1
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
2
|
Srivastava R, Sarkar K, Bonnerjee D, Bagh S. Synthetic Genetic Reversible Feynman Gate in a Single E. coli Cell and Its Application in Bacterial to Mammalian Cell Information Transfer. ACS Synth Biol 2022; 11:1040-1048. [PMID: 35179369 DOI: 10.1021/acssynbio.1c00392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reversible computing is a nonconventional form of computing where the inputs and outputs are mapped in a unique one-to-one fashion. Reversible logic gates in single living cells have not been demonstrated. Here, we constructed a synthetic genetic reversible Feynman gate in single E. coli cells, and the input-output relations were measured in a clonal population. The inputs were extracellular chemicals, isopropyl β-d-1-thiogalactopyranoside (IPTG), and anhydrotetracycline (aTc), and the outputs were two fluorescence proteins. We developed a simple mathematical model and simulation to capture the essential features of the circuit and experimentally demonstrated that the behavior of the circuit was ultrasensitive and predictive. We showed an application by creating an intercellular Feynman gate, where input information from bacteria was computed and transferred to HeLa cells through shRNAs delivery and the output signals were observed as silencing of native AKT1 and CTNNB1 genes. The introduction of reversible logics in synthetic biology is new, and given that one-to-one input-output mapping, such reversible genetic systems might have applications in sensing, diagnostics, cellular computing, and synthetic biology.
Collapse
Affiliation(s)
- Rajkamal Srivastava
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Homi Bhabha National Institute (HBNI), Block A/F, Sector-I, Bidhannagar, Kolkata 700064, India
| | - Kathakali Sarkar
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Homi Bhabha National Institute (HBNI), Block A/F, Sector-I, Bidhannagar, Kolkata 700064, India
| | - Deepro Bonnerjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Homi Bhabha National Institute (HBNI), Block A/F, Sector-I, Bidhannagar, Kolkata 700064, India
| | - Sangram Bagh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Homi Bhabha National Institute (HBNI), Block A/F, Sector-I, Bidhannagar, Kolkata 700064, India
| |
Collapse
|
3
|
Bacteria and bacterial derivatives as delivery carriers for immunotherapy. Adv Drug Deliv Rev 2022; 181:114085. [PMID: 34933064 DOI: 10.1016/j.addr.2021.114085] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
There is growing interest in the role of microorganisms in human health and disease, with evidence showing that new types of biotherapy using engineered bacterial therapeutics, including bacterial derivatives, can address specific mechanisms of disease. The complex interactions between microorganisms and metabolic/immunologic pathways underlie many diseases with unmet medical needs, suggesting that targeting these interactions may improve patient treatment. Using tools from synthetic biology and chemical engineering, non-pathogenic bacteria or bacterial products can be programmed and designed to sense and respond to environmental signals to deliver therapeutic effectors. This review describes current progress in biotherapy using live bacteria and their derivatives to achieve therapeutic benefits against various diseases.
Collapse
|
4
|
Allemailem KS. Innovative Approaches of Engineering Tumor-Targeting Bacteria with Different Therapeutic Payloads to Fight Cancer: A Smart Strategy of Disease Management. Int J Nanomedicine 2021; 16:8159-8184. [PMID: 34938075 PMCID: PMC8687692 DOI: 10.2147/ijn.s338272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Conventional therapies for cancer eradication like surgery, radiotherapy, and chemotherapy, even though most widely used, still suffer from some disappointing outcomes. The limitations of these therapies during cancer recurrence and metastasis demonstrate the need for better alternatives. Some bacteria preferentially colonize and proliferate inside tumor mass; thus these bacteria can be used as ideal candidates to deliver antitumor therapeutic agents. The bacteria like Bacillus spp., Clostridium spp., E. coli, Listeria spp., and Salmonella spp. can be reprogrammed to produce, transport, and deliver anticancer agents, eg, cytotoxic agents, prodrug converting enzymes, immunomodulators, tumor stroma targeting agents, siRNA, and drug-loaded nanoformulations based on clinical requirements. In addition, these bacteria can be genetically modified to express various functional proteins and targeting ligands that can enhance the targeting approach and controlled drug-delivery. Low tumor-targeting and weak penetration power deep inside the tumor mass limits the use of anticancer drug-nanoformulations. By using anticancer drug nanoformulations and other therapeutic payloads in combination with antitumor bacteria, it makes a synergistic effect against cancer by overcoming the individual limitations. The tumor-targeting bacteria can be either used as a monotherapy or in addition with other anticancer therapies like photothermal therapy, photodynamic therapy, and magnetic field therapy to accomplish better clinical outcomes. The toxicity issues on normal tissues is the main concern regarding the use of engineered antitumor bacteria, which requires deeper research. In this article, the mechanism by which bacteria sense tumor microenvironment, role of some anticancer agents, and the recent advancement of engineering bacteria with different therapeutic payloads to combat cancers has been reviewed. In addition, future prospective and some clinical trials are also discussed.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
5
|
Mughal MJ, Kwok HF. Multidimensional role of bacteria in cancer: Mechanisms insight, diagnostic, preventive and therapeutic potential. Semin Cancer Biol 2021; 86:1026-1044. [PMID: 34119644 DOI: 10.1016/j.semcancer.2021.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The active role of bacteria in oncogenesis has long been a topic of debate. Although, it was speculated to be a transmissible cause of cancer as early as the 16th-century, yet the idea about the direct involvement of bacteria in cancer development has only been explored in recent decades. More recently, several studies have uncovered the mechanisms behind the carcinogenic potential of bacteria which are inflammation, immune evasion, pro-carcinogenic metabolite production, DNA damage and genomic instability. On the other side, the recent development on the understanding of tumor microenvironment and technological advancements has turned this enemy into an ally. Studies using bacteria for cancer treatment and detection have shown noticeable effects. Therapeutic abilities of bioengineered live bacteria such as high specificity, selective cytotoxicity to cancer cells, responsiveness to external signals and control after ingestion have helped to overcome the challenges faced by conventional cancer therapies and highlighted the bacterial based therapy as an ideal approach for cancer treatment. In this review, we have made an effort to compile substantial evidence to support the multidimensional role of bacteria in cancer. We have discussed the multifaceted role of bacteria in cancer by highlighting the wide impact of bacteria on different cancer types, their mechanisms of actions in inducing carcinogenicity, followed by the diagnostic and therapeutic potential of bacteria in cancers. Moreover, we have also highlighted the existing gaps in the knowledge of the association between bacteria and cancer as well as the limitation and advantage of bacteria-based therapies in cancer. A better understanding of these multidimensional roles of bacteria in cancer can open up the new doorways to develop early detection strategies, prevent cancer, and develop therapeutic tactics to cure this devastating disease.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau.
| |
Collapse
|
6
|
Sultana A, Tiash S. Improved DNA delivery using invasive E. coli DH10B in human cells by modified bactofection method. J Control Release 2021; 332:233-244. [PMID: 33561481 DOI: 10.1016/j.jconrel.2021.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/16/2021] [Accepted: 02/02/2021] [Indexed: 11/15/2022]
Abstract
E. coli mediated gene delivery faces a major drawback of low efficiency despite of being a safer alternative to viral vectors. This study showed a novel, simple and effective strategy to enhance invasive E. coli DH10B vector's efficiency in human epithelial cells. The bactofection efficiency of invasive E .coli vector was analyzed in nine cell lines. It demonstrated highest (16%) reporter gene (GFP) expression in cervical cells. Methods were employed to further enhance its efficiency by adding transfection reagents (trans-bactofection method) to promote entry into host cells, lysosomotropic reagents for escape from lysosomal degradation or antibiotics to lyse internalized bacteria. Increased bacterial entry, as elucidated from nil to 3% expression in liver cells, was obtained upon complexing bacteria with PULSin. Chloroquine mediated endosomal escape resulted in 7.2 folds increase whereas tetracycline addition to lyse internalized bacteria caused ≈90% of GFP in HeLa. Eventually, the combined effect of these three methods exhibited close to 100% GFP in cervical and remarkable increase of 138 folds in breast cells. This is the first study showing comparative study of vector's gene delivery ability in various epithelial cells of the human body with improving its delivery efficiency. These data demonstrated the potential of developed bactofection method to boost up the efficiency of other bacterial vectors also, which could further be used for effectual therapeutic gene delivery in human cells.
Collapse
Affiliation(s)
- Alviya Sultana
- School of Science, Monash University, Bandar Sunway, Malaysia.
| | - Snigdha Tiash
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia.
| |
Collapse
|
7
|
Min JJ, Thi-Quynh Duong M, Ramar T, You SH, Kang SR. Theranostic Approaches Using Live Bacteria. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00056-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
8
|
Bacteria-cancer interactions: bacteria-based cancer therapy. Exp Mol Med 2019; 51:1-15. [PMID: 31827064 PMCID: PMC6906302 DOI: 10.1038/s12276-019-0297-0] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer therapeutics, such as targeted therapy and immunotherapy, have raised the hope for cures for many cancer types. However, there are still ongoing challenges to the pursuit of novel therapeutic approaches, including high toxicity to normal tissue and cells, difficulties in treating deep tumor tissue, and the possibility of drug resistance in tumor cells. The use of live tumor-targeting bacteria provides a unique therapeutic option that meets these challenges. Compared with most other therapeutics, tumor-targeting bacteria have versatile capabilities for suppressing cancer. Bacteria preferentially accumulate and proliferate within tumors, where they can initiate antitumor immune responses. Bacteria can be further programmed via simple genetic manipulation or sophisticated synthetic bioengineering to produce and deliver anticancer agents based on clinical needs. Therapeutic approaches using live tumor-targeting bacteria can be applied either as a monotherapy or in combination with other anticancer therapies to achieve better clinical outcomes. In this review, we introduce and summarize the potential benefits and challenges of this anticancer approach. We further discuss how live bacteria interact with tumor microenvironments to induce tumor regression. We also provide examples of different methods for engineering bacteria to improve efficacy and safety. Finally, we introduce past and ongoing clinical trials involving tumor-targeting bacteria. Live tumor-targeting bacteria can selectively induce cancer regression and, with the help of genetic engineering, be made safe and effective vehicles for delivering drugs to tumor cells. In a review article, Jung-Joon Min and colleagues from Chonnam National University Medical School in Hwasun, South Korea, discuss the clinical history of using natural or engineered bacterial strains to suppress cancer growth. Because bacteria such as Salmonella and Listeria preferentially home in on tumors or their surrounding microenvironments, researchers have harnessed these microbial agents to attack cancer cells without causing collateral damage to normal tissues. Bioengineers have also armed bacteria with stronger tumor-sensing and more targeted drug delivery capabilities, and improved control of off-target toxicities. An increasing number of therapeutic bacterial strains are now entering clinical testing, promising to enhance the efficacy of more conventional anticancer treatments.
Collapse
|
9
|
Zhang Z, Luo Y, Zhang Y, Guo K. Enhanced protective immune response to PCV2 adenovirus vaccine by fusion expression of Cap protein with InvC in pigs. J Vet Sci 2019; 20:e35. [PMID: 31364320 PMCID: PMC6669209 DOI: 10.4142/jvs.2019.20.e35] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/19/2019] [Accepted: 05/20/2019] [Indexed: 01/21/2023] Open
Abstract
The major immunogenic protein capsid (Cap) of porcine circovirus type 2 (PCV2) is critical to induce neutralizing antibodies and protective immune response against PCV2 infection. This study was conducted to investigate the immune response of recombinant adenovirus expressing PCV2b Cap and C-terminal domain of Yersinia pseudotuberculosis invasin (Cap-InvC) fusion protein in pigs. The recombinant adenovirus rAd-Cap-InvC, rAd-Cap and rAd were generated and used to immunize pigs. The phosphate-buffered saline was used as negative control. The specific antibodies levels in rAd-Cap-InvC and ZJ/C-strain vaccine groups were higher than that of rAd-Cap group (p < 0.05), and the neutralization antibody titer in rAd-Cap-InvC group was significantly higher than those of other groups during 21–42 days post-immunization (DPI). Moreover, lymphocyte proliferative level, interferon-γ and interleukin-13 levels in rAd-Cap-InvC group were increased compared to rAd-Cap group (p < 0.05). After virulent challenge, viruses were not detected from the blood samples in rAd-Cap-InvC and ZJ/C-strain vaccine groups after 49 DPI. And the respiratory symptom, rectal temperature, lung lesion and lymph node lesion were minimal and similar in the ZJ/C-strain and rAd-Cap-InVC groups. In conclusion, our results demonstrated that rAd-Cap-InvC was more efficiently to stimulate the production of antibody and protect pigs from PCV2 infection. We inferred that InvC is a good candidate gene for further development and application of PCV2 genetic engineering vaccine.
Collapse
Affiliation(s)
- Zhencang Zhang
- Department of Animal Engineering, Yangling Vocational and Technical College, Yangling 712100, China.,College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yan Luo
- Department of Animal Engineering, Yangling Vocational and Technical College, Yangling 712100, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
10
|
Abstract
Many options now exist for constructing oral vaccines which, in experimental systems, have shown themselves to be able to generate highly effective immunity against infectious diseases. Their suitability for implementation in clinical practice, however, for prevention of outbreaks, particularly in low- and middle-income countries (LMIC), is not always guaranteed, because of factors such as cost, logistics and cultural and environmental conditions. This brief overview provides a summary of the various approaches which can be adopted, and evaluates them from a pharmaceutical point, taking into account potential regulatory issues, expense, manufacturing complexity, etc., all of which can determine whether a vaccine approach will be successful in the late stages of development. Attention is also drawn to problems arising from inadequate diet, which impacts upon success in stimulating effective immunity, and identifies the use of lipid-based carriers as a way to counteract the problem of nutritional deficiencies in vaccination campaigns.
Collapse
Affiliation(s)
- R. R. C. New
- Middlesex UniversityHendon, LondonUK
- Vaxcine (UK) Limited, London Bioscience Innovation CentreLondonUK
| |
Collapse
|
11
|
Caballero I, Riou M, Hacquin O, Chevaleyre C, Barc C, Pezant J, Pinard A, Fassy J, Rezzonico R, Mari B, Heuzé-Vourc'h N, Pitard B, Vassaux G. Tetrafunctional Block Copolymers Promote Lung Gene Transfer in Newborn Piglets. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:186-193. [PMID: 30897407 PMCID: PMC6426709 DOI: 10.1016/j.omtn.2019.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022]
Abstract
Tetrafunctional block copolymers are molecules capable of complexing DNA. Although ineffective in vitro, studies in mice have shown that the tetrafunctional block copolymer 704 is a more efficient lung gene transfer agent than the cationic liposome GL67A, previously used in a phase II clinical trial in cystic fibrosis patients. In the present study, we compared the gene transfer capacity of the 704-DNA formulation and a cationic liposome-DNA formulation equivalent to GL67A in a larger-animal model, the newborn piglet. Our results indicate an efficacy of the 704-DNA formulation well above one order of magnitude higher than that of the cationic liposome-DNA formulation, with no elevated levels of interleukin-6 (IL-6), taken as a marker of inflammation. Transgene expression was heterogeneous within lung lobes, with expression levels that were below the detection threshold in some samples, while high in other samples. This heterogeneity is likely to be due to the bolus injection procedure as well as to the small volume of injection. The present study highlights the potential of tetrafunctional block copolymers as non-viral vectors for lung gene therapy.
Collapse
Affiliation(s)
- Ignacio Caballero
- INRA Centre Val de Loire - Université de Tours, UMR-1282 Infectiologie et Santé Publique (ISP), 37380 Nouzilly, France
| | - Mickaël Riou
- INRA Centre Val de Loire, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), 37380 Nouzilly, France
| | - Océane Hacquin
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France; FHU-OncoAge, Nice, France
| | - Claire Chevaleyre
- INRA Centre Val de Loire - Université de Tours, UMR-1282 Infectiologie et Santé Publique (ISP), 37380 Nouzilly, France
| | - Céline Barc
- INRA Centre Val de Loire, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), 37380 Nouzilly, France
| | - Jérémy Pezant
- INRA Centre Val de Loire, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), 37380 Nouzilly, France
| | - Anne Pinard
- INRA Centre Val de Loire, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), 37380 Nouzilly, France
| | - Julien Fassy
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France; FHU-OncoAge, Nice, France
| | - Roger Rezzonico
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France; FHU-OncoAge, Nice, France
| | - Bernard Mari
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France; FHU-OncoAge, Nice, France
| | | | - Bruno Pitard
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Georges Vassaux
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France; FHU-OncoAge, Nice, France.
| |
Collapse
|
12
|
Deng L, Roose K, Job ER, De Rycke R, Van Hamme E, Gonçalves A, Parthoens E, Cicchelero L, Sanders N, Fiers W, Saelens X. Oral delivery of Escherichia coli persistently infected with M2e-displaying bacteriophages partially protects against influenza A virus. J Control Release 2017; 264:55-65. [PMID: 28842314 DOI: 10.1016/j.jconrel.2017.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 01/22/2023]
Abstract
We describe a novel live oral vaccine type. Conceptually, this vaccine is based on a non-lytic, recombinant filamentous bacteriophage that displays an antigen of interest. To provide proof of concept we used the amino-terminal part of a conserved influenza A virus epitope, i.e. matrix protein 2 ectodomain (M2e) residues 2 to 16, as the antigen of interest. Rather than using the phages as purified virus-like particles as a vaccine, these phages were delivered to intestinal Peyer's patches as a live bacterium-phage combination that comprises Escherichia coli cells that conditionally express invasin derived from Yersinia pseudotuberculosis. Invasin-expressing E. coli cells were internalized by mammalian Hep-2 cells in vitro and adhered to mouse intestinal microfold (M) cells ex vivo. Invasin-expressing E. coli cells were permissive for recombinant filamentous bacteriophage f88 that displays M2e and became persistently infected. Oral administration of the live engineered E. coli-invasin-phage combination to mice induced M2e-specific serum IgG antibodies. Mice that had been immunized with invasin-expressing E. coli cells that carried M2e2-16 displaying fd phages seroconverted to M2e and showed partial protection against challenge with influenza A virus. Oral delivery of a live vaccine comprising a bacterial host that is targeted to Peyer's patches and is persistently infected with an antigen-displaying phage, can thus be exploited as an oral vaccine.
Collapse
Affiliation(s)
- Lei Deng
- VIB-UGent Center for Medical Biotechnology, VIB, Technologiepark, 927, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium
| | - Kenny Roose
- VIB-UGent Center for Medical Biotechnology, VIB, Technologiepark, 927, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium
| | - Emma R Job
- VIB-UGent Center for Medical Biotechnology, VIB, Technologiepark, 927, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium; Inflammation Research Center, VIB, Technologiepark 927, Ghent, Belgium
| | - Evelien Van Hamme
- Inflammation Research Center, VIB, Technologiepark 927, Ghent, Belgium
| | - Amanda Gonçalves
- Inflammation Research Center, VIB, Technologiepark 927, Ghent, Belgium
| | - Eef Parthoens
- Inflammation Research Center, VIB, Technologiepark 927, Ghent, Belgium
| | - Laetitia Cicchelero
- Laboratory of Gene Therapy, Faculty of Veterinary Sciences, Ghent University, Merelbeke, Belgium
| | - Niek Sanders
- Laboratory of Gene Therapy, Faculty of Veterinary Sciences, Ghent University, Merelbeke, Belgium
| | - Walter Fiers
- VIB-UGent Center for Medical Biotechnology, VIB, Technologiepark, 927, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium.
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, Technologiepark, 927, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent, Belgium.
| |
Collapse
|
13
|
Vela Ramirez JE, Sharpe LA, Peppas NA. Current state and challenges in developing oral vaccines. Adv Drug Deliv Rev 2017; 114:116-131. [PMID: 28438674 PMCID: PMC6132247 DOI: 10.1016/j.addr.2017.04.008] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
While vaccination remains the most cost effective strategy for disease prevention, communicable diseases persist as the second leading cause of death worldwide. There is a need to design safe, novel vaccine delivery methods to protect against unaddressed and emerging diseases. Development of vaccines administered orally is preferable to traditional injection-based formulations for numerous reasons including improved safety and compliance, and easier manufacturing and administration. Additionally, the oral route enables stimulation of humoral and cellular immune responses at both systemic and mucosal sites to establish broader and long-lasting protection. However, oral delivery is challenging, requiring formulations to overcome the harsh gastrointestinal (GI) environment and avoid tolerance induction to achieve effective protection. Here we address the rationale for oral vaccines, including key biological and physicochemical considerations for next-generation oral vaccine design.
Collapse
Affiliation(s)
- Julia E Vela Ramirez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Lindsey A Sharpe
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA; McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
14
|
Khan TA, Wang X, Maynard JA. Inclusion of an RGD Motif Alters Invasin Integrin-Binding Affinity and Specificity. Biochemistry 2016; 55:2078-90. [DOI: 10.1021/acs.biochem.5b01243] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tarik A. Khan
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Xianzhe Wang
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer A. Maynard
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
15
|
Kenngott EE, Kiefer R, Schneider-Daum N, Hamann A, Schneider M, Schmitt MJ, Breinig F. Surface-modified yeast cells: A novel eukaryotic carrier for oral application. J Control Release 2016; 224:1-7. [DOI: 10.1016/j.jconrel.2015.12.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/11/2022]
|
16
|
Li H, Ning P, Lin Z, Liang W, Kang K, He L, Zhang Y. Co-expression of the C-terminal domain of Yersinia enterocolitica invasin enhances the efficacy of classical swine-fever-vectored vaccine based on human adenovirus. J Biosci 2015; 40:79-90. [PMID: 25740144 DOI: 10.1007/s12038-014-9495-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of adenovirus vector-based vaccines is a promising approach for generating antigen-specific immune responses. Improving vaccine potency is necessary in other approaches to address their inadequate protection for the majority of infectious diseases. This study is the first to reconstruct a recombinant replication-defective human adenovirus co-expressing E2 and invasin C-terminal (InvC) glycoproteins (rAd-E2-InvC). rAd-E2-InvC with 2 x 10(6) TCID50 was intramuscularly administered two times to CSFV-free pigs at 14 day intervals. No adverse clinical reactions were observed in any of the pigs after the vaccination. The CSFV E2-specific antibody titer was significantly higher in the rAd-E2-InvC group than that in the rAdV-E2 group as measured by NPLA and blocking ELISA. Pigs immunized with rAd-E2-InvC were completely protected against lethal challenge. Neither CSFV RNA nor pathological changes were detected in the tissues after CSFV challenge. These results demonstrate that rAd-E2-InvC could be an alternative to the existing CSF vaccine. Moreover, InvC that acts as an adjuvant could enhance the immunogenicity of rAdV-E2 and induce high CSFV E2-specific antibody titer and protection level.
Collapse
Affiliation(s)
- Helin Li
- College of Veterinary Medicine, Northwest A and F University, Yangling 712100, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Lee TJ, Wong J, Bae S, Lee AJ, Lopatkin A, Yuan F, You L. A power-law dependence of bacterial invasion on mammalian host receptors. PLoS Comput Biol 2015; 11:e1004203. [PMID: 25879937 PMCID: PMC4399907 DOI: 10.1371/journal.pcbi.1004203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/19/2015] [Indexed: 01/04/2023] Open
Abstract
Pathogenic bacteria such as Listeria and Yersinia gain initial entry by binding to host target cells and stimulating their internalization. Bacterial uptake entails successive, increasingly strong associations between receptors on the surface of bacteria and hosts. Even with genetically identical cells grown in the same environment, there are vast differences in the number of bacteria entering any given cell. To gain insight into this variability, we examined uptake dynamics of Escherichia coli engineered to express the invasin surface receptor from Yersinia, which enables uptake via mammalian host β1-integrins. Surprisingly, we found that the uptake probability of a single bacterium follows a simple power-law dependence on the concentration of integrins. Furthermore, the value of a power-law parameter depends on the particular host-bacterium pair but not on bacterial concentration. This power-law captures the complex, variable processes underlying bacterial invasion while also enabling differentiation of cell lines. Uptake of bacteria by mammalian cells is highly variable within a population of host cells and between host cell types. A detailed but unwieldy mechanistic model describing individual host-pathogen receptor binding events is captured by a simple power-law dependence on the concentration of the host receptors. The power-law parameters capture characteristics of the host-bacterium pair interaction and can differentiate host cell lines. This study has important implications for understanding the accuracy and precision of therapeutics employing receptor-mediated transport of materials to mammalian hosts.
Collapse
Affiliation(s)
- Tae J. Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Jeffrey Wong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Sena Bae
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Anna Jisu Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Allison Lopatkin
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
- Center for Systems Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
18
|
Spisni E, Valerii MC, De Fazio L, Cavazza E, Borsetti F, Sgromo A, Candela M, Centanni M, Rizello F, Strillacci A. Cyclooxygenase-2 silencing for the treatment of colitis: a combined in vivo strategy based on RNA interference and engineered Escherichia coli. Mol Ther 2014; 23:278-89. [PMID: 25393372 DOI: 10.1038/mt.2014.222] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/09/2014] [Indexed: 02/08/2023] Open
Abstract
Nonpathogenic-invasive Escherichia coli (InvColi) bacteria are suitable for genetic transfer into mammalian cells and may act as a vehicle for RNA Interference (RNAi) in vivo. Cyclooxygenase-2 (COX-2) is overexpressed in ulcerative colitis (UC) and Crohn's disease (CD), two inflammatory conditions of the colon and small intestine grouped as inflammatory bowel disease (IBD). We engineered InvColi strains for anti-COX-2 RNAi (InvColi(shCOX2)), aiming to investigate the in vivo feasibility of a novel COX-2 silencing strategy in a murine model of colitis induced by dextran sulfate sodium (DSS). Enema administrations of InvColi(shCOX2) in DSS-treated mice led to COX-2 downregulation, colonic mucosa preservation, reduced colitis disease activity index (DAI) and increased mice survival. Moreover, DSS/InvColi(shCOX2)-treated mice showed lower levels of circulating pro-inflammatory cytokines and a reduced colitis-associated shift of gut microbiota. Considering its effectiveness and safety, we propose our InvColi(shCOX2) strategy as a promising tool for molecular therapy in intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| | - Maria C Valerii
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| | - Luigia De Fazio
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| | - Elena Cavazza
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| | - Francesca Borsetti
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| | - Annamaria Sgromo
- 1] Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy [2] Department of Biochemistry, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Manuela Centanni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Fernando Rizello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Antonio Strillacci
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Chen Q, Lee CW, Sim EUH, Narayanan K. Induction of Protein Expression Within Escherichia coli Vector for Entry into Mammalian Cells. Hum Gene Ther Methods 2014; 25:40-7. [DOI: 10.1089/hgtb.2012.188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Qingwen Chen
- School of Science, Monash University, Bandar Sunway 46150, Malaysia
| | - Choon-Weng Lee
- Institute of Biological Sciences, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Edmund Ui-Hang Sim
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Sarawak 94300, Malaysia
| | - Kumaran Narayanan
- School of Science, Monash University, Bandar Sunway 46150, Malaysia
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
20
|
Pontes D, Azevedo M, Innocentin S, Blugeon S, Lefévre F, Azevedo V, Miyoshi A, Courtin P, Chapot-Chartier MP, Langella P, Chatel JM. Immune response elicited by DNA vaccination using Lactococcus lactis is modified by the production of surface exposed pathogenic protein. PLoS One 2014; 9:e84509. [PMID: 24465412 PMCID: PMC3897362 DOI: 10.1371/journal.pone.0084509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/21/2013] [Indexed: 01/02/2023] Open
Abstract
In this study, we compared immune responses elicited by DNA immunization using Lactococcus lactis or L. lactis expressing the Staphylococcus aureus invasin Fibronectin Binding Protein A (FnBPA) at its surface. Both strains carried pValac:BLG, a plasmid containing the cDNA of Beta-Lactoglobulin (BLG), and were designated LL-BLG and LL-FnBPA+ BLG respectively. A TH2 immune response characterized by the secretion of IL-4 and IL-5 in medium of BLG reactivated splenocytes was detected after either oral or intranasal administration of LL-FnBPA+ BLG. In contrast, intranasal administration of LL-BLG elicited a TH1 immune response. After BLG sensitization, mice previously intranasally administered with LL-BLG showed a significantly lower concentration of BLG-specific IgE than the mice non-administered. Altenatively administration of LL-FnBPA+ BLG didn't modify the BLG-specific IgE concentration obtained after sensitization, thus confirming the TH2 orientation of the immune response. To determine if the TH2-skewed immune response obtained with LL-FnBpA+ BLG was FnBPA-specific or not, mice received another L. lactis strain producing a mutated form of the Listeria monocytogenes invasin Internalin A intranasally, allowing thus the binding to murine E-cadherin, and containing pValac:BLG (LL-mInlA+ BLG). As with LL-FnBPA+ BLG, LL-mInlA+ BLG was not able to elicit a TH1 immune response. Furthermore, we observed that these difference were not due to the peptidoglycan composition of the cell wall as LL-FnBPA+ BLG, LL-mInlA+ BLG and LL-BLG strains shared a similar composition. DNA vaccination using LL-BLG elicited a pro-inflammatory TH1 immune response while using LL-FnBPA+ BLG or LL-mInlA+ BLG elicited an anti-inflammatory TH2 immune response.
Collapse
MESH Headings
- Adhesins, Bacterial/genetics
- Adhesins, Bacterial/immunology
- Administration, Intranasal
- Administration, Oral
- Animals
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Cell Engineering
- Drug Delivery Systems/methods
- Female
- Immunity, Active
- Immunity, Cellular
- Lactococcus lactis/genetics
- Lactococcus lactis/immunology
- Lactoglobulins/genetics
- Lactoglobulins/immunology
- Listeriosis/immunology
- Listeriosis/prevention & control
- Mice
- Mice, Inbred BALB C
- Plasmids
- Staphylococcal Infections/immunology
- Staphylococcal Infections/prevention & control
- Th1 Cells/cytology
- Th1 Cells/immunology
- Th2 Cells/cytology
- Th2 Cells/immunology
- Vaccination
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/biosynthesis
- Vaccines, DNA/genetics
- Vaccines, Synthetic
Collapse
Affiliation(s)
- Daniela Pontes
- State University of Paraiba, Campus V, Department of Biological Sciences. João Pessoa, Paraíba, Brazil
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
| | - Marcela Azevedo
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Innocentin
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
- Lymphocyte Signaling and Development Laboratory, Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Sébastien Blugeon
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
| | | | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, Minas Gerais, Brazil
| | - Anderson Miyoshi
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, Minas Gerais, Brazil
| | - Pascal Courtin
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
| | - Marie-Pierre Chapot-Chartier
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
| | - Philippe Langella
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
| | - Jean-Marc Chatel
- INRA, UMR1319 Micalis, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR1319 Micalis, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
21
|
Gut adhesive Bacillus subtilis spores as a platform for mucosal delivery of antigens. Infect Immun 2014; 82:1414-23. [PMID: 24421038 DOI: 10.1128/iai.01255-13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacillus subtilis spores have been used as safe and heat-resistant antigen delivery vectors. Nonetheless, the oral administration of spores typically induces weak immune responses to the passenger antigens, which may be attributed to the fast transit through the gastrointestinal tract. To overcome this limitation, we have developed B. subtilis spores capable of binding to the gut epithelium by means of expressing bacterial adhesins on the spore surface. The resulting spores bound to in vitro intestinal cells, showed a longer transit through the mouse intestinal tract, and interacted with Peyer's patch cells. The adhesive spores increased the systemic and secreted antibody responses to the Streptococcus mutans P1 protein, used as a model antigen, following oral, intranasal, and sublingual administration. Additionally, P1-specific antibodies efficiently inhibited the adhesion of the oral pathogen Streptococcus mutans to abiotic surfaces. These results support the use of gut-colonizing B. subtilis spores as a new platform for the mucosal delivery of vaccine antigens.
Collapse
|
22
|
Bernardes N, Chakrabarty AM, Fialho AM. Engineering of bacterial strains and their products for cancer therapy. Appl Microbiol Biotechnol 2013; 97:5189-99. [DOI: 10.1007/s00253-013-4926-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/10/2013] [Accepted: 04/10/2013] [Indexed: 01/20/2023]
|
23
|
de Azevedo M, Karczewski J, Lefévre F, Azevedo V, Miyoshi A, Wells JM, Langella P, Chatel JM. In vitro and in vivo characterization of DNA delivery using recombinant Lactococcus lactis expressing a mutated form of L. monocytogenes Internalin A. BMC Microbiol 2012; 12:299. [PMID: 23253484 PMCID: PMC3541092 DOI: 10.1186/1471-2180-12-299] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/14/2012] [Indexed: 12/20/2022] Open
Abstract
Background The use of food-grade Lactic Acid Bacteria (LAB) as DNA delivery vehicles represents an attractive strategy to deliver DNA vaccines at the mucosal surfaces as they are generally regarded as safe (GRAS). We previously showed that either native Lactococcus lactis (LL) or recombinant invasive LL expressing Fibronectin Binding Protein A of Staphylococcus aureus (LL-FnBPA+) or Internalin A of Listeria monocytogenes (LL-InlA+), were able to deliver and trigger DNA expression by epithelial cells, either in vitro or in vivo. InlA does not bind to its receptor, the murine E-cadherin, thus limiting the use of LL-InlA+ in in vivo murine models. Moreover, FnBPA binds to its receptors, integrins, via fibronectin introducing another limiting factor. In order to avoid the limitations of LL-InlA+ and LL-FnBPA+, a new L. lactis strain was engineered to produce a previously described mutated form of InlA (LL-mInlA+) allowing the binding of mInlA on murine E-cadherin. Results After showing the expression of mInLA at the surface of LL-mInlA+ strain, in vitro gentamycin survival assay in Caco-2 cells showed that LL-mInlA+ is 1000 times more invasive than LL. LL-mInlA+ invasivity was also validated by fluorescence microscopy. LL and LL-mInlA+ were transformed with pValacBLG, a plasmid containing the cDNA of bovine β-Lactoglobulin (BLG), resulting in strains LL-BLG and LL-mInlA+BLG. The plasmid transfer in vitro using LL-mInlA+BLG was increased 10 times compared to LL-BLG. Moreover, the number of mice producing BLG in isolated enterocytes after oral administration of LL-mInlA+BLG in vivo was slightly higher than after oral administration of LL-BLG. Conclusions We confirmed in this study that the production of mInlA at the surface of L. lactis is a promising strategy for plasmid transfer in vitro and in vivo.
Collapse
Affiliation(s)
- Marcela de Azevedo
- INRA, UMR1319 Micalis, Commensals and Probiotics-Host Interactions Laboratory, Jouy-en-Josas, France
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Early Treg suppression by a listeriolysin-O-expressing E. coli vaccine in heterologous prime–boost vaccination against cancer. Vaccine 2012; 30:6903-11. [DOI: 10.1016/j.vaccine.2012.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 08/30/2012] [Accepted: 09/02/2012] [Indexed: 11/30/2022]
|
25
|
Surface display of N-terminally anchored invasin by Lactobacillus plantarum activates NF-κB in monocytes. Appl Environ Microbiol 2012; 78:5864-71. [PMID: 22706054 DOI: 10.1128/aem.01227-12] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The probiotic lactic acid bacterium Lactobacillus plantarum is a potential delivery vehicle for mucosal vaccines because of its generally regarded as safe (GRAS) status and ability to persist at the mucosal surfaces of the human intestine. However, the inherent immunogenicity of vaccine antigens is in many cases insufficient to elicit an efficient immune response, implying that additional adjuvants are needed to enhance the antigen immunogenicity. The goal of the present study was to increase the proinflammatory properties of L. plantarum by expressing a long (D1 to D5 [D1-D5]) and a short (D4-D5) version of the extracellular domain of invasin from the human pathogen Yersinia pseudotuberculosis. To display these proteins on the bacterial surface, four different N-terminal anchoring motifs from L. plantarum were used, comprising two different lipoprotein anchors, a transmembrane signal peptide anchor, and a LysM-type anchor. All these anchors mediated surface display of invasin, and several of the engineered strains were potent activators of NF-κB when interacting with monocytes in cell culture. The most distinct NF-κB responses were obtained with constructs in which the complete invasin extracellular domain was fused to a lipoanchor. The proinflammatory L. plantarum strains constructed here represent promising mucosal delivery vehicles for vaccine antigens.
Collapse
|
26
|
Crossing the barrier: Targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release 2012; 160:431-9. [DOI: 10.1016/j.jconrel.2012.02.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/02/2012] [Indexed: 01/09/2023]
|
27
|
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: 111] [Impact Index Per Article: 8.5] [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
|
28
|
Gardlik R, Hodosy J, Palffy R, Behuliak M, Janega P, Celec P. Effects of orally administered bacteria carrying HIF-1α gene in an experimental model of intestinal ischemia. Arch Med Res 2011; 41:332-7. [PMID: 20851289 DOI: 10.1016/j.arcmed.2010.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 07/16/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS Bactofection is delivery of therapeutic genes into target cells using bacteria penetrating the target cell membrane and releasing the gene into the cell. Hypoxia-inducible factor 1α (HIF-1α) represents a potential therapeutic gene to be used for gene delivery in ischemic diseases. The aim of this study was to prove the effects of bacteria-mediated transfer of hypoxia-inducible factor 1α (HIF-1α) in an experimental model of intestinal ischemia in rats. METHODS Male Wistar rats with a surgically induced ischemia of colon (cecum) or sham-operated rats were treated by per os application of E. coli carrying therapeutic genes. After 1 week, samples were taken for measurement of oxidative stress markers and expression analyses. RESULTS According to our observation, there were no signs or symptoms of ongoing ischemia in gastrointestinal tissue. Interestingly, all experimental groups treated by bacteria, regardless of their ability to invade cells or the presence of HIF-1α gene, showed decreased levels of vascular endothelial growth factor (VEGF) compared to control groups. Similarly, all treatment groups showed increased hematocrit. CONCLUSIONS We conclude ineffectiveness of the bacterial gene delivery system. However, the effect of bacteria themselves was obvious. HIF-1 can be activated hypoxia-independently by the action of pathogenic bacteria in the rat intestine. We hypothesize that therapeutic bacterial strain used may compete with siderophore-expressing bacteria present in the gut of rats to force them out and prevent their ability to activate HIF-1 in a hypoxia-independent manner. This phenomenon should be analyzed in detail in further studies.
Collapse
Affiliation(s)
- Roman Gardlik
- Institute of Pathophysiology, Comenius University, Bratislava, Slovakia.
| | | | | | | | | | | |
Collapse
|
29
|
Development of an Escherichia coli Expressing Listeriolysin-O Vaccine Against Wilms Tumor Gene 1-expressing Tumors. J Immunother 2009; 32:845-55. [DOI: 10.1097/cji.0b013e3181aee259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
30
|
Xu M, Dai MS, Mi C. Recombinant E. coli LLO/OVA vaccination effectively inhibits murine melanoma metastasis to lung by CD8+T cells immunity. Chin J Cancer Res 2009. [DOI: 10.1007/s11670-009-0044-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
31
|
Harms JS, Durward MA, Magnani DM, Splitter GA. Evaluation of recombinant invasive, non-pathogenic Eschericia coli as a vaccine vector against the intracellular pathogen, Brucella. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2009; 7:1. [PMID: 19126207 PMCID: PMC2633335 DOI: 10.1186/1476-8518-7-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 01/06/2009] [Indexed: 01/18/2023]
Abstract
Background There is no safe, effective human vaccine against brucellosis. Live attenuated Brucella strains are widely used to vaccinate animals. However these live Brucella vaccines can cause disease and are unsafe for humans. Killed Brucella or subunit vaccines are not effective in eliciting long term protection. In this study, we evaluate an approach using a live, non-pathogenic bacteria (E. coli) genetically engineered to mimic the brucellae pathway of infection and present antigens for an appropriate cytolitic T cell response. Methods E. coli was modified to express invasin of Yersinia and listerialysin O (LLO) of Listeria to impart the necessary infectivity and antigen releasing traits of the intracellular pathogen, Brucella. This modified E. coli was considered our vaccine delivery system and was engineered to express Green Fluorescent Protein (GFP) or Brucella antigens for in vitro and in vivo immunological studies including cytokine profiling and cytotoxicity assays. Results The E. coli vaccine vector was able to infect all cells tested and efficiently deliver therapeutics to the host cell. Using GFP as antigen, we demonstrate that the E. coli vaccine vector elicits a Th1 cytokine profile in both primary and secondary immune responses. Additionally, using this vector to deliver a Brucella antigen, we demonstrate the ability of the E. coli vaccine vector to induce specific Cytotoxic T Lymphocytes (CTLs). Conclusion Protection against most intracellular bacterial pathogens can be obtained mostly through cell mediated immunity. Data presented here suggest modified E. coli can be used as a vaccine vector for delivery of antigens and therapeutics mimicking the infection of the pathogen and inducing cell mediated immunity to that pathogen.
Collapse
Affiliation(s)
- Jerome S Harms
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA.
| | | | | | | |
Collapse
|
32
|
Peerlinck I, Amini-Nik S, Phillips RK, Iggo R, Lemoine NR, Tejpar S, Vassaux G. Therapeutic potential of replication-selective oncolytic adenoviruses on cells from familial and sporadic desmoid tumors. Clin Cancer Res 2008; 14:6187-92. [PMID: 18829497 DOI: 10.1158/1078-0432.ccr-08-0410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Constitutive activation of the Wnt signaling pathway is a hallmark of many cancers and has been associated with familial and sporadic desmoid tumors. The aim of the present study is to assess the therapeutic potential of oncolytic adenoviruses selectively replicating in cells in which the Wnt signaling pathway is active on primary cells from desmoid tumors. EXPERIMENTAL DESIGN Primary cells extracted from familial (n = 3) or sporadic (n = 3) desmoid tumors were cultured short term. Cancer cell survival and viral replication were measured in vitro upon infection with two different oncolytic adenoviruses targeting a constitutive activation of the Wnt signaling pathway. Adenoviral infectivity was also assessed. RESULTS Although cells extracted from one sporadic desmoid tumor responded very well to the oncolytic action of the adenoviruses (<20% of viable cells upon infection at a multiplicity of infection of 10), cells from two tumor samples were totally resistant to the viral action. Cells from the remaining samples showed intermediate sensitivity to the oncolytic viruses. These effects were correlated to the level of infectivity of the cells. Finally, in responder cells, evidences of viral replication was observed. CONCLUSIONS Our experimental data suggest that the response of desmoid tumor cells to oncolytic adenovirus is neither correlated to the type of mutation activating the Wnt signaling pathway nor to the familial or sporadic nature of the tumor. In addition, they highlight the variability of infectivity of individual tumors and predict a great variability in the response to oncolytic adenoviruses.
Collapse
Affiliation(s)
- Inge Peerlinck
- Institute of Cancer and the CR-UK Clinical Centre, Barts and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
Nitcheu-Tefit J, Dai MS, Critchley-Thorne RJ, Ramirez-Jimenez F, Xu M, Conchon S, Ferry N, Stauss HJ, Vassaux G. Listeriolysin O expressed in a bacterial vaccine suppresses CD4+CD25high regulatory T cell function in vivo. THE JOURNAL OF IMMUNOLOGY 2007; 179:1532-41. [PMID: 17641019 DOI: 10.4049/jimmunol.179.3.1532] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4(+)CD25(high) regulatory T cells (Treg) protect the host from autoimmune diseases but are also obstacles against cancer therapies. An ideal cancer vaccine would stimulate specific cytotoxic responses and reduce/suppress Treg function. In this study, we showed that Escherichia coli expressing listeriolysin O and OVA (E. coli LLO/OVA) demonstrated remarkable levels of protection against OVA-expressing tumor cells. By contrast, E. coli expressing OVA only (E. coli OVA) showed poor protection. High-avidity OVA-specific CTL were induced in E. coli LLO/OVA-vaccinated mice, and CD8(+) depletion--but not NK cell depletion, abolished the antitumor activity of the E. coli LLO/OVA vaccine. Phenotypic analysis of T cells following vaccination with either vaccine revealed preferential generation of CD44(high)CD62L(low) CD8(+) effector memory T cells over CD44(high)CD62L(high) central memory T cells. Unexpectedly, CD4(+) depletion turned E. coli OVA into a vaccine as effective as E. coli LLO/OVA suggesting that a subset of CD4(+) cells suppressed the CD8(+) T cell-mediated antitumor response. Further depletion experiments demonstrated that these suppressive cells consisted of CD4(+)CD25(high) regulatory T cells. We therefore assessed these vaccines for Treg function and found that although CD4(+)CD25(high) expansion and Foxp3 expression within this population was similar in all groups of mice, Treg cells from E. coli LLO/OVA-vaccinated animals were unable to suppress conventional T cells proliferation. These findings provide the first evidence that LLO expression affects Treg cell function and may have important implications for enhancing antitumor vaccination strategies in humans.
Collapse
MESH Headings
- Animals
- Bacterial Toxins/administration & dosage
- Bacterial Toxins/biosynthesis
- Bacterial Toxins/genetics
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Adhesion/immunology
- Cell Proliferation
- Egg Proteins/biosynthesis
- Egg Proteins/genetics
- Egg Proteins/immunology
- Escherichia coli Vaccines/administration & dosage
- Escherichia coli Vaccines/genetics
- Escherichia coli Vaccines/immunology
- Female
- Heat-Shock Proteins/administration & dosage
- Heat-Shock Proteins/biosynthesis
- Heat-Shock Proteins/genetics
- Hemolysin Proteins/administration & dosage
- Hemolysin Proteins/biosynthesis
- Hemolysin Proteins/genetics
- Immunologic Memory
- Injections, Intravenous
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Ovalbumin/biosynthesis
- Ovalbumin/genetics
- Ovalbumin/immunology
- Peptide Fragments
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- Tumor Cells, Cultured
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/genetics
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Josianne Nitcheu-Tefit
- Centre for Molecular Oncology, Institute of Cancer, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Autenrieth SE, Autenrieth IB. Yersinia enterocolitica: subversion of adaptive immunity and implications for vaccine development. Int J Med Microbiol 2007; 298:69-77. [PMID: 17702651 DOI: 10.1016/j.ijmm.2007.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Enteric Yersinia spp. invade Peyer's patches, disseminate to lymphoid tissues, and induce mucosal and systemic immune responses. Many virulence factors of Yersinia enterocolitica have been investigated in detail and were found to act on host cells involved in innate and adaptive immunity. Recent work explored as to whether attenuated Y. enterocolitica or recombinant components of Y. enterocolitica can be used as tools for vaccination. We and others have tested whether by means of the type three secretion system in attenuated Y. enterocolitica strains antigens might be delivered to antigen-presenting cells in order to induce CD8 and CD4 T cell responses. Alternatively, recombinant components of Y. enterocolitica such as invasin protein which binds to beta1 integrins of host cells have been tested for their ability to target antigen along with microparticles (fused to invasin) to antigen-presenting cells and to act as adjuvant. The work summarized in this article demonstrates that Y. enterocolitica and its components might be useful tools for novel vaccination strategies; in fact, invasin when fused to antigen and coated to microparticles might induce both CD4 and CD8 T cell responses. Likewise, attenuated Y. enterocolitica live carrier strains were reported to induce both CD8 and some CD4 T cell responses. However, we need to know more about how Y. enterocolitica subverts functions of antigen-presenting cells in order to design mutants with optimized antigen delivery features and deletion in those virulence factor that contribute to subversion of innate or adaptive immune responses.
Collapse
Affiliation(s)
- Stella E Autenrieth
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen, Elfriede-Aulhorn-Strasse 6, D-72076 Tübingen, Germany
| | | |
Collapse
|
35
|
Thorne SH. Strategies to achieve systemic delivery of therapeutic cells and microbes to tumors. Expert Opin Biol Ther 2007; 7:41-51. [PMID: 17150018 DOI: 10.1517/14712598.7.1.41] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In order to more effectively treat cancer, targeted delivery of therapeutic agents will be needed. The creation of delivery vehicles capable of locating and entering tumors before delivering a therapeutic payload will, therefore, enable the design of more beneficial and less toxic treatment platforms. Although nanoparticles, microbubbles and liposomes may also partially address these issues, the use of biological agents as delivery vehicles presently holds much promise. Through the hijacking of natural pathogen or cell trafficking pathways it is possible to actively target such agents to the tumor; they are then capable of selective replication (multiplying their therapeutic potential) and may be directly cytolytic themselves and/or may be utilized to deliver therapeutic genes. These agents, such as oncolytic viruses, attenuated bacteria and eukaryotic cells (cellular immunotherapeutics and progenitor and stem cells) will be discussed along with the mechanisms employed to deliver them systemically to tumors, including disseminated disease and micrometsastases.
Collapse
Affiliation(s)
- Steve H Thorne
- Stanford University, Bio-X Programme and Department of Pediatrics, Clark Center, California 94305, USA.
| |
Collapse
|
36
|
Detmer A, Glenting J. Live bacterial vaccines--a review and identification of potential hazards. Microb Cell Fact 2006; 5:23. [PMID: 16796731 PMCID: PMC1538998 DOI: 10.1186/1475-2859-5-23] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 06/23/2006] [Indexed: 12/20/2022] Open
Abstract
The use of live bacteria to induce an immune response to itself or to a carried vaccine component is an attractive vaccine strategy. Advantages of live bacterial vaccines include their mimicry of a natural infection, intrinsic adjuvant properties and their possibility to be administered orally. Derivatives of pathogenic and non-pathogenic food related bacteria are currently being evaluated as live vaccines. However, pathogenic bacteria demands for attenuation to weaken its virulence. The use of bacteria as vaccine delivery vehicles implies construction of recombinant strains that contain the gene cassette encoding the antigen. With the increased knowledge of mucosal immunity and the availability of genetic tools for heterologous gene expression the concept of live vaccine vehicles gains renewed interest. However, administration of live bacterial vaccines poses some risks. In addition, vaccination using recombinant bacteria results in the release of live recombinant organisms into nature. This places these vaccines in the debate on application of genetically modified organisms. In this review we give an overview of live bacterial vaccines on the market and describe the development of new live vaccines with a focus on attenuated bacteria and food-related lactic acid bacteria. Furthermore, we outline the safety concerns and identify the hazards associated with live bacterial vaccines and try to give some suggestions of what to consider during their development.
Collapse
Affiliation(s)
- Ann Detmer
- Danish Toxicology Centre, Hørsholm, Denmark
| | | |
Collapse
|