1
|
Suchithra KV, Hameed A, Surya S, Mahammad S, Arun AB. Dual phage-incorporated electrospun polyvinyl alcohol-eudragit nanofiber matrix for rapid healing of diabetic wound infected by Pseudomonas aeruginosa and Staphylococcus aureus. Drug Deliv Transl Res 2025; 15:1092-1108. [PMID: 38980574 DOI: 10.1007/s13346-024-01660-4] [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] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Diabetic wound healing remains a healthcare challenge due to co-occurring multidrug-resistant (MDR) bacterial infections and the constraints associated with sustained drug delivery. Here, we integrate two new species of phages designated as PseuPha1 and RuSa1 respectively lysing multiple clinical MDR strains of P. aeruginosa and S. aureus into a novel polyvinyl alcohol-eudragit (PVA-EU†) nanofiber matrix through electrospinning for rapid diabetic wound healing. PVA-EU† evaluated for characteristic changes that occurred due to electrospinning and subjected to elution, stability and antibacterial assays. The biocompatibility and wound healing ability of PVA-EU† were assessed through mouse fibroblast cell line NIH3T3, followed by validation through diabetic mice excision wound co-infected with P. aeruginosa and S. aureus. The electrospinning resulted in the incorporation of ~ 75% active phages at PVA-EU†, which were stable at 25 °C for 30 days and at 4 °C for 90 days. PVA-EU† showed sustained release of phages for 18 h and confirmed to be detrimental to both mono- and mixed-cultures of target pathogens. The antibacterial activity of PVA-EU† remained unaltered in the presence of high amounts of glucose, whereas alkaline pH promoted the activity. The matrix exerted no cytotoxicity on NIH3T3, but showed significant (p < 0.0001) wound healing in vitro and the process was rapid as validated through a diabetic mice model. The sustained release, quick wound closure, declined abundance of target MDR bacteria in situ and histopathological signs of recovery corroborated the therapeutic efficacy of PVA-EU†. Taken together, our data signify the potential application of PVA-EU† in the rapid treatment of diabetic wounds without the aid of antibiotics.
Collapse
Affiliation(s)
- Kokkarambath Vannadil Suchithra
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Asif Hameed
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India.
| | - Suprith Surya
- Advanced Surgical Skill Enhancement Division (ASSEND), Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Sajida Mahammad
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Ananthapadmanabha Bhagwath Arun
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India.
- Yenepoya Institute of Arts, Science, Commerce and Management, Balmatta, Mangalore-575002, India.
| |
Collapse
|
2
|
Yang P, Li J, Ma X, Hu N, Song Z, Chen B, Li S. Novel delivery systems for phages and lysins in the topical management of wound infections: a narrative review. Front Microbiol 2025; 16:1526096. [PMID: 39931378 PMCID: PMC11808012 DOI: 10.3389/fmicb.2025.1526096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Currently, multidrug-resistant (MDR) bacterial wound infections (WIs) are an extremely challenging clinical problem for physicians. Recently, compared to traditional single liquid delivery drugs, the study of five novel drug delivery systems (i.e., hydrogel, liposomes, electrospun fibers, nanoparticles and nanoemulsion) for phages and their encoded lysins in WI management has become a hot topic. To assess the current landscape of these emerging technologies, we conducted a comprehensive literature search across PubMed, Scopus and Web of Science up to July 2024, using terms such as "phage," "lysin," "wound," "hydrogel," "liposomes," "fibers," "nanoparticles," and "nanoemulsion." The criteria included original studies of five novel delivery systems for phages and lysins in WI management. The findings highlighted the positive effects of the five novel delivery systems for phages and lysins in WI management, significantly reducing wound bacterial populations, and accelerating healing at the injury site. However, the available literature on novel delivery systems for phages and lysins remains limited, particularly for lysins. In conclusion, the application of novel drug delivery systems for phages and lysins showed great potential in combating MDR bacterial WIs.
Collapse
Affiliation(s)
- Pan Yang
- Postdoctoral Research Station, Guangzhou Bay Area Institute of Biomedicine, Guangzhou, China
| | - Jing Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiumei Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Nan Hu
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Bin Chen
- Postdoctoral Research Station, Guangzhou Bay Area Institute of Biomedicine, Guangzhou, China
| | - Shizhu Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Ma S, Han C, Chen X, Li L, Chen X, Zhang W, Wang J, Han F, Yan L, Shi X. Self-healing adhesive hydrogels for sustained ozone release: Enhanced antibacterial properties and improved wound healing. J Control Release 2025; 377:212-222. [PMID: 39522563 DOI: 10.1016/j.jconrel.2024.10.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Antibacterial hydrogels have generated significant interest for their potential therapeutic applications. Ozone (O₃) is recognized for its antibacterial, anti-inflammatory, immunomodulatory, and anti-hypoxic properties, along with its minimal residual impact. However, the development of sustained O₃-release antibacterial hydrogels has been challenging due to the low solubility and short lifespan of ozone. We present an ozone-loaded emulsion hydrogel (ozonegel), which encapsulates ozonized oil within a nanoclay-poly(methacryloxyethyl sulfobetaine) supramolecular network. This adhesive, self-healing ozonegel achieves high ozone loading (91.3 mmol/kg) and releases O₃ and reactive oxygen species (ROS) over 36 h. It demonstrates broad antibacterial and anti-inflammatory effects, promoting wound healing. The remarkable properties of ozonegels suggest significant potential for advanced biomedical applications.
Collapse
Affiliation(s)
- Shanbo Ma
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an 710032, PR China
| | - Conghui Han
- School of Materials Science and Engineering, Chang'an University, Xi'an 710064, PR China
| | - Xi Chen
- School of Materials Science and Engineering, Chang'an University, Xi'an 710064, PR China
| | - Long Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an 710032, PR China
| | - Xushuai Chen
- School of Materials Science and Engineering, Chang'an University, Xi'an 710064, PR China
| | - Wei Zhang
- Military Physical Education Teaching and Research Section of Air Force Medical Service Training Base, Air Force Medical University, Xi'an, China
| | - Jin Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an 710032, PR China
| | - Fu Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| | - Luke Yan
- School of Materials Science and Engineering, Chang'an University, Xi'an 710064, PR China.
| | - Xiaopeng Shi
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an 710032, PR China.
| |
Collapse
|
4
|
Sikora M, Wąsik S, Semaniak J, Drulis-Kawa Z, Wiśniewska-Wrona M, Arabski M. Chitosan-based matrix as a carrier for bacteriophages. Appl Microbiol Biotechnol 2024; 108:6. [PMID: 38165478 PMCID: PMC10761466 DOI: 10.1007/s00253-023-12838-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024]
Abstract
Wound healing is a dynamic and complex process where infection prevention is essential. Chitosan, thanks to its bactericidal activity against gram-positive and gram-negative bacteria, as well as anti-inflammatory and hemostatic properties, is an excellent candidate to design dressings for difficult-to-heal wound treatment. The great advantage of this biopolymer is its capacity to be chemically modified, which allows for the production of various functional forms, depending on the needs and subsequent use. Moreover, chitosan can be an excellent polymer matrix for bacteriophage (phage) packing as a novel alternative/supportive antibacterial therapy approach. This study is focused on the preparation and characteristics of chitosan-based material in the form of a film with the addition of Pseudomonas lytic phages (KTN4, KT28, and LUZ19), which would exhibit antibacterial activity as a potential dressing that accelerates the wound healing. We investigated the method of producing a polymer based on microcrystalline chitosan (MKCh) to serve as the matrix for phage deposition. We described some important parameters such as average molar mass, swelling capacity, surface morphology, phage release profile, and antibacterial activity tested in the Pseudomonas aeruginosa bacterial model. The chitosan polysaccharide turned out to interact with phage particles immobilizing them within a material matrix. Nevertheless, with the high hydrophilicity and swelling features of the prepared material, the external solution of bacterial culture was absorbed and phages went in direct contact with bacteria causing their lysis in the polymer matrix. KEY POINTS: • A novel chitosan-based matrix with the addition of active phages was prepared • Phage interactions with the chitosan matrix were determined as electrostatic • Phages in the matrix work through direct contact with the bacterial cells.
Collapse
Affiliation(s)
- Monika Sikora
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University in Kielce, Kielce, Poland
- Lukasiewicz Research Network-Lodz Institute of Technology, Lodz, Poland
| | - Sławomir Wąsik
- Institute of Physics, Jan Kochanowski University in Kielce, Kielce, Poland
- Central Office of Measures, Warsaw, Poland
| | - Jacek Semaniak
- Institute of Physics, Jan Kochanowski University in Kielce, Kielce, Poland
- Central Office of Measures, Warsaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | | | - Michał Arabski
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University in Kielce, Kielce, Poland.
- Central Office of Measures, Warsaw, Poland.
| |
Collapse
|
5
|
Shiue SJ, Wu MS, Chiang YH, Lin HY. Bacteriophage-cocktail hydrogel dressing to prevent multiple bacterial infections and heal diabetic ulcers in mice. J Biomed Mater Res A 2024; 112:1846-1859. [PMID: 38706446 DOI: 10.1002/jbm.a.37728] [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: 11/13/2023] [Revised: 03/28/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024]
Abstract
Bacteriophage (phage) has been reported to reduce the bacterial infection in delayed-healing wounds and, as a result, aiding in the healing of said wounds. In this study we investigated whether the presence of phage itself could help repair delayed-healing wounds in diabetic mice. Three strains of phage that target Salmonella enterica, Escherichia coli, and Pseudomonas aeruginosa were used. To prevent the phage liquid from running off the wound, the mixture of phage (phage-cocktail) was encapsulated in a porous hydrogel dressing made with three-dimensional printing. The phage-cocktail dressing was tested for its phage preservation and release efficacy, bacterial reduction, cytotoxicity with 3T3 fibroblast, and performance in repairing a sterile full-thickness skin wound in diabetic mice. The phage-cocktail dressing released 1.7%-5.7% of the phages embedded in 24 h, and reduced between 37%-79% of the surface bacteria compared with the blank dressing (p <.05). The phage-cocktail dressing exhibited no sign of cytotoxicity after 3 days (p <.05). In vivo studies showed that 14 days after incision, the full-thickness wound treated with a phage-cocktail dressing had a higher wound healing ratio compared with the blank dressing and control (p <.01). Histological analysis showed that the structure of the skin layers in the group treated with phage-cocktail dressing was restored in an orderly fashion. Compared with the blank dressing and control, the repaired tissue in the phage-cocktail dressing group had new capillary vessels and no sign of inflammation in its dermis, and its epidermis had a higher degree of re-epithelialization (p <.05). The slow-released phage has demonstrated positive effects in repairing diabetic skin wounds.
Collapse
Affiliation(s)
- Sheng-Jie Shiue
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Shun Wu
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hsien Chiang
- Graduate Institute of Chemical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Hsin-Yi Lin
- Graduate Institute of Chemical Engineering, National Taipei University of Technology, Taipei, Taiwan
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei, Taiwan
| |
Collapse
|
6
|
Mohan N, Bosco K, Peter A, Abhitha K, Bhat SG. Bacteriophage entrapment strategies for the treatment of chronic wound infections: a comprehensive review. Arch Microbiol 2024; 206:443. [PMID: 39443305 DOI: 10.1007/s00203-024-04168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
The growing threat of antimicrobial resistance has made the quest for antibiotic alternatives or synergists one of the most pressing priorities of the 21st century. The emergence of multidrug-resistance in most of the common wound pathogens has amplified the risk of antibiotic-resistant wound infections. Bacteriophages, with their self-replicating ability and targeted specificity, can act as suitable antibiotic alternatives. Nevertheless, targeted delivery of phages to infection sites remains a crucial issue, specifically in the case of topical infections. Hence, different phage delivery systems have been studied in recent years. However, there have been no recent reviews of phage delivery systems focusing exclusively on phage application on wounds. This review provides a compendium of all the major delivery systems that have been used to deliver phages to wound infection sites. Special focus has also been awarded to phage-embedded hydrogels with a discussion on the different aspects to be considered during their preparation.
Collapse
Affiliation(s)
- Nivedya Mohan
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
| | - Kiran Bosco
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Center for Infectious Diseases and Microbiology, Westmead, NSW, Australia
| | - Anmiya Peter
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
| | - K Abhitha
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
- Inter University Centre for Nanomaterials and Devices (IUCND), Cochin University of Science and Technology, Kerala, 682022, India
| | - Sarita G Bhat
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India.
- Inter University Centre for Nanomaterials and Devices (IUCND), Cochin University of Science and Technology, Kerala, 682022, India.
| |
Collapse
|
7
|
Gao M, Wang Y, Zhuang H, Zhu Y, Chen N, Teng T. Insights into the Preparation of and Evaluation of the Bactericidal Effects of Phage-Based Hydrogels. Int J Mol Sci 2024; 25:9472. [PMID: 39273419 PMCID: PMC11394800 DOI: 10.3390/ijms25179472] [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: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The rise of antibiotic-resistant strains demands new alternatives in antibacterial treatment. Bacteriophages, with their precise host specificity and ability to target and eliminate bacteria safely, present a valuable option. Meanwhile, hydrogels, known for their excellent biodegradability and biocompatibility, serve as ideal carriers for bacteriophages. The combination of bacteriophages and hydrogels ensures heightened phage activity, concentration, controlled release, and strong antibacterial properties, making it a promising avenue for antibacterial treatment. This article provides a comprehensive review of different crosslinking methods for phage hydrogels, focusing on their application in treating infections caused by various drug-resistant bacteria and highlighting their effective antibacterial properties and controlled release capabilities.
Collapse
Affiliation(s)
- Mengyuan Gao
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yuhan Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Hanyue Zhuang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yanxia Zhu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Na Chen
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Tieshan Teng
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
8
|
Lin YH, Dharmaraj T, Chen Q, Echterhof A, Manasherob R, Zhang LJ, de Leeuw C, Peterson NA, Stannard W, Li Z, Hajfathalian M, Hargil A, Martinez HA, Pourtois J, Chang THW, Blankenberg FG, Amanatullah D, Chaudhuri O, Bollyky PL. Optimized Dosing and Delivery of Bacteriophage Therapy for Wound Infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593005. [PMID: 38766200 PMCID: PMC11100690 DOI: 10.1101/2024.05.07.593005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Lytic bacteriophages, viruses that lyse (kill) bacteria, hold great promise for treating infections, including wound infections caused by antimicrobial-resistant Pseudomonas aeruginosa. However, the optimal dosing and delivery strategies for phage therapy remain unclear. In a mouse wound infection model, we investigated the impact of dose, frequency, and administration route on the efficacy of phage therapy. We find that topical but not intravenous delivery is effective in this model. High-doses of phage reduces bacterial burden more effectively than low-doses, and repeated dosing achieves the highest eradication rates. Building on these insights, we developed "HydroPhage", a hyaluronan-based hydrogel system that uses dynamic covalent crosslinking to deliver high-titre phages over one week. HydroPhage eradicates infections five times more effectively than intravenous injection. We conclude that hydrogel-based sustained phage delivery enhances the efficacy of phage therapy and offers a practical, well-tolerated option for topical application.
Collapse
Affiliation(s)
- Yung-Hao Lin
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Institute of Medical Microbiology, University Hospital of Muenster, Muenster, Germany
| | - Robert Manasherob
- Department of Orthopaedic Surgery, Stanford Hospital and Clinics, Redwood City, CA, USA
| | - Lucy J. Zhang
- Department of Material Science and Engineering, Stanford University, Stanford, CA, USA
| | - Cas de Leeuw
- Institute for Molecules and Materials, Radboud University, Nijmegen, Netherlands
| | - Nana A. Peterson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Whitney Stannard
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Zhiwei Li
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Maryam Hajfathalian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ
| | - Aviv Hargil
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Hunter A. Martinez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Julie Pourtois
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Tony H. W. Chang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Francis G. Blankenberg
- Division of Pediatric Radiology and Nuclear Medicine, Department of Radiology, Lucile Packard Children’s Hospital, Stanford, CA, USA
| | - Derek Amanatullah
- Department of Orthopaedic Surgery, Stanford Hospital and Clinics, Redwood City, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
9
|
Yarali E, Mirzaali MJ, Ghalayaniesfahani A, Accardo A, Diaz-Payno PJ, Zadpoor AA. 4D Printing for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402301. [PMID: 38580291 DOI: 10.1002/adma.202402301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Indexed: 04/07/2024]
Abstract
4D (bio-)printing endows 3D printed (bio-)materials with multiple functionalities and dynamic properties. 4D printed materials have been recently used in biomedical engineering for the design and fabrication of biomedical devices, such as stents, occluders, microneedles, smart 3D-cell engineered microenvironments, drug delivery systems, wound closures, and implantable medical devices. However, the success of 4D printing relies on the rational design of 4D printed objects, the selection of smart materials, and the availability of appropriate types of external (multi-)stimuli. Here, this work first highlights the different types of smart materials, external stimuli, and design strategies used in 4D (bio-)printing. Then, it presents a critical review of the biomedical applications of 4D printing and discusses the future directions of biomedical research in this exciting area, including in vivo tissue regeneration studies, the implementation of multiple materials with reversible shape memory behaviors, the creation of fast shape-transformation responses, the ability to operate at the microscale, untethered activation and control, and the application of (machine learning-based) modeling approaches to predict the structure-property and design-shape transformation relationships of 4D (bio)printed constructs.
Collapse
Affiliation(s)
- Ebrahim Yarali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Mohammad J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Ava Ghalayaniesfahani
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Chemistry, Materials and Chemical Engineering, Giulio Natta, Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, Italy
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Pedro J Diaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, 3015 CN, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| |
Collapse
|
10
|
Vacek L, Polaštík Kleknerová D, Lipový B, Holoubek J, Matysková D, Černá E, Brtníková J, Jeklová E, Kobzová Š, Janda L, Lišková L, Diabelko D, Botka T, Pantůček R, Růžička F, Vojtová L. Phage therapy combined with Gum Karaya injectable hydrogels for treatment of methicillin-resistant Staphylococcus aureus deep wound infection in a porcine model. Int J Pharm 2024; 660:124348. [PMID: 38885776 DOI: 10.1016/j.ijpharm.2024.124348] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Skin and soft tissue infections (SSTIs) represent a significant healthcare challenge, particularly in the context of increasing antibiotic resistance. This study investigates the efficacy of a novel therapeutic approach combining bacteriophage (phage) therapy with a gum Karaya (GK)-based hydrogel delivery system in a porcine model of deep staphylococcal SSTIs. The study exploits the lytic activity and safety of the Staphylococcus phage 812K1/420 of the Kayvirus genus, which is active against methicillin-resistant Staphylococcus aureus (MRSA). The GK injectable hydrogels and hydrogel films, developed by our research group, serve as effective, non-toxic, and easy-to-apply delivery systems, supporting moist wound healing and re-epithelialization. In the porcine model, the combined treatment showed asynergistic effect, leading to a significant reduction in bacterial load (2.5 log CFU/gram of tissue) within one week. Local signs of inflammation were significantly reduced by day 8, with clear evidence of re-epithelialization and wound contraction. Importantly, no adverse effects of the GK-based delivery system were observed throughout the study. The results highlight the potential of this innovative therapeutic approach to effectively treat deep staphylococcal SSTIs, providing a promising avenue for further research and clinical application in the field of infections caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- L Vacek
- Department of Microbiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 602 00 Brno, Czech Republic
| | - D Polaštík Kleknerová
- Department of Microbiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 602 00 Brno, Czech Republic
| | - B Lipový
- Department of Burns and Plastic Surgery, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Advanced Biomaterials Group, Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic
| | - J Holoubek
- Department of Burns and Plastic Surgery, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - D Matysková
- Department of Burns and Plastic Surgery, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - E Černá
- Advanced Biomaterials Group, Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic
| | - J Brtníková
- Advanced Biomaterials Group, Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic
| | - E Jeklová
- Clinical Immunology and Immunology of Infectious Diseases, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - Š Kobzová
- Clinical Immunology and Immunology of Infectious Diseases, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - L Janda
- Clinical Immunology and Immunology of Infectious Diseases, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - L Lišková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - D Diabelko
- Department of Microbiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 602 00 Brno, Czech Republic
| | - T Botka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - R Pantůček
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - F Růžička
- Department of Microbiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 602 00 Brno, Czech Republic.
| | - L Vojtová
- Advanced Biomaterials Group, Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic
| |
Collapse
|
11
|
Xu K, Zhang Q, Zhu D, Jiang Z. Hydrogels in Gene Delivery Techniques for Regenerative Medicine and Tissue Engineering. Macromol Biosci 2024; 24:e2300577. [PMID: 38265144 DOI: 10.1002/mabi.202300577] [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: 12/16/2023] [Revised: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Hydrogels are 3D networks swollen with water. They are biocompatible, strong, and moldable and are emerging as a promising biomedical material for regenerative medicine and tissue engineering to deliver therapeutic genes. The excellent natural extracellular matrix simulation properties of hydrogels enable them to be co-cultured with cells or enhance the expression of viral or non-viral vectors. Its biocompatibility, high strength, and degradation performance also make the action process of carriers in tissues more ideal, making it an ideal biomedical material. It has been shown that hydrogel-based gene delivery technologies have the potential to play therapy-relevant roles in organs such as bone, cartilage, nerve, skin, reproductive organs, and liver in animal experiments and preclinical trials. This paper reviews recent articles on hydrogels in gene delivery and explains the manufacture, applications, developmental timeline, limitations, and future directions of hydrogel-based gene delivery techniques.
Collapse
Affiliation(s)
- Kexing Xu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Qinmeng Zhang
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Danji Zhu
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhiwei Jiang
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
12
|
de Kergariou C, Day GJ, Perriman AW, Armstrong JPK, Scarpa F. Flax fibre reinforced alginate poloxamer hydrogel: assessment of mechanical and 4D printing potential. SOFT MATTER 2024; 20:4021-4034. [PMID: 38695256 PMCID: PMC11095501 DOI: 10.1039/d4sm00135d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
The mechanical and printing performance of a new biomaterial, flax fibre-reinforced alginate-poloxamer based hydrogel, for load-bearing and 4D printing biomedical applications is described in this study. The-self suspendable ability of the material was evaluated by optimising the printing parameters and conducting a collapse test. 1% of the flax fibre weight fraction was sufficient to obtain an optimum hydrogel composite from a mechanical perspective. The collapse test showed that the addition of flax fibres allowed a consistent print without support over longer distances (8 and 10 mm) than the unreinforced hydrogel. The addition of 1% of flax fibres increased the viscosity by 39% and 129% at strain rates of 1 rad s-1 and 5 rad s-1, respectively, compared to the unreinforced hydrogel. The distributions of fibre size and orientation inside the material were also evaluated to identify the internal morphology of the material. The difference of coefficients of moisture expansion between the printing direction (1.29 × 10-1) and the transverse direction (6.03 × 10-1) showed potential for hygromorphic actuation in 4D printing. The actuation authority was demonstrated by printing a [0°; 90°] stacking sequence and rosette-like structures, which were then actuated using humidity gradients. Adding fibres to the hydrogel improved the repeatability of the actuation, while lowering the actuation authority from 0.11 mm-1 to 0.08 mm-1. Overall, this study highlighted the structural and actuation-related benefits of adding flax fibres to hydrogels.
Collapse
Affiliation(s)
- Charles de Kergariou
- Bristol Composites Institute, School of Civil, Aerospace and Design Engineering (CADE), University of Bristol, University Walk, Bristol BS8 1TR, UK.
| | - Graham J Day
- Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, UK
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD Bristol, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD Bristol, UK
- Research School of Chemistry and John Curtin School of Medical Research, Australian National University, Canberra ACT2601, Australia
| | - James P K Armstrong
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, BS1 3NY Bristol, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute, School of Civil, Aerospace and Design Engineering (CADE), University of Bristol, University Walk, Bristol BS8 1TR, UK.
| |
Collapse
|
13
|
Bai H, Borjihan Q, Li Z, Qin P, Cheng J, Xiao D, Dong A. Phage-Based antibacterial hydrogels for bacterial targeting and Ablation: Progress and perspective. Eur J Pharm Biopharm 2024; 198:114258. [PMID: 38479561 DOI: 10.1016/j.ejpb.2024.114258] [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: 12/22/2023] [Revised: 02/04/2024] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
The emergence of drug-resistant bacteria makes antibiotics inadequate to treat bacterial infections, which is now a global problem. Phage as a virus with specific recognition ability can effectively kill the bacteria, which is an efficacious antibacterial material to replace antibiotics. Phage-based hydrogels have good biocompatibility and antibacterial effect at the site of infection. Phage hydrogels have remarkable antibacterial effects on targeted bacteria because of their specific targeted bactericidal ability, but there are few reports and reviews on phage hydrogels. This paper discusses the construction method of phage-based antibacterial hydrogels (PAGs), summarizes the advantages related to PAGs and their applications in the direction of wound healing, treating bone bacterial infections, gastrointestinal infection treatment and other application, and finally gives an outlook on the development and research of PAGs.
Collapse
Affiliation(s)
- Haoran Bai
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, PR China; Engineering Research Center of Dairy Products Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China
| | - Qinggele Borjihan
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, Inner Mongolia, PR China
| | - Zheng Li
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, PR China; Engineering Research Center of Dairy Products Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China
| | - Peiran Qin
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, PR China; Engineering Research Center of Dairy Products Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China
| | - Jingli Cheng
- Institute of Pesticide and Environmental Toxicology, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, Hangzhou 310058, PR China
| | - Douxin Xiao
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, PR China; Engineering Research Center of Dairy Products Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China.
| | - Alideertu Dong
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, PR China; Engineering Research Center of Dairy Products Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China.
| |
Collapse
|
14
|
Kammona O, Tsanaktsidou E, Kiparissides C. Recent Developments in 3D-(Bio)printed Hydrogels as Wound Dressings. Gels 2024; 10:147. [PMID: 38391477 PMCID: PMC10887944 DOI: 10.3390/gels10020147] [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: 01/22/2024] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 02/24/2024] Open
Abstract
Wound healing is a physiological process occurring after the onset of a skin lesion aiming to reconstruct the dermal barrier between the external environment and the body. Depending on the nature and duration of the healing process, wounds are classified as acute (e.g., trauma, surgical wounds) and chronic (e.g., diabetic ulcers) wounds. The latter take several months to heal or do not heal (non-healing chronic wounds), are usually prone to microbial infection and represent an important source of morbidity since they affect millions of people worldwide. Typical wound treatments comprise surgical (e.g., debridement, skin grafts/flaps) and non-surgical (e.g., topical formulations, wound dressings) methods. Modern experimental approaches include among others three dimensional (3D)-(bio)printed wound dressings. The present paper reviews recently developed 3D (bio)printed hydrogels for wound healing applications, especially focusing on the results of their in vitro and in vivo assessment. The advanced hydrogel constructs were printed using different types of bioinks (e.g., natural and/or synthetic polymers and their mixtures with biological materials) and printing methods (e.g., extrusion, digital light processing, coaxial microfluidic bioprinting, etc.) and incorporated various bioactive agents (e.g., growth factors, antibiotics, antibacterial agents, nanoparticles, etc.) and/or cells (e.g., dermal fibroblasts, keratinocytes, mesenchymal stem cells, endothelial cells, etc.).
Collapse
Affiliation(s)
- Olga Kammona
- Chemical Process & Energy Resources Research Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57001 Thessaloniki, Greece
| | - Evgenia Tsanaktsidou
- Chemical Process & Energy Resources Research Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57001 Thessaloniki, Greece
| | - Costas Kiparissides
- Chemical Process & Energy Resources Research Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57001 Thessaloniki, Greece
- Department of Chemical Engineering, Aristotle University of Thessaloniki, P.O. Box 472, 54124 Thessaloniki, Greece
| |
Collapse
|
15
|
Chen C, Xie M, Yan Y, Li Y, Li Z, Zhang T, Gao Z, Deng L, Wang H. Preparation of berberine hydrochloride-Ag nanoparticle composite antibacterial dressing based on 3D printing technology. J Biomater Appl 2024; 38:808-820. [PMID: 38086527 DOI: 10.1177/08853282231222191] [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] [Indexed: 02/06/2024]
Abstract
In recent years, Ag nanoparticle (Ag NP)-loaded antibacterial dressings have attracted much attention in high-level medical dressings. However, the high cytotoxicity of Ag NP has always been a problem. In this paper, we examined the improvement of antibacterial activity of berberine hydrochloride (BBR) with Ag NP, the results showed that the combined use of BBR and Ag NP can effectively reduce the dosage of Ag NP while ensuring the inhibition of bacterial growth, thus an intermediate layer dressing containing combined drugs were prepared. At the same time, the top dressing of polyvinyl alcohol (PVA) solid film and the PVA bottom dressings with three kinds of leakage structures were prepared by 3D printing technology. Three kinds of PVA bottom dressings showed high quality consistency, and the greater the number of leak holes, the higher the porosity value of the dressing, while the swelling ratio value of the bottom layer dressing with three holes was the lowest. Finally, three types of BBR-Ag NP composite antibacterial dressings (3D-BBR-Ag NP) can be obtained by self-assembling of the top dressing, the intermediate layer dressing, and the bottom dressings with three kinds of leakage structures. The cumulative drug release results showed that dressing with more holes had a faster drug release rate compared to the other two ones with fewer leakage holes. Besides, five drug release kinetic models were used to investigate the cumulative BBR release profiles for three types of 3D-BBR-Ag NP. And the three types of composite dressings showed strong antibacterial activity after 6 h of cultivation with staphylococcus aureus. The study showed that the antibacterial activity of the self-assembled dressing prepared by combination of BBR with Ag NP can be improved, and the drug release rate of the hydrogel dressing can be flexibly controlled through 3D printing technology.
Collapse
Affiliation(s)
- Chen Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Maomei Xie
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueling Yan
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongyuan Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhiyao Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tong Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zanyan Gao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | - Haixia Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Lu A, Williams RO, Maniruzzaman M. 3D printing of biologics-what has been accomplished to date? Drug Discov Today 2024; 29:103823. [PMID: 37949427 DOI: 10.1016/j.drudis.2023.103823] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Three-dimensional (3D) printing is a promising approach for the stabilization and delivery of non-living biologics. This versatile tool builds complex structures and customized resolutions, and has significant potential in various industries, especially pharmaceutics and biopharmaceutics. Biologics have become increasingly prevalent in the field of medicine due to their diverse applications and benefits. Stability is the main attribute that must be achieved during the development of biologic formulations. 3D printing could help to stabilize biologics by entrapment, support binding, or crosslinking. Furthermore, gene fragments could be transited into cells during co-printing, when the pores on the membrane are enlarged. This review provides: (i) an introduction to 3D printing technologies and biologics, covering genetic elements, therapeutic proteins, antibodies, and bacteriophages; (ii) an overview of the applications of 3D printing of biologics, including regenerative medicine, gene therapy, and personalized treatments; (iii) information on how 3D printing could help to stabilize and deliver biologics; and (iv) discussion on regulations, challenges, and future directions, including microneedle vaccines, novel 3D printing technologies and artificial-intelligence-facilitated research and product development. Overall, the 3D printing of biologics holds great promise for enhancing human health by providing extended longevity and enhanced quality of life, making it an exciting area in the rapidly evolving field of biomedicine.
Collapse
Affiliation(s)
- Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
17
|
Wang R, Yeh YJ, An YN, Virly. Engineering pH-sensitive erodible chitosan hydrogel composite containing bacteriophage: An interplay between hydrogel and bacteriophage against Staphylococcus aureus. Int J Biol Macromol 2023; 253:127371. [PMID: 37827407 DOI: 10.1016/j.ijbiomac.2023.127371] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Encapsulation of phages represents a key approach for improving phage stability and controlling phage delivery dosage. The hydrogel made from positively charged quaternized chitosan (QCS) and multivalent crosslinker, aldehyde-modified poly(xylitol sebacate)-co-poly(ethylene glycol) (APP) was introduced for the first time for drug (phage 44AHJD) delivery. The freeze-thawing (FT) treatment enhanced the porous structure and the stress resistance of native hydrogel with increased compression stress (stiffness) from 10 to 20 kPa. The stiffness of the phage-loaded hydrogel (FTP) was suitable for the proper release of phage particles and polymer chains, both working synergistically against bacterial growth. The FTP followed the Korsmeyer-Peppas model's anomalous diffusion of phage particles at different temperatures (30-45 °C) and pH (6.6-8.5) conditions. FTP was sensitive to pH, which released more phage particles at pH-neutral conditions, while the release under acidic and alkaline conditions was more based on gel degradation. The high biocompatibility of FTP hydrogel at its working concentration of 30 mg mL-1 was demonstrated through a hemolysis ratio of <2 %. Sixty percent of the total encapsulated phages and 6 mg mL-1 of hydrogel debris were released after 10 h of hydrogel submerge treatment, which can fight the growing bacteria and the emergence of phage-resistant bacteria.
Collapse
Affiliation(s)
- Reuben Wang
- Institute of Food Safety and Health, National Taiwan University, Taipei City, Taiwan; Master of Public Health (MPH) Program, National Taiwan University, Taipei City, Taiwan; GIP-TRIAD Master's Degree in Agro-Biomedical Science, National Taiwan University, Taipei City, Taiwan.
| | - Yu-Jia Yeh
- Institute of Food Safety and Health, National Taiwan University, Taipei City, Taiwan
| | - Yu-Ning An
- Institute of Food Safety and Health, National Taiwan University, Taipei City, Taiwan
| | - Virly
- Global Health Program, College of Public Health, National Taiwan University, Taipei City, Taiwan; Department of Food Technology, Faculty of Agricultural Technology, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| |
Collapse
|
18
|
Mirhaj M, Varshosaz J, Labbaf S, Emadi R, Seifalian AM, Sharifianjazi F, Tavakoli M. Mupirocin loaded core-shell pluronic-pectin-keratin nanofibers improve human keratinocytes behavior, angiogenic activity and wound healing. Int J Biol Macromol 2023; 253:126700. [PMID: 37673152 DOI: 10.1016/j.ijbiomac.2023.126700] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 08/11/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
In the current study, a core-shell nanofibrous wound dressing based on Pluronic-F127 (F127) containing 2 wt% mupirocin (Mup) core and pectin (Pec)-keratin (Kr) shell was fabricated through coaxial electrospinning technique, and the blended nanofibers were also fabricated from the same materials. The fiber diameter and specific surface area of the blended nanofibers were about 101.56 nm and 20.16 m2/g, while for core-shell nanofibers they were about 97.32 nm and 25.26 m2/g, respectively. The resultant blended and core-shell nanofibers experienced a degradation of 27.65 % and 32.28 % during 7 days, respectively. The drug release profile of core-shell nanofibers revealed a sustained release of Mup over 7 days (87.66 %), while the blended F127-Pec-Kr-Mup nanofibers had a burst release within the first few hours (89.38 % up to 48 h) and a cumulative release of 91.36 % after 7 days. Due to the controlled release of Mup, the core-shell structure significantly improved the human keratinocytes behavior, angiogenic potential and wound healing in a rat model compared to the blended structure. In conclusion, the F127-Mup/Pec-Kr core-shell nanofibrous wound dressing appears to be a promising candidate for the prevention of infection, and can potentially accelerate the recovery and healing of chronic and ischemic wounds.
Collapse
Affiliation(s)
- Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Centre, Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Alexander Marcus Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd, Nanoloom Ltd, Liberum Health Ltd), London BioScience Innovation Centre, London, United Kingdom
| | - Fariborz Sharifianjazi
- Department of Natural Sciences, School of Science and Technology, University of Georgia, Tbilisi 0171, Georgia.
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
19
|
Malik DJ, Goncalves-Ribeiro H, GoldSchmitt D, Collin J, Belkhiri A, Fernandes D, Weichert H, Kirpichnikova A. Advanced Manufacturing, Formulation and Microencapsulation of Therapeutic Phages. Clin Infect Dis 2023; 77:S370-S383. [PMID: 37932112 DOI: 10.1093/cid/ciad555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023] Open
Abstract
Manufacturing and formulation of stable, high purity, and high dose bacteriophage drug products (DPs) suitable for clinical usage would benefit from improved process monitoring and control of critical process parameters that affect product quality attributes. Chemistry, Manufacturing, and Controls (CMC) for both upstream (USP) and downstream processes (DSP) need mapping of critical process parameters (CPP) and linking these to critical quality attributes (CQA) to ensure quality and consistency of phage drug substance (DS) and DPs development. Single-use technologies are increasingly becoming the go-to manufacturing option with benefits both for phage bioprocess development at the engineering run research stage and for final manufacture of the phage DS. Future phage DPs under clinical development will benefit from implementation of process analytical technologies (PAT) for better process monitoring and control. These are increasingly being used to improve process robustness (to reduce batch-to-batch variability) and productivity (yielding high phage titers). Precise delivery of stable phage DPs that are suitably formulated as liquids, gels, solid-oral dosage forms, and so forth, could significantly enhance efficacy of phage therapy outcomes. Pre-clinical development of phage DPs must include at an early stage of development, considerations for their formulation including their characterization of physiochemical properties (size, charge, etc.), buffer pH and osmolality, compatibility with regulatory approved excipients, storage stability (packaging, temperature, humidity, etc.), ease of application, patient compliance, ease of manufacturability using scalable manufacturing unit operations, cost, and regulatory requirements.
Collapse
Affiliation(s)
- Danish J Malik
- Chemical Engineering Department, Loughborough University, Loughborough, United Kingdom
| | | | - Dirk GoldSchmitt
- Division of Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
- Department of Psychology, University of Sheffield, Sheffield, United Kingdom
| | - Joe Collin
- Chemical Engineering Department, Loughborough University, Loughborough, United Kingdom
| | - Aouatif Belkhiri
- Chemical Engineering Department, Loughborough University, Loughborough, United Kingdom
| | - Diogo Fernandes
- Nanomaterials Characterisation, Malvern Panalytical, Malvern, United Kingdom
| | - Henry Weichert
- Process Analytical Technology, Sartorius Stedim Biotech GmbH, Germany
| | - Anya Kirpichnikova
- Division of Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| |
Collapse
|
20
|
Ruan H, Aulova A, Ghai V, Pandit S, Lovmar M, Mijakovic I, Kádár R. Polysaccharide-based antibacterial coating technologies. Acta Biomater 2023; 168:42-77. [PMID: 37481193 DOI: 10.1016/j.actbio.2023.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/16/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
To tackle antimicrobial resistance, a global threat identified by the United Nations, is a common cause of healthcare-associated infections (HAI) and is responsible for significant costs on healthcare systems, a substantial amount of research has been devoted to developing polysaccharide-based strategies that prevent bacterial attachment and biofilm formation on surfaces. Polysaccharides are essential building blocks for life and an abundant renewable resource that have attracted much attention due to their intrinsic remarkable biological potential antibacterial activities. If converted into efficient antibacterial coatings that could be applied to a broad range of surfaces and applications, polysaccharide-based coatings could have a significant potential global impact. However, the ultimate success of polysaccharide-based antibacterial materials will be determined by their potential for use in manufacturing processes that are scalable, versatile, and affordable. Therefore, in this review we focus on recent advances in polysaccharide-based antibacterial coatings from the perspective of fabrication methods. We first provide an overview of strategies for designing polysaccharide-based antimicrobial formulations and methods to assess the antibacterial properties of coatings. Recent advances on manufacturing polysaccharide-based coatings using some of the most common polysaccharides and fabrication methods are then detailed, followed by a critical comparative overview of associated challenges and opportunities for future developments. STATEMENT OF SIGNIFICANCE: Our review presents a timely perspective by being the first review in the field to focus on advances on polysaccharide-based antibacterial coatings from the perspective of fabrication methods along with an overview of strategies for designing polysaccharide-based antimicrobial formulations, methods to assess the antibacterial properties of coatings as well as a critical comparative overview of associated challenges and opportunities for future developments. Meanwhile this work is specifically targeted at an audience focused on featuring critical information and guidelines for developing polysaccharide-based coatings. Including such a complementary work in the journal could lead to further developments on polysaccharide antibacterial applications.
Collapse
Affiliation(s)
- Hengzhi Ruan
- Department of Industrial and Materials Science, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Alexandra Aulova
- Department of Industrial and Materials Science, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Viney Ghai
- Department of Industrial and Materials Science, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Santosh Pandit
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Martin Lovmar
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; Wellspect Healthcare AB, 431 21 Mölndal, Sweden
| | - Ivan Mijakovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Roland Kádár
- Department of Industrial and Materials Science, Chalmers University of Technology, 412 96 Göteborg, Sweden; Wallenberg Wood Science Centre (WWSC), Chalmers University of Technology, 412 96 Göteborg, Sweden.
| |
Collapse
|
21
|
Choi V, Rohn JL, Stoodley P, Carugo D, Stride E. Drug delivery strategies for antibiofilm therapy. Nat Rev Microbiol 2023; 21:555-572. [PMID: 37258686 DOI: 10.1038/s41579-023-00905-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Although new antibiofilm agents have been developed to prevent and eliminate pathogenic biofilms, their widespread clinical use is hindered by poor biocompatibility and bioavailability, unspecific interactions and insufficient local concentrations. The development of innovative drug delivery strategies can facilitate penetration of antimicrobials through biofilms, promote drug dispersal and synergistic bactericidal effects, and provide novel paradigms for clinical application. In this Review, we discuss the potential benefits of such emerging techniques for improving the clinical efficacy of antibiofilm agents, as well as highlighting the existing limitations and future prospects for these therapies in the clinic.
Collapse
Affiliation(s)
- Victor Choi
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Centre for Urological Biology, Division of Medicine, University College London, London, UK
| | - Paul Stoodley
- Departments of Microbial Infection and Immunity, Microbiology and Orthopaedics, The Ohio State University, Columbus, OH, USA
- Department of Mechanical Engineering, National Centre for Advanced Tribology at Southampton (nCATS) and National Biofilm Innovation Centre (NBIC), University of Southampton, Southampton, UK
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Eleanor Stride
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
22
|
Wang Z, Liang X, Wang G, Wang X, Chen Y. Emerging Bioprinting for Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2304738. [PMID: 37566537 DOI: 10.1002/adma.202304738] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/05/2023] [Indexed: 08/13/2023]
Abstract
Bioprinting has attracted much attention due to its suitability for fabricating biomedical devices. In particular, bioprinting has become one of the growing centers in the field of wound healing, with various types of bioprinted devices being developed, including 3D scaffolds, microneedle patches, and flexible electronics. Bioprinted devices can be designed with specific biostructures and biofunctions that closely match the shape of wound sites and accelerate the regeneration of skin through various approaches. Herein, a comprehensive review of the bioprinting of smart wound dressings is presented, emphasizing the crucial effect of bioprinting in determining biostructures and biofunctions. The review begins with an overview of bioprinting techniques and bioprinted devices, followed with an in-depth discussion of polymer-based inks, modification strategies, additive ingredients, properties, and applications. The strategies for the modification of bioprinted devices are divided into seven categories, including chemical synthesis of novel inks, physical blending, coaxial bioprinting, multimaterial bioprinting, physical absorption, chemical immobilization, and hybridization with living cells, and examples are presented. Thereafter, the frontiers of bioprinting and wound healing, including 4D bioprinting, artificial intelligence-assisted bioprinting, and in situ bioprinting, are discussed from a perspective of interdisciplinary sciences. Finally, the current challenges and future prospects in this field are highlighted.
Collapse
Affiliation(s)
- Zijian Wang
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
- Department of Urology, Hubei Province Key Laboratory of Urinary System Diseases, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiao Liang
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Guanyi Wang
- Department of Urology, Hubei Province Key Laboratory of Urinary System Diseases, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinghuan Wang
- Department of Urology, Hubei Province Key Laboratory of Urinary System Diseases, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yun Chen
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| |
Collapse
|
23
|
Mutalik C, Saukani M, Khafid M, Krisnawati DI, Darmayanti R, Puspitasari B, Cheng TM, Kuo TR. Gold-Based Nanostructures for Antibacterial Application. Int J Mol Sci 2023; 24:10006. [PMID: 37373154 DOI: 10.3390/ijms241210006] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Bacterial infections have become a fatal threat because of the abuse of antibiotics in the world. Various gold (Au)-based nanostructures have been extensively explored as antibacterial agents to combat bacterial infections based on their remarkable chemical and physical characteristics. Many Au-based nanostructures have been designed and their antibacterial activities and mechanisms have been further examined and demonstrated. In this review, we collected and summarized current developments of antibacterial agents of Au-based nanostructures, including Au nanoparticles (AuNPs), Au nanoclusters (AuNCs), Au nanorods (AuNRs), Au nanobipyramids (AuNBPs), and Au nanostars (AuNSs) according to their shapes, sizes, and surface modifications. The rational designs and antibacterial mechanisms of these Au-based nanostructures are further discussed. With the developments of Au-based nanostructures as novel antibacterial agents, we also provide perspectives, challenges, and opportunities for future practical clinical applications.
Collapse
Affiliation(s)
- Chinmaya Mutalik
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Muhammad Saukani
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Islam Kalimantan MAB, Banjarmasin 70124, Kalimantan Selatan, Indonesia
| | - Muhamad Khafid
- Department of Nursing, Faculty of Nursing and Midwifery, Universitas Nahdlatul Ulama Surabaya, Surabaya 60237, East Java, Indonesia
| | | | - Rofik Darmayanti
- Dharma Husada Nursing Academy, Kediri 64117, East Java, Indonesia
| | | | - Tsai-Mu Cheng
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-Rong Kuo
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Yu Q, Wang Q, Zhang L, Deng W, Cao X, Wang Z, Sun X, Yu J, Xu X. The applications of 3D printing in wound healing: the external delivery of stem cells and antibiosis. Adv Drug Deliv Rev 2023; 197:114823. [PMID: 37068658 DOI: 10.1016/j.addr.2023.114823] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
As the global number of chronic wound patients rises, the financial burden and social pressure on patients increase daily. Stem cells have emerged as promising tissue engineering seed cells due to their enriched sources, multidirectional differentiation ability, and high proliferation rate. However, delivering them in vitro for the treatment of skin injury is still challenging. In addition, bacteria from the wound site and the environment can significantly impact wound healing. In the last decade, 3D bioprinting has dramatically enriched cell delivery systems. The produced scaffolds by this technique can be precisely localized within cells and perform antibacterial actions. In this review, we summarized the 3D bioprinting-based external delivery of stem cells and their antibiosis to improve wound healing.
Collapse
Affiliation(s)
- Qingtong Yu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Qilong Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Linzhi Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Xia Cao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Zhe Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
25
|
Li B, Wang W, Zhao L, Yan D, Li X, Gao Q, Zheng J, Zhou S, Lai S, Feng Y, Zhang J, Jiang H, Long C, Gan W, Chen X, Wang D, Tang BZ, Liao Y. Multifunctional AIE Nanosphere-Based "Nanobomb" for Trimodal Imaging-Guided Photothermal/Photodynamic/Pharmacological Therapy of Drug-Resistant Bacterial Infections. ACS NANO 2023; 17:4601-4618. [PMID: 36826229 DOI: 10.1021/acsnano.2c10694] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Injudicious or inappropriate use of antibiotics has led to the prevalence of drug-resistant bacteria, posing a huge menace to global health. Here, a self-assembled aggregation-induced emission (AIE) nanosphere (AIE-PEG1000 NPs) that simultaneously possesses near-infrared region II (NIR-II) fluorescence emissive, photothermal, and photodynamic properties is prepared using a multifunctional AIE luminogen (AIE-4COOH). The AIE-PEG1000 NPs were encapsulated with teicoplanin (Tei) and ammonium bicarbonate (AB) into lipid nanovesicles to form a laser-activated "nanobomb" (AIE-Tei@AB NVs) for the multimodal theranostics of drug-resistant bacterial infections. In vivo experiments validate that the "nanobomb" enables high-performance NIR-II fluorescence, infrared thermal, and ultrasound (AB decomposition during the photothermal process to produce numerous CO2/NH3 bubbles, which is an efficient ultrasound contrast agent) imaging of multidrug-resistant bacteria-infected foci after intravenous administration of AIE-Tei@AB NVs followed by 660 nm laser stimulation. The highly efficient photothermal and photodynamic features of AIE-Tei@AB NVs, combined with the excellent pharmacological property of rapidly released Tei during bubble generation and NV disintegration, collectively promote broad-spectrum eradication of three clinically isolated multidrug-resistant bacteria strains and rapid healing of infected wounds. This multimodal imaging-guided synergistic therapeutic strategy can be extended for the theranostics of superbugs.
Collapse
Affiliation(s)
- Bin Li
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Wei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Lu Zhao
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Dingyuan Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Xiaoxue Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Qiuxia Gao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Sitong Zhou
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Shanshan Lai
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Yi Feng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Jie Zhang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Hang Jiang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
| | - Chengmin Long
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Wenjun Gan
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Xiaodong Chen
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Ben Zhong Tang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Yuhui Liao
- Department of Burn Surgery & Department of Clinical Laboratory, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, Guangdong, China
- Center for Infection and Immunity, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| |
Collapse
|
26
|
Jaberifard F, Ramezani S, Ghorbani M, Arsalani N, Mortazavi Moghadam F. Investigation of wound healing efficiency of multifunctional eudragit/soy protein isolate electrospun nanofiber incorporated with ZnO loaded halloysite nanotubes and allantoin. Int J Pharm 2022; 630:122434. [PMID: 36435502 DOI: 10.1016/j.ijpharm.2022.122434] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022]
Abstract
One significant aspect of the current therapeutic agents employed in wound healing involves the engineering of nano polymeric scaffolds to mimic the properties of extracellular matrix (ECM). The present work aimed to prepare and evaluate Eudragit® L100 (EU) nanofibers in combination with soy protein isolate (SPI). Allantoin (Ala) with a 2 wt% was encapsulated as a model drug renowned for its anti-inflammatory and antioxidant agents. Moreover, the synthesized ZnO-halloysite nanotubes (ZHNTs) with different concentrations of 1, 3, and 5 wt% were incorporated into the EU/SPI/Ala nanofiber as a reinforcing filler and a remarkable antibacterial agent. The scanning electron microscope (SEM) analysis showed that by increasing the weight percentage of SPI from 1 % to 2.5 %, the average diameter of nanofibers decreased from 132.3 ± 51.3 nm to 126.7 ± 47.2 nm. It was 223.5 ± 95.6 nm for nanofibers containing 5 wt% ZHNTs (the optimal sample). The evaluation of in vitro release kinetics of Ala for 24 h, showed a burst release during the first 2 h and a sustained release during the subsequent times. Moreover, the structure, crystallinity, and thermal stability of synthesized nanofibers were characterized by Fourier Transform Infrared Spectrometry (FTIR), X-ray diffraction (XRD), and Thermo gravimetric analysis (TGA), respectively. In vitro degradation and mechanical characteristics of these nanofibers were studied. Furthermore, the capability of the nanofibers for cell proliferation was revealed through the MTT test and field emission scanning electron microscopy (FESEM) images of cell attachment. The antimicrobial activity of EU/SPI/Ala/ZHNTs showed that this sample with high ZHNTs content (5 w%t) had the most remarkable antibacterial activity against S. aureus. The results revealed that EU/SPI/Ala/ZHNTs mats could be promising potential wound dressings.
Collapse
Affiliation(s)
- Farnaz Jaberifard
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran; Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soghra Ramezani
- Nanofiber Research Center, Asian Nanostructures Technology Co. (ANSTCO), Zanjan, Iran
| | - Marjan Ghorbani
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Nasser Arsalani
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Fatemeh Mortazavi Moghadam
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, USA
| |
Collapse
|
27
|
Zhou L, Min T, Bian X, Dong Y, Zhang P, Wen Y. Rational Design of Intelligent and Multifunctional Dressing to Promote Acute/Chronic Wound Healing. ACS APPLIED BIO MATERIALS 2022; 5:4055-4085. [PMID: 35980356 DOI: 10.1021/acsabm.2c00500] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Currently, the clinic's treatment of acute/chronic wounds is still unsatisfactory due to the lack of functional and appropriate wound dressings. Intelligent and multifunctional dressings are considered the most advanced wound treatment modalities. It is essential to design and develop wound dressings with required functions according to the wound microenvironment in the clinical treatment. This work summarizes microenvironment characteristics of various common wounds, such as acute wound, diabetic wound, burns wound, scalded wound, mucosal wound, and ulcers wound. Furthermore, the factors of transformation from acute wounds to chronic wounds were analyzed. Then we focused on summarizing how researchers fully and thoroughly combined the complex microenvironment with modern advanced technology to ensure the usability and value of the dressing, such as photothermal-sensitive dressings, microenvironment dressing (pH-sensitive dressings, ROS-sensitive dressings, and osmotic pressure dressings), hemostatic dressing, guiding tissue regeneration dressing, microneedle dressings, and 3D/4D printing dressings. Finally, the revolutionary development of wound dressings and how to transform the existing advanced functional dressings into clinical needs as soon as possible have carried out a reasonable and meaningful outlook.
Collapse
Affiliation(s)
- Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
- Department of Orthopaedics and Trauma, Key Laboratory of Trauma and Neural Regeneration, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Tiantian Min
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | | | - Peixun Zhang
- Department of Orthopaedics and Trauma, Key Laboratory of Trauma and Neural Regeneration, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
28
|
|
29
|
Liu ZH, Chiang MT, Lin HY. Lytic Bacteriophage as a Biomaterial to Prevent Biofilm Formation and Promote Neural Growth. Tissue Eng Regen Med 2022; 19:987-1000. [PMID: 35648339 DOI: 10.1007/s13770-022-00462-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Although non-lytic filamentous bacteriophages have been made into biomaterial to guide tissue growth, they had limited ability to prevent bacterial infection. In this work a lytic bacteriophage was used to make an antibacterial biomaterial for neural tissue repair. METHODS Lytic phages were chemically bound to the surface of a chitosan film through glutaraldehyde crosslinking. After the chemical reaction, the contact angle of the sample surface and the remaining lytic potential of the phages were measured. The numbers of bacteria on the samples were measured and examined under scanning electron microscopy. Transmission electron microscopy (TEM) was used to observe the phages and phage-infected bacteria. A neuroblast cell line was cultured on the samples to evaluate the sample's biocompatibility. RESULTS The phages conjugated to the chitosan film preserved their lytic potential and reduced 68% of bacterial growth on the sample surface at 120 min (p < 0.001). The phage-linked surface had a significantly higher contact angle than that of the control chitosan (p < 0.05). After 120 min a bacterial biofilm appeared on the control chitosan, while the phage-linked sample effectively prevented biofilm formation. The TEM images demonstrated that the phage attached and lysed the bacteria on the phage-linked sample at 120 min. The phage-linked sample significantly promoted the neuroblast cell attachment (p < 0.05) and proliferation (p < 0.01). The neuroblast on the phage-linked sample demonstrated more cell extensions after day 1. CONCLUSION The purified lytic phages were proven to be a highly bioactive nanomaterial. The phage-chitosan composite material not only promoted neural cell proliferation but also effectively prevent bacterial growth, a major cause of implant failure and removal.
Collapse
Affiliation(s)
- Zi-Hao Liu
- Graduate Institute of Chemical Engineering, National Taipei University of Technology, 3, Zhongxiao E Rd, Taipei, 106, Taiwan
| | - Ming-Tse Chiang
- Graduate Institute of Chemical Engineering, National Taipei University of Technology, 3, Zhongxiao E Rd, Taipei, 106, Taiwan
| | - Hsin-Yi Lin
- Graduate Institute of Chemical Engineering, National Taipei University of Technology, 3, Zhongxiao E Rd, Taipei, 106, Taiwan.
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, 3, Zhongxiao E Rd, Taipei, 106, Taiwan.
| |
Collapse
|
30
|
Briot T, Kolenda C, Ferry T, Medina M, Laurent F, Leboucher G, Pirot F. Paving the way for phage therapy using novel drug delivery approaches. J Control Release 2022; 347:414-424. [PMID: 35569589 DOI: 10.1016/j.jconrel.2022.05.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 12/24/2022]
Abstract
Bacterial resistance against antibiotics is an emergent medical issue. The development of novel therapeutic approaches is urgently needed and, in this context, bacteriophages represent a promising strategy to fight multi resistant bacteria. However, for some applications, bacteriophages cannot be used without an appropriate drug delivery system which increases their stability or provides an adequate targeting to the site of infection. This review summarizes the main application routes for bacteriophages and presents the new delivery approaches designed to increase phage's activity. Clinical successes of these formulations are also highlighted. Globally, this work paves the way for the design and optimization of nano and micro delivery systems for phage therapy.
Collapse
Affiliation(s)
- Thomas Briot
- Pharmacy department, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France.
| | - Camille Kolenda
- Laboratory of bacteriology, French National Reference Centre for Staphylococci, Hospices Civils de Lyon, Lyon, France; Reference Center for Complex Bone and Joint Infection (CRIOAc), Hospices Civils de Lyon, Lyon, France; International Centre for Research in Infectiology, INSERM U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Tristan Ferry
- Reference Center for Complex Bone and Joint Infection (CRIOAc), Hospices Civils de Lyon, Lyon, France; International Centre for Research in Infectiology, INSERM U1111, Université Claude Bernard Lyon 1, Lyon, France; Infectious and Tropical Diseases unit, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| | - Mathieu Medina
- Laboratory of bacteriology, French National Reference Centre for Staphylococci, Hospices Civils de Lyon, Lyon, France; Reference Center for Complex Bone and Joint Infection (CRIOAc), Hospices Civils de Lyon, Lyon, France; International Centre for Research in Infectiology, INSERM U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Frederic Laurent
- Laboratory of bacteriology, French National Reference Centre for Staphylococci, Hospices Civils de Lyon, Lyon, France; Reference Center for Complex Bone and Joint Infection (CRIOAc), Hospices Civils de Lyon, Lyon, France; International Centre for Research in Infectiology, INSERM U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Gilles Leboucher
- Pharmacy department, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France
| | - Fabrice Pirot
- Plateforme FRIPHARM, Service pharmaceutique, Groupement Hospitalier Edouard Herriot, Hospices Civils de Lyon, Lyon, France; Laboratoire de Recherche et Développement de Pharmacie Galénique Industrielle, Plateforme FRIPHARM, Faculté de Pharmacie, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique - UMR 5305, Université Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
31
|
Chen Y, Hao Y, Mensah A, Lv P, Wei Q. Bio-inspired hydrogels with fibrous structure: A review on design and biomedical applications. BIOMATERIALS ADVANCES 2022; 136:212799. [PMID: 35929334 DOI: 10.1016/j.bioadv.2022.212799] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Numerous tissues in the human body have fibrous structures, including the extracellular matrix, muscles, and heart, which perform critical biological functions and have exceptional mechanical strength. Due to their high-water content, softness, biocompatibility and elastic nature, hydrogels resemble biological tissues. Traditional hydrogels, on the other hand, have weak mechanical properties and lack tissue-like fibrous structures, limiting their potential applications. Thus, bio-inspired hydrogels with fibrous architectures have piqued the curiosity of biomedical researchers. Here, we review fabrication strategies for fibrous hydrogels and their recent progress in the biomedical fields of wound dressings, drug delivery, tissue engineering scaffolds and bioadhesives. Challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Yajun Chen
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Yi Hao
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Alfred Mensah
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Pengfei Lv
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Qufu Wei
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China.
| |
Collapse
|
32
|
Shiue SJ, Syu FS, Lin HY. Two types of bacteriophage-modified alginate hydrogels as antibacterial coatings for implants. J Taiwan Inst Chem Eng 2022. [DOI: 10.1016/j.jtice.2022.104353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Zyman A, Górski A, Międzybrodzki R. Phage therapy of wound-associated infections. Folia Microbiol (Praha) 2022; 67:193-201. [PMID: 35028881 PMCID: PMC8933295 DOI: 10.1007/s12223-021-00946-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/28/2021] [Indexed: 11/26/2022]
Abstract
Phages are viruses which can specifically infect bacteria, resulting in their destruction. Bacterial infections are a common complication of wound healing, and experimental evidence from animal models demonstrates promising potential for phage-dependent eradication of wound-associated infections. The studies discussed suggest that phage therapy may be an effective treatment, with important advantages over some current antibacterial treatments. Phage cocktails, as well as co-administration of phages and antibiotics, have been reported to minimise bacterial resistance. Further, phage-antibiotic synergism has been reported in some studies. The ideal dose of phages is still subject to debate, with evidence for both high and low doses to yield therapeutic effects. Novel delivery methods, such as hydrogels, are being explored for their advantages in topical wound healing. There are more and more Good Manufacturing Practice facilities dedicated to manufacturing phage products and phage therapy units across the world, showing the changing perception of phages which is occurring. However, further research is needed to secure the place of phages in modern medicine, with some scientists calling upon the World Health Organisation to help promote phage therapy.
Collapse
Affiliation(s)
- Anna Zyman
- Pharmacology Undergraduate Programme, School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, University Walk, Bristol, BS8 1TD UK
| | - Andrzej Górski
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- 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
| | - Ryszard Międzybrodzki
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, 02-006 Warsaw, Poland
| |
Collapse
|
34
|
Zamboulis A, Michailidou G, Koumentakou I, Bikiaris DN. Polysaccharide 3D Printing for Drug Delivery Applications. Pharmaceutics 2022; 14:145. [PMID: 35057041 PMCID: PMC8778081 DOI: 10.3390/pharmaceutics14010145] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/19/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
3D printing, or additive manufacturing, has gained considerable interest due to its versatility regarding design as well as in the large choice of materials. It is a powerful tool in the field of personalized pharmaceutical treatment, particularly crucial for pediatric and geriatric patients. Polysaccharides are abundant and inexpensive natural polymers, that are already widely used in the food industry and as excipients in pharmaceutical and cosmetic formulations. Due to their intrinsic properties, such as biocompatibility, biodegradability, non-immunogenicity, etc., polysaccharides are largely investigated as matrices for drug delivery. Although an increasing number of interesting reviews on additive manufacturing and drug delivery are being published, there is a gap concerning the printing of polysaccharides. In this article, we will review recent advances in the 3D printing of polysaccharides focused on drug delivery applications. Among the large family of polysaccharides, the present review will particularly focus on cellulose and cellulose derivatives, chitosan and sodium alginate, printed by fused deposition modeling and extrusion-based printing.
Collapse
Affiliation(s)
- Alexandra Zamboulis
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (G.M.); (I.K.)
| | | | | | - Dimitrios N. Bikiaris
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (G.M.); (I.K.)
| |
Collapse
|
35
|
de Oliveira RS, Fantaus SS, Guillot AJ, Melero A, Beck RCR. 3D-Printed Products for Topical Skin Applications: From Personalized Dressings to Drug Delivery. Pharmaceutics 2021; 13:1946. [PMID: 34834360 PMCID: PMC8625283 DOI: 10.3390/pharmaceutics13111946] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/13/2021] [Accepted: 11/14/2021] [Indexed: 01/05/2023] Open
Abstract
3D printing has been widely used for the personalization of therapies and on-demand production of complex pharmaceutical forms. Recently, 3D printing has been explored as a tool for the development of topical dosage forms and wound dressings. Thus, this review aims to present advances related to the use of 3D printing for the development of pharmaceutical and biomedical products for topical skin applications, covering plain dressing and products for the delivery of active ingredients to the skin. Based on the data acquired, the important growth in the number of publications over the last years confirms its interest. The semisolid extrusion technique has been the most reported one, probably because it allows the use of a broad range of polymers, creating the most diverse therapeutic approaches. 3D printing has been an excellent field for customizing dressings, according to individual needs. Studies discussed here imply the use of metals, nanoparticles, drugs, natural compounds and proteins and peptides for the treatment of wound healing, acne, pain relief, and anti-wrinkle, among others. The confluence of 3D printing and topical applications has undeniable advantages, and we would like to encourage the research groups to explore this field to improve the patient's life quality, adherence and treatment efficacy.
Collapse
Affiliation(s)
- Rafaela Santos de Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre 90610-000, Brazil;
| | - Stephani Silva Fantaus
- Departamento de Produção e Controle de Medicamentos, Universidade Federal do Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre 90610-000, Brazil;
| | - Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, School of Pharmacy, University of Valencia, Avenida Vicente Andres Estelles SN, 46100 Burjassot, Spain;
| | - Ana Melero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, School of Pharmacy, University of Valencia, Avenida Vicente Andres Estelles SN, 46100 Burjassot, Spain;
| | - Ruy Carlos Ruver Beck
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre 90610-000, Brazil;
- Departamento de Produção e Controle de Medicamentos, Universidade Federal do Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre 90610-000, Brazil;
| |
Collapse
|
36
|
Kudzin MH, Giełdowska M, Mrozińska Z, Boguń M. Poly(lactic acid)/Zinc/Alginate Complex Material: Preparation and Antimicrobial Properties. Antibiotics (Basel) 2021; 10:1327. [PMID: 34827265 PMCID: PMC8614701 DOI: 10.3390/antibiotics10111327] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to investigate an antimicrobial and degradable composite material consisting of melt-blown poly(lactic acid) nonwoven fabrics, alginate, and zinc. This paper describes the method of preparation and the characterization of the physicochemical and antimicrobial properties of the new fibrous composite material. The procedure consists of fabrication of nonwoven fabric and two steps of dip-coating modification: (1) impregnation of nonwoven samples in the solution of alginic sodium salt and (2) immersion in a solution of zinc (II) chloride. The characterization and analysis of new material included scanning electron microscopy (SEM), specific surface area (SSA), and total/average pore volume (BET). The polylactide/alginate/Zn fibrous composite were subjected to microbial activity tests against colonies of Gram-positive (Staphylococcus aureus), Gram-negative (Escherichia coli) bacterial strains, and the following fungal strains: Aspergillus niger van Tieghem and Chaetomium globosum. These results lay a technical foundation for the development and potential application of new composite as an antibacterial/antifungal material in biomedical areas.
Collapse
Affiliation(s)
- Marcin H. Kudzin
- Lukasiewicz Research Network-Textile Research Institute, Brzezinska 5/15, 92-103 Lodz, Poland; (M.G.); (Z.M.); (M.B.)
| | | | | | | |
Collapse
|
37
|
Li X, Yang X, Wang Z, Liu Y, Guo J, Zhu Y, Shao J, Li J, Wang L, Wang K. Antibacterial, antioxidant and biocompatible nanosized quercetin-PVA xerogel films for wound dressing. Colloids Surf B Biointerfaces 2021; 209:112175. [PMID: 34740095 DOI: 10.1016/j.colsurfb.2021.112175] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/09/2023]
Abstract
Topical use of antimicrobial agents to treat wounds to inhibit bacterial invasion and facilitate wound healing is an effective strategy. In this work, an antibacterial xerogel film for potential applications in wound dressings was developed. First, a natural antibacterial agent, quercetin (Qu), was made into water-soluble quercetin-borate (QuB) nanoparticles by merging a solvent exchange method with the borate esterification reaction. QuB nanoparticles were then employed as the cross-linking agent to achieve gelation of poly(vinyl alcohol) (PVA) to obtain antimicrobial QuB-PVA composite microgels. Furthermore, QuB-PVA microgels were utilized as raw materials to produce xerogel films via an electrospray technique. The as-prepared QuB-PVA xerogel films exhibited excellent bacteriostasis, antioxidation, biocompatibility, self-healing, accelerated skin regeneration and functional restoration, and promoted skin wound healing. The QuB-PVA films significantly facilitated the in vivo healing speed of full-thickness skin wounds compared to commercial dressings. We believe that the present multifunctional QuB-PVA xerogel film is an excellent candidate for the wound dressings.
Collapse
Affiliation(s)
- Xiaozhou Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xuxuan Yang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zicheng Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanxiang Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiaxiang Guo
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yu Zhu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiaxing Shao
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiage Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lin Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Ke Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
38
|
Formulation strategies for bacteriophages to target intracellular bacterial pathogens. Adv Drug Deliv Rev 2021; 176:113864. [PMID: 34271022 DOI: 10.1016/j.addr.2021.113864] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
Bacteriophages (Phages) are antibacterial viruses that are unaffected by antibiotic drug resistance. Many Phase I and Phase II phage therapy clinical trials have shown acceptable safety profiles. However, none of the completed trials could yield data supporting the promising observations noted in the experimental phage therapy. These trials have mainly focused on phage suspensions without enough attention paid to the stability of phage during processing, storage, and administration. This is important because in vivo studies have shown that the effectiveness of phage therapy greatly depends on the ratio of phage to bacterial concentrations (multiplicity of infection) at the infection site. Additionally, bacteria can evade phages through the development of phage-resistance and intracellular residence. This review focuses on the use of phage therapy against bacteria that survive within the intracellular niches. Recent research on phage behavior reveals that some phage can directly interact with, get internalized into, and get transcytosed across mammalian cells, prompting further research on the governing mechanisms of these interactions and the feasibility of harnessing therapeutic phage to target intracellular bacteria. Advances to improve the capability of phage attacking intracellular bacteria using formulation approaches such as encapsulating/conjugating phages into/with vector carriers via liposomes, polymeric particles, inorganic nanoparticles, and cell penetrating peptides, are summarized. While promising progress has been achieved, research in this area is still in its infancy and warrants further attention.
Collapse
|