1
|
Li X, Wang Y, Wang Y, Xie H, Gong R, Wu X, Chen J, Sun C, Gu Y. Anti-tumor activity of an αPD-L1-PE38 immunotoxin delivered by engineered Nissle 1917. Int J Biol Macromol 2025; 295:139537. [PMID: 39788238 DOI: 10.1016/j.ijbiomac.2025.139537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Although immune checkpoint inhibitors specifically targeting the PD-1/PD-L1 axis have exhibited remarkable clinical success, they are not uniformly effective across all patient cohorts. Immunotoxins, a novel class of cancer therapeutics, offering a promising alternative. PD-L1, which is also present in certain normal tissues, limits its suitability as an ideal target for immunotoxins. The probiotic strain of E. coli Nissle 1917 (EcN) could target and colonize to solid tumors, which positions it as a promising candidate for tumor tissue-specific delivery of anti-tumor proteins. In this study, we constructed a PD-L1-targeted immunotoxin, designated as αPD-L1-PE38, by fusing an anti-PD-L1 nanobody and a clinically validated PE38 toxin. This immunotoxin exhibited potent cytotoxic activity against tumor cells while showed slightly cytotoxic activity against normal cells. To effectively deliver the αPD-L1-PE38 to tumor tissues, we engineered the EcN strain to release the immunotoxin induced by L-arabinose. Upon induction, the immunotoxin was efficiently secreted, and exhibited robust anti-tumor activity mainly by inducing cell apoptosis both in vitro and in vivo. Furthermore, we enhanced the immunotoxin's affinity for PD-L1 by optimizing the linker between the nanobody and PE38 toxin. The engineered EcN expressing the optimized immunotoxin, achieved superior anti-tumor activity. Collectively, our study suggests that the delivery of immunotoxins through live bacteria to improve safety and efficacy is a promising option in cancer therapeutics.
Collapse
Affiliation(s)
- Xinrui Li
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yangui Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yanqing Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Huilin Xie
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Ruxin Gong
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, China
| | - Jin Chen
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Changning Sun
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Yuchao Gu
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
2
|
Kavianpour A, Behbahani M, Mohabatkar H. Bioinformatics comparison of hemolysin in different bacteria and experimental evaluation of anti-cancer properties of extracts of some hemolysin-producing bacteria. Microb Pathog 2024; 194:106815. [PMID: 39032674 DOI: 10.1016/j.micpath.2024.106815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/07/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Cancer is one of the main causes of death in the world. Resistance to anticancer treatments in patients with advanced solid tumors leads to new treatments. Therefore, more alternative anticancer methods have been found over time with greater specificity against tumor cells and with less or no adverse effects on normal cells. Bacterial spores of obligate anaerobes exclusively germinate in the hypoxic/necrotic areas and not in the well oxygenated areas of the body. This unique phenomenon has been exploited in using bacterial spores as a remedy for cancer. Bacterial toxins also play a significant role in either directly killing tumor cells or altering the cellular processes of the tumor cells which ultimately leads to the inhibition and regression of the solid tumor. In the microbial environment, pathogens such as Staphylococcus aureus, Bacillus cereus, or Streptococcus pyogenes produce hemolysin. This protein is used as an anti-cancer protein. To identify the production of hemolysin by bacteria, which can destroy cancer cells more effectively, different bacterial strains were first cultured in blood agar culture medium. The Strains that completely lysed red blood cells, creating transparent zones, were selected for further investigation. Then, to find out which strains have more ability to lyse red blood cells, the qualitative method of halo diameter measurement was used. Also, using quantitative methods, hemolysin strength in microtubes was determined compared to control samples. The results of the hemolysis in the microtube and the qualitative test results showed similar results. In the next step, the cell viability test was performed with the partially purified proteins. Then, bioinformatics studies such as secondary structure investigation, physicochemical properties, pseudo amino acid composition, and molecular docking were performed. The results of molecular docking showed that the hemolysin protein has the highest affinity for the cholesterol of the cytoplasmic membrane, respectively, of Bacillus subtilis, Bacillus cereus, and Staphylococcus aureus bacteria which play a significant role in either directly killing tumor cells or altering the cellular processes of the tumor cells which ultimately leads to the inhibition and regression of the solid tumor.
Collapse
|
3
|
Zhou Y, Li Q, Wu Y, Zhang W, Ding L, Ji C, Li P, Chen T, Feng L, Tang BZ, Huang X. Synergistic Brilliance: Engineered Bacteria and Nanomedicine Unite in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313953. [PMID: 38400833 DOI: 10.1002/adma.202313953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Engineered bacteria are widely used in cancer treatment because live facultative/obligate anaerobes can selectively proliferate at tumor sites and reach hypoxic regions, thereby causing nutritional competition, enhancing immune responses, and producing anticancer microbial agents in situ to suppress tumor growth. Despite the unique advantages of bacteria-based cancer biotherapy, the insufficient treatment efficiency limits its application in the complete ablation of malignant tumors. The combination of nanomedicine and engineered bacteria has attracted increasing attention owing to their striking synergistic effects in cancer treatment. Engineered bacteria that function as natural vehicles can effectively deliver nanomedicines to tumor sites. Moreover, bacteria provide an opportunity to enhance nanomedicines by modulating the TME and producing substrates to support nanomedicine-mediated anticancer reactions. Nanomedicine exhibits excellent optical, magnetic, acoustic, and catalytic properties, and plays an important role in promoting bacteria-mediated biotherapies. The synergistic anticancer effects of engineered bacteria and nanomedicines in cancer therapy are comprehensively summarized in this review. Attention is paid not only to the fabrication of nanobiohybrid composites, but also to the interpromotion mechanism between engineered bacteria and nanomedicine in cancer therapy. Additionally, recent advances in engineered bacteria-synergized multimodal cancer therapies are highlighted.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Qianying Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Wan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Lu Ding
- Department of Cardiology, Jiangxi Hypertension Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, 310030, P. R. China
| | - Ping Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330036, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, P. R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| |
Collapse
|
4
|
Sharma D, Gajjar D, Seshadri S. Understanding the role of gut microfloral bifidobacterium in cancer and its potential therapeutic applications. MICROBIOME RESEARCH REPORTS 2023; 3:3. [PMID: 38455077 PMCID: PMC10917622 DOI: 10.20517/mrr.2023.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 03/09/2024]
Abstract
Gut microbiota research has gained a tremendous amount of attention from the scientific community because of its contribution to gut homeostasis, human health, and various pathophysiological conditions. The early colonizer of the human gut, i.e., bifidobacteria, has emerged as an efficient probiotic in various diseased conditions, including cancer. This review explores the pros and cons of Bifidobacterium in various malignancies and various therapeutic strategies. We have illustrated the controversial role of bifidobacteria participating in various malignancies as well as described the current knowledge regarding its use in anticancer therapies. Ultimately, this article also addresses the need for further extensive research in elucidating the mechanism of how bifidobacteria is involved and is indirectly affecting the tumor microenvironment. Exhaustive and large-scale research is also required to solve the controversial questions regarding the involvement of bifidobacteria in cancer research.
Collapse
Affiliation(s)
| | | | - Sriram Seshadri
- Institute of Science, Nirma University, 382481 Ahmedabad, Gujarat, India
| |
Collapse
|
5
|
Ikryannikova LN, Gorokhovets NV, Belykh DA, Kurbatov LK, Zamyatnin AA. Bacterial Therapy of Cancer: A Way to the Dustbin of History or to the Medicine of the Future? Int J Mol Sci 2023; 24:ijms24119726. [PMID: 37298677 DOI: 10.3390/ijms24119726] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Bacteria are the constant companions of the human body throughout its life and even after its death. The history of a human disease such as cancer and the history of microorganisms, particularly bacteria, are believed to closely intertwined. This review was conceived to highlight the attempts of scientists from ancient times to the present day to discover the relationship between bacteria and the emergence or development of tumors in the human body. Challenges and achievements of 21st century science in forcing bacteria to serve for cancer treatment are considered. The future possibilities of bacterial cancer therapy, including the creation of bacterial microrobots, or "bacteriobots", are also discussed.
Collapse
Affiliation(s)
- Larisa N Ikryannikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Neonila V Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Darya A Belykh
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Leonid K Kurbatov
- Orekhovich Research Institute of Biomedical Chemistry, Pogodinskaya 10/8, 119991 Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1/73, 119234 Moscow, Russia
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, 119992 Moscow, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
| |
Collapse
|
6
|
Takahashi M, Sukowati EW, Nomura S, Kato A, Mizuseki K, Watanabe Y, Mukai H. Impact of tumoral structure and bacterial species on growth and biodistribution of live bacterial therapeutics in xenografted tumours. J Drug Target 2023; 31:194-205. [PMID: 36097977 DOI: 10.1080/1061186x.2022.2122477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Live bacterial therapeutics is gaining attention, especially for cancer therapy, because anaerobic bacteria selectively grow inside the solid tumours. However, the effect of tumour structure and bacterial characteristics on the pharmacokinetics of tumours is unclear; therefore, we aimed to elucidate the effects of tumour structure and types of bacteria on tumoral bacterial growth. Using six mouse xenograft models, including stroma-rich tumours similar to clinical tumours, and two models of live bacterial therapeutics, Salmonella typhimurium VNP20009 and Escherichia coli DH5α, we investigated bacterial growth and distribution in tumours after intravenous administration. Rapid growth of E. coli was observed in HCT116 and other tumours with few collagens, blood vessels not covered by mural cells, and a cancer cell area proliferated disorderly, whereas tumours with contrasting features, such as BxPC-3, showed lower bacterial growth and a limited intratumor distribution. Alternatively, Salmonella typhimurium VNP20009, when successfully proliferated (the probability was approximately 50%), grew to 108 colony forming units/g tissue even in BxPC-3 tumours, and its intratumor distribution was extensive. This study suggests that the development of new methods to modify tumour structure will be essential for the development of anti-tumour clinical therapies based on live bacterial therapeutics.
Collapse
Affiliation(s)
- Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Physiology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Erike Widyasari Sukowati
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Shoko Nomura
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Akari Kato
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kenji Mizuseki
- Department of Physiology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
7
|
Menati Rashno M, Mehraban H, Naji B, Radmehr M. Microbiome in human cancers. Access Microbiol 2021; 3:000247. [PMID: 34888478 PMCID: PMC8650843 DOI: 10.1099/acmi.0.000247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
A microbiome is defined as the aggregate of all microbiota that reside in human digestive system and other tissues. This microbiota includes viruses, bacteria, fungi that live in various human organs and tissues like stomach, guts, oesophagus, mouth cavity, urinary tract, vagina, lungs, and skin. Almost 20 % of malignant cancers worldwide are related to microbial infections including bacteria, parasites, and viruses. The human body is constantly being attacked by microbes during its lifetime and microbial pathogens that have tumorigenic effects in 15-20 % of reported cancer cases. Recent scientific advances and the discovery of the effect of microbes on cancer as a pathogen or as a drug have significantly contributed to our understanding of the complex relationship between microbiome and cancer. The aim of this study is to overview some microbiomes that reside in the human body and their roles in cancer.
Collapse
Affiliation(s)
| | - Hamed Mehraban
- Department of Biology, Payame Noor University (PNU), Tehran, Iran
| | - Behnaz Naji
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mohadeseh Radmehr
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
8
|
Rottinghaus AG, Amrofell MB, Moon TS. Biosensing in Smart Engineered Probiotics. Biotechnol J 2020; 15:e1900319. [PMID: 31860168 PMCID: PMC7305048 DOI: 10.1002/biot.201900319] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/05/2019] [Indexed: 01/01/2023]
Abstract
Engineered microbes are exciting alternatives to current diagnostics and therapeutics. Researchers have developed a wide range of genetic tools and parts to engineer probiotic and commensal microbes. Among these tools and parts, biosensors allow the microbes to sense and record or to sense and respond to chemical and environmental signals in the body, enabling them to report on health conditions of the animal host and/or deliver therapeutics in a controlled manner. This review focuses on how biosensing is applied to engineer "smart" microbes for in vivo diagnostic, therapeutic, and biocontainment goals. Hurdles that need to be overcome when transitioning from high-throughput in vitro systems to low-throughput in vivo animal models, new technologies that can be implemented to alleviate this experimental gap, and areas where future advancements can be made to maximize the utility of biosensing for medical applications are also discussed. As technologies for engineering microbes continue to be developed, these engineered organisms will be used to address many medical challenges.
Collapse
Affiliation(s)
- Austin G. Rottinghaus
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, United States
| | - Matthew B. Amrofell
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, United States
| | - Tae Seok Moon
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, United States
- Division of Biology and Biomedical Sciences, Washington University in St Louis, St. Louis, United States
| |
Collapse
|
9
|
Mansour M, Ismail S, Abou-Aisha K. Bacterial delivery of the anti-tumor azurin-like protein Laz to glioblastoma cells. AMB Express 2020; 10:59. [PMID: 32221741 PMCID: PMC7099546 DOI: 10.1186/s13568-020-00995-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/18/2020] [Indexed: 02/02/2023] Open
Abstract
Salmonella typhimurium VNP-20009 (VNP) is a non-pathogenic attenuated strain, which, as a facultative anaerobe, preferentially accumulates in hypoxic regions of solid tumors. Here, VNP was utilized as a delivery vehicle of the anti-tumor protein Lipidated azurin, Laz, which is produced by the meningitis-causing bacterium Neisseria meningitides. In brain cancer cells, Laz has been demonstrated to induce apoptosis through an interaction with the tumor suppressor protein p53. In this study, the laz gene, including its signal sequence, was cloned downstream of a hypoxia inducible promoter (HIP-1), before being electroporated into VNP. Successful ectopic expression and export of the Laz protein by VNP under hypoxic conditions were confirmed by Western blot analysis of the cell-free culture medium. Effective expression of Laz by VNP was investigated in two glioblastoma cell lines: LN-229 and U-373, with the latter line carrying a mutated version of p53; as well as in the breast cancer line MCF-7. Cytotoxicity of the VNP-Laz was assessed by determining the fluorescence of the apoptotic marker caspases 3/7. Compared to the purified Laz, VNP-Laz, significantly induced apoptosis in MCF-7, LN-229 and, to a much lower extent in U-373 cells, suggesting a p53-linked mechanism. Our results might represent a new approach of targeted gene delivery and suggest a potential application in brain tumor therapy.
Collapse
Affiliation(s)
- Manar Mansour
- Department of Microbiology and Immunology, The German University in Cairo (GUC), Main Entrance Fifth Settlement, Cairo, Egypt.
| | - Shehab Ismail
- The Cancer Research Institute CRUK Beatson Institute, Glasgow, UK
| | - Khaled Abou-Aisha
- Department of Microbiology and Immunology, The German University in Cairo (GUC), Main Entrance Fifth Settlement, Cairo, Egypt
| |
Collapse
|
10
|
He L, Yang H, Tang J, Liu Z, Chen Y, Lu B, He H, Tang S, Sun Y, Liu F, Ding X, Zhang Y, Hu S, Xia L. Intestinal probiotics E. coli Nissle 1917 as a targeted vehicle for delivery of p53 and Tum-5 to solid tumors for cancer therapy. J Biol Eng 2019; 13:58. [PMID: 31297149 PMCID: PMC6599283 DOI: 10.1186/s13036-019-0189-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/17/2019] [Indexed: 01/19/2023] Open
Abstract
Traditional cancer therapies, such as surgery treatment, radiotherapy, and chemotherapy, often fail to completely eliminate tumor cells in an anaerobic microenvironment of tumor regions. In contrast to these traditional cancer therapies, the use of targeted delivery vectors to deliver anticancer genes or antitumor drugs to hypoxic areas in tumors is the most clinically promising cancer treatment with rapid development in recent years. In this study, E.coli Nissle 1917 (EcN), an intestinal probiotic, was utilized as a targeted transport vector to deliver p53 and Tum-5 protein to tumor hypoxic regions. The tumor-targeting characteristics of EcN were investigated using luciferase LuxCDABE operon, and the results demonstrated that EcN could specifically accumulate in the solid tumor areas of SMMC-7721 tumor-bearing BALB/c nude mice. The Tum 5-p53 bifunctional proteins were initially constructed and then delivered to solid tumor regions by using the targeted transporter EcN for cancer therapy. The antitumor effect and safety of three engineered bacteria, namely, EcN (Tum-5), EcN (p53), and EcN (Tum 5-p53), were also examined. The calculated tumor volume and tumor weight indicated that these three engineered bacteria could inhibit the growth of human hepatoma SMMC-7721 cells, and the antitumor effect of EcN (Tum 5-p53) expressing the Tum 5-p53 fusion protein was significantly better than those of EcN (Tum-5) and EcN (p53) alone. Immunofluorescence demonstrated that the expression of Ki-67, a nuclear proliferation-related protein, was inhibited in the tumor areas of the groups treated with the engineered bacteria, whereas the expression of caspase-3 was upregulated. The expression trends of Ki-67 and caspase-3 were consistent with the different antitumor efficacies of these three engineered bacteria. EcN did not elicit obvious side effects on mice. This research not only provids a foundation for tumor-targeted therapy but also contributes greatly to the development of antitumor agents and anticancer proteins.
Collapse
Affiliation(s)
- Lian He
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China.,2School of Basic Medical Science, Changsha Medical University, Changsha, 410298 People's Republic of China
| | - Huijun Yang
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Jianli Tang
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Zhudong Liu
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Yiyan Chen
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Binghua Lu
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Haocheng He
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Sijia Tang
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Yunjun Sun
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Fei Liu
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Xuezhi Ding
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Youming Zhang
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Shengbiao Hu
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| | - Liqiu Xia
- 1Hunan Provincial Key Laboratory of Microbial Molecular Biology, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081 People's Republic of China
| |
Collapse
|
11
|
Gao X, Zou W, Jiang B, Xu D, Luo Y, Xiong J, Yan S, Wang Y, Tang Y, Chen C, Li H, Qiao H, Wang Q, Zou J. Experimental Study of Retention on the Combination of Bifidobacterium with High-Intensity Focused Ultrasound (HIFU) Synergistic Substance in Tumor Tissues. Sci Rep 2019; 9:6423. [PMID: 31015517 PMCID: PMC6478724 DOI: 10.1038/s41598-019-42832-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/09/2019] [Indexed: 01/04/2023] Open
Abstract
High intensity focused ultrasound (HIFU) has been recently regarded to be a new type of technique for non-invasive ablation of local tumors and HIFU synergists could significantly improve its therapeutic efficiency. The therapeutic efficiency of HIFU is greatly limited by the low retention of HIFU synergists in the target area and short residence time. This study aimed to explore a method to increase the deposition of HIFU synergists in tumors. Cationic lipid nanoparticle can be used to enhance the HIFU ablation effect, but there is still a problem for it that the deposition amount in the tumor tissue is small and the residence time is short. Bifidobacterium is highly biosafe and can be selectively colonized in the hypoxic zone of tumor tissue. Cationic lipid nanoparticles can be observed in vitro by attachment to bifidobacterium by electrostatic adsorption. And the effect of the proliferation of bifidobacterium in tumor tissues on the retention amount and retention time of cationic lipid nanoparticles in vivo was evaluated. Results showed that the cationic lipid nanoparticles were linked to the surface of Bifidobacterium effectively in vitro, while in vivo, the retention amount and retention time of cationic lipid nanoparticles could be increased by Bifidobacterium in tumor tissues, which provided a new method for improving the therapeutic efficiency of HIFU.
Collapse
Affiliation(s)
- Xuan Gao
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Wenjuan Zou
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Binglei Jiang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Die Xu
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yong Luo
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Xiong
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Sijing Yan
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Yaotai Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Chun Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Huanan Li
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Hai Qiao
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Qi Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jianzhong Zou
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
12
|
Sahan AZ, Hazra TK, Das S. The Pivotal Role of DNA Repair in Infection Mediated-Inflammation and Cancer. Front Microbiol 2018; 9:663. [PMID: 29696001 PMCID: PMC5904280 DOI: 10.3389/fmicb.2018.00663] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/21/2018] [Indexed: 12/19/2022] Open
Abstract
Pathogenic and commensal microbes induce various levels of inflammation and metabolic disease in the host. Inflammation caused by infection leads to increased production of reactive oxygen species (ROS) and subsequent oxidative DNA damage. These in turn cause further inflammation and exacerbation of DNA damage, and pose a risk for cancer development. Helicobacter pylori-mediated inflammation has been implicated in gastric cancer in many previously established studies, and Fusobacterium nucleatum presence has been observed with greater intensity in colorectal cancer patients. Despite ambiguity in the exact mechanism, infection-mediated inflammation may have a link to cancer development through an accumulation of potentially mutagenic DNA damage in surrounding cells. The multiple DNA repair pathways such as base excision, nucleotide excision, and mismatch repair that are employed by cells are vital in the abatement of accumulated mutations that can lead to carcinogenesis. For this reason, understanding the role of DNA repair as an important cellular mechanism in combatting the development of cancer will be essential to characterizing the effect of infection on DNA repair proteins and to identifying early cancer biomarkers that may be targeted for cancer therapies and treatments.
Collapse
Affiliation(s)
- Ayse Z Sahan
- Department of Pathology, University of California, San Diego, San Diego, CA, United States
| | - Tapas K Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Soumita Das
- Department of Pathology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
13
|
Sun M, Wang K, Oupický D. Advances in Stimulus-Responsive Polymeric Materials for Systemic Delivery of Nucleic Acids. Adv Healthc Mater 2018; 7:10.1002/adhm.201701070. [PMID: 29227047 PMCID: PMC5821579 DOI: 10.1002/adhm.201701070] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/13/2017] [Indexed: 01/02/2023]
Abstract
Polymeric materials that respond to a variety of endogenous and external stimuli are actively developed to overcome the main barriers to successful systemic delivery of therapeutic nucleic acids. Here, an overview of viable stimuli that are proved to improve systemic delivery of nucleic acids is provided. The main focus is placed on nucleic acid delivery systems (NADS) based on polymers that respond to pathological or physiological changes in pH, redox state, enzyme levels, hypoxia, and reactive oxygen species levels. Additional discussion is focused on NADS suitable for applications that use external stimuli, such as light, ultrasound, and local hyperthermia.
Collapse
Affiliation(s)
- Minjie Sun
- State Key Laboratory of Natural Medicines, Key Laboratory on Protein Chemistry and Structural Biology, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Kaikai Wang
- State Key Laboratory of Natural Medicines, Key Laboratory on Protein Chemistry and Structural Biology, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - David Oupický
- State Key Laboratory of Natural Medicines, Key Laboratory on Protein Chemistry and Structural Biology, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P.R. China
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
14
|
Bifidobacterium breve as a delivery vector of IL-24 gene therapy for head and neck squamous cell carcinoma in vivo. Gene Ther 2017; 24:699-705. [DOI: 10.1038/gt.2017.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 12/17/2022]
|
15
|
|
16
|
Raj R, Das S. Development and application of anticancer fluorescent CdS nanoparticles enriched Lactobacillus bacteria as therapeutic microbots for human breast carcinoma. Appl Microbiol Biotechnol 2017; 101:5439-5451. [PMID: 28455616 DOI: 10.1007/s00253-017-8298-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/06/2017] [Accepted: 04/11/2017] [Indexed: 01/21/2023]
Abstract
Applications of probiotic bacteria and nanoparticles (NPs) as therapeutic agents have great importance. This study demonstrates a combinatorial approach of both the probiotic Lactobacillus spp. (Lactobacillus fermentum and Lactobacillus plantarum) with fluorescent cadmium sulfide (CdS) NPs as therapeutic agents to target MCF-7 cancer cells (human breast cancer cells). In this study, facultative anaerobic Lactobacillus was successfully used as a vehicle to transport NPs into MCF-7 cancer cells. The cell viability assay and invasion study along with confocal and field emission scanning electron microscopy (FESEM) confirmed the release of payload (CdS NPs) into cytoplasm without any external stimuli. The biosynthesized CdS NPs of ∼22 nm were characterized by FESEM, transmission electron microscopy (TEM), atomic force microscopy (AFM), and fluorescence spectroscopy. The bacteria-NPs (microbots) interaction was investigated by growth curve studies, attenuated total reflection Fourier transform infrared spectroscopy (ATR-FTIR), FESEM, energy dispersive X-ray spectroscopy (EDX), and fluorescence and confocal microscopy. This alternative approach showed an approved and inexpensive delivering mode of specific functional cargos or therapeutic agents into the cancer cells.
Collapse
Affiliation(s)
- Ritu Raj
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Surajit Das
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
17
|
Qian C, Feng P, Yu J, Chen Y, Hu Q, Sun W, Xiao X, Hu X, Bellotti A, Shen QD, Gu Z. Anaerobe-Inspired Anticancer Nanovesicles. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201611783] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chenggen Qian
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE; School of Chemistry and Chemical Engineering; Nanjing University, Jiangsu; Nanjing 210023 P.R. China
| | - Peijian Feng
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE; School of Chemistry and Chemical Engineering; Nanjing University, Jiangsu; Nanjing 210023 P.R. China
| | - Jicheng Yu
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Yulei Chen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE; School of Chemistry and Chemical Engineering; Nanjing University, Jiangsu; Nanjing 210023 P.R. China
| | - Quanyin Hu
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Wujin Sun
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Xuanzhong Xiao
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE; School of Chemistry and Chemical Engineering; Nanjing University, Jiangsu; Nanjing 210023 P.R. China
| | - Xiuli Hu
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Qun-Dong Shen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE; School of Chemistry and Chemical Engineering; Nanjing University, Jiangsu; Nanjing 210023 P.R. China
| | - Zhen Gu
- Joint Department of Biomedical Engineering; University of North Carolina at Chapel Hill and North Carolina State University; Raleigh NC 27695 USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
- Department of Medicine; University of North Carolina School of Medicine; Chapel Hill NC 27599 USA
| |
Collapse
|
18
|
Qian C, Feng P, Yu J, Chen Y, Hu Q, Sun W, Xiao X, Hu X, Bellotti A, Shen QD, Gu Z. Anaerobe-Inspired Anticancer Nanovesicles. Angew Chem Int Ed Engl 2017; 56:2588-2593. [PMID: 28140504 DOI: 10.1002/anie.201611783] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/07/2017] [Indexed: 11/07/2022]
Abstract
Anaerobic bacteria, such as Clostridium and Salmonella, can selectively invade and colonize in tumor hypoxic regions (THRs) and deliver therapeutic products to destroy cancer cells. Herein, we present an anaerobe nanovesicle mimic that can not only be activated in THRs but also induce hypoxia in tumors by themselves. Moreover, inspired by the oxygen metabolism of anaerobes, we construct a light-induced hypoxia-responsive modality to promote dissociation of vehicles and activation of bioreductive prodrugs simultaneously. In vitro and in vivo experiments indicate that this anaerobe-inspired nanovesicle can efficiently induce apoptotic cell death and significantly inhibit tumor growth. Our work provides a new strategy for engineering stimuli-responsive drug delivery systems in a bioinspired and synergistic fashion.
Collapse
Affiliation(s)
- Chenggen Qian
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu, Nanjing, 210023, P.R. China
| | - Peijian Feng
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu, Nanjing, 210023, P.R. China
| | - Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yulei Chen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu, Nanjing, 210023, P.R. China
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xuanzhong Xiao
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu, Nanjing, 210023, P.R. China
| | - Xiuli Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Qun-Dong Shen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu, Nanjing, 210023, P.R. China
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA.,Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
19
|
Mowday AM, Guise CP, Ackerley DF, Minton NP, Lambin P, Dubois LJ, Theys J, Smaill JB, Patterson AV. Advancing Clostridia to Clinical Trial: Past Lessons and Recent Progress. Cancers (Basel) 2016; 8:cancers8070063. [PMID: 27367731 PMCID: PMC4963805 DOI: 10.3390/cancers8070063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/15/2016] [Accepted: 06/22/2016] [Indexed: 01/19/2023] Open
Abstract
Most solid cancers contain regions of necrotic tissue. The extent of necrosis is associated with poor survival, most likely because it reflects aggressive tumour outgrowth and inflammation. Intravenously injected spores of anaerobic bacteria from the genus Clostridium infiltrate and selectively germinate in these necrotic regions, providing cancer-specific colonisation. The specificity of this system was first demonstrated over 60 years ago and evidence of colonisation has been confirmed in multiple tumour models. The use of "armed" clostridia, such as in Clostridium Directed Enzyme Prodrug Therapy (CDEPT), may help to overcome some of the described deficiencies of using wild-type clostridia for treatment of cancer, such as tumour regrowth from a well-vascularised outer rim of viable cells. Successful preclinical evaluation of a transferable gene that metabolises both clinical stage positron emission tomography (PET) imaging agents (for whole body vector visualisation) as well as chemotherapy prodrugs (for conditional enhancement of efficacy) would be a valuable early step towards the prospect of "armed" clostridia entering clinical evaluation. The ability to target the immunosuppressive hypoxic tumour microenvironment using CDEPT may offer potential for synergy with recently developed immunotherapy strategies. Ultimately, clostridia may be most efficacious when combined with conventional therapies, such as radiotherapy, that sterilise viable aerobic tumour cells.
Collapse
Affiliation(s)
- Alexandra M Mowday
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Christopher P Guise
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - David F Ackerley
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand.
- School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand.
| | - Nigel P Minton
- The Clostridia Research Group, BBSRC/EPSRC Synthetic Biology Research Centre (SBRC) School of Life Sciences, University of Nottingham, Nottingham NG72RD, UK.
| | - Philippe Lambin
- Maastro (Maastricht Radiation Oncology), GROW School for Oncology and Development Biology, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands.
| | - Ludwig J Dubois
- Maastro (Maastricht Radiation Oncology), GROW School for Oncology and Development Biology, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands.
| | - Jan Theys
- Maastro (Maastricht Radiation Oncology), GROW School for Oncology and Development Biology, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands.
| | - Jeff B Smaill
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Adam V Patterson
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
20
|
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: 37] [Impact Index Per Article: 4.1] [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
|
21
|
Hernandez-Alcoceba R, Poutou J, Ballesteros-Briones MC, Smerdou C. Gene therapy approaches against cancer using in vivo and ex vivo gene transfer of interleukin-12. Immunotherapy 2016; 8:179-98. [PMID: 26786809 DOI: 10.2217/imt.15.109] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IL-12 is an immunostimulatory cytokine with strong antitumor properties. Systemic administration of IL-12 in cancer patients led to severe toxic effects, prompting the development of gene therapy vectors able to express this cytokine locally in tumors. Both nonviral and viral vectors have demonstrated a high antitumor efficacy in preclinical tumor models. Some of these vectors, including DNA electroporation, adenovirus and ex vivo transduced dendritic cells, were tested in patients, showing low toxicity and moderate antitumor efficacy. IL-12 activity can be potentiated by molecules with immunostimulatory, antiangiogenic or cytotoxic activity. These combination therapies are of clinical interest because they could lower the threshold for IL-12 efficacy, increasing the therapeutic potential of gene therapy and preventing the toxicity mediated by this cytokine.
Collapse
Affiliation(s)
- Ruben Hernandez-Alcoceba
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Joanna Poutou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - María Cristina Ballesteros-Briones
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Cristian Smerdou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| |
Collapse
|
22
|
Kramer MG, Masner M, Casales E, Moreno M, Smerdou C, Chabalgoity JA. Neoadjuvant administration of Semliki Forest virus expressing interleukin-12 combined with attenuated Salmonella eradicates breast cancer metastasis and achieves long-term survival in immunocompetent mice. BMC Cancer 2015; 15:620. [PMID: 26347489 PMCID: PMC4562361 DOI: 10.1186/s12885-015-1618-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metastatic breast cancer is a major cause of death among women worldwide; therefore efficient therapeutic strategies are extremely needed. In this work we have developed a gene therapy- and bacteria-based combined neoadjuvant approach and evaluated its antitumor effect in a clinically relevant animal model of metastatic breast cancer. METHODS 2×10(8) particles of a Semliki Forest virus vector expressing interleukin-12 (SFV-IL-12) and/or 2×10(7) units of an aroC (-) Samonella Typhimurium strain (LVR01) were injected into 4T1 tumor nodules orthotopically implanted in mice. Tumors were surgically resected and long-term survival was determined. IL-12 and interferon-γ were quantified by Enzyme-Linked ImmunoSorbent Assay, bacteria was visualized by inmunohistochemistry and the number of lung metastasis was calculated with a clonogenic assay. RESULTS SFV-IL-12 and LVR01 timely inoculated and followed by surgical resection of tumors succeeded in complete inhibition of lethal lung metastasis and long-term survival in 90% of treated mice. The combined therapy was markedly synergistic compared to each treatment alone, since SFV-IL-12 monotherapy showed a potent antiangiogenic effect, being able to inhibit tumor growth and extend survival, but could not prevent establishment of distant metastasis and death of tumor-excised animals. On the other hand, LVR01 alone also showed a significant, although limited, antitumor potential, despite its ability to invade breast cancer cells and induce granulocyte recruitment. The efficacy of the combined therapy depended on the order in which both factors were administered; inasmuch the therapeutic effect was only observed when SFV-IL-12 was administered previous to LVR01, whereas administration of LVR01 before SFV-IL-12 had negligible antitumor activity. Moreover, pre-treatment with LVR01 seemed to suppress SFV-IL-12 antiangiogenic effects associated to lower IL-12 expression in this group. Re-challenged mice were unable to reject a second 4T1 tumor; however 100% of them could be totally cured by applying the same neoadjuvant combined regimen. To our knowledge, these are the most encouraging results obtained to date in a post-operatory setting using the highly aggressive 4T1 animal model. CONCLUSIONS SFV-IL-12-based gene therapy combined with Salmonella LVR01 neoadjuvant administration has a synergic antitumor effect and may be a promising therapeutic option to prevent and/or eradicate pre-operatory metastasis in locally advanced breast cancer.
Collapse
Affiliation(s)
- M Gabriela Kramer
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Martín Masner
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Erkuden Casales
- Division Gene Therapy, Center for Applied Medical Research, University of Navarra, Av. Pio XII 55, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, c/Irunlarrea 3, 31008, Pamplona, Spain.
| | - María Moreno
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Cristian Smerdou
- Division Gene Therapy, Center for Applied Medical Research, University of Navarra, Av. Pio XII 55, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, c/Irunlarrea 3, 31008, Pamplona, Spain.
| | - José A Chabalgoity
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| |
Collapse
|
23
|
Belcheva A, Irrazabal T, Martin A. Gut microbial metabolism and colon cancer: Can manipulations of the microbiota be useful in the management of gastrointestinal health? Bioessays 2015; 37:403-12. [DOI: 10.1002/bies.201400204] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Alberto Martin
- Department of Immunology; University of Toronto; ON Canada
| |
Collapse
|
24
|
Ninomiya K, Yamada R, Meisaku H, Shimizu N. Effect of ultrasound irradiation on bacterial internalization and bacteria-mediated gene transfer to cancer cells. ULTRASONICS SONOCHEMISTRY 2014; 21:1187-1193. [PMID: 24373691 DOI: 10.1016/j.ultsonch.2013.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/04/2013] [Accepted: 12/08/2013] [Indexed: 06/03/2023]
Abstract
The present study demonstrates that ultrasound irradiation can facilitate bacteria-mediated gene delivery (bactofection). Escherichia coli modified with avidin were employed as a vehicle for delivery of the green fluorescent protein (GFP) gene, a model heterologous gene, into the breast cancer cell line MCF-7. Avidin-mediated binding of E. coli to MCF-7 cells enhanced the internalization of E. coli by approximately 17%, irrespective of the use of ultrasound irradiation. Furthermore, the use of ultrasound irradiation increased the internalization by approximately 5%, irrespective of the presence of avidin on the E. coli cell surface. The percentages of GFP-expressing MCF-7 cells at 24h after bactofection were below 0.5% and 2% for the case with only avidin-modification of E. coli cell surface and only ultrasound irradiation, respectively. However, combining avidin modification with the ultrasound treatment increased this value to 8%. Thus, the use of avidin-modified bacteria in conjunction with ultrasound irradiation has potential as an effective strategy for tumor-targeted bactofection.
Collapse
Affiliation(s)
- Kazuaki Ninomiya
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | - Ryuji Yamada
- Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | - Hitomi Meisaku
- Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | - Nobuaki Shimizu
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa 920-1192, Japan.
| |
Collapse
|
25
|
Mesa-Pereira B, Medina C, Camacho EM, Flores A, Santero E. Novel tools to analyze the function of Salmonella effectors show that SvpB ectopic expression induces cell cycle arrest in tumor cells. PLoS One 2013; 8:e78458. [PMID: 24205236 PMCID: PMC3804527 DOI: 10.1371/journal.pone.0078458] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 09/12/2013] [Indexed: 01/26/2023] Open
Abstract
In order to further characterize its role in pathogenesis and to establish whether its overproduction can lead to eukaryotic tumor cell death, Salmonella strains able to express its virulence factor SpvB (an ADP-ribosyl transferase enzyme) in a salicylate-inducible way have been constructed and analyzed in different eukaryotic tumor cell lines. To do so, the bacterial strains bearing the expression system have been constructed in a ∆purD background, which allows control of bacterial proliferation inside the eukaryotic cell. In the absence of bacterial proliferation, salicylate-induced SpvB production resulted in activation of caspases 3 and 7 and apoptotic cell death. The results clearly indicated that controlled SpvB production leads to F-actin depolimerization and either G1/S or G2/M phase arrest in all cell lines tested, thus shedding light on the function of SpvB in Salmonella pathogenesis. In the first place, the combined control of protein production by salicylate regulated vectors and bacterial growth by adenine concentration offers the possibility to study the role of Salmonella effectors during eukaryotic cells infection. In the second place, the salicylate-controlled expression of SpvB by the bacterium provides a way to evaluate the potential of other homologous or heterologous proteins as antitumor agents, and, eventually to construct novel potential tools for cancer therapy, given that Salmonella preferentially proliferates in tumors.
Collapse
Affiliation(s)
- Beatriz Mesa-Pereira
- Centro Andaluz de Biología del Desarrollo/ CSIC/ Universidad Pablo de Olavide/ Junta de Andalucía. Departamento de Biología Molecular e Ingeniería Bioquímica, Seville, Spain
| | - Carlos Medina
- Centro Andaluz de Biología del Desarrollo/ CSIC/ Universidad Pablo de Olavide/ Junta de Andalucía. Departamento de Biología Molecular e Ingeniería Bioquímica, Seville, Spain
| | - Eva María Camacho
- Centro Andaluz de Biología del Desarrollo/ CSIC/ Universidad Pablo de Olavide/ Junta de Andalucía. Departamento de Biología Molecular e Ingeniería Bioquímica, Seville, Spain
| | - Amando Flores
- Centro Andaluz de Biología del Desarrollo/ CSIC/ Universidad Pablo de Olavide/ Junta de Andalucía. Departamento de Biología Molecular e Ingeniería Bioquímica, Seville, Spain
- * E-mail:
| | - Eduardo Santero
- Centro Andaluz de Biología del Desarrollo/ CSIC/ Universidad Pablo de Olavide/ Junta de Andalucía. Departamento de Biología Molecular e Ingeniería Bioquímica, Seville, Spain
| |
Collapse
|
26
|
Chen Q, Tarr AJ, Liu X, Wang Y, Reed NS, Demarsh CP, Sheridan JF, Quan N. Controlled progressive innate immune stimulation regimen prevents the induction of sickness behavior in the open field test. J Inflamm Res 2013; 6:91-8. [PMID: 23950656 PMCID: PMC3741056 DOI: 10.2147/jir.s45111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Peripheral immune activation by bacterial mimics or live replicating pathogens is well known to induce central nervous system activation. Sickness behavior alterations are often associated with inflammation-induced increases in peripheral proinflammatory cytokines (eg, interleukin [IL]-1β and IL-6). However, most researchers have used acute high dose endotoxin/bacterial challenges to observe these outcomes. Using this methodology may pose inherent risks in the translational interpretation of the experimental data in these studies. Studies using Escherichia coli have yet to establish the full kinetics of repeated E. coli peripheral injections. Therefore, we sought to examine the effects of repeated low dose E. coli on sickness behavior and local peripheral inflammation in the open field test. Results from the current experiments showed a behavioral dose response, where increased amounts of E. coli resulted in correspondingly increased sickness behavior. Furthermore, animals that received a subthreshold dose (ie, one that did not cause sickness behavior) of E. coli 24 hours prior were able to withstand a larger dose of E. coli on the second day (a dose that would normally cause sickness behavior in mice without prior exposure) without inducing sickness behavior. In addition, animals that received escalating subthreshold doses of E. coli on days 1 and 2 behaviorally tolerated a dose of E. coli 25 times higher than what would normally cause sickness behavior if given acutely. Lastly, increased levels of E. coli caused increased IL-6 and IL-1β protein expression in the peritoneal cavity, and this increase was blocked by administering a subthreshold dose of E. coli 24 hours prior. These data show that progressive challenges with subthreshold levels of E. coli may obviate the induction of sickness behavior and proinflammatory cytokine expression.
Collapse
Affiliation(s)
- Qun Chen
- Division of Oral Biology, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Park SJ, Bae H, Ko SY, Min JJ, Park JO, Park S. Selective bacterial patterning using the submerged properties of microbeads on agarose gel. Biomed Microdevices 2013; 15:793-9. [DOI: 10.1007/s10544-013-9765-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
28
|
Tirandaz H, Mohammadi E. Efficient tumor targeting by anaerobic butyrate-producing bacteria. Med Hypotheses 2013; 80:675-8. [PMID: 23410499 DOI: 10.1016/j.mehy.2013.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Revised: 01/13/2013] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
Abstract
Butyrate as an important short chain fatty acid has been shown to affect different kinds of cancer cells. Butyrate exerts its anti-cancerous effects by several mechanisms and has lead to successful outcomes in phase I and II clinical trials. Moreover, since solid tumors grow rapidly, multiple regions of hypoxia and anoxia forms within them that provide good niches for the growth of anaerobic bacteria. It has been shown that bacterial tumor targeting is an applicable strategy for tumor-selective therapy. Therefore, we propose that nonpathogenic anaerobic butyrate-producing bacteria may be a versatile tool in tumor therapy as they can grow in anoxic and hypoxic regions of tumors and influence tumor cells by producing butyric acid. Moreover, this approach may overcome the existing problems of butyrate delivery to the sites of tumor and enhance its bioavailability. Also reversion of cancer drug resistance by butyrate will be plausible. Tumor targeting with nonpathogenic anaerobic bacteria with a higher capacity to produce butyrate could be the focus of future research.
Collapse
Affiliation(s)
- Hassan Tirandaz
- Biotechnology Research Center, Research Institute of Petroleum Industry, Tehran, Iran.
| | | |
Collapse
|
29
|
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: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
Guan GF, Zhao M, Liu LM, Jin CS, Sun K, Zhang DJ, Yu DJ, Cao HW, Lu YQ, Wen LJ. Salmonella typhimurium mediated delivery of Apoptin in human laryngeal cancer. Int J Med Sci 2013; 10:1639-48. [PMID: 24155656 PMCID: PMC3805922 DOI: 10.7150/ijms.6960] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/30/2013] [Indexed: 01/26/2023] Open
Abstract
An effective cancer therapeutic should target tumours specifically with limited systemic toxicity. Here, we transformed an attenuated Salmonella typhimurium (S. typhimurium) with an Apoptin expressing plasmid into a human laryngeal carcinoma cell line. The expression of the inserted gene was measured using fluorescence and immunoblotting assays. The attenuated S. typhimurium-mediated Apoptin significantly decreased cytotoxicity and strongly increased cell apoptosis through the activation of caspase-3. The process was mediated by Bax, cytochrome c and caspase-9. A syngeneic nude murine tumour model was used to determine the anti-tumour effects of the recombinant bacteria in vivo. Systemic injection of the recombinant bacteria with and without re-dosing caused significant tumour growth delay and reduced tumour microvessel density, thereby extending host survival. Our findings indicated that the use of recombinant Salmonella typhimurium as an Apoptin expression vector has potential cancer therapeutic benefits.
Collapse
Affiliation(s)
- Guo-fang Guan
- 1. Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Letzel AC, Pidot SJ, Hertweck C. A genomic approach to the cryptic secondary metabolome of the anaerobic world. Nat Prod Rep 2012; 30:392-428. [PMID: 23263685 DOI: 10.1039/c2np20103h] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A total of 211 complete and published genomes from anaerobic bacteria are analysed for the presence of secondary metabolite biosynthesis gene clusters, in particular those tentatively coding for polyketide synthases (PKS) and non-ribosomal peptide synthetases (NRPS). We investigate the distribution of these gene clusters according to bacterial phylogeny and, if known, correlate these to the type of metabolic pathways they encode. The potential of anaerobes as secondary metabolite producers is highlighted.
Collapse
Affiliation(s)
- Anne-Catrin Letzel
- Leibniz Institute for Natural Product Research and Infection Biology HKI, Beutenbergstr. 11a, Jena, 07745, Germany
| | | | | |
Collapse
|
32
|
Cho S, Park SJ, Ko SY, Park JO, Park S. Development of bacteria-based microrobot using biocompatible poly(ethylene glycol). Biomed Microdevices 2012; 14:1019-25. [DOI: 10.1007/s10544-012-9704-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
33
|
Walter J, Hausmann S, Drepper T, Puls M, Eggert T, Dihné M. Flavin mononucleotide-based fluorescent proteins function in mammalian cells without oxygen requirement. PLoS One 2012; 7:e43921. [PMID: 22984451 PMCID: PMC3439463 DOI: 10.1371/journal.pone.0043921] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/27/2012] [Indexed: 11/23/2022] Open
Abstract
Usage of the enhanced green fluorescent protein (eGFP) in living mammalian cells is limited to aerobic conditions due to requirement of oxygen during chromophore formation. Since many diseases or disease models are associated with acute or chronic hypoxia, eGFP-labeling of structures of interest in experimental studies might be unreliable leading to biased results. Thus, a chromophore yielding a stable fluorescence under hypoxic conditions is desirable. The fluorescence of flavin mononucleotide (FMN)-based fluorescent proteins (FbFPs) does not require molecular oxygen. Recently, the advantages of FbFPs for several bacterial strains and yeasts were described, specifically, their usage as a real time fluorescence marker in bacterial expression studies and their ability of chromophore formation under anaerobic conditions. Our objective was to verify if FbFPs also function in mammalian cells in order to potentially broaden the repertoire of chromophores with ones that can reliably be used in mammalian studies under hypoxic conditions. In the present study, we demonstrate for the first time, that FbFPs can be expressed in different mammalian cells, among them murine neural stem cells during proliferative and differentiated stages. Fluorescence intensities were comparable to eGFP. In contrast to eGFP, the FbFP fluorescence did not decrease when cells were exposed to defined hypoxic conditions neither in proliferating nor in differentiated cells. Thus, FbFPs can be regarded as an alternative to eGFP in studies that target cellular structures which are exposed to hypoxic conditions.
Collapse
Affiliation(s)
- Janine Walter
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Baden-Württemberg, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
Liu Y, Zhou M, Luo D, Wang L, Hong Y, Yang Y, Sha Y. Bacteria-mediated in vivo delivery of quantum dots into solid tumor. Biochem Biophys Res Commun 2012; 425:769-74. [DOI: 10.1016/j.bbrc.2012.07.150] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 07/27/2012] [Indexed: 11/16/2022]
|
35
|
Garai P, Gnanadhas DP, Chakravortty D. Salmonella enterica serovars Typhimurium and Typhi as model organisms: revealing paradigm of host-pathogen interactions. Virulence 2012; 3:377-388. [PMID: 22722237 PMCID: PMC3478240 DOI: 10.4161/viru.21087] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The lifestyle of intracellular pathogens has always questioned the skill of a microbiologist in the context of finding the permanent cure to the diseases caused by them. The best tool utilized by these pathogens is their ability to reside inside the host cell, which enables them to easily bypass the humoral immunity of the host, such as the complement system. They further escape from the intracellular immunity, such as lysosome and inflammasome, mostly by forming a protective vacuole-bound niche derived from the host itself. Some of the most dreadful diseases are caused by these vacuolar pathogens, for example, tuberculosis by Mycobacterium or typhoid fever by Salmonella. To deal with such successful pathogens therapeutically, the knowledge of a host-pathogen interaction system becomes primarily essential, which further depends on the use of a model system. A well characterized pathogen, namely Salmonella, suits the role of a model for this purpose, which can infect a wide array of hosts causing a variety of diseases. This review focuses on various such aspects of research on Salmonella which are useful for studying the pathogenesis of other intracellular pathogens.
Collapse
Affiliation(s)
- Preeti Garai
- Department of Microbiology and Cell Biology; Centre for Infectious Disease Research and Biosafety Laboratories; Indian Institute of Science; Bangalore, India
| | - Divya Prakash Gnanadhas
- Department of Microbiology and Cell Biology; Centre for Infectious Disease Research and Biosafety Laboratories; Indian Institute of Science; Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology; Centre for Infectious Disease Research and Biosafety Laboratories; Indian Institute of Science; Bangalore, India
| |
Collapse
|
36
|
Yun M, Pan S, Jiang SN, Nguyen VH, Park SH, Jung CH, Kim HS, Min JJ, Choy HE, Hong Y. Effect of Salmonella treatment on an implanted tumor (CT26) in a mouse model. J Microbiol 2012; 50:502-10. [PMID: 22752915 DOI: 10.1007/s12275-012-2090-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 03/14/2012] [Indexed: 01/08/2023]
Abstract
The use of bacteria has contributed to recent advances in targeted cancer therapy especially for its tumor-specific accumulation and proliferation. In this study, we investigated the molecular events following bacterial therapy using an attenuated Salmonella Typhimurium defective in ppGpp synthesis (ΔppGpp), by analyzing those proteins differentially expressed in tumor tissues from treated and untreated mice. CT26 murine colon cancer cells were implanted in BALB/c mice and allowed to form tumors. The tumor-bearing mice were treated with the attenuated Salmonella Typhimurium. Tumor tissues were analyzed by 2D-PAGE. Fourteen differentially expressed proteins were identified by mass spectrometry. The analysis revealed that cytoskeletal components, including vimentin, drebrin-like protein, and tropomyosin-alpha 3, were decreased while serum proteins related to heme or iron metabolism, including transferrin, hemopexin, and haptoglobin were increased. Subsequent studies revealed that the decrease in cytoskeletal components occurred at the transcriptional level and that the increase in heme and iron metabolism proteins occurred in liver. Most interestingly, the same pattern of increased expression of transferrin, hemopexin, and haptoglobin was observed following radiotherapy at the dosage of 14 Gy.
Collapse
Affiliation(s)
- Misun Yun
- Departments of Microbiology, Chonnam National University Medical School, Gwangju, 501-746, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen J, Yang B, Cheng X, Qiao Y, Tang B, Chen G, Wei J, Liu X, Cheng W, Du P, Huang X, Jiang W, Hu Q, Hu Y, Li J, Hua ZC. Salmonella-mediated tumor-targeting TRAIL gene therapy significantly suppresses melanoma growth in mouse model. Cancer Sci 2011; 103:325-33. [DOI: 10.1111/j.1349-7006.2011.02147.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
38
|
Yu H. Bacteria-mediated disease therapy. Appl Microbiol Biotechnol 2011; 92:1107-13. [DOI: 10.1007/s00253-011-3648-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 09/29/2011] [Accepted: 10/16/2011] [Indexed: 12/19/2022]
|
39
|
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
|
40
|
Magnetic resonance imaging of tumors colonized with bacterial ferritin-expressing Escherichia coli. PLoS One 2011; 6:e25409. [PMID: 21984917 PMCID: PMC3184983 DOI: 10.1371/journal.pone.0025409] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/02/2011] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have shown that human ferritin can be used as a reporter of gene expression for magnetic resonance imaging (MRI). Bacteria also encode three classes of ferritin-type molecules with iron accumulation properties. Methods and Findings Here, we investigated whether these bacterial ferritins can also be used as MRI reporter genes and which of the bacterial ferritins is the most suitable reporter. Bacterial ferritins were overexpressed in probiotic E. coli Nissle 1917. Cultures of these bacteria were analyzed and those generating highest MRI contrast were further investigated in tumor bearing mice. Among members of three classes of bacterial ferritin tested, bacterioferritin showed the most promise as a reporter gene. Although all three proteins accumulated similar amounts of iron when overexpressed individually, bacterioferritin showed the highest contrast change. By site-directed mutagenesis we also show that the heme iron, a unique part of the bacterioferritin molecule, is not critical for MRI contrast change. Tumor-specific induction of bacterioferritin-expression in colonized tumors resulted in contrast changes within the bacteria-colonized tumors. Conclusions Our data suggest that colonization and gene expression by live vectors expressing bacterioferritin can be monitored by MRI due to contrast changes.
Collapse
|
41
|
Keates AC, Fruehauf J, Xiang S, Li CJ. TransKingdom RNA interference: a bacterial approach to challenges in RNAi therapy and delivery. Biotechnol Genet Eng Rev 2011; 25:113-27. [PMID: 21412352 DOI: 10.5661/bger-25-113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Since its discovery in 1998 RNA interference (RNAi), a potent and highly selective gene silencing mechanism, has revolutionized the field of biological science. The ability of RNAi to specifically down-regulate the expression of any cellular protein has had a profound impact on the study of gene function in vitro. This property of RNAi also holds great promise for in vivo functional genomics and interventions against a wide spectrum of diseases, especially those with "undruggable" therapeutic targets. Despite the enormous potential of RNAi for medicine, development of in vivo applications has met with significant problems, particularly in terms of delivery. For effective gene silencing to occur, silencing RNA must reach the cytoplasm of the target cell. Consequently, various strategies using chemically modified siRNA, liposomes, nanoparticles and viral vectors are being developed to deliver silencing RNA. These approaches, however, can be expensive and in many cases they lack target cell specificity or clinical compatibility. Recently, we have shown that RNAi can be activated in vitro and in vivo by non-pathogenic bacteria engineered to manufacture and deliver silencing shRNA to target cells. This new approach, termed TransKingdom RNAi (tkRNAi), has several key advantages. First, tkRNAi may provide a viable means to accomplish therapeutic RNAi since non-pathogenic bacteria have a proven safety record in clinical applications. Second, tkRNAi eliminates the cost of siRNA manufacture since silencing shRNA are produced inside bacteria. Moreover, the intracellular mechanism of shRNA release inherent to tkRNAi may circumvent, or mitigate, the activation of host immune responses. Finally, tkRNAi may facilitate high-throughput in vivo functional genomics screening since bacteria-based RNAi libraries can be easily constructed, stored, reproduced and amplified, thereby allowing for the creation of a stable gene silencing system.
Collapse
Affiliation(s)
- Andrew C Keates
- Skip Ackerman Center for Molecular Therapeutics, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
42
|
Taniguchi S, Fujimori M, Sasaki T, Tsutsui H, Shimatani Y, Seki K, Amano J. Targeting solid tumors with non-pathogenic obligate anaerobic bacteria. Cancer Sci 2010; 101:1925-32. [PMID: 20579076 PMCID: PMC11158574 DOI: 10.1111/j.1349-7006.2010.01628.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecular-targeting drugs with fewer severe adverse effects are attracting great attention as the next wave of cancer treatment. There exist, however, populations of cancer cells resistant to these drugs that stem from the instability of tumor cells and/or the existence of cancer stem cells, and thus specific toxicity is required to destroy them. If such selectivity is not available, these targets may be sought out not by the cancer cell types themselves, but rather in their adjacent cancer microenvironments by means of hypoxia, low pH, and so on. The anaerobic conditions present in malignant tumor tissues have previously been regarded as a source of resistance in cancer cells against conventional therapy. However, there now appears to be a way to make use of these limiting factors as a selective target. In this review, we will refer to several trials, including our own, to direct attention to the utilizable anaerobic conditions present in malignant tumor tissues and the use of bacteria as carriers to target them. Specifically, we have been developing a method to attack solid cancers using the non-pathogenic obligate anaerobic bacterium Bifidobacterium longum as a vehicle to selectively recognize and target the anaerobic conditions in solid cancer tissues. We will also discuss the existence of low oxygen pressure in tumor masses in spite of generally enhanced angiogenesis, overview current cancer therapies, especially the history and present situation of bacterial utility to treat solid tumors, and discuss the rationality and future possibilities of this novel mode of cancer treatment.
Collapse
Affiliation(s)
- Shun'ichiro Taniguchi
- Department of Molecular Oncology, Institute on Aging and Adaptation, Shinshu University Graduate School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Park SJ, Bae H, Kim J, Lim B, Park J, Park S. Motility enhancement of bacteria actuated microstructures using selective bacteria adhesion. LAB ON A CHIP 2010; 10:1706-1711. [PMID: 20422075 DOI: 10.1039/c000463d] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Microrobots developed by the technological advances are useful for application in various fields. Nevertheless, they have limitations with respect to their actuator and motility. Our experiments aim to determine whether a bioactuator using the flagellated bacteria Serratia marcescens would enhance the motility of microrobots. In this study, we investigate that the flagellated bacteria Serratia marcescens could be utilized as actuators for SU-8 microstructures by bovine serum albumin-selective patterning. Firstly, we analyze the adherence of the bacteria to the SU-8 micro cube by selective patterning using 5% BSA. The results show that number of attached-bacteria in the uncoated side of the selectively- coated micro cube with BSA increased by 200% compared with that in all sides of the non treated micro cube. Secondly, the selectively BSA coated micro cube had 210% higher motility than the uncoated micro cube. The results revealed that the bacteria patterned to a specific site using 5% BSA significantly increase the motility of the bacteria actuated microstructure.
Collapse
Affiliation(s)
- Sung Jun Park
- School of Mechanical Systems Engineering, Chonnam National University, Gwangju 500-757, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Sottnik JL, U'Ren LW, Thamm DH, Withrow SJ, Dow SW. Chronic bacterial osteomyelitis suppression of tumor growth requires innate immune responses. Cancer Immunol Immunother 2010; 59:367-78. [PMID: 19701748 PMCID: PMC11030164 DOI: 10.1007/s00262-009-0755-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 08/04/2009] [Indexed: 01/01/2023]
Abstract
Clinical studies over the past several years have reported that metastasis-free survival times in humans and dogs with osteosarcoma are significantly increased in patients that develop chronic bacterial osteomyelitis at their surgical site. However, the immunological mechanism by which osteomyelitis may suppress tumor growth has not been investigated. Therefore, we used a mouse model of osteomyelitis to assess the effects of bone infection on innate immunity and tumor growth. A chronic Staphylococcal osteomyelitis model was established in C3H mice and the effects of infection on tumor growth of syngeneic DLM8 osteosarcoma were assessed. The effects of infection on tumor angiogenesis and innate immunity, including NK cell and monocyte responses, were assessed. We found that osteomyelitis significantly inhibited the growth of tumors in mice, and that the effect was independent of the infecting bacterial type, tumor type, or mouse strain. Depletion of NK cells or monocytes reversed the antitumor activity elicited by infection. Moreover, infected mice had a significant increase in circulating monocytes and numbers of tumor associated macrophages. Infection suppressed tumor angiogenesis but did not affect the numbers of circulating endothelial cells. Therefore, we concluded that chronic localized bacterial infection could elicit significant systemic antitumor activity dependent on NK cells and macrophages.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Clinical Sciences, Animal Cancer Center, Colorado State University, Ft. Collins, 80523, USA.
| | | | | | | | | |
Collapse
|
45
|
Min JJ, Nguyen VH, Gambhir SS. Molecular imaging of biological gene delivery vehicles for targeted cancer therapy: beyond viral vectors. Nucl Med Mol Imaging 2010; 44:15-24. [PMID: 24899933 DOI: 10.1007/s13139-009-0006-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 11/29/2009] [Accepted: 11/30/2009] [Indexed: 12/17/2022] Open
Abstract
Cancer persists as one of the most devastating diseases in the world. Problems including metastasis and tumor resistance to chemotherapy and radiotherapy have seriously limited the therapeutic effects of present clinical treatments. To overcome these limitations, cancer gene therapy has been developed over the last two decades for a broad spectrum of applications, from gene replacement and knockdown to vaccination, each with different requirements for gene delivery. So far, a number of genes and delivery vectors have been investigated, and significant progress has been made with several gene therapy modalities in clinical trials. Viral vectors and synthetic liposomes have emerged as the vehicles of choice for many applications. However, both have limitations and risks that restrict gene therapy applications, including the complexity of production, limited packaging capacity, and unfavorable immunological features. While continuing to improve these vectors, it is important to investigate other options, particularly nonviral biological agents such as bacteria, bacteriophages, and bacteria-like particles. Recently, many molecular imaging techniques for safe, repeated, and high-resolution in vivo imaging of gene expression have been employed to assess vector-mediated gene expression in living subjects. In this review, molecular imaging techniques for monitoring biological gene delivery vehicles are described, and the specific use of these methods at different steps is illustrated. Linking molecular imaging to gene therapy will eventually help to develop novel gene delivery vehicles for preclinical study and support the development of future human applications.
Collapse
Affiliation(s)
- Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School, 5 Hak1 dong, Dong-gu, Gwangju, 501-746 Republic of Korea
| | - Vu H Nguyen
- Department of Nuclear Medicine, Chonnam National University Medical School, 5 Hak1 dong, Dong-gu, Gwangju, 501-746 Republic of Korea
| | - Sanjiv S Gambhir
- Molecular Imaging Program, Department of Radiology & Bioengineering, Bio-X Program, Stanford University, Stanford, CA 94305 USA
| |
Collapse
|
46
|
Abstract
The tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a potent inducer of tumor cell apoptosis, but concerns of considerable liver toxicity limit its uses in human cancer therapy. Here, we show that i.v. injected Escherichia coli DH5α (E. coli DH5α) specifically replicates in solid tumors and metastases in live animals. E. coli DH5α does not enter tumor cells and suits for being the vector for soluble TRAIL (sTRAIL), which induces apoptosis by activating cell-surface death receptors. With the high ‘tumor-targeting' nature, we demonstrate that intratumoral (i.t.) and intravenous injection of sTRAIL-expressing E. coli DH5α results in the tumor-targeted release of biologically active molecules, which leads to a dramatic reduction in the tumor growth rate and the prolonged survival of tumor-bearing mice. TRAIL delivery by E. coli DH5α did not cause any detectable toxicity to any organs, suggesting that E. coli DH5α-delivered sTRAIL protein therapy may provide a feasible and effective form of treatment for solid tumors.
Collapse
|
47
|
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
|
48
|
Malathi M, Mohan PS, Butcher RJ, Venil CK. Benzimidazole quinoline derivatives — An effective green fluorescent dye for bacterial imaging. CAN J CHEM 2009. [DOI: 10.1139/v09-139] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A one-pot synthesis of benzimidazoles by condensing naphthyl or quinoline aldehyde with benzene-1,2-diamine has been reported. IR, 1H and 13C NMR, mass spectral, and CHN analyses were used to elucidate the structures of the products. The molecular structural correlation in the optical properties of the quinoline and naphthalene benzimidazoles was explored. The fluorescence quantum yield ([Formula: see text]) and time-resolved fluorescent lifetime of the quinoline benzimidazoles derivatives were estimated. The influence of solvent polarity and pH on the optical property of quinoline derivatives was illustrated. To explore the bioanalytical applicability, the thermal stability by TG–DTA analysis and the cytogenetic analysis of 3-(1H-benzoimidazol-2-yl)-2-chloro-8-methyl-quinoline (1b) compound were carried out. The fluorescent staining ability of 1b was analyzed and also compared with the normal Gram staining in the bacterium.
Collapse
Affiliation(s)
- Mahalingam Malathi
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
- Department of Chemistry, Howard University, WA, USA
- Department of Environmental Science, Division of Microbiology, Bharathiar University, Coimbatore 641 046, India
| | - Palathurai Subramaniam Mohan
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
- Department of Chemistry, Howard University, WA, USA
- Department of Environmental Science, Division of Microbiology, Bharathiar University, Coimbatore 641 046, India
| | - Raymond J. Butcher
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
- Department of Chemistry, Howard University, WA, USA
- Department of Environmental Science, Division of Microbiology, Bharathiar University, Coimbatore 641 046, India
| | - Chidambaram Kulandaisamy Venil
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
- Department of Chemistry, Howard University, WA, USA
- Department of Environmental Science, Division of Microbiology, Bharathiar University, Coimbatore 641 046, India
| |
Collapse
|
49
|
Aigner A. Transkingdom RNA interference (tkRNAi) as a new delivery tool for therapeutic RNA. Expert Opin Biol Ther 2009; 9:1533-42. [DOI: 10.1517/14712590903307354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Brennan DF, Barford D. Eliminylation: a post-translational modification catalyzed by phosphothreonine lyases. Trends Biochem Sci 2009; 34:108-14. [DOI: 10.1016/j.tibs.2008.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 11/09/2008] [Accepted: 11/12/2008] [Indexed: 12/23/2022]
|