1
|
Castillo JG, Fernandez S, Campbell T, Gonzalez-Ventura D, Williams J, Ybarra J, Hernandez NF, Wells E, Portnoy DA, DuPage M. Cellular mechanisms underlying beneficial versus detrimental effects of bacterial antitumor immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.15.580555. [PMID: 39975413 PMCID: PMC11838217 DOI: 10.1101/2024.02.15.580555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Bacteria engineered to express tumor antigens as a cancer vaccine have yielded mixed results. Here, we utilized an attenuated strain of Listeria monocytogenes ( ΔactA, Lm ) that does not express tumor antigen to explore the immunological response to Listeria itself in the context of intravenous (IV), intratumoral (IT), or a combination of IV+IT administration into tumor-bearing mice. Unexpectedly, we found that Lm persisted in tumors of immune competent mice, regardless of the administration route. While IT Lm alone led to the recruitment of immunosuppressive immune cells that promoted tumor growth, IV Lm followed by IT Lm controlled tumor growth. IV Lm vaccination generated a pool of anti- Lm cytotoxic CD8 T cells that killed Lm -infected non-tumor cells to control tumor growth both indirectly, by limiting cancer cell proliferation, and directly, by enhancing tumor-specific T cell responses. Our findings reveal a differential impact of IT Lm administration on tumor progression that depends on the presence of anti- Lm CD8 T cells, which alone are sufficient to promote therapeutic efficacy.
Collapse
|
2
|
Sun J, Wang J, Jiang X, Xia J, Han Y, Chen M, Xu J, Deng S, Cheng C, Song H. LADS: a powerful vaccine platform for cancer immunotherapy and prevention. BMC Biol 2024; 22:291. [PMID: 39696249 DOI: 10.1186/s12915-024-02086-7] [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: 05/07/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The intracellular bacterium Listeria monocytogenes is an attractive vector for cancer immunotherapy as it can effectively deliver tumor antigens to antigen-presenting cells, leading to a robust antitumor response. RESULTS In this study, we developed a novel vaccine platform called Listeria-based Live Attenuated Double Substitution (LADS), which involves introducing two amino acid substitutions (N478AV479A) into the virulence factor listeriolysin O (LLO). LADS is a safe vaccine platform, with an attenuation of nearly 7000-fold, while retaining complete immunogenicity due to the absence of deletion of any virulence factors. We developed two LADS-based vaccines, LADS-E7 and LADS-AH1, which deliver the human papillomavirus (HPV) type 16 E7 oncoprotein and murine colon carcinoma immunodominant antigen AH1, respectively. Treatment with LADS-E7 or LADS-AH1 significantly inhibited and regressed established tumors, while also dramatically increasing the populations of tumor-infiltrated antigen-specific CD8+ T cells. RNA-sequencing analysis of tumor tissue samples revealed that LADS-E7 altered the expression of genes related to the immune response. Moreover, intratumoral injection of LADS-based vaccines induced strong antitumor responses, generating systemic antitumor responses to control distant tumor growth. Encouragingly, LADS-E7 or LADS-AH1 immunization effectively prevented tumor formation and growth. CONCLUSIONS Our findings demonstrate that LADS-based vaccines represent a more powerful platform for the development of immunotherapeutic and preventive vaccines against cancers and infectious diseases.
Collapse
Affiliation(s)
- Jing Sun
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Jing Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Xin Jiang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Jing Xia
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Yue Han
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Mianmian Chen
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Jiali Xu
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Simin Deng
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China
| | - Changyong Cheng
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China.
| | - Houhui Song
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine, Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province, 311300, China.
| |
Collapse
|
3
|
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
|
4
|
欧 倩, 陈 昭, 唐 静, 陈 梦, 张 云, 汪 川. [ Listeria Balanced Lethal Systems Expressing Cervical Cancer Antigen Genes: Construction and Basic Biological Characteristics]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1159-1166. [PMID: 38162066 PMCID: PMC10752777 DOI: 10.12182/20231160210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Indexed: 01/03/2024]
Abstract
Objective To construct Listeria monocytogenes (LM) and Listeria ivanovii (LI) balanced lethal systems expressing cervical cancer antigens, to study their basic biological characteristics, and to provide reference data for the immunotherapy of cervical cancer. Methods Through seamless cloning via in vitro ligation kit, the HPV16 E6E7 fusion protein antigen gene constructed in our lab was spliced to the complement plasmid pCWgfp-LM dal-Amp that contained the nutritional gene dal. Then, we replaced the ampicillin (Amp) resistance gene of the complement plasmid with the asd nutrition gene. The ligation reaction mixture was transformed into Escherichia coli (E. coli) recipient bacteria DH5αΔasd and the complement plasmid pCWgfp-E6E7-LM dal-Ampfree, which expressed cervical cancer antigens and had no Amp resistance, was obtained by nutrition screening from the E. coli DH5αΔasd. The plasmid pCWgfp-E6E7-LM dal-Ampfree was complemented into LMΔdd and LIΔdd, the attenuated nutrition-deficient Listeria strains with the virulence genes actA and plcB and nutrition genes dal and dat deleted by electroporation, thereby obtaining LM and LI balanced lethal systems expressing cervical cancer antigen genes. The in vitro growth of the strains was observed. Western blot was performed to examine the status of antigen protein expression. PCR was performed to measure the in vitro passage stability of complement plasmid pCWgfp-E6E7-LM dal-Ampfree. Their basic biological characteristics were examined by biochemical reaction tests and hemolysis assay. Results Two Listeria balanced lethal systems expressing cervical cancer antigen were successfully constructed. The HPV16 type E6E7 fusion protein was successfully expressed in the two Listeria balanced lethal systems. pCWgfp-E6E7-LM dal-Ampfree, the positive plasmid expressing cervical cancer antigen, maintained stable existence in the two Listeria balanced lethal systems. The two Listeria balanced lethal systems expressing cervical cancer antigen showed significantly better recovery growth in comparison with Listeria nutrition deficiency strains. The results of biochemical reaction tests showed that most of the biochemical reaction of the two Listeria balanced lethal systems expressing cervical cancer antigen were consistent with those of Listeria attenuated strains. The two Listeria balanced lethal systems expressing cervical cancer antigen still maintained the hemolytic ability, although their hemolytic ability was slightly inferior to that of the Listeria balanced lethal systems not expressing cervical cancer antigen and the Listeria attenuated strains. Conclusion The two Listeria balanced lethal systems expressing cervical cancer antigen genes are constructed successfully. They display normal in vitro growth. The complement plasmid pCWgfp-E6E7-LM dal-Ampfree can maintain stable existence in vitro, showing little change in its biochemical characteristics and hemolytic ability. Further research should be conducted to investigate the potential of these two recombinant strains to be used as candidate strains for cervical cancer therapeutic vaccine.
Collapse
Affiliation(s)
- 倩 欧
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
- 深圳市生医联盟生物科技集团有限公司 (深圳 518057)Shenzhen Biomed Alliance Biotech Group Co., Ltd, Shenzhen 518057, China
| | - 昭斌 陈
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 静 唐
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 梦蝶 陈
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 云雯 张
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 川 汪
- 四川大学华西公共卫生学院/四川大学华西第四医院 卫生检验与检疫系 (成都 610041)Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Isticato R. Bacterial Spore-Based Delivery System: 20 Years of a Versatile Approach for Innovative Vaccines. Biomolecules 2023; 13:947. [PMID: 37371527 DOI: 10.3390/biom13060947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/25/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Mucosal vaccines offer several advantages over injectable conventional vaccines, such as the induction of adaptive immunity, with secretory IgA production at the entry site of most pathogens, and needle-less vaccinations. Despite their potential, only a few mucosal vaccines are currently used. Developing new effective mucosal vaccines strongly relies on identifying innovative antigens, efficient adjuvants, and delivery systems. Several approaches based on phages, bacteria, or nanoparticles have been proposed to deliver antigens to mucosal surfaces. Bacterial spores have also been considered antigen vehicles, and various antigens have been successfully exposed on their surface. Due to their peculiar structure, spores conjugate the advantages of live microorganisms with synthetic nanoparticles. When mucosally administered, spores expressing antigens have been shown to induce antigen-specific, protective immune responses. This review accounts for recent progress in the formulation of spore-based mucosal vaccines, describing a spore's structure, specifically the spore surface, and the diverse approaches developed to improve its efficiency as a vehicle for heterologous antigen presentation.
Collapse
Affiliation(s)
- Rachele Isticato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Via Cinthia 4, 80126 Naples, Italy
- Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), 80055 Naples, Italy
| |
Collapse
|
6
|
Yu S, Sun H, Li Y, Wei S, Xu J, Liu J. Hydrogels as promising platforms for engineered living bacteria-mediated therapeutic systems. Mater Today Bio 2022; 16:100435. [PMID: 36164505 PMCID: PMC9508596 DOI: 10.1016/j.mtbio.2022.100435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
The idea of using engineered bacteria as prospective living therapeutic agents for the treatment of different diseases has been raised. Nevertheless, the development of safe and effective treatment strategies remains essential to the success of living bacteria-mediated therapy. Hydrogels have presented great promise for the delivery of living bacterial therapeutics due to their tunable physicochemical properties, good bioactivities, and excellent protection of labile payloads. In this review, we summarize the hydrogel design strategies for living bacteria-mediated therapy and review the recent advances in hydrogel-based living bacterial agent delivery for the treatment of typical diseases, including those for digestive health, skin fungal infections, wound healing, vaccines, and cancer, and discuss the current challenges and future perspectives of these strategies in the field. It is believed that the importance of hydrogel-based living bacteria-mediated therapy is expected to further increase with the development of both synthetic biology and biomaterials science in the future.
Collapse
Affiliation(s)
- Shuangjiang Yu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, People's Republic of China
| | - Hongcheng Sun
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, People's Republic of China
| | - Yongguang Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, People's Republic of China
| | - Shu Wei
- Jing Hengyi School of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, People's Republic of China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, People's Republic of China
| |
Collapse
|
7
|
García-Álvarez R, Vallet-Regí M. Bacteria and cells as alternative nano-carriers for biomedical applications. Expert Opin Drug Deliv 2022; 19:103-118. [PMID: 35076351 PMCID: PMC8802895 DOI: 10.1080/17425247.2022.2029844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Nano-based systems have received a lot of attention owing to their particular properties and, hence, have been proposed for a wide variety of biomedical applications. These nanosystems could be potentially employed for diagnosis and therapy of different medical issues. Although these nanomaterials are designed for specific tasks, interactions, and transformations when administered to the human body affect their performance and behavior. In this regard, bacteria and other cells have been presented as alternative nanocarriers. These microorganisms can be genetically modified and customized for a more specific therapeutic action and, in combination with nanomaterials, can lead to bio-hybrids with a unique potential for biomedical purposes. AREAS COVERED Literature regarding bacteria and cells employed in combination with nanomaterials for biomedical applications is revised and discussed in this review. The potential as well as the limitations of these novel bio-hybrid systems are evaluated. Several examples are presented to show the performance of these alternative nanocarriers. EXPERT OPINION Bio-hybrid systems have shown their potential as alternative nanocarriers as they contribute to better performance than traditional nano-based systems. Nevertheless, their limitations must be studied, and advantages and drawbacks assessed before their application to medicine.
Collapse
Affiliation(s)
- Rafaela García-Álvarez
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| |
Collapse
|
8
|
Wang S, Ma J, Ji Q, Liu Q. Evaluation of an attenuated Listeria monocytogenes as a vaccine vector to control Helicobacter pylori infection. Immunol Lett 2021; 238:68-74. [PMID: 34363896 DOI: 10.1016/j.imlet.2021.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/29/2021] [Indexed: 12/01/2022]
Abstract
The increasing resistance of Helicobacter pylori (H. pylori) to antibiotics has limited the efficacy of antibiotic therapy in the treatment of H. pylori-associated gastric diseases. The vaccine as an alternative method is becoming a safe and effective way to address this problem. In previous studies, live vector vaccines have proved to be effective in controlling H. pylori infection. Attenuated Listeria monocytogenes (L. monocytogenes) is a potential candidate vector applied in clinical trials, which can deliver foreign antigens and induce a broad immune response. To further explore the effectiveness of L. monocytogenes as a vaccine vector against H. pylori, attenuated L. monocytogenes-based vaccine EGDeΔactA/inlB(EGDeAB)-MECU was constructed to secrete a multi-epitope chimeric antigen (MECU) containing multiple B cell epitopes from H. pylori antigens. EGDeAB-MECU could secrete MECU stably. After immunized by gavage and intravenous injection, both EGDeAB and EGDeAB-MECU could significantly decrease gastric H. pylori colonization and induce a high level of specific antibodies against H. pylori. In conclusion, attenuated L. monocytogenes had an immunotherapeutic effect on H. pylori-infected mice, indicating its further development as a promising candidate vaccine vector for the H. pylori vaccine.
Collapse
Affiliation(s)
- Shuying Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qianyu Ji
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
9
|
Ji Q, Ma J, Wang S, Liu Q. Systematic identification of a panel of strong promoter regions from Listeria monocytogenes for fine-tuning gene expression. Microb Cell Fact 2021; 20:132. [PMID: 34247599 PMCID: PMC8273982 DOI: 10.1186/s12934-021-01628-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/05/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Attenuated Listeria monocytogenes (Lm) has been widely used as a vaccine vector in the prevention and treatment of pathogen infection and tumor diseases. In addition, previous studies have proved that the attenuated Lm can protect zebrafish from Vibrio infections, indicating that the attenuated Lm has a good application prospect in the field of aquatic vaccines. However, the limitation mainly lies in the lack of a set of well-characterized natural promoters for the expression of target antigens in attenuated Lm. RESULTS In our study, candidate strong promoters were identified through RNA-seq analysis, and characterized in Lm through enhanced green fluorescent protein (EGFP). Nine native promoters that showed stronger activities than that of the known strong promoter P36 under two tested temperatures (28 and 37 °C) were selected from the set, and P29 with the highest activity was 24-fold greater than P36. Furthermore, we demonstrated that P29 could initiate EGFP expression in ZF4 cells and zebrafish embryos. CONCLUSIONS This well-characterized promoter library can be used to fine-tune the expression of different proteins in Lm. The availability of a well-characterized promoter toolbox of Lm is essential for the analysis of yield increase for biotechnology applications.
Collapse
Affiliation(s)
- Qianyu Ji
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Shuying Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China.
| |
Collapse
|
10
|
Alginate-chitosan microcapsules improve vaccine potential of gamma-irradiated Listeria monocytogenes against listeriosis in murine model. Int J Biol Macromol 2021; 176:567-577. [PMID: 33581203 DOI: 10.1016/j.ijbiomac.2021.02.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/27/2022]
Abstract
Listeria monocytogenes is a cause of infectious food-borne disease in humans, characterized by neurological manifestations, abortion, and neonatal septicemia. It is intracellular bacterium, which limits the development of protective inactivated vacines. Adjuvants capable of stimulating cellular immune response are important tools for developing novel vaccines against intracellular bacteria. The aim of this study was to evaluate the vaccine potential of L. monocytogenes inactivated by gamma irradiation (KLM-γ) encapsulated in alginate microcapsules associated or not with chitosan against listeriosis in the murine model. At the fourth day after challenge there was a reduction in bacterial recovery in mice vaccinated with KLM-γ encapsulated with alginate or alginate-chitosan, with lower bacterial loads in the spleen (10 fold) and liver (100 fold) when compared to non-vaccinated mice. In vitro stimulation of splenocytes from mice vaccinated with alginate-chitosan-encapsulated KLM-γ resulted in lymphocyte proliferation, increase of proportion of memory CD4+ and CD8+ T cell and production of IL-10 and IFN-γ. Interestingly, the group vaccinated with alginate-chitosan-encapsulated KLM-γ had increased survival to lethal infection with lower L. monocytogenes-induced hepatic inflammation and necrosis. Therefore, KLM-γ encapsulation with alginate-chitosan proved to have potential for development of novel and safe inactivated vaccine formulations against listeriosis.
Collapse
|
11
|
Gilley RP, Dube PH. Checkpoint blockade inhibitors enhances the effectiveness of a Listeria monocytogenes-based melanoma vaccine. Oncotarget 2020; 11:740-754. [PMID: 32133048 PMCID: PMC7041938 DOI: 10.18632/oncotarget.27490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Melanoma continues to be a significant health concern worldwide despite recent improvements in treatment. Unlike many other prominent cancers, melanoma incidence in both men and women increased over the past decade in the U. S. and much of the developed world. The single greatest risk factor for melanoma is damage from ultraviolet radiation associated with lifestyle. The lifestyle component suggests that although melanoma risk can be minimized with behavioral changes, vaccinating high-risk individuals against melanoma may be the most efficacious preventative method. Accordingly, using a highly attenuated, double-mutant L. monocytogenes strain expressing a tumor-associated antigen, we obtained significant protection against melanoma in a mouse model. The Listeria-based vaccine induced protection through antigen-specific CD8+ T-cells inducing both a protective primary and a memory T-cell response. Vaccinated animals were significantly protected from melanoma. When used in conjunction with checkpoint blockade treatment, the vaccine substantially reduced tumor size and number relative to animals receiving checkpoint blockade (CPB) alone. This study provides evidence that CPB treatment synergizes with a L. monocytogenes-based melanoma vaccine to enhance vaccine-mediated protection.
Collapse
Affiliation(s)
- Ryan P. Gilley
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Peter H. Dube
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| |
Collapse
|
12
|
Garza-Morales R, Rendon BE, Malik MT, Garza-Cabrales JE, Aucouturier A, Bermúdez-Humarán LG, McMasters KM, McNally LR, Gomez-Gutierrez JG. Targeting Melanoma Hypoxia with the Food-Grade Lactic Acid Bacterium Lactococcus Lactis. Cancers (Basel) 2020; 12:cancers12020438. [PMID: 32069844 PMCID: PMC7072195 DOI: 10.3390/cancers12020438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Hypoxia is a feature of the tumor microenvironment that reduces efficacy of immuno- and chemotherapies, resulting in poor clinical outcomes. Lactococcus lactis is a facultative anaerobic gram-positive lactic acid bacterium (LAB) that is Generally Recognized as Safe (GRAS). Recently, the use of LAB as a delivery vehicle has emerged as an alternative strategy to deliver therapeutic molecules; therefore, we investigated whether L. lactis can target and localize within melanoma hypoxic niches. To simulate hypoxic conditions in vitro, melanoma cells A2058, A375 and MeWo were cultured in a chamber with a gas mixture of 5% CO2, 94% N2 and 1% O2. Among the cell lines tested, MeWo cells displayed greater survival rates when compared to A2058 and A375 cells. Co-cultures of L. lactis expressing GFP or mCherry and MeWo cells revealed that L. lactis efficiently express the transgenes under hypoxic conditions. Moreover, multispectral optoacoustic tomography (MSOT), and near infrared (NIR) imaging of tumor-bearing BALB/c mice revealed that the intravenous injection of either L. lactis expressing β-galactosidase (β-gal) or infrared fluorescent protein (IRFP713) results in the establishment of the recombinant bacteria within tumor hypoxic niches. Overall, our data suggest that L. lactis represents an alternative strategy to target and deliver therapeutic molecules into the tumor hypoxic microenvironment.
Collapse
Affiliation(s)
- Rodolfo Garza-Morales
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Beatriz E. Rendon
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA;
| | - Mohammad Tariq Malik
- Department of Microbiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Jeannete E. Garza-Cabrales
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Anne Aucouturier
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (A.A.); (L.G.B.-H.)
| | - Luis G. Bermúdez-Humarán
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (A.A.); (L.G.B.-H.)
| | - Kelly M. McMasters
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Lacey R. McNally
- Department of Bioengineering, Stephenson Cancer Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Jorge G. Gomez-Gutierrez
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
- Correspondence: ; Tel.: +1-(502)-852-5745
| |
Collapse
|
13
|
Mahdy SE, Sijing L, Lin S, Xiang Z, Hao-Tai C, Xiaofang P, Chuan W. Development of a recombinant vaccine against foot and mouth disease utilizing mutant attenuated Listeria ivanovii strain as a live vector. J Virol Methods 2019; 273:113722. [PMID: 31422118 DOI: 10.1016/j.jviromet.2019.113722] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 07/16/2019] [Accepted: 08/14/2019] [Indexed: 12/31/2022]
Abstract
The drawbacks of conventional inactivated Foot and Mouth Disease (FMD) vaccine, such as escaping of the virus during manufacture processes prompted researchers to explore novel types of vaccine to overcome these disadvantages. Listeria ivanovii (LI) is an intracellular microorganism that possesses immune-stimulatory properties, making it appropriate for use as a live bacterial vaccine vector. The Foot and mouth disease virus (FMDV) VP1 protein is the most immunogenic part of FMDV capsid, it has most of the antigenic sites for viral neutralization. The expression of antigen gene cassette in vitro was confirmed by Western blot analysis. Mice were able to eliminate LI△actAplcB-vp1 from the liver and spleen within few days revealed a safety of the candidate vaccine. Two doses of LI△actAplcB-vp1 with 14 days of interval were injected into mice. High levels of specific IgG antibodies and CD8+ and CD4+ T cells secreted cytokines including IFN-γ, TNF-α and IL-2 against FMDV-VP1 were achieved. Based on the obtained results, LI△actAplcB-vp1 candidate vaccine utilizing Listeria ivanovii as a live vector-based vaccine could enhance a specific cellular and humoral immune responses against the inserted FMDV-vp1 heterologous genes. LI△actAplcB-vp1 candidate vaccine could be a modern tool to overcome the disadvantages of the traditional inactivated FMD vaccine.
Collapse
Affiliation(s)
- S E Mahdy
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Liu Sijing
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Su Lin
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Zhang Xiang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Chen Hao-Tai
- Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Pei Xiaofang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Wang Chuan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Côté-Gravel J, Malouin F. Symposium review: Features of Staphylococcus aureus mastitis pathogenesis that guide vaccine development strategies. J Dairy Sci 2018; 102:4727-4740. [PMID: 30580940 DOI: 10.3168/jds.2018-15272] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/07/2018] [Indexed: 12/25/2022]
Abstract
Bovine mastitis affects animal health and welfare and milk production and quality, and it challenges the economic success of dairy farms. Staphylococcus aureus is one of the most commonly found pathogens in clinical mastitis but it also causes subclinical, persistent, and difficult-to-treat intramammary infections. Because of the failure of conventional antibiotic treatments and increasing pressure and concern from experts and consumers over the use of antibiotics in the dairy industry, many attempts have been made over the years to develop a vaccine for the prevention and control of Staph. aureus intramammary infections. Still, no commercially available vaccine formulation demonstrates sufficient protection and cost-effective potential. Multiple factors account for the lack of protection, including inadequate vaccine targets, high diversity among mastitis-provoking strains, cow-to-cow variation in immune response, and a failure to elicit an immune response that is appropriate for protection against a highly complex pathogen. The purpose of this review is to summarize key concepts related to the pathogenesis of Staph. aureus, and its interaction with the host, as well as to describe recent vaccine development strategies for prevention and control of Staph. aureus mastitis.
Collapse
Affiliation(s)
- Julie Côté-Gravel
- Centre d'Étude et de Valorisation de la Diversité Microbienne (CEVDM), Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Canada, J1K 2R1
| | - François Malouin
- Centre d'Étude et de Valorisation de la Diversité Microbienne (CEVDM), Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Canada, J1K 2R1.
| |
Collapse
|
15
|
Ding C, Wang X, Ma J, Xie M, Dong Q, Liu Q. Exploration of the bacterial invasion capacity of Listeria monocytogenes in ZF4 cells. Microb Pathog 2018; 124:238-243. [PMID: 30145253 DOI: 10.1016/j.micpath.2018.08.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 08/15/2018] [Accepted: 08/23/2018] [Indexed: 11/19/2022]
Abstract
Despite the results from zebrafish challenged model have demonstrated that Listeria monocytogenes (Lm) has strong adjuvant effects when this attenuated pathogenic bacteria is viewed as aquaculture vaccine vector, the underlying mechanism is not clear and extensive investigations are required. To further explore the potential of Lm in the field of aquaculture vaccine, zebrafish embryonic fibroblast cell line (ZF4) was used to evaluate the invasion ability of Lm. The data from cellular level showed that Lm had the lower invasion tendentiousness in ZF4 cells while bacterial invasion capacity was compared between zebrafish embryos cell line and human intestinal epithelial cell line. In ZF4 cells, there is no significant difference in bacterial invasion capacity between wild strain EGD-e and double-deleted strain ΔactA/inlB, which suggested that this attenuated effect was not showed in zebrafish cells. In addition, translation analysis indicated that the expressions of CD4 and CD8a in ZF4 cells increased after 2-h infection of the two Lm strains. These results further demonstrated that Lm presented multiple advantages including lower pathogenicity and antigen presentation when attenuated stain was viewed as aquaculture vaccine vector.
Collapse
Affiliation(s)
- Chengchao Ding
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Xiang Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Manman Xie
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Qingli Dong
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
16
|
Single vector platform vaccine protects against lethal respiratory challenge with Tier 1 select agents of anthrax, plague, and tularemia. Sci Rep 2018; 8:7009. [PMID: 29725025 PMCID: PMC5934503 DOI: 10.1038/s41598-018-24581-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/04/2018] [Indexed: 01/26/2023] Open
Abstract
Bacillus anthracis, Yersinia pestis, and Francisella tularensis are the causative agents of Tier 1 Select Agents anthrax, plague, and tularemia, respectively. Currently, there are no licensed vaccines against plague and tularemia and the licensed anthrax vaccine is suboptimal. Here we report F. tularensis LVS ΔcapB (Live Vaccine Strain with a deletion in capB)- and attenuated multi-deletional Listeria monocytogenes (Lm)-vectored vaccines against all three aforementioned pathogens. We show that LVS ΔcapB- and Lm-vectored vaccines express recombinant B. anthracis, Y. pestis, and F. tularensis immunoprotective antigens in broth and in macrophage-like cells and are non-toxic in mice. Homologous priming-boosting with the LVS ΔcapB-vectored vaccines induces potent antigen-specific humoral and T-cell-mediated immune responses and potent protective immunity against lethal respiratory challenge with all three pathogens. Protection against anthrax was far superior to that obtained with the licensed AVA vaccine and protection against tularemia was comparable to or greater than that obtained with the toxic and unlicensed LVS vaccine. Heterologous priming-boosting with LVS ΔcapB- and Lm-vectored B. anthracis and Y. pestis vaccines also induced potent protective immunity against lethal respiratory challenge with B. anthracis and Y. pestis. The single vaccine platform, especially the LVS ΔcapB-vectored vaccine platform, can be extended readily to other pathogens.
Collapse
|
17
|
Yurina V. Live Bacterial Vectors-A Promising DNA Vaccine Delivery System. Med Sci (Basel) 2018; 6:E27. [PMID: 29570602 PMCID: PMC6024733 DOI: 10.3390/medsci6020027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccination is one of the most successful immunology applications that has considerably improved human health. The DNA vaccine is a new vaccine being developed since the early 1990s. Although the DNA vaccine is promising, no human DNA vaccine has been approved to date. The main problem facing DNA vaccine efficacy is the lack of a DNA vaccine delivery system. Several studies explored this limitation. One of the best DNA vaccine delivery systems uses a live bacterial vector as the carrier. The live bacterial vector induces a robust immune response due to its natural characteristics that are recognized by the immune system. Moreover, the route of administration used by the live bacterial vector is through the mucosal route that beneficially induces both mucosal and systemic immune responses. The mucosal route is not invasive, making the vaccine easy to administer, increasing the patient's acceptance. Lactic acid bacterium is one of the most promising bacteria used as a live bacterial vector. However, some other attenuated pathogenic bacteria, such as Salmonella spp. and Shigella spp., have been used as DNA vaccine carriers. Numerous studies showed that live bacterial vectors are a promising candidate to deliver DNA vaccines.
Collapse
Affiliation(s)
- Valentina Yurina
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, East Java 65145, Malang, Indonesia.
| |
Collapse
|
18
|
Ding C, Ma J, Dong Q, Liu Q. Live bacterial vaccine vector and delivery strategies of heterologous antigen: A review. Immunol Lett 2018; 197:70-77. [PMID: 29550258 DOI: 10.1016/j.imlet.2018.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/13/2018] [Indexed: 02/06/2023]
Abstract
Live bacteria, including attenuated bacteria and probiotics, can be engineered to deliver target antigen to excite the host immune system. The preponderance of these live bacterial vaccine vectors is that they can stimulate durable humoral and cellular immunity. Moreover, delivery strategies of heterologous antigen in live bacterial promote the applications of new vaccine development. Genetic technologies are evolving, which potentiate the developing of heterologous antigen delivery systems, including bacterial surface display system, bacterial secretion system and balanced lethal vector system. Although the live bacterial vaccine vector is a powerful adjuvant, certain disadvantages, such as safety risk, must also be taken into account. In this review, we compare the development of representative live bacterial vectors, and summarize the main characterizations of the various delivery strategies of heterologous antigen in live vector vaccines.
Collapse
Affiliation(s)
- Chengchao Ding
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Qingli Dong
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China.
| |
Collapse
|
19
|
Shirakawa T, Kitagawa K. Antitumor effect of oral cancer vaccine with Bifidobacterium delivering WT1 protein to gut immune system is superior to WT1 peptide vaccine. Hum Vaccin Immunother 2018; 14:159-162. [PMID: 29048978 PMCID: PMC5791589 DOI: 10.1080/21645515.2017.1382787] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/08/2017] [Accepted: 09/18/2017] [Indexed: 12/30/2022] Open
Abstract
Despite the revolutionary progress of immune checkpoint inhibitors (CPIs) for cancer immunotherapy, CPIs are effective only in a subset of patients. Combining CPIs and cancer vaccines to achieve better clinical outcomes is a reasonable approach since CPI enhances cancer vaccine-induced tumor-associated antigen (TAA) specific CTL. Among the various TAAs so far identified, WT1 protein is one of the most promising TAAs as a cancer vaccine target. Until now clinical trials of WT1 vaccine have demonstrated only modest clinical efficacy. These WT1 vaccines were based on peptides or dendritic cells (DCs), and there was no oral cancer vaccine. Recently, we developed a WT1 oral cancer vaccine using a recombinant Bifidobacterium displaying WT1 protein, which can efficiently deliver WT1 protein to the gut immune system, and we demonstrated that this oral cancer vaccine had a significant anti-tumor effect in a C1498-WT1 murine leukemia syngeneic tumor model. The WT1 protein displayed in this vaccine consists of about 70% of the WT1 amino acid sequence including multiple known CD4 and CD8 T-cell epitopes of WT1. In this commentary, we introduce our recent data indicating the superior anti-tumor effect of a WT1 oral cancer vaccine delivering WT1 protein to the gut immune system compared to a peptide vaccine.
Collapse
Affiliation(s)
- Toshiro Shirakawa
- Division of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
- Division of Translational Research for Biologics, Department of Internal Medicine Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Kitagawa
- Division of Translational Research for Biologics, Department of Internal Medicine Related, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
20
|
Sensing of Bacterial Cyclic Dinucleotides by the Oxidoreductase RECON Promotes NF-κB Activation and Shapes a Proinflammatory Antibacterial State. Immunity 2017; 46:433-445. [PMID: 28329705 DOI: 10.1016/j.immuni.2017.02.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/20/2016] [Accepted: 01/13/2017] [Indexed: 12/25/2022]
Abstract
Bacterial and host cyclic dinucleotides (cdNs) mediate cytosolic immune responses through the STING signaling pathway, although evidence suggests that alternative pathways exist. We used cdN-conjugated beads to biochemically isolate host receptors for bacterial cdNs, and we identified the oxidoreductase RECON. High-affinity cdN binding inhibited RECON enzyme activity by simultaneously blocking the substrate and cosubstrate sites, as revealed by structural analyses. During bacterial infection of macrophages, RECON antagonized STING activation by acting as a molecular sink for cdNs. Bacterial infection of hepatocytes, which do not express STING, revealed that RECON negatively regulates NF-κB activation. Loss of RECON activity, via genetic ablation or inhibition by cdNs, increased NF-κB activation and reduced bacterial survival, suggesting that cdN inhibition of RECON promotes a proinflammatory, antibacterial state that is distinct from the antiviral state associated with STING activation. Thus, RECON functions as a cytosolic sensor for bacterial cdNs, shaping inflammatory gene activation via its effects on STING and NF-κB.
Collapse
|
21
|
Lehouritis P, Hogan G, Tangney M. Designer bacteria as intratumoural enzyme biofactories. Adv Drug Deliv Rev 2017; 118:8-23. [PMID: 28916496 DOI: 10.1016/j.addr.2017.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/18/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Bacterial-directed enzyme prodrug therapy (BDEPT) is an emerging form of treatment for cancer. It is a biphasic variant of gene therapy in which a bacterium, armed with an enzyme that can convert an inert prodrug into a cytotoxic compound, induces tumour cell death following tumour-specific prodrug activation. BDEPT combines the innate ability of bacteria to selectively proliferate in tumours, with the capacity of prodrugs to undergo contained, compartmentalised conversion into active metabolites in vivo. Although BDEPT has undergone clinical testing, it has received limited clinical exposure, and has yet to achieve regulatory approval. In this article, we review BDEPT from the system designer's perspective, and provide detailed commentary on how the designer should strategize its development de novo. We report on contemporary advancements in this field which aim to enhance BDEPT in terms of safety and efficacy. Finally, we discuss clinical and regulatory barriers facing BDEPT, and propose promising approaches through which these hurdles may best be tackled.
Collapse
|
22
|
Listeria-Vectored Vaccine Expressing the Mycobacterium tuberculosis 30-Kilodalton Major Secretory Protein via the Constitutively Active prfA* Regulon Boosts Mycobacterium bovis BCG Efficacy against Tuberculosis. Infect Immun 2017. [PMID: 28630063 DOI: 10.1128/iai.00245-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A potent vaccine against tuberculosis, one of the world's deadliest diseases, is needed to enhance the immunity of people worldwide, most of whom have been vaccinated with the partially effective Mycobacterium bovis BCG vaccine. Here we investigate novel live attenuated recombinant Listeria monocytogenes (rLm) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein (r30/antigen 85B [Ag85B]) (rLm30) as heterologous booster vaccines in animals primed with BCG. Using three attenuated L. monocytogenes vectors, L. monocytogenes ΔactA (LmI), L. monocytogenes ΔactA ΔinlB (LmII), and L. monocytogenes ΔactA ΔinlB prfA* (LmIII), we constructed five rLm30 vaccine candidates expressing r30 linked in frame to the L. monocytogenes listeriolysin O signal sequence and driven by the hly promoter (h30) or linked in frame to the ActA N-terminal 100 amino acids and driven by the actA promoter (a30). All five rLm30 vaccines secreted r30 in broth and macrophages; while rLm30 expressing r30 via a constitutively active prfA* regulon (rLmIII/a30) expressed the largest amount of r30 in broth culture, all five rLm30 vaccines expressed equivalent amounts of r30 in infected macrophages. In comparative studies, boosting of BCG-immunized mice with rLmIII/a30 induced the strongest antigen-specific T-cell responses, including splenic and lung polyfunctional CD4+ T cells expressing the three cytokines interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-2 (IL-2) (P < 0.001) and splenic and lung CD8+ T cells expressing IFN-γ (P < 0.0001). In mice and guinea pigs, the rLmIII/a30 and rLmI/h30 vaccines were generally more potent booster vaccines than r30 with an adjuvant and a recombinant adenovirus vaccine expressing r30. In a setting in which BCG alone was highly immunoprotective, boosting of mice with rLmIII/a30, the most potent of the vaccines, significantly enhanced protection against aerosolized M. tuberculosis (P < 0.01).
Collapse
|
23
|
Xiong X, Jiao J, Gregory AE, Wang P, Bi Y, Wang X, Jiang Y, Wen B, Portnoy DA, Samuel JE, Chen C. Identification of Coxiella burnetii CD8+ T-Cell Epitopes and Delivery by Attenuated Listeria monocytogenes as a Vaccine Vector in a C57BL/6 Mouse Model. J Infect Dis 2017; 215:1580-1589. [PMID: 27703037 DOI: 10.1093/infdis/jiw470] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/25/2016] [Indexed: 01/26/2023] Open
Abstract
Coxiella burnetii is a gram-negative bacterium that causes acute and chronic Q fever. Because of the severe adverse effect of whole-cell vaccination, identification of immunodominant antigens of C. burnetii has become a major focus of Q fever vaccine development. We hypothesized that secreted C. burnetii type IV secretion system (T4SS) effectors may represent a major class of CD8+ T-cell antigens, owing to their cytosolic localization. Twenty-nine peptides were identified that elicited robust CD8+ T-cell interferon γ (IFN-γ) recall responses from mice infected with C. burnetii. Interestingly, 22 of 29 epitopes were derived from 17 T4SS-related proteins, none of which were identified as immunodominant antigens by using previous antibody-guided approaches. These epitopes were expressed in an attenuated Listeria monocytogenes vaccine strain. Immunization with recombinant L. monocytogenes vaccines induced a robust CD8+ T-cell response and conferred measurable protection against C. burnetii infection in mice. These data suggested that T4SS effectors represent an important class of C. burnetii antigens that can induce CD8+ T-cell responses. We also showed that attenuated L. monocytogenes vaccine vectors are an efficient antigen-delivery platform that can be used to induce robust protective CD8+ T-cell immune responses against C. burnetii infection.
Collapse
Affiliation(s)
- Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Anthony E Gregory
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan
| | - Pengcheng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology.,Department of Clinical Laboratory, 105th Hospital of the People's Liberation Army, Hefei, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Xiaoyi Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - James E Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan
| | - Chen Chen
- Department of Molecular and Cell Biology, University of California, Berkeley
| |
Collapse
|
24
|
Kunda NK, Wafula D, Tram M, Wu TH, Muttil P. A stable live bacterial vaccine. Eur J Pharm Biopharm 2016; 103:109-117. [PMID: 27020530 DOI: 10.1016/j.ejpb.2016.03.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/04/2016] [Accepted: 03/23/2016] [Indexed: 01/31/2023]
Abstract
Formulating vaccines into a dry form enhances its thermal stability. This is critical to prevent administering damaged and ineffective vaccines, and to reduce its final cost. A number of vaccines in the market as well as those being evaluated in the clinical setting are in a dry solid state; yet none of these vaccines have achieved long-term stability at high temperatures. We used spray-drying to formulate a recombinant live attenuated Listeria monocytogenes (Lm; expressing Francisella tularensis immune protective antigen pathogenicity island protein IglC) bacterial vaccine into a thermostable dry powder using various sugars and an amino acid. Lm powder vaccine showed minimal loss in viability when stored for more than a year at ambient room temperature (∼23°C) or for 180days at 40°C. High temperature viability was achieved by maintaining an inert atmosphere in the storage container and removing oxygen free radicals that damage bacterial membranes. Further, in vitro antigenicity was confirmed by infecting a dendritic cell line with cultures derived from spray dried Lm and detection of an intracellularly expressed protective antigen. A combination of stabilizing excipients, a cost effective one-step drying process, and appropriate storage conditions could provide a viable option for producing, storing and transporting heat-sensitive vaccines, especially in regions of the world that require them the most.
Collapse
Affiliation(s)
- Nitesh K Kunda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA
| | - Denis Wafula
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, MD, USA
| | - Meilinn Tram
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA
| | - Terry H Wu
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA; Center for Infectious Disease and Immunity, University of New Mexico, Albuquerque, NM, USA
| | - Pavan Muttil
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
25
|
Lin IYC, Van TTH, Smooker PM. Live-Attenuated Bacterial Vectors: Tools for Vaccine and Therapeutic Agent Delivery. Vaccines (Basel) 2015; 3:940-72. [PMID: 26569321 PMCID: PMC4693226 DOI: 10.3390/vaccines3040940] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022] Open
Abstract
Genetically attenuated microorganisms, including pathogenic and commensal bacteria, can be engineered to carry and deliver heterologous antigens to elicit host immunity against both the vector as well as the pathogen from which the donor gene is derived. These live attenuated bacterial vectors have been given much attention due to their capacity to induce a broad range of immune responses including localized mucosal, as well as systemic humoral and/or cell-mediated immunity. In addition, the unique tumor-homing characteristics of these bacterial vectors has also been exploited for alternative anti-tumor vaccines and therapies. In such approach, tumor-associated antigen, immunostimulatory molecules, anti-tumor drugs, or nucleotides (DNA or RNA) are delivered. Different potential vectors are appropriate for specific applications, depending on their pathogenic routes. In this review, we survey and summarize the main features of the different types of live bacterial vectors and discussed the clinical applications in the field of vaccinology. In addition, different approaches for using live attenuated bacterial vectors for anti-cancer therapy is discussed, and some promising pre-clinical and clinical studies in this field are outlined.
Collapse
Affiliation(s)
- Ivan Y C Lin
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Thi Thu Hao Van
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Peter M Smooker
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| |
Collapse
|
26
|
da Silva AJ, Zangirolami TC, Novo-Mansur MTM, Giordano RDC, Martins EAL. Live bacterial vaccine vectors: an overview. Braz J Microbiol 2015; 45:1117-29. [PMID: 25763014 PMCID: PMC4323283 DOI: 10.1590/s1517-83822014000400001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/17/2014] [Indexed: 02/07/2023] Open
Abstract
Genetically attenuated microorganisms, pathogens, and some commensal bacteria can be engineered to deliver recombinant heterologous antigens to stimulate the host immune system, while still offering good levels of safety. A key feature of these live vectors is their capacity to stimulate mucosal as well as humoral and/or cellular systemic immunity. This enables the use of different forms of vaccination to prevent pathogen colonization of mucosal tissues, the front door for many infectious agents. Furthermore, delivery of DNA vaccines and immune system stimulatory molecules, such as cytokines, can be achieved using these special carriers, whose adjuvant properties and, sometimes, invasive capacities enhance the immune response. More recently, the unique features and versatility of these vectors have also been exploited to develop anti-cancer vaccines, where tumor-associated antigens, cytokines, and DNA or RNA molecules are delivered. Different strategies and genetic tools are constantly being developed, increasing the antigenic potential of agents delivered by these systems, opening fresh perspectives for the deployment of vehicles for new purposes. Here we summarize the main characteristics of the different types of live bacterial vectors and discuss new applications of these delivery systems in the field of vaccinology.
Collapse
Affiliation(s)
- Adilson José da Silva
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Teresa Cristina Zangirolami
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Maria Teresa Marques Novo-Mansur
- Departamento de Genética e Evolução Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Roberto de Campos Giordano
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Elizabeth Angélica Leme Martins
- Centro de Biotecnologia Instituto Butantan São PauloSP Brazil Centro de Biotecnologia, Instituto Butantan, São Paulo, SP, Brazil
| |
Collapse
|
27
|
Calderón-González R, Frande-Cabanes E, Bronchalo-Vicente L, Lecea-Cuello MJ, Pareja E, Bosch-Martínez A, Fanarraga ML, Yañez-Díaz S, Carrasco-Marín E, Alvarez-Domínguez C. Cellular vaccines in listeriosis: role of the Listeria antigen GAPDH. Front Cell Infect Microbiol 2014; 4:22. [PMID: 24600592 PMCID: PMC3930854 DOI: 10.3389/fcimb.2014.00022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 02/05/2014] [Indexed: 11/13/2022] Open
Abstract
The use of live Listeria-based vaccines carries serious difficulties when administrated to immunocompromised individuals. However, cellular carriers have the advantage of inducing multivalent innate immunity as well as cell-mediated immune responses, constituting novel and secure vaccine strategies in listeriosis. Here, we compare the protective efficacy of dendritic cells (DCs) and macrophages and their safety. We examined the immune response of these vaccine vectors using two Listeria antigens, listeriolysin O (LLO) and glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), and several epitopes such as the LLO peptides, LLO189−201 and LLO91−99 and the GAPDH peptide, GAPDH1−22. We discarded macrophages as safe vaccine vectors because they show anti-Listeria protection but also high cytotoxicity. DCs loaded with GAPDH1−22 peptide conferred higher protection and security against listeriosis than the widely explored LLO91−99 peptide. Anti-Listeria protection was related to the changes in DC maturation caused by these epitopes, with high production of interleukin-12 as well as significant levels of other Th1 cytokines such as monocyte chemotactic protein-1, tumor necrosis factor-α, and interferon-γ, and with the induction of GAPDH1−22-specific CD4+ and CD8+ immune responses. This is believed to be the first study to explore the use of a novel GAPDH antigen as a potential DC-based vaccine candidate for listeriosis, whose efficiency appears to highlight the relevance of vaccine designs containing multiple CD4+ and CD8+ epitopes.
Collapse
Affiliation(s)
- Ricardo Calderón-González
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain ; Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria Santander, Spain
| | - Elisabet Frande-Cabanes
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain
| | - Lucía Bronchalo-Vicente
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain ; Servicio de Dermatología, Hospital Universitario Marqués de Valdecilla Santander, Spain
| | - M Jesús Lecea-Cuello
- Servicio de Pediatría, Hospital Universitario Marqués de Valdecilla-IFIMAV Santander, Spain
| | - Eduardo Pareja
- Information Technologies Research Group, Era7 Bioinformatics Granada, Spain
| | - Alexandre Bosch-Martínez
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain
| | - Mónica L Fanarraga
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria Santander, Spain
| | - Sonsoles Yañez-Díaz
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain ; Servicio de Dermatología, Hospital Universitario Marqués de Valdecilla Santander, Spain
| | - Eugenio Carrasco-Marín
- Servicio de Pediatría, Hospital Universitario Marqués de Valdecilla-IFIMAV Santander, Spain
| | - Carmen Alvarez-Domínguez
- Grupo de Genómica, Proteómica y Vacunas, Primera Planta-Laboratorio 124, Fundación Marqués de Valdecilla-IFIMAV Santander, Spain
| |
Collapse
|
28
|
Shollenberger LM, Bui CT, Paterson Y, Nyhoff L, Harn DA. HIV-1 vaccine-specific responses induced by Listeria vector vaccines are maintained in mice subsequently infected with a model helminth parasite, Schistosoma mansoni. Vaccine 2013; 31:5651-8. [PMID: 24120546 DOI: 10.1016/j.vaccine.2013.09.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/30/2013] [Accepted: 09/27/2013] [Indexed: 01/07/2023]
Abstract
In areas co-endemic for helminth parasites and HIV/AIDS, infants are often administered vaccines prior to infection with immune modulatory helminth parasites. Systemic Th2 biasing and immune suppression caused by helminth infection reduces cell-mediated responses to vaccines such as tetanus toxoid and BCG. Therefore, we asked if infection with helminthes post-vaccination, alters already established vaccine induced immune responses. In our model, mice are vaccinated against HIV-1 Gag using a Listeria vaccine vector (Lm-Gag) in a prime-boost manner, then infected with the human helminth parasite Schistosoma mansoni. This allows us to determine if established vaccine responses are maintained or altered after helminth infection. Our second objective asked if helminth infection post-vaccination alters the recipient's ability to respond to a second boost. Here we compared responses between uninfected mice, schistosome infected mice, and infected mice that were given an anthelminthic, which occurred coincident with the boost or four weeks prior, as well as comparing to un-boosted mice. We report that HIV-1 vaccine-specific responses generated by Listeria vector HIV-1 vaccines are maintained following subsequent chronic schistosome infection, providing further evidence that Listeria vector vaccines induce potent vaccine-specific responses that can withstand helminth infection. We also were able to demonstrate that administration of a second Listeria boost, which markedly enhanced the immune response, was minimally impacted by schistosome infection, or anthelminthic therapy. Surprisingly, we also observed enhanced antibody responses to HIV Gag in vaccinated mice subsequently infected with schistosomes.
Collapse
Affiliation(s)
- Lisa M Shollenberger
- Department of Infectious Diseases, College of Veterinary Medicine and the Center for Tropical and Emerging Global Diseases, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602-7387, USA.
| | | | | | | | | |
Collapse
|
29
|
Sun R, Liu Y. Listeriolysin O as a strong immunogenic molecule for the development of new anti-tumor vaccines. Hum Vaccin Immunother 2013; 9:1058-68. [PMID: 23399758 PMCID: PMC3899140 DOI: 10.4161/hv.23871] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/23/2013] [Accepted: 02/03/2013] [Indexed: 11/19/2022] Open
Abstract
The pore-forming toxin listeriolysin O (LLO), which is produced by Listeria monocytogenes, mediates bacterial phagosomal escape and facilitates bacterial multiplication during infection. This toxin has recently gained attention because of its confirmed role in the controlled and specific modulation of the immune response. Currently, cancer immunotherapies are focused on conquering the immune tolerance induced by poorly immunogenic tumor antigens and eliciting strong, lasting immunological memory. An effective way to achieve these goals is the co-administration of potent immunomodulatory adjuvant components with vaccine vectors. LLO, a toxin that belongs to the family of cholesterol-dependent cytolysins (CDCs), exhibits potent cell type-non-specific toxicity and is a source of dominant CD4(+) and CD8(+) T cell epitopes. According to recent research, in addition to its effective cytotoxicity as a cancer immunotherapeutic drug, the non-specific adjuvant property of LLO makes it promising for the development of efficacious anti-tumor vaccines.
Collapse
Affiliation(s)
- Rui Sun
- Department of Pathology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
| | - Yuqin Liu
- Department of Pathology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
- Cell Resource Center; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
| |
Collapse
|
30
|
Shollenberger LM, Bui C, Paterson Y, Allen K, Harn D. Successful vaccination of immune suppressed recipients using Listeria vector HIV-1 vaccines in helminth infected mice. Vaccine 2013; 31:2050-6. [PMID: 23470236 DOI: 10.1016/j.vaccine.2013.02.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 01/22/2013] [Accepted: 02/19/2013] [Indexed: 10/27/2022]
Abstract
Vaccines for HIV, malaria and TB remain high priorities, especially for sub-Saharan populations. The question is: will vaccines currently in development for these diseases function in populations that have a high prevalence of helminth infection? Infection with helminth parasites causes immune suppression and a CD4+ Th2 skewing of the immune system, thereby impairing Th1-type vaccine efficacy. In this study, we conduct HIV vaccine trials in mice with and without chronic helminth infection to mimic the human vaccine recipient populations in Sub-Saharan Africa and other helminth parasite endemic regions of the world, as there is large overlap in global prevalence for HIV and helminth infection. Here, we demonstrate that Listeria monocytogenes functions as a vaccine vector to drive robust and functional HIV-specific cellular immune responses, irrespective of chronic helminth infection. This observation represents a significant advance in the field of vaccine research and underscores the concept that vaccines in the developmental pipeline should be effective in the target populations.
Collapse
Affiliation(s)
- Lisa M Shollenberger
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602-7387, USA.
| | | | | | | | | |
Collapse
|
31
|
A heterologous prime-boost vaccination strategy comprising the Francisella tularensis live vaccine strain capB mutant and recombinant attenuated Listeria monocytogenes expressing F. tularensis IglC induces potent protective immunity in mice against virulent F. tularensis aerosol challenge. Infect Immun 2013; 81:1550-61. [PMID: 23439306 DOI: 10.1128/iai.01013-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis, the causative agent of tularemia, is a category A bioterrorism agent. A vaccine that is safer and more effective than the currently available unlicensed F. tularensis live vaccine strain (LVS) is needed to protect against intentional release of aerosolized F. tularensis, the most dangerous type of exposure. In this study, we employed a heterologous prime-boost vaccination strategy comprising intradermally administered LVS ΔcapB (highly attenuated capB-deficient LVS mutant) as the primer vaccine and rLm/iglC (recombinant attenuated Listeria monocytogenes expressing the F. tularensis immunoprotective antigen IglC) as the booster vaccine. Boosting LVS ΔcapB-primed mice with rLm/iglC significantly enhanced T cell immunity; their splenic T cells secreted significantly more gamma interferon (IFN-γ) and had significantly more cytokine (IFN-γ and/or tumor necrosis factor [TNF] and/or interleukin-2 [IL-2])-producing CD4(+) and CD8(+) T cells upon in vitro IglC stimulation. Importantly, mice primed with LVS ΔcapB or rLVS ΔcapB/IglC, boosted with rLm/iglC, and subsequently challenged with 10 50% lethal doses (LD50) of aerosolized highly virulent F. tularensis Schu S4 had a significantly higher survival rate and mean survival time than mice immunized with only LVS ΔcapB (P < 0.0001); moreover, compared with mice immunized once with LVS, primed-boosted mice had a higher survival rate (75% versus 62.5%) and mean survival time during the first 21 days postchallenge (19 and 20 days for mice boosted after being primed with LVS ΔcapB and rLVS ΔcapB/IglC, respectively, versus 17 days for mice immunized with LVS) and maintained their weight significantly better (P < 0.01). Thus, the LVS ΔcapB-rLm/iglC prime-boost vaccination strategy holds substantial promise for a vaccine that is safer and at least as potent as LVS.
Collapse
|
32
|
Systemic infection of Mice with Listeria monocytogenes to characterize host immune responses. Methods Mol Biol 2013; 1031:125-44. [PMID: 23824896 DOI: 10.1007/978-1-62703-481-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Listeria monocytogenes is a Gram-positive facultative intracellular bacterium that is widely used to characterize bacterial pathogenesis and host immunity. Here, we describe a set of basic methods and techniques to infect mice with L. monocytogenes, measure bacterial load in tissues, and analyze immune cell subsets responding to infection in the spleen and liver. In addition, a specialized method for immune cell depletion is incorporated within the overall protocol, along with suggestions at various points in the protocol for minimizing experimental variability in mouse infection studies using L. monocytogenes. Finally, we highlight a number of experimental strategies for which L. monocytogenes has facilitated research into host immune responses and bacterial pathogenesis.
Collapse
|
33
|
Jia Y, Yin Y, Duan F, Fu H, Hu M, Gao Y, Pan Z, Jiao X. Prophylactic and therapeutic efficacy of an attenuated Listeria monocytogenes-based vaccine delivering HPV16 E7 in a mouse model. Int J Mol Med 2012; 30:1335-42. [PMID: 23027427 DOI: 10.3892/ijmm.2012.1136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/02/2012] [Indexed: 11/06/2022] Open
Abstract
Listeria monocytogenes (L. monocytogenes) has been developed as a cancer vaccine vector due to its ability to elicit strong innate and adaptive immune responses. For clinical application, it is necessary to exploit a Listeria platform strain that is safe and that also retains its immunogenicity to develop vaccine candidates against cancer. In this study, a highly attenuated strain with a deletion of actA/plcB was employed as a vector to deliver the human papillomavirus type 16 (HPV16) E7 antigen, which was stably inserted into the chromosome of L. monocytogenes. The prophylactic and therapeutic efficacy of the recombinant L. monocytogenes strain expressing E7 (LM1-2-E7) were evaluated in C57BL/6 mice. In prophylactic tumor challenge assays, immunization with the recombinant strain LM1-2-E7 was able to protect against tumor formation in 87.5% of the mice, even after a second challenge, suggesting that this prophylactic immunization can provide long-lasting immunity. In the therapeutic setting, immunization with LM1-2-E7 led to tumor regression in 50% of the mice and suppressed tumor growth in the remaining mice. The results showed that the recombinant strain was cleared by the immune system within 5 days after immunization and induced a Th1 immune response against E7 peptide and E7-specific cytotoxic T-lymphocyte (CTL) killing activity without severe inflammatory responses in the spleen and liver. Markedly, recombinant Listeria strain resulted in preferential accumulation within tumor tissues and induced higher numbers of CD8+ T cells that infiltrated into the tumor, which were associated with retardation of tumor growth. Collectively, these data indicate that LM1-2-E7 is a possible vaccine candidate against cervical cancer.
Collapse
Affiliation(s)
- Yanyan Jia
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Nascimento IP, Leite LCC. Recombinant vaccines and the development of new vaccine strategies. Braz J Med Biol Res 2012; 45:1102-11. [PMID: 22948379 PMCID: PMC3854212 DOI: 10.1590/s0100-879x2012007500142] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/22/2012] [Indexed: 11/22/2022] Open
Abstract
Vaccines were initially developed on an empirical basis, relying mostly on attenuation or inactivation of pathogens. Advances in immunology, molecular biology, biochemistry, genomics, and proteomics have added new perspectives to the vaccinology field. The use of recombinant proteins allows the targeting of immune responses focused against few protective antigens. There are a variety of expression systems with different advantages, allowing the production of large quantities of proteins depending on the required characteristics. Live recombinant bacteria or viral vectors effectively stimulate the immune system as in natural infections and have intrinsic adjuvant properties. DNA vaccines, which consist of non-replicating plasmids, can induce strong long-term cellular immune responses. Prime-boost strategies combine different antigen delivery systems to broaden the immune response. In general, all of these strategies have shown advantages and disadvantages, and their use will depend on the knowledge of the mechanisms of infection of the target pathogen and of the immune response required for protection. In this review, we discuss some of the major breakthroughs that have been achieved using recombinant vaccine technologies, as well as new approaches and strategies for vaccine development, including potential shortcomings and risks.
Collapse
Affiliation(s)
- I P Nascimento
- Centro de Biotecnologia, Instituto Butantan, São Paulo, SP, Brasil
| | | |
Collapse
|
35
|
Warren SE, Duong H, Mao DP, Armstrong A, Rajan J, Miao EA, Aderem A. Generation of a Listeria vaccine strain by enhanced caspase-1 activation. Eur J Immunol 2011; 41:1934-40. [PMID: 21538346 DOI: 10.1002/eji.201041214] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 03/09/2011] [Accepted: 04/26/2011] [Indexed: 11/08/2022]
Abstract
The immunostimulatory properties conferred by vaccine adjuvants require caspase-1 for processing of IL-1β and IL-18. Caspase-1 is activated in response to a breach of the cytosolic compartment by microbes and the process is initiated by intracellular pattern recognition receptors within inflammasomes. Listeria monocytogenes is detected in the cytosol by the NLRC4, NLRP3 and AIM2 inflammasomes. NLRC4 is activated by flagellin, and L. monocytogenes evades NLRC4 by repressing flagellin expression. We generated an L. monocytogenes strain that was forced to express flagellin in the host cell cytosol. This strain hyperactivated caspase-1 and was preferentially cleared via NLRC4 detection in an IL-1β/IL-18 independent manner. We also created a strain of L. monocytogenes with forced expression of another NLRC4 agonist, PrgJ, from the Type III secretion system of Salmonella typhimurium. Forced expression of flagellin or PrgJ resulted in attenuation, yet both strains conferred protective immunity in mice against lethal challenge with L. monocytogenes. This work is the first demonstration of specific targeting of the caspase-1 activation pathway to generate a safe and potent L. monocytogenes-based vaccine. Moreover, the attenuated strains with embedded flagellin or PrgJ adjuvants represent attractive vectors for vaccines aimed at eliciting T-cell responses.
Collapse
|
36
|
Velge P, Roche SM. Variability of Listeria monocytogenes virulence: a result of the evolution between saprophytism and virulence? Future Microbiol 2011; 5:1799-821. [PMID: 21155663 DOI: 10.2217/fmb.10.134] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The genus Listeria consists of eight species but only two are pathogenic. Human listeriosis due to Listeria monocytogenes is a foodborne disease. L. monocytogenes is widespread in the environment living as a saprophyte, but is also capable of making the transition into a pathogen following its ingestion by susceptible humans or animals. It is now known that many distinct strains of L. monocytogenes differ in their virulence and epidemic potential. Unfortunately, there is currently no standard definition of virulence levels and no complete comprehensive overview of the evolution of Listeria species and L. monocytogenes strains taking into account the presence of both epidemic and low-virulence strains. This article focuses on the methods and genes allowing us to determine the pathogenic potential of Listeria strains, and the evolution of Listeria virulence. The presence of variable levels of virulence within L. monocytogenes has important consequences on detection of Listeria strains and risk analysis but also on our comprehension of how certain pathogens will behave in a population over evolutionary time.
Collapse
Affiliation(s)
- Philippe Velge
- INRA de tours, UR1282, Infectiologie Animale et Santé Publique, 37380 Nouzilly, France.
| | | |
Collapse
|
37
|
Priming and activation of human ovarian and breast cancer-specific CD8+ T cells by polyvalent Listeria monocytogenes-based vaccines. J Immunother 2010; 32:856-69. [PMID: 19752748 DOI: 10.1097/cji.0b013e3181b0b125] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapeutic vaccine is potentially an effective strategy to combat cancer. Essential components of an effective vaccine must include antigens that are processed by the major histocompatibility complex class I pathway, presented by the tumor major histocompatibility complex molecules, and an effective antigen delivery platform that is capable of breaking self-tolerance. In this study, we characterized a set of ovarian cancer-specific T-cell epitopes delivered by live-attenuated recombinant Listeria monocytogenes (Lm DeltaactADeltainlB) as a vaccine vector. We present data that peptide-specific T cells recognize the human monocytic cell line THP-1 infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Furthermore, we demonstrate that recombinant L. monocytogenes (Lm)-infected antigen-presenting cells can prime and expand epitope-specific CD8 T cells in vitro and such CD8 T cells recognize not only peptide-loaded targets but also ovarian and breast tumor cells presenting endogenous epitopes. Finally, peptide-specific T cells generated using peripheral blood mononuclear cell from ovarian cancer patients recognize target cells infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Our results demonstrate that live-attenuated recombinant Lm can be used effectively as a vehicle to deliver cancer peptide antigens singly or as a multiepitope construct. Thus, the use of recombinant live-attenuated Lm strains encoding endogenously processed and presented tumor epitopes/antigens represents an attractive strategy for active cancer immunotherapy in a clinical setting.
Collapse
|
38
|
Freitag NE, Port GC, Miner MD. Listeria monocytogenes - from saprophyte to intracellular pathogen. Nat Rev Microbiol 2009; 7:623-8. [PMID: 19648949 DOI: 10.1038/nrmicro2171] [Citation(s) in RCA: 417] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Listeria monocytogenes is a bacterium that lives in the soil as a saprophyte but is capable of making the transition into a pathogen following its ingestion by susceptible humans or animals. Recent studies suggest that L. monocytogenes mediates its saprophyte-to-cytosolic-parasite transition through the careful modulation of the activity of a virulence regulatory protein known as PrfA, using a range of environmental cues that include available carbon sources. In this Progress article we describe the regulation of PrfA and its role in the L. monocytogenes transition from the saprophytic stage to the virulent intracellular stage.
Collapse
Affiliation(s)
- Nancy E Freitag
- Department of Microbiology and Immunology (MC790), University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois 606127344, USA.
| | | | | |
Collapse
|
39
|
Abstract
Tularemia, caused by the Gram-negative bacterium Francisella tularensis, can be contracted by the bite of an arthropod vector or by inhalation. This disease occurs relatively infrequently but can be severe and even life-threatening if untreated. Until recently, there were few laboratories studying this organism; however, concerns over its potential use as a biological weapon have led to renewed attention to F. tularensis research, particularly in the area of vaccine development. Advances in the ability to genetically manipulate F. tularensis, along with knowledge gained from the creation and refinement of attenuated bacterial vaccines for other diseases, continue to foster significant progress in the development of live-attenuated bacterial vaccines, as well as defined antigen and subunit vaccines.
Collapse
Affiliation(s)
- Barbara J Mann
- Departments of Medicine & Microbiology, University of Virginia Health Systems, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
40
|
Abstract
Although HER2/neu-targeted cancer vaccines have shown initial promise in the adjuvant setting, a therapeutic vaccine remains elusive due to the tumor escape mechanisms of established cancer. As described by Seavey et al. in this issue of CCR, a Listeria-delivered vaccine may help overcome immune tolerance, leading to an effective therapeutic vaccine.
Collapse
Affiliation(s)
- Guy T Clifton
- General Surgery, Brooke Army Medical Center, Fort Sam Houston, Texas 78234, USA
| | | |
Collapse
|
41
|
Seavey MM, Maciag PC, Al-Rawi N, Sewell D, Paterson Y. An anti-vascular endothelial growth factor receptor 2/fetal liver kinase-1 Listeria monocytogenes anti-angiogenesis cancer vaccine for the treatment of primary and metastatic Her-2/neu+ breast tumors in a mouse model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:5537-46. [PMID: 19380802 PMCID: PMC2850569 DOI: 10.4049/jimmunol.0803742] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thirty years after angiogenesis was shown to play an enabling role in cancer, modern medicine is still trying to develop novel compounds and therapeutics to target the tumor vasculature. However, most therapeutics require multiple rounds of administration and can have toxic side effects. In this study, we use anti-angiogenesis immunotherapy to target cells actively involved in forming new blood vessels that support the growth and spread of breast cancer. Targeting a central cell type involved in angiogenesis, endothelial cells, we immunized against host vascular endothelial growth factor receptor 2 to fight the growth of Her-2/neu(+) breast tumors. Using the bacterial vector, Listeria monocytogenes (Lm), we fused polypeptides from the mouse vascular endothelial growth factor receptor 2 molecule (fetal liver kinase-1) to the microbial adjuvant, listeriolysin-O, and used Lm to deliver the Ags and elicit potent antitumor CTL responses. Lm-listeriolysin-O-fetal liver kinase-1 was able to eradicate some established breast tumors, reduce microvascular density in the remaining tumors, protect against tumor rechallenge and experimental metastases, and induce epitope spreading to various regions of the tumor-associated Ag Her-2/neu. Tumor eradication was found to be dependent on epitope spreading to HER-2/neu and was not solely due to the reduction of tumor vasculature. However, vaccine efficacy did not affect normal wound healing nor have toxic side effects on pregnancy. We show that an anti-angiogenesis vaccine can overcome tolerance to the host vasculature driving epitope spreading to an endogenous tumor protein and drive active tumor regression.
Collapse
MESH Headings
- Amino Acid Sequence
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/genetics
- Angiogenesis Inhibitors/immunology
- Animals
- Bacterial Toxins/administration & dosage
- Bacterial Toxins/genetics
- Bacterial Toxins/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Female
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/genetics
- Growth Inhibitors/immunology
- Heat-Shock Proteins/administration & dosage
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/immunology
- Hemolysin Proteins/administration & dosage
- Hemolysin Proteins/genetics
- Hemolysin Proteins/immunology
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Lung Neoplasms/blood supply
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Molecular Sequence Data
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Pathologic/prevention & control
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Vascular Endothelial Growth Factor Receptor-2/administration & dosage
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/immunology
Collapse
Affiliation(s)
- Matthew M. Seavey
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | | | | | | | - Yvonne Paterson
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
42
|
Abstract
The care of patients with breast cancer has become increasingly complex with advancements in diagnostic modalities, surgical approaches, and adjuvant treatments. A multidisciplinary approach to breast cancer care is essential to the successful integration of available therapies. This article addresses the key components of multidisciplinary breast cancer care, with a special emphasis on new and emerging approaches over the past 10 years in the fields of diagnostics, surgery, radiation, medical oncology, and plastic surgery.
Collapse
|
43
|
Abstract
Pathogen recognition and induction of immune responses are important for efficient elimination of infection. However, pathogens such as Listeria monocytogenes employ strategies to evade or modulate these defences, thus creating a more favourable environment that ensures their survival and pathogenesis. New insights into these strategies, particularly those targeting innate immunity, have recently emerged. L. monocytogenes is initially detected at the cell surface or in phagosomes by toll-like receptor 2 and in the cytosol by nuclear oligodimerization domain (NOD)-like receptors (NOD1, NOD2) and NALP3 and Ipaf. It carries out N-deacetylation of peptidoglycan to avoid this detection by toll-like receptor 2 and NOD-like receptors. L. monocytogenes modulates transcription of host immunity genes through modification of histones and chromatin remodelling. Furthermore, L. monocytogenes has recently been shown to avoid autophagy and induce apoptosis in immune effector cells. In this review we discuss some of these strategies, which have provided new insights into the interaction between L. monocytogenes and the immune response at a crucial stage of infection.
Collapse
Affiliation(s)
- Sinead C Corr
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
| | | |
Collapse
|
44
|
Jia Q, Lee BY, Clemens DL, Bowen RA, Horwitz MA. Recombinant attenuated Listeria monocytogenes vaccine expressing Francisella tularensis IglC induces protection in mice against aerosolized Type A F. tularensis. Vaccine 2009; 27:1216-29. [PMID: 19126421 DOI: 10.1016/j.vaccine.2008.12.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 12/11/2008] [Accepted: 12/11/2008] [Indexed: 10/21/2022]
Abstract
Fransicella tularensis, the causative agent of tularemia, is in the top category (Category A) of potential agents of bioterrorism. To develop a safer vaccine against aerosolized F. tularensis, we have employed an attenuated Listeria monocytogenes, which shares with F. tularensis an intracellular and extraphagosomal lifestyle, as a delivery vehicle for F. tularensis antigens. We constructed recombinant L. monocytogenes (rLm) vaccines stably expressing seven F. tularensis proteins including IglC (rLm/iglC), and tested their immunogenicity and protective efficacy against lethal F. tularensis challenge in mice. Mice immunized intradermally with rLm/iglC developed significant cellular immune responses to F. tularensis IglC as evidenced by lymphocyte proliferation and CD4+ and CD8+ T-cell intracellular expression of interferon gamma. Moreover, mice immunized with rLm/iglC were protected against lethal challenge with F. tularensis LVS administered by the intranasal route, a route chosen to mimic airborne infection, and, most importantly, against aerosol challenge with the highly virulent Type A F. tularensis SchuS4 strain.
Collapse
Affiliation(s)
- Qingmei Jia
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States
| | | | | | | | | |
Collapse
|
45
|
Loeffler DIM, Smolen K, Aplin L, Cai B, Kollmann TR. Fine-tuning the safety and immunogenicity of Listeria monocytogenes-based neonatal vaccine platforms. Vaccine 2008; 27:919-27. [PMID: 19059297 DOI: 10.1016/j.vaccine.2008.11.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2008] [Revised: 11/14/2008] [Accepted: 11/14/2008] [Indexed: 10/21/2022]
Abstract
We have developed virulence-attenuated strains of Listeria monocytogenes (Lm) that can be used as safe yet effective vaccine carriers for neonatal vaccination. Here we compare the vaccine efficacy of Lm based vaccine carrier candidates after only a single immunization in murine neonates and adults: Lm Delta(trpS actA) based strains that express and secrete multiple copies of the model antigen ovalbumin (OVA) either under the control of a phagosomal (P(hly)) or cytosolic (P(actA))-driven listerial promoter. While both strains induced high levels of antigen-specific primary and secondary CD8 and CD4 T cell responses, both neonatal and adult mice immunized with the phagosomal driven strain were significantly better protected against wildtype Lm challenge as compared to the naïve control group than mice immunized with the cytosolic driven strains. Interestingly, only neonatal mice immunized with the phagosomal driven strains generated high IgG antibody responses against OVA. Our phagosomal driven Lm-based vaccine platform presents the broadest (cellular & humoral response) and most efficient (highly protective) vaccine platform for neonatal vaccination yet described.
Collapse
Affiliation(s)
- Daniela I M Loeffler
- Department of Paediatrics, Child and Family Research Institute, Vancouver, BC, Canada.
| | | | | | | | | |
Collapse
|
46
|
Construction and characterization of an attenuated Listeria monocytogenes strain for clinical use in cancer immunotherapy. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 16:96-103. [PMID: 19020110 DOI: 10.1128/cvi.00274-08] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Listeria monocytogenes has been exploited previously as a vaccine vector for the delivery of heterologous proteins such as tumor-specific antigens for active cancer immunotherapy. However, for effective use of live vector in clinics, safety is a major concern. In the present study, we describe an irreversibly attenuated and highly immunogenic L. monocytogenes platform, the L. monocytogenes dal-, dat-, and actA-deleted strain that expresses the human prostate-specific antigen (PSA) using an antibiotic resistance marker-free plasmid (the dal dat DeltaactA 142 strain expressing PSA). Despite limited in vivo survival, the dal dat DeltaactA 142 strain was able to elicit efficient immune responses required for tumor clearance. Our results showed that immunization of mice with the dal dat DeltaactA 142 strain caused the regression of the tumors established by the prostate adenocarcinoma cell line expressing PSA. An evaluation of immunologic potency indicated that the dal dat DeltaactA 142 strain elicits a high frequency of PSA-specific immune responses. Interestingly, immunization with the dal dat DeltaactA 142 strain induced significant infiltration of PSA-specific T cells in the intratumoral milieu. Collectively, our data suggest that the dal dat DeltaactA 142 strain is a safe and potent vector for clinical use and that this platform may be further exploited as a potential candidate to express other single or multiple antigens for cancer immunotherapy.
Collapse
|
47
|
Constitutive Activation of the PrfA regulon enhances the potency of vaccines based on live-attenuated and killed but metabolically active Listeria monocytogenes strains. Infect Immun 2008; 76:3742-53. [PMID: 18541651 DOI: 10.1128/iai.00390-08] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recombinant vaccines derived from the facultative intracellular bacterium Listeria monocytogenes are presently undergoing early-stage clinical evaluation in oncology treatment settings. This effort has been stimulated in part due to preclinical results that illustrate potent activation of innate and adaptive immune effectors by L. monocytogenes vaccines, combined with efficacy in rigorous animal models of malignant and infectious disease. Here, we evaluated the immunologic potency of a panel of isogenic vaccine strains that varied only in prfA. PrfA is an intracellularly activated transcription factor that induces expression of virulence genes and encoded heterologous antigens (Ags) in appropriately engineered vaccine strains. Mutant strains with PrfA locked into a constitutively active state are known as PrfA* mutants. We assessed the impacts of three PrfA* mutants, G145S, G155S, and Y63C, on the immunologic potencies of live-attenuated and photochemically inactivated nucleotide excision repair mutant (killed but metabolically active [KBMA]) vaccines. While PrfA* substantially increased Ag expression in strains grown in broth culture, Ag expression levels were equivalent in infected macrophage and dendritic cell lines, conditions that more closely parallel those in the immunized host. However, only the prfA(G155S) allele conferred significantly enhanced vaccine potency to KBMA vaccines. In the KBMA vaccine background, we show that PrfA*(G155S) enhanced functional cellular immunity following an intravenous or intramuscular prime-boost immunization regimen. These results form the basis of a rationale for including the prfA(G155S) allele in future live-attenuated or KBMA L. monocytogenes vaccines advanced to the clinical setting.
Collapse
|
48
|
Selected prfA* mutations in recombinant attenuated Listeria monocytogenes strains augment expression of foreign immunogens and enhance vaccine-elicited humoral and cellular immune responses. Infect Immun 2008; 76:3439-50. [PMID: 18474644 DOI: 10.1128/iai.00245-08] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While recombinant Listeria monocytogenes strains can be explored as vaccine candidates, it is important to develop attenuated but highly immunogenic L. monocytogenes vaccine vectors. Here, prfA* mutations selected on the basis of upregulated expression of L. monocytogenes PrfA-dependent genes and proteins were assessed to determine their abilities to augment expression of foreign immunogens in recombinant L. monocytogenes vectors and therefore enhance vaccine-elicited immune responses (a prfA* mutation is a mutation that results in constitutive overexpression of PrfA and PrfA-dependent virulence genes; the asterisk distinguishes the mutation from inactivation or stop mutations). A total of 63 recombinant L. monocytogenes vaccine vectors expressing seven individual viral or bacterial immunogens each in nine different L. monocytogenes strains carrying wild-type prfA or having prfA* mutations were constructed and investigated. Mutations selected on the basis of increased PrfA activation in recombinant L. monocytogenes prfA* vaccine vectors augmented expression of seven individual protein immunogens remarkably. Consistently, prime and boost vaccination studies with mice indicated that the prfA(G155S) mutation in recombinant L. monocytogenes DeltaactA prfA* strains enhanced vaccine-elicited cellular immune responses. Surprisingly, the prfA(G155S) mutation was found to enhance vaccine-elicited humoral immune responses as well. The highly immunogenic recombinant L. monocytogenes DeltaactA prfA* vaccine strains were as attenuated as the recombinant parent L. monocytogenes DeltaactA vaccine vector. Thus, recombinant attenuated L. monocytogenes DeltaactA prfA* vaccine vectors potentially are better antimicrobial and anticancer vaccines.
Collapse
|
49
|
Abstract
Listeria monocytogenes is an ubiquitous gram positive bacterium responsible for a severe food borne disease in human and animals which has become a multifaceted model. Indeed, it is a facultative intracellular bacterium that induces an amazingly rapid and sterilizing T-cell response and has been--and still is--widely used by immunologists. Listeria's strategies to invade non-phagocytic cells and spread from cell to cell have been and still are outstandingly instrumental to address key questions in cell biology. Orally acquired listeriosis can now be studied in relevant animal models. Finally, Listeria is among the few bacteria which have so rapidly benefited from comparative genomics. It now ranks among the most documented pathogens.
Collapse
Affiliation(s)
- Pascale Cossart
- Unité des interactions Bactéries-Cellules, INSERM U604, INRA USC 2020, Institut Pasteur, 28 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|