1
|
Arboleda-García A, Alarcon-Ruiz I, Boada-Acosta L, Boada Y, Vignoni A, Jantus-Lewintre E. Advancements in synthetic biology-based bacterial cancer therapy: A modular design approach. Crit Rev Oncol Hematol 2023; 190:104088. [PMID: 37541537 DOI: 10.1016/j.critrevonc.2023.104088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
Synthetic biology aims to program living bacteria cells with artificial genetic circuits for user-defined functions, transforming them into powerful tools with numerous applications in various fields, including oncology. Cancer treatments have serious side effects on patients due to the systemic action of the drugs involved. To address this, new systems that provide localized antitumoral action while minimizing damage to healthy tissues are required. Bacteria, often considered pathogenic agents, have been used as cancer treatments since the early 20th century. Advances in genetic engineering, synthetic biology, microbiology, and oncology have improved bacterial therapies, making them safer and more effective. Here we propose six modules for a successful synthetic biology-based bacterial cancer therapy, the modules include Payload, Release, Tumor-targeting, Biocontainment, Memory, and Genetic Circuit Stability Module. These will ensure antitumor activity, safety for the environment and patient, prevent bacterial colonization, maintain cell stability, and prevent loss or defunctionalization of the genetic circuit.
Collapse
Affiliation(s)
- Andrés Arboleda-García
- Systems Biology and Biosystems Control Lab, Instituto de Automática e Informática Industrial, Universitat Politècnica de València, Spain
| | - Ivan Alarcon-Ruiz
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lissette Boada-Acosta
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain; TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación Investigación del Hospital General Universitario de Valencia, Valencia, Spain; Molecular Oncology Laboratory, Fundación Investigación del Hospital General Universitario de Valencia, Valencia, Spain
| | - Yadira Boada
- Systems Biology and Biosystems Control Lab, Instituto de Automática e Informática Industrial, Universitat Politècnica de València, Spain
| | - Alejandro Vignoni
- Systems Biology and Biosystems Control Lab, Instituto de Automática e Informática Industrial, Universitat Politècnica de València, Spain.
| | - Eloisa Jantus-Lewintre
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain; TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación Investigación del Hospital General Universitario de Valencia, Valencia, Spain; Molecular Oncology Laboratory, Fundación Investigación del Hospital General Universitario de Valencia, Valencia, Spain; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
2
|
Zhang H, Wang Y, Li M, Cao K, Qi Z, Zhu L, Zhang Z, Hou L. A self-guidance biological hybrid drug delivery system driven by anaerobes to inhibit the proliferation and metastasis of colon cancer. Asian J Pharm Sci 2022; 17:892-907. [PMID: 36600894 PMCID: PMC9800957 DOI: 10.1016/j.ajps.2022.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/17/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer is often accompanied by multiple organ metastasis. Anaerobic Bifidobacterium Infantis (BI) bacterial can selectively grow in hypoxic colorectal tumor microenvironment (TME), to own the natural advantage of preferentially colorectal tumor targeting. Herein, a self-guidance biological hybrid drug delivery system (BI-ES-FeAlg/DOX) based on BI was constructed to inhibit the proliferation and metastasis of colon cancer. Results demonstrated that BI-ES-FeAlg/DOX could overcome physical barriers to target and accumulate in colon tumor tissues. Then DOX was released to kill tumor cells along with the phase transition (solid to liquid) of FeAlg hydrogel, due to Fe3+ was reduced to Fe2+by intracellular GSH. Meanwhile, BI-ES selectively colonized into tumors and expressed endostatin (ES) protein to down-regulate VEGF and bFGF expression, exerting anti-angiogenic effect. Moreover, FeAlg catalyzed H2O2 in the local tumor to generate cytotoxic ·OH, further enhancing the antitumor effect. The pharmacodynamic result in AOM/DSS model proved that BI-ES-FeAlg/DOX had the best therapeutic effect, with the final V/V0 of 2.19 ± 0.57, which was significantly lower than the other groups. Meanwhile, on CT-26 tumor-bearing model, it also showed an outstanding anti-tumor effect with inhibition rate of 82.12% ± 3.08%. In addition, lung metastases decreased significantly in tumor metastasis model after BI-ES-FeAlg/DOX treatment.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450000, China,Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou 450000, China
| | - Yaping Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengting Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kexuan Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zijun Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ling Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450000, China,Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou 450000, China,Corresponding authors.
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450000, China,Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou 450000, China,Corresponding authors.
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450000, China,Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou 450000, China,Corresponding authors.
| |
Collapse
|
3
|
Khan FH, Bhat BA, Sheikh BA, Tariq L, Padmanabhan R, Verma JP, Shukla AC, Dowlati A, Abbas A. Microbiome dysbiosis and epigenetic modulations in lung cancer: From pathogenesis to therapy. Semin Cancer Biol 2022; 86:732-742. [PMID: 34273520 DOI: 10.1016/j.semcancer.2021.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/25/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023]
Abstract
The lung microbiome plays an essential role in maintaining healthy lung function, including host immune homeostasis. Lung microbial dysbiosis or disruption of the gut-lung axis can contribute to lung carcinogenesis by causing DNA damage, inducing genomic instability, or altering the host's susceptibility to carcinogenic insults. Thus far, most studies have reported the association of microbial composition in lung cancer. Mechanistic studies describing host-microbe interactions in promoting lung carcinogenesis are limited. Considering cancer as a multifaceted disease where epigenetic dysregulation plays a critical role, epigenetic modifying potentials of microbial metabolites and toxins and their roles in lung tumorigenesis are not well studied. The current review explains microbial dysbiosis and epigenetic aberrations in lung cancer and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Faizan Haider Khan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | | | - Lubna Tariq
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Roshan Padmanabhan
- Department of Medicine, Case Western Reserve University, and University Hospital, Cleveland, OH, 44106, USA
| | - Jay Prakash Verma
- Institute of Environment and Sustainable Development, Banaras Hindu University Varanasi, India
| | | | - Afshin Dowlati
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA; University Hospitals Seidman Cancer Center, Cleveland, OH, 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44116, USA
| | - Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44116, USA.
| |
Collapse
|
4
|
The Oncobiome in Gastroenteric and Genitourinary Cancers. Int J Mol Sci 2022; 23:ijms23179664. [PMID: 36077063 PMCID: PMC9456244 DOI: 10.3390/ijms23179664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Early evidence suggests a strong association of microorganisms with several human cancers, and great efforts have been made to understand the pathophysiology underlying microbial carcinogenesis. Bacterial dysbiosis causes epithelial barrier failure, immune dysregulation and/or genotoxicity and, consequently, creates a tumor-permissive microenvironment. The majority of the bacteria in our body reside in the gastrointestinal tract, known as gut microbiota, which represents a complex and delicate ecosystem. Gut microbes can reach the pancreas, stomach and colon via the bloodstream. Oral bacterial translocations can also occur. In the stomach, pancreas and colon, low microbial diversity is associated with cancer, in particular with a bad prognosis. The urogenital tract also harbors unique microbiota, distinct from the gut microbiota, which might have a role in the urinary and female/male reproductive cancers’ pathogenesis. In healthy women, the majority of bacteria reside in the vagina and cervix and unlike other mucosal sites, the vaginal microbiota exhibits low microbial diversity. Genital dysbiosis might have an active role in the development and/or progression of gynecological malignancies through mechanisms including modulation of oestrogen metabolism. Urinary dysbiosis may influence the pathogenesis of bladder cancer and prostate cancer in males. Modulation of the microbiome via pre, pro and postbiotics, fecal or vaginal microbiota transplantation and engineering bacteria might prove useful in improving cancer treatment response and quality of life. Elucidating the complex host-microbiome interactions will result in prevention and therapeutic efficacy interventions.
Collapse
|
5
|
Levit R, Cortes-Perez NG, de Moreno de Leblanc A, Loiseau J, Aucouturier A, Langella P, LeBlanc JG, Bermúdez-Humarán LG. Use of genetically modified lactic acid bacteria and bifidobacteria as live delivery vectors for human and animal health. Gut Microbes 2022; 14:2110821. [PMID: 35960855 PMCID: PMC9377234 DOI: 10.1080/19490976.2022.2110821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
There is now strong evidence to support the interest in using lactic acid bacteria (LAB)in particular, strains of lactococci and lactobacilli, as well as bifidobacteria, for the development of new live vectors for human and animal health purposes. LAB are Gram-positive bacteria that have been used for millennia in the production of fermented foods. In addition, numerous studies have shown that genetically modified LAB and bifodobacteria can induce a systemic and mucosal immune response against certain antigens when administered mucosally. They are therefore good candidates for the development of new mucosal delivery strategies and are attractive alternatives to vaccines based on attenuated pathogenic bacteria whose use presents health risks. This article reviews the most recent research and advances in the use of LAB and bifidobacteria as live delivery vectors for human and animal health.
Collapse
Affiliation(s)
- Romina Levit
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Naima G. Cortes-Perez
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 0496, 78350 Jouy-en-Josas, France
| | - Alejandra de Moreno de Leblanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Jade Loiseau
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Anne Aucouturier
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Jean Guy LeBlanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Luis G. Bermúdez-Humarán
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France,CONTACT Luis G. Bermúdez-Humarán Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| |
Collapse
|
6
|
Bi Z, Cui E, Yao Y, Chang X, Wang X, Zhang Y, Xu GX, Zhuang H, Hua ZC. Recombinant Bifidobacterium longum Carrying Endostatin Protein Alleviates Dextran Sodium Sulfate-Induced Colitis and Colon Cancer in Rats. Front Microbiol 2022; 13:927277. [PMID: 35847065 PMCID: PMC9280188 DOI: 10.3389/fmicb.2022.927277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
Bifidobacterium has been widely administrated orally as probiotics to prevent pathogen colonization and modulate the gut microbiome balance. Endostatin is an endogenous inhibitor of angiogenesis and has been shown to inhibit tumor growth, invasion, and metastasis. At present, the combination of endostatin and chemotherapeutic drugs has been regarded as a promising antitumor treatment strategy. In this study, we selected a safe strain of Bifidobacterium longum as a delivery system to transport endostatin to the gastrointestinal tract and explored their combined effect on inflammatory bowel disease (IBD) and colitis-associated cancer. The results indicated that B. longum-Endo relieved dextran sulfate sodium-induced body weight loss, diarrhea, colon shortening, and epithelium damage. Long-term oral administration of B. longum-Endo significantly decreased tumor formation rate, tumor number, and tumor size. Moreover, the effect of B. longum-Endo on gut microbiota dysbiosis was also confirmed by 16S rRNA sequencing analysis. The levels of potentially beneficial bacteria, such as Lactobacillus, Bifidobacterium, Allobaculum, and Parabateroides, were increased in the B. longum-Endo group compared to the model and B. longum groups. Meanwhile, levels of potentially pathogenic bacteria including Desulfovibrio, Helicobacter, and Enterorhabdus were decreased. Taken together, these results suggested that oral administration of recombinant B. longum-Endo strain may be a promising therapeutic strategy for IBD and colitis-associated cancer.
Collapse
Affiliation(s)
- Zhiqian Bi
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Enqing Cui
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Yingying Yao
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Xiaoyao Chang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Xiaoyang Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Yuhui Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Gen-Xing Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
- *Correspondence: Gen-Xing Xu,
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
- Hongqin Zhuang,
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
- Changzhou High-Tech Research Institute of Nanjing University, Changzhou, China
- Jiangsu Target Pharma Laboratories Inc., Changzhou, China
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- Zi-Chun Hua,
| |
Collapse
|
7
|
Emerging applications of bacteria as antitumor agents. Semin Cancer Biol 2021; 86:1014-1025. [PMID: 33989734 DOI: 10.1016/j.semcancer.2021.05.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Bacteria are associated with the human body and colonize the gut, skin, and mucous membranes. These associations can be either symbiotic or pathogenic. In either case, bacteria derive more benefit from their host. The ability of bacteria to enter and survive within the human body can be exploited for human benefit. They can be used as a vehicle for delivering or producing bioactive molecules, such as toxins and lytic enzymes, and eventually for killing tumor cells. Clostridium and Salmonella have been shown to infect and survive within the human body, including in tumors. There is a need to develop genetic circuits, which enable bacterial cells to carry out the following activities: (i) escape the human immune system, (ii) invade tumors, (iii) multiply within the tumorous cells, (iv) produce toxins via quorum sensing at low cell densities, and (v) express suicide genes to undergo cell death or cell lysis after the tumor has been lysed. Thus, bacteria have the potential to be exploited as anticancer agents.
Collapse
|
8
|
Zuo F, Marcotte H. Advancing mechanistic understanding and bioengineering of probiotic lactobacilli and bifidobacteria by genome editing. Curr Opin Biotechnol 2021; 70:75-82. [PMID: 33445135 DOI: 10.1016/j.copbio.2020.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/02/2020] [Accepted: 12/17/2020] [Indexed: 12/01/2022]
Abstract
Typical traditional probiotics lactobacilli and bifidobacteria are gaining great interest to be developed as living diagnostics and therapeutics for improving human health. However, the mechanistic basis underlying their inherent health beneficial property remain incompletely understood which can slow down the translational pipeline in the functional food and pharmaceutical field. Efficient genome editing will advance the understanding of the molecular mechanism of the probiotics' physiological properties and their interaction with the host and the host microbiota, thereby further promote the development of next-generation designer probiotics with improved robustness and tailored functionalities. With the expansion of genome editing strategies such as CRISPR-Cas-based tools and IPSD assisted genome engineering as well as other synthetic biology technologies, the research and application of these health-promoting bacteria for the food and pharmaceutical industry will be further enhanced.
Collapse
Affiliation(s)
- Fanglei Zuo
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden; Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm SE-106 91, Sweden.
| | - Harold Marcotte
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| |
Collapse
|
9
|
Zou Y, Chen T. Engineered Akkermansia muciniphila: A promising agent against diseases (Review). Exp Ther Med 2020; 20:285. [PMID: 33209129 PMCID: PMC7668130 DOI: 10.3892/etm.2020.9415] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/15/2020] [Indexed: 12/23/2022] Open
Abstract
Achieving a harmonious gut microbial ecosystem has been hypothesized to be a successful method for alleviating metabolic disorders. The administration of probiotics, such as Lactobacillus and Bifidobacteria, is a known traditional and safe pathway to regulate human commensal microbes. With advancements in genetic sequencing and genetic editing tools, more bacteria are able to function as engineered probiotics with multiple therapeutic properties. As one of the next-generation probiotic candidates, Akkermansia muciniphila (A. muciniphila) has been discovered to enhance the gut barrier function and moderate inflammatory responses, exhibit improved effects with pasteurization and display beneficial probiotic effects in individuals with obesity, type 2 diabetes, atherosclerosis and autism-related gastrointestinal disturbances. In view of this knowledge, the present review aimed to summarize the effects of A. muciniphila in the treatment of metabolic disorders and to discuss several mature recombination systems for the genetic modification of A. muciniphila. From gaining an enhanced understanding of its genetic background, ingested A. muciniphila is expected to be used in various applications, including as a diagnostic tool, and in the site-specific delivery of therapeutic drugs.
Collapse
Affiliation(s)
- Yixuan Zou
- Institute of Translational Medicine, National Engineering Research Center for Bioengineering Drugs and Technologies, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Tingtao Chen
- Institute of Translational Medicine, National Engineering Research Center for Bioengineering Drugs and Technologies, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| |
Collapse
|
10
|
Anti-tumor effect of a recombinant Bifidobacterium strain secreting a claudin-targeting molecule in a mouse breast cancer model. Eur J Pharmacol 2020; 887:173596. [PMID: 32979353 DOI: 10.1016/j.ejphar.2020.173596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/02/2023]
Abstract
Bifidobacterium is a nonpathogenic strain of anaerobic bacteria that selectively localizes and proliferates in tumors. It has emerged as a specific carrier of anticancer proteins against malignant tumors. Claudins are tetraspanin transmembrane proteins that form tight junctions. Claudin-4 is overexpressed in certain epithelial malignant cancers. The C-terminal fragment of the Clostridium perfringens enterotoxin (C-CPE), an exotoxin without the cytotoxic domain, strongly binds to claudin-4. The C-CPE fusion toxin (C-CPE-PE23), which targets claudin-4, strongly suppresses tumor growth; however, C-CPE fusion toxins exhibit hepatic toxicity. In this study, we successfully generated a strain of Bifidobacterium longum that secreted C-CPE-PE23 (B. longum-C-CPE-PE23) and was specific to and cross reactive with human and mouse claudin-4. We evaluated the therapeutic potential of this strain against triple-negative breast cancer using a mouse model. C-CPE-PE23 decreased cell viability in a dose-dependent manner in human and mouse breast cancer cell lines. After intravenous injection, Bifidobacterium was specifically distributed in the tumors of mice bearing breast cancer tumors. Moreover, B. longum-C-CPE-PE23 significantly suppressed tumor growth in mice with breast cancer without serious side effects, such as weight loss or hepatic and renal damage. We suggest that B. longum-C-CPE-PE23 is a good candidate for breast cancer treatment. Bifidobacterium could also be used as a drug delivery system for hepatotoxic agents.
Collapse
|
11
|
Yang R, Wang D, Chu T. Synthesis and bioevaluation of radioiodinated nitroimidazole hypoxia imaging agents by one-pot click reaction. Bioorg Med Chem Lett 2020; 30:127386. [PMID: 32738994 DOI: 10.1016/j.bmcl.2020.127386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 02/02/2023]
Abstract
Eight radioiodinated 2-nitroimidazole derivatives for use as hypoxia imaging agents were synthesized by one-pot click reaction using four azides, two alkynes, and [131I]iodide ions and evaluated by hypoxic cellular uptake and biodistribution experiments. The results suggested that radiotracers with suitable partition coefficients (log P: -0.2-1.2) were more likely to have higher hypoxic cellular uptake. Among these eight molecules, [131I]15 ([131I]-(5-iodo-1-(2-(2-(2-nitro-1H-imidazol-1-yl)ethoxy)ethyl)-4-((2-nitro-1H-imidazol-1-yl)methyl)-1H-1,2,3-triazole)) had a suitable log P (0.05 ± 0.03) and contained two 2-nitroimidazole groups. The hypoxic/aerobic cellular uptake ratio of [131I]15 was 4.4 ± 0.5, and the tumor/blood (T/B) and tumor/muscle (T/M) ratios were 2.03 ± 0.45 and 6.82 ± 1.70, respectively. These results suggested that [131I]15 was a potential hypoxia imaging agent.
Collapse
Affiliation(s)
- Renyu Yang
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Damin Wang
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Taiwei Chu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
12
|
Shanmugaraj B, Priya LB, Mahalakshmi B, Subbiah S, Hu RM, Velmurugan BK, Baskaran R. Bacterial and viral vectors as vaccine delivery vehicles for breast cancer therapy. Life Sci 2020; 250:117550. [PMID: 32179071 DOI: 10.1016/j.lfs.2020.117550] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/06/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
|
13
|
Ngo N, Choucair K, Creeden JF, Qaqish H, Bhavsar K, Murphy C, Lian K, Albrethsen MT, Stanbery L, Phinney RC, Brunicardi FC, Dworkin L, Nemunaitis J. Bifidobacterium spp: the promising Trojan Horse in the era of precision oncology. Future Oncol 2019; 15:3861-3876. [PMID: 31668087 DOI: 10.2217/fon-2019-0374] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Selective delivery of therapeutic agents into solid tumors has been a major challenge impeding the achievement of long-term disease remission and cure. The need to develop alternative drug delivery routes to achieve higher drug concentration in tumor tissue, reduce unwanted off-target side effects and thus achieve greater therapeutic efficacy, has resulted in an explosive body of research. Bifidobacterium spp. are anaerobic, nonpathogenic, Gram-positive bacteria, commensal to the human gut that are a possible anticancer drug-delivery vehicle. In this review, we describe Bifidobacterium's microbiology, current clinical applications, overview of the preclinical work investigating Bifidobacterium's potential to deliver anticancer therapy, and review the different strategies used up to date. Finally, we discuss both current challenges and future prospects.
Collapse
Affiliation(s)
- Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Khalil Choucair
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Justin F Creeden
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Hanan Qaqish
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Krupa Bhavsar
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Chantal Murphy
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Kendra Lian
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Mary T Albrethsen
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Laura Stanbery
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | | | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
- ProMedica Health System, Toledo, OH 43606, USA
| |
Collapse
|
14
|
Zhang Y, Fang Z, Li R, Huang X, Liu Q. Design of Outer Membrane Vesicles as Cancer Vaccines: A New Toolkit for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11091314. [PMID: 31500086 PMCID: PMC6769604 DOI: 10.3390/cancers11091314] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/27/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer vaccines have been extensively studied in recent years and have contributed to exceptional achievements in cancer treatment. They are some of the most newly developed vaccines, although only two are currently approved for use, Provenge and Talimogene laherparepvec (T-VEC). Despite the approval of these two vaccines, most vaccines have been terminated at the clinical trial stage, which indicates that although they are effective in theory, concerns still exist, including low antigenicity of targeting antigens and tumor heterogeneity. In recent years, with new understanding of the biological function and vaccine potential of outer membrane vesicles (OMVs), their potential application in cancer vaccine design deserves our attention. Therefore, this review focuses on the mechanisms, advantages, and prospects of OMVs as antigen-carrier vaccines in cancer vaccine development. We believe that OMV-based vaccines present a safe and effective cancer therapeutic option with broad application prospects.
Collapse
Affiliation(s)
- Yingxuan Zhang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Zheyan Fang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Ruizhen Li
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China
- Key Laboratory of Tumor Pathogenesis and Molecular Pathology, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China.
- Key Laboratory of Tumor Pathogenesis and Molecular Pathology, School of Medicine, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
15
|
Abstract
Humans are a colonized with trillions of commensal microorganisms which exert a profound effect on normal host physiology and immune function through an abundance of genetic and metabolic by-products. Although the commensal microbiome has beneficial functions to host physiology, perturbations of the composition of the commensal microbiome or the homeostatic mucosal environment can lead to the induction of immune pathology and systemic inflammation. In the context of cancer progression or response to immune therapy, this inflammation can be detrimental, resulting in tumor growth and the promotion of immune suppression. On the other hand, significant associations have been identified whereby certain commensal microorganisms are able to enhance T cell function or are required for tumor control in cancer patients treated with certain immune therapies and chemotherapies. The focus of this chapter is to highlight the role of the commensal microbiome during tumor progression and in response to immune therapies.
Collapse
|
16
|
Song S, Vuai MS, Zhong M. The role of bacteria in cancer therapy - enemies in the past, but allies at present. Infect Agent Cancer 2018; 13:9. [PMID: 29568324 PMCID: PMC5856380 DOI: 10.1186/s13027-018-0180-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/15/2018] [Indexed: 12/20/2022] Open
Abstract
In recent decades, bacteria’s therapeutic role has aroused attention in medicinal and pharmaceutical research. While bacteria are considered among the primary agents for causing cancer, recent research has shown intriguing results suggesting that bacteria can be effective agents for cancer treatment – they are the perfect vessels for targeted cancer therapy. Several bacterial strains/species have been discovered to possess inherent oncolytic potentials to invade and colonize solid tumors in vivo. The therapeutic strategy of using bacteria for treating cancer is considered to be effective; however, the severe side effects encountered during the treatment resulted in the abandonment of the therapy. State-of-the-art genetic engineering has been recently applied to bacteria therapy and resulted in a greater efficacy with minimum side effects. In addition, the anti-cancer potential of tumor-targeting bacteria through oral administration circumvents the use of the intravenous route and the associated adverse effects. This review aims to provide a comprehensive summary of the latest literature on the role of bacteria in cancer treatment.
Collapse
Affiliation(s)
- Shiyu Song
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China
| | - Miza S Vuai
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China.,2Department of Natural Science, State University of Zanzibar (SUZA), P.O Box 146, Zanzibar, Tanzania
| | - Mintao Zhong
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China
| |
Collapse
|
17
|
Takano S, Ishikawa E, Matsuda M, Sakamoto N, Akutsu H, Yamamoto T, Matsumura A. The anti-angiogenic role of soluble-form VEGF receptor in malignant gliomas. Int J Oncol 2016; 50:515-524. [PMID: 28000842 DOI: 10.3892/ijo.2016.3810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/21/2016] [Indexed: 11/05/2022] Open
|
18
|
Shi L, Yu B, Cai CH, Huang JD. Angiogenic inhibitors delivered by the type III secretion system of tumor-targeting Salmonella typhimurium safely shrink tumors in mice. AMB Express 2016; 6:56. [PMID: 27558018 PMCID: PMC4996802 DOI: 10.1186/s13568-016-0226-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/10/2016] [Indexed: 12/19/2022] Open
Abstract
Despite of a growing number of bacterial species that apparently exhibit intrinsic tumor-targeting properties, no bacterium is able to inhibit tumor growth completely in the immunocompetent hosts, due to its poor dissemination inside the tumors. Oxygen and inflammatory reaction form two barriers and restrain the spread of the bacteria inside the tumors. Here, we engineered a Salmonella typhimurium strain named ST8 which is safe and has limited ability to spread beyond the anaerobic regions of tumors. When injected systemically to tumor-bearing immunocompetent mice, ST8 accumulated in tumors at levels at least 100-fold greater than parental obligate anaerobic strain ST4. ST8/pSEndo harboring therapeutic plasmids encoding Endostatin fused with a secreted protein SopA could target vasculature at the tumor periphery, can stably maintain and safely deliver a therapeutic vector, release angiogenic inhibitors through a type III secretion system (T3SS) to interfere with the pro-angiogenic action of growth factors in tumors. Mice with murine CT26 colon cancer that had been injected with ST8/pSEndo showed efficient tumor suppression by inducing more severe necrosis and inhibiting blooding vessel density within tumors. Our findings provide a therapeutic platform for indirectly acting therapeutic strategies such as anti-angiogenesis and immune therapy.
Collapse
|
19
|
Nguyen VH, Min JJ. Salmonella-Mediated Cancer Therapy: Roles and Potential. Nucl Med Mol Imaging 2016; 51:118-126. [PMID: 28559936 DOI: 10.1007/s13139-016-0415-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/29/2016] [Accepted: 03/28/2016] [Indexed: 01/21/2023] Open
Abstract
The use of bacteria for cancer therapy, which was proposed many years ago, was not recognized as a potential therapeutic strategy until recently. Technological advances and updated knowledge have enabled the genetic engineering of bacteria for their safe and effective application in the treatment of cancer. The efficacy of radiotherapy depends mainly on tissue oxygen levels, and low oxygen concentrations in necrotic and hypoxic regions are a common cause of treatment failure. In addition, the distribution of a drug is important for the therapeutic effect of chemotherapy, and the poor vasculature in tumors impairs drug delivery, limiting the efficacy of a drug, especially in necrotic and hypoxic regions. Bacteria-mediated cancer therapy (BMCT) relies on facultative anaerobes that can survive in well or poorly oxygenated regions, and it therefore improves the therapeutic efficacy drug distribution throughout the tumor mass. Since the mid-1990s, the number of published bacterial therapy papers has increased rapidly, with a doubling time of 2.5 years in which the use of Salmonella increased significantly. BMCT is being reevaluated to overcome some of the drawbacks of conventional therapies. This review focuses on Salmonella-mediated cancer therapy as the most widely used type of BMCT.2.
Collapse
Affiliation(s)
- Vu Hong Nguyen
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California, 1500 East Duarte Road, Duarte, CA 91010 USA
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School, 5 Hak1 dong, Dong-gu, Gwangju, 501-746 Republic of Korea
| |
Collapse
|
20
|
Osswald A, Sun Z, Grimm V, Ampem G, Riegel K, Westendorf AM, Sommergruber W, Otte K, Dürre P, Riedel CU. Three-dimensional tumor spheroids for in vitro analysis of bacteria as gene delivery vectors in tumor therapy. Microb Cell Fact 2015; 14:199. [PMID: 26655167 PMCID: PMC4676896 DOI: 10.1186/s12934-015-0383-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022] Open
Abstract
Background Several studies in animal models demonstrated that obligate and facultative anaerobic bacteria of the genera Bifidobacterium, Salmonella, or Clostridium specifically colonize solid tumors. Consequently, these and other bacteria are discussed as live vectors to deliver therapeutic genes to inhibit tumor growth. Therapeutic approaches for cancer treatment using anaerobic bacteria have been investigated in different mouse models. In the present study, solid three-dimensional (3D) multicellular tumor spheroids (MCTS) of the colorectal adenocarcinoma cell line HT-29 were generated and tested for their potential to study prodrug-converting enzyme therapies using bacterial vectors in vitro. Results HT-29 MCTS resembled solid tumors displaying all relevant features with an outer zone of proliferating cells and hypoxic and apoptotic regions in the core. Upon incubation with HT-29 MCTS, Bifidobacterium bifidum S17 and Salmonella typhimurium YB1 selectively localized, survived and replicated in hypoxic areas inside MCTS. Furthermore, spores of the obligate anaerobe Clostridium sporogenes germinated in these hypoxic areas. To further evaluate the potential of MCTS to investigate therapeutic approaches using bacteria as gene delivery vectors, recombinant bifidobacteria expressing prodrug-converting enzymes were used. Expression of a secreted cytosine deaminase in combination with 5-fluorocytosine had no effect on growth of MCTS due to an intrinsic resistance of HT-29 cells to 5-fluorouracil, i.e. the converted drug. However, a combination of the prodrug CB1954 and a strain expressing a secreted chromate reductase effectively inhibited MCTS growth. Conclusions Collectively, the presented results indicate that MCTS are a suitable and reliable model to investigate live bacteria as gene delivery vectors for cancer therapy in vitro. Electronic supplementary material The online version of this article (doi:10.1186/s12934-015-0383-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Osswald
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Zhongke Sun
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,College of Life Sciences and Agriculture, Zhoukou Normal University, Chuanhui District, Zhoukou, 466001, People's Republic of China.
| | - Verena Grimm
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Grace Ampem
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Karin Riegel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Wolfgang Sommergruber
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400, Biberach, Germany.
| | - Peter Dürre
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Christian U Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| |
Collapse
|
21
|
Lin IYC, Van TTH, Smooker PM. Live-Attenuated Bacterial Vectors: Tools for Vaccine and Therapeutic Agent Delivery. Vaccines (Basel) 2015; 3:940-72. [PMID: 26569321 PMCID: PMC4693226 DOI: 10.3390/vaccines3040940] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022] Open
Abstract
Genetically attenuated microorganisms, including pathogenic and commensal bacteria, can be engineered to carry and deliver heterologous antigens to elicit host immunity against both the vector as well as the pathogen from which the donor gene is derived. These live attenuated bacterial vectors have been given much attention due to their capacity to induce a broad range of immune responses including localized mucosal, as well as systemic humoral and/or cell-mediated immunity. In addition, the unique tumor-homing characteristics of these bacterial vectors has also been exploited for alternative anti-tumor vaccines and therapies. In such approach, tumor-associated antigen, immunostimulatory molecules, anti-tumor drugs, or nucleotides (DNA or RNA) are delivered. Different potential vectors are appropriate for specific applications, depending on their pathogenic routes. In this review, we survey and summarize the main features of the different types of live bacterial vectors and discussed the clinical applications in the field of vaccinology. In addition, different approaches for using live attenuated bacterial vectors for anti-cancer therapy is discussed, and some promising pre-clinical and clinical studies in this field are outlined.
Collapse
Affiliation(s)
- Ivan Y C Lin
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Thi Thu Hao Van
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Peter M Smooker
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| |
Collapse
|
22
|
A Phytase-Based Reporter System for Identification of Functional Secretion Signals in Bifidobacteria. PLoS One 2015; 10:e0128802. [PMID: 26086721 PMCID: PMC4472781 DOI: 10.1371/journal.pone.0128802] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 04/30/2015] [Indexed: 11/19/2022] Open
Abstract
Health-promoting effects have been attributed to a number of Bifidobacterium sp. strains. These effects as well as the ability to colonise the host depend on secreted proteins. Moreover, rational design of protein secretion systems bears the potential for the generation of novel probiotic bifidobacteria with improved health-promoting or therapeutic properties. To date, there is only very limited data on secretion signals of bifidobacteria available. Using in silico analysis, we demonstrate that all bifidobacteria encode the major components of Sec-dependent secretion machineries but only B. longum strains harbour Tat protein translocation systems. A reporter plasmid for secretion signals in bifidobacteria was established by fusing the coding sequence of the signal peptide of a sialidase of Bifidobacterium bifidum S17 to the phytase gene appA of E. coli. The recombinant strain showed increased phytase activity in spent culture supernatants and reduced phytase levels in crude extracts compared to the control indicating efficient phytase secretion. The reporter plasmid was used to screen seven predicted signal peptides in B. bifidum S17 and B. longum E18. The tested signal peptides differed substantially in their efficacy to mediate protein secretion in different host strains. An efficient signal peptide was used for expression and secretion of a therapeutically relevant protein in B. bifidum S17. Expression of a secreted cytosine deaminase led to a 100-fold reduced sensitivity of B. bifidum S17 to 5-fluorocytosine compared to the non-secreted cytosine deaminase suggesting efficient conversion of 5-fluorocytosine to the cytotoxic cancer drug 5-fluorouracil by cytosine deaminase occurred outside the bacterial cell. Selection of appropriate signal peptides for defined protein secretion might improve therapeutic efficacy as well as probiotic properties of bifidobacteria.
Collapse
|
23
|
Xiao X, Jin R, Li J, Bei Y, Wei T. The antitumor effect of suicide gene therapy using Bifidobacterium infantis-mediated herpes simplex virus thymidine kinase/ganciclovir in a nude mice model of renal cell carcinoma. Urology 2014; 84:982.e15-20. [PMID: 25123427 DOI: 10.1016/j.urology.2014.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/08/2014] [Accepted: 05/13/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To confirm the effectivity of Bifidobacterium infantis-mediated herpes simplex virus thymidine kinase/ganciclovir suicide gene system on the treatment of renal cell carcinoma in nude mice and further explore the mechanisms. MATERIALS AND METHODS A B infantis thymidine kinase (B infantis-TK) suicide gene system was constructed in our previous study. Tumor-bearing nude mice were randomized into 4 groups and injected with normal saline, B infantis, B infantis/pGEX-1λT, and B infantis-TK, respectively, via tail vein, followed by intraperitoneal injection of ganciclovir. The treatment effects were evaluated by the terminal deoxynucleotidyl transferase-mediated deoxynucleotide triphosphate nick end labeling assay, quantitative reverse transcriptase polymerase chain reaction, and Western blotting. Side effects were also recorded. RESULTS Compared with the other 3 treatments, the treatment with B infantis-TK resulted in a significant effective antitumor activity and stronger apoptotic response. Western blot analysis showed that the expression levels of Rel A and Bcl-xL were significantly lower, whereas those of caspase 3 and Bax were significantly higher in tumor tissues resected from group B infantis-TK, which were consistent with the quantitative reverse transcriptase-polymerase chain reaction results. CONCLUSION The B infantis-TK/ganciclovir therapy system exhibits an effective antitumor activity by promoting tumor cell apoptosis through both the intrinsic and the extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ren Jin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiang Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Bei
- Department of Urology, The Affiliated Hospital of Chengdu University, Chengdu, China
| | - Tang Wei
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
24
|
Zhang S, Chen Y, Wang J, Tang F, Miao T, Li M. Role of nontoxigenic Clostridium novyi in solid tumor therapy. REVIEWS IN MEDICAL MICROBIOLOGY 2014; 25:71-76. [DOI: 10.1097/mrm.0000000000000005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
25
|
Guglielmetti S, Mayo B, Álvarez-Martín P. Mobilome and genetic modification of bifidobacteria. Benef Microbes 2013; 4:143-66. [PMID: 23271067 DOI: 10.3920/bm2012.0031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Until recently, proper development of molecular studies in Bifidobacterium species has been hampered by growth difficulties, because of their exigent nutritive requirements, oxygen sensitivity and lack of efficient genetic tools. These studies, however, are critical to uncover the cross-talk between bifidobacteria and their hosts' cells and to prove unequivocally the supposed beneficial effects provided through the endogenous bifidobacterial populations or after ingestion as probiotics. The genome sequencing projects of different bifidobacterial strains have provided a wealth of genetic data that will be of much help in deciphering the molecular basis of the physiological properties of bifidobacteria. To this end, the purposeful development of stable cloning and expression vectors based on robust replicons - either from temperate phages or resident plasmids - is still needed. This review addresses the current knowledge on the mobile genetic elements of bifidobacteria (prophages, plasmids and transposons) and summarises the different types of vectors already available, together with the transformation procedures for introducing DNA into the cells. It also covers recent molecular studies performed with such vectors and incipient results on the genetic modification of these organisms, establishing the basis that would allow the use of bifidobacteria for future biotechnological applications.
Collapse
Affiliation(s)
- S Guglielmetti
- Dipartimento di Scienze e Tecnologie Alimentari e Microbiologiche, Sezione di Microbiologia Industriale, Università degli studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | | | | |
Collapse
|
26
|
Ruiz L, Motherway MO, Lanigan N, van Sinderen D. Transposon mutagenesis in Bifidobacterium breve: construction and characterization of a Tn5 transposon mutant library for Bifidobacterium breve UCC2003. PLoS One 2013; 8:e64699. [PMID: 23737995 PMCID: PMC3667832 DOI: 10.1371/journal.pone.0064699] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/17/2013] [Indexed: 01/20/2023] Open
Abstract
Bifidobacteria are claimed to contribute positively to human health through a range of beneficial or probiotic activities, including amelioration of gastrointestinal and metabolic disorders, and therefore this particular group of gastrointestinal commensals has enjoyed increasing industrial and scientific attention in recent years. However, the molecular mechanisms underlying these probiotic mechanisms are still largely unknown, mainly due to the fact that molecular tools for bifidobacteria are rather poorly developed, with many strains lacking genetic accessibility. In this work, we describe the generation of transposon insertion mutants in two bifidobacterial strains, B. breve UCC2003 and B. breve NCFB2258. We also report the creation of the first transposon mutant library in a bifidobacterial strain, employing B. breve UCC2003 and a Tn5-based transposome strategy. The library was found to be composed of clones containing single transposon insertions which appear to be randomly distributed along the genome. The usefulness of the library to perform phenotypic screenings was confirmed through identification and analysis of mutants defective in D-galactose, D-lactose or pullulan utilization abilities.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Microbiology and Alimentary Pharmabiotic Centre, National University of Ireland, Cork, Ireland
| | - Mary O’Connell Motherway
- Department of Microbiology and Alimentary Pharmabiotic Centre, National University of Ireland, Cork, Ireland
| | - Noreen Lanigan
- Department of Microbiology and Alimentary Pharmabiotic Centre, National University of Ireland, Cork, Ireland
| | - Douwe van Sinderen
- Department of Microbiology and Alimentary Pharmabiotic Centre, National University of Ireland, Cork, Ireland
- * E-mail:
| |
Collapse
|
27
|
Yin X, Yu B, Tang Z, He B, Ren J, Xiao X, Tang W. Bifidobacterium infantis-mediated HSV-TK/GCV suicide gene therapy induces both extrinsic and intrinsic apoptosis in a rat model of bladder cancer. Cancer Gene Ther 2012; 20:77-81. [PMID: 23258087 DOI: 10.1038/cgt.2012.86] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bladder cancer is the most common cancer in the urological tract. Here, we investigated the molecular pathways involved in the apoptosis of rodent bladder cancer, which was treated with Bifidobacterium infantis (BI)-mediated herpes simplex virus thymidine kinase (HSV-TK)/ganciclovir (GCV) suicide gene therapy. We engineered the BI-TK system, which consists of BI with the recombinant plasmid PGEX-1λT carrying HSV-TK (that is, TK) gene. Tumor-bearing rats were randomly divided into three groups and tail vein injected with normal saline (group A), BI/PGEX-1λT (group B) or BI-TK (group C), followed by intraperitoneal injection of GCV. The BI-TK/GCV system (group C) was shown to inhibit tumor growth and caused the apoptosis of tumor cells as assessed by in situ terminal transferase dUTP nick-end labeling assay. While varied extents of apoptosis were detected in tumor cells from all groups, tumor cells treated with BI-TK/GCV system (group C) exhibited the highest level of apoptosis (P < 0.05), consistent with our previous studies. Furthermore, we found that the expression levels of Fas, FasL, Cyt-C and caspase-9 in tumor tissues derived from group C were significantly higher than the other two groups (P < 0.001). Therefore, our results have demonstrated that the BI-TK/GCV therapy system exhibits a sustainable antitumor growth activity in the rodent model of bladder cancer. Mechanistically, both extrinsic and intrinsic apoptosis pathways are involved in the BI-TK/GCV antitumor functionality.
Collapse
Affiliation(s)
- X Yin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Mei L, Wang Y, Chu T. ⁹⁹mTc/Re complexes bearing bisnitroimidazole or mononitroimidazole as potential bioreductive markers for tumor: synthesis, physicochemical characterization and biological evaluation. Eur J Med Chem 2012; 58:50-63. [PMID: 23088932 DOI: 10.1016/j.ejmech.2012.09.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 10/27/2022]
Abstract
Four monoamine-monoamide dithiol (MAMA) ligands containing two or one nitroimidazole moieties were synthesized and labeled with (99m)Tc (labeling yield > 95%). The proposed structures of (99m)Tc-complexes are identified by comparison with analogous Re-MAMA complexes. (99m)Tc-MAMA complexes show better physicochemical characters than (99m)TcO-(PnAO-1-(2-nitroimidazole)). Reduction potentials of nitro groups of the rhenium complexes are within the range for bioreductive compounds. As expected, biodistribution studies demonstrate that the 2-nitroimidazole complex shows better tumor-to-tissue ratios than 4-nitroimidazole analog for mononitroimidazole complexes, but not for MAMA-bisnitroimidazoles due to higher lipophilicity. Both the bisnitroimidazole compounds show rapider excretion, lower background activity in liver and higher tumor-to-tissue ratios than the mononitroimidazoles. Better biodistribution characteristic makes both the MAMA-bisnitroimidazole complexes, especially (99m)Tc-15, be potential tumor hypoxia marker.
Collapse
Affiliation(s)
- Lei Mei
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, PR China
| | | | | |
Collapse
|
29
|
Accessing the inaccessible: molecular tools for bifidobacteria. Appl Environ Microbiol 2012; 78:5035-42. [PMID: 22582076 DOI: 10.1128/aem.00551-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bifidobacteria are an important group of the human intestinal microbiota that have been shown to exert a number of beneficial probiotic effects on the health status of their host. Due to these effects, bifidobacteria have attracted strong interest in health care and food industries for probiotic applications and several species are listed as so-called "generally recognized as safe" (GRAS) microorganisms. Moreover, recent studies have pointed out their potential as an alternative or supplementary strategy in tumor therapy or as live vaccines. In order to study the mechanisms by which these organisms exert their beneficial effects and to generate recombinant strains that can be used as drug delivery vectors or live vaccines, appropriate molecular tools are indispensable. This review provides an overview of the currently available methods and tools to generate recombinant strains of bifidobacteria. The currently used protocols for transformation of bifidobacteria, as well as replicons, selection markers, and determinants of expression, will be summarized. We will further discuss promoters, terminators, and localization signals that have been used for successful generation of expression vectors.
Collapse
|
30
|
Li ZJ, Zhu H, Ma BY, Zhao F, Mao SH, Liu TG, He JP, Deng LC, Yi C, Huang Y. Inhibitory effect of Bifidobacterium infantis-mediated sKDR prokaryotic expression system on angiogenesis and growth of Lewis lung cancer in mice. BMC Cancer 2012; 12:155. [PMID: 22536942 PMCID: PMC3404897 DOI: 10.1186/1471-2407-12-155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 04/26/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To construct the Bifidobacterium infantis-mediated soluble kinase insert domain receptor (sKDR) prokaryotic expression system and to observe its inhibitory effect on growth of human umbilicus vessel endothelial cells (HUVECs) in vitro and Lewis lung cancer (LLC) on mice in vivo. METHODS The Bifidobacterium infantis-mediated sKDR prokaryotic expression system was constructed through electroporation and subsequently identified through PCR and Western blot analysis. HUVECs were added to the products of this system to evaluate the anti-angiogenesis effect through MTT assay in vitro. The LLC mice models were divided into three groups: one group treated with saline (group a); one group treated with recombinant Bifidobacterium infantis containing pTRKH2-PsT plasmid group (group b); and one group treated with recombinant Bifidobacterium infantis containing pTRKH2-PsT/sKDR plasmid group (group c). The quality of life and survival of mice were recorded. Tumor volume, tumor weight, inhibitive rate, and necrosis rate of tumor were also evaluated. Necrosis of tumor and signals of blood flow in tumors were detected through color Doppler ultrasound. In addition, microvessel density (MVD) of the tumor tissues was assessed through CD31 immunohistochemical analysis. RESULTS The positively transformed Bifidobacterium infantis with recombinant pTRKH2-PsT/sKDR plasmid was established, and was able to express sKDR at gene and protein levels. The proliferation of HUVECs cultivated with the extract of positively transformed bacteria was inhibited significantly compared with other groups (P < 0. 05). The quality of life of mice in group c was better than in group a and b. The recombinant Bifidobacterium infantis containing pTRKH2-PsT/sKDR plasmid enhanced the efficacy of tumor growth suppression and prolongation of survival, increased the necrosis rate of tumor significantly, and could obviously decrease MVD and the signals of blood flow in tumors. CONCLUSION The Bifidobacterium infantis-mediated sKDR prokaryotic expression system was constructed successfully. This system could express sKDR at gene and protein levels and significantly inhibit the growth of HUVECs induced by VEGF in vitro. Moreover, it could inhibit tumor growth and safely prolong the survival time of LLC C57BL/6 mice.
Collapse
Affiliation(s)
- Zhao-Jun Li
- Department of Abdominal Cancer, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fukiya S, Hirayama Y, Sakanaka M, Kano Y, Yokota A. Technological advances in bifidobacterial molecular genetics: application to functional genomics and medical treatments. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2012; 31:15-25. [PMID: 24936345 PMCID: PMC4034290 DOI: 10.12938/bmfh.31.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/17/2012] [Indexed: 11/29/2022]
Abstract
Bifidobacteria are well known as beneficial intestinal bacteria that exert
health-promoting effects in humans. In addition to physiological and immunological
investigations, molecular genetic technologies have been developed and have recently
started to be applied to clarify the molecular bases of
host-Bifidobacterium interactions. These technologies include
transformation technologies and Escherichia coli-Bifidobacterium shuttle
vectors that enable heterologous gene expression. In this context, a plasmid artificial
modification method that protects the introduced plasmid from the restriction system in
host bifidobacteria has recently been developed to increase transformation efficiency. On
the other hand, targeted gene inactivation systems, which are vital for functional
genomics, seemed far from being practically applicable in bifidobacteria. However,
remarkable progress in this technology has recently been achieved, enabling functional
genomics in bifidobacteria. Integrated use of these molecular genetic technologies with
omics-based analyses will surely boost characterization of the molecular basis underlying
beneficial effects of bifidobacteria. Applications of recombinant bifidobacteria to
medical treatments have also progressed.
Collapse
Affiliation(s)
- Satoru Fukiya
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| | - Yosuke Hirayama
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| | - Mikiyasu Sakanaka
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| | - Yasunobu Kano
- Department of Molecular Genetics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Atsushi Yokota
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| |
Collapse
|
32
|
Yin Y, Kou L, Wang JJ, Xu GX. Therapeutic efficacy of Bifidobacterium longum-mediated human interleukin-2 with endostatin or TRAIL in transplanted tumors in mice. Exp Ther Med 2011; 3:481-486. [PMID: 22969915 DOI: 10.3892/etm.2011.421] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 11/28/2011] [Indexed: 11/06/2022] Open
Abstract
Interleukin-2 (IL-2), as an important cytokine in immune response, has been demonstrated to have therapeutic activity in several cancer models. In our previous study, we showed that the pBV22210 vector containing a chloramphenicol resistance gene and the cryptic plasmid, pMB1, from the Bifidobacterium longum (B. longum) strain could stably replicate and did not significantly affect the biological characteristics of B. longum. In this study, B. longum was transfected by electroporation with pBV22210 containing IL-2 (B. longum-pBV22210-IL-2), its growth curve was determined, and its inhibitory effect on tumor xenografts in mice was examined. The results showed that B. longum-pBV22210-IL-2 reduced the tumor size and prolonged the survival time of H22 tumor-bearing mice. In addition, when cyclophosphamide (CTX), B. longum-pBV22210-endostatin, or B. longum-pBV22210-TRAIL was combined with B. longum-pBV22210-IL-2, the antitumor effect was significantly enhanced. The survival times of the mice in the combination groups of B. longum-pBV22210-endostatin or B. longum-pBV22210-TRAIL were longer than those of the mice in the B. longum-pBV22210-IL-2 alone group. However, when CTX was added, the survival times of the mice showed no statistically significant difference compared with those of the mice in the dextrose-saline solution group. These results suggest that B. longum-pBV22210-IL-2 has potent antitumor effects that could be enhanced when combined with chemotherapeutic drugs or other antitumor genes.
Collapse
Affiliation(s)
- Yan Yin
- Department of Biological Science and Technology and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences
| | | | | | | |
Collapse
|
33
|
Yu Z, Huang Z, Shao C, Huang Y, Zhang F, Yang J, Deng L, Zeng Z, Deng Q, Zeng W. Oral administration of interferon-α2b-transformed Bifidobacterium longum protects BALB/c mice against coxsackievirus B3-induced myocarditis. Virol J 2011; 8:525. [PMID: 22151967 DOI: 10.1186/1743-422x-8-525] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 12/08/2011] [Indexed: 11/10/2022] Open
Abstract
Multiple reports have claimed that low-dose orally administered interferon (IFN)-α is beneficial in the treatment of many infectious diseases and provides a viable alternative to high-dose intramuscular treatment. However, research is needed on how to express IFN stably in the gut. Bifidobacterium may be a suitable carrier for human gene expression and secretion in the intestinal tract for the treatment of gastrointestinal diseases. We reported previously that Bifidobacterium longum can be used as a novel oral delivery of IFN-α. IFN-transformed B. longum can exert an immunostimulatory role in mice; however the answer to whether this recombinant B. longum can be used to treat virus infection still remains elusive. Here, we investigated the efficacy of IFN-transformed B. longum administered orally on coxsackie virus B3 (CVB3)-induced myocarditis in BALB/c mice. Our data indicated that oral administration of IFN-transformed B. longum for 2 weeks after virus infection reduced significantly the severity of virus-induced myocarditis, markedly down regulated virus titers in the heart, and induced a T helper 1 cell pattern in the spleen and heart compared with controls. Oral administration of the IFN-transformed B. longum, therefore, may play a potential role in the treatment of CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Zhijian Yu
- Department of Infectious Diseases, the Affiliated Shenzhen Nanshan Hospital of Guangdong Medical College, No 89 Taoyuan Road, Nanshan district, Shenzhen, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Baban CK, Cronin M, O'Hanlon D, O'Sullivan GC, Tangney M. Bacteria as vectors for gene therapy of cancer. Bioeng Bugs 2011; 1:385-94. [PMID: 21468205 DOI: 10.4161/bbug.1.6.13146] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/23/2010] [Accepted: 07/26/2010] [Indexed: 12/13/2022] Open
Abstract
Anti-cancer therapy faces major challenges, particularly in terms of specificity of treatment. The ideal therapy would eradicate tumor cells selectively with minimum side effects on normal tissue. Gene or cell therapies have emerged as realistic prospects for the treatment of cancer, and involve the delivery of genetic information to a tumor to facilitate the production of therapeutic proteins. However, there is still much to be done before an efficient and safe gene medicine is achieved, primarily developing the means of targeting genes to tumors safely and efficiently. An emerging family of vectors involves bacteria of various genera. It has been shown that bacteria are naturally capable of homing to tumors when systemically administered resulting in high levels of replication locally. Furthermore, invasive species can deliver heterologous genes intra-cellularly for tumor cell expression. Here, we review the use of bacteria as vehicles for gene therapy of cancer, detailing the mechanisms of action and successes at preclinical and clinical levels.
Collapse
Affiliation(s)
- Chwanrow K Baban
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
35
|
Zhu H, Li Z, Mao S, Ma B, Zhou S, Deng L, Liu T, Cui D, Zhao Y, He J, Yi C, Huang Y. Antitumor effect of sFlt-1 gene therapy system mediated by Bifidobacterium Infantis on Lewis lung cancer in mice. Cancer Gene Ther 2011; 18:884-96. [PMID: 21921942 PMCID: PMC3215997 DOI: 10.1038/cgt.2011.57] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Soluble fms-like tyrosine kinase receptor (sFlt-1) is a soluble form of extramembrane part of vascular endothelial growth factor receptor-1 (VEGFR-1) that has antitumor effects. Bifidobacterium Infantis is a kind of non-pathogenic and anaerobic bacteria that may have specific targeting property of hypoxic environment inside of solid tumors. The aim of this study was to construct Bifidobacterium Infantis-mediated sFlt-1 gene transferring system and investigate its antitumor effect on Lewis lung cancer (LLC) in mice. Our results demonstrated that the Bifidobacterium Infantis-mediated sFlt-1 gene transferring system was constructed successfully and the system could express sFlt-1 at the levels of gene and protein. This system could not only significantly inhibit growth of human umbilical vein endothelial cells induced by VEGF in vitro, but also inhibit the tumor growth and prolong survival time of LLC C57BL/6 mice safely. These data suggest that Bifidobacterium Infantis-mediated sFlt-1 gene transferring system presents a promising therapeutic approach for the treatment of cancer.
Collapse
Affiliation(s)
- H Zhu
- Department of Abdominal Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
When one considers the organism Salmonella enterica serotype Typhimurium (S. Typhimurium), one usually thinks of the Gram-negative enteric pathogen that causes the severe food borne illness, gastroentertitis. In this context, the idea of Salmonella being exploited as a cancer therapeutic seems pretty remote. However, there has been an escalating interest in the development of tumor-therapeutic bacteria for use in the treatment of a variety of cancers. This strategy takes advantage of the remarkable ability of certain bacteria to preferentially replicate and accumulate within tumors. In the case of S. Typhimurium, this organism infects and selectively grows within implanted tumors, achieving tumor/normal tissue ratios of approximately 1,000:1. Salmonella also has some attractive properties well suited for the design of a chemotherapeutic agent. In particular, this pathogen can easily be manipulated to carry foreign genes, and since this species is a facultative anaerobe, it is able to survival in both oxygenated and hypoxic conditions, implying this organism could colonize both small metastatic lesions as well as larger tumors. These observations are the impetus to a burgeoning field focused on the development of Salmonella as a clinically useful anti-cancer agent. We will discuss three cutting edge technologies employing Salmonella to target tumors.
Collapse
|
37
|
Cronin M, Ventura M, Fitzgerald GF, van Sinderen D. Progress in genomics, metabolism and biotechnology of bifidobacteria. Int J Food Microbiol 2011; 149:4-18. [PMID: 21320731 DOI: 10.1016/j.ijfoodmicro.2011.01.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 01/06/2011] [Accepted: 01/10/2011] [Indexed: 12/16/2022]
Abstract
Members of the genus Bifidobacterium were first described over a century ago and were quickly associated with a healthy intestinal tract due to their numerical dominance in breast-fed babies as compared to bottle-fed infants. Health benefits elicited by bifidobacteria to its host, as supported by clinical trials, have led to their wide application as probiotic components of health-promoting foods, especially in fermented dairy products. However, the relative paucity of genetic tools available for bifidobacteria has impeded development of a comprehensive molecular understanding of this genus. In this review we present a summary of current knowledge on bifidobacterial metabolism, classification, physiology and genetics and outline the currently available methods for genetically accessing and manipulating the genus.
Collapse
Affiliation(s)
- Michelle Cronin
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jnr. Laboratory, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
38
|
Abstract
Several bacterial species have inherent ability to colonize solid tumors in vivo. However, their natural anti-tumor activity can be enhanced by genetic engineering that enables these bacteria express or transfer therapeutic molecules into target cells. In this review, we summarize latest research on cancer therapy using genetically modified bacteria with particular emphasis on blocking tumor angiogenesis. Despite recent progress, only a few recent studies on bacterial tumor therapy have focused on anti-angiogenesis. Bacteria-mediated anti-angiogenesis therapy for cancer, however, is an attractive approach given that solid tumors are often characterized by increased vascularization. Here, we discuss four different approaches for using modified bacteria as anti-cancer therapeutics--bactofection, DNA vaccination, alternative gene therapy and transkingdom RNA interference--with a specific focus on angiogenesis suppression. Critical areas and future directions for this field are also outlined.
Collapse
|
39
|
Wall DM, Srikanth C, McCormick BA. Targeting tumors with salmonella Typhimurium- potential for therapy. Oncotarget 2010; 1:721-728. [PMID: 21321381 PMCID: PMC3157733 DOI: 10.18632/oncotarget.206] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 12/29/2010] [Indexed: 11/25/2022] Open
Abstract
When one considers the organism Salmonella enterica serotype Typhimurium (S. Typhimurium), one usually thinks of the Gram-negative enteric pathogen that causes the severe food borne illness, gastroentertitis. In this context, the idea of Salmonella being exploited as a cancer therapeutic seems pretty remote. However, there has been an escalating interest in the development of tumor-therapeutic bacteria for use in the treatment of a variety of cancers. This strategy takes advantage of the remarkable ability of certain bacteria to preferentially replicate and accumulate within tumors. In the case of S. Typhimurium, this organism infects and selectively grows within implanted tumors, achieving tumor/normal tissue ratios of approximately 1,000:1. Salmonella also has some attractive properties well suited for the design of a chemotherapeutic agent. In particular, this pathogen can easily be manipulated to carry foreign genes, and since this species is a facultative anaerobe, it is able to survival in both oxygenated and hypoxic conditions, implying this organism could colonize both small metastatic lesions as well as larger tumors. These observations are the impetus to a burgeoning field focused on the development of Salmonella as a clinically useful anti-cancer agent. We will discuss three cutting edge technologies employing Salmonella to target tumors.
Collapse
Affiliation(s)
- Daniel M. Wall
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, United Kingdom
| | - C.V. Srikanth
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, United States of America
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, United States of America
| |
Collapse
|
40
|
Fu G, Yin Y, Hu B, Xu G. Bifidobacteriumas a Delivery System of Functional Genes for Cancer Gene Therapy. EMERGING CANCER THERAPY 2010:99-117. [DOI: 10.1002/9780470626528.ch5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
41
|
Increased mRNA expression of interferon-induced Mx1 and immunomodulation following oral administration of IFN-α2b-transformed B. longum to mice. Arch Microbiol 2010; 192:633-8. [PMID: 20535450 DOI: 10.1007/s00203-010-0589-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 05/04/2010] [Accepted: 05/12/2010] [Indexed: 10/19/2022]
|
42
|
Leschner S, Weiss S. Salmonella—allies in the fight against cancer. J Mol Med (Berl) 2010; 88:763-73. [PMID: 20526574 DOI: 10.1007/s00109-010-0636-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 05/06/2010] [Accepted: 05/14/2010] [Indexed: 01/30/2023]
|
43
|
Bernardes N, Seruca R, Chakrabarty AM, Fialho AM. Microbial-based therapy of cancer: current progress and future prospects. Bioeng Bugs 2009; 1:178-90. [PMID: 21326924 DOI: 10.4161/bbug.1.3.10903] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 12/02/2009] [Indexed: 12/12/2022] Open
Abstract
The use of bacteria in the regression of certain forms of cancer has been recognized for more than a century. Much effort, therefore, has been spent over the years in developing wild-type or modified bacterial strains to treat cancer. However, their use at the dose required for therapeutic efficacy has always been associated with toxicity problems and other deleterious effects. Recently, the old idea of using bacteria in the treatment of cancer has attracted considerable interest and new genetically engineered attenuated strains as well as microbial compounds that might have specific anticancer activity without side effects are being evaluated for their ability to act as new anticancer agents. This involves the use of attenuated bacterial strains and expressing foreign genes that encode the ability to convert non-toxic prodrugs to cytotoxic drugs. Novel strategies also include the use of bacterial products such as proteins, enzymes, immunotoxins and secondary metabolites, which specifically target cancer cells and cause tumor regression through growth inhibition, cell cycle arrest or apoptosis induction. In this review we describe the current knowledge and discuss the future directions regarding the use of bacteria or their products, in cancer therapy.
Collapse
Affiliation(s)
- Nuno Bernardes
- Institute for Biotechnology and Bioengineering (IBB), Center for Biological and Chemical Engineering, Instituto Superior Tecnico, Lisbon, Portugal
| | | | | | | |
Collapse
|