, and immunogenicity, making them ideal delivery strains. Overall, the results clearly showed that strain JOL2782 with sifA deletion had higher invasiveness, demonstrating superior vaccine deliverability, while JOL2837 with lpxE substitution for pagL and rfaL deletion had outstanding safety potential with drastically abridged endotoxicity.
Collapse
Affiliation(s)
- Ram Prasad Aganja
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Chandran Sivasankar
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea.
| |
Collapse
|