1
|
Ibrahim R, Aranjani JM, Kalikot Valappil V, Nair G. Unveiling the potential bacteriophage therapy: a systematic review. Future Sci OA 2025; 11:2468114. [PMID: 39976508 PMCID: PMC11845108 DOI: 10.1080/20565623.2025.2468114] [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: 08/06/2024] [Accepted: 01/21/2025] [Indexed: 02/23/2025] Open
Abstract
INTRODUCTION Antimicrobial resistance renders conventional therapy, demanding the need for alternative therapeutic techniques. A potential strategy for treating infections caused by multi-drug-resistant bacteria is using bacteriophages, viruses that only multiply and infect specific bacteria. This review aims to evaluate the findings of clinical studies on phage therapy for bacterial illnesses. METHODS A comprehensive search method was utilized to identify 11 appropriate trials, which were then assessed for safety, efficacy, and treatment outcomes. The Joann-Briggs-Institute checklist and PRISMA criteria were used to evaluate these studies thoroughly. The results were summarized by extracting and analyzing data on trial design, treatment outcomes, safety profiles, and therapeutic effectiveness. RESULTS Phage treatment had a strong safety profile, with few side effects recorded across many routes, including oral, intravenous, and topical. Clinical studies demonstrated its effectiveness in lowering bacterial loads, resolving infections, and destroying biofilms. However, diversity in trial designs hampered the generalizability of the findings. CONCLUSION This study emphasizes the promise of phage therapy as a safe and efficient treatment for bacterial-illnesses. Despite its potential, there are still significant gaps in clinical application, long-term efficacy assessment, and trial standardization. Addressing these issues is critical to developing phage therapy as an effective alternative treatment for multidrug-resistant-illnesses.
Collapse
Affiliation(s)
- Rafwana Ibrahim
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Vipin Kalikot Valappil
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Government Medical College, Kannur, India
| | - Gouri Nair
- Department of Pharmacology, Faculty of Pharmacy, Ramaiah University of Applied Sciences, Bengaluru, India
| |
Collapse
|
2
|
Rodríguez-Arellano SN, González-Gómez JP, Gomez-Gil B, González-Ávila M, Palomera-Hernández JR, Barrón-Cabrera E, Vergara-Jiménez MDJ, Chaidez C. A Two-Phage Cocktail Modulates Gut Microbiota Composition and Metabolic Profiles in an Ex Vivo Colon Model. Int J Mol Sci 2025; 26:2805. [PMID: 40141446 PMCID: PMC11942677 DOI: 10.3390/ijms26062805] [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: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Bacteriophage therapy is a promising approach for targeting antibiotic-resistant bacteria and modulating gut microbiota in metabolic diseases such as obesity. This study evaluated the impact of a two-phage cocktail on an ex vivo colonic simulation model of gut microbiota derived from obese individuals, both in its normalized state and after enrichment with Enterobacter cloacae, an obesity-related bacteria. Microbiological analyses confirmed that the phage cocktail remained active throughout the colonic regions over three digestion cycles and effectively reduced enterobacterial populations in the enriched microbiota. Metabarcoding of the 16S rRNA gene revealed that phage therapy did not significantly alter the abundance of dominant genera, but selectively reduced E. cloacae across all colonic regions. Alpha diversity was significantly affected only in the enriched microbiota, while beta diversity analysis indicated significant compositional shifts during therapy, with reduced dispersion in the final treatment stage. Short-chain fatty acid profiling demonstrated region- and group-specific metabolic responses, with increased lactic and butyric acid concentrations in the ascending colon of the enriched microbiota following phage treatment. This study provides the first ex vivo evidence that a two-phage cocktail can selectively eliminate E. cloacae while preserving overall microbiota structure and functionality. These findings establish a foundation for future in vivo studies exploring the role of phage therapy in reshaping gut microbial communities and metabolic profiles, highlighting its potential as a precision tool for managing gut dysbiosis in metabolic disorders.
Collapse
Affiliation(s)
| | - Jean Pierre González-Gómez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Culiacan 80110, Sinaloa, Mexico
| | - Bruno Gomez-Gil
- Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlan 82112, Sinaloa, Mexico
| | - Marisela González-Ávila
- Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Juan Ramón Palomera-Hernández
- Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Elisa Barrón-Cabrera
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacan 80019, Sinaloa, Mexico
| | | | - Cristobal Chaidez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Culiacan 80110, Sinaloa, Mexico
| |
Collapse
|
3
|
Lee SY, Michell KA, Butler MM, Smith BT, Woolf EK, Holmes SC, Grabos LE, Vazquez AR, Isweiri H, Bunning M, Uchanski ME, Rao S, Newman SE, Weir TL, Johnson SA. Feasibility and Tolerability of Daily Microgreen Consumption in Healthy Middle-Aged/Older Adults: A Randomized, Open-Label, Controlled Crossover Trial. Nutrients 2025; 17:467. [PMID: 39940327 PMCID: PMC11820112 DOI: 10.3390/nu17030467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Microgreens are rich in nutrients and phytochemicals that can support healthy aging, including attenuation of cardiovascular disease risk. The nutrient and phytochemical contents of red beet (i.e., bull's blood' beet, Beta vulgaris) and red cabbage (Brassica oleracea var capitate) microgreens, as well as existing preclinical evidence suggest their cardioprotective effects, but the feasibility, gastrointestinal tolerability, and human health effects of daily microgreen consumption are unknown. This study aimed to evaluate the feasibility and gastrointestinal tolerability of 2 weeks of daily microgreen consumption in healthy middle-aged/older (MA/O) adults. A secondary aim was to characterize potential health effects. Methods: Healthy MA/O adults (initial n = 26) were randomized to consume either 2 cups of 'bull's blood' beet or red cabbage microgreens daily for 2 weeks in a crossover design, with each treatment period separated by 2 weeks. Feasibility was determined through participant retention and intervention compliance (i.e., total doses consumed divided by 14 days), while gastrointestinal tolerability was determined by a gastrointestinal health questionnaire and bowel movement log. Impacts of microgreen consumption on brachial and aortic hemodynamic parameters, and gut microbiota composition were evaluated. Results: Daily consumption for 2 weeks of 'bull's blood' beet and red cabbage microgreens was found to be feasible as indicated by high participant retention (final n = 24) and overall treatment compliance of 95.6%. Gastrointestinal symptom severity was not impacted overall, though an improvement in gastrointestinal inflammation-associated symptom severity scores following the red cabbage microgreen intervention (p = 0.047) was observed. There were no changes in bowel movement quality, hemodynamic parameters, or on alpha or beta diversity of the gut microbiota. Conclusions: Daily consumption of 'bull's blood' beet and red cabbage microgreens is feasible and tolerable in healthy MA/O adults. Future studies designed to evaluate their health impacts are needed.
Collapse
Affiliation(s)
- Sylvia Y. Lee
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Kiri A. Michell
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Michelle M. Butler
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Brayden T. Smith
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Emily K. Woolf
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Sydney C. Holmes
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Lauren E. Grabos
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Allegra R. Vazquez
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Hanan Isweiri
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, CO 80523, USA (M.E.U.)
- Department of Biology, University of Benghazi, Benghazi 16063, Libya
| | - Marisa Bunning
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Mark E. Uchanski
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, CO 80523, USA (M.E.U.)
| | - Sangeeta Rao
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
| | - Steven E. Newman
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, CO 80523, USA (M.E.U.)
| | - Tiffany L. Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| | - Sarah A. Johnson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA; (S.Y.L.); (K.A.M.); (E.K.W.); (T.L.W.)
| |
Collapse
|
4
|
Palma M, Qi B. Advancing Phage Therapy: A Comprehensive Review of the Safety, Efficacy, and Future Prospects for the Targeted Treatment of Bacterial Infections. Infect Dis Rep 2024; 16:1127-1181. [PMID: 39728014 DOI: 10.3390/idr16060092] [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: 09/04/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Phage therapy, a treatment utilizing bacteriophages to combat bacterial infections, is gaining attention as a promising alternative to antibiotics, particularly for managing antibiotic-resistant bacteria. This study aims to provide a comprehensive review of phage therapy by examining its safety, efficacy, influencing factors, future prospects, and regulatory considerations. The study also seeks to identify strategies for optimizing its application and to propose a systematic framework for its clinical implementation. METHODS A comprehensive analysis of preclinical studies, clinical trials, and regulatory frameworks was undertaken to evaluate the therapeutic potential of phage therapy. This included an in-depth assessment of key factors influencing clinical outcomes, such as infection site, phage-host specificity, bacterial burden, and immune response. Additionally, innovative strategies-such as combination therapies, bioengineered phages, and phage cocktails-were explored to enhance efficacy. Critical considerations related to dosing, including inoculum size, multiplicity of infection, therapeutic windows, and personalized medicine approaches, were also examined to optimize treatment outcomes. RESULTS Phage therapy has demonstrated a favorable safety profile in both preclinical and clinical settings, with minimal adverse effects. Its ability to specifically target harmful bacteria while preserving beneficial microbiota underpins its efficacy in treating a range of infections. However, variable outcomes in some studies highlight the importance of addressing critical factors that influence therapeutic success. Innovative approaches, including combination therapies, bioengineered phages, expanded access to diverse phage banks, phage cocktails, and personalized medicine, hold significant promise for improving efficacy. Optimizing dosing strategies remains a key area for enhancement, with critical considerations including inoculum size, multiplicity of infection, phage kinetics, resistance potential, therapeutic windows, dosing frequency, and patient-specific factors. To support the clinical application of phage therapy, a streamlined four-step guideline has been developed, providing a systematic framework for effective treatment planning and implementation. CONCLUSION Phage therapy offers a highly adaptable, targeted, and cost-effective approach to addressing antibiotic-resistant infections. While several critical factors must be thoroughly evaluated to optimize treatment efficacy, there remains significant potential for improvement through innovative strategies and refined methodologies. Although phage therapy has yet to achieve widespread approval in the U.S. and Europe, its accessibility through Expanded Access programs and FDA authorizations for food pathogen control underscores its promise. Established practices in countries such as Poland and Georgia further demonstrate its clinical feasibility. To enable broader adoption, regulatory harmonization and advancements in production, delivery, and quality control will be essential. Notably, the affordability and scalability of phage therapy position it as an especially valuable solution for developing regions grappling with escalating rates of antibiotic resistance.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
- R&D Drug Discovery, Protheragen Inc., Holbrook, NY 11741, USA
| | - Bowen Qi
- Drug Discovery and Development, Creative Biolabs Inc., Shirley, NY 11967, USA
| |
Collapse
|
5
|
Deng Y, Jiang S, Duan H, Shao H, Duan Y. Bacteriophages and their potential for treatment of metabolic diseases. J Diabetes 2024; 16:e70024. [PMID: 39582431 PMCID: PMC11586638 DOI: 10.1111/1753-0407.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Recent advances highlight the role of gut virome, particularly phageome, in metabolic disorders such as obesity, type 2 diabetes mellitus, metabolic dysfunction-associated fatty liver disease, and cardiovascular diseases, including hypertension, stroke, coronary heart disease, and hyperlipidemia. While alterations in gut bacteria are well-documented, emerging evidence suggests that changes in gut viruses also contribute to these disorders. Bacteriophages, the most abundant gut viruses, influence bacterial populations through their lytic and lysogenic cycles, potentially modulating the gut ecosystem and metabolic pathways. Phage therapy, previously overshadowed by antibiotics, is experiencing renewed interest due to rising antibiotic resistance. It offers a novel approach to precisely edit the gut microbiota, with promising applications in metabolic diseases. In this review, we summarize recent discoveries about gut virome in metabolic disease patients, review preclinical and clinical studies of phage therapy on metabolic diseases as well as the breakthroughs and currently faced problems and concerns.
Collapse
Affiliation(s)
- Youpeng Deng
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Shouwei Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hanyu Duan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Haonan Shao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yi Duan
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
6
|
Amarillas L, Padilla-Lafarga F, León Chan RG, Padilla J, Lugo-Melchor Y, López Avendaño JE, Lightbourn-Rojas L, Estrada-Acosta M. Isolation and Characterization of a Bacteriophage with Potential for the Control of Multidrug-Resistant Salmonella Strains Encoding Virulence Factors Associated with the Promotion of Precancerous Lesions. Viruses 2024; 16:1711. [PMID: 39599826 PMCID: PMC11598880 DOI: 10.3390/v16111711] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Antimicrobial-resistant bacteria represent a serious threat to public health. Among these bacteria, Salmonella is of high priority because of its morbidity levels and its ability to induce different types of cancer. AIM This study aimed to identify Salmonella strains encoding genes linked to the promotion of precancerous lesions and to isolate a bacteriophage to evaluate its preclinical potential against these bacteria. METHODOLOGY An epidemiological approach based on wastewater analysis was employed to isolate Salmonella strains and detect genes associated with the induction of precancerous lesions. Antimicrobial susceptibility was assessed by the disk diffusion method. A bacteriophage was isolated via the double agar technique, and its morphological characteristics, stability, host range, replication dynamics, and ability to control Salmonella under different conditions were evaluated. The bacteriophage genome was sequenced and analyzed using bioinformatics tools. RESULTS Thirty-seven Salmonella strains were isolated, seventeen of which contained the five genes associated with precancerous lesions' induction. These strains exhibited resistance to multiple antimicrobials, including fluoroquinolones. A bacteriophage from the Autographiviridae family with lytic activity against 21 bacterial strains was isolated. This phage exhibited a 20 min replication cycle, releasing 52 ± 3 virions per infected cell. It demonstrated stability and efficacy in reducing the Salmonella concentration in simulated gastrointestinal conditions, and its genome lacked genes that represent a biosafety risk. CONCLUSION This bacteriophage shows promising preclinical potential as a biotherapeutic agent against Salmonella.
Collapse
Affiliation(s)
- Luis Amarillas
- Instituto de Investigación Lightbourn, Jimenez 33981, Mexico; (L.A.); (R.G.L.C.)
- Facultad de Agronomía de la Universidad Autónoma de Sinaloa, Culiacán 80000, Mexico
| | | | | | - Jorge Padilla
- Facultad de Agronomía de la Universidad Autónoma de Sinaloa, Culiacán 80000, Mexico
| | - Yadira Lugo-Melchor
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara 44270, Mexico
| | | | | | - Mitzi Estrada-Acosta
- Facultad de Agronomía de la Universidad Autónoma de Sinaloa, Culiacán 80000, Mexico
| |
Collapse
|
7
|
Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, Akanyibah FA, Mao F. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother 2024; 179:117302. [PMID: 39163678 DOI: 10.1016/j.biopha.2024.117302] [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: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex disorder with an unknown cause. However, the dysbiosis of the gut microbiome has been found to play a role in IBD etiology, including exacerbated immune responses and defective intestinal barrier integrity. The gut microbiome can also be a potential biomarker for several diseases, including IBD. Currently, conventional treatments targeting pro-inflammatory cytokines and pathways in IBD-associated dysbiosis do not yield effective results. Other therapies that directly target the dysbiotic microbiome for effective outcomes are emerging. We review the role of the gut microbiome in health and IBD and its potential as a diagnostic, prognostic, and therapeutic target for IBD. This review also explores emerging therapeutic advancements that target gut microbiome-associated alterations in IBD, such as nanoparticle or encapsulation delivery, fecal microbiota transplantation, nutritional therapies, microbiome/probiotic engineering, phage therapy, mesenchymal stem cells (MSCs), gut proteins, and herbal formulas.
Collapse
Affiliation(s)
- Xiu Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu 212300, China
| | - Peipei Cai
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang 212028, China
| | - Anquan Shang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China.
| |
Collapse
|
8
|
Bernardi F, Ungaro F, D’Amico F, Zilli A, Parigi TL, Massimino L, Allocca M, Danese S, Furfaro F. The Role of Viruses in the Pathogenesis of Immune-Mediated Gastro-Intestinal Diseases. Int J Mol Sci 2024; 25:8301. [PMID: 39125870 PMCID: PMC11313478 DOI: 10.3390/ijms25158301] [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: 06/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Immune-mediated gastrointestinal (GI) diseases, including achalasia, celiac disease, and inflammatory bowel diseases, pose significant challenges in diagnosis and management due to their complex etiology and diverse clinical manifestations. While genetic predispositions and environmental factors have been extensively studied in the context of these conditions, the role of viral infections and virome dysbiosis remains a subject of growing interest. This review aims to elucidate the involvement of viral infections in the pathogenesis of immune-mediated GI diseases, focusing on achalasia and celiac disease, as well as the virome dysbiosis in IBD. Recent evidence suggests that viral pathogens, ranging from common respiratory viruses to enteroviruses and herpesviruses, may trigger or exacerbate achalasia and celiac disease by disrupting immune homeostasis in the GI tract. Furthermore, alterations in the microbiota and, specifically, in the virome composition and viral-host interactions have been implicated in perpetuating chronic intestinal inflammation in IBD. By synthesizing current knowledge on viral contributions to immune-mediated GI diseases, this review aims to provide insights into the complex interplay between viral infections, host genetics, and virome dysbiosis, shedding light on novel therapeutic strategies aimed at mitigating the burden of these debilitating conditions on patients' health and quality of life.
Collapse
Affiliation(s)
- Francesca Bernardi
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy
| | - Federica Ungaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Ferdinando D’Amico
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Alessandra Zilli
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Tommaso Lorenzo Parigi
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Luca Massimino
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Mariangela Allocca
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy
| | - Federica Furfaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| |
Collapse
|
9
|
Guo Z, Yuan M, Chai J. Mini review advantages and limitations of lytic phages compared with chemical antibiotics to combat bacterial infections. Heliyon 2024; 10:e34849. [PMID: 39148970 PMCID: PMC11324966 DOI: 10.1016/j.heliyon.2024.e34849] [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: 05/22/2023] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
The overuse of antibiotics has caused the emergence of antibiotic-resistant strains, such as multidrug-resistant, extensively drug-resistant, and pandrug-resistant bacteria. The treatment of infections caused by such strains has become a formidable challenge. In the post-antibiotic era, phage therapy is an attractive solution for this problem and some successful phase 1 and 2 studies have demonstrated the efficacy and safety of phage therapy over the last decade. It is a form of evolutionary medicine, phages exhibit immunomodulatory and anti-inflammatory properties. However, phage therapy is limited by factors, such as the narrow spectrum of host strains, the special pharmacokinetics and pharmacodynamics in vivo, immune responses, and the development of phage resistance. The aim of this minireview was to compare the potencies of lytic phages and chemical antibiotics to treat bacterial infections. The advantages of phage therapy has fewer side effects, self-replication, evolution, bacterial biofilms eradication, immunomodulatory and anti-inflammatory properties compared with chemical antibiotics. Meanwhile, the disadvantages of phage therapy include the narrow spectrum of available host strains, the special pharmacokinetics and pharmacodynamics in vivo, immune responses, and phage resistance hurdles. Recently, some researchers continue to make efforts to overcome these limitations of phage therapy. Phage therapy will be a welcome addition to the gamut of options available for treating antibiotic-resistant bacterial infections. We focus on the advantages and limitations of phage therapy with the intention of exploiting the advantages and overcoming the limitations.
Collapse
Affiliation(s)
- Zhimin Guo
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mengyao Yuan
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiannan Chai
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
10
|
Liu K, Wang C, Zhou X, Guo X, Yang Y, Liu W, Zhao R, Song H. Bacteriophage therapy for drug-resistant Staphylococcus aureus infections. Front Cell Infect Microbiol 2024; 14:1336821. [PMID: 38357445 PMCID: PMC10864608 DOI: 10.3389/fcimb.2024.1336821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Drug-resistant Staphylococcus aureus stands as a prominent pathogen in nosocomial and community-acquired infections, capable of inciting various infections at different sites in patients. This includes Staphylococcus aureus bacteremia (SaB), which exhibits a severe infection frequently associated with significant mortality rate of approximately 25%. In the absence of better alternative therapies, antibiotics is still the main approach for treating infections. However, excessive use of antibiotics has, in turn, led to an increase in antimicrobial resistance. Hence, it is imperative that new strategies are developed to control drug-resistant S. aureus infections. Bacteriophages are viruses with the ability to infect bacteria. Bacteriophages, were used to treat bacterial infections before the advent of antibiotics, but were subsequently replaced by antibiotics due to limited theoretical understanding and inefficient preparation processes at the time. Recently, phages have attracted the attention of many researchers again because of the serious problem of antibiotic resistance. This article provides a comprehensive overview of phage biology, animal models, diverse clinical case treatments, and clinical trials in the context of drug-resistant S. aureus phage therapy. It also assesses the strengths and limitations of phage therapy and outlines the future prospects and research directions. This review is expected to offer valuable insights for researchers engaged in phage-based treatments for drug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Kaixin Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Xudong Zhou
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
- College of Public Health, China Medical University, Shenyang, China
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Hongbin Song
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
- College of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
11
|
Tun HM, Peng Y, Massimino L, Sin ZY, Parigi TL, Facoetti A, Rahman S, Danese S, Ungaro F. Gut virome in inflammatory bowel disease and beyond. Gut 2024; 73:350-360. [PMID: 37949638 PMCID: PMC10850733 DOI: 10.1136/gutjnl-2023-330001] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The gut virome is a dense community of viruses inhabiting the gastrointestinal tract and an integral part of the microbiota. The virome coexists with the other components of the microbiota and with the host in a dynamic equilibrium, serving as a key contributor to the maintenance of intestinal homeostasis and functions. However, this equilibrium can be interrupted in certain pathological states, including inflammatory bowel disease, causing dysbiosis that may participate in disease pathogenesis. Nevertheless, whether virome dysbiosis is a causal or bystander event requires further clarification. DESIGN This review seeks to summarise the latest advancements in the study of the gut virome, highlighting its cross-talk with the mucosal microenvironment. It explores how cutting-edge technologies may build upon current knowledge to advance research in this field. An overview of virome transplantation in diseased gastrointestinal tracts is provided along with insights into the development of innovative virome-based therapeutics to improve clinical management. RESULTS Gut virome dysbiosis, primarily driven by the expansion of Caudovirales, has been shown to impact intestinal immunity and barrier functions, influencing overall intestinal homeostasis. Although emerging innovative technologies still need further implementation, they display the unprecedented potential to better characterise virome composition and delineate its role in intestinal diseases. CONCLUSIONS The field of gut virome is progressively expanding, thanks to the advancements of sequencing technologies and bioinformatic pipelines. These have contributed to a better understanding of how virome dysbiosis is linked to intestinal disease pathogenesis and how the modulation of virome composition may help the clinical intervention to ameliorate gut disease management.
Collapse
Affiliation(s)
- Hein Min Tun
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- JC School of Public Health and Primary Care, Faculty of medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Peng
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- JC School of Public Health and Primary Care, Faculty of medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Luca Massimino
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Zhen Ye Sin
- JC School of Public Health and Primary Care, Faculty of medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tommaso Lorenzo Parigi
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele Facoltà di Medicina e Chirurgia, Milano, Italy
| | - Amanda Facoetti
- Università Vita-Salute San Raffaele Facoltà di Medicina e Chirurgia, Milano, Italy
| | | | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele Facoltà di Medicina e Chirurgia, Milano, Italy
| | - Federica Ungaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele Facoltà di Medicina e Chirurgia, Milano, Italy
| |
Collapse
|
12
|
Mahmud MR, Tamanna SK, Akter S, Mazumder L, Akter S, Hasan MR, Acharjee M, Esti IZ, Islam MS, Shihab MMR, Nahian M, Gulshan R, Naser S, Pirttilä AM. Role of bacteriophages in shaping gut microbial community. Gut Microbes 2024; 16:2390720. [PMID: 39167701 PMCID: PMC11340752 DOI: 10.1080/19490976.2024.2390720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Phages are the most diversified and dominant members of the gut virobiota. They play a crucial role in shaping the structure and function of the gut microbial community and consequently the health of humans and animals. Phages are found mainly in the mucus, from where they can translocate to the intestinal organs and act as a modulator of gut microbiota. Understanding the vital role of phages in regulating the composition of intestinal microbiota and influencing human and animal health is an emerging area of research. The relevance of phages in the gut ecosystem is supported by substantial evidence, but the importance of phages in shaping the gut microbiota remains unclear. Although information regarding general phage ecology and development has accumulated, detailed knowledge on phage-gut microbe and phage-human interactions is lacking, and the information on the effects of phage therapy in humans remains ambiguous. In this review, we systematically assess the existing data on the structure and ecology of phages in the human and animal gut environments, their development, possible interaction, and subsequent impact on the gut ecosystem dynamics. We discuss the potential mechanisms of prophage activation and the subsequent modulation of gut bacteria. We also review the link between phages and the immune system to collect evidence on the effect of phages on shaping the gut microbial composition. Our review will improve understanding on the influence of phages in regulating the gut microbiota and the immune system and facilitate the development of phage-based therapies for maintaining a healthy and balanced gut microbiota.
Collapse
Affiliation(s)
- Md. Rayhan Mahmud
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | | | - Sharmin Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Lincon Mazumder
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Sumona Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Mrityunjoy Acharjee
- Department of Microbiology, Stamford University Bangladesh, Dhaka, Bangladesh
| | - Israt Zahan Esti
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
- Department of Molecular Systems Biology, Faculty of Technology, University of Turku, Turku, Finland
| | - Md. Saidul Islam
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Md. Nahian
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Rubaiya Gulshan
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Sadia Naser
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | |
Collapse
|
13
|
Fujiki J, Schnabl B. Phage therapy: Targeting intestinal bacterial microbiota for the treatment of liver diseases. JHEP Rep 2023; 5:100909. [PMID: 37965159 PMCID: PMC10641246 DOI: 10.1016/j.jhepr.2023.100909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 11/16/2023] Open
Abstract
Phage therapy has been overshadowed by antibiotics for decades. However, it is being revisited as a powerful approach against antimicrobial-resistant bacteria. As bacterial microbiota have been mechanistically linked to gastrointestinal and liver diseases, precise editing of the gut microbiota via the selective bactericidal action of phages has prompted renewed interest in phage therapy. In this review, we summarise the basic virological properties of phages and the latest findings on the composition of the intestinal phageome and the changes associated with liver diseases. We also review preclinical and clinical studies assessing phage therapy for the treatment of gastrointestinal and liver diseases, as well as future prospects and challenges.
Collapse
Affiliation(s)
- Jumpei Fujiki
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
14
|
Simpson EA, Stacey HJ, Langley RJ, Jones JD. Phage therapy: Awareness and demand among clinicians in the United Kingdom. PLoS One 2023; 18:e0294190. [PMID: 37956142 PMCID: PMC10642789 DOI: 10.1371/journal.pone.0294190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Bacterial resistance or tolerance to antibiotics is costly to patients and healthcare providers. With the impact of antibiotic resistance forecast to grow, alternative antimicrobial approaches are needed to help treat patients with antibiotic refractory infections and reduce reliance upon existing antibiotics. There is renewed interest in bacteriophage (phage) therapy as a promising antimicrobial strategy. We therefore performed the first multi-specialty survey about phage therapy and the first such survey among clinicians in the United Kingdom. An anonymous 10-question survey of clinicians from medical and surgical specialties in two Scottish Health Boards was performed. The 90 respondents spanned 26 specialties and were predominantly consultants (73.3%). The respondents were concerned about antibiotic resistance in their clinical practice; 83 respondents estimated having seen 711 patients in the last 12 months whose infections were refractory to antibiotics (delaying or preventing resolution). Over half (58.8%) of the respondents had previously heard of phage therapy. Staphylococci, Pseudomonas and E. coli were identified as the highest cross-specialty priorities for the development of phage therapy. Together, 77 respondents estimated seeing 300 patients in the last 12 months for whom phage therapy may have been appropriate (an average of 3.9 patients per clinician). Most respondents (71.1%, n = 90) were already willing to consider using phage therapy in appropriate cases. Additional comments from the respondents affirmed the potential utility of phage therapy and highlighted a need for more information. The results of this survey demonstrate substantial demand for and willingness to use phage therapy in appropriate cases, both from individual clinicians and across specialties. Demand from a wide range of specialties illustrates the broad clinical utility of phage therapy and potential scope of impact. Widening access to phage therapy could deliver substantial clinical and financial benefits for patients and health authorities alike.
Collapse
Affiliation(s)
- Emily A Simpson
- Medical Microbiology, Ninewells Hospital, NHS Tayside, Dundee, United Kingdom
| | - Helen J Stacey
- Public Health, Kings Cross Hospital, NHS Tayside, Dundee, United Kingdom
| | - Ross J Langley
- Department of Paediatric Respiratory and Sleep Medicine, Royal Hospital for Children, Glasgow, United Kingdom
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Joshua D Jones
- Medical Microbiology, Ninewells Hospital, NHS Tayside, Dundee, United Kingdom
- Infection Medicine, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Lorenzo-Rebenaque L, Casto-Rebollo C, Diretto G, Frusciante S, Rodríguez JC, Ventero MP, Molina-Pardines C, Vega S, Marin C, Marco-Jiménez F. Modulation of Caecal Microbiota and Metabolome Profile in Salmonella-Infected Broilers by Phage Therapy. Int J Mol Sci 2023; 24:15201. [PMID: 37894882 PMCID: PMC10607084 DOI: 10.3390/ijms242015201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteriophage therapy is considered one of the most promising tools to control zoonotic bacteria, such as Salmonella, in broiler production. Phages exhibit high specificity for their targeted bacterial hosts, causing minimal disruption to the niche microbiota. However, data on the gut environment's response to phage therapy in poultry are limited. This study investigated the influence of Salmonella phage on host physiology through caecal microbiota and metabolome modulation using high-throughput 16S rRNA gene sequencing and an untargeted metabolomics approach. We employed 24 caecum content samples and 24 blood serum samples from 4-, 5- and 6-week-old broilers from a previous study where Salmonella phages were administered via feed in Salmonella-infected broilers, which were individually weighed weekly. Phage therapy did not affect the alpha or beta diversity of the microbiota. Specifically, we observed changes in the relative abundance of 14 out of the 110 genera using the PLS-DA and Bayes approaches. On the other hand, we noted changes in the caecal metabolites (63 up-accumulated and 37 down-accumulated out of the 1113 caecal metabolites). Nevertheless, the minimal changes in blood serum suggest a non-significant physiological response. The application of Salmonella phages under production conditions modulates the caecal microbiome and metabolome profiles in broilers without impacting the host physiology in terms of growth performance.
Collapse
Affiliation(s)
- Laura Lorenzo-Rebenaque
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Cristina Casto-Rebollo
- Institute for Animal Science and Technology, Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Gianfranco Diretto
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Via Anguillarese, 301, Santa Maria di Galeria, 00123 Rome, Italy; (G.D.); (S.F.)
| | - Sarah Frusciante
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Via Anguillarese, 301, Santa Maria di Galeria, 00123 Rome, Italy; (G.D.); (S.F.)
| | - Juan Carlos Rodríguez
- Microbiology Department, Dr. Balmis University General Hospital, Microbiology Division, Miguel Hernández University, ISABIAL, 03010 Alicante, Spain;
| | - María-Paz Ventero
- Microbiology Department, Dr. Balmis University General Hospital, ISABIAL, 03010 Alicante, Spain; (M.-P.V.); (C.M.-P.)
| | - Carmen Molina-Pardines
- Microbiology Department, Dr. Balmis University General Hospital, ISABIAL, 03010 Alicante, Spain; (M.-P.V.); (C.M.-P.)
| | - Santiago Vega
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Clara Marin
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Francisco Marco-Jiménez
- Institute for Animal Science and Technology, Universitat Politècnica de València, 46022 Valencia, Spain;
| |
Collapse
|
16
|
Yang Q, Le S, Zhu T, Wu N. Regulations of phage therapy across the world. Front Microbiol 2023; 14:1250848. [PMID: 37869667 PMCID: PMC10588630 DOI: 10.3389/fmicb.2023.1250848] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/06/2023] [Indexed: 10/24/2023] Open
Abstract
Phage therapy, a century-long treatment targeting bacterial infection, was widely abandoned after the clinical availability of antibiotics in the mid-20th century. However, the crisis of antimicrobial resistance today led to its revival in many countries. While many articles dive into its clinical application now, little research is presenting phage therapy from a regulatory perspective. Here, we focus on the regulations of phage therapy by dividing sections into Eastern Europe where it was never abandoned and Western Europe, Australia, the United States, India, and China where it only re-attracted researchers' attention in recent decades. New insights about its regulations in China are provided as little English literature has specifically discussed this previously. Ultimately, by introducing the regulations in phage therapy for human health across representative countries, we hope to provide ideas of how countries may borrow each other's adapting legislation in phage therapy to best overcome the current regulatory hurdles.
Collapse
Affiliation(s)
- Qimao Yang
- CreatiPhage Biotechnology Co., Ltd., Shanghai, China
| | - Shuai Le
- Department of Microbiology, Army Medical University, Chongqing, China
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tongyu Zhu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Nannan Wu
- CreatiPhage Biotechnology Co., Ltd., Shanghai, China
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Emencheta SC, Olovo CV, Eze OC, Kalu CF, Berebon DP, Onuigbo EB, Vila MMDC, Balcão VM, Attama AA. The Role of Bacteriophages in the Gut Microbiota: Implications for Human Health. Pharmaceutics 2023; 15:2416. [PMID: 37896176 PMCID: PMC10609668 DOI: 10.3390/pharmaceutics15102416] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/18/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteriophages (phages) are nano-sized viruses characterized by their inherent ability to live off bacteria. They utilize diverse mechanisms to absorb and gain entry into the bacterial cell wall via the release of viral genetic material, which uses the replication mechanisms of the host bacteria to produce and release daughter progeny virions that attack the surrounding host cells. They possess specific characteristics, including specificity for particular or closely related bacterial species. They have many applications, including as potential alternatives to antibiotics against multi-resistant bacterial pathogens and as control agents in bacteria-contaminated environments. They are ubiquitously abundant in nature and have diverse biota, including in the gut. Gut microbiota describes the community and interactions of microorganisms within the intestine. As with bacteria, parasitic bacteriophages constantly interact with the host bacterial cells within the gut system and have obvious implications for human health. However, it is imperative to understand these interactions as they open up possible applicable techniques to control gut-implicated bacterial diseases. Thus, this review aims to explore the interactions of bacteriophages with bacterial communities in the gut and their current and potential impacts on human health.
Collapse
Affiliation(s)
- Stephen C. Emencheta
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
| | - Chinasa V. Olovo
- Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria;
| | - Osita C. Eze
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Chisom F. Kalu
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Dinebari P. Berebon
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Ebele B. Onuigbo
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Marta M. D. C. Vila
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
| | - Victor M. Balcão
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, P-3810-193 Aveiro, Portugal
| | - Anthony A. Attama
- Department of Pharmaceutics, University of Nigeria, Nsukka 410001, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka 410001, Nigeria
| |
Collapse
|
18
|
Zou G, He L, Rao J, Song Z, Du H, Li R, Wang W, Zhou Y, Liang L, Chen H, Li J. Improving the safety and efficacy of phage therapy from the perspective of phage-mammal interactions. FEMS Microbiol Rev 2023; 47:fuad042. [PMID: 37442611 DOI: 10.1093/femsre/fuad042] [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: 12/14/2022] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/15/2023] Open
Abstract
Phage therapy has re-emerged as a promising solution for combating antimicrobial-resistant bacterial infections. Increasingly, studies have revealed that phages possess therapeutic potential beyond their antimicrobial properties, including regulating the gut microbiome and maintain intestinal homeostasis, as a novel nanocarrier for targeted drug delivery. However, the complexity and unpredictability of phage behavior during treatment pose a significant challenge in clinical practice. The intricate interactions established between phages, humans, and bacteria throughout their long coexistence in the natural ecosystem contribute to the complexity of phage behavior in therapy, raising concerns about their efficacy and safety as therapeutic agents. Revealing the mechanisms by which phages interact with the human body will provide a theoretical basis for increased application of promising phage therapy. In this review, we provide a comprehensive summary of phage-mammal interactions, including signaling pathways, adaptive immunity responses, and phage-mediated anti-inflammatory responses. Then, from the perspective of phage-mammalian immune system interactions, we present the first systematic overview of the factors affecting phage therapy, such as the mode of administration, the physiological status of the patient, and the biological properties of the phage, to offer new insights into phage therapy for various human diseases.
Collapse
Affiliation(s)
- Geng Zou
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Lijun He
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Rao
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhiyong Song
- College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Hu Du
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Runze Li
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenjing Wang
- College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Zhou
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Lu Liang
- School of Bioscience, University of Nottingham, Sutton Bonington LE12 5RD, United Kingdom
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinquan Li
- National Key Laboratory of Agricultural Microbiology, Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, College of Veterinary Medicine, College of Biomedicine and Health, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| |
Collapse
|
19
|
Singh J, Fitzgerald DA, Jaffe A, Hunt S, Barr JJ, Iredell J, Selvadurai H. Single-arm, open-labelled, safety and tolerability of intrabronchial and nebulised bacteriophage treatment in children with cystic fibrosis and Pseudomonas aeruginosa. BMJ Open Respir Res 2023; 10:e001360. [PMID: 37160359 PMCID: PMC10173968 DOI: 10.1136/bmjresp-2022-001360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
INTRODUCTION Cystic fibrosis (CF) is a multisystem condition that is complicated by recurrent pulmonary infections requiring aggressive antibiotic treatment. This predisposes the patient to complications such as sensorineural hearing loss, renal impairment, hypersensitivity and the development of antibiotic resistance. Pseudomonas aeruginosa is one of the more common organisms which cause recurrent infections and result in greater morbidity and mortality in people living with CF. Bacteriophages have been identified as a potential alternative or adjunct to antibiotics. We hypothesise that bacteriophage therapy is a safe and well-tolerated treatment in children with CF infected with P. aeruginosa infection in their airways. METHODS This single-arm, open-labelled, non-randomised trial will run for a maximum period of 36 months with up to 10 participants. Adolescents (≥12 years and <18 years of age) who continue to shed P.aeruginosa (within 3 months of enrolment) despite undergoing eradication therapy previously, will be considered for this trial. Non-genetically modified bacteriophages that have demonstrated obligate lytic activity against each of the study participants' P. aeruginosa strains will be selected and prepared according to a combination of established protocols (isolation, purification, sterility testing and packaging) to achieve close to good manufacturing practice recommendations. The selected bacteriophage will be administered endo-bronchially first under direct vision, followed by two times a day nebulisation for 7 days in addition to standard CF treatment (intravenous antibiotics, physiotherapy to be completed as inpatient for 10-14 days). Safety and tolerability will be defined as the absence of (1) fever above 38.5°C occurring within 1 hour of the administration of the nebulised bacteriophage, (2) a 10% decline in spirometry (forced expiratory volume in 1 s %) measured preadministration and postadministration of the first dose of nebulised bacteriophage. Clinical reviews including repeat sputum cultures and spirometry will be performed at 3, 6, 9 and 12 months following bacteriophage treatment. ETHICS AND DISSEMINATION Our clinical trial is conducted in accordance with (1) good clinical practice, (2) Australian legislation, (3) National Health and Medical Research Council guidelines for the ethical conduct of research. TRIAL REGISTRATION NUMBER Australia and New Zealand Clinical Trial Registry (ACTRN12622000767707).
Collapse
Affiliation(s)
- Jagdev Singh
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Dominic A Fitzgerald
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sharon Hunt
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | | | - Jonathan Iredell
- Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Department of Infectious Diseases, Westmead Hospital, Westmead, New South Wales, Australia
| | - Hiran Selvadurai
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
20
|
Chen L, Hou X, Chu H. The Novel Role of Phage Particles in Chronic Liver Diseases. Microorganisms 2023; 11:1181. [PMID: 37317156 PMCID: PMC10220600 DOI: 10.3390/microorganisms11051181] [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: 03/20/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 06/16/2023] Open
Abstract
The gut microbiome is made up of bacteria, fungi, viruses and archaea, all of which are closely related with human health. As the main component of enterovirus, the role of bacteriophages (phages) in chronic liver disease has been gradually recognized. Chronic liver diseases, including alcohol-related liver disease and nonalcoholic fatty liver disease, exhibit alterations of the enteric phages. Phages shape intestinal bacterial colonization and regulate bacterial metabolism. Phages adjoining to intestinal epithelial cells prevent bacteria from invading the intestinal barrier, and mediate intestinal inflammatory response. Phages are also observed increasing intestinal permeability and migrating to peripheral blood and organs, likely contributing to inflammatory injury in chronic liver diseases. By preying on harmful bacteria, phages can improve the gut microbiome of patients with chronic liver disease and thus act as an effective treatment method.
Collapse
Affiliation(s)
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
21
|
Diallo K, Dublanchet A. A Century of Clinical Use of Phages: A Literature Review. Antibiotics (Basel) 2023; 12:751. [PMID: 37107113 PMCID: PMC10135294 DOI: 10.3390/antibiotics12040751] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Growing antibiotic resistance and the broken antibiotic market have renewed interest in the use of phages, a century-old therapy that fell into oblivion in the West after two decades of promising results. This literature review with a particular focus on French literature aims to complement current scientific databases with medical and non-medical publications on the clinical use of phages. While several cases of successful treatment with phages have been reported, prospective randomized clinical trials are needed to confirm the efficacy of this therapy.
Collapse
Affiliation(s)
- Kevin Diallo
- Department of Infective and Tropical Diseases and Internal Medicine, University Hospital of la Reunion, 97448 Saint-Pierre, France
| | - Alain Dublanchet
- Independent Researcher, 2465 Rue Céline Robert, 94300 Vincennes, France
| |
Collapse
|
22
|
Mehmood Khan F, Manohar P, Singh Gondil V, Mehra N, Kayode Oyejobi G, Odiwuor N, Ahmad T, Huang G. The applications of animal models in phage therapy: An update. Hum Vaccin Immunother 2023; 19:2175519. [PMID: 36935353 PMCID: PMC10072079 DOI: 10.1080/21645515.2023.2175519] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
The rapid increase in antibiotic resistance presents a dire situation necessitating the need for alternative therapeutic agents. Among the current alternative therapies, phage therapy (PT) is promising. This review extensively summarizes preclinical PT approaches in various in-vivo models. PT has been evaluated in several recent clinical trials. However, there are still several unanswered concerns due to a lack of appropriate regulation and pharmacokinetic data regarding the application of phages in human therapeutic procedures. In this review, we also presented the current state of PT and considered how animal models can be used to adapt these therapies for humans. The development of realistic solutions to circumvent these constraints is critical for advancing this technology.
Collapse
Affiliation(s)
- Fazal Mehmood Khan
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.,Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Prasanth Manohar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Vijay Singh Gondil
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Nancy Mehra
- Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Greater Kayode Oyejobi
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,Department of Microbiology, Osun State University, Osogbo, Nigeria.,School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Nelson Odiwuor
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,International College, University of Chinese Academy of Sciences, Beijing, China.,Microbiology, Sino-Africa Joint Research Centre, Nairobi, Kenya
| | - Tauseef Ahmad
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Guangtao Huang
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Jones JD, Trippett C, Suleman M, Clokie MRJ, Clark JR. The Future of Clinical Phage Therapy in the United Kingdom. Viruses 2023; 15:721. [PMID: 36992430 PMCID: PMC10053292 DOI: 10.3390/v15030721] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Bacteriophage (phage) therapy is a promising alternative antimicrobial strategy with the potential to transform the way bacterial infections are treated. In the United Kingdom, phages are classed as a biological medicine. Although no phages are licensed for UK use, they may be used as unlicensed medicinal products where licensed alternatives cannot meet a patient's clinical needs. In the last 2 years, 12 patients in the UK have received phage therapy, and there is burgeoning clinical interest. Currently, clinical phage provision in the UK is ad hoc and relies upon networking with international sources of phages. The provision of phage therapy in the UK will not progress beyond an increasing number of ad hoc cases until an onshore sustainable and scalable source of well-characterised phages manufactured in accordance with Good Manufacturing Practice (GMP) is established. Here, we present an exciting new collaboration between UK Phage Therapy, the Centre for Phage Research at University of Leicester, CPI, and Fixed Phage. These partners, and others as we develop, will establish sustainable, scalable, and equitable phage therapy provision in the UK. We set out a vision for how phage therapy will be integrated into the NHS and healthcare more broadly, including the complementarity between licensed (cocktail) and unlicensed (personalised) phage preparations. Key elements of phage therapy infrastructure in the UK will be GMP phage manufacturing, a national phage library, and a national clinical phage centre. Together, this infrastructure will support NHS microbiology departments to develop and oversee phage therapy provision across the UK. As it will take time to deliver this, we also describe considerations for clinicians seeking to use unlicensed phage therapy in the interim. In summary, this review sets out a roadmap for the delivery of clinical phage therapy to the UK, the benefits of which we hope will reverberate for patients for decades to come.
Collapse
Affiliation(s)
| | - Clare Trippett
- CPI, 1 Union Square, Central Park, Darlington DL1 1GL, UK
| | - Mehrunisha Suleman
- The Ethox Centre, University of Oxford, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, Oxford OX3 7LF, UK
| | - Martha R. J. Clokie
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Jason R. Clark
- Fixed Phage, West of Scotland Science Park, Block 2, Kelvin Campus, 2317 Maryhill Road, Glasgow G20 0SP, UK
| |
Collapse
|
24
|
Maimaiti Z, Li Z, Xu C, Chen J, Chai W. Global trends and hotspots of phage therapy for bacterial infection: A bibliometric visualized analysis from 2001 to 2021. Front Microbiol 2023; 13:1067803. [PMID: 36699585 PMCID: PMC9868171 DOI: 10.3389/fmicb.2022.1067803] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Background Antibiotic resistance is one of the main global threats to human health, and just the development of new antimicrobial medications is not enough to solve the crisis. Phage therapy (PT), a safe and effective treatment method, has reignited the interest of researchers due to its efficacy in the clinical treatment of drug-resistant bacterial infections. There is, however, no bibliometric analysis of the overall trends on this topic. Therefore, this study aims to provide an overview of the current state of development and research in this area. Methods We extracted all relevant publications from the Web of Science Core Collection (WoSCC) database between 2001 and 2021. We performed bibliometric analysis and visualization using CiteSpace, VOS viewer, and R software. Annual trends of publications, countries/regions distributions, institutions, funding agencies, co-cited journals, author contributions, core journals, references, and keywords were analyzed. Results A total of 6,538 papers were enrolled in this study, including 5,364 articles and 1,174 reviews. Publications have increased drastically from 61 in 2001 to 937 in 2021, with 3,659 articles published in the last 5 years. North America, Western Europe, and East Asia were significant contributor regions. The United States, China, and the United Kingdom were the most productive countries. The Polish Academy of Sciences was the most contributive institution. Frontiers in Microbiology and Applied and Environmental Microbiology were the most productive and co-cited journals. A. Gorski and R. Lavigne published most articles in this field, while V. A. Fischetti was the author with the most cited. Regarding keywords, research focuses include phage biology, phage against clinically important pathogens, phage lysis proteins, phage therapy, biofilm-related research, and recent clinical applications. Conclusion Phage therapy is a potential strategy for combating antibiotic resistance, and it will provide us with an alternative therapeutic option for bacterial infection. According to global trends, the scientific output of PT in bacterial infections is increasing, with developed countries such as the United States leading the way in this area. Although the safety and efficacy of PT have been proven, more clinical trials on the phages against infectious diseases caused by various pathogens are still needed.
Collapse
Affiliation(s)
- Zulipikaer Maimaiti
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhuo Li
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China,School of Medicine, Nankai University, Tianjin, China
| | - Chi Xu
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiying Chen
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Jiying Chen, ; Wei Chai,
| | - Wei Chai
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Jiying Chen, ; Wei Chai,
| |
Collapse
|
25
|
Cervantes-Echeverría M, Gallardo-Becerra L, Cornejo-Granados F, Ochoa-Leyva A. The Two-Faced Role of crAssphage Subfamilies in Obesity and Metabolic Syndrome: Between Good and Evil. Genes (Basel) 2023; 14:139. [PMID: 36672880 PMCID: PMC9858991 DOI: 10.3390/genes14010139] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/27/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Viral metagenomic studies of the human gut microbiota have unraveled the differences in phage populations between health and disease, stimulating interest in phages' role on bacterial ecosystem regulation. CrAssphage is a common and abundant family in the gut virome across human populations. Therefore, we explored its role in obesity (O) and obesity with metabolic syndrome (OMS) in a children's cohort. We found a significantly decreased prevalence, diversity, and richness of the crAssphage Alpha subfamily in OMS mainly driven by a decrease in the Alpha_1 and Alpha_4 genera. On the contrary, there was a significant increase in the Beta subfamily in OMS, mainly driven by an increase in Beta_6. Additionally, an overabundance of the Delta_8 genus was observed in OMS. Notably, a decreased abundance of crAssphages was significantly correlated with the overabundance of Bacilli in the same group. The Bacilli class is a robust taxonomical biomarker of O and was also significantly abundant in our OMS cohort. Our results suggest that a loss of stability in the Alpha subfamily of crAssphages is associated with O and OMS. Contrary, an overabundance of the Delta subfamily was found in OMS. Our study advises the importance of considering the dual role (good and evil) of crAssphage subfamilies and their participation in conditions such as O, where we suggest that Alpha loss and Delta gain are associated with obese individuals.
Collapse
Affiliation(s)
| | | | | | - Adrian Ochoa-Leyva
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Avenida Universidad 2001, Cuernavaca 62210, Morelos, Mexico
| |
Collapse
|
26
|
Marongiu L, Burkard M, Lauer UM, Hoelzle LE, Venturelli S. Reassessment of Historical Clinical Trials Supports the Effectiveness of Phage Therapy. Clin Microbiol Rev 2022; 35:e0006222. [PMID: 36069758 PMCID: PMC9769689 DOI: 10.1128/cmr.00062-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Phage therapy has become a hot topic in medical research due to the increasing prevalence of antibiotic-resistant bacteria strains. In the treatment of bacterial infections, bacteriophages have several advantages over antibiotics, including strain specificity, lack of serious side effects, and low development costs. However, scientists dismissed the clinical success of early clinical trials in the 1940s, slowing the adoption of this promising antibacterial application in Western countries. The current study used statistical methods commonly used in modern meta-analysis to reevaluate early 20th-century studies and compare them with clinical trials conducted in the last 20 years. Using a random effect model, the development of disease after treatment with or without phages was measured in odds ratios (OR) with 95% confidence intervals (CI). Based on the findings of 17 clinical trials conducted between 1921 and 1940, phage therapy was effective (OR = 0.21, 95% CI = 0.10 to 0.44, P value < 0.0001). The current study includes a topic review on modern clinical trials; four could be analyzed, indicating a noneffective therapy (OR = 2.84, 95% CI = 1.53 to 5.27, P value = 0.0009). The results suggest phage therapy was surprisingly less effective than standard treatments in resolving bacterial infections. However, the results were affected by the small sample set size. This work also contextualizes the development of phage therapy in the early 20th century and highlights the expansion of phage applications in the last few years. In conclusion, the current review shows phage therapy is no longer an underestimated tool in the treatment of bacterial infections.
Collapse
Affiliation(s)
- Luigi Marongiu
- Department of Biochemistry of Nutrition, University of Hohenheim, Stuttgart, Germany
- Department of Internal Medicine VIII, University Hospital Tuebingen, Tuebingen, Germany
| | - Markus Burkard
- Department of Biochemistry of Nutrition, University of Hohenheim, Stuttgart, Germany
| | - Ulrich M. Lauer
- Department of Internal Medicine VIII, University Hospital Tuebingen, Tuebingen, Germany
| | - Ludwig E. Hoelzle
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
- HoLMiR-Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany
| | - Sascha Venturelli
- Department of Biochemistry of Nutrition, University of Hohenheim, Stuttgart, Germany
- Institute of Physiology, Department of Vegetative and Clinical Physiology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
27
|
Jaglan AB, Anand T, Verma R, Vashisth M, Virmani N, Bera BC, Vaid RK, Tripathi BN. Tracking the phage trends: A comprehensive review of applications in therapy and food production. Front Microbiol 2022; 13:993990. [PMID: 36504807 PMCID: PMC9730251 DOI: 10.3389/fmicb.2022.993990] [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: 07/14/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
In the present scenario, the challenge of emerging antimicrobial resistance is affecting human health globally. The increasing incidences of multidrug-resistant infections have become harder to treat, causing high morbidity, and mortality, and are posing extensive financial loss. Limited discovery of new antibiotic molecules has further complicated the situation and has forced researchers to think and explore alternatives to antibiotics. This has led to the resurgence of the bacteriophages as an effective alternative as they have a proven history in the Eastern world where lytic bacteriophages have been used since their first implementation over a century ago. To help researchers and clinicians towards strengthening bacteriophages as a more effective, safe, and economical therapeutic alternative, the present review provides an elaborate narrative about the important aspects of bacteriophages. It abridges the prerequisite essential requirements of phage therapy, the role of phage biobank, and the details of immune responses reported while using bacteriophages in the clinical trials/compassionate grounds by examining the up-to-date case reports and their effects on the human gut microbiome. This review also discusses the potential of bacteriophages as a biocontrol agent against food-borne diseases in the food industry and aquaculture, in addition to clinical therapy. It finishes with a discussion of the major challenges, as well as phage therapy and phage-mediated biocontrols future prospects.
Collapse
Affiliation(s)
- Anu Bala Jaglan
- Department of Zoology and Aquaculture, Chaudhary Charan Singh Haryana Agricultural University, Hisar, India
| | - Taruna Anand
- ICAR – National Research Centre on Equines, Hisar, India,*Correspondence: Taruna Anand,
| | - Ravikant Verma
- Department of Zoology and Aquaculture, Chaudhary Charan Singh Haryana Agricultural University, Hisar, India
| | - Medhavi Vashisth
- Department of Molecular Biology, Biotechnology, and Bioinformatics, Chaudhary Charan Singh Haryana Agricultural University, Hisar, India
| | - Nitin Virmani
- ICAR – National Research Centre on Equines, Hisar, India
| | - B. C. Bera
- ICAR – National Research Centre on Equines, Hisar, India
| | - R. K. Vaid
- ICAR – National Research Centre on Equines, Hisar, India
| | - B. N. Tripathi
- Animal Science Division, Indian Council of Agricultural Research, Krishi Bhawan, New Delhi, India
| |
Collapse
|
28
|
Lorenzo-Rebenaque L, Casto-Rebollo C, Diretto G, Frusciante S, Rodríguez JC, Ventero MP, Molina-Pardines C, Vega S, Marin C, Marco-Jiménez F. Examining the effects of Salmonella phage on the caecal microbiota and metabolome features in Salmonella-free broilers. Front Genet 2022; 13:1060713. [PMID: 36437955 PMCID: PMC9691336 DOI: 10.3389/fgene.2022.1060713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/26/2022] [Indexed: 10/29/2023] Open
Abstract
Bacteriophages selectively infect and kill their target bacterial host, being a promising approach to controlling zoonotic bacteria in poultry production. To ensure confidence in its use, fundamental questions of safety and toxicity monitoring of phage therapy should be raised. Due to its high specificity, a minimal impact on the gut ecology is expected; however, more in-depth research into key parameters that influence the success of phage interventions has been needed to reach a consensus on the impact of bacteriophage therapy in the gut. In this context, this study aimed to investigate the interaction of phages with animals; more specifically, we compared the caecum microbiome and metabolome after a Salmonella phage challenge in Salmonella-free broilers, evaluating the role of the phage administration route. To this end, we employed 45 caecum content samples from a previous study where Salmonella phages were administered via drinking water or feed for 24 h from 4, 5 to 6-weeks-old broilers. High-throughput 16S rRNA gene sequencing showed a high level of similarity (beta diversity) but revealed a significant change in alpha diversity between broilers with Salmonella-phage administered in the drinking water and control. Our results showed that the phages affected only a few genera of the microbiota's structure, regardless of the administration route. Among these, we found a significant increase in Streptococcus and Sellimonas in the drinking water and Lactobacillus, Anaeroplasma and Clostridia_vadinBB60_group in the feed. Nevertheless, the LC-HRMS-based metabolomics analyses revealed that despite few genera were significantly affected, a substantial number of metabolites, especially in the phage administered in the drinking water were significantly altered (64 and 14 in the drinking water and feed groups, respectively). Overall, our study shows that preventive therapy with bacteriophages minimally alters the caecal microbiota but significantly impacts their metabolites, regardless of the route of administration.
Collapse
Affiliation(s)
- Laura Lorenzo-Rebenaque
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain
| | - Cristina Casto-Rebollo
- Institute of Science and Animal Technology, Universitat Politècnica de València, Valencia, Spain
| | - Gianfranco Diretto
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Santa Maria di Galeria, Rome, Italy
| | - Sarah Frusciante
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Santa Maria di Galeria, Rome, Italy
| | - Juan Carlos Rodríguez
- Microbiology Department, Balmis General University Hospital, Microbiology Division, Miguel Hernández University, ISABIAL, Alicante, Spain
| | - María-Paz Ventero
- Microbiology Department, Balmis General University Hospital, ISABIAL, Alicante, Spain
| | | | - Santiago Vega
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain
| | - Clara Marin
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain
| | - Francisco Marco-Jiménez
- Institute of Science and Animal Technology, Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
29
|
Stacey HJ, De Soir S, Jones JD. The Safety and Efficacy of Phage Therapy: A Systematic Review of Clinical and Safety Trials. Antibiotics (Basel) 2022; 11:1340. [PMID: 36289998 PMCID: PMC9598614 DOI: 10.3390/antibiotics11101340] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
Trials of phage therapy have not consistently reported efficacy. This contrasts with promising efficacy rates from a sizeable and compelling body of observational literature. This systematic review explores the reasons why many phage trials have not demonstrated efficacy. Four electronic databases were systematically searched for safety and/or efficacy trials of phage therapy. Sixteen trials of phage therapy were included, in which 378 patients received phage. These were divided into historical (pre-2000; N = 3; n = 76) and modern (post-2000; N = 13; n = 302) trials. All 13 modern trials concluded that phage therapy was safe. Six of the 13 modern trials were exclusively safety trials. Seven modern trials investigated both safety and efficacy; efficacy was observed in two. Two of three historical trials did not comment on safety, while adverse effects in the third likely reflected the use of phage preparations contaminated with bacterial debris. None of the historical trials contained evidence of efficacy. The evidence from trials is that phage therapy is safe. For efficacy to be observed a therapeutic amount of the right phage(s) must be delivered to the right place to treat infections containing enough susceptible bacterial cells. Trials that have not demonstrated efficacy have not fulfilled one or more elements of this principle.
Collapse
Affiliation(s)
- Helen J. Stacey
- Public Health, Kings Cross Hospital, Clepington Road, Dundee DD3 8EA, UK
| | - Steven De Soir
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Rue Bruyn, 1120 Brussels, Belgium
- Cellular & Molecular Pharmacology, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier 73, 1200 Brussels, Belgium
| | - Joshua D. Jones
- Infection Medicine, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Clinical Microbiology, NHS Tayside, Dundee DD2 1SG, UK
| |
Collapse
|
30
|
Boix-Amorós A, Monaco H, Sambataro E, Clemente JC. Novel technologies to characterize and engineer the microbiome in inflammatory bowel disease. Gut Microbes 2022; 14:2107866. [PMID: 36104776 PMCID: PMC9481095 DOI: 10.1080/19490976.2022.2107866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We present an overview of recent experimental and computational advances in technology used to characterize the microbiome, with a focus on how these developments improve our understanding of inflammatory bowel disease (IBD). Specifically, we present studies that make use of flow cytometry and metabolomics assays to provide a functional characterization of microbial communities. We also describe computational methods for strain-level resolution, temporal series, mycobiome and virome data, co-occurrence networks, and compositional data analysis. In addition, we review novel techniques to therapeutically manipulate the microbiome in IBD. We discuss the benefits and drawbacks of these technologies to increase awareness of specific biases, and to facilitate a more rigorous interpretation of results and their potential clinical application. Finally, we present future lines of research to better characterize the relation between microbial communities and IBD pathogenesis and progression.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Department of Genetics and Genomic Sciences, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai. New York, NY, USA
| | - Hilary Monaco
- Department of Genetics and Genomic Sciences, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai. New York, NY, USA
| | - Elisa Sambataro
- Department of Biological Sciences, CUNY Hunter College, New York, NY, USA
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai. New York, NY, USA,CONTACT Jose C. Clemente Department of Genetics and Genomic Sciences, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai. New York, NY10029USA
| |
Collapse
|
31
|
Zhao H, Li Y, Lv P, Huang J, Tai R, Jin X, Wang J, Wang X. Salmonella Phages Affect the Intestinal Barrier in Chicks by Altering the Composition of Early Intestinal Flora: Association With Time of Phage Use. Front Microbiol 2022; 13:947640. [PMID: 35910610 PMCID: PMC9329052 DOI: 10.3389/fmicb.2022.947640] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/15/2022] [Indexed: 01/03/2023] Open
Abstract
Phages show promise in replacing antibiotics to treat or prevent bacterial diseases in the chicken breeding industry. Chicks are easily affected by their environment during early growth. Thus, this study investigated whether oral phages could affect the intestinal barrier function of chicks with a focus on the cecal microbiome. In a two-week trial, forty one-day-old hens were randomly divided into four groups: (1) NC, negative control; (2) Phage 1, 109 PFU phage/day (days 3–5); (3) Phage 2, 109 PFU phage/day (days 8–10); and (4) AMX, 1 mg/mL amoxicillin/day (days 8–10). High-throughput sequencing results of cecal contents showed that oral administration of phages significantly affected microbial community structure and community composition, and increased the relative abundance of Enterococcus. The number of different species in the Phage 1 group was much higher than that in the Phage 2 group, and differences in alpha and beta diversity also indicated that the magnitude of changes in the composition of the cecal microbiota correlated with the time of phage use. Particularly in the first stage of cecal microbiota development, oral administration of bacteriophages targeting Salmonella may cause substantial changes in chicks, as evidenced by the results of the PICRUSt2 software function prediction, reminding us to be cautious about the time of phage use in chicks and to avoid high oral doses of phages during the first stage. Additionally, the Phage 2 samples not only showed a significant increase in the relative abundance of Bifidobacterium and Subdoligranulum, but also improved the intestinal morphology (jejunum) and increased the mRNA expression level of occludin and ZO-1. We concluded that phages do not directly interact with eukaryotic cells. The enhancement of intestinal barrier function by phages in chicks may be related to changes in the intestinal flora induced by phages. This implies that phages may affect intestinal health by regulating the intestinal flora. This study provides new ideas for phage prevention of intestinal bacterial infections and promotes large-scale application of phages in the poultry industry.
Collapse
Affiliation(s)
- Hongze Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yue Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Peilin Lv
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinmei Huang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rong Tai
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiue Jin
- Hubei Provincial Institute of Veterinary Drug Control, Wuhan, China
| | - Jianhua Wang
- Hubei Provincial Institute of Veterinary Drug Control, Wuhan, China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Xiliang Wang,
| |
Collapse
|
32
|
Uyttebroek S, Chen B, Onsea J, Ruythooren F, Debaveye Y, Devolder D, Spriet I, Depypere M, Wagemans J, Lavigne R, Pirnay JP, Merabishvili M, De Munter P, Peetermans WE, Dupont L, Van Gerven L, Metsemakers WJ. Safety and efficacy of phage therapy in difficult-to-treat infections: a systematic review. THE LANCET INFECTIOUS DISEASES 2022; 22:e208-e220. [DOI: 10.1016/s1473-3099(21)00612-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/26/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022]
|
33
|
Duan Y, Young R, Schnabl B. Bacteriophages and their potential for treatment of gastrointestinal diseases. Nat Rev Gastroenterol Hepatol 2022; 19:135-144. [PMID: 34782783 PMCID: PMC8966578 DOI: 10.1038/s41575-021-00536-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 02/08/2023]
Abstract
Although bacteriophages have been overshadowed as therapeutic agents by antibiotics for decades, the emergence of multidrug-resistant bacteria and a better understanding of the role of the gut microbiota in human health and disease have brought them back into focus. In this Perspective, we briefly introduce basic phage biology and summarize recent discoveries about phages in relation to their role in the gut microbiota and gastrointestinal diseases, such as inflammatory bowel disease and chronic liver disease. In addition, we review preclinical studies and clinical trials of phage therapy for enteric disease and explore current challenges and potential future directions.
Collapse
Affiliation(s)
- Yi Duan
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Ry Young
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
- Center for Phage Technology, Texas A&M AgriLife Research and Texas A&M University, College Station, TX, USA
- Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
- Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
34
|
El Haddad L, Mendoza JF, Jobin C. Bacteriophage-mediated manipulations of microbiota in gastrointestinal diseases. Front Microbiol 2022; 13:1055427. [PMID: 36466675 PMCID: PMC9714271 DOI: 10.3389/fmicb.2022.1055427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
Although some gastrointestinal diseases could be managed using various antibiotics regimen, this therapeutic approach lacks precision and damages the microbiota. Emerging literature suggests that phages may play a key role in restoring the gut microbiome balance and controlling disease progression either with exogenous phage intervention or filtered fecal transplantation or even engineered phages. In this review, we will discuss the current phage applications aiming at controlling the bacterial population and preventing infection, inflammation, and cancer progression in the context of gastrointestinal diseases.
Collapse
Affiliation(s)
- Lynn El Haddad
- Department of Medicine, University of Florida, Gainesville, FL, United States.,Department of Molecular Genetics and Microbiology, Gainesville, FL, United States
| | - Jesus F Mendoza
- Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, FL, United States.,Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, United States.,Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
35
|
Lood C, Haas PJ, van Noort V, Lavigne R. Shopping for phages? Unpacking design rules for therapeutic phage cocktails. Curr Opin Virol 2021; 52:236-243. [PMID: 34971929 DOI: 10.1016/j.coviro.2021.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
In bacteriophage therapy, the combination of different phages into a single cocktail is of critical importance to overcome the narrow host range of single phage isolates. Today, the design of therapeutic cocktails is often akin to a black box and relies largely on intuition and (pre-)availability of isolates in local collections. Here we show that straightforward host range analysis can disclose design rules and we propose to apply/translate a data mining approach, historically applied in the field of marketing ('shopping cart analysis') to explore patterns in phage combinations. The technique is broadly applicable to host range datasets and can serve in combination with other molecular-based approaches to propose rationales for phage cocktail design.
Collapse
Affiliation(s)
- Cédric Lood
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, Leuven, Belgium; Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics, Laboratory of Computational Systems Biology, KU Leuven, Leuven, Belgium.
| | - Pieter-Jan Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vera van Noort
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics, Laboratory of Computational Systems Biology, KU Leuven, Leuven, Belgium; Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Rob Lavigne
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| |
Collapse
|
36
|
Abedon ST, Danis-Wlodarczyk KM, Alves DR. Phage Therapy in the 21st Century: Is There Modern, Clinical Evidence of Phage-Mediated Efficacy? Pharmaceuticals (Basel) 2021; 14:1157. [PMID: 34832939 PMCID: PMC8625828 DOI: 10.3390/ph14111157] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Many bacteriophages are obligate killers of bacteria. That this property could be medically useful was first recognized over one hundred years ago, with 2021 being the 100-year anniversary of the first clinical phage therapy publication. Here we consider modern use of phages in clinical settings. Our aim is to answer one question: do phages serve as effective anti-bacterial infection agents when used clinically? An important emphasis of our analyses is on whether phage therapy-associated anti-bacterial infection efficacy can be reasonably distinguished from that associated with often coadministered antibiotics. We find that about half of 70 human phage treatment reports-published in English thus far in the 2000s-are suggestive of phage-mediated anti-bacterial infection efficacy. Two of these are randomized, double-blinded, infection-treatment studies while 14 of those studies, in our opinion, provide superior evidence of a phage role in observed treatment successes. Roughly three-quarters of these potentially phage-mediated outcomes are based on microbiological as well as clinical results, with the rest based on clinical success. Since many of these phage treatments are of infections for which antibiotic therapy had not been successful, their collective effectiveness is suggestive of a valid utility in employing phages to treat otherwise difficult-to-cure bacterial infections.
Collapse
Affiliation(s)
- Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906, USA;
| | | | - Diana R. Alves
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906, USA;
| |
Collapse
|
37
|
Cookson TA. Using bacteriophages to characterize gut microbe interactions in situ. Med Hypotheses 2021; 158:110715. [PMID: 34753010 DOI: 10.1016/j.mehy.2021.110715] [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: 08/24/2021] [Accepted: 10/17/2021] [Indexed: 11/25/2022]
Abstract
As bacteriophage therapy is being investigated more as an alternative to antibiotics, laboratories are isolating and characterizing the functions of bacteriophages. Additionally, with large variations between gut microbiome studies and inconsistencies in results, there is a need for discrete characterization of specific gut microbes in situ. This hypothesis paper describes a method to utilize bacteriophages in order to outline the functions of specific gut bacteria in existing biological systems with minimal disturbance. Further, the effects of specific microbe depletion on gut bacterial composition and host health can theoretically also be measured.
Collapse
|
38
|
Isolation of a Virulent Aeromonas salmonicida subsp. masoucida Bacteriophage and Its Application in Phage Therapy in Turbot ( Scophthalmus maximus). Appl Environ Microbiol 2021; 87:e0146821. [PMID: 34406829 DOI: 10.1128/aem.01468-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aeromonas salmonicida is an aquatic pathogen that can infect a variety of fish. Phage therapy has been applied to treat bacterial infections. In this study, we obtained three A. salmonicida subsp. masoucida phage isolates from sewage, and one phage (vB_AsM_ZHF) exhibited the best antibacterial effect, based on in vitro kinetics experiments. Sequencing indicated that the vB_AsM_ZHF genome is 161,887 bp (41.24% C+G content) with 237 predicted open reading frames. No antibiotic resistance or virulence genes were detected in the complete genome, which is a requirement for phage therapy safety. Intraperitoneal injection of phage vB_AsM_ZHF into turbot at 8 × 104 PFU/fish rescued turbot from A. salmonicida subsp. masoucida injection and reduced the bacterial burden by 1 order of magnitude. Injection of vB_AsM_ZHF also decreased levels of inflammatory cell infiltration in muscle tissue, cytokines interleukin-1β (IL-1β), tumor necrosis factor alpha (TNF-α), and gamma interferon (IFN-γ) in serum and the expression of the inflammatory factors IL-1β, IL-6, IFN-γ, transforming growth factor β, TNF-α, and hepcidin in the liver, spleen, and head kidney of turbot. Phage vB_AsM_ZHF demonstrated antibacterial ability in vitro and in vivo and significantly reduced mortality in turbot challenged by A. salmonicida subsp. masoucida. This study revealed that phage vB_AsM_ZHF can effectively treat the infection caused by A. salmonicida subsp. masoucida in turbot. IMPORTANCE A. salmonicida is an aquatic pathogen that can infect different fish and causes economic loss to the global aquaculture industry. Clinical strains of A. salmonicida have developed multidrug resistance, and phage therapy is being evaluated for controlling bacterial infections. Phages are biological antibacterial agents and have the potential to be therapeutic agents against multidrug-resistant bacteria. In this study, three A. salmonicida subsp. masoucida phages were isolated from sewage, and their biological behaviors were characterized. The newly isolated phage vB_AsM_ZHF could inhibit A. salmonicida subsp. masoucida infection in vitro and in vivo, suggesting that it may be an alternative strategy to antibiotics for protecting fish against multidrug-resistant A. salmonicida subsp. masoucida in the aquaculture industry.
Collapse
|
39
|
Pallavali RR, Degati VL, Narala VR, Velpula KK, Yenugu S, Durbaka VRP. Lytic Bacteriophages Against Bacterial Biofilms Formed by Multidrug-Resistant Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, and Staphylococcus aureus Isolated from Burn Wounds. PHAGE (NEW ROCHELLE, N.Y.) 2021; 2:120-130. [PMID: 36161242 PMCID: PMC9041503 DOI: 10.1089/phage.2021.0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Background: Use of bacteriophages as antibiofilm agents to tackle multidrug-resistant bacteria has gained importance in recent years. Materials and Methods: In this study, biofilm formation by Staphylococcus aureus, Pseudomona aeruginosa, Klebsiella pneumoniae, and Escherichia coli under different growth conditions was studied. Furthermore, the ability of bacteriophages to inhibit biofilm formation was analyzed. Results: Under dynamic growth condition, wherein the medium is renewed for every 12 h, the amount of biomass produced and log10 colony-forming unit counts of all bacterial species studied was highest when compared with other growth conditions tested. Biomass of biofilms produced was drastically reduced when incubated for 2 or 4 h with bacteriophages vB_SAnS_SADP1, vB_PAnP_PADP4, vB_KPnM_KPDP1, and vB_ECnM_ECDP3. Scanning electron microscopy and confocal laser scanning microscopy analyses indicated that the reduction in biomass was due to the lytic action of the bacteriophages. Conclusions: Results of our study reinforce the concept of developing bacteriophages as alternatives to antibiotics to treat bacterial infections.
Collapse
Affiliation(s)
| | | | | | - Kiran Kumar Velpula
- Department of Cancer Biology and Pharmacology (Peoria), University of Illinois, Chicago, Peoria, Illinois, USA
| | - Suresh Yenugu
- Department of Animal Biology, University of Hyderabad, Hyderabad, India
- Address correspondence to: Suresh Yenugu, PhD, Department of Animal Biology, University of Hyderabad, Hyderabad 500046, India
| | - Vijaya Raghava Prasad Durbaka
- Department of Microbiology, Yogi Vemana University, Kadapa, India
- Address correspondence to: Vijaya Raghava Prasad Durbaka, PhD, Department of Microbiology, Yogi Vemana University, Kadapa 516005, India
| |
Collapse
|
40
|
Vlassov VV, Tikunova NV, Morozova VV. Bacteriophages as Therapeutic Preparations: What Restricts Their Application in Medicine. BIOCHEMISTRY (MOSCOW) 2021; 85:1350-1361. [PMID: 33280578 DOI: 10.1134/s0006297920110061] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The increasing prevalence of bacterial pathogens with multiple antibiotic resistance requires development of new approaches to control infections. Phage therapy is one of the most promising approaches. In recent years, research organizations and a number of pharmaceutical companies have intensified investigations aimed at developing bacteriophage-based therapeutics. In the United States and European countries, special centers have been established that experimentally apply phage therapy to treat patients who do not respond to antibiotic therapy. This review describes the features of bacteriophages as therapeutic tools, critically discusses the results of clinical trials of bacteriophage preparations, and assesses the prospects for using phage therapy to treat certain types of infectious diseases.
Collapse
Affiliation(s)
- V V Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - N V Tikunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - V V Morozova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
41
|
Figueiredo CM, Malvezzi Karwowski MS, da Silva Ramos RCP, de Oliveira NS, Peña LC, Carneiro E, Freitas de Macedo RE, Rosa EAR. Bacteriophages as tools for biofilm biocontrol in different fields. BIOFOULING 2021; 37:689-709. [PMID: 34304662 DOI: 10.1080/08927014.2021.1955866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
Microbial biofilms are difficult to control due to the limited accessibility that antimicrobial drugs and chemicals have to the entrapped inner cells. The extracellular matrix, binds water, contributes to altered cell physiology within biofilms and act as a barrier for most antiproliferative molecules. Thus, new strategies need to be developed to overcome biofilm vitality. In this review, based on 223 documents, the advantages, recommendations, and limitations of using bacteriophages as 'biofilm predators' are presented. The plausibility of using phages (bacteriophages and mycoviruses) to control biofilms grown in different environments is also discussed. The topics covered here include recent historical experiences in biofilm control/eradication using phages in medicine, dentistry, veterinary, and food industries, the pros and cons of their use, and the development of microbial resistance/immunity to such viruses.
Collapse
Affiliation(s)
| | | | | | | | - Lorena Caroline Peña
- Xenobiotics Research Unit, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Everdan Carneiro
- Graduate Program in Dentistry, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | | | - Edvaldo Antonio Ribeiro Rosa
- Graduate Program in Dentistry, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
- Graduate Program in Animal Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
- Xenobiotics Research Unit, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
42
|
Liu D, Van Belleghem JD, de Vries CR, Burgener E, Chen Q, Manasherob R, Aronson JR, Amanatullah DF, Tamma PD, Suh GA. The Safety and Toxicity of Phage Therapy: A Review of Animal and Clinical Studies. Viruses 2021; 13:1268. [PMID: 34209836 PMCID: PMC8310247 DOI: 10.3390/v13071268] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing rates of infection by antibiotic resistant bacteria have led to a resurgence of interest in bacteriophage (phage) therapy. Several phage therapy studies in animals and humans have been completed over the last two decades. We conducted a systematic review of safety and toxicity data associated with phage therapy in both animals and humans reported in English language publications from 2008-2021. Overall, 69 publications met our eligibility criteria including 20 animal studies, 35 clinical case reports or case series, and 14 clinical trials. After summarizing safety and toxicity data from these publications, we discuss potential approaches to optimize safety and toxicity monitoring with the therapeutic use of phage moving forward. In our systematic review of the literature, we found some adverse events associated with phage therapy, but serious events were extremely rare. Comprehensive and standardized reporting of potential toxicities associated with phage therapy has generally been lacking in the published literature. Structured safety and tolerability endpoints are necessary when phages are administered as anti-infective therapeutics.
Collapse
Affiliation(s)
- Dan Liu
- Department of Burn, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Jonas D. Van Belleghem
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Christiaan R. de Vries
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Elizabeth Burgener
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA;
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Robert Manasherob
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Jenny R. Aronson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Pranita D. Tamma
- Division of Pediatric Infectious Diseases, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Gina A. Suh
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
43
|
Huang Z, Zhang Z, Tong J, Malakar PK, Chen L, Liu H, Pan Y, Zhao Y. Phages and their lysins: Toolkits in the battle against foodborne pathogens in the postantibiotic era. Compr Rev Food Sci Food Saf 2021; 20:3319-3343. [PMID: 33938116 DOI: 10.1111/1541-4337.12757] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022]
Abstract
Worldwide, foods waste caused by putrefactive organisms and diseases caused by foodborne pathogens persist as public health problems even with a plethora of modern antimicrobials. Our over reliance on antimicrobials use in agriculture, medicine, and other fields will lead to a postantibiotic era where bacterial genotypic resistance, phenotypic adaptation, and other bacterial evolutionary strategies cause antimicrobial resistance (AMR). This AMR is evidenced by the emergence of multiple drug-resistant (MDR) bacteria and pan-resistant (PDR) bacteria, which produces cross-contamination in multiple fields and poses a more serious threat to food safety. A "red queen premise" surmises that the coevolution of phages and bacteria results in an evolutionary arms race that compels phages to adapt and survive bacterial antiphage strategies. Phages and their lysins are therefore useful toolkits in the design of novel antimicrobials in food protection and foodborne pathogens control, and the modality of using phages as a targeted vector against foodborne pathogens is gaining momentum based on many encouraging research outcomes. In this review, we discuss the rationale of using phages and their lysins as weapons against spoilage organisms and foodborne pathogens, and outline the targeted conquest or dodge mechanism of phages and the development of novel phage prospects. We also highlight the implementation of phages and their lysins to control foodborne pathogens in a farm-table-hospital domain in the postantibiotic era.
Collapse
Affiliation(s)
- Zhenhua Huang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Zhaohuan Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Jinrong Tong
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Pradeep K Malakar
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Liangbiao Chen
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Haiquan Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China.,Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Yingjie Pan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China.,Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China.,Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| |
Collapse
|
44
|
Marongiu L, Burkard M, Venturelli S, Allgayer H. Dietary Modulation of Bacteriophages as an Additional Player in Inflammation and Cancer. Cancers (Basel) 2021; 13:cancers13092036. [PMID: 33922485 PMCID: PMC8122878 DOI: 10.3390/cancers13092036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 01/06/2023] Open
Abstract
Natural compounds such as essential oils and tea have been used successfully in naturopathy and folk medicine for hundreds of years. Current research is unveiling the molecular role of their antibacterial, anti-inflammatory, and anticancer properties. Nevertheless, the effect of these compounds on bacteriophages is still poorly understood. The application of bacteriophages against bacteria has gained a particular interest in recent years due to, e.g., the constant rise of antimicrobial resistance to antibiotics, or an increasing awareness of different types of microbiota and their potential contribution to gastrointestinal diseases, including inflammatory and malignant conditions. Thus, a better knowledge of how dietary products can affect bacteriophages and, in turn, the whole gut microbiome can help maintain healthy homeostasis, reducing the risk of developing diseases such as diverse types of gastroenteritis, inflammatory bowel disease, or even cancer. The present review summarizes the effect of dietary compounds on the physiology of bacteriophages. In a majority of works, the substance class of polyphenols showed a particular activity against bacteriophages, and the primary mechanism of action involved structural damage of the capsid, inhibiting bacteriophage activity and infectivity. Some further dietary compounds such as caffeine, salt or oregano have been shown to induce or suppress prophages, whereas others, such as the natural sweeter stevia, promoted species-specific phage responses. A better understanding of how dietary compounds could selectively, and specifically, modulate the activity of individual phages opens the possibility to reorganize the microbial network as an additional strategy to support in the combat, or in prevention, of gastrointestinal diseases, including inflammation and cancer.
Collapse
Affiliation(s)
- Luigi Marongiu
- Department of Experimental Surgery—Cancer Metastasis, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany;
| | - Markus Burkard
- Department of Biochemistry of Nutrition, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany;
| | - Sascha Venturelli
- Department of Biochemistry of Nutrition, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany;
- Department of Vegetative and Clinical Physiology, University Hospital of Tuebingen, Otfried-Müllerstr. 27, 72076 Tuebingen, Germany
- Correspondence: (S.V.); (H.A.); Tel.: +49-(0)711-459-24113 (ext. 24195) (S.V.); +49-(0)621-383-71630 (ext. 71635) (H.A.); Fax: +49-(0)-711-459-23822 (S.V.); +49-(0)-621-383-71631 (H.A.)
| | - Heike Allgayer
- Department of Experimental Surgery—Cancer Metastasis, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany;
- Correspondence: (S.V.); (H.A.); Tel.: +49-(0)711-459-24113 (ext. 24195) (S.V.); +49-(0)621-383-71630 (ext. 71635) (H.A.); Fax: +49-(0)-711-459-23822 (S.V.); +49-(0)-621-383-71631 (H.A.)
| |
Collapse
|
45
|
Pinto AM, Silva MD, Pastrana LM, Bañobre-López M, Sillankorva S. The clinical path to deliver encapsulated phages and lysins. FEMS Microbiol Rev 2021; 45:6204673. [PMID: 33784387 DOI: 10.1093/femsre/fuab019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
The global emergence of multidrug-resistant pathogens is shaping the current dogma regarding the use of antibiotherapy. Many bacteria have evolved to become resistant to conventional antibiotherapy, representing a health and economic burden for those afflicted. The search for alternative and complementary therapeutic approaches has intensified and revived phage therapy. In recent decades, the exogenous use of lysins, encoded in phage genomes, has shown encouraging effectiveness. These two antimicrobial agents reduce bacterial populations; however, many barriers challenge their prompt delivery at the infection site. Encapsulation in delivery vehicles provides targeted therapy with a controlled compound delivery, surpassing chemical, physical and immunological barriers that can inactivate and eliminate them. This review explores phages and lysins' current use to resolve bacterial infections in the respiratory, digestive, and integumentary systems. We also highlight the different challenges they face in each of the three systems and discuss the advances towards a more expansive use of delivery vehicles.
Collapse
Affiliation(s)
- Ana Mafalda Pinto
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Maria Daniela Silva
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Lorenzo M Pastrana
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Manuel Bañobre-López
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Sanna Sillankorva
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| |
Collapse
|
46
|
Wu N, Zhu T. Potential of Therapeutic Bacteriophages in Nosocomial Infection Management. Front Microbiol 2021; 12:638094. [PMID: 33633717 PMCID: PMC7901949 DOI: 10.3389/fmicb.2021.638094] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/11/2021] [Indexed: 01/05/2023] Open
Abstract
Nosocomial infections (NIs) are hospital-acquired infections which pose a high healthcare burden worldwide. The impact of NIs is further aggravated by the global spread of antimicrobial resistance (AMR). Conventional treatment and disinfection agents are often insufficient to catch up with the increasing AMR and tolerance of the pathogenic bacteria. This has resulted in a need for alternative approaches and raised new interest in therapeutic bacteriophages (phages). In contrast to the limited clinical options available against AMR bacteria, the extreme abundance and biodiversity of phages in nature provides an opportunity to establish an ever-expanding phage library that collectively provides sustained broad-spectrum and poly microbial coverage. Given the specificity of phage-host interactions, phage susceptibility testing can serve as a rapid and cost-effective method for bacterial subtyping. The library can also provide a database for routine monitoring of nosocomial infections as a prelude to preparing ready-to-use phages for patient treatment and environmental sterilization. Despite the remaining obstacles for clinical application of phages, the establishment of phage libraries, pre-stocked phage vials prepared to good manufacturing practice (GMP) standards, and pre-optimized phage screening technology will facilitate efforts to make phages available as modern medicine. This may provide the breakthrough needed to demonstrate the great potential in nosocomial infection management.
Collapse
Affiliation(s)
- Nannan Wu
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tongyu Zhu
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Penziner S, Schooley RT, Pride DT. Animal Models of Phage Therapy. Front Microbiol 2021; 12:631794. [PMID: 33584632 PMCID: PMC7876411 DOI: 10.3389/fmicb.2021.631794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/11/2021] [Indexed: 01/21/2023] Open
Abstract
Amidst the rising tide of antibiotic resistance, phage therapy holds promise as an alternative to antibiotics. Most well-designed studies on phage therapy exist in animal models. In order to progress to human clinical trials, it is important to understand what these models have accomplished and determine how to improve upon them. Here we provide a review of the animal models of phage therapy in Western literature and outline what can be learned from them in order to bring phage therapy closer to becoming a feasible alternative to antibiotics in clinical practice.
Collapse
Affiliation(s)
- Samuel Penziner
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Robert T Schooley
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - David T Pride
- Department of Medicine, University of California, San Diego, San Diego, CA, United States.,Department of Pathology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
48
|
Low HZ, Böhnlein C, Sprotte S, Wagner N, Fiedler G, Kabisch J, Franz CMAP. Fast and Easy Phage-Tagging and Live/Dead Analysis for the Rapid Monitoring of Bacteriophage Infection. Front Microbiol 2021; 11:602444. [PMID: 33391221 PMCID: PMC7775415 DOI: 10.3389/fmicb.2020.602444] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/25/2020] [Indexed: 11/23/2022] Open
Abstract
Use of bacteriophages, which are viruses that kill bacteria, for biocontrol of pathogens and antimicrobial resistant bacteria has become increasingly important in recent years. As traditional culture-based methods are laborious and time-consuming, practicable use of bacteriophages will hinge on development of rapid and high throughput methods to analyze, characterize and screen large bacteriophage libraries. We thus established a novel method to fluorescently tag bacteriophages for virus screening and interaction studies, without the need for complicated and laborious purification procedures or genetic engineering of viruses to express fluorescent proteins. Bacteriophage PMBT14 was tagged using DNA dye Syto 13. Simply by using a membrane filter, tagged bacteriophages can be separated from non-sequestered excess dye rapidly, effortlessly, and cheaply. The procedure takes less than 30 min and makes use of simple laboratory consumables that are already commonly used for bacteriophage preparations. As proof of concept, we present here flow cytometric methods to analyze bacteriophage binding, infection and killing that are very accessible for high throughput analysis. We show that the resulting fluorescently tagged bacteriophage can be used to specifically stain its host bacterium Pseudomonas fluorescens DSM 50090. Individual fluorescent bacteriophages, their binding to and initial infection of bacteria could also be observed using confocal microscopy. The infection process was halted by the metabolic inhibitor sodium azide, suggesting a requirement of host metabolic processes for penetration by PMBT14. Flow cytometric live/dead assays was used as a complementary method to determine bacteriophage infection of its host. We made preliminary efforts to adapt the tagging method to two other bacteriophages and discuss potential pitfalls and solutions in the use of tagged phages. Fluorescent phage tagging has previously been demonstrated to facilitate analysis of bacteriophage–host interactions. The method adopted in this study makes it fast, easy as well as cost effective.
Collapse
Affiliation(s)
- Hui Zhi Low
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Christina Böhnlein
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Sabrina Sprotte
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Natalia Wagner
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Gregor Fiedler
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Jan Kabisch
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Charles M A P Franz
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| |
Collapse
|
49
|
Łusiak-Szelachowska M, Weber-Dąbrowska B, Żaczek M, Borysowski J, Górski A. The Presence of Bacteriophages in the Human Body: Good, Bad or Neutral? Microorganisms 2020; 8:E2012. [PMID: 33339331 PMCID: PMC7767151 DOI: 10.3390/microorganisms8122012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
The presence of bacteriophages (phages) in the human body may impact bacterial microbiota and modulate immunity. The role of phages in human microbiome studies and diseases is poorly understood. However, the correlation between a greater abundance of phages in the gut in ulcerative colitis and diabetes has been suggested. Furthermore, most phages found at different sites in the human body are temperate, so their therapeutic effects and their potential beneficial effects remain unclear. Hence, far, no correlation has been observed between the presence of widespread crAssphage in the human population and human health and diseases. Here, we emphasize the beneficial effects of phage transfer in fecal microbiota transplantation (FMT) in Clostridioides difficile infection. The safety of phage use in gastrointestinal disorders has been demonstrated in clinical studies. The significance of phages in the FMT as well as in gastrointestinal disorders remains to be established. An explanation of the multifaceted role of endogenous phages for the development of phage therapy is required.
Collapse
Affiliation(s)
- Marzanna Łusiak-Szelachowska
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ł.-S.); (B.W.-D.); (M.Ż.)
| | - Beata Weber-Dąbrowska
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ł.-S.); (B.W.-D.); (M.Ż.)
- Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
| | - Maciej Żaczek
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ł.-S.); (B.W.-D.); (M.Ż.)
| | - Jan Borysowski
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, 02-006 Warsaw, Poland;
| | - Andrzej Górski
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ł.-S.); (B.W.-D.); (M.Ż.)
- Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Infant Jesus Hospital, Medical University of Warsaw, 02-005 Warsaw, Poland
| |
Collapse
|
50
|
Gutiérrez B, Domingo-Calap P. Phage Therapy in Gastrointestinal Diseases. Microorganisms 2020; 8:microorganisms8091420. [PMID: 32947790 PMCID: PMC7565598 DOI: 10.3390/microorganisms8091420] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal tract microbiota plays a key role in the regulation of the pathogenesis of several gastrointestinal diseases. In particular, the viral fraction, composed essentially of bacteriophages, influences homeostasis by exerting a selective pressure on the bacterial communities living in the tract. Gastrointestinal inflammatory diseases are mainly induced by bacteria, and have risen due to the emergence of antibiotic resistant strains. In the lack of effective treatments, phage therapy has been proposed as a clinical alternative to restore intestinal eubiosis, thanks to its immunomodulatory and bactericidal effect against bacterial pathogens, such as Clostridioides difficile in ulcerative colitis and invasive adherent Escherichia coli in Crohn’s disease. In addition, genetically modified temperate phages could be used to suppress the transcription of bacterial virulence factors. In this review, we will highlight the latest advances in research in the field, as well as the clinical trials based on phage therapy in the area of gastroenterology.
Collapse
Affiliation(s)
- Beatriz Gutiérrez
- Department of Genetics, Universitat de València, 46100 Valencia, Spain;
| | - Pilar Domingo-Calap
- Department of Genetics, Universitat de València, 46100 Valencia, Spain;
- Institute for Integrative Systems Biology, ISysBio, Universitat de València-CSIC, 46980 Valencia, Spain
- Correspondence: ; Tel.: +34-963-543-261
| |
Collapse
|