Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (85)

Search Parameters:
Keywords = therapeutic phage products

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 442 KiB  
Review
Enhancing Agricultural Productivity in Dairy Cow Mastitis Management: Innovations in Non-Antibiotic Treatment Technologies
by Lijie Jiang, Qi Li, Huiqing Liao, Hourong Liu and Zhiqiang Wang
Vet. Sci. 2025, 12(7), 662; https://doi.org/10.3390/vetsci12070662 - 12 Jul 2025
Cited by 1 | Viewed by 568
Abstract
Dairy mastitis is a common dairy farming disease. It severely affects the health of dairy cows and the quality and yield of dairy products. This paper reviews the main current mastitis treatments and associated bacterial resistance. It emphasizes the importance of integrated resistance [...] Read more.
Dairy mastitis is a common dairy farming disease. It severely affects the health of dairy cows and the quality and yield of dairy products. This paper reviews the main current mastitis treatments and associated bacterial resistance. It emphasizes the importance of integrated resistance and treatment management. The therapeutic efficacy and resistance associated with commonly used antibiotics such as penicillin, cephalosporins, macrolides and fluoroquinolones are analyzed. The principles, application effects and benefits of non-antibiotic therapies are also discussed, including those of immunotherapy, herbal therapy, probiotic therapy and phage therapy. The paper presents the latest gene editing and nanotechnology advances in the contexts of big data and artificial intelligence. It suggests future research directions such as developing new antibiotics, optimizing treatment and enhancing immunity. In conclusion, effective treatment and management can control dairy cow mastitis. It can guarantee cow health, improve dairy product quality and promote sustainable dairy industry development. Full article
(This article belongs to the Special Issue Exploring Innovative Approaches in Veterinary Health)
Show Figures

Figure 1

27 pages, 1648 KiB  
Review
Carbapenem Resistance in Acinetobacter baumannii: Mechanisms, Therapeutics, and Innovations
by Joyce de Souza, Helena Regina Salomé D’Espindula, Isabel de Farias Ribeiro, Geiziane Aparecida Gonçalves, Marcelo Pillonetto and Helisson Faoro
Microorganisms 2025, 13(7), 1501; https://doi.org/10.3390/microorganisms13071501 - 27 Jun 2025
Viewed by 1513
Abstract
The global rise of carbapenem-resistant Acinetobacter baumannii (CRAB) strains poses a critical challenge to healthcare systems due to limited therapeutic options and high mortality rates, especially in intensive care settings. This review explores the epidemiological landscape and molecular mechanisms driving carbapenem resistance, including [...] Read more.
The global rise of carbapenem-resistant Acinetobacter baumannii (CRAB) strains poses a critical challenge to healthcare systems due to limited therapeutic options and high mortality rates, especially in intensive care settings. This review explores the epidemiological landscape and molecular mechanisms driving carbapenem resistance, including the production of diverse beta-lactamases (particularly OXA-type enzymes), porin loss, efflux pump overexpression, and mutations in antibiotic targets. Emerging treatment strategies are discussed, such as the use of new beta-lactam–beta-lactamase inhibitor combinations (e.g., sulbactam–durlobactam), siderophore cephalosporins, next-generation polymyxins, as well as novel agents like zosurabalpin and rifabutin (BV100). Alternative approaches—including phage therapy, antimicrobial peptides, CRISPR-based gene editing, and nanoparticle-based delivery systems—are also evaluated for their potential to bypass traditional resistance mechanisms. Furthermore, advances in artificial intelligence and multi-omics integration are highlighted as tools for identifying novel drug targets and predicting resistance profiles. Together, these innovations represent a multifaceted strategy to overcome CRAB infections, yet their successful implementation requires further clinical validation and coordinated surveillance efforts. This analysis highlights the urgent need for continued investment in innovative treatments and effective resistance monitoring to limit the spread of CRAB and protect the effectiveness of last-line antibiotics. Full article
(This article belongs to the Special Issue Combating Antimicrobial Resistance: Innovations and Strategies)
Show Figures

Figure 1

23 pages, 3549 KiB  
Article
Immunomodulatory Effects of Escherichia coli Phage GADS24 on Human Dendritic Cells
by Alia M. Aldahlawi, Ghadah A. Alsubhi, Jehan S. Alrahimi, Fatemah S. Basingab and Kawther A. Zaher
Biomedicines 2025, 13(7), 1519; https://doi.org/10.3390/biomedicines13071519 - 21 Jun 2025
Viewed by 383
Abstract
Background: Multidrug-resistant (MDR) Escherichia coli (E. coli) strains pose a significant public health challenge, which has led to the exploration of alternative therapeutic strategies. Due to their antibacterial and immunomodulatory properties, bacteriophages have emerged as promising therapeutic agents. Methods: This study [...] Read more.
Background: Multidrug-resistant (MDR) Escherichia coli (E. coli) strains pose a significant public health challenge, which has led to the exploration of alternative therapeutic strategies. Due to their antibacterial and immunomodulatory properties, bacteriophages have emerged as promising therapeutic agents. Methods: This study investigates the effects of GADS24, a novel lytic bacteriophage of E. coli, on human-monocyte-derived dendritic cells (DCs). DCs are exposed to purified GADS24 phage, bacterial lysate, or a combination of both. Flow cytometry was used to assess the expression of surface markers (HLA-DR, CD80, CD83, and CD86), and ELISA was used to measure cytokine production (IL-10 and IL-12p70). Results: Following treatment with bacterial lysate, a significant increase in DC maturation markers was observed. The GADS24 phage alone induced a moderate upregulation of these markers, decreased IL-10 secretion, and increased IL-12p70. Combining bacterial lysate and phage tempered the maturation response compared to the lysate treatment alone. Conclusion: These findings suggest that GADS24 exerts antibacterial activity and modulates host immunity by influencing DC maturation and cytokine production. Due to its dual antimicrobial and immunomodulatory functions, GADS24 is likely to be a valuable adjunctive therapy for multidrug-resistant (MDR) bacterial infections. Furthermore, in vivo studies are necessary to confirm these promising in vitro results. Full article
Show Figures

Figure 1

22 pages, 2019 KiB  
Article
A Single-Domain VNAR Nanobody Binds with High-Affinity and Selectivity to the Heparin Pentasaccharide Fondaparinux
by Martha Gschwandtner, Rupert Derler, Elisa Talker, Christina Trojacher, Nina Gubensäk, Walter Becker, Tanja Gerlza, Zangger Klaus, Pawel Stocki, Frank S. Walsh, Julia Lynn Rutkowski and Andreas Kungl
Int. J. Mol. Sci. 2025, 26(9), 4045; https://doi.org/10.3390/ijms26094045 - 24 Apr 2025
Viewed by 877
Abstract
Glycosaminoglycans (GAGs) are key ligands for proteins involved in physiological and pathological processes. Specific GAG-binding patterns are rarely identified, with the heparin pentasaccharide as an Antithrombin-III ligand being the best characterized. Generating glycan-specific antibodies is difficult due to their size, pattern dispersion, and [...] Read more.
Glycosaminoglycans (GAGs) are key ligands for proteins involved in physiological and pathological processes. Specific GAG-binding patterns are rarely identified, with the heparin pentasaccharide as an Antithrombin-III ligand being the best characterized. Generating glycan-specific antibodies is difficult due to their size, pattern dispersion, and flexibility. Single-domain variable new antigen receptors (VNAR nanobodies) from nurse sharks are highly soluble, stable, and versatile. Their unique properties suggest advantages over conventional antibodies, particularly for challenging biotherapeutic targets. Here we have used VNAR semi-synthetic phage libraries to select high-affinity fondaparinux-binding VNARs that did not show cross-reactivity with other GAG species. Competition ELISA and surface plasmon resonance identified a single fondaparinux-selective VNAR clone. This VNAR exhibited an extraordinarily stable protein fold: the beta-strands are stabilized by a robust hydrophobic network, as revealed by heteronuclear NMR. Docking fondaparinux to the VNAR structure revealed a large contact surface area between the CDR3 loop of the antibody and the glycan. Fusing the VNAR with a human Fc domain resulted in a stable product with a high affinity for fondaparinux (Kd = 9.3 × 10−8 M) that could efficiently discriminate between fondaparinux and other glycosaminoglycans. This novel glycan-targeting screening technology represents a promising therapeutic strategy for addressing GAG-related diseases. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

32 pages, 1181 KiB  
Review
Skin Microbiota: Mediator of Interactions Between Metabolic Disorders and Cutaneous Health and Disease
by Magdalini Kreouzi, Nikolaos Theodorakis, Maria Nikolaou, Georgios Feretzakis, Athanasios Anastasiou, Konstantinos Kalodanis and Aikaterini Sakagianni
Microorganisms 2025, 13(1), 161; https://doi.org/10.3390/microorganisms13010161 - 14 Jan 2025
Cited by 6 | Viewed by 3467
Abstract
Metabolic disorders, including type 2 diabetes mellitus (T2DM), obesity, and metabolic syndrome, are systemic conditions that profoundly impact the skin microbiota, a dynamic community of bacteria, fungi, viruses, and mites essential for cutaneous health. Dysbiosis caused by metabolic dysfunction contributes to skin barrier [...] Read more.
Metabolic disorders, including type 2 diabetes mellitus (T2DM), obesity, and metabolic syndrome, are systemic conditions that profoundly impact the skin microbiota, a dynamic community of bacteria, fungi, viruses, and mites essential for cutaneous health. Dysbiosis caused by metabolic dysfunction contributes to skin barrier disruption, immune dysregulation, and increased susceptibility to inflammatory skin diseases, including psoriasis, atopic dermatitis, and acne. For instance, hyperglycemia in T2DM leads to the formation of advanced glycation end products (AGEs), which bind to the receptor for AGEs (RAGE) on keratinocytes and immune cells, promoting oxidative stress and inflammation while facilitating Staphylococcus aureus colonization in atopic dermatitis. Similarly, obesity-induced dysregulation of sebaceous lipid composition increases saturated fatty acids, favoring pathogenic strains of Cutibacterium acnes, which produce inflammatory metabolites that exacerbate acne. Advances in metabolomics and microbiome sequencing have unveiled critical biomarkers, such as short-chain fatty acids and microbial signatures, predictive of therapeutic outcomes. For example, elevated butyrate levels in psoriasis have been associated with reduced Th17-mediated inflammation, while the presence of specific Lactobacillus strains has shown potential to modulate immune tolerance in atopic dermatitis. Furthermore, machine learning models are increasingly used to integrate multi-omics data, enabling personalized interventions. Emerging therapies, such as probiotics and postbiotics, aim to restore microbial diversity, while phage therapy selectively targets pathogenic bacteria like Staphylococcus aureus without disrupting beneficial flora. Clinical trials have demonstrated significant reductions in inflammatory lesions and improved quality-of-life metrics in patients receiving these microbiota-targeted treatments. This review synthesizes current evidence on the bidirectional interplay between metabolic disorders and skin microbiota, highlighting therapeutic implications and future directions. By addressing systemic metabolic dysfunction and microbiota-mediated pathways, precision strategies are paving the way for improved patient outcomes in dermatologic care. Full article
(This article belongs to the Special Issue Human Skin Microbiota, 2nd Edition)
Show Figures

Figure 1

34 pages, 4591 KiB  
Review
Phytochemicals Controlling Enterohemorrhagic Escherichia coli (EHEC) Virulence—Current Knowledge of Their Mechanisms of Action
by Patryk Strzelecki, Monika Karczewska, Agnieszka Szalewska-Pałasz and Dariusz Nowicki
Int. J. Mol. Sci. 2025, 26(1), 381; https://doi.org/10.3390/ijms26010381 - 4 Jan 2025
Cited by 3 | Viewed by 2296
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a common pathotype of E. coli that causes numerous outbreaks of foodborne illnesses. EHEC is a zoonotic pathogen that is transmitted from animals to humans. Ruminants, particularly cattle, are considered important reservoirs for virulent EHEC strains. Humans can [...] Read more.
Enterohemorrhagic Escherichia coli (EHEC) is a common pathotype of E. coli that causes numerous outbreaks of foodborne illnesses. EHEC is a zoonotic pathogen that is transmitted from animals to humans. Ruminants, particularly cattle, are considered important reservoirs for virulent EHEC strains. Humans can become infected with EHEC through the consumption of contaminated food and water or through direct contact with infected animals or humans. E. coli O157:H7 is one of the most commonly reported causes of foodborne illnesses in developed countries. The formation of attaching and effacing (A/E) lesions on the intestinal epithelium, combined with Shiga toxin production, is a hallmark of EHEC infection and can lead to lethal hemolytic–uremic syndrome (HUS). For the phage-dependent regulation of Shiga toxin production, antibiotic treatment is contraindicated, as it may exacerbate toxin production, limiting therapeutic options to supportive care. In response to this challenge and the growing threat of antibiotic resistance, phytochemicals have emerged as promising antivirulence agents. These plant-derived compounds target bacterial virulence mechanisms without promoting resistance. Therefore, the aim of this study is to summarize the recent knowledge on the use of phytochemicals targeting EHEC. We focused on the molecular basis of their action, targeting the principal virulence determinants of EHEC. Full article
(This article belongs to the Special Issue Biological Research on Plant Bioactive Compounds)
Show Figures

Figure 1

55 pages, 1734 KiB  
Review
Advancing Phage Therapy: A Comprehensive Review of the Safety, Efficacy, and Future Prospects for the Targeted Treatment of Bacterial Infections
by Marco Palma and Bowen Qi
Infect. Dis. Rep. 2024, 16(6), 1127-1181; https://doi.org/10.3390/idr16060092 - 28 Nov 2024
Cited by 17 | Viewed by 7756
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, [...] Read more.
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. Full article
(This article belongs to the Section Bacterial Diseases)
Show Figures

Figure 1

16 pages, 301 KiB  
Review
Phage-Based Therapy in Combination with Antibiotics: A Promising Alternative against Multidrug-Resistant Gram-Negative Pathogens
by Cleo Anastassopoulou, Stefanos Ferous, Aikaterini Petsimeri, Georgia Gioula and Athanasios Tsakris
Pathogens 2024, 13(10), 896; https://doi.org/10.3390/pathogens13100896 - 14 Oct 2024
Cited by 8 | Viewed by 6006
Abstract
The continued rise in antimicrobial resistance poses a serious threat to public health worldwide. The use of phages that can have bactericidal activity without disrupting the normal flora represents a promising alternative treatment method. This practice has been successfully applied for decades, mainly [...] Read more.
The continued rise in antimicrobial resistance poses a serious threat to public health worldwide. The use of phages that can have bactericidal activity without disrupting the normal flora represents a promising alternative treatment method. This practice has been successfully applied for decades, mainly in Eastern Europe, and has recently been used as an emergency therapy for compassionate care in the United States. Here, we provide a comprehensive review of the pre-clinical and clinical applications of phage therapy concerning three major Gram-negative pathogens: Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The advantages and the challenges of expanding the usage of phages as an alternative or adjunctive treatment for antimicrobial-resistant bacterial infections are discussed. We emphasize the virologic complexities of using the highly adaptable phage populations as molecular tools, along with antibiotic chemical compounds, to effectively combat rapidly coevolving pathogenic bacteria in the host microenvironment. Pre-clinical studies, isolated clinical reports and a few randomized clinical trials have shown that bacteriophages can be effective in treating multidrug-resistant bacterial infections. The ability of some phages to revert the resistance against antibiotics, and possibly also against the human complement and other phages, appears to be a great advantage of phage therapy despite the inevitable emergence of phage-resistant strains. Bacteriophages (or specific phage-derived products) can enhance antimicrobial efficacy by reducing bacterial virulence via the alteration of basic bacterial structures, primarily of the cellular wall and membrane. Although several issues remain open regarding their effective clinical application, it appears that phage-based therapeutics in combination with antibiotics can provide an effective solution to the spread of antimicrobial resistance. Full article
17 pages, 1771 KiB  
Review
Mycobacteriophages and Their Applications
by Andrea Bonacorsi, Caterina Ferretti, Mariagrazia Di Luca and Laura Rindi
Antibiotics 2024, 13(10), 926; https://doi.org/10.3390/antibiotics13100926 - 27 Sep 2024
Cited by 3 | Viewed by 3080
Abstract
Mycobacterial infections caused by tuberculous and non-tuberculous strains pose significant treatment challenges, especially among immunocompromised patients. Conventional antibiotic therapies often fail due to bacterial resistance, highlighting the need for alternative therapeutic strategies. Mycobacteriophages are emerging as promising candidates for the treatment of mycobacteria. [...] Read more.
Mycobacterial infections caused by tuberculous and non-tuberculous strains pose significant treatment challenges, especially among immunocompromised patients. Conventional antibiotic therapies often fail due to bacterial resistance, highlighting the need for alternative therapeutic strategies. Mycobacteriophages are emerging as promising candidates for the treatment of mycobacteria. This review comprehensively explores phage isolation, characterization, and clinical applications. Despite the need for more extensive in vitro and in vivo studies, existing evidence shows their efficacy against both sensitive and antibiotic-resistant mycobacterial strains, even under disease-mimicking conditions, particularly when used in cocktails to minimize resistance development. Mycobacteriophages can be engineered and evolved to overcome limitations associated with lysogeny and narrow host range. Furthermore, they exhibit activity in ex vivo and in vivo infection models, successfully targeting mycobacteria residing within macrophages. Delivery methods such as bacterial and liposomal vectors facilitate their entry into human cells. Considering the potential for phage-treatment-induced bacterial resistance, as described in this review, the combination of mycobacteriophages with antibiotics shows efficacy in countering mycobacterial growth, both in the laboratory setting and in animal models. Interestingly, phage-encoded products can potentiate the activity of relevant antibiotics. Finally, the application of phages in different compassionate cases is reported. The positive outcomes indicate that phage therapy represents a promising solution for the treatment of antibiotic-resistant mycobacteria. Full article
Show Figures

Graphical abstract

22 pages, 976 KiB  
Article
Comprehensive Genomic Analysis of Uropathogenic E. coli: Virulence Factors, Antimicrobial Resistance, and Mobile Genetic Elements
by Kidon Sung, Mohamed Nawaz, Miseon Park, Jungwhan Chon, Saeed A. Khan, Khulud Alotaibi and Ashraf A. Khan
Pathogens 2024, 13(9), 794; https://doi.org/10.3390/pathogens13090794 - 13 Sep 2024
Cited by 3 | Viewed by 2915
Abstract
Our whole-genome sequencing analysis of sixteen uropathogenic E. coli isolates revealed a concerning picture of multidrug resistance and potentially virulent bacteria. All isolates belonged to four distinct clonal groups, with the highly prevalent ST131 lineage being associated with extensive antibiotic resistance and virulence [...] Read more.
Our whole-genome sequencing analysis of sixteen uropathogenic E. coli isolates revealed a concerning picture of multidrug resistance and potentially virulent bacteria. All isolates belonged to four distinct clonal groups, with the highly prevalent ST131 lineage being associated with extensive antibiotic resistance and virulence factors. Notably, all isolates exhibited multidrug resistance, with some resistant to as many as 12 antibiotics. Fluoroquinolone resistance stemmed primarily from efflux pumps and mutations in gyrase and topoisomerase genes. Additionally, we identified genes encoding resistance to extended-spectrum cephalosporins, trimethoprim/sulfamethoxazole, and various heavy metals. The presence of diverse plasmids and phages suggests the potential for horizontal gene transfer and the dissemination of virulence factors. All isolates harbored genomic islands containing virulence factors associated with adhesion, biofilm formation, and invasion. Genes essential for iron acquisition, flagella biosynthesis, secretion systems, and toxin production were also prevalent. Adding further complexity to understanding the isolates’ genetic makeup, we identified CRISPR-Cas systems. This study underscores the need for continued genomic surveillance in understanding the pathogenic mechanisms and resistance profiles of uropathogenic E. coli to aid in developing targeted therapeutic strategies. Full article
(This article belongs to the Special Issue Antimicrobial Resistance of Pathogens Causing Nosocomial Infections)
Show Figures

Figure 1

14 pages, 2589 KiB  
Article
Identification of a Fully Human Antibody VH Domain Targeting Anaplastic Lymphoma Kinase (ALK) with Applications in ALK-Positive Solid Tumor Immunotherapy
by Chuan Chen, Zehua Sun, Zening Wang, Seungmin Shin, Abigail Berrios, John W. Mellors, Dimiter S. Dimitrov and Wei Li
Antibodies 2024, 13(2), 39; https://doi.org/10.3390/antib13020039 - 7 May 2024
Cited by 2 | Viewed by 2920
Abstract
The anaplastic lymphoma kinase (ALK, CD247) is a potential target for antibody-based therapy. However, no antibody-based therapeutics targeting ALK have entered clinical trials, necessitating the development of novel antibodies with unique therapeutic merits. Single-domain antibodies (sdAb) bear therapeutic advantages compared to the full-length [...] Read more.
The anaplastic lymphoma kinase (ALK, CD247) is a potential target for antibody-based therapy. However, no antibody-based therapeutics targeting ALK have entered clinical trials, necessitating the development of novel antibodies with unique therapeutic merits. Single-domain antibodies (sdAb) bear therapeutic advantages compared to the full-length antibody including deeper tumor penetration, cost-effective production and fast washout from normal tissues. In this study, we identified a human immunoglobulin heavy chain variable domain (VH domain) (VH20) from an in-house phage library. VH20 exhibits good developability and high specificity with no off-target binding to ~6000 human membrane proteins. VH20 efficiently bound to the glycine-rich region of ALK with an EC50 of 0.4 nM and a KD of 6.54 nM. Both VH20-based bispecific T cell engager (TCE) and chimeric antigen receptor T cells (CAR Ts) exhibited potent cytolytic activity to ALK-expressing tumor cells in an ALK-dependent manner. VH20 CAR Ts specifically secreted proinflammatory cytokines including IL-2, TNFα and IFNγ after incubation with ALK-positive cells. To our knowledge, this is the first reported human single-domain antibody against ALK. Our in vitro characterization data indicate that VH20 could be a promising ALK-targeting sdAb with potential applications in ALK-expressing tumors, including neuroblastoma (NBL) and non-small cell lung cancer. Full article
Show Figures

Figure 1

19 pages, 750 KiB  
Review
The Profound Influence of Gut Microbiome and Extracellular Vesicles on Animal Health and Disease
by Muttiah Barathan, Sook Luan Ng, Yogeswaran Lokanathan, Min Hwei Ng and Jia Xian Law
Int. J. Mol. Sci. 2024, 25(7), 4024; https://doi.org/10.3390/ijms25074024 - 4 Apr 2024
Cited by 28 | Viewed by 6045
Abstract
The animal gut microbiota, comprising a diverse array of microorganisms, plays a pivotal role in shaping host health and physiology. This review explores the intricate dynamics of the gut microbiome in animals, focusing on its composition, function, and impact on host–microbe interactions. The [...] Read more.
The animal gut microbiota, comprising a diverse array of microorganisms, plays a pivotal role in shaping host health and physiology. This review explores the intricate dynamics of the gut microbiome in animals, focusing on its composition, function, and impact on host–microbe interactions. The composition of the intestinal microbiota in animals is influenced by the host ecology, including factors such as temperature, pH, oxygen levels, and nutrient availability, as well as genetic makeup, diet, habitat, stressors, and husbandry practices. Dysbiosis can lead to various gastrointestinal and immune-related issues in animals, impacting overall health and productivity. Extracellular vesicles (EVs), particularly exosomes derived from gut microbiota, play a crucial role in intercellular communication, influencing host health by transporting bioactive molecules across barriers like the intestinal and brain barriers. Dysregulation of the gut–brain axis has implications for various disorders in animals, highlighting the potential role of microbiota-derived EVs in disease progression. Therapeutic approaches to modulate gut microbiota, such as probiotics, prebiotics, microbial transplants, and phage therapy, offer promising strategies for enhancing animal health and performance. Studies investigating the effects of phage therapy on gut microbiota composition have shown promising results, with potential implications for improving animal health and food safety in poultry production systems. Understanding the complex interactions between host ecology, gut microbiota, and EVs provides valuable insights into the mechanisms underlying host–microbe interactions and their impact on animal health and productivity. Further research in this field is essential for developing effective therapeutic interventions and management strategies to promote gut health and overall well-being in animals. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

32 pages, 407 KiB  
Article
The Medicinal Phage—Regulatory Roadmap for Phage Therapy under EU Pharmaceutical Legislation
by Timo Faltus
Viruses 2024, 16(3), 443; https://doi.org/10.3390/v16030443 - 12 Mar 2024
Cited by 23 | Viewed by 6442
Abstract
Bacteriophage therapy is a promising approach to treating bacterial infections. Research and development of bacteriophage therapy is intensifying due to the increase in antibiotic resistance and the faltering development of new antibiotics. Bacteriophage therapy uses bacteriophages (phages), i.e., prokaryotic viruses, to specifically target [...] Read more.
Bacteriophage therapy is a promising approach to treating bacterial infections. Research and development of bacteriophage therapy is intensifying due to the increase in antibiotic resistance and the faltering development of new antibiotics. Bacteriophage therapy uses bacteriophages (phages), i.e., prokaryotic viruses, to specifically target and kill pathogenic bacteria. The legal handling of this type of therapy raises several questions. These include whether phage therapeutics belong to a specially regulated class of medicinal products, and which legal framework should be followed with regard to the various technical ways in which phage therapeutics can be manufactured and administered. The article shows to which class of medicinal products phage therapeutics from wild type phages and from genetically modified (designer) phages do or do not belong. Furthermore, the article explains which legal framework is relevant for the manufacture and administration of phage therapeutics, which are manufactured in advance in a uniform, patient-independent manner, and for tailor-made patient-specific phage therapeutics. For the systematically coherent, successful translation of phage therapy, the article considers pharmaceutical law and related legal areas, such as genetic engineering law. Finally, the article shows how the planned legislative revisions of Directive 2001/83/EC and Regulation (EC) No 726/2004 may affect the legal future of phage therapy. Full article
(This article belongs to the Section Bacterial Viruses)
15 pages, 3888 KiB  
Article
A Novel Anti-CD47 Nanobody Tetramer for Cancer Therapy
by Nataliya M. Ratnikova, Yulia Kravchenko, Anna Ivanova, Vladislav Zhuchkov, Elena Frolova and Stepan Chumakov
Antibodies 2024, 13(1), 2; https://doi.org/10.3390/antib13010002 - 2 Jan 2024
Cited by 7 | Viewed by 4560
Abstract
CD47 acts as a defense mechanism for tumor cells by sending a “don’t eat me” signal via its bond with SIRPα. With CD47’s overexpression linked to poor cancer outcomes, its pathway has become a target in cancer immunotherapy. Though monoclonal antibodies offer specificity, [...] Read more.
CD47 acts as a defense mechanism for tumor cells by sending a “don’t eat me” signal via its bond with SIRPα. With CD47’s overexpression linked to poor cancer outcomes, its pathway has become a target in cancer immunotherapy. Though monoclonal antibodies offer specificity, they have limitations like the large size and production costs. Nanobodies, due to their small size and unique properties, present a promising therapeutic alternative. In our study, a high-affinity anti-CD47 nanobody was engineered from an immunized alpaca. We isolated a specific VHH from the phage library, which has nanomolar affinity to SIRPα, and constructed a streptavidin-based tetramer. The efficacy of the nanobody and its derivative was evaluated using various assays. The new nanobody demonstrated higher affinity than the monoclonal anti-CD47 antibody, B6H12.2. The nanobody and its derivatives also stimulated substantial phagocytosis of tumor cell lines and induced apoptosis in U937 cells, a response confirmed in both in vitro and in vivo settings. Our results underscore the potential of the engineered anti-CD47 nanobody as a promising candidate for cancer immunotherapy. The derived nanobody could offer a more effective, cost-efficient alternative to conventional antibodies in disrupting the CD47–SIRPα axis, opening doors for its standalone or combinatorial therapeutic applications in oncology. Full article
Show Figures

Figure 1

16 pages, 1816 KiB  
Article
Prophage Carriage and Genetic Diversity within Environmental Isolates of Clostridioides difficile
by Khald Blau and Claudia Gallert
Int. J. Mol. Sci. 2024, 25(1), 2; https://doi.org/10.3390/ijms25010002 - 19 Dec 2023
Cited by 2 | Viewed by 2101
Abstract
Clostridioides difficile is an important human pathogen causing antibiotic-associated diarrhoea worldwide. Besides using antibiotics for treatment, the interest in bacteriophages as an alternative therapeutic option has increased. Prophage abundance and genetic diversity are well-documented in clinical strains, but the carriage of prophages in [...] Read more.
Clostridioides difficile is an important human pathogen causing antibiotic-associated diarrhoea worldwide. Besides using antibiotics for treatment, the interest in bacteriophages as an alternative therapeutic option has increased. Prophage abundance and genetic diversity are well-documented in clinical strains, but the carriage of prophages in environmental strains of C. difficile has not yet been explored. Thus, the prevalence and genetic diversity of integrated prophages in the genomes of 166 environmental C. difficile isolates were identified. In addition, the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas systems were determined in the genomes of prophage regions. Predicted prophages and CRISPR-Cas systems were identified by using the PHASTER web server and CRISPRCasFinder, respectively. Phylogenetic relationships among predicated prophages were also constructed based on phage-related genes, terminase large (TerL) subunits and LysM. Among 372 intact prophages, the predominant prophages were phiCDHM1, phiCDHM19, phiMMP01, phiCD506, phiCD27, phiCD211, phiMMP03, and phiC2, followed by phiMMP02, phiCDKM9, phiCD6356, phiCDKM15, and phiCD505. Two newly discovered siphoviruses, phiSM101- and phivB_CpeS-CP51-like Clostridium phages, were identified in two C. difficile genomes. Most prophages were found in sequence types (STs) ST11, ST3, ST8, ST109, and ST2, followed by ST6, ST17, ST4, ST5, ST44, and ST58. An obvious correlation was found between prophage types and STs/ribotypes. Most predicated prophages carry CRISPR arrays. Some prophages carry several gene products, such as accessory gene regulator (Agr), putative spore protease, and abortive infection (Abi) systems. This study shows that prophage carriage, along with genetic diversity and their CRISPR arrays, may play a role in the biology, lifestyle, and fitness of their host strains. Full article
(This article belongs to the Special Issue Bacteriophage—Molecular Studies 5.0)
Show Figures

Figure 1

Back to TopTop