1
|
Du J, Zhou T, Peng W. Functional polysaccharide-based hydrogel in bone regeneration: From fundamentals to advanced applications. Carbohydr Polym 2025; 352:123138. [PMID: 39843049 DOI: 10.1016/j.carbpol.2024.123138] [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: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
Bone regeneration is limited and generally requires external intervention to promote effective repair. Autografts, allografts, and xenografts as traditional methods for addressing bone defects have been widely utilized, their clinical applicability is limited due to their respective disadvantages. Fortunately, functional polysaccharide hydrogels have gained significant attention in bone regeneration due to their exceptional drug-loading capacity, biocompatibility, and ease of chemical modification. They also provide an optimal microenvironment for bone repair and regeneration. This review provides an overview of various functional polysaccharide hydrogels derived from biocompatible materials, focusing on their applications in intelligent delivery systems, bone tissue regeneration, and cartilage defect repair. Particularly, the incorporation of bioactive molecules into the design of functional polysaccharide hydrogels has been shown to significantly enhance bone regeneration. Additionally, this review emphasizes the preparation methods for functional polysaccharide hydrogels and associated the bone healing mechanisms. Finally, the limitations and future prospects of functional polysaccharide hydrogels are thoroughly evaluated.
Collapse
Affiliation(s)
- Jian Du
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100048, China; Hebei North University, Zhangjiakou, 075000, China
| | - Tian Zhou
- Hebei North University, Zhangjiakou, 075000, China
| | - Wei Peng
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100048, China.
| |
Collapse
|
2
|
Liu L, Jing R, Yao L, Wang Y, Mu L, Hu Y. Hemostasis Strategies and Recent Advances in Hydrogels for Managing Uncontrolled Hemorrhage. ACS APPLIED BIO MATERIALS 2025; 8:42-61. [PMID: 39745272 DOI: 10.1021/acsabm.4c01221] [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: 01/21/2025]
Abstract
Hemorrhage continues to pose a significant challenge in various medical contexts, underscoring the need for advanced hemostatic materials. Hemostatic hydrogels have gained recognition as innovative tools for addressing uncontrollable bleeding, attributed to their distinctive features including biological compatibility, tunable mechanical properties, and exceptional hemostatic performance. This review provides a comprehensive overview of hemostatic hydrogels that offer rapid and effective bleeding control. Particularly, this review focuses on hemostatic hydrogel design and associated hemostatic mechanisms. Additionally, recent advancements in the application of these materials are discussed in detail, especially in clinical trials. Finally, the challenges and potential advancements of hemostatic hydrogels are analyzed and assessed. This review seeks to emphasize the role of hydrogels in biomedical applications for hemorrhage control and provide perspectives on the innovation of clinically applicable hemostatic materials.
Collapse
Affiliation(s)
- Lijun Liu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Rui Jing
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Lei Yao
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yanbo Wang
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Lihua Mu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuan Hu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
3
|
Hong G, Khazaee T, Cobos SF, Christiansen SD, Liu J, Drangova M, Holdsworth DW. Characterizing diffusion-controlled release of small-molecules using quantitative MRI in view of applications to orthopedic infection. NMR IN BIOMEDICINE 2024; 37:e5254. [PMID: 39358036 DOI: 10.1002/nbm.5254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024]
Abstract
Calcium sulfate is an established carrier for localized drug delivery, but a means to non-invasively measure drug release, which would improve our understanding of localized delivery, remains an unmet need. We aim to quantitatively estimate the diffusion-controlled release of small molecules loaded into a calcium sulfate carrier through a gadobutrol-based contrast agent, which acts as a surrogate small molecule. A central cylindrical core made of calcium sulfate, either alone or within a metal scaffold, is loaded with contrast agents that release into agar. Multi-echo scans are acquired at multiple time points over 4 weeks and processed into R2* and quantitative susceptibility mapping (QSM) maps. Mean R2* values are fit to a known drug delivery model, which are then compared with the decrease in core QSM. Fitting R2* measurements of calcium sulfate core while constraining constants to a drug release model results in an R2-value of 0.991, yielding a diffusion constant of 4.59 × 10-11 m2 s-1. Incorporating the carrier within a metal scaffold results in a slower release. QSM shows the resulting loss of susceptibility in the non-metal core but is unreliable around metal. R2* characterizes the released gadobutrol, and QSM detects the resulting decrease in core susceptibility. The addition of a porous metal scaffold slows the release of gadobutrol, as expected.
Collapse
Affiliation(s)
- Greg Hong
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Tina Khazaee
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Santiago F Cobos
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Spencer D Christiansen
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Junmin Liu
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
| | - Maria Drangova
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - David W Holdsworth
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
4
|
Ansari M, Shahlaei M, Hosseinzadeh S, Moradi S. Recent advances in nanostructured delivery systems for vancomycin. Nanomedicine (Lond) 2024; 19:1931-1951. [PMID: 39143926 PMCID: PMC11457640 DOI: 10.1080/17435889.2024.2377063] [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: 03/04/2024] [Accepted: 06/30/2024] [Indexed: 08/16/2024] Open
Abstract
Despite the development of new generations of antibiotics, vancomycin remained as a high-efficacy antibiotic for treating the infections caused by MRSA. Researchers have explored various nanoformulations, aiming to enhance the therapeutic efficacy of vancomycin. Such novel formulations improve the effectiveness of drug cargoes in treating bacterial infections and minimizing the risk of adverse effects. The vast of researches have focuses on enhancing the permeation ability of vancomycin through different biological barriers especially those of gastrointestinal tract. Increasing the drug loading and tuning the drug release from nanocarrier are other important goal for many conducted studies. This study reviews the newest nano-based formulations for vancomycin as a key antibiotic in treating hospitalized bacterial infections.
Collapse
Affiliation(s)
- Mohabbat Ansari
- Department of Tissue Engineering & Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering & Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Xu W, Huang W, Cai X, Dang Z, Hao L, Wang L. Dexamethasone Long-Term Controlled Release from Injectable Dual-Network Hydrogels with Porous Microspheres Immunomodulation Promotes Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40581-40601. [PMID: 39074361 PMCID: PMC11311136 DOI: 10.1021/acsami.4c06661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Long-lasting, controlled-release, and minimally invasive injectable platforms that provide a stable blood concentration to promote bone regeneration are less well developed. Using hexagonal mesoporous silica (HMS) loaded with dexamethasone (DEX) and poly(lactic-co-glycolic acid) (PLGA), we prepared porous DEX/HMS/PLGA microspheres (PDHP). In contrast to HMS/PLGA microspheres (HP), porous HMS/PLGA microspheres (PHP), DEX/PLGA microspheres (DP), and DEX/HMS/PLGA microspheres (DHP), PDHP showed notable immuno-coordinated osteogenic capabilities and were best at promoting bone mesenchymal stem cell proliferation and osteogenic differentiation. PDHP were combined with methacrylated silk (SilMA) and sodium alginate (SA) to form an injectable photocurable dual-network hydrogel platform that could continuously release the drug for more than 4 months. By adjusting the content of the microspheres in the hydrogel, a zero-order release hydrogel platform was obtained in vitro for 48 days. When the microsphere content was 1%, the hydrogel platform exhibited the best biocompatibility and osteogenic effects. The expression levels of the osteogenic gene alkaline phosphatases, BMP-2 and OPN were 10 to 15 times higher in the 1% group than in the 0% group, respectively. In addition, the 1% microsphere hydrogel strongly stimulated macrophage polarization to the M2 phenotype, establishing an immunological milieu that supports bone regrowth. The aforementioned outcomes were also observed in vivo. The most successful method for correcting cranial bone abnormalities in SD rats was to use a hydrogel called SilMA/SA containing 1% drug-loaded porous microspheres (PDHP/SS). The angiogenic and osteogenic effects of this treatment were also noticeably greater in the PDHP/SS group than in the control and blank groups. In addition, PDHP/SS polarized M2 macrophages and suppressed M1 macrophages in vivo, which reduced the local immune-inflammatory response, promoted angiogenesis, and cooperatively aided in situ bone healing. This work highlights the potential application of an advanced hydrogel platform for long-term, on-demand, controlled release for bone tissue engineering.
Collapse
Affiliation(s)
- Weikang Xu
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- Guangdong
Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology
Research Centre, No.
10 Shiliugang Road, Jianghai Avenue Central, Haizhu
District, Guangzhou 510316, China
| | - Weihua Huang
- Affiliated
Qingyuan Hospital, Guangzhou Medical University,
Qingyuan People’s Hospital, No. 35, Yinquan North Road, Qingcheng District, Qingyuan 511518, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- Department
of Orthopaedic Surgery, the Second Affiliated Hospital of Guangzhou
Medical University, the Second Clinical
Medicine School of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu
District, Guangzhou 510260, China
| | - Xiayu Cai
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
| | - Zhaohui Dang
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- National
Engineering Research Centre for Human Tissue Restoration and Function
Reconstruction, South China University of
Technology, No. 381 Wushan Road, Guangzhou 510275, China
| | - Lijing Hao
- National
Engineering Research Centre for Human Tissue Restoration and Function
Reconstruction, South China University of
Technology, No. 381 Wushan Road, Guangzhou 510275, China
| | - Liyan Wang
- Department
of Stomatology, Foshan Women’s and Children’s Hospital, No. 11 Renmin Xi Road, Chancheng
District, Foshan 528000, China
| |
Collapse
|
6
|
Liang S, Xiao L, Chen T, Roa P, Cocco E, Peng Z, Yu L, Wu M, Liu J, Zhao X, Deng W, Wang X, Zhao C, Deng Y, Mai Y. Injectable Nanocomposite Hydrogels Improve Intraperitoneal Co-delivery of Chemotherapeutics and Immune Checkpoint Inhibitors for Enhanced Peritoneal Metastasis Therapy. ACS NANO 2024; 18:18963-18979. [PMID: 39004822 DOI: 10.1021/acsnano.4c02312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Intraperitoneal co-delivery of chemotherapeutic drugs (CDs) and immune checkpoint inhibitors (ICIs) brings hope to improve treatment outcomes in patients with peritoneal metastasis from ovarian cancer (OC). However, current intraperitoneal drug delivery systems face issues such as rapid drug clearance from lymphatic drainage, heterogeneous drug distribution, and uncontrolled release of therapeutic agents into the peritoneal cavity. Herein, we developed an injectable nanohydrogel by combining carboxymethyl chitosan (CMCS) with bioadhesive nanoparticles (BNPs) based on polylactic acid-hyperbranched polyglycerol. This system enables the codelivery of CD and ICI into the intraperitoneal space to extend drug retention. The nanohydrogel is formed by cross-linking of aldehyde groups on BNPs with amine groups on CMCS via reversible Schiff base bonds, with CD and ICI loaded separately into BNPs and CMCS network. BNP/CMCS nanohydrogel maintained the activity of the biomolecules and released drugs in a sustained manner over a 7 day period. The adhesive property, through the formation of Schiff bases with peritoneal tissues, confers BNPs with an extended residence time in the peritoneal cavity after being released from the nanohydrogel. In a mouse model, BNP/CMCS nanohydrogel loaded with paclitaxel (PTX) and anti-PD-1 antibodies (αPD-1) significantly suppressed peritoneal metastasis of OC compared to all other tested groups. In addition, no systemic toxicity of nanohydrogel-loaded PTX and αPD-1 was observed during the treatment, which supports potential translational applications of this delivery system.
Collapse
Affiliation(s)
- Shu Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen 518020, China
| | - Tian Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Paola Roa
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Jie Liu
- ISCTE Business School, BRU-IUL, University Institute of Lisbon, Avenida das Armadas, Lisbon 1649-026, Portugal
| | - Xizhe Zhao
- Department of Chemistry, College of Staten Island, City University of New York, New York, New York 10314, United States
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Chao Zhao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35487, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| |
Collapse
|
7
|
Farasati
Far B, Safaei M, Nahavandi R, Gholami A, Naimi-Jamal MR, Tamang S, Ahn JE, Ramezani Farani M, Huh YS. Hydrogel Encapsulation Techniques and Its Clinical Applications in Drug Delivery and Regenerative Medicine: A Systematic Review. ACS OMEGA 2024; 9:29139-29158. [PMID: 39005800 PMCID: PMC11238230 DOI: 10.1021/acsomega.3c10102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 07/16/2024]
Abstract
Hydrogel encapsulation is a promising carrier for cell and drug delivery due to its ability to protect the encapsulated entities from harsh physiological conditions and enhance their therapeutic efficacy and bioavailability. However, there is not yet consensus on the optimal hydrogel type, encapsulation method, and clinical application. Therefore, a systematic review of hydrogel encapsulation techniques and their potential for clinical application is needed to provide a comprehensive and up-to-date overview. In this systematic review, we searched electronic databases for articles published between 2008 and 2023 that described the encapsulation of cells or drug molecules within hydrogels. Herein, we identified 9 relevant studies that met the inclusion and exclusion criteria of our study. Our analysis revealed that the physicochemical properties of the hydrogel, such as its porosity, swelling behavior, and degradation rate, play a critical role in the encapsulation of cells or drug molecules. Furthermore, the encapsulation method, including physical, chemical, or biological methods, can affect the encapsulated entities' stability, bioavailability, and therapeutic efficacy. Challenges of hydrogel encapsulation include poor control over the release of encapsulated entities, limited shelf life, and potential immune responses. Future directions of hydrogel encapsulation include the development of novel hydrogel and encapsulation methods and the integration of hydrogel encapsulation with other technologies, such as 3D printing and gene editing. In conclusion, this review is useful for researchers, clinicians, and policymakers who are interested in this field of drug delivery and regenerative medicine that can serve as a guide for the future development of novel technologies that can be applied into clinical practice.
Collapse
Affiliation(s)
- Bahareh Farasati
Far
- Department
of Chemistry, Iran University of Science
and Technology, Tehran 13114-16846, Iran
| | - Maryam Safaei
- Department
of Pharmacology, Faculty of Pharmacy, Eastern
Mediterranean University, via Mersin 10, Famagusta, TR. North Cyprus 99628, Turkey
| | - Reza Nahavandi
- School
of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Amir Gholami
- Faculty
of Medicine, Kurdistan University of Medical
Science, Sanandaj 6618634683, Iran
| | | | - Sujina Tamang
- NanoBio
High-Tech Materials Research Center, Department of Biological Sciences
and Bioengineering, Inha University, Incheon, 402-751, Republic of Korea
| | - Jung Eun Ahn
- NanoBio
High-Tech Materials Research Center, Department of Biological Sciences
and Bioengineering, Inha University, Incheon, 402-751, Republic of Korea
| | - Marzieh Ramezani Farani
- NanoBio
High-Tech Materials Research Center, Department of Biological Sciences
and Bioengineering, Inha University, Incheon, 402-751, Republic of Korea
| | - Yun Suk Huh
- NanoBio
High-Tech Materials Research Center, Department of Biological Sciences
and Bioengineering, Inha University, Incheon, 402-751, Republic of Korea
| |
Collapse
|
8
|
Fan S, Liu Q, Dong J, Ai X, Li J, Huang W, Sun T. In situ forming an injectable hyaluronic acid hydrogel for drug delivery and synergistic tumor therapy. Heliyon 2024; 10:e32135. [PMID: 38867981 PMCID: PMC11168435 DOI: 10.1016/j.heliyon.2024.e32135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Stimulus-responsive injectable hydrogel has the key characteristics of in situ drug-loading ability and the controlled drug release, enabling efficient delivery and precise release of chemotherapy drugs at the tumor site, thereby being used as a local drug delivery system for sustained tumor treatment. This article designed a smart responsive injectable hydrogel loaded with anti-tumor drugs and nanoparticles to achieve efficient and specific synergistic treatment of tumors. Hyaluronic acid (HA) hydrogel obtained by cross-linking HA-SH (HS) and HA-Tyr (HT) through horseradish peroxidase (HRP), and doxorubicin hydrochloride (DOX) and folic acid-polyethylene glycol-amine (FA-PEG-NH2) modified PDA (denoted as PPF) were encapsulated to construct the HS/HT@PPF/D hydrogel. The hydrogel had good biocompatibility, injectability, and could respond to multiple stimuli at the tumor site, thereby achieving controlled drug release. At the same time, PPF gave it excellent photothermal efficiency, photothermal stability and tumor targeting. In vitro and in vivo experimental results showed that the HS/HT@PPF/D hydrogel combined with near-infrared laser irradiation could significantly improve its anti-tumor effect and could almost eliminate the entire tumor mass without obvious adverse reactions. The HS/HT@PPF/D hydrogel could achieve multi-stimulus-responsive drug delivery and be used for precise chemo-photothermal synergistic tumor treatment, thus providing a new platform for local synergistic tumor treatment.
Collapse
Affiliation(s)
- Sisi Fan
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Qinghuan Liu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Jia Dong
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Xiaorui Ai
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Jing Li
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Wei Huang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan, 430070, China
| |
Collapse
|
9
|
Britton D, Legocki J, Paul D, Katsara O, Aristizabal O, Pandya N, Mishkit O, Xiao Y, Aristizabal M, Rahman N, Schneider R, Wadghiri YZ, Montclare JK. Coiled-Coil Protein Hydrogels Engineered with Minimized Fiber Diameters for Sustained Release of Doxorubicin in Triple-Negative Breast Cancer. ACS Biomater Sci Eng 2024; 10:3425-3437. [PMID: 38622760 PMCID: PMC11094684 DOI: 10.1021/acsbiomaterials.4c00349] [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: 02/23/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks expressed protein targets, making therapy development challenging. Hydrogels offer a promising new route in this regard by improving the chemotherapeutic efficacy through increased solubility and sustained release. Moreover, subcutaneous hydrogel administration reduces patient burden by requiring less therapy and shorter treatment times. We recently established the design principles for the supramolecular assembly of single-domain coiled-coils into hydrogels. Using a modified computational design algorithm, we designed Q8, a hydrogel with rapid assembly for faster therapeutic hydrogel preparation. Q8 encapsulates and releases doxorubicin (Dox), enabling localized sustained release via subcutaneous injection. Remarkably, a single subcutaneous injection of Dox-laden Q8 (Q8•Dox) significantly suppresses tumors within just 1 week. This work showcases the bottom-up engineering of a fully protein-based drug delivery vehicle for improved TBNC treatment via noninvasive localized therapy.
Collapse
Affiliation(s)
- Dustin Britton
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Jakub Legocki
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Deven Paul
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Olga Katsara
- Department
of Microbiology, New York University Grossman
School of Medicine, New York, New York 10016, United States
| | - Orlando Aristizabal
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Neelam Pandya
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Orin Mishkit
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Yingxin Xiao
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Matias Aristizabal
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Neha Rahman
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Robert Schneider
- Department
of Microbiology, New York University Grossman
School of Medicine, New York, New York 10016, United States
- Department
of Radiation Oncology, New York University
Grossman School of Medicine, New
York, New York 10016, United States
| | - Youssef Z. Wadghiri
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Jin Kim Montclare
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
- Department
of Biomedical Engineering, New York University
Tandon School of Engineering, Brooklyn ,New York11201, United States
- Department
of Chemistry, New York University, New York, New York 10012, United States
- Department
of Biomaterials, New York University College
of Dentistry, New York, New York 10010, United States
| |
Collapse
|
10
|
Khairnar P, Phatale V, Shukla S, Tijani AO, Hedaoo A, Strauss J, Verana G, Vambhurkar G, Puri A, Srivastava S. Nanocarrier-Integrated Microneedles: Divulging the Potential of Novel Frontiers for Fostering the Management of Skin Ailments. Mol Pharm 2024; 21:2118-2147. [PMID: 38660711 DOI: 10.1021/acs.molpharmaceut.4c00144] [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: 04/26/2024]
Abstract
The various kinds of nanocarriers (NCs) have been explored for the delivery of therapeutics designed for the management of skin manifestations. The NCs are considered as one of the promising approaches for the skin delivery of therapeutics attributable to sustained release and enhanced skin penetration. Despite the extensive applications of the NCs, the challenges in their delivery via skin barrier (majorly stratum corneum) have persisted. To overcome all the challenges associated with the delivery of NCs, the microneedle (MN) technology has emerged as a beacon of hope. Programmable drug release, being painless, and its minimally invasive nature make it an intriguing strategy to circumvent the multiple challenges associated with the various drug delivery systems. The integration of positive traits of NCs and MNs boosts therapeutic effectiveness by evading stratum corneum, facilitating the delivery of NCs through the skin and enhancing their targeted delivery. This review discusses the barrier function of skin, the importance of MNs, the types of MNs, and the superiority of NC-loaded MNs. We highlighted the applications of NC-integrated MNs for the management of various skin ailments, combinational drug delivery, active targeting, in vivo imaging, and as theranostics. The clinical trials, patent portfolio, and marketed products of drug/NC-integrated MNs are covered. Finally, regulatory hurdles toward benchtop-to-bedside translation, along with promising prospects needed to scale up NC-integrated MN technology, have been deliberated. The current review is anticipated to deliver thoughtful visions to researchers, clinicians, and formulation scientists for the successful development of the MN-technology-based product by carefully optimizing all the formulation variables.
Collapse
Affiliation(s)
- Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, Tennessee 37614, United States
| | - Aachal Hedaoo
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Jordan Strauss
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, Tennessee 37614, United States
| | - Gabrielle Verana
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, Tennessee 37614, United States
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, Tennessee 37614, United States
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| |
Collapse
|
11
|
Lin TW, Sing CE. Effect of penetrant-polymer interactions and shape on the motion of molecular penetrants in dense polymer networks. J Chem Phys 2024; 160:114905. [PMID: 38511661 DOI: 10.1063/5.0197140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024] Open
Abstract
The diffusion of dilute molecular penetrants within polymers plays a crucial role in the advancement of material engineering for applications such as coatings and membrane separations. The potential of highly cross-linked polymer networks in these applications stems from their capacity to adjust the size and shape selectivity through subtle changes in network structures. In this paper, we use molecular dynamics simulation to understand the role of penetrant shape (aspect ratios) and its interaction with polymer networks on its diffusivity. We characterize both local penetrant hopping and the long-time diffusive motion for penetrants and consider different aspect ratios and penetrant-network interaction strengths at a variety of cross-link densities and temperatures. The shape affects the coupling of penetrant motion to the cross-link density- and temperature-dependent structural relaxation of networks and also affects the way a penetrant experiences the confinement from the network meshes. The attractive interaction between the penetrant and network primarily affects the former since only the system of dilute limit is of present interest. These results offer fundamental insights into the intricate interplay between penetrant characteristics and polymer network properties and also suggest future directions for manipulating polymer design to enhance the separation efficiency.
Collapse
Affiliation(s)
- Tsai-Wei Lin
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Charles E Sing
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
12
|
Vu TT, Jo SH, Kim SH, Kim BK, Park SH, Lim KT. Injectable and Multifunctional Hydrogels Based on Poly( N-acryloyl glycinamide) and Alginate Derivatives for Antitumor Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38470564 DOI: 10.1021/acsami.4c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Chemotherapy is a conventional treatment that uses drugs to kill cancer cells; however, it may induce side effects and may be incompletely effective, leading to the risk of tumor recurrence. To address this issue, we developed novel injectable thermal/near-infrared (NIR)-responsive hydrogels to control drug release. The injectable hydrogel formulation was composed of biocompatible alginates, poly(N-acryloyl glycinamide) (PNAGA) copolymers with an upper critical solution temperature, and NIR-responsive cross-linkers containing coumarin groups, which were gelated through bioorthogonal inverse electron demand Diels-Alder reactions. The hydrogels exhibited quick gelation times (120-800 s) and high drug loading efficiencies (>90%). The hydrogels demonstrated a higher percentage of drug release at 37 °C than that at 25 °C due to the enhanced swelling behavior of temperature-responsive PNAGA moieties. Upon NIR irradiation, the hydrogels released most of the entrapped doxorubicin (DOX) (97%) owing to the cleavage of NIR-sensitive coumarin ester groups. The hydrogels displayed biocompatibility with normal cells, while induced antitumor activity toward cancer cells. DOX/hydrogels treated with NIR light inhibited tumor growth in nude mice bearing tumors. In addition, the injected hydrogels emitted red fluorescence upon excitation at a green wavelength, so that the drug delivery and hydrogel degradation in vivo could be tracked in the xenograft model.
Collapse
Affiliation(s)
- Trung Thang Vu
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, South Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, South Korea
| | - Sung-Han Jo
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, South Korea
| | - Seon-Hwa Kim
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, South Korea
| | - Byeong Kook Kim
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, South Korea
| | - Sang-Hyug Park
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, South Korea
| | - Kwon Taek Lim
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, South Korea
- Institute of Display Semiconductor Technology, Pukyong National University, Busan 48513, South Korea
| |
Collapse
|
13
|
Goshtasbi H, Abdolahinia ED, Fathi M, Movafeghi A, Omidian H, Barar J, Omidi Y. Astaxanthin-loaded alginate-chitosan gel beads activate Nrf2 and pro-apoptotic signalling pathways against oxidative stress. J Microencapsul 2024; 41:140-156. [PMID: 38410930 DOI: 10.1080/02652048.2024.2319048] [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: 04/23/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
Oxidative stress (OS) plays a crucial role in disease development. Astaxanthin (ATX), a valuable natural compound, may reduce OS and serve as a treatment for diseases like neurodegenerative disorders and cancer. Nuclear factor-erythroid 2-related factor 2 (Nrf2) regulates antioxidant enzymes and OS management. We evaluated ATX's antioxidant activity via Alg-CS/ATX gel beads in vitro. ATX-encapsulated alginate-chitosan (Alg-CS/ATX) gel beads were synthesized and structurally/morphologically characterized by SEM, FT-IR, and XRD. Their biological effects were examined in human umbilical vein endothelial cells (HUVECs) treated with H2O2 through MTT assay, Annexin V/PI, cell cycle studies, and western blotting. Alg-CS effectively carried ATX, with high capacity and reduced pore size. Alg-CS/ATX displayed an 84% encapsulation efficiency, maintaining stability for 30 days. In vitro studies showed a 1.4-fold faster release at pH 5.4 than at neutral pH, improving ATX's therapeutic potential. HUVECs treated with Alg-CS/ATX showed enhanced viability via increased Nrf2 expression. Alg-CS gel beads exhibit significant potential as a biocompatible vehicle for delivering ATX to combat OS with considerable opportunity for clinical applications.
Collapse
Affiliation(s)
- Hamieh Goshtasbi
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Movafeghi
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Hossein Omidian
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
14
|
Pérez-Flores JG, García-Curiel L, Pérez-Escalante E, Contreras-López E, Olloqui EJ. Arabinoxylans matrixes as a potential material for drug delivery systems development - A bibliometric analysis and literature review. Heliyon 2024; 10:e25445. [PMID: 38352745 PMCID: PMC10862686 DOI: 10.1016/j.heliyon.2024.e25445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Arabinoxylans (AX) have become a focal point in the pharmaceutical sector owing to their physicochemical, biological, and functional properties. The purpose of this paper was to present a summary of the utilization of AX as drug release matrices through a bibliometric analysis (BA) and a literature review to spotlight the AX functional characteristics and their technological applications to promote this line of research. The BA was carried out using data from a Web of Science database research, specifically emphasizing the analysis of authors' keywords. This approach was chosen due to its significance in comprehensively understanding a particular research field and its relevance for in-depth knowledge of a research field. The BA outcomes revealed limited information concerning the AX applications in both release matrices and as excipients in the formulation and development of drug delivery systems (DDS), so there is a need for additional scientific and technological research in these areas to address the existing information gaps. However, the literature review shows that the native and modified AX from different delivery release systems, such as macrogels (including films, tablets, and hard gelatin capsules) and multi-particulate systems (including micro and nanogels), present an excellent potential as release matrices of biomolecules and drugs, such as doxorubicin, diclofenac sodium, caffeine, gentamicin, tizanidine hydrochloride, and insulin. In conclusion, AX have a wide potential for application in the pharmaceutical industry, so this work is expected to be a reference point for future research by scientists, technologists, and entrepreneurs who cope with the subject.
Collapse
Affiliation(s)
- Jesús Guadalupe Pérez-Flores
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción s/n, Carretera Pachuca-Actopan, 42060, San Agustín Tlaxiaca, Hidalgo, Mexico
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo km 4.5, 42184, Mineral de la Reforma, Hidalgo, Mexico
| | - Laura García-Curiel
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción s/n, Carretera Pachuca-Actopan, 42060, San Agustín Tlaxiaca, Hidalgo, Mexico
| | - Emmanuel Pérez-Escalante
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo km 4.5, 42184, Mineral de la Reforma, Hidalgo, Mexico
| | - Elizabeth Contreras-López
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo km 4.5, 42184, Mineral de la Reforma, Hidalgo, Mexico
| | - Enrique J. Olloqui
- CONAHCyT, Colegio de Postgraduados, Campus Puebla, Boulevard Forjadores, 72760, Puebla, Puebla, Mexico
| |
Collapse
|
15
|
Koshani R, Nia MH, Ataie Z, Wang Y, Kakkar A, van de Ven TGM. Multifunctional self-healing hydrogels via nanoengineering of colloidal and polymeric cellulose. Int J Biol Macromol 2024; 259:129181. [PMID: 38184036 DOI: 10.1016/j.ijbiomac.2023.129181] [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: 09/27/2023] [Revised: 12/13/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
The unique features of self-healing hydrogels hold great potential for biomedical applications including injectable hydrogels for cancer treatment, procedures for tumor removal or resection. However, the fabrication of durable and multifunctional self-healing hydrogels composed of biocompatible, green building blocks via versatile synthetic methodology continues to pose a significant challenge. Here, we engineered dialdehyde cellulose (DAC, as a macromolecular bio-crosslinker), and electrosterically stabilized nanocrystalline cellulose (ENCC, as a ligand-targeted drug carrier) to facilitate a strategy for the construction of self-healing hydrogels. Benefiting from its high carboxyl group density, ENCC was functionalized with folic acid (FA) using a non-toxic DMTMM coupling agent and loaded with doxorubicin (DOX, a model drug) through electrostatic interactions. A natural self-healing hydrogel was prepared from carboxymethyl chitosan (CCTS) and DAC mixed with DOX-loaded FA-ENCC using dynamic Schiff-base and hydrogen linkages. A combination of active supramolecular and vital covalent junctions led to a soft (storage modulus ∼500 Pa) and durable material, with rapid (< 5 min) reconstruction of molecular structure from fractured and injected to intact forms. The DAC-CCTS hydrogel showed an appreciable loading capacity of ∼5 mg g-1. Biocompatibility of the hydrogels was evaluated using cell viability and metabolic activity assays, showing lower metabolic activity due to sustained release of its cargo. These materials offer a versatile, sustainable, and green platform for the efficient construction of hydrogels, based on macro- and nano-engineered cellulose, the most abundant and easily accessible biopolymer.
Collapse
Affiliation(s)
- Roya Koshani
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Department of Food Science and Agricultural Chemistry, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada; Pulp and Paper Research Centre, McGill University, 3420 University Street, Montreal, QC H3A 2A7, Canada; Quebec Centre for Advanced Materials (QCAM), 3420 University Street, Montreal, QC H3A 2A7, Canada; Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, SK S7N 5C9, Canada.
| | - Marzieh Heidari Nia
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Pulp and Paper Research Centre, McGill University, 3420 University Street, Montreal, QC H3A 2A7, Canada; Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, SK S7N 5C9, Canada.
| | - Zaman Ataie
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Yixiang Wang
- Department of Food Science and Agricultural Chemistry, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada; Quebec Centre for Advanced Materials (QCAM), 3420 University Street, Montreal, QC H3A 2A7, Canada.
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Quebec Centre for Advanced Materials (QCAM), 3420 University Street, Montreal, QC H3A 2A7, Canada.
| | - Theo G M van de Ven
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Pulp and Paper Research Centre, McGill University, 3420 University Street, Montreal, QC H3A 2A7, Canada; Quebec Centre for Advanced Materials (QCAM), 3420 University Street, Montreal, QC H3A 2A7, Canada.
| |
Collapse
|
16
|
He W, Zhang Y, Qu Y, Liu M, Li G, Pan L, Xu X, Shi G, Hao Q, Liu F, Gao Y. Research progress on hydrogel-based drug therapy in melanoma immunotherapy. BMB Rep 2024; 57:71-78. [PMID: 38053295 PMCID: PMC10910090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Melanoma is one of the most aggressive skin tumors, and conventional treatment modalities are not effective in treating advanced melanoma. Although immunotherapy is an effective treatment for melanoma, it has disadvantages, such as a poor response rate and serious systemic immune-related toxic side effects. The main solution to this problem is the use of biological materials such as hydrogels to reduce these side effects and amplify the immune killing effect against tumor cells. Hydrogels have great advantages as local slow-release drug carriers, including the ability to deliver antitumor drugs directly to the tumor site, enhance the local drug concentration in tumor tissue, reduce systemic drug distribution and exhibit good degradability. Despite these advantages, there has been limited research on the application of hydrogels in melanoma treatment. Therefore, this article provides a comprehensive review of the potential application of hydrogels in melanoma immunotherapy. Hydrogels can serve as carriers for sustained drug delivery, enabling the targeted and localized delivery of drugs with minimal systemic side effects. This approach has the potential to improve the efficacy of immunotherapy for melanoma. Thus, the use of hydrogels as drug delivery vehicles for melanoma immunotherapy has great potential and warrants further exploration. [BMB Reports 2024; 57(2): 71-78].
Collapse
Affiliation(s)
- Wei He
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yanqin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yi Qu
- Department of Xi’an Shunmei Medical Cosmetology Outpatient, Xi’an 710075, China
| | - Mengmeng Liu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Guodong Li
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Luxiang Pan
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Xinyao Xu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Gege Shi
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen 510515, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
17
|
Garshasbi HR, Soleymani S, Naghib SM, Mozafari MR. Multi-stimuli-responsive Hydrogels for Therapeutic Systems: An Overview on Emerging Materials, Devices, and Drugs. Curr Pharm Des 2024; 30:2027-2046. [PMID: 38877860 DOI: 10.2174/0113816128304924240527113111] [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: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 09/21/2024]
Abstract
The rising interest in hydrogels nowadays is due to their usefulness in physiological conditions as multi-stimuli-responsive hydrogels. To reply to the prearranged stimuli, including chemical triggers, light, magnetic field, electric field, ionic strength, temperature, pH, and glucose levels, dual/multi-stimuli-sensitive gels/hydrogels display controllable variations in mechanical characteristics and swelling. Recent attention has focused on injectable hydrogel-based drug delivery systems (DDS) because of its promise to offer regulated, controlled, and targeted medication release to the tumor site. These technologies have great potential to improve treatment outcomes and lessen side effects from prolonged chemotherapy exposure.
Collapse
Affiliation(s)
- Hamid Reza Garshasbi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Sina Soleymani
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
- Biomaterials and Tissue Engineering Research Group, Department of Interdisciplinary Technologies, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
18
|
Chen B, Zhu D, Li Q, Wang C, Cui J, Zheng Z, Wang X. Mechanically Reinforced and Injectable Universal Adhesive Based on a PEI-PAA/Alg Dual-Network Hydrogel Designed by Topological Entanglement and Catechol Chemistry. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59826-59837. [PMID: 38098133 DOI: 10.1021/acsami.3c14743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Universal adhesion of hydrogels to diverse materials is essential to their extensive applications. Unfortunately, tough adhesion of wet surfaces remains an urgent challenge so far, requiring robust cohesion strength for effective stress dissipation. In this work, a dual-network hydrogel polyethylenimine-poly(acrylic acid)/alginate (PEI-PAA/Alg) with excellent mechanical strength is realized via PEI-PAA complex and calcium alginate coordination for universal adhesion by the synergistic effort of topological entanglement and catechol chemistry. The dual networks of PEI-PAA/Alg provide mechanically reinforced cohesion strength, which is sufficient for energy dissipation during adhesion with universal materials. After the integration of mussel-inspired dopamine into PAA or Alg, the adhesive demonstrates further improved adhesion performance with a solid adherend and capability to bond cancellous bones. Notably, the dopamine-modified adhesive exhibits better instant adhesion and reversibility with wet surfaces compared with commercial fibrin. Adhesion interfaces are investigated by SEM and micro-FTIR to verify the effectiveness of strategies of topological entanglement. Furthermore, the adhesive also possesses great injectability, stability, tissue adhesion, and biocompatibility. In vivo wound healing and histological analysis indicate that the hydrogel can promote wound closure, epidermis regeneration, and tissue refunctionalization, implying its potential application for bioadhesive and wound dressing.
Collapse
Affiliation(s)
- Buyun Chen
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dandan Zhu
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiang Li
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chenhao Wang
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiahua Cui
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhen Zheng
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinling Wang
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
19
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Dinakar YH, Rajana N, Kumari NU, Jain V, Mehra NK. Recent Advances of Multifunctional PLGA Nanocarriers in the Management of Triple-Negative Breast Cancer. AAPS PharmSciTech 2023; 24:258. [PMID: 38097825 DOI: 10.1208/s12249-023-02712-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Even though chemotherapy stands as a standard option in the therapy of TNBC, problems associated with it such as anemia, bone marrow suppression, immune suppression, toxic effects on healthy cells, and multi-drug resistance (MDR) can compromise their effects. Nanoparticles gained paramount importance in overcoming the limitations of conventional chemotherapy. Among the various options, nanotechnology has appeared as a promising path in preclinical and clinical studies for early diagnosis of primary tumors and metastases and destroying tumor cells. PLGA has been extensively studied amongst various materials used for the preparation of nanocarriers for anticancer drug delivery and adjuvant therapy because of their capability of higher encapsulation, easy surface functionalization, increased stability, protection of drugs from degradation versatility, biocompatibility, and biodegradability. Furthermore, this review also provides an overview of PLGA-based nanoparticles including hybrid nanoparticles such as the inorganic PLGA nanoparticles, lipid-coated PLGA nanoparticles, cell membrane-coated PLGA nanoparticles, hydrogels, exosomes, and nanofibers. The effects of all these systems in various in vitro and in vivo models of TNBC were explained thus pointing PLGA-based NPs as a strategy for the management of TNBC.
Collapse
Affiliation(s)
- Yirivinti Hayagreeva Dinakar
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Nalla Usha Kumari
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India.
| |
Collapse
|
21
|
Yu YH, Lee CH, Hsu YH, Chou YC, Hong BK, Huang CT, Liu SJ. Novel CO 2-encapsulated Pluronic F127 hydrogel for the treatment of Achilles tendon injury. Sci Rep 2023; 13:21895. [PMID: 38081952 PMCID: PMC10713641 DOI: 10.1038/s41598-023-49339-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Nonsurgical treatment and surgical repairment of injured Achilles tendons seldom restore the wounded tendon to its original elasticity and stiffness. Therefore, we hypothesized that the surgically repaired Achilles tendon can achieve satisfactory regeneration by applying multi-drug encapsulated hydrogels. In this study, a novel bupivacaine-eluting carbon dioxide-encapsulated Pluronic F127 hydrogel (BC-hydrogel) was developed for the treatment of Achilles tendon injuries. The rheological properties of BC-hydrogel were measured. A high-performance liquid chromatography assay was used to assess the release characteristics of bupivacaine in both in vitro and in vivo settings. Furthermore, the effectiveness of BC-hydrogel in treating torn tendons was examined in a rat model, and histological analyses were conducted. Evidently, the degradable hydrogels continuously eluted bupivacaine for more than 14 days. The animal study results revealed that the BC-hydrogel improved the post-surgery mobility of the animals compared with pristine hydrogels. Histological assay results demonstrated a significant reaction to high vascular endothelial growth factor in the surrounding tissues and expression of collagen I within the repaired tendon. This demonstrates the potential of this novel BC-hydrogel as an effective treatment method for Achilles tendon injuries.
Collapse
Affiliation(s)
- Yi-Hsun Yu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 33305, Taiwan
| | - Chen-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Tao-Yuan, 33305, Taiwan
| | - Yung-Heng Hsu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 33305, Taiwan
| | - Ying-Chao Chou
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 33305, Taiwan
| | - Bo-Kui Hong
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, 33302, Taiwan
| | - Chao-Tsai Huang
- Department of Chemical and Materials Engineering, Tamkang University, New Taipei City, 251301, Taiwan
| | - Shih-Jung Liu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 33305, Taiwan.
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, 33302, Taiwan.
- Biomaterials Lab, Mechanical Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302, Taiwan.
| |
Collapse
|
22
|
Dibazar ZE, Zarei M, Mohammadikhah M, Oudah SK, Elyasi M, Kokabi H, Shahgolzari M, Asl LD, Azizy M. Crosslinking strategies for biomimetic hydrogels in bone tissue engineering. Biophys Rev 2023; 15:2027-2040. [PMID: 38192345 PMCID: PMC10771399 DOI: 10.1007/s12551-023-01141-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/03/2023] [Indexed: 01/10/2024] Open
Abstract
Bone tissue engineering has become a popular area of study for making biomimetic hydrogels to treat bone diseases. In this work, we looked at biocompatible hydrogels that can be injected into bone defects that require the smallest possible surgery. Mineral ions can be attached to polymer chains to make useful hydrogels that help bones heal faster. These ions are very important for the balance of the body. In the chemically-triggered sector, advanced hydrogels cross-linked by different molecular agents have many advantages, such as being selective, able to form gels, and having mechanical properties that can be modified. In addition, different photo-initiators can be used to make photo cross linkable hydrogels react quickly and moderately under certain light bands. Enzyme-triggered hydrogels are another type of hydrogel that can be used to repair bone tissue because they are biocompatible and gel quickly. We also look at some of the important factors mentioned above that could change how well bone tissue engineering works as a therapy. Finally, this review summarizes the problems that still need to be solved to make clinically relevant hydrogels.
Collapse
Affiliation(s)
- Zahra Ebrahimvand Dibazar
- Department of Oral and Maxillo Facial Medicine, Faculty of Dentistry, Tabriz Azad University of Medical Sciences, Tabriz, 5165687386 Iran
| | - Mahdi Zarei
- Student Research Committee, Tabriz university of medical sciences, Tabriz, Iran
| | - Meysam Mohammadikhah
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Milad Elyasi
- Otolaryngology department, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Hadi Kokabi
- Department of Periodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, 65175-4171 Iran
| | - Mehdi Shahgolzari
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, 65175-4171 Iran
| | - Leila Delnabi Asl
- Department of Internal Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Azizy
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
23
|
Ni H, Qian S, Lu J, Feng J, Mou XZ, Zhang J. Natural Polysaccharide Delivery Platforms with Multiscale Structure Used for Cancer Chemoimmunotherapy. Mol Pharm 2023; 20:5778-5789. [PMID: 37752866 DOI: 10.1021/acs.molpharmaceut.3c00633] [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: 09/28/2023]
Abstract
Chemoimmunotherapy is an effective cancer treatment method. Drugs are always combined and used in treating cancer. However, the characteristic of drugs varies, making it challenging to control their release kinetics utilizing delivery devices with a single microstructure. In this study, we attempted to uniformly size drugs of varying molecular weights and confine them in a compartment where immune cells may be recruited and moved freely. Dextran microgels were created as modular drug libraries to address the cryogel burst release of small molecule drugs. Then, modular drug libraries and granulocyte-macrophage colony-stimulating factor (GM-CSF) were integrated into cryogels for a combined treatment. Herein, alginate was zwitterion modified to avoid the immune reaction generated by the material. Because of its macroporous structure, the cryogel could be injected into the body, eliminating invasive surgical procedures. Results demonstrated that multiscale delivery platforms could improve the synergistic effect of various medications on tumor treatment.
Collapse
Affiliation(s)
- Haifeng Ni
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Sunxiang Qian
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Lu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Feng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang 310014, P. R. China
| | - Jing Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| |
Collapse
|
24
|
Liu J, Zhou J, Meng Y, Zhu L, Xu J, Huang Z, Wang S, Xia Y. Artificial Skin with Patterned Stripes for Color Camouflage and Thermoregulation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48601-48612. [PMID: 37787638 DOI: 10.1021/acsami.3c08872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Chameleons are famous for their quick color changing abilities, and it is commonly assumed that they do this for camouflage. However, recent reports revealed that chameleons also change color for body temperature regulation. Inspired by the structure of the panther chameleon's skin, a stripe-patterned poly(N-isopropylacrylamide) (PNIPAM) and polyacrylamide (PAM) hydrogel film with a laminated structure is fabricated in this work; thus, both camouflage and thermoregulation can be achieved through controlling Vis and NIR light effectively. For the PNIPAM stripe, the upper layer is the native PNIPAM hydrogel and the lower layer is the carbon nanotube-composited PNIPAM hydrogel. Thus, the PNIPAM stripe is capable of reaching 28 °C at a low environmental temperature (12 °C) and a low radiation intensity (20 mW cm-2), while preventing the body temperature from rising by changing to white under a strong radiation intensity (100 mW cm-2). For the PAM stripe, the upper layer combines colloidal photonic crystals and displays a tunable structural color by stretching, and the lower layer is mixed with PNIPAM microgels for thermal regulation. Through the fabrication of multifunctional patterns, the film can achieve both dynamic structural color and thermoregulation by precisely controlling solar radiation absorption, scattering, and reflection. More importantly, in the stripe-patterned system, the shrinkage of the PNIPAM stripes can effectively trigger the elongation of the PAM stripe, which endows the structural color changing process to be self-powered completely. The performances show that the stripe-patterned film may have potential applications in intelligent coatings, especially in areas with large temperature differences during the day such as high plains.
Collapse
Affiliation(s)
- Jiahui Liu
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Jie Zhou
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Yaru Meng
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Liqian Zhu
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Jintao Xu
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Zehua Huang
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Shengjie Wang
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Yongqing Xia
- Department of Biological and Bioenergy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
25
|
Bhattacharya S, Bhattacharyya T, Khanra S, Banerjee R, Dash J. Nucleoside-Derived Metallohydrogel Induces Cell Death in Leishmania Parasites. ACS Infect Dis 2023; 9:1676-1684. [PMID: 37606735 DOI: 10.1021/acsinfecdis.2c00635] [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] [Indexed: 08/23/2023]
Abstract
Self-assembled hydrogels by virtue of their unique 3D network and tunability have extensively been explored for bio-medical applications like tissue engineering, delivery and release of therapeutic agents, etc. Herein, we demonstrate for the first-time nucleoside-based biocompatible hydrogels with a remarkable leishmanicidal effect against both Leishmania major promastigotes and amastigotes and no cytotoxic effect on the macrophage cell line. In this work, a series of biocompatible hydrogels have been synthesized by silver ion-driven self-assembly of natural nucleoside and nucleotide-like cytidine and 5'-GMP. The supramolecular metallogel obtained from the assembly of cytidine and boronic acid is capable of inducing apoptotic-like cell death of protozoan parasite by causing damage to the membrane as well as DNA. These hydrogels could find promising applications in combating cutaneous leishmaniasis by topical treatment.
Collapse
Affiliation(s)
- Semantee Bhattacharya
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Tanima Bhattacharyya
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Supriya Khanra
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata 700064, India
| | - Rahul Banerjee
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata 700064, India
| | - Jyotirmayee Dash
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
26
|
Remlova E, Feig VR, Kang Z, Patel A, Ballinger I, Ginzburg A, Kuosmanen J, Fabian N, Ishida K, Jenkins J, Hayward A, Traverso G. Activated Metals to Generate Heat for Biomedical Applications. ACS MATERIALS LETTERS 2023; 5:2508-2517. [PMID: 37680546 PMCID: PMC10481395 DOI: 10.1021/acsmaterialslett.3c00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023]
Abstract
Delivering heat in vivo could enhance a wide range of biomedical therapeutic and diagnostic technologies, including long-term drug delivery devices and cancer treatments. To date, providing thermal energy is highly power-intensive, rendering it oftentimes inaccessible outside of clinical settings. We developed an in vivo heating method based on the exothermic reaction between liquid-metal-activated aluminum and water. After establishing a method for consistent activation, we characterized the heat generation capabilities with thermal imaging and heat flux measurements. We then demonstrated one application of this reaction: to thermally actuate a gastric resident device made from a shape-memory alloy called Nitinol. Finally, we highlight the advantages and future directions for leveraging this novel in situ heat generation method beyond the showcased example.
Collapse
Affiliation(s)
- Eva Remlova
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Health Sciences and Technology, Eidgenössische
Technische Hochschule Zürich, Universitätstrasse 2, 8092 Zürich, Switzerland
| | - Vivian Rachel Feig
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ziliang Kang
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ashka Patel
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ian Ballinger
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Anna Ginzburg
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cell/Cellular and Molecular Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Johannes Kuosmanen
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Niora Fabian
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Comparative Medicine, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Keiko Ishida
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Joshua Jenkins
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alison Hayward
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Comparative Medicine, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Giovanni Traverso
- Division
of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
27
|
Pourmadadi M, Aslani A, Abdouss M. Synthesis and characterization of biological macromolecules double emulsion based on carboxymethylcellulose/gelatin hydrogel incorporated with ZIF-8 as metal organic frameworks for sustained anti-cancer drug release. Int J Biol Macromol 2023:125168. [PMID: 37270138 DOI: 10.1016/j.ijbiomac.2023.125168] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
The field of nanotechnology has introduced novel prospects for drug delivery systems, which have the potential to supplant conventional chemotherapy with reduced adverse effects. Despite being a promising porous material, ZIF-8, a metal-organic framework, tends to agglomerate in water, which limits its applicability. In order to resolve this problem, we added ZIF-8 to hydrogels consisting of gelatin and carboxymethylcellulose. This improved their mechanical strength and stability while avoiding aggregation. We utilized double emulsions with the hydrogels' biological macromolecules to construct drug carriers with enhanced control over drug release. The nanocarriers were subjected to various analytical techniques for characterization, such as Fourier-transform infrared (FTIR) spectroscopy, X-ray diffraction (XRD), field-emission scanning electron microscopy (FESEM), zeta potential, and dynamic light scattering (DLS). The findings of our study revealed that the mean size of the produced nanocarriers were 250 nm, and their zeta potential was -40.1 mV, which suggests favorable stability. The synthesized nanocarriers were found to exhibit cytotoxicity towards cancer cells, as evidenced by the results of MTT assays and flow cytometry tests. The cell viability percentage was determined to be 55 % for the prepared nanomedicine versus 70 % for the free drug. In summary, our study illustrates that the integration of ZIF-8 into hydrogels produces drug delivery systems with improved characteristics. Furthermore, the prepared nanocarriers exhibit potential for future investigation and advancement.
Collapse
Affiliation(s)
- Mehrab Pourmadadi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Ali Aslani
- Chemistry Department, Amirkabir University of Technology
| | - Majid Abdouss
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| |
Collapse
|
28
|
Luo Y, Li J, Ding Q, Wang H, Liu C, Wu J. Functionalized Hydrogel-Based Wearable Gas and Humidity Sensors. NANO-MICRO LETTERS 2023; 15:136. [PMID: 37225851 PMCID: PMC10209388 DOI: 10.1007/s40820-023-01109-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023]
Abstract
Breathing is an inherent human activity; however, the composition of the air we inhale and gas exhale remains unknown to us. To address this, wearable vapor sensors can help people monitor air composition in real time to avoid underlying risks, and for the early detection and treatment of diseases for home healthcare. Hydrogels with three-dimensional polymer networks and large amounts of water molecules are naturally flexible and stretchable. Functionalized hydrogels are intrinsically conductive, self-healing, self-adhesive, biocompatible, and room-temperature sensitive. Compared with traditional rigid vapor sensors, hydrogel-based gas and humidity sensors can directly fit human skin or clothing, and are more suitable for real-time monitoring of personal health and safety. In this review, current studies on hydrogel-based vapor sensors are investigated. The required properties and optimization methods of wearable hydrogel-based sensors are introduced. Subsequently, existing reports on the response mechanisms of hydrogel-based gas and humidity sensors are summarized. Related works on hydrogel-based vapor sensors for their application in personal health and safety monitoring are presented. Moreover, the potential of hydrogels in the field of vapor sensing is elucidated. Finally, the current research status, challenges, and future trends of hydrogel gas/humidity sensing are discussed.
Collapse
Affiliation(s)
- Yibing Luo
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Jianye Li
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Qiongling Ding
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Hao Wang
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Chuan Liu
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Jin Wu
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China.
| |
Collapse
|
29
|
Xia W, Lai G, Li Y, Zeng C, Sun C, Zhang P, Zhu G, Li L, Wu L. Photo-crosslinked adhesive hydrogel loaded with extracellular vesicles promoting hemostasis and liver regeneration. Front Bioeng Biotechnol 2023; 11:1170212. [PMID: 37234477 PMCID: PMC10208220 DOI: 10.3389/fbioe.2023.1170212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Hepatectomy is an effective surgical method for the treatment of liver diseases, but intraoperative bleeding and postoperative liver function recovery are still key issues. This study aims to develop a composite hydrogel dressing with excellent hemostatic properties, biocompatibility, and ability to promote liver cell regeneration. The modified gelatin matrix (GelMA, 10%) was mixed with equal volumes of sodium alginate-dopamine (Alg-DA) at concentrations of 0.5%, 1%, and 2%. Then a cross-linking agent (0.1%) was added to prepare different composite hydrogels under UV light, named GelMA/Alg-DA-0.5, GelMA/Alg-DA-1 and GelMA/Alg-DA-2, respectively. All the prepared hydrogel has a porous structure with a porosity greater than 65%, and could be stabilized in a gel state after being cross-linked by ultraviolet light. Physicochemical characterization showed that the elastic modulus, water absorption, adhesion, and compressibility of the composite hydrogels were improved with increasing Alg-DA content. Furthermore, the prepared hydrogel exhibits in vitro degradability, excellent biocompatibility, and good hemostatic function. Among all tested groups, the group of GelMA/Alg-DA-1 hydrogel performed the best. To further enhance its application potential in the field of liver regeneration, adipose-derived mesenchymal stem cell exosomes (AD-MSC-Exo) were loaded into GelMA/Alg-DA-1 hydrogel. Under the same conditions, GelMA/Alg-DA-1/Exo promoted cell proliferation and migration more effectively than hydrogels without extracellular vesicles. In conclusion, the prepared GelMA/Alg-DA-1 composite hydrogel loaded with AD-MSC-Exo has great application potential in liver wound hemostasis and liver regeneration.
Collapse
Affiliation(s)
- Wuzheng Xia
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guanzhi Lai
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yichuan Li
- Department of Hepatobiliary and Pancreatic Surgery, People’s Hospital of Guang’an City, West China-Guang’an Hospital, Sichuan University, Guang’an, China
| | - Cong Zeng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of General Practice, Hospital of South China Normal University, Guangzhou, China
| | - Chengjun Sun
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Pinzhe Zhang
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Guanghao Zhu
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Leping Li
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Linwei Wu
- Department of Organ Transplantation, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Affonso de Oliveira JF, Zhao Z, Xiang Y, Shin MD, Villaseñor KE, Deng X, Shukla S, Chen S, Steinmetz NF. COVID-19 vaccines based on viral nanoparticles displaying a conserved B-cell epitope show potent immunogenicity and a long-lasting antibody response. Front Microbiol 2023; 14:1117494. [PMID: 37152732 PMCID: PMC10157238 DOI: 10.3389/fmicb.2023.1117494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 05/09/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 sparked intensive research into the development of effective vaccines, 50 of which have been approved thus far, including the novel mRNA-based vaccines developed by Pfizer and Moderna. Although limiting the severity of the disease, the mRNA-based vaccines presented drawbacks, such as the cold chain requirement. Moreover, antibody levels generated by these vaccines decline significantly after 6 months. These vaccines deliver mRNA encoding the full-length spike (S) glycoprotein of SARS-CoV-2, but must be updated as new strains and variants of concern emerge, creating a demand for adjusted formulations and booster campaigns. To overcome these challenges, we have developed COVID-19 vaccine candidates based on the highly conserved SARS CoV-2, 809-826 B-cell peptide epitope (denoted 826) conjugated to cowpea mosaic virus (CPMV) nanoparticles and bacteriophage Qβ virus-like particles, both platforms have exceptional thermal stability and facilitate epitope delivery with inbuilt adjuvant activity. We evaluated two administration methods: subcutaneous injection and an implantable polymeric scaffold. Mice received a prime-boost regimen of 100 μg per dose (2 weeks apart) or a single dose of 200 μg administered as a liquid formulation, or a polymer implant. Antibody titers were evaluated longitudinally over 50 weeks. The vaccine candidates generally elicited an early Th2-biased immune response, which stimulates the production of SARS-CoV-2 neutralizing antibodies, followed by a switch to a Th1-biased response for most formulations. Exceptionally, vaccine candidate 826-CPMV (administered as prime-boost, soluble injection) elicited a balanced Th1/Th2 immune response, which is necessary to prevent pulmonary immunopathology associated with Th2 bias extremes. While the Qβ-based vaccine elicited overall higher antibody titers, the CPMV-induced antibodies had higher avidity. Regardless of the administration route and formulation, our vaccine candidates maintained high antibody titers for more than 50 weeks, confirming a potent and durable immune response against SARS-CoV-2 even after a single dose.
Collapse
Affiliation(s)
| | - Zhongchao Zhao
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Yi Xiang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Matthew D. Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | | | - Xinyi Deng
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Center for Engineering in Cancer, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
31
|
Meng R, Zhu H, Deng P, Li M, Ji Q, He H, Jin L, Wang B. Research progress on albumin-based hydrogels: Properties, preparation methods, types and its application for antitumor-drug delivery and tissue engineering. Front Bioeng Biotechnol 2023; 11:1137145. [PMID: 37113668 PMCID: PMC10127125 DOI: 10.3389/fbioe.2023.1137145] [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: 01/04/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Albumin is derived from blood plasma and is the most abundant protein in blood plasma, which has good mechanical properties, biocompatibility and degradability, so albumin is an ideal biomaterial for biomedical applications, and drug-carriers based on albumin can better reduce the cytotoxicity of drug. Currently, there are numerous reviews summarizing the research progress on drug-loaded albumin molecules or nanoparticles. In comparison, the study of albumin-based hydrogels is a relatively small area of research, and few articles have systematically summarized the research progress of albumin-based hydrogels, especially for drug delivery and tissue engineering. Thus, this review summarizes the functional features and preparation methods of albumin-based hydrogels, different types of albumin-based hydrogels and their applications in antitumor drugs, tissue regeneration engineering, etc. Also, potential directions for future research on albumin-based hydrogels are discussed.
Collapse
Affiliation(s)
- Run Meng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Huimin Zhu
- Sheyang County Comprehensive Inspection and Testing Center, Yancheng, China
| | - Peiying Deng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Minghui Li
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qingzhi Ji
- School of Pharmacy, Yancheng Teachers’ University, Yancheng, China
| | - Hao He
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
32
|
Salehi S, Naghib SM, Garshasbi HR, Ghorbanzadeh S, Zhang W. Smart stimuli-responsive injectable gels and hydrogels for drug delivery and tissue engineering applications: A review. Front Bioeng Biotechnol 2023; 11:1104126. [PMID: 36911200 PMCID: PMC9992555 DOI: 10.3389/fbioe.2023.1104126] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Hydrogels are widely used biomaterials in the delivery of therapeutic agents, including drugs, genes, proteins, etc., as well as tissue engineering, due to obvious properties such as biocompatibility and their similarity to natural body tissues. Some of these substances have the feature of injectability, which means that the substance is injected into the desired place in the solution state and then turns into the gel, which makes it possible to administer them from a way with a minimal amount of invasion and eliminate the need for surgery to implant pre-formed materials. Gelation can be caused by a stimulus and/or spontaneously. Suppose this induces due to the effect of one or many stimuli. In that case, the material in question is called stimuli-responsive because it responds to the surrounding conditions. In this context, we introduce the different stimuli that cause gelation and investigate the different mechanisms of the transformation of the solution into the gel in them. Also, we study special structures, such as nano gels or nanocomposite gels.
Collapse
Affiliation(s)
- Saba Salehi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, Iran University of Science and Technology (IUST), ACECR, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, Iran University of Science and Technology (IUST), ACECR, Tehran, Iran
| | - Hamid Reza Garshasbi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, Iran University of Science and Technology (IUST), ACECR, Tehran, Iran
| | - Sadegh Ghorbanzadeh
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Wei Zhang
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| |
Collapse
|
33
|
Liu W, Li Z, Wang Z, Huang Z, Sun C, Liu S, Jiang Y, Yang H. Functional System Based on Glycyrrhizic Acid Supramolecular Hydrogel: Toward Polymorph Control, Stabilization, and Controlled Release. ACS APPLIED MATERIALS & INTERFACES 2023; 15:7767-7776. [PMID: 36732699 DOI: 10.1021/acsami.2c19903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Developments of a drug delivery system (DDS) based on a natural supramolecular hydrogel have been of wide interest due to its biocompatibility, efficacy, and adjustable performance. However, a simple and efficient design of functional hydrogel DDS based on the templated interplay of gelator and model drug is still a challenge. In this work, natural glycyrrhetinic acid (GA) gel was selected as a carrier to encapsulate the model drug pyrazinamide (PZA). It was found that the carboxyl-amide interaction at the interface of gel-drug achieved polymorph control, stabilization, and pH-responsive release. Powder X-ray diffraction confirmed that the metastable γ form of PZA was obtained from the GA gel. Spectral analysis and molecular dynamics simulation showed that the protonation at the amide-O promoted the discretization of PZA molecules in solution, resulting in the polymorphism. Furthermore, the gel-drug interplay increased the stability of the γ form significantly from 2 days to 3 months by in situ encapsulation in the GA gel. In vitro release study indicated that the GA gel achieved targeted control release of PZA due to the pH-responsiveness property of GA. This work provides a promising option for hydrogel-based DDS design combined with polymorph control and stabilization.
Collapse
Affiliation(s)
- Weiqi Liu
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Zhiqiang Li
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Zixuan Wang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Ziyin Huang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Chenbo Sun
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Shiyuan Liu
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
| | - Yanbin Jiang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou510640, China
- School of Chemical Engineering, Guangdong University of Petrochemical Technology, Maoming525000, China
| | - Huaiyu Yang
- Department of Chemical Engineering, Loughborough University, LoughboroughLE11 3TU, Leicestershire, U.K
| |
Collapse
|
34
|
Khakpour E, Salehi S, Naghib SM, Ghorbanzadeh S, Zhang W. Graphene-based nanomaterials for stimuli-sensitive controlled delivery of therapeutic molecules. Front Bioeng Biotechnol 2023; 11:1129768. [PMID: 36845181 PMCID: PMC9947473 DOI: 10.3389/fbioe.2023.1129768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Stimuli-responsive drug delivery has attracted tremendous attention in the past decades. It provides a spatial- and temporal-controlled release in response to different triggers, thus enabling highly efficient drug delivery and minimizing drug side effects. Graphene-based nanomaterials have been broadly explored, and they show great potential in smart drug delivery due to their stimuli-responsive behavior and high loading capacity for an extended range of drug molecules. These characteristics are a result of high surface area, mechanical stability and chemical stability, and excellent optical, electrical, and thermal properties. Their great and infinite functionalization potential also allows them to be integrated into several types of polymers, macromolecules, or other nanoparticles, leading to the fabrication of novel nanocarriers with enhanced biocompatibility and trigger-sensitive properties. Thus, numerous studies have been dedicated to graphene modification and functionalization. In the current review, we introduce graphene derivatives and different graphene-based nanomaterials utilized in drug delivery and discuss the most important advances in their functionalization and modification. Also, their potential and progress in an intelligent drug release in response to different types of stimuli either endogenous (pH, redox conditions, and reactive oxygen species (ROS)) or exogenous (temperature, near-infrared (NIR) radiation, and electric field) will be debated.
Collapse
Affiliation(s)
- Elnaz Khakpour
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran
| | - Saba Salehi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran,*Correspondence: Seyed Morteza Naghib, ; Wei Zhang,
| | - Sadegh Ghorbanzadeh
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Wei Zhang
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China,*Correspondence: Seyed Morteza Naghib, ; Wei Zhang,
| |
Collapse
|
35
|
Pogostin BH, Saenz G, Cole CC, Euliano EM, Hartgerink JD, McHugh KJ. Dynamic Imine Bonding Facilitates Mannan Release from a Nanofibrous Peptide Hydrogel. Bioconjug Chem 2023; 34:193-203. [PMID: 36580277 PMCID: PMC10061233 DOI: 10.1021/acs.bioconjchem.2c00461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recently, there has been increased interest in using mannan as an immunomodulatory bioconjugate. Despite notable immunological and functional differences between the reduced (R-Man) and oxidized (O-Man) forms of mannan, little is known about the impact of mannan oxidation state on its in vivo persistence or its potential controlled release from biomaterials that may improve immunotherapeutic or prophylactic efficacy. Here, we investigate the impact of oxidation state on the in vitro and in vivo release of mannan from a biocompatible and immunostimulatory multidomain peptide hydrogel, K2(SL)6K2 (abbreviated as K2), that has been previously used for the controlled release of protein and small molecule payloads. We observed that O-Man released more slowly from K2 hydrogels in vitro than R-Man. In vivo, the clearance of O-Man from K2 hydrogels was slower than O-Man alone. We attributed the slower release rate to the formation of dynamic imine bonds between reactive aldehyde groups on O-Man and the lysine residues on K2. This imine interaction was also observed to improve K2 + O-Man hydrogel strength and shear recovery without significantly influencing secondary structure or peptide nanofiber formation. There were no observed differences in the in vivo release rates of O-Man loaded in K2, R-Man loaded in K2, and R-Man alone. These data suggest that, after subcutaneous injection, R-Man naturally persists longer in vivo than O-Man and minimally interacts with the peptide hydrogel. These results highlight a potentially critical, but previously unreported, difference in the in vivo behavior of O-Man and R-Man and demonstrate that K2 can be used to normalize the release of O-Man to that of R-Man. Further, since K2 itself is an adjuvant, a combination of O-Man and K2 could be used to enhance the immunostimulatory effects of O-Man for applications such as infectious disease vaccines and cancer immunotherapy.
Collapse
Affiliation(s)
- Brett H Pogostin
- Department of Bioengineering, Rice University, Houston, Texas77005, United States
| | - Gabriel Saenz
- Department of Chemistry, Rice University, Houston, Texas77005, United States
| | - Carson C Cole
- Department of Chemistry, Rice University, Houston, Texas77005, United States
| | - Erin M Euliano
- Department of Bioengineering, Rice University, Houston, Texas77005, United States
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, Texas77005, United States
- Department of Chemistry, Rice University, Houston, Texas77005, United States
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, Texas77005, United States
- Department of Chemistry, Rice University, Houston, Texas77005, United States
| |
Collapse
|
36
|
Kola P, Nagesh PKB, Roy PK, Deepak K, Reis RL, Kundu SC, Mandal M. Innovative nanotheranostics: Smart nanoparticles based approach to overcome breast cancer stem cells mediated chemo- and radioresistances. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1876. [PMID: 36600447 DOI: 10.1002/wnan.1876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Prithwish Kola
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rui Luis Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
37
|
Younas F, Zaman M, Aman W, Farooq U, Raja MAG, Amjad MW. Thiolated Polymeric Hydrogels for Biomedical Applications: A Review. Curr Pharm Des 2023; 29:3172-3186. [PMID: 37622704 DOI: 10.2174/1381612829666230825100859] [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: 03/23/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023]
Abstract
Hydrogels are a three-dimensional (3D) network of hydrophilic polymers. The physical and chemical crosslinking of polymeric chains maintains the structure of the hydrogels even when they are swollen in water. They can be modified with thiol by thiol epoxy, thiol-ene, thiol-disulfide, or thiol-one reactions. Their application as a matrix for protein and drug delivery, cellular immobilization, regenerative medicine, and scaffolds for tissue engineering was initiated in the early 21st century. This review focuses on the ingredients, classification techniques, and applications of hydrogels, types of thiolation by different thiol-reducing agents, along with their mechanisms. In this study, different applications for polymers used in thiolated hydrogels, including dextran, gelatin, polyethylene glycol (PEG), cyclodextrins, chitosan, hyaluronic acid, alginate, poloxamer, polygalacturonic acid, pectin, carrageenan gum, arabinoxylan, carboxymethyl cellulose (CMC), gellan gum, and polyvinyl alcohol (PVA) are reviewed.
Collapse
Affiliation(s)
- Farhan Younas
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | - Muhammad Zaman
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | - Waqar Aman
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | - Umer Farooq
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | | | - Muhammad Wahab Amjad
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, PA 15213, USA
| |
Collapse
|
38
|
Kumar D, Dua K, Tiwari S. Localized Delivery of Bioactives using Structured Liposomal Gels. Curr Pharm Des 2023; 29:3206-3220. [PMID: 37974442 DOI: 10.2174/0113816128263001231102053654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/03/2023] [Indexed: 11/19/2023]
Abstract
Liposomes have gained a lot of interest for drug delivery applications, and some of these preparations have been commercialized. These are formulated with biocompatible components and can be used for delivering a wide range of payloads differing in aqueous solubility and molecular weight. Liposome-based delivery approaches are limited mainly by two factors: (a) poor dispersion stability, and (b) pre-mature leakage of payloads. In this review, we have discussed the stabilization of liposomal vesicles by their entrapment in hydrogels. Studies reveal that such hydrogels can maintain the structural integrity of liposomes. Release of liposomes from the hydrogel network can be modulated through careful screening of matrix former and degree of its cross-linking. Accordingly, we have reviewed the approaches of stabilizing liposomal vesicles through entrapment in hydrogels. Application of liposome-embedded hydrogels has been reviewed in context of localized drug delivery. Our discussion is focussed on the delivery of bioactives to the skin. Such an approach appears alluring from the standpoint of minimizing the undesirable distribution of payload(s) the systemic circulation and off-target sites.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, New South Wales 2007, Australia
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| |
Collapse
|
39
|
Meissner S, Akepogu JH, Arnet SM, Dean MZ, Ji J, Wright G, Harland B, Raos B, Svirskis D, Thakur SS. Investigating the influence of ultrasound parameters on ibuprofen drug release from hydrogels. Drug Deliv Transl Res 2022; 13:1390-1404. [PMID: 36509966 DOI: 10.1007/s13346-022-01277-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Hydrogels are promising ultrasound-responsive drug delivery systems. In this study, we investigated how different ultrasound parameters affected drug release and structural integrity of self-healing hydrogels composed of alginate or poloxamers. The effects of amplitude and duty cycle at low frequency (24 kHz) ultrasound stimulation were first investigated using alginate hydrogels at 2% w/v and 2.5% w/v. Increasing ultrasound amplitude increased drug release from these gels, although high amplitudes caused large variations in release and damaged the gel structure. Increasing duty cycle also increased drug release, although a threshold was observed with the lower pulsed 50% duty cycle achieving similar levels of drug release to a continuous 100% duty cycle. Poloxamer-based hydrogels were also responsive to the optimised parameters at low frequency (24 kHz, 20% amplitude, 50% duty cycle for 30 s) and showed similar drug release results to a 2.5% w/v alginate hydrogel. Weight loss studies demonstrated that the 2% w/v alginate hydrogel underwent significant erosion following ultrasound application, whereas the 2.5% w/v alginate and the poloxamer gels were unaffected by application of the same parameters (24 kHz, 20% amplitude, 50% duty cycle for 30 s). The rheological properties of the hydrogels were also unaffected and the FTIR spectra remained unchanged after low frequency ultrasound stimulation (24 kHz, 20% amplitude, 50% duty cycle for 30 s). Finally, high-frequency ultrasound stimulation (1 MHz, 3 W.cm-2, 50% duty cycle) was also trialled; the alginate gels were less responsive to this frequency, while no statistically significant impact on drug release was observed from the poloxamer gels. This study demonstrates the importance of ultrasound parameters and polymer selection in designing ultrasound-responsive hydrogels.
Collapse
Affiliation(s)
- Svenja Meissner
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Jhansi H Akepogu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Samantha M Arnet
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mohammed Z Dean
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Jessie Ji
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Grace Wright
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Bruce Harland
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Brad Raos
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Sachin S Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
40
|
Zaszczyńska A, Niemczyk-Soczynska B, Sajkiewicz P. A Comprehensive Review of Electrospun Fibers, 3D-Printed Scaffolds, and Hydrogels for Cancer Therapies. Polymers (Basel) 2022; 14:5278. [PMID: 36501672 PMCID: PMC9736375 DOI: 10.3390/polym14235278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/09/2022] Open
Abstract
Anticancer therapies and regenerative medicine are being developed to destroy tumor cells, as well as remodel, replace, and support injured organs and tissues. Nowadays, a suitable three-dimensional structure of the scaffold and the type of cells used are crucial for creating bio-inspired organs and tissues. The materials used in medicine are made of non-degradable and degradable biomaterials and can serve as drug carriers. Developing flexible and properly targeted drug carrier systems is crucial for tissue engineering, regenerative medicine, and novel cancer treatment strategies. This review is focused on presenting innovative biomaterials, i.e., electrospun nanofibers, 3D-printed scaffolds, and hydrogels as a novel approach for anticancer treatments which are still under development and awaiting thorough optimization.
Collapse
Affiliation(s)
| | | | - Paweł Sajkiewicz
- Laboratory of Polymers & Biomaterials, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| |
Collapse
|
41
|
Vu ND, Crozzolo J, Guérinot A, Nicolaÿ R. Injectable and Self-Healing Thiazolidine-Crosslinked Hydrogels: Synthesis and Characterization. Macromolecules 2022. [DOI: 10.1021/acs.macromol.2c01714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Nam Duc Vu
- Molecular, Macromolecular Chemistry and Materials (C3M), ESPCI Paris, CNRS, PSL University, 10 rue Vauquelin, 75005 Paris, France
| | - Julien Crozzolo
- Molecular, Macromolecular Chemistry and Materials (C3M), ESPCI Paris, CNRS, PSL University, 10 rue Vauquelin, 75005 Paris, France
| | - Amandine Guérinot
- Molecular, Macromolecular Chemistry and Materials (C3M), ESPCI Paris, CNRS, PSL University, 10 rue Vauquelin, 75005 Paris, France
| | - Renaud Nicolaÿ
- Molecular, Macromolecular Chemistry and Materials (C3M), ESPCI Paris, CNRS, PSL University, 10 rue Vauquelin, 75005 Paris, France
| |
Collapse
|
42
|
Preparation and Properties of Egg White Dual Cross-Linked Hydrogel with Potential Application for Bone Tissue Engineering. Polymers (Basel) 2022; 14:polym14235116. [PMID: 36501519 PMCID: PMC9735576 DOI: 10.3390/polym14235116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022] Open
Abstract
In this study, an egg white dual cross-linked hydrogel was developed based on the principle that the external stimulus can denature proteins and cause them to aggregate, forming hydrogel. The sodium hydroxide was used to induce gelation of the egg white protein, subsequently introducing calcium ions to cross-link with protein chains, thereby producing a dual cross-linked hydrogel. The characteristics of the dual cross-linked hydrogels-including the secondary structure, stability, microstructure, swelling performance, texture properties, and biosafety-were investigated to determine the effects of calcium ion on the egg white hydrogel (EWG) and evaluate the potential application in the field of tissue engineering. Results showed that calcium ions could change the β-sheet content of the protein in EWG after soaking it in different concentrations of CaCl2 solution, leading to changes in the hydrogen bonds and the secondary structure of polypeptide chains. It was confirmed that calcium ions promoted the secondary cross-linking of the protein chain, which facilitated polypeptide folding and aggregation, resulting in enhanced stability of the egg white dual cross-linked hydrogel. Furthermore, the swelling capacity of the EWG decreased with increasing concentration of calcium ions, and the texture properties including hardness, cohesiveness and springiness of the hydrogels were improved. In addition, the calcium cross-linked EWG hydrogels exhibited biocompatibility and cell-surface adhesion in vitro. Hence, this work develops a versatile strategy to fabricate dual cross-linked protein hydrogel with biosafety and cell-surface adhesion, and both the strategy and calcium-egg white cross-linked hydrogels have potential for use in bone tissue engineering.
Collapse
|
43
|
Chafran L, Carfagno A, Altalhi A, Bishop B. Green Hydrogel Synthesis: Emphasis on Proteomics and Polymer Particle-Protein Interaction. Polymers (Basel) 2022; 14:4755. [PMID: 36365747 PMCID: PMC9656617 DOI: 10.3390/polym14214755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 08/26/2023] Open
Abstract
The field of drug discovery has seen significant progress in recent years. These advances drive the development of new technologies for testing compound's effectiveness, as well as their adverse effects on organs and tissues. As an auxiliary tool for drug discovery, smart biomaterials and biopolymers produced from biodegradable monomers allow the manufacture of multifunctional polymeric devices capable of acting as biosensors, of incorporating bioactives and biomolecules, or even mimicking organs and tissues through self-association and organization between cells and biopolymers. This review discusses in detail the use of natural monomers for the synthesis of hydrogels via green routes. The physical, chemical and morphological characteristics of these polymers are described, in addition to emphasizing polymer-particle-protein interactions and their application in proteomics studies. To highlight the diversity of green synthesis methodologies and the properties of the final hydrogels, applications in the areas of drug delivery, antibody interactions, cancer therapy, imaging and biomarker analysis are also discussed, as well as the use of hydrogels for the discovery of antimicrobial and antiviral peptides with therapeutic potential.
Collapse
Affiliation(s)
- Liana Chafran
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110 , USA
| | | | | | - Barney Bishop
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110 , USA
| |
Collapse
|
44
|
Oliveira CBP, Gomes V, Ferreira PMT, Martins JA, Jervis PJ. Peptide-Based Supramolecular Hydrogels as Drug Delivery Agents: Recent Advances. Gels 2022; 8:706. [PMID: 36354614 PMCID: PMC9689023 DOI: 10.3390/gels8110706] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 08/19/2023] Open
Abstract
Supramolecular peptide hydrogels have many important applications in biomedicine, including drug delivery applications for the sustained release of therapeutic molecules. Targeted and selective drug administration is often preferential to systemic drug delivery, as it can allow reduced doses and can avoid the toxicity and side-effects caused by off-target binding. New discoveries are continually being reported in this rapidly developing field. In this review, we report the latest developments in supramolecular peptide-based hydrogels for drug delivery, focusing primarily on discoveries that have been reported in the last four years (2018-present). We address clinical points, such as peptide self-assembly and drug release, mechanical properties in drug delivery, peptide functionalization, bioadhesive properties and drug delivery enhancement strategies, drug release profiles, and different hydrogel matrices for anticancer drug loading and release.
Collapse
Affiliation(s)
| | | | | | | | - Peter J. Jervis
- Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
45
|
Katopodi T, Petanidis S, Tsavlis D, Anestakis D, Charalampidis C, Chatziprodromidou I, Eskitzis P, Zarogoulidis P, Kosmidis C, Matthaios D, Porpodis K. Engineered multifunctional nanocarriers for controlled drug delivery in tumor immunotherapy. Front Oncol 2022; 12:1042125. [PMID: 36338748 PMCID: PMC9634039 DOI: 10.3389/fonc.2022.1042125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
The appearance of chemoresistance in cancer is a major issue. The main barriers to conventional tumor chemotherapy are undesirable toxic effects and multidrug resistance. Cancer nanotherapeutics were developed to get around the drawbacks of conventional chemotherapy. Through clinical evaluation of thoughtfully developed nano delivery systems, cancer nanotherapeutics have recently offered unmatched potential to comprehend and combat drug resistance and toxicity. In different design approaches, including passive targeting, active targeting, nanomedicine, and multimodal nanomedicine combination therapy, were successful in treating cancer in this situation. Even though cancer nanotherapy has achieved considerable technological development, tumor biology complexity and heterogeneity and a lack of full knowledge of nano-bio interactions remain important hurdles to future clinical translation and commercialization. The recent developments and advancements in cancer nanotherapeutics utilizing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are covered in this article. Additionally, an evaluation of different nanotherapeutics-based approaches to cancer treatment, such as tumor microenvironment targeted techniques, sophisticated delivery methods for the precise targeting of cancer stem cells, as well as an update on clinical studies are discussed. Lastly, the potential for cancer nanotherapeutics to overcome tumor relapse and the therapeutic effects and targeted efficacies of modern nanosystems are analyzed.
Collapse
Affiliation(s)
- Theodora Katopodi
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Savvas Petanidis
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Drosos Tsavlis
- Department of Medicine, Laboratory of Experimental Physiology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Doxakis Anestakis
- Department of Histology, Medical School, University of Cyprus, Nicosia, Cyprus
| | | | | | | | - Paul Zarogoulidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christoforos Kosmidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Konstantinos Porpodis
- Pulmonary Department-Oncology Unit, “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
46
|
Unger K, Anzengruber M, Coclite AM. Measurements of Temperature and Humidity Responsive Swelling of Thin Hydrogel Films by Interferometry in an Environmental Chamber. Polymers (Basel) 2022; 14:polym14193987. [PMID: 36235931 PMCID: PMC9573520 DOI: 10.3390/polym14193987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/14/2022] [Accepted: 09/18/2022] [Indexed: 11/23/2022] Open
Abstract
Thin film thermo-responsive hydrogels have become a huge interest in applications such as smart drug-delivery systems or sensor/actuator technology. So far, mostly, the response of such hydrogels has been measured only by varying the temperature in a liquid environment, but studies of the response towards humidity and temperature are rare because of experimental limitations. Often the swelling measurements are performed on samples placed on a stage that can be heated/cooled, while vapors enter the permeation chamber at their own temperature. This thermal difference leads to some uncertainties on the exact relative humidity to which the sample is exposed to. In this study, we explored the possibility of performing swelling measurements under thermal equilibrium by placing the sample and an interferometer, as a detector, in an environmental chamber and therefore exposing the smart hydrogel to adjustable temperatures and relative humidity conditions while measuring the hydrogel’s thin film thickness changes. As a case study, we used thin films of the thermo-responsive hydrogel, poly N-vinylcaprolactam deposited by initiated chemical vapor deposition (iCVD). Similar thin films were previously characterized by in situ ellipsometry while the sample was heated on a stage and exposed to humid air produced at room temperature. The comparison between the two measurement methods showed that while measurements in the presence of thermal gradients are limited mostly to low humidity, measurements in thermal equilibrium are restricted only by the operation limits of the used environmental chamber.
Collapse
|
47
|
Guo Y, Sun L, Wang Y, Wang Q, Jing D, Liu S. Nanomaterials based on thermosensitive polymer in biomedical field. Front Chem 2022; 10:946183. [PMID: 36212064 PMCID: PMC9532752 DOI: 10.3389/fchem.2022.946183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/30/2022] [Indexed: 11/27/2022] Open
Abstract
The progress of nanotechnology enables us to make use of the special properties of materials on the nanoscale and open up many new fields of biomedical research. Among them, thermosensitive nanomaterials stand out in many biomedical fields because of their “intelligent” behavior in response to temperature changes. However, this article mainly reviews the research progress of thermosensitive nanomaterials, which are popular in biomedical applications in recent years. Here, we simply classify the thermally responsive nanomaterials according to the types of polymers, focusing on the mechanisms of action and their advantages and potential. Finally, we deeply investigate the applications of thermosensitive nanomaterials in drug delivery, tissue engineering, sensing analysis, cell culture, 3D printing, and other fields and probe the current challenges and future development prospects of thermosensitive nanomaterials.
Collapse
Affiliation(s)
- Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- *Correspondence: Yingshu Guo,
| | - Li Sun
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Yajing Wang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Qianqian Wang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Dan Jing
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Shiwei Liu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| |
Collapse
|
48
|
Pasek-Allen JL, Kantesaria S, Gangwar L, Shao Q, Gao Z, Idiyatullin D, Han Z, Etheridge ML, Garwood M, Jagadeesan BD, Bischof JC. Injectable and Repeatable Inductive Heating of Iron Oxide Nanoparticle-Enhanced "PHIL" Embolic toward Tumor Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:41659-41670. [PMID: 36070361 DOI: 10.1021/acsami.2c05941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Deep-seated tumors of the liver, brain, and other organ systems often recur after initial surgical, chemotherapeutic, radiation, or focal treatments. Repeating these treatments is often invasive and traumatic. We propose an iron oxide nanoparticle (IONP)-enhanced precipitating hydrophobic injectable liquid (PHIL, MicroVention inc.) embolic as a localized dual treatment implant for nutrient deprivation and multiple repeatable thermal ablation. Following a single injection, multiple thermal treatments can be repeated as needed, based on monitoring of tumor growth/recurrence. Herein we show the ability to create an injectable stable PHIL-IONP solution, monitor deposition of the PHIL-IONP precipitate dispersion by μCT, and gauge the IONP distribution within the embolic by magnetic resonance imaging. Once precipitated, the implant could be heated to reach therapeutic temperatures >8 °C for thermal ablation (clinical temperature of ∼45 °C), in a model disk and a 3D tumor bed model. Heat output was not affected by physiological conditions, multiple heating sessions, or heating at intervals over a 1 month duration. Further, in ex vivo mice hind-limb tumors, we could noninvasively heat the embolic to an "ablative" temperature elevation of 17 °C (clinically 54 °C) in the first 5 min and maintain the temperature rise over +8 °C (clinically a temperature of 45 °C) for longer than 15 min.
Collapse
Affiliation(s)
- Jacqueline L Pasek-Allen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Saurin Kantesaria
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Lakshya Gangwar
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zhe Gao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Djaudat Idiyatullin
- Department of Radiology, Neurology and Neurosurgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael L Etheridge
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael Garwood
- Department of Radiology, Neurology and Neurosurgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Bharathi D Jagadeesan
- Department of Radiology, Neurology and Neurosurgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Radiology, Neurology and Neurosurgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
49
|
Niu Q, Sun Q, Bai R, Zhang Y, Zhuang Z, Zhang X, Xin T, Chen S, Han B. Progress of Nanomaterials-Based Photothermal Therapy for Oral Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:10428. [PMID: 36142341 PMCID: PMC9499573 DOI: 10.3390/ijms231810428] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/06/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the top 15 most prevalent cancers worldwide. However, the current treatment models for OSCC (e.g., surgery, chemotherapy, radiotherapy, and combination therapy) present several limitations: damage to adjacent healthy tissue, possible recurrence, low efficiency, and severe side effects. In this context, nanomaterial-based photothermal therapy (PTT) has attracted extensive research attention. This paper reviews the latest progress in the application of biological nanomaterials for PTT in OSCC. We divide photothermal nanomaterials into four categories (noble metal nanomaterials, carbon-based nanomaterials, metal compounds, and organic nanomaterials) and introduce each category in detail. We also mention in detail the drug delivery systems for PTT of OSCC and briefly summarize the applications of hydrogels, liposomes, and micelles. Finally, we note the challenges faced by the clinical application of PTT nanomaterials and the possibility of further improvement, providing direction for the future research of PTT in OSCC treatment.
Collapse
Affiliation(s)
- Qin Niu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Qiannan Sun
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Rushui Bai
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunfan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zimeng Zhuang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Xin Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianyi Xin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Si Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Bing Han
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
50
|
Luo Y, Pauer W, Luinstra GA. Fabrication of Thermo-Responsive Controllable Shape-Changing Hydrogel. Gels 2022; 8:531. [PMID: 36135243 PMCID: PMC9498808 DOI: 10.3390/gels8090531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/03/2022] Open
Abstract
Temperature response double network (DN) hydrogels comprising a network formed by polymerization of methacrylic acid (MA) modified PVA, N,N'-methylene bis(acrylamide), N-isopropylacryl amide (NIPAM), and one formed from crystalline polyvinyl alcohol (PVA) are prepared in a 3D printed tailor-made mold. The (PVA-MA)-g-PNIPAAm thermoset intermediate is formed in water by a radical, photo-initiated process, and in the presence of dissolved PVA polymers. A subsequent freezing-thawing sequence induces the crystallization of the PVA network, which forms a second network inside the thermoset NIPAM polymer. The prepared hydrogel is thermoresponsive by the phase transition of PNIPAAm segments (T ≈ 32 °C) and has good mechanical properties (tensile strength 1.23 MPa, compressive strength 1.47 MPa). Thermal cycling between room temperature at 40 or 50 °C shows the product converses from a virgin-state to a steady-state, which most likely involves the reorganization of PVA crystals. The swelling-deswelling cycles remain clear at a length change of about 13%.
Collapse
Affiliation(s)
| | | | - Gerrit A. Luinstra
- Institut für Technische und Makromolekulare Chemie, Universität Hamburg, 20146 Hamburg, Germany
| |
Collapse
|