1
|
Gao S, Zuo W, Kang C, Zou Z, Zhang K, Qiu J, Shang X, Li J, Zhang Y, Zuo Q, Zhao Y, Jin M. Saccharomyces cerevisiae oral immunization in mice using multi-antigen of the African swine fever virus elicits a robust immune response. Front Immunol 2024; 15:1373656. [PMID: 38742108 PMCID: PMC11089227 DOI: 10.3389/fimmu.2024.1373656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
African swine fever virus (ASFV) is one of the most complex viruses. ASFV is a serious threat to the global swine industry because no commercial vaccines against this virus are currently available except in Vietnam. Moreover, ASFV is highly stable in the environment and can survive in water, feed, and aerosols for a long time. ASFV is transmitted through the digestive and respiratory tract. Mucosal immunity is the first line of defense against ASFV. Saccharomyces cerevisiae (SC), which has been certified by the U.S. Food and Drug Administration and has a generally recognized as safe status in the food industry, was used for oral immunization in this study. ASFV antigens were effectively expressed in recombinant SC strains with high DNA copy numbers and stable growth though surface display technology and chromosome engineering (δ-integration). The recombinant SC strains containing eight ASFV antigens-KP177R, E183L, E199L, CP204L, E248R, EP402R, B602L, and B646L- induced strong humoral and mucosal immune responses in mice. There was no antigenic competition, and these antigens induced Th1 and Th2 cellular immune responses. Therefore, the oral immunization strategy using recombinant SC strains containing multiple ASFV antigens demonstrate potential for future testing in swine, including challenge studies to evaluate its efficacy as a vaccine against ASFV.
Collapse
Affiliation(s)
- Shuo Gao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Wenfeng Zuo
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Chao Kang
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Zhong Zou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Kaiqi Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jun Qiu
- College of Animal Sciences, Yangtze University, Jingzhou, Hubei, China
| | - Xiaomin Shang
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Jingjing Li
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Yuanfeng Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Qi Zuo
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Ya Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Meilin Jin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
2
|
Zhao B, Guo Y, Sun R, Zhang L, Yang L, Mei X, Zhang L, Huang J. Quadrivalent hemagglutinin and adhesion expressed on Saccharomyces cerevisiae induce protective immunity against Mycoplasma gallisepticum infection and improve gut microbiota. Microb Pathog 2024; 187:106511. [PMID: 38168552 DOI: 10.1016/j.micpath.2023.106511] [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: 10/14/2023] [Revised: 12/01/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Mycoplasma gallisepticum (MG) infection causes infectious respiratory diseases in poultry, causing economic losses to the poultry industry. Therefore, this study aims to develop a safe, convenient, and effective multivalent recombinant Saccharomyces cerevisiae vaccine candidate and to explore its potential for oral immunization as a subunit vaccine. Mycoplasma gallisepticum Cytadhesin (MGC) and variable lipoprotein and hemagglutinin (vlhA) are associated with the pathogenesis of MG. In this study, a quadrivalent recombinant Saccharomyces cerevisiae (ST1814G-MG) displaying on MGC2, MGC3, VLH5, and VLH3, proteins was innovatively constructed, and its protective efficiency was evaluated in birds. The results showed that oral immunization with ST1814G-MG stimulates specific antibodies in chickens, reshapes the composition of the gut microbiota, reduces the Mycoplasma loading and pulmonary disease injury in the lungs. In addition, we found that oral ST1814G-MG had better protection against MG infection than an inactivated vaccine, and co-administration with the inactivated vaccine was even more effective. The results suggest that ST1814G-MG is a potentially safer and effective agent for controlling MG infection.
Collapse
Affiliation(s)
- Baiping Zhao
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Liu Yang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Xuefeng Mei
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Lei Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
3
|
Austriaco N. Yeast oral vaccines against infectious diseases. Front Microbiol 2023; 14:1150412. [PMID: 37138614 PMCID: PMC10149678 DOI: 10.3389/fmicb.2023.1150412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Vaccines that are delivered orally have several advantages over their counterparts that are administered via injection. Despite the advantages of oral delivery, however, approved oral vaccines are currently limited either to diseases that affect the gastrointestinal tract or to pathogens that have a crucial life cycle stage in the gut. Moreover, all of the approved oral vaccines for these diseases involve live-attenuated or inactivated pathogens. This mini-review summarizes the potential and challenges of yeast oral vaccine delivery systems for animal and human infectious diseases. These delivery systems utilize whole yeast recombinant cells that are consumed orally to transport candidate antigens to the immune system of the gut. This review begins with a discussion of the challenges associated with oral administration of vaccines and the distinct benefits offered by whole yeast delivery systems over other delivery systems. It then surveys the emerging yeast oral vaccines that have been developed over the past decade to combat animal and human diseases. In recent years, several candidate vaccines have emerged that can elicit the necessary immune response to provide significant protection against challenge by pathogen. They serve as proof of principle to show that yeast oral vaccines hold much promise.
Collapse
|
4
|
How CW, Ong YS, Low SS, Pandey A, Show PL, Foo JB. How far have we explored fungi to fight cancer? Semin Cancer Biol 2022; 86:976-989. [PMID: 33737109 DOI: 10.1016/j.semcancer.2021.03.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/21/2021] [Accepted: 03/13/2021] [Indexed: 01/01/2023]
Abstract
The use of fungal cultures have been well documented in human history. Although its used in healthcare, like penicillin and statins, have saved countless of lives, but there is still no fungal products that are specifically indicated for cancers. Research into fungal-derived materials to curb cancers in the recent decades have made a considerable progress in terms of drug delivery vehicles, anticancer active ingredients and cancer immunotherapy. Various parts of the organisms have successfully been exploited to achieve specific tasks. Apart from the identification of novel anticancer compound from fungi, its native capsular structure can also be used as drug cargo to achieve higher oral bioavailability. This review summarises the anticancer potential of fungal-derived materials, highlighting the role of capsular polysaccharides, proteins, and other structures in variety of innovative utilities to fit the current pharmaceutical technology. Many bioactive compounds isolated from fungi have also been formulated into nanoparticles to achieve greater anticancer activity. The progress of fungal compounds and their analogues in clinical trials is also highlighted. In addition, the potential of various fungal species to be developed for anticancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Chee Wun How
- School of Pharmacy, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Sze Shin Low
- Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor Darul Ehsan, Malaysia
| | - Ashok Pandey
- Centre for Innovation and Translational Research, CSIR-Indian Institute of Toxicology Research, Lucknow, 226 001, India
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor Darul Ehsan, Malaysia.
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia; Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
5
|
So KK, Chun J, Luong NN, Seo HW, Kim DH. Expression of an immunocomplex consisting of Fc fragment fused with a consensus dengue envelope domain III in Saccharomyces cerevisiae. Biotechnol Lett 2021; 43:1895-1904. [PMID: 34245387 PMCID: PMC8272446 DOI: 10.1007/s10529-021-03161-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
Objectives To explore Saccharomyces cerevisiae as an expression platform for dengue oral immune complex vaccine development. Results Molecular engineering was applied to create a fusion gene construct (scEDIII-PIGS) consisting of a yeast codon optimized sequence encoding for a synthetic consensus dengue envelope domain III (scEDIII) followed by a modified IgG Fc domain (PIGS). Northern blot showed transcription of the target gene, with a temporal expression pattern similar to those from previous work. Western blot showed assembly of various immune complexes from monomer to hexamer. Partial purification of scEDIII-PIGS was also attempted to demonstrate the feasibility of yeast system for immune complex vaccine development. Approximately 1 mg of scEDIII-PIGS can be produced from 1 l culture. Conclusion This work demonstrated for the first time that various immunocomplex structures of our target protein could be efficiently produced in S. cerevisiae for future application in developing oral and injectable vaccines against various pathogens. Supplementary Information The online version contains supplementary material available at 10.1007/s10529-021-03161-7.
Collapse
Affiliation(s)
- Kum-Kang So
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Jeesun Chun
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Nguyen Ngoc Luong
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Hee-Won Seo
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
6
|
Muscarella P, Bekaii-Saab T, McIntyre K, Rosemurgy A, Ross SB, Richards DA, Fisher WE, Flynn PJ, Mattson A, Coeshott C, Roder H, Roder J, Harrell FE, Cohn A, Rodell TC, Apelian D. A Phase 2 Randomized Placebo-Controlled Adjuvant Trial of GI-4000, a Recombinant Yeast Expressing Mutated RAS Proteins in Patients with Resected Pancreas Cancer. J Pancreat Cancer 2021; 7:8-19. [PMID: 33786412 PMCID: PMC7997807 DOI: 10.1089/pancan.2020.0021] [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] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose: GI-4000, a series of recombinant yeast expressing four different mutated RAS proteins, was evaluated in subjects with resected ras-mutated pancreas cancer. Methods: Subjects (n = 176) received GI-4000 or placebo plus gemcitabine. Subjects' tumors were genotyped to identify which matched GI-4000 product to administer. Immune responses were measured by interferon-γ (IFNγ) ELISpot assay and by regulatory T cell (Treg) frequencies on treatment. Pretreatment plasma was retrospectively analyzed by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-ToF) mass spectrometry for proteomic signatures predictive of GI-4000 responsiveness. Results: GI-4000 was well tolerated, with comparable safety findings between treatment groups. The GI-4000 group showed a similar pattern of median recurrence-free and overall survival (OS) compared with placebo. For the prospectively defined and stratified R1 resection subgroup, there was a trend in 1 year OS (72% vs. 56%), an improvement in OS (523.5 vs. 443.5 days [hazard ratio (HR) = 1.06 [confidence interval (CI): 0.53-2.13], p = 0.872), and increased frequency of immune responders (40% vs. 8%; p = 0.062) for GI-4000 versus placebo and a 159-day improvement in OS for R1 GI-4000 immune responders versus placebo (p = 0.810). For R0 resection subjects, no increases in IFNγ responses in GI-4000-treated subjects were observed. A higher frequency of R0/R1 subjects with a reduction in Tregs (CD4+/CD45RA+/Foxp3low) was observed in GI-4000-treated subjects versus placebo (p = 0.033). A proteomic signature was identified that predicted response to GI-4000/gemcitabine regardless of resection status. Conclusion: These results justify continued investigation of GI-4000 in studies stratified for likely responders or in combination with immune check-point inhibitors or other immunomodulators, which may provide optimal reactivation of antitumor immunity. ClinicalTrials.gov Number: NCT00300950.
Collapse
Affiliation(s)
- Peter Muscarella
- Department of Surgery, Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | | | | | - Sharona B Ross
- Digestive Disorders Institute, AdventHealth Tampa, Tampa, Florida, USA
| | | | | | - Patrick J Flynn
- Minnesota Oncology, US Oncology Research, Minneapolis, Minnesota, USA
| | - Alicia Mattson
- Smuggler Mountain Group (SMG, Inc.), Aspen, Colorado, USA
| | | | | | | | - Frank E Harrell
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Allen Cohn
- Rocky Mountain Cancer Center, Denver, Colorado, USA
| | | | - David Apelian
- Smuggler Mountain Group (SMG, Inc.), Aspen, Colorado, USA
| |
Collapse
|
7
|
Liu DQ, Lu S, Zhang L, Zhang LX, Ji M, Liu XG, Yu Z, Liu RT. A biomimetic yeast shell vaccine coated with layered double hydroxides induces a robust humoral and cellular immune response against tumors. NANOSCALE ADVANCES 2020; 2:3494-3506. [PMID: 36134256 PMCID: PMC9419453 DOI: 10.1039/d0na00249f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/25/2020] [Indexed: 05/05/2023]
Abstract
Enhancing both the humoral and cellular immune response for tumor vaccination remains a challenge. Inspired by natural pathogen structures, we took β-glucan particles derived from a baker's yeast cell shell (YS) as a vaccine carrier and danger signal for dendritic cells (DCs), and coated the YS with catanionic layered double hydroxides (LDH) by electrostatic adsorption to form a biomimetic yeast cell particle (YSL). Our experimental results showed that the YSL vaccine efficiently targeted antigen-presenting cells (APCs) and remarkably enhanced antigen cross-presentation, and strongly improved the activation and maturation of DCs. Moreover, the YSL vaccine elicited an extremely high antibody titer and strong antigen-specific cytotoxic T lymphocyte together with mixed Th1/Th17 cellular immune responses and induced marked prophylactic and therapeutic effects against E.G7-OVA tumors in mouse models. These results suggest that YSL, integrating a yeast shell to mimic natural pathogens and LDH with high antigen-loading capacity and lysosome escape, is a promising tumor vaccine platform for rapid, effective and strong induction of both humoral and cellular immune responses.
Collapse
Affiliation(s)
- Dong-Qun Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
- School of Chemical Engineering, University of Chinese Academy of Science Beijing 100049 China
| | - Shuai Lu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
| | - Lun Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
- School of Chemical Engineering, University of Chinese Academy of Science Beijing 100049 China
| | - Ling-Xiao Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
- School of Chemical Engineering, University of Chinese Academy of Science Beijing 100049 China
| | - Mei Ji
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
- School of Chemical Engineering, University of Chinese Academy of Science Beijing 100049 China
| | - Xiao-Ge Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
- School of Chemical Engineering, University of Chinese Academy of Science Beijing 100049 China
| | - Zhuo Yu
- Beijing Tsinghua Changgung Hospital Changping District Beijing. 102218 China +86 10 56118500 +86 10 56119544
| | - Rui-Tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Haidian District Beijing 100190 China +86 10 82545025 +86 10 82545017
| |
Collapse
|
8
|
Whole Recombinant Saccharomyces cerevisiae Yeast Expressing Ras Mutations as Treatment for Patients With Solid Tumors Bearing Ras Mutations: Results From a Phase 1 Trial. J Immunother 2019. [PMID: 29528991 PMCID: PMC5895167 DOI: 10.1097/cji.0000000000000219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are developing whole, heat-killed, recombinant Saccharomyces cerevisiae yeast, engineered to encode target proteins, which stimulate immune responses against malignant cells expressing those targets. This phase 1 trial, enrolling patients with advanced colorectal or pancreas cancer, was designed to evaluate safety, immunogenicity, response, and overall survival of ascending doses of the GI-4000 series of products, which express 3 different forms of mutated Ras proteins. The study enrolled 33 heavily pretreated subjects (14 with pancreas and 19 with colorectal cancer), whose tumors were genotyped before enrollment to identify the specific ras mutation and thereby to identify which GI-4000 product to administer. No dose limiting toxicities were observed and no subject discontinued treatment due to a GI-4000 related adverse event (AE). The majority of AEs and all fatal events were due to underlying disease progression and AE frequencies were not significantly different among dose groups. GI-4000 was immunogenic, as Ras mutation-specific immune responses were detected on treatment in ∼60% of subjects. No objective tumor responses were observed but based on imaging, clinical status and/or biochemical markers, stable disease was observed in 6 subjects (18%) on day 29, while 1 subject had stable disease at days 57 and 85 follow-up visits. The median overall survival was 3.3 months (95% confidence interval, 2.3–5.3 mo), and 5 subjects survived past the 48-week follow-up period. No significant dose-dependent trends for survival were observed. This first clinical trial in humans with GI-4000 demonstrated a favorable safety profile and immunogenicity in the majority of subjects.
Collapse
|
9
|
Xv Z, Lv J, Jiang J, Wang W, Feng F, Zhang L, Xue X, Li W. Effective Neutralizing Antibody Produced in Mice Directly Immunized with Integrated Pichia pastoris Expressing HPV16L1 Protein. Viral Immunol 2019; 32:308-317. [DOI: 10.1089/vim.2019.0055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Zhen Xv
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Jinhui Lv
- Research Center for Translational Medicine, East Hospital Tongji University School of Medicine, Shanghai, China
| | - Jie Jiang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Wenhuan Wang
- Key Laboratory of Uterology of Wenzhou City People's Hospital, Wenzhou, China
| | - Fangfang Feng
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Lifang Zhang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Wenshu Li
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Sneaking Out for Happy Hour: Yeast-Based Approaches to Explore and Modulate Immune Response and Immune Evasion. Genes (Basel) 2019; 10:genes10090667. [PMID: 31480411 PMCID: PMC6770942 DOI: 10.3390/genes10090667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 01/09/2023] Open
Abstract
Many pathogens (virus, bacteria, fungi, or parasites) have developed a wide variety of mechanisms to evade their host immune system. The budding yeast Saccharomyces cerevisiae has successfully been used to decipher some of these immune evasion strategies. This includes the cis-acting mechanism that limits the expression of the oncogenic Epstein–Barr virus (EBV)-encoded EBNA1 and thus of antigenic peptides derived from this essential but highly antigenic viral protein. Studies based on budding yeast have also revealed the molecular bases of epigenetic switching or recombination underlying the silencing of all except one members of extended families of genes that encode closely related and highly antigenic surface proteins. This mechanism is exploited by several parasites (that include pathogens such as Plasmodium, Trypanosoma, Candida, or Pneumocystis) to alternate their surface antigens, thereby evading the immune system. Yeast can itself be a pathogen, and pathogenic fungi such as Candida albicans, which is phylogenetically very close to S. cerevisiae, have developed stealthiness strategies that include changes in their cell wall composition, or epitope-masking, to control production or exposure of highly antigenic but essential polysaccharides in their cell wall. Finally, due to the high antigenicity of its cell wall, yeast has been opportunistically exploited to create adjuvants and vectors for vaccination.
Collapse
|
11
|
Kumar R, Kumar P. Yeast-based vaccines: New perspective in vaccine development and application. FEMS Yeast Res 2019; 19:5298404. [PMID: 30668686 DOI: 10.1093/femsyr/foz007] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
In presently licensed vaccines, killed or attenuated organisms act as a source of immunogens except for peptide-based vaccines. These conventional vaccines required a mass culture of associated or related organisms and long incubation periods. Special requirements during storage and transportation further adds to the cost of vaccine preparations. Availability of complete genome sequence, well-established genetic, inherent natural adjuvant and non-pathogenic nature of yeast species viz. Saccharomyces cerevisiae, Pichia pastoris makes them an ideal model system for the development of vaccines both for public health and for on-farm consumption. In this review, we compile the work in this emerging field during last two decades with major emphases on S. cerevisiae and P. pastoris which are routinely used worldwide for expression of heterologous proteins with therapeutic value against infectious diseases along with possible use in cancer therapy. We also pointed towards the developments in use of whole recombinant yeast, yeast surface display and virus-like particles as a novel strategy in the fight against infectious diseases and cancer along with other aspects including suitability of yeast in vaccines preparations, yeast cell wall component as an immune stimulator or modulator and present status of yeast-based vaccines in clinical trials.
Collapse
Affiliation(s)
- Ravinder Kumar
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Piyush Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400 076, Maharashtra, India
| |
Collapse
|
12
|
Tackling Cancer with Yeast-Based Technologies. Trends Biotechnol 2019; 37:592-603. [DOI: 10.1016/j.tibtech.2018.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/24/2018] [Accepted: 11/30/2018] [Indexed: 12/19/2022]
|
13
|
Bal J, Jung HY, Nguyen LN, Park J, Jang YS, Kim DH. Evaluation of cell-surface displayed synthetic consensus dengue EDIII cells as a potent oral vaccine candidate. Microb Cell Fact 2018; 17:146. [PMID: 30217208 PMCID: PMC6138890 DOI: 10.1186/s12934-018-0994-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/10/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Dengue is a rapidly spreading mosquito borne tropical viral disease affecting hundreds of millions of people across the globe annually. The dengue virus (DENV) includes four genetically distinct serotypes that cause serious life-threatening infections, including dengue hemorrhagic fever/dengue shock syndrome. Dengue vaccine development is complicated by the possibility of vaccine-enhanced severe dengue disease due to antibody-dependent enhancement by pre-existing cross-reactivity, as well as homotypic antibodies. Thus, the development of an efficacious dengue vaccine conferring simultaneous and durable immunity to each of the four DENV serotypes has not yet been developed despite years of research. For mass immunization in deeply affected resource-limited countries, oral vaccination is considered more beneficial than conventional approaches. Therefore, in a continuing effort towards designing economical and potent vaccine candidates, the current study applied yeast surface display technology to develop an oral dengue vaccine candidate using whole recombinant yeast cells displaying the recombinant fusion protein of M cell targeting ligand Co1 fused to the synthetic consensus dengue envelope domain III (scEDIII). Female Balb/c mice were orally fed with recombinant yeast cells and immunogenicity in terms of systemic and mucosal immune responses was monitored. RESULTS Immunofluorescence microscopy with dengue specific antibody and fluorescein isothiocyanate-conjugated anti-mouse IgG antibody clearly showed that recombinant protein Co1-scEDIII-AGA was localized on the cell surface of the respective clones in comparison with scEDIII-Co1 and Mock cells with no fluorescence. Oral dosage applications of surface displayed Co1-scEDIII-AGA stimulated a systemic humoral immune response in the form of dengue-specific serum IgG, as well as a mucosal immune response in the form of secretory immunoglobulin A (sIgA). Antigen-specific B cell responses in isolated lymphoid cells from the spleen and Peyer's patches further supported an elevated mucosal immune response. In addition, surface displayed Co1-scEDIII-AGA feeding elicited strong immune responses in comparison with scEDIII-Co1 and Mock following intraperitoneal booster with purified scEDIII antigen. CONCLUSIONS Surface displayed preparations of Co1-scEDIII-AGA induced strong immunogenicity compared with non-displayed scEDIII-Co1. Prior studies have supported the neutralization potential of scEDIII constructs against all four serotypes. Thus, the oral administration of genetically engineered yeast whole cells displaying biologically active Co1-scEDIII fusion protein without any further processing shows prospective as a potent oral vaccine candidate against dengue viral infection.
Collapse
Affiliation(s)
- Jyotiranjan Bal
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Hee-Young Jung
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Luong Ngoc Nguyen
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Jisang Park
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Yong-Suk Jang
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| |
Collapse
|
14
|
Liu DQ, Lu S, Zhang LX, Ji M, Liu SY, Wang SW, Liu RT. An indoleamine 2, 3-dioxygenase siRNA nanoparticle-coated and Trp2-displayed recombinant yeast vaccine inhibits melanoma tumor growth in mice. J Control Release 2018; 273:1-12. [DOI: 10.1016/j.jconrel.2018.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 10/18/2022]
|
15
|
GI-19007, a Novel Saccharomyces cerevisiae-Based Therapeutic Vaccine against Tuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00245-17. [PMID: 29046306 PMCID: PMC5717186 DOI: 10.1128/cvi.00245-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/05/2017] [Indexed: 01/05/2023]
Abstract
As yet, very few vaccine candidates with activity in animals against Mycobacterium tuberculosis infection have been tested as therapeutic postexposure vaccines. We recently described two pools of mycobacterial proteins with this activity, and here we describe further studies in which four of these proteins (Rv1738, Rv2032, Rv3130, and Rv3841) were generated as a fusion polypeptide and then delivered in a novel yeast-based platform (Tarmogen) which itself has immunostimulatory properties, including activation of Toll-like receptors. This platform can deliver antigens into both the class I and class II antigen presentation pathways and stimulate strong Th1 and Th17 responses. In mice this fusion vaccine, designated GI-19007, was immunogenic and elicited strong gamma interferon (IFN-γ) and interleukin-17 (IL-17) responses; despite this, they displayed minimal prophylactic activity in mice that were subsequently infected with a virulent clinical strain. In contrast, in a therapeutic model in the guinea pig, GI-19007 significantly reduced the lung bacterial load and reduced lung pathology, particularly in terms of secondary lesion development, while significantly improving survival in one-third of these animals. In further studies in which guinea pigs were vaccinated with BCG before challenge, therapeutic vaccination with GI-19007 initially improved survival versus that of animals given BCG alone, although this protective effect was gradually lost at around 400 days after challenge. Given its apparent ability to substantially limit bacterial dissemination within and from the lungs, GI-19007 potentially can be used to limit lung damage as well as facilitating chemotherapeutic regimens in infected individuals.
Collapse
|
16
|
Hdeib A, Wright JM. Brachyury-targeting immunotherapy for chordomas: treatment with GI-6301, a Saccharomyces cerevisiae yeast-based cancer vaccine. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1331126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alia Hdeib
- Department of Neurosurgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - James M. Wright
- Department of Neurosurgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
17
|
A poxviral-based cancer vaccine the transcription factor twist inhibits primary tumor growth and metastases in a model of metastatic breast cancer and improves survival in a spontaneous prostate cancer model. Oncotarget 2016; 6:28194-210. [PMID: 26317648 PMCID: PMC4695054 DOI: 10.18632/oncotarget.4442] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/23/2015] [Indexed: 01/11/2023] Open
Abstract
Several transcription factors play a role in the alteration of gene expression that occurs during cancer metastasis. Twist expression has been shown to be associated with the hallmarks of the metastatic process, as well as poor prognosis and drug resistance in many tumor types. However, primarily due to their location within the cell and the lack of a hydrophobic groove required for drug attachment, transcription factors such as Twist are difficult to target with conventional therapies. An alternative therapeutic strategy is a vaccine comprised of a Modified vaccinia Ankara (MVA), incorporating the Twist transgene and a TRIad of COstimulatory Molecules (B7-1, ICAM-1, LFA-3; TRICOM). Here we characterize an MVA-TWIST/TRICOM vaccine that induced both CD4+ and CD8+ Twist-specific T-cell responses in vivo. In addition, administration of this vaccine reduced both the primary tumor growth and metastasis in the 4T1 model of metastatic breast cancer. In the TRAMP transgenic model of spontaneous prostate cancer, MVA-TWIST/TRICOM alone significantly improved survival, and when combined with the androgen receptor antagonist enzalutamide, the vaccine further improved survival. These studies thus provide a rationale for the use of active immunotherapy targeting transcription factors involved in the metastatic process and for the combination of cancer vaccines with androgen deprivation.
Collapse
|
18
|
SiRNA In Vivo-Targeted Delivery to Murine Dendritic Cells by Oral Administration of Recombinant Yeast. Methods Mol Biol 2016; 1364:165-81. [PMID: 26472450 DOI: 10.1007/978-1-4939-3112-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
SiRNA therapeutics promise a future where any target in the transcriptome could be potentially addressed. However, the delivery of SiRNAs and targeting of particular cell types or organs are major challenges. A novel, efficient, and safe delivery system for promising the introduction of SiRNAs into particular cell types within living organisms is of great significance. Our previous studies have proved that recombinant protein (MSTN) and exogenous gene (EGFP) as vaccines, and furthermore functional CD40 shRNA expression can be delivered into dendritic cells (DCs) in mouse by oral administration of recombinant yeast (Saccharomyces cerevisiae). Here, we describe the details of the promising and innovative approach based on oral administration of recombinant yeast that allows in vivo-targeted delivery of functional SiRNA to murine intestinal DCs.
Collapse
|
19
|
Ahn DH, Williams TM, Goldstein DA, El-Rayes B, Bekaii-Saab T. Adjuvant therapy for pancreas cancer in an era of value based cancer care. Cancer Treat Rev 2016; 42:10-7. [PMID: 26620819 PMCID: PMC4976619 DOI: 10.1016/j.ctrv.2015.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/08/2015] [Accepted: 11/12/2015] [Indexed: 12/16/2022]
Abstract
In resected pancreas cancer, adjuvant therapy improves outcomes and is considered the standard of care for patients who recover sufficiently post operatively. Chemotherapy or combined chemotherapy and radiation therapy (chemoradiation; CRT) are strategies used in the adjuvant setting. However, there is a lack of evidence to suggest whether the addition of RT to chemotherapy translates to an improvement in clinical outcomes. This is true even when accounting for the subset of patients with a higher risk for recurrence, such as those with R1 and lymph node positive disease. When considering the direct and indirect costs, impact on quality of life and questionable added clinical benefit, the true "net health benefit" from added RT to chemotherapy becomes more uncertain. Future directions, including the utilization of modern RT, integration of novel therapies, and intensifying chemotherapy regimens may improve outcomes in resected pancreas cancer.
Collapse
Affiliation(s)
- Daniel H Ahn
- The Ohio State University Wexner Medical Center, 310 W. 10th Ave, Columbus, OH, United States
| | - Terence M Williams
- The Ohio State University Wexner Medical Center, 310 W. 10th Ave, Columbus, OH, United States
| | - Daniel A Goldstein
- Winship Cancer Institute, Emory University, 1365-C Clifton Rd NE, Atlanta, GA, United States
| | - Bassel El-Rayes
- Winship Cancer Institute, Emory University, 1365-C Clifton Rd NE, Atlanta, GA, United States
| | - Tanios Bekaii-Saab
- The Ohio State University Wexner Medical Center, 310 W. 10th Ave, Columbus, OH, United States.
| |
Collapse
|
20
|
Yan N, Xu K, Li X, Liu Y, Bai Y, Zhang X, Han B, Chen Z, Zhang Z. Recombinant Saccharomyces cerevisiae serves as novel carrier for oral DNA vaccines in Carassius auratus. FISH & SHELLFISH IMMUNOLOGY 2015; 47:758-765. [PMID: 26481518 DOI: 10.1016/j.fsi.2015.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
Oral delivery of DNA vaccines represents a promising vaccinating method for fish. Recombinant yeast has been proved to be a safe carrier for delivering antigen proteins and DNAs to some species in vivo. However, whether recombinant yeast can be used to deliver functional DNAs for vaccination to fish is still unknown. In this study, red crucian carp (Carassius auratus) was orally administrated with recombinant Saccharomyces cerevisiae harboring CMV-EGFP expression cassette. On day 5 post the first vaccination, EGFP expression in the hindgut was detected under fluorescence microscope. To further study whether the delivered gene could induce specific immune responses, the model antigen ovalbumin (OVA) was used as immunogen, and oral administrations were conducted with recombinant S. cerevisiae harboring pCMV-OVA mammalian gene expression cassette as gene delivery or pADH1-OVA yeast gene expression cassette as protein delivery. Each administration was performed with three different doses, and the OVA-specific serum antibody was detected in all the experimental groups by western blotting and enzyme-linked immunosorbent assay (ELISA). ELISA assay also revealed that pCMV-OVA group with lower dose (pCMV-OVA-L) and pADH1-OVA group with moderate dose (pADH1-OVA-M) triggered relatively stronger antibody response than the other two doses. Moreover, the antibody level induced by pCMV-OVA-L group was significantly higher than pADH1-OVA-M group at the same serum dilutions. All the results suggested that recombinant yeast can be used as a potential carrier for oral DNA vaccines and would help to develop more practical strategies to control infectious diseases in aquaculture.
Collapse
Affiliation(s)
- Nana Yan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Kun Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xinyi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yuwan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yichun Bai
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaohan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Baoquan Han
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhilong Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhiying Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
21
|
Bolhassani A, Muller M, Roohvand F, Motevalli F, Agi E, Shokri M, Rad MM, Hosseinzadeh S. Whole recombinant Pichia pastoris expressing HPV16 L1 antigen is superior in inducing protection against tumor growth as compared to killed transgenic Leishmania. Hum Vaccin Immunother 2015; 10:3499-508. [PMID: 25668661 DOI: 10.4161/21645515.2014.979606] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The development of an efficient vaccine against high-risk HPV types can reduce the incidence rates of cervical cancer by generating anti-tumor protective responses. Traditionally, the majority of prophylactic viral vaccines are composed of live, attenuated or inactivated viruses. Among them, the design of an effective and low-cost vaccine is critical. Inactivated vaccines especially heat-killed yeast cells have emerged as a promising approach for generating antigen-specific immunotherapy. Recent studies have indicated that yeast cell wall components possess adjuvant activities. Moreover, a non-pathogenic protozoan, Leishmania tarentolae (L.tar) has attracted a great attention as a live candidate vaccine. In current study, immunological and protective efficacy of whole recombinant killed Pichia pastoris and Leishmania tarentolae expressing HPV16 L1 capsid protein was evaluated in tumor mice model. We found that Pichia-L1, L.tar-L1 and Gardasil groups increase the IgG2a/IgG1 ratio, indicating a relative preference for the induction of Th1 immune responses. Furthermore, subcutaneous injection of killed Pichia-L1 generated the significant L1-specific IFN-γ immune response as well as the best protective effects in vaccinated mice as compared to killed L.tar-L1, killed Pichia pastoris, killed L.tar and PBS groups. Indeed, whole recombinant Leishmania tarentolae could not protect mice against C3 tumor mice model. These data suggest that Pichia-L1 may be a candidate for the control of HPV infections.
Collapse
Key Words
- 2-ME, mercaptoethanol
- AOX1, alcohol oxidase I gene
- ConA, concanavalin A
- DAB, 3,3′-diaminobenzidine
- FACS, fluorescence-activated cell sorting
- GFP, green fluorescent protein
- HPV, human papillomaviruses
- KBMA, killed but metabolicallyactive
- L.tar, Leishmania tarentolae
- L1 capsid protein
- Leishmania tarentolae expression system
- Pichia pastoris expression system
- SD, standard deviation
- Yeast-HBsAg, yeast expressing hepatitis B surface antigen
- cervical cancer
- human papillomavirus
- killed vaccine
- rL1, recombinant L1
Collapse
Affiliation(s)
- Azam Bolhassani
- a Department of Hepatitis and AIDs; Pasteur Institute of Iran ; Tehran , Iran.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Palena C, Hamilton DH. Immune Targeting of Tumor Epithelial-Mesenchymal Transition via Brachyury-Based Vaccines. Adv Cancer Res 2015. [PMID: 26216630 DOI: 10.1016/bs.acr.2015.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As a manifestation of their inherent plasticity, carcinoma cells undergo profound phenotypic changes during progression toward metastasis. One such phenotypic modulation is the epithelial-mesenchymal transition (EMT), an embryonically relevant process that can be reinstated by tumor cells, resulting in the acquisition of metastatic propensity, stem-like cell properties, and resistance to a variety of anticancer therapies, including chemotherapy, radiation, and some small-molecule targeted therapies. Targeting of the EMT is emerging as a novel intervention against tumor progression. This review focuses on the potential use of cancer vaccine strategies targeting tumor cells that exhibit mesenchymal-like features, with an emphasis on the current status of development of vaccine platforms directed against the T-box transcription factor brachyury, a novel cancer target involved in tumor EMT, stemness, and resistance to therapies. Also presented is a summary of potential mechanisms of resistance to immune-mediated attack driven by EMT and the development of novel combinatorial strategies based on the use of agents that alleviate tumor EMT for an optimized targeting of plastic tumor cells that are responsible for tumor recurrence and the establishment of therapeutic refractoriness.
Collapse
Affiliation(s)
- Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - Duane H Hamilton
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
23
|
Bloy N, Buqué A, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautès-Fridman C, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Naked and vectored DNA-based anticancer vaccines. Oncoimmunology 2015; 4:e1026531. [PMID: 26155408 PMCID: PMC4485755 DOI: 10.1080/2162402x.2015.1026531] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 12/28/2022] Open
Abstract
One type of anticancer vaccine relies on the administration of DNA constructs encoding one or multiple tumor-associated antigens (TAAs). The ultimate objective of these preparations, which can be naked or vectored by non-pathogenic viruses, bacteria or yeast cells, is to drive the synthesis of TAAs in the context of an immunostimulatory milieu, resulting in the (re-)elicitation of a tumor-targeting immune response. In spite of encouraging preclinical results, the clinical efficacy of DNA-based vaccines employed as standalone immunotherapeutic interventions in cancer patients appears to be limited. Thus, efforts are currently being devoted to the development of combinatorial regimens that allow DNA-based anticancer vaccines to elicit clinically relevant immune responses. Here, we discuss recent advances in the preclinical and clinical development of this therapeutic paradigm.
Collapse
Key Words
- AFP, α-fetoprotein
- APC, antigen-presenting cell
- CDR, complementarity-determining region
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CTLA4, cytotoxic T lymphocyte protein 4
- DAMP, damage-associated molecular pattern
- DC, dendritic cell
- FDA, Food and Drug Administration
- GM-CSF, granulocyte macrophage colony-stimulating factor
- GX-188E
- HCC, hepatocellular carcinoma
- HNSCC, head and neck squamous cell carcinoma
- HPV, human papillomavirus
- IL, interleukin
- OS, overall survival
- OVA, ovalbumin
- PAP, prostate acid phosphatase
- SCGB2A2, secretoglobin, family 2A, member 2
- SOX2, SRY (sex determining region Y)-box 2
- T, brachyury homolog
- TAA, tumor-associated antigen
- TLR, Toll-like receptor
- TRA, tumor rejection antigen
- Treg, regulatory T cell
- VGX-3100
- WT1, Wilms tumor 1
- adjuvants
- dendritic cell
- electroporation
- mucosal immunity
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
| | - Aitziber Buqué
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System; Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS); Barcelona, Spain
| | - Francesca Castoldi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Faculté de Medicine; Université Paris Sud/Paris XI; Le Kremlin-Bicêtre, France
- Sotio a.c; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM, U1138; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, U1138; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Jitka Fucikova
- Sotio a.c; Prague, Czech Republic
- Dept. of Immunology; 2 Faculty of Medicine and University Hospital Motol; Charles University; Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Radek Spisek
- Sotio a.c; Prague, Czech Republic
- Dept. of Immunology; 2 Faculty of Medicine and University Hospital Motol; Charles University; Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- INSERM, U970; Paris, France
- Paris-Cardiovascular Research Center (PARCC); Paris, France
- Service d'Immunologie Biologique; Hôpital Européen Georges Pompidou (HEGP); AP-HP; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
24
|
Assis-Marques MA, Oliveira AF, Ruas LP, dos Reis TF, Roque-Barreira MC, Coelho PSR. Saccharomyces cerevisiae expressing Gp43 protects mice against Paracoccidioides brasiliensis infection. PLoS One 2015; 10:e0120201. [PMID: 25790460 PMCID: PMC4366343 DOI: 10.1371/journal.pone.0120201] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/26/2015] [Indexed: 01/28/2023] Open
Abstract
The dimorphic fungus Paracoccidioides brasiliensis is the etiological agent of paracoccidioidomycosis (PCM). It is believed that approximately 10 million people are infected with the fungus and approximately 2% will eventually develop the disease. Unlike viral and bacterial diseases, fungal diseases are the ones against which there is no commercially available vaccine. Saccharomyces cerevisiae may be a suitable vehicle for immunization against fungal infections, as they require the stimulation of different arms of the immune response. Here we evaluated the efficacy of immunizing mice against PCM by using S. cerevisiae yeast expressing gp43. When challenged by inoculation of P. brasiliensis yeasts, immunized animals showed a protective profile in three different assays. Their lung parenchyma was significantly preserved, exhibiting fewer granulomas with fewer fungal cells than found in non-immunized mice. Fungal burden was reduced in the lung and spleen of immunized mice, and both organs contained higher levels of IL-12 and IFN-γ compared to those of non-vaccinated mice, a finding that suggests the occurrence of Th1 immunity. Taken together, our results indicate that the recombinant yeast vaccine represents a new strategy to confer protection against PCM.
Collapse
Affiliation(s)
- Mariana Aprigio Assis-Marques
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
| | - Aline Ferreira Oliveira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
| | - Luciana Pereira Ruas
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
| | - Thaila Fernanda dos Reis
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
| | - Maria Cristina Roque-Barreira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
| | - Paulo Sergio Rodrigues Coelho
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049–900, Brasil
- * E-mail:
| |
Collapse
|
25
|
Zhang X, Hu S, Du X, Li T, Han L, Kong J. Heterologous expression of carcinoembryonic antigen in Lactococcus lactis via LcsB-mediated surface displaying system for oral vaccine development. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2014; 49:851-858. [PMID: 25641594 DOI: 10.1016/j.jmii.2014.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/22/2014] [Accepted: 11/05/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND/PURPOSE Carcinoembryonic antigen (CEA) is an attractive target for immunotherapy because it is expressed minimally in normal tissue, but is overexpressed in a wide variety of malignant epithelial tissues. Lactic acid bacteria (LABs), widely used in food processes, are attractive candidates for oral vaccination. Thus, we examined whether LABs could be used as a live vaccine vector to deliver CEA antigen. METHODS CEA was cloned into an Escherichia coli/Lactococcus lactis shuttle vector pSEC:LEISS under the control of a nisin promoter. For displaying the CEA on the cell surface of the L. lactis strain, the anchor motif LcsB from the S-layer protein of Lactobacillus crispatus was fused with CEA. Intracellular and cell surface expression of the CEA-LcsB fusion was confirmed by western blot analysis. RESULTS Significantly higher levels of CEA-specific secretory immunoglobulin A in the sera of mice were observed upon oral administration of strain cultures containing the CEA-LcsB fused protein. In addition, the CEA-LcsB antigen group showed a higher spleen index compared to the CEA antigen alone or negative control, demonstrating that surface-displayed CEA antigen could induce a higher immune response. CONCLUSION These results provided the first evidence for displaying CEA antigen on the cell surfaces of LABs as oral vaccines against cancer or infectious diseases.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Health Science Exchange and Service Center of Jinan, Jinan 250013, China.
| | - Shumin Hu
- State Key Laboratory of Biological Fermentation Engineering of Beer, Tsingtao Brewery Co. Ltd., Qingdao, China; State Key Laboratory of Microbial Technology, Shandong University, Jinan 250100, China
| | - Xue Du
- Health Science Exchange and Service Center of Jinan, Jinan 250013, China
| | - Tiejun Li
- Health Science Exchange and Service Center of Jinan, Jinan 250013, China
| | - Lanlan Han
- State Key Laboratory of Microbial Technology, Shandong University, Jinan 250100, China
| | - Jian Kong
- State Key Laboratory of Microbial Technology, Shandong University, Jinan 250100, China.
| |
Collapse
|
26
|
Tucker JA, Jochems C, Boyerinas B, Fallon J, Greiner JW, Palena C, Rodell TC, Schlom J, Tsang KY. Identification and characterization of a cytotoxic T-lymphocyte agonist epitope of brachyury, a transcription factor involved in epithelial to mesenchymal transition and metastasis. Cancer Immunol Immunother 2014; 63:1307-17. [PMID: 25186612 DOI: 10.1007/s00262-014-1603-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/12/2014] [Indexed: 12/31/2022]
Abstract
The transcription factor brachyury is a major driver of epithelial to mesenchymal transition in human carcinoma cells. It is overexpressed in several human tumor types versus normal adult tissues, except for testes and thyroid. Overexpression is associated with drug resistance and poor prognosis. Previous studies identified a brachyury HLA-A2 cytotoxic T-lymphocyte epitope. The studies reported here describe an enhancer epitope of brachyury. Compared to the native epitope, the agonist epitope: (a) has enhanced binding to MHC class I, (b) increased the IFN-γ production from brachyury-specific T cells, (c) generated brachyury-specific T cells with greater levels of perforin and increased proliferation, (d) generated T cells more proficient at lysing human carcinoma cells endogenously expressing the native epitope, and (e) achieved greater brachyury-specific T-cell responses in vivo in HLA-A2 transgenic mice. These studies also report the generation of a heat-killed recombinant Saccharomyces cerevisiae (yeast) vector expressing the full-length brachyury gene encoding the agonist epitope. Compared to yeast-brachyury (native) devoid of the agonist epitope, the yeast-brachyury (agonist) enhanced the activation of brachyury-specific T cells, which efficiently lysed human carcinoma cells. In addition to providing the rationale for the recombinant yeast-brachyury (agonist) as a potential vaccine in cancer therapy, these studies also provide the rationale for the use of the agonist in (a) dendritic cell (DC) vaccines, (b) adjuvant or liposomal vaccines, (c) recombinant viral and/or bacterial vaccines, (d) protein/polypeptide vaccines, (e) activation of T cells ex vivo in adoptive therapy protocols, and (f) generation of genetically engineered targeted T cells.
Collapse
Affiliation(s)
- Jo A Tucker
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD, 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Immunological targeting of tumor cells undergoing an epithelial-mesenchymal transition via a recombinant brachyury-yeast vaccine. Oncotarget 2014; 4:1777-90. [PMID: 24125763 PMCID: PMC3858563 DOI: 10.18632/oncotarget.1295] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The embryonic T-box transcription factor brachyury is aberrantly expressed in a range of human tumors. Previous studies have demonstrated that brachyury is a driver of the epithelial-mesenchymal transition (EMT), a process associated with cancer progression. Brachyury expression in human tumor cells enhances tumor invasiveness in vitro and metastasis in vivo, and induces resistance to various conventional therapeutics including chemotherapy and radiation. These characteristics, and the selective expression of brachyury for a range of human tumor types vs. normal adult tissues, make brachyury an attractive tumor target. Due to its intracellular localization and the “undruggable” character of transcription factors, available options to target brachyury are currently limited. Here we report on the development and characterization of an immunological platform for the efficient targeting of brachyury-positive tumors consisting of a heat-killed, recombinant Saccharomyces cerevisiae (yeast)–brachyury vector-based vaccine (designated as GI-6301) that expresses the full-length human brachyury protein. We demonstrate that human dendritic cells treated with recombinant yeast-brachyury can activate and expand brachyury-specific CD4+ and CD8+ T cells in vitro that, in turn, can effectively lyse human tumor cells expressing the brachyury protein. Vaccination of mice with recombinant yeast-brachyury is also shown here to elicit brachyury-specific CD4+ and CD8+ T-cell responses, and to induce anti-tumor immunity in the absence of toxicity. Based on these results, a Phase I clinical trial of GI-6301 is currently ongoing in patients with advanced tumors; to our knowledge, this is the first vaccine platform aimed at targeting a driver of tumor EMT that has successfully reached the clinical stage.
Collapse
|
28
|
King TH, Kemmler CB, Guo Z, Mann D, Lu Y, Coeshott C, Gehring AJ, Bertoletti A, Ho ZZ, Delaney W, Gaggar A, Subramanian GM, McHutchison JG, Shrivastava S, Lee YJL, Kottilil S, Bellgrau D, Rodell T, Apelian D. A whole recombinant yeast-based therapeutic vaccine elicits HBV X, S and Core specific T cells in mice and activates human T cells recognizing epitopes linked to viral clearance. PLoS One 2014; 9:e101904. [PMID: 25051027 PMCID: PMC4106793 DOI: 10.1371/journal.pone.0101904] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/12/2014] [Indexed: 12/17/2022] Open
Abstract
Chronic hepatitis B infection (CHB) is characterized by sub-optimal T cell responses to viral antigens. A therapeutic vaccine capable of restoring these immune responses could potentially improve HBsAg seroconversion rates in the setting of direct acting antiviral therapies. A yeast-based immunotherapy (Tarmogen) platform was used to make a vaccine candidate expressing hepatitis B virus (HBV) X, surface (S), and Core antigens (X-S-Core). Murine and human immunogenicity models were used to evaluate the type and magnitude of HBV-Ag specific T cell responses elicited by the vaccine. C57BL/6J, BALB/c, and HLA-A*0201 transgenic mice immunized with yeast expressing X-S-Core showed T cell responses to X, S and Core when evaluated by lymphocyte proliferation assay, ELISpot, intracellular cytokine staining (ICS), or tumor challenge assays. Both CD4+ and CD8+ T cell responses were observed. Human T cells transduced with HBc18-27 and HBs183-91 specific T cell receptors (TCRs) produced interferon gamma (IFNγ following incubation with X-S-Core-pulsed dendritic cells (DCs). Furthermore, stimulation of peripheral blood mononuclear cells (PBMCs) isolated from CHB patients or from HBV vaccine recipients with autologous DCs pulsed with X-S-Core or a related product (S-Core) resulted in pronounced expansions of HBV Ag-specific T cells possessing a cytolytic phenotype. These data indicate that X-S-Core-expressing yeast elicit functional adaptive immune responses and supports the ongoing evaluation of this therapeutic vaccine in patients with CHB to enhance the induction of HBV-specific T cell responses.
Collapse
Affiliation(s)
- Thomas H. King
- GlobeImmune, Inc., Louisville, Colorado, United States of America
- * E-mail:
| | | | - Zhimin Guo
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| | - Derrick Mann
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| | - Yingnian Lu
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| | - Claire Coeshott
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| | - Adam J. Gehring
- Molecular Microbiology and Immunology & Saint Louis University Liver Center, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - Antonio Bertoletti
- Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - Zi Z. Ho
- Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - William Delaney
- Gilead Sciences Inc., Foster City, California, United States of America
| | - Anuj Gaggar
- Gilead Sciences Inc., Foster City, California, United States of America
| | | | | | - Shikha Shrivastava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu-Jin L. Lee
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shyamasundaran Kottilil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Donald Bellgrau
- GlobeImmune, Inc., Louisville, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Timothy Rodell
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| | - David Apelian
- GlobeImmune, Inc., Louisville, Colorado, United States of America
| |
Collapse
|
29
|
Zhang L, Zhang T, Wang L, Shao S, Chen Z, Zhang Z. In vivo targeted delivery of CD40 shRNA to mouse intestinal dendritic cells by oral administration of recombinant Sacchromyces cerevisiae. Gene Ther 2014; 21:709-14. [PMID: 24871580 PMCID: PMC4086734 DOI: 10.1038/gt.2014.50] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/07/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022]
Abstract
Short hairpin RNA (shRNA)-mediated gene regulation is a commonly used technique for gene manipulation. An efficient and safe delivery system is indispensable when shRNA is delivered into living organisms for gene therapy. Previous studies have proved that DNA and protein can be delivered into dendritic cells (DCs) by non-pathogenic Saccharomyces cerevisiae without being degraded. CD40 is closely related to apoptosis of tumor cells and some immune mechanisms. In this study, we demonstrated that recombinant yeast S. cerevisiae efficiently delivered the shRNA of immune-associated gene (CD40) into mouse intestinal DCs via oral administration. Western blot analysis of isolated intestinal DCs indicated that the inhibition of CD40 gene expression reached up to 56-91%. The secretion of cytokines such as interleukin-2 (IL-2), IL-6, IL-10, IL-12, tumor necrosis factor-α and interferon-γ in intestinal DCs had varying degrees of changes. In conclusion, we found that orally administered recombinant yeast can be used as an efficient shRNA delivery system for intestinal DC-specific gene silencing and immunomodulation in vivo.
Collapse
Affiliation(s)
- L Zhang
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| | - T Zhang
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| | - L Wang
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| | - S Shao
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| | - Z Chen
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| | - Z Zhang
- College of Animal Science and Technology, Shaan'xi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, YangLing, Shaan'xi, People's Republic of China
| |
Collapse
|
30
|
Ardiani A, Gameiro SR, Palena C, Hamilton DH, Kwilas A, King TH, Schlom J, Hodge JW. Vaccine-mediated immunotherapy directed against a transcription factor driving the metastatic process. Cancer Res 2014; 74:1945-57. [PMID: 24520078 DOI: 10.1158/0008-5472.can-13-2045] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Numerous reports have now demonstrated that the epithelial-to-mesenchymal transition (EMT) process is involved in solid tumor progression, metastasis, and drug resistance. Several transcription factors have been implicated as drivers of EMT and metastatic progression, including Twist. Overexpression of Twist has been shown to be associated with poor prognosis and drug resistance for many carcinomas and other tumor types. The role of Twist in experimental cancer metastases has been principally studied in the 4T1 mammary tumor model, where silencing of Twist in vitro has been shown to greatly reduce in vivo metastatic spread. Transcription factors such as Twist are generally believed to be "undruggable" because of their nuclear location and lack of a specific groove for tight binding of a small molecule inhibitor. An alternative approach to drug therapy targeting transcription factors driving the metastatic process is T-cell-mediated immunotherapy. A therapeutic vaccine platform that has been previously characterized consists of heat-killed recombinant Saccharomyces cerevisiae (yeast) capable of expressing tumor-associated antigen protein. We report here the construction and characterization of a recombinant yeast expressing the entire Twist protein, which is capable of inducing both CD8(+) and CD4(+) Twist-specific T-cell responses in vivo. Vaccination of mice reduced the size of primary transplanted 4T1 tumors and had an even greater antitumor effect on lung metastases of the same mice, which was dependent on Twist-specific CD8(+) T cells. These studies provide the rationale for vaccine-induced T-cell-mediated therapy of transcription factors involved in driving the metastatic process.
Collapse
Affiliation(s)
- Andressa Ardiani
- Authors' Affiliations: Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland; and GlobeImmune Inc., Louisville, Colorado
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhao H, Wang Y, Ma Z, Wang Y, Feng WH. Recombinant Kluyveromyces lactis expressing highly pathogenic porcine reproductive and respiratory syndrome virus GP5 elicits mucosal and cell-mediated immune responses in mice. J Vet Sci 2013; 15:199-208. [PMID: 24378591 PMCID: PMC4087221 DOI: 10.4142/jvs.2014.15.2.199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/23/2013] [Indexed: 11/25/2022] Open
Abstract
Currently, killed-virus and modified-live porcine reproductive and respiratory syndrome virus (PRRSV) vaccines are used to control porcine reproductive and respiratory syndrome. However, both types of vaccines have inherent drawbacks; accordingly, the development of novel PRRSV vaccines is urgently needed. Previous studies have suggested that yeast possesses adjuvant activities, and it has been used as an expression vehicle to elicit immune responses to foreign antigens. In this report, recombinant Kluyveromyces lactis expressing GP5 of HP-PRRSV (Yeast-GP5) was generated and immune responses to this construct were analyzed in mice. Intestinal mucosal PRRSV-specific sIgA antibody and higher levels of IFN-γ in spleen CD4+ and CD8+ T cells were induced by oral administration of Yeast-GP5. Additionally, Yeast-GP5 administered subcutaneously evoked vigorous cell-mediated immunity, and PRRSV-specific lymphocyte proliferation and IFN-γ secretion were detected in the splenocytes of mice. These results suggest that Yeast-GP5 has the potential for use as a vaccine for PRRSV in the future.
Collapse
Affiliation(s)
- Haiyan Zhao
- State Key Laboratory of Agrobiotechnology, Key Laboratory of Soil Microbiology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | | | | | | | | |
Collapse
|
32
|
Gameiro SR, Jammeh ML, Hodge JW. Cancer vaccines targeting carcinoembryonic antigen: state-of-the-art and future promise. Expert Rev Vaccines 2013; 12:617-29. [PMID: 23750792 DOI: 10.1586/erv.13.40] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Concurrent with the US FDA's approval of the first therapeutic cancer vaccine, and supported by mounting clinical evidence indicating that targeting carcinoembryonic antigen (CEA) can safely overcome pre-existing tolerance, a multitude of novel CEA cancer vaccines are now in various stages of development. Since cancer-driven immune suppression often limits the efficacy of vaccines, numerous strategies are being examined in both preclinical and clinical settings to overcome immunosuppressive elements, including the combined use of vaccines with certain chemotherapies, immune checkpoint inhibitors, small-molecule targeted therapies and radiation. This review discusses the current state and future direction of therapeutic cancer vaccines targeting CEA, based on advances achieved over the last 5 years.
Collapse
Affiliation(s)
- Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
33
|
Phase I trial of a recombinant yeast-CEA vaccine (GI-6207) in adults with metastatic CEA-expressing carcinoma. Cancer Immunol Immunother 2013; 63:225-34. [PMID: 24327292 DOI: 10.1007/s00262-013-1505-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 11/24/2013] [Indexed: 12/22/2022]
Abstract
Yeast-CEA (GI-6207) is a therapeutic cancer vaccine genetically modified to express recombinant carcinoembryonic antigen (CEA) protein, using heat-killed yeast (Saccharomyces cerevisiae) as a vector. In preclinical studies, yeast-CEA induced a strong immune response to CEA and antitumor responses. Patients received subcutaneous vaccines every 2 weeks for 3 months and then monthly. Patients were enrolled at 3 sequential dose levels: 4, 16, and 40 yeast units (10(7) yeast particles/unit). Eligible patients were required to have serum CEA > 5 ng/mL or > 20 % CEA(+) tumor block, ECOG PS 0-2, and no history of autoimmunity. Restaging scans were performed at 3 months and then bimonthly. Peripheral blood was collected for the analysis of immune response (e.g., by ELISPOT assay). Twenty-five patients with metastatic CEA-expressing carcinomas were enrolled. Median patient age was 52 (range 39-81). A total of 135 vaccines were administered. The vaccine was well tolerated, and the most common adverse event was grade 1/2 injection-site reaction. Five patients had stable disease beyond 3 months (range 3.5-18 months), and each had CEA stabilization while on-study. Some patients showed evidence post-vaccination of increases in antigen-specific CD8(+) T cells and CD4(+) T lymphocytes and decreases in regulatory T cells. Of note, a patient with medullary thyroid cancer had substantial T cell responses and a vigorous inflammatory reaction at sites of metastatic disease. Yeast-CEA vaccination had minimal toxicity and induced some antigen-specific T cell responses and CEA stabilization in a heterogeneous, heavily pre-treated patient population. Further studies are required to determine the clinical benefit of yeast-CEA vaccination.
Collapse
|
34
|
Ardiani A, Farsaci B, Rogers CJ, Protter A, Guo Z, King TH, Apelian D, Hodge JW. Combination therapy with a second-generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model. Clin Cancer Res 2013; 19:6205-18. [PMID: 24048332 PMCID: PMC3833876 DOI: 10.1158/1078-0432.ccr-13-1026] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Enzalutamide, a second-generation androgen antagonist, was approved by the U.S. Food and Drug Administration (FDA) for castration-resistant prostate cancer (CRPC) treatment. Immunotherapy has been shown to be a promising strategy for prostate cancer. This study was performed to provide data to support the combination of enzalutamide and immunotherapy for CRPC treatment. EXPERIMENTAL DESIGN Male C57BL/6 or TRAMP (transgenic adenocarcinoma of the mouse prostate) prostate cancer model mice were exposed to enzalutamide and/or a therapeutic vaccine targeting Twist, an antigen involved in epithelial-to-mesenchymal transition and metastasis. The physiologic and immunologic effects of enzalutamide were characterized. The generation of Twist-specific immunity by Twist-vaccine was assessed. Finally, the combination of enzalutamide and Twist-vaccine to improve TRAMP mice overall survival was evaluated. RESULTS Enzalutamide mediated immunogenic modulation in TRAMP-C2 cells. In vivo, enzalutamide mediated reduced genitourinary tissue weight, enlargement of the thymus, and increased levels of T-cell excision circles. Because no changes were seen in T-cell function, as determined by CD4(+) T-cell proliferation and regulatory T cell (Treg) functional assays, enzalutamide was determined to be immune inert. Enzalutamide did not diminish the ability of Twist-vaccine to generate Twist-specific immunity. Twist was confirmed as a valid tumor antigen in TRAMP mice by immunohistochemistry. The combination of enzalutamide and Twist-vaccine resulted in significantly increased overall survival of TRAMP mice compared with other treatment groups (27.5 vs. 10.3 weeks). Notably, the effectiveness of the combination therapy increased with disease stage, i.e., the greatest survival benefit was seen in mice with advanced-stage prostate tumors. CONCLUSIONS These data support the combination of enzalutamide and immunotherapy as a promising treatment strategy for CRPC. Clin Cancer Res; 19(22); 6205-18. ©2013 AACR.
Collapse
Affiliation(s)
- Andressa Ardiani
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Connie J. Rogers
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Gene delivery to dendritic cells by orally administered recombinant Saccharomyces cerevisiae in mice. Vaccine 2013. [DOI: 10.1016/j.vaccine.2012.11.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Nguyen NL, Kim JM, Park JA, Park SM, Jang YS, Yang MS, Kim DH. Expression and purification of an immunogenic dengue virus epitope using a synthetic consensus sequence of envelope domain III and Saccharomyces cerevisiae. Protein Expr Purif 2013; 88:235-42. [PMID: 23376461 DOI: 10.1016/j.pep.2013.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 01/06/2013] [Accepted: 01/20/2013] [Indexed: 12/24/2022]
Abstract
A synthetic consensus gene was designed based on residues of the amino acid sequences of dengue envelope domain III (scEDIII) from all four serotypes, and codon optimization for expression was conducted using baker's yeast, Saccharomyces cerevisiae. The synthetic gene was cloned into a yeast episomal expression vector, pYEGPD-TER, which was designed to direct cloned gene expression using the glyceraldehyde-3-phosphate dehydrogenase (GPD) promoter, a functional signal peptide of the amylase 1A protein from rice, and the GAL7 terminator. PCR and back-transformation into Escherichia coli confirmed the presence of the scEDIII gene-containing plasmid in the transformants. Northern blot analysis showed the presence of the scEDIII-specific transcript. Western blot analysis indicated that expressed scEDIII, with mobility similar to purified EDIII from E. coli, was successfully secreted into the culture media. Quantitative ELISA revealed that the recombinant scEDIII comprised approximately 0.1-0.6% of cell-free extract. In addition, 0.1-0.6 mg of scEDIII protein per liter of culture filtrate was detected on day 1 and peaked on day 3 after cultivation. The secreted scEDIII protein can be purified to ≥90% purity with 85% recovery using a simple ion-exchange FPLC followed by molecular weight cut-off. Upon administration of the purified protein to mice, mouse sera contained antibodies that were specific to all four serotypes of dengue virus. Moreover, a balanced immune response against all four serotypes was observed, suggesting that it may be possible to develop an effective tetravalent dengue vaccine using S. cerevisiae.
Collapse
Affiliation(s)
- Ngoc-Luong Nguyen
- Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
Hepatitis C vaccines. Vaccines (Basel) 2013. [DOI: 10.1016/b978-1-4557-0090-5.00051-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
IL-6-inducing whole yeast-based immunotherapy directly controls IL-12-dependent CD8 T-cell responses. J Immunother 2012; 35:14-22. [PMID: 22130158 DOI: 10.1097/cji.0b013e3182356888] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In current clinical trails, whole yeast-based immunotherapy expressing hepatitis C viral antigens demonstrated statistically significant improvement in end of treatment responses when combined with type I interferon based standard of care, even in standard of care resistant patients. Although preclinical data suggest yeast vaccination, such as type I interferon, facilitates CD8 T-cell immunity, the capacity of yeast to generate immunity in patients resistant to type I interferon calls into question the mechanism(s) underpinning the efficacy of this approach. We show yeast and a Toll-like receptor exclusive agonist, Pam3Cys, differ in CD8 T-cell generation when combined with an agonistic CD40 antibody. Although both yeast and PamCys were largely Toll-like receptor dependent, the primary CD8 response generated by yeast was significantly less than Pam3Cys in wild-type hosts even in a CD4 T-cell-deficient setting. In addition, immunization of IL6 mice with yeast produced a 3-fold to 6-fold increased CD8 response while the Pam3Cys response was unaffected. The yeast but not Pam3Cys-driven CD8 response was inhibited in both wild-type and IL-6 hosts by blocking interleukin (IL)-12. In addition, IL6 mice had increased CD86 expression on their dendritic cells after yeast immunization also inhibited by IL-12 blockade. Collectively, our results indicate the CD8 T-cell response to yeast but not Pam3Cys is influenced by IL-6-mediated control of IL-12 critical for dendritic cell activation. To our knowledge this is the first demonstration that yeast directly influence IL-12-associated CD8 T-cell immunity providing an additional route whereby recombinant yeast may provide efficacy independent of type I interferon.
Collapse
|
39
|
Yeast-based protein delivery to mammalian phagocytic cells is increased by coexpression of bacterial listeriolysin. Microbes Infect 2011; 13:908-13. [DOI: 10.1016/j.micinf.2011.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 05/09/2011] [Accepted: 05/13/2011] [Indexed: 11/23/2022]
|
40
|
Bazan SB, Geginat G, Breinig T, Schmitt MJ, Breinig F. Uptake of various yeast genera by antigen-presenting cells and influence of subcellular antigen localization on the activation of ovalbumin-specific CD8 T lymphocytes. Vaccine 2011; 29:8165-73. [DOI: 10.1016/j.vaccine.2011.07.141] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 07/21/2011] [Accepted: 07/25/2011] [Indexed: 02/08/2023]
|
41
|
Liu M, Capilla J, Johansen ME, Alvarado D, Martinez M, Chen V, Clemons KV, Stevens DA. Saccharomyces as a vaccine against systemic aspergillosis: ‘the friend of man’ a friend again? J Med Microbiol 2011; 60:1423-1432. [DOI: 10.1099/jmm.0.033290-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Min Liu
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- California Institute for Medical Research, San Jose, CA, USA
| | - Javier Capilla
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- California Institute for Medical Research, San Jose, CA, USA
| | - Maria E. Johansen
- California Institute for Medical Research, San Jose, CA, USA
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Danielle Alvarado
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- California Institute for Medical Research, San Jose, CA, USA
| | - Marife Martinez
- California Institute for Medical Research, San Jose, CA, USA
| | - Vicky Chen
- California Institute for Medical Research, San Jose, CA, USA
| | - Karl V. Clemons
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- California Institute for Medical Research, San Jose, CA, USA
| | - David A. Stevens
- Stanford University, Stanford, CA, USA
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- California Institute for Medical Research, San Jose, CA, USA
| |
Collapse
|
42
|
Delivery of functional DNA and messenger RNA to mammalian phagocytic cells by recombinant yeast. Gene Ther 2011; 19:237-45. [DOI: 10.1038/gt.2011.121] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Dodson LF, Hawkins WG, Goedegebuure P. Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 2011; 3:517-37. [PMID: 21463193 DOI: 10.2217/imt.11.10] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death with an overall 5-year survival of less than 5%. As there is ample evidence that pancreatic adenocarcinomas elicit antitumor immune responses, identification of pancreatic cancer-associated antigens has spurred the development of vaccination-based strategies for treatment. While promising results have been observed in animal tumor models, most clinical studies have found only limited success. As most trials were performed in patients with advanced pancreatic cancer, the contribution of immune suppressor mechanisms should be taken into account. In this article, we detail recent work in tumor antigen vaccination and the recently identified mechanisms of immune suppression in pancreatic cancer. We offer our perspective on how to increase the clinical efficacy of vaccines for pancreatic cancer.
Collapse
Affiliation(s)
- Lindzy F Dodson
- Washington University School of Medicine, Department of Surgery, Saint Louis, MO 63110, USA.
| | | | | |
Collapse
|
44
|
Torresi J, Johnson D, Wedemeyer H. Progress in the development of preventive and therapeutic vaccines for hepatitis C virus. J Hepatol 2011; 54:1273-85. [PMID: 21236312 DOI: 10.1016/j.jhep.2010.09.040] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 08/27/2010] [Accepted: 09/07/2010] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a blood borne disease estimated to chronically infect 3% of the worlds' population causing significant morbidity and mortality. Current medical therapy is curative in approximately 50% of patients. While recent treatment advances of genotype 1 infection using directly acting antiviral agents (DAAs) are encouraging, there is still a need to develop vaccine strategies capable of preventing infection. Moreover, vaccines may also be used in future in combination with DAAs enabling interferon-free treatment regimens. Viral and host specific factors contribute to viral evasion and present important impediments to vaccine development. Both, innate and adaptive immune responses are of major importance for the control of HCV infection. However, HCV has evolved ways of evading the host's immune response in order to establish persistent infection. For example, HCV inhibits intracellular interferon signalling pathways, impairs the activation of dendritic cells, CD8(+) and CD4(+) T cell responses, induces a state of T-cell exhaustion and selects escape variants with mutations CD8(+) T cell epitopes. An effective vaccine will need to produce strong and broadly cross-reactive CD4(+), CD8(+) T cell and neutralising antibody (NAb) responses to be successful in preventing or clearing HCV. Vaccines in clinical trials now include recombinant proteins, synthetic peptides, virosome based vaccines, tarmogens, modified vaccinia Ankara based vaccines, and DNA based vaccines. Several preclinical vaccine strategies are also under development and include recombinant adenoviral vaccines, virus like particles, and synthetic peptide vaccines. This paper will review the vaccines strategies employed, their success to date and future directions of vaccine design.
Collapse
Affiliation(s)
- Joseph Torresi
- Austin Centre for Infection Research, Department of Infectious Diseases Austin Hospital, Heidelberg, Victoria 3084, Australia.
| | | | | |
Collapse
|
45
|
Exploitation of differential homeostatic proliferation of T-cell subsets following chemotherapy to enhance the efficacy of vaccine-mediated antitumor responses. Cancer Immunol Immunother 2011; 60:1227-42. [PMID: 21544650 DOI: 10.1007/s00262-011-1020-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 04/07/2011] [Indexed: 12/22/2022]
Abstract
The 5-year survival rate for stage IB-III non-small-cell lung cancer (NSCLC) remains 15%. Surgical resection followed by adjuvant chemotherapy with cisplatin and vinorelbine is one standard-of-care. We sought to determine in a preclinical model whether (a) the combination of cisplatin and vinorelbine could positively modulate components of the immune system independent of antitumor activity, and (b) there were synergistic effects of this drug combination and vaccine immunotherapy. We examined the effect of cisplatin/vinorelbine on gene expression, cell-surface phenotype, and CTL-mediated cytolysis of murine lung carcinoma cells in vitro; we also assessed the effects of cisplatin/vinorelbine on immune subsets and function of Tregs in vivo. Finally, we evaluated the potential synergy between chemotherapy and a recombinant yeast-CEA vaccine in a murine model transgenic for CEA with mice bearing lung tumors. These studies demonstrate that exposure of lung tumor cells to the platinum doublet cisplatin/vinorelbine modulates tumor cell phenotype and increases sensitivity to CTL-mediated cytolysis. These studies also demonstrate that cisplatin/vinorelbine (a) induces sub-myeloablative leucopenia that differentially modulates reconstitution of Treg versus effector T-cell subsets and (b) can be employed synergistically with vaccine, exploiting homeostatic peripheral expansion of T cells. Antitumor studies show for the first time that cisplatin/vinorelbine combined with vaccine increases the survival of mice with established NSCLC. These findings provide the rationale for the potential clinical benefit of the combined use of vaccine with cisplatin/vinorelbine chemotherapy regimens.
Collapse
|
46
|
Czepczyńska-Krężel H, Czerwinski M, Krężel A, Krop-Watorek A. Isolation of carcinoembryonic antigen N-terminal domains (N-A1) from soluble aggregates. Protein Expr Purif 2011; 78:78-85. [PMID: 21458574 DOI: 10.1016/j.pep.2011.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 03/24/2011] [Accepted: 03/25/2011] [Indexed: 11/16/2022]
Abstract
Carcinoembryonic antigen (CEA) was identified as a prominent tumor-associated antigen in human colorectal cancer and it is still intensively investigated. However, its physiological role remains unclear. The CEA molecule is composed of seven highly hydrophobic, immunoglobulin-like domains, six of which contain a single disulphide bridge. The production of recombinant protein containing Ig-like domains in bacterial expression systems often results in partial degradation or insolubility due to aggregation hampering the analysis of their native structure and function. Here, we present a new method of expression and purification of CEA N-terminal domains (N-A1) fused to MBP in Escherichia coli. In order to optimize the expression and purification of CEA N-A1 domains we evaluated bacteria cultivation conditions, the length of N-A1 domains, fusion systems (GST- and MBP-tag), IPTG concentrations and protein purification conditions. We have found that MBP-N-A1 fusion protein digested with TEV protease forms soluble aggregates composed of N-A1 domains and incompletely digested MBP-N-A1 fusion protein. Using 1.25 M guanidinium chloride (GdmCl) as a component of the elution buffer we were able to achieve an almost complete dissociation of the aggregates. The dissociation was monitored by circular dichroism and fluorescence measurements. The CD spectra and Ellman's assay suggest that the conformation of N-A1 domains and their disulphide bonds are correct.
Collapse
Affiliation(s)
- Hanna Czepczyńska-Krężel
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, PL-53-114 Wroclaw, Poland
| | | | | | | |
Collapse
|
47
|
Ardiani A, Higgins JP, Hodge JW. Vaccines based on whole recombinant Saccharomyces cerevisiae cells. FEMS Yeast Res 2011; 10:1060-9. [PMID: 20707820 DOI: 10.1111/j.1567-1364.2010.00665.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The ultimate goal of therapeutic vaccines is to activate and exploit the patient's own immune system to vigorously and dynamically seek and eradicate established malignant or virally infected cells. Therapeutic vaccines also offer the potential for preventing disease recurrence. Saccharomyces cerevisiae-based vaccines, where the yeast is engineered to express viral or tumor antigens, represent an ideal therapeutic approach due to their ability to stimulate tumor- or viral-specific CD4(+) and CD8(+) T-cell responses that are capable of reducing disease burden. This review describes preclinical and clinical studies supporting the development of S. cerevisiae-based therapeutic vaccines for the treatment of cancer and viral diseases, as well as multimodal strategies in which therapeutic vaccines are combined with cytotoxic drugs to achieve a greater clinical response.
Collapse
Affiliation(s)
- Andressa Ardiani
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
48
|
Liu M, Clemons KV, Bigos M, Medovarska I, Brummer E, Stevens DA. Immune responses induced by heat killed Saccharomyces cerevisiae: a vaccine against fungal infection. Vaccine 2011; 29:1745-53. [PMID: 21219976 DOI: 10.1016/j.vaccine.2010.12.119] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/04/2010] [Accepted: 12/23/2010] [Indexed: 01/30/2023]
Abstract
Heat-killed Saccharomyces cerevisiae (HKY) used as a vaccine protects mice against systemic aspergillosis and coccidioidomycosis. Little is known about the immune response induced by HKY vaccination, consequently our goal was to do an analysis of HKY-induced immune responses involved in protection. BALB/c mice were vaccinated subcutaneously 3 times with HKY, a protective reagent, and bronchoalveolar lavage fluid, spleen, lymph nodes, and serum collected 2-5 weeks later. Cultured spleen or lymph node cells were stimulated with HKY. Proliferation of HKY-stimulated spleen or lymph node cells was tested by Alamar Blue reduction and flow cytometry. Cytokines from lymphocyte supernatants and antibody to glycans in serum collected from HKY-vaccinated mice were measured by ELISA. The results show that HKY promoted spleen cell and lymph node cell proliferation from HKY-vaccinated mice but not from PBS-vaccinated control mice (all P<0.05). Cytokine measurement showed HKY significantly promoted IFNγ, IL-6 and IL-17A production by spleen cells and lymph node cells (all P<0.05 and P<0.01, respectively). Cytokine production by HKY-stimulated cells from PBS-vaccinated mice was lower than those from HKY-vaccinated (P<0.05). Cytokines in BAL from HKY-vaccinated were higher, 1.7-fold for IFNγ and 2.1-fold for TNFα, than in BAL from PBS-vaccinated. Flow cytometry of lymphocytes from HKY-vaccinated showed 52% of CD3(+) or 56% of CD8(+) cells exhibited cell division after stimulation with HKY, compared to non-stimulated controls (26 or 23%, respectively) or HKY-stimulated cells from PBS-vaccinated (31 or 34%). HKY also induced antibody against Saccharomyces glucan and mannan with titers 4- or 2-fold, respectively, above that in unvaccinated. Taken together, the results suggested that HKY vaccination induces significant and specific Th1 type cellular immune responses and antibodies to glucan and mannan.
Collapse
Affiliation(s)
- Min Liu
- California Institute for Medical Research, San Jose, CA, USA
| | | | | | | | | | | |
Collapse
|
49
|
Andrukhov O, Matejka M, Rausch-Fan X. Effect of cyclosporin A on proliferation and differentiation of human periodontal ligament cells. Acta Odontol Scand 2010; 68:329-34. [PMID: 20818914 DOI: 10.3109/00016357.2010.514717] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Cyclosporin A (CsA) is widely used to prevent rejection after organ transplantation. However, it also causes several side-effects, including gingival overgrowth and bone resorption. Cellular mechanisms underlying the effect of CsA on periodontal tissue remain unclear. In this study, we investigated the effect of CsA on the proliferation and expression of characteristic markers in periodontal ligament cells (PDLs). MATERIAL AND METHODS The proliferation and viability of PDLs were measured by direct cell counting and 3,4,5-dimethylthiazol-2-yl-2,5-diphenyl tetrazolium bromide assay, respectively. mRNA expression levels of the specific proteins alkaline phosphatase (ALP), osteocalcin (OC) and collagen type 1 (Coll-1) were quantified using real-time polymerase chain reaction. Finally, ALP activity of PDLs was investigated using a specific colorimetric assay. RESULTS We found that proliferation of PDLs was stimulated by 0.01–0.1 μg/ml CsA and unaffected by 1 μg/ml CsA. The viability of PDLs was increased by 0.1 μg/ml CsA and not affected by 0.01 μg/ml and 1 μg/ml CsA. Furthermore, the mRNA expression levels of ALP, OC and Coll-1 in PDLs were significantly increased upon stimulation with 0.1 μg/ml CsA for 24 h or by stimulation with 0.01 μg/ml CsA for 48 h. In contrast, significantly lower expression levels of all three proteins in PDLs were observed upon stimulation with 1 μg/ml CsA for 48 h. The ALP activity of PDLs exhibited a similar pattern of changes upon CsA stimulation. CONCLUSION Our data demonstrated that CsA may influence both the proliferation and differentiation of human PDLs, which may play an important role in the homeostasis of periodontal tissue.
Collapse
Affiliation(s)
- Oleh Andrukhov
- Department of Periodontology, Bernhard Gottlieb School of Dentistry, Medical University of Vienna, Vienna, Austria.
| | | | | |
Collapse
|
50
|
Whole recombinant Hansenula polymorpha expressing hepatitis B virus surface antigen (yeast-HBsAg) induces potent HBsAg-specific Th1 and Th2 immune responses. Vaccine 2009; 28:187-94. [DOI: 10.1016/j.vaccine.2009.09.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/07/2009] [Accepted: 09/22/2009] [Indexed: 11/23/2022]
|