1
|
Wu X, Meng Y, Yao Z, Lin X, Hu M, Cai S, Gao S, Zhang H. Extracellular vesicles as nature's nano carriers in cancer therapy: insights toward preclinical studies and clinical applications. Pharmacol Res 2025:107751. [PMID: 40345354 DOI: 10.1016/j.phrs.2025.107751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/14/2025] [Accepted: 04/27/2025] [Indexed: 05/11/2025]
Abstract
Extracellular vesicles (EVs), which are secreted by various cell types, hold significant potential for cancer therapy. However, there are several challenges and difficulties that limit their application in clinical settings. This review, which integrates the work of our team and recent advancements in this research field, discusses EV-based cancer treatment strategies to guide their clinical application. The following treatment strategies are discussed: 1) leveraging the inherent properties of EVs for the development of cancer treatments; 2) modifying EVs using EV engineering methods to improve drug loading and delivery; 3) targeting key molecules in tumor-derived EV (TDE) synthesis to inhibit their production; and 4) clearing TDEs from the tumor microenvironment. Additionally, on the basis of research into EV-based vaccines and bispecific antibodies, this review elaborates on strategies to enhance antitumor immunity via EVs and discusses engineering modifications that can improve EV targeting ability and stability and the research progress of AI technology in targeted delivery of EV drugs. Although there are limited strategies for enhancing EV targeting abilities, this review provides an in-depth discussion of prior studies. Finally, this review summarizes the clinical progress on the use of EVs in cancer therapy and highlights challenges that need to be addressed.
Collapse
Affiliation(s)
- Xiaotong Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yuhua Meng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaona Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Thoracic Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Mengyuan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Songwang Cai
- Department of Thoracic Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shegan Gao
- College of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China.
| | - Hao Zhang
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China.
| |
Collapse
|
2
|
Gomari MM, Ghantabpour T, Pourgholam N, Rostami N, Hatfield SM, Namazifar F, Abkhiz S, Eslami SS, Ramezanpour M, Darestanifarahani M, Astsaturov I, Bencherif SA. Breaking barriers: Smart vaccine platforms for cancer immunomodulation. Cancer Commun (Lond) 2025; 45:529-571. [PMID: 39901621 PMCID: PMC12067400 DOI: 10.1002/cac2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite significant advancements in cancer treatment, current therapies often fail to completely eradicate malignant cells. This shortfall underscores the urgent need to explore alternative approaches such as cancer vaccines. Leveraging the immune system's natural ability to target and kill cancer cells holds great therapeutic potential. However, the development of cancer vaccines is hindered by several challenges, including low stability, inadequate immune response activation, and the immunosuppressive tumor microenvironment, which limit their efficacy. Recent progress in various fields, such as click chemistry, nanotechnology, exosome engineering, and neoantigen design, offer innovative solutions to these challenges. These achievements have led to the emergence of smart vaccine platforms (SVPs), which integrate protective carriers for messenger ribonucleic acid (mRNA) with functionalization strategies to optimize targeted delivery. Click chemistry further enhances SVP performance by improving the encapsulation of mRNA antigens and facilitating their precise delivery to target cells. This review highlights the latest developments in SVP technologies for cancer therapy, exploring both their opportunities and challenges in advancing these transformative approaches.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Taha Ghantabpour
- Department of AnatomySchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Nima Pourgholam
- School of Nursing and MidwiferyIran University of Medical ScienceTehranIran
| | - Neda Rostami
- Department of Chemical EngineeringArak UniversityArakIran
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection InstituteDepartment of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
| | | | - Shadi Abkhiz
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Seyed Sadegh Eslami
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
- Molecular Proteomics LaboratoryBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Mahsa Ramezanpour
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | | | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer InstituteFox Chase Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Polymers, Biopolymers Surfaces (PBS) LaboratoryNational Center for Scientific Research (CNRS) Mixed Research Unit (UMR) 6270University Rouen NormandieRouenFrance
| |
Collapse
|
3
|
Ahmadishoar S, Mones Saeed S, Salih Mahdi M, Mohammed Taher W, Alwan M, Jasem Jawad M, Khdyair Hamad A, Gandomkar H. The potential use of bacteria and their derivatives as delivery systems for nanoparticles in the treatment of cancer. J Drug Target 2025:1-34. [PMID: 40186857 DOI: 10.1080/1061186x.2025.2489979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Nanomaterials, unique optical, magnetic, and electrical properties at the nanoscale (1-100 nm), have been engineered to improve drug capacity, bioavailability, and specificity in cancer treatment. These advancements address toxicity and lack of selectivity in conventional therapies, enabling precise targeting of cancer cells, the tumour microenvironment, and the immune system. Among emerging approaches, bacterial treatment shows promise due to its natural ability to target cancer and its diverse therapeutic mechanisms, which nanotechnology can further enhance. Bacteria-based drug delivery systems leverage bacteria's adaptability and survival strategies within the human body. Bacterial derivatives, such as bacterial ghosts (BGs), bacterial extracellular vesicles (BEVs), and dietary toxins, are recognised as effective biological nanomaterials capable of carrying nanoparticles (NPs). These systems have attracted increasing attention for their potential in targeted NP delivery for cancer treatment. This study explores the use of various bacteria and their byproducts as NP delivery vehicles, highlighting their potential in treating different types of cancer. By combining the strengths of nanotechnology and bacterial therapy, these innovative approaches aim to revolutionise cancer treatment with improved precision and efficacy.
Collapse
Affiliation(s)
- Shiva Ahmadishoar
- Department of Microbiology, Male.C., Islamic Azad University, Malekan, Iran
| | - Samaa Mones Saeed
- Dental Prosthetics Techniques Department, Health and Medical Techniques College/AlNoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | | - Hossein Gandomkar
- Department of Surgical Oncology, Tehran University of Medical Medicine, Tehran, Iran
| |
Collapse
|
4
|
Fu Z, Lin S, Chen H, Guo H, Li J, Chen Y, Lu Y, Liu J, Huang W, Pang Y. Generating Self-Adjuvated Nanofiber Vaccines by Coating Bacterial Flagella with Antigens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415887. [PMID: 39981905 DOI: 10.1002/adma.202415887] [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: 10/17/2024] [Revised: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Bacteria-based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection-associated side effects due to the living characteristics. Here, the use of bacteria-derived flagella is described to construct self-adjuvated nanofiber vaccines. With the help of charge-reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self-adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen-specific T-cell and B-cell immune responses. In ovalbumin-overexpressing melanoma-bearing mice, immunization with ovalbumin-carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS-CoV-2 Spike protein S1 subunit. Bacterial flagella-based self-adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Chen
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Lu
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Xiangfu Laboratory, Jiaxing, Zhejiang, 314102, China
| | - Yan Pang
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
5
|
Oyler HJ, Callister AW, Kutch MN, Wakefield MR, Fang Y. Leveraging Microorganisms to Combat Skin Cancer. Microorganisms 2025; 13:462. [PMID: 40005824 PMCID: PMC11858759 DOI: 10.3390/microorganisms13020462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Skin cancer, including melanoma and non-melanoma types, presents a significant and growing global health challenge due to its increasing incidence and mortality rates. While conventional treatments such as surgical excision, immunotherapy, and targeted therapies are well-established, microorganism-based approaches represent an innovative and promising alternative. These therapies employ live, genetically engineered, or commensal bacteria, viral vectors, or bacterial components to achieve various therapeutic mechanisms, including tumor targeting, immune system modulation, vascular disruption, competitive exclusion, drug delivery, and direct oncolysis. Despite their potential, these approaches require further investigation to address safety concerns, optimize treatment protocols, and gain a comprehensive understanding of their long-term outcomes.
Collapse
Affiliation(s)
- Hayden J. Oyler
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA;
| | - Austen W. Callister
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT 84132, USA;
| | - Makenzi N. Kutch
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65211, USA (M.R.W.)
| | - Mark R. Wakefield
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65211, USA (M.R.W.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA;
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65211, USA (M.R.W.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Gao S, Li X, Han B. Bacterial and bacterial derivatives-based drug delivery systems: a novel approach for treating central nervous system disorders. Expert Opin Drug Deliv 2025; 22:163-180. [PMID: 39688950 DOI: 10.1080/17425247.2024.2444364] [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: 12/06/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Bacteria and their derivatives show great potential as drug delivery systems due to their unique chemotaxis, biocompatibility, and targeting abilities. In CNS disease treatment, bacterial carriers can cross the blood-brain barrier (BBB) and deliver drugs precisely, overcoming limitations of traditional methods. Advances in genetic engineering, synthetic biology, and nanotechnology have transformed these systems into multifunctional platforms for personalized CNS treatment. AREAS COVERED This review examines the latest research on bacterial carriers for treating ischemic brain injury, neurodegenerative diseases, and gliomas. Bacteria efficiently cross the blood-brain barrier via active targeting, endocytosis, paracellular transport, and the nose-to-brain route for precise drug delivery. Various bacterial drug delivery systems, such as OMVs and bacterial ghosts, are explored for their design and application. Databases were searched in Google Scholar for the period up to December 2024. EXPERT OPINION Future developments in bacterial drug delivery will rely on AI-driven design and high-throughput engineering, enhancing treatment precision. Personalized medicine will further optimize bacterial carriers for individual patients, but challenges such as biosafety, immune rejection, and scalability must be addressed. As multimodal diagnostic and therapeutic strategies advance, bacterial carriers are expected to play a central role in CNS disease treatment, offering novel precision medicine solutions.
Collapse
Affiliation(s)
- Shizhu Gao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Li
- Orthopedic Medical Center, 2nd hospital of Jilin University, Changchun, PR China
| | - Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
7
|
Lloren KKS, Senevirathne A, Lee JH. Advancing vaccine technology through the manipulation of pathogenic and commensal bacteria. Mater Today Bio 2024; 29:101349. [PMID: 39850273 PMCID: PMC11754135 DOI: 10.1016/j.mtbio.2024.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/30/2024] [Accepted: 11/15/2024] [Indexed: 01/25/2025] Open
Abstract
Advancements in vaccine technology are increasingly focused on leveraging the unique properties of both pathogenic and commensal bacteria. This revolutionary approach harnesses the diverse immune modulatory mechanisms and bacterial biology inherent in different bacterial species enhancing vaccine efficacy and safety. Pathogenic bacteria, known for their ability to induce robust immune responses, are being studied for their potential to be engineered into safe, attenuated vectors that can target specific diseases with high precision. Concurrently, commensal bacteria, which coexist harmlessly with their hosts and contribute to immune system regulation, are also being explored as novel delivery systems and in microbiome-based therapy. These bacteria can modulate immune responses, offering a promising avenue for developing effective and personalized vaccines. Integrating the distinctive characteristics of pathogenic and commensal bacteria with advanced bacterial engineering techniques paves the way for innovative vaccine and therapeutic platforms that could address a wide range of infectious diseases and potentially non-infectious conditions. This holistic approach signifies a paradigm shift in vaccine development and immunotherapy, emphasizing the intricate interplay between the bacteria and the immune systems to achieve optimal immunological outcomes.
Collapse
Affiliation(s)
- Khristine Kaith S. Lloren
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| |
Collapse
|
8
|
Renteria-Flores FI, García-Chagollán M, Jave-Suárez LF. Bactofection, Bacterial-Mediated Vaccination, and Cancer Therapy: Current Applications and Future Perspectives. Vaccines (Basel) 2024; 12:968. [PMID: 39340000 PMCID: PMC11435753 DOI: 10.3390/vaccines12090968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
From the first report in 1891 by Dr. Coley of the effective treatment of tumors in 1000 patients with Streptococcus and the first successful use of bacterial vectors for transferring therapeutic genes in 1980 by Dr. Schnaffer, bactofection has been shown to be a promising strategy in the fields of vaccination, gene therapy, and cancer therapy. This review describes the general theory of bactofection and its advantages, disadvantages, challenges, and expectations, compiling the most notable advances in 14 vaccination studies, 27 cancer therapy studies, and 13 clinical trials. It also describes the current scope of bactofection and promising results. The extensive knowledge of Salmonella biology, as well as the multiple adequacies of the Ty21a vaccination platform, has allowed notable developments worldwide that have mainly been reflected in therapeutic efforts against cancer. In this regard, we strongly recommend the creation of a recombinant Ty21a model that constitutively expresses the GtgE protease from S. typhimurium, allowing this vector to be used in animal trials, thus enhancing the likelihood of favorable results that could quickly transition to clinical trials. From the current perspective, it is necessary to explore a greater diversity of bacterial vectors and find the best combination of implemented attenuations, generating personalized models that guarantee the maximum effectiveness in cancer therapy and vaccination.
Collapse
Affiliation(s)
- Francisco Israel Renteria-Flores
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Mariel García-Chagollán
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis Felipe Jave-Suárez
- Division of Immunology, Biomedical Research Centre of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
9
|
Salasar Moghaddam F, Tabibian M, Absalan M, Tavoosidana G, Ghahremani MH, Tabatabaei N, Abdolhosseini M, Shafiee Sabet M, Motevaseli E. Comparative analysis of Escherichia coli Nissle 1917 ghosts quality: a study of two chemical methods. Arch Microbiol 2024; 206:386. [PMID: 39190149 DOI: 10.1007/s00203-024-04095-0] [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: 01/06/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/28/2024]
Abstract
The gram-negative bacterium Escherichia coli Nissle 1917 (EcN) has long been recognized for its therapeutic potential in treating various intestinal diseases. Bacterial ghosts (BGs) are empty shells of non-living bacterial cells that demonstrate enormous potential for medicinal applications. Genetic and chemical techniques can create these BGs. In the current study, we produced Escherichia coli Nissle 1917 ghosts (EcNGs) for the first time using benzoic acid (BA) and sodium hydroxide (SH). BA is a feeble acidic chemical that enhances gram-negative bacteria's external membrane permeability, reduces energy production, and decreases internal pH. SH has shown success in producing BGs from some gram-negative and gram-positive organisms. This research aims to produce EcNGs using the minimum inhibitory concentration (MIC) of SH and BA, specifically 3.125 mg/mL. We assessed the bacterial quality of the BGs produced using quantitative PCR (qPCR) and Bradford protein assays. Field emission scanning electron microscopy (FE-SEM) showed the three-dimensional structure of EcNGs. The study confirmed the presence of tunnel-like pores on the outer surface, indicating the preservation of cell membrane integrity. Importantly, this investigation introduces BA as a novel chemical inducer of EcNGs, suggesting its potential alongside SH for efficient EcNG formation.
Collapse
Affiliation(s)
- Fahimeh Salasar Moghaddam
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mobina Tabibian
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnologies, Shahid Beheshti University, Tehran, Iran
| | - Moloud Absalan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Tavoosidana
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology-Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrollah Tabatabaei
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansoreh Abdolhosseini
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Shafiee Sabet
- Department of Neurology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Ijaz M, Hasan I, Chaudhry TH, Huang R, Zhang L, Hu Z, Tan Q, Guo B. Bacterial derivatives mediated drug delivery in cancer therapy: a new generation strategy. J Nanobiotechnology 2024; 22:510. [PMID: 39182109 PMCID: PMC11344338 DOI: 10.1186/s12951-024-02786-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
Cancer is measured as a major threat to human life and is a leading cause of death. Millions of cancer patients die every year, although a burgeoning number of researchers have been making tremendous efforts to develop cancer medicine to fight against cancer. Owing to the complexity and heterogeneity of cancer, lack of ability to treat deep tumor tissues, and high toxicity to the normal cells, it complicates the therapy of cancer. However, bacterial derivative-mediated drug delivery has raised the interest of researchers in overcoming the restrictions of conventional cancer chemotherapy. In this review, we show various examples of tumor-targeting bacteria and bacterial derivatives for the delivery of anticancer drugs. This review also describes the advantages and limitations of delivering anticancer treatment drugs under regulated conditions employing these tumor-targeting bacteria and their membrane vesicles. This study highlights the substantial potential for clinical translation of bacterial-based drug carriers, improve their ability to work with other treatment modalities, and provide a more powerful, dependable, and distinctive tumor therapy.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518060, Guangdong, China
| | - Tamoor Hamid Chaudhry
- Antimicrobial Resistance (AMR) Containment & Infection Prevention & Control (IPC) Program, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Rui Huang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Lan Zhang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Ziwei Hu
- Institute of Otolaryngology Head and Neck Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, China.
| | - Qingqin Tan
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Narayanan S, Baburajan AP, Muhammad M, Joseph A, Vemula PK, Bhat SG. Demonstrating the immunostimulatory and cytokine-augmentation effects of bacterial ghosts on natural killer cells and Caenorhabditis Elegans. Biotechnol Bioeng 2024; 121:959-970. [PMID: 38059432 PMCID: PMC7615764 DOI: 10.1002/bit.28619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
The potential of bacteria-based immunotherapy lies in its ability to inherently enhance immune responses. However, the "liveness" of bacteria poses risks of bacterial escape, nonspecific immuno-stimulation, and ethical concerns, limiting their acceptability in immunotherapy. In this scenario, nonliving empty bacterial-cell envelopes, named bacterial ghosts (BGs), have emerged as immuno-stimulants with the potential to side-step the limitations of live bacterial therapies. This study demonstrates the capability of BGs in modulating the functionality of NK-92 cells and Caenorhabditis elegans (C. elegans), as well as perform as cytokine-therapy adjuvants. BGs were obtained through a pH-driven culture method, and were validated for their structural and chemical integrity via electron microscopy and spectroscopy. In NK-92 cells, BGs have shown significant immuno-stimulation by boosting the gene-expression of perforin, granzyme-B, Fas-L, and interferon-gamma by factors of 3.5-, 1.5-, 12.5-, and 8.6-folds, respectively. Combined BG and IL-12 treatment yielded a notable 10.2-fold increase in interferon-gamma protein expression in 24 h. The BGs also significantly influenced the innate immune response in C. elegans through the upregulation of lysozyme genes viz., ilys-3 (8.8-fold) and lys-2 (3.1-fold). Our investigation into the impact of BGs on natural killer cells and C. elegans highlights its potential as a valid alternative approach for new-age immunotherapy and cytokine augmentation.
Collapse
Affiliation(s)
- Sreeja Narayanan
- Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| | | | - Mumtaz Muhammad
- Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| | - Andrea Joseph
- Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| | - Praveen Kumar Vemula
- Chemical Biology Approaches for Stem Cells and Therapeutics, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, Karnataka, India
| | - Sarita Ganapathy Bhat
- Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| |
Collapse
|
13
|
Zhang H, Li Z, Li W, Jin Y, Li Y, Xiao Q, Tong D, Zhou J. Comparison of the immune effects of the Chlamydia abortus MOMP antigen displayed in different parts of bacterial ghosts. Front Microbiol 2024; 15:1349746. [PMID: 38389524 PMCID: PMC10883653 DOI: 10.3389/fmicb.2024.1349746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Bacterial ghosts (BGs) are promising vaccine platforms owing to their high adjuvant properties and delivery efficiency. Heterologous antigens can be anchored to different parts of BGs using genetic engineering strategies to prepare vaccines. However, several key issues need to be resolved, including the efficient preparation of BGs and determining the optimal anchoring position of exogenous antigens in the BGs. Here, we prepared an efficient temperature-controlled lysis system using lysis gene E of phage PhiX174 and used the major outer membrane protein (MOMP) of Chlamydia abortus (C. abortus) as a model antigen to explore the optimal display location of exogenous antigens in BGs. We demonstrated that the constructed recombinant temperature-controlled lysis plasmid can still stably inhibit E gene expression at 37°C, and the lysis efficiency of E. coli can reach above 99.9%. Four recombinant MOMP Escherichia coli (E. coli) ghost vaccines were constructed using different anchor sequences. These vaccines all induced strong specific antibody responses and secrete high levels of IFN-γ in immunized mice and significantly increased the clearance of C. abortus in a mouse infection model. Notably, the strongest immune effect was observed when MOMP was displayed on the surface of E. coli ghosts (rECG-InpN-M), which resulted in the clearance of C. abortus in mice 6 days earlier than that with the recombinant MOMP vaccine. Altogether, we constructed an efficient BG temperature-controlled lysis system and provided a feasible strategy for developing a BG delivery platform with enhanced immune effects.
Collapse
Affiliation(s)
- Huaiyu Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zhaocai Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wei Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Youshun Jin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yunhui Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Xiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jizhang Zhou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| |
Collapse
|
14
|
Zhao L, Chen J, Bai B, Song G, Zhang J, Yu H, Huang S, Wang Z, Lu G. Topical drug delivery strategies for enhancing drug effectiveness by skin barriers, drug delivery systems and individualized dosing. Front Pharmacol 2024; 14:1333986. [PMID: 38293666 PMCID: PMC10825035 DOI: 10.3389/fphar.2023.1333986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Topical drug delivery is widely used in various diseases because of the advantages of not passing through the gastrointestinal tract, avoiding gastrointestinal irritation and hepatic first-pass effect, and reaching the lesion directly to reduce unnecessary adverse reactions. The skin helps the organism to defend itself against a huge majority of external aggressions and is one of the most important lines of defense of the body. However, the skin's strong barrier ability is also a huge obstacle to the effectiveness of topical medications. Allowing the bioactive, composition in a drug to pass through the stratum corneum barrier as needed to reach the target site is the most essential need for the bioactive, composition to exert its therapeutic effect. The state of the skin barrier, the choice of delivery system for the bioactive, composition, and individualized disease detection and dosing planning influence the effectiveness of topical medications. Nowadays, enhancing transdermal absorption of topically applied drugs is the hottest research area. However, enhancing transdermal absorption of drugs is not the first choice to improve the effectiveness of all drugs. Excessive transdermal absorption enhances topical drug accumulation at non-target sites and the occurrence of adverse reactions. This paper introduces topical drug delivery strategies to improve drug effectiveness from three perspectives: skin barrier, drug delivery system and individualized drug delivery, describes the current status and shortcomings of topical drug research, and provides new directions and ideas for topical drug research.
Collapse
Affiliation(s)
- Lin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiamei Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bai Bai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guili Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiwei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guanghua Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Faghihkhorasani A, Ahmed HH, Mashool NM, Alwan M, Assefi M, Adab AH, Yasamineh S, Gholizadeh O, Baghani M. The potential use of bacteria and bacterial derivatives as drug delivery systems for viral infection. Virol J 2023; 20:222. [PMID: 37789431 PMCID: PMC10548687 DOI: 10.1186/s12985-023-02183-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 10/05/2023] Open
Abstract
Viral infections in humans are responsible for fatalities worldwide and contribute to the incidence of various human ailments. Controllable targeted medicine delivery against many illnesses, including viral infection, may be significantly aided by using bacteria and bacteria-derived products. They may accumulate in diseased tissues despite physical obstacles, where they can launch antiviral immunity. The ability to genetically and chemically modify them means that vaccinations against viral infections may be manufactured and delivered to affected tissues more safely and effectively. The objective of this study is to provide an overview of the latest advancements in the field of utilizing bacteria and bacterial derivatives as carriers for administering medication to treat viral diseases such as SARS-CoV-2, hepatitis B virus, hepatitis C virus, human immunodeficiency virus, human papillomavirus, influenza, and Ebola virus.
Collapse
Affiliation(s)
| | | | | | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | - Marjan Assefi
- University of North Carolina at Greensboro, Greensboro, USA
| | - Aya Hussein Adab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Omid Gholizadeh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Moein Baghani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Halder P, Maiti S, Banerjee S, Das S, Dutta M, Dutta S, Koley H. Bacterial ghost cell based bivalent candidate vaccine against Salmonella Typhi and Salmonella Paratyphi A: A prophylactic study in BALB/c mice. Vaccine 2023; 41:5994-6007. [PMID: 37625993 DOI: 10.1016/j.vaccine.2023.08.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/06/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Typhoid and emerging paratyphoid fever are a severe enteric disease worldwide with high morbidity and mortality. Licensed typhoid vaccines are in the market, but no paratyphoid vaccine is currently available. In the present study we developed a bivalent vaccine against Salmonella Typhi and Paratyphi A using a bacterial ghost platform. Bacterial ghost cells (BGs) are bacteria-derived cell membranes without cytoplasmic contents that retain their cellular morphology, including all cell surface features. Furthermore, BGs have inherent adjuvant properties that promote an enhanced humoral and cellular immune reaction to the target antigen. Sodium hydroxide was used to prepare ghost cells of Salmonella Typhi and Paratyphi A. The bacterial ghost cells were characterised using electron microscopy. Then BALB/c mice were immunized three times (0th, 14th and 28th day) with the bivalent typhoidal bacterial ghost cells. Haematological study of adult mice throughout immunization period reflected that the immunogen was safe to administer and does not affect the animals' health. After complete immunization, we found significant serum antibody titter against whole cell lysate, outer membrane protein and lipopolysaccharide of both bacteria, and cell-mediated immunity was observed in an ex-vivo experiment. CD4+, CD8a+ and CD19+ splenic cell populations were increased in immunized animals. Bivalent Typhoidal ghost cell immunized mice showed better survival, less bacterial colonization in systemic organs, and less inflammation and/or destruction of tissue in histopathological analysis than non-immunized control mice.Serum antibodies of immunized animals can significantly inhibit bacterial motility and mucin penetration ability with better killing properties against Salmonella Typhi and Paratyphi A. Furthermore, significant passive protection was observed through the adoptive transfer of serum antibody and lymphocytes of immunized animals to naïve animals after bacterial infection. In summary, Bivalent Typhoidal Bacterial Ghost cells (BTBGs) enhances immunogenic properties and serves as a safe and effective prevention strategy against Salmonella Typhi and Paratyphi A.
Collapse
Affiliation(s)
- Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Suhrid Maiti
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Soumalya Banerjee
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Sanjib Das
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Moumita Dutta
- Division of Electron Microscopy, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India.
| |
Collapse
|
17
|
Abdelfattah A, Samir R, Amin HM. Production of highly immunogenic and safe Triton X-100 produced bacterial ghost vaccine against Shigella flexneri 2b serotype. Gut Pathog 2023; 15:41. [PMID: 37679798 PMCID: PMC10483756 DOI: 10.1186/s13099-023-00568-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Bacterial ghost cells (BGCs) are cells were drained of their genetic and cytoplasmic components. This work aimed to develop vaccine candidates against the Shigella flexneri (S. flexneri) 2b serotype using the BGCs approach. For the first time, (S. flexneri) 2b serotype BGCs vaccine was prepared by incubation with Triton X-100 (TX100) for only 12 h. Its safety and immunogenicity were compared to another vaccine produced using a previously used surfactant, namely Tween 80 (TW80). Scanning electron microscopy (SEM), cellular DNA, protein contents measurements, and ghost cell re-cultivation were used to confirm the successful generation of the BGCs. Immunogenicity was assessed through mice's intraperitoneal (IP) immunization followed by infection with S. flexneri ATCC 12022. Finally, histopathological examination was carried out. RESULTS Viable colony forming units (CFUs) of S. flexneri were counted from stool samples as well as homogenized colon tissues of the non-immunized challenged group. Immunized mice sera showed a significant increase in serum bactericidal activity of both preparations (TX100 = 40% and TW80 = 56%) compared to the non-immunized challenged group (positive control). The IgG levels of the bacterial ghost-vaccinated groups were four and three times greater for the TX100 and TW80 ghost vaccines, respectively, compared to that of the positive control; both bacterial ghost vaccines (BGVs) were safe and effective, according to the results of the safety check tests and histopathological analysis. CONCLUSIONS When comparing the BGVs prepared using TX100 and TW80 methods, the use of TX100 as a new chemical treating agent for BGC production attained robust results in terms of shorter incubation time with the targeted cells and a strong immune response against S. flexneri 2b serotype ATCC 12022 in the IP challenge test. However, a clinical study is needed to confirm the efficacy and total safety of this novel vaccine.
Collapse
Affiliation(s)
- Amany Abdelfattah
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 26 July Mehwar Road Intersection With Wahat Road, 6Th of October, 12451 Giza Egypt
| | - Reham Samir
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Nile Corniche, El Sayeda Zeinab, Cairo, 11562 Egypt
| | - Heba M. Amin
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 26 July Mehwar Road Intersection With Wahat Road, 6Th of October, 12451 Giza Egypt
| |
Collapse
|
18
|
Orta AK, Riera N, Li YE, Tanaka S, Yun HG, Klaic L, Clemons WM. The mechanism of the phage-encoded protein antibiotic from ΦX174. Science 2023; 381:eadg9091. [PMID: 37440661 DOI: 10.1126/science.adg9091] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/23/2023] [Indexed: 07/15/2023]
Abstract
The historically important phage ΦX174 kills its host bacteria by encoding a 91-residue protein antibiotic called protein E. Using single-particle electron cryo-microscopy, we demonstrate that protein E bridges two bacterial proteins to form the transmembrane YES complex [MraY, protein E, sensitivity to lysis D (SlyD)]. Protein E inhibits peptidoglycan biosynthesis by obstructing the MraY active site leading to loss of lipid I production. We experimentally validate this result for two different viral species, providing a clear model for bacterial lysis and unifying previous experimental data. Additionally, we characterize the Escherichia coli MraY structure-revealing features of this essential enzyme-and the structure of the chaperone SlyD bound to a protein. Our structures provide insights into the mechanism of phage-mediated lysis and for structure-based design of phage therapeutics.
Collapse
Affiliation(s)
- Anna K Orta
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Nadia Riera
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yancheng E Li
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shiho Tanaka
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Hyun Gi Yun
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lada Klaic
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - William M Clemons
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
19
|
Fan S, Han H, Yan Z, Lu Y, He B, Zhang Q. Lipid-based nanoparticles for cancer immunotherapy. MEDICAL REVIEW (2021) 2023; 3:230-269. [PMID: 37789955 PMCID: PMC10542882 DOI: 10.1515/mr-2023-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/07/2023] [Indexed: 10/05/2023]
Abstract
As the fourth most important cancer management strategy except surgery, chemotherapy and radiotherapy, cancer immunotherapy has been confirmed to elicit durable antitumor effects in the clinic by leveraging the patient's own immune system to eradicate the cancer cells. However, the limited population of patients who benefit from the current immunotherapies and the immune related adverse events hinder its development. The immunosuppressive microenvironment is the main cause of the failure, which leads to cancer immune evasion and immunity cycle blockade. Encouragingly, nanotechnology has been engineered to enhance the efficacy and reduce off-target toxicity of their therapeutic cargos by spatiotemporally controlling the biodistribution and release kinetics. Among them, lipid-based nanoparticles are the first nanomedicines to make clinical translation, which are now established platforms for diverse areas. In this perspective, we discuss the available lipid-based nanoparticles in research and market here, then describe their application in cancer immunotherapy, with special emphasis on the T cells-activated and macrophages-targeted delivery system. Through perpetuating each step of cancer immunity cycle, lipid-based nanoparticles can reduce immunosuppression and promote drug delivery to trigger robust antitumor response.
Collapse
Affiliation(s)
- Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Huize Han
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhicheng Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yao Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo, Zhejiang Province, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo, Zhejiang Province, China
| |
Collapse
|
20
|
Dranchak PK, Oliphant E, Queme B, Lamy L, Wang Y, Huang R, Xia M, Tao D, Inglese J. In vivo quantitative high-throughput screening for drug discovery and comparative toxicology. Dis Model Mech 2023; 16:dmm049863. [PMID: 36786055 PMCID: PMC10067442 DOI: 10.1242/dmm.049863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
Quantitative high-throughput screening (qHTS) pharmacologically evaluates chemical libraries for therapeutic uses, toxicological risk and, increasingly, for academic probe discovery. Phenotypic high-throughput screening assays interrogate molecular pathways, often relying on cell culture systems, historically less focused on multicellular organisms. Caenorhabditis elegans has served as a eukaryotic model organism for human biology by virtue of genetic conservation and experimental tractability. Here, a paradigm enabling C. elegans qHTS using 384-well microtiter plate laser-scanning cytometry is described, in which GFP-expressing organisms revealing phenotype-modifying structure-activity relationships guide subsequent life-stage and proteomic analyses, and Escherichia coli bacterial ghosts, a non-replicating nutrient source, allow compound exposures over two life cycles, mitigating bacterial overgrowth complications. We demonstrate the method with libraries of anti-infective agents, or substances of toxicological concern. Each was tested in seven-point titration to assess the feasibility of nematode-based in vivo qHTS, and examples of follow-up strategies were provided to study organism-based chemotype selectivity and subsequent network perturbations with a physiological impact. We anticipate that this qHTS approach will enable analysis of C. elegans orthologous phenotypes of human pathologies to facilitate drug library profiling for a range of therapeutic indications.
Collapse
Affiliation(s)
- Patricia K. Dranchak
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Erin Oliphant
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Bryan Queme
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Laurence Lamy
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Yuhong Wang
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ruili Huang
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Menghang Xia
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Dingyin Tao
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - James Inglese
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20817, USA
| |
Collapse
|
21
|
Wang Y, Li Z, Mo F, Chen-Mayfield TJ, Saini A, LaMere AM, Hu Q. Chemically engineering cells for precision medicine. Chem Soc Rev 2023; 52:1068-1102. [PMID: 36633324 DOI: 10.1039/d2cs00142j] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cell-based therapy holds great potential to address unmet medical needs and revolutionize the healthcare industry, as demonstrated by several therapeutics such as CAR-T cell therapy and stem cell transplantation that have achieved great success clinically. Nevertheless, natural cells are often restricted by their unsatisfactory in vivo trafficking and lack of therapeutic payloads. Chemical engineering offers a cost-effective, easy-to-implement engineering tool that allows for strengthening the inherent favorable features of cells and confers them new functionalities. Moreover, in accordance with the trend of precision medicine, leveraging chemical engineering tools to tailor cells to accommodate patients individual needs has become important for the development of cell-based treatment modalities. This review presents a comprehensive summary of the currently available chemically engineered tools, introduces their application in advanced diagnosis and precision therapy, and discusses the current challenges and future opportunities.
Collapse
Affiliation(s)
- Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fanyi Mo
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Ting-Jing Chen-Mayfield
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Aryan Saini
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Afton Martin LaMere
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
22
|
Chen Y, Li ZH, Zeng X, Zhang XZ. Bacteria-based bioactive materials for cancer imaging and therapy. Adv Drug Deliv Rev 2023; 193:114696. [PMID: 36632868 DOI: 10.1016/j.addr.2023.114696] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/02/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Owing to the unique biological functions, bacteria as biological materials have been widely used in biomedical field. With advances in biotechnology and nanotechnology, various bacteria-based bioactive materials were developed for cancer imaging and therapy. In this review, different types of bacteria-based bioactive materials and their construction strategies were summarized. The advantages and property-function relationship of bacteria-based bioactive materials were described. Representative researches of bacteria-based bioactive materials in cancer imaging and therapy were illustrated, revealing general ideas for their construction. Also, limitation and challenges of bacteria-based bioactive materials in cancer research were discussed.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Zi-Hao Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China; Wuhan Research Centre for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430071, PR China.
| |
Collapse
|
23
|
Bahy R, Gaber A, Zedan H, Mabrook M. New typhoid vaccine using sponge-like reduced protocol: development and evaluation. Clin Exp Vaccine Res 2023; 12:70-76. [PMID: 36844684 PMCID: PMC9950223 DOI: 10.7774/cevr.2023.12.1.70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/22/2022] [Indexed: 02/19/2023] Open
Abstract
Purpose Typhoid remains a major health problem, especially in the developing world. Furthermore, the emergence of multidrug-resistant and extensively drug-resistant strains of Salmonella typhi added a sense of urgency to develop more effective typhoid vaccines, one of which is bacterial ghosts (BGs), prepared by both genetic and chemical means. The chemical method includes incubation with numerous agents for a short time at their minimum inhibitory or minimum growth concentrations. This study included the preparation of BGs by a sponge-like reduced protocol (SLRP). Materials and Methods Critical concentrations of sodium dodecyl sulfate, NaOH, and H2O2 were used. Moreover, high-quality BGs were visualized by scanning electron microscope (SEM). Subculturing was used to confirm the absence of vital cells. Besides, the concentrations of the released DNA and protein were estimated spectrophotometrically. In addition, the integrity of cells was proved by visualizing Gram-stained cells using a light microscope. Furthermore, a comparison between the immunogenicity and safety of the prepared vaccine and the available whole-cell killed vaccine was established. Results Improved preparation of high-quality BGs of S. typhi, visualized by SEM, revealed punctured cells with intact outer shells. Moreover, the absence of vital cells was confirmed by subculturing. At the same time, the release of respective amounts of proteins and DNA is another evidence of BGs' production. Additionally, the challenge test provided evidence that the prepared BGs are immunogenic and have the same efficacy as the whole cell vaccine. Conclusion The SLRP provided a simple, economical, and feasible method for BGs preparation.
Collapse
Affiliation(s)
- Rehab Bahy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Asmaa Gaber
- Department of Microbiology, General Division of Basic Medical Sciences, Egyptian Drug Authority (EDA), formerly National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Hamdallah Zedan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mona Mabrook
- Department of Microbiology, General Division of Basic Medical Sciences, Egyptian Drug Authority (EDA), formerly National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
24
|
Mougenot MF, Pereira VS, Costa ALR, Lancellotti M, Porcionatto MA, da Silveira JC, de la Torre LG. Biomimetic Nanovesicles—Sources, Design, Production Methods, and Applications. Pharmaceutics 2022; 14:pharmaceutics14102008. [PMID: 36297442 PMCID: PMC9610935 DOI: 10.3390/pharmaceutics14102008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Despite all the progress in the field of liposomes and nanoparticles for applications as drug and gene delivery systems, the specific targeting and immune system escape capabilities of these systems are still limited. Biomimetic nanovesicles emerged as a strategy to overcome these and other limitations associated with synthetic carriers, such as short circulation time, cytotoxicity, and difficulty in crossing biological barriers, since many of the desirable abilities of drug delivery systems are innate characteristics of biological vesicles. Thus, the question arises: would biomimetic nanovesicles be responsible for addressing these advances? It is currently known that biomimetic nanovesicles (BNV) can combine the intrinsic advantages of natural materials with the well-known production methods and controllability of synthetic systems. Besides, the development of the biotechnology and nanotechnology fields has provided a better understanding of the functionalities of biological vesicles and the means for the design and production of biomimetic nanovesicles (BNV). Based on this, this work will focus on tracking the main research on biomimetic nanovesicles (BNV) applied as drug and gene delivery systems, and for vaccines applications. In addition, it will describe the different sources of natural vesicles, the technical perspectives on obtaining them, and the possibility of their hybridization with synthetic liposomes.
Collapse
Affiliation(s)
- Marcel Franco Mougenot
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Vanessa Sousa Pereira
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Ana Letícia Rodrigues Costa
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Institute of Exact and Technological Sciences, Campus Florestal, Federal University of Viçosa (UFV), Florestal 35690-000, Brazil
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | | | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Correspondence: ; Tel.: +55-19-3521-0397
| |
Collapse
|
25
|
Construction and In Vitro Evaluation of a Tumor Acidic pH-Targeting Drug Delivery System Based on Escherichia coli Nissle 1917 Bacterial Ghosts. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9090433. [PMID: 36134979 PMCID: PMC9495381 DOI: 10.3390/bioengineering9090433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/06/2022]
Abstract
Synthetic nanocarriers are a promising therapeutic delivery strategy. However, these systems are often hampered by inherent disadvantages such as strong biotoxicity and poor biocompatibility. To overcome these issues, biological carriers with commonly used chemotherapy drugs have been developed. In this work, engineered bacterial ghosts (BGs) originated from probiotic Escherichia coli Nissle 1917 (EcN) were devised to specifically target acidic extracellular environments of tumor tissue. To improve the production efficiency and safety, a novel lysis protein E from phage α3 was applied to produce EcN BGs under high growth densities in high quality. In addition, the acidity-triggered rational membrane (ATRAM) peptides were displayed in EcN BGs to facilitate specific cancer cell internalization within the acidic tumor microenvironment before drug release. In conclusion, the engineered EcN BGs offer a promising means for bionic bacteria construction for hepatocellular carcinoma therapy.
Collapse
|
26
|
Ma Y, Zhu W, Zhu G, Xu Y, Li S, Chen R, Chen L, Wang J. Efficient Robust Yield Method for Preparing Bacterial Ghosts by Escherichia coli Phage ID52 Lysis Protein E. Bioengineering (Basel) 2022; 9:300. [PMID: 35877351 PMCID: PMC9311611 DOI: 10.3390/bioengineering9070300] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Bacterial ghosts (BGs) are nonliving empty bacterial shells without cytoplasm retaining original morphology and identical antigenicity of natural bacteria, making them high potential and promising vaccine candidates and delivery vehicles. However, the low yield of commonly used BGs preparation methods limits its mass production and widely application. In order to improve BGs production, E. coli phage ID52 lysis protein E was introduced to generating BGs for the first time. Above all, we compared the lysis activity of lysis protein of E. coli phage φX174 and E. coli phage ID52 as well as the effects of promoters on the lysis activity of ID52-E, which shown that the lysis activity and BGs formation rate of protein ID52-E was significantly higher than protein φX174-E. Further, the lysis activity of ID52-E was significantly improved under the control of L-arabinose inducible promoter which initial induction OD600 reached as high as 2.0. The applicability of lysis protein ID52-E induced by L-arabinose was proved by preparing probiotic E. coli Nissle 1917 BGs and pathogenic Salmonella typhimurium BGs in mass production. This paper introduced a novel and highly efficient method for BGs preparation depending on recombinant expression of E. coli phage ID52-E under eco-friendly and reasonable price inducer L-arabinose.
Collapse
Affiliation(s)
- Yi Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wenjun Zhu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
| | - Guanshu Zhu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
| | - Yue Xu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
| | - Shuyu Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
| | - Rui Chen
- Bionavi Life Sciences Co., Ltd., Shenzhen 518118, China;
| | - Lidan Chen
- Department of Laboratory Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China;
| | - Jufang Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (W.Z.); (G.Z.); (Y.X.); (S.L.)
| |
Collapse
|
27
|
Xie YJ, Huang M, Li D, Hou JC, Liang HH, Nasim AA, Huang JM, Xie C, Leung ELH, Fan XX. Bacteria-based nanodrug for anticancer therapy. Pharmacol Res 2022; 182:106282. [PMID: 35662630 DOI: 10.1016/j.phrs.2022.106282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022]
Abstract
Bacteria-based immunotherapy has become a promising strategy to induce innate and adaptive responses for fighting cancer. The advantages of bacteriolytic tumor therapy mainly lie in stimulation of innate immunity and colonization of some bacteria targeting the tumor microenvironment (TME). These bacteria have cytotoxic proteins and immune modulating factors that can effectively restrain tumor growth. However, cancer is a multifactorial disease and single therapy is typically unable to eradicate tumors. Rapid progress has been made in combining bacteria with nanotechnology. Using the nanomolecular properties of bacterial products for tumor treatment preserves many features from the original bacteria while providing some unique advantages. Nano-bacterial therapy can enhance permeability and retention of drugs, increase the tolerability of the targeted drugs, promote the release of immune cell mediators, and induce immunogenic cell death pathways. In addition, combining nano-bacterial mediated antitumor therapeutic systems with modern therapy is an effective strategy for overcoming existing barriers in antitumor treatment and can achieve satisfactory therapeutic efficacy. Overall, exploring the immune antitumor characteristics of adjuvant clinical treatment with bacteria can provide potential efficacious treatment strategies for combatting cancer.
Collapse
Affiliation(s)
- Ya-Jia Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Dan Li
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Jin-Cai Hou
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Hai-Hai Liang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ali Adnan Nasim
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ju-Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chun Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Elaine Lai-Han Leung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
28
|
Yu X, Wang L, Yang X, Zhang S, Li G, Zhang L, Li J, Wang X, Zhou H, Jiang Y, Cui W, Li Y, Tang L, Qiao X. Lactobacillus casei Ghosts as a Vehicle for the Delivery of DNA Vaccines Mediate Immune Responses. Front Immunol 2022; 13:849409. [PMID: 35711427 PMCID: PMC9193971 DOI: 10.3389/fimmu.2022.849409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022] Open
Abstract
We developed Lactobacillus casei bacterial ghosts (BGs) as vehicles for delivering DNA vaccines and analyzed their effects on immune responses. Uptake of the plasmids encoding the enhanced green fluorescent protein (pCI-EGFP) and BGs loaded with pCI-EGFP by macrophages was investigated using fluorescence microscopy and flow cytometry. The results showed that pCI-EGFP-loaded L. casei BGs were efficiently taken up by macrophages. Lactobacillus casei BGs loaded with plasmids encoding VP6 protein of PoRV (pCI-PoRV-VP6) significantly upregulated the mRNA expression of interleukin (IL)-1β, IL-10, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS), arginase-1 (Arg-1), Mannose receptor (CD206) toll-like receptor (TLR)-2, TLR4, and TLR9 in macrophages. The levels of markers of M1 polarization (IL-10 and TNF-α) and M2 polarization (Arg-1 and CD206) were increased in macrophages incubated with pCI-PoRV-VP6-loaded BGs compared with the control group. The results of the enzyme-linked immunosorbent assay showed that the secretion of IL-1β, IL-10, and TNF-α in macrophages was significantly upregulated compared with the control group. Flow cytometry demonstrated that L. casei BGs loaded with pCI-PoRV-VP6 promoted the maturation of dendritic cells (DCs). Following incubation with pCI-PoRV-VP6-loaded BGs, the mRNA expression levels of IL-1β, IL-6 and interferon (IFN)-γ in DCs were significantly increased. ELISA assay showed the secretion of the IL-1β, IL-6, IFN-γ IL-10 and TNF-α in DCs were upregulated significantly. Thus, L. casei BGs promoted the maturation and activation of DCs. We analyzed the stimulatory capacity of DCs in a mixed lymphocyte reaction with allogeneic T cells. T cell proliferation increased upon incubation with DCs stimulated by BGs. After immunizing mice with BGs loaded with pCI-PoRV-VP6, the specific IgG levels in the serum were higher than those elicited by BGs loaded with pCI-PoRV-VP6. BGs loaded with pCI-PoRV-VP6 on Th1 and Th2 cytokines polarized T cells into the Th1 type and increased the proportion of CD4+/CD8+ T cells. These results indicate L. casei BGs effectively mediate immune responses and can be used as delivery system for DNA vaccination.
Collapse
Affiliation(s)
- Xiaoli Yu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Li Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinru Yang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Songsong Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Guiwei Li
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Lanlan Zhang
- Heilongjiang Fishery Technology Extension Station , Harbin, China
| | - Jiaxuan Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaona Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Han Zhou
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanping Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wen Cui
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yijing Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Lijie Tang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyuan Qiao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- *Correspondence: Xinyuan Qiao,
| |
Collapse
|
29
|
He CY, Yang JH, Ye YB, Zhao HL, Liu MZ, Yang QL, Liu BS, He S, Chen ZL. Proteomic and Antibody Profiles Reveal Antigenic Composition and Signatures of Bacterial Ghost Vaccine of Brucella abortus A19. Front Immunol 2022; 13:874871. [PMID: 35529865 PMCID: PMC9074784 DOI: 10.3389/fimmu.2022.874871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
Brucellosis is an important zoonotic disease that causes great economic losses. Vaccine immunisation is the main strategy for the prevention and control of brucellosis. Although live attenuated vaccines play important roles in the prevention of this disease, they also have several limitations, such as residual virulence and difficulty in the differentiation of immunisation and infection. We developed and evaluated a new bacterial ghost vaccine of Brucella abortus A19 by a new double inactivation method. The results showed that the bacterial ghost vaccine of Brucella represents a more safe and efficient vaccine for brucellosis. We further characterised the antigenic components and signatures of the vaccine candidate A19BG. Here, we utilised a mass spectrometry-based label-free relative quantitative proteomics approach to investigate the global proteomics changes in A19BGs compared to its parental A19. The proteomic analysis identified 2014 proteins, 1116 of which were differentially expressed compared with those in A19. The common immunological proteins of OMPs (Bcsp31, Omp25, Omp10, Omp19, Omp28, and Omp2a), HSPs (DnaK, GroS, and GroL), and SodC were enriched in the proteome of A19BG. By protein micro array-based antibody profiling, significant differences were observed between A19BG and A19 immune response, and a number of signature immunogenic proteins were identified. Two of these proteins, the BMEII0032 and BMEI0892 proteins were significantly different (P < 0.01) in distinguishing between A19 and A19BG immune sera and were identified as differential diagnostic antigens for the A19BG vaccine candidate. In conclusion, using comparative proteomics and antibody profiling, protein components and signature antigens were identified for the ghost vaccine candidate A19BG, which are valuable for further developing the vaccine and its monitoring assays.
Collapse
Affiliation(s)
- Chuan-Yu He
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
| | - Jiang-Hua Yang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Yin-Bo Ye
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Hai-Long Zhao
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
| | - Meng-Zhi Liu
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
| | - Qi-Lin Yang
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
| | - Bao-Shan Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Sun He
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
| | - Ze-Liang Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
- Technology Center, Tecon Biological Co., Ltd., Urumqi, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Innovative Institute of Zoonoses, Inner Mongolia Minzu University, Tongliao, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Key Laboratory of Tropical Diseases Control, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
30
|
Pandey M, Choudhury H, Vijayagomaran PA, Lian PNP, Ning TJ, Wai NZ, Xian-Zhuang N, Le Er C, Rahmah NSN, Kamaruzzaman NDB, Mayuren J, Candasamy M, Gorain B, Chawla PA, Amin MCIM. Recent Update on Bacteria as a Delivery Carrier in Cancer Therapy: From Evil to Allies. Pharm Res 2022; 39:1115-1134. [PMID: 35386012 PMCID: PMC8985562 DOI: 10.1007/s11095-022-03240-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/18/2022] [Indexed: 01/19/2023]
Abstract
Cancer is associated with a comprehensive burden that significantly affects patient’s quality of life. Even though patients’ disease condition is improving following conventional therapies, researchers are studying alternative tools that can penetrate solid tumours to deliver the therapeutics due to issues of developing resistance by the cancer cells. Treating cancer is not the only the goal in cancer therapy; it also includes protecting non-cancerous cells from the toxic effects of anti-cancer agents. Thus, various advanced techniques, such as cell-based drug delivery, bacteria-mediated therapy, and nanoparticles, are devised for site-specific delivery of drugs. One of the novel methods that can be targeted to deliver anti-cancer agents is by utilising genetically modified non-pathogenic bacterial species. This is due to the ability of bacterial species to multiply selectively or non-selectively on tumour cells, resulting in biofilms that leads to disruption of metastasis process. In preclinical studies, this technology has shown significant results in terms of efficacy, and some are currently under investigation. Therefore, researchers have conducted studies on bacteria transporting the anti-cancer drug to targeted tumours. Alternatively, bacterial ghosts and bacterial spores are utilised to deliver anti-cancer drugs. Although in vivo studies of bacteria-mediated cancer therapy have shown successful outcome, further research on bacteria, specifically their targeting mechanism, is required to establish a complete clinical approach in cancer treatment. This review has focused on the up-to-date understanding of bacteria as a therapeutic carrier in the treatment of cancer as an emerging field.
Collapse
Affiliation(s)
- Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia.
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | | | - Pauline Ng Poh Lian
- School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Tan Jing Ning
- School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Ng Zing Wai
- School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Ng Xian-Zhuang
- School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Chong Le Er
- School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | | | | | - Jayashree Mayuren
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, 57000 Bukit Jalil, Kuala Lumpur, Malaysia
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Pooja A Chawla
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy Moga, Ghall Kalan, Punjab, India
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| |
Collapse
|
31
|
Yin T, Diao Z, Blum NT, Qiu L, Ma A, Huang P. Engineering Bacteria and Bionic Bacterial Derivatives with Nanoparticles for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104643. [PMID: 34908239 DOI: 10.1002/smll.202104643] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/25/2021] [Indexed: 06/14/2023]
Abstract
Natural bacteria are interesting subjects for cancer treatments owing to their unique autonomy-driven and hypoxic target properties. Genetically modified bacteria (such as bacteria with msbB gene and aroA gene modifications) can effectively cross sophisticated physiological barriers and transport antitumor agents into deep tumor tissues, and they have good biosafety. Additionally, bacteria can secrete cytokines (such as interleukin-224, interferon-gamma [IFN-γ], and interleukin-1β) and activate antitumor immune responses in the tumor microenvironment, resulting in tumor inhibition. All of these characteristics can be easily utilized to develop synergistic antitumor strategies by combining bacteria-based agents with other therapeutic approaches. Herein, representative studies of bacteria-instructed multimodal synergistic cancer therapy are introduced (e.g., photothermal therapy, chemoimmunotherapy, photodynamic therapy, and photocontrolled bacterial metabolite therapy), and their key advantages are systematically expounded. The current challenges and future prospects in advancing the development of bacteria-based micro/nanomedicines in the field of synthetic biology research are also emphasized, which will hopefully promote the development of related bacteria-based cancer therapies.
Collapse
Affiliation(s)
- Ting Yin
- Guangdong Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Zhenying Diao
- Guangdong Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Nicholas Thomas Blum
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, P. R. China
| | - Long Qiu
- Guangdong Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Aiqing Ma
- Guangdong Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, P. R. China
| |
Collapse
|
32
|
Rabea S, Yassin AS, Mohammed AF, Salem-Bekhit MM, Alanazi FK, Esmail EA, Moneib NA, Hashem AEM. Immunological characterization of the chemically prepared ghosts of Salmonella Typhimurium as a vaccine candidate. BMC Vet Res 2022; 18:72. [PMID: 35180858 PMCID: PMC8855557 DOI: 10.1186/s12917-021-03112-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Bacterial ghosts are the evacuated bacterial cellular membranes from most of the genetic and protein contents which preserved their surface characters. Recently, bacterial ghosts exploited for different biomedical applications, for instance, vaccination. The purpose of this study is to measure the immunogenic protective response of bacterial ghosts of Salmonella Typhimurium in animals and to allow future testing this response in humans. The immunologic response was qualitatively, quantitatively, and functionally measured. We have measured the humoral and cellular immune responses, such as immunoglobulins elevation (IgG), increased granulocytes, serum antibacterial activity, clearance of virulence in feces and liver, and the survival rate. RESULTS The bacterial ghosts' vaccine was able to protect 100% of subcutaneously vaccinated rats and 75% of adjuvant subcutaneously vaccinated rats. The lowest survival rate was in the orally vaccinated group (25%). The maximum level of serum IgG titers, as well as serum and feces bactericidal activity (100% eradication), was exhibited in the subcutaneously vaccinated group with adjuvant vaccines followed by the subcutaneously vaccinated one. Additionally, the highest granulocytes' number was observed in the adjuvant vaccine subcutaneously immunized group. The bacterial load in liver homogenate was eliminated in the subcutaneously vaccinated rats after the virulence challenge. CONCLUSIONS The bacterial ghosts of Salmonella enterica serovar Typhimurium that prepared by Tween 80 Protocol showed an effective vaccine candidate that protected animals, eliminated the virulence in feces and liver. These findings show that chemically induced bacterial ghosts of Salmonella Typhimurium can be a promising vaccine.
Collapse
Affiliation(s)
- Sameh Rabea
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia.
| | - Aymen S Yassin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11652, Egypt
| | - Aly Fahmy Mohammed
- The International Center for Advanced Researchers (ICTAR-Egypt), Cairo, Egypt
| | - Mounir M Salem-Bekhit
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11651, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Eman Amin Esmail
- The Holding Company for Production of Vaccines, Sera And Drugs (VACSERA), Cairo, Egypt
| | - Nayera A Moneib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11652, Egypt
| | - Abd Elgawad M Hashem
- Faculty of Pharmacy, British University in Egypt (BUE), Cairo, 11837 - P.O. Box 43, Egypt
| |
Collapse
|
33
|
Abdelnaby MA, Shoueir KR, Ghazy AA, Abdelhamid SM, El Kemary MA, Mahmoud HE, Baraka K, Abozahra RR. Synthesis and evaluation of metallic nanoparticles-based vaccines against Candida albicans infections. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
34
|
Johnson AP, Sabu C, Nivitha K, Sankar R, Shirin VA, Henna T, Raphey V, Gangadharappa H, Kotta S, Pramod K. Bioinspired and biomimetic micro- and nanostructures in biomedicine. J Control Release 2022; 343:724-754. [DOI: 10.1016/j.jconrel.2022.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
|
35
|
García-Álvarez R, Vallet-Regí M. Bacteria and cells as alternative nano-carriers for biomedical applications. Expert Opin Drug Deliv 2022; 19:103-118. [PMID: 35076351 PMCID: PMC8802895 DOI: 10.1080/17425247.2022.2029844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Nano-based systems have received a lot of attention owing to their particular properties and, hence, have been proposed for a wide variety of biomedical applications. These nanosystems could be potentially employed for diagnosis and therapy of different medical issues. Although these nanomaterials are designed for specific tasks, interactions, and transformations when administered to the human body affect their performance and behavior. In this regard, bacteria and other cells have been presented as alternative nanocarriers. These microorganisms can be genetically modified and customized for a more specific therapeutic action and, in combination with nanomaterials, can lead to bio-hybrids with a unique potential for biomedical purposes. AREAS COVERED Literature regarding bacteria and cells employed in combination with nanomaterials for biomedical applications is revised and discussed in this review. The potential as well as the limitations of these novel bio-hybrid systems are evaluated. Several examples are presented to show the performance of these alternative nanocarriers. EXPERT OPINION Bio-hybrid systems have shown their potential as alternative nanocarriers as they contribute to better performance than traditional nano-based systems. Nevertheless, their limitations must be studied, and advantages and drawbacks assessed before their application to medicine.
Collapse
Affiliation(s)
- Rafaela García-Álvarez
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| |
Collapse
|
36
|
Salem-Bekhit MM, Youssof AME, Alanazi FK, Aleanizy FS, Abdulaziz A, Taha EI, Amara AAAF. Bacteria from Infectious Particles to Cell Based Anticancer Targeted Drug Delivery Systems. Pharmaceutics 2021; 13:1984. [PMID: 34959266 PMCID: PMC8706210 DOI: 10.3390/pharmaceutics13121984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Bacterial ghosts (BGs) are empty cell envelopes of nonliving evacuated bacterial cells. They are free from their cytoplasmic contents; however, they sustain their cellular 3D morphology and antigenic structures, counting on bioadhesive properties. Lately, they have been tested as an advanced drug delivery system (DDS) for different materials like DNA, peptides, or drugs, either single components or combinations. Different studies have revealed that, BG DDS were paid the greatest attention in recent years. The current review explores the impact of BGs on the field of drug delivery and drug targeting. BGs have a varied area of applications, including vaccine and tumor therapy. Moreover, the use of BGs, their synthesis, their uniqueness as a delivery system and application principles in cancer are discussed. Furthermore, the safety issues of BGs and stability aspects of using ghost bacteria as delivery systems are discussed. Future perspective efforts that must be followed for this important system to continue to grow are important and promising.
Collapse
Affiliation(s)
- Mounir M. Salem-Bekhit
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah M. E. Youssof
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fars K. Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fadilah Sfouq Aleanizy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
| | - Alsuwyeh Abdulaziz
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ehab I. Taha
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
| | - Amro Abd Al Fattah Amara
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria 21934, Egypt;
| |
Collapse
|
37
|
Xie S, Zhang P, Zhang Z, Liu Y, Chen M, Li S, Li X. Bacterial navigation for tumor targeting and photothermally-triggered bacterial ghost transformation for spatiotemporal drug release. Acta Biomater 2021; 131:172-184. [PMID: 34171461 DOI: 10.1016/j.actbio.2021.06.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Cancer chemotherapy is confronted with challenges regarding the effective delivery of chemotherapeutics into tumor cells after systemic administration. Herein, we propose a strategy to load drugs into probiotic E. coli Nissle 1917 (EcN) for self-guided navigation to tumor tissues and subsequently release the drugs with in situ transformation into bacterial ghosts (BGs). Chemotherapeutic agent 5-fluorouracil (FU) and macrophage phenotype regulator zoledronic acid (ZOL) are loaded into EcN through electroporation, followed by decoration of Au nanorods on the ECN surface to construct EcNZ/F@Au. High loading levels of 5FU (8.8%) and ZOL (10.5%) are achieved as well as high retention rates of bacterial viability (87%) and motion velocity (88%). Under near infrared (NIR) illumination the photothermal effect of Au nanorods elevates the local temperature to induce the transformation of live EcN into BGs. The created transmembrane channels initiate the gradual drug release from BGs, thus representing the first attempt to control the drug release via a biological evolution. An intermittent NIR illumination causes stepwise increases in the BG formation and drug release, which could implement an external on-off control and spatiotemporal drug release. Self-guided motion of EcN promotes efficient extravasation across blood vessels and preferential accumulation of drugs in tumors. In addition to the chemotherapeutic effect of FU, the local release of ZOL from EcNZ/F@Au enhances valid polarization of tumor-associated macrophages toward the M1 phenotype and an effective production of proinflammatory cytokines, leading to a synergistic efficacy on tumor growth inhibition. Thus, this study demonstrates a feasible strategy to integrate chemotherapy, immunotherapy, and photothermal effects in a concise manner for effective cancer treatment with few side effects. STATEMENT OF SIGNIFICANCE: Bacteria are capable to trace and colonize in hypoxic tumor tissues. Bacterial drug carriers indicate limitations in efficient drug loading and effective release modulation. Herein, we propose a strategy to load drugs into bacteria for self-guided delivery and subsequently release the drugs in tumors with in situ transformation into bacterial ghost (BGs). Drugs are loaded into live bacteria through electroporation and Au nanorods are decorated on the bacterial surface, wherein the photothermal effect, chemotherapy, and immunotherapy are integrated in a concise manner. NIR illmumination of Au nanorods elevates the local temparature, induces the BG tranformation, and activates the spatiotemporal drug release, representing the first attempt of release modulation via a biological evolution.
Collapse
|
38
|
Li Z, Wang Y, Liu J, Rawding P, Bu J, Hong S, Hu Q. Chemically and Biologically Engineered Bacteria-Based Delivery Systems for Emerging Diagnosis and Advanced Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102580. [PMID: 34347325 DOI: 10.1002/adma.202102580] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/10/2021] [Indexed: 06/13/2023]
Abstract
Bacteria are one of the main groups of organisms, which dynamically and closely participate in human health and disease development. With the integration of chemical biotechnology, bacteria have been utilized as an emerging delivery system for various biomedical applications. Given the unique features of bacteria such as their intrinsic biocompatibility and motility, bacteria-based delivery systems have drawn wide interest in the diagnosis and treatment of various diseases, including cancer, infectious diseases, kidney failure, and hyperammonemia. Notably, at the interface of chemical biotechnology and bacteria, many research opportunities have been initiated, opening a promising frontier in biomedical application. Herein, the current synergy of chemical biotechnology and bacteria, the design principles for bacteria-based delivery systems, the microbial modulation, and the clinical translation are reviewed, with a special focus on the emerging advances in diagnosis and therapy.
Collapse
Affiliation(s)
- Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Jun Liu
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Piper Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, Wisconsin Center for NanoBioSystems, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison (UW-Madison), Madison, Wisconsin, 53705, USA
| |
Collapse
|
39
|
Liang J, Zhao X. Nanomaterial-based delivery vehicles for therapeutic cancer vaccine development. Cancer Biol Med 2021; 18:j.issn.2095-3941.2021.0004. [PMID: 33979069 PMCID: PMC8185868 DOI: 10.20892/j.issn.2095-3941.2021.0004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022] Open
Abstract
Nanomaterial-based delivery vehicles such as lipid-based, polymer-based, inorganics-based, and bio-inspired vehicles often carry distinct and attractive advantages in the development of therapeutic cancer vaccines. Based on various delivery vehicles, specifically designed nanomaterials-based vaccines are highly advantageous in boosting therapeutic and prophylactic antitumor immunities. Specifically, therapeutic vaccines featuring unique properties have made major contributions to the enhancement of antigen immunogenicity, encapsulation efficiency, biocompatibility, and stability, as well as promoting antigen cross-presentation and specific CD8+ T cell responses. However, for clinical applications, tumor-associated antigen-derived vaccines could be an obstacle, involving immune tolerance and deficiency of tumor specificities, in achieving maximum therapeutic indices. However, when using bioinformatics predictions with emerging innovations of in silico tools, neoantigen-based therapeutic vaccines might become potent personalized vaccines for tumor treatments. In this review, we summarize the development of preclinical therapeutic cancer vaccines and the advancements of nanomaterial-based delivery vehicles for cancer immunotherapies, which provide the basis for a personalized vaccine delivery platform. Moreover, we review the existing challenges and future perspectives of nanomaterial-based personalized vaccines for novel tumor immunotherapies.
Collapse
Affiliation(s)
- Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Svenskaya Y, Garello F, Lengert E, Kozlova A, Verkhovskii R, Bitonto V, Ruggiero MR, German S, Gorin D, Terreno E. Biodegradable polyelectrolyte/magnetite capsules for MR imaging and magnetic targeting of tumors. Nanotheranostics 2021; 5:362-377. [PMID: 33850694 PMCID: PMC8040826 DOI: 10.7150/ntno.59458] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/15/2021] [Indexed: 01/14/2023] Open
Abstract
Rationale: The tireless research for effective drug delivery approaches is prompted by poor target tissue penetration and limited selectivity against diseased cells. To overcome these issues, various nano- and micro-carriers have been developed so far, but some of them are characterized by slow degradation time, thus hampering repeated drug administrations. The aim of this study was to pursue a selective delivery of magnetic biodegradable polyelectrolyte capsules in a mouse breast cancer model, using an external magnetic field. Methods: Four different kinds of magnetic polyelectrolyte capsules were fabricated via layer-by-layer assembly of biodegradable polymers on calcium carbonate templates. Magnetite nanoparticles were embedded either into the capsules' shell (sample S) or both into the shell and the inner volume of the capsules (samples CnS, where n is the number of nanoparticle loading cycles). Samples were first characterized in terms of their relaxometric and photosedimentometric properties. In vitro magnetic resonance imaging (MRI) experiments, carried out on RAW 264.7 cells, allowed the selection of two lead samples that proceeded for the in vivo testing on a mouse breast cancer model. In the set of in vivo experiments, an external magnet was applied for 1 hour following the intravenous injection of the capsules to improve their delivery to tumor, and MRI scans were acquired at different time points post administration. Results: All samples were considered non-cytotoxic as they provided more than 76% viability of RAW 264.7 cells upon 2 h incubation. Sample S appeared to be the most efficient in terms of T2-MRI contrast, but the less sensitive to external magnet navigation, since no difference in MRI signal with and without the magnet was observed. On the other side, sample C6S was efficiently delivered to the tumor tissue, with a three-fold T2-MRI contrast enhancement upon the external magnet application. The effective magnetic targeting of C6S capsules was also confirmed by the reduction in T2-MRI contrast in spleen if compared with the untreated with magnet mice values, and the presence of dense and clustered iron aggregates in tumor histology sections even 48 h after the magnetic targeting. Conclusion: The highlighted strategy of magnetic biodegradable polyelectrolyte capsules' design allows for the development of an efficient drug delivery system, which through an MRI-guided externally controlled navigation may lead to a significant improvement of the anticancer chemotherapy performance.
Collapse
Affiliation(s)
- Yulia Svenskaya
- Remote Controlled Systems for Theranostics laboratory, Research and Educational Institute of Nanostructures and Biosystems, Saratov State University, 410012 Saratov, Russia
| | - Francesca Garello
- Molecular and Preclinical Imaging Centres, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Ekaterina Lengert
- Remote Controlled Systems for Theranostics laboratory, Research and Educational Institute of Nanostructures and Biosystems, Saratov State University, 410012 Saratov, Russia
| | - Anastasiia Kozlova
- Biomedical Photoacoustics Laboratory, Saratov State University, 410012 Saratov, Russia
| | - Roman Verkhovskii
- Biomedical Photoacoustics Laboratory, Saratov State University, 410012 Saratov, Russia
| | - Valeria Bitonto
- Molecular and Preclinical Imaging Centres, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Maria Rosaria Ruggiero
- Molecular and Preclinical Imaging Centres, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Sergey German
- Laboratory of Optics and Spectroscopy of Nanoobjects, Institute of Spectroscopy of the RAS, Troitsk 108840, Russia.,Center of Photonics and Quantum Materials, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Dmitry Gorin
- Center of Photonics and Quantum Materials, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Enzo Terreno
- Molecular and Preclinical Imaging Centres, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
41
|
Soleymani S, Tavassoli A, Hashemi Tabar G, Kalidari GA, Dehghani H. Design, development, and evaluation of the efficacy of a nucleic acid-free version of a bacterial ghost candidate vaccine against avian pathogenic E. coli (APEC) O78:K80 serotype. Vet Res 2020; 51:144. [PMID: 33298146 PMCID: PMC7724879 DOI: 10.1186/s13567-020-00867-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
One of the major bacterial infectious diseases in the poultry industry is avian pathogenic Escherichia coli (APEC), which causes colibacillosis in chickens. To develop a novel nucleic acid-free bacterial ghost (BG) vaccine against the O78:K80 serotype of APEC, in this study we constructed a plasmid that harbored E-lysis and S nuclease (SNUC). Following the expression, the O78:K80 bacteria lost all of their cytoplasmic content and nucleic acids by enzymatic digestion. The functionality of these two proteins in the production procedure of bacterial ghosts was confirmed by monitoring the number of colonies, scanning electron microscopy imaging, gel electrophoresis of genomic DNA, and qPCR on the plasmid content of bacterial ghosts. The protective efficacy of the ghost vaccine generated from O78:K80 serotype of APEC was tested in chickens by injection and inhalation routes and compared with that in chickens that received the injection of a killed vaccine. The O78:K80 BG vaccine candidate, used as injection and inhalation, in comparison with the killed vaccine, triggered higher proinflammatory cytokine expression including IL-6, IL-1β, and TNFSF15; a higher level of antibody-dependent humoral (IgY and IgA) and cellular immune responses (IFNγ and lymphocyte proliferation); and lower lesion scores. According to the results of this study, we suggest that the bacterial ghost technology has the potential to be applied for the development of novel vaccines against avian colibacillosis. This technology provides an effective and reliable approach to make multivalent vaccines for more prevalent APEC strains involved in the establishment of this infectious disease in the poultry industry.
Collapse
Affiliation(s)
- Safoura Soleymani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amin Tavassoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Gholamreza Hashemi Tabar
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Gholam Ali Kalidari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran. .,Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran. .,Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
42
|
Ji S, Gong Q, Zhang W, Zheng J, Peng B, Yang M. Recombinant Vibrio parahaemolyticus ghosts protect zebrafish against infection by Vibrio species. FISH & SHELLFISH IMMUNOLOGY 2020; 107:64-72. [PMID: 33038509 DOI: 10.1016/j.fsi.2020.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/07/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Aquatic animals are frequently threated by bacterial pathogens. The most economic and efficient protection against bacterial infection are through vaccine immunization. The various serotypes of the pathogens, such as Vibrios, hurdle the development of the vaccines, especially polyvalent vaccines. Here, we demonstrate that recombinant bacterial ghost is a good candidate for multivalent vaccine. By expressing PhiX174 gene E alone or co-expressing the gene E with two genes encoding outer membrane proteins (VP1667 and VP2369) in V. parahaemolyticus, we generated the recombinant V. parahaemolyticus ghosts VPG and rVPGs respectively. Fish immunized with either VPG or rVPG showed increased survival against the infection by either V. parahaemolyticus or V. alginolyticus, with a better protective effect by immunization with rVPG. Our furthermore studies show that rVPG stimulates stronger innate immune responses by increasing the expression of tnfα, il1β, il6, il8 and il10 as well as that of c3b, lyz, and tlr5, the key players linking the innate and adaptive immune responses upon microbial stimulation. In summary, VPG and rVPG can protect zebrafish against the infection from at least two Vibrio species, suggesting its potential value for further aquaculture vaccines development.
Collapse
Affiliation(s)
- Shengle Ji
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University, Hangzhou, 311300, China
| | - Qiyang Gong
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Wenwen Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Bo Peng
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Menghua Yang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University, Hangzhou, 311300, China.
| |
Collapse
|
43
|
Alanazi FK, Alsuwyeh AA, Haq N, Salem-Bekhit MM, Al-Dhfyan A, Shakeel F. Vision of bacterial ghosts as drug carriers mandates accepting the effect of cell membrane on drug loading. Drug Dev Ind Pharm 2020; 46:1716-1725. [PMID: 32893682 DOI: 10.1080/03639045.2020.1820039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/27/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
Abstract
The use of bacterial ghosts (BGs) for drug delivery is an extremely fascinating perspective especially with the inherited efficient target-ability to specialized tissues. Trafficking of drug molecules across the outer membrane of Gram-negative bacteria are important to be understood for both loading (influx) and drug release (efflux). In this study, Escherichia coli (E. coli) BGs were prepared using modified protocol sponge-like reduced protocol (SLRP) which was used for loading of doxorubicin (DOX). First time in the literature, different possible factors affecting DOX loading from BGs were examined in this study. These factors including drug concentration, temperature, pH gradient, incubation time and tonicity, are proposed to effect on drug loading into E. coli BGs. Results of optimum effect from accompanied factors were found to be 10 mg/mL as DOX concentration at pH 6 with tonicity of 0.7% incubated overnight at 4 °C. After gather all factors, the amount of DOX loaded inside the BGs was recorded as 37.58%. The in vitro release studies of DOX loaded BGs over time showed a burst initial release rate of 26.75% at the first 12 h followed by a period of sustained release lasting for 16 days to give maximum release rate of 58.04%. Remarkably, DOX loaded in BG showed more apoptosis (55%) than control and DOX solution. Overall, the results indicated the presence of some important factors to be controlled when loading drugs into BGs. Also, data showed the future possibility of utilizing BGs to deliver DOX to colon cancer cells.
Collapse
Affiliation(s)
- Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz A Alsuwyeh
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nazrul Haq
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mounir M Salem-Bekhit
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Abdullah Al-Dhfyan
- Stem Cell Therapy Program, King Faisal Specialized Hospital and Research Center, Riyadh, Saudi Arabia
| | - Faiyaz Shakeel
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
Sherif AY, Harisa GI, Alanazi FK, Youssof AME. Engineering of Exosomes: Steps Towards Green Production of Drug Delivery System. Curr Drug Targets 2020; 20:1537-1549. [PMID: 31309889 DOI: 10.2174/1389450120666190715104100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/09/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022]
Abstract
Targeting of therapeutic agents to their specific site of action not only increases the treatment efficacy, but also reduces systemic toxicity. Therefore, various drug delivery systems (DDSs) have been developed to achieve this target. However, most of those DDSs have several issues regarding biocompatibility and environmental hazard. In contrast to the synthetic DDSs, exosome-based natural carriers are biocompatible, biodegradable and safe for the environment. Since exosomes play a role in intercellular communication, they have been widely utilized as carriers for different therapeutic agents. This article was aimed to provide an overview of exosomes as an environment-friendly DDS in terms of engineering, isolation, characterization, application and limitation.
Collapse
Affiliation(s)
- Abdelrahman Y Sherif
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Biochemistry, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah M E Youssof
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Yaghoubi A, Khazaei M, Jalili S, Hasanian SM, Avan A, Soleimanpour S, Cho WC. Bacteria as a double-action sword in cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188388. [PMID: 32589907 DOI: 10.1016/j.bbcan.2020.188388] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023]
Abstract
Bacteria have long been known as one of the primary causative agents of cancer, however, recent studies suggest that they can be used as a promising agent in cancer therapy. Because of the limitations that conventional treatment faces due to the specific pathophysiology and the tumor environment, there is a great need for the new anticancer therapeutic agents. Bacteriotherapy utilizes live, attenuated strains or toxins, peptides, bacteriocins of the bacteria in the treatment of cancer. Moreover, they are widely used as a vector for delivering genes, peptides, or drugs to the tumor target. Interestingly, it was found that their combination with the conventional therapeutic approaches may enhance the treatment outcome. In the genome editing era, it is feasible to develop a novel generation of therapeutic bacteria with fewer side effects and more efficacy for cancer therapy. Here we review the current knowledge on the dual role of bacteria in the development of cancer as well as cancer therapy.
Collapse
Affiliation(s)
- Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saba Jalili
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hasanian
- Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, SAR, China.
| |
Collapse
|
46
|
Alizadeh S, Esmaeili A, Barzegari A, Rafi MA, Omidi Y. Bioengineered smart bacterial carriers for combinational targeted therapy of solid tumours. J Drug Target 2020; 28:700-713. [PMID: 32116051 DOI: 10.1080/1061186x.2020.1737087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite many endeavours for the development of new anticancer drugs, effective therapy of solid tumours remains a challenging issue. The current cancer chemotherapies may associate with two important limitations, including the lack/trivial specificity of treatment modalities towards diseased cells/tissues resulting in undesired side effects, and the emergence of drug-resistance mechanisms by tumour cells causing the failure of the treatment. Much attention, therefore, has currently been paid to develop smart and highly specific anticancer agents with maximal therapeutic impacts and minimal side effects. Among various strategies used to target cancer cells, bacteria-based cancer therapies (BCTs) have been validated as potential gene/drug delivery carriers, which can also be engineered to be used in diagnosis processes. They can be devised to selectively target the tumour microenvironment (TME), within which they may preferentially proliferate in the necrotic and anaerobic parts - often inaccessible to other therapeutics. BCTs are capable to sense and respond to the environmental signals, upon which they are considered as smart microrobots applicable in the controlled delivery of therapeutic agents to the TME. In this review, we aimed to provide comprehensive insights into the potentials of the bioengineered bacteria as smart and targeted bio-carriers and discuss their applications in cancer therapy.
Collapse
Affiliation(s)
- Siamak Alizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.,Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad A Rafi
- Department of Neurology, Sidney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Qin M, Du G, Sun X. Biomimetic cell-derived nanocarriers for modulating immune responses. Biomater Sci 2020; 8:530-543. [PMID: 31750453 DOI: 10.1039/c9bm01444f] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we summarize various applications of biomimetic carriers in modulating immune responses and discuss the future perspectives.
Collapse
Affiliation(s)
- Ming Qin
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| |
Collapse
|
48
|
Liu L, He H, Liu J. Advances on Non-Genetic Cell Membrane Engineering for Biomedical Applications. Polymers (Basel) 2019; 11:E2017. [PMID: 31817418 PMCID: PMC6961000 DOI: 10.3390/polym11122017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapeutics are very promising modalities to address many unmet medical needs, including genetic engineering, drug delivery, and regenerative medicine as well as bioimaging. To enhance the function and improve the efficacy of cell-based therapeutics, a variety of cell surface engineering strategies (genetic engineering and non-genetic engineering) are developed to modify the surface of cells or cell-based therapeutics with some therapeutic molecules, artificial receptors, and multifunctional nanomaterials. In comparison to complicated procedures and potential toxicities associated with genetic engineering, non-genetic engineering strategies have emerged as a powerful and compatible complement to traditional genetic engineering strategies for enhancing the function of cells or cell-based therapeutics. In this review, we will first briefly summarize key non-genetic methodologies including covalent chemical conjugation (surface reactive groups-direct conjugation, and enzymatically mediated and metabolically mediated indirect conjugation) and noncovalent physical bioconjugation (biotinylation, electrostatic interaction, and lipid membrane fusion as well as hydrophobic insertion), which have been developed to engineer the surface of cell-based therapeutics with various materials. Next, we will comprehensively highlight the latest advances in non-genetic cell membrane engineering surrounding different cells or cell-based therapeutics, including whole-cell-based therapeutics, cell membrane-derived therapeutics, and extracellular vesicles. Advances will be focused specifically on cells that are the most popular types in this field, including erythrocytes, platelets, cancer cells, leukocytes, stem cells, and bacteria. Finally, we will end with the challenges, future trends, and our perspectives of this relatively new and fast-developing research field.
Collapse
Affiliation(s)
- Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
49
|
Construction of a novel tetravalent dengue vaccine with a Salmonella Typhimurium bacterial ghost and evaluation of its immunogenicity and protective efficacy using a murine model. Vaccine 2019; 38:916-924. [PMID: 31706812 DOI: 10.1016/j.vaccine.2019.10.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 01/27/2023]
Abstract
Efforts to develop a safe, effective, and affordable dengue vaccine have focused on providing simultaneous immunity against all four serotypes of the dengue virus (DENV). In the current study, Salmonella Typhimurium (ST) lysed by gene E activation was genetically constructed to deliver the envelope protein domain III (EDIII) of all four serotypes of DENV using a foreign antigen delivery and expression vector, pJHL184. Each DENV-EDIII protein expressed in the constructed strain was validated by immunoblot analysis. To assess the immunogenicity and protective efficacy of the constructs against dengue infection, BALB/c mice were injected once orally with either the individual ST-EDIII constructs or a mix of all four ST-EDIII constructs followed by intramuscular administration of the purified EDIII protein. Significantly elevated titers of EDIII-specific IgG, IgG1, and IgG2a were observed in the immunized mice (P < 0.01). Furthermore, lymphocyte proliferative activity and CD3+CD4+ T-cell subpopulations increased significantly in vitro in re-pulsed splenic T cells compared with those from non-immunized mice. In addition, a lower viral load was detected in the BG-EDIII vaccinated group after challenge with DENV-infected K562 cells. Collectively, the results demonstrate that DENV-EDIII expressed in the inactivated ST strain could induce robust humoral and cell-mediated immunity specific to the target antigen and could provide significant protective potential.
Collapse
|
50
|
Hoseini Shahidi R, Hashemi Tabar G, Bassami MR, Jamshidi A, Dehghani H. The design and application of a bacterial ghost vaccine to evaluate immune response and defense against avian pathogenic Escherichia coli O2:K1 serotype. Res Vet Sci 2019; 125:153-161. [PMID: 31228739 DOI: 10.1016/j.rvsc.2019.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 05/10/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022]
Abstract
An Escherichia coli (E. coli) O2:K1 bacterial ghost was produced by controlled expression of bacteriophage PhiX 174 lysis gene E. Temperature controlled expression of this gene caused tunnels and holes in the cell wall of E. coli O2:K1 bacterium, leading to loss of cytoplasmic contents. Formation of E. coli O2:K1 ghost was confirmed by scanning electron microscopy and determination of colony forming units. To evaluate the efficiency of this bacterial ghost vaccine to elicit cellular and humoral immune responses, 85 one day old chickens from Ross 308 breed were divided into the following 5 groups; group 1 (non-immunized control), group 2 (vaccine administered by injection of E. coli O2:K1 killed vaccine), group 3 (vaccine administered by injection of E. coli O2:K1 ghost), group 4 (vaccine administered by inhalation of E. coli O2:K1 ghost), and group 5 (neither immunized, nor challenged as negative control). The groups of 2, 3, and 4 were received vaccines at days 7, 14, and 22. Groups 1 to 4 were challenged with the wild type at day 33. Evaluation of post-mortem lesions and immune responses in all groups showed that chicken injected with the killed vaccine and the bacterial ghost had the best protection. These findings suggest that this bacterial ghost has the potential to be used as a poultry colibacillosis vaccine.
Collapse
Affiliation(s)
- Reza Hoseini Shahidi
- Biotechnology Division, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Gholamreza Hashemi Tabar
- Biotechnology Division, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mohammad Reza Bassami
- Biotechnology Division, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Division of Poultry Diseases, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abdollah Jamshidi
- Division of Food Hygiene, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Biotechnology Division, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Division of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|